WO2013090648A1 - Modified nucleoside, nucleotide, and nucleic acid compositions - Google Patents

Modified nucleoside, nucleotide, and nucleic acid compositions Download PDF

Info

Publication number
WO2013090648A1
WO2013090648A1 PCT/US2012/069610 US2012069610W WO2013090648A1 WO 2013090648 A1 WO2013090648 A1 WO 2013090648A1 US 2012069610 W US2012069610 W US 2012069610W WO 2013090648 A1 WO2013090648 A1 WO 2013090648A1
Authority
WO
WIPO (PCT)
Prior art keywords
optionally substituted
modified
lipid
nucleic acid
formulation
Prior art date
Application number
PCT/US2012/069610
Other languages
French (fr)
Inventor
Antonin De Fougerolles
Kristy M. WOOD
Sayda M. ELBASHIR
Noubar B. Afeyan
Pedro Valencia
Jason P. SCHRUM
Original Assignee
modeRNA Therapeutics
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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=48610363&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO2013090648(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority claimed from PCT/US2012/058519 external-priority patent/WO2013052523A1/en
Application filed by modeRNA Therapeutics filed Critical modeRNA Therapeutics
Priority to AU2012352180A priority Critical patent/AU2012352180A1/en
Priority to DE12858350.7T priority patent/DE12858350T1/en
Priority to ES12858350T priority patent/ES2923757T3/en
Priority to EP12858350.7A priority patent/EP2791160B1/en
Priority to SG11201402666WA priority patent/SG11201402666WA/en
Priority to LTEPPCT/US2012/069610T priority patent/LT2791160T/en
Priority to DK12858350.7T priority patent/DK2791160T3/en
Priority to KR1020147019601A priority patent/KR20140102759A/en
Priority to MX2014007233A priority patent/MX2014007233A/en
Priority to EP22159380.9A priority patent/EP4144378A1/en
Priority to HRP20220717TT priority patent/HRP20220717T1/en
Priority to CN201280069609.3A priority patent/CN104114572A/en
Priority to RS20220503A priority patent/RS63244B1/en
Priority to JP2014547454A priority patent/JP2015501844A/en
Priority to PL12858350T priority patent/PL2791160T3/en
Priority to CA2859387A priority patent/CA2859387A1/en
Priority to RU2014129004A priority patent/RU2649364C2/en
Priority to SI201232001T priority patent/SI2791160T1/en
Publication of WO2013090648A1 publication Critical patent/WO2013090648A1/en
Priority to US14/103,188 priority patent/US9220792B2/en
Priority to US14/104,568 priority patent/US9095552B2/en
Priority to US14/104,591 priority patent/US9233141B2/en
Priority to US14/104,585 priority patent/US20140193482A1/en
Priority to US14/104,556 priority patent/US9114113B2/en
Priority to US14/104,531 priority patent/US9107886B2/en
Priority to US14/105,214 priority patent/US20140171485A1/en
Priority to US14/105,210 priority patent/US20150044277A1/en
Priority to US14/105,217 priority patent/US20140255467A1/en
Priority to US14/105,221 priority patent/US20140255468A1/en
Priority to US14/105,224 priority patent/US9220755B2/en
Priority to US14/106,957 priority patent/US9050297B2/en
Priority to US14/107,029 priority patent/US20140113959A1/en
Priority to US14/106,988 priority patent/US9301993B2/en
Priority to US14/107,053 priority patent/US9216205B2/en
Priority to US14/107,079 priority patent/US9149506B2/en
Priority to US14/107,105 priority patent/US9255129B2/en
Priority to US14/170,744 priority patent/US20140148502A1/en
Priority to US14/170,910 priority patent/US20140155474A1/en
Priority to US14/170,903 priority patent/US20140155473A1/en
Priority to US14/171,119 priority patent/US20140179771A1/en
Priority to US14/170,751 priority patent/US20140200263A1/en
Priority to US14/170,914 priority patent/US20140155475A1/en
Priority to US14/170,897 priority patent/US20140155472A1/en
Priority to US14/170,747 priority patent/US20140194494A1/en
Priority to US14/171,226 priority patent/US9061059B2/en
Priority to US14/171,242 priority patent/US20140200264A1/en
Priority to US14/171,249 priority patent/US9089604B2/en
Priority to US14/171,235 priority patent/US20140206755A1/en
Priority to US14/280,867 priority patent/US20140275229A1/en
Priority to IL232749A priority patent/IL232749A0/en
Priority to ZA2014/03783A priority patent/ZA201403783B/en
Priority to HK15103589.4A priority patent/HK1203077A1/en
Priority to US14/694,357 priority patent/US9283287B2/en
Priority to US14/750,004 priority patent/US9828416B2/en
Priority to US14/975,141 priority patent/US9504734B2/en
Priority to US15/015,684 priority patent/US9587003B2/en
Priority to US15/060,707 priority patent/US9782462B2/en
Priority to US15/174,219 priority patent/US20160289674A1/en
Priority to US15/425,813 priority patent/US10501513B2/en
Priority to US15/440,523 priority patent/US20180086807A1/en
Priority to US16/665,985 priority patent/US20210115101A1/en
Priority to US16/707,998 priority patent/US20200247861A1/en
Priority to US16/860,121 priority patent/US20220273555A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/193Colony stimulating factors [CSF]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/7105Natural ribonucleic acids, i.e. containing only riboses attached to adenine, guanine, cytosine or uracil and having 3'-5' phosphodiester links
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/7115Nucleic acids or oligonucleotides having modified bases, i.e. other than adenine, guanine, cytosine, uracil or thymine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/46Hydrolases (3)
    • A61K38/48Hydrolases (3) acting on peptide bonds (3.4)
    • A61K38/482Serine endopeptidases (3.4.21)
    • A61K38/4833Thrombin (3.4.21.5)
    • 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
    • A61K48/0008Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'non-active' part of the composition delivered, e.g. wherein such 'non-active' part is not delivered simultaneously with the 'active' part of the composition
    • A61K48/0025Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'non-active' part of the composition delivered, e.g. wherein such 'non-active' part is not delivered simultaneously with the 'active' part of the composition wherein the non-active part clearly interacts with the delivered nucleic acid
    • A61K48/0033Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'non-active' part of the composition delivered, e.g. wherein such 'non-active' part is not delivered simultaneously with the 'active' part of the composition wherein the non-active part clearly interacts with the delivered nucleic acid the non-active part being non-polymeric
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/0008Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'non-active' part of the composition delivered, e.g. wherein such 'non-active' part is not delivered simultaneously with the 'active' part of the composition
    • A61K48/0025Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'non-active' part of the composition delivered, e.g. wherein such 'non-active' part is not delivered simultaneously with the 'active' part of the composition wherein the non-active part clearly interacts with the delivered nucleic acid
    • A61K48/0041Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'non-active' part of the composition delivered, e.g. wherein such 'non-active' part is not delivered simultaneously with the 'active' part of the composition wherein the non-active part clearly interacts with the delivered nucleic acid the non-active part being polymeric
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/005Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'active' part of the composition delivered, i.e. the nucleic acid delivered
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/005Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'active' part of the composition delivered, i.e. the nucleic acid delivered
    • A61K48/0066Manipulation of the nucleic acid to modify its expression pattern, e.g. enhance its duration of expression, achieved by the presence of particular introns in the delivered nucleic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • A61K9/0024Solid, semi-solid or solidifying implants, which are implanted or injected in body tissue
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0048Eye, e.g. artificial tears
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/127Liposomes
    • A61K9/1271Non-conventional liposomes, e.g. PEGylated liposomes, liposomes coated with polymers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/127Liposomes
    • A61K9/1271Non-conventional liposomes, e.g. PEGylated liposomes, liposomes coated with polymers
    • A61K9/1272Non-conventional liposomes, e.g. PEGylated liposomes, liposomes coated with polymers with substantial amounts of non-phosphatidyl, i.e. non-acylglycerophosphate, surfactants as bilayer-forming substances, e.g. cationic lipids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/16Agglomerates; Granulates; Microbeadlets ; Microspheres; Pellets; Solid products obtained by spray drying, spray freeze drying, spray congealing,(multiple) emulsion solvent evaporation or extraction
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/16Agglomerates; Granulates; Microbeadlets ; Microspheres; Pellets; Solid products obtained by spray drying, spray freeze drying, spray congealing,(multiple) emulsion solvent evaporation or extraction
    • A61K9/1605Excipients; Inactive ingredients
    • A61K9/1629Organic macromolecular compounds
    • A61K9/1641Organic macromolecular compounds obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyethylene glycol, poloxamers
    • A61K9/1647Polyesters, e.g. poly(lactide-co-glycolide)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/53Colony-stimulating factor [CSF]
    • C07K14/535Granulocyte CSF; Granulocyte-macrophage CSF
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2/00Peptides of undefined number of amino acids; Derivatives 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
    • 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
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/117Nucleic acids having immunomodulatory properties, e.g. containing CpG-motifs
    • 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
    • C12N15/67General methods for enhancing the expression
    • 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
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • C12N15/88Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation using microencapsulation, e.g. using amphiphile liposome vesicle
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12PFERMENTATION OR ENZYME-USING PROCESSES TO SYNTHESISE A DESIRED CHEMICAL COMPOUND OR COMPOSITION OR TO SEPARATE OPTICAL ISOMERS FROM A RACEMIC MIXTURE
    • C12P21/00Preparation of peptides or proteins
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12PFERMENTATION OR ENZYME-USING PROCESSES TO SYNTHESISE A DESIRED CHEMICAL COMPOUND OR COMPOSITION OR TO SEPARATE OPTICAL ISOMERS FROM A RACEMIC MIXTURE
    • C12P21/00Preparation of peptides or proteins
    • C12P21/02Preparation of peptides or proteins having a known sequence of two or more amino acids, e.g. glutathione

Definitions

  • exogenous unmodified nucleic acid molecules particularly viral nucleic acids
  • IFN interferon
  • a nucleic acid e.g., a ribonucleic acid (RNA)
  • RNA ribonucleic acid
  • formulation compositions comprising a delivery agent that can effectively facilitate the in vivo delivery of nucleic acids to targeted cells without generating an innate immune response.
  • the present disclosure provides, inter alia, formulation compositions comprising modified nucleic acid molecules which may encode a protein, a protein precursor, or a partially or fully processed form of the protein or a protein precursor.
  • the formulation compositions may further include a modified nucleic acid molecule and a delivery agent.
  • the present invention further provides nucleic acids useful for encoding polypeptides capable of modulating a cell's function and/or activity.
  • a method of producing a polypeptide of interest in a mammalian cell or tissue comprises contacting the mammalian cell or tissue with a formulation comprising a modified mRNA encoding a polypeptide of interest.
  • the formulation may be, but is not limited to, nanoparticles, poly(lactic-co-glycolic acid)(PLGA) microspheres, lipidoids, lipoplex, liposome, polymers, carbohydrates (including simple sugars), cationic lipids, fibrin gel, fibrin hydrogel, fibrin glue, fibrin sealant, fibrinogen, thrombin, rapidly eliminated lipid nanoparticles (reLNPs) and combinations thereof.
  • the modified mRNA may comprise a purified IVT transcript.
  • the formulation comprising the modified mRNA is a nanoparticle which may comprise at least one lipid.
  • the lipid may be selected from, but is not limited to, DLin- DMA, DLin-K-DMA, 98N12-5, C12-200, DLin-MC3 -DMA, DLin-KC2-DMA, DODMA, PLGA, PEG, PEG-DMG and PEGylated lipids.
  • the lipid may be a cationic lipid such as, but not limited to, DLin-DMA, DLin-D-DMA, DLin-MC3 -DMA, DLin-KC2-DMA and DODMA.
  • the lipid to modified mRNA ration in the formulation may be between 10: 1 and 30: 10.
  • the mean size of the nanoparticle formulation may comprise the modified mRNA between 60 and 225 nm.
  • the PDI of the nanoparticle formulation comprising the modified mRNA is between 0.03 and 0.15.
  • the zeta potential of the lipid may be from -10 to +10 at a pH of 7.4
  • the formulations of modified mRNA may comprise a fusogenic lipid, cholesterol and a PEG lipid.
  • the formulation may have a molar ratio 50: 10:38.5:1.5-3.0 (cationic lipid:fusogenic lipid: cholesterol: PEG lipid).
  • the PEG lipid may be selected from, but is not limited to PEG-c- DOMG, PEG-DMG.
  • the fusogenic lipid may be DSPC.
  • the mammalian cell or tissue may be contacted using a device such as, but not limited to, a syringe pump, internal osmotic pump and external osmotic pump.
  • the formulation of modified mR A may be a PLGA microsphere which may be between 4 and 20 ⁇ in size.
  • the modified mRNA may be released from the formulation at less than 50% in a 48 hour time period.
  • the PLGA microsphere formulation may be stable in serum. Stability may be determined relative to unformulated modified mRNA in 90%.
  • the loading weight percent of the modified mRNA PLGA microsphere may be at least 0.05%, at least 0.1%, at least 0.2%, at least 0.3%, at least 0.4% or at least 0.5%.
  • the encapsulation efficiency of the modified mRNA in the PLGA microsphere may be at least 50%, at least 70%, at least 90% or at least 97%.
  • a lipid nanoparticle of the present invention may be formulated in a sealant such as, but not limited to, a fibrin sealant.
  • the mammalian cells or tissues may be contacted by a route of administration such as, but not limited to, intravenous, intramuscular, intravitreal, intrathecal, intratumoral, pulmonary and subcutaneous.
  • the mammalian cells or tissues may be contacted using a split dosing schedule.
  • the mammalian cell or tissue may be contacted by injection.
  • the injection may be made to tissue selected from the group consisting of intradermal space, epidermis, subcutaneous tissue and muscle.
  • the polypeptide of interest may be produced in the cell or tissue in a location systemic from the location of contacting.
  • the polypeptide of interest may be detectable in serum for up to 72 hours after contacting.
  • the level of the polypeptide of interest can be higher than the levels prior to dosing.
  • the level of the polypeptide of interest may be greater in the serum of female subjects than in the serum of male subjects.
  • the formulation of modified mRNA may comprise more than one modified mRNA.
  • the formulation may have two or three modified mRNA.
  • the formulation comprising the modified mRNA may comprise a rapidly eliminated lipid nanoparticle (reLNP) which may comprise a reLNP lipid, fusogenic lipid, cholesterol and a PEG lipid at a molar ratio of 50: 10: 38.5: 1.5 (reLNP lipid:fusogenic lipid: cholesterol: PEG lipid).
  • the fusogenic lipid may be DSPC and the PEG lipid may be PEG-c-DOMG.
  • the reLNP lipid may be DLin-DMA with an internal or terminal ester or DLin-MC3-DMA with an internal or terminal ester.
  • the total lipid to modified mRNA weight ration may be between 10: 1 and 30: 1.
  • the formulation comprising modified mRNA may comprise a fibrin sealant.
  • the formulation comprising modified mRNA may comprise a lipidoid where the lipid is selected from the group consisting of CI 2-200 and 98N12-5.
  • the formulation comprising modified mRNA may include a polymer.
  • the polymer may be coated, covered, surrounded, enclosed or comprise a layer of a hydrogel or surgical sealant.
  • the polymer may be selected from the group consisting of PLGA, ethylene vinyl acetate, poloxamer and GELSITE®.
  • a polypeptide of interest may be produced in a mammalian cell or tissue by contacting the mammalian cell or tissue with a buffer formulation comprising a modified mRNA encoding the polypeptide of interest.
  • the buffer formulation may be selected from, but is not limited to, slaine, phosphate buffered saline and Ringer's lactate.
  • the buffer formulation may comprise a calcium concentration of between 1 to 10 mM.
  • the modified mRNA in the buffer formulation may comprise a purified IVT transcript.
  • a pharmacologic effect in a primate may be produced by contacting the primate with a composition comprising a formulated modified mRNA encoding a polypeptide of interest.
  • the modified mRNA may comprise a purified IVT transcript and/or may be formulated in nanoparticles, poly(lactic-co-glycolic acid)(PLGA) microspheres, lipidoids, lipoplex, liposome, polymers, carbohydrates (including simple sugars), cationic lipids, fibrin gel, fibrin hydrogel, fibrin glue, fibrin sealant, fibrinogen, thrombin, rapidly eliminated lipid nanoparticles (reLNPs) and combinations thereof.
  • the pharmacological effect may be greater than the pharmacologic effect associated with a therapeutic agent and/or composition known to produce said pharmacologic effect.
  • the composition may comprise a formulated or unformulated modified mRNA.
  • the pharmacologic effect may result in a therapeutically effective outcome of a disease, disorder, condition or infection. Such therapeutically effective outcome may include, but is not limited to, treatment, improvement of one or more symptoms, diagnosis, prevention, and delay of onset.
  • the pharmacologic effect may include, but is not limited to, change in cell count, alteration in serum chemistry, alteration of enzyme activity, increase in hemoglobin, and increase in hematocrit.
  • the present disclosure provides a formulation composition which comprises a modified nucleic acid molecule and a delivery agent.
  • the modified nucleic acid molecule may be selected from the group consisting of DNA, complimentary DNA (cDNA), RNA, messenger RNA (mRNA), RNAi-inducing agents, RNAi agents, siRNA, shRNA, miRNA, antisense R A, ribozymes, catalytic DNA, R A that induce triple helix formation, aptamers, vectors and combinations thereof. If the modified nucleic acid molecule is mR A the mR A may be derived from cDNA.
  • the modified nucleic acid molecule may comprise at least one modification and a translatable region. In some instances, the modified nucleic acid comprises at least two modifications and a translatable region.
  • the modification may be located on the backbone and/or a nucleoside of the nucleic acid molecule. The modification may be located on both a nucleoside and a backbone linkage.
  • a modification may be located on the backbone linkage of the modified nucleic acid molecule.
  • the backbone linkage may be modified by replacing of one or more oxygen atoms.
  • the modification of the backbone linkage may comprise replacing at least one phosphodiester linkage with a phosphorothioate linkage.
  • a modification may be located on a nucleoside of the modified nucleic acid molecule.
  • the modification on the nucleoside may be located on the sugar of said nucleoside.
  • the modification of the nucleoside may occur at the 2' position on the nucleoside.
  • the nucleoside modification may include a compound selected from the group consisting of pyridin-4-one ribonucleoside, 5-aza-uridine, 2-thio-5-aza-uridine, 2-thiouridine, 4-thio- pseudouridine, 2-thio-pseudouridine, 5-hydroxyuridine, 3-methyluridine, 5-carboxymethyl-uridine, 1-carboxymethyl-pseudouridine, 5-propynyl-uridine, 1-propynyl-pseudouridine, 5- taurinomethyluridine, 1-taurinomethyl-pseudouridine, 5-taurinomethyl-2-thio-uridine, 1- taurinomethyl-4-thio-uridine, 5-methyl-uridine, 1-methyl-pseudouridine, 4-thio-l-methyl- pseudouridine, 2-thio- 1 -methyl-pseudouridine, 1 -methyl- 1 -deaza-pseudouridine,
  • a modification may be located on a nucleobase of the modified nucleic acid molecule.
  • the modification on the nucleobase may be selected from the group consisting of cytosine, guanine, adenine, thymine and uracil.
  • the modification on the nucleobase may be selected from the group consisting of deaza-adenosine and deaza-guanosine, and the linker may be attached at a C-7 or C-8 position of said deaza-adenosine or deaza-guanosine.
  • the modified nucleobase may be selected from the group consisting of cytosine and uracil, and the linker may be attached to the modified nucleobase at an N-3 or C-5 position.
  • the linker attached to the nucleobase may be selected from the group consisting of diethylene glycol, dipropylene glycol, triethylene glycol, tripropylene glycol, tetraethylene glycol, tetraethylene glycol, divalent alkyl, alkenyl, alkynyl moiety, ester, amide, and ether moiety.
  • two modifications of the nucleic acid molecule may be located on nucleosides of the modified nucleic acid molecule.
  • the modified nucleosides may be selected from 5-methylcytosine and pseudouridine.
  • two modifications of the modified nucleic acid molecule may be located on a nucleotide or a nucleoside.
  • the present disclosure provides a formulation comprising a nucleic acid molecule such as, but not limited to, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 9 and SEQ ID NO: 10 and a delivery agent.
  • the nucleic acid molecule may comprise a polyA tail about 160 nucleotides in length.
  • the nucleic acid molecule may comprise at least one 5' terminal cap such as, but not limited to, CapO, Capl, ARC A, inosine, Nl- methyl-guanosine, 2'fluoro-guanosine, 7-deaza-guanosine, 8-oxo-guanosine, 2-amino-guanosine, LNA-guanosine, and 2-azido-guanosine.
  • 5' terminal cap such as, but not limited to, CapO, Capl, ARC A, inosine, Nl- methyl-guanosine, 2'fluoro-guanosine, 7-deaza-guanosine, 8-oxo-guanosine, 2-amino-guanosine, LNA-guanosine, and 2-azido-guanosine.
  • the present disclosure provides a nucleic acid of SEQ ID NO: 6, a 5' terminal cap which is Capl, a poly A tail of approximately 160 nucleotides in length and a delivery agent.
  • the present disclosure provides a nucleic acid of SEQ ID NO: 7, a 5' terminal cap which is Capl, a poly A tail of approximately 160 nucleotides in length and a delivery agent.
  • the present disclosure provides a nucleic acid of SEQ ID NO: 9, a 5' terminal cap which is Capl, a poly A tail of approximately 160 nucleotides in length and a delivery agent.
  • the present disclosure provides a nucleic acid of SEQ ID NO: 10, a 5' terminal cap which is Capl, a poly A tail of approximately 160 nucleotides in length and a delivery agent.
  • the delivery agent comprises at least one method to improve delivery selected from the group consisting of lipidoids, liposomes, lipid nanoparticles, rapidly eliminated lipid nanoparticles (reLNPs), polymers, lipoplexes, peptides, proteins, hydrogels, sealants, chemical modifications, conjugation, cells and enhancers.
  • lipidoids liposomes
  • lipid nanoparticles rapidly eliminated lipid nanoparticles (reLNPs)
  • reLNPs rapidly eliminated lipid nanoparticles
  • polymers lipoplexes
  • peptides proteins
  • hydrogels hydrogels
  • sealants chemical modifications, conjugation, cells and enhancers.
  • the lipidoid, lipid nanoparticle and rapidly eliminated lipid nanoparticles which may be used as a delivery agent may include a lipid which may be selected from the group consisting of C12-200, MD1, 98N12-5, DLin-DMA, DLin-K-DMA, DLin-KC2-DMA, DLin-MC3 -DMA, PLGA, PEG, PEG-DMG, PEGylated lipids and analogs thereof.
  • the rapidly eliminated lipid nanoparticle may have an ester linkage at the terminal end of the lipid chain, or an ester linkage may be an internal linkage located to the right or left of a saturated carbon in the lipid chain.
  • the rapidly eliminated lipid nanoparticle which may be used as a delivery agent may be, but is not limited to, DLin-MC3-DMA and DLin-DMA.
  • the lipid nanoparticle may comprise PEG and at least one component such as, but not limited to, cholesterol, cationic lipid and fusogenic lipid.
  • the lipid nanoparticle may comprise at least one of a PEG, cholesterol, cationic lipid and fusogenic lipid.
  • the fusogenic lipid is disteroylphophatidyl choline (DSPC).
  • the PEG lipid is PEG-DMG.
  • the cationic lipid may be, but not limited to, DLin-DMA, DLin-MC3 -DM A, C12-200, 98N12-5 and DLin-KC2- DMA.
  • the lipid nanoparticle composition may comprise 50 mol% cationic lipid, 10 mol% DSPC, 1.5-3.0 mol% PEG and 37-38.5 mol% cholesterol.
  • a modified nucleic acid may be formulated with PLGA to form a sustained release formulation.
  • a modified nucleic acid may be formulated with PLGA and other active and/or inactive components to form a sustained release formulation.
  • the modified nucleic acid molecule may include, but is not limited to, SEQ ID NO: 9 and SEQ ID NO: 10.
  • a sustained release formulation may comprise a sustained release microsphere.
  • the sustained release microsphere may be about 10 to about 50 um in diameter.
  • the sustained release microsphere may contain about 0.001 to about 1.0 weight percent of at least one modified nucleic acid molecule.
  • the modified nucleic acids of the present invention may include at least one stop codon before the 3' untranslated region (UTR).
  • the stop codon may be selected from TGA, TAA and TAG.
  • the modified nucleic acids of the present invention include the stop codon TGA and one additional stop codon.
  • the addition stop codon may be TAA.
  • the modified nucleic acid of the present invention includes three stop codons.
  • the present disclosure provides a controlled release formulation comprising a modified nucleic acid which may encode a polypeptide of interest.
  • the modified nucleic acid may be encapsulated or substantially encapsulated in a delivery agent.
  • the delivery agent may be coated, covered, surrounded, enclosed or comprise a layer of polymer, hydrogel and/or surgical sealant.
  • the controlled release formulation may comprise a second layer of polymer, hydrogel and/or surgical sealant.
  • the delivery agent of the controlled release formulation may include, but is not limited to, lipidoids, liposomes, lipid nanoparticles, rapidly eliminated lipid nanoparticles, lipoplexes and self-assembled lipid nanoparticles.
  • the polymer which may be used in the controlled release formulation may include, but is not limited to, PLGA, ethylene vinyl acetate, poloxamer and GELSITE®.
  • the surgical sealant which may be used in the controlled release formulation may include, but is not limited to, fibrinogen polymers, TISSEELL®, PEG-based sealants and COSEAL®.
  • the delivery agent of the controlled release formulation comprises a lipid nanoparticle or a rapidly eliminated lipid nanoparticle delivery agent.
  • the lipid nanoparticle or rapidly eliminated lipid nanoparticle may be coated, substantially coated, covered, substantially covered, surrounded, substanitally surrounded, enclosed, substantially enclosed or comprises a layer of polymer, hydrogel and/or surgical sealant.
  • the delivery agent may be a lipid nanoparticle which may be coated, substantially coated, covered, substantially covered, surrounded, substantially surrounded, enclosed, substantially enclosed or comprises a layer of PLGA.
  • FIG. 1 illustrates lipid structures in the prior art useful in the present invention. Shown are the structures for 98N12-5 (TETA5-LAP), DLin-DMA, DLin-K-DMA (2,2-Dilinoleyl-4- dimethylaminomethyl-[l,3]-dioxolane), DLin-KC2-DMA, DLin-MC3-DMA and C12-200.
  • FIG. 2 is a representative plasmid useful in the IVT reactions taught herein.
  • the plasmid contains Insert 64818, designed by the instant inventors.
  • FIG. 3 is a gel profile of modified mR A encapsulated in PLGA microspheres. DETAILED DESCRIPTION
  • nucleic acids and proteins of the present disclosure are capable of reducing the innate immune activity of a population of cells into which they are introduced, thus increasing the efficiency of protein production in that cell population. Further, one or more additional advantageous activities and/or properties of the nucleic acids and proteins of the present disclosure are described herein.
  • kits for treating a subject having or being suspected of having a disease, disorder and/or condition comprising administering to a subject in need of such treatment a composition described herein in an amount sufficient to treat the disease, disorder and/or condition.
  • the present disclosure provides nucleic acids, including RNA such as mRNA, which contain one or more modified nucleosides or nucleotides (termed “modified nucleic acid molecules,” “modified mRNA” or “modified mRNA molecules”) as described herein.
  • modified nucleic acid molecules of the present invention may have useful properties including, but not limited to, a significant decrease in or a lack of a substantial induction of the innate immune response of a cell into which the modified mRNA is introduced.
  • the modified nucleic acid molecules may also exhibit enhanced efficiency of protein production, intracellular retention of nucleic acids, and viability of contacted cells, as well as having reduced immunogenicity as compared to unmodified nucleic acid molecules.
  • modified nucleic acid molecules containing a translatable region and one, two, or more than two different nucleoside modifications
  • Exemplary nucleic acids for use in this disclosure include ribonucleic acids (RNA), deoxyribonucleic acids (DNAs), threose nucleic acids (TNAs), glycol nucleic acids (GNAs), locked nucleic acids (LNAs) or a hybrid thereof.
  • the modified nucleic acid molecules include messenger RNA (mRNA).
  • mRNA messenger RNA
  • the modified nucleic acid molecules of the present disclosure may not substantially induce an innate immune response of a cell into which the modified mRNA is introduced.
  • the modified nucleic acid molecule may exhibit reduced degradation, as compared to a nucleic acid that has not been modified, in a cell where the modified nucleic acid molecule is introduced.
  • nucleic acid includes any compound and/or substance that is or can be incorporated into an oligonucleotide chain.
  • exemplary nucleic acids for use in accordance with the present disclosure include, but are not limited to, one or more of DNA, cDNA, RNA including messenger RNA (mRNA), hybrids thereof, RNAi-inducing agents, RNAi agents, siRNA, shRNA, miRNA, antisense RNA, ribozymes, catalytic DNA, RNA that induce triple helix formation, aptamers, vectors and the like.
  • the present disclosure provides a modified nucleic acid molecule containing a degradation domain, which is capable of being acted on in a directed manner within a cell.
  • the modified nucleic acid molecules may be chemically modified on the sugar, nucleobase (e.g., in the 5 ' position of the nucleobase), or phosphate backbone (e.g., replacing the phosphate with another moiety such as a thiophospate).
  • the modification may result in a disruption of a major groove binding partner interaction, which may contribute to an innate immune response.
  • the formulation composition when administered to a subject, can result in improved bioavailability, therapeutic window, or volume of distribution of the modified nucleic acid molecule relative to administration of the modified nucleic acid molecule without the incorporation of the delivery agent.
  • the modified nucleosides and nucleotides of the modified nucleic acid molecules of the present invention may be synthesized using the O-protected compounds described in International Pub. No. WO2012138530, the contents of which is herein incorporated by reference in its entirety.
  • the modified nucleic acid molecule may comprise mRNA.
  • the modified mRNA may be derived from cDNA.
  • mmRNA may comprise at least two nucleoside modifications.
  • the nucleoside modifications may be selected from 5-methylcytosine and pseudouridine. In another embodiment, at least one of the nucleoside modifications is not 5-methylcytosine and/or
  • the delivery agent may comprise formulations allowing for localized and systemic delivery of mmRNA.
  • the formulations of the modified nucleic acids molecules and/or mmR A may be selected from, but are not limited to, lipidoids, liposomes and lipid nanoparticles, rapidly eliminated lipid nanoparticles, polymers, lipoplexes, peptides and proteins, at least one chemical modification and conjugation, enhancers, and/or cells.
  • the modified nucleic acid molecules of the present invention may include at least two stop codons before the 3' untranslated region (UTR).
  • the stop codon may be selected from TGA, TAA and TAG.
  • the nucleic acids of the present invention include the stop codon TGA and one additional stop codon.
  • the addition stop codon may be TAA.
  • the modified nucleic acid molecules may comprise three stop codons.
  • nucleic acid is optional in a modified nucleic acid molecule but these components may be beneficial in some embodiments.
  • Untranslated regions (UTRs) of a gene are transcribed but not translated.
  • the 5' UTR starts at the transcription start site and continues to the start codon but does not include the start codon; whereas, the 3' UTR starts immediately following a stop codon and continues until the
  • UTR transcriptional termination signal.
  • the regulatory features of a UTR can be incorporated into the modified mRNA molecules of the present invention to enhance the stability of the molecule.
  • the specific features can also be incorporated to ensure controlled down-regulation of the transcript in case they are misdirected to undesired organs sites. 5' UTR and Translation Initiation
  • Natural 5' UTRs bear features which play roles in for translation initiation. They harbor signatures like Kozak sequences which are commonly known to be involved in the process by which the ribosome initiates translation of many genes. Kozak sequences have the consensus
  • CCR(A/G)CCAUGG (SEQ ID NO: 1), where R is a purine (adenine or guanine) three bases upstream of the start codon (AUG), which is followed by another 'G'.
  • R is a purine (adenine or guanine) three bases upstream of the start codon (AUG), which is followed by another 'G'.
  • 5' UTR also have been known to form secondary structures which are involved in elongation factor binding.
  • modified mRNA molecules of the invention By engineering the features typically found in abundantly expressed genes of specific target organs, one can enhance the stability and protein production of the modified mRNA molecules of the invention.
  • introduction of 5' UTR of liver-expressed mRNA such as albumin, serum amyloid A, Apolipoprotein A/B/E, transferrin, alpha fetoprotein, erythropoietin, or Factor VIII, could be used to enhance expression of a modified nucleic acid molecule, such as a mmRNA, in hepatic cell lines or liver.
  • tissue-specific mRNA to improve expression in that tissue is possible for muscle (MyoD, Myosin, Myoglobin, Myogenin, Herculin), for endothelial cells (Tie-1, CD36), for myeloid cells (C/EBP, AML1, G-CSF, GM-CSF, CD1 lb, MSR, Fr-1, i-NOS), for leukocytes (CD45, CD18), for adipose tissue (CD36, GLUT4, ACRP30, adiponectin) and for lung epithelial cells (SP-A/B/C/D).
  • non-UTR sequences may be incorporated into the 5' (or 3' UTR) UTRs of the modified nucleic acid molecules of the present invention.
  • introns or portions of introns sequences may be incorporated into the flanking regions of the modified mRNA of the invention. Incorporation of intronic sequences may increase protein production as well as mRNA levels.
  • AU rich elements can be separated into three classes (Chen et al, 1995): Class I AREs contain several dispersed copies of an AUUUA motif within U-rich regions. C-Myc and MyoD contain class I AREs. Class II AREs possess two or more overlapping UUAUUUA(U/A)(U/A) (SEQ ID NO: 2) nonamers. Molecules containing this type of AREs include GM-CSF and TNF-a. Class III ARES are less well defined.
  • AREs 3' UTR AU rich elements
  • AREs 3' UTR AU rich elements
  • one or more copies of an ARE can be introduced to make modified mRNA of the invention less stable and thereby curtail translation and decrease production of the resultant protein.
  • AREs can be identified and removed or mutated to increase the intracellular stability and thus increase translation and production of the resultant protein.
  • Transfection experiments can be conducted in relevant cell lines, using modified mRNAof the invention and protein production can be assayed at various time points post-transfection.
  • cells can be transfected with different ARE-engineering molecules and by using an ELISA kit to the relevant protein and assaying protein produced at 6 hours, 12 hours, 24 hours, 48 hours, and 7 days post- transfection.
  • microRNAs are 19-25 nucleotide long noncoding RNAs that bind to the 3' UTR of nucleic acid molecules and down-regulate gene expression either by reducing nucleic acid molecule stability or by inhibiting translation.
  • the modified mRNA of the invention may comprise one or more microRNA target sequences, microRNA sequences, or microRNA seeds. Such sequences may correspond to any known microRNA such as those taught in US Publication
  • a microRNA sequence comprises a "seed" region, i.e., a sequence in the region of positions 2-8 of the mature microRNA, which sequence has perfect Watson-Crick complementarity to the miRNA target sequence.
  • a microRNA seed may comprise positions 2-8 or 2-7 of the mature microRNA.
  • a microRNA seed may comprise 7 nucleotides (e.g., nucleotides 2-8 of the mature microRNA), wherein the seed-complementary site in the corresponding miRNA target is flanked by an adenine (A) opposed to microRNA position 1.
  • a microRNA seed may comprise 6 nucleotides (e.g., nucleotides 2-7 of the mature microRNA), wherein the seed-complementary site in the corresponding miRNA target is flanked byan adenine (A) opposed to microRNA position 1.
  • A an adenine
  • the bases of the microRNA seed have complete complementarity with the target sequence.
  • microRNA target sequences By engineering microRNA target sequences into the 3 'UTR of modified mRNA of the invention one can target the molecule for degradation or reduced translation, provided the microRNA in question is available. This process will reduce the hazard of off target effects upon nucleic acid molecule delivery. Identification of microRNA, microRNA target regions, and their expression patterns and role in biology have been reported (Bonauer et al, Curr Drug Targets 2010 11 :943-949; Anand and Cheresh Curr Opin Hematol 2011 18 : 171 - 176; Contreras and Rao Leukemia 2012 26:404-413 (2011 Dec 20.
  • the modified nucleic acid molecule is a modified mRNA and is not intended to be delivered to the liver but ends up there, then miR-122, a microRNA abundant in liver, can inhibit the expression of the gene of interest if one or multiple target sites of miR-122 are engineered into the 3' UTR of the modified mRNA.
  • Introduction of one or multiple binding sites for different microRNA can be engineered to further decrease the longevity, stability, and protein translation of a modified nucleic acid molecule and/or modified mRNA.
  • microRNA site refers to a microRNA target site or a microRNA recognition site, or any nucleotide sequence to which a microRNA binds or associates. It should be understood that “binding” may follow traditional Watson-Crick hybridization rules or may reflect any stable association of the microRNA with the target sequence at or adjacent to the microRNA site.
  • microRNA binding sites can be engineered out of (i.e. removed from) sequences in which they naturally occur in order to increase protein expression in specific tissues.
  • miR-122 binding sites may be removed to improve protein expression in the liver. Regulation of expression in multiple tissues can be accomplished through introduction or removal or one or several microRNA binding sites.
  • tissues where microRNA are known to regulate mRNA, and thereby protein expression include, but are not limited to, liver (miR-122), muscle (miR-133, miR-206, miR-208), endothelial cells (miR- 17-92, miR-126), myeloid cells (miR-142-3p, miR-142-5p, miR-16, miR-21, miR-223, miR-24, miR-27), adipose tissue (let-7, miR-30c), heart (miR-ld, miR-149), kidney (miR- 192, miR-194, miR-204), and lung epithelial cells (let-7, miR-133, miR-126).
  • liver miR-122
  • muscle miR-133, miR-206, miR-208
  • endothelial cells miR- 17-92, miR-126
  • myeloid cells miR-142-3p, miR-142-5p, miR-16, miR-21, miR
  • MicroRNA can also regulate complex biological processes such as angiogenesis (miR- 132) (Anand and Cheresh Curr Opin Hematol 2011 18: 171-176; herein incorporated by reference in its entirety).
  • angiogenesis miR- 132
  • binding sites for microRNAs that are involved in such processes may be removed or introduced, in order to tailor the expression of the modified mRNA expression to biologically relevant cell types or to the context of relevant biological processes.
  • modified mRNA can be engineered for more targeted expression in specific cell types or only under specific biological conditions. Through introduction of tissue-specific microRNA binding sites, modified mRNA could be designed that would be optimal for protein expression in a tissue or in the context of a biological condition.
  • Transfection experiments can be conducted in relevant cell lines, using engineered modified mRNA and protein production can be assayed at various time points post-transfection. For example, cells can be transfected with different micro RNA binding site-engineering modified mRNA and by using an ELISA kit to the relevant protein and assaying protein produced at 6 hour, 12 hour, 24 hour, 48 hour, 72 hour and 7 days post-transfection. In vivo experiments can also be conducted using microRNA-binding site-engineered molecules to examine changes in tissue-specific expression of formulated modified mRNA.
  • the 5' cap structure of an mRNA is involved in nuclear export, increasing mRNA stability and binds the mRNA Cap Binding Protein (CBP), which is responsible for mRNA stability in the cell and translation competency through the association of CBP with poly(A) binding protein to form the mature cyclic mRNA species.
  • CBP mRNA Cap Binding Protein
  • the cap further assists the removal of 5' proximal introns removal during mRNA splicing.
  • Endogenous mRNA molecules may be 5 '-end capped generating a 5 '-ppp-5 '-triphosphate linkage between a terminal guanosine cap residue and the 5 '-terminal transcribed sense nucleotide of the mRNA molecule.
  • This 5'-guanylate cap may then be methylated to generate an N7-methyl- guanylate residue.
  • the ribose sugars of the terminal and/or anteterminal transcribed nucleotides of the 5' end of the mRNA may optionally also be 2'-0-methylated.
  • 5'-decapping through hydrolysis and cleavage of the guanylate cap structure may target a nucleic acid molecule, such as an mRNA molecule, for degradation.
  • Modifications to the modified mRNA of the present invention may generate a non- hydrolyzable cap structure preventing decapping and thus increasing mRNA half-life. Because cap structure hydrolysis requires cleavage of 5 '-ppp-5' phosphorodiester linkages, modified nucleotides may be used during the capping reaction. For example, a Vaccinia Capping Enzyme from New England Biolabs (Ipswich, MA) may be used with a-thio-guanosine nucleotides according to the manufacturer's instructions to create a phosphorothioate linkage in the 5 '-ppp-5' cap. Additional modified guanosine nucleotides may be used such as a-methyl-phosphonate and seleno-phosphate nucleotides.
  • Additional modifications include, but are not limited to, 2'-0-methylation of the ribose sugars of 5 '-terminal and/or 5 '-anteterminal nucleotides of the mRNA (as mentioned above) on the 2'-hydroxyl group of the sugar ring.
  • Multiple distinct 5 '-cap structures can be used to generate the 5 '-cap of a nucleic acid molecule, such as an m NA molecule.
  • Cap analogs which herein are also referred to as synthetic cap analogs, chemical caps, chemical cap analogs, or structural or functional cap analogs, differ from natural (i.e. endogenous, wild-type or physiological) 5 '-caps in their chemical structure, while retaining cap function. Cap analogs may be chemically (i.e. non-enzymatically) or enzymatically synthesized and/or linked to a nucleic acid molecule.
  • the Anti-Reverse Cap Analog (ARCA) cap contains two guanines linked by a 5 '-5 '-triphosphate group, wherein one guanine contains an N7 methyl group as well as a 3'-0-methyl group (i.e., N7,3'-0-dimethyl-guanosine-5'-triphosphate-5'-guanosine (m 7 G-3'mppp-G; which may equivalently be designated 3' 0-Me-m7G(5')ppp(5')G).
  • the 3'-0 atom of the other, unmodified, guanine becomes linked to the 5'-terminal nucleotide of the capped nucleic acid molecule (e.g. an mRNA or mmRNA).
  • the N7- and 3'-0-methlyated guanine provides the terminal moiety of the capped nucleic acid molecule (e.g. mRNA or mmRNA).
  • mCAP which is similar to ARCA but has a 2'-0-methyl group on guanosine (i.e., N7,2'-0-dimethyl-guanosine-5'-triphosphate-5'-guanosine, m 7 Gm-ppp-G).
  • cap analogs allow for the concomitant capping of a nucleic acid molecule in an in vitro transcription reaction, up to 20% of transcripts can remain uncapped. This, as well as the structural differences of a cap analog from an endogenous 5 '-cap structures of nucleic acids produced by the endogenous, cellular transcription machinery, may lead to reduced translational competency and reduced cellular stability.
  • Modified mRNA of the present invention may also be capped post-transcriptionally, using enzymes, in order to generate more authentic 5 '-cap structures.
  • the phrase "more authentic” refers to a feature that closely mirrors or mimics, either structurally or functionally, an endogenous or wild type feature. That is, a "more authentic" feature is better representative of an endogenous, wild-type, natural or physiological cellular function and/or structure as compared to synthetic features or analogs, etc., of the prior art, or which outperforms the corresponding endogenous, wild-type, natural or physiological feature in one or more respects.
  • Non-limiting examples of more authentic 5 'cap structures of the present invention are those which, among other things, have enhanced binding of cap binding proteins, increased half life, reduced susceptibility to 5' endonucleases and/or reduced 5'decapping, as compared to synthetic 5 'cap structures known in the art (or to a wild-type, natural or physiological 5'cap structure).
  • recombinant Vaccinia Virus Capping Enzyme and recombinant 2'-0-methyltransferase enzyme can create a canonical 5'- 5 '-triphosphate linkage between the 5 '-terminal nucleotide of an mRNA and a guanine cap nucleotide wherein the cap guanine contains an N7 methylation and the 5 '-terminal nucleotide of the mRNA contains a 2 '-0 -methyl.
  • Capl structure Such a structure is termed the Capl structure.
  • Cap structures include, but are not limited to, 7mG(5')ppp(5')N,pN2p (cap 0), 7mG(5')ppp(5')NlmpNp (cap 1), and 7mG(5')-ppp(5')NlmpN2mp (cap 2).
  • the modified mRNA may be capped post-transcriptionally, and because this process is more efficient, nearly 100% of the modified mRNA may be capped. This is in contrast to -80% when a cap analog is linked to an mRNA in the course of an in vitro transcription reaction.
  • 5' terminal caps may include endogenous caps or cap analogs.
  • a 5' terminal cap may comprise a guanine analog.
  • Useful guanine analogs include, but are not limited to, inosine, Nl-methyl-guanosine, 2'fluoro- guanosine, 7-deaza-guanosine, 8-oxo-guanosine, 2-amino-guanosine, LNA-guanosine, and 2-azido- guanosine.
  • Additional viral sequences such as, but not limited to, the translation enhancer sequence of the barley yellow dwarf virus (BYDV-PAV) can be engineered and inserted in the 3' UTR of the modified mRNA of the invention and can stimulate the translation of the mRNA in vitro and in vivo.
  • Transfection experiments can be conducted in relevant cell lines at and protein production can be assayed by ELISA at 12 hour, 24 hour, 48 hour, 72 hour and day 7 post-transfection.
  • modified mRNA which may contain an internal ribosome entry site (IRES).
  • IRES internal ribosome entry site
  • An IRES may act as the sole ribosome binding site, or may serve as one of multiple ribosome binding sites of an mRNA.
  • Modified mRNA containing more than one functional ribosome binding site may encode several peptides or polypeptides that are translated independently by the ribosomes ("multicistronic nucleic acid molecules").
  • IRES sequences that can be used according to the invention include without limitation, those from picornaviruses (e.g. FMDV), pest viruses (CFFV), polio viruses (PV), encephalomyocarditis viruses (ECMV), foot-and-mouth disease viruses (FMDV), hepatitis C viruses (HCV), classical swine fever viruses (CSFV), murine leukemia virus (MLV), simian immune deficiency viruses (SIV) or cricket paralysis viruses (CrPV).
  • picornaviruses e.g. FMDV
  • CFFV pest viruses
  • PV polio viruses
  • ECMV encephalomyocarditis viruses
  • FMDV foot-and-mouth disease viruses
  • HCV hepatitis C viruses
  • CSFV classical swine fever viruses
  • MLV murine leukemia virus
  • SIV simian immune deficiency viruses
  • CrPV cricket paralysis viruses
  • a long chain of adenine nucleotides may be added to a modified nucleic acid molecule such as a modified mRNA molecules in order to increase stability.
  • a modified nucleic acid molecule such as a modified mRNA molecules
  • poly-A polymerase adds a chain of adenine nucleotides to the RNA.
  • the length of a poly-A tail of the present invention is greater than 30 nucleotides in length.
  • the poly-A tail is greater than 35 nucleotides in length (e.g., at least or greater than about 35, 40, 45, 50, 55, 60, 70, 80, 90, 100, 120, 140, 160, 180, 200, 250, 300, 350, 400, 450, 500, 600, 700, 800, 900, 1,000, 1,100, 1,200, 1,300, 1,400, 1,500, 1,600, 1,700, 1,800, 1,900, 2,000, 2,500, and 3,000 nucleotides).
  • the modified mRNA includes from about 30 to about 3,000 nucleotides (e.g., from 30 to 50, from 30 to 100, from 30 to 250, from 30 to 500, from 30 to 750, from 30 to 1,000, from 30 to 1,500, from 30 to 2,000, from 30 to 2,500, from 50 to 100, from 50 to 250, from 50 to 500, from 50 to 750, from 50 to 1,000, from 50 to 1,500, from 50 to 2,000, from 50 to 2,500, from 50 to 3,000, from 100 to 500, from 100 to 750, from 100 to 1,000, from 100 to 1,500, from 100 to 2,000, from 100 to 2,500, from 100 to 3,000, from 500 to 750, from 500 to 1,000, from 500 to 1,500, from 500 to 2,000, from 500 to 2,500, from 500 to 3,000, from 1,000 to 1,500, from 1,000 to 2,000, from 1,000 to 2,500, from 1,000 to 3,000, from 1,500 to 2,000, from 1,500 to 2,500, from 1,500 to 3,000, from 1,000 to 1,500, from 1,000 to 2,000
  • the poly-A tail is designed relative to the length of the overall modified mRNA. This design may be based on the length of the coding region, the length of a particular feature or region (such as the flanking regions), or based on the length of the ultimate product expressed from the modified mRNA.
  • the poly-A tail may be 10, 20, 30, 40, 50, 60, 70, 80, 90, or 100% greater in length than the modified mRNA, region or feature thereof.
  • the poly-A tail may also be designed as a fraction of modified mRNA to which it belongs.
  • the poly-A tail may be 10, 20, 30, 40, 50, 60, 70, 80, or 90% or more of the total length of the molecule or the total length of the molecule minus the poly-A tail.
  • engineered binding sites and conjugation of modified mRNA for Poly-A binding protein may enhance expression.
  • multiple distinct modified mRNA may be linked together to the PABP (Poly- A binding protein) through the 3 '-end using modified nucleotides at the 3 '-terminus of the poly-A tail.
  • Transfection experiments can be conducted in relevant cell lines at and protein production can be assayed by ELISA at 12 hour, 24 hour, 48 hour, 72 hour and day 7 post-transfection.
  • the modified mRNA of the present invention are designed to include a polyA-G quartet.
  • the G-quartet is a cyclic hydrogen bonded array of four guanine nucleotides that can be formed by G-rich sequences in both DNA and RNA.
  • the G-quartet is incorporated at the end of the poly-A tail.
  • the resultant mmRNA molecule is assayed for stability, protein production and other parameters including half- life at various time points. It has been discovered that the polyA-G quartet results in protein production equivalent to at least 75% of that seen using a poly-A tail of 120 nucleotides alone.
  • modified nucleic acids and modified mRNA (mmRNA) of the invention may contain one, two, or more different modifications.
  • modified nucleic acids and mmRNA may contain one, two, or more different nucleoside or nucleotide modifications.
  • a modified nucleic acid or mmRNA (e.g., having one or more mmRNA molecules) introduced to a cell may exhibit reduced degradation in the cell, as compared to an unmodified nucleic acid or mmRNA.
  • the modified nucleic acids and mmRNA can include any useful modification, such as to the sugar, the nucleobase (e.g., one or more modifications of a nucleobase, such as by replacing or substituting an atom of a pyrimidine nucleobase with optionally substituted amino, optionally substituted thiol, optionally substituted alkyl (e.g., methyl or ethyl), or halo (e.g., chloro or fluoro), or the internucleoside linkage (e.g., one or more modification to the phosphodiester backbone).
  • the nucleobase e.g., one or more modifications of a nucleobase, such as by replacing or substituting an atom of a pyrimidine nucleobase with optionally substituted amino, optionally substituted thiol, optionally substituted alkyl (e.g., methyl or ethyl), or halo (e.g., chloro or fluoro), or the internu
  • modifications are present in both the sugar and the internucleoside linkage (e.g., one or modifications, such as those present in ribonucleic acids (RNA), deoxyribonucleic acids (DNAs), threose nucleic acids (TNAs), glycol nucleic acids (GNAs), peptide nucleic acids (PNAs), locked nucleic acids (LNAs) or hybrids thereof). Additional modifications are described herein.
  • RNA ribonucleic acids
  • DNAs deoxyribonucleic acids
  • TAAs threose nucleic acids
  • GNAs glycol nucleic acids
  • PNAs peptide nucleic acids
  • LNAs locked nucleic acids
  • the modified nucleic acids and mmRNA of the invention do not substantially induce an innate immune response of a cell into which the mRNA is introduced.
  • degradation of a modified nucleic acid molecule or modified mRNA may be preferable if precise timing of protein production is desired.
  • the invention provides a modified nucleic acid molecule containing a degradation domain, which is capable of being acted on in a directed manner within a cell.
  • nucleic acids comprising a nucleoside or nucleotide that can disrupt the binding of a major groove interacting, e.g. binding, partner with the nucleic acid (e.g., where the modified nucleotide has decreased binding affinity to major groove interacting partner, as compared to an unmodified nucleotide).
  • the modified nucleic acid and mmRNA can optionally include other agents (e.g., RNAi- inducing agents, RNAi agents, siRNA, shRNA, miRNA, antisense RNA, ribozymes, catalytic DNA, tRNA, RNA that induce triple helix formation, aptamers, vectors, etc.).
  • the modified nucleic acids or mmRNA may include one or more messenger RNA (mRNA) and one or more modified nucleoside or nucleotides (e.g., mmRNA molecules). Details for these modified nucleic acids and mmRNA follow.
  • the modified nucleic acids or mmRNA of the invention may include a first region of linked nucleosides encoding a polypeptide of interest, a first flanking region located at the 5' terminus of the first region, and a second flanking region located at the 3' terminus of the first region.
  • the modified nucleic acids or mmRNA includes n number of linked nucleosides having Formula (la) or Formula (Ia-1): (la) or a pharmaceutically acceptable salt or stereoisomer thereof, wherein
  • U is O, S, N(R u ) nu , or C(R u ) nu , wherein nu is an integer from 0 to 2 and each R u is, independently, H, halo, or optionally substituted alkyl;
  • each of R 1' , R 2 , R 1" , R 2" , R 1 , R 2 , R 3 , R 4 , and R 5 is, if present, independently, H, halo, hydroxy, thiol, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, optionally substituted hydroxyalkoxy, optionally substituted amino, azido, optionally substituted aryl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, optionally substituted aminoalkynyl,or absent; wherein the combination of R 3 with one or more of
  • R , R , R , R , or R can join together to form optionally substituted alkylene or optionally substituted heteroalkylene and, taken together with the carbons to which they are attached, provide an optionally substituted heterocyclyl (e.g., a bicyclic, tricyclic, or tetracyclic heterocyclyl); wherein the combination of R 5 with one or
  • R , R , or R e.g., the combination of R and R , the combination of R and R , the combination of R 2 and R 5 , or the combination of R 2 and R 5
  • R 4 and one or more of R ⁇ R ⁇ R ⁇ R 2 ' , R 3 , or R 5 can join together to form optionally substituted alkylene or optionally substituted heteroalkylene and, taken together with the carbons to which they are attached, provide an optionally substituted heterocyclyl (e.g., a bicyclic, tricyclic, or tetracyclic heterocyclyl); and wherein the combination of R 4 and one or more of R ⁇ R ⁇ R ⁇ R 2 ' , R 3 , or R 5 can join together to form optionally substituted alkylene or optionally substituted heteroalkylene and, taken together with the carbons to which they are attached, provide an optionally substituted heterocyclyl (e.g., a bicyclic, tricyclic, or tetracyclic heterocyclyl
  • each of Y 1 , Y 2 , and Y 3 is, independently, O, S, Se, -NR N1 -, optionally substituted alkylene, or optionally substituted heteroalkylene, wherein R N1 is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, or absent;
  • each Y 4 is, independently, H, hydroxy, thiol, boranyl, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted thioalkoxy, optionally substituted alkoxyalkoxy, or optionally substituted amino;
  • each Y 5 is, independently, O, S, Se, optionally substituted alkylene (e.g., methylene), or optionally substituted heteroalkylene;
  • n is an integer from 1 to 100,000.
  • B is a nucleobase (e.g., a purine, a pyrimidine, or derivatives thereof), wherein the combination of B and R 1 , the combination of B and R 2 , the combination of B and R 1 , or the combination of B and R 2 can, taken together with the carbons to which they are attached, optionally form a bicyclic group (e.g., a bicyclic heterocyclyl) or wherein the combination of B, R 1 , and R 3 or the combination of B, R 2 , and R 3 can optionally form a tricyclic or tetracyclic group (e.g., a tricyclic or tetracyclic heterocyclyl, such as in Formula (IIo)-(IIp) herein).
  • the modified nucleic acid or mmRNA includes a modified ribose.
  • the modified nucleic acid or mmRNA includes n number of linked nucleosides having Formula (Ia-2)-(Ia-5) or a pharmaceutically acceptable salt or stereoisomer thereof.
  • the modified nucleic acid or mmRNA includes n number of linked nucleosides having Formula (lb) or Formula (Ib-1):
  • U is O, S, N(R u ) nu , or C(R u ) nu , wherein nu is an integer from 0 to 2 and each R u is, independently, H, halo, or optionally substituted alkyl;
  • each of R 1 , R 3 , R 3 , and R 4 is, independently, H, halo, hydroxy, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, optionally substituted hydroxyalkoxy, optionally substituted amino, azido, optionally substituted aryl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, or absent; and wherein the combination of R 1 and R 3 or the combination of R 1 and R 3" can be taken together to form optionally substituted alkylene or optionally substituted heteroalkylene (e.g., to produce a locked nucleic acid);
  • each R 5 is, independently, H, halo, hydroxy, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, or absent;
  • each of Y 1 , Y 2 , and Y 3 is, independently, O, S, Se, -NR N1 -, optionally substituted alkylene, or optionally substituted heteroalkylene, wherein R N1 is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, or optionally substituted aryl;
  • each Y 4 is, independently, H, hydroxy, thiol, boranyl, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted alkoxyalkoxy, or optionally substituted amino;
  • n is an integer from 1 to 100,000.
  • B is a nucleobase
  • the modified nucleic acid or mmRNA includes n number of linked nucleosides having Formula (Ic):
  • U is O, S, N(R ) nu , or C(R ) nu , wherein nu is an integer from 0 to 2 and each R is, independently, H, halo, or optionally substituted alkyl;
  • each of B 1 , B 2 , and B 3 is, independently, a nucleobase (e.g., a purine, a pyrimidine, or derivatives thereof, as described herein), H, halo, hydroxy, thiol, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, optionally substituted hydroxyalkoxy, optionally substituted amino, azido, optionally substituted aryl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, or optionally substituted aminoalkynyl, wherein one and only one of B 1 , B 2 , and B 3 is a nucleobase;
  • a nucleobase e.g., a purine, a pyrimidine, or derivatives thereof, as described herein
  • H halo, hydroxy, thi
  • each of R bl , R b2 , R b3 , R 3 , and R 5 is, independently, H, halo, hydroxy, thiol, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, optionally substituted hydroxyalkoxy, optionally substituted amino, azido, optionally substituted aryl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl or optionally substituted aminoalkynyl;
  • each of Y 1 , Y 2 , and Y 3 is, independently, O, S, Se, -NR N1 -, optionally substituted alkylene, or optionally substituted heteroalkylene, wherein R N1 is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, or optionally substituted aryl;
  • each Y 4 is, independently, H, hydroxy, thiol, boranyl, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted thioalkoxy, optionally substituted alkoxyalkoxy, or optionally substituted amino;
  • each Y 5 is, independently, O, S, Se, optionally substituted alkylene (e.g., methylene), or optionally substituted heteroalkylene;
  • n is an integer from 1 to 100,000.
  • the ring including U does not have a double bond between U-
  • the modified nucleic acid or mmRNA includes n number of linked nucleosides having Formula (Id):
  • U is O, S, N(R u ) nu , or C(R u ) nu , wherein nu is an integer from 0 to 2 and each R u is, independently, H, halo, or optionally substituted alkyl;
  • each R 3 is, independently, H, halo, hydroxy, thiol, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, optionally substituted hydroxyalkoxy, optionally substituted amino, azido, optionally substituted aryl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl,or optionally substituted aminoalkynyl;
  • each of Y 1 , Y 2 , and Y 3 is, independently, O, S, Se, -NR N1 -, optionally substituted alkylene, or optionally substituted heteroalkylene, wherein R N1 is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, or optionally substituted aryl;
  • each Y 4 is, independently, H, hydroxy, thiol, boranyl, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted thioalkoxy, optionally substituted alkoxyalkoxy, or optionally substituted amino;
  • each Y 5 is, independently, O, S, optionally substituted alkylene (e.g., methylene), or optionally substituted heteroalkylene;
  • n is an integer from 1 to 100,000.
  • B is a nucleobase (e.g., a purine, a pyrimidine, or derivatives thereof).
  • the modified nucleic acid molecules or modified mRNA includes n number of linked nucleosides having Formula (Ie):
  • each of U' and U" is, independently, O, S, N(R ) nu , or C(R ) nu , wherein nu is an integer from 0 to 2 and each R u is, independently, H, halo, or optionally substituted alkyl;
  • each R 6 is, independently, H, halo, hydroxy, thiol, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, optionally substituted hydroxyalkoxy, optionally substituted amino, azido, optionally substituted aryl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl,or optionally substituted aminoalkynyl;
  • each Y 5 is, independently, O, S, optionally substituted alkylene (e.g., methylene or ethylene), or optionally substituted heteroalkylene;
  • n is an integer from 1 to 100,000;
  • B is a nucleobase (e.g., a purine, a pyrimidine, or derivatives thereof).
  • the modified nucleic acid or mmRNA includes n number of linked nucleosides having Formula (If) or If-1):
  • each of U' and U" is, independently, O, S, N, N(R u ) nu , or C(R u ) nu , wherein nu is an integer from 0 to 2 and each R u is, independently, H, halo, or optionally substituted alkyl (e.g., U' is O and
  • is a single bond or absent
  • each of R 1 , R 2 , R 1 , R 2 , R 3 , and R 4 is, independently, H, halo, hydroxy, thiol, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, optionally substituted hydroxyalkoxy, optionally substituted amino, azido, optionally substituted aryl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, optionally substituted aminoalkynyl,or absent; and wherein the combination of R 1 and R 3 , the combination of R 1 and R 3 , the combination of R 2 and R 3 , or the combination of R 2 and R 3 can be taken together to form optionally substituted alkylene or optionally substituted heteroalkylene (e.g., to produce a locked nucleic acid);each of m' and m"
  • each of Y 1 , Y 2 , and Y 3 is, independently, O, S, Se, -NR N1 -, optionally substituted alkylene, or optionally substituted heteroalkylene, wherein R N1 is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, or absent;
  • each Y 4 is, independently, H, hydroxy, thiol, boranyl, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted thioalkoxy, optionally substituted alkoxyalkoxy, or optionally substituted amino;
  • each Y 5 is, independently, O, S, Se, optionally substituted alkylene (e.g., methylene), or optionally substituted heteroalkylene;
  • n is an integer from 1 to 100,000.
  • B is a nucleobase (e.g., a purine, a pyrimidine, or derivatives thereof).
  • the ring including U has one or two double bonds.
  • the modified nucleic acid or mmRNA e.g., Formulas (Ia)-Ia-5), (Ib)-(If-l), (Ila)-(IIp), (IIb-1), (IIb-2), (IIc-l)-(IIc-2), (IIn-1), (IIn-2), (IVa)-(IVl), and (IXa)-(IXr)), each of R 1 , R 1 , and R 1 , if present, is H.
  • each of R 2 , R 2 , and R 2 is, independently, H, halo (e.g., fluoro), hydroxy, optionally substituted alkoxy (e.g., methoxy or ethoxy), or optionally substituted alkoxyalkoxy.
  • alkoxyalkoxy is - (CH 2 )s2(OCH 2 CH 2 )si(CH 2 )s30R', wherein si is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R' is H or Ci_ 2 o alkyl).
  • s2 is 0, si is 1 or 2
  • s3 is 0 or 1
  • R' is Ci_ 6 alkyl.
  • the modified nucleic acid or mmRNA e.g., Formulas (Ia)-Ia-5), (Ib)-(If-l), (Ila)-(IIp), (IIb-1), (IIb-2), (IIc-l)-(IIc-2), (IIn-1), (IIn-2), (IVa)-(IVl), and (IXa)-(IXr)), each of R 2 , R 2 , and R 2 , if present, is H.
  • each of R 1 , R 1 , and R 1 is, independently, H, halo (e.g., fluoro), hydroxy, optionally substituted alkoxy (e.g., methoxy or ethoxy), or optionally substituted alkoxyalkoxy.
  • alkoxyalkoxy is - (CH 2 )s2(OCH 2 CH 2 )si(CH 2 )s30R', wherein si is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R' is H or Ci_ 2 o alkyl).
  • s2 is 0, si is 1 or 2
  • s3 is 0 or 1
  • R' is Ci_ 6 alkyl.
  • each of R 3 , R 4 , and R 5 is, independently, H, halo (e.g., fluoro), hydroxy, optionally substituted alkyl, optionally substituted alkoxy (e.g., methoxy or ethoxy), or optionally substituted alkoxyalkoxy.
  • R 3 is H, R 4 is H, R 5 is H, or R 3 , R 4 , and R 5 are all H.
  • R 3 is Ci_ 6 alkyl
  • R 4 is Ci_ 6 alkyl
  • R 5 is Ci_ 6 alkyl
  • R 3 and R 4 are both H
  • R 5 is Ci_ 6 alkyl.
  • modified nucleic acids or mmRNA e.g., Formulas (Ia)-Ia-5), (Ib)-(If-l), (Ila)-(IIp), (IIb-1), (IIb-2), (IIc-l)-(IIc-2), (IIn-1), (IIn-2), (IVa)-(IVl), and (IXa)-(IXr)
  • R 3 and R 5 join together to form optionally substituted alkylene or optionally substituted
  • heteroalkylene and, taken together with the carbons to which they are attached, provide an optionally substituted heterocyclyl (e.g., a bicyclic, tricyclic, or tetracyclic heterocyclyl, such as trans-3',4' analogs, wherein R 3 and R 5 join together to form heteroalkylene (e.g., -(CH2)biO(CH 2 )b20(CFl2)b3-, wherein each of bl, b2, and b3 are, independently, an integer from 0 to 3).
  • a bicyclic, tricyclic, or tetracyclic heterocyclyl such as trans-3',4' analogs, wherein R 3 and R 5 join together to form heteroalkylene (e.g., -(CH2)biO(CH 2 )b20(CFl2)b3-, wherein each of bl, b2, and b3 are, independently, an integer from 0 to 3).
  • R 3 and one or more of R 1 , R 1 , R 2 , R 2 , or R 5 join together to form optionally substituted alkylene or optionally substituted heteroalkylene and, taken together with the carbons to which they are attached, provide an optionally substituted heterocyclyl (e.g., a bicyclic, tricyclic, or tetracyclic heterocyclyl, R 3 and one or more of R 1 , R 1 , R 2 , R 2 , or R 5 join together to form heteroalkylene (e.g., -(
  • R 5 and one or more of R 1 , R 1 , R 2 , or R 2 join together to form optionally substituted alkylene or optionally substituted heteroalkylene and, taken together with the carbons to which they are attached, provide an optionally substituted heterocyclyl (e.g., a bicyclic, tricyclic, or tetracyclic heterocyclyl, R 5 and one or more of R 1 , R 1 , R 2 , or R 2 join together to form heteroalkylene (e.g., - (CH2)biO(CH 2
  • each Y 2 is, independently, O, S, or -NR N1 -, wherein R N1 is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, or optionally substituted aryl.
  • Y 2 is NR N1 -, wherein R N1 is H or optionally substituted alkyl (e.g., Ci_ 6 alkyl, such as methyl, ethyl, isopropyl, or n-propyl).
  • R N1 is H or optionally substituted alkyl (e.g., Ci_ 6 alkyl, such as methyl, ethyl, isopropyl, or n-propyl).
  • each Y 3 is, independently, O or S.
  • R 1 is H; each R 2 is, independently, H, halo (e.g., fluoro), hydroxy, optionally substituted alkoxy (e.g., methoxy or ethoxy), or optionally substituted alkoxyalkoxy (e.g., -
  • each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R' is H or Ci_2o alkyl, such as wherein s2 is 0, si is 1 or 2, s3 is 0 or 1, and R' is Ci_ 6 alkyl); each Y 2 is, independently, O or -NR N1 -, wherein R N1 is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, or optionally substituted aryl (e.g., wherein R N1 is H or optionally substituted alkyl (e.g., Ci_ 6 alkyl, such as methyl, e
  • R 3 is H, halo (e.g., fluoro), hydroxy, optionally substituted alkyl, optionally substituted alkoxy (e.g., methoxy or ethoxy), or optionally substituted alkoxyalkoxy.
  • halo e.g., fluoro
  • hydroxy optionally substituted alkyl
  • optionally substituted alkoxy e.g., methoxy or ethoxy
  • optionally substituted alkoxyalkoxy optionally substituted alkoxyalkoxy.
  • each Y 1 is , independently, O or -NR N1 -, wherein R N1 is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, or optionally substituted aryl (e.g., wherein R N1 is H or optionally substituted alkyl (e.g., Ci_ 6 alkyl, such as methyl, ethyl, isopropyl, or n-propyl)); and each Y 4 is, independently, H, hydroxy, thiol, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted thioalkoxy, optionally substituted alkoxyalkoxy, or optionally substituted amino.
  • R N1 is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, or optionally substituted aryl (e.g., wherein R N1 is H or optionally substituted alkyl (e.g
  • each R 1 is, independently, H, halo (e.g., fluoro), hydroxy, optionally substituted alkoxy (e.g., methoxy or ethoxy), or optionally substituted alkoxyalkoxy (e.g., -(CH 2 ) s2 (OCH 2 CH 2 )si(CH 2 )s30R', wherein si is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3,
  • R 2 is H; each Y 2 is, independently, O or -NR N1 -, wherein R N1 is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, or optionally substituted aryl
  • R > N1 i ⁇ s H or optionally substituted alkyl e.g., Ci_ 6 alkyl, such as methyl, ethyl, isopropyl, or n-propyl
  • each Y 3 is, independently, O or S (e.g., S).
  • R 3 is H, halo (e.g., fluoro), hydroxy, optionally substituted alkyl, optionally substituted alkoxy (e.g., methoxy or ethoxy), or optionally substituted alkoxyalkoxy.
  • each Y 1 is , independently, O or -NR N1 -, wherein R N1 is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, or optionally substituted aryl (e.g., wherein R N1 is H or optionally substituted alkyl (e.g., Ci_ 6 alkyl, such as methyl, ethyl, isopropyl, or n-propyl)); and each Y 4 is, independently, H, hydroxy, thiol, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted thioalkoxy, optionally substituted alkoxyalkoxy, or optionally substituted amino.
  • R N1 is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, or optionally substituted aryl (e.g., wherein R N1 is H or optionally substituted alkyl (e.g
  • the ring including U is in the ⁇ -D (e.g., ⁇ -D-ribo) configuration.
  • the ring including U is in the a-L (e.g., a-L-ribo) configuration.
  • one or more B is not pseudouridine ( ⁇ ) or 5-methyl-cytidine (m 5 C).
  • about 10% to about 100% of B nucleobases is not ⁇ or m 5 C (e.g., from 10% to 20%, from 10% to 35%, from 10% to 50%, from 10% to 60%, from 10% to 75%, from 10% to 90%, from 10% to 95%, from 10% to 98%, from 10% to 99%, from 20% to 35%, from 20% to 50%, from 20% to 60%, from 20% to 75%, from 20% to 90%, from 20% to 95%, from 20% to 98%, from 20% to 99%, from 20% to 100%, from 50% to 60%, from 50% to 75%, from 50% to 90%, from 50% to 95%, from 50% to 98%, from 50% to 99%, from 50% to 100%, from 75% to 90%, from 75% to 95%, from 75% to 98%, from 75% to 99%, and from 75% to 100% of n number of B is not ⁇ or m 5 C). In some embodiments, B is not ⁇ or m 5 C.
  • modified nucleic acids or mmRNA e.g., Formulas (Ia)-(Ia-5), (Ib)-(If-l), (Ila)-(IIp), (IIb-1), (IIb-2), (IIc-l)-(IIc-2), (IIn-1), (IIn-2), (IVa)-(IVl), and (IXa)-(IXr)
  • B is an unmodified nucleobase selected from cytosine, guanine, uracil and adenine
  • at least one of Y 1 , Y 2 , or Y 3 is not O.
  • the modified nucleic acids or mmRNA includes a modified ribose. In some embodiments, modified nucleic acids or mmRNA includes n number of linked nucleosides having Formula (Ila)-(IIc):
  • U is O or C(R u ) nu , wherein nu is an integer from 0 to 2 and each R u is, independently, H, halo, or optionally substituted alkyl (e.g., U is -CH 2 - or -CH-).
  • each of R 1 , R 2 , R 3 , R 4 , and R 5 is, independently, H, halo, hydroxy, thiol, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, optionally substituted hydroxyalkoxy, optionally substituted amino, azido, optionally substituted aryl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, or absent (e.g., each R 1 and R 2 is, independently, H, halo, hydroxy, optionally substituted alkyl, or optionally substituted alkoxy; each
  • R 3 and R 4 is, independently, H or optionally substituted alkyl; and R 5 is H or hydroxy), and is a single bond or double bond.
  • the modified nucleic acid or mmRNA includes n number of linked nucleosides having Formula lib- 1 )-(IIb-2) :
  • U is O or C(R ) nu , wherein nu is an integer from 0 to 2 and each R is, independently, H, halo, or optionally substituted alkyl (e.g., U is -CH 2 - or - CH-).
  • each of R 1 and R 2 is, independently, H, halo, hydroxy, thiol, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, optionally substituted hydroxyalkoxy, optionally substituted amino, azido, optionally substituted aryl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, or absent (e.g., each R 1 and R 2 is, independently, H, halo, hydroxy, optionally substituted alkyl, or optionally substituted alkoxy, e.g., H, halo, hydroxy, alkyl, or alkoxy).
  • R 2 is hydroxy or optionally substituted alkoxy (e.g., methoxy, ethoxy, or any described herein).
  • the modified nucleic acid or mmRNA includes n number of linked
  • U is O or C(R u ) nu , wherein nu is an integer from 0 to 2 and each R 1 is, independently, H, halo, or optionally substituted alkyl (e.g., U is -CH 2 - or -CH-).
  • each of R 1 , R 2 , and R 3 is, independently, H, halo, hydroxy, thiol, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, optionally substituted hydroxyalkoxy, optionally substituted amino, azido, optionally substituted aryl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, or absent (e.g., each R 1 and R 2 is, independently, H, halo, hydroxy, optionally substituted alkyl, or optionally substituted alkoxy, e.g., H, halo, hydroxy, alkyl, or alkoxy; and each R 3 is, independently, H or optionally substituted alkyl)).
  • R 2 is optionally substituted alkoxy (e.g., methoxy or ethoxy, or any described herein).
  • R 1 is optionally substituted alkyl
  • R 2 is hydroxy.
  • R 1 is hydroxy
  • R 2 is optionally substituted alkyl.
  • R 3 is optionally substituted alkyl.
  • the modified nucleic acids or mmRNA includes an acyclic modified ribose. In some embodiments, the modified nucleic acids or mmRNA includes n number of linked nucleosides having Formula (Ild)-(IIf):
  • the modified nucleic acids or mmRNA includes an acyclic modified hexitol. In some embodiments, the modified nucleic acids or mmRNA includes n number of linked nucleosides having Formula (Ilg)-(IIj):
  • the modified nucleic acids or mmRNA includes a sugar moiety having a contracted or an expanded ribose ring. In some embodiments, the modified nucleic acids or mmRNA includes n number of linked nucleosides havin Formula Ilk - IIm):
  • each of R 1 , R 1 , R 2 , and R 2 is, independently, H, halo, hydroxy, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, or absent; and wherein the combination of R 2 and R 3 or the combination of R 2 and R 3 can be taken together to form optionally substituted alkylene or optionally substituted heteroalkylene.
  • the modified nucleic acids or mmRNA includes a locked modified ribose. In some embodiments, the modified nucleic acids or mmRNA includes n number of linked nucleosides havin Formula (Iln):
  • R 3 is O, S, or -NR N1 -, wherein R N1 is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, or optionally substituted aryl and R 3 is optionally substituted alkylene (e.g., -CH 2 -, -CH 2 CH 2 -, or -CH 2 CH 2 CH 2 -) or optionally substituted heteroalkylene (e.g., - CH 2 NH-, -CH 2 CH 2 NH-, -CH 2 OCH 2 -, or -CH 2 CH 2 OCH 2 -)(e.g., R 3' is O and R 3" is optionally substituted alkylene (e.g., -CH 2 -, -CH 2 CH 2 -, or -CH 2 CH 2 CH 2 -)).
  • the modified nucleic acid or mmRNA includes n number of linked nucleosides havin Formula (IIn-l)- II-n2):
  • R 3 is O, S, or -NR N1 -, wherein R N1 is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, or optionally substituted aryl and R 3 is optionally substituted alkylene (e.g., -CH 2 -, -CH 2 CH 2 -, or -CH 2 CH 2 CH 2 -) or optionally substituted heteroalkylene (e.g., -CH 2 NH-, -CH 2 CH 2 NH-, -CH 2 OCH 2 -, or -CH 2 CH 2 OCH 2 -) (e.g., R 3' is O and R 3 is optionally substituted alkylene (e.g., -CH 2 -, -CH 2 CH 2 -, or -CH 2 CH 2 CH 2 -)).
  • R N1 is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, or optional
  • the modified nucleic acids or mmRNA includes a locked modified ribose that forms a tetracyclic heterocyclyl. In some embodiments, the modified nucleic acids or mmRNA includes n number of linked nucleosides havin Formula Ho :
  • R 12a , R 12c , T 1 , T 1 ,T 2 , T 2 , V 1 , and V 3 are as described herein.
  • any of the formulas for the modified nucleic acids or mmRNA can include one or more nucleobases described herein (e.g., Formulas (bl)-(b43)).
  • the present invention provides methods of preparing a modified nucleic acids or mmRNA comprising at least one nucleotide (e.g., mmRNA molecule), wherein the modified nucleic acid comprises n number of nucleosides having Formula (la), as defined herein:
  • the present invention provides methods of amplifying a modified nucleic acids or mmRNA comprising at least one nucleotide (e.g., mmRNA molecule), the method comprising: reacting a compound of Formula (Ilia), as defined herein, with a primer, a cDNA template, and an RNA polymerase.
  • a compound of Formula (Ilia) as defined herein, with a primer, a cDNA template, and an RNA polymerase.
  • the present invention provides methods of preparing a modified nucleic acids or mmRNA comprising at least one nucleotide (e.g., mmRNA molecule), wherein the modified nucleic acid comprises n number of nucleosides having Formula (Ia-1), as defined herein:
  • the present invention provides methods of amplifying a modified nucleic acids or mmRNA comprising at least one nucleotide (e.g., mmRNA molecule), the method comprising reacting a compound of Formula (IIIa-1), as defined herein, with a primer, a cDNA template, and an RNA polymerase.
  • a nucleotide e.g., mmRNA molecule
  • the present invention provides methods of preparing a modified mRNA comprising at least one nucleotide (e.g., mmRNA molecule), wherein the polynucleotide comprises n number of nucleosides having Formula (Ia-2), as defined herein:
  • the present invention provides methods of amplifying a modified mR A comprising at least one nucleotide (e.g., mmR A molecule), the method comprising:
  • reaction may be repeated from 1 to about 7,000 times.
  • B may be a nucleobase of Formula (bl)-(b43).
  • modified nucleic acids and mmRNA can optionally include 5' and/or 3' flanking regions, which are described herein.
  • RNA e.g. mmRNA
  • the present invention also includes building blocks, e.g., modified ribonucleosides, modified ribonucleotides, of modified RNA (mmRNA) molecules.
  • mmRNA modified RNA
  • these mmRNA can be useful for preparing the modified nucleic acids or mmRNA of the invention.
  • the buildin block molecule has Formula Ilia or (Ilia- 1 ) :
  • the building block molecule which may be incorporated into a modified nucleic acid or mmRNA has Formula IVa -(IVb):
  • B is as described herein (e.g., any one of (bl)-(b43)).
  • Formula (IVa) or (IVb) is combined with a modified uracil (e.g., any one of formulas (bl)-(b9), (b21)-(b23), and (b28)-(b31), such as formula (bl), (b8), (b28), (b29), or (b30)).
  • Formula (IVa) or (IVb) is combined with a modified cytosine (e.g., any one of formulas (M0)-(bl4), (b24), (b25), and (b32)-(b36), such as formula (blO) or (b32)).
  • a modified guanine e.g., any one of formulas (bl5)-(bl7) and (b37)-(b40)
  • Formula (IVa) or (IVb) is combined with a modified adenine (e.g., any one of formulas (M8)-(b20) and (b41)-(b43)).
  • the building block molecule which may be incorporated into a modified nucleic acid molecule or mmRNA, has Formula (IVc)-(IVk):
  • B is as described herein (e.g., any one of (bl)-(b43)).
  • one of Formulas (IVc)-(IVk) is combined with a modified uracil (e.g., any one of formulas (bl)-(b9), (b21)- (b23), and (b28)-(b31), such as formula (bl), (b8), (b28), (b29), or (b30)).
  • a modified uracil e.g., any one of formulas (bl)-(b9), (b21)- (b23), and (b28)-(b31), such as formula (bl), (b8), (b28), (b29), or (b30).
  • a modified uracil e.g., any one of
  • one of Formulas (IVc)-(IVk) is combined with a modified cytosine (e.g., any one of formulas (M0)-(bl4), (b24), (b25), and (b32)-(b36), such as formula (blO) or (b32)).
  • one of Formulas (IVc)-(IVk) is combined with a modified guanine (e.g., any one of formulas (bl5)-(bl7) and (b37)-(b40)).
  • one of Formulas (IVc)-(IVk) is combined with a modified adenine (e.g., any one of formulas (M8)-(b20) and (b41)-(b43)).
  • the building block molecule which may be incorporated into a modified nucleic acid molecule or mmR A, has Formula (Va) or (Vb):
  • the building block molecule which may be incorporated into a modified nucleic acid molecule or mmRNA, has Formula (IXa)-(IXd):
  • one of Formulas (IXa)-(IXd) is combined with a modified uracil (e.g., any one of formulas (bl)-(b9), (b21)- (b23), and (b28)-(b31), such as formula (bl), (b8), (b28), (b29), or (b30)).
  • a modified uracil e.g., any one of formulas (bl)-(b9), (b21)- (b23), and (b28)-(b31), such as formula (bl), (b8), (b28), (b29), or (b30)
  • one of Formulas (IXa)-(IXd) is combined with a modified cytosine (e.g., any one of formulas (M0)-(bl4), (b24), (b25), and (b32)-(b36), such as formula (blO) or (b32)).
  • a modified cytosine e.g., any one of formulas (M0)-(bl4), (b24), (b25), and (b32)-(b36), such as formula (blO) or (b32)
  • one of Formulas (IXa)-(IXd) is combined with a modified guanine (e.g., any one of formulas (bl5)-(bl7) and (b37)-(b40)).
  • a modified adenine e.g., any one of formulas (M8)-(b20) and (b41)-(b43)).
  • the building block molecule which may be incorporated into a modified nucleic acid molecule or mmRNA, has Formula (IXe)-(IXg):
  • one of Formulas (IXe)-(IXg) is combined with a modified uracil (e.g., any one of formulas (bl)-(b9), (b21)-(b23), and (b28)-(b31), such as formula (bl), (b8), (b28), (b29), or (b30)).
  • a modified uracil e.g., any one of formulas (bl)-(b9), (b21)-(b23), and (b28)-(b31), such as formula (bl), (b8), (b28), (b29), or (b30)
  • one of Formulas (IXe)-(IXg) is combined with a modified cytosine (e.g., any one of formulas (M0)-(bl4), (b24), (b25), and (b32)- (b36), such as formula (blO) or (b32)).
  • one of Formulas (IXe)-(IXg) is combined with a modified guanine (e.g., any one of formulas (bl5)-(bl7) and (b37)-(b40)).
  • one of Formulas (IXe)-(IXg) is combined with a modified adenine (e any one of formulas (bl8)-(b20) and (b41)-(b43)).
  • the building block molecule which may be incorporated into
  • B is as described herein (e.g., any one of (bl)-(b43)).
  • one of Formulas (IXh)-(IXk) is combined with a modified uracil (e.g., any one of formulas (bl)-(b9), (b21)- (b23), and (b28)-(b31), such as formula (bl), (b8), (b28), (b29), or (b30)).
  • a modified uracil e.g., any one of formulas (bl)-(b9), (b21)- (b23), and (b28)-(b31), such as formula (bl), (b8), (b28), (b29), or (b30).
  • one of Formulas (IXh)-(IXk) is combined with a modified cytosine (e.g., any one of formulas (M0)-(bl4), (b24), (b25), and (b32)-(b36), such as formula (blO) or (b32)).
  • a modified cytosine e.g., any one of formulas (M0)-(bl4), (b24), (b25), and (b32)-(b36), such as formula (blO) or (b32)
  • one of Formulas (IXh)-(IXk) is combined with a modified guanine (e.g., any one of formulas (bl5)-(bl7) and (b37)-(b40)).
  • a modified adenine e.g., any one of formulas (M8)-(b20) and (b41)-(b43)).
  • the building block molecule which may be incorporated into a modified nucleic acid molecule or mmRNA, has Formula (IXl)-(IXr):
  • each rl and r2 is, independently, an integer from 0 to 5 (e.g., from 0 to 3, from 1 to 3, or from 1 to 5) and B is as described herein (e.g., any one of (bl)-(b43)).
  • one of Formulas (IXl)-(IXr) is combined with a modified uracil (e.g., any one of formulas (bl)-(b9), (b21)-(b23), and (b28)-(b31), such as formula (bl), (b8), (b28), (b29), or (b30)).
  • a modified uracil e.g., any one of formulas (bl)-(b9), (b21)-(b23), and (b28)-(b31), such as formula (bl), (b8), (b28), (b29), or (b30)
  • one of Formulas (IXl)-(IXr) is combined with a modified cytosine (e.g., any one of formulas (M0)-(bl4), (b24), (b25), and (b32)-(b36), such as formula (blO) or (b32)).
  • one of Formulas (IXl)-(IXr) is combined with a modified guanine (e.g., any one of formulas (bl5)-(bl7) and (b37)-(b40)).
  • one of Formulas (IXl)-(IXr) is combined with a modified adenine (e.g., any one of formulas (M8)-(b20) and (b41)-(b43)).
  • the building block molecule which may be incorporated into a modified nucleic acid molecules or mmR A, can be selected from the group consisting of: B- 10), BB- 12), or a pharmaceutically acceptable salt or stereoisomer thereof, wherein each r is, independently, an integer from 0 to 5 (e.g., from 0 to 3, from 1 to 3, or from 1 to 5).
  • the building block molecule which may be incorporated into a modified nucleic acid molecule or mmRNA, can be selected from the group consisting of:
  • HO OH (BB- 20), or a pharmaceutically acceptable salt or stereoisomer thereof, wherein each r is, independently, an integer from 0 to 5 (e.g., from 0 to 3, from 1 to 3, or from 1 to 5) and si is as described herein.
  • the building block molecule which may be incorporated into a nucleic acid (e.g., RNA, mRNA, or mmRNA), is a modified uridine (e.g., selected from the group consisting of:
  • the building block molecule which may be incorporated into a modified nucleic acid molecule or mmRNA, is a modified cytidine (e.g., selected from the group consisting of:
  • BB- 153 (BB- 155), eutically acceptable salt or stereoisomer thereof, wherein Y 1 , Y Y 4 , Y 6 , and r are as described herein (e.g., each r is, independently, an integer from 0 to 5, such as from 0 to 3, from 1 to 3, or from 1 to 5)).
  • the building block molecule which may be incorporated into a modified nucleic acid
  • molecule or mmR A can be:
  • the building block molecule which may be incorporated into a modified nucleic acid molecule or mmRNA, is a modified adenosine (e.g., selected from the group consisting of:
  • HO OH (BB- 192), HO OH (BB- 193), and (BB- 200) or a pharmaceutically acceptable salt or stereoisomer thereof, wherein Y 1 , Y 3 , Y 4 , Y 6 , and r are as described herein (e.g., each r is, independently, an integer from 0 to 5, such as from 0 to 3, from 1 to 3, or from 1 to 5)).
  • the building block molecule which may be incorporated into a modified nucleic acid molecule or mmRNA, is a modified guanosine (e.g., selected from the group consisting of:
  • BB- 237) or a pharmaceutically acceptable salt or stereoisomer thereof, wherein Y 1 , Y 3 , Y 4 , Y 6 , and r are as described herein (e.g., each r is, independently, an integer from 0 to 5, such as from 0 to 3, from 1 to 3, or from 1 to 5)).
  • the chemical modification can include replacement of C group at C- 5 of the ring (e.g., for a pyrimidine nucleoside, such as cytosine or uracil) with N (e.g., replacement of the >CH group at C-5 with >NR N1 group, wherein R N 1 is H or optionally substituted alkyl).
  • the mmRNA molecule which may be incorporated into a modified nucleic acid molecule or mmRNA, can be:
  • each r is, independently, an integer from 0 to 5 (e.g., from 0 to 3, from 1 to 3, or from 1 to 5).
  • the chemical modification can include replacement of the hydrogen at C-5 of cytosine with halo (e.g., Br, CI, F, or I) or optionally substituted alkyl (e.g., methyl).
  • halo e.g., Br, CI, F, or I
  • optionally substituted alkyl e.g., methyl
  • the mmRNA molecule which may be incorporated into a modified nucleic acid or mmRNA, can be:
  • each r is, independently, an integer from 0 to 5 (e.g., from 0 to 3, from 1 to 3, or from 1 to 5).
  • the chemical modification can include a fused ring that is formed by the NH 2 at the C-4 position and the carbon atom at the C-5 position.
  • the building block molecule which may be incorporated into a modified nucleic acid molecule or mmRNA, can be: (BB- 246), or a pharmaceutically acceptable salt or stereoisomer thereof, wherein each r is, independently, an integer from 0 to 5 (e.g., from 0 to 3, from 1 to 3, or from 1 to 5).
  • modified nucleosides and nucleotides which may be incorporated into a modified nucleic acid or mmR A (e.g., R A or mR A, as described herein), can be modified on the sugar of the ribonucleic acid.
  • the 2' hydroxyl group (OH) can be modified or replaced with a number of different substituents.
  • Exemplary substitutions at the 2'- position include, but are not limited to, H, halo, optionally substituted Ci_ 6 alkyl; optionally substituted Ci_ 6 alkoxy; optionally substituted C 6-1 o aryloxy; optionally substituted C3-8 cycloalkyl; optionally substituted C 3 _ 8 cycloalkoxy; optionally substituted C 6 _io aryloxy; optionally substituted C 6 -io aryl-Ci_6 alkoxy, optionally substituted C 1-12 (heterocyclyl)oxy; a sugar (e.g., ribose, pentose, or any described herein); a polyethyleneglycol (PEG), -0(CH 2 CH 2 0) n CH 2 CH 2 OR, where R is H or optionally substituted alkyl, and n is an integer from 0 to 20 (e.g., from 0 to 4, from 0 to 8, from 0 to 10, from 0 to 16, from 1 to 4,
  • RNA includes the sugar group ribose, which is a 5-membered ring having an oxygen.
  • modified nucleotides include replacement of the oxygen in ribose (e.g., with S, Se, or alkylene, such as methylene or ethylene); addition of a double bond (e.g., to replace ribose with cyclopentenyl or cyclohexenyl); ring contraction of ribose (e.g., to form a 4- membered ring of cyclobutane or oxetane); ring expansion of ribose (e.g., to form a 6- or 7- membered ring having an additional carbon or heteroatom, such as for anhydrohexitol, altritol, mannitol, cyclohexanyl, cyclohexenyl, and morpholino that also has a phosphoramidate backbone); multicyclic
  • the sugar group can also contain one or more carbons that possess the opposite stereochemical configuration than that of the corresponding carbon in ribose.
  • a modified nucleic acid molecule or mmRNA can include nucleotides containing, e.g., arabinose, as the sugar.
  • nucleoside is defined as a compound containing a sugar molecule (e.g., a pentose or ribose) or a derivative thereof in combination with an organic base (e.g., a purine or pyrimidine) or a derivative thereof.
  • organic base e.g., a purine or pyrimidine
  • nucleotide is defined as a nucleoside including a phosphate group.
  • modified nucleotides e.g., modified mRNA
  • modified mRNA may by synthesized by any useful method, as described herein (e.g., chemically, enzymatically, or recombinantly to include one or more modified or non-natural nucleosides).
  • the modified nucleotide base pairing encompasses not only the standard adenosine- thymine, adenosine-uracil, or guanosine-cytosine base pairs, but also base pairs formed between nucleotides and/or modified nucleotides comprising non-standard or modified bases, wherein the arrangement of hydrogen bond donors and hydrogen bond acceptors permits hydrogen bonding between a non-standard base and a standard base or between two complementary non-standard base structures.
  • non-standard base pairing is the base pairing between the modified nucleotide inosine and adenine, cytosine or uracil.
  • the modified nucleosides and nucleotides can include a modified nucleobase.
  • nucleobases found in RNA include, but are not limited to, adenine, guanine, cytosine, and uracil.
  • nucleobase found in DNA include, but are not limited to, adenine, guanine, cytosine, and thymine.
  • These nucleobases can be modified or wholly replaced to provide modified nucleic acids or mmRNA molecules having enhanced properties, e.g., resistance to nucleases through disruption of the binding of a major groove binding partner.
  • Table 1 below identifies the chemical faces of each canonical nucleotide. Circles identify the atoms comprising the respective chemical regions. Table 1
  • B is a modified uracil.
  • exemplary modified uracils include those in
  • is a single or double bond;
  • each of T 1 , T 1 , T 2 , and T 2 is, independently, H, optionally substituted alkyl, optionally substituted alkoxy, or optionally substituted thioalkoxy, or the combination of T 1 and T 1 or the combination of T 2 and T 2 join together (e.g., as in T 2 ) to form O (oxo), S (thio), or Se (seleno);
  • each of V 1 and V 2 is, independently, O, S, N(R vb ) nv , or C(R vb ) nv , wherein nv is an integer from 0 to 2 and each R ⁇ is, independently, H, halo, optionally substituted amino acid, optionally substituted alkyl, optionally substituted haloalkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted hydroxyalkyl, optionally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally substituted aminoalkyl (e.g., substituted with an N- protecting group, such as any described herein, e.g., trifluoroacetyl), optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, optionally substituted acylamino
  • R 10 is H, halo, optionally substituted amino acid, hydroxy, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aminoalkyl, optionally substituted hydroxyalkyl, optionally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, optionally substituted alkoxy, optionally substituted alkoxycarbonylalkyl, optionally substituted alkoxycarbonylalkenyl, optionally substituted alkoxycarbonylalkynyl, optionally substituted alkoxycarbonylalkoxy, optionally substituted carboxyalkoxy, optionally substituted carboxyalkyl, or optionally substituted carbamoylalkyl;
  • R 11 is H or optionally substituted alkyl
  • R 12a is H, optionally substituted alkyl, optionally substituted hydroxyalkyl, optionally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally substituted
  • R c is H, halo, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted thioalkoxy, optionally substituted amino, optionally substituted hydroxyalkyl, optionally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, or optionally substituted aminoalkynyl.
  • is a single or double bond
  • each of T 1 , T 1 , T 2 , and T 2 is, independently, H, optionally substituted alkyl, optionally substituted alkoxy, or optionally substituted thioalkoxy, or the combination of T 1 and T 1 join together (e.g., as in T 1 ) or the combination of T 2 and T 2 join together (e.g., as in T 2 ) to form O (oxo), S (thio), or Se (seleno), or each T 1 and T 2 is, independently, O (oxo), S (thio), or Se (seleno);
  • each of W 1 and W 2 is, independently, N(R Wa ) nw or C(R Wa ) nw , wherein nw is an integer from 0 to 2 and each R Wa is, independently, H, optionally substituted alkyl, or optionally substituted alkoxy;
  • each V 3 is, independently, O, S, N(R Va ) nv , or C(R Va ) nv , wherein nv is an integer from 0 to 2 and each R Va is, independently, H, halo, optionally substituted amino acid, optionally substituted alkyl, optionally substituted hydroxyalkyl, optionally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted heterocyclyl, optionally substituted alkheterocyclyl, optionally substituted alkoxy, optionally substituted alkenyloxy, or optionally substituted alkynyloxy, optionally substituted aminoalkyl (e.g., substituted with an N-protecting group, such as any described herein, e.g., trifluoroacetyl, or sulfoalkyl), optionally substituted aminoalkenyl, optionally substituted
  • R 12a is H, optionally substituted alkyl, optionally substituted hydroxyalkyl, optionally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, optionally substituted carboxyalkyl (e.g., optionally substituted with hydroxy and/or an O-protecting group), optionally substituted carboxyalkoxy, optionally substituted carboxyaminoalkyl, optionally substituted carbamoylalkyl, or absent;
  • R 12b is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted hydroxyalkyl, optionally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, optionally substituted alkaryl, optionally substituted heterocyclyl, optionally substituted alkheterocyclyl,
  • optionally substituted amino acid optionally substituted alkoxycarbonylacyl, optionally substituted alkoxycarbonylalkoxy, optionally substituted alkoxycarbonylalkyl, optionally substituted alkoxycarbonylalkenyl, optionally substituted alkoxycarbonylalkynyl, optionally substituted alkoxycarbonylalkoxy, optionally substituted carboxyalkyl (e.g., optionally substituted with hydroxy and/or an O-protecting group), optionally substituted carboxyalkoxy, optionally substituted carboxyaminoalkyl, or optionally substituted carbamoylalkyl,
  • R 12c is H, halo, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted thioalkoxy, optionally substituted amino, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, or optionally substituted aminoalkynyl.
  • modified uracils include those having Formula (b28)-(b31): (b31), or a pharmaceutically acceptable salt or stereoisomer thereof, wherein
  • each of T 1 and T 2 is, independently, O (oxo), S (thio), or Se (seleno);
  • each R ⁇ and R ⁇ is, independently, H, halo, optionally substituted amino acid, optionally substituted alkyl, optionally substituted haloalkyl, optionally substituted hydroxyalkyl, optionally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkyl (e.g., substituted with an N- protecting group, such as any described herein, e.g., trifluoroacetyl, or sulfoalkyl), optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, optionally substituted acylaminoalkyl (e.g., substituted with an N-protecting group, such as any described herein, e.g., trifluoroacetyl), optionally
  • R 12a is H, optionally substituted alkyl, optionally substituted carboxyaminoalkyl, optionally substituted aminoalkyl (e.g., e.g., substituted with an N-protecting group, such as any described herein, e.g., trifluoroacetyl, or sulfoalkyl), optionally substituted aminoalkenyl, or optionally substituted aminoalkynyl; and [00232] R is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted hydroxyalkyl, optionally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, optionally substituted aminoalkynyl (e.g., e.g., substituted aminoalkenyl, optionally substituted aminoalkynyl (e.g., e.g., substituted with
  • optionally substituted alkoxycarbonylacyl optionally substituted alkoxycarbonylalkoxy, optionally substituted alkoxycarbonylalkyl, optionally substituted alkoxycarbonylalkenyl, optionally substituted alkoxycarbonylalkynyl, optionally substituted alkoxycarbonylalkoxy, optionally substituted carboxyalkoxy, optionally substituted carboxyalkyl, or optionally substituted
  • T 1 is O (oxo), and T 2 is S (thio) or Se (seleno). In other embodiments, T 1 is S (thio), and T 2 is O (oxo) or Se (seleno).
  • R ⁇ is H, optionally substituted alkyl, or optionally substituted alkoxy.
  • each R 12a and R 12b is, independently, H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, or optionally substituted hydroxyalkyl.
  • R 12a is H.
  • both R 12a and R 12b are H.
  • each R ⁇ of R 12b is, independently, optionally substituted aminoalkyl (e.g., substituted with an N-protecting group, such as any described herein, e.g., trifluoroacetyl, or sulfoalkyl), optionally substituted aminoalkenyl, optionally substituted
  • aminoalkynyl or optionally substituted acylaminoalkyl (e.g., substituted with an N-protecting group, such as any described herein, e.g., trifluoroacetyl).
  • the amino and/or alkyl of the optionally substituted aminoalkyl is substituted with one or more of optionally substituted alkyl, optionally substituted alkenyl, optionally substituted sulfoalkyl, optionally substituted carboxy (e.g., substituted with an O-protecting group), optionally substituted hydroxy (e.g., substituted with an O- protecting group), optionally substituted carboxyalkyl (e.g., substituted with an O-protecting group), optionally substituted alkoxycarbonylalkyl (e.g., substituted with an O-protecting group), or N- protecting group.
  • optionally substituted alkyl optionally substituted alkenyl
  • optionally substituted sulfoalkyl optionally substituted carb
  • optionally substituted aminoalkyl is substituted with an optionally substituted sulfoalkyl or optionally substituted alkenyl.
  • R 12a and R ⁇ are both H.
  • T 1 is O (oxo)
  • T 2 is S (thio) or Se (seleno).
  • R ⁇ is optionally substituted alkoxycarbonylalkyl or optionally substituted carbamoylalkyl.
  • the optional substituent for R a , R , R c , or R a is a polyethylene glycol group (e.g., -(CH 2 ) S2 (OCH 2 CH 2 ) s i(CH 2 ) S3 0R', wherein si is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R' is H or Ci_ 2 o alkyl); or an amino-polyethylene glycol group (e.g., -NR N1 (CH 2 ) s2 (CH 2 CH 2 0) s i(CH 2 )
  • B is a modified cytosine.
  • exemplary modified cytosines include compounds (bl0)-(bl4):
  • each of T 3 and T 3 is, independently, H, optionally substituted alkyl, optionally substituted alkoxy, or optionally substituted thioalkoxy, or the combination of T 3 and T 3 join together (e.g., as in T 3 ) to form O (oxo), S (thio), or Se (seleno);
  • each V 4 is, independently, O, S, N(R Vc ) nv , or C(R Vc ) nv , wherein nv is an integer from 0 to 2 and each R Vc is, independently, H, halo, optionally substituted amino acid, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted heterocyclyl, optionally substituted alkheterocyclyl, or optionally substituted alkynyloxy (e.g., optionally substituted with any substituent described herein, such as those selected from (1)-(21) for alkyl), wherein the combination of R 13b and R Vc can be taken together to form optionally substituted heterocyclyl;
  • each V 5 is, independently, N(R vd ) nv , or C(R vd ) nv , wherein nv is an integer from 0 to 2 and each R vd is, independently, H, halo, optionally substituted amino acid, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted heterocyclyl, optionally substituted alkheterocyclyl, or optionally substituted alkynyloxy (e.g., optionally substituted with any substituent described herein, such as those selected from (1)-(21) for alkyl) (e.g., V 5 is -CH or N);
  • each of R 13a and R 13b is, independently, H, optionally substituted acyl, optionally substituted acyloxyalkyl, optionally substituted alkyl, or optionally substituted alkoxy, wherein the combination of R 13b and R 14 can be taken together to form optionally substituted heterocyclyl;
  • each R 14 is, independently, H, halo, hydroxy, thiol, optionally substituted acyl, optionally substituted amino acid, optionally substituted alkyl, optionally substituted haloalkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted hydroxyalkyl (e.g., substituted with an O-protecting group), optionally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl,, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, optionally substituted acyloxyalkyl, optionally substituted amino (e.g., -NHR, wherein R is H, alkyl, aryl, or phosphoryl), azido, optionally substituted aryl, optionally substituted heterocyclyl, optionally substituted alkheterocycl
  • each of R 15 and R 16 is, independently, H, optionally substituted alkyl, optionally substituted alkenyl, or optionally substituted alkynyl.
  • modified cytosines include those having Formula (b32)-(b35):
  • each of T 1 and T 3 is, independently, O (oxo), S (thio), or Se (seleno);
  • each of R 13a and R 13b is, independently, H, optionally substituted acyl, optionally substituted acyloxyalkyl, optionally substituted alkyl, or optionally substituted alkoxy, wherein the combination of R 13b and R 14 can be taken together to form optionally substituted heterocyclyl;
  • each R 14 is, independently, H, halo, hydroxy, thiol, optionally substituted acyl, optionally substituted amino acid, optionally substituted alkyl, optionally substituted haloalkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted hydroxyalkyl (e.g., substituted with an O-protecting group), optionally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, optionally substituted acyloxyalkyl, optionally substituted amino (e.g., -NHR, wherein R is H, alkyl, aryl, or phosphoryl), azido, optionally substituted aryl, optionally substituted heterocyclyl, optionally substituted alkheterocycly
  • each of R 15 and R 16 is, independently, H, optionally substituted alkyl, optionally substituted alkenyl, or optionally substituted alkynyl (e.g., R 15 is H, and R 16 is H or optionally substituted alkyl).
  • R 15 is H, and R 16 is H or optionally substituted alkyl.
  • R 14 is H, acyl, or hydroxyalkyl.
  • R 14 is halo.
  • both R 14 and R 15 are H.
  • both R 15 and R 16 are H.
  • each of R 14 and R 15 and R 16 is H.
  • each of R 13a and R 13b is independently, H or optionally substituted alkyl.
  • modified cytosines include compounds of Formula (b36):
  • each R 13b is, independently, H, optionally substituted acyl, optionally substituted acyloxyalkyl, optionally substituted alkyl, or optionally substituted alkoxy, wherein the combination of R 13b and R 14b can be taken together to form optionally substituted heterocyclyl;
  • each R 14a and R 14b is, independently, H, halo, hydroxy, thiol, optionally substituted acyl, optionally substituted amino acid, optionally substituted alkyl, optionally substituted haloalkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted hydroxyalkyl (e.g., substituted with an O-protecting group), optionally substituted hydroxyalkenyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, optionally substituted acyloxyalkyl, optionally substituted amino (e.g., -NHR, wherein R is H, alkyl, aryl, phosphoryl, optionally substituted aminoalkyl, or optionally substituted carboxyaminoalkyl), azido, optionally substituted aryl, optionally substituted hetero
  • each of R 15 is, independently, H, optionally substituted alkyl, optionally substituted alkenyl, or optionally substituted alkynyl.
  • R 14b is an optionally substituted amino acid (e.g., optionally substituted lysine). In some embodiments, R 14a is H.
  • B is a modified guanine.
  • exemplary modified guanines include compounds of Formula (bl5)-(bl7):
  • each of T 4 , T 4 , T 5 , T 5 , T 6 , and T 6 is, independently, H, optionally substituted alkyl, or optionally substituted alkoxy, and wherein the combination of T 4 and T 4 (e.g., as in T 4 ) or the combination of T 5 and T 5 (e.g., as in T 5 ) or the combination of T 6 and T 6 join together (e.g., as in T 6 ) form O (oxo), S (thio), or Se (seleno);
  • each of V 5 and V 6 is, independently, O, S, N(R vd ) nv , or C(R vd ) nv , wherein nv is an integer from 0 to 2 and each R vd is, independently, H, halo, thiol, optionally substituted amino acid, cyano, amidine, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy (e.g., optionally substituted with any substituent described herein, such as those selected from (1)-(21) for alkyl), optionally substituted thioalkoxy, or optionally substituted amino; and
  • each of R 17 , R 18 , R 19a , R 19b , R 21 , R 22 , R 23 , and R 24 is, independently, H, halo, thiol, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted thioalkoxy, optionally substituted amino, or optionally substituted amino acid.
  • Exemplary modified guanosines include compounds of Formula (b37)-(b40): (b40), or a pharmaceutically acceptable salt or stereoisomer thereof, wherein
  • each of T is, independently, H, optionally substituted alkyl, or optionally substituted alkoxy, and each T 4 is, independently, O (oxo), S (thio), or Se (seleno);
  • each of R 18 , R 19a , R 19b , and R 21 is, independently, H, halo, thiol, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted thioalkoxy, optionally substituted amino, or optionally substituted amino acid.
  • R 18 is H or optionally substituted alkyl.
  • T 4 is oxo.
  • each of R 19a and R 19b is, independently, H or optionally substituted alkyl.
  • B is a modified adenine.
  • exemplary modified adenines include compounds of Formula (M8)-(b20):
  • each V 7 is, independently, O, S, N(R Ve ) nv , or C(R Ve ) nv , wherein nv is an integer from 0 to 2 and each R Ve is, independently, H, halo, optionally substituted amino acid, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted alkoxy, optionally substituted alkenyloxy, or optionally substituted alkynyloxy (e.g., optionally substituted with any substituent described herein, such as those selected from (1)-(21) for alkyl); [00267] each R is, independently, H, halo, thiol, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted thioalkoxy, or optionally substituted amino;
  • each of R 26a and R 26b is, independently, H, optionally substituted acyl, optionally substituted amino acid, optionally substituted carbamoylalkyl, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted hydroxyalkyl, optionally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally substituted alkoxy, or polyethylene glycol group (e.g., -(CH 2 )s2(OCH 2 CH 2 )si(CH 2 )s30R', wherein si is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R' is H or Ci_ 2 o alkyl); or an amino-polyethylene glycol
  • each R 27 is, independently, H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted alkoxy, optionally substituted thioalkoxy or optionally substituted amino;
  • each R 28 is, independently, H, optionally substituted alkyl, optionally substituted alkenyl, or optionally substituted alkynyl;
  • each R 29 is, independently, H, optionally substituted acyl, optionally substituted amino acid, optionally substituted carbamoylalkyl, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted hydroxyalkyl, optionally substituted hydroxyalkenyl, optionally substituted alkoxy, or optionally substituted amino.
  • Exemplary modified adenines include compounds of Formula (b41 )-(b43) :
  • each R is, independently, H, halo, thiol, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted thioalkoxy, or optionally substituted amino;
  • each of R 26a and R 26b is, independently, H, optionally substituted acyl, optionally substituted amino acid, optionally substituted carbamoylalkyl, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted hydroxyalkyl, optionally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally substituted alkoxy, or polyethylene glycol group (e.g., -(CH 2 )s2(OCH 2 CH 2 )si(CH 2 )s30R', wherein si is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R' is H or Ci_ 2 o alkyl); or an amino-polyethylene glycol
  • each R 27 is, independently, H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted alkoxy, optionally substituted thioalkoxy, or optionally substituted amino.
  • R 26a is H, and R 26b is optionally substituted alkyl.
  • each of R 26a and R 26b is, independently, optionally substituted alkyl.
  • R 27 is optionally substituted alkyl, optionally substituted alkoxy, or optionally substituted thioalkoxy.
  • R 25 is optionally substituted alkyl, optionally substituted alkoxy, or optionally substituted thioalkoxy.
  • the optional substituent for R 26a , R 26b , or R 29 is a polyethylene glycol group (e.g., -(CH 2 ) S2 (OCH 2 CH 2 )si(CH 2 ) S 30R', wherein si is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R' is H or Ci_ 2 o alkyl); or an amino- polyethylene glycol group (e.g., -NR N1 (CH 2 ) S2 (CH 2 CH 2 0) s i(CH 2 ) S3 NR N1 , wherein si is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 1 from 1 to 10 (e.g.
  • B may have Formula (b21): (b21), wherein X is, independently, O, S, optionally substituted alkylene (e.g., methylene), or optionally substituted heteroalkylene, xa is an integer from 0 to 3, and R 12a and T 2 are as described herein.
  • X is, independently, O, S, optionally substituted alkylene (e.g., methylene), or optionally substituted heteroalkylene
  • xa is an integer from 0 to 3
  • R 12a and T 2 are as described herein.
  • B may have Formula (b22):
  • R 10 is, independently, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted heterocyclyl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, optionally substituted alkoxy, optionally substituted alkoxycarbonylalkyl, optionally substituted alkoxycarbonylalkenyl, optionally substituted alkoxycarbonylalkynyl, optionally substituted alkoxycarbonylalkoxy, optionally substituted carboxyalkoxy, optionally substituted carboxyalkyl, or optionally substituted carbamoylalkyl, and R 11 , R 12a , T 1 , and T 2 are as described herein.
  • B may have Formula (b23):
  • R 1U is optionally substituted heterocyclyl (e.g., optionally substituted furyl, optionally substitued thienyl, or optionally substitued pyrrolyl), optionally substituted aryl (e.g., optionally substituted phenyl or optionally substituted naphthyl), or any substituent described herein (e.g., for R 10 ) ;and wherein R 11 (e.g., H or any substituent described herein), R 12a (e.g., H or any substituent described herein), T 1 (e.g., oxo or any substituent described herein), and T 2 (e.g., oxo or any substituent described herein) are as described herein.
  • R 11 e.g., H or any substituent described herein
  • R 12a e.g., H or any substituent described herein
  • T 1 e.g., oxo or any substituent described herein
  • T 2 e.g.,
  • B may have Formula (b24): (b24), wherein R 14 is, independently, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted heterocyclyl, optionally substituted alkaryl, optionally substituted alkheterocyclyl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, optionally substituted alkoxy, optionally substituted alkoxycarbonylalkyl, optionally substituted alkoxycarbonylalkenyl, optionally substituted alkoxycarbonylalkynyl, optionally substituted alkoxycarbonylalkoxy, optionally substituted carboxyalkoxy, optionally substituted carboxyalkyl, or optionally substituted carbamoylalkyl, and R 13a , R 13b , R 15 , and T 3 are as described herein.
  • B may have Formula (b25):
  • R 14 is optionally substituted heterocyclyl (e.g., optionally substituted furyl, optionally substitued thienyl, or optionally substitued pyrrolyl), optionally substituted aryl (e.g., optionally substituted phenyl or optionally substituted naphthyl), or any substituent described herein (e.g., for R 14 or R 14 ); and wherein R 13a (e.g., H or any substituent described herein), R 13b (e.g., H or any substituent described herein), R 15 (e.g., H or any substituent described herein), and T 3 (e.g., oxo or any substituent described herein) are as described herein.
  • R 14 is optionally substituted heterocyclyl (e.g., optionally substituted furyl, optionally substitued thienyl, or optionally substitued pyrrolyl), optionally substituted aryl (e.g., optionally substituted phenyl or
  • B is a nucleobase selected from the group consisting of cytosine, guanine, adenine, and uracil. In some embodiments, B may be:
  • the modified nucleobase is a modified uracil.
  • exemplary nucleobases and nucleosides having a modified uracil include pseudouridine ( ⁇ ), pyridin-4-one ribonucleoside, 5-aza-uridine, 6-aza-uridine, 2-thio-5-aza-uridine, 2-thio-uridine (s 2 U), 4-thio- uridine (s 4 U), 4-thio-pseudouridine, 2-thio-pseudouridine, 5 -hydroxy-uridine (ho 5 U), 5-aminoallyl- uridine, 5-halo-uridine (e.g., 5-iodo ⁇ uridine or 5-bromo-uridine), 3-methyl-uridine (m 3 U), 5- methoxy-uridine (mo 5 U), uridine 5-oxyacetic acid (cmo 5 U), uridine 5-oxyacetic acid methyl ester (mcmo 5 U), 5-carboxymethyl-uridine
  • the modified nucleobase is a modified cytosine.
  • exemplary nucleobases and nucleosides having a modified cytosine include 5-aza-cytidine, 6-aza-cytidine, pseudoisocytidine, 3-methyl-cytidine (m 3 C), N4-acetyl-cytidine (ac 4 C), 5-formyl-cytidine (f 5 C), N4- methyl-cytidine (m 4 C), 5-methyl-cytidine (m 5 C), 5-halo-cytidine (e.g., 5-iodo-cytidine), 5- hydroxymethyl-cytidine (hm 5 C), 1-methyl-pseudoisocytidine, pyrrolo-cytidine, pyrrolo- pseudoisocytidine, 2-thio-cytidine (s 2 C), 2-thio-5-methyl-cytidine, 4-thio-pseudois
  • the modified nucleobase is a modified adenine.
  • exemplary nucleobases and nucleosides having a modified adenine include 2-amino-purine, 2, 6-diaminopurine,
  • 2- amino-6-halo-purine e.g., 2-amino-6-chloro-purine
  • 6-halo-purine e.g., 6-chloro-purine
  • 2- amino-6-methyl-purine 8-azido-adenosine, 7-deaza-adenine, 7-deaza-8-aza-adenine, 7-deaza-2- amino-purine, 7-deaza-8-aza-2-amino-purine, 7-deaza-2, 6-diaminopurine, 7-deaza-8-aza-2,6- diaminopurine, 1-methyl-adenosine (m x A), 2-methyl-adenine (m 2 A), N6-methyl-adenosine(m 6 A), 2- methylthio-N6-methyl-adenosine (ms 2 m 6 A), N6-isopentenyl-adenosine (i 6 A), 2-methylthio-N6- isopentenyl-adenos
  • the modified nucleobase is a modified guanine.
  • exemplary nucleobases and nucleosides having a modified guanine include inosine (I), 1-methyl-inosine (m 1 !), wyosine (imG), methylwyosine (mimG), 4-demethyl-wyosine (imG-14), isowyosine (imG2), wybutosine (yW), peroxywybutosine (o 2 yW), hydroxywybutosine (OHyW), undermodified hydroxywybutosine (OHyW*), 7-deaza-guanosine, queuosine (Q), epoxyqueuosine (oQ), galactosyl- queuosine (galQ), mannosyl-queuosine (manQ), 7-cyano-7-deaza-guanosine (preQ 0 ), 7- aminomethyl-7-deaza-guanosine (pre
  • the nucleobase of the nucleotide can be independently selected from a purine, a pyrimidine, a purine or pyrimidine analog.
  • the nucleobase can each be independently selected from adenine, cytosine, guanine, uracil, or hypoxanthine.
  • the nucleobase can also include, for example, naturally-occurring and synthetic derivatives of a base, including pyrazolo[3,4-d]pyrimidines, 5-methylcytosine (5-me-C), 5-hydroxymethyl cytosine, xanthine, hypoxanthine, 2-aminoadenine, 6-methyl and other alkyl derivatives of adenine and guanine, 2-propyl and other alkyl derivatives of adenine and guanine, 2-thiouracil, 2-thiothymine and 2-thiocytosine, 5-propynyl uracil and cytosine, 6-azo uracil, cytosine and thymine, 5 -uracil (pseudouracil), 4-thiouracil, 8-halo (e.g., 8-bromo), 8-amino, 8-thiol, 8-thioalkyl, 8-hydroxyl and other 8-substituted aden
  • each letter refers to the representative base and/or derivatives thereof, e.g., A includes adenine or adenine analogs, e.g., 7-deaza adenine).
  • the modified nucleosides and nucleotides which may be incorporated into a modified nucleic acid or mmRNA molecule, can be modified on the internucleoside linkage (e.g., phosphate backbone).
  • the phosphate groups of the backbone can be modified by replacing one or more of the oxygen atoms with a different substituent.
  • the modified nucleosides and nucleotides can include the wholesale replacement of an unmodified phosphate moiety with a modified phosphate as described herein.
  • modified phosphate groups include, but are not limited to, phosphorothioate, phosphoroselenates, boranophosphates, boranophosphate esters, hydrogen phosphonates, phosphoramidates, phosphorodiamidates, alkyl or aryl phosphonates, and
  • Phosphorodithioates have both non-linking oxygens replaced by sulfur.
  • the phosphate linker can also be modified by the replacement of a linking oxygen with nitrogen (bridged phosphoramidates), sulfur (bridged phosphorothioates), and carbon (bridged methylene- phosphonates).
  • a-thio substituted phosphate moiety is provided to confer stability to RNA and DNA polymers through the unnatural phosphorothioate backbone linkages. Phosphorothioate DNA and RNA have increased nuclease resistance and subsequently a longer half-life in a cellular
  • Phosphorothioate linked modified nucleic acids or mmRNA molecules are expected to also reduce the innate immune response through weaker binding/activation of cellular innate immune molecules.
  • a modified nucleoside includes an alpha-thio-nucleoside (e.g., 5'- 0-(l-thiophosphate)-adenosine, 5'-0-(l-thiophosphate)-cytidine (a-thio-cytidine), 5'-0-(l- thiophosphate)-guanosine, 5'-0-(l-thiophosphate)-uridine, or 5'-0-(l-thiophosphate)- pseudouridine).
  • alpha-thio-nucleoside e.g., 5'- 0-(l-thiophosphate)-adenosine, 5'-0-(l-thiophosphate)-cytidine (a-thio-cytidine), 5'-0-(l- thiophosphate)-guanosine, 5'-0-(l-thiophosphate)-uridine, or 5'-0-(l-thiophosphate)- pseudouridine).
  • the modified nucleic acids and mmRNA of the invention can include a combination of modifications to the sugar, the nucleobase, and/or the internucleoside linkage. These combinations can include any one or more modifications described herein.
  • any of the nucleotides described herein in Formulas (la), (Ia-l)-(Ia-3), (Ib)-(If), (Ila)-(IIp), (IIb-1), (IIb-2), (IIc-l)-(IIc-2), (IIn-1), (IIn-2), (IVa)-(IVl), and (IXa)-(IXr) can be combined with any of the nucleobases described herein (e.g., in Formulas (bl)-(b43) or any other described herein).
  • modified nucleic acid and mmRNA molecules for use in accordance with the invention may be prepared according to any useful technique, as described herein.
  • the modified nucleosides and nucleotides used in the synthesis of modified nucleic acid and mmRNA molecules disclosed herein can be prepared from readily available starting materials using the following general methods and procedures. Where typical or preferred process conditions (e.g., reaction temperatures, times, mole ratios of reactants, solvents, pressures, etc.) are provided, a skilled artisan would be able to optimize and develop additional process conditions. Optimum reaction conditions may vary with the particular reactants or solvent used, but such conditions can be determined by one skilled in the art by routine optimization procedures.
  • spectroscopic means such as nuclear magnetic resonance spectroscopy (e.g., 1H or 13 C) infrared spectroscopy, spectrophotometry (e.g., UV-visible), or mass spectrometry, or by chromatography such as high performance liquid chromatography (HPLC) or thin layer chromatography.
  • HPLC high performance liquid chromatography
  • Preparation of modified nucleic acid and mmRNA molecules of the present invention can involve the protection and deprotection of various chemical groups.
  • the need for protection and deprotection, and the selection of appropriate protecting groups can be readily determined by one skilled in the art.
  • the chemistry of protecting groups can be found, for example, in Greene, et al., Protective Groups in Organic Synthesis, 2d. Ed., Wiley & Sons, 1991, which is incorporated herein by reference in its entirety.
  • Suitable solvents can be substantially nonreactive with the starting materials (reactants), the intermediates, or products at the temperatures at which the reactions are carried out, i.e., temperatures which can range from the solvent's freezing temperature to the solvent's boiling temperature.
  • a given reaction can be carried out in one solvent or a mixture of more than one solvent.
  • suitable solvents for a particular reaction step can be selected.
  • Suitable resolving agents for fractional recrystallization methods are, for example, optically active acids, such as the D and L forms of tartaric acid, diacetyltartaric acid, dibenzoyltartaric acid, mandelic acid, malic acid, lactic acid or the various optically active camphorsulfonic acids.
  • Resolution of racemic mixtures can also be carried out by elution on a column packed with an optically active resolving agent (e.g., dinitrobenzoylphenylglycine).
  • an optically active resolving agent e.g., dinitrobenzoylphenylglycine
  • Suitable elution solvent composition can be determined by one skilled in the art.
  • Modified nucleosides and nucleotides can be prepared according to the synthetic methods described in Ogata et al, J. Org. Chem. 74:2585-2588 (2009); Purmal et al, Nucl. Acids Res. 22(1): 72-78, (1994); Fukuhara et al, Biochemistry, 1(4): 563-568 (1962); and Xu et al, Tetrahedron, 48(9): 1729-1740 (1992), each of which are incorporated by reference in their entirety.
  • the modified nucleic acid and mmRNA of the invention need not be uniformly modified along the entire length of the molecule.
  • one or more or all types of nucleotide e.g., purine or pyrimidine, or any one or more or all of A, G, U, C
  • nucleotides X in a polynucleotide of the invention are modified, wherein X may any one of nucleotides A, G, U, C, or any one of the combinations A+G, A+U, A+C, G+U, G+C, U+C, A+G+U, A+G+C, G+U+C or A+G+C
  • nucleotide modifications may exist at various positions in the modified nucleic acid or mmRNA.
  • nucleotide analogs or other modification(s) may be located at any position(s) of a modified nucleic acid or mmRNA such that the function of the modified nucleic acid or mmRNA is not substantially decreased.
  • a modification may also be a 5 ' or 3 ' terminal modification.
  • the modified nucleic acid or mmRNA may contain from about 1 % to about 100% modified nucleotides, or any intervening percentage (e.g., from 1% to 20%>, from 1% to 25%, from 1% to 50%, from 1% to 60%, from 1% to 70%, from 1% to 80%, from 1% to 90%, from 1% to 95%, from 10% to 20%, from 10% to 25%, from 10% to 50%, from 10% to 60%, from 10% to 70%, from 10% to 80%, from 10% to 90%, from 10% to 95%, from 10% to 100%, from 20% to 25%, from 20% to 50%, from 20% to 60%, from 20% to 70%, from 20% to 80%, from 20% to 90%, from 20% to 95%, from 20% to 100%, from 50% to 60%, from 50% to 70%, from 50% to 80%, from 50% to 90%, from 50% to 95%, from 50% to 100%, from 70% to 80%, from 70% to 90%, from 70% to 95%, from 70% to 100%, from 80% to 90%, from 80% to 95%, from 90% to 95%, from 70% to
  • the modified nucleic acid or mmR A includes a modified pyrimidine (e.g., a modified uracil/uridine or modified cytosine/cytidine).
  • the uracil or uridine in the modified nucleic acid or mmRNA molecule may be replaced with from about P/o to about 100% of a modified uracil or modified uridine (e.g., from 1% to 20%>, from 1% to 25%, from 1% to 50%, from 1% to 60%, from 1% to 70%, from 1% to 80%, from 1% to 90%, from 1% to 95%, from 10% to 20%, from 10% to 25%, from 10% to 50%, from 10% to 60%, from 10% to 70%, from 10% to 80%, from 10% to 90%, from 10% to 95%, from 10% to 100%, from 20% to 25%, from 20% to 50%, from 20% to 60%, from 20% to 70%, from 20% to 80%, from 20% to 90%, from 20% to 95%, from 20% to 100%, from 20% to 25%, from 20% to 50%, from 20%
  • the modified uracil or uridine can be replaced by a compound having a single unique structure or by a plurality of compounds having different structures (e.g., 2, 3, 4 or more unique structures, as described herein).
  • the cytosine or cytidine in the modified nucleic acid or mmRNA molecule may be replaced with from about 1% to about 100% of a modified cytosine or modified cytidine (e.g., from 1% to 20%>, from 1% to 25%, from 1% to 50%, from 1% to 60%, from 1% to 70%, from 1% to 80%, from 1% to 90%, from 1% to 95%, from 10% to 20%, from 10% to 25%, from 10% to 50%, from 10% to 60%, from 10% to 70%, from 10% to 80%, from 10% to 90%, from 10% to 95%, from 10% to 100%, from 20% to 25%, from 20% to 50%, from 20% to 60%, from 20% to 70%, from 20% to 80%, from 20% to 90%, from 20% to 95%, from 20% to 100%, from 20% to
  • the modified cytosine or cytidine can be replaced by a compound having a single unique structure or by a plurality of compounds having different structures (e.g., 2, 3, 4 or more unique structures, as described herein).
  • the present disclosure provides methods of synthesizing a modified nucleic a id or mmRNA including n number of linked nucleosides having Formula (Ia-1):
  • Y 9 is H, hydroxy, phosphoryl, pyrophosphate, sulfate, amino, thiol, optionally substituted amino acid, or a peptide (e.g., including from 2 to 12 amino acids); and each P 1 , P 2 , and P 3 is, independently, a suitable protecting group; and @ denotes a solid support;
  • steps a) and b) are repeated from 1 to about 10,000 times.
  • the methods further comprise a nucleotide (e.g., building block molecule) selected from the group consisting of adenosine, cytosine, guanosine, and uracil.
  • the nucleobase may be a pyrimidine or derivative thereof.
  • the modified nucleic acid or mmRNA is translatable.
  • modified nucleic acids and mmRNA are optional, and are beneficial in some embodiments.
  • a 5' untranslated region (UTR) and/or a 3'UTR are provided, wherein either or both may independently contain one or more different nucleoside modifications.
  • nucleoside modifications may also be present in the translatable region.
  • modified nucleic acids and mmRNA containing a Kozak sequence are provided below in Scheme 1 through Scheme 11.
  • Scheme 1 provides a general method for phosphorylation of nucleosides, including modified nucleosides.
  • Scheme 2 provides the use of multiple protecting and deprotecting steps to promote phosphorylation at the 5' position of the sugar, rather than the 2' and 3' hydroxyl groups.
  • Modified nucleotides can be synthesized in any useful manner.
  • Schemes 3, 4, and 7 provide exemplary methods for synthesizing modified nucleotides having a modified purine nucleobase; and
  • Schemes 5 and 6 provide exemplary methods for synthesizing modified nucleotides having a modified pseudouridine or pseudoisocytidine, respectively.
  • Schemes 8 and 9 provide exemplary syntheses of modified nucleotides.
  • Scheme 10 provides a non-limiting biocatalytic method for producing nucleotides.
  • Scheme 11 provides an exemplary synthesis of a modified uracil, where the Nl position is modified with R 12b , as provided elsewhere, and the 5'-position of ribose is phosphorylated.
  • T 1 , T 2 , R 12a , R 12b , and r are as provided herein.
  • This synthesis, as well as optimized versions thereof, can be used to modify other pyrimidine nucleobases and purine nucleobases (see e.g., Formulas (bl)- (b43)) and/or to install one or more phosphate groups (e.g., at the 5' position of the sugar).
  • This alkylating reaction can also be used to include one or more optionally substituted alkyl group at any reactive group (e.g., amino group) in any nucleobase described herein (e.g., the amino groups in the Watson-Crick base-pairing face for cytosine, uracil, adenine, and guanine).
  • any reactive group e.g., amino group
  • nucleobase described herein e.g., the amino groups in the Watson-Crick base-pairing face for cytosine, uracil, adenine, and guanine.
  • modified nucleotides and modified nucleotide combinations are provided below in Table 2. These combinations of modified nucleotides can be used to form the modified nucleic acids or mmRNA of the invention. Unless otherwise noted, the modified nucleotides may be completely substituted for the natural nucleotides of the modified nucleic acids or mmRNA of the invention. As a non-limiting example, the natural nucleotide uridine may be substituted with a modified nucleoside described herein.
  • the natural nucleotide uridine may be partially substituted (e.g., about 0.1%, 1%, 5%, 10%>, 15%, 20%>, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 99.9%) with at least one of the modified nucleoside disclosed herein.
  • cytosines are pyrrolo-cytidine
  • cytosines are 5 -methyl-cytidine
  • cytosines are 5 -methyl-cytidine
  • uridines are 5 -methyl-cytidine/ about 50% of uridines are 2-thio- uridine
  • cytosines are N4-acetyl-cytidine
  • cytosines are N4-acetyl-cytidine
  • cytosines are N4-acetyl-cytidine/ about 50% of uridines are 2- thio-uridine
  • modified nucleotide combinations are provided below in Table 3. These combinations of modified nucleotides can be used to form the modified nucleic acid molecules or mmRNA of the invention.
  • At least 25% of the cytosines are replaced by a compound of Formula (blO)-(M4) (e.g., at least about 30%>, at least about 35%>, at least about 40%>, at least about 45%), at least about 50%>, at least about 55%>, at least about 60%>, at least about 65%>, at least about 70%), at least about 75%>, at least about 80%>, at least about 85%>, at least about 90%>, at least about 95%, or about 100%).
  • a compound of Formula (blO)-(M4) e.g., at least about 30%>, at least about 35%>, at least about 40%>, at least about 45%
  • at least about 50%> at least about 55%>, at least about 60%>, at least about 65%>, at least about 70%
  • at least about 75%> at least about 80%>, at least about 85%>, at least about 90%>, at least about 95%, or about 100%.
  • At least 25% of the uracils are replaced by a compound of Formula (bl)-(b9) (e.g., at least about 30%>, at least about 35%>, at least about 40%>, at least about 45%>, at least about 50%), at least about 55%>, at least about 60%>, at least about 65%>, at least about 70%>, at least about 75%), at least about 80%>, at least about 85%>, at least about 90%>, at least about 95%>, or about 100%).
  • a compound of Formula (bl)-(b9) e.g., at least about 30%>, at least about 35%>, at least about 40%>, at least about 45%>, at least about 50%
  • at least about 55%> at least about 60%>, at least about 65%>, at least about 70%>, at least about 75
  • at least about 80%>, at least about 85%>, at least about 90%>, at least about 95%>, or about 100% e.g., at least about 30%>, at least about
  • At least 25%> of the cytosines are replaced by a compound of Formula (bl0)-(bl4), and at least 25% of the uracils are replaced by a compound of Formula (bl)- (b9) (e.g., at least about 30%>, at least about 35%>, at least about 40%>, at least about 45%>, at least about 50%), at least about 55%>, at least about 60%>, at least about 65%>, at least about 70%>, at least about 75%), at least about 80%>, at least about 85%>, at least about 90%>, at least about 95%>, or about 100%).
  • Modified nucleic acid molecules for use in accordance with the present disclosure may be prepared according to any available technique including, but not limited to, in vitro transcription such as chemical synthesis and enzymatic synthesis, or enzymatic and chemical cleavage of a longer precursor, etc.
  • Methods of synthesizing RNA are known in the art (see, e.g., Gait, M.J. (ed.) Oligonucleotide synthesis: a practical approach, Oxford [Oxfordshire], Washington, DC: IRL Press, 1984; and Herdewijn, P. (ed.) Oligonucleotide synthesis: methods and applications, Methods in Molecular Biology, v. 288 (Clifton, N.J.) Totowa, N.J.: Humana Press, 2005; both of which are incorporated herein by reference).
  • modified nucleic acid molecules disclosed herein can be prepared from readily available starting materials using the following general methods and procedures. It is understood that where typical or preferred process conditions (i.e., reaction temperatures, times, mole ratios of reactants, solvents, pressures, etc.) are given other process conditions can also be used unless otherwise stated. Optimum reaction conditions may vary with the particular reactants or solvent used, but such conditions can be determined by one skilled in the art by routine optimization procedures.
  • spectroscopic means such as nuclear magnetic resonance spectroscopy (e.g., 1H or 13 C) infrared spectroscopy, spectrophotometry (e.g., UV-visible), mass spectrometry, or by chromatography such as high performance liquid chromatography (HPLC) or thin layer chromatography.
  • HPLC high performance liquid chromatography
  • Modification of modified nucleic acid molecules can involve the protection and deprotection of various chemical groups.
  • the need for protection and deprotection, and the selection of appropriate protecting groups can be readily determined by one skilled in the art.
  • the chemistry of protecting groups can be found, for example, in Greene, et al, Protective Groups in Organic
  • the reactions of the processes described herein can be carried out in suitable solvents, which can be readily selected by one of skill in the art of organic synthesis.
  • suitable solvents can be substantially nonreactive with the starting materials (reactants), the intermediates, or products at the temperatures at which the reactions are carried out, i.e., temperatures which can range from the solvent's freezing temperature to the solvent's boiling temperature.
  • a given reaction can be carried out in one solvent or a mixture of more than one solvent.
  • suitable solvents for a particular reaction step can be selected.
  • Resolution of racemic mixtures of modified nucleic acid molecules can be carried out by any of numerous methods known in the art.
  • An example method includes, but is not limited to, fractional recrystallization using a "chiral resolving acid" which is an optically active, salt-forming organic acid.
  • Suitable resolving agents for fractional recrystallization methods are, for example, optically active acids, such as the D and L forms of tartaric acid, diacetyltartaric acid,
  • dibenzoyltartaric acid mandelic acid, malic acid, lactic acid or the various optically active camphorsulfonic acids.
  • Resolution of racemic mixtures can also be carried out by elution on a column packed with an optically active resolving agent (e.g., dinitrobenzoylphenylglycine).
  • an optically active resolving agent e.g., dinitrobenzoylphenylglycine
  • Suitable elution solvent composition can be determined by one skilled in the art.
  • Modified nucleic acid molecules need not be uniformly modified along the entire length of the molecule. Different nucleic acid modifications and/or backbone structures may exist at various positions in the nucleic acid. One of ordinary skill in the art will appreciate that the nucleotide analogs or other modification(s) may be located at any position(s) of a nucleic acid such that the function of the nucleic acid is not substantially decreased. A modification may also be a 5 Or 3' terminal modification.
  • the nucleic acids may contain at a minimum one modified nucleotide and at maximum 100% modified nucleotides, or any intervening percentage, such as at least 5% modified nucleotides, at least 10% modified nucleotides, at least 25% modified nucleotides, at least 50% modified nucleotides, at least 80% modified nucleotides, or at least 90% modified nucleotides.
  • the nucleic acids may contain a modified pyrimidine such as uracil or cytosine.
  • at least 5%, at least 10%>, at least 25%, at least 50%, at least 80%, at least 90% or 100% of the uracil in the nucleic acid may be replaced with a modified uracil.
  • the modified uracil can be replaced by a compound having a single unique structure, or can be replaced by a plurality of compounds having different structures (e.g., 2, 3, 4 or more unique structures).
  • At least 5%, at least 10%, at least 25%, at least 50%, at least 80%, at least 90% or 100% of the cytosine in the nucleic acid may be replaced with a modified cytosine.
  • the modified cytosine can be replaced by a compound having a single unique structure, or can be replaced by a plurality of compounds having different structures (e.g., 2, 3, 4 or more unique structures).
  • the shortest length of a modified mRNA, herein "mmRNA,” of the present disclosure can be the length of an mRNA sequence that may be sufficient to encode for a dipeptide. In another embodiment, the length of the mRNA sequence may be sufficient to encode for a tripeptide. In another embodiment, the length of an mRNA sequence may be sufficient to encode for a tetrapeptide. In another embodiment, the length of an mRNA sequence may be sufficient to encode for a pentapeptide. In another embodiment, the length of an mRNA sequence may be sufficient to encode for a hexapeptide. In another embodiment, the length of an mRNA sequence may be sufficient to encode for a heptapeptide.
  • the length of an mRNA sequence may be sufficient to encode for an octapeptide. In another embodiment, the length of an mRNA sequence may be sufficient to encode for a nonapeptide. In another embodiment, the length of an mRNA sequence may be sufficient to encode for a decapeptide.
  • Examples of dipeptides that the modified nucleic acid molecule sequences can encode for include, but are not limited to, carnosine and anserine.
  • the mRNA may be greater than 30 nucleotides in length. In another embodiment, the RNA molecule may be greater than 35 nucleotides in length (e.g., at least or greater than about 35, 40, 45, 50, 55, 60, 70, 80, 90, 100, 120, 140, 160, 180, 200, 250, 300, 350, 400, 450, 500, 600, 700, 800, 900, 1,000, 1,100, 1,200, 1,300, 1,400, 1,500, 1,600, 1,700, 1,800, 1,900, 2,000, 2,500, 3,000, 4,000 and 5,000 nucleotides).
  • the RNA molecule may be greater than 35 nucleotides in length (e.g., at least or greater than about 35, 40, 45, 50, 55, 60, 70, 80, 90, 100, 120, 140, 160, 180, 200, 250, 300, 350, 400, 450, 500, 600, 700, 800, 900, 1,000, 1,100, 1,200, 1,300, 1,400, 1,500, 1,600,
  • modified nucleic acid molecules e.g., modified mRNA (mmRNA)
  • mmRNA modified mRNA
  • RNA ligands comprising modified nucleotides or modified nucleic acid molecules, as described herein, decrease interactions with major groove binding partners, and therefore decrease an innate immune response, or expression and secretion of pro-inflammatory cytokines, or both.
  • Example major groove interacting, e.g. binding, partners include, but are not limited to, the following nucleases and helicases.
  • TLRs Toll-like Receptors
  • helicases Within membranes, TLRs (Toll-like Receptors) 3, 7, and 8 can respond to single- and double-stranded RNA.
  • members of the superfamily 2 class of DE (D/H) helicases and ATPases can sense RNA to initiate antiviral responses.
  • These helicases include the RIG-I (retinoic acid-inducible gene I) and MDA5 (melanoma differentiation- associated gene 5).
  • Other examples include laboratory of genetics and physiology 2 (LGP2), HIN- 200 domain containing proteins, or Helicase-domain containing proteins.
  • the modified nucleic acid molecules decrease the innate immune response in a cell.
  • innate immune response includes a cellular response to exogenous nucleic acids, including, but not limited to, single stranded nucleic acids, generally of viral or bacterial origin, which involve the induction of cytokine expression and release, particularly the interferons, and cell death. Protein synthesis may also be reduced during the innate cellular immune response. While it is advantageous to eliminate the innate immune response in a cell, the present disclosure provides modified mRNA that substantially reduce the immune response, including interferon signaling, without entirely eliminating such a response.
  • the immune response may be reduced by 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99%), 99.9%), or greater than 99.9% as compared to the immune response induced by a corresponding unmodified nucleic acid molecule.
  • a reduction can be measured by the expression or activity level of Type 1 interferons or the expression of interferon-regulated genes such as the toll-like receptors ⁇ e.g., TLR7 and TLR8).
  • Reduction of the innate immune response can also be measured by decreased cell death following one or more administrations of modified RNA to a cell population; e.g., cell death is 10%, 25%, 50%, 75%, 85%, 90%, 95%, or over 95% less than the cell death frequency observed with a corresponding unmodified nucleic acid molecule.
  • cell death may affect fewer than 50%, 40%, 30%, 20%, 10%, 5%, 1%, 0.1%, 0.01% or fewer than 0.01% of cells contacted with the modified nucleic acid molecules.
  • the present disclosure provides for the repeated introduction ⁇ e.g. , transfection) of modified nucleic acid molecules into a target cell population, e.g., in vitro, ex vivo, or in vivo.
  • the step of contacting the cell population may be repeated one or more times (such as two, three, four, five or more than five times).
  • the step of contacting the cell population with the modified nucleic acid molecules may be repeated a number of times sufficient such that a predetermined efficiency of protein translation in the cell population is achieved. Given the reduced cytotoxicity of the target cell population by the nucleic acid modifications, such repeated
  • transfections are achievable in a variety of cell types.
  • modified nucleic acids of the invention may have superior properties making them more suitable as therapeutic modalities.
  • the present inventors have determined that to improve protein production, one may consider the nature of the modification, or combination of modifications, the percent modification and survey more than one cytokine or metric to determine the efficacy and risk profile of a particular modified mRNA.
  • methods of determining the effectiveness of a modified mRNA as compared to unmodified involves the measure and analysis of one or more cytokines whose expression is triggered by the administration of the exogenous nucleic acid of the invention. These values are compared to administration of an umodified nucleic acid or to a standard metric such as cytokine response, PolylC, R-848 or other standard known in the art.
  • One example of a standard metric developed herein is the measure of the ratio of the level or amount of encoded polypeptide (protein) produced in the cell, tissue or organism to the level or amount of one or more (or a panel) of cytokines whose expression is triggered in the cell, tissue or organism as a result of administration or contact with the modified nucleic acid.
  • Such ratios are referred to herein as the Protein: Cytokine Ratio or "PC" Ratio.
  • PC ratio Cytokine Ratio
  • the higher the PC ratio the more efficacioius the modified nucleic acid (polynucleotide encoding the protein measured).
  • Preferred PC Ratios, by cytokine, of the present invention may be greater than 1, greater than 10, greater than 100, greater than 1000, greater than 10,000 or more. Modified nucleic acids having higher PC Ratios than a modified nucleic acid of a different or unmodified construct are preferred.
  • the PC ratio may be further qualified by the percent modification present in the polynucleotide. For example, normalized to a 100% modified nucleic acid, the protein production as a function of cytokine (or risk) or cytokine profile can be determined.
  • the present invention provides a method for determining, across chemistries, cytokines or percent modification, the relative efficacy of any particular modified polynucleotide by comparing the PC Ratio of the modified nucleic acid (polynucleotide).
  • mRNA molecules may be used to elicit or provoke an immune response in an organism.
  • the mRNA molecules to be delivered may encode an immunogenic peptide or polypeptide and may encode more than one such peptide or polypeptide.
  • certain modified nucleosides, or combinations thereof, when introduced into the modified nucleic acid molecules or mmRNA of the invention will activate the innate immune response.
  • Such activating molecules are useful as adjuvants when combined with polypeptides and/or other vaccines.
  • the activating molecules contain a translatable region which encodes for a polypeptide sequence useful as a vaccine, thus providing the ability to be a self-adjuvant.
  • the modified nucleic acid molecules and/or mmRNA of the invention may encode an immunogen.
  • the delivery of modified nucleic acid molecules and/or mmRNA encoding an immunogen may activate the immune response.
  • the modified nucleic acid molecules and/or mmRNA encoding an immunogen may be delivered to cells to trigger multiple innate response pathways (see International Pub. No. WO2012006377; herein incorporated by reference in its entirety).
  • the modified nucleic acid molecules and mmRNA of the present invention encoding an immunogen may be delivered to a vertebrate in a dose amount large enough to be immunogenic to the vertebrate (see International Pub. No. WO2012006372 and WO2012006369; each of which is herein incorporated by reference in their entirety).
  • the modified nucleic acid molecules or mmRNA of invention may encode a polypeptide sequence for a vaccine and may further comprise an inhibitor.
  • the inhibitor may impair antigen presentation and/or inhibit various pathways known in the art.
  • the modified nucleic acid molecules or mmRNA of the invention may be used for a vaccine in combination with an inhibitor which can impair antigen presentation (see International Pub. No. WO2012089225 and WO2012089338; each of which is herein incorporated by reference in their entirety).
  • the modified nucleic acid molecules or mmRNA of the invention may be self-replicating RNA.
  • Self-replicating RNA molecules can enhance efficiency of RNA delivery and expression of the enclosed gene product.
  • the modified nucleic acid molecules or mmRNA may comprise at least one modification described herein and/or known in the art.
  • the self-replicating RNA can be designed so that the self-replicating RNA does not induce production of infectious viral particles.
  • the self- replicating RNA may be designed by the methods described in US Pub. No. US20110300205 and International Pub. No. WO2011005799, each of which is herein incorporated by reference in their entirety.
  • the self-replicating modified nucleic acid molecules or mmRNA of the invention may encode a protein which may raise the immune response.
  • the modified nucleic acid molecules and/or mmRNA may be self-replicating mRNA may encode at least one antigen (see US Pub. No. US20110300205 and International Pub. No. WO2011005799; each of which is herein incorporated by reference in their entirety).
  • the self-replicating modified nucleic acids or mmRNA of the invention may be formulated using methods described herein or known in the art.
  • the self-replicating RNA may be formulated for delivery by the methods described in Geall et al (Nonviral delivery of self-amplifying RNA vaccines, PNAS 2012; PMID: 22908294; herein incorporated by reference in its entirety ).
  • the modified nucleic acid molecules or mmRNA of the present invention may encode amphipathic and/or immunogenic amphipathic peptides.
  • a formulation of the modified nucleic acid molecules or mmRNA of the present invention may further comprise an amphipathic and/or immunogenic amphipathic peptide.
  • the modified nucleic acid molecule or mmRNA comprising an amphipathic and/or immunogenic amphipathic peptide may be formulated as described in US. Pub. No. US20110250237 and International Pub. Nos. WO2010009277 and WO2010009065; each of which is herein incorporated by reference in their entirety.
  • the modified nucleic acid molecules and mmRNA of the present invention may be immunostimultory.
  • the modified nucleic acid molecules and mmRNA may encode all or a part of a positive-sense or a negative-sense stranded RNA virus genome (see International Pub No. WO2012092569 and US Pub No. US20120177701, each of which is herein incorporated by reference in their entirety).
  • the immunostimultory modified nucleic acid molecules or mmRNA of the present invention may be formulated with an excipient for administration as described herein and/or known in the art (see International Pub No. WO2012068295 and US Pub No. US20120213812, each of which is herein incorporated by reference in their entirety).
  • the response of the vaccine formulated by the methods described herein may be enhanced by the addition of various compounds to induce the therapeutic effect.
  • the vaccine formulation may include a MHC II binding peptide or a peptide having a similar sequence to a MHC II binding peptide (see International Pub Nos. WO2012027365, WO201 1031298 and US Pub No. US20120070493, US201 101 10965, each of which is herein incorporated by reference in their entirety).
  • the vaccine formulations may comprise modified nicotinic compounds which may generate an antibody response to nicotine residue in a subject (see International Pub No. WO2012061717 and US Pub No. US201201 14677, each of which is herein incorporated by reference in their entirety).
  • the modified nucleic acid molecules encode polypeptides, e.g., a variant polypeptides, which have a certain identity to a reference polypeptide sequence.
  • identity refers to a relationship between the sequences of two or more peptides, determined by comparing the sequences. In the art, “identity” also refers to the degree of sequence relatedness between peptides, as determined by the number of matches between strings of two or more amino acid residues. Identity measures the percent of identical matches between the smaller of two or more sequences with gap alignments (if any) addressed by a particular mathematical model or computer program (i.e., "algorithms"). Identity of related peptides can be readily calculated by known methods.
  • Such methods include, but are not limited to, those described in Computational Molecular Biology, Lesk, A. M., ed., Oxford University Press, New York, 1988; Biocomputing: Informatics and Genome Projects, Smith, D. W., ed., Academic Press, New York, 1993; Computer Analysis of Sequence Data, Part 1 , Griffin, A. M., and Griffin, H. G., eds., Humana Press, New Jersey, 1994; Sequence Analysis in Molecular Biology, von Heinje, G., Academic Press, 1987; Sequence Analysis Primer, Gribskov, M. and Devereux, J., eds., M. Stockton Press, New York, 1991 ; and Carillo et al, SIAM J. Applied Math. 48, 1073 (1988); all of which are herein incorporated by reference in their entirety.
  • the polypeptide variant may have the same or a similar activity as the reference polypeptide.
  • the variant may have an altered activity (e.g., increased or decreased) relative to a reference polypeptide.
  • variants of a particular polynucleotide or polypeptide of the present disclosure will have at least about 40%, 45%, 50%>, 55%>, 60%>, 65%>, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity to that particular reference polynucleotide or polypeptide as determined by sequence alignment programs and parameters described herein and known to those skilled in the art.
  • protein fragments, functional protein domains, and homologous proteins are also considered to be within the scope of this present disclosure.
  • a protein fragment of a reference protein meaning a polypeptide sequence which is at least one amino acid residue shorter than a reference polypeptide sequence but otherwise identical
  • 10, 20, 30, 40, 50, 60, 70, 80, 90, 100 or greater than 100 amino acids in length is provided herein.
  • any protein that includes a stretch of about 20, about 30, about 40, about 50, or about 100 amino acids which are about 40%, about 50%, about 60%>, about 70%>, about 80%>, about 90%), about 95%o, or about 100% identical to any of the sequences described herein can be utilized in accordance with the present disclosure.
  • a protein sequence to be utilized in accordance with the present disclosure includes 2, 3, 4, 5, 6, 7, 8, 9, 10, or more mutations as shown in any of the sequences provided or referenced herein.
  • Proper protein translation involves the physical aggregation of a number of polypeptides and nucleic acids associated with the mRNA.
  • Provided by the present disclosure are protein-nucleic acid complexes, containing a translatable mRNA having one or more nucleoside modifications (e.g., at least two different nucleoside modifications) and one or more polypeptides bound to the mRNA.
  • the proteins are provided in an amount effective to prevent or to reduce an innate immune response of a cell into which the complex is introduced.
  • mRNA having sequences that are substantially not translatable may be effective as a vaccine when administered to a subject. It is further provided that the subject administered the vaccine may be a mammal, more preferably a human and most preferably a patient.
  • modified nucleic acid molecules that contain one or more noncoding regions. Such modified nucleic acid molecules are generally not translated, but are capable of binding to and sequestering one or more translational machinery component such as a ribosomal protein or a transfer RNA (tRNA), thereby effectively reducing the protein expression in the cell.
  • the modified nucleic acid molecule may contain a small nucleolar RNA (sno-RNA), micro RNA (miRNA), small interfering RNA (siRNA) or Piwi-interacting RNA (piRNA).
  • the present invention provides modified nucleic acids and mmRNA compositions and complexes in combination with one or more pharmaceutically acceptable excipients.
  • Pharmaceutical compositions may optionally comprise one or more additional active substances, e.g. therapeutically and/or prophylactically active substances.
  • additional active substances e.g. therapeutically and/or prophylactically active substances.
  • General considerations in the formulation and/or manufacture of pharmaceutical agents may be found, for example, in Remington: The Science and Practice of Pharmacy 21 st ed., Lippincott Williams & Wilkins, 2005 (incorporated herein by reference in its entirety).
  • compositions are administered to humans, human patients or subjects.
  • active ingredient generally refers to modified nucleic acids and mmRNA to be delivered as described herein.
  • compositions are principally directed to pharmaceutical compositions which are suitable for administration to humans, it will be understood by the skilled artisan that such compositions are generally suitable for administration to any other animal, e.g., to non-human animals, e.g. non-human mammals. Modification of pharmaceutical compositions suitable for administration to humans in order to render the compositions suitable for administration to various animals is well understood, and the ordinarily skilled veterinary pharmacologist can design and/or perform such modification with merely ordinary, if any, experimentation. Subjects to which administration of the pharmaceutical compositions is contemplated include, but are not limited to, humans and/or other primates;
  • mammals including commercially relevant mammals such as cattle, pigs, horses, sheep, cats, dogs, mice, and/or rats; and/or birds, including commercially relevant birds such as poultry, chickens, ducks, geese, and/or turkeys.
  • Formulations of the pharmaceutical compositions described herein may be prepared by any method known or hereafter developed in the art of pharmacology. In general, such preparatory methods include the step of bringing the active ingredient into association with an excipient and/or one or more other accessory ingredients, and then, if necessary and/or desirable, dividing, shaping and/or packaging the product into a desired single- or multi-dose unit.
  • a pharmaceutical composition in accordance with the invention may be prepared, packaged, and/or sold in bulk, as a single unit dose, and/or as a plurality of single unit doses.
  • a "unit dose" is discrete amount of the pharmaceutical composition comprising a predetermined amount of the active ingredient.
  • the amount of the active ingredient is generally equal to the dosage of the active ingredient which would be administered to a subject and/or a convenient fraction of such a dosage such as, for example, one-half or one -third of such a dosage.
  • compositions in accordance with the invention will vary, depending upon the identity, size, and/or condition of the subject treated and further depending upon the route by which the composition is to be administered.
  • the composition may comprise between 0.1% and 100%, e.g., between .5 and 50%>, between 1-30%, between 5-80%>, at least 80%> (w/w) active ingredient.
  • the modified nucleic acid, and mmRNA of the invention can be formulated using one or more excipients to: (1) increase stability; (2) increase cell transfection; (3) permit the sustained or delayed release (e.g., from a depot formulation of the modified nucleic acid, or mmRNA); (4) alter the biodistribution (e.g., target the modified nucleic acid, or mmRNA to specific tissues or cell types); (5) increase the translation of encoded protein in vivo; and/or (6) alter the release profile of encoded protein in vivo.
  • excipients to: (1) increase stability; (2) increase cell transfection; (3) permit the sustained or delayed release (e.g., from a depot formulation of the modified nucleic acid, or mmRNA); (4) alter the biodistribution (e.g., target the modified nucleic acid, or mmRNA to specific tissues or cell types); (5) increase the translation of encoded protein in vivo; and/or (6) alter the release profile of encoded protein in vivo.
  • excipients of the present invention can include, without limitation, lipidoids, liposomes, lipid nanoparticles, polymers, lipoplexes, core- shell nanoparticles, peptides, proteins, cells transfected with modified nucleic acid, or mmRNA (e.g., for transplantation into a subject), hyaluronidase, nanoparticle mimics and combinations thereof.
  • the formulations of the invention can include one or more excipients, each in an amount that together increases the stability of the modified nucleic acid, or mmRNA, increases cell transfection by the modified nucleic acid, or mmRNA, increases the expression of modified nucleic acid, or mmRNA encoded protein, and/or alters the release profile of modified nucleic acid, or mmRNA encoded proteins.
  • the modified nucleic acids and mmRNA of the present invention may be formulated using self-assembled nucleic acid nanoparticles.
  • Formulations of the pharmaceutical compositions described herein may be prepared by any method known or hereafter developed in the art of pharmacology. In general, such preparatory methods include the step of associating the active ingredient with an excipient and/or one or more other accessory ingredients.
  • a pharmaceutical composition in accordance with the present disclosure may be prepared, packaged, and/or sold in bulk, as a single unit dose, and/or as a plurality of single unit doses.
  • a "unit dose" refers to a discrete amount of the pharmaceutical composition comprising a predetermined amount of the active ingredient.
  • the amount of the active ingredient may generally be equal to the dosage of the active ingredient which would be administered to a subject and/or a convenient fraction of such a dosage including, but not limited to, one-half or one-third of such a dosage.
  • Relative amounts of the active ingredient, the pharmaceutically acceptable excipient, and/or any additional ingredients in a pharmaceutical composition in accordance with the present disclosure may vary, depending upon the identity, size, and/or condition of the subject being treated and further depending upon the route by which the composition is to be administered.
  • the composition may comprise between 0.1% and 99% (w/w) of the active ingredient.
  • the modified mRNA formulations described herein may contain at least one modified mRNA.
  • the formulations may contain 1, 2, 3, 4 or 5 modified mRNA.
  • the formulation contains at least three modified mRNA encoding proteins.
  • the formulation contains at least five modified mRNA encoding proteins.
  • compositions may additionally comprise a pharmaceutically acceptable excipient, which, as used herein, includes, but is not limited to, any and all solvents, dispersion media, diluents, or other liquid vehicles, dispersion or suspension aids, surface active agents, isotonic agents, thickening or emulsifying agents, preservatives, and the like, as suited to the particular dosage form desired.
  • a pharmaceutically acceptable excipient includes, but is not limited to, any and all solvents, dispersion media, diluents, or other liquid vehicles, dispersion or suspension aids, surface active agents, isotonic agents, thickening or emulsifying agents, preservatives, and the like, as suited to the particular dosage form desired.
  • excipients for formulating pharmaceutical compositions and techniques for preparing the composition are known in the art (see Remington: The Science and Practice of Pharmacy, 21 st Edition, A. R. Gennaro, Lippincott, Williams & Wilkins, Baltimore, MD,
  • any conventional excipient medium may be contemplated within the scope of the present disclosure, except insofar as any conventional excipient medium may be incompatible with a substance or its derivatives, such as by producing any undesirable biological effect or otherwise interacting in a deleterious manner with any other component(s) of the pharmaceutical composition.
  • the particle size of the lipid nanoparticle may be increased and/or decreased.
  • the change in particle size may be able to help counter biological reaction such as, but not limited to, inflammation or may increase the biological effect of the modified mRNA delivered to mammals.
  • compositions include, but are not limited to, inert diluents, surface active agents and/or emulsifiers, preservatives, buffering agents, lubricating agents, and/or oils. Such excipients may optionally be included in the pharmaceutical formulations of the invention.
  • the present disclosure describes their formulation and use in delivering single stranded modified nucleic acid molecules or mmRNA.
  • Complexes, micelles, liposomes or particles can be prepared containing these lipidoids and therefore, can result in an effective delivery of the modified nucleic acid molecules or mmRNA, as judged by the production of an encoded protein, following the injection of a lipidoid formulation via localized and/or systemic routes of
  • Lipidoid complexes of modified nucleic acid molecules or mmRNA can be administered by various means including, but not limited to, intravenous, intramuscular, or subcutaneous routes.
  • nucleic acids may be affected by many parameters, including, but not limited to, the formulation composition, nature of particle PEGylation, degree of loading, oligonucleotide to lipid ratio, and biophysical parameters such as, but not limited to, particle size (Akinc et al., Mol Ther. 2009 17:872-879; herein incorporated by reference in its entirety).
  • particle size Akinc et al., Mol Ther. 2009 17:872-879; herein incorporated by reference in its entirety.
  • small changes in the anchor chain length of poly(ethylene glycol) (PEG) lipids may result in significant effects on in vivo efficacy.
  • Formulations with the different lipidoids including, but not limited to penta[3-(l-laurylaminopropionyl)]-triethylenetetramine hydrochloride (TETA-5LAP; aka 98N12-5, see Murugaiah et al., Analytical Biochemistry, 401 :61 (2010); herein incorporated by reference in its entirety), CI 2-200 (including derivatives and variants), and MDl, can be tested for in vivo activity.
  • TETA-5LAP penta[3-(l-laurylaminopropionyl)]-triethylenetetramine hydrochloride
  • CI 2-200 including derivatives and variants
  • MDl can be tested for in vivo activity.
  • the lipidoid referred to herein as "CI 2-200" is disclosed by Love et al, Proc Natl Acad Sci U S A. 2010 107: 1864-1869 and Liu and Huang, Molecular Therapy. 2010 669-670 (see Figure 1); both of which are herein incorporated by reference in their entirety.
  • the lipidoid formulations can include particles comprising either 3 or 4 or more components in addition to modified nucleic acid molecules or mmRNA.
  • formulations with certain lipidoids include, but are not limited to, 98N12-5 and may contain 42% lipidoid, 48% cholesterol and 10% PEG (CI 4 alkyl chain length).
  • formulations with certain lipidoids include, but are not limited to, CI 2-200 and may contain 50%> lipidoid, 10%> disteroylphosphatidyl choline, 38.5% cholesterol, and 1.5% PEG-DMG.
  • a modified nucleic acid molecule or mmRNA formulated with a lipidoid for systemic intravenous administration can target the liver.
  • a final optimized intravenous formulation using modified nucleic acid molecule or mmRNA, and comprising a lipid molar composition of 42% 98N12-5, 48% cholesterol, and 10% PEG-lipid with a final weight ratio of about 7.5 to 1 total lipid to modified nucleic acid, or mmRNA, and a C14 alkyl chain length on the PEG lipid, with a mean particle size of roughly 50-60 nm can result in the distribution of the formulation to be greater than 90% to the liver.
  • an intravenous formulation using a C12-200 see US provisional application 61/175,770 and published international application
  • lipidoid may have a molar ratio of 50/10/38.5/1.5 of C12-200/disteroylphosphatidyl choline/cholesterol/PEG- DMG, with a weight ratio of 7 to 1 total lipid to modified nucleic acid molecule or mmRNA, and a mean particle size of 80 nm may be effective to deliver modified nucleic acid molecule or mmRNA to hepatocytes (see, Love et al, Proc Natl Acad Sci U S A. 2010 107: 1864-1869 herein incorporated by reference in its entirety).
  • an MDl lipidoid-containing formulation may be used to effectively deliver modified nucleic acid molecule or mmRNA to hepatocytes in vivo.
  • the characteristics of optimized lipidoid formulations for intramuscular or subcutaneous routes may vary significantly depending on the target cell type and the ability of formulations to diffuse through the extracellular matrix into the blood stream. While a particle size of less than 150 nm may be desired for effective hepatocyte delivery due to the size of the endothelial fenestrae (see, Akinc et al., Mol Ther.
  • lipidoid- formulated modified nucleic acid molecules or mmRNA to deliver the formulation to other cells types including, but not limited to, endothelial cells, myeloid cells, and muscle cells may not be similarly size-limited.
  • Use of lipidoid formulations to deliver siRNA in vivo to other non-hepatocyte cells such as myeloid cells and endothelium has been reported (see Akinc et al., Nat Biotechnol. 2008 26:561-569; Leuschner et al, Nat Biotechnol. 2011 29: 1005-1010; Cho et al. Adv. Funct. Mater.
  • lipidoid formulations may have a similar component molar ratio. Different ratios of lipidoids and other components including, but not limited to, disteroylphosphatidyl choline, cholesterol and PEG-DMG, may be used to optimize the formulation of the modified nucleic acid, or mmRNA for delivery to different cell types including, but not limited to, hepatocytes, myeloid cells, muscle cells, etc.
  • the component molar ratio may include, but is not limited to, 50% C12-200, 10% disteroylphosphatidyl choline, 38.5% cholesterol, and %1.5 PEG-DMG (see
  • lipidoid formulations for the localized delivery of nucleic acids to cells (such as, but not limited to, adipose cells and muscle cells) via either subcutaneous or intramuscular delivery, may not require all of the formulation components desired for systemic delivery, and as such may comprise only the lipidoid and the modified nucleic acid molecule or mmRNA.
  • Combinations of different lipidoids may be used to improve the efficacy of modified nucleic acid molecule or mmRNA directed protein production as the lipidoids may be able to increase cell transfection by the modified nucleic acid molecule or mmRNA; and/or increase the translation of encoded protein (see Whitehead et al., Mol. Ther. 2011, 19: 1688-1694, herein incorporated by reference in its entirety).
  • Liposomes Liposomes, Lipoplexes, and Lipid Nanoparticles
  • modified nucleic acid molecules and mmRNA of the invention can be formulated using one or more liposomes, lipoplexes, or lipid nanoparticles.
  • pharmaceutical compositions of modified nucleic acid molecule or mmRNA include liposomes. Liposomes are artificially-prepared vesicles which may primarily be composed of a lipid bilayer and may be used as a delivery vehicle for the administration of nutrients and pharmaceutical formulations.
  • Liposomes can be of different sizes such as, but not limited to, a multilamellar vesicle (MLV) which may be hundreds of nanometers in diameter and may contain a series of concentric bilayers separated by narrow aqueous compartments, a small unicellular vesicle (SUV) which may be smaller than 50 nm in diameter, and a large unilamellar vesicle (LUV) which may be between 50 and 500 nm in diameter.
  • MLV multilamellar vesicle
  • SUV small unicellular vesicle
  • LUV large unilamellar vesicle
  • Liposome design may include, but is not limited to, opsonins or ligands in order to improve the attachment of liposomes to unhealthy tissue or to activate events such as, but not limited to, endocytosis.
  • Liposomes may contain a low or a high pH in order to improve the delivery of the pharmaceutical formulations.
  • liposomes may depend on the physicochemical characteristics such as, but not limited to, the pharmaceutical formulation entrapped and the liposomal ingredients , the nature of the medium in which the lipid vesicles are dispersed, the effective concentration of the entrapped substance and its potential toxicity, any additional processes involved during the application and/or delivery of the vesicles, the optimization size, polydispersity and the shelf-life of the vesicles for the intended application, and the batch-to-batch reproducibility and possibility of large-scale production of safe and efficient liposomal products.
  • compositions described herein may include, without limitation, liposomes such as those formed from 1 ,2-dioleyloxy-N,N-dimethylaminopropane (DODMA) liposomes, DiLa2 liposomes from Marina Biotech (Bothell, WA), l,2-dilinoleyloxy-3- dimethylaminopropane (DLin-DMA), 2,2-dilinoleyl-4-(2-dimethylaminoethyl)-[ 1 ,3]-dioxolane (DLin-KC2-DMA), and MC3 (US20100324120; herein incorporated by reference in its entirety) and liposomes which may deliver small molecule drugs such as, but not limited to, DOXIL® from Janssen Biotech, Inc.
  • DODMA dioleyloxy-N,N-dimethylaminopropane
  • DLin-DMA dimethylaminopropane
  • DLin-KC2-DMA 2,2-dilin
  • compositions described herein may include, without limitation, liposomes such as those formed from the synthesis of stabilized plasmid-lipid particles (SPLP) or stabilized nucleic acid lipid particle (SNALP) that have been previously described and shown to be suitable for oligonucleotide delivery in vitro and in vivo (see Wheeler et al. Gene Therapy. 1999 6:271-281; Zhang et al. Gene Therapy. 1999 6: 1438-1447; Jeffs et al. Pharm Res. 2005 22:362-372; Morrissey et al, Nat Biotechnol. 2005 2: 1002-1007;
  • liposomes such as those formed from the synthesis of stabilized plasmid-lipid particles (SPLP) or stabilized nucleic acid lipid particle (SNALP) that have been previously described and shown to be suitable for oligonucleotide delivery in vitro and in vivo (see Wheeler et al. Gene Therapy. 1999 6:271-281; Zhang et al
  • the liposome formulations are composed of 3 to 4 lipid components in addition to the modified nucleic acid molecule or mrnRNA.
  • a liposome can contain, but is not limited to, 55% cholesterol, 20% disteroylphosphatidyl choline (DSPC), 10% PEG-S-DSG, and 15% 1 ,2-dioleyloxy- N,N-dimethylaminopropane (DODMA), as described by Jeffs et al.
  • certain liposome formulations may contain, but are not limited to, 48% cholesterol, 20% DSPC, 2% PEG-c- DMA, and 30% cationic lipid, where the cationic lipid can be 1 ,2-distearloxy-N,N- dimethylaminopropane (DSDMA), DODMA, DLin-DMA, or l,2-dilinolenyloxy-3- dimethylaminopropane (DLenDMA), as described by Heyes et al.
  • DSDMA 1,2-distearloxy-N,N- dimethylaminopropane
  • DODMA DODMA
  • DLin-DMA 1,2-dilinolenyloxy-3- dimethylaminopropane
  • compositions may include liposomes which may be formed to deliver mrnRNA which may encode at least one immunogen.
  • the mrnRNA may be encapsulated by the liposome and/or it may be contained in an aqueous core which may then be encapsulated by the liposome (see International Pub. Nos. WO2012031046, WO2012031043, WO2012030901 and WO2012006378; each of which is herein incorporated by reference in their entirety).
  • the mrnRNA which may encode an immunogen may be formulated in a cationic oil-in- water emulsion where the emulsion particle comprises an oil core and a cationic lipid which can interact with the mrnRNA anchoring the molecule to the emulsion particle (see International Pub. No. WO2012006380; herein incorporated by reference in its entirety).
  • the lipid formulation may include at least cationic lipid, a lipid which may enhance transfection and a least one lipid which contains a hydrophilic head group linked to a lipid moiety (International Pub. No. WO2011076807 and U.S. Pub. No. 20110200582; each of which is herein incorporated by reference in their entirety).
  • the modified mRNA encoding an immunogen may be formulated in a lipid vesicle which may have crosslinks between functionalized lipid bilayers (see U.S. Pub. No. 20120177724, herein incorporated by reference in its entirety).
  • the modified mRNA may be formulated in a lipid vesicle which may have crosslinks between functionalized lipid bilayers.
  • the modified mRNA may be formulated in a lipid-polycation complex.
  • the formation of the lipid-polycation complex may be accomplished by methods known in the art and/or as described in U.S. Pub. No. 20120178702, herein incorporated by reference in its entirety.
  • the polycation may include a cationic peptide or a polypeptide such as, but not limited to, polylysine, polyornithine and/or polyarginine and the cationic peptides described in International Pub. No. WO2012013326; herein incorporated by reference in its entirety.
  • the modified mRNA may be formulated in a lipid-polycation complex which may further include a neutral lipid such as, but not limited to, cholesterol or dioleoyl
  • DOPE phosphatidylethanolamine
  • the liposome formulation may be influenced by, but not limited to, the selection of the cationic lipid component, the degree of cationic lipid saturation, the nature of the PEGylation, ratio of all components and biophysical parameters such as size.
  • the selection of the cationic lipid component the degree of cationic lipid saturation, the nature of the PEGylation, ratio of all components and biophysical parameters such as size.
  • the liposome formulation was composed of 57.1 % cationic lipid, 7.1% dipalmitoylphosphatidylcholme, 34.3 % cholesterol, and 1.4% PEG-c-DMA.
  • changing the composition of the cationic lipid could more effectively deliver siRNA to various antigen presenting cells (Basha et al. Mol Ther. 2011 19:2186-2200; herein incorporated by reference in its entirety).
  • the ratio of PEG in the lipid nanoparticle (LNP) formulations may be increased or decreased and/or the carbon chain length of the PEG lipid may be modified from C14 to C18 to alter the pharmacokinetics and/or biodistribution of the LNP formulations.
  • LNP formulations may contain 1-5% of the lipid molar ratio of PEG-c-DOMG as compared to the cationic lipid, DSPC and cholesterol.
  • the PEG-c-DOMG may be replaced with a PEG lipid such as, but not limited to, PEG- DSG (1,2-Distearoyl-sn-glycerol, methoxypolyethylene glycol) or PEG-DPG (1,2-Dipalmitoyl-sn-glycerol, methoxypolyethylene glycol).
  • PEG- DSG (1,2-Distearoyl-sn-glycerol, methoxypolyethylene glycol
  • PEG-DPG 1,2-Dipalmitoyl-sn-glycerol, methoxypolyethylene glycol.
  • the cationic lipid may be selected from any lipid known in the art such as, but not limited to, DLin-MC3-DMA, DLin-DMA, C12-200 and DLin-KC2-DMA.
  • the cationic lipid may be selected from, but not limited to, a cationic lipid described in International Publication Nos. WO2012040184, WO2011153120,
  • the cationic lipid may be selected from, but not limited to, formula A described in International Publication Nos. WO2012040184, WO2011153120, WO2011149733, WO2011090965, WO2011043913, WO2011022460, WO2012061259, WO2012054365 and
  • the cationic lipid may be selected from, but not limited to, formula CLI-CLXXIX of International Publication No. WO2008103276, formula CLI-CLXXIX of US Patent No. 7,893,302, formula CLI-CLXXXXII of US Patent No. 7,404,969 and formula I-VI of US Patent Publication No. US20100036115; each of which is herein incorporated by reference in their entirety.
  • the cationic lipid may be selected from (20Z,23Z)-N,N-dimethylnonacosa-20,23- dien- 10-amine, ( 17Z,20Z)-N,N-dimemylhexacosa- 17,20-dien-9-amine, ( 1 Z, 19Z)-N5N- dimethylpentacosa-1 6, 19-dien-8-amine, (13Z,16Z)-N,N-dimethyldocosa-13,16-dien-5-amine, ( 12Z, 15Z)-N,N-dimethylhenicosa- 12, 15-dien-4-amine, (14Z, 17Z)-N,N-dimethyltricosa- 14,17-dien- 6-amine, (15Z, 18Z)-N,N-dimethyltetracosa- 15,18-dien-7-amine, ( 18Z,21 Z)-N,N-dimethylheptacosa- 18,21 -dien
  • the cationic lipid may be synthesized by methods known in the art and/or as described in International Publication Nos. WO2012040184, WO2011153120,
  • the LNP formulation may contain PEG-c-DOMG at 3% lipid molar ratio. In another embodiment, the LNP formulation may contain PEG-c-DOMG at 1.5% lipid molar ratio.
  • the LNP formulation may contain PEG-DMG 2000 (1,2-dimyristoyl- sn-glycero-3-phophoethanolamine-N-[methoxy(poly ethylene glycol)-2000).
  • the LNP formulation may contain PEG-DMG 2000, a cationic lipid known in the art and at least one other component.
  • the LNP formulation may contain PEG-DMG 2000, a cationic lipid known in the art, DSPC and cholesterol.
  • the LNP formulation may contain PEG-DMG 2000, DLin-DMA, DSPC and cholesterol.
  • the LNP formulation may contain PEG-DMG 2000, DLin-DMA, DSPC and cholesterol in a molar ratio of 2:40: 10:48 (see e.g. Geall et al., Nonviral delivery of self-amplifying RNA vaccines, PNAS 2012; PMID: 22908294; herein incorporated by reference in its entirety).
  • the LNP formulation may be formulated by the methods described in International Publication Nos. WO2011127255 or WO2008103276, each of which is herein incorporated by reference in their entirety.
  • modified RNA described herein may be encapsulated in LNP formulations as described in WO2011127255 and/or
  • modified RNA described herein may be formulated in a nanoparticle to be delivered by a parenteral route as described in U.S. Pub. No. 20120207845; herein incorporated by reference in its entirety.
  • LNP formulations described herein may comprise a polycationic composition.
  • the polycationic composition may be selected from formula 1-60 of US Patent Publication No. US20050222064; herein incorporated by reference in its entirety.
  • the LNP formulations comprising a polycationic composition may be used for the delivery of the modified RNA described herein in vivo and/or in vitro.
  • the LNP formulations described herein may additionally comprise a permeability enhancer molecule.
  • a permeability enhancer molecule are described in US
  • Patent Publication No. US20050222064 herein incorporated by reference in its entirety.
  • the pharmaceutical compositions may be formulated in liposomes such as, but not limited to, DiLa2 liposomes (Marina Biotech, Bothell, WA), SMARTICLES® (Marina
  • the nanoparticle formulations may be a carbohydrate nanoparticle comprising a carbohydrate carrier and a modified nucleic acid molecule (e.g., mmRNA).
  • the carbohydrate carrier may include, but is not limited to, an anhydride-modified phytoglycogen or glycogen-type material, phtoglycogen octenyl succinate, phytoglycogen beta- dextrin, anhydride-modified phytoglycogen beta-dextrin. (See e.g., International Publication No. WO2012109121; herein incorporated by reference in its entirety).
  • Lipid nanoparticle formulations may be improved by replacing the cationic lipid with a biodegradable cationic lipid which is known as a rapidly eliminated lipid nanoparticle (reLNP).
  • Ionizable cationic lipids such as, but not limited to, DLinDMA, DLin-KC2-DMA, and DLin-MC3- DMA, have been shown to accumulate in plasma and tissues over time and may be a potential source of toxicity.
  • the rapid metabolism of the rapidly eliminated lipids can improve the tolerability and therapeutic index of the lipid nanoparticles by an order of magnitude from a 1 mg/kg dose to a 10 mg/kg dose in rat.
  • ester linkage can improve the degradation and metabolism profile of the cationic component, while still maintaining the activity of the reLNP formulation.
  • the ester linkage can be internally located within the lipid chain or it may be terminally located at the terminal end of the lipid chain.
  • the internal ester linkage may replace any carbon in the lipid chain.
  • the internal ester linkage may be located on either side of the saturated carbon.
  • reLNPs include,
  • an immune response may be elicited by delivering a lipid nanoparticle which may include a nanospecies, a polymer and an immunogen.
  • a lipid nanoparticle which may include a nanospecies, a polymer and an immunogen.
  • the polymer may encapsulate the nanospecies or partially encapsulate the nanospecies.
  • the immunogen may be a recombinant protein, a modified R A described herein.
  • the lipid nanoparticle may be formulated for use in a vaccine such as, but not limited to, against a pathogen.
  • Lipid nanoparticles may be engineered to alter the surface properties of particles so the lipid nanoparticles may penetrate the mucosal barrier.
  • Mucus is located on mucosal tissue such as, but not limted to, oral (e.g., the buccal and esophageal membranes and tonsil tissue), ophthalmic, gastrointestinal (e.g., stomach, small intestine, large intestine, colon, rectum), nasal, respiratory (e.g., nasal, pharyngeal, tracheal and bronchial membranes), genital (e.g., vaginal, cervical and urethral membranes). Nanoparticles larger than 10-200 nm which are preferred for higher drug
  • the transport of nanoparticles may be determined using rates of permeation and/or fluorescent microscopy techniques including, but not limited to, fluorescence recovery after photobleaching (FRAP) and high resolution multiple particle tracking (MPT).
  • FRAP fluorescence recovery after photobleaching
  • MPT high resolution multiple particle tracking
  • compositions which can penetrate a mucosal barrier may be made as described in U.S. Pat. No. 8,241,670, herein incorporated by reference in its entirety.
  • the lipid nanoparticle engineered to penetrate mucus may comprise a polymeric material (i.e. a polymeric core) and/or a polymer-vitamin conjugate and/or a tri-block co-polymer.
  • the polymeric material may include, but is not limited to, polyamines, polyethers, polyamides, polyesters, polycarbamates, polyureas, polycarbonates, poly(styrenes), polyimides, polysulfones, polyurethanes, polyacetylenes, polyethylenes, polyethyeneimines, polyisocyanates, polyacrylates, polymethacrylates, polyacrylonitriles, and polyarylates.
  • the polymeric material may be
  • the polymeric material may additionally be irradiated.
  • the polymeric material may be gamma irradiated (See e.g., International App. No. WO201282165, herein incorporated by reference in its entirety).
  • Non-limiting examples of specific polymers include poly(caprolactone) (PCL), ethylene vinyl acetate polymer (EVA), poly(lactic acid) (PLA), poly(L-lactic acid) (PLLA), poly(glycolic acid) (PGA), poly(lactic acid-co- glycolic acid) (PLGA), poly(L-lactic acid-co-glycolic acid) (PLLGA), poly(D,L-lactide) (PDLA), poly(L-lactide) (PLLA), poly(D,L-lactide-co-caprolactone), poly(D,L-lactide-co-caprolactone-co- glycolide), poly(D,L-lactide-co-PEO-co-D,L-lactide), poly(D,L-lactide-co-PPO-co-D,L-lactide), polyalkyl cyanoacralate, polyurethane, poly-L-lysine (PLL), hydroxypropyl methacrylate (HPMA),
  • the lipid nanoparticle may be coated or associated with a co-polymer such as, but not limited to, a block copolymer, and (poly(ethylene glycol))-(poly(propylene oxide))-(poly(ethylene glycol)) triblock copolymer (see e.g., US Publication 20120121718 and US Publication 20100003337 and U.S. Pat. No. 8,263,665; each of which is herein incorporated by reference in their entirety).
  • the co-polymer may be a polymer that is generally regarded as safe (GRAS) and the formation of the lipid nanoparticle may be in such a way that no new chemical entities are created.
  • the lipid nanoparticle may comprise poloxamers coating PLGA nanoparticles without forming new chemical entities which are still able to rapidly penetrate human mucus (Yang et al. Angew. Chem. Int. Ed. 2011 50:2597-2600; herein incorporated by reference in its entirety).
  • the vitamin of the polymer-vitamin conjugate may be vitamin E.
  • the vitamin portion of the conjugate may be substituted with other suitable components such as, but not limited to, vitamin A, vitamin E, other vitamins, cholesterol, a hydrophobic moiety, or a hydrophobic component of other surfactants (e.g., sterol chains, fatty acids, hydrocarbon chains and alkylene oxide chains).
  • the lipid nanoparticle engineered to penetrate mucus may include surface altering agents such as, but not limited to, mmRNA, anionic proteins (e.g., bovine serum albumin), surfactants (e.g., cationic surfactants such as for example dimethyldioctadecyl-ammonium bromide), sugars or sugar derivatives (e.g., cyclodextrin), nucleic acids, polymers (e.g., heparin, polyethylene glycol and poloxamer), mucolytic agents (e.g., N-acetylcysteine, mugwort, bromelain, papain, clerodendrum, acetylcysteine, bromhexine, carbocisteine, eprazinone, mesna, ambroxol, sobrerol, domiodol, letosteine, stepronin, tiopronin, gelsolin, thymosin
  • anionic proteins
  • the surface altering agent may be embedded or enmeshed in the particle's surface or disposed (e.g., by coating, adsorption, covalent linkage, or other process) on the surface of the lipid nanoparticle.
  • the mucus penetrating lipid nanoparticles may comprise at least one mmRNA described herein.
  • the mmRNA may be encapsulated in the lipid nanoparticle and/or disposed on the surface of the paricle.
  • the mmRNA may be covalently coupled to the lipid nanoparticle.
  • Formulations of mucus penetrating lipid nanoparticles may comprise a plurality of nanoparticles. Further, the formulations may contain particles which may interact with the mucus and alter the structural and/or adhesive properties of the surrounding mucus to decrease mucoadhesion which may increase the delivery of the mucus penetrating lipid nanoparticles to the mucosal tissue.
  • the modified nucleic acid molecule or mmRNA is formulated as a lipoplex, such as, without limitation, the ATUPLEXTM system, the DACC system, the DBTC system and other siRNA-lipoplex technology from Silence Therapeutics (London, United Kingdom), STEMFECTTM from STEMGENT® (Cambridge, MA), and polyethylenimine (PEI) or protamine- based targeted and non-targeted delivery of nucleic acids acids (Aleku et al. Cancer Res. 2008 68:9788-9798; Strumberg et al.
  • a lipoplex such as, without limitation, the ATUPLEXTM system, the DACC system, the DBTC system and other siRNA-lipoplex technology from Silence Therapeutics (London, United Kingdom), STEMFECTTM from STEMGENT® (Cambridge, MA), and polyethylenimine (PEI) or protamine- based targeted and non-targeted delivery of nucleic acids acids (Aleku et al. Cancer Res
  • such formulations may also be constructed or compositions altered such that they passively or actively are directed to different cell types in vivo, including but not limited to hepatocytes, immune cells, tumor cells, endothelial cells, antigen presenting cells, and leukocytes (Akinc et al. Mol Ther. 2010 18: 1357-1364; Song et al, Nat Biotechnol. 2005 23:709- 717; Judge et al, J Clin Invest. 2009 119:661-673; Kaufmann et al, Microvasc Res 2010 80:286- 293; Santel et al, Gene Ther 2006 13:1222-1234; Santel et al, Gene Ther 2006 13:1360-1370;
  • DLin-DMA, DLin-KC2-DMA and DLin-MC3 -DMA-based lipid nanoparticle formulations which have been shown to bind to apolipoprotein E and promote binding and uptake of these formulations into hepatocytes in vivo (Akinc et al. Mol Ther. 2010 18: 1357-1364; herein incorporated by reference in its entirety).
  • Formulations can also be selectively targeted through expression of different ligands on their surface as exemplified by, but not limited by, folate, transferrin, N- acetylgalactosamine (GalNAc), and antibody targeted approaches (Kolhatkar et al, Curr Drug Discov Technol. 2011 8: 197-206; Musacchio and Torchilin, Front Biosci. 2011 16:1388-1412; Yu et al, Mol Membr Biol. 2010 27:286-298; Patil et al, Crit Rev Ther Drug Carrier Syst. 2008 25: 1-61; Benoit et al, Biomacromolecules.
  • the modified nucleic acid molecules or mmRNA are formulated as a solid lipid nanoparticle.
  • a solid lipid nanoparticle may be spherical with an average diameter between 10 to 1000 nm. SLN possess a solid lipid core matrix that can solubilize lipophilic molecules and may be stabilized with surfactants and/or emulsifiers.
  • the lipid nanoparticle may be a self-assembly lipid-polymer nanoparticle (see Zhang et al., ACS Nano, 2008, 2 (8), pp 1696-1702; herein incorporated by reference in its entirety).
  • Liposomes, lipoplexes, or lipid nanoparticles may be used to improve the efficacy of modified nucleic acid molecules or mmRNA directed protein production as these formulations may be able to increase cell transfection by the modified nucleic acid molecule or mmRNA; and/or increase the translation of encoded protein.
  • One such example involves the use of lipid encapsulation to enable the effective systemic delivery of polyplex plasmid DNA (Heyes et al., Mol Ther. 2007 15:713-720; herein incorporated by reference in its entirety).
  • the liposomes, lipoplexes, or lipid nanoparticles may also be used to increase the stability of the modified nucleic acid molecules or mmRNA.
  • the modified nucleic acid molecules and/or the mmRNA of the present invention can be formulated for controlled release and/or targeted delivery.
  • the modified nucleic acid molecules and/or the mmRNA of the present invention can be formulated for controlled release and/or targeted delivery.
  • controlled release refers to a pharmaceutical composition or compound release profile that conforms to a particular pattern of release to effect a therapeutic outcome.
  • the modified nucleic acids molecules or the mmRNA may be encapsulated into a delivery agent described herein and/or known in the art for controlled release and/or targeted delivery.
  • encapsulate means to enclose, surround or encase. As it relates to the formulation of the compounds of the invention, encapsulation may be substantial, complete or partial.
  • substantially encapsulated means that at least greater than 50, 60, 70, 80, 85, 90, 95, 96, 97, 98, 99, 99.9, 99.9 or greater than 99.999% of the pharmaceutical composition or compound of the invention may be enclosed, surrounded or encased within the delivery agent.
  • encapsulation means that less than 10, 10, 20, 30, 40 50 or less of the pharmaceutical composition or compound of the invention may be enclosed, surrounded or encased within the delivery agent.
  • encapsulation may be determined by measuring the escape or the activity of the pharmaceutical composition or compound of the invention using fluorescence and/or electron micrograph. For example, at least 1, 5, 10, 20, 30, 40, 50, 60, 70, 80, 85, 90, 95, 96, 97, 98, 99, 99.9, 99.99 or greater than 99.99% of the pharmaceutical composition or compound of the invention are encapsulated in the delivery agent.
  • the controlled release formulation may include, but is not limited to, tri-block co-polymers.
  • the formulation may include two different types of tri-block co-polymers (International Pub. No. WO2012131104 and WO2012131106; each of which is herein incorporated by reference in its entirety).
  • the modified nucleic acid molecules or the mmRNA may be encapsulated into a lipid nanoparticle or a rapidly eliminated lipid nanoparticle and the lipid nanoparticles or a rapidly eliminated lipid nanoparticle may then be encapsulated into a polymer, hydrogel and/or surgical sealant described herein and/or known in the art.
  • the polymer, hydrogel or surgical sealant may be PLGA, ethylene vinyl acetate (EVAc), poloxamer, GELSITE® (Nanotherapeutics, Inc. Alachua, FL), HYLENEX® (Halozyme
  • surgical sealants such as fibrinogen polymers (Ethicon Inc. Cornelia, GA), TISSELL® (Baxter International, Inc Deerfield, IL), PEG-based sealants, and COSEAL® (Baxter International, Inc Deerfield, IL).
  • the lipid nanoparticle may be encapsulated into any polymer known in the art which may form a gel when injected into a subject.
  • the lipid nanoparticle may be encapsulated into a polymer matrix which may be biodegradable.
  • the modified nucleic acid molecules or mmRNA formulation for controlled release and/or targeted delivery may also include at least one controlled release coating.
  • Controlled release coatings include, but are not limited to, OPADRY®, polyvinylpyrrolidone/vinyl acetate copolymer, polyvinylpyrrolidone, hydroxypropyl methylcellulose, hydroxypropyl cellulose, hydroxyethyl cellulose, EUDRAGIT RL®, EUDRAGIT RS® and cellulose derivatives such as ethylcellulose aqueous dispersions (AQUACOAT® and SURELEASE®).
  • the controlled release and/or targeted delivery formulation may comprise at least one degradable polyester which may contain polycationic side chains.
  • Degradable polyesters include, but are not limited to, poly(serine ester), poly(L-lactide-co-L-lysine), poly(4- hydroxy-L-proline ester), and combinations thereof.
  • the degradable polyesters may include a PEG conjugation to form a PEGylated polymer.
  • the modified nucleic acid molecules and/or the mmRNA of the present invention may be encapsulated in a therapeutic nanoparticle.
  • Therapeutic nanoparticles may be formulated by methods described herein and known in the art such as, but not limited to,
  • therapeutic polymer nanoparticles may be identified by the methods described in US Pub No. US20120140790, herein incorporated by reference in its entirety.
  • the therapeutic nanoparticle may be formulated for sustained release.
  • sustained release refers to a pharmaceutical composition or compound that conforms to a release rate over a specific period of time. The period of time may include, but is not limited to, hours, days, weeks, months and years.
  • the sustained release nanoparticle may comprise a polymer and a therapeutic agent such as, but not limited to, the modified nucleic acid molecules and mmRNA of the present invention (see International Pub No. 2010075072 and US Pub No. US20100216804, US20110217377 and US20120201859, each of which is herein incorporated by reference in their entirety).
  • the therapeutic nanoparticles may be formulated to be target specific.
  • the thereapeutic nanoparticles may include a corticosteroid (see International Pub. No. WO2011084518 herein incorporated by reference in its entirety).
  • the therapeutic nanoparticles of the present invention may be formulated to be cancer specific.
  • the therapeutic nanoparticles may be formulated in
  • the nanoparticles of the present invention may comprise a polymeric matrix.
  • the nanoparticle may comprise two or more polymers such as, but not limited to, polyethylenes, polycarbonates, polyanhydrides, polyhydroxyacids,
  • polypropylfumerates polycaprolactones, polyamides, polyacetals, polyethers, polyesters, poly(orthoesters), polycyanoacrylates, polyvinyl alcohols, polyurethanes, polyphosphazenes, polyacrylates, polymethacrylates, polycyanoacrylates, polyureas, polystyrenes, polyamines, polylysine, poly(ethylene imine), poly(serine ester), poly(L-lactide-co-L-lysine), poly(4-hydroxy-L- proline ester) or combinations thereof.
  • the therapeutic nanoparticle comprises a diblock copolymer.
  • the diblock copolymer may include PEG in combination with a polymer such as, but not limited to, polyethylenes, polycarbonates, polyanhydrides, polyhydroxyacids,
  • polypropylfumerates polycaprolactones, polyamides, polyacetals, polyethers, polyesters, poly(orthoesters), polycyanoacrylates, polyvinyl alcohols, polyurethanes, polyphosphazenes, polyacrylates, polymethacrylates, polycyanoacrylates, polyureas, polystyrenes, polyamines, polylysine, poly(ethylene imine), poly(serine ester), poly(L-lactide-co-L-lysine), poly(4-hydroxy-L- proline ester) or combinations thereof.
  • the therapeutic nanoparticle comprises a PLGA-PEG block copolymer (see US Pub. No. US20120004293 and US Pat No. 8,236,330, each of which is herein incorporated by reference in their entirety).
  • the therapeutic nanoparticle is a stealth nanoparticle comprising a diblock copolymer of PEG and PLA or PEG and PLGA (see US Pat No 8,246,968, herein incorporated by reference in its entirety).
  • the therapeutic nanoparticle may comprise a multiblock copolymer (See e.g., U.S. Pat. No. 8,263,665 and 8,287,910; each of which is herein incorporated by reference in its entirety).
  • the block copolymers described herein may be included in a polyion complex comprising a non-polymeric micelle and the block copolymer.
  • a polyion complex comprising a non-polymeric micelle and the block copolymer.
  • the therapeutic nanoparticle may comprise at least one acrylic polymer.
  • Acrylic polymers include but are not limited to, acrylic acid, methacrylic acid, acrylic acid and methacrylic acid copolymers, methyl methacrylate copolymers, ethoxyethyl methacrylates, cyanoethyl methacrylate, amino alkyl methacrylate copolymer, poly(acrylic acid), poly(methacrylic acid), polycyanoacrylates and combinations thereof.
  • the therapeutic nanoparticles may comprise at least one cationic polymer described herein and/or known in the art.
  • the therapeutic nanoparticles may comprise at least one amine- containing polymer such as, but not limited to polylysine, polyethylene imine, poly(amidoamine) dendrimers, poly(beta-amino esters) (See e.g., U.S. Pat. No. 8,287,849; herein incorporated by reference in its entirety) and combinations thereof.
  • amine- containing polymer such as, but not limited to polylysine, polyethylene imine, poly(amidoamine) dendrimers, poly(beta-amino esters) (See e.g., U.S. Pat. No. 8,287,849; herein incorporated by reference in its entirety) and combinations thereof.
  • the therapeutic nanoparticles may comprise at least one degradable polyester which may contain polycationic side chains.
  • Degradeable polyesters include, but are not limited to, poly(serine ester), poly(L-lactide-co-L-lysine), poly(4-hydroxy-L-proline ester), and combinations thereof.
  • the degradable polyesters may include a PEG conjugation to form a PEGylated polymer.
  • the therapeutic nanoparticle may include a conjugation of at least one targeting ligand.
  • the targeting ligand may be any ligand known in the art such as, but not limited to, a monoclonal antibody. (Kirpotin et al, Cancer Res. 2006 66:6732-6740; herein incorporated by reference in its entirety).
  • the therapeutic nanoparticle may be formulated in an aqueous solution which may be used to target cancer (see International Pub No. WO2011084513 and US Pub No. US20110294717, each of which is herein incorporated by reference in their entirety).
  • the modified nucleic acid molecules or mmRNA may be encapsulated in, linked to and/or associated with synthetic nanocarriers.
  • Synthetic nanocarriers include, but are not limited to, those described in International Pub. Nos. WO2010005740, WO2010030763, WO201213501, WO2012149252, WO2012149255, WO2012149259, WO2012149265,
  • the synthetic nanocarriers may be formulated using methods known in the art and/or described herein. As a non-limiting example, the synthetic nanocarriers may be formulated by the methods described in International Pub Nos. WO2010005740, WO2010030763 and WO201213501 and US Pub. Nos. US20110262491, US20100104645, US20100087337 and US20120244222, each of which is herein incorporated by reference in their entirety. In another embodiment, the synthetic nanocarrier formulations may be lyophilized by methods described in International Pub. No.
  • the synthetic nanocarriers may contain reactive groups to release the modified nucleic acid molecules and/or mmRNA described herein (see International Pub. No.
  • the synthetic nanocarriers may contain an immunostimulatory agent to enhance the immune response from delivery of the synthetic nanocarrier.
  • the synthetic nanocarrier may comprise a Thl immunostimulatory agent which may enhance a Thl -based response of the immune system (see International Pub No. WO2010123569 and US Pub. No. US20110223201, each of which is herein incorporated by reference in its entirety).
  • the synthetic nanocarriers may be formulated for targeted release.
  • the synthetic nanocarrier is formulated to release the modified nucleic acid molecules and/or mmRNA at a specified pH and/or after a desired time interval.
  • the synthetic nanoparticle may be formulated to release the modified mRNA molecules and/or mmRNA after 24 hours and/or at a pH of 4.5 (see International Pub. Nos. WO2010138193 and WO2010138194 and US Pub Nos. US20110020388 and US20110027217, each of which is herein incorporated by reference in their entirety).
  • the synthetic nanocarriers may be formulated for controlled and/or sustained release of the modified nucleic acid molecules and/or mmRNA described herein.
  • the synthetic nanocarriers for sustained release may be formulated by methods known in the art, described herein and/or as described in International Pub No.
  • the synthetic nanocarrier may be formulated for use as a vaccine.
  • the synthetic nanocarrier may encapsulate at least one modified nucleic acid molecule and/or mmRNA which encodes at least one antigen.
  • the synthetic nanocarrier may include at least one antigen and an excipient for a vaccine dosage form (see International Pub No. WO2011150264 and US Pub No. US20110293723, each of which is herein incorporated by reference in their entirety).
  • a vaccine dosage form may include at least two synthetic nanocarriers with the same or different antigens and an excipient (see International Pub No. WO2011150249 and US Pub No.
  • the vaccine dosage form may be selected by methods described herein, known in the art and/or described in International Pub No. WO2011150258 and US Pub No. US20120027806, each of which is herein incorporated by reference in their entirety).
  • the synthetic nanocarrier may comprise at least one modified nucleic acid molecule and/or mmRNA which encodes at least one adjuvant.
  • the synthetic nanocarrier may comprise at least one modified nucleic molecule acid and/or mmRNA and an adjuvant.
  • the synthetic nanocarrier comprising and adjuvant may be formulated by the methods described in International Pub No. WO2011150240 and US Pub No. US20110293700, each of which is herein incorporated by reference in its entirety.
  • the synthetic nanocarrier may encapsulate at least one modified nucleic acid molecule and/or mmRNA which encodes a peptide, fragment or region from a virus.
  • the synthetic nanocarrier may include, but is not limited to, the nanocarriers described in International Pub No. WO2012024621, WO201202629, WO2012024632 and US Pub No. US20120064110, US20120058153 and US20120058154, each of which is herein incorporated by reference in their entirety.
  • the nanoparticle may be optimized for oral administration.
  • the nanoparticle may comprise at least one cationic biopolymer such as, but not limited to, chitosan or a derivative thereof.
  • the nanoparticle may be formulated by the methods described in U.S. Pub. No. 20120282343; herein incorporated by reference in its entirety.
  • the modified nucleic acid molecules and mmRNA of the invention can be formulated using natural and/or synthetic polymers.
  • polymers which may be used for delivery include, but are not limited to, DYNAMIC POLYCONJUGATE® (Arrowhead Research Corp., Pasadena, CA) formulations from MIRUS® Bio (Madison, WI) and Roche Madison
  • PHASERXTM polymer formulations such as, without limitation, SMARTT
  • POLYMER TECHNOLOGYTM (Seattle, WA), DMRI/DOPE, poloxamer, VAXFECTIN® adjuvant from Vical (San Diego, CA), chitosan, cyclodextrin from Calando Pharmaceuticals (Pasadena, CA), dendrimers and poly(lactic-co-glycolic acid) (PLGA) polymers, RONDELTM
  • RNAi/Oligonucleotide Nanoparticle Delivery polymers (Arrowhead Research Corporation, Pasadena, CA) and pH responsive co-block polymers such as, but not limited to, PHASERXTM (Seattle, WA).
  • a non-limiting example of chitosan formulation includes a core of positively charged chitosan and an outer portion of negatively charged substrate (U.S. Pub. No. 20120258176; herein incorporated by reference in its entirety).
  • Chitosan includes, but is not limited to N-trimethyl chitosan, mono-N-carboxymethyl chitosan (MCC), N-palmitoyl chitosan (NPCS), EDTA-chitosan, low molecular weight chitosan, chitosan derivatives, or combinations thereof.
  • the polymers used in the present invention have undergone processing to reduce and/or inhibit the attachement of unwanted substances such as, but not limited to, bacteria, to the surface of the polymer.
  • the polymer may be processed by methods known and/or described in the art and/or described in International Pub. No. WO2012150467, herein incorporated by reference in its entirety.
  • a non- limiting example of PLGA formulations include, but are not limited to, PLGA injectable depots (e.g., ELIGARD® which is formed by dissolving PLGA in 66% N-methyl-2- pyrrolidone (NMP) and the remainder being aqueous solvent and leuprolide. Once injected, the PLGA and leuprolide peptide precipitates into the subcutaneous space).
  • PLGA injectable depots e.g., ELIGARD® which is formed by dissolving PLGA in 66% N-methyl-2- pyrrolidone (NMP) and the remainder being aqueous solvent and leuprolide. Once injected, the PLGA and leuprolide peptide precipitates into the subcutaneous space).
  • oligonucleotides in vivo into the cell cytoplasm are dynamic polyconjugates and cyclodextrin-based nanoparticles.
  • the first of these delivery approaches uses dynamic polyconjugates and has been shown in vivo in mice to effectively deliver siRNA and silence endogenous target mRNA in hepatocytes (Rozema et al, Proc Natl Acad Sci U S A.
  • the polymer complex On binding to the hepatocyte and entry into the endosome, the polymer complex disassembles in the low-pH environment, with the polymer exposing its positive charge, leading to endosomal escape and cytoplasmic release of the siRNA from the polymer.
  • the polymer Through replacement of the ⁇ -acetylgalactosamine group with a mannose group, it was shown one could alter targeting from asialoglycoprotein receptor-expressing hepatocytes to sinusoidal endothelium and Kupffer cells.
  • Another polymer approach involves using transferrin- targeted cyclodextrin-containing polycation nanoparticles.
  • the polymer formulation can permit the sustained or delayed release of modified nucleic acid molecules or mmRNA (e.g., following intramuscular or subcutaneous injection).
  • the altered release profile for the modified nucleic acid molecule or mmRNA can result in, for example, translation of an encoded protein over an extended period of time.
  • the polymer formulation may also be used to increase the stability of the modified nucleic acid molecule or mmRNA.
  • Biodegradable polymers have been previously used to protect nucleic acids other than mmRNA from degradation and been shown to result in sustained release of payloads in vivo (Rozema et al, Proc Natl Acad Sci U S A. 2007 104: 12982-12887; Sullivan et al, Expert Opin Drug Deliv. 2010 7: 1433-1446; Convertine et al., Biomacromolecules. 2010 Oct 1; Chu et al., Acc Chem Res. 2012 Jan 13; Manganiello et al., Biomaterials. 2012 33:2301-2309; Benoit et al, Biomacromolecules. 2011 12:2708-2714; Singha et al, Nucleic Acid Ther.
  • the pharmaceutical compositions may be sustained release
  • the sustained release formulations may be for subcutaneous delivery.
  • Sustained release formulations may include, but are not limited to, PLGA microspheres, ethylene vinyl acetate (EVAc), poloxamer, GELSITE® (Nanotherapeutics, Inc. Alachua, FL), HYLENEX® (Halozyme Therapeutics, San Diego CA), surgical sealants such as fibrinogen polymers (Ethicon Inc. Cornelia, GA), TISSELL® (Baxter International, Inc Deerfield, IL), PEG- based sealants, and COSEAL® (Baxter International, Inc Deerfield, IL).
  • modified mRNA may be formulated in PLGA microspheres by preparing the PLGA microspheres with tunable release rates (e.g., days and weeks) and
  • EVAc are non-biodegradeable, biocompatible polymers which are used extensively in pre-clinical sustained release implant applications (e.g., extended release products Ocusert a pilocarpine ophthalmic insert for glaucoma or progestasert a sustained release progesterone intrauterine deivce; transdermal delivery systems Testoderm, Duragesic and Selegiline; catheters).
  • Poloxamer F-407 NF is a hydrophilic, non-ionic surfactant triblock copolymer of polyoxyethylene-polyoxypropylene-polyoxyethylene having a low viscosity at temperatures less than 5°C and forms a solid gel at temperatures greater than 15°C.
  • PEG-based surgical sealants comprise two synthetic PEG components mixed in a delivery device which can be prepared in one minute, seals in 3 minutes and is reabsorbed within 30 days.
  • GELSITE® and natural polymers are capable of in-situ gelation at the site of administration. They have been shown to interact with protein and peptide therapeutic candidates through ionic ineraction to provide a stabilizing effect.
  • Polymer formulations can also be selectively targeted through expression of different ligands as exemplified by, but not limited by, folate, transferrin, and N-acetylgalactosamine
  • the modified nucleic acid molecules and mmRNA of the invention may be formulated with or in a polymeric compound.
  • the polymer may include at least one polymer such as, but not limited to, polyethenes, polyethylene glycol (PEG), poly(l-lysine)(PLL), PEG grafted to PLL, cationic lipopolymer, biodegradable cationic lipopolymer, polyethyleneimine (PEI), cross-linked branched poly(alkylene imines), a polyamine derivative, a modified poloxamer, a biodegradable polymer, elastic biodegradable polymer, biodegradable block copolymer, biodegradable random copolymer, biodegradable polyester copolymer, biodegradable polyester block copolymer, biodegradable polyester block random copolymer, multiblock copolymers, linear biodegradable copolymer, poly[a-(4-aminobutyl)-L-glycolic acid) (PAGA)
  • polycyanoacrylates polyvinyl alcohols, polyurethanes, polyphosphazenes, polyacrylates, polymethacrylates, polycyanoacrylates, polyureas, polystyrenes, polyamines, polylysine,
  • the modified nucleic acid molecules or mmRNA of the invention may be formulated with the polymeric compound of PEG grafted with PLL as described in U.S. Pat. No. 6,177,274; herein incorporated by reference in its entirety.
  • the formulation may be used for transfecting cells in vitro or for in vivo delivery of the modified nucleic acid molecules and mmRNA.
  • the modified nucleic acid molecules and mmRNA may be suspended in a solution or medium with a cationic polymer, in a dry pharmaceutical composition or in a solution that is capable of being dried as described in U.S. Pub. Nos. 20090042829 and
  • modified nucleic acid molecules or mmRNA of the invention may be formulated with a PLGA-PEG block copolymer (see US Pub. No.
  • modified nucleic acid molecules or mmRNA of the invention may be formulated with a diblock copolymer of PEG and PLA or PEG and PLGA (see US Pat No 8,246,968, herein incorporated by reference in its entirety).
  • a polyamine derivative may be used to deliver nucleic acid molecules and/or mmRNA or to treat and/or prevent a disease or to be included in an implantable or injectable device (U.S. Pub. No. 20100260817 herein incorporated by reference in its entirety).
  • a pharmaceutical composition may include the modified nucleic acid molecules and mmRNA and the polyamine derivative described in U.S. Pub. No. 20100260817 (the contents of which are
  • the modified nucleic acids or mmRNA of the present invention may be delivered using a polyaminde polymer such as, but not limited to, a polymer comprising a 1,3-dipolar addition polymer prepared by combining a carbohydrate diazide monomer with a dilkyne unite comprising oligoamines (U.S. Pat. No.
  • the modified nucleic acid molecules and/or mmRNA of the invention may be formulated with at least one acrylic polymer.
  • Acrylic polymers include but are not limited to, acrylic acid, methacrylic acid, acrylic acid and methacrylic acid copolymers, methyl methacrylate copolymers, ethoxyethyl methacrylates, cyanoethyl methacrylate, amino alkyl methacrylate copolymer, poly(acrylic acid), poly(methacrylic acid), polycyanoacrylates and combinations thereof.
  • the modified nucleic acid molecules and/or mmRNA of the present invention may be formulated with at least one polymer and/or derivatives thereof described in International Publication Nos. WO2011115862, WO2012082574 and WO2012068187 and U.S. Pub. No. 20120283427, each of which are herein incorporated by reference in their entireties.
  • the modified nucleic acid molecules or mmRNA of the present invention may be formulated with a polymer of formula Z as described in WO2011115862, herein incorporated by reference in its entirety.
  • the modified nucleic acid molecules or mmRNA may be formulated with a polymer of formula Z, Z' or Z" as described in International Pub. Nos.
  • WO2012082574 or WO2012068187 each of which are herein incorporated by reference in their entireties.
  • the polymers formulated with the modified nucleic acids and/or modified mRNA of the present invention may be synthesized by the methods described in International Pub. Nos. WO2012082574 or WO2012068187, each of which are herein incorporated by reference in their entireties.
  • Formulations of modified nucleic acid molecules and/or mmRNA of the invention may include at least one amine-containing polymer such as, but not limited to polylysine, polyethylene imine, poly(amidoamine) dendrimers or combinations thereof.
  • the modified nucleic acid molecules and/or mmRNA of the invention may be formulated in a pharmaceutical compound including a poly(alkylene imine), a biodegradable cationic lipopolymer, a biodegradable block copolymer, a biodegradable polymer, or a
  • biodegradable random copolymer a biodegradable polyester block copolymer, a biodegradable polyester polymer, a biodegradable polyester random copolymer, a linear biodegradable copolymer, PAGA, a biodegradable cross-linked cationic multi-block copolymer or combinations thereof.
  • the biodegradable cationic lipopolymer may be made by methods known in the art and/or described in U.S. Pat. No. 6,696,038, U.S. App. Nos. 20030073619 and 20040142474 each of which is herein incorporated by reference in their entireties.
  • the poly(alkylene imine) may be made using methods known in the art and/or as described in U.S. Pub. No.
  • biodegradabale polymer, biodegradable block copolymer, the biodegradable random copolymer, biodegradable polyester block copolymer, biodegradable polyester polymer, or biodegradable polyester random copolymer may be made using methods known in the art and/or as described in U.S. Pat. Nos. 6,517,869 and 6,267,987, the contents of which are each incorporated herein by reference in their entirety.
  • the linear biodegradable copolymer may be made using methods known in the art and/or as described in U.S. Pat. No.
  • the PAGA polymer may be made using methods known in the art and/or as described in U.S. Pat. No. 6,217,912 herein incorporated by reference in its entirety.
  • the PAGA polymer may be copolymerized to form a copolymer or block copolymer with polymers such as but not limited to, poly-L-lysine, polyargine, polyornithine, histones, avidin, protamines, polylactides and poly(lactide- co-glycolides).
  • the biodegradable cross-linked cationic multi-block copolymers may be made my methods known in the art and/or as described in U.S. Pat. No. 8,057,821 or U.S. Pub. No.
  • the multi-block copolymers may be synthesized using linear polyethyleneimine (LPEI) blocks which have distinct patterns as compared to branched polyethyleneimines.
  • LPEI linear polyethyleneimine
  • the composition or pharmaceutical composition may be made by the methods known in the art, described herein, or as described in U.S. Pub. No. 20100004315 or U.S. Pat. Nos. 6,267,987 and 6,217,912 each of which are herein incorporated by reference in their entireties.
  • the modified nucleic acid molecules and mmRNA of the invention may be formulated with at least one degradable polyester which may contain polycationic side chains.
  • Degradable polyesters include, but are not limited to, poly(serine ester), poly(L-lactide-co-L-lysine), poly(4- hydroxy-L-proline ester), and combinations thereof.
  • the degradable polyesters may include a PEG conjugation to form a PEGylated polymer.
  • the modified nucleic acid molecules and mmRNA of the invention may be formulated with at least one crosslinkable polyester.
  • Crosslinkable polyesters include those known in the art and described in US Pub. No. 20120269761, herein incorporated by reference in its entirety.
  • the polymers described herein may be conjugated to a lipid- terminating PEG.
  • PLGA may be conjugated to a lipid-terminating PEG forming PLGA-DSPE-PEG.
  • PEG conjugates for use with the present invention are described in International Publication No. WO2008103276, herein
  • the polymers may be conjugated using a ligand conjugate such as, but not limited to, the conjugates described in U.S. Pat. No. 8,273,363, herein incorporated by reference in its entirety.
  • the modified nucleic acid molecules and/or mmRNA described herein may be conjugated with another compound.
  • conjugates are described in US Patent Nos. 7,964,578 and 7,833,992, each of which are herein incorporated by reference in their entireties.
  • modified RNA of the present invention may be conjugated with conjugates of formula 1-122 as described in US Patent Nos. 7,964,578 and 7,833,992, each of which are herein incorporated by reference in their entireties.
  • the modified RNA described herein may be conjugated with a metal such as, but not limited to, gold. (See e.g., Giljohann et al. Journ. Amer. Chem. Soc.
  • modified nucleic acid molecules and/or mmRNA described herein may be conjugated and/or encapsulated in gold-nanoparticles.
  • a gene delivery composition may include a nucleotide sequence and a poloxamer.
  • the modified nucleic acid and mmRNA of the present inveition may be used in a gene delivery composition with the poloxamer described in U.S. Pub. No. 20100004313.
  • the polymer formulation of the present invention may be stabilized by contacting the polymer formulation, which may include a cationic carrier, with a cationic lipopolymer which may be covalently linked to cholesterol and polyethylene glycol groups.
  • the polymer formulation may be contacted with a cationic lipopolymer using the methods described in U.S. Pub. No. 20090042829 herein incorporated by reference in its entirety.
  • the cationic carrier may include, but is not limited to, polyethylenimine, poly(trimethylenimine),
  • the modified nucleic acid molecules and/or mmRNA of the invention may be formulated in a polyplex of one or more polymers (U.S. Pub. No. 20120237565 and 20120270927; each of which is herein incorporated by reference in its entirety).
  • the polyplex comprises two or more cationic polymers.
  • the catioinic polymer may comprise a poly(ethylene imine) (PEI) such as linear PEL
  • the modified nucleic acid molecules and mmRNA of the invention can also be formulated as a nanoparticle using a combination of polymers, lipids, and/or other biodegradable agents, such as, but not limited to, calcium phosphate.
  • Components may be combined in a core-shell, hybrid, and/or layer-by-layer architecture, to allow for fine-tuning of the nanoparticle so to delivery of the modified nucleic acid molecule and mmRNA may be enhanced (Wang et al., Nat Mater. 2006 5:791-796; Fuller et al, Biomaterials. 2008 29: 1526-1532; DeKoker et al, Adv Drug Deliv Rev.
  • the nanoparticle may comprise a plurality of polymers such as, but not limited to hydrophilic-hydrophobic polymers (e.g., PEG-PLGA), hydrophobic polymers (e.g., PEG) and/or hydrophilic polymers (International Pub. No. WO20120225129; herein incorporated by reference in its entirety).
  • hydrophilic-hydrophobic polymers e.g., PEG-PLGA
  • hydrophobic polymers e.g., PEG
  • hydrophilic polymers International Pub. No. WO20120225129
  • Biodegradable calcium phosphate nanoparticles in combination with lipids and/or polymers have been shown to deliver modified nucleic acid molecules and mmRNA in vivo.
  • a lipid coated calcium phosphate nanoparticle which may also contain a targeting ligand such as anisamide, may be used to deliver the modified nucleic acid molecule and mmRNA of the present invention.
  • a targeting ligand such as anisamide
  • a lipid coated calcium phosphate nanoparticle was used (Li et al., J Contr Rel. 2010 142: 416- 421; Li et al, J Contr Rel. 2012 158: 108-114; Yang et al, Mol Ther. 2012 20:609-615; herein incorporated by refereince in its entirety).
  • This delivery system combines both a targeted nanoparticle and a component to enhance the endosomal escape, calcium phosphate, in order to improve delivery of the siRNA.
  • calcium phosphate with a PEG-polyanion block copolymer may be used to deliver modified nucleic acid molecules and mmRNA (Kazikawa et al., J Contr Rel. 2004 97:345-356; Kazikawa et al., J Contr Rel. 2006 111 :368-370; herein incorporated by reference in its entirety).
  • a PEG-charge-conversional polymer (Pitella et al., Biomaterials. 2011 32:3106-3114) may be used to form a nanoparticle to deliver the modified nucleic acid molecules and mmRNA of the present invention.
  • the PEG-charge-conversional polymer may improve upon the PEG-polyanion block copolymers by being cleaved into a polycation at acidic pH, thus enhancing endosomal escape.
  • core-shell nanoparticles have additionally focused on a high-throughput approach to synthesize cationic cross-linked nanogel cores and various shells (Siegwart et al., Proc Natl Acad Sci U S A. 2011 108: 12996-13001).
  • the complexation, delivery, and internalization of the polymeric nanoparticles can be precisely controlled by altering the chemical composition in both the core and shell components of the nanoparticle.
  • the core-shell nanoparticles may efficiently deliver siR A to mouse hepatocytes after they covalently attach cholesterol to the nanoparticle.
  • a hollow lipid core comprising a middle PLGA layer and an outer neutral lipid layer containg PEG may be used to delivery of the modified nucleic acid molecules and mmRNA of the present invention.
  • a luciferease-expressing tumor it was determined that the lipid-polymer-lipid hybrid nanoparticle significantly suppressed luciferase expression, as compared to a conventional lipoplex (Shi et al, Angew Chem Int Ed. 2011 50:7027-7031; herein incorporated by reference in its entirety).
  • the lipid nanoparticles may comprise a core of the modified nucleic acid molecules disclosed herein and a polymer shell.
  • the polymer shell may be any of the polymers described herein and are known in the art.
  • the polymer shell may be used to protect the modified nucleic acids in the core.
  • Core-shell nanoparticles for use with the modified nucleic acid molecules of the present invention are described and may be formed by the methods described in U.S. Pat. No. 8,313,777 herein incorporated by reference in its entirety.
  • the core-shell nanoparticles may comprise a core of the modified nucleic acid molecules disclosed herein and a polymer shell.
  • the polymer shell may be any of the polymers described herein and are known in the art.
  • the polymer shell may be used to protect the modified nucleic acid molecules in the core.
  • modified nucleic acid molecules and mmRNA of the invention can be formulated with peptides and/or proteins in order to increase transfection of cells by the modified nucleic acid molecules or mmRNA.
  • peptides such as, but not limited to, cell penetrating peptides and proteins and peptides that enable intracellular delivery may be used to deliver pharmaceutical formulations.
  • a non-limiting example of a cell penetrating peptide which may be used with the pharmaceutical formulations of the present invention include a cell-penetrating peptide sequence attached to polycations that facilitates delivery to the intracellular space, e.g., HIV- derived TAT peptide, penetratins, transportans, or hCT derived cell-penetrating peptides (see, e.g., Caron et al, Mol. Ther. 3(3):310-8 (2001); Langel, Cell-Penetrating Peptides: Processes and Applications (CRC Press, Boca Raton FL, 2002); El-Andaloussi et al., Curr. Pharm. Des.
  • compositions can also be formulated to include a cell penetrating agent, e.g., liposomes, which enhance delivery of the compositions to the intracellular space.
  • a cell penetrating agent e.g., liposomes
  • Modified nucleic acid molecules and mmRNA of the invention may be complexed to peptides and/or proteins such as, but not limited to, peptides and/or proteins from Aileron Therapeutics (Cambridge, MA) and Permeon Biologies (Cambridge, MA) in order to enable intracellular delivery (Cronican et al., ACS Chem.
  • the cell-penetrating polypeptide may comprise a first domain and a second domain.
  • the first domain may comprise a supercharged polypeptide.
  • the second domain may comprise a protein-binding partner.
  • protein-binding partner includes, but are not limited to, antibodies and functional fragments thereof, scaffold proteins, or peptides.
  • the cell- penetrating polypeptide may further comprise an intracellular binding partner for the protein-binding partner.
  • the cell-penetrating polypeptide may be capable of being secreted from a cell where the modified nucleic acid molecules or mmRNA may be introduced.
  • Formulations of the including peptides or proteins may be used to increase cell transfection by the modified nucleic acid molecule or mmRNA, alter the biodistribution of the modified nucleic acid molecule or mmRNA (e.g., by targeting specific tissues or cell types), and/or increase the translation of encoded protein.
  • alter the biodistribution of the modified nucleic acid molecule or mmRNA e.g., by targeting specific tissues or cell types
  • increase the translation of encoded protein See e.g., International Pub. No. WO2012110636; herein
  • the modified nucleic acid moleclue and mmRNA of the invention can be transfected ex vivo into cells, which are subsequently transplanted into a subject.
  • the pharmaceutical compositions may include red blood cells to deliver modified RNA to liver and myeloid cells, virosomes to deliver modified nucleic acid molecules and mmRNA in virus-like particles (VLPs), and electroporated cells such as, but not limited to, from MAXCYTE®
  • modified nucleic acid molecules and mmRNA may be delivered in synthetic VLPs synthesized by the methods described in International Pub No.
  • Cell-based formulations of the modified nucleic acid molecules and mmRNA of the invention may be used to ensure cell transfection (e.g., in the cellular carrier), alter the
  • modified nucleic acid molecule or mmRNA e.g., by targeting the cell carrier to specific tissues or cell types, and/or increase the translation of encoded protein.
  • nucleic acid into a cell includes viral and non-viral mediated techniques.
  • non-viral mediated techniques include, but are not limited to, electroporation, calcium phosphate mediated transfer, nucleofection, sonoporation, heat shock, magneto fection, liposome mediated transfer,
  • microinjection microprojectile mediated transfer (nanoparticles), cationic polymer mediated transfer (DEAE-dextran, polyethylenimine, polyethylene glycol (PEG) and the like) or cell fusion.
  • nanoparticles nanoparticles
  • cationic polymer mediated transfer DEE-dextran, polyethylenimine, polyethylene glycol (PEG) and the like
  • the technique of sonoporaiton, or cellular sonication is the use of sound (e.g., ultrasonic frequencies) for modifying the permeability of the cell plasma membrane.
  • Sonoporation methods are known to those in the art and are taught for example as it relates to bacteria in US Patent Publication 20100196983 and as it relates to other cell types in, for example, US Patent Publication 20100009424, each of which are incorporated herein by reference in their entirety.
  • modified nucleic acid molecules or mmRNA may be delivered by electroporation as described in Example 8.
  • modified nucleic acid molecules or mmRNA of the invention can include hyaluronidase, which catalyzes the hydrolysis of hyaluronan.
  • hyaluronidase catalyzes the hydrolysis of hyaluronan.
  • hyaluronidase By catalyzing the hydrolysis of hyaluronan, a constituent of the interstitial barrier, hyaluronidase lowers the viscosity of hyaluronan, thereby increasing tissue permeability (Frost, Expert Opin. Drug Deliv. (2007) 4:427-440; herein incorporated by reference in its entirety). It is useful to speed their dispersion and systemic distribution of encoded proteins produced by transfected cells.
  • the hyaluronidase can be used to increase the number of cells exposed to a modified nucleic acid molecule or mmR A of the invention administered
  • the modified nucleic acid molecules and mmRNA of the invention may be encapsulated within and/or absorbed to a nanoparticle mimic.
  • a nanoparticle mimic can mimic the delivery function organisms or particles such as, but not limited to, pathogens, viruses, bacteria, fungus, parasites, prions and cells.
  • the modified mRNA of the invention may be encapsulated in a non-viron particle which can mimic the delivery function of a virus (see
  • the modified nucleic acid molecules or mmRNA of the invention can be attached or otherwise bound to at least one nanotube such as, but not limited to, rosette nanotubes, rosette nanotubes having twin bases with a linker, carbon nanotubes and/or single-walled carbon nanotubes,
  • the modified nucleic acid molecules or mmRNA may be bound to the nanotubes through forces such as, but not limited to, steric, ionic, covalent and/or other forces.
  • the nanotube can release one or more modified nucleic acid molecule or mmRNA into cells.
  • the size and/or the surface structure of at least one nanotube may be altered so as to govern the interaction of the nanotubes within the body and/or to attach or bind to the modified nucleic acid molecule or mmRNA disclosed herein.
  • the building block and/or the functional groups attached to the building block of the at least one nanotube may be altered to adjust the dimensions and/or properties of the nanotube.
  • the length of the nanotubes may be altered to hinder the nanotubes from passing through the holes in the walls of normal blood vessels but still small enough to pass through the larger holes in the blood vessels of tumor tissue.
  • At least one nanotube may also be coated with delivery enhancing compounds including polymers, such as, but not limited to, polyethylene glycol.
  • at least one nanotube and/or the modified mRNA may be mixed with pharmaceutically acceptable excipients and/or delivery vehicles.
  • the modified mRNA are attached and/or otherwise bound to at least one rosette nanotube.
  • the rosette nanotubes may be formed by a process known in the art and/or by the process described in International Publication No. WO2012094304, herein incorporated by reference in its entirety.
  • At least one modified mRNA may be attached and/or otherwise bound to at least one rosette nanotube by a process as described in International Publication No.
  • rosette nanotubes or modules forming rosette nanotubes are mixed in aqueous media with at least one modified mRNA under conditions which may cause at least one modified mRNA to attach or otherwise bind to the rosette nanotubes.
  • the modified nucleic acid molecule or mmRNA may be attached to and/or otherwise bound to at least one carbon nanotube.
  • the modified nucleic acid molecule or mmRNA may be bound to a linking agent and the linked agent may be bound to the carbon nanotube (See e.g., U.S. Pat No. 8,246,995; herein incorporated by reference in its entirety).
  • the carbon nanotube may be a single -walled nanotube (See e.g., U.S. Pat No.
  • the modified nucleic acids molecules and mmRNA of the invention include conjugates, such as a modified nucleic acid molecule or mmRNA covalently linked to a carrier or targeting group, or including two encoding regions that together produce a fusion protein (e.g., bearing a targeting group and therapeutic protein or peptide).
  • conjugates such as a modified nucleic acid molecule or mmRNA covalently linked to a carrier or targeting group, or including two encoding regions that together produce a fusion protein (e.g., bearing a targeting group and therapeutic protein or peptide).
  • the conjugates of the invention include a naturally occurring substance, such as a protein (e.g., human serum albumin (HSA), low-density lipoprotein (LDL), high-density lipoprotein (HDL), or globulin); an carbohydrate (e.g., a dextran, pullulan, chitin, chitosan, inulin, cyclodextrin or hyaluronic acid); or a lipid.
  • the ligand may also be a recombinant or synthetic molecule, such as a synthetic polymer, e.g., a synthetic polyamino acid, an oligonucleotide (e.g. an aptamer).
  • polyamino acids examples include polyamino acid is a polylysine (PLL), poly L-aspartic acid, poly L- glutamic acid, styrene-maleic acid anhydride copolymer, poly(L-lactide-co-glycolied) copolymer, divinyl ether-maleic anhydride copolymer, N-(2-hydroxypropyl)methacrylamide copolymer (HMPA), polyethylene glycol (PEG), polyvinyl alcohol (PVA), polyurethane, poly(2-ethylacryllic acid), N-isopropylacrylamide polymers, or polyphosphazine.
  • polyamines include:
  • polyethylenimine polylysine (PLL)
  • PLL polylysine
  • spermine spermidine
  • polyamine pseudopeptide-polyamine
  • peptidomimetic polyamine dendrimer polyamine
  • arginine amidine
  • protamine cationic lipid
  • cationic porphyrin quaternary salt of a polyamine, or an alpha helical peptide.
  • the conjugate of the present invention may function as a carrier for the modified nucleic acid molecules and mmRNA of the present invention.
  • the conjugate may comprise a cationic polymer such as, but not limited to, polyamine, polylysine, polyalkylenimine, and polyethylenimine which may be grafted to with poly(ethylene glycol).
  • the conjugate may be similar to the polymeric conjugate and the method of synthesizing the polymeric conjugate described in U.S. Pat. No. 6,586,524 herein incorporated by reference in its entirety.
  • the conjugates can also include targeting groups, e.g., a cell or tissue targeting agent, e.g., a lectin, glycoprotein, lipid or protein, e.g., an antibody, that binds to a specified cell type such as a kidney cell.
  • a cell or tissue targeting agent e.g., a lectin, glycoprotein, lipid or protein, e.g., an antibody, that binds to a specified cell type such as a kidney cell.
  • a targeting group can be a thyrotropin, melanotropin, lectin, glycoprotein, surfactant protein A, Mucin carbohydrate, multivalent lactose, multivalent galactose, N-acetyl-galactosamine, N-acetyl-gulucosamine multivalent mannose, multivalent fucose, glycosylated polyaminoacids, multivalent galactose, transferrin, bisphosphonate, polyglutamate, polyaspartate, a lipid, cholesterol, a steroid, bile acid, folate, vitamin B12, biotin, an RGD peptide, an RGD peptide mimetic or an aptamer.
  • Targeting groups can be proteins, e.g., glycoproteins, or peptides, e.g., molecules having a specific affinity for a co-ligand, or antibodies e.g., an antibody, that binds to a specified cell type such as a cancer cell, endothelial cell, or bone cell.
  • Targeting groups may also include hormones and hormone receptors. They can also include non-peptidic species, such as lipids, lectins, carbohydrates, vitamins, cofactors, multivalent lactose, multivalent galactose, N-acetyl- galactosamine, N-acetyl-gulucosamine multivalent mannose, multivalent fucose, or aptamers.
  • the ligand can be, for example, a lipopolysaccharide, or an activator of p38 MAP kinase.
  • the targeting group can be any ligand that is capable of targeting a specific receptor.
  • the targeting group is an aptamer.
  • the aptamer can be unmodified or have any combination of modifications disclosed herein.
  • compositions of the present invention may include chemical modifications such as, but not limited to, modifications similar to locked nucleic acids.
  • LNA locked nucleic acid
  • Representative U.S. patents that teach the preparation of PNA compounds include, but are not limited to, U.S. Pat. Nos. 5,539,082; 5,714,331; and 5,719,262, each of which is herein incorporated by reference. Further teaching of PNA compounds can be found, for example, in Nielsen et al, Science, 1991, 254, 1497-1500.
  • Some embodiments featured in the invention include modified nucleic acids or mmRNA with phosphorothioate backbones and oligonucleosides with other modified backbones, and in particular— CH 2 — NH— CH 2 — , ⁇ CH 2 ⁇ N(CH 3 ) ⁇ 0 ⁇ CH 2 ⁇ [known as a methylene (methylimino) or MMI backbone], -CH 2 -0-N(CH 3 ) ⁇ CH 2 -, --CH 2 --N(CH 3 )--N(CH 3 )-CH 2 -- and -N(CH 3 )-CH 2 - CH 2 ⁇ [wherein the native phosphodiester backbone is represented as— O— P(0) 2 — O— CH 2 — ] of the above-referenced U.S.
  • the polynucletotides featured herein have morpholino backbone structures of the above-referenced U.S. Pat. No. 5,034,506.
  • Modifications at the 2' position may also aid in delivery.
  • modifications at the 2' position are not located in a polypeptide-coding sequence, i.e., not in a translatable region.
  • Modifications at the 2' position may be located in a 5' UTR, a 3' UTR and/or a tailing region. Modifications at the 2' position can include one of the following at the 2' position: H (i.e., 2'-deoxy); F; 0-, S-, or N-alkyl; 0-, S-, or N-alkenyl; 0-, S- or N-alkynyl; or O-alkyl-O-alkyl, wherein the alkyl, alkenyl and alkynyl may be substituted or unsubstituted Ci to Cio alkyl or C 2 to Cio alkenyl and alkynyl. Exemplary suitable modifications include 0[(CH 2 ) n O] m CH 3 , 0(CH 2 ). n OCH 3 ,
  • n and m are from 1 to about 10.
  • the modified nucleic acids or mmRNA include one of the following at the 2' position: Ci to Cio lower alkyl, substituted lower alkyl, alkaryl, aralkyl, O-alkaryl or O-aralkyl, SH, SCH 3 , OCN, CI, Br, CN, CF 3 , OCF 3 , SOCH 3 , S0 2 CH 3 , ON0 2 , N0 2 , N 3 , NH 2 , heterocycloalkyl, heterocycloalkaryl, aminoalkylamino, polyalkylamino, substituted silyl, an RNA cleaving group, a reporter group, an intercalator, a group for improving the pharmacokinetic properties, or a group for improving the pharmacodynamic properties, and other substituents having similar properties.
  • the modification includes a 2'-methoxyethoxy (2'-0— CH 2 CH 2 OCH 3 , also known as 2'-0-(2-methoxyethyl) or 2'-MOE) (Martin et al, Helv. Chim. Acta, 1995, 78:486-504) i.e., an alkoxy-alkoxy group.
  • Another exemplary modification is 2'- dimethylaminooxyethoxy, i.e., a 0(CH 2 ) 2 ON(CH 3 ) 2 group, also known as 2'-DMAOE, as described in examples herein below, and 2'-dimethylaminoethoxyethoxy (also known in the art as 2'-0- dimethylaminoethoxyethyl or 2'-DMAEOE), i.e., 2'-0 ⁇ CH 2 -0 ⁇ CH 2 -N(CH 2 ) 2 , also described in examples herein below.
  • the modified nucleic acid molecule or mmRNA is covalently conjugated to a cell-penetrating polypeptide.
  • the cell-penetrating peptide may also include a signal sequence.
  • the conjugates of the invention can be designed to have increased stability; increased cell transfection; and/or altered the biodistribution (e.g., targeted to specific tissues or cell types). Self-Assembled Nanoparticles
  • Self-assembled nanoparticles have a well-defined size which may be precisely controlled as the nucleic acid strands may be easily reprogrammable.
  • the optimal particle size for a cancer-targeting nanodelivery carrier is 20-100 nm as a diameter greater than 20 nm avoids renal clearance and enhances delivery to certain tumors through enhanced permeability and retention effect.
  • Using self-assembled nucleic acid nanoparticles a single uniform population in size and shape having a precisely controlled spatial orientation and density of cancer-targeting ligands for enhanced delivery.
  • oligonucleotide nanoparticles were prepared using programmable self-assembly of short DNA fragments and therapeutic siRNAs.
  • nanoparticles are molecularly identical with controllable particle size and target ligand location and density.
  • the DNA fragments and siRNAs self-assembled into a one-step reaction to generate DNA/siRNA tetrahedral nanoparticles for targeted in vivo delivery. (Lee et al., Nature Nanotechnology 2012 7:389-393; herein incorporated by reference in its entirety).
  • the modified nucleic acid molecules and mmRNA disclosed herein may be formulated as self-assembled nanoparticles.
  • nucleic acids may be used to make nanoparticles which may be used in a delivery system for the modified nucleic acid molecules and/or mmRNA of the present invention (See e.g., International Pub. No.
  • the nucleic acid self-assembled nanoparticles may comprise a core of the modified nucleic acid molecules or mmRNA disclosed herein and a polymer shell.
  • the polymer shell may be any of the polymers described herein and are known in the art.
  • the polymer shell may be used to protect the modified nucleic acid molecules and mmRNA in the core.
  • Polymers may be used to form sheets which self-assembled into nanoparticles. These nanoparticles may be used to deliver the modified nucleic acids and mmRNA of the present invention.
  • these self-assembled nanoparticles may be microsponges formed of long polymers of RNA hairpins which form into crystalline 'pleated' sheets before self-assembling into microsponges.
  • These microsponges are densely-packed sponge like microparticles which may function as an efficient carrier and may be able to deliver cargo to a cell.
  • the microsponges may be from lum to 300 nm in diameter.
  • the microsponges may be complexed with other agents known in the art to form larger microsponges.
  • the microsponge may be complexed with an agent to form an outer layer to promote cellular uptake such as polycation polyethyleneime (PEI).
  • PEI polycation polyethyleneime
  • This complex can form a 250-nm diameter particle that can remain stable at high temperatures (150°C) (Grabow and Jaegar, Nature Materials 2012, 11 :269-269; herein incorporated by reference in its entirety). Additionally these microsponges may be able to exhibit an extraordinary degree of protection from degradation by ribonucleases.
  • the polymer-based self-assembled nanoparticles such as, but not limited to, microsponges, may be fully programmable nanoparticles.
  • the geometry, size and stoichiometry of the nanoparticle may be precisely controlled to create the optimal nanoparticle for delivery of cargo such as, but not limited to, modified nucleic acid molecules and mmRNA.
  • the polymer based nanoparticles may comprise a core of the modified nucleic acid molecules and mmRNA disclosed herein and a polymer shell.
  • the polymer shell may be any of the polymers described herein and are known in the art.
  • the polymer shell may be used to protect the modified nucleic acid molecules and mmRNA in the core.
  • the modified nucleic acid molecules or mmRNAs of the present invention may be formulated in inorganic nanoparticles (U.S. Pat. No. 8,257,745, herein incorporated by reference in its entirety).
  • the inorganic nanoparticles may include, but are not limited to, clay substances that are water swellable.
  • the inorganic nanoparticle may include synthetic smectite clays which are made from simple silicates (See e.g., U.S. Pat. No. 5,585,108 and 8,257,745 each of which are herein incorporated by reference in their entirety).
  • the inorganic nanoparticles may comprise a core of the modified nucleic acids disclosed herein and a polymer shell.
  • the polymer shell may be any of the polymers described herein and are known in the art.
  • the polymer shell may be used to protect the modified nucleic acids in the core.
  • the modified nucleic acid molecules or mmRNAs of the present invention may be formulated in water-dispersible nanoparticle comprising a semiconductive or metallic material (U.S. Pub. No. 20120228565; herein incorporated by reference in its entirety) or formed in a magnetic nanoparticle (U.S. Pub. No. 20120265001 and 20120283503; each of which is herein incorporated by reference in its entirety).
  • the water-dispersible nanoparticles may be hydrophobic nanoparticles or hydrophilic nanoparticles.
  • the semi-conductive and/or metallic nanoparticles may comprise a core of the modified nucleic acids disclosed herein and a polymer shell.
  • the polymer shell may be any of the polymers described herein and are known in the art.
  • the polymer shell may be used to protect the modified nucleic acids in the core.
  • the modified mR A disclosed herein may be encapsulated into any hydrogel known in the art which may form a gel when injected into a subject.
  • Hydrogels are a network of polymer chains that are hydrophilic, and are sometimes found as a colloidal gel in which water is the dispersion medium. Hydrogels are highly absorbent (they can contain over 99% water) natural or synthetic polymers. Hydrogels also possess a degree of flexibility very similar to natural tissue, due to their significant water content.
  • the hydrogel described herein may used to encapsulate lipid nanoparticles which are biocompatible, biodegradable and/or porous.
  • the hydrogel may be an aptamer-functionalized hydrogel.
  • the aptamer-functionalized hydrogel may be programmed to release one or more modified nucleic acid molecules and/or mmRNA using nucleic acid hybridization.
  • the hydrogel may be a shaped as an inverted opal.
  • the opal hydrogels exhibit higher swelling ratios and the swelling kinetics is an order of magnitude faster as well.
  • Methods of producing opal hydrogels and description of opal hydrogels are described in International Pub. No. WO2012148684, herein incorporated by reference in its entirety.
  • the hydrogel may be an antibacterial hydrogel.
  • the antibacterial hydrogel may comprise a pharmaceutical acceptable salt or organic material such as, but not limited to pharmaceutical grade and/or medical grade silver salt and aloe vera gel or extract. (International Pub. No. WO2012151438, herein incorporated by reference in its entirety).
  • the modified mRNA may be encapsulated in a lipid nanoparticle and then the lipid nanoparticle may be encapsulated into a hyrdogel.
  • the modified mRNA disclosed herein may be encapsulated into any gel known in the art.
  • the gel may be a fluorouracil injectable gel or a fluorouracil injectable gel containing a chemical compound and/or drug known in the art.
  • the modified mRNA may be encapsulated in a fluorouracil gel containing epinephrine (See e.g., Smith et al. Cancer Chemotherapty and Pharmacology, 1999 44(4):267-274; herein incorporated by reference in its entirety).
  • the modified nucleic acid molecules and/or mmRNA disclosed herein may be encapsulated into a fibrin gel, fibrin hydrogel or fibrin glue.
  • the modified nucleic acid molecules and/or mmRNA may be formulated in a lipid nanoparticle or a rapidly eliminated lipid nanoparticle prior to being encapsulated into a fibrin gel, fibrin hydrogel or a fibrin glue.
  • the modified nucleic acid molecules and/or mmRNA may be formulated as a lipoplex prior to being encapsulated into a fibrin gel, hydrogel or a fibrin glue.
  • Fibrin gels, hydrogels and glues comprise two components, a fibrinogen solution and a thrombin solution which is rich in calcium (See e.g., Spicer and Mikos, Journal of Controlled Release 2010. 148: 49-55; Kidd et al. Journal of Controlled Release 2012. 157:80-85; each of which is herein incorporated by reference in its entirety).
  • the concentration of the components of the fibrin gel, hydrogel and/or glue can be altered to change the characteristics, the network mesh size, and/or the degradation characteristics of the gel, hydrogel and/or glue such as, but not limited to changing the release characteristics of the fibrin gel, hydrogel and/or glue. (See e.g., Spicer and Mikos, Journal of Controlled Release 2010.
  • This feature may be advantageous when used to deliver the modified mRNA disclosed herein. (See e.g., Kidd et al. Journal of Controlled Release 2012. 157:80-85;
  • Formulations of modified nucleic acid molecules disclosed herein may include cations or anions.
  • the formulations include metal cations such as, but not limited to, Zn2+, Ca2+, Cu2+, Mg+ and combinations thereof.
  • formulations may include polymers and a modified mRNA complexed with a metal cation (See e.g., U.S. Pat. Nos. 6,265,389 and 6,555,525, each of which is herein incorporated by reference in its entirety).
  • the modified nucleic acid molecules and/or mmRNA disclosed herein may be formulated in nanoparticles and/or microparticles. These nanoparticles and/or microparticles may be molded into any size shape and chemistry. As an example, the nanoparticles and/or microparticles may be made using the PRINT® technology by LIQUIDA TECHNOLOGIES® (Morrisville, NC) (See e.g., International Pub. No. WO2007024323; herein incorporated by reference in its entirety).
  • the molded nanoparticles may comprise a core of the modified nucleic acid molecules and/or mmRNA disclosed herein and a polymer shell.
  • the polymer shell may be any of the polymers described herein and are known in the art.
  • the polymer shell may be used to protect the modified nucleic acid molecules and/or mmRNA in the core.
  • the modified nucleic acid molecules and/or mmRNA disclosed herein may be formulated in NanoJackets and NanoLiposomes by Keystone Nano (State College, PA).
  • NanoJackets are made of compounds that are naturally found in the body including calcium, phosphate and may also include a small amount of silicates.
  • Nanojackets may range in size from 5 to 50 nm and may be used to deliver hydrophilic and hydrophobic compounds such as, but not limited to, modified nucleic acid molecules and/or mmRNA.
  • NanoLiposomes are made of lipids such as, but not limited to, lipids which naturally occur in the body. NanoLiposomes may range in size from 60-80 nm and may be used to deliver hydrophilic and hydrophobic compounds such as, but not limited to, modified nucleic acid molecules and/or mmRNA. In one aspect, the modified nucleic acids disclosed herein are formulated in a NanoLiposome such as, but not limited to, Ceramide NanoLiposomes.
  • compositions may additionally comprise a pharmaceutically acceptable excipient, which, as used herein, includes, but are not limited to, any and all solvents, dispersion media, diluents, or other liquid vehicles, dispersion or suspension aids, surface active agents, isotonic agents, thickening or emulsifying agents, preservatives, solid binders, lubricants and the like, as suited to the particular dosage form desired.
  • a pharmaceutically acceptable excipient includes, but are not limited to, any and all solvents, dispersion media, diluents, or other liquid vehicles, dispersion or suspension aids, surface active agents, isotonic agents, thickening or emulsifying agents, preservatives, solid binders, lubricants and the like, as suited to the particular dosage form desired.
  • a pharmaceutically acceptable excipient includes, but are not limited to, any and all solvents, dispersion media, diluents, or other liquid vehicles, dispersion or suspension aid
  • compositions and techniques for preparing the composition are known in the art (see Remington: The Science and Practice of Pharmacy, 21 st Edition, A. R. Gennaro, Lippincott, Williams & Wilkins, Baltimore, MD, 2006; incorporated herein by reference in its entirety).
  • the use of a conventional excipient medium may be contemplated within the scope of the present disclosure, except insofar as any conventional excipient medium may be incompatible with a substance or its derivatives, such as by producing any undesirable biological effect or otherwise interacting in a deleterious manner with any other component(s) of the pharmaceutical composition.
  • a pharmaceutically acceptable excipient may be at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% pure.
  • an excipient may be approved for use for humans and for veterinary use.
  • an excipient may be approved by United States Food and Drug Administration.
  • an excipient may be of pharmaceutical grade.
  • an excipient may meet the standards of the United States Pharmacopoeia (USP), the European Pharmacopoeia (EP), the British Pharmacopoeia, and/or the International Pharmacopoeia.
  • compositions include, but are not limited to, inert diluents, dispersing and/or granulating agents, surface active agents and/or emulsifiers, disintegrating agents, binding agents, preservatives, buffering agents, lubricating agents, and/or oils. Such excipients may optionally be included in pharmaceutical formulations.
  • the composition may also include excipients such as cocoa butter and suppository waxes, coloring agents, coating agents, sweetening, flavoring, and/or perfuming agents.
  • Exemplary diluents include, but are not limited to, calcium carbonate, sodium carbonate, calcium phosphate, dicalcium phosphate, calcium sulfate, calcium hydrogen phosphate, sodium phosphate lactose, sucrose, cellulose, microcrystalline cellulose, kaolin, mannitol, sorbitol, inositol, sodium chloride, dry starch, cornstarch, powdered sugar, etc., and/or combinations thereof.
  • Exemplary granulating and/or dispersing agents include, but are not limited to, potato starch, corn starch, tapioca starch, sodium starch glycolate, clays, alginic acid, guar gum, citrus pulp, agar, bentonite, cellulose and wood products, natural sponge, cation-exchange resins, calcium carbonate, silicates, sodium carbonate, cross-linked poly(vinyl-pyrrolidone) (crospovidone), sodium carboxymethyl starch (sodium starch glycolate), carboxymethyl cellulose, cross-linked sodium carboxymethyl cellulose (croscarmellose), methylcellulose, pregelatinized starch (starch 1500), microcrystalline starch, water insoluble starch, calcium carboxymethyl cellulose, magnesium aluminum silicate (VEEGUM®), sodium lauryl sulfate, quaternary ammonium compounds, etc., and/or combinations thereof.
  • crospovidone cross-linked poly(vinyl-pyrrolidone)
  • Exemplary surface active agents and/or emulsifiers include, but are not limited to, natural emulsifiers ⁇ e.g. acacia, agar, alginic acid, sodium alginate, tragacanth, chondrux, cholesterol, xanthan, pectin, gelatin, egg yolk, casein, wool fat, cholesterol, wax, and lecithin), colloidal clays (e.g. bentonite [aluminum silicate] and VEEGUM [magnesium aluminum silicate]), long chain amino acid derivatives, high molecular weight alcohols (e.g.
  • stearyl alcohol cetyl alcohol, oleyl alcohol, triacetin monostearate, ethylene glycol distearate, glyceryl monostearate, and propylene glycol monostearate, polyvinyl alcohol), carbomers (e.g. carboxy polymethylene, polyacrylic acid, acrylic acid polymer, and carboxyvinyl polymer), carrageenan, cellulosic derivatives (e.g.
  • carboxymethylcellulose sodium powdered cellulose, hydroxymethyl cellulose, hydroxypropyl cellulose, hydroxypropyl methylcellulose, methylcellulose), sorbitan fatty acid esters (e.g.
  • polyoxyethylene esters e.g. polyoxyethylene monostearate [MYRJ ® 45], polyoxyethylene hydrogenated castor oil, polyethoxylated castor oil, polyoxymethylene stearate, and SOLUTOL ®
  • sucrose fatty acid esters e.g. CREMOPHOR ®
  • polyoxyethylene ethers e.g.
  • polyoxyethylene lauryl ether [BRIJ ® 30]), poly(vinyl-pyrrolidone), diethylene glycol monolaurate, triethanolamine oleate, sodium oleate, potassium oleate, ethyl oleate, oleic acid, ethyl laurate, sodium lauryl sulfate, PLUORINC ® F 68, POLOXAMER ® 188, cetrimonium bromide, cetylpyridinium chloride, benzalkonium chloride, docusate sodium, etc. and/or
  • Exemplary binding agents include, but are not limited to, starch (e.g. cornstarch and starch paste); gelatin; sugars (e.g. sucrose, glucose, dextrose, dextrin, molasses, lactose, lactitol, mannitol,); natural and synthetic gums (e.g. acacia, sodium alginate, extract of Irish moss, panwar gum, ghatti gum, mucilage of isapol husks, carboxymethylcellulose, methylcellulose, ethylcellulose,
  • starch e.g. cornstarch and starch paste
  • gelatin e.g. sucrose, glucose, dextrose, dextrin, molasses, lactose, lactitol, mannitol
  • natural and synthetic gums e.g. acacia, sodium alginate, extract of Irish moss, panwar gum, ghatti gum, mucilage of isapol husk
  • hydroxyethylcellulose hydroxypropyl cellulose, hydroxypropyl methylcellulose, microcrystalline cellulose, cellulose acetate, poly(vinyl-pyrrolidone), magnesium aluminum silicate (VEEGUM ® ), and larch arabogalactan); alginates; polyethylene oxide; polyethylene glycol; inorganic calcium salts; silicic acid; polymethacrylates; waxes; water; alcohol; etc.; and combinations thereof.
  • Exemplary preservatives may include, but are not limited to, antioxidants, chelating agents, antimicrobial preservatives, antifungal preservatives, alcohol preservatives, acidic preservatives, and/or other preservatives.
  • Exemplary antioxidants include, but are not limited to, alpha tocopherol, ascorbic acid, acorbyl palmitate, butylated hydroxyanisole, butylated hydroxytoluene,
  • chelating agents include ethylenediaminetetraacetic acid (EDTA), citric acid monohydrate, disodium edetate, dipotassium edetate, edetic acid, fumaric acid, malic acid, phosphoric acid, sodium edetate, tartaric acid, and/or trisodium edetate.
  • EDTA ethylenediaminetetraacetic acid
  • Exemplary antimicrobial preservatives include, but are not limited to, benzalkonium chloride, benzethonium chloride, benzyl alcohol, bronopol, cetrimide,
  • cetylpyridinium chloride chlorhexidine, chlorobutanol, chlorocresol, chloroxylenol, cresol, ethyl alcohol, glycerin, hexetidine, imidurea, phenol, phenoxyethanol, phenylethyl alcohol,
  • Exemplary antifungal preservatives include, but are not limited to, butyl paraben, methyl paraben, ethyl paraben, propyl paraben, benzoic acid, hydroxybenzoic acid, potassium benzoate, potassium sorbate, sodium benzoate, sodium propionate, and/or sorbic acid.
  • Exemplary alcohol preservatives include, but are not limited to, ethanol, polyethylene glycol, phenol, phenolic compounds, bisphenol, chlorobutanol, hydroxybenzoate, and/or phenylethyl alcohol.
  • Exemplary acidic preservatives include, but are not limited to, vitamin A, vitamin C, vitamin E, beta-carotene, citric acid, acetic acid, dehydroacetic acid, ascorbic acid, sorbic acid, and/or phytic acid.
  • preservatives include, but are not limited to, tocopherol, tocopherol acetate, deteroxime mesylate, cetrimide, butylated hydroxyanisol (BHA), butylated hydroxytoluened (BHT), ethylenediamine, sodium lauryl sulfate (SLS), sodium lauryl ether sulfate (SLES), sodium bisulfite, sodium metabisulfite, potassium sulfite, potassium metabisulfite, GLYDANT PLUS ® , PHENONIP ® , methylparaben, GERM ALL ® 115,
  • GERMABEN ® II NEOLONE TM , KATHON TM , and/or EUXYL ® .
  • Exemplary buffering agents include, but are not limited to, citrate buffer solutions, acetate buffer solutions, phosphate buffer solutions, ammonium chloride, calcium carbonate, calcium chloride, calcium citrate, calcium glubionate, calcium gluceptate, calcium gluconate, d-gluconic acid, calcium glycerophosphate, calcium lactate, propanoic acid, calcium levulinate, pentanoic acid, dibasic calcium phosphate, phosphoric acid, tribasic calcium phosphate, calcium hydroxide phosphate, potassium acetate, potassium chloride, potassium gluconate, potassium mixtures, dibasic potassium phosphate, monobasic potassium phosphate, potassium phosphate mixtures, sodium acetate, sodium bicarbonate, sodium chloride, sodium citrate, sodium lactate, dibasic sodium phosphate, monobasic sodium phosphate, sodium phosphate mixtures, tromethamine, magnesium hydroxide, aluminum hydroxide, alginic acid, pyrogen-free water
  • Exemplary lubricating agents include, but are not limited to, magnesium stearate, calcium stearate, stearic acid, silica, talc, malt, glyceryl behanate, hydrogenated vegetable oils, polyethylene glycol, sodium benzoate, sodium acetate, sodium chloride, leucine, magnesium lauryl sulfate, sodium lauryl sulfate, etc., and combinations thereof.
  • oils include, but are not limited to, almond, apricot kernel, avocado, babassu, bergamot, black current seed, borage, cade, camomile, canola, caraway, carnauba, castor, cinnamon, cocoa butter, coconut, cod liver, coffee, corn, cotton seed, emu, eucalyptus, evening primrose, fish, flaxseed, geraniol, gourd, grape seed, hazel nut, hyssop, isopropyl myristate, jojoba, kukui nut, lavandin, lavender, lemon, litsea cubeba, macademia nut, mallow, mango seed, meadowfoam seed, mink, nutmeg, olive, orange, orange roughy, palm, palm kernel, peach kernel, peanut, poppy seed, pumpkin seed, rapeseed, rice bran, rosemary, safflower, sandalwood, sasquana,
  • oils include, but are not limited to, butyl stearate, caprylic triglyceride, capric triglyceride, cyclomethicone, diethyl sebacate, dimethicone 360, isopropyl myristate, mineral oil, octyldodecanol, oleyl alcohol, silicone oil, and/or combinations thereof.
  • Excipients such as cocoa butter and suppository waxes, coloring agents, coating agents, sweetening, flavoring, and/or perfuming agents can be present in the composition, according to the judgment of the formulator.
  • the present disclosure encompasses the delivery of modified nucleic acid molecules or mmRNA for any of therapeutic, pharmaceutical, diagnostic or imaging by any appropriate route taking into consideration likely advances in the sciences of drug delivery. Delivery may be naked or formulated.
  • the modified nucleic acid molecules or mmRNA of the present invention may be delivered to a cell naked.
  • naked refers to delivering modified nucleic acid molecules or mmRNA free from agents which promote transfection.
  • the modified nucleic acid molecules or mmRNA delivered to the cell may contain no modifications.
  • the naked modified nucleic acid molecules or mmRNA may be delivered to the cell using routes of administration known in the art and described herein.
  • the modified nucleic acid molecules or mmR A of the present invention may be formulated, using the methods described herein.
  • the formulations may contain modified nucleic acid molecules or mmRNA which may be modified and/or unmodified.
  • the formulations may further include, but are not limited to, cell penetration agents, a pharmaceutically acceptable carrier, a delivery agent, a bioerodible or biocompatible polymer, a solvent, and a sustained-release delivery depot.
  • the formulated modified nucleic acid molecules or mmRNA may be delivered to the cell using routes of administration known in the art and described herein.
  • compositions may also be formulated for direct delivery to an organ or tissue in any of several ways in the art including, but not limited to, direct soaking or bathing, via a catheter, by gels, powder, ointments, creams, gels, lotions, and/or drops, by using substrates such as fabric or biodegradable materials coated or impregnated with the compositions, and the like.
  • the modified nucleic acid molecules or mmRNA of the present invention may be administered by any route which results in a therapeutically effective outcome. These include, but are not limited to enteral, gastroenteral, epidural, oral, transdermal, epidural (peridural), intracerebral (into the cerebrum), intracerebroventricular (into the cerebral ventricles), epicutaneous (application onto the skin), intradermal, (into the skin itself), subcutaneous (under the skin), nasal administration (through the nose), intravenous (into a vein), intraarterial (into an artery), intramuscular (into a muscle), intracardiac (into the heart), intraosseous infusion (into the bone marrow), intrathecal (into the spinal canal), intraperitoneal, (infusion or injection into the peritoneum), intravesical infusion, intravitreal, (through the eye), intracavernous injection, ( into the base of the penis), intravaginal administration, intrauterine, extra-am
  • Liquid dosage forms for parenteral administration include, but are not limited to, pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups, and/or elixirs.
  • liquid dosage forms may comprise inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, dimethylformamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof.
  • inert diluents commonly used in the art such as, for example, water or
  • oral compositions can include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and/or perfuming agents.
  • adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and/or perfuming agents.
  • compositions are mixed with solubilizing agents such as CREMOPHOR ® , alcohols, oils, modified oils, glycols, polysorbates, cyclodextrins, polymers, and/or combinations thereof.
  • Injectable preparations for example, sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing agents, wetting agents, and/or suspending agents.
  • Sterile injectable preparations may be sterile injectable solutions, suspensions, and/or emulsions in nontoxic parenterally acceptable diluents and/or solvents, for example, as a solution in 1,3-butanediol.
  • the acceptable vehicles and solvents that may be employed are water, Ringer's solution, U.S.P., and isotonic sodium chloride solution.
  • Sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil can be employed including synthetic mono- or diglycerides.
  • Fatty acids such as oleic acid can be used in the preparation of injectables.
  • Injectable formulations can be sterilized, for example, by filtration through a bacterial- retaining filter, and/or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved or dispersed in sterile water or other sterile injectable medium prior to use.
  • Depot injectable formulations are prepared by entrapping the drug in liposomes or microemulsions which are compatible with body tissues.
  • compositions for rectal or vaginal administration are typically suppositories which can be prepared by mixing compositions with suitable non-irritating excipients such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active ingredient.
  • suitable non-irritating excipients such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active ingredient.
  • Liquid dosage forms for oral administration include, but are not limited to,
  • liquid dosage forms may comprise inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, dimethylformamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof.
  • inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl
  • oral compositions can include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and/or perfuming agents.
  • adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and/or perfuming agents.
  • compositions are mixed with solubilizing agents such as CREMOPHOR ® , alcohols, oils, modified oils, glycols, polysorbates, cyclodextrins, polymers, and/or combinations thereof.
  • Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules.
  • an active ingredient is mixed with at least one inert, pharmaceutically acceptable excipient such as sodium citrate or dicalcium phosphate and/or fillers or extenders ⁇ e.g. starches, lactose, sucrose, glucose, mannitol, and silicic acid), binders ⁇ e.g.
  • the dosage form may comprise buffering agents.
  • compositions containing the modified nucleic acid molecules or mmRNA of the invention may be formulated for administration topically.
  • the skin may be an ideal target site for delivery as it is readily accessible. Gene expression may be restricted not only to the skin, potentially avoiding nonspecific toxicity, but also to specific layers and cell types within the skin.
  • the site of cutaneous expression of the delivered compositions will depend on the route of nucleic acid delivery.
  • Three routes are commonly considered to deliver modified nucleic acid molecules or mmRNA to the skin: (i) topical application (e.g. for local/regional treatment); (ii) intradermal injection (e.g. for local/regional treatment); and (iii) systemic delivery (e.g. for treatment of dermato logic diseases that affect both cutaneous and extracutaneous regions).
  • Modified nucleic acid molecules or mmRNA can be delivered to the skin by several different approaches known in the art.
  • the invention provides for a variety of dressings (e.g., wound dressings) or bandages (e.g., adhesive bandages) for conveniently and/or effectively carrying out methods of the present invention.
  • dressing or bandages may comprise sufficient amounts of pharmaceutical compositions and/or modified nucleic acid molecules or mmRNA described herein to allow a user to perform multiple treatments of a subject(s).
  • the invention provides for the modified nucleic acid molecules or mmRNA compositions to be delivered in more than one injection.
  • tissue before topical and/or transdermal administration at least one area of tissue, such as skin, may be subjected to a device and/or solution which may increase permeability.
  • the tissue may be subjected to an abrasion device to increase the permeability of the skin (see U.S. Patent Publication No. 20080275468, herein incorporated by reference in its entirety).
  • the tissue may be subjected to an ultrasound enhancement device.
  • An ultrasound enhancement device may include, but is not limited to, the devices described in U.S. Publication No. 20040236268 and U.S. Patent Nos. 6,491,657 and 6,234,990; each of which are herein incorporated by reference in their entireties.
  • a device may be used to increase permeability of tissue before delivering formulations of modified mRNA described herein.
  • the permeability of skin may be measured by methods known in the art and/or described in U.S. Patent No. 6,190,315, herein incorporated by reference in its entirety.
  • a modified mRNA formulation may be delivered by the drug delivery methods described in U.S. Patent No. 6,190,315, herein incorporated by reference in its entirety.
  • tissue may be treated with a eutectic mixture of local anesthetics (EMLA) cream before, during and/or after the tissue may be subjected to a device which may increase permeability.
  • EMLA local anesthetics

Abstract

The present disclosure provides, inter alia, formulation compositions comprising modified nucleic acid molecules which may encode a protein, a protein precursor, or a partially or fully processed form of the protein or a protein precursor. The formulation composition may further include a modified nucleic acid molecule and a delivery agent. The present invention further provides nucleic acids useful for encoding polypeptides capable of modulating a cell's function and/or activity.

Description

MODIFIED NUCLEOSIDE, NUCLEOTIDE, AND NUCLEIC ACID COMPOSITIONS
REFERENCE TO SEQUENCE LISTING
[0001] The present application is being filed along with a Sequence Listing in electronic format. The Sequence Listing file, entitled Ml lPCTSQLST.txt, was created on December 14, 2012 and is 25,579 bytes in size. The information in electronic format of the Sequence Listing is incorporated herein by reference in its entirety.
CROSS REFERENCE TO RELATED APPLICATIONS
[0002] This application claims the benefit of U.S. Provisional Patent Application No. 61/576,705, filed December 16, 2011, entitled Modified Nucleoside, Nucleotide, and Nucleic Acid
Compositions, U.S. Provisional Patent Application No. 61/618,957, filed April 2, 2012, entitled Modified Nucleoside, Nucleotide, and Nucleic Acid Compositions, U.S. Provisional Patent
Application No. 61/648,244, filed May 17, 2012, entitled Modified Nucleoside, Nucleotide, and Nucleic Acid Compositions, U.S. Provisional Patent Application No. 61/681,712, filed August 10,2012, entitled Modified Nucleoside, Nucleotide, Nucleic Acid Compositions and U.S. Provisional Patent Application No. 61/696,381 filed September 4, 2012, entitled Modified Nucleoside,
Nucleotide and Nucleic Acid Compositions, and Nucleic Acid Compositions, U.S. Provisional Patent Application No. 61/709,303 filed October 3, 2012, entitled Modified Nucleoside, Nucleotide and Nucleic Acid Compositions, U.S. Provisional Patent Application No. 61/712,490 filed October 11, 2012, entitled Modified Nucleoside, Nucleotide and Nucleic Acid Compositions and
International Pub. No. PCT/US2012/058519 filed October 3, 2012 Modified Nucleosides,
Nucleotides, and Nucleic Acids, And Uses Thereof, the contents of which are incorporated herein by reference in their entirety.
BACKGROUND OF THE INVENTION
[0003] In general, exogenous unmodified nucleic acid molecules, particularly viral nucleic acids, introduced into the cell induce an innate immune response which results in cytokine and interferon (IFN) production and ultimately cell death. It is of great interest for therapeutics, diagnostics, reagents and for biological assays to be able to deliver a nucleic acid, e.g., a ribonucleic acid (RNA), into a cell, such as to cause intracellular translation of the nucleic acid and production of the encoded protein instead of generating an innate immune response. Thus, there is a need to develop formulation compositions comprising a delivery agent that can effectively facilitate the in vivo delivery of nucleic acids to targeted cells without generating an innate immune response.
SUMMARY OF THE INVENTION
[0004] The present disclosure provides, inter alia, formulation compositions comprising modified nucleic acid molecules which may encode a protein, a protein precursor, or a partially or fully processed form of the protein or a protein precursor. The formulation compositions may further include a modified nucleic acid molecule and a delivery agent. The present invention further provides nucleic acids useful for encoding polypeptides capable of modulating a cell's function and/or activity.
[0005] In one aspect a method of producing a polypeptide of interest in a mammalian cell or tissue is described. The method comprises contacting the mammalian cell or tissue with a formulation comprising a modified mRNA encoding a polypeptide of interest. The formulation may be, but is not limited to, nanoparticles, poly(lactic-co-glycolic acid)(PLGA) microspheres, lipidoids, lipoplex, liposome, polymers, carbohydrates (including simple sugars), cationic lipids, fibrin gel, fibrin hydrogel, fibrin glue, fibrin sealant, fibrinogen, thrombin, rapidly eliminated lipid nanoparticles (reLNPs) and combinations thereof. The modified mRNA may comprise a purified IVT transcript.
[0006] In one embodiment, the formulation comprising the modified mRNA is a nanoparticle which may comprise at least one lipid. The lipid may be selected from, but is not limited to, DLin- DMA, DLin-K-DMA, 98N12-5, C12-200, DLin-MC3 -DMA, DLin-KC2-DMA, DODMA, PLGA, PEG, PEG-DMG and PEGylated lipids. In another aspect, the lipid may be a cationic lipid such as, but not limited to, DLin-DMA, DLin-D-DMA, DLin-MC3 -DMA, DLin-KC2-DMA and DODMA.
[0007] The lipid to modified mRNA ration in the formulation may be between 10: 1 and 30: 10. The mean size of the nanoparticle formulation may comprise the modified mRNA between 60 and 225 nm. The PDI of the nanoparticle formulation comprising the modified mRNA is between 0.03 and 0.15. The zeta potential of the lipid may be from -10 to +10 at a pH of 7.4
[0008] The formulations of modified mRNA may comprise a fusogenic lipid, cholesterol and a PEG lipid. The formulation may have a molar ratio 50: 10:38.5:1.5-3.0 (cationic lipid:fusogenic lipid: cholesterol: PEG lipid). The PEG lipid may be selected from, but is not limited to PEG-c- DOMG, PEG-DMG. The fusogenic lipid may be DSPC. [0009] The mammalian cell or tissue may be contacted using a device such as, but not limited to, a syringe pump, internal osmotic pump and external osmotic pump.
[0010] The formulation of modified mR A may be a PLGA microsphere which may be between 4 and 20 μιη in size. The modified mRNA may be released from the formulation at less than 50% in a 48 hour time period. The PLGA microsphere formulation may be stable in serum. Stability may be determined relative to unformulated modified mRNA in 90%.
[0011] The loading weight percent of the modified mRNA PLGA microsphere may be at least 0.05%, at least 0.1%, at least 0.2%, at least 0.3%, at least 0.4% or at least 0.5%. The encapsulation efficiency of the modified mRNA in the PLGA microsphere may be at least 50%, at least 70%, at least 90% or at least 97%.
[0012] A lipid nanoparticle of the present invention may be formulated in a sealant such as, but not limited to, a fibrin sealant.
[0013] The mammalian cells or tissues may be contacted by a route of administration such as, but not limited to, intravenous, intramuscular, intravitreal, intrathecal, intratumoral, pulmonary and subcutaneous. The mammalian cells or tissues may be contacted using a split dosing schedule. The mammalian cell or tissue may be contacted by injection. The injection may be made to tissue selected from the group consisting of intradermal space, epidermis, subcutaneous tissue and muscle. The polypeptide of interest may be produced in the cell or tissue in a location systemic from the location of contacting.
[0014] The polypeptide of interest may be detectable in serum for up to 72 hours after contacting. The level of the polypeptide of interest can be higher than the levels prior to dosing. The level of the polypeptide of interest may be greater in the serum of female subjects than in the serum of male subjects.
[0015] The formulation of modified mRNA may comprise more than one modified mRNA. The formulation may have two or three modified mRNA.
[0016] The formulation comprising the modified mRNA may comprise a rapidly eliminated lipid nanoparticle (reLNP) which may comprise a reLNP lipid, fusogenic lipid, cholesterol and a PEG lipid at a molar ratio of 50: 10: 38.5: 1.5 (reLNP lipid:fusogenic lipid: cholesterol: PEG lipid). The fusogenic lipid may be DSPC and the PEG lipid may be PEG-c-DOMG. The reLNP lipid may be DLin-DMA with an internal or terminal ester or DLin-MC3-DMA with an internal or terminal ester. The total lipid to modified mRNA weight ration may be between 10: 1 and 30: 1. [0017] The formulation comprising modified mRNA may comprise a fibrin sealant.
[0018] The formulation comprising modified mRNA may comprise a lipidoid where the lipid is selected from the group consisting of CI 2-200 and 98N12-5.
[0019] The formulation comprising modified mRNA may include a polymer. The polymer may be coated, covered, surrounded, enclosed or comprise a layer of a hydrogel or surgical sealant. The polymer may be selected from the group consisting of PLGA, ethylene vinyl acetate, poloxamer and GELSITE®.
[0020] A polypeptide of interest may be produced in a mammalian cell or tissue by contacting the mammalian cell or tissue with a buffer formulation comprising a modified mRNA encoding the polypeptide of interest. The buffer formulation may be selected from, but is not limited to, slaine, phosphate buffered saline and Ringer's lactate. The buffer formulation may comprise a calcium concentration of between 1 to 10 mM. The modified mRNA in the buffer formulation may comprise a purified IVT transcript.
[0021] A pharmacologic effect in a primate may be produced by contacting the primate with a composition comprising a formulated modified mRNA encoding a polypeptide of interest. The modified mRNA may comprise a purified IVT transcript and/or may be formulated in nanoparticles, poly(lactic-co-glycolic acid)(PLGA) microspheres, lipidoids, lipoplex, liposome, polymers, carbohydrates (including simple sugars), cationic lipids, fibrin gel, fibrin hydrogel, fibrin glue, fibrin sealant, fibrinogen, thrombin, rapidly eliminated lipid nanoparticles (reLNPs) and combinations thereof. The pharmacological effect may be greater than the pharmacologic effect associated with a therapeutic agent and/or composition known to produce said pharmacologic effect. The composition may comprise a formulated or unformulated modified mRNA. The pharmacologic effect may result in a therapeutically effective outcome of a disease, disorder, condition or infection. Such therapeutically effective outcome may include, but is not limited to, treatment, improvement of one or more symptoms, diagnosis, prevention, and delay of onset. The pharmacologic effect may include, but is not limited to, change in cell count, alteration in serum chemistry, alteration of enzyme activity, increase in hemoglobin, and increase in hematocrit.
[0022] In one embodiment, the present disclosure provides a formulation composition which comprises a modified nucleic acid molecule and a delivery agent. The modified nucleic acid molecule may be selected from the group consisting of DNA, complimentary DNA (cDNA), RNA, messenger RNA (mRNA), RNAi-inducing agents, RNAi agents, siRNA, shRNA, miRNA, antisense R A, ribozymes, catalytic DNA, R A that induce triple helix formation, aptamers, vectors and combinations thereof. If the modified nucleic acid molecule is mR A the mR A may be derived from cDNA.
[0023] In one embodiment, the modified nucleic acid molecule may comprise at least one modification and a translatable region. In some instances, the modified nucleic acid comprises at least two modifications and a translatable region. The modification may be located on the backbone and/or a nucleoside of the nucleic acid molecule. The modification may be located on both a nucleoside and a backbone linkage.
[0024] In one embodiment, a modification may be located on the backbone linkage of the modified nucleic acid molecule. The backbone linkage may be modified by replacing of one or more oxygen atoms. The modification of the backbone linkage may comprise replacing at least one phosphodiester linkage with a phosphorothioate linkage.
[0025] In one embodiment, a modification may be located on a nucleoside of the modified nucleic acid molecule. The modification on the nucleoside may be located on the sugar of said nucleoside. The modification of the nucleoside may occur at the 2' position on the nucleoside.
[0026] The nucleoside modification may include a compound selected from the group consisting of pyridin-4-one ribonucleoside, 5-aza-uridine, 2-thio-5-aza-uridine, 2-thiouridine, 4-thio- pseudouridine, 2-thio-pseudouridine, 5-hydroxyuridine, 3-methyluridine, 5-carboxymethyl-uridine, 1-carboxymethyl-pseudouridine, 5-propynyl-uridine, 1-propynyl-pseudouridine, 5- taurinomethyluridine, 1-taurinomethyl-pseudouridine, 5-taurinomethyl-2-thio-uridine, 1- taurinomethyl-4-thio-uridine, 5-methyl-uridine, 1-methyl-pseudouridine, 4-thio-l-methyl- pseudouridine, 2-thio- 1 -methyl-pseudouridine, 1 -methyl- 1 -deaza-pseudouridine, 2-thio- 1 -methyl- 1 - deaza-pseudouridine, dihydrouridine, dihydropseudouridine, 2-thio-dihydrouridine, 2-thio- dihydropseudouridine, 2-methoxyuridine, 2-methoxy-4-thio-uridine, 4-methoxy-pseudouridine, 4- methoxy-2-thio-pseudouridine, 5-aza-cytidine, pseudoisocytidine, 3-methyl-cytidine, N4- acetylcytidine, 5-formylcytidine, N4-methylcytidine, 5-hydroxymethylcytidine, 1-methyl- pseudoisocytidine, pyrrolo-cytidine, pyrrolo-pseudoisocytidine, 2-thio-cytidine, 2-thio-5-methyl- cytidine, 4-thio-pseudoisocytidine, 4-thio-l-methyl-pseudoisocytidine, 4-thio-l -methyl- 1-deaza- pseudoisocytidine, 1 -methyl- 1-deaza-pseudoisocytidine, zebularine, 5-aza-zebularine, 5-methyl- zebularine, 5-aza-2-thio-zebularine, 2-thio-zebularine, 2-methoxy-cytidine, 2-methoxy-5-methyl- cytidine, 4-methoxy-pseudoisocytidine, 4-methoxy-l-methyl-pseudoisocytidine, 2-aminopurine, 2, 6-diaminopurine, 7-deaza-adenine, 7-deaza-8-aza-adenine, 7-deaza-2-aminopurine, 7-deaza-8-aza-2- aminopurine, 7-deaza-2,6-diaminopurine, 7-deaza-8-aza-2,6-diaminopurine, 1-methyladenosine, N6- methyladenosine, N6-isopentenyladenosine, N6-(cis-hydroxyisopentenyl)adenosine, 2-methylthio- N6-(cis-hydroxyisopentenyl) adenosine, N6-glycinylcarbamoyladenosine, N6- threonylcarbamoyladenosine, 2-methylthio-N6-threonyl carbamoyladenosine, N6,N6- dimethyladenosine, 7-methyladenine, 2-methylthio-adenine, 2-methoxy-adenine, inosine, 1-methyl- inosine, wyosine, wybutosine, 7-deaza-guanosine, 7-deaza-8-aza-guanosine, 6-thio-guanosine, 6- thio-7-deaza-guanosine, 6-thio-7-deaza-8-aza-guanosine, 7-methyl-guanosine, 6-thio-7-methyl- guanosine, 7-methylinosine, 6-methoxy-guanosine, 1-methylguanosine, N2-methylguanosine, N2,N2-dimethylguanosine, 8-oxo-guanosine, 7-methyl-8-oxo-guanosine, l-methyl-6-thio-guanosine, N2-methyl-6-thio-guanosine, and N2,N2-dimethyl-6-thio-guanosine. In another embodiment, the modifications are independently selected from the group consisting of 5-methylcytosine,
pseudouridine and 1-methylpseudouridine
[0027] In one embodiment, a modification may be located on a nucleobase of the modified nucleic acid molecule. The modification on the nucleobase may be selected from the group consisting of cytosine, guanine, adenine, thymine and uracil. The modification on the nucleobase may be selected from the group consisting of deaza-adenosine and deaza-guanosine, and the linker may be attached at a C-7 or C-8 position of said deaza-adenosine or deaza-guanosine. The modified nucleobase may be selected from the group consisting of cytosine and uracil, and the linker may be attached to the modified nucleobase at an N-3 or C-5 position. The linker attached to the nucleobase may be selected from the group consisting of diethylene glycol, dipropylene glycol, triethylene glycol, tripropylene glycol, tetraethylene glycol, tetraethylene glycol, divalent alkyl, alkenyl, alkynyl moiety, ester, amide, and ether moiety.
[0028] In one embodiment, two modifications of the nucleic acid molecule may be located on nucleosides of the modified nucleic acid molecule. The modified nucleosides may be selected from 5-methylcytosine and pseudouridine.
[0029] In one embodiment, two modifications of the modified nucleic acid molecule may be located on a nucleotide or a nucleoside. In one embodiment, the present disclosure provides a formulation comprising a nucleic acid molecule such as, but not limited to, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 9 and SEQ ID NO: 10 and a delivery agent. The nucleic acid molecule may comprise a polyA tail about 160 nucleotides in length. Further, the nucleic acid molecule may comprise at least one 5' terminal cap such as, but not limited to, CapO, Capl, ARC A, inosine, Nl- methyl-guanosine, 2'fluoro-guanosine, 7-deaza-guanosine, 8-oxo-guanosine, 2-amino-guanosine, LNA-guanosine, and 2-azido-guanosine.
[0030] In one embodiment, the present disclosure provides a nucleic acid of SEQ ID NO: 6, a 5' terminal cap which is Capl, a poly A tail of approximately 160 nucleotides in length and a delivery agent.
[0031] In one embodiment, the present disclosure provides a nucleic acid of SEQ ID NO: 7, a 5' terminal cap which is Capl, a poly A tail of approximately 160 nucleotides in length and a delivery agent.
[0032] In one embodiment, the present disclosure provides a nucleic acid of SEQ ID NO: 9, a 5' terminal cap which is Capl, a poly A tail of approximately 160 nucleotides in length and a delivery agent.
[0033] In one embodiment, the present disclosure provides a nucleic acid of SEQ ID NO: 10, a 5' terminal cap which is Capl, a poly A tail of approximately 160 nucleotides in length and a delivery agent.
[0034] In one embodiment, the delivery agent comprises at least one method to improve delivery selected from the group consisting of lipidoids, liposomes, lipid nanoparticles, rapidly eliminated lipid nanoparticles (reLNPs), polymers, lipoplexes, peptides, proteins, hydrogels, sealants, chemical modifications, conjugation, cells and enhancers. The lipidoid, lipid nanoparticle and rapidly eliminated lipid nanoparticles which may be used as a delivery agent may include a lipid which may be selected from the group consisting of C12-200, MD1, 98N12-5, DLin-DMA, DLin-K-DMA, DLin-KC2-DMA, DLin-MC3 -DMA, PLGA, PEG, PEG-DMG, PEGylated lipids and analogs thereof. The rapidly eliminated lipid nanoparticle may have an ester linkage at the terminal end of the lipid chain, or an ester linkage may be an internal linkage located to the right or left of a saturated carbon in the lipid chain. The rapidly eliminated lipid nanoparticle which may be used as a delivery agent may be, but is not limited to, DLin-MC3-DMA and DLin-DMA.
[0035] In one embodiment, the lipid nanoparticle may comprise PEG and at least one component such as, but not limited to, cholesterol, cationic lipid and fusogenic lipid.
[0036] In one embodiment, the lipid nanoparticle may comprise at least one of a PEG, cholesterol, cationic lipid and fusogenic lipid. [0037] In one embodiment, the fusogenic lipid is disteroylphophatidyl choline (DSPC). In another embodiment, the PEG lipid is PEG-DMG. In yet another embodiment, the cationic lipid may be, but not limited to, DLin-DMA, DLin-MC3 -DM A, C12-200, 98N12-5 and DLin-KC2- DMA.
[0038] In one embodiment, the lipid nanoparticle composition may comprise 50 mol% cationic lipid, 10 mol% DSPC, 1.5-3.0 mol% PEG and 37-38.5 mol% cholesterol.
[0039] In one embodiment, a modified nucleic acid may be formulated with PLGA to form a sustained release formulation. In another embodiment, a modified nucleic acid may be formulated with PLGA and other active and/or inactive components to form a sustained release formulation. In one embodiment, the modified nucleic acid molecule may include, but is not limited to, SEQ ID NO: 9 and SEQ ID NO: 10.
[0040] In one embodiment, a sustained release formulation may comprise a sustained release microsphere. The sustained release microsphere may be about 10 to about 50 um in diameter. In another embodiment, the sustained release microsphere may contain about 0.001 to about 1.0 weight percent of at least one modified nucleic acid molecule.
[0041] In one embodiment, the modified nucleic acids of the present invention may include at least one stop codon before the 3' untranslated region (UTR). The stop codon may be selected from TGA, TAA and TAG. In one embodiment, the modified nucleic acids of the present invention include the stop codon TGA and one additional stop codon. In a further embodiment the addition stop codon may be TAA. In another embodiment, the modified nucleic acid of the present invention includes three stop codons.
[0042] In one embodiment, the present disclosure provides a controlled release formulation comprising a modified nucleic acid which may encode a polypeptide of interest. The modified nucleic acid may be encapsulated or substantially encapsulated in a delivery agent. The delivery agent may be coated, covered, surrounded, enclosed or comprise a layer of polymer, hydrogel and/or surgical sealant. In a further embodiment, the controlled release formulation may comprise a second layer of polymer, hydrogel and/or surgical sealant.
[0043] In one embodiment, the delivery agent of the controlled release formulation may include, but is not limited to, lipidoids, liposomes, lipid nanoparticles, rapidly eliminated lipid nanoparticles, lipoplexes and self-assembled lipid nanoparticles.
[0044] [0045] The polymer which may be used in the controlled release formulation may include, but is not limited to, PLGA, ethylene vinyl acetate, poloxamer and GELSITE®. The surgical sealant which may be used in the controlled release formulation may include, but is not limited to, fibrinogen polymers, TISSEELL®, PEG-based sealants and COSEAL®.
[0046] In one embodiment, the delivery agent of the controlled release formulation comprises a lipid nanoparticle or a rapidly eliminated lipid nanoparticle delivery agent. In one aspect, the lipid nanoparticle or rapidly eliminated lipid nanoparticle may be coated, substantially coated, covered, substantially covered, surrounded, substanitally surrounded, enclosed, substantially enclosed or comprises a layer of polymer, hydrogel and/or surgical sealant. In another aspect, the delivery agent may be a lipid nanoparticle which may be coated, substantially coated, covered, substantially covered, surrounded, substantially surrounded, enclosed, substantially enclosed or comprises a layer of PLGA.
BRIEF DESCRIPTION OF THE DRAWINGS
[0047] The foregoing and other objects, features and advantages will be apparent from the following description of particular embodiments of the invention, as illustrated in the accompanying drawings in which like reference characters refer to the same parts throughout the different views. The drawings are not necessarily to scale, emphasis instead being placed upon illustrating the principles of various embodiments of the invention.
[0048] FIG. 1 illustrates lipid structures in the prior art useful in the present invention. Shown are the structures for 98N12-5 (TETA5-LAP), DLin-DMA, DLin-K-DMA (2,2-Dilinoleyl-4- dimethylaminomethyl-[l,3]-dioxolane), DLin-KC2-DMA, DLin-MC3-DMA and C12-200.
[0049] FIG. 2 is a representative plasmid useful in the IVT reactions taught herein. The plasmid contains Insert 64818, designed by the instant inventors.
[0050] FIG. 3 is a gel profile of modified mR A encapsulated in PLGA microspheres. DETAILED DESCRIPTION
[0051] The delivery of nucleic acids into cells has many undesired complications including the integration of the nucleic acid into the target cell genome which may result in imprecise expression levels, the deleterious transfer of the nucleic acid to progeny and neighbor cells and a substantial risk of causing mutations. The modified nucleic acid molecules of the present disclosure are capable of reducing the innate immune activity of a population of cells into which they are introduced, thus increasing the efficiency of protein production in that cell population. Further, one or more additional advantageous activities and/or properties of the nucleic acids and proteins of the present disclosure are described herein.
[0052] In addition, provided herein are methods of treating a subject having or being suspected of having a disease, disorder and/or condition the methods comprising administering to a subject in need of such treatment a composition described herein in an amount sufficient to treat the disease, disorder and/or condition.
[0053] Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of methods featured in the invention, suitable methods and materials are described below.
Modified Nucleic Acid Molecules
[0054] The present disclosure provides nucleic acids, including RNA such as mRNA, which contain one or more modified nucleosides or nucleotides (termed "modified nucleic acid molecules," "modified mRNA" or "modified mRNA molecules") as described herein. The modification of the nucleic acid molecules of the present invention may have useful properties including, but not limited to, a significant decrease in or a lack of a substantial induction of the innate immune response of a cell into which the modified mRNA is introduced. The modified nucleic acid molecules may also exhibit enhanced efficiency of protein production, intracellular retention of nucleic acids, and viability of contacted cells, as well as having reduced immunogenicity as compared to unmodified nucleic acid molecules.
[0055] Provided are modified nucleic acid molecules containing a translatable region and one, two, or more than two different nucleoside modifications Exemplary nucleic acids for use in this disclosure include ribonucleic acids (RNA), deoxyribonucleic acids (DNAs), threose nucleic acids (TNAs), glycol nucleic acids (GNAs), locked nucleic acids (LNAs) or a hybrid thereof. In preferred embodiments, the modified nucleic acid molecules include messenger RNA (mRNA). As described herein, the modified nucleic acid molecules of the present disclosure may not substantially induce an innate immune response of a cell into which the modified mRNA is introduced. In another embodiment, the modified nucleic acid molecule may exhibit reduced degradation, as compared to a nucleic acid that has not been modified, in a cell where the modified nucleic acid molecule is introduced.
[0056] The term "nucleic acid" includes any compound and/or substance that is or can be incorporated into an oligonucleotide chain. Exemplary nucleic acids for use in accordance with the present disclosure include, but are not limited to, one or more of DNA, cDNA, RNA including messenger RNA (mRNA), hybrids thereof, RNAi-inducing agents, RNAi agents, siRNA, shRNA, miRNA, antisense RNA, ribozymes, catalytic DNA, RNA that induce triple helix formation, aptamers, vectors and the like.
[0057] In certain embodiments, it is desirable to intracellularly degrade a modified nucleic acid molecule introduced into the cell. For example it would be desirable to degrade a modified nucleic acid molecule if precise timing of protein production was desired. Thus, the present disclosure provides a modified nucleic acid molecule containing a degradation domain, which is capable of being acted on in a directed manner within a cell.
[0058] In some embodiments, the modified nucleic acid molecules may be chemically modified on the sugar, nucleobase (e.g., in the 5 ' position of the nucleobase), or phosphate backbone (e.g., replacing the phosphate with another moiety such as a thiophospate). In some embodiments, the modification may result in a disruption of a major groove binding partner interaction, which may contribute to an innate immune response. In some embodiments, the formulation composition, when administered to a subject, can result in improved bioavailability, therapeutic window, or volume of distribution of the modified nucleic acid molecule relative to administration of the modified nucleic acid molecule without the incorporation of the delivery agent. In some embodiments, the modified nucleosides and nucleotides of the modified nucleic acid molecules of the present invention may be synthesized using the O-protected compounds described in International Pub. No. WO2012138530, the contents of which is herein incorporated by reference in its entirety.
[0059] In certain embodiments, the modified nucleic acid molecule may comprise mRNA. In particular embodiments, the modified mRNA (mmRNA) may be derived from cDNA. In certain embodiments, mmRNA may comprise at least two nucleoside modifications. In one embodiment, the nucleoside modifications may be selected from 5-methylcytosine and pseudouridine. In another embodiment, at least one of the nucleoside modifications is not 5-methylcytosine and/or
pseudouridine. In certain embodiments the delivery agent may comprise formulations allowing for localized and systemic delivery of mmRNA. The formulations of the modified nucleic acids molecules and/or mmR A may be selected from, but are not limited to, lipidoids, liposomes and lipid nanoparticles, rapidly eliminated lipid nanoparticles, polymers, lipoplexes, peptides and proteins, at least one chemical modification and conjugation, enhancers, and/or cells.
[0060] In one embodiment, the modified nucleic acid molecules of the present invention may include at least two stop codons before the 3' untranslated region (UTR). The stop codon may be selected from TGA, TAA and TAG. In one embodiment, the nucleic acids of the present invention include the stop codon TGA and one additional stop codon. In a further embodiment the addition stop codon may be TAA. In another embodiment, the modified nucleic acid molecules may comprise three stop codons.
[0061] Other components of a nucleic acid are optional in a modified nucleic acid molecule but these components may be beneficial in some embodiments.
Untranslated Regions (UTRs)
[0062] Untranslated regions (UTRs) of a gene are transcribed but not translated. The 5' UTR starts at the transcription start site and continues to the start codon but does not include the start codon; whereas, the 3' UTR starts immediately following a stop codon and continues until the
transcriptional termination signal. There is growing body of evidence about the regulatory roles played by the UTRs in terms of stability of the nucleic acid molecule and translation. The regulatory features of a UTR can be incorporated into the modified mRNA molecules of the present invention to enhance the stability of the molecule. The specific features can also be incorporated to ensure controlled down-regulation of the transcript in case they are misdirected to undesired organs sites. 5' UTR and Translation Initiation
[0063] Natural 5' UTRs bear features which play roles in for translation initiation. They harbor signatures like Kozak sequences which are commonly known to be involved in the process by which the ribosome initiates translation of many genes. Kozak sequences have the consensus
CCR(A/G)CCAUGG (SEQ ID NO: 1), where R is a purine (adenine or guanine) three bases upstream of the start codon (AUG), which is followed by another 'G'. 5' UTR also have been known to form secondary structures which are involved in elongation factor binding.
[0064] By engineering the features typically found in abundantly expressed genes of specific target organs, one can enhance the stability and protein production of the modified mRNA molecules of the invention. For example, introduction of 5' UTR of liver-expressed mRNA, such as albumin, serum amyloid A, Apolipoprotein A/B/E, transferrin, alpha fetoprotein, erythropoietin, or Factor VIII, could be used to enhance expression of a modified nucleic acid molecule, such as a mmRNA, in hepatic cell lines or liver. Likewise, use of 5' UTR from other tissue-specific mRNA to improve expression in that tissue is possible for muscle (MyoD, Myosin, Myoglobin, Myogenin, Herculin), for endothelial cells (Tie-1, CD36), for myeloid cells (C/EBP, AML1, G-CSF, GM-CSF, CD1 lb, MSR, Fr-1, i-NOS), for leukocytes (CD45, CD18), for adipose tissue (CD36, GLUT4, ACRP30, adiponectin) and for lung epithelial cells (SP-A/B/C/D).
[0065] Other non-UTR sequences may be incorporated into the 5' (or 3' UTR) UTRs of the modified nucleic acid molecules of the present invention. For example, introns or portions of introns sequences may be incorporated into the flanking regions of the modified mRNA of the invention. Incorporation of intronic sequences may increase protein production as well as mRNA levels.
3' UTR and the AU Rich Elements
[0066] 3' UTRs are known to have stretches of Adenosines and Uridines embedded in them. These AU rich signatures are particularly prevalent in genes with high rates of turnover. Based on their sequence features and functional properties, the AU rich elements (AREs) can be separated into three classes (Chen et al, 1995): Class I AREs contain several dispersed copies of an AUUUA motif within U-rich regions. C-Myc and MyoD contain class I AREs. Class II AREs possess two or more overlapping UUAUUUA(U/A)(U/A) (SEQ ID NO: 2) nonamers. Molecules containing this type of AREs include GM-CSF and TNF-a. Class III ARES are less well defined. These U rich regions do not contain an AUUUA motif. c-Jun and Myogenin are two well-studied examples of this class. Most proteins binding to the AREs are known to destabilize the messenger, whereas members of the ELAV family, most notably HuR, have been documented to increase the stability of mRNA. HuR binds to AREs of all the three classes. Engineering the HuR specific binding sites into the 3' UTR of nucleic acid molecules will lead to HuR binding and thus, stabilization of the message in vivo.
[0067] Introduction, removal or modification of 3' UTR AU rich elements (AREs) can be used to modulate the stability of modified mRNA of the invention. When engineering specific modified mRNA, one or more copies of an ARE can be introduced to make modified mRNA of the invention less stable and thereby curtail translation and decrease production of the resultant protein.
[0068] Likewise, AREs can be identified and removed or mutated to increase the intracellular stability and thus increase translation and production of the resultant protein. Transfection experiments can be conducted in relevant cell lines, using modified mRNAof the invention and protein production can be assayed at various time points post-transfection. For example, cells can be transfected with different ARE-engineering molecules and by using an ELISA kit to the relevant protein and assaying protein produced at 6 hours, 12 hours, 24 hours, 48 hours, and 7 days post- transfection.
Incorporating microRNA Binding Sites
[0069] microRNAs (or miRNA) are 19-25 nucleotide long noncoding RNAs that bind to the 3' UTR of nucleic acid molecules and down-regulate gene expression either by reducing nucleic acid molecule stability or by inhibiting translation. The modified mRNA of the invention may comprise one or more microRNA target sequences, microRNA sequences, or microRNA seeds. Such sequences may correspond to any known microRNA such as those taught in US Publication
US2005/0261218 and US Publication US2005/0059005, the contents of which are incorporated herein by reference in their entirety.
[0070] A microRNA sequence comprises a "seed" region, i.e., a sequence in the region of positions 2-8 of the mature microRNA, which sequence has perfect Watson-Crick complementarity to the miRNA target sequence. A microRNA seed may comprise positions 2-8 or 2-7 of the mature microRNA. In some embodiments, a microRNA seed may comprise 7 nucleotides (e.g., nucleotides 2-8 of the mature microRNA), wherein the seed-complementary site in the corresponding miRNA target is flanked by an adenine (A) opposed to microRNA position 1. In some embodiments, a microRNA seed may comprise 6 nucleotides (e.g., nucleotides 2-7 of the mature microRNA), wherein the seed-complementary site in the corresponding miRNA target is flanked byan adenine (A) opposed to microRNA position 1. See for example, Grimson A, Farh K , Johnston WK, Garrett-Engele P, Lim LP, Barrel DP; Mol Cell. 2007 Jul 6;27(1):91-105; each of which is herein incorporated by reference in their entirety. The bases of the microRNA seed have complete complementarity with the target sequence. By engineering microRNA target sequences into the 3 'UTR of modified mRNA of the invention one can target the molecule for degradation or reduced translation, provided the microRNA in question is available. This process will reduce the hazard of off target effects upon nucleic acid molecule delivery. Identification of microRNA, microRNA target regions, and their expression patterns and role in biology have been reported (Bonauer et al, Curr Drug Targets 2010 11 :943-949; Anand and Cheresh Curr Opin Hematol 2011 18 : 171 - 176; Contreras and Rao Leukemia 2012 26:404-413 (2011 Dec 20. doi: 10.1038/leu.2011.356); Barrel Cell 2009 136:215-233; Landgraf et al, Cell, 2007 129:1401-1414; each of which is herein incorporated by reference in its entirety). [0071] For example, if the modified nucleic acid molecule is a modified mRNA and is not intended to be delivered to the liver but ends up there, then miR-122, a microRNA abundant in liver, can inhibit the expression of the gene of interest if one or multiple target sites of miR-122 are engineered into the 3' UTR of the modified mRNA. Introduction of one or multiple binding sites for different microRNA can be engineered to further decrease the longevity, stability, and protein translation of a modified nucleic acid molecule and/or modified mRNA.
[0072] As used herein, the term "microRNA site" refers to a microRNA target site or a microRNA recognition site, or any nucleotide sequence to which a microRNA binds or associates. It should be understood that "binding" may follow traditional Watson-Crick hybridization rules or may reflect any stable association of the microRNA with the target sequence at or adjacent to the microRNA site.
[0073] Conversely, for the purposes of the modified mRNA of the present invention, microRNA binding sites can be engineered out of (i.e. removed from) sequences in which they naturally occur in order to increase protein expression in specific tissues. For example, miR-122 binding sites may be removed to improve protein expression in the liver. Regulation of expression in multiple tissues can be accomplished through introduction or removal or one or several microRNA binding sites.
[0074] Examples of tissues where microRNA are known to regulate mRNA, and thereby protein expression, include, but are not limited to, liver (miR-122), muscle (miR-133, miR-206, miR-208), endothelial cells (miR- 17-92, miR-126), myeloid cells (miR-142-3p, miR-142-5p, miR-16, miR-21, miR-223, miR-24, miR-27), adipose tissue (let-7, miR-30c), heart (miR-ld, miR-149), kidney (miR- 192, miR-194, miR-204), and lung epithelial cells (let-7, miR-133, miR-126). MicroRNA can also regulate complex biological processes such as angiogenesis (miR- 132) (Anand and Cheresh Curr Opin Hematol 2011 18: 171-176; herein incorporated by reference in its entirety). In the modified mRNA of the present invention, binding sites for microRNAs that are involved in such processes may be removed or introduced, in order to tailor the expression of the modified mRNA expression to biologically relevant cell types or to the context of relevant biological processes.
[0075] Lastly, through an understanding of the expression patterns of microRNA in different cell types, modified mRNA can be engineered for more targeted expression in specific cell types or only under specific biological conditions. Through introduction of tissue-specific microRNA binding sites, modified mRNA could be designed that would be optimal for protein expression in a tissue or in the context of a biological condition. [0076] Transfection experiments can be conducted in relevant cell lines, using engineered modified mRNA and protein production can be assayed at various time points post-transfection. For example, cells can be transfected with different micro RNA binding site-engineering modified mRNA and by using an ELISA kit to the relevant protein and assaying protein produced at 6 hour, 12 hour, 24 hour, 48 hour, 72 hour and 7 days post-transfection. In vivo experiments can also be conducted using microRNA-binding site-engineered molecules to examine changes in tissue-specific expression of formulated modified mRNA.
5' Capping
[0077] The 5' cap structure of an mRNA is involved in nuclear export, increasing mRNA stability and binds the mRNA Cap Binding Protein (CBP), which is responsible for mRNA stability in the cell and translation competency through the association of CBP with poly(A) binding protein to form the mature cyclic mRNA species. The cap further assists the removal of 5' proximal introns removal during mRNA splicing.
[0078] Endogenous mRNA molecules may be 5 '-end capped generating a 5 '-ppp-5 '-triphosphate linkage between a terminal guanosine cap residue and the 5 '-terminal transcribed sense nucleotide of the mRNA molecule. This 5'-guanylate cap may then be methylated to generate an N7-methyl- guanylate residue. The ribose sugars of the terminal and/or anteterminal transcribed nucleotides of the 5' end of the mRNA may optionally also be 2'-0-methylated. 5'-decapping through hydrolysis and cleavage of the guanylate cap structure may target a nucleic acid molecule, such as an mRNA molecule, for degradation.
[0079] Modifications to the modified mRNA of the present invention may generate a non- hydrolyzable cap structure preventing decapping and thus increasing mRNA half-life. Because cap structure hydrolysis requires cleavage of 5 '-ppp-5' phosphorodiester linkages, modified nucleotides may be used during the capping reaction. For example, a Vaccinia Capping Enzyme from New England Biolabs (Ipswich, MA) may be used with a-thio-guanosine nucleotides according to the manufacturer's instructions to create a phosphorothioate linkage in the 5 '-ppp-5' cap. Additional modified guanosine nucleotides may be used such as a-methyl-phosphonate and seleno-phosphate nucleotides.
[0080] Additional modifications include, but are not limited to, 2'-0-methylation of the ribose sugars of 5 '-terminal and/or 5 '-anteterminal nucleotides of the mRNA (as mentioned above) on the 2'-hydroxyl group of the sugar ring. Multiple distinct 5 '-cap structures can be used to generate the 5 '-cap of a nucleic acid molecule, such as an m NA molecule.
[0081] Cap analogs, which herein are also referred to as synthetic cap analogs, chemical caps, chemical cap analogs, or structural or functional cap analogs, differ from natural (i.e. endogenous, wild-type or physiological) 5 '-caps in their chemical structure, while retaining cap function. Cap analogs may be chemically (i.e. non-enzymatically) or enzymatically synthesized and/or linked to a nucleic acid molecule.
[0082] For example, the Anti-Reverse Cap Analog (ARCA) cap contains two guanines linked by a 5 '-5 '-triphosphate group, wherein one guanine contains an N7 methyl group as well as a 3'-0-methyl group (i.e., N7,3'-0-dimethyl-guanosine-5'-triphosphate-5'-guanosine (m7G-3'mppp-G; which may equivalently be designated 3' 0-Me-m7G(5')ppp(5')G). The 3'-0 atom of the other, unmodified, guanine becomes linked to the 5'-terminal nucleotide of the capped nucleic acid molecule (e.g. an mRNA or mmRNA). The N7- and 3'-0-methlyated guanine provides the terminal moiety of the capped nucleic acid molecule (e.g. mRNA or mmRNA).
[0083] Another exemplary cap is mCAP, which is similar to ARCA but has a 2'-0-methyl group on guanosine (i.e., N7,2'-0-dimethyl-guanosine-5'-triphosphate-5'-guanosine, m7Gm-ppp-G).
[0084] While cap analogs allow for the concomitant capping of a nucleic acid molecule in an in vitro transcription reaction, up to 20% of transcripts can remain uncapped. This, as well as the structural differences of a cap analog from an endogenous 5 '-cap structures of nucleic acids produced by the endogenous, cellular transcription machinery, may lead to reduced translational competency and reduced cellular stability.
[0085] Modified mRNA of the present invention may also be capped post-transcriptionally, using enzymes, in order to generate more authentic 5 '-cap structures. As used herein, the phrase "more authentic" refers to a feature that closely mirrors or mimics, either structurally or functionally, an endogenous or wild type feature. That is, a "more authentic" feature is better representative of an endogenous, wild-type, natural or physiological cellular function and/or structure as compared to synthetic features or analogs, etc., of the prior art, or which outperforms the corresponding endogenous, wild-type, natural or physiological feature in one or more respects. Non-limiting examples of more authentic 5 'cap structures of the present invention are those which, among other things, have enhanced binding of cap binding proteins, increased half life, reduced susceptibility to 5' endonucleases and/or reduced 5'decapping, as compared to synthetic 5 'cap structures known in the art (or to a wild-type, natural or physiological 5'cap structure). For example, recombinant Vaccinia Virus Capping Enzyme and recombinant 2'-0-methyltransferase enzyme can create a canonical 5'- 5 '-triphosphate linkage between the 5 '-terminal nucleotide of an mRNA and a guanine cap nucleotide wherein the cap guanine contains an N7 methylation and the 5 '-terminal nucleotide of the mRNA contains a 2 '-0 -methyl. Such a structure is termed the Capl structure. This cap results in a higher translational-competency and cellular stability and a reduced activation of cellular proinflammatory cytokines, as compared, e.g., to other 5'cap analog structures known in the art. Cap structures include, but are not limited to, 7mG(5')ppp(5')N,pN2p (cap 0), 7mG(5')ppp(5')NlmpNp (cap 1), and 7mG(5')-ppp(5')NlmpN2mp (cap 2).
[0086] Because the modified mRNA may be capped post-transcriptionally, and because this process is more efficient, nearly 100% of the modified mRNA may be capped. This is in contrast to -80% when a cap analog is linked to an mRNA in the course of an in vitro transcription reaction.
[0087] According to the present invention, 5' terminal caps may include endogenous caps or cap analogs. According to the present invention, a 5' terminal cap may comprise a guanine analog. Useful guanine analogs include, but are not limited to, inosine, Nl-methyl-guanosine, 2'fluoro- guanosine, 7-deaza-guanosine, 8-oxo-guanosine, 2-amino-guanosine, LNA-guanosine, and 2-azido- guanosine.
Viral Sequences
[0088] Additional viral sequences such as, but not limited to, the translation enhancer sequence of the barley yellow dwarf virus (BYDV-PAV) can be engineered and inserted in the 3' UTR of the modified mRNA of the invention and can stimulate the translation of the mRNA in vitro and in vivo. Transfection experiments can be conducted in relevant cell lines at and protein production can be assayed by ELISA at 12 hour, 24 hour, 48 hour, 72 hour and day 7 post-transfection.
IRES Sequences
[0089] Further, provided are modified mRNA which may contain an internal ribosome entry site (IRES). First identified as a feature Picorna virus RNA, IRES plays an important role in initiating protein synthesis in absence of the 5' cap structure. An IRES may act as the sole ribosome binding site, or may serve as one of multiple ribosome binding sites of an mRNA. Modified mRNA containing more than one functional ribosome binding site may encode several peptides or polypeptides that are translated independently by the ribosomes ("multicistronic nucleic acid molecules"). When modified mRNA are provided with an IRES, further optionally provided is a second translatable region. Examples of IRES sequences that can be used according to the invention include without limitation, those from picornaviruses (e.g. FMDV), pest viruses (CFFV), polio viruses (PV), encephalomyocarditis viruses (ECMV), foot-and-mouth disease viruses (FMDV), hepatitis C viruses (HCV), classical swine fever viruses (CSFV), murine leukemia virus (MLV), simian immune deficiency viruses (SIV) or cricket paralysis viruses (CrPV).
Poly-A tails
[0090] During RNA processing, a long chain of adenine nucleotides (poly-A tail) may be added to a modified nucleic acid molecule such as a modified mRNA molecules in order to increase stability. Immediately after transcription, the 3' end of the transcript may be cleaved to free a 3' hydroxyl. Then poly-A polymerase adds a chain of adenine nucleotides to the RNA. The process, called polyadenylation, adds a poly-A tail that can be between, for example, approximately 100 and 250 residues long.
[0091] It has been discovered that unique poly-A tail lengths provide certain advantages to the modified mRNA of the present invention.
[0092] Generally, the length of a poly-A tail of the present invention is greater than 30 nucleotides in length. In another embodiment, the poly-A tail is greater than 35 nucleotides in length (e.g., at least or greater than about 35, 40, 45, 50, 55, 60, 70, 80, 90, 100, 120, 140, 160, 180, 200, 250, 300, 350, 400, 450, 500, 600, 700, 800, 900, 1,000, 1,100, 1,200, 1,300, 1,400, 1,500, 1,600, 1,700, 1,800, 1,900, 2,000, 2,500, and 3,000 nucleotides). In some embodiments, the modified mRNA includes from about 30 to about 3,000 nucleotides (e.g., from 30 to 50, from 30 to 100, from 30 to 250, from 30 to 500, from 30 to 750, from 30 to 1,000, from 30 to 1,500, from 30 to 2,000, from 30 to 2,500, from 50 to 100, from 50 to 250, from 50 to 500, from 50 to 750, from 50 to 1,000, from 50 to 1,500, from 50 to 2,000, from 50 to 2,500, from 50 to 3,000, from 100 to 500, from 100 to 750, from 100 to 1,000, from 100 to 1,500, from 100 to 2,000, from 100 to 2,500, from 100 to 3,000, from 500 to 750, from 500 to 1,000, from 500 to 1,500, from 500 to 2,000, from 500 to 2,500, from 500 to 3,000, from 1,000 to 1,500, from 1,000 to 2,000, from 1,000 to 2,500, from 1,000 to 3,000, from 1,500 to 2,000, from 1,500 to 2,500, from 1,500 to 3,000, from 2,000 to 3,000, from 2,000 to 2,500, and from 2,500 to 3,000).
[0093] In one embodiment, the poly-A tail is designed relative to the length of the overall modified mRNA. This design may be based on the length of the coding region, the length of a particular feature or region (such as the flanking regions), or based on the length of the ultimate product expressed from the modified mRNA.
[0094] In this context the poly-A tail may be 10, 20, 30, 40, 50, 60, 70, 80, 90, or 100% greater in length than the modified mRNA, region or feature thereof. The poly-A tail may also be designed as a fraction of modified mRNA to which it belongs. In this context, the poly-A tail may be 10, 20, 30, 40, 50, 60, 70, 80, or 90% or more of the total length of the molecule or the total length of the molecule minus the poly-A tail. Further, engineered binding sites and conjugation of modified mRNA for Poly-A binding protein may enhance expression.
[0095] Additionally, multiple distinct modified mRNA may be linked together to the PABP (Poly- A binding protein) through the 3 '-end using modified nucleotides at the 3 '-terminus of the poly-A tail. Transfection experiments can be conducted in relevant cell lines at and protein production can be assayed by ELISA at 12 hour, 24 hour, 48 hour, 72 hour and day 7 post-transfection.
[0096] In one embodiment, the modified mRNA of the present invention are designed to include a polyA-G Quartet. The G-quartet is a cyclic hydrogen bonded array of four guanine nucleotides that can be formed by G-rich sequences in both DNA and RNA. In this embodiment, the G-quartet is incorporated at the end of the poly-A tail. The resultant mmRNA molecule is assayed for stability, protein production and other parameters including half- life at various time points. It has been discovered that the polyA-G quartet results in protein production equivalent to at least 75% of that seen using a poly-A tail of 120 nucleotides alone.
Modifications
[0097] The modified nucleic acids and modified mRNA (mmRNA) of the invention may contain one, two, or more different modifications. In some embodiments, modified nucleic acids and mmRNA may contain one, two, or more different nucleoside or nucleotide modifications. In some embodiments, a modified nucleic acid or mmRNA (e.g., having one or more mmRNA molecules) introduced to a cell may exhibit reduced degradation in the cell, as compared to an unmodified nucleic acid or mmRNA.
[0098] The modified nucleic acids and mmRNA can include any useful modification, such as to the sugar, the nucleobase (e.g., one or more modifications of a nucleobase, such as by replacing or substituting an atom of a pyrimidine nucleobase with optionally substituted amino, optionally substituted thiol, optionally substituted alkyl (e.g., methyl or ethyl), or halo (e.g., chloro or fluoro), or the internucleoside linkage (e.g., one or more modification to the phosphodiester backbone). In certain embodiments, modifications are present in both the sugar and the internucleoside linkage (e.g., one or modifications, such as those present in ribonucleic acids (RNA), deoxyribonucleic acids (DNAs), threose nucleic acids (TNAs), glycol nucleic acids (GNAs), peptide nucleic acids (PNAs), locked nucleic acids (LNAs) or hybrids thereof). Additional modifications are described herein.
[0099] As described herein, the modified nucleic acids and mmRNA of the invention do not substantially induce an innate immune response of a cell into which the mRNA is introduced. In certain embodiments, it may desirable to intracellularly degrade a modified nucleic acid molecule or modified nucleic acid molecule introduced into the cell. For example, degradation of a modified nucleic acid molecule or modified mRNA may be preferable if precise timing of protein production is desired. Thus, in some embodiments, the invention provides a modified nucleic acid molecule containing a degradation domain, which is capable of being acted on in a directed manner within a cell. In another aspect, the present disclosure provides nucleic acids comprising a nucleoside or nucleotide that can disrupt the binding of a major groove interacting, e.g. binding, partner with the nucleic acid (e.g., where the modified nucleotide has decreased binding affinity to major groove interacting partner, as compared to an unmodified nucleotide).
[00100] The modified nucleic acid and mmRNA can optionally include other agents (e.g., RNAi- inducing agents, RNAi agents, siRNA, shRNA, miRNA, antisense RNA, ribozymes, catalytic DNA, tRNA, RNA that induce triple helix formation, aptamers, vectors, etc.). In some embodiments, the modified nucleic acids or mmRNA may include one or more messenger RNA (mRNA) and one or more modified nucleoside or nucleotides (e.g., mmRNA molecules). Details for these modified nucleic acids and mmRNA follow.
Modified Nucleic Acids
[00101] The modified nucleic acids or mmRNA of the invention may include a first region of linked nucleosides encoding a polypeptide of interest, a first flanking region located at the 5' terminus of the first region, and a second flanking region located at the 3' terminus of the first region.
[00102] In some embodiments, the modified nucleic acids or mmRNA includes n number of linked nucleosides having Formula (la) or Formula (Ia-1):
Figure imgf000024_0001
(la) or a pharmaceutically acceptable salt or stereoisomer thereof, wherein
[00103] U is O, S, N(Ru)nu, or C(Ru)nu, wherein nu is an integer from 0 to 2 and each Ru is, independently, H, halo, or optionally substituted alkyl;
[00104] — is a single bond or absent;
[00105] each of R1', R2 , R1", R2", R1, R2, R3, R4, and R5 is, if present, independently, H, halo, hydroxy, thiol, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, optionally substituted hydroxyalkoxy, optionally substituted amino, azido, optionally substituted aryl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, optionally substituted aminoalkynyl,or absent; wherein the combination of R3 with one or more of
1 ' 1" 2' 2" 5 1 ' 3 1" 3
R , R , R , R , or R (e.g., the combination of R and R , the combination of R and R , the combination of R 2' and R 3 , the combination of R 2" and R 3 , or the combination of R 5 and R 3 ) can join together to form optionally substituted alkylene or optionally substituted heteroalkylene and, taken together with the carbons to which they are attached, provide an optionally substituted heterocyclyl (e.g., a bicyclic, tricyclic, or tetracyclic heterocyclyl); wherein the combination of R5 with one or
1 ' 1 " 2' 2" 1 ' 5 1" 5 more of R , R , R , or R (e.g., the combination of R and R , the combination of R and R , the combination of R2 and R5, or the combination of R2 and R5) can join together to form optionally substituted alkylene or optionally substituted heteroalkylene and, taken together with the carbons to which they are attached, provide an optionally substituted heterocyclyl (e.g., a bicyclic, tricyclic, or tetracyclic heterocyclyl); and wherein the combination of R4 and one or more of R^ R^ R^ R2 ', R3, or R5 can join together to form optionally substituted alkylene or optionally substituted heteroalkylene and, taken together with the carbons to which they are attached, provide an optionally substituted heterocyclyl (e.g., a bicyclic, tricyclic, or tetracyclic heterocyclyl); [00106] each of m' and m" is, independently, an integer from 0 to 3 (e.g., from 0 to 2, from 0 to 1, from 1 to 3, or from 1 to 2);
[00107] each of Y1, Y2, and Y3, is, independently, O, S, Se, -NRN1-, optionally substituted alkylene, or optionally substituted heteroalkylene, wherein RN1 is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, or absent;
[00108] each Y4 is, independently, H, hydroxy, thiol, boranyl, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted thioalkoxy, optionally substituted alkoxyalkoxy, or optionally substituted amino;
[00109] each Y5 is, independently, O, S, Se, optionally substituted alkylene (e.g., methylene), or optionally substituted heteroalkylene;
[00110] n is an integer from 1 to 100,000; and
[00111] B is a nucleobase (e.g., a purine, a pyrimidine, or derivatives thereof), wherein the combination of B and R1 , the combination of B and R2 , the combination of B and R1 , or the combination of B and R2 can, taken together with the carbons to which they are attached, optionally form a bicyclic group (e.g., a bicyclic heterocyclyl) or wherein the combination of B, R1 , and R3 or the combination of B, R2 , and R3 can optionally form a tricyclic or tetracyclic group (e.g., a tricyclic or tetracyclic heterocyclyl, such as in Formula (IIo)-(IIp) herein). In some embodiments, the modified nucleic acid or mmRNA includes a modified ribose.
[00112] In some embodiments, the modified nucleic acid or mmRNA includes n number of linked nucleosides having Formula (Ia-2)-(Ia-5) or a pharmaceutically acceptable salt or stereoisomer thereof.
Figure imgf000026_0001
[00113] In some embodiments, the modified nucleic acid or mmRNA includes n number of linked nucleosides having Formula (lb) or Formula (Ib-1):
Figure imgf000026_0002
or a pharmaceutically acceptable salt or stereoisomer thereof, wherein
[00114] U is O, S, N(Ru)nu, or C(Ru)nu, wherein nu is an integer from 0 to 2 and each Ru is, independently, H, halo, or optionally substituted alkyl;
[00115] — is a single bond or absent;
[00116] each of R1, R3 , R3 , and R4 is, independently, H, halo, hydroxy, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, optionally substituted hydroxyalkoxy, optionally substituted amino, azido, optionally substituted aryl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, or absent; and wherein the combination of R1 and R3 or the combination of R1 and R3" can be taken together to form optionally substituted alkylene or optionally substituted heteroalkylene (e.g., to produce a locked nucleic acid);
[00117] each R5 is, independently, H, halo, hydroxy, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, or absent;
[00118] each of Y1, Y2, and Y3 is, independently, O, S, Se, -NRN1-, optionally substituted alkylene, or optionally substituted heteroalkylene, wherein RN1 is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, or optionally substituted aryl;
each Y4 is, independently, H, hydroxy, thiol, boranyl, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted alkoxyalkoxy, or optionally substituted amino;
[00119] n is an integer from 1 to 100,000; and
[00120] B is a nucleobase.
[00121] In some embodiments, the modified nucleic acid or mmRNA includes n number of linked nucleosides having Formula (Ic):
Figure imgf000027_0001
or a pharmaceutically acceptable salt or stereoisomer thereof, wherein
[00122] U is O, S, N(R )nu, or C(R )nu, wherein nu is an integer from 0 to 2 and each R is, independently, H, halo, or optionally substituted alkyl;
[00123] — is a single bond or absent;
[00124] each of B1, B2, and B3 is, independently, a nucleobase (e.g., a purine, a pyrimidine, or derivatives thereof, as described herein), H, halo, hydroxy, thiol, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, optionally substituted hydroxyalkoxy, optionally substituted amino, azido, optionally substituted aryl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, or optionally substituted aminoalkynyl, wherein one and only one of B1, B2, and B3 is a nucleobase;
[00125] each of Rbl, Rb2, Rb3, R3, and R5 is, independently, H, halo, hydroxy, thiol, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, optionally substituted hydroxyalkoxy, optionally substituted amino, azido, optionally substituted aryl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl or optionally substituted aminoalkynyl;
[00126] each of Y1, Y2, and Y3, is, independently, O, S, Se, -NRN1-, optionally substituted alkylene, or optionally substituted heteroalkylene, wherein RN1 is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, or optionally substituted aryl;
[00127] each Y4 is, independently, H, hydroxy, thiol, boranyl, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted thioalkoxy, optionally substituted alkoxyalkoxy, or optionally substituted amino;
[00128] each Y5 is, independently, O, S, Se, optionally substituted alkylene (e.g., methylene), or optionally substituted heteroalkylene;
[00129] n is an integer from 1 to 100,000; and
[00130] wherein the ring including U can include one or more double bonds.
[00131] In particular embodiments, the ring including U does not have a double bond between U-
CB3Rb3 or between CB3Rb3-CB2R'
[00132] In some embodiments, the modified nucleic acid or mmRNA includes n number of linked nucleosides having Formula (Id):
Figure imgf000028_0001
(Id), or a pharmaceutically acceptable salt or stereoisomer thereof, wherein
[00133] U is O, S, N(Ru)nu, or C(Ru)nu, wherein nu is an integer from 0 to 2 and each Ru is, independently, H, halo, or optionally substituted alkyl; [00134] each R3 is, independently, H, halo, hydroxy, thiol, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, optionally substituted hydroxyalkoxy, optionally substituted amino, azido, optionally substituted aryl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl,or optionally substituted aminoalkynyl;
[00135] each of Y1, Y2, and Y3, is, independently, O, S, Se, -NRN1-, optionally substituted alkylene, or optionally substituted heteroalkylene, wherein RN1 is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, or optionally substituted aryl;
[00136] each Y4 is, independently, H, hydroxy, thiol, boranyl, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted thioalkoxy, optionally substituted alkoxyalkoxy, or optionally substituted amino;
[00137] each Y5 is, independently, O, S, optionally substituted alkylene (e.g., methylene), or optionally substituted heteroalkylene;
[00138] n is an integer from 1 to 100,000; and
[00139] B is a nucleobase (e.g., a purine, a pyrimidine, or derivatives thereof).
[00140] In some embodiments, the modified nucleic acid molecules or modified mRNA includes n number of linked nucleosides having Formula (Ie):
Figure imgf000029_0001
(Ie), or a pharmaceutically acceptable salt or stereoisomer thereof, wherein
[00141] each of U' and U" is, independently, O, S, N(R )nu, or C(R )nu, wherein nu is an integer from 0 to 2 and each Ru is, independently, H, halo, or optionally substituted alkyl;
each R6 is, independently, H, halo, hydroxy, thiol, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, optionally substituted hydroxyalkoxy, optionally substituted amino, azido, optionally substituted aryl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl,or optionally substituted aminoalkynyl;
[00142] each Y5 is, independently, O, S, optionally substituted alkylene (e.g., methylene or ethylene), or optionally substituted heteroalkylene; [00143] n is an integer from 1 to 100,000; and
[00144] B is a nucleobase (e.g., a purine, a pyrimidine, or derivatives thereof).
[00145] In some embodiments, the modified nucleic acid or mmRNA includes n number of linked nucleosides having Formula (If) or If-1):
Figure imgf000030_0001
(If), or a pharmaceutically acceptable salt or stereoisomer thereof, wherein
[00146] each of U' and U" is, independently, O, S, N, N(Ru)nu, or C(Ru)nu, wherein nu is an integer from 0 to 2 and each Ru is, independently, H, halo, or optionally substituted alkyl (e.g., U' is O and
U" is N);
[00147] — is a single bond or absent;
[00148] each of R1 , R2 , R1 , R2 , R3, and R4 is, independently, H, halo, hydroxy, thiol, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, optionally substituted hydroxyalkoxy, optionally substituted amino, azido, optionally substituted aryl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, optionally substituted aminoalkynyl,or absent; and wherein the combination of R1 and R3, the combination of R1 and R3, the combination of R2 and R3, or the combination of R2 and R3 can be taken together to form optionally substituted alkylene or optionally substituted heteroalkylene (e.g., to produce a locked nucleic acid);each of m' and m" is, independently, an integer from 0 to 3 (e.g., from 0 to 2, from 0 to 1, from 1 to 3, or from 1 to 2);
[00149] each of Y1, Y2, and Y3, is, independently, O, S, Se, -NRN1-, optionally substituted alkylene, or optionally substituted heteroalkylene, wherein RN1 is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, or absent;
[00150] each Y4 is, independently, H, hydroxy, thiol, boranyl, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted thioalkoxy, optionally substituted alkoxyalkoxy, or optionally substituted amino;
[00151] each Y5 is, independently, O, S, Se, optionally substituted alkylene (e.g., methylene), or optionally substituted heteroalkylene;
[00152] n is an integer from 1 to 100,000; and
[00153] B is a nucleobase (e.g., a purine, a pyrimidine, or derivatives thereof).
[00154] In some embodiments of the modified nucleic acid or mmRNA (e.g., (Ia)-(Ia-5), (Ib)-(If-l), (Ila)-(IIp), (IIb-1), (IIb-2), (IIc-l)-(IIc-2), (IIn-1), (IIn-2), (IVa)-(IVl), and (IXa)-(IXr)), the ring including U has one or two double bonds.
[00155] In some embodiments of the modified nucleic acid or mmRNA (e.g., Formulas (Ia)-Ia-5), (Ib)-(If-l), (Ila)-(IIp), (IIb-1), (IIb-2), (IIc-l)-(IIc-2), (IIn-1), (IIn-2), (IVa)-(IVl), and (IXa)-(IXr)), each of R1, R1 , and R1 , if present, is H. In further embodiments, each of R2, R2 , and R2 , if present, is, independently, H, halo (e.g., fluoro), hydroxy, optionally substituted alkoxy (e.g., methoxy or ethoxy), or optionally substituted alkoxyalkoxy. In particular embodiments, alkoxyalkoxy is - (CH2)s2(OCH2CH2)si(CH2)s30R', wherein si is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R' is H or Ci_2o alkyl). In some embodiments, s2 is 0, si is 1 or 2, s3 is 0 or 1, and R' is Ci_6 alkyl.
[00156] In some embodiments of the modified nucleic acid or mmRNA (e.g., Formulas (Ia)-Ia-5), (Ib)-(If-l), (Ila)-(IIp), (IIb-1), (IIb-2), (IIc-l)-(IIc-2), (IIn-1), (IIn-2), (IVa)-(IVl), and (IXa)-(IXr)), each of R2, R2 , and R2 , if present, is H. In further embodiments, each of R1, R1 , and R1 , if present, is, independently, H, halo (e.g., fluoro), hydroxy, optionally substituted alkoxy (e.g., methoxy or ethoxy), or optionally substituted alkoxyalkoxy. In particular embodiments, alkoxyalkoxy is - (CH2)s2(OCH2CH2)si(CH2)s30R', wherein si is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R' is H or Ci_2o alkyl). In some embodiments, s2 is 0, si is 1 or 2, s3 is 0 or 1, and R' is Ci_6 alkyl.
[00157] In some embodiments of the modified nucleic acids or mmRNA (e.g., Formulas (Ia)-Ia-5), (Ib)-(If-l), (Ila)-(IIp), (IIb-1), (IIb-2), (IIc-l)-(IIc-2), (IIn-1), (IIn-2), (IVa)-(IVl), and (IXa)-(IXr)), each of R3, R4, and R5 is, independently, H, halo (e.g., fluoro), hydroxy, optionally substituted alkyl, optionally substituted alkoxy (e.g., methoxy or ethoxy), or optionally substituted alkoxyalkoxy. In particular embodiments, R3 is H, R4 is H, R5 is H, or R3, R4, and R5 are all H. In particular embodiments, R3 is Ci_6 alkyl, R4 is Ci_6 alkyl, R5 is Ci_6 alkyl, or R3, R4, and R5 are all Ci_6 alkyl. In particular embodiments, R3 and R4 are both H, and R5 is Ci_6 alkyl.
[00158] In some embodiments of the modified nucleic acids or mmRNA (e.g., Formulas (Ia)-Ia-5), (Ib)-(If-l), (Ila)-(IIp), (IIb-1), (IIb-2), (IIc-l)-(IIc-2), (IIn-1), (IIn-2), (IVa)-(IVl), and (IXa)-(IXr)), R3 and R5 join together to form optionally substituted alkylene or optionally substituted
heteroalkylene and, taken together with the carbons to which they are attached, provide an optionally substituted heterocyclyl (e.g., a bicyclic, tricyclic, or tetracyclic heterocyclyl, such as trans-3',4' analogs, wherein R3 and R5 join together to form heteroalkylene (e.g., -(CH2)biO(CH2)b20(CFl2)b3-, wherein each of bl, b2, and b3 are, independently, an integer from 0 to 3).
[00159] In some embodiments of the modified nucleic acids or mmRNA (e.g., Formulas (Ia)-Ia-5), (Ib)-(If-l), (Ila)-(IIp), (IIb-1), (IIb-2), (IIc-l)-(IIc-2), (IIn-1), (IIn-2), (IVa)-(IVl), and (IXa)-(IXr)), R3 and one or more of R1 , R1 , R2 , R2 , or R5 join together to form optionally substituted alkylene or optionally substituted heteroalkylene and, taken together with the carbons to which they are attached, provide an optionally substituted heterocyclyl (e.g., a bicyclic, tricyclic, or tetracyclic heterocyclyl, R3 and one or more of R1 , R1 , R2 , R2 , or R5 join together to form heteroalkylene (e.g., -(CH2)biO(CH2)b20(CFl2)b3-, wherein each of bl, b2, and b3 are, independently, an integer from 0 to 3).
[00160] In some embodiments of the modified nucleic acids or mmRNA (e.g., Formulas (Ia)-Ia-5), (Ib)-(If-l), (Ila)-(IIp), (IIb-1), (IIb-2), (IIc-l)-(IIc-2), (IIn-1), (IIn-2), (IVa)-(IVl), and (IXa)-(IXr)), R5 and one or more of R1 , R1 , R2 , or R2 join together to form optionally substituted alkylene or optionally substituted heteroalkylene and, taken together with the carbons to which they are attached, provide an optionally substituted heterocyclyl (e.g., a bicyclic, tricyclic, or tetracyclic heterocyclyl, R5 and one or more of R1 , R1 , R2 , or R2 join together to form heteroalkylene (e.g., - (CH2)biO(CH2)b20(CFl2)b3-, wherein each of bl, b2, and b3 are, independently, an integer from 0 to 3).
[00161] In some embodiments of the modified nucleic acids or mmRNA (e.g., Formulas (Ia)-Ia-5), (Ib)-(If-l), (Ila)-(IIp), (IIb-1), (IIb-2), (IIc-l)-(IIc-2), (IIn-1), (IIn-2), (IVa)-(IVl), and (IXa)-(IXr)), each Y2 is, independently, O, S, or -NRN1-, wherein RN1 is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, or optionally substituted aryl. In particular embodiments, Y2 is NRN1-, wherein RN1 is H or optionally substituted alkyl (e.g., Ci_6 alkyl, such as methyl, ethyl, isopropyl, or n-propyl).
[00162] In some embodiments of the modified nucleic acids or mmRNA (e.g., Formulas (Ia)-Ia-5), (Ib)-(If-l), (Ila)-(IIp), (IIb-1), (IIb-2), (IIc-l)-(IIc-2), (IIn-1), (IIn-2), (IVa)-(IVl), and (IXa)-(IXr)), each Y3 is, independently, O or S.
[00163] In some embodiments of the modified nucleic acids or mmRNA (e.g., Formulas (Ia)-Ia-5), (Ib)-(If-l), (Ila)-(IIp), (IIb-1), (IIb-2), (IIc-l)-(IIc-2), (IIn-1), (IIn-2), (IVa)-(IVl), and (IXa)-(IXr)), R1 is H; each R2 is, independently, H, halo (e.g., fluoro), hydroxy, optionally substituted alkoxy (e.g., methoxy or ethoxy), or optionally substituted alkoxyalkoxy (e.g., -
(CH2)S2(OCH2CH2)si(CH2)S30R', wherein si is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R' is H or Ci_2o alkyl, such as wherein s2 is 0, si is 1 or 2, s3 is 0 or 1, and R' is Ci_6 alkyl); each Y2 is, independently, O or -NRN1-, wherein RN1 is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, or optionally substituted aryl (e.g., wherein RN1 is H or optionally substituted alkyl (e.g., Ci_6 alkyl, such as methyl, ethyl, isopropyl, or n-propyl)); and each Y3 is, independently, O or S (e.g., S). In further embodiments, R3 is H, halo (e.g., fluoro), hydroxy, optionally substituted alkyl, optionally substituted alkoxy (e.g., methoxy or ethoxy), or optionally substituted alkoxyalkoxy. In yet further embodiments, each Y1 is , independently, O or -NRN1-, wherein RN1 is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, or optionally substituted aryl (e.g., wherein RN1 is H or optionally substituted alkyl (e.g., Ci_6 alkyl, such as methyl, ethyl, isopropyl, or n-propyl)); and each Y4 is, independently, H, hydroxy, thiol, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted thioalkoxy, optionally substituted alkoxyalkoxy, or optionally substituted amino.
[00164] In some embodiments of the modified nucleic acids or mmRNA (e.g., Formulas (Ia)-(Ia-5), (Ib)-(If-l), (Ila)-(IIp), (IIb-1), (IIb-2), (IIc-l)-(IIc-2), (IIn-1), (IIn-2), (IVa)-(IVl), and (IXa)-(IXr)), each R1 is, independently, H, halo (e.g., fluoro), hydroxy, optionally substituted alkoxy (e.g., methoxy or ethoxy), or optionally substituted alkoxyalkoxy (e.g., -(CH2)s2(OCH2CH2)si(CH2)s30R', wherein si is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3,
independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R' is H or Ci_2o alkyl, such as wherein s2 is 0, si is 1 or 2, s3 is 0 or 1, and R' is Ci_6 alkyl); R2 is H; each Y2 is, independently, O or -NRN1-, wherein RN1 is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, or optionally substituted aryl
(e.g., wherein R > N1 i ·s H or optionally substituted alkyl (e.g., Ci_6 alkyl, such as methyl, ethyl, isopropyl, or n-propyl)); and each Y3 is, independently, O or S (e.g., S). In further embodiments, R3 is H, halo (e.g., fluoro), hydroxy, optionally substituted alkyl, optionally substituted alkoxy (e.g., methoxy or ethoxy), or optionally substituted alkoxyalkoxy. In yet further embodiments, each Y1 is , independently, O or -NRN1-, wherein RN1 is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, or optionally substituted aryl (e.g., wherein RN1 is H or optionally substituted alkyl (e.g., Ci_6 alkyl, such as methyl, ethyl, isopropyl, or n-propyl)); and each Y4 is, independently, H, hydroxy, thiol, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted thioalkoxy, optionally substituted alkoxyalkoxy, or optionally substituted amino.
[00165] In some embodiments of the modified nucleic acids or mmRNA (e.g., Formulas (Ia)-(Ia-5), (Ib)-(If-l), (Ila)-(IIp), (IIb-1), (IIb-2), (IIc-l)-(IIc-2), (IIn-1), (IIn-2), (IVa)-(IVl), and (IXa)-(IXr)), the ring including U is in the β-D (e.g., β-D-ribo) configuration.
[00166] In some embodiments of the modified nucleic acids or mmRNA (e.g Formulas (Ia)-(Ia-5), (Ib)-(If-l), (Ila)-(IIp), (IIb-1), (IIb-2), (IIc-l)-(IIc-2), (IIn-1), (IIn-2), (IVa)-(IVl), and (IXa)-(IXr)), the ring including U is in the a-L (e.g., a-L-ribo) configuration.
[00167] In some embodiments of the modified nucleic acids or mmRNA (e.g., Formulas (Ia)-(Ia-5), (Ib)-(If-l), (Ila)-(IIp), (IIb-1), (IIb-2), (IIc-l)-(IIc-2), (IIn-1), (IIn-2), (IVa)-(IVl), and (IXa)-(IXr)), one or more B is not pseudouridine (ψ) or 5-methyl-cytidine (m5C). In some embodiments, about 10% to about 100% of B nucleobases is not ψ or m5C (e.g., from 10% to 20%, from 10% to 35%, from 10% to 50%, from 10% to 60%, from 10% to 75%, from 10% to 90%, from 10% to 95%, from 10% to 98%, from 10% to 99%, from 20% to 35%, from 20% to 50%, from 20% to 60%, from 20% to 75%, from 20% to 90%, from 20% to 95%, from 20% to 98%, from 20% to 99%, from 20% to 100%, from 50% to 60%, from 50% to 75%, from 50% to 90%, from 50% to 95%, from 50% to 98%, from 50% to 99%, from 50% to 100%, from 75% to 90%, from 75% to 95%, from 75% to 98%, from 75% to 99%, and from 75% to 100% of n number of B is not ψ or m5C). In some embodiments, B is not ψ or m5C.
[00168] In some embodiments of the modified nucleic acids or mmRNA (e.g., Formulas (Ia)-(Ia-5), (Ib)-(If-l), (Ila)-(IIp), (IIb-1), (IIb-2), (IIc-l)-(IIc-2), (IIn-1), (IIn-2), (IVa)-(IVl), and (IXa)-(IXr)), when B is an unmodified nucleobase selected from cytosine, guanine, uracil and adenine, then at least one of Y1, Y2, or Y3 is not O.
[00169] In some embodiments, the modified nucleic acids or mmRNA includes a modified ribose. In some embodiments, modified nucleic acids or mmRNA includes n number of linked nucleosides having Formula (Ila)-(IIc):
Figure imgf000035_0001
a pharmaceutically acceptable salt or stereoisomer thereof. In particular embodiments, U is O or C(Ru)nu, wherein nu is an integer from 0 to 2 and each Ru is, independently, H, halo, or optionally substituted alkyl (e.g., U is -CH2- or -CH-). In other embodiments, each of R1, R2, R3, R4, and R5 is, independently, H, halo, hydroxy, thiol, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, optionally substituted hydroxyalkoxy, optionally substituted amino, azido, optionally substituted aryl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, or absent (e.g., each R1 and R2 is, independently, H, halo, hydroxy, optionally substituted alkyl, or optionally substituted alkoxy; each
R3 and R4 is, independently, H or optionally substituted alkyl; and R5 is H or hydroxy), and is a single bond or double bond.
[00170] In particular embodiments, the modified nucleic acid or mmRNA includes n number of linked nucleosides having Formula lib- 1 )-(IIb-2) :
Figure imgf000035_0002
or a pharmaceutically acceptable salt or stereoisomer thereof. In some embodiments, U is O or C(R )nu, wherein nu is an integer from 0 to 2 and each R is, independently, H, halo, or optionally substituted alkyl (e.g., U is -CH2- or - CH-). In other embodiments, each of R1 and R2 is, independently, H, halo, hydroxy, thiol, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, optionally substituted hydroxyalkoxy, optionally substituted amino, azido, optionally substituted aryl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, or absent (e.g., each R1 and R2 is, independently, H, halo, hydroxy, optionally substituted alkyl, or optionally substituted alkoxy, e.g., H, halo, hydroxy, alkyl, or alkoxy). In particular embodiments, R2 is hydroxy or optionally substituted alkoxy (e.g., methoxy, ethoxy, or any described herein).
[00171] In particular embodiments, the modified nucleic acid or mmRNA includes n number of linked
(IIc-3)
Figure imgf000036_0001
thereof. In some embodiments, U is O or C(Ru)nu, wherein nu is an integer from 0 to 2 and each R1 is, independently, H, halo, or optionally substituted alkyl (e.g., U is -CH2- or -CH-). In some embodiments, each of R1, R2, and R3 is, independently, H, halo, hydroxy, thiol, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, optionally substituted hydroxyalkoxy, optionally substituted amino, azido, optionally substituted aryl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, or absent (e.g., each R1 and R2 is, independently, H, halo, hydroxy, optionally substituted alkyl, or optionally substituted alkoxy, e.g., H, halo, hydroxy, alkyl, or alkoxy; and each R3 is, independently, H or optionally substituted alkyl)). In particular embodiments, R2 is optionally substituted alkoxy (e.g., methoxy or ethoxy, or any described herein). In particular embodiments, R1 is optionally substituted alkyl, and R2 is hydroxy. In other embodiments, R1 is hydroxy, and R2 is optionally substituted alkyl. In further embodiments, R3 is optionally substituted alkyl.
[00172] In some embodiments, the modified nucleic acids or mmRNA includes an acyclic modified ribose. In some embodiments, the modified nucleic acids or mmRNA includes n number of linked nucleosides having Formula (Ild)-(IIf):
Figure imgf000037_0001
(Ilf), or a pharmaceutically acceptable salt or stereoisomer thereof.
[00173] In some embodiments, the modified nucleic acids or mmRNA includes an acyclic modified hexitol. In some embodiments, the modified nucleic acids or mmRNA includes n number of linked nucleosides having Formula (Ilg)-(IIj):
Figure imgf000038_0001
(Hi), or (Ilj), or a pharmaceutically acceptable salt or stereoisomer thereof.
[00174] In some embodiments, the modified nucleic acids or mmRNA includes a sugar moiety having a contracted or an expanded ribose ring. In some embodiments, the modified nucleic acids or mmRNA includes n number of linked nucleosides havin Formula Ilk - IIm):
Figure imgf000038_0002
(Urn), or a pharmaceutically acceptable salt or stereoisomer thereof, wherein each of R1 , R1 , R2 , and R2 is, independently, H, halo, hydroxy, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, or absent; and wherein the combination of R2 and R3 or the combination of R2 and R3 can be taken together to form optionally substituted alkylene or optionally substituted heteroalkylene.
[00175] In some embodiments, the modified nucleic acids or mmRNA includes a locked modified ribose. In some embodiments, the modified nucleic acids or mmRNA includes n number of linked nucleosides havin Formula (Iln):
Figure imgf000039_0001
(Iln), or a pharmaceutically acceptable salt or stereoisomer thereof, wherein R3 is O, S, or -NRN1-, wherein RN1 is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, or optionally substituted aryl and R3 is optionally substituted alkylene (e.g., -CH2-, -CH2CH2-, or -CH2CH2CH2-) or optionally substituted heteroalkylene (e.g., - CH2NH-, -CH2CH2NH-, -CH2OCH2-, or -CH2CH2OCH2-)(e.g., R3' is O and R3" is optionally substituted alkylene (e.g., -CH2-, -CH2CH2-, or -CH2CH2CH2-))..
[00176] In some embodiments, the modified nucleic acid or mmRNA includes n number of linked nucleosides havin Formula (IIn-l)- II-n2):
Figure imgf000039_0002
(Iln- 1) or (IIn-2), or a pharmaceutically acceptable salt or stereoisomer thereof, wherein R3 is O, S, or -NRN1-, wherein RN1 is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, or optionally substituted aryl and R3 is optionally substituted alkylene (e.g., -CH2-, -CH2CH2-, or -CH2CH2CH2-) or optionally substituted heteroalkylene (e.g., -CH2NH-, -CH2CH2NH-, -CH2OCH2-, or -CH2CH2OCH2-) (e.g., R3' is O and R3 is optionally substituted alkylene (e.g., -CH2-, -CH2CH2-, or -CH2CH2CH2-)). [00177] In some embodiments, the modified nucleic acids or mmRNA includes a locked modified ribose that forms a tetracyclic heterocyclyl. In some embodiments, the modified nucleic acids or mmRNA includes n number of linked nucleosides havin Formula Ho :
Figure imgf000040_0001
(Hp), or a
pharmaceutically acceptable salt or stereoisomer thereof, wherein R12a, R12c, T1 , T1 ,T2 , T2 , V1, and V3 are as described herein.
[00178] Any of the formulas for the modified nucleic acids or mmRNA can include one or more nucleobases described herein (e.g., Formulas (bl)-(b43)).
[00179] In one embodiment, the present invention provides methods of preparing a modified nucleic acids or mmRNA comprising at least one nucleotide (e.g., mmRNA molecule), wherein the modified nucleic acid comprises n number of nucleosides having Formula (la), as defined herein:
Figure imgf000040_0002
a), the method comprising reacting a compound of Formula (Ilia), as defined herein:
Figure imgf000041_0001
with an RNA polymerase, and a cDNA template.
[00180] In a further embodiment, the present invention provides methods of amplifying a modified nucleic acids or mmRNA comprising at least one nucleotide (e.g., mmRNA molecule), the method comprising: reacting a compound of Formula (Ilia), as defined herein, with a primer, a cDNA template, and an RNA polymerase.
[00181] In one embodiment, the present invention provides methods of preparing a modified nucleic acids or mmRNA comprising at least one nucleotide (e.g., mmRNA molecule), wherein the modified nucleic acid comprises n number of nucleosides having Formula (Ia-1), as defined herein:
Figure imgf000041_0002
the method comprising reacting a compound of Formula (IIIa-1), as defined herein:
Figure imgf000042_0001
(IIIa-1), with an RNA polymerase, and a cDNA template.
[00182] In a further embodiment, the present invention provides methods of amplifying a modified nucleic acids or mmRNA comprising at least one nucleotide (e.g., mmRNA molecule), the method comprising reacting a compound of Formula (IIIa-1), as defined herein, with a primer, a cDNA template, and an RNA polymerase.
[00183] In one embodiment, the present invention provides methods of preparing a modified mRNA comprising at least one nucleotide (e.g., mmRNA molecule), wherein the polynucleotide comprises n number of nucleosides having Formula (Ia-2), as defined herein:
Figure imgf000042_0002
(Ia-2),
the method comprising reacting a compound of Formula (IIIa-2), as defined herein:
Figure imgf000042_0003
(IIIa-2), with an RNA polymerase, and a cDNA template. [00184] In a further embodiment, the present invention provides methods of amplifying a modified mR A comprising at least one nucleotide (e.g., mmR A molecule), the method comprising:
[00185] reacting a compound of Formula (IIIa-2), as defined herein, with a primer, a cDNA template, and an RNA polymerase.
[00186] In some embodiments, the reaction may be repeated from 1 to about 7,000 times. In any of the embodiments herein, B may be a nucleobase of Formula (bl)-(b43).
[00187] The modified nucleic acids and mmRNA can optionally include 5' and/or 3' flanking regions, which are described herein.
Modified RNA (e.g. mmRNA) Molecules
[00188] The present invention also includes building blocks, e.g., modified ribonucleosides, modified ribonucleotides, of modified RNA (mmRNA) molecules. For example, these mmRNA can be useful for preparing the modified nucleic acids or mmRNA of the invention.
[00189] In some embodiments, the buildin block molecule has Formula Ilia or (Ilia- 1 ) :
Figure imgf000043_0001
or a
pharmaceutically acceptable salt or stereoisomer thereof, wherein the substituents are as described herein (e.g., for Formula (la) and (Ia-1)), and wherein when B is an unmodified nucleobase selected from cytosine, guanine, uracil and adenine, then at least one of Y1, Y2, or Y3 is not O.
[00190] In some embodiments, the building block molecule, which may be incorporated into a modified nucleic acid or mmRNA has Formula IVa -(IVb):
Figure imgf000043_0002
(IV a) or (IVb), or a pharmaceutically acceptable salt or stereoisomer thereof , wherein B is as described herein (e.g., any one of (bl)-(b43)). In particular embodiments, Formula (IVa) or (IVb) is combined with a modified uracil (e.g., any one of formulas (bl)-(b9), (b21)-(b23), and (b28)-(b31), such as formula (bl), (b8), (b28), (b29), or (b30)). In particular embodiments, Formula (IVa) or (IVb) is combined with a modified cytosine (e.g., any one of formulas (M0)-(bl4), (b24), (b25), and (b32)-(b36), such as formula (blO) or (b32)). In particular embodiments, Formula (IVa) or (IVb) is combined with a modified guanine (e.g., any one of formulas (bl5)-(bl7) and (b37)-(b40)). In particular embodiments, Formula (IVa) or (IVb) is combined with a modified adenine (e.g., any one of formulas (M8)-(b20) and (b41)-(b43)).
[00191] In some embodiments, the building block molecule , which may be incorporated into a modified nucleic acid molecule or mmRNA, has Formula (IVc)-(IVk):
Figure imgf000044_0001
Figure imgf000045_0001
HO I (IVl), or a pharmaceutically acceptable salt or stereoisomer thereof, wherein B is as described herein (e.g., any one of (bl)-(b43)). In particular embodiments, one of Formulas (IVc)-(IVk) is combined with a modified uracil (e.g., any one of formulas (bl)-(b9), (b21)- (b23), and (b28)-(b31), such as formula (bl), (b8), (b28), (b29), or (b30)). In particular
embodiments, one of Formulas (IVc)-(IVk) is combined with a modified cytosine (e.g., any one of formulas (M0)-(bl4), (b24), (b25), and (b32)-(b36), such as formula (blO) or (b32)). In particular embodiments, one of Formulas (IVc)-(IVk) is combined with a modified guanine (e.g., any one of formulas (bl5)-(bl7) and (b37)-(b40)). In particular embodiments, one of Formulas (IVc)-(IVk) is combined with a modified adenine (e.g., any one of formulas (M8)-(b20) and (b41)-(b43)).
[00192] In other embodiments, the building block molecule, which may be incorporated into a modified nucleic acid molecule or mmR A, has Formula (Va) or (Vb):
Figure imgf000045_0002
(Vb), or a
pharmaceutically acceptable salt or stereoisomer thereof, wherein B is as described herein (e.g., any one of (bl)-(b43)).
[00193] In other embodiments, the building block molecule, which may be incorporated into a modified nucleic acid molecule or mmRNA, has Formula (IXa)-(IXd):
Figure imgf000046_0001
or
Figure imgf000046_0002
(IXd), or a pharmaceutically acceptable salt or stereoisomer thereof, wherein B is as described herein (e.g., any one of (bl)-(b43)). In particular embodiments, one of Formulas (IXa)-(IXd) is combined with a modified uracil (e.g., any one of formulas (bl)-(b9), (b21)- (b23), and (b28)-(b31), such as formula (bl), (b8), (b28), (b29), or (b30)). In particular
embodiments, one of Formulas (IXa)-(IXd) is combined with a modified cytosine (e.g., any one of formulas (M0)-(bl4), (b24), (b25), and (b32)-(b36), such as formula (blO) or (b32)). In particular embodiments, one of Formulas (IXa)-(IXd) is combined with a modified guanine (e.g., any one of formulas (bl5)-(bl7) and (b37)-(b40)). In particular embodiments, one of Formulas (IXa)-(IXd) is combined with a modified adenine (e.g., any one of formulas (M8)-(b20) and (b41)-(b43)).
[00194] In other embodiments, the building block molecule, which may be incorporated into a modified nucleic acid molecule or mmRNA, has Formula (IXe)-(IXg):
Figure imgf000046_0003
pharmaceutically acceptable salt or stereoisomer thereof, wherein B is as described herein (e.g., any one of (bl)-(b43)). In particular embodiments, one of Formulas (IXe)-(IXg) is combined with a modified uracil (e.g., any one of formulas (bl)-(b9), (b21)-(b23), and (b28)-(b31), such as formula (bl), (b8), (b28), (b29), or (b30)). In particular embodiments, one of Formulas (IXe)-(IXg) is combined with a modified cytosine (e.g., any one of formulas (M0)-(bl4), (b24), (b25), and (b32)- (b36), such as formula (blO) or (b32)). In particular embodiments, one of Formulas (IXe)-(IXg) is combined with a modified guanine (e.g., any one of formulas (bl5)-(bl7) and (b37)-(b40)). In particular embodiments, one of Formulas (IXe)-(IXg) is combined with a modified adenine (e any one of formulas (bl8)-(b20) and (b41)-(b43)).
[00195] In other embodiments, the building block molecule, which may be incorporated into
Figure imgf000047_0001
wherein B is as described herein (e.g., any one of (bl)-(b43)). In particular embodiments, one of Formulas (IXh)-(IXk) is combined with a modified uracil (e.g., any one of formulas (bl)-(b9), (b21)- (b23), and (b28)-(b31), such as formula (bl), (b8), (b28), (b29), or (b30)). In particular
embodiments, one of Formulas (IXh)-(IXk) is combined with a modified cytosine (e.g., any one of formulas (M0)-(bl4), (b24), (b25), and (b32)-(b36), such as formula (blO) or (b32)). In particular embodiments, one of Formulas (IXh)-(IXk) is combined with a modified guanine (e.g., any one of formulas (bl5)-(bl7) and (b37)-(b40)). In particular embodiments, one of Formulas (IXh)-(IXk) is combined with a modified adenine (e.g., any one of formulas (M8)-(b20) and (b41)-(b43)).
[00196] In other embodiments, the building block molecule, which may be incorporated into a modified nucleic acid molecule or mmRNA, has Formula (IXl)-(IXr):
Figure imgf000048_0001
HO Br(IXq), or H3(jxr) or a pharmaceutically acceptable salt or stereoisomer thereof, wherein each rl and r2 is, independently, an integer from 0 to 5 (e.g., from 0 to 3, from 1 to 3, or from 1 to 5) and B is as described herein (e.g., any one of (bl)-(b43)). In particular embodiments, one of Formulas (IXl)-(IXr) is combined with a modified uracil (e.g., any one of formulas (bl)-(b9), (b21)-(b23), and (b28)-(b31), such as formula (bl), (b8), (b28), (b29), or (b30)). In particular embodiments, one of Formulas (IXl)-(IXr) is combined with a modified cytosine (e.g., any one of formulas (M0)-(bl4), (b24), (b25), and (b32)-(b36), such as formula (blO) or (b32)). In particular embodiments, one of Formulas (IXl)-(IXr) is combined with a modified guanine (e.g., any one of formulas (bl5)-(bl7) and (b37)-(b40)). In particular embodiments, one of Formulas (IXl)-(IXr) is combined with a modified adenine (e.g., any one of formulas (M8)-(b20) and (b41)-(b43)).
[00197] In some embodiments, the building block molecule, which may be incorporated into a modified nucleic acid molecules or mmR A, can be selected from the group consisting of:
Figure imgf000049_0001
B- 10),
Figure imgf000050_0001
BB- 12), or a pharmaceutically acceptable salt or stereoisomer thereof, wherein each r is, independently, an integer from 0 to 5 (e.g., from 0 to 3, from 1 to 3, or from 1 to 5).
[00198] In some embodiments, the building block molecule, which may be incorporated into a modified nucleic acid molecule or mmRNA, can be selected from the group consisting of:
Figure imgf000050_0002
Figure imgf000051_0001
HO OH (BB- 20), or a pharmaceutically acceptable salt or stereoisomer thereof, wherein each r is, independently, an integer from 0 to 5 (e.g., from 0 to 3, from 1 to 3, or from 1 to 5) and si is as described herein.
[00199] In some embodiments, the building block molecule, which may be incorporated into a nucleic acid (e.g., RNA, mRNA, or mmRNA), is a modified uridine (e.g., selected from the group consisting of:
(BB- 22),
Figure imgf000051_0002
Figure imgf000052_0001
B- 33),
Figure imgf000053_0001
Figure imgf000054_0001
Figure imgf000055_0001
Figure imgf000056_0001
HO OH (BB- 65),
Figure imgf000057_0001
H (BB- 68),
Figure imgf000057_0002
(BB- 69),
Figure imgf000058_0001
HO OCH3 (BB- 70), HO OH (BB-71),
Figure imgf000058_0002
- 72), - 73),
Figure imgf000058_0003
77),
Figure imgf000059_0001
HO OH (BB- 78), (BB- 79),
Figure imgf000059_0002
HO OH (BB- 84), HO OH (BB- 85),
Figure imgf000060_0001
Figure imgf000061_0001
Figure imgf000061_0002
OH (BB- 96), HO OH (BB- 97),
Figure imgf000061_0003
OH (BB- 98), HO OH (BB- 99),
Figure imgf000061_0004
H (BB-100), HO OH (BB-101)
Figure imgf000062_0001
H (BB- 105),
Figure imgf000063_0001
11),
Figure imgf000064_0001
Figure imgf000065_0001
HO O (BB- 125), or a pharmaceutically acceptable salt or stereoisomer thereof, wherein Y1, Y3, Y4, Y6, and r are as described herein (e.g., each r is, independently, an integer from 0 to 5, such as from 0 to 3, from 1 to 3, or from 1 to 5)). [00200] In some embodiments, the building block molecule, which may be incorporated into a modified nucleic acid molecule or mmRNA, is a modified cytidine (e.g., selected from the group consisting of:
(BB- 127),
BB- 129),
Figure imgf000066_0001
BB-
B- 133),
Figure imgf000067_0001
HO OCH 3 (BB- 134), HO OH (BB- 135),
BB-137),
Figure imgf000067_0002
HO OH (BB- 138), HO OH (BB- 139),
Figure imgf000068_0001
HO Br (BB- 146), HO OH (BB- 147),
Figure imgf000069_0001
HO CH3 (BB- 148), HO OH (BB- 149),
Figure imgf000069_0002
HO OCH3 (BB- 150), HO OH (BB- 151),
(BB- 153),
Figure imgf000069_0003
(BB- 155),
Figure imgf000070_0001
eutically acceptable salt or stereoisomer thereof, wherein Y1, Y Y4, Y6, and r are as described herein (e.g., each r is, independently, an integer from 0 to 5, such as from 0 to 3, from 1 to 3, or from 1 to 5)). For example, the building block molecule, which may be incorporated into a modified nucleic acid
molecule or mmR A, can be:
Figure imgf000070_0002
(BB- 161), or a pharmaceutically acceptable salt or stereoisomer thereof, wherein each r independently, an integer from 0 to 5 (e.g., from 0 to 3, from 1 to 3, or from 1 to 5). [00201] In some embodiments, the building block molecule, which may be incorporated into a modified nucleic acid molecule or mmRNA, is a modified adenosine (e.g., selected from the group consisting of:
Figure imgf000071_0001
HO Br (BB- 168), HO I (BB- 169),
Figure imgf000072_0001
Figure imgf000073_0001
(BB- 185),
Figure imgf000074_0001
HO OH (BB- 186), HO OH (BB- 187),
Figure imgf000074_0002
HO OH (BB- 188), (BB- 189),
Figure imgf000074_0003
HO OH (BB- 190), HO OH (BB- 191),
Figure imgf000075_0001
HO OH (BB- 192), HO OH (BB- 193),
Figure imgf000075_0002
and
Figure imgf000076_0001
(BB- 200) or a pharmaceutically acceptable salt or stereoisomer thereof, wherein Y1, Y3, Y4, Y6, and r are as described herein (e.g., each r is, independently, an integer from 0 to 5, such as from 0 to 3, from 1 to 3, or from 1 to 5)).
[00202] In some embodiments, the building block molecule, which may be incorporated into a modified nucleic acid molecule or mmRNA, is a modified guanosine (e.g., selected from the group consisting of:
Figure imgf000076_0002
,
Figure imgf000077_0001
,
Figure imgf000078_0001
,
Figure imgf000079_0001
, B- 236), and
Figure imgf000080_0001
(BB- 237), or a pharmaceutically acceptable salt or stereoisomer thereof, wherein Y1, Y3, Y4, Y6, and r are as described herein (e.g., each r is, independently, an integer from 0 to 5, such as from 0 to 3, from 1 to 3, or from 1 to 5)).
[00203] In some embodiments, the chemical modification can include replacement of C group at C- 5 of the ring (e.g., for a pyrimidine nucleoside, such as cytosine or uracil) with N (e.g., replacement of the >CH group at C-5 with >NRN1 group, wherein RN1 is H or optionally substituted alkyl). For example, the mmRNA molecule, which may be incorporated into a modified nucleic acid molecule or mmRNA, can be:
Figure imgf000080_0002
a pharmaceutically acceptable salt or stereoisomer thereof, wherein each r is, independently, an integer from 0 to 5 (e.g., from 0 to 3, from 1 to 3, or from 1 to 5).
[00204] In another embodiment, the chemical modification can include replacement of the hydrogen at C-5 of cytosine with halo (e.g., Br, CI, F, or I) or optionally substituted alkyl (e.g., methyl). For example, the mmRNA molecule, which may be incorporated into a modified nucleic acid or mmRNA, can be:
Figure imgf000081_0001
pharmaceutically acceptable salt or stereoisomer thereof, wherein each r is, independently, an integer from 0 to 5 (e.g., from 0 to 3, from 1 to 3, or from 1 to 5).
[00205] In yet a further embodiment, the chemical modification can include a fused ring that is formed by the NH2 at the C-4 position and the carbon atom at the C-5 position. For example, the building block molecule, which may be incorporated into a modified nucleic acid molecule or mmRNA, can be:
Figure imgf000082_0001
(BB- 246), or a pharmaceutically acceptable salt or stereoisomer thereof, wherein each r is, independently, an integer from 0 to 5 (e.g., from 0 to 3, from 1 to 3, or from 1 to 5).
Modifications on the Sugar
[00206] The modified nucleosides and nucleotides (e.g., building block molecules), which may be incorporated into a modified nucleic acid or mmR A (e.g., R A or mR A, as described herein), can be modified on the sugar of the ribonucleic acid. For example, the 2' hydroxyl group (OH) can be modified or replaced with a number of different substituents. Exemplary substitutions at the 2'- position include, but are not limited to, H, halo, optionally substituted Ci_6 alkyl; optionally substituted Ci_6 alkoxy; optionally substituted C6-1o aryloxy; optionally substituted C3-8 cycloalkyl; optionally substituted C3_8 cycloalkoxy; optionally substituted C6_io aryloxy; optionally substituted C6-io aryl-Ci_6 alkoxy, optionally substituted C1-12 (heterocyclyl)oxy; a sugar (e.g., ribose, pentose, or any described herein); a polyethyleneglycol (PEG), -0(CH2CH20)nCH2CH2OR, where R is H or optionally substituted alkyl, and n is an integer from 0 to 20 (e.g., from 0 to 4, from 0 to 8, from 0 to 10, from 0 to 16, from 1 to 4, from 1 to 8, from 1 to 10, from 1 to 16, from 1 to 20, from 2 to 4, from 2 to 8, from 2 to 10, from 2 to 16, from 2 to 20, from 4 to 8, from 4 to 10, from 4 to 16, and from 4 to 20); "locked" nucleic acids (LNA) in which the 2 '-hydroxyl is connected by a Ci_6 alkylene or Ci_ 6 heteroalkylene bridge to the 4 '-carbon of the same ribose sugar, where exemplary bridges included methylene, propylene, ether, or amino bridges; aminoalkyl, as defined herein; aminoalkoxy, as defined herein; amino as defined herein; and amino acid, as defined herein.
[00207] Generally, RNA includes the sugar group ribose, which is a 5-membered ring having an oxygen. Exemplary, non-limiting modified nucleotides include replacement of the oxygen in ribose (e.g., with S, Se, or alkylene, such as methylene or ethylene); addition of a double bond (e.g., to replace ribose with cyclopentenyl or cyclohexenyl); ring contraction of ribose (e.g., to form a 4- membered ring of cyclobutane or oxetane); ring expansion of ribose (e.g., to form a 6- or 7- membered ring having an additional carbon or heteroatom, such as for anhydrohexitol, altritol, mannitol, cyclohexanyl, cyclohexenyl, and morpholino that also has a phosphoramidate backbone); multicyclic forms (e.g., tricyclo; and "unlocked" forms, such as glycol nucleic acid (GNA) (e.g., R- GNA or S-GNA, where ribose is replaced by glycol units attached to phosphodiester bonds), threose nucleic acid (TNA, where ribose is replace with a-L-threofuranosyl-(3'→2')) , and peptide nucleic acid (PNA, where 2-amino-ethyl-glycine linkages replace the ribose and phosphodiester backbone). The sugar group can also contain one or more carbons that possess the opposite stereochemical configuration than that of the corresponding carbon in ribose. Thus, a modified nucleic acid molecule or mmRNA can include nucleotides containing, e.g., arabinose, as the sugar.
Modifications on the Nucleobase
[00208] The present disclosure provides for modified nucleosides and nucleotides. As described herein "nucleoside" is defined as a compound containing a sugar molecule (e.g., a pentose or ribose) or a derivative thereof in combination with an organic base (e.g., a purine or pyrimidine) or a derivative thereof. As described herein, "nucleotide" is defined as a nucleoside including a phosphate group. The modified nucleotides (e.g., modified mRNA) may by synthesized by any useful method, as described herein (e.g., chemically, enzymatically, or recombinantly to include one or more modified or non-natural nucleosides).
[00209] The modified nucleotide base pairing encompasses not only the standard adenosine- thymine, adenosine-uracil, or guanosine-cytosine base pairs, but also base pairs formed between nucleotides and/or modified nucleotides comprising non-standard or modified bases, wherein the arrangement of hydrogen bond donors and hydrogen bond acceptors permits hydrogen bonding between a non-standard base and a standard base or between two complementary non-standard base structures. One example of such non-standard base pairing is the base pairing between the modified nucleotide inosine and adenine, cytosine or uracil.
[00210] The modified nucleosides and nucleotides can include a modified nucleobase. Examples of nucleobases found in RNA include, but are not limited to, adenine, guanine, cytosine, and uracil. Examples of nucleobase found in DNA include, but are not limited to, adenine, guanine, cytosine, and thymine. These nucleobases can be modified or wholly replaced to provide modified nucleic acids or mmRNA molecules having enhanced properties, e.g., resistance to nucleases through disruption of the binding of a major groove binding partner. Table 1 below identifies the chemical faces of each canonical nucleotide. Circles identify the atoms comprising the respective chemical regions. Table 1
Watson-Crick ajor Groove Minor Groove
Face Face
Pyrimidines
Purines
Figure imgf000084_0001
[00211] In some embodiments, B is a modified uracil. Exemplary modified uracils include those
Figure imgf000084_0002
in
[00212] ^ is a single or double bond; [00213] each of T1 , T1 , T2 , and T2 is, independently, H, optionally substituted alkyl, optionally substituted alkoxy, or optionally substituted thioalkoxy, or the combination of T1 and T1 or the combination of T2 and T2 join together (e.g., as in T2) to form O (oxo), S (thio), or Se (seleno);
[00214] each of V1 and V2 is, independently, O, S, N(Rvb)nv, or C(Rvb)nv, wherein nv is an integer from 0 to 2 and each R^ is, independently, H, halo, optionally substituted amino acid, optionally substituted alkyl, optionally substituted haloalkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted hydroxyalkyl, optionally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally substituted aminoalkyl (e.g., substituted with an N- protecting group, such as any described herein, e.g., trifluoroacetyl), optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, optionally substituted acylaminoalkyl (e.g., substituted with an N-protecting group, such as any described herein, e.g., trifluoroacetyl), optionally substituted alkoxycarbonylalkyl, optionally substituted alkoxycarbonylalkenyl, optionally substituted alkoxycarbonylalkynyl, or optionally substituted alkoxycarbonylalkoxy (e.g., optionally substituted with any substituent described herein, such as those selected from (1)-(21) for alkyl);
[00215] R10 is H, halo, optionally substituted amino acid, hydroxy, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aminoalkyl, optionally substituted hydroxyalkyl, optionally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, optionally substituted alkoxy, optionally substituted alkoxycarbonylalkyl, optionally substituted alkoxycarbonylalkenyl, optionally substituted alkoxycarbonylalkynyl, optionally substituted alkoxycarbonylalkoxy, optionally substituted carboxyalkoxy, optionally substituted carboxyalkyl, or optionally substituted carbamoylalkyl;
[00216] R11 is H or optionally substituted alkyl;
[00217] R12a is H, optionally substituted alkyl, optionally substituted hydroxyalkyl, optionally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally substituted
aminoalkyl, optionally substituted aminoalkenyl, or optionally substituted aminoalkynyl, optionally substituted carboxyalkyl (e.g., optionally substituted with hydroxy), optionally substituted carboxyalkoxy, optionally substituted carboxyaminoalkyl, or optionally substituted carbamoylalkyl; and [00218] R c is H, halo, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted thioalkoxy, optionally substituted amino, optionally substituted hydroxyalkyl, optionally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, or optionally substituted aminoalkynyl.
[00219] Other exemplary modified uracils include those having Formula (b6)-(b9):
Figure imgf000086_0001
pharmaceutically acceptable salt or stereoisomer thereof, wherein
[00220] ^ is a single or double bond;
[00221] each of T1 , T1 , T2 , and T2 is, independently, H, optionally substituted alkyl, optionally substituted alkoxy, or optionally substituted thioalkoxy, or the combination of T1 and T1 join together (e.g., as in T1) or the combination of T2 and T2 join together (e.g., as in T2) to form O (oxo), S (thio), or Se (seleno), or each T1 and T2 is, independently, O (oxo), S (thio), or Se (seleno);
[00222] each of W1 and W2 is, independently, N(RWa)nw or C(RWa)nw, wherein nw is an integer from 0 to 2 and each RWa is, independently, H, optionally substituted alkyl, or optionally substituted alkoxy;
[00223] each V3 is, independently, O, S, N(RVa)nv, or C(RVa)nv, wherein nv is an integer from 0 to 2 and each RVa is, independently, H, halo, optionally substituted amino acid, optionally substituted alkyl, optionally substituted hydroxyalkyl, optionally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted heterocyclyl, optionally substituted alkheterocyclyl, optionally substituted alkoxy, optionally substituted alkenyloxy, or optionally substituted alkynyloxy, optionally substituted aminoalkyl (e.g., substituted with an N-protecting group, such as any described herein, e.g., trifluoroacetyl, or sulfoalkyl), optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, optionally substituted acylaminoalkyl (e.g., substituted with an N-protecting group, such as any described herein, e.g., trifluoroacetyl), optionally substituted alkoxycarbonylalkyl, optionally substituted alkoxycarbonylalkenyl, optionally substituted alkoxycarbonylalkynyl, optionally substituted alkoxycarbonylacyl, optionally substituted alkoxycarbonylalkoxy, optionally substituted carboxyalkyl (e.g., optionally substituted with hydroxy and/or an O-protecting group), optionally substituted carboxyalkoxy, optionally substituted carboxyaminoalkyl, or optionally substituted carbamoylalkyl (e.g., optionally substituted with any substituent described herein, such as those selected from (1)-(21) for alkyl), and wherein RVa and R12c taken together with the carbon atoms to which they are attached can form optionally substituted cycloalkyl, optionally substituted aryl, or optionally substituted heterocyclyl (e.g., a 5- or 6-membered ring);
[00224] R12a is H, optionally substituted alkyl, optionally substituted hydroxyalkyl, optionally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, optionally substituted carboxyalkyl (e.g., optionally substituted with hydroxy and/or an O-protecting group), optionally substituted carboxyalkoxy, optionally substituted carboxyaminoalkyl, optionally substituted carbamoylalkyl, or absent;
[00225] R12b is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted hydroxyalkyl, optionally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, optionally substituted alkaryl, optionally substituted heterocyclyl, optionally substituted alkheterocyclyl,
optionally substituted amino acid, optionally substituted alkoxycarbonylacyl, optionally substituted alkoxycarbonylalkoxy, optionally substituted alkoxycarbonylalkyl, optionally substituted alkoxycarbonylalkenyl, optionally substituted alkoxycarbonylalkynyl, optionally substituted alkoxycarbonylalkoxy, optionally substituted carboxyalkyl (e.g., optionally substituted with hydroxy and/or an O-protecting group), optionally substituted carboxyalkoxy, optionally substituted carboxyaminoalkyl, or optionally substituted carbamoylalkyl,
[00226] wherein the combination of R12b and T1 or the combination of R12b and R12c can join together to form optionally substituted heterocyclyl; and
[00227] R12c is H, halo, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted thioalkoxy, optionally substituted amino, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, or optionally substituted aminoalkynyl.
[00228] Further exemplary modified uracils include those having Formula (b28)-(b31):
Figure imgf000088_0001
(b31), or a pharmaceutically acceptable salt or stereoisomer thereof, wherein
[00229] each of T1 and T2 is, independently, O (oxo), S (thio), or Se (seleno);
[00230] each R^ and R^ is, independently, H, halo, optionally substituted amino acid, optionally substituted alkyl, optionally substituted haloalkyl, optionally substituted hydroxyalkyl, optionally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkyl (e.g., substituted with an N- protecting group, such as any described herein, e.g., trifluoroacetyl, or sulfoalkyl), optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, optionally substituted acylaminoalkyl (e.g., substituted with an N-protecting group, such as any described herein, e.g., trifluoroacetyl), optionally substituted alkoxycarbonylalkyl, optionally substituted alkoxycarbonylalkenyl, optionally substituted alkoxycarbonylalkynyl, optionally substituted alkoxycarbonylacyl, optionally substituted alkoxycarbonylalkoxy, optionally substituted carboxyalkyl (e.g., optionally substituted with hydroxy and/or an O-protecting group), optionally substituted carboxyalkoxy, optionally substituted carboxyaminoalkyl, or optionally substituted carbamoylalkyl (e.g., optionally substituted with any substituent described herein, such as those selected from (1)-(21) for alkyl) (e.g., R^ is optionally substituted alkyl, optionally substituted alkenyl, or optionally substituted aminoalkyl, e.g., substituted with an N-protecting group, such as any described herein, e.g., trifluoroacetyl, or sulfoalkyl);
[00231] R12a is H, optionally substituted alkyl, optionally substituted carboxyaminoalkyl, optionally substituted aminoalkyl (e.g., e.g., substituted with an N-protecting group, such as any described herein, e.g., trifluoroacetyl, or sulfoalkyl), optionally substituted aminoalkenyl, or optionally substituted aminoalkynyl; and [00232] R is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted hydroxyalkyl, optionally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, optionally substituted aminoalkynyl (e.g., e.g., substituted with an N-protecting group, such as any described herein, e.g., trifluoroacetyl, or sulfoalkyl),
[00233] optionally substituted alkoxycarbonylacyl, optionally substituted alkoxycarbonylalkoxy, optionally substituted alkoxycarbonylalkyl, optionally substituted alkoxycarbonylalkenyl, optionally substituted alkoxycarbonylalkynyl, optionally substituted alkoxycarbonylalkoxy, optionally substituted carboxyalkoxy, optionally substituted carboxyalkyl, or optionally substituted
carbamoylalkyl.
[00234] In particular embodiments, T1 is O (oxo), and T2 is S (thio) or Se (seleno). In other embodiments, T1 is S (thio), and T2 is O (oxo) or Se (seleno). In some embodiments, R^ is H, optionally substituted alkyl, or optionally substituted alkoxy.
[00235] In other embodiments, each R12a and R12b is, independently, H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, or optionally substituted hydroxyalkyl. In particular embodiments, R12a is H. In other embodiments, both R12a and R12b are H.
[00236] In some embodiments, each R^ of R12b is, independently, optionally substituted aminoalkyl (e.g., substituted with an N-protecting group, such as any described herein, e.g., trifluoroacetyl, or sulfoalkyl), optionally substituted aminoalkenyl, optionally substituted
aminoalkynyl, or optionally substituted acylaminoalkyl (e.g., substituted with an N-protecting group, such as any described herein, e.g., trifluoroacetyl). In some embodiments, the amino and/or alkyl of the optionally substituted aminoalkyl is substituted with one or more of optionally substituted alkyl, optionally substituted alkenyl, optionally substituted sulfoalkyl, optionally substituted carboxy (e.g., substituted with an O-protecting group), optionally substituted hydroxy (e.g., substituted with an O- protecting group), optionally substituted carboxyalkyl (e.g., substituted with an O-protecting group), optionally substituted alkoxycarbonylalkyl (e.g., substituted with an O-protecting group), or N- protecting group. In some embodiments, optionally substituted aminoalkyl is substituted with an optionally substituted sulfoalkyl or optionally substituted alkenyl. In particular embodiments, R12a and R^ are both H. In particular embodiments, T1 is O (oxo), and T2 is S (thio) or Se (seleno).
[00237] In some embodiments, R^ is optionally substituted alkoxycarbonylalkyl or optionally substituted carbamoylalkyl. [00238] In particular embodiments, the optional substituent for R a, R , R c, or R a is a polyethylene glycol group (e.g., -(CH2)S2(OCH2CH2)si(CH2)S30R', wherein si is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R' is H or Ci_2o alkyl); or an amino-polyethylene glycol group (e.g., -NRN1(CH2)s2(CH2CH20)si(CH2)s3NRN1, wherein si is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and each RN1 is, independently, hydrogen or optionally substituted Ci_6 alkyl).
[00239] In some embodiments, B is a modified cytosine. Exemplary modified cytosines include compounds (bl0)-(bl4):
(bl3), or
Figure imgf000090_0001
(bl4), or a pharmaceutically acceptable salt or stereoisomer thereof, wherein
[00240] each of T3 and T3 is, independently, H, optionally substituted alkyl, optionally substituted alkoxy, or optionally substituted thioalkoxy, or the combination of T3 and T3 join together (e.g., as in T3) to form O (oxo), S (thio), or Se (seleno);
[00241] each V4 is, independently, O, S, N(RVc)nv, or C(RVc)nv, wherein nv is an integer from 0 to 2 and each RVc is, independently, H, halo, optionally substituted amino acid, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted heterocyclyl, optionally substituted alkheterocyclyl, or optionally substituted alkynyloxy (e.g., optionally substituted with any substituent described herein, such as those selected from (1)-(21) for alkyl), wherein the combination of R13b and RVc can be taken together to form optionally substituted heterocyclyl;
[00242] each V5 is, independently, N(Rvd)nv, or C(Rvd)nv, wherein nv is an integer from 0 to 2 and each Rvd is, independently, H, halo, optionally substituted amino acid, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted heterocyclyl, optionally substituted alkheterocyclyl, or optionally substituted alkynyloxy (e.g., optionally substituted with any substituent described herein, such as those selected from (1)-(21) for alkyl) (e.g., V5 is -CH or N);
[00243] each of R13a and R13b is, independently, H, optionally substituted acyl, optionally substituted acyloxyalkyl, optionally substituted alkyl, or optionally substituted alkoxy, wherein the combination of R13b and R14 can be taken together to form optionally substituted heterocyclyl;
[00244] each R14 is, independently, H, halo, hydroxy, thiol, optionally substituted acyl, optionally substituted amino acid, optionally substituted alkyl, optionally substituted haloalkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted hydroxyalkyl (e.g., substituted with an O-protecting group), optionally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl,, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, optionally substituted acyloxyalkyl, optionally substituted amino (e.g., -NHR, wherein R is H, alkyl, aryl, or phosphoryl), azido, optionally substituted aryl, optionally substituted heterocyclyl, optionally substituted alkheterocyclyl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, or optionally substituted aminoalkyl; and
[00245] each of R15 and R16 is, independently, H, optionally substituted alkyl, optionally substituted alkenyl, or optionally substituted alkynyl.
[00246] Further exemplary modified cytosines include those having Formula (b32)-(b35):
Figure imgf000091_0001
(b32), (b33), or (b35), or a pharmaceutically acceptable salt or stereoisomer thereof, wherein
[00247] each of T1 and T3 is, independently, O (oxo), S (thio), or Se (seleno);
[00248] each of R13a and R13b is, independently, H, optionally substituted acyl, optionally substituted acyloxyalkyl, optionally substituted alkyl, or optionally substituted alkoxy, wherein the combination of R13b and R14 can be taken together to form optionally substituted heterocyclyl;
[00249] each R14 is, independently, H, halo, hydroxy, thiol, optionally substituted acyl, optionally substituted amino acid, optionally substituted alkyl, optionally substituted haloalkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted hydroxyalkyl (e.g., substituted with an O-protecting group), optionally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, optionally substituted acyloxyalkyl, optionally substituted amino (e.g., -NHR, wherein R is H, alkyl, aryl, or phosphoryl), azido, optionally substituted aryl, optionally substituted heterocyclyl, optionally substituted alkheterocyclyl, optionally substituted aminoalkyl (e.g., hydroxyalkyl, alkyl, alkenyl, or alkynyl), optionally substituted aminoalkenyl, or optionally substituted aminoalkynyl; and
[00250] each of R15 and R16 is, independently, H, optionally substituted alkyl, optionally substituted alkenyl, or optionally substituted alkynyl (e.g., R15 is H, and R16 is H or optionally substituted alkyl).
[00251] In some embodiments, R15 is H, and R16 is H or optionally substituted alkyl. In particular embodiments, R14 is H, acyl, or hydroxyalkyl. In some embodiments, R14 is halo. In some embodiments, both R14 and R15 are H. In some embodiments, both R15 and R16 are H. In some embodiments, each of R14 and R15 and R16 is H. In further embodiments, each of R13a and R13b is independently, H or optionally substituted alkyl.
n- limiting examples of modified cytosines include compounds of Formula (b36):
Figure imgf000092_0001
(b36) or a pharmaceutically acceptable salt or stereoisomer thereof, wherein
[00253] each R13b is, independently, H, optionally substituted acyl, optionally substituted acyloxyalkyl, optionally substituted alkyl, or optionally substituted alkoxy, wherein the combination of R13b and R14b can be taken together to form optionally substituted heterocyclyl;
[00254] each R14a and R14b is, independently, H, halo, hydroxy, thiol, optionally substituted acyl, optionally substituted amino acid, optionally substituted alkyl, optionally substituted haloalkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted hydroxyalkyl (e.g., substituted with an O-protecting group), optionally substituted hydroxyalkenyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, optionally substituted acyloxyalkyl, optionally substituted amino (e.g., -NHR, wherein R is H, alkyl, aryl, phosphoryl, optionally substituted aminoalkyl, or optionally substituted carboxyaminoalkyl), azido, optionally substituted aryl, optionally substituted heterocyclyl, optionally substituted alkheterocyclyl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, or optionally substituted aminoalkynyl; and
[00255] each of R15 is, independently, H, optionally substituted alkyl, optionally substituted alkenyl, or optionally substituted alkynyl.
[00256] In particular embodiments, R14b is an optionally substituted amino acid (e.g., optionally substituted lysine). In some embodiments, R14a is H.
[00257] In some embodiments, B is a modified guanine. Exemplary modified guanines include compounds of Formula (bl5)-(bl7):
Figure imgf000093_0001
or a pharmaceutically acceptable salt or stereoisomer thereof, wherein
[00258] each of T4 , T4 , T5 , T5 , T6 , and T6 is, independently, H, optionally substituted alkyl, or optionally substituted alkoxy, and wherein the combination of T4 and T4 (e.g., as in T4) or the combination of T5 and T5 (e.g., as in T5) or the combination of T6 and T6 join together (e.g., as in T6) form O (oxo), S (thio), or Se (seleno);
[00259] each of V5 and V6 is, independently, O, S, N(Rvd)nv, or C(Rvd)nv, wherein nv is an integer from 0 to 2 and each Rvd is, independently, H, halo, thiol, optionally substituted amino acid, cyano, amidine, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy (e.g., optionally substituted with any substituent described herein, such as those selected from (1)-(21) for alkyl), optionally substituted thioalkoxy, or optionally substituted amino; and
[00260] each of R17, R18, R19a, R19b, R21, R22, R23, and R24 is, independently, H, halo, thiol, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted thioalkoxy, optionally substituted amino, or optionally substituted amino acid.
[00261] Exemplary modified guanosines include compounds of Formula (b37)-(b40):
Figure imgf000094_0001
(b40), or a pharmaceutically acceptable salt or stereoisomer thereof, wherein
[00262] each of T is, independently, H, optionally substituted alkyl, or optionally substituted alkoxy, and each T4 is, independently, O (oxo), S (thio), or Se (seleno);
[00263] each of R18, R19a, R19b, and R21 is, independently, H, halo, thiol, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted thioalkoxy, optionally substituted amino, or optionally substituted amino acid.
[00264] In some embodiments, R18 is H or optionally substituted alkyl. In further embodiments, T4 is oxo. In some embodiments, each of R19a and R19b is, independently, H or optionally substituted alkyl.
[00265] In some embodiments, B is a modified adenine. Exemplary modified adenines include compounds of Formula (M8)-(b20):
Figure imgf000094_0002
or a pharmaceutically acceptable salt or stereoisomer thereof, wherein
[00266] each V7 is, independently, O, S, N(RVe)nv, or C(RVe)nv, wherein nv is an integer from 0 to 2 and each RVe is, independently, H, halo, optionally substituted amino acid, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted alkoxy, optionally substituted alkenyloxy, or optionally substituted alkynyloxy (e.g., optionally substituted with any substituent described herein, such as those selected from (1)-(21) for alkyl); [00267] each R is, independently, H, halo, thiol, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted thioalkoxy, or optionally substituted amino;
[00268] each of R26a and R26b is, independently, H, optionally substituted acyl, optionally substituted amino acid, optionally substituted carbamoylalkyl, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted hydroxyalkyl, optionally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally substituted alkoxy, or polyethylene glycol group (e.g., -(CH2)s2(OCH2CH2)si(CH2)s30R', wherein si is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R' is H or Ci_2o alkyl); or an amino-polyethylene glycol group (e.g., -NRN1(CH2)s2(CH2CH20)si(CH2)s3NRN1 , wherein si is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and each RN1 is, independently, hydrogen or optionally substituted Ci_6 alkyl);
[00269] each R27 is, independently, H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted alkoxy, optionally substituted thioalkoxy or optionally substituted amino;
[00270] each R28 is, independently, H, optionally substituted alkyl, optionally substituted alkenyl, or optionally substituted alkynyl; and
[00271] each R29 is, independently, H, optionally substituted acyl, optionally substituted amino acid, optionally substituted carbamoylalkyl, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted hydroxyalkyl, optionally substituted hydroxyalkenyl, optionally substituted alkoxy, or optionally substituted amino.
[00272] Exemplary modified adenines include compounds of Formula (b41 )-(b43) :
Figure imgf000095_0001
pharmaceutically acceptable salt or stereoisomer thereof, wherein [00273] each R is, independently, H, halo, thiol, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted thioalkoxy, or optionally substituted amino;
[00274] each of R26a and R26b is, independently, H, optionally substituted acyl, optionally substituted amino acid, optionally substituted carbamoylalkyl, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted hydroxyalkyl, optionally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally substituted alkoxy, or polyethylene glycol group (e.g., -(CH2)s2(OCH2CH2)si(CH2)s30R', wherein si is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R' is H or Ci_2o alkyl); or an amino-polyethylene glycol group (e.g., -NRN1(CH2)s2(CH2CH20)si(CH2)s3NRN1 , wherein si is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and each RN1 is, independently, hydrogen or optionally substituted Ci_6 alkyl); and
[00275] each R27 is, independently, H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted alkoxy, optionally substituted thioalkoxy, or optionally substituted amino.
[00276] In some embodiments, R26a is H, and R26b is optionally substituted alkyl. In some embodiments, each of R26a and R26b is, independently, optionally substituted alkyl. In particular embodiments, R27 is optionally substituted alkyl, optionally substituted alkoxy, or optionally substituted thioalkoxy. In other embodiments, R25 is optionally substituted alkyl, optionally substituted alkoxy, or optionally substituted thioalkoxy.
[00277] In particular embodiments, the optional substituent for R26a, R26b, or R29 is a polyethylene glycol group (e.g., -(CH2)S2(OCH2CH2)si(CH2)S30R', wherein si is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R' is H or Ci_2o alkyl); or an amino- polyethylene glycol group (e.g., -NRN1(CH2)S2(CH2CH20)si(CH2)S3NRN1, wherein si is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and each RN1 is, independently, hydrogen or optionally substituted Ci_6 alkyl).
[00278] In some embodiments, B may have Formula (b21):
Figure imgf000097_0001
(b21), wherein X is, independently, O, S, optionally substituted alkylene (e.g., methylene), or optionally substituted heteroalkylene, xa is an integer from 0 to 3, and R12a and T2 are as described herein.
[00279] In some embodiments, B may have Formula (b22):
Figure imgf000097_0002
(b22), wherein R10 is, independently, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted heterocyclyl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, optionally substituted alkoxy, optionally substituted alkoxycarbonylalkyl, optionally substituted alkoxycarbonylalkenyl, optionally substituted alkoxycarbonylalkynyl, optionally substituted alkoxycarbonylalkoxy, optionally substituted carboxyalkoxy, optionally substituted carboxyalkyl, or optionally substituted carbamoylalkyl, and R11, R12a, T1, and T2 are as described herein.
[00280] In some embodiments, B may have Formula (b23):
Figure imgf000097_0003
(b23), wherein R1U is optionally substituted heterocyclyl (e.g., optionally substituted furyl, optionally substitued thienyl, or optionally substitued pyrrolyl), optionally substituted aryl (e.g., optionally substituted phenyl or optionally substituted naphthyl), or any substituent described herein (e.g., for R10) ;and wherein R11 (e.g., H or any substituent described herein), R12a (e.g., H or any substituent described herein), T1 (e.g., oxo or any substituent described herein), and T2 (e.g., oxo or any substituent described herein) are as described herein.
[00281] In some embodiments, B may have Formula (b24):
Figure imgf000098_0001
(b24), wherein R14 is, independently, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted heterocyclyl, optionally substituted alkaryl, optionally substituted alkheterocyclyl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, optionally substituted alkoxy, optionally substituted alkoxycarbonylalkyl, optionally substituted alkoxycarbonylalkenyl, optionally substituted alkoxycarbonylalkynyl, optionally substituted alkoxycarbonylalkoxy, optionally substituted carboxyalkoxy, optionally substituted carboxyalkyl, or optionally substituted carbamoylalkyl, and R13a, R13b, R15, and T3 are as described herein.
[00282] In some embodiments, B may have Formula (b25):
Figure imgf000098_0002
(b25), wherein R14 is optionally substituted heterocyclyl (e.g., optionally substituted furyl, optionally substitued thienyl, or optionally substitued pyrrolyl), optionally substituted aryl (e.g., optionally substituted phenyl or optionally substituted naphthyl), or any substituent described herein (e.g., for R14 or R14 ); and wherein R13a (e.g., H or any substituent described herein), R13b (e.g., H or any substituent described herein), R15 (e.g., H or any substituent described herein), and T3 (e.g., oxo or any substituent described herein) are as described herein.
[00283] In some embodiments, B is a nucleobase selected from the group consisting of cytosine, guanine, adenine, and uracil. In some embodiments, B may be:
Figure imgf000098_0003
[00284] In some embodiments, the modified nucleobase is a modified uracil. Exemplary nucleobases and nucleosides having a modified uracil include pseudouridine (ψ), pyridin-4-one ribonucleoside, 5-aza-uridine, 6-aza-uridine, 2-thio-5-aza-uridine, 2-thio-uridine (s2U), 4-thio- uridine (s4U), 4-thio-pseudouridine, 2-thio-pseudouridine, 5 -hydroxy-uridine (ho5U), 5-aminoallyl- uridine, 5-halo-uridine (e.g., 5-iodo~uridine or 5-bromo-uridine), 3-methyl-uridine (m3U), 5- methoxy-uridine (mo5U), uridine 5-oxyacetic acid (cmo5U), uridine 5-oxyacetic acid methyl ester (mcmo5U), 5-carboxymethyl-uridine (cm5U), 1-carboxymethyl-pseudouridine, 5- carboxyhydroxymethyl-uridine (chm5U), 5-carboxyhydroxymethyl-uridine methyl ester (mchm5U), 5-methoxycarbonylmethyl-uridine (mcm5U), 5-methoxycarbonylmethyl-2-thio-uridine (mcm5s2U), 5-aminomethyl-2-thio-uridine (nmVU), 5-methylaminomethyl-uridine (mnm5U), 5- methylaminomethyl-2-thio-uridine (mnm5s2U), 5-methylaminomethyl-2-seleno-uridine (mnm5se2U), 5-carbamoylmethyl-uridine (ncm5U), 5-carboxymethylaminomethyl-uridine (cmnm5U), 5- carboxymethylaminomethyl-2-thio-uridine (cmnm5s2U), 5-propynyl-uridine, 1-propynyl- pseudouridine, 5-taurinomethyl-uridine (xm5U), 1-taurinomethyl-pseudouridine, 5-taurinomethyl-2- thio-uridine^mVU), l-taurinomethyl-4-thio-pseudouridine, 5-methyl-uridine (m5U, i.e., having the nucleobase deoxythymine), 1-methyl-pseudouridine (m^), 5-methyl-2-thio-uridine (m5s2U), 1- methyl-4-thio-pseudouridine
Figure imgf000099_0001
4-thio- 1-methyl-pseudouridine, 3-methyl-pseudouridine (m3\|/), 2-thio- 1-methyl-pseudouridine, 1 -methyl- 1-deaza-pseudouridine, 2-thio-l -methyl- 1-deaza- pseudouridine, dihydrouridine (D), dihydropseudouridine, 5,6-dihydrouridine, 5-methyl- dihydrouridine (m5D), 2-thio-dihydrouridine, 2-thio-dihydropseudouridine, 2-methoxy-uridine, 2- methoxy-4-thio-uridine, 4-methoxy-pseudouridine, 4-methoxy-2-thio-pseudouridine, N 1-methyl- pseudouridine, 3-(3-amino-3-carboxypropyl)uridine (acp3U), l-methyl-3-(3-amino-3- carboxypropyl)pseudouridine (acp3 ψ), 5-(isopentenylaminomethyl)uridine (inm5U), 5- (isopentenylaminomethyl)-2-thio-uridine (inm5s2U), a-thio-uridine, 2'-0-methyl-uridine (Um), 5,2'- O-dimethyl-uridine (m5Um), 2'-0-methyl-pseudouridine (ψτη), 2-thio-2'-0-methyl-uridine (s2Um), 5 -methoxycarbonylmethyl-2 ' -O-methyl-uridine (mcm5Um), 5 -carbamoylmethyl-2 ' -O-methyl- uridine (ncm5Um), 5 -carboxymethylaminomethyl-2' -O-methyl-uridine (cmnm5Um), 3,2'-0- dimethyl-uridine (m3Um), 5 -(isopentenylaminomethyl)-2' -O-methyl-uridine (inm5Um), 1-thio- uridine, deoxythymidine, 2'-F-ara -uridine, 2'-F-uridine, 2'-OH-ara-uridine, 5-(2-carbomethoxyvinyl) uridine, and 5-[3-(l -E-propenylamino)uridine..
[00285] In some embodiments, the modified nucleobase is a modified cytosine. Exemplary nucleobases and nucleosides having a modified cytosine include 5-aza-cytidine, 6-aza-cytidine, pseudoisocytidine, 3-methyl-cytidine (m3C), N4-acetyl-cytidine (ac4C), 5-formyl-cytidine (f5C), N4- methyl-cytidine (m4C), 5-methyl-cytidine (m5C), 5-halo-cytidine (e.g., 5-iodo-cytidine), 5- hydroxymethyl-cytidine (hm5C), 1-methyl-pseudoisocytidine, pyrrolo-cytidine, pyrrolo- pseudoisocytidine, 2-thio-cytidine (s2C), 2-thio-5-methyl-cytidine, 4-thio-pseudoisocytidine, 4-thio- 1 -methyl-pseudoisocytidine, 4-thio- 1 -methyl- 1 -deaza-pseudoisocytidine, 1 -methyl- 1 -deaza- pseudoisocytidine, zebularine, 5-aza-zebularine, 5-methyl-zebularine, 5-aza-2-thio-zebularine, 2- thio-zebularine, 2-methoxy-cytidine, 2-methoxy-5-methyl-cytidine, 4-methoxy-pseudoisocytidine, 4- methoxy- 1-methyl-pseudoisocytidine, lysidine (k2C), a-thio-cytidine, 2'-0-methyl-cytidine (Cm), 5,2'-0-dimethyl-cytidine (m5Cm), N4-acetyl-2'-0-methyl-cytidine (ac4Cm), N4,2'-0-dimethyl- cytidine (m4Cm), 5-formyl-2'-0-methyl-cytidine (f5Cm), N4,N4,2'-0-trimethyl-cytidine (m4 2Cm),
1- thio-cytidine, 2'-F-ara-cytidine, 2'-F-cytidine, and 2'-OH-ara-cytidine.
[00286] In some embodiments, the modified nucleobase is a modified adenine. Exemplary nucleobases and nucleosides having a modified adenine include 2-amino-purine, 2, 6-diaminopurine,
2- amino-6-halo-purine (e.g., 2-amino-6-chloro-purine), 6-halo-purine (e.g., 6-chloro-purine), 2- amino-6-methyl-purine, 8-azido-adenosine, 7-deaza-adenine, 7-deaza-8-aza-adenine, 7-deaza-2- amino-purine, 7-deaza-8-aza-2-amino-purine, 7-deaza-2, 6-diaminopurine, 7-deaza-8-aza-2,6- diaminopurine, 1-methyl-adenosine (mxA), 2-methyl-adenine (m2A), N6-methyl-adenosine(m6A), 2- methylthio-N6-methyl-adenosine (ms2m6A), N6-isopentenyl-adenosine (i6A), 2-methylthio-N6- isopentenyl-adenosine (ms2i6A), N6-(cis-hydroxyisopentenyl)adenosine (io6A), 2-methylthio-N6- (cis-hydroxyisopentenyl)adenosine (ms2io6A), N6-glycinylcarbamoyl-adenosine (g6A), N6- threonylcarbamoyl-adenosine (t6A), N6-methyl-N6-threonylcarbamoyl-adenosine (m6t6A), 2- methylthio-N6-threonylcarbamoyl-adenosine (ms2g6A), N6,N6-dimethyl-adenosine (m6 2A), N6- hydroxynorvalylcarbamoyl-adenosine (hn6A), 2-methylthio-N6-hydroxynorvalylcarbamoyl- adenosine (ms2hn6A), N6-acetyl-adenosine (ac6A), 7-methyl-adenine, 2-methylthio-adenine, 2- methoxy-adenine, a-thio-adenosine, 2'-0-methyl-adenosine (Am), N6,2'-0-dimethyl-adenosine (m6Am), N6,N6,2'-0-trimethyl-adenosine (m6 2Am), l,2'-0-dimethyl-adenosine (rr^Am), 2'-0- ribosyladenosine (phosphate) (Ar(p)), 2-amino-N6-methyl-purine, 1-thio-adenosine, 8-azido- adenosine, 2'-F-ara-adenosine, 2'-F-adenosine, 2'-OH-ara-adenosine, and
N6 - ( 19 - amino -pentaoxanonadecy l)-adenosine .
[00287] In some embodiments, the modified nucleobase is a modified guanine. Exemplary nucleobases and nucleosides having a modified guanine include inosine (I), 1-methyl-inosine (m1!), wyosine (imG), methylwyosine (mimG), 4-demethyl-wyosine (imG-14), isowyosine (imG2), wybutosine (yW), peroxywybutosine (o2yW), hydroxywybutosine (OHyW), undermodified hydroxywybutosine (OHyW*), 7-deaza-guanosine, queuosine (Q), epoxyqueuosine (oQ), galactosyl- queuosine (galQ), mannosyl-queuosine (manQ), 7-cyano-7-deaza-guanosine (preQ0), 7- aminomethyl-7-deaza-guanosine (preQi), archaeosine (G ), 7-deaza-8-aza-guanosine, 6-thio- guanosine, 6-thio-7-deaza-guanosine, 6-thio-7-deaza-8-aza-guanosine, 7-methyl-guanosine (m7G), 6-thio-7-methyl-guanosine, 7-methyl-inosine, 6-methoxy-guanosine, 1-methyl-guanosine ( G), N2-methyl-guanosine (m2G), N2,N2-dimethyl-guanosine (m2 2G), N2,7-dimethyl-guanosine (m2'7G), N2, N2,7-dimethyl-guanosine (m2'2'7G), 8-oxo-guanosine, 7-methyl-8-oxo-guanosine, l-methyl-6- thio-guanosine, N2-methyl-6-thio-guanosine, N2,N2-dimethyl-6-thio-guanosine, a-thio-guanosine, 2'-0-methyl-guanosine (Gm), N2-methyl-2'-0-methyl-guanosine (m2Gm), N2,N2-dimethyl-2'-0- methyl-guanosine (m2 2Gm), l-methyl-2'-0-methyl-guanosine ( Gm), N2,7-dimethyl-2'-0-methyl- guanosine (m2'7Gm), 2'-0-methyl-inosine (Im), l,2'-0-dimethyl-inosine (m^m), 2'-0- ribosylguanosine (phosphate) (Gr(p)), 1-thio-guanosine, 06-methyl-guanosine, 2'-F-ara-guanosine, and 2'-F-guanosine.
[00288] The nucleobase of the nucleotide can be independently selected from a purine, a pyrimidine, a purine or pyrimidine analog. For example, the nucleobase can each be independently selected from adenine, cytosine, guanine, uracil, or hypoxanthine. In another embodiment, the nucleobase can also include, for example, naturally-occurring and synthetic derivatives of a base, including pyrazolo[3,4-d]pyrimidines, 5-methylcytosine (5-me-C), 5-hydroxymethyl cytosine, xanthine, hypoxanthine, 2-aminoadenine, 6-methyl and other alkyl derivatives of adenine and guanine, 2-propyl and other alkyl derivatives of adenine and guanine, 2-thiouracil, 2-thiothymine and 2-thiocytosine, 5-propynyl uracil and cytosine, 6-azo uracil, cytosine and thymine, 5 -uracil (pseudouracil), 4-thiouracil, 8-halo (e.g., 8-bromo), 8-amino, 8-thiol, 8-thioalkyl, 8-hydroxyl and other 8-substituted adenines and guanines, 5-halo particularly 5-bromo, 5-trifluoromethyl and other 5-substituted uracils and cytosines, 7-methylguanine and 7-methyladenine, 8-azaguanine and 8- azaadenine, deazaguanine, 7-deazaguanine, 3-deazaguanine, deazaadenine, 7-deazaadenine, 3- deazaadenine, pyrazolo[3,4-d]pyrimidine, imidazo[l,5-a] l,3,5 triazinones, 9-deazapurines, imidazo[4,5-d]pyrazines, thiazolo[4,5-d]pyrimidines, pyrazin-2-ones, 1,2,4-triazine, pyridazine; and 1,3,5 triazine. When the nucleotides are depicted using the shorthand A, G, C, T or U, each letter refers to the representative base and/or derivatives thereof, e.g., A includes adenine or adenine analogs, e.g., 7-deaza adenine). Modifications on the Internucleoside Linkage
[00289] The modified nucleosides and nucleotides, which may be incorporated into a modified nucleic acid or mmRNA molecule, can be modified on the internucleoside linkage (e.g., phosphate backbone). The phosphate groups of the backbone can be modified by replacing one or more of the oxygen atoms with a different substituent. Further, the modified nucleosides and nucleotides can include the wholesale replacement of an unmodified phosphate moiety with a modified phosphate as described herein. Examples of modified phosphate groups include, but are not limited to, phosphorothioate, phosphoroselenates, boranophosphates, boranophosphate esters, hydrogen phosphonates, phosphoramidates, phosphorodiamidates, alkyl or aryl phosphonates, and
phosphotriesters. Phosphorodithioates have both non-linking oxygens replaced by sulfur. The phosphate linker can also be modified by the replacement of a linking oxygen with nitrogen (bridged phosphoramidates), sulfur (bridged phosphorothioates), and carbon (bridged methylene- phosphonates).
[00290] The a-thio substituted phosphate moiety is provided to confer stability to RNA and DNA polymers through the unnatural phosphorothioate backbone linkages. Phosphorothioate DNA and RNA have increased nuclease resistance and subsequently a longer half-life in a cellular
environment. Phosphorothioate linked modified nucleic acids or mmRNA molecules are expected to also reduce the innate immune response through weaker binding/activation of cellular innate immune molecules.
[00291] In specific embodiments, a modified nucleoside includes an alpha-thio-nucleoside (e.g., 5'- 0-(l-thiophosphate)-adenosine, 5'-0-(l-thiophosphate)-cytidine (a-thio-cytidine), 5'-0-(l- thiophosphate)-guanosine, 5'-0-(l-thiophosphate)-uridine, or 5'-0-(l-thiophosphate)- pseudouridine).
Combinations of Modified Sugars, Nucleobases, and Internucleoside Linkages
[00292] The modified nucleic acids and mmRNA of the invention can include a combination of modifications to the sugar, the nucleobase, and/or the internucleoside linkage. These combinations can include any one or more modifications described herein. For examples, any of the nucleotides described herein in Formulas (la), (Ia-l)-(Ia-3), (Ib)-(If), (Ila)-(IIp), (IIb-1), (IIb-2), (IIc-l)-(IIc-2), (IIn-1), (IIn-2), (IVa)-(IVl), and (IXa)-(IXr) can be combined with any of the nucleobases described herein (e.g., in Formulas (bl)-(b43) or any other described herein).
Synthesis of Modified Nucleic Acids and mmRNA Molecules [00293] The modified nucleic acid and mmRNA molecules for use in accordance with the invention may be prepared according to any useful technique, as described herein. The modified nucleosides and nucleotides used in the synthesis of modified nucleic acid and mmRNA molecules disclosed herein can be prepared from readily available starting materials using the following general methods and procedures. Where typical or preferred process conditions (e.g., reaction temperatures, times, mole ratios of reactants, solvents, pressures, etc.) are provided, a skilled artisan would be able to optimize and develop additional process conditions. Optimum reaction conditions may vary with the particular reactants or solvent used, but such conditions can be determined by one skilled in the art by routine optimization procedures.
[00294] The processes described herein can be monitored according to any suitable method known in the art. For example, product formation can be monitored by spectroscopic means, such as nuclear magnetic resonance spectroscopy (e.g., 1H or 13C) infrared spectroscopy, spectrophotometry (e.g., UV-visible), or mass spectrometry, or by chromatography such as high performance liquid chromatography (HPLC) or thin layer chromatography.
[00295] Preparation of modified nucleic acid and mmRNA molecules of the present invention can involve the protection and deprotection of various chemical groups. The need for protection and deprotection, and the selection of appropriate protecting groups can be readily determined by one skilled in the art. The chemistry of protecting groups can be found, for example, in Greene, et al., Protective Groups in Organic Synthesis, 2d. Ed., Wiley & Sons, 1991, which is incorporated herein by reference in its entirety.
[00296] The reactions of the processes described herein can be carried out in suitable solvents, which can be readily selected by one of skill in the art of organic synthesis. Suitable solvents can be substantially nonreactive with the starting materials (reactants), the intermediates, or products at the temperatures at which the reactions are carried out, i.e., temperatures which can range from the solvent's freezing temperature to the solvent's boiling temperature. A given reaction can be carried out in one solvent or a mixture of more than one solvent. Depending on the particular reaction step, suitable solvents for a particular reaction step can be selected.
[00297] Resolution of racemic mixtures of modified nucleosides and nucleotides can be carried out by any of numerous methods known in the art. An example method includes fractional
recrystallization using a "chiral resolving acid" which is an optically active, salt-forming organic acid. Suitable resolving agents for fractional recrystallization methods are, for example, optically active acids, such as the D and L forms of tartaric acid, diacetyltartaric acid, dibenzoyltartaric acid, mandelic acid, malic acid, lactic acid or the various optically active camphorsulfonic acids.
Resolution of racemic mixtures can also be carried out by elution on a column packed with an optically active resolving agent (e.g., dinitrobenzoylphenylglycine). Suitable elution solvent composition can be determined by one skilled in the art.
[00298] Modified nucleosides and nucleotides (e.g., binding block molecules) can be prepared according to the synthetic methods described in Ogata et al, J. Org. Chem. 74:2585-2588 (2009); Purmal et al, Nucl. Acids Res. 22(1): 72-78, (1994); Fukuhara et al, Biochemistry, 1(4): 563-568 (1962); and Xu et al, Tetrahedron, 48(9): 1729-1740 (1992), each of which are incorporated by reference in their entirety.
[00299] The modified nucleic acid and mmRNA of the invention need not be uniformly modified along the entire length of the molecule. For example, one or more or all types of nucleotide (e.g., purine or pyrimidine, or any one or more or all of A, G, U, C) may or may not be uniformly modified in a polynucleotide of the invention, or in a given predetermined sequence region thereof. In some embodiments, all nucleotides X in a polynucleotide of the invention (or in a given sequence region thereof) are modified, wherein X may any one of nucleotides A, G, U, C, or any one of the combinations A+G, A+U, A+C, G+U, G+C, U+C, A+G+U, A+G+C, G+U+C or A+G+C
[00300] Different sugar modifications, nucleotide modifications, and/or internucleoside linkages (e.g., backbone structures) may exist at various positions in the modified nucleic acid or mmRNA. One of ordinary skill in the art will appreciate that the nucleotide analogs or other modification(s) may be located at any position(s) of a modified nucleic acid or mmRNA such that the function of the modified nucleic acid or mmRNA is not substantially decreased. A modification may also be a 5 ' or 3 ' terminal modification. The modified nucleic acid or mmRNA may contain from about 1 % to about 100% modified nucleotides, or any intervening percentage (e.g., from 1% to 20%>, from 1% to 25%, from 1% to 50%, from 1% to 60%, from 1% to 70%, from 1% to 80%, from 1% to 90%, from 1% to 95%, from 10% to 20%, from 10% to 25%, from 10% to 50%, from 10% to 60%, from 10% to 70%, from 10% to 80%, from 10% to 90%, from 10% to 95%, from 10% to 100%, from 20% to 25%, from 20% to 50%, from 20% to 60%, from 20% to 70%, from 20% to 80%, from 20% to 90%, from 20% to 95%, from 20% to 100%, from 50% to 60%, from 50% to 70%, from 50% to 80%, from 50% to 90%, from 50% to 95%, from 50% to 100%, from 70% to 80%, from 70% to 90%, from 70% to 95%, from 70% to 100%, from 80% to 90%, from 80% to 95%, from 80% to 100%, from 90% to 95%, from 90% to 100%, and from 95% to 100%).
[00301] In some embodiments, the modified nucleic acid or mmR A includes a modified pyrimidine (e.g., a modified uracil/uridine or modified cytosine/cytidine). In some embodiments, the uracil or uridine in the modified nucleic acid or mmRNA molecule may be replaced with from about P/o to about 100% of a modified uracil or modified uridine (e.g., from 1% to 20%>, from 1% to 25%, from 1% to 50%, from 1% to 60%, from 1% to 70%, from 1% to 80%, from 1% to 90%, from 1% to 95%, from 10% to 20%, from 10% to 25%, from 10% to 50%, from 10% to 60%, from 10% to 70%, from 10% to 80%, from 10% to 90%, from 10% to 95%, from 10% to 100%, from 20% to 25%, from 20% to 50%, from 20% to 60%, from 20% to 70%, from 20% to 80%, from 20% to 90%, from 20% to 95%, from 20% to 100%, from 50% to 60%, from 50% to 70%, from 50% to 80%, from 50% to 90%, from 50% to 95%, from 50% to 100%, from 70% to 80%, from 70% to 90%, from 70% to 95%, from 70% to 100%, from 80% to 90%, from 80% to 95%, from 80% to 100%, from 90% to 95%, from 90% to 100%, and from 95% to 100% of a modified uracil or modified uridine). The modified uracil or uridine can be replaced by a compound having a single unique structure or by a plurality of compounds having different structures (e.g., 2, 3, 4 or more unique structures, as described herein). In some embodiments, the cytosine or cytidine in the modified nucleic acid or mmRNA molecule may be replaced with from about 1% to about 100% of a modified cytosine or modified cytidine (e.g., from 1% to 20%>, from 1% to 25%, from 1% to 50%, from 1% to 60%, from 1% to 70%, from 1% to 80%, from 1% to 90%, from 1% to 95%, from 10% to 20%, from 10% to 25%, from 10% to 50%, from 10% to 60%, from 10% to 70%, from 10% to 80%, from 10% to 90%, from 10% to 95%, from 10% to 100%, from 20% to 25%, from 20% to 50%, from 20% to 60%, from 20% to 70%, from 20% to 80%, from 20% to 90%, from 20% to 95%, from 20% to 100%, from 50% to 60%, from 50% to 70%, from 50% to 80%, from 50% to 90%, from 50% to 95%, from 50% to 100%, from 70% to 80%, from 70% to 90%, from 70% to 95%, from 70% to 100%, from 80% to 90%, from 80% to 95%, from 80% to 100%, from 90% to 95%, from 90% to 100%, and from 95% to 100% of a modified cytosine or modified cytidine). The modified cytosine or cytidine can be replaced by a compound having a single unique structure or by a plurality of compounds having different structures (e.g., 2, 3, 4 or more unique structures, as described herein). [00302] In some embodiments, the present disclosure provides methods of synthesizing a modified nucleic a id or mmRNA including n number of linked nucleosides having Formula (Ia-1):
Figure imgf000106_0001
(Ia-1), comprising:
[00303] a) reacting a nucleotide of Formula (IV-1):
Figure imgf000106_0002
(IV-1),
[00304] with a phosphoramidite compound of Formula (V-l):
Figure imgf000106_0003
),
[00305] wherein Y9 is H, hydroxy, phosphoryl, pyrophosphate, sulfate, amino, thiol, optionally substituted amino acid, or a peptide (e.g., including from 2 to 12 amino acids); and each P1, P2, and P3 is, independently, a suitable protecting group; and @ denotes a solid support;
[00306] to provide a modified nucleic acid or mmRNA of Formula (VI-1):
Figure imgf000107_0001
[00307] b) oxidizing or sulfurizing the modified nucleic acid or mmRNA of Formula (V) to yield a modified nucleic acid or mmRNA of Formula (VII- 1):
Figure imgf000107_0002
[00308] c) removing the protecting groups to yield the modified nucleic acid or mmRNA of Formula (la).
[00309] In some embodiments, steps a) and b) are repeated from 1 to about 10,000 times. In some embodiments, the methods further comprise a nucleotide (e.g., building block molecule) selected from the group consisting of adenosine, cytosine, guanosine, and uracil. In some embodiments, the nucleobase may be a pyrimidine or derivative thereof. In some embodiments, the modified nucleic acid or mmRNA is translatable.
[00310] Other components of modified nucleic acids and mmRNA are optional, and are beneficial in some embodiments. For example, a 5' untranslated region (UTR) and/or a 3'UTR are provided, wherein either or both may independently contain one or more different nucleoside modifications. In such embodiments, nucleoside modifications may also be present in the translatable region. Also provided are modified nucleic acids and mmRNA containing a Kozak sequence. [00311] Exemplary syntheses of modified nucleotides, which are incorporated into a modified nucleic acid or mmRNA, e.g., RNA or mRNA, are provided below in Scheme 1 through Scheme 11. Scheme 1 provides a general method for phosphorylation of nucleosides, including modified nucleosides.
Scheme 1
Figure imgf000108_0001
[00312] Various protecting groups may be used to control the reaction. For example, Scheme 2 provides the use of multiple protecting and deprotecting steps to promote phosphorylation at the 5' position of the sugar, rather than the 2' and 3' hydroxyl groups.
Scheme 2
Figure imgf000109_0001
[00313] Modified nucleotides can be synthesized in any useful manner. Schemes 3, 4, and 7 provide exemplary methods for synthesizing modified nucleotides having a modified purine nucleobase; and Schemes 5 and 6 provide exemplary methods for synthesizing modified nucleotides having a modified pseudouridine or pseudoisocytidine, respectively.
Figure imgf000110_0001
Scheme 5
Figure imgf000111_0001
- 109- Scheme 6
Figure imgf000112_0001
1) P0C13
2) Pyrophosphate
Figure imgf000112_0002
Figure imgf000113_0001
[00314] Schemes 8 and 9 provide exemplary syntheses of modified nucleotides. Scheme 10 provides a non-limiting biocatalytic method for producing nucleotides.
- I l l - Scheme 8
Figure imgf000114_0001
Figure imgf000114_0002
Scheme 9
Figure imgf000115_0001
1 ) H"
2) "OH, heat
Figure imgf000115_0002
[00315] Scheme 11 provides an exemplary synthesis of a modified uracil, where the Nl position is modified with R12b, as provided elsewhere, and the 5'-position of ribose is phosphorylated. T1, T2, R12a, R12b, and r are as provided herein. This synthesis, as well as optimized versions thereof, can be used to modify other pyrimidine nucleobases and purine nucleobases (see e.g., Formulas (bl)- (b43)) and/or to install one or more phosphate groups (e.g., at the 5' position of the sugar). This alkylating reaction can also be used to include one or more optionally substituted alkyl group at any reactive group (e.g., amino group) in any nucleobase described herein (e.g., the amino groups in the Watson-Crick base-pairing face for cytosine, uracil, adenine, and guanine).
Scheme 11
Figure imgf000116_0001
Combinations of Nucleotides in mmRNA
[00316] Further examples of modified nucleotides and modified nucleotide combinations are provided below in Table 2. These combinations of modified nucleotides can be used to form the modified nucleic acids or mmRNA of the invention. Unless otherwise noted, the modified nucleotides may be completely substituted for the natural nucleotides of the modified nucleic acids or mmRNA of the invention. As a non-limiting example, the natural nucleotide uridine may be substituted with a modified nucleoside described herein. In another non-limiting example, the natural nucleotide uridine may be partially substituted (e.g., about 0.1%, 1%, 5%, 10%>, 15%, 20%>, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 99.9%) with at least one of the modified nucleoside disclosed herein.
Table 2
Figure imgf000116_0002
pyrrolo-cytidine/pseudouridine
about 50% of the cytosines are pyrrolo-cytidine
5 -methyl-cytidine 5 -methyl-cytidine/ 5 -iodo-uridine
5-methyl-cytidine/N 1 -methyl-pseudouridine
5-methyl-cytidine/a-thio-uridine
5 -methyl-cytidine/ 5 -methyl-uridine
5-methyl-cytidine/pseudouridine
about 25% of cytosines are 5 -methyl-cytidine
about 50% of cytosines are 5 -methyl-cytidine
5 -methyl-cytidine/ 5 -methoxy-uridine
5-methyl-cytidine/5-bromo-uridine
5-methyl-cytidine/2-thio-uridine
5-methyl-cytidine/about 50% of uridines are 2-thio-uridine
about 50% of uridines are 5 -methyl-cytidine/ about 50% of uridines are 2-thio- uridine
N4-acetyl-cytidine N4-acetyl-cytidine /5-iodo-uridine
N4-acetyl-cytidine /N 1 -methyl-pseudouridine
N4-acetyl-cytidine / -thio-uridine
N4-acetyl-cytidine 15 -methyl-uridine
N4-acetyl-cytidine /pseudouridine
about 50% of cytosines are N4-acetyl-cytidine
about 25% of cytosines are N4-acetyl-cytidine
N4-acetyl-cytidine 15 -methoxy-uridine
N4-acetyl-cytidine /5-bromo-uridine
N4-acetyl-cytidine /2-thio-uridine
about 50% of cytosines are N4-acetyl-cytidine/ about 50% of uridines are 2- thio-uridine
[00317] Further examples of modified nucleotide combinations are provided below in Table 3. These combinations of modified nucleotides can be used to form the modified nucleic acid molecules or mmRNA of the invention.
Table 3
Figure imgf000117_0001
modified cytidine with (b32)/2-thio-uridine
about 50% of cytidine substituted with modified cytidine (b32)/ about 50% of uridines are 2-thio-uridine
modified uridine modified uridine with (bl)/ N4-acetyl-cytidine
having one or more modified uridine with (bl)/ 5-methyl-cytidine
nucleobases of
Formula (bl)
modified uridine modified uridine with (b8)/ N4-acetyl-cytidine
having one or more modified uridine with (b8)/ 5-methyl-cytidine
nucleobases of
Formula (b8)
modified uridine modified uridine with (b28)/ N4-acetyl-cytidine
having one or more modified uridine with (b28)/ 5-methyl-cytidine
nucleobases of
Formula (b28)
modified uridine modified uridine with (b29)/ N4-acetyl-cytidine
having one or more modified uridine with (b29)/ 5-methyl-cytidine
nucleobases of
Formula (b29)
modified uridine modified uridine with (b30)/ N4-acetyl-cytidine
having one or more modified uridine with (b30)/ 5-methyl-cytidine
nucleobases of
Formula (b30)
[00318] In some embodiments, at least 25% of the cytosines are replaced by a compound of Formula (blO)-(M4) (e.g., at least about 30%>, at least about 35%>, at least about 40%>, at least about 45%), at least about 50%>, at least about 55%>, at least about 60%>, at least about 65%>, at least about 70%), at least about 75%>, at least about 80%>, at least about 85%>, at least about 90%>, at least about 95%, or about 100%).
[00319] In some embodiments, at least 25% of the uracils are replaced by a compound of Formula (bl)-(b9) (e.g., at least about 30%>, at least about 35%>, at least about 40%>, at least about 45%>, at least about 50%), at least about 55%>, at least about 60%>, at least about 65%>, at least about 70%>, at least about 75%), at least about 80%>, at least about 85%>, at least about 90%>, at least about 95%>, or about 100%).
[00320] In some embodiments, at least 25%> of the cytosines are replaced by a compound of Formula (bl0)-(bl4), and at least 25% of the uracils are replaced by a compound of Formula (bl)- (b9) (e.g., at least about 30%>, at least about 35%>, at least about 40%>, at least about 45%>, at least about 50%), at least about 55%>, at least about 60%>, at least about 65%>, at least about 70%>, at least about 75%), at least about 80%>, at least about 85%>, at least about 90%>, at least about 95%>, or about 100%). Synthesis of Modified Nucleic Acid Molecules
[00321] Modified nucleic acid molecules for use in accordance with the present disclosure may be prepared according to any available technique including, but not limited to, in vitro transcription such as chemical synthesis and enzymatic synthesis, or enzymatic and chemical cleavage of a longer precursor, etc. Methods of synthesizing RNA are known in the art (see, e.g., Gait, M.J. (ed.) Oligonucleotide synthesis: a practical approach, Oxford [Oxfordshire], Washington, DC: IRL Press, 1984; and Herdewijn, P. (ed.) Oligonucleotide synthesis: methods and applications, Methods in Molecular Biology, v. 288 (Clifton, N.J.) Totowa, N.J.: Humana Press, 2005; both of which are incorporated herein by reference).
[00322] The modified nucleic acid molecules disclosed herein can be prepared from readily available starting materials using the following general methods and procedures. It is understood that where typical or preferred process conditions (i.e., reaction temperatures, times, mole ratios of reactants, solvents, pressures, etc.) are given other process conditions can also be used unless otherwise stated. Optimum reaction conditions may vary with the particular reactants or solvent used, but such conditions can be determined by one skilled in the art by routine optimization procedures.
[00323] The processes described herein can be monitored according to any suitable method known in the art. For example, product formation can be monitored by spectroscopic means, such as nuclear magnetic resonance spectroscopy (e.g., 1H or 13C) infrared spectroscopy, spectrophotometry (e.g., UV-visible), mass spectrometry, or by chromatography such as high performance liquid chromatography (HPLC) or thin layer chromatography.
[00324] Preparation of modified nucleic acid molecules can involve the protection and deprotection of various chemical groups. The need for protection and deprotection, and the selection of appropriate protecting groups can be readily determined by one skilled in the art. The chemistry of protecting groups can be found, for example, in Greene, et al, Protective Groups in Organic
Synthesis, 2d. Ed., Wiley & Sons, 1991, which is incorporated herein by reference in its entirety.
[00325] The reactions of the processes described herein can be carried out in suitable solvents, which can be readily selected by one of skill in the art of organic synthesis. Suitable solvents can be substantially nonreactive with the starting materials (reactants), the intermediates, or products at the temperatures at which the reactions are carried out, i.e., temperatures which can range from the solvent's freezing temperature to the solvent's boiling temperature. A given reaction can be carried out in one solvent or a mixture of more than one solvent. Depending on the particular reaction step, suitable solvents for a particular reaction step can be selected.
[00326] Resolution of racemic mixtures of modified nucleic acid molecules can be carried out by any of numerous methods known in the art. An example method includes, but is not limited to, fractional recrystallization using a "chiral resolving acid" which is an optically active, salt-forming organic acid. Suitable resolving agents for fractional recrystallization methods are, for example, optically active acids, such as the D and L forms of tartaric acid, diacetyltartaric acid,
dibenzoyltartaric acid, mandelic acid, malic acid, lactic acid or the various optically active camphorsulfonic acids. Resolution of racemic mixtures can also be carried out by elution on a column packed with an optically active resolving agent (e.g., dinitrobenzoylphenylglycine). Suitable elution solvent composition can be determined by one skilled in the art.
[00327] Modified nucleic acid molecules need not be uniformly modified along the entire length of the molecule. Different nucleic acid modifications and/or backbone structures may exist at various positions in the nucleic acid. One of ordinary skill in the art will appreciate that the nucleotide analogs or other modification(s) may be located at any position(s) of a nucleic acid such that the function of the nucleic acid is not substantially decreased. A modification may also be a 5 Or 3' terminal modification. The nucleic acids may contain at a minimum one modified nucleotide and at maximum 100% modified nucleotides, or any intervening percentage, such as at least 5% modified nucleotides, at least 10% modified nucleotides, at least 25% modified nucleotides, at least 50% modified nucleotides, at least 80% modified nucleotides, or at least 90% modified nucleotides. For example, the nucleic acids may contain a modified pyrimidine such as uracil or cytosine. In some embodiments, at least 5%, at least 10%>, at least 25%, at least 50%, at least 80%, at least 90% or 100% of the uracil in the nucleic acid may be replaced with a modified uracil. The modified uracil can be replaced by a compound having a single unique structure, or can be replaced by a plurality of compounds having different structures (e.g., 2, 3, 4 or more unique structures). In some
embodiments, at least 5%, at least 10%, at least 25%, at least 50%, at least 80%, at least 90% or 100% of the cytosine in the nucleic acid may be replaced with a modified cytosine. The modified cytosine can be replaced by a compound having a single unique structure, or can be replaced by a plurality of compounds having different structures (e.g., 2, 3, 4 or more unique structures).
[00328] Generally, the shortest length of a modified mRNA, herein "mmRNA," of the present disclosure can be the length of an mRNA sequence that may be sufficient to encode for a dipeptide. In another embodiment, the length of the mRNA sequence may be sufficient to encode for a tripeptide. In another embodiment, the length of an mRNA sequence may be sufficient to encode for a tetrapeptide. In another embodiment, the length of an mRNA sequence may be sufficient to encode for a pentapeptide. In another embodiment, the length of an mRNA sequence may be sufficient to encode for a hexapeptide. In another embodiment, the length of an mRNA sequence may be sufficient to encode for a heptapeptide. In another embodiment, the length of an mRNA sequence may be sufficient to encode for an octapeptide. In another embodiment, the length of an mRNA sequence may be sufficient to encode for a nonapeptide. In another embodiment, the length of an mRNA sequence may be sufficient to encode for a decapeptide.
[00329] Examples of dipeptides that the modified nucleic acid molecule sequences can encode for include, but are not limited to, carnosine and anserine.
[00330] In a further embodiment, the mRNA may be greater than 30 nucleotides in length. In another embodiment, the RNA molecule may be greater than 35 nucleotides in length (e.g., at least or greater than about 35, 40, 45, 50, 55, 60, 70, 80, 90, 100, 120, 140, 160, 180, 200, 250, 300, 350, 400, 450, 500, 600, 700, 800, 900, 1,000, 1,100, 1,200, 1,300, 1,400, 1,500, 1,600, 1,700, 1,800, 1,900, 2,000, 2,500, 3,000, 4,000 and 5,000 nucleotides).
Exemplary Properties of Modified Nucleic Acid Molecules
Major Groove Interacting Partners
[00331] The modified nucleic acid molecules, e.g., modified mRNA (mmRNA), described herein can disrupt interactions with recognition receptors that detect and respond to RNA ligands through interactions, e.g. binding, with the major groove face of a nucleotide or nucleic acid. As such, RNA ligands comprising modified nucleotides or modified nucleic acid molecules, as described herein, decrease interactions with major groove binding partners, and therefore decrease an innate immune response, or expression and secretion of pro-inflammatory cytokines, or both.
[00332] Example major groove interacting, e.g. binding, partners include, but are not limited to, the following nucleases and helicases. Within membranes, TLRs (Toll-like Receptors) 3, 7, and 8 can respond to single- and double-stranded RNA. Within the cytoplasm, members of the superfamily 2 class of DE (D/H) helicases and ATPases can sense RNA to initiate antiviral responses. These helicases include the RIG-I (retinoic acid-inducible gene I) and MDA5 (melanoma differentiation- associated gene 5). Other examples include laboratory of genetics and physiology 2 (LGP2), HIN- 200 domain containing proteins, or Helicase-domain containing proteins. Prevention or Reduction of Innate Cellular Immune Response Activation Using Modified Nucleic Acid Molecules
[00333] The modified nucleic acid molecules, e.g., mmRNA, described herein, decrease the innate immune response in a cell. The term "innate immune response" includes a cellular response to exogenous nucleic acids, including, but not limited to, single stranded nucleic acids, generally of viral or bacterial origin, which involve the induction of cytokine expression and release, particularly the interferons, and cell death. Protein synthesis may also be reduced during the innate cellular immune response. While it is advantageous to eliminate the innate immune response in a cell, the present disclosure provides modified mRNA that substantially reduce the immune response, including interferon signaling, without entirely eliminating such a response. In some embodiments, the immune response may be reduced by 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99%), 99.9%), or greater than 99.9% as compared to the immune response induced by a corresponding unmodified nucleic acid molecule. Such a reduction can be measured by the expression or activity level of Type 1 interferons or the expression of interferon-regulated genes such as the toll-like receptors {e.g., TLR7 and TLR8). Reduction of the innate immune response can also be measured by decreased cell death following one or more administrations of modified RNA to a cell population; e.g., cell death is 10%, 25%, 50%, 75%, 85%, 90%, 95%, or over 95% less than the cell death frequency observed with a corresponding unmodified nucleic acid molecule. Moreover, cell death may affect fewer than 50%, 40%, 30%, 20%, 10%, 5%, 1%, 0.1%, 0.01% or fewer than 0.01% of cells contacted with the modified nucleic acid molecules.
[00334] The present disclosure provides for the repeated introduction {e.g. , transfection) of modified nucleic acid molecules into a target cell population, e.g., in vitro, ex vivo, or in vivo. The step of contacting the cell population may be repeated one or more times (such as two, three, four, five or more than five times). In some embodiments, the step of contacting the cell population with the modified nucleic acid molecules may be repeated a number of times sufficient such that a predetermined efficiency of protein translation in the cell population is achieved. Given the reduced cytotoxicity of the target cell population by the nucleic acid modifications, such repeated
transfections are achievable in a variety of cell types.
[00335] The modified nucleic acids of the invention, including the combination of modifications taught herein may have superior properties making them more suitable as therapeutic modalities. [00336] It has been determined that the "all or none" model in the art is sorely insufficient to describe the biological phenomena associated with the therapeutic utility of modified mRNA. The present inventors have determined that to improve protein production, one may consider the nature of the modification, or combination of modifications, the percent modification and survey more than one cytokine or metric to determine the efficacy and risk profile of a particular modified mRNA.
[00337] In one aspect of the invention, methods of determining the effectiveness of a modified mRNA as compared to unmodified involves the measure and analysis of one or more cytokines whose expression is triggered by the administration of the exogenous nucleic acid of the invention. These values are compared to administration of an umodified nucleic acid or to a standard metric such as cytokine response, PolylC, R-848 or other standard known in the art.
[00338] One example of a standard metric developed herein is the measure of the ratio of the level or amount of encoded polypeptide (protein) produced in the cell, tissue or organism to the level or amount of one or more (or a panel) of cytokines whose expression is triggered in the cell, tissue or organism as a result of administration or contact with the modified nucleic acid. Such ratios are referred to herein as the Protein: Cytokine Ratio or "PC" Ratio. The higher the PC ratio, the more efficacioius the modified nucleic acid (polynucleotide encoding the protein measured). Preferred PC Ratios, by cytokine, of the present invention may be greater than 1, greater than 10, greater than 100, greater than 1000, greater than 10,000 or more. Modified nucleic acids having higher PC Ratios than a modified nucleic acid of a different or unmodified construct are preferred.
[00339] The PC ratio may be further qualified by the percent modification present in the polynucleotide. For example, normalized to a 100% modified nucleic acid, the protein production as a function of cytokine (or risk) or cytokine profile can be determined.
[00340] In one embodiment, the present invention provides a method for determining, across chemistries, cytokines or percent modification, the relative efficacy of any particular modified polynucleotide by comparing the PC Ratio of the modified nucleic acid (polynucleotide).
Activation of the immune response: Vaccines
[00341] In one embodiment of the present invention, mRNA molecules may be used to elicit or provoke an immune response in an organism. The mRNA molecules to be delivered may encode an immunogenic peptide or polypeptide and may encode more than one such peptide or polypeptide.
[00342] Additionally, certain modified nucleosides, or combinations thereof, when introduced into the modified nucleic acid molecules or mmRNA of the invention will activate the innate immune response. Such activating molecules are useful as adjuvants when combined with polypeptides and/or other vaccines. In certain embodiments, the activating molecules contain a translatable region which encodes for a polypeptide sequence useful as a vaccine, thus providing the ability to be a self-adjuvant.
[00343] In one embodiment, the modified nucleic acid molecules and/or mmRNA of the invention may encode an immunogen. The delivery of modified nucleic acid molecules and/or mmRNA encoding an immunogen may activate the immune response. As a non-limiting example, the modified nucleic acid molecules and/or mmRNA encoding an immunogen may be delivered to cells to trigger multiple innate response pathways (see International Pub. No. WO2012006377; herein incorporated by reference in its entirety). As another non-limiting example, the modified nucleic acid molecules and mmRNA of the present invention encoding an immunogen may be delivered to a vertebrate in a dose amount large enough to be immunogenic to the vertebrate (see International Pub. No. WO2012006372 and WO2012006369; each of which is herein incorporated by reference in their entirety).
[00344] The modified nucleic acid molecules or mmRNA of invention may encode a polypeptide sequence for a vaccine and may further comprise an inhibitor. The inhibitor may impair antigen presentation and/or inhibit various pathways known in the art. As a non-limiting example, the modified nucleic acid molecules or mmRNA of the invention may be used for a vaccine in combination with an inhibitor which can impair antigen presentation (see International Pub. No. WO2012089225 and WO2012089338; each of which is herein incorporated by reference in their entirety).
[00345] In one embodiment, the modified nucleic acid molecules or mmRNA of the invention may be self-replicating RNA. Self-replicating RNA molecules can enhance efficiency of RNA delivery and expression of the enclosed gene product. In one embodiment, the modified nucleic acid molecules or mmRNA may comprise at least one modification described herein and/or known in the art. In one embodiment, the self-replicating RNA can be designed so that the self-replicating RNA does not induce production of infectious viral particles. As a non- limiting example the self- replicating RNA may be designed by the methods described in US Pub. No. US20110300205 and International Pub. No. WO2011005799, each of which is herein incorporated by reference in their entirety. [00346] In one embodiment, the self-replicating modified nucleic acid molecules or mmRNA of the invention may encode a protein which may raise the immune response. As a non-limiting example, the modified nucleic acid molecules and/or mmRNA may be self-replicating mRNA may encode at least one antigen (see US Pub. No. US20110300205 and International Pub. No. WO2011005799; each of which is herein incorporated by reference in their entirety).
[00347] In one embodiment, the self-replicating modified nucleic acids or mmRNA of the invention may be formulated using methods described herein or known in the art. As a non-limiting example, the self-replicating RNA may be formulated for delivery by the methods described in Geall et al (Nonviral delivery of self-amplifying RNA vaccines, PNAS 2012; PMID: 22908294; herein incorporated by reference in its entirety ).
[00348] In one embodiment, the modified nucleic acid molecules or mmRNA of the present invention may encode amphipathic and/or immunogenic amphipathic peptides.
[00349] In on embodiment, a formulation of the modified nucleic acid molecules or mmRNA of the present invention may further comprise an amphipathic and/or immunogenic amphipathic peptide. As a non-limiting example, the modified nucleic acid molecule or mmRNA comprising an amphipathic and/or immunogenic amphipathic peptide may be formulated as described in US. Pub. No. US20110250237 and International Pub. Nos. WO2010009277 and WO2010009065; each of which is herein incorporated by reference in their entirety.
[00350] In one embodiment, the modified nucleic acid molecules and mmRNA of the present invention may be immunostimultory. As a non-limiting example, the modified nucleic acid molecules and mmRNA may encode all or a part of a positive-sense or a negative-sense stranded RNA virus genome (see International Pub No. WO2012092569 and US Pub No. US20120177701, each of which is herein incorporated by reference in their entirety). In another non-limiting example, the immunostimultory modified nucleic acid molecules or mmRNA of the present invention may be formulated with an excipient for administration as described herein and/or known in the art (see International Pub No. WO2012068295 and US Pub No. US20120213812, each of which is herein incorporated by reference in their entirety).
[00351] In one embodiment, the response of the vaccine formulated by the methods described herein may be enhanced by the addition of various compounds to induce the therapeutic effect. As a non-limiting example, the vaccine formulation may include a MHC II binding peptide or a peptide having a similar sequence to a MHC II binding peptide (see International Pub Nos. WO2012027365, WO201 1031298 and US Pub No. US20120070493, US201 101 10965, each of which is herein incorporated by reference in their entirety). As another example, the vaccine formulations may comprise modified nicotinic compounds which may generate an antibody response to nicotine residue in a subject (see International Pub No. WO2012061717 and US Pub No. US201201 14677, each of which is herein incorporated by reference in their entirety).
Polypeptide variants
[00352] The modified nucleic acid molecules encode polypeptides, e.g., a variant polypeptides, which have a certain identity to a reference polypeptide sequence. The term "identity," as known in the art, refers to a relationship between the sequences of two or more peptides, determined by comparing the sequences. In the art, "identity" also refers to the degree of sequence relatedness between peptides, as determined by the number of matches between strings of two or more amino acid residues. Identity measures the percent of identical matches between the smaller of two or more sequences with gap alignments (if any) addressed by a particular mathematical model or computer program (i.e., "algorithms"). Identity of related peptides can be readily calculated by known methods. Such methods include, but are not limited to, those described in Computational Molecular Biology, Lesk, A. M., ed., Oxford University Press, New York, 1988; Biocomputing: Informatics and Genome Projects, Smith, D. W., ed., Academic Press, New York, 1993; Computer Analysis of Sequence Data, Part 1 , Griffin, A. M., and Griffin, H. G., eds., Humana Press, New Jersey, 1994; Sequence Analysis in Molecular Biology, von Heinje, G., Academic Press, 1987; Sequence Analysis Primer, Gribskov, M. and Devereux, J., eds., M. Stockton Press, New York, 1991 ; and Carillo et al, SIAM J. Applied Math. 48, 1073 (1988); all of which are herein incorporated by reference in their entirety.
[00353] In some embodiments, the polypeptide variant may have the same or a similar activity as the reference polypeptide. Alternatively, the variant may have an altered activity (e.g., increased or decreased) relative to a reference polypeptide. Generally, variants of a particular polynucleotide or polypeptide of the present disclosure will have at least about 40%, 45%, 50%>, 55%>, 60%>, 65%>, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity to that particular reference polynucleotide or polypeptide as determined by sequence alignment programs and parameters described herein and known to those skilled in the art.
[00354] As recognized by those skilled in the art, protein fragments, functional protein domains, and homologous proteins are also considered to be within the scope of this present disclosure. For example, provided herein is any protein fragment of a reference protein (meaning a polypeptide sequence which is at least one amino acid residue shorter than a reference polypeptide sequence but otherwise identical) 10, 20, 30, 40, 50, 60, 70, 80, 90, 100 or greater than 100 amino acids in length. In another example, any protein that includes a stretch of about 20, about 30, about 40, about 50, or about 100 amino acids which are about 40%, about 50%, about 60%>, about 70%>, about 80%>, about 90%), about 95%o, or about 100% identical to any of the sequences described herein can be utilized in accordance with the present disclosure. In certain embodiments, a protein sequence to be utilized in accordance with the present disclosure includes 2, 3, 4, 5, 6, 7, 8, 9, 10, or more mutations as shown in any of the sequences provided or referenced herein.
Polvpeptide-nucleic acid complexes
[00355] Proper protein translation involves the physical aggregation of a number of polypeptides and nucleic acids associated with the mRNA. Provided by the present disclosure are protein-nucleic acid complexes, containing a translatable mRNA having one or more nucleoside modifications (e.g., at least two different nucleoside modifications) and one or more polypeptides bound to the mRNA. Generally, the proteins are provided in an amount effective to prevent or to reduce an innate immune response of a cell into which the complex is introduced.
Untranslatable Modified Nucleic Acid Molecules
[00356] As described herein, provided are mRNA having sequences that are substantially not translatable. Such mRNA may be effective as a vaccine when administered to a subject. It is further provided that the subject administered the vaccine may be a mammal, more preferably a human and most preferably a patient.
[00357] Also provided are modified nucleic acid molecules that contain one or more noncoding regions. Such modified nucleic acid molecules are generally not translated, but are capable of binding to and sequestering one or more translational machinery component such as a ribosomal protein or a transfer RNA (tRNA), thereby effectively reducing the protein expression in the cell. The modified nucleic acid molecule may contain a small nucleolar RNA (sno-RNA), micro RNA (miRNA), small interfering RNA (siRNA) or Piwi-interacting RNA (piRNA).
Pharmaceutical Compositions
Formulation, Administration, Delivery and Dosing
[00358] The present invention provides modified nucleic acids and mmRNA compositions and complexes in combination with one or more pharmaceutically acceptable excipients. Pharmaceutical compositions may optionally comprise one or more additional active substances, e.g. therapeutically and/or prophylactically active substances. General considerations in the formulation and/or manufacture of pharmaceutical agents may be found, for example, in Remington: The Science and Practice of Pharmacy 21st ed., Lippincott Williams & Wilkins, 2005 (incorporated herein by reference in its entirety).
[00359] In some embodiments, compositions are administered to humans, human patients or subjects. For the purposes of the present disclosure, the phrase "active ingredient" generally refers to modified nucleic acids and mmRNA to be delivered as described herein.
[00360] Although the descriptions of pharmaceutical compositions provided herein are principally directed to pharmaceutical compositions which are suitable for administration to humans, it will be understood by the skilled artisan that such compositions are generally suitable for administration to any other animal, e.g., to non-human animals, e.g. non-human mammals. Modification of pharmaceutical compositions suitable for administration to humans in order to render the compositions suitable for administration to various animals is well understood, and the ordinarily skilled veterinary pharmacologist can design and/or perform such modification with merely ordinary, if any, experimentation. Subjects to which administration of the pharmaceutical compositions is contemplated include, but are not limited to, humans and/or other primates;
mammals, including commercially relevant mammals such as cattle, pigs, horses, sheep, cats, dogs, mice, and/or rats; and/or birds, including commercially relevant birds such as poultry, chickens, ducks, geese, and/or turkeys.
[00361] Formulations of the pharmaceutical compositions described herein may be prepared by any method known or hereafter developed in the art of pharmacology. In general, such preparatory methods include the step of bringing the active ingredient into association with an excipient and/or one or more other accessory ingredients, and then, if necessary and/or desirable, dividing, shaping and/or packaging the product into a desired single- or multi-dose unit.
[00362] A pharmaceutical composition in accordance with the invention may be prepared, packaged, and/or sold in bulk, as a single unit dose, and/or as a plurality of single unit doses. As used herein, a "unit dose" is discrete amount of the pharmaceutical composition comprising a predetermined amount of the active ingredient. The amount of the active ingredient is generally equal to the dosage of the active ingredient which would be administered to a subject and/or a convenient fraction of such a dosage such as, for example, one-half or one -third of such a dosage. [00363] Relative amounts of the active ingredient, the pharmaceutically acceptable excipient, and/or any additional ingredients in a pharmaceutical composition in accordance with the invention will vary, depending upon the identity, size, and/or condition of the subject treated and further depending upon the route by which the composition is to be administered. By way of example, the composition may comprise between 0.1% and 100%, e.g., between .5 and 50%>, between 1-30%, between 5-80%>, at least 80%> (w/w) active ingredient.
Formulations
[00364] The modified nucleic acid, and mmRNA of the invention can be formulated using one or more excipients to: (1) increase stability; (2) increase cell transfection; (3) permit the sustained or delayed release (e.g., from a depot formulation of the modified nucleic acid, or mmRNA); (4) alter the biodistribution (e.g., target the modified nucleic acid, or mmRNA to specific tissues or cell types); (5) increase the translation of encoded protein in vivo; and/or (6) alter the release profile of encoded protein in vivo. In addition to traditional excipients such as any and all solvents, dispersion media, diluents, or other liquid vehicles, dispersion or suspension aids, surface active agents, isotonic agents, thickening or emulsifying agents, preservatives, excipients of the present invention can include, without limitation, lipidoids, liposomes, lipid nanoparticles, polymers, lipoplexes, core- shell nanoparticles, peptides, proteins, cells transfected with modified nucleic acid, or mmRNA (e.g., for transplantation into a subject), hyaluronidase, nanoparticle mimics and combinations thereof. Accordingly, the formulations of the invention can include one or more excipients, each in an amount that together increases the stability of the modified nucleic acid, or mmRNA, increases cell transfection by the modified nucleic acid, or mmRNA, increases the expression of modified nucleic acid, or mmRNA encoded protein, and/or alters the release profile of modified nucleic acid, or mmRNA encoded proteins. Further, the modified nucleic acids and mmRNA of the present invention may be formulated using self-assembled nucleic acid nanoparticles.
[00365] Formulations of the pharmaceutical compositions described herein may be prepared by any method known or hereafter developed in the art of pharmacology. In general, such preparatory methods include the step of associating the active ingredient with an excipient and/or one or more other accessory ingredients.
[00366] A pharmaceutical composition in accordance with the present disclosure may be prepared, packaged, and/or sold in bulk, as a single unit dose, and/or as a plurality of single unit doses. As used herein, a "unit dose" refers to a discrete amount of the pharmaceutical composition comprising a predetermined amount of the active ingredient. The amount of the active ingredient may generally be equal to the dosage of the active ingredient which would be administered to a subject and/or a convenient fraction of such a dosage including, but not limited to, one-half or one-third of such a dosage.
[00367] Relative amounts of the active ingredient, the pharmaceutically acceptable excipient, and/or any additional ingredients in a pharmaceutical composition in accordance with the present disclosure may vary, depending upon the identity, size, and/or condition of the subject being treated and further depending upon the route by which the composition is to be administered. For example, the composition may comprise between 0.1% and 99% (w/w) of the active ingredient.
[00368] In some embodiments, the modified mRNA formulations described herein may contain at least one modified mRNA. The formulations may contain 1, 2, 3, 4 or 5 modified mRNA. In one embodiment, the formulation contains at least three modified mRNA encoding proteins. In one embodiment, the formulation contains at least five modified mRNA encoding proteins.
[00369] Pharmaceutical formulations may additionally comprise a pharmaceutically acceptable excipient, which, as used herein, includes, but is not limited to, any and all solvents, dispersion media, diluents, or other liquid vehicles, dispersion or suspension aids, surface active agents, isotonic agents, thickening or emulsifying agents, preservatives, and the like, as suited to the particular dosage form desired. Various excipients for formulating pharmaceutical compositions and techniques for preparing the composition are known in the art (see Remington: The Science and Practice of Pharmacy, 21st Edition, A. R. Gennaro, Lippincott, Williams & Wilkins, Baltimore, MD, 2006; incorporated herein by reference in its entirety). The use of a conventional excipient medium may be contemplated within the scope of the present disclosure, except insofar as any conventional excipient medium may be incompatible with a substance or its derivatives, such as by producing any undesirable biological effect or otherwise interacting in a deleterious manner with any other component(s) of the pharmaceutical composition.
[00370] In some embodiments, the particle size of the lipid nanoparticle may be increased and/or decreased. The change in particle size may be able to help counter biological reaction such as, but not limited to, inflammation or may increase the biological effect of the modified mRNA delivered to mammals.
[00371] Pharmaceutically acceptable excipients used in the manufacture of pharmaceutical compositions include, but are not limited to, inert diluents, surface active agents and/or emulsifiers, preservatives, buffering agents, lubricating agents, and/or oils. Such excipients may optionally be included in the pharmaceutical formulations of the invention.
Lipidoids
[00372] The synthesis of lipidoids has been extensively described and formulations containing these compounds are particularly suited for delivery of modified nucleic acid molecules or mmR A (see Mahon et al, Bioconjug Chem. 2010 21 : 1448-1454; Schroeder et al., J Intern Med. 2010 267:9-21; Akinc et al, Nat Biotechnol. 2008 26:561-569; Love et al, Proc Natl Acad Sci U S A. 2010
107: 1864-1869; Siegwart et al, Proc Natl Acad Sci U S A. 2011 108: 12996-3001; all of which are incorporated herein in their entireties).
[00373] While these lipidoids have been used to effectively deliver double stranded small interfering RNA molecules in rodents and non-human primates (see Akinc et al., Nat Biotechnol. 2008 26:561-569; Frank-Kamenetsky et al, Proc Natl Acad Sci U S A. 2008 105: 11915-11920; Akinc et al, Mol Ther. 2009 17:872-879; Love et al, Proc Natl Acad Sci U S A. 2010 107:1864- 1869; Leuschner et al., Nat Biotechnol. 2011 29: 1005-1010; all of which is incorporated herein in their entirety), the present disclosure describes their formulation and use in delivering single stranded modified nucleic acid molecules or mmRNA. Complexes, micelles, liposomes or particles can be prepared containing these lipidoids and therefore, can result in an effective delivery of the modified nucleic acid molecules or mmRNA, as judged by the production of an encoded protein, following the injection of a lipidoid formulation via localized and/or systemic routes of
administration. Lipidoid complexes of modified nucleic acid molecules or mmRNA can be administered by various means including, but not limited to, intravenous, intramuscular, or subcutaneous routes.
[00374] In vivo delivery of nucleic acids may be affected by many parameters, including, but not limited to, the formulation composition, nature of particle PEGylation, degree of loading, oligonucleotide to lipid ratio, and biophysical parameters such as, but not limited to, particle size (Akinc et al., Mol Ther. 2009 17:872-879; herein incorporated by reference in its entirety). As an example, small changes in the anchor chain length of poly(ethylene glycol) (PEG) lipids may result in significant effects on in vivo efficacy. Formulations with the different lipidoids, including, but not limited to penta[3-(l-laurylaminopropionyl)]-triethylenetetramine hydrochloride (TETA-5LAP; aka 98N12-5, see Murugaiah et al., Analytical Biochemistry, 401 :61 (2010); herein incorporated by reference in its entirety), CI 2-200 (including derivatives and variants), and MDl, can be tested for in vivo activity.
[00375] The lipidoid referred to herein as "98N12-5" is disclosed by Akinc et al, Mol Ther. 2009 17:872-879 and is incorporated by reference in its entirety (See Figure 1).
[00376] The lipidoid referred to herein as "CI 2-200" is disclosed by Love et al, Proc Natl Acad Sci U S A. 2010 107: 1864-1869 and Liu and Huang, Molecular Therapy. 2010 669-670 (see Figure 1); both of which are herein incorporated by reference in their entirety. The lipidoid formulations can include particles comprising either 3 or 4 or more components in addition to modified nucleic acid molecules or mmRNA. As an example, formulations with certain lipidoids, include, but are not limited to, 98N12-5 and may contain 42% lipidoid, 48% cholesterol and 10% PEG (CI 4 alkyl chain length). As another example, formulations with certain lipidoids, include, but are not limited to, CI 2-200 and may contain 50%> lipidoid, 10%> disteroylphosphatidyl choline, 38.5% cholesterol, and 1.5% PEG-DMG.
[00377] In one embodiment, a modified nucleic acid molecule or mmRNA formulated with a lipidoid for systemic intravenous administration can target the liver. For example, a final optimized intravenous formulation using modified nucleic acid molecule or mmRNA, and comprising a lipid molar composition of 42% 98N12-5, 48% cholesterol, and 10% PEG-lipid with a final weight ratio of about 7.5 to 1 total lipid to modified nucleic acid, or mmRNA, and a C14 alkyl chain length on the PEG lipid, with a mean particle size of roughly 50-60 nm, can result in the distribution of the formulation to be greater than 90% to the liver.(see, Akinc et al., Mol Ther. 2009 17:872-879; herein incorporated by reference in its entirety). In another example, an intravenous formulation using a C12-200 (see US provisional application 61/175,770 and published international application
WO2010129709, each of which is herein incorporated by reference in their entirety) lipidoid may have a molar ratio of 50/10/38.5/1.5 of C12-200/disteroylphosphatidyl choline/cholesterol/PEG- DMG, with a weight ratio of 7 to 1 total lipid to modified nucleic acid molecule or mmRNA, and a mean particle size of 80 nm may be effective to deliver modified nucleic acid molecule or mmRNA to hepatocytes (see, Love et al, Proc Natl Acad Sci U S A. 2010 107: 1864-1869 herein incorporated by reference in its entirety). In another embodiment, an MDl lipidoid-containing formulation may be used to effectively deliver modified nucleic acid molecule or mmRNA to hepatocytes in vivo. The characteristics of optimized lipidoid formulations for intramuscular or subcutaneous routes may vary significantly depending on the target cell type and the ability of formulations to diffuse through the extracellular matrix into the blood stream. While a particle size of less than 150 nm may be desired for effective hepatocyte delivery due to the size of the endothelial fenestrae (see, Akinc et al., Mol Ther. 2009 17:872-879 herein incorporated by reference in its entirety), use of a lipidoid- formulated modified nucleic acid molecules or mmRNA to deliver the formulation to other cells types including, but not limited to, endothelial cells, myeloid cells, and muscle cells may not be similarly size-limited. Use of lipidoid formulations to deliver siRNA in vivo to other non-hepatocyte cells such as myeloid cells and endothelium has been reported (see Akinc et al., Nat Biotechnol. 2008 26:561-569; Leuschner et al, Nat Biotechnol. 2011 29: 1005-1010; Cho et al. Adv. Funct. Mater. 2009 19:31 12-3118; 8th International Judah Folkman Conference, Cambridge, MA October 8-9, 2010; each of which is herein incorporated by reference in its entirety). Effective delivery to myeloid cells, such as monocytes, lipidoid formulations may have a similar component molar ratio. Different ratios of lipidoids and other components including, but not limited to, disteroylphosphatidyl choline, cholesterol and PEG-DMG, may be used to optimize the formulation of the modified nucleic acid, or mmRNA for delivery to different cell types including, but not limited to, hepatocytes, myeloid cells, muscle cells, etc. For example, the component molar ratio may include, but is not limited to, 50% C12-200, 10% disteroylphosphatidyl choline, 38.5% cholesterol, and %1.5 PEG-DMG (see
Leuschner et al., Nat Biotechnol 2011 29: 1005-1010; herein incorporated by reference in its entirety). The use of lipidoid formulations for the localized delivery of nucleic acids to cells (such as, but not limited to, adipose cells and muscle cells) via either subcutaneous or intramuscular delivery, may not require all of the formulation components desired for systemic delivery, and as such may comprise only the lipidoid and the modified nucleic acid molecule or mmRNA.
[00378] Combinations of different lipidoids may be used to improve the efficacy of modified nucleic acid molecule or mmRNA directed protein production as the lipidoids may be able to increase cell transfection by the modified nucleic acid molecule or mmRNA; and/or increase the translation of encoded protein (see Whitehead et al., Mol. Ther. 2011, 19: 1688-1694, herein incorporated by reference in its entirety).
Liposomes, Lipoplexes, and Lipid Nanoparticles
[00379] The modified nucleic acid molecules and mmRNA of the invention can be formulated using one or more liposomes, lipoplexes, or lipid nanoparticles. In one embodiment, pharmaceutical compositions of modified nucleic acid molecule or mmRNA include liposomes. Liposomes are artificially-prepared vesicles which may primarily be composed of a lipid bilayer and may be used as a delivery vehicle for the administration of nutrients and pharmaceutical formulations. Liposomes can be of different sizes such as, but not limited to, a multilamellar vesicle (MLV) which may be hundreds of nanometers in diameter and may contain a series of concentric bilayers separated by narrow aqueous compartments, a small unicellular vesicle (SUV) which may be smaller than 50 nm in diameter, and a large unilamellar vesicle (LUV) which may be between 50 and 500 nm in diameter. Liposome design may include, but is not limited to, opsonins or ligands in order to improve the attachment of liposomes to unhealthy tissue or to activate events such as, but not limited to, endocytosis. Liposomes may contain a low or a high pH in order to improve the delivery of the pharmaceutical formulations.
[00380] The formation of liposomes may depend on the physicochemical characteristics such as, but not limited to, the pharmaceutical formulation entrapped and the liposomal ingredients , the nature of the medium in which the lipid vesicles are dispersed, the effective concentration of the entrapped substance and its potential toxicity, any additional processes involved during the application and/or delivery of the vesicles, the optimization size, polydispersity and the shelf-life of the vesicles for the intended application, and the batch-to-batch reproducibility and possibility of large-scale production of safe and efficient liposomal products.
[00381] In one embodiment, pharmaceutical compositions described herein may include, without limitation, liposomes such as those formed from 1 ,2-dioleyloxy-N,N-dimethylaminopropane (DODMA) liposomes, DiLa2 liposomes from Marina Biotech (Bothell, WA), l,2-dilinoleyloxy-3- dimethylaminopropane (DLin-DMA), 2,2-dilinoleyl-4-(2-dimethylaminoethyl)-[ 1 ,3]-dioxolane (DLin-KC2-DMA), and MC3 (US20100324120; herein incorporated by reference in its entirety) and liposomes which may deliver small molecule drugs such as, but not limited to, DOXIL® from Janssen Biotech, Inc. (Horsham, PA). In one embodiment, pharmaceutical compositions described herein may include, without limitation, liposomes such as those formed from the synthesis of stabilized plasmid-lipid particles (SPLP) or stabilized nucleic acid lipid particle (SNALP) that have been previously described and shown to be suitable for oligonucleotide delivery in vitro and in vivo (see Wheeler et al. Gene Therapy. 1999 6:271-281; Zhang et al. Gene Therapy. 1999 6: 1438-1447; Jeffs et al. Pharm Res. 2005 22:362-372; Morrissey et al, Nat Biotechnol. 2005 2: 1002-1007;
Zimmermann et al, Nature. 2006 441 : 111-114; Heyes et al. J Contr Rel. 2005 107:276-287; Semple et al. Nature Biotech. 2010 28: 172-176; Judge et al. J Clin Invest. 2009 119:661-673; deFougeroUes Hum Gene Ther. 2008 19: 125-132; all of which are incorporated herein in their entireties.) The original manufacture method by Wheeler et al. was a detergent dialysis method, which was later improved by Jeffs et al. and is referred to as the spontaneous vesicle formation method. The liposome formulations are composed of 3 to 4 lipid components in addition to the modified nucleic acid molecule or mrnRNA. As an example a liposome can contain, but is not limited to, 55% cholesterol, 20% disteroylphosphatidyl choline (DSPC), 10% PEG-S-DSG, and 15% 1 ,2-dioleyloxy- N,N-dimethylaminopropane (DODMA), as described by Jeffs et al. As another example, certain liposome formulations may contain, but are not limited to, 48% cholesterol, 20% DSPC, 2% PEG-c- DMA, and 30% cationic lipid, where the cationic lipid can be 1 ,2-distearloxy-N,N- dimethylaminopropane (DSDMA), DODMA, DLin-DMA, or l,2-dilinolenyloxy-3- dimethylaminopropane (DLenDMA), as described by Heyes et al.
[00382] In one embodiment, pharmaceutical compositions may include liposomes which may be formed to deliver mrnRNA which may encode at least one immunogen. The mrnRNA may be encapsulated by the liposome and/or it may be contained in an aqueous core which may then be encapsulated by the liposome (see International Pub. Nos. WO2012031046, WO2012031043, WO2012030901 and WO2012006378; each of which is herein incorporated by reference in their entirety). In another embodiment, the mrnRNA which may encode an immunogen may be formulated in a cationic oil-in- water emulsion where the emulsion particle comprises an oil core and a cationic lipid which can interact with the mrnRNA anchoring the molecule to the emulsion particle (see International Pub. No. WO2012006380; herein incorporated by reference in its entirety). In yet another embodiment, the lipid formulation may include at least cationic lipid, a lipid which may enhance transfection and a least one lipid which contains a hydrophilic head group linked to a lipid moiety (International Pub. No. WO2011076807 and U.S. Pub. No. 20110200582; each of which is herein incorporated by reference in their entirety). In another embodiment, the modified mRNA encoding an immunogen may be formulated in a lipid vesicle which may have crosslinks between functionalized lipid bilayers (see U.S. Pub. No. 20120177724, herein incorporated by reference in its entirety).
[00383] In one embodiment, the modified mRNA may be formulated in a lipid vesicle which may have crosslinks between functionalized lipid bilayers.
[00384] In one embodiment, the modified mRNA may be formulated in a lipid-polycation complex. The formation of the lipid-polycation complex may be accomplished by methods known in the art and/or as described in U.S. Pub. No. 20120178702, herein incorporated by reference in its entirety. As a non-limiting example, the polycation may include a cationic peptide or a polypeptide such as, but not limited to, polylysine, polyornithine and/or polyarginine and the cationic peptides described in International Pub. No. WO2012013326; herein incorporated by reference in its entirety. In another embodiment, the modified mRNA may be formulated in a lipid-polycation complex which may further include a neutral lipid such as, but not limited to, cholesterol or dioleoyl
phosphatidylethanolamine (DOPE).
[00385] The liposome formulation may be influenced by, but not limited to, the selection of the cationic lipid component, the degree of cationic lipid saturation, the nature of the PEGylation, ratio of all components and biophysical parameters such as size. In one example by Semple et al.
(Semple et al. Nature Biotech. 2010 28: 172-176; herein incorporated by reference in its entirety), the liposome formulation was composed of 57.1 % cationic lipid, 7.1% dipalmitoylphosphatidylcholme, 34.3 % cholesterol, and 1.4% PEG-c-DMA. As another example, changing the composition of the cationic lipid could more effectively deliver siRNA to various antigen presenting cells (Basha et al. Mol Ther. 2011 19:2186-2200; herein incorporated by reference in its entirety).
[00386] In some embodiments, the ratio of PEG in the lipid nanoparticle (LNP) formulations may be increased or decreased and/or the carbon chain length of the PEG lipid may be modified from C14 to C18 to alter the pharmacokinetics and/or biodistribution of the LNP formulations. As a non- limiting example, LNP formulations may contain 1-5% of the lipid molar ratio of PEG-c-DOMG as compared to the cationic lipid, DSPC and cholesterol. In another embodiment the PEG-c-DOMG may be replaced with a PEG lipid such as, but not limited to, PEG- DSG (1,2-Distearoyl-sn-glycerol, methoxypolyethylene glycol) or PEG-DPG (1,2-Dipalmitoyl-sn-glycerol, methoxypolyethylene glycol). The cationic lipid may be selected from any lipid known in the art such as, but not limited to, DLin-MC3-DMA, DLin-DMA, C12-200 and DLin-KC2-DMA.
[00387] In one embodiment, the cationic lipid may be selected from, but not limited to, a cationic lipid described in International Publication Nos. WO2012040184, WO2011153120,
WO2011149733, WO2011090965, WO2011043913, WO2011022460, WO2012061259,
WO2012054365, WO2012044638, WO2010080724, WO201021865 and WO2008103276, US Patent Nos. 7,893,302, 7,404,969 and 8,283,333 and US Patent Publication No. US20100036115 and US20120202871; each of which is herein incorporated by reference in their entirety. In another embodiment, the cationic lipid may be selected from, but not limited to, formula A described in International Publication Nos. WO2012040184, WO2011153120, WO2011149733, WO2011090965, WO2011043913, WO2011022460, WO2012061259, WO2012054365 and
WO2012044638; each of which is herein incorporated by reference in their entirety. In yet another embodiment, the cationic lipid may be selected from, but not limited to, formula CLI-CLXXIX of International Publication No. WO2008103276, formula CLI-CLXXIX of US Patent No. 7,893,302, formula CLI-CLXXXXII of US Patent No. 7,404,969 and formula I-VI of US Patent Publication No. US20100036115; each of which is herein incorporated by reference in their entirety. As a non- limiting example, the cationic lipid may be selected from (20Z,23Z)-N,N-dimethylnonacosa-20,23- dien- 10-amine, ( 17Z,20Z)-N,N-dimemylhexacosa- 17,20-dien-9-amine, ( 1 Z, 19Z)-N5N- dimethylpentacosa-1 6, 19-dien-8-amine, (13Z,16Z)-N,N-dimethyldocosa-13,16-dien-5-amine, ( 12Z, 15Z)-N,N-dimethylhenicosa- 12, 15-dien-4-amine, (14Z, 17Z)-N,N-dimethyltricosa- 14,17-dien- 6-amine, (15Z, 18Z)-N,N-dimethyltetracosa- 15,18-dien-7-amine, ( 18Z,21 Z)-N,N-dimethylheptacosa- 18,21 -dien- 10-amine, (15Z, 18Z)-N,N-dimethyltetracosa- 15,18-dien-5-amine, (14Z, 17Z)-N,N- dimethyltricosa- 14,17-dien-4-amine, ( 19Z,22Z)-N,N-dimeihyloctacosa- 19,22-dien-9-amine, (18Z,21 Z)-N,N-dimethylheptacosa- 18 ,21 -dien-8 -amine, (17Z,20Z)-N,N-dimethylhexacosa- 17,20-dien-7- amine, ( 16Z, 19Z)-N,N-dimethylpentacosa- 16,19-dien-6-amine, (22Z,25Z)-N,N- dimethylhentriaconta-22,25-dien-10-amine, (21 Z ,24Z)-N,N-dimethyltriaconta-21,24-dien-9-amine, ( 18Z)-N,N-dimetylheptacos- 18-en- 10-amine, ( 17Z)-N,N-dimethylhexacos- 17-en-9-amine,
( 19Z,22Z)-N,N-dimethyloctacosa- 19,22-dien-7-amine, N,N-dimethylheptacosan- 10-amine,
(20Z,23Z)-N-ethyl-N-methylnonacosa-20,23-dien-10-amine, 1-[(1 lZ,14Z)-l-nonylicosa-l 1,14-dien-
1- yl] pyrrolidine, (20Z)-N,N-dimethylheptacos-20-en-l 0-amine, (15Z)-N,N-dimethyl eptacos-15-en- 1 0-amine, (14Z)-N,N-dimethylnonacos-14-en-10-amine, (17Z)-N,N-dimethylnonacos-17-en-10- amine, (24Z)-N,N-dimethyltritriacont-24-en-10-amine, (20Z)-N,N-dimethylnonacos-20-en-l 0- amine, (22Z)-N,N-dimethylhentriacont-22-en-10-amine, ( 16Z)-N,N-dimethylpentacos- 16-en-8- amine, ( 12Z, 15Z)-N,N-dimethyl-2-nonylhenicosa- 12, 15-dien- 1-amine, (13Z, 16Z)-N,N-dimethyl-3- nonyldocosa-13,16-dien-l-amine, N,N-dimethyl-l-[(lS,2R)-2-octylcyclopropyl] eptadecan-8-amine,
1 -[(1 S,2R)-2-hexylcyclopropyl]-N,N-dimethylnonadecan- 10-amine, Ν,Ν-dimethyl- 1 -[(1 S ,2R)-2- octylcyclopropyl]nonadecan-l 0-amine, N,N-dimethyl-21-[(lS,2R)-2-octylcyclopropyl]henicosan-10- amine,N,N-dimethyl-l-[(lS,2S)-2-{[(lR,2R)-2-pentylcycIopropyl]methyl}cyclopropyl]nonadecan- 10-amine,N,N-dimethyl-l-[(lS,2R)-2-octylcyclopropyl]hexadecan-8-amine, N,N-dimethyl-[(lR,2S)-
2- undecyIcyclopropyl]tetradecan-5-amine, N,N-dimethyl-3-{7-[(lS,2R)-2-octylcyclopropyl]heptyl} dodecan- 1-amine, l-[(lR,2S)-2-hepty lcyclopropyl]-N,N-dimethyloctadecan-9-amine, 1-[(1S,2R)- 2-decylcyclopropyl]-N,N-dimethylpentadecan-6-amine, N,N-dimethyl-l-[(lS,2R)-2- octylcyclopropyl]pentadecan-8-amine, R-N,N-dimethyl-l-[(9Z,12Z)-octadeca-9,12-dien-l-yloxy]-3- (octyloxy)propan-2-amine, S-N,N-dimethyl-l-[(9Z,12Z)-octadeca-9,12-dien-l-yloxy]-3- (octyloxy)propan-2-amine, 1 - {2-[(9Z, 12Z)-octadeca-9, 12-dien- 1 -yloxy]- 1 -
[(octyloxy)methyl]ethyl}pyrrolidine, (2S)-N,N-dimethyl-l-[(9Z,12Z)-octadeca-9,12-dien-l-yloxy]- 3 - [(5Z)-oct-5 -en- 1 -yloxy]propan-2-amine, 1 - {2- [(9Z, 12Z)-octadeca-9, 12-dien- 1 -yloxy] - 1 - [(octyloxy)methyl]ethyl}azetidine, (2S)-l-(hexyloxy)-N,N-dimethyl-3-[(9Z,12Z)-octadeca-9,12- dien- 1 -yloxy]propan-2-amine, (2S)- 1 -(heptyloxy)-N,N-dimethyl-3-[(9Z, 12Z)-octadeca-9, 12-dien- 1 - yloxy]propan-2-amine, Ν,Ν-dimethyl- 1 -(nonyloxy)-3-[(9Z, 12Z)-octadeca-9, 12-dien- 1 - yloxy]propan-2-amine, N,N-dimethyl-l-[(9Z)-octadec-9-en-l-yloxy]-3-(octyloxy)propan-2-amine; (2S)-N,N-dimethyl- 1 -[(6Z,9Z, 12Z)-octadeca-6,9, 12-trien- 1 -yloxy]-3-(octyloxy)propan-2-amine, (2S)- 1 -[( 11 Z, 14Z)-icosa- 11,14-dien- 1 -yloxy]-N,N-dimethyl-3-(pentyloxy)propan-2-amine, (2S)- 1 - (hexyloxy)-3-[(l lZ,14Z)-icosa-l 1,14-dien- l-yloxy]-N,N-dimethylpropan-2-amine, 1-[(11Z,14Z)- icosa- 11 , 14-dien- 1 -yloxy]-N,N-dimethy 1 -3 -(octyloxy)propan-2-amine, 1 - [( 13Z, 16Z)-docosa-13 ,16- dien-l-yloxy]-N,N-dimethyl-3-(octyloxy)propan-2-amine, (2S)- 1 -[(13Z, 16Z)-docosa- 13,16-dien- 1 - yloxy]-3-(hexyloxy)-N,N-dimethylpropan-2-amine, (2S)- 1 -[(13Z)-docos-l 3 -en- 1 -yloxy] -3- (hexyloxy)-N,N-dimethylpropan-2-amine, 1 -[(13Z)-docos-l 3 -en- 1 -yloxy] -N,N-dimethyl-3- (octyloxy)propan-2-amine, 1 -[(9Z)-hexadec-9-en- 1 -yloxy]-N,N-dimethyl-3-(octyloxy)propan-2- amine, (2R)-N,N-dimethyl-H(l-metoylo ctyl)oxy]-3-[(9Z,12Z)-octadeca-9,12-dien-l-yloxy]propan- 2-amine, (2R)-l-[(3,7-dimethyloctyl)oxy]-N,N-dimethyl-3-[(9Z,12Z)-octadeca-9,12-dien-l- yloxy]propan-2-amine, N,N-dimethyl-l-(octyloxy)-3-({8-[(lS,2S)-2-{[(lR,2R)-2- pentylcyclopropyl]methyl} cyclopropyl]octyl} oxy)propan-2-amine, Ν,Ν-dimethyl- 1 - { [8-(2- oclylcyclopropyl)octyl]oxy}-3-(octyloxy)propan-2-amine and (11E,20Z,23Z)-N,N- dimethylnonacosa-ll,20,2-trien-10-amine or a pharmaceutically acceptable salt or stereoisomer thereof.
[00388] In one embodiment, the cationic lipid may be synthesized by methods known in the art and/or as described in International Publication Nos. WO2012040184, WO2011153120,
WO2011149733, WO2011090965, WO2011043913, WO2011022460, WO2012061259,
WO2012054365, WO2012044638, WO2010080724 and WO201021865; each of which is herein incorporated by reference in their entirety. [00389] In one embodiment, the LNP formulation may contain PEG-c-DOMG at 3% lipid molar ratio. In another embodiment, the LNP formulation may contain PEG-c-DOMG at 1.5% lipid molar ratio.
[00390] In one embodiment, the LNP formulation may contain PEG-DMG 2000 (1,2-dimyristoyl- sn-glycero-3-phophoethanolamine-N-[methoxy(poly ethylene glycol)-2000). In one embodiment, the LNP formulation may contain PEG-DMG 2000, a cationic lipid known in the art and at least one other component. In another embodiment, the LNP formulation may contain PEG-DMG 2000, a cationic lipid known in the art, DSPC and cholesterol. As a non-limiting example, the LNP formulation may contain PEG-DMG 2000, DLin-DMA, DSPC and cholesterol. As another non- limiting example the LNP formulation may contain PEG-DMG 2000, DLin-DMA, DSPC and cholesterol in a molar ratio of 2:40: 10:48 (see e.g. Geall et al., Nonviral delivery of self-amplifying RNA vaccines, PNAS 2012; PMID: 22908294; herein incorporated by reference in its entirety).
[00391] In one embodiment, the LNP formulation may be formulated by the methods described in International Publication Nos. WO2011127255 or WO2008103276, each of which is herein incorporated by reference in their entirety. As a non-limiting example, modified RNA described herein may be encapsulated in LNP formulations as described in WO2011127255 and/or
WO2008103276; each of which is herein incorporated by reference in their entirety. As another non-limiting example, modified RNA described herein may be formulated in a nanoparticle to be delivered by a parenteral route as described in U.S. Pub. No. 20120207845; herein incorporated by reference in its entirety.
[00392] In one embodiment, LNP formulations described herein may comprise a polycationic composition. As a non-limiting example, the polycationic composition may be selected from formula 1-60 of US Patent Publication No. US20050222064; herein incorporated by reference in its entirety. In another embodiment, the LNP formulations comprising a polycationic composition may be used for the delivery of the modified RNA described herein in vivo and/or in vitro.
[00393] In one embodiment, the LNP formulations described herein may additionally comprise a permeability enhancer molecule. Non- limiting permeability enhancer molecules are described in US
Patent Publication No. US20050222064; herein incorporated by reference in its entirety.
[00394] In one embodiment, the pharmaceutical compositions may be formulated in liposomes such as, but not limited to, DiLa2 liposomes (Marina Biotech, Bothell, WA), SMARTICLES® (Marina
Biotech, Bothell, WA), neutral DOPC (l,2-dioleoyl-sn-glycero-3-phosphocholine) based liposomes (e.g., siRNA delivery for ovarian cancer (Landen et al. Cancer Biology & Therapy 2006 5(12)1708- 1713); herein incorporated by reference in its entirety) and hyaluronan-coated liposomes (Quiet Therapeutics, Israel).
[00395] The nanoparticle formulations may be a carbohydrate nanoparticle comprising a carbohydrate carrier and a modified nucleic acid molecule (e.g., mmRNA). As a non-limiting example, the carbohydrate carrier may include, but is not limited to, an anhydride-modified phytoglycogen or glycogen-type material, phtoglycogen octenyl succinate, phytoglycogen beta- dextrin, anhydride-modified phytoglycogen beta-dextrin. (See e.g., International Publication No. WO2012109121; herein incorporated by reference in its entirety).
[00396] Lipid nanoparticle formulations may be improved by replacing the cationic lipid with a biodegradable cationic lipid which is known as a rapidly eliminated lipid nanoparticle (reLNP). Ionizable cationic lipids, such as, but not limited to, DLinDMA, DLin-KC2-DMA, and DLin-MC3- DMA, have been shown to accumulate in plasma and tissues over time and may be a potential source of toxicity. The rapid metabolism of the rapidly eliminated lipids can improve the tolerability and therapeutic index of the lipid nanoparticles by an order of magnitude from a 1 mg/kg dose to a 10 mg/kg dose in rat. Inclusion of an enzymatically degraded ester linkage can improve the degradation and metabolism profile of the cationic component, while still maintaining the activity of the reLNP formulation. The ester linkage can be internally located within the lipid chain or it may be terminally located at the terminal end of the lipid chain. The internal ester linkage may replace any carbon in the lipid chain.
[00397] In one embodiment, the internal ester linkage may be located on either side of the saturated carbon. Non-limiting examples of reLNPs include,
Figure imgf000140_0001
[00398] In one embodiment, an immune response may be elicited by delivering a lipid nanoparticle which may include a nanospecies, a polymer and an immunogen. (U.S. Publication No.
20120189700 and International Publication No. WO2012099805; each of which is herein incorporated by reference in their entirety). The polymer may encapsulate the nanospecies or partially encapsulate the nanospecies. The immunogen may be a recombinant protein, a modified R A described herein. In one embodiment, the lipid nanoparticle may be formulated for use in a vaccine such as, but not limited to, against a pathogen.
[00399] Lipid nanoparticles may be engineered to alter the surface properties of particles so the lipid nanoparticles may penetrate the mucosal barrier. Mucus is located on mucosal tissue such as, but not limted to, oral (e.g., the buccal and esophageal membranes and tonsil tissue), ophthalmic, gastrointestinal (e.g., stomach, small intestine, large intestine, colon, rectum), nasal, respiratory (e.g., nasal, pharyngeal, tracheal and bronchial membranes), genital (e.g., vaginal, cervical and urethral membranes). Nanoparticles larger than 10-200 nm which are preferred for higher drug
encapsulation efficiency and the ability to provide the sustained delivery of a wide array of drugs have been thought to be too large to rapidly diffuse through mucosal barriers. Mucus is
continuously secreted, shed, discarded or digested and recycled so most of the trapped particles may be removed from the mucosla tissue within seconds or within a few hours. Large polymeric nanoparticles (200nm -500nm in diameter) which have been coated densely with a low molecular weight polyethylene glycol (PEG) diffused through mucus only 4 to 6-fold lower than the same particles diffusing in water (Lai et al. PNAS 2007 104(5): 1482-487; Lai et al. Adv Drug Deliv Rev. 2009 61(2): 158-171; each of which is herein incorporated by reference in their entirety). The transport of nanoparticles may be determined using rates of permeation and/or fluorescent microscopy techniques including, but not limited to, fluorescence recovery after photobleaching (FRAP) and high resolution multiple particle tracking (MPT). As a non- limiting example, compositions which can penetrate a mucosal barrier may be made as described in U.S. Pat. No. 8,241,670, herein incorporated by reference in its entirety.
[00400] The lipid nanoparticle engineered to penetrate mucus may comprise a polymeric material (i.e. a polymeric core) and/or a polymer-vitamin conjugate and/or a tri-block co-polymer. The polymeric material may include, but is not limited to, polyamines, polyethers, polyamides, polyesters, polycarbamates, polyureas, polycarbonates, poly(styrenes), polyimides, polysulfones, polyurethanes, polyacetylenes, polyethylenes, polyethyeneimines, polyisocyanates, polyacrylates, polymethacrylates, polyacrylonitriles, and polyarylates. The polymeric material may be
biodegradable and/or biocompatible. The polymeric material may additionally be irradiated. As a non-limiting example, the polymeric material may be gamma irradiated (See e.g., International App. No. WO201282165, herein incorporated by reference in its entirety). Non-limiting examples of specific polymers include poly(caprolactone) (PCL), ethylene vinyl acetate polymer (EVA), poly(lactic acid) (PLA), poly(L-lactic acid) (PLLA), poly(glycolic acid) (PGA), poly(lactic acid-co- glycolic acid) (PLGA), poly(L-lactic acid-co-glycolic acid) (PLLGA), poly(D,L-lactide) (PDLA), poly(L-lactide) (PLLA), poly(D,L-lactide-co-caprolactone), poly(D,L-lactide-co-caprolactone-co- glycolide), poly(D,L-lactide-co-PEO-co-D,L-lactide), poly(D,L-lactide-co-PPO-co-D,L-lactide), polyalkyl cyanoacralate, polyurethane, poly-L-lysine (PLL), hydroxypropyl methacrylate (HPMA), polyethyleneglycol, poly-L-glutamic acid, poly(hydroxy acids), polyanhydrides, polyorthoesters, poly(ester amides), polyamides, poly(ester ethers), polycarbonates, polyalkylenes such as polyethylene and polypropylene, polyalkylene glycols such as poly(ethylene glycol) (PEG), polyalkylene oxides (PEO), polyalkylene terephthalates such as poly(ethylene terephthalate), polyvinyl alcohols (PVA), polyvinyl ethers, polyvinyl esters such as poly( vinyl acetate), polyvinyl halides such as poly(vinyl chloride) (PVC), polyvinylpyrrolidone, polysiloxanes, polystyrene (PS), polyurethanes, derivatized celluloses such as alkyl celluloses, hydroxyalkyl celluloses, cellulose ethers, cellulose esters, nitro celluloses, hydroxypropylcellulose, carboxymethylcellulose, polymers of acrylic acids, such as poly(methyl(meth)acrylate) (PMMA), poly(ethyl(meth)acrylate), poly(butyl(meth)acrylate), poly(isobutyl(meth)acrylate), poly(hexyl(meth)acrylate),
poly(isodecyl(meth)acrylate), poly(lauryl(meth)acrylate), poly(phenyl(meth)acrylate), poly(methyl acrylate), poly(isopropyl acrylate), poly(isobutyl acrylate), poly(octadecyl acrylate) and copolymers and mixtures thereof, polydioxanone and its copolymers, polyhydroxyalkanoates, polypropylene fumarate, polyoxymethylene, poloxamers, poly(ortho)esters, poly(butyric acid), poly(valeric acid), poly(lactide-co-caprolactone), and trimethylene carbonate, polyvinylpyrrolidone. The lipid nanoparticle may be coated or associated with a co-polymer such as, but not limited to, a block copolymer, and (poly(ethylene glycol))-(poly(propylene oxide))-(poly(ethylene glycol)) triblock copolymer (see e.g., US Publication 20120121718 and US Publication 20100003337 and U.S. Pat. No. 8,263,665; each of which is herein incorporated by reference in their entirety). The co-polymer may be a polymer that is generally regarded as safe (GRAS) and the formation of the lipid nanoparticle may be in such a way that no new chemical entities are created. For example, the lipid nanoparticle may comprise poloxamers coating PLGA nanoparticles without forming new chemical entities which are still able to rapidly penetrate human mucus (Yang et al. Angew. Chem. Int. Ed. 2011 50:2597-2600; herein incorporated by reference in its entirety). [00401] The vitamin of the polymer-vitamin conjugate may be vitamin E. The vitamin portion of the conjugate may be substituted with other suitable components such as, but not limited to, vitamin A, vitamin E, other vitamins, cholesterol, a hydrophobic moiety, or a hydrophobic component of other surfactants (e.g., sterol chains, fatty acids, hydrocarbon chains and alkylene oxide chains).
[00402] The lipid nanoparticle engineered to penetrate mucus may include surface altering agents such as, but not limited to, mmRNA, anionic proteins (e.g., bovine serum albumin), surfactants (e.g., cationic surfactants such as for example dimethyldioctadecyl-ammonium bromide), sugars or sugar derivatives (e.g., cyclodextrin), nucleic acids, polymers (e.g., heparin, polyethylene glycol and poloxamer), mucolytic agents (e.g., N-acetylcysteine, mugwort, bromelain, papain, clerodendrum, acetylcysteine, bromhexine, carbocisteine, eprazinone, mesna, ambroxol, sobrerol, domiodol, letosteine, stepronin, tiopronin, gelsolin, thymosin β4 dornase alfa, neltenexine, erdosteine) and various DNases including rhDNase.. The surface altering agent may be embedded or enmeshed in the particle's surface or disposed (e.g., by coating, adsorption, covalent linkage, or other process) on the surface of the lipid nanoparticle. (see e.g., US Publication 20100215580 and US Publication 20080166414; each of which is herein incorporated by reference in their entirety).
[00403] The mucus penetrating lipid nanoparticles may comprise at least one mmRNA described herein. The mmRNA may be encapsulated in the lipid nanoparticle and/or disposed on the surface of the paricle. The mmRNA may be covalently coupled to the lipid nanoparticle. Formulations of mucus penetrating lipid nanoparticles may comprise a plurality of nanoparticles. Further, the formulations may contain particles which may interact with the mucus and alter the structural and/or adhesive properties of the surrounding mucus to decrease mucoadhesion which may increase the delivery of the mucus penetrating lipid nanoparticles to the mucosal tissue.
[00404] In one embodiment, the modified nucleic acid molecule or mmRNA is formulated as a lipoplex, such as, without limitation, the ATUPLEX™ system, the DACC system, the DBTC system and other siRNA-lipoplex technology from Silence Therapeutics (London, United Kingdom), STEMFECT™ from STEMGENT® (Cambridge, MA), and polyethylenimine (PEI) or protamine- based targeted and non-targeted delivery of nucleic acids acids (Aleku et al. Cancer Res. 2008 68:9788-9798; Strumberg et al. Int J Clin Pharmacol Ther 2012 50:76-78; Santel et al, Gene Ther 2006 13:1222-1234; Santel et al, Gene Ther 2006 13: 1360-1370; Gutbier et al, Pulm Pharmacol. Ther. 2010 23:334-344; Kaufmann et al. Microvasc Res 2010 80:286-293Weide et al. J Immunother. 2009 32:498-507; Weide et al. J Immunother. 2008 31 : 180-188; Pascolo Expert Opin. Biol. Ther. 4: 1285-1294; Fotin-Mleczek et al, 2011 J. Immunother. 34: 1-15; Song et al, Nature Biotechnol. 2005, 23:709-717; Peer et al, Proc Natl Acad Sci U S A. 2007 6;104:4095-4100; deFougerolles Hum Gene Ther. 2008 19: 125-132; all of which are incorporated herein by reference in its entirety).
[00405] In one embodiment such formulations may also be constructed or compositions altered such that they passively or actively are directed to different cell types in vivo, including but not limited to hepatocytes, immune cells, tumor cells, endothelial cells, antigen presenting cells, and leukocytes (Akinc et al. Mol Ther. 2010 18: 1357-1364; Song et al, Nat Biotechnol. 2005 23:709- 717; Judge et al, J Clin Invest. 2009 119:661-673; Kaufmann et al, Microvasc Res 2010 80:286- 293; Santel et al, Gene Ther 2006 13:1222-1234; Santel et al, Gene Ther 2006 13:1360-1370;
Gutbier et al, Pulm Pharmacol. Ther. 2010 23:334-344; Basha et al, Mol. Ther. 2011 19:2186-2200; Fenske and Cullis, Expert Opin Drug Deliv. 2008 5:25-44; Peer et al, Science. 2008 319:627-630; Peer and Lieberman, Gene Ther. 2011 18:1127-1133; all of which are incorporated herein by reference in its entirety). One example of passive targeting of formulations to liver cells includes the DLin-DMA, DLin-KC2-DMA and DLin-MC3 -DMA-based lipid nanoparticle formulations which have been shown to bind to apolipoprotein E and promote binding and uptake of these formulations into hepatocytes in vivo (Akinc et al. Mol Ther. 2010 18: 1357-1364; herein incorporated by reference in its entirety). Formulations can also be selectively targeted through expression of different ligands on their surface as exemplified by, but not limited by, folate, transferrin, N- acetylgalactosamine (GalNAc), and antibody targeted approaches (Kolhatkar et al, Curr Drug Discov Technol. 2011 8: 197-206; Musacchio and Torchilin, Front Biosci. 2011 16:1388-1412; Yu et al, Mol Membr Biol. 2010 27:286-298; Patil et al, Crit Rev Ther Drug Carrier Syst. 2008 25: 1-61; Benoit et al, Biomacromolecules. 2011 12:2708-2714; Zhao et al., Expert Opin Drug Deliv. 2008 5:309-319; Akinc et al, Mol Ther. 2010 18: 1357-1364; Srinivasan et al, Methods Mol Biol. 2012 820: 105-116; Ben-Arie et al, Methods Mol Biol. 2012 757:497-507; Peer 2010 J Control Release. 20:63-68; Peer et al, Proc Natl Acad Sci U S A. 2007 104:4095-4100; Kim et al, Methods Mol Biol. 2011 721 :339-353; Subramanya et al, Mol Ther. 2010 18:2028-2037; Song et al., Nat
Biotechnol. 2005 23:709-717; Peer et al, Science. 2008 319:627-630; Peer and Lieberman, Gene Ther. 2011 18: 1127-1133; all of which are incorporated herein by reference in its entirety)..
[00406] In one embodiment, the modified nucleic acid molecules or mmRNA are formulated as a solid lipid nanoparticle. A solid lipid nanoparticle (SLN) may be spherical with an average diameter between 10 to 1000 nm. SLN possess a solid lipid core matrix that can solubilize lipophilic molecules and may be stabilized with surfactants and/or emulsifiers. In a further embodiment, the lipid nanoparticle may be a self-assembly lipid-polymer nanoparticle (see Zhang et al., ACS Nano, 2008, 2 (8), pp 1696-1702; herein incorporated by reference in its entirety).
[00407] Liposomes, lipoplexes, or lipid nanoparticles may be used to improve the efficacy of modified nucleic acid molecules or mmRNA directed protein production as these formulations may be able to increase cell transfection by the modified nucleic acid molecule or mmRNA; and/or increase the translation of encoded protein. One such example involves the use of lipid encapsulation to enable the effective systemic delivery of polyplex plasmid DNA (Heyes et al., Mol Ther. 2007 15:713-720; herein incorporated by reference in its entirety). The liposomes, lipoplexes, or lipid nanoparticles may also be used to increase the stability of the modified nucleic acid molecules or mmRNA.
[00408] In one embodiment, the modified nucleic acid molecules and/or the mmRNA of the present invention can be formulated for controlled release and/or targeted delivery. As used herein,
"controlled release" refers to a pharmaceutical composition or compound release profile that conforms to a particular pattern of release to effect a therapeutic outcome. In one embodiment, the modified nucleic acids molecules or the mmRNA may be encapsulated into a delivery agent described herein and/or known in the art for controlled release and/or targeted delivery. As used herein, the term "encapsulate" means to enclose, surround or encase. As it relates to the formulation of the compounds of the invention, encapsulation may be substantial, complete or partial. The term "substantially encapsulated" means that at least greater than 50, 60, 70, 80, 85, 90, 95, 96, 97, 98, 99, 99.9, 99.9 or greater than 99.999% of the pharmaceutical composition or compound of the invention may be enclosed, surrounded or encased within the delivery agent. "Partially
encapsulation" means that less than 10, 10, 20, 30, 40 50 or less of the pharmaceutical composition or compound of the invention may be enclosed, surrounded or encased within the delivery agent. Advantageously, encapsulation may be determined by measuring the escape or the activity of the pharmaceutical composition or compound of the invention using fluorescence and/or electron micrograph. For example, at least 1, 5, 10, 20, 30, 40, 50, 60, 70, 80, 85, 90, 95, 96, 97, 98, 99, 99.9, 99.99 or greater than 99.99% of the pharmaceutical composition or compound of the invention are encapsulated in the delivery agent.
[00409] In one embodiment, the controlled release formulation may include, but is not limited to, tri-block co-polymers. As a non- limiting example, the formulation may include two different types of tri-block co-polymers (International Pub. No. WO2012131104 and WO2012131106; each of which is herein incorporated by reference in its entirety).
[00410] In another embodiment, the modified nucleic acid molecules or the mmRNA may be encapsulated into a lipid nanoparticle or a rapidly eliminated lipid nanoparticle and the lipid nanoparticles or a rapidly eliminated lipid nanoparticle may then be encapsulated into a polymer, hydrogel and/or surgical sealant described herein and/or known in the art. As a non-limiting example, the polymer, hydrogel or surgical sealant may be PLGA, ethylene vinyl acetate (EVAc), poloxamer, GELSITE® (Nanotherapeutics, Inc. Alachua, FL), HYLENEX® (Halozyme
Therapeutics, San Diego CA), surgical sealants such as fibrinogen polymers (Ethicon Inc. Cornelia, GA), TISSELL® (Baxter International, Inc Deerfield, IL), PEG-based sealants, and COSEAL® (Baxter International, Inc Deerfield, IL).
[00411] In another embodiment, the lipid nanoparticle may be encapsulated into any polymer known in the art which may form a gel when injected into a subject. As a non-limiting example, the lipid nanoparticle may be encapsulated into a polymer matrix which may be biodegradable.
[00412] In one embodiment, the modified nucleic acid molecules or mmRNA formulation for controlled release and/or targeted delivery may also include at least one controlled release coating. Controlled release coatings include, but are not limited to, OPADRY®, polyvinylpyrrolidone/vinyl acetate copolymer, polyvinylpyrrolidone, hydroxypropyl methylcellulose, hydroxypropyl cellulose, hydroxyethyl cellulose, EUDRAGIT RL®, EUDRAGIT RS® and cellulose derivatives such as ethylcellulose aqueous dispersions (AQUACOAT® and SURELEASE®).
[00413] In one embodiment, the controlled release and/or targeted delivery formulation may comprise at least one degradable polyester which may contain polycationic side chains. Degradeable polyesters include, but are not limited to, poly(serine ester), poly(L-lactide-co-L-lysine), poly(4- hydroxy-L-proline ester), and combinations thereof. In another embodiment, the degradable polyesters may include a PEG conjugation to form a PEGylated polymer.
[00414] In one embodiment, the modified nucleic acid molecules and/or the mmRNA of the present invention may be encapsulated in a therapeutic nanoparticle. Therapeutic nanoparticles may be formulated by methods described herein and known in the art such as, but not limited to,
International Pub Nos. WO2010005740, WO2010030763, WO2010005721, WO2010005723, WO2012054923, US Pub. Nos. US20110262491, US20100104645, US20100087337,
US20100068285, US20110274759, US20100068286 and US20120288541, and US Pat No. 8,206,747, 8,293,276 8,318,208 and 8,318,211; each of which is herein incorporated by reference in their entirety. In another embodiment, therapeutic polymer nanoparticles may be identified by the methods described in US Pub No. US20120140790, herein incorporated by reference in its entirety.
[00415] In one embodiment, the therapeutic nanoparticle may be formulated for sustained release. As used herein, "sustained release" refers to a pharmaceutical composition or compound that conforms to a release rate over a specific period of time. The period of time may include, but is not limited to, hours, days, weeks, months and years. As a non-limiting example, the sustained release nanoparticle may comprise a polymer and a therapeutic agent such as, but not limited to, the modified nucleic acid molecules and mmRNA of the present invention (see International Pub No. 2010075072 and US Pub No. US20100216804, US20110217377 and US20120201859, each of which is herein incorporated by reference in their entirety).
[00416] In one embodiment, the therapeutic nanoparticles may be formulated to be target specific. As a non-limiting example, the thereapeutic nanoparticles may include a corticosteroid (see International Pub. No. WO2011084518 herein incorporated by reference in its entirety). In one embodiment, the therapeutic nanoparticles of the present invention may be formulated to be cancer specific. As a non- limiting example, the therapeutic nanoparticles may be formulated in
nanoparticles described in International Pub No. WO2008121949, WO2010005726,
WO2010005725, WO2011084521 and US Pub No. US20100069426, US20120004293 and
US20100104655, each of which is herein incorporated by reference in their entirety.
[00417] In one embodiment, the nanoparticles of the present invention may comprise a polymeric matrix. As a non-limiting example, the nanoparticle may comprise two or more polymers such as, but not limited to, polyethylenes, polycarbonates, polyanhydrides, polyhydroxyacids,
polypropylfumerates, polycaprolactones, polyamides, polyacetals, polyethers, polyesters, poly(orthoesters), polycyanoacrylates, polyvinyl alcohols, polyurethanes, polyphosphazenes, polyacrylates, polymethacrylates, polycyanoacrylates, polyureas, polystyrenes, polyamines, polylysine, poly(ethylene imine), poly(serine ester), poly(L-lactide-co-L-lysine), poly(4-hydroxy-L- proline ester) or combinations thereof.
[00418] In one embodiment, the therapeutic nanoparticle comprises a diblock copolymer. In one embodiment, the diblock copolymer may include PEG in combination with a polymer such as, but not limited to, polyethylenes, polycarbonates, polyanhydrides, polyhydroxyacids,
polypropylfumerates, polycaprolactones, polyamides, polyacetals, polyethers, polyesters, poly(orthoesters), polycyanoacrylates, polyvinyl alcohols, polyurethanes, polyphosphazenes, polyacrylates, polymethacrylates, polycyanoacrylates, polyureas, polystyrenes, polyamines, polylysine, poly(ethylene imine), poly(serine ester), poly(L-lactide-co-L-lysine), poly(4-hydroxy-L- proline ester) or combinations thereof.
[00419] As a non- limiting example the therapeutic nanoparticle comprises a PLGA-PEG block copolymer (see US Pub. No. US20120004293 and US Pat No. 8,236,330, each of which is herein incorporated by reference in their entirety). In another non-limiting example, the therapeutic nanoparticle is a stealth nanoparticle comprising a diblock copolymer of PEG and PLA or PEG and PLGA (see US Pat No 8,246,968, herein incorporated by reference in its entirety).
[00420] In one embodiment, the therapeutic nanoparticle may comprise a multiblock copolymer (See e.g., U.S. Pat. No. 8,263,665 and 8,287,910; each of which is herein incorporated by reference in its entirety).
[00421] In one embodiment, the block copolymers described herein may be included in a polyion complex comprising a non-polymeric micelle and the block copolymer. (See e.g., U.S. Pub. No. 20120076836; herein incorporated by reference in its entirety).
[00422] In one embodiment, the therapeutic nanoparticle may comprise at least one acrylic polymer. Acrylic polymers include but are not limited to, acrylic acid, methacrylic acid, acrylic acid and methacrylic acid copolymers, methyl methacrylate copolymers, ethoxyethyl methacrylates, cyanoethyl methacrylate, amino alkyl methacrylate copolymer, poly(acrylic acid), poly(methacrylic acid), polycyanoacrylates and combinations thereof.
[00423] In one embodiment, the therapeutic nanoparticles may comprise at least one cationic polymer described herein and/or known in the art.
[00424] In one embodiment, the therapeutic nanoparticles may comprise at least one amine- containing polymer such as, but not limited to polylysine, polyethylene imine, poly(amidoamine) dendrimers, poly(beta-amino esters) (See e.g., U.S. Pat. No. 8,287,849; herein incorporated by reference in its entirety) and combinations thereof.
[00425] In one embodiment, the therapeutic nanoparticles may comprise at least one degradable polyester which may contain polycationic side chains. Degradeable polyesters include, but are not limited to, poly(serine ester), poly(L-lactide-co-L-lysine), poly(4-hydroxy-L-proline ester), and combinations thereof. In another embodiment, the degradable polyesters may include a PEG conjugation to form a PEGylated polymer. [00426] In another embodiment, the therapeutic nanoparticle may include a conjugation of at least one targeting ligand. The targeting ligand may be any ligand known in the art such as, but not limited to, a monoclonal antibody. (Kirpotin et al, Cancer Res. 2006 66:6732-6740; herein incorporated by reference in its entirety).
[00427] In one embodiment, the therapeutic nanoparticle may be formulated in an aqueous solution which may be used to target cancer (see International Pub No. WO2011084513 and US Pub No. US20110294717, each of which is herein incorporated by reference in their entirety).
[00428] In one embodiment, the modified nucleic acid molecules or mmRNA may be encapsulated in, linked to and/or associated with synthetic nanocarriers. Synthetic nanocarriers include, but are not limited to, those described in International Pub. Nos. WO2010005740, WO2010030763, WO201213501, WO2012149252, WO2012149255, WO2012149259, WO2012149265,
WO2012149268, WO2012149282, WO2012149301, WO2012149393, WO2012149405,
WO2012149411 and WO2012149454 and US Pub. Nos. US20110262491, US20100104645, US20100087337 and US20120244222, each of which is herein incorporated by reference in their entirety. The synthetic nanocarriers may be formulated using methods known in the art and/or described herein. As a non-limiting example, the synthetic nanocarriers may be formulated by the methods described in International Pub Nos. WO2010005740, WO2010030763 and WO201213501 and US Pub. Nos. US20110262491, US20100104645, US20100087337 and US20120244222, each of which is herein incorporated by reference in their entirety. In another embodiment, the synthetic nanocarrier formulations may be lyophilized by methods described in International Pub. No.
WO2011072218 and US Pat No. 8,211,473; each of which is herein incorporated by reference in their entirety.
[00429] In one embodiment, the synthetic nanocarriers may contain reactive groups to release the modified nucleic acid molecules and/or mmRNA described herein (see International Pub. No.
WO20120952552 and US Pub No. US20120171229, each of which is herein incorporated by reference in their entirety).
[00430] In one embodiment, the synthetic nanocarriers may contain an immunostimulatory agent to enhance the immune response from delivery of the synthetic nanocarrier. As a non-limiting example, the synthetic nanocarrier may comprise a Thl immunostimulatory agent which may enhance a Thl -based response of the immune system (see International Pub No. WO2010123569 and US Pub. No. US20110223201, each of which is herein incorporated by reference in its entirety). [00431] In one embodiment, the synthetic nanocarriers may be formulated for targeted release. In one embodiment, the synthetic nanocarrier is formulated to release the modified nucleic acid molecules and/or mmRNA at a specified pH and/or after a desired time interval. As a non-limiting example, the synthetic nanoparticle may be formulated to release the modified mRNA molecules and/or mmRNA after 24 hours and/or at a pH of 4.5 (see International Pub. Nos. WO2010138193 and WO2010138194 and US Pub Nos. US20110020388 and US20110027217, each of which is herein incorporated by reference in their entirety).
[00432] In one embodiment, the synthetic nanocarriers may be formulated for controlled and/or sustained release of the modified nucleic acid molecules and/or mmRNA described herein. As a non-limiting example, the synthetic nanocarriers for sustained release may be formulated by methods known in the art, described herein and/or as described in International Pub No.
WO2010138192 and US Pub No. 20100303850, each of which is herein incorporated by reference in their entirety.
[00433] In one embodiment, the synthetic nanocarrier may be formulated for use as a vaccine. In one embodiment, the synthetic nanocarrier may encapsulate at least one modified nucleic acid molecule and/or mmRNA which encodes at least one antigen. As a non-limiting example, the synthetic nanocarrier may include at least one antigen and an excipient for a vaccine dosage form (see International Pub No. WO2011150264 and US Pub No. US20110293723, each of which is herein incorporated by reference in their entirety). As another non-limiting example, a vaccine dosage form may include at least two synthetic nanocarriers with the same or different antigens and an excipient (see International Pub No. WO2011150249 and US Pub No. US20110293701, each of which is herein incorporated by reference in their entirety). The vaccine dosage form may be selected by methods described herein, known in the art and/or described in International Pub No. WO2011150258 and US Pub No. US20120027806, each of which is herein incorporated by reference in their entirety).
[00434] In one embodiment, the synthetic nanocarrier may comprise at least one modified nucleic acid molecule and/or mmRNA which encodes at least one adjuvant. In another embodiment, the synthetic nanocarrier may comprise at least one modified nucleic molecule acid and/or mmRNA and an adjuvant. As a non-limiting example, the synthetic nanocarrier comprising and adjuvant may be formulated by the methods described in International Pub No. WO2011150240 and US Pub No. US20110293700, each of which is herein incorporated by reference in its entirety. [00435] In one embodiment, the synthetic nanocarrier may encapsulate at least one modified nucleic acid molecule and/or mmRNA which encodes a peptide, fragment or region from a virus. As a non-limiting example, the synthetic nanocarrier may include, but is not limited to, the nanocarriers described in International Pub No. WO2012024621, WO201202629, WO2012024632 and US Pub No. US20120064110, US20120058153 and US20120058154, each of which is herein incorporated by reference in their entirety.
[00436] In one embodiment, the nanoparticle may be optimized for oral administration. The nanoparticle may comprise at least one cationic biopolymer such as, but not limited to, chitosan or a derivative thereof. As a non-limiting example, the nanoparticle may be formulated by the methods described in U.S. Pub. No. 20120282343; herein incorporated by reference in its entirety.
Polymers, Biodegradable Nanoparticles, and Core-Shell Nanoparticles
[00437] The modified nucleic acid molecules and mmRNA of the invention can be formulated using natural and/or synthetic polymers. Non-limiting examples of polymers which may be used for delivery include, but are not limited to, DYNAMIC POLYCONJUGATE® (Arrowhead Research Corp., Pasadena, CA) formulations from MIRUS® Bio (Madison, WI) and Roche Madison
(Madison, WI), PHASERX™ polymer formulations such as, without limitation, SMARTT
POLYMER TECHNOLOGY™ (Seattle, WA), DMRI/DOPE, poloxamer, VAXFECTIN® adjuvant from Vical (San Diego, CA), chitosan, cyclodextrin from Calando Pharmaceuticals (Pasadena, CA), dendrimers and poly(lactic-co-glycolic acid) (PLGA) polymers, RONDEL™
(RNAi/Oligonucleotide Nanoparticle Delivery) polymers (Arrowhead Research Corporation, Pasadena, CA) and pH responsive co-block polymers such as, but not limited to, PHASERX™ (Seattle, WA).
[00438] A non-limiting example of chitosan formulation includes a core of positively charged chitosan and an outer portion of negatively charged substrate (U.S. Pub. No. 20120258176; herein incorporated by reference in its entirety). Chitosan includes, but is not limited to N-trimethyl chitosan, mono-N-carboxymethyl chitosan (MCC), N-palmitoyl chitosan (NPCS), EDTA-chitosan, low molecular weight chitosan, chitosan derivatives, or combinations thereof.
[00439] In one embodiment, the polymers used in the present invention have undergone processing to reduce and/or inhibit the attachement of unwanted substances such as, but not limited to, bacteria, to the surface of the polymer. The polymer may be processed by methods known and/or described in the art and/or described in International Pub. No. WO2012150467, herein incorporated by reference in its entirety.
[00440] A non- limiting example of PLGA formulations include, but are not limited to, PLGA injectable depots (e.g., ELIGARD® which is formed by dissolving PLGA in 66% N-methyl-2- pyrrolidone (NMP) and the remainder being aqueous solvent and leuprolide. Once injected, the PLGA and leuprolide peptide precipitates into the subcutaneous space).
[00441] Many of these polymer approaches have demonstrated efficacy in delivering
oligonucleotides in vivo into the cell cytoplasm (reviewed in deFougerolles Hum Gene Ther. 2008 19: 125-132; herein incorporated by reference in its entirety). Two polymer approaches that have yielded robust in vivo delivery of nucleic acids, in this case with small interfering RNA (siRNA), are dynamic polyconjugates and cyclodextrin-based nanoparticles. The first of these delivery approaches uses dynamic polyconjugates and has been shown in vivo in mice to effectively deliver siRNA and silence endogenous target mRNA in hepatocytes (Rozema et al, Proc Natl Acad Sci U S A. 2007 104: 12982-12887; herein incorporated by reference in its entirety). This particular approach is a multicomponent polymer system whose key features include a membrane-active polymer to which nucleic acid, in this case siRNA, is covalently coupled via a disulfide bond and where both PEG (for charge masking) and ^-acetylgalactosamine (for hepatocyte targeting) groups are linked via pH- sensitive bonds (Rozema et al, Proc Natl Acad Sci U S A. 2007 104: 12982-12887; herein incorporated by reference in its entirety). On binding to the hepatocyte and entry into the endosome, the polymer complex disassembles in the low-pH environment, with the polymer exposing its positive charge, leading to endosomal escape and cytoplasmic release of the siRNA from the polymer. Through replacement of the ^-acetylgalactosamine group with a mannose group, it was shown one could alter targeting from asialoglycoprotein receptor-expressing hepatocytes to sinusoidal endothelium and Kupffer cells. Another polymer approach involves using transferrin- targeted cyclodextrin-containing polycation nanoparticles. These nanoparticles have demonstrated targeted silencing of the EWS-FLI1 gene product in transferrin receptor-expressing Ewing's sarcoma tumor cells (Hu-Lieskovan et al., Cancer Res.2005 65: 8984-8982; herein incorporated by reference in its entirety) and siRNA formulated in these nanoparticles was well tolerated in non-human primates (Heidel et al., Proc Natl Acad Sci USA 2007 104:5715-21; herein incorporated by reference in its entirety). Both of these delivery strategies incorporate rational approaches using both targeted delivery and endosomal escape mechanisms. [00442] The polymer formulation can permit the sustained or delayed release of modified nucleic acid molecules or mmRNA (e.g., following intramuscular or subcutaneous injection). The altered release profile for the modified nucleic acid molecule or mmRNA can result in, for example, translation of an encoded protein over an extended period of time. The polymer formulation may also be used to increase the stability of the modified nucleic acid molecule or mmRNA.
Biodegradable polymers have been previously used to protect nucleic acids other than mmRNA from degradation and been shown to result in sustained release of payloads in vivo (Rozema et al, Proc Natl Acad Sci U S A. 2007 104: 12982-12887; Sullivan et al, Expert Opin Drug Deliv. 2010 7: 1433-1446; Convertine et al., Biomacromolecules. 2010 Oct 1; Chu et al., Acc Chem Res. 2012 Jan 13; Manganiello et al., Biomaterials. 2012 33:2301-2309; Benoit et al, Biomacromolecules. 2011 12:2708-2714; Singha et al, Nucleic Acid Ther. 2011 2:133-147; deFougerolles Hum Gene Ther. 2008 19: 125-132; Schaffert and Wagner, Gene Ther. 2008 16:1131-1138; Chaturvedi et al, Expert Opin Drug Deliv. 2011 8: 1455-1468; Davis, Mol Pharm. 2009 6:659-668; Davis, Nature 2010 464:1067-1070; each of which is herein incorporated by reference in its entirety).
[00443] In one embodiment, the pharmaceutical compositions may be sustained release
formulations. In a further embodiment, the sustained release formulations may be for subcutaneous delivery. Sustained release formulations may include, but are not limited to, PLGA microspheres, ethylene vinyl acetate (EVAc), poloxamer, GELSITE® (Nanotherapeutics, Inc. Alachua, FL), HYLENEX® (Halozyme Therapeutics, San Diego CA), surgical sealants such as fibrinogen polymers (Ethicon Inc. Cornelia, GA), TISSELL® (Baxter International, Inc Deerfield, IL), PEG- based sealants, and COSEAL® (Baxter International, Inc Deerfield, IL).
[00444] As a non-limiting example modified mRNA may be formulated in PLGA microspheres by preparing the PLGA microspheres with tunable release rates (e.g., days and weeks) and
encapsulating the modified mRNA in the PLGA microspheres while maintaining the integrity of the modified mRNA during the encapsulation process. EVAc are non-biodegradeable, biocompatible polymers which are used extensively in pre-clinical sustained release implant applications (e.g., extended release products Ocusert a pilocarpine ophthalmic insert for glaucoma or progestasert a sustained release progesterone intrauterine deivce; transdermal delivery systems Testoderm, Duragesic and Selegiline; catheters). Poloxamer F-407 NF is a hydrophilic, non-ionic surfactant triblock copolymer of polyoxyethylene-polyoxypropylene-polyoxyethylene having a low viscosity at temperatures less than 5°C and forms a solid gel at temperatures greater than 15°C. PEG-based surgical sealants comprise two synthetic PEG components mixed in a delivery device which can be prepared in one minute, seals in 3 minutes and is reabsorbed within 30 days. GELSITE® and natural polymers are capable of in-situ gelation at the site of administration. They have been shown to interact with protein and peptide therapeutic candidates through ionic ineraction to provide a stabilizing effect.
[00445] Polymer formulations can also be selectively targeted through expression of different ligands as exemplified by, but not limited by, folate, transferrin, and N-acetylgalactosamine
(GalNAc) (Benoit et al., Biomacromolecules. 2011 12:2708-2714; Rozema et al., Proc Natl Acad Sci U S A. 2007 104: 12982-12887; Davis, Mol Pharm. 2009 6:659-668; Davis, Nature 2010 464:1067- 1070; each of which is herein incorporated by reference in its entirety).
[00446] The modified nucleic acid molecules and mmRNA of the invention may be formulated with or in a polymeric compound. The polymer may include at least one polymer such as, but not limited to, polyethenes, polyethylene glycol (PEG), poly(l-lysine)(PLL), PEG grafted to PLL, cationic lipopolymer, biodegradable cationic lipopolymer, polyethyleneimine (PEI), cross-linked branched poly(alkylene imines), a polyamine derivative, a modified poloxamer, a biodegradable polymer, elastic biodegradable polymer, biodegradable block copolymer, biodegradable random copolymer, biodegradable polyester copolymer, biodegradable polyester block copolymer, biodegradable polyester block random copolymer, multiblock copolymers, linear biodegradable copolymer, poly[a-(4-aminobutyl)-L-glycolic acid) (PAGA), biodegradable cross-linked cationic multi-block copolymers, polycarbonates, polyanhydrides, polyhydroxyacids, polypropylfumerates, polycaprolactones, polyamides, polyacetals, polyethers, polyesters, poly(orthoesters),
polycyanoacrylates, polyvinyl alcohols, polyurethanes, polyphosphazenes, polyacrylates, polymethacrylates, polycyanoacrylates, polyureas, polystyrenes, polyamines, polylysine,
poly(ethylene imine), poly(serine ester), poly(L-lactide-co-L-lysine), poly(4-hydroxy-L-proline ester), acrylic polymers, amine-containing polymers, dextran polymers, dextran polymer derivatives or combinations thereof .
[00447] As a non-limiting example, the modified nucleic acid molecules or mmRNA of the invention may be formulated with the polymeric compound of PEG grafted with PLL as described in U.S. Pat. No. 6,177,274; herein incorporated by reference in its entirety. The formulation may be used for transfecting cells in vitro or for in vivo delivery of the modified nucleic acid molecules and mmRNA. In another example, the modified nucleic acid molecules and mmRNA may be suspended in a solution or medium with a cationic polymer, in a dry pharmaceutical composition or in a solution that is capable of being dried as described in U.S. Pub. Nos. 20090042829 and
20090042825; each of which are herein incorporated by reference in their entireties.
[00448] As another non-limiting example the modified nucleic acid molecules or mmRNA of the invention may be formulated with a PLGA-PEG block copolymer (see US Pub. No.
US20120004293 and US Pat No. 8,236,330, each of which are herein incorporated by reference in their entireties) or PLGA-PEG-PLGA block copolymers (See U.S. Pat. No. 6,004,573, herein incorporated by reference in its entirety). As a non- limiting example, the modified nucleic acid molecules or mmRNA of the invention may be formulated with a diblock copolymer of PEG and PLA or PEG and PLGA (see US Pat No 8,246,968, herein incorporated by reference in its entirety).
[00449] A polyamine derivative may be used to deliver nucleic acid molecules and/or mmRNA or to treat and/or prevent a disease or to be included in an implantable or injectable device (U.S. Pub. No. 20100260817 herein incorporated by reference in its entirety). As a non-limiting example, a pharmaceutical composition may include the modified nucleic acid molecules and mmRNA and the polyamine derivative described in U.S. Pub. No. 20100260817 (the contents of which are
incorporated herein by reference in its entirety). As a non-limiting example the modified nucleic acids or mmRNA of the present invention may be delivered using a polyaminde polymer such as, but not limited to, a polymer comprising a 1,3-dipolar addition polymer prepared by combining a carbohydrate diazide monomer with a dilkyne unite comprising oligoamines (U.S. Pat. No.
8,236,280; herein incorporated by reference in its entirety).
[00450] The modified nucleic acid molecules and/or mmRNA of the invention may be formulated with at least one acrylic polymer. Acrylic polymers include but are not limited to, acrylic acid, methacrylic acid, acrylic acid and methacrylic acid copolymers, methyl methacrylate copolymers, ethoxyethyl methacrylates, cyanoethyl methacrylate, amino alkyl methacrylate copolymer, poly(acrylic acid), poly(methacrylic acid), polycyanoacrylates and combinations thereof.
[00451] In one embodiment, the modified nucleic acid molecules and/or mmRNA of the present invention may be formulated with at least one polymer and/or derivatives thereof described in International Publication Nos. WO2011115862, WO2012082574 and WO2012068187 and U.S. Pub. No. 20120283427, each of which are herein incorporated by reference in their entireties. In another embodiment, the modified nucleic acid molecules or mmRNA of the present invention may be formulated with a polymer of formula Z as described in WO2011115862, herein incorporated by reference in its entirety. In yet another embodiment, the modified nucleic acid molecules or mmRNA may be formulated with a polymer of formula Z, Z' or Z" as described in International Pub. Nos. WO2012082574 or WO2012068187, each of which are herein incorporated by reference in their entireties. The polymers formulated with the modified nucleic acids and/or modified mRNA of the present invention may be synthesized by the methods described in International Pub. Nos. WO2012082574 or WO2012068187, each of which are herein incorporated by reference in their entireties.
[00452] Formulations of modified nucleic acid molecules and/or mmRNA of the invention may include at least one amine-containing polymer such as, but not limited to polylysine, polyethylene imine, poly(amidoamine) dendrimers or combinations thereof.
[00453] For example, the modified nucleic acid molecules and/or mmRNA of the invention may be formulated in a pharmaceutical compound including a poly(alkylene imine), a biodegradable cationic lipopolymer, a biodegradable block copolymer, a biodegradable polymer, or a
biodegradable random copolymer, a biodegradable polyester block copolymer, a biodegradable polyester polymer, a biodegradable polyester random copolymer, a linear biodegradable copolymer, PAGA, a biodegradable cross-linked cationic multi-block copolymer or combinations thereof. The biodegradable cationic lipopolymer may be made by methods known in the art and/or described in U.S. Pat. No. 6,696,038, U.S. App. Nos. 20030073619 and 20040142474 each of which is herein incorporated by reference in their entireties. The poly(alkylene imine) may be made using methods known in the art and/or as described in U.S. Pub. No. 20100004315, herein incorporated by reference in its entirety. The biodegradabale polymer, biodegradable block copolymer, the biodegradable random copolymer, biodegradable polyester block copolymer, biodegradable polyester polymer, or biodegradable polyester random copolymer may be made using methods known in the art and/or as described in U.S. Pat. Nos. 6,517,869 and 6,267,987, the contents of which are each incorporated herein by reference in their entirety. The linear biodegradable copolymer may be made using methods known in the art and/or as described in U.S. Pat. No.
6,652,886. The PAGA polymer may be made using methods known in the art and/or as described in U.S. Pat. No. 6,217,912 herein incorporated by reference in its entirety. The PAGA polymer may be copolymerized to form a copolymer or block copolymer with polymers such as but not limited to, poly-L-lysine, polyargine, polyornithine, histones, avidin, protamines, polylactides and poly(lactide- co-glycolides). The biodegradable cross-linked cationic multi-block copolymers may be made my methods known in the art and/or as described in U.S. Pat. No. 8,057,821 or U.S. Pub. No. 2012009145 each of which are herein incorporated by reference in their entireties. For example, the multi-block copolymers may be synthesized using linear polyethyleneimine (LPEI) blocks which have distinct patterns as compared to branched polyethyleneimines. Further, the composition or pharmaceutical composition may be made by the methods known in the art, described herein, or as described in U.S. Pub. No. 20100004315 or U.S. Pat. Nos. 6,267,987 and 6,217,912 each of which are herein incorporated by reference in their entireties.
[00454] The modified nucleic acid molecules and mmRNA of the invention may be formulated with at least one degradable polyester which may contain polycationic side chains. Degradeable polyesters include, but are not limited to, poly(serine ester), poly(L-lactide-co-L-lysine), poly(4- hydroxy-L-proline ester), and combinations thereof. In another embodiment, the degradable polyesters may include a PEG conjugation to form a PEGylated polymer.
[00455] The modified nucleic acid molecules and mmRNA of the invention may be formulated with at least one crosslinkable polyester. Crosslinkable polyesters include those known in the art and described in US Pub. No. 20120269761, herein incorporated by reference in its entirety.
[00456] In one embodiment, the polymers described herein may be conjugated to a lipid- terminating PEG. As a non-limiting example, PLGA may be conjugated to a lipid-terminating PEG forming PLGA-DSPE-PEG. As another non-limiting example, PEG conjugates for use with the present invention are described in International Publication No. WO2008103276, herein
incorporated by reference in its entirety. The polymers may be conjugated using a ligand conjugate such as, but not limited to, the conjugates described in U.S. Pat. No. 8,273,363, herein incorporated by reference in its entirety.
[00457] In one embodiment, the modified nucleic acid molecules and/or mmRNA described herein may be conjugated with another compound. Non-limiting examples of conjugates are described in US Patent Nos. 7,964,578 and 7,833,992, each of which are herein incorporated by reference in their entireties. In another embodiment, modified RNA of the present invention may be conjugated with conjugates of formula 1-122 as described in US Patent Nos. 7,964,578 and 7,833,992, each of which are herein incorporated by reference in their entireties. The modified RNA described herein may be conjugated with a metal such as, but not limited to, gold. (See e.g., Giljohann et al. Journ. Amer. Chem. Soc. 2009 131(6): 2072-2073; herein incorporated by reference in its entirety). In another embodiment, the modified nucleic acid molecules and/or mmRNA described herein may be conjugated and/or encapsulated in gold-nanoparticles. (Interantional Pub. No. WO201216269 and U.S. Pub. No. 20120302940; each of which is herein incorporated by reference in its entirety).
[00458] As described in U.S. Pub. No. 20100004313, herein incorporated by reference in its entirety, a gene delivery composition may include a nucleotide sequence and a poloxamer. For example, the modified nucleic acid and mmRNA of the present inveition may be used in a gene delivery composition with the poloxamer described in U.S. Pub. No. 20100004313.
[00459] In one embodiment, the polymer formulation of the present invention may be stabilized by contacting the polymer formulation, which may include a cationic carrier, with a cationic lipopolymer which may be covalently linked to cholesterol and polyethylene glycol groups. The polymer formulation may be contacted with a cationic lipopolymer using the methods described in U.S. Pub. No. 20090042829 herein incorporated by reference in its entirety. The cationic carrier may include, but is not limited to, polyethylenimine, poly(trimethylenimine),
poly(tetramethylenimine), polypropylenimine, aminoglycoside-polyamine, dideoxy-diamino-b- cyclodextrin, spermine, spermidine, poly(2-dimethylamino)ethyl methacrylate, poly(lysine), poly(histidine), poly(arginine), cationized gelatin, dendrimers, chitosan, l,2-Dioleoyl-3- Trimethylammonium-Propane (DOTAP), N-[ 1 -(2,3-dioleoyloxy)propyl]-N,N,N- trimethylammonium chloride (DOTMA), l-[2-(oleoyloxy)ethyl]-2-oleyl-3-(2- hydroxyethyl)imidazolinium chloride (DOTIM), 2,3-dioleyloxy-N-[2(sperminecarboxamido)ethyl]- N,N-dimethyl-l-propanaminium trifluoroacetate (DOSPA), 3B-[N— (Ν',Ν'-Dimethylaminoethane)- carbamoyl]Cholesterol Hydrochloride (DC-Cholesterol HC1) diheptadecylamidoglycyl spermidine (DOGS), N,N-distearyl-N,N-dimethylammonium bromide (DDAB), N-(l,2-dimyristyloxyprop-3- yl)-N,N-dimethyl-N-hydroxyethyl ammonium bromide (DMRIE), N,N-dioleyl-N,N- dimethylammonium chloride DODAC) and combinations thereof.
[00460] The modified nucleic acid molecules and/or mmRNA of the invention may be formulated in a polyplex of one or more polymers (U.S. Pub. No. 20120237565 and 20120270927; each of which is herein incorporated by reference in its entirety). In one embodiment, the polyplex comprises two or more cationic polymers. The catioinic polymer may comprise a poly(ethylene imine) (PEI) such as linear PEL
[00461] The modified nucleic acid molecules and mmRNA of the invention can also be formulated as a nanoparticle using a combination of polymers, lipids, and/or other biodegradable agents, such as, but not limited to, calcium phosphate. Components may be combined in a core-shell, hybrid, and/or layer-by-layer architecture, to allow for fine-tuning of the nanoparticle so to delivery of the modified nucleic acid molecule and mmRNA may be enhanced (Wang et al., Nat Mater. 2006 5:791-796; Fuller et al, Biomaterials. 2008 29: 1526-1532; DeKoker et al, Adv Drug Deliv Rev. 2011 63:748-761; Endres et al, Biomaterials. 2011 32:7721-7731; Su et al, Mol Pharm. 2011 Jun 6;8(3):774-87; each of which is herein incorporated by reference in its entirety). As a non-limiting example, the nanoparticle may comprise a plurality of polymers such as, but not limited to hydrophilic-hydrophobic polymers (e.g., PEG-PLGA), hydrophobic polymers (e.g., PEG) and/or hydrophilic polymers (International Pub. No. WO20120225129; herein incorporated by reference in its entirety).
[00462] Biodegradable calcium phosphate nanoparticles in combination with lipids and/or polymers have been shown to deliver modified nucleic acid molecules and mmRNA in vivo. In one embodiment, a lipid coated calcium phosphate nanoparticle, which may also contain a targeting ligand such as anisamide, may be used to deliver the modified nucleic acid molecule and mmRNA of the present invention. For example, to effectively deliver siRNA in a mouse metastatic lung model a lipid coated calcium phosphate nanoparticle was used (Li et al., J Contr Rel. 2010 142: 416- 421; Li et al, J Contr Rel. 2012 158: 108-114; Yang et al, Mol Ther. 2012 20:609-615; herein incorporated by refereince in its entirety). This delivery system combines both a targeted nanoparticle and a component to enhance the endosomal escape, calcium phosphate, in order to improve delivery of the siRNA.
[00463] In one embodiment, calcium phosphate with a PEG-polyanion block copolymer may be used to deliver modified nucleic acid molecules and mmRNA (Kazikawa et al., J Contr Rel. 2004 97:345-356; Kazikawa et al., J Contr Rel. 2006 111 :368-370; herein incorporated by reference in its entirety).
[00464] In one embodiment, a PEG-charge-conversional polymer (Pitella et al., Biomaterials. 2011 32:3106-3114) may be used to form a nanoparticle to deliver the modified nucleic acid molecules and mmRNA of the present invention. The PEG-charge-conversional polymer may improve upon the PEG-polyanion block copolymers by being cleaved into a polycation at acidic pH, thus enhancing endosomal escape.
[00465] The use of core-shell nanoparticles has additionally focused on a high-throughput approach to synthesize cationic cross-linked nanogel cores and various shells (Siegwart et al., Proc Natl Acad Sci U S A. 2011 108: 12996-13001). The complexation, delivery, and internalization of the polymeric nanoparticles can be precisely controlled by altering the chemical composition in both the core and shell components of the nanoparticle. For example, the core-shell nanoparticles may efficiently deliver siR A to mouse hepatocytes after they covalently attach cholesterol to the nanoparticle.
[00466] In one embodiment, a hollow lipid core comprising a middle PLGA layer and an outer neutral lipid layer containg PEG may be used to delivery of the modified nucleic acid molecules and mmRNA of the present invention. As a non-limiting example, in mice bearing a luciferease- expressing tumor, it was determined that the lipid-polymer-lipid hybrid nanoparticle significantly suppressed luciferase expression, as compared to a conventional lipoplex (Shi et al, Angew Chem Int Ed. 2011 50:7027-7031; herein incorporated by reference in its entirety).
[00467] In one embodiment, the lipid nanoparticles may comprise a core of the modified nucleic acid molecules disclosed herein and a polymer shell. The polymer shell may be any of the polymers described herein and are known in the art. In an additional embodiment, the polymer shell may be used to protect the modified nucleic acids in the core.
[00468] Core-shell nanoparticles for use with the modified nucleic acid molecules of the present invention are described and may be formed by the methods described in U.S. Pat. No. 8,313,777 herein incorporated by reference in its entirety.
[00469] In one embodiment, the core-shell nanoparticles may comprise a core of the modified nucleic acid molecules disclosed herein and a polymer shell. The polymer shell may be any of the polymers described herein and are known in the art. In an additional embodiment, the polymer shell may be used to protect the modified nucleic acid molecules in the core.
Peptides and Proteins
[00470] The modified nucleic acid molecules and mmRNA of the invention can be formulated with peptides and/or proteins in order to increase transfection of cells by the modified nucleic acid molecules or mmRNA. In one embodiment, peptides such as, but not limited to, cell penetrating peptides and proteins and peptides that enable intracellular delivery may be used to deliver pharmaceutical formulations. A non-limiting example of a cell penetrating peptide which may be used with the pharmaceutical formulations of the present invention include a cell-penetrating peptide sequence attached to polycations that facilitates delivery to the intracellular space, e.g., HIV- derived TAT peptide, penetratins, transportans, or hCT derived cell-penetrating peptides (see, e.g., Caron et al, Mol. Ther. 3(3):310-8 (2001); Langel, Cell-Penetrating Peptides: Processes and Applications (CRC Press, Boca Raton FL, 2002); El-Andaloussi et al., Curr. Pharm. Des.
11(28):3597-611 (2003); and Deshayes et al, Cell. Mol. Life Sci. 62(16): 1839-49 (2005), all of which are incorporated herein by reference). The compositions can also be formulated to include a cell penetrating agent, e.g., liposomes, which enhance delivery of the compositions to the intracellular space. Modified nucleic acid molecules and mmRNA of the invention may be complexed to peptides and/or proteins such as, but not limited to, peptides and/or proteins from Aileron Therapeutics (Cambridge, MA) and Permeon Biologies (Cambridge, MA) in order to enable intracellular delivery (Cronican et al., ACS Chem. Biol. 2010 5:747-752; McNaughton et al., Proc. Natl. Acad. Sci. USA 2009 106:6111-6116; Sawyer, Chem Biol Drug Des. 2009 73:3-6; Verdine and Hilinski, Methods Enzymol. 2012;503:3-33; all of which are herein incorporated by reference in its entirety).
[00471] In one embodiment, the cell-penetrating polypeptide may comprise a first domain and a second domain. The first domain may comprise a supercharged polypeptide. The second domain may comprise a protein-binding partner. As used herein, "protein-binding partner" includes, but are not limited to, antibodies and functional fragments thereof, scaffold proteins, or peptides. The cell- penetrating polypeptide may further comprise an intracellular binding partner for the protein-binding partner. The cell-penetrating polypeptide may be capable of being secreted from a cell where the modified nucleic acid molecules or mmRNA may be introduced.
[00472] Formulations of the including peptides or proteins may be used to increase cell transfection by the modified nucleic acid molecule or mmRNA, alter the biodistribution of the modified nucleic acid molecule or mmRNA (e.g., by targeting specific tissues or cell types), and/or increase the translation of encoded protein. (See e.g., International Pub. No. WO2012110636; herein
incorporated by reference in its entirety).
Cells
[00473] The modified nucleic acid moleclue and mmRNA of the invention can be transfected ex vivo into cells, which are subsequently transplanted into a subject. As non-limiting examples, the pharmaceutical compositions may include red blood cells to deliver modified RNA to liver and myeloid cells, virosomes to deliver modified nucleic acid molecules and mmRNA in virus-like particles (VLPs), and electroporated cells such as, but not limited to, from MAXCYTE®
(Gaithersburg, MD) and from ERYTECH® (Lyon, France) to deliver modified RNA. Examples of use of red blood cells, viral particles and electroporated cells to deliver payloads other than mmRNA have been documented (Godfrin et al., Expert Opin Biol Ther. 2012 12: 127-133; Fang et al., Expert Opin Biol Ther. 2012 12:385-389; Hu et al, Proc Natl Acad Sci U S A. 2011 108: 10980-10985; Lund et al, Pharm Res. 2010 27:400-420; Huckriede et al, J Liposome Res. 2007;17:39-47; Cusi, Hum Vaccin. 2006 2: 1-7; de Jonge et al., Gene Ther. 2006 13:400-41 1; all of which are herein incorporated by reference in its entirety). The modified nucleic acid molecules and mmRNA may be delivered in synthetic VLPs synthesized by the methods described in International Pub No.
WO2011085231 and US Pub No. 20110171248, each of which are herein incorporated by reference in their entireties.
[00474] Cell-based formulations of the modified nucleic acid molecules and mmRNA of the invention may be used to ensure cell transfection (e.g., in the cellular carrier), alter the
biodistribution of the modified nucleic acid molecule or mmRNA (e.g., by targeting the cell carrier to specific tissues or cell types), and/or increase the translation of encoded protein.
Introduction into cells
[00475] A variety of methods are known in the art and suitable for introduction of nucleic acid into a cell, including viral and non-viral mediated techniques. Examples of typical non-viral mediated techniques include, but are not limited to, electroporation, calcium phosphate mediated transfer, nucleofection, sonoporation, heat shock, magneto fection, liposome mediated transfer,
microinjection, microprojectile mediated transfer (nanoparticles), cationic polymer mediated transfer (DEAE-dextran, polyethylenimine, polyethylene glycol (PEG) and the like) or cell fusion.
[00476] The technique of sonoporaiton, or cellular sonication, is the use of sound (e.g., ultrasonic frequencies) for modifying the permeability of the cell plasma membrane. Sonoporation methods are known to those in the art and are taught for example as it relates to bacteria in US Patent Publication 20100196983 and as it relates to other cell types in, for example, US Patent Publication 20100009424, each of which are incorporated herein by reference in their entirety.
[00477] Electroporation techniques are also well known in the art. In one embodiment, modified nucleic acid molecules or mmRNA may be delivered by electroporation as described in Example 8. Hyaluronidase
[00478] The intramuscular or subcutaneous localized injection of modified nucleic acid molecules or mmRNA of the invention can include hyaluronidase, which catalyzes the hydrolysis of hyaluronan. By catalyzing the hydrolysis of hyaluronan, a constituent of the interstitial barrier, hyaluronidase lowers the viscosity of hyaluronan, thereby increasing tissue permeability (Frost, Expert Opin. Drug Deliv. (2007) 4:427-440; herein incorporated by reference in its entirety). It is useful to speed their dispersion and systemic distribution of encoded proteins produced by transfected cells. Alternatively, the hyaluronidase can be used to increase the number of cells exposed to a modified nucleic acid molecule or mmR A of the invention administered
intramuscularly or subcutaneously.
Nanoparticle Mimics
[00479] The modified nucleic acid molecules and mmRNA of the invention may be encapsulated within and/or absorbed to a nanoparticle mimic. A nanoparticle mimic can mimic the delivery function organisms or particles such as, but not limited to, pathogens, viruses, bacteria, fungus, parasites, prions and cells. As a non-limiting example the modified mRNA of the invention may be encapsulated in a non-viron particle which can mimic the delivery function of a virus (see
International Pub. No. WO2012006376 herein incorporated by reference in its entirety).
Nanotubes
[00480] The modified nucleic acid molecules or mmRNA of the invention can be attached or otherwise bound to at least one nanotube such as, but not limited to, rosette nanotubes, rosette nanotubes having twin bases with a linker, carbon nanotubes and/or single-walled carbon nanotubes, The modified nucleic acid molecules or mmRNA may be bound to the nanotubes through forces such as, but not limited to, steric, ionic, covalent and/or other forces.
[00481] In one embodiment, the nanotube can release one or more modified nucleic acid molecule or mmRNA into cells. The size and/or the surface structure of at least one nanotube may be altered so as to govern the interaction of the nanotubes within the body and/or to attach or bind to the modified nucleic acid molecule or mmRNA disclosed herein. In one embodiment, the building block and/or the functional groups attached to the building block of the at least one nanotube may be altered to adjust the dimensions and/or properties of the nanotube. As a non-limiting example, the length of the nanotubes may be altered to hinder the nanotubes from passing through the holes in the walls of normal blood vessels but still small enough to pass through the larger holes in the blood vessels of tumor tissue.
[00482] In one embodiment, at least one nanotube may also be coated with delivery enhancing compounds including polymers, such as, but not limited to, polyethylene glycol. In another embodiment, at least one nanotube and/or the modified mRNA may be mixed with pharmaceutically acceptable excipients and/or delivery vehicles. [00483] In one embodiment, the modified mRNA are attached and/or otherwise bound to at least one rosette nanotube. The rosette nanotubes may be formed by a process known in the art and/or by the process described in International Publication No. WO2012094304, herein incorporated by reference in its entirety. At least one modified mRNA may be attached and/or otherwise bound to at least one rosette nanotube by a process as described in International Publication No.
WO2012094304, herein incorporated by reference in its entirety, where rosette nanotubes or modules forming rosette nanotubes are mixed in aqueous media with at least one modified mRNA under conditions which may cause at least one modified mRNA to attach or otherwise bind to the rosette nanotubes.
[00484] In one embodiment, the modified nucleic acid molecule or mmRNA may be attached to and/or otherwise bound to at least one carbon nanotube. As a non-limiting example, the modified nucleic acid molecule or mmRNA may be bound to a linking agent and the linked agent may be bound to the carbon nanotube (See e.g., U.S. Pat No. 8,246,995; herein incorporated by reference in its entirety). The carbon nanotube may be a single -walled nanotube (See e.g., U.S. Pat No.
8,246,995; herein incorporated by reference in its entirety).
Conjugates
[00485] The modified nucleic acids molecules and mmRNA of the invention include conjugates, such as a modified nucleic acid molecule or mmRNA covalently linked to a carrier or targeting group, or including two encoding regions that together produce a fusion protein (e.g., bearing a targeting group and therapeutic protein or peptide).
[00486] The conjugates of the invention include a naturally occurring substance, such as a protein (e.g., human serum albumin (HSA), low-density lipoprotein (LDL), high-density lipoprotein (HDL), or globulin); an carbohydrate (e.g., a dextran, pullulan, chitin, chitosan, inulin, cyclodextrin or hyaluronic acid); or a lipid. The ligand may also be a recombinant or synthetic molecule, such as a synthetic polymer, e.g., a synthetic polyamino acid, an oligonucleotide (e.g. an aptamer). Examples of polyamino acids include polyamino acid is a polylysine (PLL), poly L-aspartic acid, poly L- glutamic acid, styrene-maleic acid anhydride copolymer, poly(L-lactide-co-glycolied) copolymer, divinyl ether-maleic anhydride copolymer, N-(2-hydroxypropyl)methacrylamide copolymer (HMPA), polyethylene glycol (PEG), polyvinyl alcohol (PVA), polyurethane, poly(2-ethylacryllic acid), N-isopropylacrylamide polymers, or polyphosphazine. Example of polyamines include:
polyethylenimine, polylysine (PLL), spermine, spermidine, polyamine, pseudopeptide-polyamine, peptidomimetic polyamine, dendrimer polyamine, arginine, amidine, protamine, cationic lipid, cationic porphyrin, quaternary salt of a polyamine, or an alpha helical peptide.
[00487] Representative U.S. patents that teach the preparation of polynucleotide conjugates, particularly to RNA, include, but are not limited to, U.S. Pat. Nos. 4,828,979; 4,948,882; 5,218,105;
5,525,465; 5,541,313; 5,545,730; 5,552,538; 5,578,717, 5,580,731; 5,591,584 5,109,124; 5,118,802; 5,138,045; 5,414,077; 5,486,603; 5,512,439; 5,578,718; 5,608,046; 4,587,044 4,605,735;
4,667,025; 4,762,779; 4,789,737; 4,824,941 ; 4,835,263; 4,876,335; 4,904,582 4,958,013;
5,082,830; 5,112,963; 5,214,136; 5,082,830; 5,112,963; 5,214,136; 5,245,022 5,254,469;
5,258,506; 5,262,536; 5,272,250; 5,292,873; 5,317,098; 5,371,241, 5,391,723 5,416,203, 5,451,463; 5,510,475; 5,512,667; 5,514,785; 5,565,552; 5,567,810; 5,574,142; 5,585,481; 5,587,371;
5,595,726; 5,597,696; 5,599,923; 5,599,928 and 5,688,941; 6,294,664; 6,320,017; 6,576,752;
6,783,931; 6,900,297; 7,037,646; each of which is herein incorporated by reference in their entireties.
[00488] In one embodiment, the conjugate of the present invention may function as a carrier for the modified nucleic acid molecules and mmRNA of the present invention. The conjugate may comprise a cationic polymer such as, but not limited to, polyamine, polylysine, polyalkylenimine, and polyethylenimine which may be grafted to with poly(ethylene glycol). As a non- limiting example, the conjugate may be similar to the polymeric conjugate and the method of synthesizing the polymeric conjugate described in U.S. Pat. No. 6,586,524 herein incorporated by reference in its entirety.
[00489] The conjugates can also include targeting groups, e.g., a cell or tissue targeting agent, e.g., a lectin, glycoprotein, lipid or protein, e.g., an antibody, that binds to a specified cell type such as a kidney cell. A targeting group can be a thyrotropin, melanotropin, lectin, glycoprotein, surfactant protein A, Mucin carbohydrate, multivalent lactose, multivalent galactose, N-acetyl-galactosamine, N-acetyl-gulucosamine multivalent mannose, multivalent fucose, glycosylated polyaminoacids, multivalent galactose, transferrin, bisphosphonate, polyglutamate, polyaspartate, a lipid, cholesterol, a steroid, bile acid, folate, vitamin B12, biotin, an RGD peptide, an RGD peptide mimetic or an aptamer.
[00490] Targeting groups can be proteins, e.g., glycoproteins, or peptides, e.g., molecules having a specific affinity for a co-ligand, or antibodies e.g., an antibody, that binds to a specified cell type such as a cancer cell, endothelial cell, or bone cell. Targeting groups may also include hormones and hormone receptors. They can also include non-peptidic species, such as lipids, lectins, carbohydrates, vitamins, cofactors, multivalent lactose, multivalent galactose, N-acetyl- galactosamine, N-acetyl-gulucosamine multivalent mannose, multivalent fucose, or aptamers. The ligand can be, for example, a lipopolysaccharide, or an activator of p38 MAP kinase.
[00491] The targeting group can be any ligand that is capable of targeting a specific receptor.
Examples include, without limitation, folate, GalNAc, galactose, mannose, mannose-6P, apatamers, integrin receptor ligands, chemokine receptor ligands, transferrin, biotin, serotonin receptor ligands, PSMA, endothelin, GCPII, somatostatin, LDL, and HDL ligands. In particular embodiments, the targeting group is an aptamer. The aptamer can be unmodified or have any combination of modifications disclosed herein.
[00492] In one embodiment, pharmaceutical compositions of the present invention may include chemical modifications such as, but not limited to, modifications similar to locked nucleic acids.
[00493] Representative U.S. Patents that teach the preparation of locked nucleic acid (LNA) such as those from Santaris, include, but are not limited to, the following: U.S. Pat. Nos. 6,268,490;
6,670,461; 6,794,499; 6,998,484; 7,053,207; 7,084,125; and 7,399,845, each of which is herein incorporated by reference in its entirety.
[00494] Representative U.S. patents that teach the preparation of PNA compounds include, but are not limited to, U.S. Pat. Nos. 5,539,082; 5,714,331; and 5,719,262, each of which is herein incorporated by reference. Further teaching of PNA compounds can be found, for example, in Nielsen et al, Science, 1991, 254, 1497-1500.
[00495] Some embodiments featured in the invention include modified nucleic acids or mmRNA with phosphorothioate backbones and oligonucleosides with other modified backbones, and in particular— CH2— NH— CH2— , ~CH2~N(CH3)~0~CH2~[known as a methylene (methylimino) or MMI backbone], -CH2-0-N(CH3)~CH2-, --CH2--N(CH3)--N(CH3)-CH2-- and -N(CH3)-CH2- CH2~[wherein the native phosphodiester backbone is represented as— O— P(0)2— O— CH2— ] of the above-referenced U.S. Pat. No. 5,489,677, and the amide backbones of the above -referenced U.S. Pat. No. 5,602,240. In some embodiments, the polynucletotides featured herein have morpholino backbone structures of the above-referenced U.S. Pat. No. 5,034,506.
[00496] Modifications at the 2' position may also aid in delivery. Preferably, modifications at the 2' position are not located in a polypeptide-coding sequence, i.e., not in a translatable region.
Modifications at the 2' position may be located in a 5' UTR, a 3' UTR and/or a tailing region. Modifications at the 2' position can include one of the following at the 2' position: H (i.e., 2'-deoxy); F; 0-, S-, or N-alkyl; 0-, S-, or N-alkenyl; 0-, S- or N-alkynyl; or O-alkyl-O-alkyl, wherein the alkyl, alkenyl and alkynyl may be substituted or unsubstituted Ci to Cio alkyl or C2 to Cio alkenyl and alkynyl. Exemplary suitable modifications include 0[(CH2)nO] mCH3, 0(CH2).nOCH3,
0(CH2)„NH2, 0(CH2) nCH3, 0(CH2)nONH2, and 0(CH2)nON[(CH2)nCH3)]2, where n and m are from 1 to about 10. In other embodiments, the modified nucleic acids or mmRNA include one of the following at the 2' position: Ci to Cio lower alkyl, substituted lower alkyl, alkaryl, aralkyl, O-alkaryl or O-aralkyl, SH, SCH3, OCN, CI, Br, CN, CF3, OCF3, SOCH3, S02CH3, ON02, N02, N3, NH2, heterocycloalkyl, heterocycloalkaryl, aminoalkylamino, polyalkylamino, substituted silyl, an RNA cleaving group, a reporter group, an intercalator, a group for improving the pharmacokinetic properties, or a group for improving the pharmacodynamic properties, and other substituents having similar properties. In some embodiments, the modification includes a 2'-methoxyethoxy (2'-0— CH2CH2OCH3, also known as 2'-0-(2-methoxyethyl) or 2'-MOE) (Martin et al, Helv. Chim. Acta, 1995, 78:486-504) i.e., an alkoxy-alkoxy group. Another exemplary modification is 2'- dimethylaminooxyethoxy, i.e., a 0(CH2)2ON(CH3)2 group, also known as 2'-DMAOE, as described in examples herein below, and 2'-dimethylaminoethoxyethoxy (also known in the art as 2'-0- dimethylaminoethoxyethyl or 2'-DMAEOE), i.e., 2'-0~CH2-0~CH2-N(CH2)2, also described in examples herein below. Other modifications include 2'-methoxy (2'-OCH3), 2'-aminopropoxy (2'- OCH2CH2CH2NH2) and 2'-fluoro (2'-F). Similar modifications may also be made at other positions, particularly the 3' position of the sugar on the 3' terminal nucleotide or in 2'-5' linked dsRNAs and the 5' position of 5' terminal nucleotide. Polynucleotides of the invention may also have sugar mimetics such as cyclobutyl moieties in place of the pentofuranosyl sugar. Representative U.S. patents that teach the preparation of such modified sugar structures include, but are not limited to, U.S. Pat. Nos. 4,981 ,957; 5, 1 18,800; 5,319,080; 5,359,044; 5,393,878; 5,446,137; 5,466,786;
5,514,785; 5,519, 134; 5,567,81 1 ; 5,576,427; 5,591 ,722; 5,597,909; 5,610,300; 5,627,053;
5,639,873; 5,646,265; 5,658,873; 5,670,633; and 5,700,920 and each of which is herein incorporated by reference.
[00497] In still other embodiments, the modified nucleic acid molecule or mmRNA is covalently conjugated to a cell-penetrating polypeptide. The cell-penetrating peptide may also include a signal sequence. The conjugates of the invention can be designed to have increased stability; increased cell transfection; and/or altered the biodistribution (e.g., targeted to specific tissues or cell types). Self-Assembled Nanoparticles
Nucleic Acid Self-Assembled Nanoparticles
[00498] Self-assembled nanoparticles have a well-defined size which may be precisely controlled as the nucleic acid strands may be easily reprogrammable. For example, the optimal particle size for a cancer-targeting nanodelivery carrier is 20-100 nm as a diameter greater than 20 nm avoids renal clearance and enhances delivery to certain tumors through enhanced permeability and retention effect. Using self-assembled nucleic acid nanoparticles a single uniform population in size and shape having a precisely controlled spatial orientation and density of cancer-targeting ligands for enhanced delivery. As a non-limiting example, oligonucleotide nanoparticles were prepared using programmable self-assembly of short DNA fragments and therapeutic siRNAs. These nanoparticles are molecularly identical with controllable particle size and target ligand location and density. The DNA fragments and siRNAs self-assembled into a one-step reaction to generate DNA/siRNA tetrahedral nanoparticles for targeted in vivo delivery. (Lee et al., Nature Nanotechnology 2012 7:389-393; herein incorporated by reference in its entirety).
[00499] In one embodiment, the modified nucleic acid molecules and mmRNA disclosed herein may be formulated as self-assembled nanoparticles. As a non- limiting example, nucleic acids may be used to make nanoparticles which may be used in a delivery system for the modified nucleic acid molecules and/or mmRNA of the present invention (See e.g., International Pub. No.
WO2012125987; herein incorporated by reference in its entirety).
[00500] In one embodiment, the nucleic acid self-assembled nanoparticles may comprise a core of the modified nucleic acid molecules or mmRNA disclosed herein and a polymer shell. The polymer shell may be any of the polymers described herein and are known in the art. In an additional embodiment, the polymer shell may be used to protect the modified nucleic acid molecules and mmRNA in the core.
Polymer-Based Self-Assembled Nanoparticles
[00501] Polymers may be used to form sheets which self-assembled into nanoparticles. These nanoparticles may be used to deliver the modified nucleic acids and mmRNA of the present invention. In one embodiment, these self-assembled nanoparticles may be microsponges formed of long polymers of RNA hairpins which form into crystalline 'pleated' sheets before self-assembling into microsponges. These microsponges are densely-packed sponge like microparticles which may function as an efficient carrier and may be able to deliver cargo to a cell. The microsponges may be from lum to 300 nm in diameter. The microsponges may be complexed with other agents known in the art to form larger microsponges. As a non- limiting example, the microsponge may be complexed with an agent to form an outer layer to promote cellular uptake such as polycation polyethyleneime (PEI). This complex can form a 250-nm diameter particle that can remain stable at high temperatures (150°C) (Grabow and Jaegar, Nature Materials 2012, 11 :269-269; herein incorporated by reference in its entirety). Additionally these microsponges may be able to exhibit an extraordinary degree of protection from degradation by ribonucleases.
[00502] In another embodiment, the polymer-based self-assembled nanoparticles such as, but not limited to, microsponges, may be fully programmable nanoparticles. The geometry, size and stoichiometry of the nanoparticle may be precisely controlled to create the optimal nanoparticle for delivery of cargo such as, but not limited to, modified nucleic acid molecules and mmRNA.
[00503] In one embodiment, the polymer based nanoparticles may comprise a core of the modified nucleic acid molecules and mmRNA disclosed herein and a polymer shell. The polymer shell may be any of the polymers described herein and are known in the art. In an additional embodiment, the polymer shell may be used to protect the modified nucleic acid molecules and mmRNA in the core. Inorganic Nanoparticles
[00504] The modified nucleic acid molecules or mmRNAs of the present invention may be formulated in inorganic nanoparticles (U.S. Pat. No. 8,257,745, herein incorporated by reference in its entirety). The inorganic nanoparticles may include, but are not limited to, clay substances that are water swellable. As a non-limiting example, the inorganic nanoparticle may include synthetic smectite clays which are made from simple silicates (See e.g., U.S. Pat. No. 5,585,108 and 8,257,745 each of which are herein incorporated by reference in their entirety).
[00505] In one embodiment, the inorganic nanoparticles may comprise a core of the modified nucleic acids disclosed herein and a polymer shell. The polymer shell may be any of the polymers described herein and are known in the art. In an additional embodiment, the polymer shell may be used to protect the modified nucleic acids in the core.
Semi-conductive and Metallic Nanoparticles
[00506] The modified nucleic acid molecules or mmRNAs of the present invention may be formulated in water-dispersible nanoparticle comprising a semiconductive or metallic material (U.S. Pub. No. 20120228565; herein incorporated by reference in its entirety) or formed in a magnetic nanoparticle (U.S. Pub. No. 20120265001 and 20120283503; each of which is herein incorporated by reference in its entirety). The water-dispersible nanoparticles may be hydrophobic nanoparticles or hydrophilic nanoparticles.
[00507] In one embodiment, the semi-conductive and/or metallic nanoparticles may comprise a core of the modified nucleic acids disclosed herein and a polymer shell. The polymer shell may be any of the polymers described herein and are known in the art. In an additional embodiment, the polymer shell may be used to protect the modified nucleic acids in the core.
Gels and Hydrogels
[00508] In one embodiment, the modified mR A disclosed herein may be encapsulated into any hydrogel known in the art which may form a gel when injected into a subject. Hydrogels are a network of polymer chains that are hydrophilic, and are sometimes found as a colloidal gel in which water is the dispersion medium. Hydrogels are highly absorbent (they can contain over 99% water) natural or synthetic polymers. Hydrogels also possess a degree of flexibility very similar to natural tissue, due to their significant water content. The hydrogel described herein may used to encapsulate lipid nanoparticles which are biocompatible, biodegradable and/or porous.
[00509] As a non-limiting example, the hydrogel may be an aptamer-functionalized hydrogel. The aptamer-functionalized hydrogel may be programmed to release one or more modified nucleic acid molecules and/or mmRNA using nucleic acid hybridization. (Battig et al., J. Am. Chem. Society. 2012 134: 12410-12413; herein incorporated by reference in its entirety).
[00510] As another non-limiting example, the hydrogel may be a shaped as an inverted opal.
The opal hydrogels exhibit higher swelling ratios and the swelling kinetics is an order of magnitude faster as well. Methods of producing opal hydrogels and description of opal hydrogels are described in International Pub. No. WO2012148684, herein incorporated by reference in its entirety.
[00511] In yet another non-limiting example, the hydrogel may be an antibacterial hydrogel. The antibacterial hydrogel may comprise a pharmaceutical acceptable salt or organic material such as, but not limited to pharmaceutical grade and/or medical grade silver salt and aloe vera gel or extract. (International Pub. No. WO2012151438, herein incorporated by reference in its entirety).
[00512] In one embodiment, the modified mRNA may be encapsulated in a lipid nanoparticle and then the lipid nanoparticle may be encapsulated into a hyrdogel.
[00513] In one embodiment, the modified mRNA disclosed herein may be encapsulated into any gel known in the art. As a non-limiting example the gel may be a fluorouracil injectable gel or a fluorouracil injectable gel containing a chemical compound and/or drug known in the art. As another example, the modified mRNA may be encapsulated in a fluorouracil gel containing epinephrine (See e.g., Smith et al. Cancer Chemotherapty and Pharmacology, 1999 44(4):267-274; herein incorporated by reference in its entirety).
[00514] In one embodiment, the modified nucleic acid molecules and/or mmRNA disclosed herein may be encapsulated into a fibrin gel, fibrin hydrogel or fibrin glue. In another embodiment, the modified nucleic acid molecules and/or mmRNA may be formulated in a lipid nanoparticle or a rapidly eliminated lipid nanoparticle prior to being encapsulated into a fibrin gel, fibrin hydrogel or a fibrin glue. In yet another embodiment, the modified nucleic acid molecules and/or mmRNA may be formulated as a lipoplex prior to being encapsulated into a fibrin gel, hydrogel or a fibrin glue. Fibrin gels, hydrogels and glues comprise two components, a fibrinogen solution and a thrombin solution which is rich in calcium (See e.g., Spicer and Mikos, Journal of Controlled Release 2010. 148: 49-55; Kidd et al. Journal of Controlled Release 2012. 157:80-85; each of which is herein incorporated by reference in its entirety). The concentration of the components of the fibrin gel, hydrogel and/or glue can be altered to change the characteristics, the network mesh size, and/or the degradation characteristics of the gel, hydrogel and/or glue such as, but not limited to changing the release characteristics of the fibrin gel, hydrogel and/or glue. (See e.g., Spicer and Mikos, Journal of Controlled Release 2010. 148: 49-55; Kidd et al. Journal of Controlled Release 2012. 157:80-85; Catelas et al. Tissue Engineering 2008. 14: 119-128; each of which is herein incorporated by reference in its entirety). This feature may be advantageous when used to deliver the modified mRNA disclosed herein. (See e.g., Kidd et al. Journal of Controlled Release 2012. 157:80-85;
Catelas et al. Tissue Engineering 2008. 14: 119-128; each of which is herein incorporated by reference in its entirety).
Cations and Anions
[00515] Formulations of modified nucleic acid molecules disclosed herein may include cations or anions. In one embodiment, the formulations include metal cations such as, but not limited to, Zn2+, Ca2+, Cu2+, Mg+ and combinations thereof. As a non-limiting example, formulations may include polymers and a modified mRNA complexed with a metal cation (See e.g., U.S. Pat. Nos. 6,265,389 and 6,555,525, each of which is herein incorporated by reference in its entirety).
Molded Nanoparticles and Microparticles
[00516] The modified nucleic acid molecules and/or mmRNA disclosed herein may be formulated in nanoparticles and/or microparticles. These nanoparticles and/or microparticles may be molded into any size shape and chemistry. As an example, the nanoparticles and/or microparticles may be made using the PRINT® technology by LIQUIDA TECHNOLOGIES® (Morrisville, NC) (See e.g., International Pub. No. WO2007024323; herein incorporated by reference in its entirety).
[00517] In one embodiment, the molded nanoparticles may comprise a core of the modified nucleic acid molecules and/or mmRNA disclosed herein and a polymer shell. The polymer shell may be any of the polymers described herein and are known in the art. In an additional embodiment, the polymer shell may be used to protect the modified nucleic acid molecules and/or mmRNA in the core.
NanoJackets and NanoLiposomes
[00518] The modified nucleic acid molecules and/or mmRNA disclosed herein may be formulated in NanoJackets and NanoLiposomes by Keystone Nano (State College, PA). NanoJackets are made of compounds that are naturally found in the body including calcium, phosphate and may also include a small amount of silicates. Nanojackets may range in size from 5 to 50 nm and may be used to deliver hydrophilic and hydrophobic compounds such as, but not limited to, modified nucleic acid molecules and/or mmRNA.
[00519] NanoLiposomes are made of lipids such as, but not limited to, lipids which naturally occur in the body. NanoLiposomes may range in size from 60-80 nm and may be used to deliver hydrophilic and hydrophobic compounds such as, but not limited to, modified nucleic acid molecules and/or mmRNA. In one aspect, the modified nucleic acids disclosed herein are formulated in a NanoLiposome such as, but not limited to, Ceramide NanoLiposomes.
Excipients
[00520] Pharmaceutical formulations may additionally comprise a pharmaceutically acceptable excipient, which, as used herein, includes, but are not limited to, any and all solvents, dispersion media, diluents, or other liquid vehicles, dispersion or suspension aids, surface active agents, isotonic agents, thickening or emulsifying agents, preservatives, solid binders, lubricants and the like, as suited to the particular dosage form desired. Various excipients for formulating
pharmaceutical compositions and techniques for preparing the composition are known in the art (see Remington: The Science and Practice of Pharmacy, 21st Edition, A. R. Gennaro, Lippincott, Williams & Wilkins, Baltimore, MD, 2006; incorporated herein by reference in its entirety). The use of a conventional excipient medium may be contemplated within the scope of the present disclosure, except insofar as any conventional excipient medium may be incompatible with a substance or its derivatives, such as by producing any undesirable biological effect or otherwise interacting in a deleterious manner with any other component(s) of the pharmaceutical composition.
[00521] In some embodiments, a pharmaceutically acceptable excipient may be at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% pure. In some embodiments, an excipient may be approved for use for humans and for veterinary use. In some embodiments, an excipient may be approved by United States Food and Drug Administration. In some embodiments, an excipient may be of pharmaceutical grade. In some embodiments, an excipient may meet the standards of the United States Pharmacopoeia (USP), the European Pharmacopoeia (EP), the British Pharmacopoeia, and/or the International Pharmacopoeia.
[00522] Pharmaceutically acceptable excipients used in the manufacture of pharmaceutical compositions include, but are not limited to, inert diluents, dispersing and/or granulating agents, surface active agents and/or emulsifiers, disintegrating agents, binding agents, preservatives, buffering agents, lubricating agents, and/or oils. Such excipients may optionally be included in pharmaceutical formulations. The composition may also include excipients such as cocoa butter and suppository waxes, coloring agents, coating agents, sweetening, flavoring, and/or perfuming agents.
[00523] Exemplary diluents include, but are not limited to, calcium carbonate, sodium carbonate, calcium phosphate, dicalcium phosphate, calcium sulfate, calcium hydrogen phosphate, sodium phosphate lactose, sucrose, cellulose, microcrystalline cellulose, kaolin, mannitol, sorbitol, inositol, sodium chloride, dry starch, cornstarch, powdered sugar, etc., and/or combinations thereof.
[00524] Exemplary granulating and/or dispersing agents include, but are not limited to, potato starch, corn starch, tapioca starch, sodium starch glycolate, clays, alginic acid, guar gum, citrus pulp, agar, bentonite, cellulose and wood products, natural sponge, cation-exchange resins, calcium carbonate, silicates, sodium carbonate, cross-linked poly(vinyl-pyrrolidone) (crospovidone), sodium carboxymethyl starch (sodium starch glycolate), carboxymethyl cellulose, cross-linked sodium carboxymethyl cellulose (croscarmellose), methylcellulose, pregelatinized starch (starch 1500), microcrystalline starch, water insoluble starch, calcium carboxymethyl cellulose, magnesium aluminum silicate (VEEGUM®), sodium lauryl sulfate, quaternary ammonium compounds, etc., and/or combinations thereof.
[00525] Exemplary surface active agents and/or emulsifiers include, but are not limited to, natural emulsifiers {e.g. acacia, agar, alginic acid, sodium alginate, tragacanth, chondrux, cholesterol, xanthan, pectin, gelatin, egg yolk, casein, wool fat, cholesterol, wax, and lecithin), colloidal clays (e.g. bentonite [aluminum silicate] and VEEGUM [magnesium aluminum silicate]), long chain amino acid derivatives, high molecular weight alcohols (e.g. stearyl alcohol, cetyl alcohol, oleyl alcohol, triacetin monostearate, ethylene glycol distearate, glyceryl monostearate, and propylene glycol monostearate, polyvinyl alcohol), carbomers (e.g. carboxy polymethylene, polyacrylic acid, acrylic acid polymer, and carboxyvinyl polymer), carrageenan, cellulosic derivatives (e.g.
carboxymethylcellulose sodium, powdered cellulose, hydroxymethyl cellulose, hydroxypropyl cellulose, hydroxypropyl methylcellulose, methylcellulose), sorbitan fatty acid esters (e.g.
polyoxyethylene sorbitan monolaurate [TWEEN®20], polyoxyethylene sorbitan [TWEEN®60], polyoxyethylene sorbitan monooleate [TWEEN®80], sorbitan monopalmitate [SPAN®40], sorbitan monostearate [SPAN®60], sorbitan tristearate [SPAN®65], glyceryl monooleate, sorbitan
monooleate [SPAN®80]), polyoxyethylene esters (e.g. polyoxyethylene monostearate [MYRJ®45], polyoxyethylene hydrogenated castor oil, polyethoxylated castor oil, polyoxymethylene stearate, and SOLUTOL®), sucrose fatty acid esters, polyethylene glycol fatty acid esters (e.g. CREMOPHOR®), polyoxyethylene ethers, (e.g. polyoxyethylene lauryl ether [BRIJ®30]), poly(vinyl-pyrrolidone), diethylene glycol monolaurate, triethanolamine oleate, sodium oleate, potassium oleate, ethyl oleate, oleic acid, ethyl laurate, sodium lauryl sulfate, PLUORINC®F 68, POLOXAMER®188, cetrimonium bromide, cetylpyridinium chloride, benzalkonium chloride, docusate sodium, etc. and/or
combinations thereof.
[00526] Exemplary binding agents include, but are not limited to, starch (e.g. cornstarch and starch paste); gelatin; sugars (e.g. sucrose, glucose, dextrose, dextrin, molasses, lactose, lactitol, mannitol,); natural and synthetic gums (e.g. acacia, sodium alginate, extract of Irish moss, panwar gum, ghatti gum, mucilage of isapol husks, carboxymethylcellulose, methylcellulose, ethylcellulose,
hydroxyethylcellulose, hydroxypropyl cellulose, hydroxypropyl methylcellulose, microcrystalline cellulose, cellulose acetate, poly(vinyl-pyrrolidone), magnesium aluminum silicate (VEEGUM®), and larch arabogalactan); alginates; polyethylene oxide; polyethylene glycol; inorganic calcium salts; silicic acid; polymethacrylates; waxes; water; alcohol; etc.; and combinations thereof.
[00527] Exemplary preservatives may include, but are not limited to, antioxidants, chelating agents, antimicrobial preservatives, antifungal preservatives, alcohol preservatives, acidic preservatives, and/or other preservatives. Exemplary antioxidants include, but are not limited to, alpha tocopherol, ascorbic acid, acorbyl palmitate, butylated hydroxyanisole, butylated hydroxytoluene,
monothioglycerol, potassium metabisulfite, propionic acid, propyl gallate, sodium ascorbate, sodium bisulfite, sodium metabisulfite, and/or sodium sulfite. Exemplary chelating agents include ethylenediaminetetraacetic acid (EDTA), citric acid monohydrate, disodium edetate, dipotassium edetate, edetic acid, fumaric acid, malic acid, phosphoric acid, sodium edetate, tartaric acid, and/or trisodium edetate. Exemplary antimicrobial preservatives include, but are not limited to, benzalkonium chloride, benzethonium chloride, benzyl alcohol, bronopol, cetrimide,
cetylpyridinium chloride, chlorhexidine, chlorobutanol, chlorocresol, chloroxylenol, cresol, ethyl alcohol, glycerin, hexetidine, imidurea, phenol, phenoxyethanol, phenylethyl alcohol,
phenylmercuric nitrate, propylene glycol, and/or thimerosal. Exemplary antifungal preservatives include, but are not limited to, butyl paraben, methyl paraben, ethyl paraben, propyl paraben, benzoic acid, hydroxybenzoic acid, potassium benzoate, potassium sorbate, sodium benzoate, sodium propionate, and/or sorbic acid. Exemplary alcohol preservatives include, but are not limited to, ethanol, polyethylene glycol, phenol, phenolic compounds, bisphenol, chlorobutanol, hydroxybenzoate, and/or phenylethyl alcohol. Exemplary acidic preservatives include, but are not limited to, vitamin A, vitamin C, vitamin E, beta-carotene, citric acid, acetic acid, dehydroacetic acid, ascorbic acid, sorbic acid, and/or phytic acid. Other preservatives include, but are not limited to, tocopherol, tocopherol acetate, deteroxime mesylate, cetrimide, butylated hydroxyanisol (BHA), butylated hydroxytoluened (BHT), ethylenediamine, sodium lauryl sulfate (SLS), sodium lauryl ether sulfate (SLES), sodium bisulfite, sodium metabisulfite, potassium sulfite, potassium metabisulfite, GLYDANT PLUS®, PHENONIP®, methylparaben, GERM ALL® 115,
GERMABEN®II, NEOLONE, KATHON, and/or EUXYL®.
[00528] Exemplary buffering agents include, but are not limited to, citrate buffer solutions, acetate buffer solutions, phosphate buffer solutions, ammonium chloride, calcium carbonate, calcium chloride, calcium citrate, calcium glubionate, calcium gluceptate, calcium gluconate, d-gluconic acid, calcium glycerophosphate, calcium lactate, propanoic acid, calcium levulinate, pentanoic acid, dibasic calcium phosphate, phosphoric acid, tribasic calcium phosphate, calcium hydroxide phosphate, potassium acetate, potassium chloride, potassium gluconate, potassium mixtures, dibasic potassium phosphate, monobasic potassium phosphate, potassium phosphate mixtures, sodium acetate, sodium bicarbonate, sodium chloride, sodium citrate, sodium lactate, dibasic sodium phosphate, monobasic sodium phosphate, sodium phosphate mixtures, tromethamine, magnesium hydroxide, aluminum hydroxide, alginic acid, pyrogen-free water, isotonic saline, Ringer's solution, ethyl alcohol, etc., and/or combinations thereof. [00529] Exemplary lubricating agents include, but are not limited to, magnesium stearate, calcium stearate, stearic acid, silica, talc, malt, glyceryl behanate, hydrogenated vegetable oils, polyethylene glycol, sodium benzoate, sodium acetate, sodium chloride, leucine, magnesium lauryl sulfate, sodium lauryl sulfate, etc., and combinations thereof.
[00530] Exemplary oils include, but are not limited to, almond, apricot kernel, avocado, babassu, bergamot, black current seed, borage, cade, camomile, canola, caraway, carnauba, castor, cinnamon, cocoa butter, coconut, cod liver, coffee, corn, cotton seed, emu, eucalyptus, evening primrose, fish, flaxseed, geraniol, gourd, grape seed, hazel nut, hyssop, isopropyl myristate, jojoba, kukui nut, lavandin, lavender, lemon, litsea cubeba, macademia nut, mallow, mango seed, meadowfoam seed, mink, nutmeg, olive, orange, orange roughy, palm, palm kernel, peach kernel, peanut, poppy seed, pumpkin seed, rapeseed, rice bran, rosemary, safflower, sandalwood, sasquana, savoury, sea buckthorn, sesame, shea butter, silicone, soybean, sunflower, tea tree, thistle, tsubaki, vetiver, walnut, and wheat germ oils. Exemplary oils include, but are not limited to, butyl stearate, caprylic triglyceride, capric triglyceride, cyclomethicone, diethyl sebacate, dimethicone 360, isopropyl myristate, mineral oil, octyldodecanol, oleyl alcohol, silicone oil, and/or combinations thereof.
[00531] Excipients such as cocoa butter and suppository waxes, coloring agents, coating agents, sweetening, flavoring, and/or perfuming agents can be present in the composition, according to the judgment of the formulator.
Delivery
[00532] The present disclosure encompasses the delivery of modified nucleic acid molecules or mmRNA for any of therapeutic, pharmaceutical, diagnostic or imaging by any appropriate route taking into consideration likely advances in the sciences of drug delivery. Delivery may be naked or formulated.
Naked Delivery
[00533] The modified nucleic acid molecules or mmRNA of the present invention may be delivered to a cell naked. As used herein in, "naked" refers to delivering modified nucleic acid molecules or mmRNA free from agents which promote transfection. For example, the modified nucleic acid molecules or mmRNA delivered to the cell may contain no modifications. The naked modified nucleic acid molecules or mmRNA may be delivered to the cell using routes of administration known in the art and described herein.
Formulated Delivery [00534] The modified nucleic acid molecules or mmR A of the present invention may be formulated, using the methods described herein. The formulations may contain modified nucleic acid molecules or mmRNA which may be modified and/or unmodified. The formulations may further include, but are not limited to, cell penetration agents, a pharmaceutically acceptable carrier, a delivery agent, a bioerodible or biocompatible polymer, a solvent, and a sustained-release delivery depot. The formulated modified nucleic acid molecules or mmRNA may be delivered to the cell using routes of administration known in the art and described herein.
[00535] The compositions may also be formulated for direct delivery to an organ or tissue in any of several ways in the art including, but not limited to, direct soaking or bathing, via a catheter, by gels, powder, ointments, creams, gels, lotions, and/or drops, by using substrates such as fabric or biodegradable materials coated or impregnated with the compositions, and the like.
Administration
[00536] The modified nucleic acid molecules or mmRNA of the present invention may be administered by any route which results in a therapeutically effective outcome. These include, but are not limited to enteral, gastroenteral, epidural, oral, transdermal, epidural (peridural), intracerebral (into the cerebrum), intracerebroventricular (into the cerebral ventricles), epicutaneous (application onto the skin), intradermal, (into the skin itself), subcutaneous (under the skin), nasal administration (through the nose), intravenous (into a vein), intraarterial (into an artery), intramuscular (into a muscle), intracardiac (into the heart), intraosseous infusion (into the bone marrow), intrathecal (into the spinal canal), intraperitoneal, (infusion or injection into the peritoneum), intravesical infusion, intravitreal, (through the eye), intracavernous injection, ( into the base of the penis), intravaginal administration, intrauterine, extra-amniotic administration, transdermal (diffusion through the intact skin for systemic distribution), transmucosal (diffusion through a mucous membrane), insufflation (snorting), sublingual, sublabial, enema, eye drops (onto the conjunctiva), or in ear drops. In specific embodiments, compositions may be administered in a way which allows them cross the blood-brain barrier, vascular barrier, or other epithelial barrier. Non-limiting routes of
administration for the modified nucleic acids or mmRNA of the present invention are described below.
Parenteral and Injectible Administration
[00537] Liquid dosage forms for parenteral administration include, but are not limited to, pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups, and/or elixirs. In addition to active ingredients, liquid dosage forms may comprise inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, dimethylformamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof. Besides inert diluents, oral compositions can include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and/or perfuming agents. In certain embodiments for parenteral administration, compositions are mixed with solubilizing agents such as CREMOPHOR®, alcohols, oils, modified oils, glycols, polysorbates, cyclodextrins, polymers, and/or combinations thereof.
[00538] Injectable preparations, for example, sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing agents, wetting agents, and/or suspending agents. Sterile injectable preparations may be sterile injectable solutions, suspensions, and/or emulsions in nontoxic parenterally acceptable diluents and/or solvents, for example, as a solution in 1,3-butanediol. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution, U.S.P., and isotonic sodium chloride solution. Sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose any bland fixed oil can be employed including synthetic mono- or diglycerides. Fatty acids such as oleic acid can be used in the preparation of injectables.
[00539] Injectable formulations can be sterilized, for example, by filtration through a bacterial- retaining filter, and/or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved or dispersed in sterile water or other sterile injectable medium prior to use.
[00540] In order to prolong the effect of an active ingredient, it is often desirable to slow the absorption of the active ingredient from subcutaneous or intramuscular injection. This may be accomplished by the use of a liquid suspension of crystalline or amorphous material with poor water solubility. The rate of absorption of the drug then depends upon its rate of dissolution which, in turn, may depend upon crystal size and crystalline form. Alternatively, delayed absorption of a parenterally administered drug form is accomplished by dissolving or suspending the drug in an oil vehicle. Injectable depot forms are made by forming microencapsule matrices of the drug in biodegradable polymers such as polylactide-polyglycolide. Depending upon the ratio of drug to polymer and the nature of the particular polymer employed, the rate of drug release can be controlled. Examples of other biodegradable polymers include poly(orthoesters) and
poly(anhydrides). Depot injectable formulations are prepared by entrapping the drug in liposomes or microemulsions which are compatible with body tissues.
Rectal and Vaginal Administration
[00541] Compositions for rectal or vaginal administration are typically suppositories which can be prepared by mixing compositions with suitable non-irritating excipients such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active ingredient. Oral Administration
[00542] Liquid dosage forms for oral administration include, but are not limited to,
pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups, and/or elixirs. In addition to active ingredients, liquid dosage forms may comprise inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, dimethylformamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof. Besides inert diluents, oral compositions can include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and/or perfuming agents. In certain embodiments for parenteral administration, compositions are mixed with solubilizing agents such as CREMOPHOR®, alcohols, oils, modified oils, glycols, polysorbates, cyclodextrins, polymers, and/or combinations thereof.
[00543] Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules. In such solid dosage forms, an active ingredient is mixed with at least one inert, pharmaceutically acceptable excipient such as sodium citrate or dicalcium phosphate and/or fillers or extenders {e.g. starches, lactose, sucrose, glucose, mannitol, and silicic acid), binders {e.g.
carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidinone, sucrose, and acacia), humectants {e.g. glycerol), disintegrating agents {e.g. agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate), solution retarding agents {e.g. paraffin), absorption accelerators {e.g. quaternary ammonium compounds), wetting agents {e.g. cetyl alcohol and glycerol monostearate), absorbents {e.g. kaolin and bentonite clay), and lubricants {e.g. talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate), and mixtures thereof. In the case of capsules, tablets and pills, the dosage form may comprise buffering agents.
Topical or Transdermal Administration
[00544] As described herein, compositions containing the modified nucleic acid molecules or mmRNA of the invention may be formulated for administration topically. The skin may be an ideal target site for delivery as it is readily accessible. Gene expression may be restricted not only to the skin, potentially avoiding nonspecific toxicity, but also to specific layers and cell types within the skin.
[00545] The site of cutaneous expression of the delivered compositions will depend on the route of nucleic acid delivery. Three routes are commonly considered to deliver modified nucleic acid molecules or mmRNA to the skin: (i) topical application (e.g. for local/regional treatment); (ii) intradermal injection (e.g. for local/regional treatment); and (iii) systemic delivery (e.g. for treatment of dermato logic diseases that affect both cutaneous and extracutaneous regions). Modified nucleic acid molecules or mmRNA can be delivered to the skin by several different approaches known in the art. Most topical delivery approaches have been shown to work for delivery of DNA, such as but not limited to, topical application of non-cationic liposome-DNA complex, cationic liposome-DNA complex, particle-mediated (gene gun), puncture-mediated gene transfections, and viral delivery approaches. After delivery of the nucleic acid, gene products have been detected in a number of different skin cell types, including, but not limited to, basal keratinocytes, sebaceous gland cells, dermal fibroblasts and dermal macrophages.
[00546] In one embodiment, the invention provides for a variety of dressings (e.g., wound dressings) or bandages (e.g., adhesive bandages) for conveniently and/or effectively carrying out methods of the present invention. Typically dressing or bandages may comprise sufficient amounts of pharmaceutical compositions and/or modified nucleic acid molecules or mmRNA described herein to allow a user to perform multiple treatments of a subject(s).
[00547] In one embodiment, the invention provides for the modified nucleic acid molecules or mmRNA compositions to be delivered in more than one injection.
[00548] In one embodiment, before topical and/or transdermal administration at least one area of tissue, such as skin, may be subjected to a device and/or solution which may increase permeability. In one embodiment, the tissue may be subjected to an abrasion device to increase the permeability of the skin (see U.S. Patent Publication No. 20080275468, herein incorporated by reference in its entirety). In another embodiment, the tissue may be subjected to an ultrasound enhancement device. An ultrasound enhancement device may include, but is not limited to, the devices described in U.S. Publication No. 20040236268 and U.S. Patent Nos. 6,491,657 and 6,234,990; each of which are herein incorporated by reference in their entireties. Methods of enhancing the permeability of tissue are described in U.S. Publication Nos. 20040171980 and 20040236268 and U.S. Pat. No. 6,190,315; each of which are herein incorporated by reference in their entireties.
[00549] In one embodiment, a device may be used to increase permeability of tissue before delivering formulations of modified mRNA described herein. The permeability of skin may be measured by methods known in the art and/or described in U.S. Patent No. 6,190,315, herein incorporated by reference in its entirety. As a non-limiting example, a modified mRNA formulation may be delivered by the drug delivery methods described in U.S. Patent No. 6,190,315, herein incorporated by reference in its entirety.
[00550] In another non- limiting example tissue may be treated with a eutectic mixture of local anesthetics (EMLA) cream before, during and/or after the tissue may be subjected to a device which may increase permeability. Katz et al. (Anesth Analg (2004); 98:371-76; herein incorporated by reference in its entirety) showed that using the EMLA cream in combination with a low energy, an onset of superficial cutaneous analgesia was seen as fast as 5 minutes after a pretreatment with a low energy ultrasound.
[00551] In one embodiment, enhancers may be applied to the tissue before, during, and/or after the tissue has been treated to increase permeability. Enhancers include, but are not limited to, transport enhancers, physical enhancers, and cavitation enhancers. Non-limiting examples of enhancers are described in U.S. Patent No. 6,190,315, herein incorporated by reference in its entirety.
[00552] In one embodiment, a device may be used to increase permeability of tissue before delivering formulations of modified mRNA described herein, which may further contain a substance that invokes an immune response. In another non-limiting example, a formulation containing a substance to invoke an immune response may be delivered by the methods described in U.S.
Publication Nos. 20040171980 and 20040236268; each of which are herein incorporated by reference in their entireties.
[00553] Dosage forms for topical and/or transdermal administration of a composition may include ointments, pastes, creams, lotions, gels, powders, solutions, sprays, inhalants and/or patches. Generally, an active ingredient is admixed under sterile conditions with a pharmaceutically acceptable excipient and/or any needed preservatives and/or buffers as may be required.
[00554] Additionally, the present invention contemplates the use of transdermal patches, which often have the added advantage of providing controlled delivery of a compound to the body. Such dosage forms may be prepared, for example, by dissolving and/or dispensing the compound in the proper medium. Alternatively or additionally, rate may be controlled by either providing a rate controlling membrane and/or by dispersing the compound in a polymer matrix and/or gel.
[00555] Formulations suitable for topical administration include, but are not limited to, liquid and/or semi liquid preparations such as liniments, lotions, oil in water and/or water in oil emulsions such as creams, ointments and/or pastes, and/or solutions and/or suspensions. Topically- administrable formulations may, for example, comprise from about 0.1% to about 10% (w/w) active ingredient, although the concentration of active ingredient may be as high as the solubility limit of the active ingredient in the solvent. Formulations for topical administration may further comprise one or more of the additional ingredients described herein.
Depot Administration
[00556] As described herein, in some embodiments, the composition is formulated in depots for extended release. Generally, a specific organ or tissue (a "target tissue") is targeted for
administration.
[00557] In some aspects of the invention, the modified nucleic acid molecules or mmRNA are spatially retained within or proximal to a target tissue. Provided are method of providing a composition to a target tissue of a mammalian subject by contacting the target tissue (which contains one or more target cells) with the composition under conditions such that the composition, in particular the nucleic acid component(s) of the composition, is substantially retained in the target tissue, meaning that at least 10, 20, 30, 40, 50, 60, 70, 80, 85, 90, 95, 96, 97, 98, 99, 99.9, 99.99 or greater than 99.99% of the composition is retained in the target tissue. Advantageously, retention is determined by measuring the amount of the nucleic acid present in the composition that enters one or more target cells. For example, at least 1, 5, 10, 20, 30, 40, 50, 60, 70, 80, 85, 90, 95, 96, 97, 98, 99, 99.9, 99.99 or greater than 99.99%> of the nucleic acids administered to the subject are present intracellularly at a period of time following administration. For example, intramuscular injection to a mammalian subject is performed using an aqueous composition containing a ribonucleic acid and a transfection reagent, and retention of the composition is determined by measuring the amount of the ribonucleic acid present in the muscle cells.
[00558] Aspects of the invention are directed to methods of providing a composition to a target tissue of a mammalian subject, by contacting the target tissue (containing one or more target cells) with the composition under conditions such that the composition is substantially retained in the target tissue. The composition contains an effective amount of a nucleic acid molecules or mmRNA such that the polypeptide of interest is produced in at least one target cell. The compositions generally contain a cell penetration agent, although "naked" nucleic acid (such as nucleic acids without a cell penetration agent or other agent) is also contemplated, and a pharmaceutically acceptable carrier.
[00559] In some circumstances, the amount of a protein produced by cells in a tissue is desirably increased. Preferably, this increase in protein production is spatially restricted to cells within the target tissue. Thus, provided are methods of increasing production of a protein of interest in a tissue of a mammalian subject. A composition is provided that contains modified nucleic acid molecule or mmRNA characterized in that a unit quantity of composition has been determined to produce the polypeptide of interest in a substantial percentage of cells contained within a predetermined volume of the target tissue.
[00560] In some embodiments, the composition includes a plurality of different modified nucleic acid molecules or mmRNA, where one or more than one of the modified nucleic acid molecules or mmRNA encodes a polypeptide of interest. Optionally, the composition also contains a cell penetration agent to assist in the intracellular delivery of the composition. A determination is made of the dose of the composition required to produce the polypeptide of interest in a substantial percentage of cells contained within the predetermined volume of the target tissue (generally, without inducing significant production of the polypeptide of interest in tissue adjacent to the predetermined volume, or distally to the target tissue). Subsequent to this determination, the determined dose is introduced directly into the tissue of the mammalian subject.
[00561] In one embodiment, the invention provides for the modified nucleic acid molecules or mmRNA to be delivered in more than one injection or by split dose injections.
[00562] In one embodiment, the invention may be retained near target tissue using a small disposable drug reservoir, patch pump or osmotic pump. Non-limiting examples of patch pumps include those manufactured and/or sold by BD®, (Franklin Lakes, NJ), Insulet Corporation (Bedford, MA) , SteadyMed Therapeutics (San Francisco, CA) , Medtronic (Minneapolis, MN) (e.g., MiniMed), UniLife (York, PA), Valeritas (Bridgewater, NJ), and SpringLeaf Therapeutics (Boston, MA). A non-limiting example of an osmotic pump include those manufactured by DURECT® (Cupertino, CA) (e.g., DUROS® and ALZET ®).
Pulmonary Administration
[00563] A pharmaceutical composition may be prepared, packaged, and/or sold in a formulation suitable for pulmonary administration via the buccal cavity. Such a formulation may comprise dry particles which comprise the active ingredient and which have a diameter in the range from about 0.5 nm to about 7 nm or from about 1 nm to about 6 nm. Such compositions are suitably in the form of dry powders for administration using a device comprising a dry powder reservoir to which a stream of propellant may be directed to disperse the powder and/or using a self propelling solvent/powder dispensing container such as a device comprising the active ingredient dissolved and/or suspended in a low-boiling propellant in a sealed container. Such powders comprise particles wherein at least 98% of the particles by weight have a diameter greater than 0.5 nm and at least 95% of the particles by number have a diameter less than 7 nm. Alternatively, at least 95% of the particles by weight have a diameter greater than 1 nm and at least 90% of the particles by number have a diameter less than 6 nm. Dry powder compositions may include a solid fine powder diluent such as sugar and are conveniently provided in a unit dose form.
[00564] Low boiling propellants generally include liquid propellants having a boiling point of below 65 °F at atmospheric pressure. Generally the propellant may constitute 50%> to 99.9%> (w/w) of the composition, and active ingredient may constitute 0.1% to 20% (w/w) of the composition. A propellant may further comprise additional ingredients such as a liquid non-ionic and/or solid anionic surfactant and/or a solid diluent (which may have a particle size of the same order as particles comprising the active ingredient).
[00565] As a non-limiting example, the modified nucleic acid molecules or mmRNA described herein may be formulated for pulmonary delivery by the methods described in U.S. Pat. No.
8,257,685; herein incorporated by reference in its entirety.
[00566] Pharmaceutical compositions formulated for pulmonary delivery may provide an active ingredient in the form of droplets of a solution and/or suspension. Such formulations may be prepared, packaged, and/or sold as aqueous and/or dilute alcoholic solutions and/or suspensions, optionally sterile, comprising active ingredient, and may conveniently be administered using any nebulization and/or atomization device. Such formulations may further comprise one or more additional ingredients including, but not limited to, a flavoring agent such as saccharin sodium, a volatile oil, a buffering agent, a surface active agent, and/or a preservative such as
methylhydroxybenzoate. Droplets provided by this route of administration may have an average diameter in the range from about 0.1 nm to about 200 nm.
Intranasal, nasal and buccal Administration
[00567] Formulations described herein as being useful for pulmonary delivery are useful for intranasal delivery of a pharmaceutical composition. Another formulation suitable for intranasal administration is a coarse powder comprising the active ingredient and having an average particle from about 0.2 μιη to 500 μιη. Such a formulation is administered in the manner in which snuff is taken, i.e. by rapid inhalation through the nasal passage from a container of the powder held close to the nose.
[00568] Formulations suitable for nasal administration may, for example, comprise from about as little as 0.1% (w/w) and as much as 100%) (w/w) of active ingredient, and may comprise one or more of the additional ingredients described herein. A pharmaceutical composition may be prepared, packaged, and/or sold in a formulation suitable for buccal administration. Such formulations may, for example, be in the form of tablets and/or lozenges made using conventional methods, and may, for example, 0.1% to 20% (w/w) active ingredient, the balance comprising an orally dissolvable and/or degradable composition and, optionally, one or more of the additional ingredients described herein. Alternately, formulations suitable for buccal administration may comprise a powder and/or an aerosolized and/or atomized solution and/or suspension comprising active ingredient. Such powdered, aerosolized, and/or aerosolized formulations, when dispersed, may have an average particle and/or droplet size in the range from about 0.1 nm to about 200 nm, and may further comprise one or more of any additional ingredients described herein.
Ophthalmic Administration
[00569] A pharmaceutical composition may be prepared, packaged, and/or sold in a formulation suitable for ophthalmic administration. Such formulations may, for example, be in the form of eye drops including, for example, a 0.1/1.0%) (w/w) solution and/or suspension of the active ingredient in an aqueous or oily liquid excipient. Such drops may further comprise buffering agents, salts, and/or one or more other of any additional ingredients described herein. Other ophthalmically- administrable formulations which are useful include those which comprise the active ingredient in microcrystalline form and/or in a liposomal preparation. Ear drops and/or eye drops are contemplated as being within the scope of this invention. A multilayer thin film device may be prepared to contain a pharmaceutical composition for delivery to the eye and/or surrounding tissue. Payload Administration: Detectable Agents and Therapeutic Agents
[00570] The modified nucleic acid molecules or mmRNA described herein can be used in a number of different scenarios in which delivery of a substance (the "payload") to a biological target is desired, for example delivery of detectable substances for detection of the target, or delivery of a therapeutic agent. Detection methods can include, but are not limited to, both imaging in vitro and in vivo imaging methods, e.g., immunohistochemistry, bio luminescence imaging (BLI), Magnetic Resonance Imaging (MRI), positron emission tomography (PET), electron microscopy, X-ray computed tomography, Raman imaging, optical coherence tomography, absorption imaging, thermal imaging, fluorescence reflectance imaging, fluorescence microscopy, fluorescence molecular tomographic imaging, nuclear magnetic resonance imaging, X-ray imaging, ultrasound imaging, photoacoustic imaging, lab assays, or in any situation where tagging/staining/imaging is required.
[00571] The modified nucleic acid molecules or mmRNA can be designed to include both a linker and a payload in any useful orientation. For example, a linker having two ends is used to attach one end to the payload and the other end to the nucleobase, such as at the C-7 or C-8 positions of the deaza-adenosine or deaza-guanosine or to the N-3 or C-5 positions of cytosine or uracil. The polynucleotide of the invention can include more than one payload (e.g., a label and a transcription inhibitor), as well as a cleavable linker.
[00572] In one embodiment, the modified nucleotide is a modified 7-deaza-adenosine triphosphate, where one end of a cleavable linker is attached to the C7 position of 7-deaza-adenine, the other end of the linker is attached to an inhibitor (e.g., to the C5 position of the nucleobase on a cytidine), and a label (e.g., Cy5) is attached to the center of the linker (see, e.g., compound 1 of A*pCp C5 Parg Capless in Fig. 5 and columns 9 and 10 of U.S. Pat. No. 7,994,304, incorporated herein by reference). Upon incorporation of the modified 7-deaza-adenosine triphosphate to an encoding region, the resulting polynucleotide having a cleavable linker attached to a label and an inhibitor (e.g., a polymerase inhibitor). Upon cleavage of the linker (e.g., with reductive conditions to reduce a linker having a cleavable disulfide moiety), the label and inhibitor are released. Additional linkers and payloads (e.g., therapeutic agents, detectable labels, and cell penetrating payloads) are described herein. [00573] Scheme 12 below depicts an exemplary modified nucleotide wherein the nucleobase, adenine, is attached to a linker at the C-7 carbon of 7-deaza adenine. In addition, Scheme 12 depicts the modified nucleotide with the linker and payload, e.g., a detectable agent, incorporated onto the 3' end of the mR A. Disulfide cleavage and 1 ,2-addition of the thiol group onto the propargyl ester releases the detectable agent. The remaining structure (depicted, for example, as pApC5Parg in Scheme 12) is the inhibitor. The rationale for the structure of the modified nucleotides is that the tethered inhibitor sterically interferes with the ability of the polymerase to incorporate a second base. Thus, it is critical that the tether be long enough to affect this function and that the inhibiter be in a stereochemical orientation that inhibits or prohibits second and follow on nucleotides into the growing polynucleotide strand.
Scheme 12
Figure imgf000187_0001
Figure imgf000188_0001
[00574] For example, the modified nucleic acid molecules or mmRNA described herein can be used in reprogramming induced pluripotent stem cells (iPS cells), which can directly track cells that are transfected compared to total cells in the cluster. In another example, a drug that may be attached to the modified nucleic acid molecules or mmRNA via a linker and may be fluorescently labeled can be used to track the drug in vivo, e.g. intracellularly. Other examples include, but are not limited to, the use of modified nucleic acid molecules or mmRNA in reversible drug delivery into cells.
[00575] The modified nucleic acid molecules or mmRNA described herein can be used in intracellular targeting of a payload, e.g., detectable or therapeutic agent, to specific organelle.
Exemplary intracellular targets can include, but are not limited to, the nuclear localization for advanced mRNA processing, or a nuclear localization sequence (NLS) linked to the mRNA containing an inhibitor. [00576] In addition, the modified nucleic acid molecules or mmRNA described herein can be used to deliver therapeutic agents to cells or tissues, e.g., in living animals. For example, the modified nucleic acids or mmRNA described herein can be used to deliver highly polar chemotherapeutics agents to kill cancer cells. The modified nucleic acid molecules or mmRNA attached to the therapeutic agent through a linker can facilitate member permeation allowing the therapeutic agent to travel into a cell to reach an intracellular target.
[00577] In one example, the linker is attached at the 2'-position of the ribose ring and/or at the 3' and/or 5' positionof the modified nucleic acid molecule or mmRNA (See e.g., International Pub. No. WO2012030683, herein incorporated by reference in its entirety). The linker may be any linker disclosed herein, known in the art and/or disclosed in International Pub. No. WO2012030683, herein incorporated by reference in its entirety.
[00578] In another example, the modified nucleic acid molecules or mmRNA can be attached to the modified nucleic acid molecules or mmRNA a viral inhibitory peptide (VIP) through a cleavable linker. The cleavable linker can release the VIP and dye into the cell. In another example, the modified nucleic acid molecules or mmRNA can be attached through the linker to an ADP- ribosylate, which is responsible for the actions of some bacterial toxins, such as cholera toxin, diphtheria toxin, and pertussis toxin. These toxin proteins are ADP-ribosyltransferases that modify target proteins in human cells. For example, cholera toxin ADP-ribosylates G proteins modifies human cells by causing massive fluid secretion from the lining of the small intestine, which results in life-threatening diarrhea.
[00579] In some embodiments, the payload may be a therapeutic agent such as a cytotoxin, radioactive ion, chemotherapeutic, or other therapeutic agent. A cytotoxin or cytotoxic agent includes any agent that may be detrimental to cells. Examples include, but are not limited to, taxol, cytochalasin B, gramicidin D, ethidium bromide, emetine, mitomycin, etoposide, teniposide, vincristine, vinblastine, colchicine, doxorubicin, daunorubicin, dihydroxyanthracinedione, mitoxantrone, mithramycin, actinomycin D, 1-dehydrotestosterone, glucocorticoids, procaine, tetracaine, lidocaine, propranolol, puromycin, maytansinoids, e.g., maytansinol (see U.S. Pat. No. 5,208,020 incorporated herein in its entirety), rachelmycin (CC-1065, see U.S. Pat. Nos. 5,475,092, 5,585,499, and 5,846,545, all of which are incorporated herein by reference), and analogs or homo logs thereof. Radioactive ions include, but are not limited to iodine {e.g., iodine 125 or iodine 131), strontium 89, phosphorous, palladium, cesium, iridium, phosphate, cobalt, yttrium 90, samarium 153, and praseodymium. Other therapeutic agents include, but are not limited to, antimetabolites (e.g., methotrexate, 6-mercaptopurine, 6-thioguanine, cytarabine, 5-fluorouracil decarbazine), alkylating agents (e.g., mechlorethamine, thiotepa chlorambucil, rachelmycin (CC- 1065), melphalan, carmustine (BSNU), lomustine (CCNU), cyclophosphamide, busulfan, dibromomannitol, streptozotocin, mitomycin C, and cis-dichlorodiamine platinum (II) (DDP) cisplatin), anthracyclines (e.g., daunorubicin (formerly daunomycin) and doxorubicin), antibiotics (e.g., dactinomycin (formerly actinomycin), bleomycin, mithramycin, and anthramycin (AMC)), and anti-mitotic agents (e.g., vincristine, vinblastine, taxol and maytansinoids).
[00580] In some embodiments, the payload may be a detectable agent, such as various organic small molecules, inorganic compounds, nanoparticles, enzymes or enzyme substrates, fluorescent materials, luminescent materials (e.g., luminol), bioluminescent materials (e.g., luciferase, luciferin, and aequorin), chemiluminescent materials, radioactive materials (e.g., 18F, 67Ga, 81mKr, 82Rb, mIn, 1231, 133Xe, 201T1, 1251, 35S, 14C, 3H, or 99mTc (e.g., as pertechnetate (technetate(VII), Tc04 ~)), and contrast agents (e.g., gold (e.g., gold nanoparticles), gadolinium (e.g., chelated Gd), iron oxides (e.g., superparamagnetic iron oxide (SPIO), monocrystalline iron oxide nanoparticles (MIONs), and ultrasmall superparamagnetic iron oxide (USPIO)), manganese chelates (e.g., Mn-DPDP), barium sulfate, iodinated contrast media (iohexol), microbubbles, or perfluorocarbons). Such optically- detectable labels include for example, without limitation, 4-acetamido-4'-isothiocyanatostilbene- 2,2'disulfonic acid; acridine and derivatives (e.g., acridine and acridine isothiocyanate); 5-(2'- aminoethyl)aminonaphthalene-l -sulfonic acid (EDANS); 4-amino-N-[3- vinylsulfonyl)phenyl]naphthalimide-3 ,5 disulfonate; N-(4-anilino-l-naphthyl)maleimide;
anthranilamide; BODIPY; Brilliant Yellow; coumarin and derivatives (e.g., coumarin, 7-amino-4- methylcoumarin (AMC, Coumarin 120), and 7-amino-4-trifluoromethylcoumarin (Coumarin 151)); cyanine dyes; cyanosine; 4',6-diaminidino-2-phenylindole (DAPI); 5 ' 5"-dibromopyrogallol- sulfonaphthalein (Bromopyrogallol Red); 7-diethylamino-3-(4'-isothiocyanatophenyl)-4- methylcoumarin; diethylenetriamine pentaacetate; 4,4'-diisothiocyanatodihydro-stilbene-2,2'- disulfonic acid; 4,4'-diisothiocyanatostilbene-2,2'-disulfonic acid; 5-[dimethylamino]-naphthalene- 1-sulfonyl chloride (DNS, dansylchloride); 4-dimethylaminophenylazophenyl-4' -isothiocyanate (DABITC); eosin and derivatives (e.g., eosin and eosin isothiocyanate); erythrosin and derivatives (e.g., erythrosin B and erythrosin isothiocyanate); ethidium; fluorescein and derivatives (e.g., 5- carboxyfluorescein (FAM), 5-(4,6-dichlorotriazin-2-yl)aminofluorescein (DTAF), 2',7'-dimethoxy- 4'5'-dichloro-6-carboxyfluorescein, fluorescein, fluorescein isothiocyanate, X-rhodamine-5-(and-6)- isothiocyanate (QFITC or XRITC), and fluorescamine); 2-[2-[3-[[l,3-dihydro-l,l-dimethyl-3-(3- sulfopropyl)-2H-benz[e]indol-2-ylidene]ethylidene]-2-[4-(ethoxycarbonyl)- 1 -piperazinyl]- 1 - cyclopenten- 1 -yl] ethenyl]- 1 , 1 -dimethyl-3 -(3 -sulforpropyl)- 1 H-benz[e]indolium hydroxide, inner salt, compound with n,n-diethylethanamine(l : 1) (IR144); 5-chloro-2-[2-[3-[(5-chloro-3-ethyl-2(3H)- benzothiazol- ylidene)ethylidene] -2-(diphenylamino)- 1 -cyclopenten- 1 -yl] ethenyl] -3 -ethyl benzothiazolium perchlorate (IR140); Malachite Green isothiocyanate; 4-methylumbelliferone orthocresolphthalein; nitrotyrosine; pararosaniline; Phenol Red; B-phycoerythrin; o- phthaldialdehyde; pyrene and derivatives(e.g., pyrene, pyrene butyrate, and succinimidyl 1- pyrene); butyrate quantum dots; Reactive Red 4 (CibacronTM Brilliant Red 3B-A); rhodamine and derivatives (e.g., 6-carboxy-X-rhodamine (ROX), 6-carboxyrhodamine (R6G), lissamine rhodamine B sulfonyl chloride rhodamine (Rhod), rhodamine B, rhodamine 123, rhodamine X
isothiocyanate, sulforhodamine B, sulforhodamine 101, sulfonyl chloride derivative of
sulforhodamine 101 (Texas Red), Ν,Ν,Ν ',Ν 'tetramethyl-6-carboxyrhodamine (TAMRA) tetramethyl rhodamine, and tetramethyl rhodamine isothiocyanate (TRITC)); riboflavin; rosolic acid; terbium chelate derivatives; Cyanine-3 (Cy3); Cyanine-5 (Cy5); cyanine-5.5 (Cy5.5), Cyanine-7 (Cy7); IRD 700; IRD 800; Alexa 647; La Jolta Blue; phthalo cyanine; and naphthalo cyanine.
[00581] In some embodiments, the detectable agent may be a non-detectable pre-cursor that becomes detectable upon activation (e.g., fluorogenic tetrazine-fluorophore constructs (e.g., tetrazine-BODIPY FL, tetrazine-Oregon Green 488, or tetrazine-BODIPY TMR-X) or enzyme activatable fluorogenic agents (e.g., PROSENSE® (VisEn Medical))). In vitro assays in which the enzyme labeled compositions can be used include, but are not limited to, enzyme linked
immunosorbent assays (ELISAs), immunoprecipitation assays, immunofluorescence, enzyme immunoassays (EIA), radioimmunoassays (RIA), and Western blot analysis. Combinations
[00582] The nucleic acid molecules or mmRNA may be used in combination with one or more other therapeutic, prophylactic, diagnostic, or imaging agents. By "in combination with," it is not intended to imply that the agents must be administered at the same time and/or formulated for delivery together, although these methods of delivery are within the scope of the present disclosure. Compositions can be administered concurrently with, prior to, or subsequent to, one or more other desired therapeutics or medical procedures. In general, each agent will be administered at a dose and/or on a time schedule determined for that agent. In some embodiments, the present disclosure encompasses the delivery of pharmaceutical, prophylactic, diagnostic, or imaging compositions in combination with agents that may improve their bioavailability, reduce and/or modify their metabolism, inhibit their excretion, and/or modify their distribution within the body. As a non- limiting example, the nucleic acid molecules or mmRNA may be used in combination with a pharmaceutical agent for the treatment of cancer or to control hyperproliferative cells. In U.S. Pat. No. 7,964,571, herein incorporated by reference in its entirety, a combination therapy for the treatment of solid primary or metastasized tumor is described using a pharmaceutical composition including a DNA plasmid encoding for interleukin-12 with a lipopolymer and also administering at least one anticancer agent or chemotherapeutic. Further, the nucleic acid molecules and mmRNA of the present invention that encodes anti-proliferative molecules may be in a pharmaceutical composition with a lipopolymer (see e.g., U.S. Pub. No. 20110218231, herein incorporated by reference in its entirety, claiming a pharmaceutical composition comprising a DNA plasmid encoding an anti-proliferative molecule and a lipopolymer) which may be administered with at least one chemotherapeutic or anticancer agent.
Cell Penetrating Payloads
[00583] In some embodiments, the modified nucleotides and modified nucleic acid molecules, which are incorporated into a nucleic acid, e.g., RNA or mRNA, can also include a payload that can be a cell penetrating moiety or agent that enhances intracellular delivery of the compositions. For example, the compositions can include, but are not limited to, a cell-penetrating peptide sequence that facilitates delivery to the intracellular space, e.g., HIV-derived TAT peptide, penetratins, transportans, or hCT derived cell-penetrating peptides, see, e.g., Caron et al, (2001) Mol Ther. 3(3):310-8; Langel, Cell-Penetrating Peptides: Processes and Applications (CRC Press, Boca Raton FL 2002); El-Andaloussi et al, (2005) Curr Pharm Des. 11(28):3597-611; and Deshayes et al, (2005) Cell Mol Life Sci. 62(16): 1839-49; all of which are incorporated herein by reference. The compositions can also be formulated to include a cell penetrating agent, e.g. , liposomes, which enhance delivery of the compositions to the intracellular space.
Biological Targets
[00584] The modified nucleotides and modified nucleic acid molecules described herein, which are incorporated into a nucleic acid, e.g., RNA or mRNA, can be used to deliver a payload to any biological target for which a specific ligand exists or can be generated. The ligand can bind to the biological target either covalently or non-covalently. [00585] Examples of biological targets include, but are not limited to, biopolymers, e.g., antibodies, nucleic acids such as R A and DNA, proteins, enzymes; examples of proteins include, but are not limited to, enzymes, receptors, and ion channels. In some embodiments the target may be a tissue - or a cell-type specific marker, e.g. , a protein that is expressed specifically on a selected tissue or cell type. In some embodiments, the target may be a receptor, such as, but not limited to, plasma membrane receptors and nuclear receptors; more specific examples include, but are not limited to, G-protein-coupled receptors, cell pore proteins, transporter proteins, surface-expressed antibodies, HLA proteins, MHC proteins and growth factor receptors.
Dosing
[00586] The present invention provides methods comprising administering modified mR As and their encoded proteins or complexes in accordance with the invention to a subject in need thereof. Nucleic acids, proteins or complexes, or pharmaceutical, imaging, diagnostic, or prophylactic compositions thereof, may be administered to a subject using any amount and any route of administration effective for preventing, treating, diagnosing, or imaging a disease, disorder, and/or condition (e.g., a disease, disorder, and/or condition relating to working memory deficits). The exact amount required will vary from subject to subject, depending on the species, age, and general condition of the subject, the severity of the disease, the particular composition, its mode of administration, its mode of activity, and the like. Compositions in accordance with the invention are typically formulated in dosage unit form for ease of administration and uniformity of dosage. It will be understood, however, that the total daily usage of the compositions of the present invention may be decided by the attending physician within the scope of sound medical judgment. The specific therapeutically effective, prophylactically effective, or appropriate imaging dose level for any particular patient will depend upon a variety of factors including the disorder being treated and the severity of the disorder; the activity of the specific compound employed; the specific composition employed; the age, body weight, general health, sex and diet of the patient; the time of
administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidental with the specific compound employed; and like factors well known in the medical arts.
[00587] In certain embodiments, compositions in accordance with the present invention may be administered at dosage levels sufficient to deliver from about 0.0001 mg/kg to about 100 mg/kg, from about 0.001 mg/kg to about 0.05 mg/kg, from about 0.005 mg/kg to about 0.05 mg/kg, from about 0.001 mg/kg to about 0.005 mg/kg, from about 0.05 mg/kg to about 0.5 mg/kg, from about 0.01 mg/kg to about 50 mg/kg, from about 0.1 mg/kg to about 40 mg/kg, from about 0.5 mg/kg to about 30 mg/kg, from about 0.01 mg/kg to about 10 mg/kg, from about 0.1 mg/kg to about 10 mg/kg, or from about 1 mg/kg to about 25 mg/kg, of subject body weight per day, one or more times a day, to obtain the desired therapeutic, diagnostic, prophylactic, or imaging effect. The desired dosage may be delivered three times a day, two times a day, once a day, every other day, every third day, every week, every two weeks, every three weeks, or every four weeks. In certain embodiments, the desired dosage may be delivered using multiple administrations (e.g., two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, fourteen, or more administrations).
[00588] According to the present invention, it has been discovered that administration of mmRNA in split-dose regimens produce higher levels of proteins in mammalian subjects. As used herein, a "split dose" is the division of single unit dose or total daily dose into two or more doses, e.g, two or more administrations of the single unit dose. As used herein, a "single unit dose" is a dose of any therapeutic administed in one dose/at one time/single route/single point of contact, i.e., single administration event. As used herein, a "total daily dose" is an amount given or prescribed in 24 hr period. It may be administered as a single unit dose. In one embodiment, the mmRNA of the present invention are administed to a subject in split doses. The mmRNA may be formulated in buffer only or in a formulation described herein.
Dosage Forms
[00589] A pharmaceutical composition described herein can be formulated into a dosage form described herein, such as a topical, intranasal, intratracheal, or injectable (e.g., intravenous, intraocular, intra vitreal, intramuscular, intracardiac, intraperitoneal, subcutaneous).
Liquid dosage forms
[00590] Liquid dosage forms for parenteral administration include, but are not limited to, pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups, and/or elixirs. In addition to active ingredients, liquid dosage forms may comprise inert diluents commonly used in the art including, but not limited to, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, dimethylformamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof. In certain embodiments for parenteral administration, compositions may be mixed with solubilizing agents such as CREMOPHOR®, alcohols, oils, modified oils, glycols, polysorbates, cyclodextrins, polymers, and/or combinations thereof.
Injectable
[00591] Injectable preparations, for example, sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art and may include suitable dispersing agents, wetting agents, and/or suspending agents. Sterile injectable preparations may be sterile injectable solutions, suspensions, and/or emulsions in nontoxic parenterally acceptable diluents and/or solvents, for example, a solution in 1,3-butanediol. Among the acceptable vehicles and solvents that may be employed include, but are not limited to, water, Ringer's solution, U.S.P., and isotonic sodium chloride solution. Sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose any bland fixed oil can be employed including synthetic mono- or diglycerides. Fatty acids such as oleic acid can be used in the preparation of injectables.
[00592] Injectable formulations can be sterilized, for example, by filtration through a bacterial- retaining filter, and/or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved or dispersed in sterile water or other sterile injectable medium prior to use.
[00593] In order to prolong the effect of an active ingredient, it may be desirable to slow the absorption of the active ingredient from subcutaneous or intramuscular injection. This may be accomplished by the use of a liquid suspension of crystalline or amorphous material with poor water solubility. The rate of absorption of modified mRNA then depends upon its rate of dissolution which, in turn, may depend upon crystal size and crystalline form. Alternatively, delayed absorption of a parenterally administered modified mRNA may be accomplished by dissolving or suspending the modified mRNA in an oil vehicle. Injectable depot forms are made by forming microencapsule matrices of the modified mRNA in biodegradable polymers such as polylactide-polyglycolide.
Depending upon the ratio of modified mRNA to polymer and the nature of the particular polymer employed, the rate of modified mRNA release can be controlled. Examples of other biodegradable polymers include, but are not limited to, poly(orthoesters) and poly(anhydrides). Depot injectable formulations may be prepared by entrapping the modified mRNA in liposomes or microemulsions which are compatible with body tissues. Pulmonary
[00594] Formulations described herein as being useful for pulmonary delivery may also be used for intranasal delivery of a pharmaceutical composition. Another formulation suitable for intranasal administration may be a coarse powder comprising the active ingredient and having an average particle from about 0.2 μιη to 500 μιη. Such a formulation may be administered in the manner in which snuff is taken, i.e. by rapid inhalation through the nasal passage from a container of the powder held close to the nose.
[00595] Formulations suitable for nasal administration may, for example, comprise from about as little as 0.1% (w/w) and as much as 100%) (w/w) of active ingredient, and may comprise one or more of the additional ingredients described herein. A pharmaceutical composition may be prepared, packaged, and/or sold in a formulation suitable for buccal administration. Such formulations may, for example, be in the form of tablets and/or lozenges made using conventional methods, and may, for example, contain about 0.1% to 20% (w/w) active ingredient, where the balance may comprise an orally dissolvable and/or degradable composition and, optionally, one or more of the additional ingredients described herein. Alternately, formulations suitable for buccal administration may comprise a powder and/or an aerosolized and/or atomized solution and/or suspension comprising active ingredient. Such powdered, aerosolized, and/or aerosolized formulations, when dispersed, may have an average particle and/or droplet size in the range from about 0.1 nm to about 200 nm, and may further comprise one or more of any additional ingredients described herein.
[00596] General considerations in the formulation and/or manufacture of pharmaceutical agents may be found, for example, in Remington: The Science and Practice of Pharmacy 21st ed.,
Lippincott Williams & Wilkins, 2005 (incorporated herein by reference in its entirety).
Coatings or Shells
[00597] Solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings and other coatings well known in the pharmaceutical formulating art. They may optionally comprise opacifying agents and can be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Examples of embedding compositions which can be used include polymeric substances and waxes. Solid compositions of a similar type may be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like. Properties of the Pharmaceutical Compositions
[00598] The pharmaceutical compositions described herein can be characterized by one or more of the following properties:
Bioavailability
[00599] The modified nucleic acid molecules and mmRNA, when formulated into a composition with a delivery agent as described herein, can exhibit an increase in bioavailability as compared to a composition lacking a delivery agent as described herein. As used herein, the term "bioavailability" refers to the systemic availability of a given amount of a modified nucleic acid molecule
administered to a mammal. Bioavailability can be assessed by measuring the area under the curve (AUC) or the maximum serum or plasma concentration (Cmax) of the unchanged form of a compound following administration of the compound to a mammal. AUC is a determination of the area under the curve plotting the serum or plasma concentration of a compound along the ordinate (Y-axis) against time along the abscissa (X-axis). Generally, the AUC for a particular compound can be calculated using methods known to those of ordinary skill in the art and as described in G. S.
Banker, Modern Pharmaceutics, Drugs and the Pharmaceutical Sciences, v. 72, Marcel Dekker, New York, Inc., 1996, herein incorporated by reference in its entirety.
[00600] The Cmax value is the maximum concentration of the compound achieved in the serum or plasma of a mammal following administration of the compound to the mammal. The Cmax value of a particular compound can be measured using methods known to those of ordinary skill in the art. The phrases "increasing bioavailability" or "improving the pharmacokinetics," as used herein mean that the systemic availability of a first modified nucleic acid molecule, measured as AUC, Cmax, or Cmin in a mammal is greater, when co-administered with a delivery agent as described herein, than when such co-administration does not take place. In some embodiments, the bioavailability of the modified nucleic acid molecule can increase by at least about 2%, at least about 5%, at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%), at least about 40%>, at least about 45%, at least about 50%>, at least about 55%, at least about 60%), at least about 65%, at least about 70%>, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, or about 100%.
Therapeutic Window
[00601] The modified nucleic acid molecules and mmRNA when formulated into a composition with a delivery agent as described herein, can exhibit an increase in the therapeutic window of the administered modified nucleic acid molecule composition as compared to the therapeutic window of the administered modified nucleic acid molecule composition lacking a delivery agent as described herein. As used herein "therapeutic window" refers to the range of plasma concentrations, or the range of levels of therapeutically active substance at the site of action, with a high probability of eliciting a therapeutic effect. In some embodiments, the therapeutic window of the modified nucleic acid molecule when co-administered with a delivery agent as described herein can increase by at least about 2%, at least about 5%, at least about 10%, at least about 15%, at least about 20%, at least about 25%o, at least about 30%>, at least about 35%, at least about 40%>, at least about 45%, at least about 50%), at least about 55%, at least about 60%>, at least about 65%, at least about 70%, at least about 75%), at least about 80%, at least about 85%, at least about 90%, at least about 95%, or about 100%.
Volume of Distribution
[00602] The modified nucleic acid molecules, when formulated into a composition with a delivery agent as described herein, can exhibit an improved volume of distribution (Vdist), e.g., reduced or targeted, relative to a modified nucleic acid molecule composition lacking a delivery agent as described herein. The volume of distribution (Vdist) relates the amount of the drug in the body to the concentration of the drug in the blood or plasma. As used herein, the term "volume of distribution" refers to the fluid volume that would be required to contain the total amount of the drug in the body at the same concentration as in the blood or plasma: Vdist equals the amount of drug in the body/concentration of drug in blood or plasma. For example, for a 10 mg dose and a plasma concentration of 10 mg/L, the volume of distribution would be 1 liter. The volume of distribution reflects the extent to which the drug is present in the extravascular tissue. A large volume of distribution reflects the tendency of a compound to bind to the tissue components compared with plasma protein binding. In a clinical setting, Vdist can be used to determine a loading dose to achieve a steady state concentration. In some embodiments, the volume of distribution of the modified nucleic acid molecule when co -administered with a delivery agent as described herein can decrease at least about 2%, at least about 5%, at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%. Biological Effect [00603] In one embodiment, the biological effect of the modified mRNA delivered to the animals may be categorized by analyzing the protein expression in the animals. The protein expression may be determined from analyzing a biological sample collected from a mammal administered the modified mRNA of the present invention. In one embodiment, the expression protein encoded by the modified mRNA administered to the mammal of at least 50 pg/ml may be preferred. For example, a protein expression of 50-200 pg/ml for the protein encoded by the modified mRNA delivered to the mammal may be seen as a therapeutically effective amount of protein in the mammal.
Detection of Modified Nucleic Acids by Mass Spectrometry
[00604] Mass spectrometry (MS) is an analytical technique that can provide structural and molecular mass/concentration information on molecules after their conversion to ions. The molecules are first ionized to acquire positive or negative charges and then they travel through the mass analyzer to arrive at different areas of the detector according to their mass/charge (m/z) ratio.
[00605] Mass spectrometry is performed using a mass spectrometer which includes an ion source for ionizing the fractionated sample and creating charged molecules for further analysis. For example ionization of the sample may be performed by electrospray ionization (ESI), atmospheric pressure chemical ionization (APCI), photoionization, electron ionization, fast atom bombardment (FAB)/liquid secondary ionization (LSIMS), matrix assisted laser desorption/ionization (MALDI), field ionization, field desorption, thermospray/plasmaspray ionization, and particle beam ionization. The skilled artisan will understand that the choice of ionization method can be determined based on the analyte to be measured, type of sample, the type of detector, the choice of positive versus negative mode, etc.
[00606] After the sample has been ionized, the positively charged or negatively charged ions thereby created may be analyzed to determine a mass-to-charge ratio (i.e., m/z). Suitable analyzers for determining mass-to-charge ratios include quadropole analyzers, ion traps analyzers, and time- of-flight analyzers. The ions may be detected using several detection modes. For example, selected ions may be detected (i.e., using a selective ion monitoring mode (SIM)), or alternatively, ions may be detected using a scanning mode, e.g., multiple reaction monitoring (MRM) or selected reaction monitoring (SRM).
[00607] Liquid chromatography-multiple reaction monitoring (LC-MS/MRM) coupled with stable isotope labeled dilution of peptide standards has been shown to be an effective method for protein verification (e.g., Keshishian et al., Mol Cell Proteomics 2009 8: 2339-2349; Kuhn et al., Clin Chem 2009 55:1108-1117; Lopez et al, Clin Chem 2010 56:281-290; each of which are herein
incorporated by reference in its entirety). Unlike untargeted mass spectrometry frequently used in biomarker discovery studies, targeted MS methods are peptide sequence-based modes of MS that focus the full analytical capacity of the instrument on tens to hundreds of selected peptides in a complex mixture. By restricting detection and fragmentation to only those peptides derived from proteins of interest, sensitivity and reproducibility are improved dramatically compared to discovery- mode MS methods. This method of mass spectrometry-based multiple reaction monitoring (MRM) quantitation of proteins can dramatically impact the discovery and quantitation of biomarkers via rapid, targeted, multiplexed protein expression profiling of clinical samples.
[00608] In one embodiment, a biological sample which may contain at least one protein encoded by at least one modified mRNA of the present invention may be analyzed by the method of MRM -MS. The quantification of the biological sample may further include, but is not limited to, isotopically labeled peptides or proteins as internal standards.
[00609] According to the present invention, the biological sample, once obtained from the subject, may be subjected to enzyme digestion. As used herein, the term "digest" means to break apart into shorter peptides. As used herein, the phrase "treating a sample to digest proteins" means
manipulating a sample in such a way as to break down proteins in a sample. These enzymes include, but are not limited to, trypsin, endoproteinase Glu-C and chymotrypsin. In one embodiment, a biological sample which may contain at least one protein encoded by at least one modified mRNA of the present invention may be digested using enzymes.
[00610] In one embodiment, a biological sample which may contain protein encoded by modified mRNA of the present invention may be analyzed for protein using electrospray ionization.
Electrospray ionization (ESI) mass spectrometry (ESIMS) uses electrical energy to aid in the transfer of ions from the solution to the gaseous phase before they are analyzed by mass spectrometry.
Samples may be analyzed using methods known in the art (e.g., Ho et al., Clin Biochem Rev. 2003 24(1):3-12; herein incorporated by reference in its entirety). The ionic species contained in solution may be transferred into the gas phase by dispersing a fine spray of charge droplets, evaporating the solvent and ejecting the ions from the charged droplets to generate a mist of highly charged droplets. The mist of highly charged droplets may be analyzed using at least 1, at least 2, at least 3 or at least 4 mass analyzers such as, but not limited to, a quadropole mass analyzer. Further, the mass spectrometry method may include a purification step. As a non- limiting example, the first quadrapole may be set to select a single m/z ratio so it may filter out other molecular ions having a different m/z ratio which may eliminate complicated and time-consuming sample purification procedures prior to MS analysis.
[00611] In one embodiment, a biological sample which may contain protein encoded by modified mR A of the present invention may be analyzed for protein in a tandem ESIMS system (e.g., MS/MS). As non-limiting examples, the droplets may be analyzed using a product scan (or daughter scan) a precursor scan (parent scan) a neutral loss or a multiple reaction monitoring.
[00612] In one embodiment, a biological sample which may contain protein encoded by modified mRNA of the present invention may be analyzed using matrix-assisted laser desorption/ionization (MALDI) mass spectrometry (MALDIMS). MALDI provides for the nondestructive vaporization and ionization of both large and small molecules, such as proteins. In MALDI analysis, the analyte is first co-crystallized with a large molar excess of a matrix compound, which may also include, but is not limited to, an ultraviolet absorbing weak organic acid. Non-limiting examples of matrices used in MALDI are a-cyano-4-hydroxycinnamic acid, 3,5-dimethoxy-4-hydroxycinnamic acid and 2,5-dihydroxybenzoic acid. Laser radiation of the analyte-matrix mixture may result in the vaporization of the matrix and the analyte. The laser induced desorption provides high ion yields of the intact analyte and allows for measurement of compounds with high accuracy. Samples may be analyzed using methods known in the art (e.g., Lewis, Wei and Siuzdak, Encyclopedia of Analytical Chemistry 2000:5880-5894; herein incorporated by reference in its entirety). As non-limiting examples, mass analyzers used in the MALDI analysis may include a linear time-of-flight (TOF), a TOF reflectron or a Fourier transform mass analyzer.
[00613] In one embodiment, the analyte-matrix mixture may be formed using the dried-droplet method. A biologic sample is mixed with a matrix to create a saturated matrix solution where the matrix-to-sample ratio is approximately 5000: 1. An aliquot (approximately 0.5-2.0 uL) of the saturated matrix solution is then allowed to dry to form the analyte-matrix mixture.
[00614] In one embodiment, the analyte-matrix mixture may be formed using the thin-layer method. A matrix homogeneous film is first formed and then the sample is then applied and may be absorbed by the matrix to form the analyte-matrix mixture.
[00615] In one embodiment, the analyte-matrix mixture may be formed using the thick-layer method. A matrix homogeneous film is formed with a nitro-cellulose matrix additive. Once the uniform nitro-cellulose matrix layer is obtained the sample is applied and absorbed into the matrix to form the analyte-matrix mixture.
[00616] In one embodiment, the analyte-matrix mixture may be formed using the sandwich method. A thin layer of matrix crystals is prepared as in the thin-layer method followed by the addition of droplets of aqueous trifluoroacetic acid, the sample and matrix. The sample is then absorbed into the matrix to form the analyte-matrix mixture.
Uses of Modified Nucleic Acid Molecules
Therapeutic Agents
[00617] The modified nucleic acid molecules and the proteins translated from the modified nucleic acid molecules described herein can be used as therapeutic agents. For example, a modified nucleic acid molecule described herein can be administered to a subject, wherein the modified nucleic acid molecule is translated in vivo to produce a therapeutic peptide in the subject. Accordingly, provided herein are compositions, methods, kits, and reagents for treatment or prevention of disease or conditions in humans and other mammals. The active therapeutic agents of the present disclosure include, but are not limited to, modified nucleic acid molecules, cells containing modified nucleic acid molecules or polypeptides translated from the modified nucleic acid molecules, polypeptides translated from modified nucleic acid molecules, and cells contacted with cells containing modified nucleic acid molecules or polypeptides translated from the modified nucleic acid molecules.
[00618] In certain embodiments, combination therapeutics are provided which may containing one or more modified nucleic acid molecules containing translatable regions along with a protein that induces antibody-dependent cellular toxicity. As used herein "translatable regions" encode for a protein or proteins that may boost a subject's immunity. For example, provided herein are therapeutics containing one or more nucleic acids that encode trastuzumab and granulocyte-colony stimulating factor (G-CSF). In particular, such combination therapeutics may be useful in Her2+ breast cancer patients who develop induced resistance to trastuzumab. (See, e.g., Albrecht,
Immunotherapy. 2(6):795-8 (2010); herein incorporated by reference in its entirety).
[00619] Methods of inducing translation of a recombinant polypeptide in a cell population using the modified nucleic acid molecules described herein are also provided. Such translation can be in vivo, ex vivo, in culture, or in vitro. The cell population may be contacted with an effective amount of a composition containing a nucleic acid that has at least one nucleoside modification, and a
translatable region encoding the recombinant polypeptide. The population may be contacted under conditions such that the nucleic acid may be localized into one or more cells of the cell population and the recombinant polypeptide may be translated in the cell from the nucleic acid.
[00620] An effective amount of the composition may be provided based, at least in part, on the target tissue, target cell type, means of administration, physical characteristics of the nucleic acid (e.g., size, and extent of modified nucleosides), and other determinants. In general, an effective amount of the composition provides efficient protein production in the cell, preferably more efficient than a composition containing a corresponding unmodified nucleic acid molecule. Increased efficiency may be demonstrated by increased cell transfection (i.e., the percentage of cells transfected with the nucleic acid), increased protein translation from the nucleic acid, decreased nucleic acid degradation (as demonstrated, e.g. , by increased duration of protein translation from a modified nucleic acid molecule), or reduced innate immune response of the host cell.
[00621] Aspects of the present disclosure are directed to methods of inducing in vivo translation of a recombinant polypeptide in a mammalian subject in need thereof. Therein, an effective amount of a composition containing a nucleic acid that has at least one nucleoside modification and a translatable region encoding the recombinant polypeptide may be administered to the subject using the delivery methods described herein. The nucleic acid may be provided in an amount and under other conditions such that the nucleic acid is localized into a cell of the subject and the recombinant polypeptide may be translated in the cell from the nucleic acid. The cell in which the nucleic acid is localized, or the tissue in which the cell is present, may be targeted with one or more than one rounds of nucleic acid administration.
[00622] Other aspects of the present disclosure relate to transplantation of cells containing modified nucleic acid molecules to a mammalian subject. Administration of cells to mammalian subjects is known to those of ordinary skill in the art, and include, but is not limited to, local implantation (e.g., topical or subcutaneous administration), organ delivery or systemic injection (e.g., intravenous injection or inhalation), and the formulation of cells in pharmaceutically acceptable carrier.
Compositions containing modified nucleic acid molecules are formulated for administration intramuscularly, transarterially, intraperitoneally, intravenously, intranasally, subcutaneously, endoscopically, transdermally, or intrathecally. In some embodiments, the composition may be formulated for extended release.
[00623] The subject to whom the therapeutic agent may be administered suffers from or may be at risk of developing a disease, disorder, or deleterious condition. Provided are methods of identifying, diagnosing, and classifying subjects on these bases, which may include clinical diagnosis, biomarker levels, genome-wide association studies (GWAS), and other methods known in the art.
[00624] In certain embodiments, the administered modified nucleic acid molecule directs production of one or more recombinant polypeptides that provide a functional activity which may be substantially absent in the cell in which the recombinant polypeptide may be translated. For example, the missing functional activity may be enzymatic, structural, or gene regulatory in nature.
[00625] In other embodiments, the administration of a modified nucleic acid molecule directs production of one or more recombinant polypeptides that replace a polypeptide (or multiple polypeptides) that may be substantially absent in the cell in which the recombinant polypeptide may be translated. Such absence may be due to a genetic mutation of the encoding gene or a regulatory pathway thereof. Alternatively, the recombinant polypeptide functions to antagonize the activity of an endogenous protein present in, on the surface of, or secreted from the cell. Usually, the activity of the endogenous protein may be deleterious to the subject, for example, due to the mutation of the endogenous protein resulting in altered activity or localization. Additionally, the recombinant polypeptide antagonizes, directly or indirectly, the activity of a biological moiety present in, on the surface of, or secreted from the cell. Examples of antagonized biological moieties include, but are not limited to, lipids (e.g., cholesterol), a lipoprotein (e.g., low density lipoprotein), a nucleic acid, a carbohydrate, or a small molecule toxin.
[00626] The recombinant proteins described herein may be engineered for localization within the cell, potentially within a specific compartment such as the nucleus, or are engineered for secretion from the cell or translocation to the plasma membrane of the cell.
[00627] As described herein, a useful feature of the modified nucleic acid molecules of the present disclosure is the capacity to reduce the innate immune response of a cell to an exogenous nucleic acid. Provided are methods for performing the titration, reduction or elimination of the immune response in a cell or a population of cells. In some embodiments, the cell may be contacted with a first composition that contains a first dose of a first exogenous nucleic acid including a translatable region and at least one nucleoside modification, and the level of the innate immune response of the cell to the first exogenous nucleic acid may be determined. Subsequently, the cell may be contacted with a second composition, which includes a second dose of the first exogenous nucleic acid, the second dose containing a lesser amount of the first exogenous nucleic acid as compared to the first dose. Alternatively, the cell may be contacted with a first dose of a second exogenous nucleic acid. The second exogenous nucleic acid may contain one or more modified nucleosides, which may be the same or different from the first exogenous nucleic acid or, alternatively, the second exogenous nucleic acid may not contain modified nucleosides. The steps of contacting the cell with the first composition and/or the second composition may be repeated one or more times. Additionally, efficiency of protein production (e.g. , protein translation) in the cell may be optionally determined, and the cell may be re-transfected with the first and/or second composition repeatedly until a target protein production efficiency is achieved.
Therapeutics for diseases and conditions
[00628] Provided herein are methods for treating or preventing a symptom of diseases,
characterized by missing or aberrant protein activity, by supplying the missing protein activity or overcoming the aberrant protein activity. Because of the rapid initiation of protein production following introduction of modified mRNA, as compared to viral DNA vectors, the compounds of the present disclosure are particularly advantageous in treating acute diseases such as sepsis, stroke, and myocardial infarction. Moreover, an accurate titration of protein may be achievable using the modified mRNA of the present disclosure as the modified mRNA may be able to alter transcription rates and thus cause changes in gene expression.
[00629] Diseases characterized by dysfunctional or aberrant protein activity include, but are not limited to, cancer and proliferative diseases, genetic diseases {e.g., cystic fibrosis), autoimmune diseases, diabetes, neurodegenerative diseases, cardiovascular diseases, and metabolic diseases. The present disclosure provides a method for treating such conditions or diseases in a subject by introducing nucleic acid or cell-based therapeutics containing the modified nucleic acid molecules provided herein, wherein the modified nucleic acid molecules encode for a protein that antagonizes or otherwise overcomes the aberrant protein activity present in the cell of the subject. Specific examples of a dysfunctional protein include, but are not limited to, the missense mutation variants of the cystic fibrosis transmembrane conductance regulator (CFTR) gene, which produce a
dysfunctional protein variant of CFTR protein, which causes cystic fibrosis.
[00630] Multiple diseases may be characterized by missing (or substantially diminished such that proper protein function does not occur) protein activity. Such proteins may not be present, or they may be essentially non-functional.
[00631] Thus, provided are methods of treating cystic fibrosis in a mammalian subject by contacting a cell of the subject with a modified nucleic acid molecule having a translatable region that encodes a functional CFTR polypeptide, under conditions such that an effective amount of the CTFR polypeptide is present in the cell. Preferred target cells are epithelial cells, such as the lung, and methods of administration are determined in view of the target tissue; i.e., for lung delivery, the RNA molecules are formulated for administration by inhalation.
[00632] In another embodiment, the present disclosure provides a method for treating
hyperlipidemia in a subject, by introducing into a cell population of the subject with a modified mRNA molecule encoding Sortilin, a protein recently characterized by genomic studies, thereby ameliorating the hyperlipidemia in a subject. The SORTl gene encodes a trans-Golgi network (TGN) transmembrane protein called Sortilin. Genetic studies have shown that one of five individuals has a single nucleotide polymorphism, rsl2740374, in the lpl3 locus of the SORTl gene that predisposes them to having low levels of low-density lipoprotein (LDL) and very-low-density lipoprotein (VLDL). Each copy of the minor allele, present in about 30% of people, alters LDL cholesterol by 8 mg/dL, while two copies of the minor allele, present in about 5% of the population, lowers LDL cholesterol 16 mg/dL. Carriers of the minor allele have also been shown to have a 40% decreased risk of myocardial infarction. Functional in vivo studies in mice describes that overexpression of SORTl in mouse liver tissue led to significantly lower LDL-cholesterol levels, as much as 80%> lower, and that silencing SORTl increased LDL cholesterol approximately 200% (Musunuru K et al. From noncoding variant to phenotype via SORTl at the lpl3 cholesterol locus. Nature 2010; 466: 714-721; herein incorporated by reference in its entirety).
Methods of cellular nucleic acid delivery
[00633] Methods of the present disclosure enhance nucleic acid delivery into a cell population, in vivo, ex vivo, or in culture. For example, a cell culture containing a plurality of host cells {e.g., eukaryotic cells such as yeast or mammalian cells) may be contacted with a composition that contains an modified nucleic acid molecule having at least one nucleoside modification and, optionally, a translatable region. The composition may also generally contain a transfection reagent or other compound that may increases the efficiency of modified nucleic acid molecule uptake into the host cells. The modified nucleic acid molecule may exhibit enhanced retention in the cell population, relative to a corresponding unmodified nucleic acid molecule. The retention of the modified nucleic acid molecule may greater than the retention of the unmodified nucleic acid molecule. In some embodiments, it is at least about 50%, 75%, 90%, 95%, 100%, 150%, 200% or more than 200% greater than the retention of the unmodified nucleic acid molecule. Such retention advantage may be achieved by one round of transfection with the modified nucleic acid molecule, or may be obtained following repeated rounds of transfection.
[00634] In some embodiments, the modified nucleic acid molecule may be delivered to a target cell population with one or more additional nucleic acids. Such delivery may be at the same time, or the modified nucleic acid molecule is delivered prior to delivery of the one or more additional nucleic acids. The additional one or more nucleic acids may be modified nucleic acid molecules or unmodified nucleic acid molecules. It is understood that the initial presence of the modified nucleic acid molecules may not substantially induce an innate immune response of the cell population and, moreover, that the innate immune response may not be activated by the later presence of the unmodified nucleic acid molecules. In this regard, the enhanced nucleic acid may not itself contain a translatable region, if the protein desired to be present in the target cell population is translated from the unmodified nucleic acid molecules.
Targeting Moieties
[00635] In some embodiments, modified nucleic acid molecules are provided to express a protein- binding partner or a receptor on the surface of the cell, which may function to target the cell to a specific tissue space or to interact with a specific moiety, either in vivo or in vitro. Suitable protein- binding partners include, but are not limited to, antibodies and functional fragments thereof, scaffold proteins, or peptides. Additionally, modified nucleic acid molecules may be employed to direct the synthesis and extracellular localization of lipids, carbohydrates, or other biological moieties.
Permanent Gene Expression Silencing
[00636] A method for epigenetically silencing gene expression in a mammalian subject, comprising a nucleic acid where the translatable region encodes a polypeptide or polypeptides capable of directing sequence-specific histone H3 methylation to initiate hetero chromatin formation and reduce gene transcription around specific genes for the purpose of silencing the gene. For example, a gain- of-function mutation in the Janus Kinase 2 gene is responsible for the family of Myeloproliferative Diseases.
Expression of Ligand or Receptor on Cell Surface
[00637] In some aspects and embodiments of the aspects described herein, the modified R A can be used to express a ligand or ligand receptor on the surface of a cell (e.g., a homing moiety). A ligand or ligand receptor moiety attached to a cell surface can permit the cell to have a desired biological interaction with a tissue or an agent in vivo. A ligand can be an antibody, an antibody fragment, an aptamer, a peptide, a vitamin, a carbohydrate, a protein or polypeptide, a receptor, e.g., cell-surface receptor, an adhesion molecule, a glycoprotein, a sugar residue, a therapeutic agent, a drug, a glycosaminoglycan, or any combination thereof. For example, a ligand can be an antibody that recognizes a cancer-cell specific antigen, rendering the cell capable of preferentially interacting with tumor cells to permit tumor-specific localization of a modified cell. A ligand can confer the ability of a cell composition to accumulate in a tissue to be treated, since a preferred ligand may be capable of interacting with a target molecule on the external face of a tissue to be treated. Ligands having limited cross-reactivity to other tissues are generally preferred.
[00638] In some cases, a ligand can act as a homing moiety which permits the cell to target to a specific tissue or interact with a specific ligand. Such homing moieties can include, but are not limited to, any member of a specific binding pair, antibodies, monoclonal antibodies, or derivatives or analogs thereof, including without limitation: Fv fragments, single chain Fv (scFv) fragments, Fab' fragments, F(ab')2 fragments, single domain antibodies, camelized antibodies and antibody fragments, humanized antibodies and antibody fragments, and multivalent versions of the foregoing; multivalent binding reagents including without limitation: monospecific or bispecific antibodies, such as disulfide stabilized Fv fragments, scFv tandems ((SCFV)2 fragments), diabodies, tribodies or tetrabodies, which typically are covalently linked or otherwise stabilized (i.e., leucine zipper or helix stabilized) scFv fragments; and other homing moieties include for example, aptamers, receptors, and fusion proteins.
[00639] In some embodiments, the homing moiety may be a surface-bound antibody, which can permit tuning of cell targeting specificity. This is especially useful since highly specific antibodies can be raised against an epitope of interest for the desired targeting site. In one embodiment, multiple antibodies are expressed on the surface of a cell, and each antibody can have a different specificity for a desired target. Such approaches can increase the avidity and specificity of homing interactions.
[00640] A skilled artisan can select any homing moiety based on the desired localization or function of the cell, for example an estrogen receptor ligand, such as tamoxifen, can target cells to estrogen- dependent breast cancer cells that have an increased number of estrogen receptors on the cell surface. Other non-limiting examples of ligand/receptor interactions include CCRI (e.g., for treatment of inflamed joint tissues or brain in rheumatoid arthritis, and/or multiple sclerosis), CCR7, CCR8 (e.g., targeting to lymph node tissue), CCR6, CCR9,CCR10 (e.g., to target to intestinal tissue), CCR4, CCR10 (e.g., for targeting to skin), CXCR4 (e.g., for general enhanced transmigration), HCELL (e.g., for treatment of inflammation and inflammatory disorders, bone marrow), Alpha4beta7 (e.g., for intestinal mucosa targeting), VLA-4/VCAM-1 (e.g., targeting to endothelium). In general, any receptor involved in targeting (e.g., cancer metastasis) can be harnessed for use in the methods and compositions described herein.
Mediators of Cell Death
[00641] In one embodiment, a modified nucleic acid molecule composition can be used to induce apoptosis in a cell (e.g., a cancer cell) by increasing the expression of a death receptor, a death receptor ligand or a combination thereof. This method can be used to induce cell death in any desired cell and has particular usefulness in the treatment of cancer where cells escape natural apoptotic signals.
[00642] Apoptosis can be induced by multiple independent signaling pathways that converge upon a final effector mechanism consisting of multiple interactions between several "death receptors" and their ligands, which belong to the tumor necrosis factor (TNF) receptor/ligand superfamily. The best-characterized death receptors are CD95 ("Fas"), TNFRI (p55), death receptor 3 (DR3 or Apo3/TRAMO), DR4 and DR5 (apo2-TRAIL-R2). The final effector mechanism of apoptosis may be the activation of a series of proteinases designated as caspases. The activation of these caspases results in the cleavage of a series of vital cellular proteins and cell death. The molecular mechanism of death receptors/ligands-induced apoptosis is well known in the art. For example, Fas/FasL- mediated apoptosis is induced by binding of three FasL molecules which induces trimerization of Fas receptor via C-terminus death domains (DDs), which in turn recruits an adapter protein FADD (Fas-associated protein with death domain) and Caspase-8. The oligomerization of this trimolecular complex, Fas/FAIDD/caspase-8, results in proteolytic cleavage of proenzyme caspase-8 into active caspase-8 that, in turn, initiates the apoptosis process by activating other downstream caspases through proteolysis, including caspase-3. Death ligands in general are apoptotic when formed into trimers or higher order of structures. As monomers, they may serve as antiapoptotic agents by competing with the trimers for binding to the death receptors.
[00643] In one embodiment, the modified nucleic acid molecule composition encodes for a death receptor (e.g., Fas, TRAIL, TRAMO, TNFR, TLR etc). Cells made to express a death receptor by transfection of modified RNA become susceptible to death induced by the ligand that activates that receptor. Similarly, cells made to express a death ligand, e.g., on their surface, will induce death of cells with the receptor when the transfected cell contacts the target cell. In another embodiment, the modified RNA composition encodes for a death receptor ligand (e.g., FasL, TNF, etc). In another embodiment, the modified RNA composition encodes a caspase (e.g., caspase 3, caspase 8, caspase 9 etc). Where cancer cells often exhibit a failure to properly differentiate to a non-pro liferative or controlled proliferative form, in another embodiment, the synthetic, modified RNA composition encodes for both a death receptor and its appropriate activating ligand. In another embodiment, the synthetic, modified RNA composition encodes for a differentiation factor that when expressed in the cancer cell, such as a cancer stem cell, will induce the cell to differentiate to a non-pathogenic or nonself-renewing phenotype (e.g., reduced cell growth rate, reduced cell division etc) or to induce the cell to enter a dormant cell phase (e.g., G0 resting phase).
[00644] One of skill in the art will appreciate that the use of apoptosis-inducing techniques may require that the modified nucleic acid molecules are appropriately targeted to e.g., tumor cells to prevent unwanted wide-spread cell death. Thus, one can use a delivery mechanism (e.g., attached ligand or antibody, targeted liposome etc) that recognizes a cancer antigen such that the modified nucleic acid molecules are expressed only in cancer cells.
Kits and Devices
Kits
[00645] The invention provides a variety of kits for conveniently and/or effectively carrying out methods of the present invention. Typically kits will comprise sufficient amounts and/or numbers of components to allow a user to perform multiple treatments of a subject(s) and/or to perform multiple experiments.
[00646] In one aspect, the present invention provides kits for protein production, comprising a first modified nucleic acid molecule or mrnRNA comprising a translatable region. The kit may further comprise packaging and instructions and/or a delivery agent to form a formulation composition. The delivery agent may comprise a saline, a buffered solution, a lipidoid or any delivery agent disclosed herein.
[00647] In one embodiment, the buffer solution may include sodium chloride, calcium chloride, phosphate and/or EDTA. In another embodiment, the buffer solution may include, but is not limited to, saline, saline with 2mM calcium, 5% sucrose, 5% sucrose with 2mM calcium, 5% Mannitol, 5% Mannitol with 2mM calcium, Ringer's lactate, sodium chloride, sodium chloride with 2mM calcium and mannose (See e.g., U.S. Pub. No. 20120258046; herein incorporated by reference in its entirety). In a futher embodiment, the buffer solutions may be precipitated or it may be lyophilized. The amount of each component may be varied to enable consistent, reproducible higher concentration saline or simple buffer formulations. The components may also be varied in order to increase the stability of modified nucleic acid molecules and mmRNA in the buffer solution over a period of time and/or under a variety of conditions.
[00648] In one aspect, the present invention provides kits for protein production, comprising: a modified nucleic acid molecule or mmRNA comprising a translatable region, provided in an amount effective to produce a desired amount of a protein encoded by the translatable region when introduced into a target cell; a second modified nucleic acid molecule or mmRNA comprising an inhibitory nucleic acid, provided in an amount effective to substantially inhibit the innate immune response of the cell; and packaging and instructions.
[00649] In one aspect, the present invention provides kits for protein production, comprising a modified nucleic acid molecule or mmRNA comprising a translatable region, wherein the nucleic acid exhibits reduced degradation by a cellular nuclease, and packaging and instructions.
[00650] In one aspect, the present invention provides kits for protein production, comprising a modified nucleic acid molecule or mmRNA comprising a translatable region, wherein the nucleic acid exhibits reduced degradation by a cellular nuclease, and a mammalian cell suitable for translation of the translatable region of the first nucleic acid.
Devices
[00651] The present invention provides for devices which may incorporate modified nucleic acid molecules or mmRNA that encode polypeptides of interest. These devices contain in a stable formulation the reagents to synthesize a nucleic acid in a formulation available to be immediately delivered to a subject in need thereof, such as a human patient. Non-limiting examples of such a polypeptide of interest include a growth factor and/or angiogenesis stimulator for wound healing, a peptide antibiotic to facilitate infection control, and an antigen to rapidly stimulate an immune response to a newly identified virus.
[00652] In some embodiments the device is self-contained, and is optionally capable of wireless remote access to obtain instructions for synthesis and/or analysis of the generated modified nucleic acid molecule or mmRNA. The device is capable of mobile synthesis of at least one modified nucleic acid molecule or mmRNA and preferably an unlimited number of different modified nucleic acid molecules or mmRNA. In certain embodiments, the device is capable of being transported by one or a small number of individuals. In other embodiments, the device is scaled to fit on a benchtop or desk. In other embodiments, the device is scaled to fit into a suitcase, backpack or similarly sized object.
[00653] In another embodiment, the device may be a point of care or handheld device. In further embodiments, the device is scaled to fit into a vehicle, such as a car, truck or ambulance, or a military vehicle such as a tank or personnel carrier. The information necessary to generate a modified mR A encoding polypeptide of interest is present within a computer readable medium present in the device.
[00654] In one embodiment, a device may be used to assess levels of a protein which has been administered in the form of a modified nucleic acid or mmRNA. The device may comprise a blood, urine or other biofluidic test.
[00655] In some embodiments, the device is capable of communication (e.g., wireless
communication) with a database of nucleic acid and polypeptide sequences. The device contains at least one sample block for insertion of one or more sample vessels. Such sample vessels are capable of accepting in liquid or other form any number of materials such as template DNA, nucleotides, enzymes, buffers, and other reagents. The sample vessels are also capable of being heated and cooled by contact with the sample block. The sample block is generally in communication with a device base with one or more electronic control units for the at least one sample block. The sample block preferably contains a heating module, such heating molecule capable of heating and/or cooling the sample vessels and contents thereof to temperatures between about -20C and above +100C. The device base is in communication with a voltage supply such as a battery or external voltage supply. The device also contains means for storing and distributing the materials for RNA synthesis.
[00656] Optionally, the sample block contains a module for separating the synthesized nucleic acids. Alternatively, the device contains a separation module operably linked to the sample block. Preferably the device contains a means for analysis of the synthesized nucleic acid. Such analysis includes sequence identity (demonstrated such as by hybridization), absence of non-desired sequences, measurement of integrity of synthesized mRNA (such has by microfluidic viscometry combined with spectrophotometry), and concentration and/or potency of modified RNA (such as by spectrophotometry). [00657] In certain embodiments, the device is combined with a means for detection of pathogens present in a biological material obtained from a subject, e.g., the IBIS PLEX-ID system (Abbott, Abbott Park, IL) for microbial identification.
[00658] Suitable devices for use in delivering intradermal pharmaceutical compositions described herein include short needle devices such as those described in U.S. Patents 4,886,499; 5,190,521; 5,328,483; 5,527,288; 4,270,537; 5,015,235; 5,141,496; and 5,417,662; each of which is herein incorporated by reference in their entirety. Intradermal compositions may be administered by devices which limit the effective penetration length of a needle into the skin, such as those described in PCT publication WO 99/34850 (herein incorporated by reference in its entirety) and functional equivalents thereof. Jet injection devices which deliver liquid compositions to the dermis via a liquid jet injector and/or via a needle which pierces the stratum corneum and produces a jet which reaches the dermis are suitable. Jet injection devices are described, for example, in U.S. Patents 5,480,381; 5,599,302; 5,334,144; 5,993,412; 5,649,912; 5,569,189; 5,704,911; 5,383,851;
5,893,397; 5,466,220; 5,339,163; 5,312,335; 5,503,627; 5,064,413; 5,520,639; 4,596,556;
4,790,824; 4,941,880; 4,940,460; and PCT publications WO 97/37705 and WO 97/13537; each of which are hrein incorporated by reference in their entirety. Ballistic powder/particle delivery devices which use compressed gas to accelerate vaccine in powder form through the outer layers of the skin to the dermis are suitable. Alternatively or additionally, conventional syringes may be used in the classical mantoux method of intradermal administration.
[00659] In some embodiments, the device may be a pump or comprise a catheter for administration of compounds or compositions of the invention across the blood brain barrier. Such devices include but are not limited to a pressurized olfactory delivery device, iontophoresis devices, multi-layered microfluidic devices, and the like. Such devices may be portable or stationary. They may be implantable or externally tethered to the body or combinations thereof.
[00660] Devices for administration may be employed to deliver the modified nucleic acid molecules or mmRNA of the present invention according to single, multi- or split-dosing regimens taught herein. Such devices are described below.
[00661] Method and devices known in the art for multi-administration to cells, organs and tissues are contemplated for use in conjunction with the methods and compositions disclosed herein as embodiments of the present invention. These include, for example, those methods and devices having multiple needles, hybrid devices employing for example lumens or catheters as well as devices utilizing heat, electric current or radiation driven mechanisms.
[00662] According to the present invention, these multi-administration devices may be utilized to deliver the single, multi- or split doses contemplated herein.
[00663] A method for delivering therapeutic agents to a solid tissue has been described by Bahrami et al. and is taught for example in US Patent Publication 20110230839, the contents of which are incorporated herein by reference in their entirety. According to Bahrami, an array of needles is incorporated into a device which delivers a substantially equal amount of fluid at any location in said solid tissue along each needle's length.
[00664] A device for delivery of biological material across the biological tissue has been described by Kodgule et al. and is taught for example in US Patent Publication 20110172610, the contents of which are incorporated herein by reference in their entirety. According to Kodgule, multiple hollow micro-needles made of one or more metals and having outer diameters from about 200 microns to about 350 microns and lengths of at least 100 microns are incorporated into the device which delivers peptides, proteins, carbohydrates, nucleic acid molecules, lipids and other pharmaceutically active ingredients or combinations thereof.
[00665] A delivery probe for delivering a therapeutic agent to a tissue has been described by Gunday et al. and is taught for example in US Patent Publication 20110270184, the contents of each of which are incorporated herein by reference in their entirety. According to Gunday, multiple needles are incorporated into the device which moves the attached capsules between an activated position and an inactivated position to force the agent out of the capsules through the needles.
[00666] A multiple-injection medical apparatus has been described by Assaf and is taught for example in US Patent Publication 20110218497, the contents of which are incorporated herein by reference in their entirety. According to Assaf, multiple needles are incorporated into the device which has a chamber connected to one or more of said needles and a means for continuously refilling the chamber with the medical fluid after each injection.
[00667] In one embodiment, the modified nucleic acid molecule or mmRNA is administered subcutaneously or intramuscularly via at least 3 needles to three different, optionally adjacent, sites simultaneously, or within a 60 minutes period (e.g., administration to 4 ,5, 6, 7, 8, 9, or 10 sites simultaneously or within a 60 minute period). The split doses can be administered simultaneously to adjacent tissue using the devices described in U.S. Patent Publication Nos. 20110230839 and 20110218497, each of which is incorporated herein by reference in their entirety.
[00668] An at least partially implantable system for injecting a substance into a patient's body, in particular a penis erection stimulation system has been described by Forsell and is taught for example in US Patent Publication 20110196198, the contents of which are incorporated herein by reference in their entirety. According to Forsell, multiple needles are incorporated into the device which is implanted along with one or more housings adjacent the patient's left and right corpora cavernosa. A reservoir and a pump are also implanted to supply drugs through the needles.
[00669] A method for the transdermal delivery of a therapeutic effective amount of iron has been described by Berenson and is taught for example in US Patent Publication 20100130910, the contents of which are incorporated herein by reference in their entirety. According to Berenson, multiple needles may be used to create multiple micro channels in stratum corneum to enhance transdermal delivery of the ionic iron on an iontophoretic patch.
[00670] A method for delivery of biological material across the biological tissue has been described by Kodgule et al and is taught for example in US Patent Publication 20110196308, the contents of which are incorporated herein by reference in their entirety. According to Kodgule, multiple biodegradable microneedles containing a therapeutic active ingredient are incorporated in a device which delivers proteins, carbohydrates, nucleic acid molecules, lipids and other pharmaceutically active ingredients or combinations thereof.
[00671] A transdermal patch comprising a botulinum toxin composition has been described by Donovan and is taught for example in US Patent Publication 20080220020, the contents of which are incorporated herein by reference in their entirety. According to Donovan, multiple needles are incorporated into the patch which delivers botulinum toxin under stratum corneum through said needles which project through the stratum corneum of the skin without rupturing a blood vessel.
[00672] A small, disposable drug reservoir, or patch pump, which can hold approximately 0.2 to 15 mL of liquid formulations can be placed on the skin and deliver the formulation continuously subcutaneously using a small bore needed (e.g., 26 to 34 gauge). As non-limiting examples, the patch pump may be 50 mm by 76 mm by 20 mm spring loaded having a 30 to 34 gauge needle (BD™ Microinfuser, Franklin Lakes NJ), 41 mm by 62 mm by 17 mm with a 2 mL reservoir used for drug delivery such as insulin (OMNIPOD®, Insulet Corporation Bedford, MA), or 43-60 mm diameter, 10 mm thick with a 0.5 to 10 mL reservoir (PATCHPUMP®, SteadyMed Therapeutics, San Francisco, CA). Further, the patch pump may be battery powered and/or rechargeable.
[00673] A cryoprobe for administration of an active agent to a location of cryogenic treatment has been described by Toubia and is taught for example in US Patent Publication 20080140061, the contents of which are incorporated herein by reference in their entirety. According to Toubia, multiple needles are incorporated into the probe which receives the active agent into a chamber and administers the agent to the tissue.
[00674] A method for treating or preventing inflammation or promoting healthy joints has been described by Stock et al and is taught for example in US Patent Publication 20090155186, the contents of which are incorporated herein by reference in their entirety. According to Stock, multiple needles are incorporated in a device which administers compositions containing signal transduction modulator compounds.
[00675] A multi-site injection system has been described by Kimmell et al. and is taught for example in US Patent Publication 20100256594, the contents of which are incorporated herein by reference in their entirety. According to Kimmell, multiple needles are incorporated into a device which delivers a medication into a stratum corneum through the needles.
[00676] A method for delivering interferons to the intradermal compartment has been described by Dekker et al. and is taught for example in US Patent Publication 20050181033, the contents of which are incorporated herein by reference in their entirety. According to Dekker, multiple needles having an outlet with an exposed height between 0 and 1 mm are incorporated into a device which improves pharmacokinetics and bioavailability by delivering the substance at a depth between 0.3 mm and 2 mm.
[00677] A method for delivering genes, enzymes and biological agents to tissue cells has described by Desai and is taught for example in US Patent Publication 20030073908, the contents of which are incorporated herein by reference in their entirety. According to Desai, multiple needles are incorporated into a device which is inserted into a body and delivers a medication fluid through said needles.
[00678] A method for treating cardiac arrhythmias with fibroblast cells has been described by Lee et al and is taught for example in US Patent Publication 20040005295, the contents of which are incorporated herein by reference in their entirety. According to Lee, multiple needles are
incorporated into the device which delivers fibroblast cells into the local region of the tissue. [00679] A method using a magnetically controlled pump for treating a brain tumor has been described by Shachar et al. and is taught for example in US Patent 7,799,012 (method) and
7,799,016 (device), the contents of which are incorporated herein by reference in their entirety. According Shachar, multiple needles were incorporated into the pump which pushes a medicating agent through the needles at a controlled rate.
[00680] Methods of treating functional disorders of the bladder in mammalian females have been described by Versi et al. and are taught for example in US Patent 8,029,496, the contents of which are incorporated herein by reference in their entirety. According to Versi, an array of micro-needles is incorporated into a device which delivers a therapeutic agent through the needles directly into the trigone of the bladder.
[00681] A micro-needle transdermal transport device has been described by Angel et al and is taught for example in US Patent 7,364,568, the contents of which are incorporated herein by reference in their entirety. According to Angel, multiple needles are incorporated into the device which transports a substance into a body surface through the needles which are inserted into the surface from different directions. The micro-needle transdermal transport device may be a solid micro-needle system or a hollow micro-needle system. As a non-limiting example, the solid microneedle system may have up to a 0.5 mg capacity, with 300-1500 solid micro-needles per cm2 about 150-700 μιη tall coated with a drug. The micro-needles penetrate the stratum corneum and remain in the skin for short duration (e.g., 20 seconds to 15 minutes). In another example, the hollow microneedle system has up to a 3 mL capacity to deliver liquid formulations using 15-20 microneedles per cm2 being approximately 950 μιη tall. The micro-needles penetrate the skin to allow the liquid formulations to flow from the device into the skin. The hollow micro-needle system may be worn from 1 to 30 minutes depending on the formulation volume and viscocity.
[00682] A device for subcutaneous infusion has been described by Dalton et al and is taught for example in US Patent 7,150,726, the contents of which are incorporated herein by reference in their entirety. According to Dalton, multiple needles are incorporated into the device which delivers fluid through the needles into a subcutaneous tissue.
[00683] A device and a method for intradermal delivery of vaccines and gene therapeutic agents through microcannula have been described by Mikszta et al. and are taught for example in US Patent 7,473,247, the contents of which are incorporated herein by reference in their entirety. According to Mitszta, at least one hollow micro-needle is incorporated into the device which delivers the vaccines to the subject's skin to a depth of between 0.025 mm and 2 mm.
[00684] A method of delivering insulin has been described by Pettis et al and is taught for example in US Patent 7,722,595, the contents of which are incorporated herein by reference in their entirety. According to Pettis, two needles are incorporated into a device wherein both needles insert essentially simultaneously into the skin with the first at a depth of less than 2.5 mm to deliver insulin to intradermal compartment and the second at a depth of greater than 2.5 mm and less than 5.0 mm to deliver insulin to subcutaneous compartment.
[00685] Cutaneous injection delivery under suction has been described by Kochamba et al. and is taught for example in US Patent 6,896,666, the contents of which are incorporated herein by reference in their entirety. According to Kochamba, multiple needles in relative adjacency with each other are incorporated into a device which injects a fluid below the cutaneous layer.
[00686] A device for withdrawing or delivering a substance through the skin has been described by Down et al and is taught for example in US Patent 6,607,513, the contents of which are incorporated herein by reference in their entirety. According to Down, multiple skin penetrating members which are incorporated into the device have lengths of about 100 microns to about 2000 microns and are about 30 to 50 gauge.
[00687] A device for delivering a substance to the skin has been described by Palmer et al and is taught for example in US Patent 6,537,242, the contents of which are incorporated herein by reference in their entirety. According to Palmer, an array of micro-needles is incorporated into the device which uses a stretching assembly to enhance the contact of the needles with the skin and provides a more uniform delivery of the substance.
[00688] A perfusion device for localized drug delivery has been described by Zamoyski and is taught for example in US Patent 6,468,247, the contents of which are incorporated herein by reference in their entirety. According to Zamoyski, multiple hypodermic needles are incorporated into the device which injects the contents of the hypodermics into a tissue as said hypodermics are being retracted.
[00689] A method for enhanced transport of drugs and biological molecules across tissue by improving the interaction between micro-needles and human skin has been described by Prausnitz et al. and is taught for example in US Patent 6,743,211, the contents of which are incorporated herein by reference in their entirety. According to Prausnitz, multiple micro-needles are incorporated into a device which is able to present a more rigid and less deformable surface to which the micro-needles are applied.
[00690] A device for intraorgan administration of medicinal agents has been described by Ting et al and is taught for example in US Patent 6,077,251, the contents of which are incorporated herein by reference in their entirety. According to Ting, multiple needles having side openings for enhanced administration are incorporated into a device which by extending and retracting said needles from and into the needle chamber forces a medicinal agent from a reservoir into said needles and injects said medicinal agent into a target organ.
[00691] A multiple needle holder and a subcutaneous multiple channel infusion port has been described by Brown and is taught for example in US Patent 4,695,273, the contents of which are incorporated herein by reference in their entirety. According to Brown, multiple needles on the needle holder are inserted through the septum of the infusion port and communicate with isolated chambers in said infusion port.
[00692] A dual hypodermic syringe has been described by Horn and is taught for example in US Patent 3,552,394, the contents of which are incorporated herein by reference in their entirety.
According to Horn, two needles incorporated into the device are spaced apart less than 68 mm and may be of different styles and lengths, thus enabling injections to be made to different depths.
[00693] A syringe with multiple needles and multiple fluid compartments has been described by Hershberg and is taught for example in US Patent 3,572,336, the contents of which are incorporated herein by reference in their entirety. According to Hershberg, multiple needles are incorporated into the syringe which has multiple fluid compartments and is capable of simultaneously administering incompatible drugs which are not able to be mixed for one injection.
[00694] A surgical instrument for intradermal injection of fluids has been described by Eliscu et al. and is taught for example in US Patent 2,588,623, the contents of which are incorporated herein by reference in their entirety. According to Eliscu, multiple needles are incorporated into the instrument which injects fluids intradermally with a wider disperse.
[00695] An apparatus for simultaneous delivery of a substance to multiple breast milk ducts has been described by Hung and is taught for example in EP 1818017, the contents of which are incorporated herein by reference in their entirety. According to Hung, multiple lumens are incorporated into the device which inserts though the orifices of the ductal networks and delivers a fluid to the ductal networks. [00696] A catheter for introduction of medications to the tissue of a heart or other organs has been described by Tkebuchava and is taught for example in WO2006138109, the contents of which are incorporated herein by reference in their entirety. According to Tkebuchava, two curved needles are incorporated which enter the organ wall in a flattened trajectory.
[00697] Devices for delivering medical agents have been described by Mckay et al. and are taught for example in WO2006118804, the content of which are incorporated herein by reference in their entirety. According to Mckay, multiple needles with multiple orifices on each needle are
incorporated into the devices to facilitate regional delivery to a tissue, such as the interior disc space of a spinal disc.
[00698] A method for directly delivering an immunomodulatory substance into an intradermal space within a mammalian skin has been described by Pettis and is taught for example in
WO2004020014, the contents of which are incorporated herein by reference in their entirety.
According to Pettis, multiple needles are incorporated into a device which delivers the substance through the needles to a depth between 0.3 mm and 2 mm.
[00699] Methods and devices for administration of substances into at least two compartments in skin for systemic absorption and improved pharmacokinetics have been described by Pettis et al. and are taught for example in WO2003094995, the contents of which are incorporated herein by reference in their entirety. According to Pettis, multiple needles having lengths between about 300 μιη and about 5 mm are incorporated into a device which delivers to intradermal and subcutaneous tissue compartments simultaneously.
[00700] A drug delivery device with needles and a roller has been described by Zimmerman et al. and is taught for example in WO2012006259, the contents of which are incorporated herein by reference in their entirety. According to Zimmerman, multiple hollow needles positioned in a roller are incorporated into the device which delivers the content in a reservoir through the needles as the roller rotates.
[00701] A drug delivery device such as a stent is known in the art and is taught for example in U.S. Pub. Nos. US20060020329, US20040172127 and US20100161032; the contents of which are herein incorporated by reference in their entirety. Formulations of the modified nucleic acid molecules and mmRNA described herein may be delivered using stents. Additionally, stents used herein may be able to deliver multiple modified nucleic acid molecules and/or formulations at the same or varied rates of delivery. Non-limiting examples of manufacturers of stents include CORDIS® (Miami, FL) (CYPHER®), Boston Scientific Corporation (Natick, MA) (TAXUS®), Medtronic (Minneapolis, MN) (ENDEAVOUR®) and Abbott (Abbott Park, IL) (XIENCE V®).
Methods and Devices utilizing catheters and/or lumens
[00702] Methods and devices using catheters and lumens may be employed to administer the mmRNA of the present invention on a single, multi- or split dosing schedule. Such methods and devices are described below.
[00703] A catheter-based delivery of skeletal myoblasts to the myocardium of damaged hearts has been described by Jacoby et al and is taught for example in US Patent Publication 20060263338, the contents of which are incorporated herein by reference in their entirety. According to Jacoby, multiple needles are incorporated into the device at least part of which is inserted into a blood vessel and delivers the cell composition through the needles into the localized region of the subject's heart.
[00704] An apparatus for treating asthma using neurotoxin has been described by Deem et al and is taught for example in US Patent Publication 20060225742, the contents of which are incorporated herein by reference in their entirety. According to Deem, multiple needles are incorporated into the device which delivers neurotoxin through the needles into the bronchial tissue.
[00705] A method for administering multiple-component therapies has been described by Nayak and is taught for example in US Patent 7,699,803, the contents of which are incorporated herein by reference in their entirety. According to Nayak, multiple injection cannulas may be incorporated into a device wherein depth slots may be included for controlling the depth at which the therapeutic substance is delivered within the tissue.
[00706] A surgical device for ablating a channel and delivering at least one therapeutic agent into a desired region of the tissue has been described by Mclntyre et al and is taught for example in US Patent 8,012,096, the contents of which are incorporated herein by reference in their entirety.
According to Mclntyre, multiple needles are incorporated into the device which dispenses a therapeutic agent into a region of tissue surrounding the channel and is particularly well suited for transmyocardial revascularization operations.
[00707] Methods of treating functional disorders of the bladder in mammalian females have been described by Versi et al and are taught for example in US Patent 8,029,496, the contents of which are incorporated herein by reference in their entirety. According to Versi, an array of micro-needles is incorporated into a device which delivers a therapeutic agent through the needles directly into the trigone of the bladder. [00708] A device and a method for delivering fluid into a flexible biological barrier have been described by Yeshurun et al. and are taught for example in US Patent 7,998,119 (device) and 8,007,466 (method), the contents of which are incorporated herein by reference in their entirety. According to Yeshurun, the micro-needles on the device penetrate and extend into the flexible biological barrier and fluid is injected through the bore of the hollow micro-needles.
[00709] A method for epicardially injecting a substance into an area of tissue of a heart having an epicardial surface and disposed within a torso has been described by Bonner et al and is taught for example in US Patent 7,628,780, the contents of which are incorporated herein by reference in their entirety. According to Bonner, the devices have elongate shafts and distal injection heads for driving needles into tissue and injecting medical agents into the tissue through the needles.
[00710] A device for sealing a puncture has been described by Nielsen et al and is taught for example in US Patent 7,972,358, the contents of which are incorporated herein by reference in their entirety. According to Nielsen, multiple needles are incorporated into the device which delivers a closure agent into the tissue surrounding the puncture tract.
[00711] A method for myogenesis and angiogenesis has been described by Chiu et al. and is taught for example in US Patent 6,551,338, the contents of which are incorporated herein by reference in their entirety. According to Chiu, 5 to 15 needles having a maximum diameter of at least 1.25 mm and a length effective to provide a puncture depth of 6 to 20 mm are incorporated into a device which inserts into proximity with a myocardium and supplies an exogeneous angiogenic or myogenic factor to said myocardium through the conduits which are in at least some of said needles.
[00712] A method for the treatment of prostate tissue has been described by Bolmsj et al. and is taught for example in US Patent 6,524,270, the contents of which are incorporated herein by reference in their entirety. According to Bolmsj, a device comprising a catheter which is inserted through the urethra has at least one hollow tip extendible into the surrounding prostate tissue. An astringent and analgesic medicine is administered through said tip into said prostate tissue.
[00713] A method for infusing fluids to an intraosseous site has been described by Findlay et al. and is taught for example in US Patent 6,761,726, the contents of which are incorporated herein by reference in their entirety. According to Findlay, multiple needles are incorporated into a device which is capable of penetrating a hard shell of material covered by a layer of soft material and delivers a fluid at a predetermined distance below said hard shell of material. [00714] A device for injecting medications into a vessel wall has been described by Vigil et al. and is taught for example in US Patent 5,713,863, the contents of which are incorporated herein by reference in their entirety. According to Vigil, multiple injectors are mounted on each of the flexible tubes in the device which introduces a medication fluid through a multi-lumen catheter, into said flexible tubes and out of said injectors for infusion into the vessel wall.
[00715] A catheter for delivering therapeutic and/or diagnostic agents to the tissue surrounding a bodily passageway has been described by Faxon et al. and is taught for example in US Patent 5,464,395, the contents of which are incorporated herein by reference in their entirety. According to Faxon, at least one needle cannula is incorporated into the catheter which delivers the desired agents to the tissue through said needles which project outboard of the catheter.
[00716] Balloon catheters for delivering therapeutic agents have been described by Orr and are taught for example in WO2010024871, the contents of which are incorporated herein by reference in their entirety. According to Orr, multiple needles are incorporated into the devices which deliver the therapeutic agents to different depths within the tissue. In another aspect, drug-eluting balloons may be used to deliver the formulations described herein. The drug-eluting balloons may be used in target lesion applications such as, but are not limited to, in-stent restenosis, treating lesion in tortuous vessels, bifurcation lesions, femoral/popliteal lesions and below the knee lesions.
[00717] A device for deliverying therapeutic agents (e.g., modified nucleic acid molecules or mmRNA) to tissue disposed about a lumin has been described by Perry et al. and is taught for example in U.S. Pat. Pub. US20100125239, the contents of which are herein incorporated by reference in their entirety. According to Perry, the catheter has a balloon which may be coated with a therapeutic agent by methods known in the art and described in Perry. When the balloon expands, the therapeutic agent will contact the surrounding tissue. The device may additionally have a heat source to change the temperature of the coating on the balloon to release the thereapeutic agent to the tissue.
Methods and Devices utilizing electrical current
[00718] Methods and devices utilizing electric current may be employed to deliver the mmRNA of the present invention according to the single, multi- or split dosing regimens taught herein. Such methods and devices are described below.
[00719] An electro collagen induction therapy device has been described by Marquez and is taught for example in US Patent Publication 20090137945, the contents of which are incorporated herein by reference in their entirety. According to Marquez, multiple needles are incorporated into the device which repeatedly pierce the skin and draw in the skin a portion of the substance which is applied to the skin first.
[00720] An electrokinetic system has been described by Etheredge et al. and is taught for example in US Patent Publication 20070185432, the contents of which are incorporated herein by reference in their entirety. According to Etheredge, micro-needles are incorporated into a device which drives by an electrical current the medication through the needles into the targeted treatment site.
[00721] An iontophoresis device has been described by Matsumura et al. and is taught for example in US Patent 7,437,189, the contents of which are incorporated herein by reference in their entirety. According to Matsumura, multiple needles are incorporated into the device which is capable of delivering ionizable drug into a living body at higher speed or with higher efficiency.
[00722] Intradermal delivery of biologically active agents by needle-free injection and
electroporation has been described by Hoffmann et al and is taught for example in US Patent 7,171,264, the contents of which are incorporated herein by reference in their entirety. According to Hoffmann, one or more needle-free injectors are incorporated into an electroporation device and the combination of needle-free injection and electroporation is sufficient to introduce the agent into cells in skin, muscle or mucosa.
[00723] A method for electropermeabilization-mediated intracellular delivery has been described by Lundkvist et al. and is taught for example in US Patent 6,625,486, the contents of which are incorporated herein by reference in their entirety. According to Lundkvist, a pair of needle electrodes is incorporated into a catheter. Said catheter is positioned into a body lumen followed by extending said needle electrodes to penetrate into the tissue surrounding said lumen. Then the device introduces an agent through at least one of said needle electrodes and applies electric field by said pair of needle electrodes to allow said agent pass through the cell membranes into the cells at the treatment site.
[00724] A delivery system for transdermal immunization has been described by Levin et al. and is taught for example in WO2006003659, the contents of which are incorporated herein by reference in their entirety. According to Levin, multiple electrodes are incorporated into the device which applies electrical energy between the electrodes to generate micro channels in the skin to facilitate transdermal delivery. [00725] A method for delivering RF energy into skin has been described by Schomacker and is taught for example in WO2011163264, the contents of which are incorporated herein by reference in their entirety. According to Schomacker, multiple needles are incorporated into a device which applies vacuum to draw skin into contact with a plate so that needles insert into skin through the holes on the plate and deliver RF energy.
Definitions
[00726] At various places in the present specification, substituents of compounds of the present disclosure are disclosed in groups or in ranges. It is specifically intended that the present disclosure include each and every individual subcombination of the members of such groups and ranges. For example, the term "Ci_6 alkyl" is specifically intended to individually disclose methyl, ethyl, C3 alkyl, C4 alkyl, C5 alkyl, and C6 alkyl.
[00727] About: As used herein, the term "about" means +/- 10% of the recited value.
[00728] Administered in combination: As used herein, the term "administered in combination" or "combined administration" means that two or more agents {e.g., a modified nucleic acid or mmRNA encoding an anti-microbial polypeptide {e.g., an anti-bacterial polypeptide), e.g., an anti-microbial polypeptide described herein and an anti-microbial agent {e.g., an anti-microbial polypeptide or a small molecule anti-microbial compound described herein)) are administered to a subject at the same time or within an interval such that there may be an overlap of an effect of each agent on the patient. In some embodiments, they are administered within about 60, 30, 15, 10, 5, or 1 minute of one another. In some embodiments, the administrations of the agents are spaced sufficiently close together such that a combinatorial {e.g. , a synergistic) effect is achieved.
[00729] Animal: As used herein, the term "animal" refers to any member of the animal kingdom. In some embodiments, "animal" refers to humans at any stage of development. In some
embodiments, "animal" refers to non-human animals at any stage of development. In certain embodiments, the non-human animal is a mammal {e.g., a rodent, a mouse, a rat, a rabbit, a monkey, a dog, a cat, a sheep, cattle, a primate, or a pig). In some embodiments, animals include, but are not limited to, mammals, birds, reptiles, amphibians, fish, and worms. In some embodiments, the animal is a transgenic animal, genetically-engineered animal, or a clone.
[00730] Antigens of interest or desired antigens: As used herein, the terms "antigens of interest" or "desired antigens" include those proteins and other biomolecules provided herein that are
immunospecifically bound by the antibodies and fragments, mutants, variants, and alterations thereof described herein. Examples of antigens of interest include, but are not limited to, insulin, insulinlike growth factor, hGH, tPA, cytokines, such as interleukins (IL), e.g., IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-11, IL-12, IL-13, IL-14, IL-15, IL-16, IL-17, IL-18, interferon (IFN) alpha, IFN beta, IFN gamma, IFN omega or IFN tau, tumor necrosis factor (TNF), such as TNF alpha and TNF beta, TNF gamma, TRAIL; G-CSF, GM-CSF, M-CSF, MCP-1 and VEGF.
[00731] Approximately: As used herein, the term "approximately" or "about," as applied to one or more values of interest, refers to a value that is similar to a stated reference value. In certain embodiments, the term "approximately" or "about" refers to a range of values that fall within 25%, 20%, 19%, 18%, 17%, 16%, 15%, 14%, 13%, 12%, 11%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, or less in either direction (greater than or less than) of the stated reference value unless otherwise stated or otherwise evident from the context (except where such number would exceed 100%) of a possible value).
[00732] Associated with: As used herein, the terms "associated with," "conjugated," "linked," "attached," and "tethered," when used with respect to two or more moieties, means that the moieties are physically associated or connected with one another, either directly or via one or more additional moieties that serves as a linking agent, to form a structure that is sufficiently stable so that the moieties remain physically associated under the conditions in which the structure is used, e.g. , physiological conditions. An "association" need not be strictly through direct covalent chemical bonding. It may also suggest ionic or hydrogen bonding or a hybridization based connectivity sufficiently stable such that the "associated" entities remain physically associated.
[00733] Bifunctional: As used herein, the term "bifunctional" refers to any substance, molecule or moiety which is capable of or maintains at least two functions. The functions may effect the same outcome or a different outcome. The structure that produces the function may be the same or different. For example, bifunctional modified RNA of the present invention may encode a cytotoxic peptide (a first function) while those nucleosides which comprise the encoding RNA are, in and of themselves, cytotoxic (second function). In this example, delivery of the bifunctional modified RNA to a cancer cell would produce not only a peptide or protein molecule which may ameliorate or treat the cancer but would also deliver a cytotoxic payload of nucleosides to the cell should degradation, instead of translation of the modified RNA, occur. [00734] Biocompatible: As used herein, the term "biocompatible" means compatible with living cells, tissues, organs or systems posing little to no risk of injury, toxicity or rejection by the immune system.
[00735] Biodegradable: As used herein, the term "biodegradable" means capable of being broken down into innocuous products by the action of living things.
[00736] Biologically active: As used herein, the phrase "biologically active" refers to a
characteristic of any substance that has activity in a biological system and/or organism. For instance, a substance that, when administered to an organism, has a biological affect on that organism, is considered to be biologically active. In particular embodiments, the modified nucleic acid or mmR A of the present invention may be considered biologically active if even a portion of the modified nucleic acid or mmRNA is biologically active or mimics an activity considered biologically relevant.
[00737] Chemical terms: The following provides the definition of various chemical terms from "acyl" to "thiol."
[00738] The term "acyl," as used herein, represents a hydrogen or an alkyl group (e.g., a haloalkyl group), as defined herein, that is attached to the parent molecular group through a carbonyl group, as defined herein, and is exemplified by formyl (i.e., a carboxyaldehyde group), acetyl, propionyl, butanoyl and the like. Exemplary unsubstituted acyl groups include from 1 to 7, from 1 to 11 , or from 1 to 21 carbons. In some embodiments, the alkyl group is further substituted with 1, 2, 3, or 4 substituents as described herein.
[00739] The term "acylamino," as used herein, represents an acyl group, as defined herein, attached to the parent molecular group though an amino group, as defined herein (i.e., -N(RN1)-C(0)-R, where R is H or an optionally substituted Ci_6, C1-10, or Ci_2o alkyl group and RN1 is as defined herein). Exemplary unsubstituted acylamino groups include from 1 to 41 carbons (e.g., from 1 to 7, from 1 to 13, from 1 to 21, from 2 to 7, from 2 to 13, from 2 to 21, or from 2 to 41 carbons). In some embodiments, the alkyl group is further substituted with 1, 2, 3, or 4 substituents as described herein, and/or the amino group is -NH2 or -NHRN1, wherein RN1 is, independently, OH, N02, NH2, NRN22, S02ORN2, S02RN2, SORN2, alkyl, or aryl, and each RN2 can be H, alkyl, or aryl.
[00740] The term "acyloxy," as used herein, represents an acyl group, as defined herein, attached to the parent molecular group though an oxygen atom (i.e., -0-C(0)-R, where R is H or an optionally substituted C1-6, C1-10, or Ci_20 alkyl group). Exemplary unsubstituted acyloxy groups include from 1 to 21 carbons (e.g., from 1 to 7 or from 1 to 1 1 carbons). In some embodiments, the alkyl group is further substituted with 1 , 2, 3, or 4 substituents as described herein, and/or the amino group is -NH2 or -NHRN1, wherein RN1 is, independently, OH, N02, NH2, NRN2 2, S02ORN2, S02RN2, SORN2, alkyl, or aryl, and each RN2 can be H, alkyl, or aryl.
[00741] The term "alkaryl," as used herein, represents an aryl group, as defined herein, attached to the parent molecular group through an alkylene group, as defined herein. Exemplary unsubstituted alkaryl groups are from 7 to 30 carbons (e.g., from 7 to 16 or from 7 to 20 carbons, such as Ci_6 alk- C6-io aryl, Ci_io alk-C6-io aryl, or Ci_2o alk-C6-io aryl). In some embodiments, the alkylene and the aryl each can be further substituted with 1 , 2, 3, or 4 substituent groups as defined herein for the respective groups. Other groups preceded by the prefix "alk-" are defined in the same manner, where "alk" refers to a Ci_6 alkylene, unless otherwise noted, and the attached chemical structure is as defined herein.
[00742] The term "alkcycloalkyl" represents a cycloalkyl group, as defined herein, attached to the parent molecular group through an alkylene group, as defined herein (e.g., an alkylene group of from 1 to 4, from 1 to 6, from 1 to 10, or form 1 to 20 carbons). In some embodiments, the alkylene and the cycloalkyl each can be further substituted with 1 , 2, 3, or 4 substituent groups as defined herein for the respective group.
[00743] The term "alkenyl," as used herein, represents monovalent straight or branched chain groups of, unless otherwise specified, from 2 to 20 carbons (e.g., from 2 to 6 or from 2 to 10 carbons) containing one or more carbon-carbon double bonds and is exemplified by ethenyl, 1- propenyl, 2-propenyl, 2-methyl-l-propenyl, 1-butenyl, 2-butenyl, and the like. Alkenyls include both cis and trans isomers. Alkenyl groups may be optionally substituted with 1 , 2, 3, or 4 substituent groups that are selected, independently, from amino, aryl, cycloalkyl, or heterocyclyl (e.g., heteroaryl), as defined herein, or any of the exemplary alkyl substituent groups described herein.
[00744] The term "alkenyloxy" represents a chemical substituent of formula -OR, where R is a C2_ 20 alkenyl group (e.g., C2_6 or C2_i0 alkenyl), unless otherwise specified. Exemplary alkenyloxy groups include ethenyloxy, propenyloxy, and the like. In some embodiments, the alkenyl group can be further substituted with 1 , 2, 3, or 4 substituent groups as defined herein (e.g., a hydroxy group).
[00745] The term "alkheteroaryl" refers to a heteroaryl group, as defined herein, attached to the parent molecular group through an alkylene group, as defined herein. Exemplary unsubstituted alkheteroaryl groups are from 2 to 32 carbons (e.g., from 2 to 22, from 2 to 18, from 2 to 17, from 2 to 16, from 3 to 15, from 2 to 14, from 2 to 13, or from 2 to 12 carbons, such as Ci_6 alk-Ci_i2 heteroaryl, C1-10 alk-Ci_i2 heteroaryl, or Ci_2o alk-Ci_i2 heteroaryl). In some embodiments, the alkylene and the heteroaryl each can be further substituted with 1 , 2, 3, or 4 substituent groups as defined herein for the respective group. Alkheteroaryl groups are a subset of alkheterocyclyl groups.
[00746] The term "alkheterocyclyl" represents a heterocyclyl group, as defined herein, attached to the parent molecular group through an alkylene group, as defined herein. Exemplary unsubstituted alkheterocyclyl groups are from 2 to 32 carbons (e.g., from 2 to 22, from 2 to 18, from 2 to 17, from 2 to 16, from 3 to 15, from 2 to 14, from 2 to 13, or from 2 to 12 carbons, such as Ci_6 alk-Ci_i2 heterocyclyl, C1-10 alk-Ci_i2 heterocyclyl, or Ci_20 alk-Ci_i2 heterocyclyl). In some embodiments, the alkylene and the heterocyclyl each can be further substituted with 1 , 2, 3, or 4 substituent groups as defined herein for the respective group.
[00747] The term "alkoxy" represents a chemical substituent of formula -OR, where R is a Ci_2o alkyl group (e.g., Ci_6 or C1-10 alkyl), unless otherwise specified. Exemplary alkoxy groups include methoxy, ethoxy, propoxy (e.g., n-propoxy and isopropoxy), t-butoxy, and the like. In some embodiments, the alkyl group can be further substituted with 1 , 2, 3, or 4 substituent groups as defined herein (e.g., hydroxy or alkoxy).
[00748] The term "alkoxyalkoxy" represents an alkoxy group that is substituted with an alkoxy group. Exemplary unsubstituted alkoxyalkoxy groups include between 2 to 40 carbons (e.g., from 2 to 12 or from 2 to 20 carbons, such as Ci_6 alkoxy-Ci_6 alkoxy, C1-10 alkoxy-Ci_io alkoxy, or Ci_2o alkoxy-Ci_2o alkoxy). In some embodiments, the each alkoxy group can be further substituted with 1 , 2, 3, or 4 substituent groups as defined herein.
[00749] The term "alkoxyalkyl" represents an alkyl group that is substituted with an alkoxy group. Exemplary unsubstituted alkoxyalkyl groups include between 2 to 40 carbons (e.g., from 2 to 12 or from 2 to 20 carbons, such as Ci_6 alkoxy-Ci_6 alkyl, C1-10 alkoxy-Ci_io alkyl, or Ci_2o alkoxy-Ci_2o alkyl). In some embodiments, the alkyl and the alkoxy each can be further substituted with 1 , 2, 3, or 4 substituent groups as defined herein for the respective group.
[00750] The term "alkoxycarbonyl," as used herein, represents an alkoxy, as defined herein, attached to the parent molecular group through a carbonyl atom (e.g., -C(0)-OR, where R is H or an optionally substituted Ci_6, C1-10, or Ci_2o alkyl group). Exemplary unsubstituted alkoxycarbonyl include from 1 to 21 carbons (e.g., from 1 to 11 or from 1 to 7 carbons). In some embodiments, the alkoxy group is further substituted with 1, 2, 3, or 4 substituents as described herein.
[00751] The term "alkoxy carbonylalkoxy," as used herein, represents an alkoxy group, as defined herein, that is substituted with an alkoxycarbonyl group, as defined herein (e.g., -0-alkyl-C(0)-OR, where R is an optionally substituted Ci_6, C1-10, or Ci_2o alkyl group). Exemplary unsubstituted alkoxy carbonylalkoxy include from 3 to 41 carbons (e.g., from 3 to 10, from 3 to 13, from 3 to 17, from 3 to 21, or from 3 to 31 carbons, such as Ci_6 alkoxycarbonyl-Ci_6 alkoxy, C1-10
alkoxycarbonyl-Ci_io alkoxy, or Ci_2o alkoxycarbonyl-Ci_2o alkoxy). In some embodiments, each alkoxy group is further independently substituted with 1, 2, 3, or 4 substituents, as described herein (e.g., a hydroxy group).
[00752] The term "alkoxy carbonylalkyl," as used herein, represents an alkyl group, as defined herein, that is substituted with an alkoxycarbonyl group, as defined herein (e.g., -alkyl-C(0)-OR, where R is an optionally substituted Ci_2o, C1-10, or Ci_6 alkyl group). Exemplary unsubstituted alkoxycarbonylalkyl include from 3 to 41 carbons (e.g., from 3 to 10, from 3 to 13, from 3 to 17, from 3 to 21, or from 3 to 31 carbons, such as Ci_6 alkoxycarbonyl-Ci_6 alkyl, C1-10 alkoxycarbonyl- Ci_io alkyl, or Ci_2o alkoxycarbonyl-Ci_2o alkyl). In some embodiments, each alkyl and alkoxy group is further independently substituted with 1, 2, 3, or 4 substituents as described herein (e.g., a hydroxy group).
[00753] The term "alkyl," as used herein, is inclusive of both straight chain and branched chain saturated groups from 1 to 20 carbons (e.g., from 1 to 10 or from 1 to 6), unless otherwise specified. Alkyl groups are exemplified by methyl, ethyl, n- and iso-propyl, n-, sec-, iso- and tert-butyl, neopentyl, and the like, and may be optionally substituted with one, two, three, or, in the case of alkyl groups of two carbons or more, four substituents independently selected from the group consisting of: (1) Ci_6 alkoxy; (2) Ci_6 alkylsulfinyl; (3) amino, as defined herein (e.g., unsubstituted amino (i.e., -NH2) or a substituted amino (i.e., -N(RN1)2, where RN1 is as defined for amino); (4) C6-io aryl-Ci_6 alkoxy; (5) azido; (6) halo; (7) (C2_9 heterocyclyl)oxy; (8) hydroxy; (9) nitro; (10) oxo (e.g., carboxyaldehyde or acyl); (11) Ci_7 spirocyclyl; (12) thioalkoxy; (13) thiol; (14) -C02RA , where RA is selected from the group consisting of (a) Ci_2o alkyl (e.g., Ci_6 alkyl), (b) C2_2o alkenyl (e.g., C2_6 alkenyl), (c) C6-io aryl, (d) hydrogen, (e) Ci_6 alk-C6-io aryl, (f) amino-Ci_2o alkyl, (g) polyethylene glycol of -(CH2)s2(OCH2CH2)si(CH2)S30R', wherein si is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R' is H or Ci_2o alkyl, and (h) amino- polyethylene glycol of -NRN1(CH2)S2(CH2CH20)si(CH2)s3NRN1, wherein si is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and each RN1 is, independently, hydrogen or optionally substituted Ci_6 alkyl; (15) -C(0)NRB Rc , where each of RB and R is, independently, selected from the group consisting of (a) hydrogen, (b) Ci_6 alkyl, (c) C6_io aryl, and (d) Ci_6 alk-C6-io aryl; (16) -S02RD , where RD is selected from the group consisting of (a) Ci_6 alkyl, (b) C6-io aryl, (c) Ci_6 alk-C6-io aryl, and (d) hydroxy; (17) -S02NRE RF , where each of RE and RF is, independently, selected from the group consisting of (a) hydrogen, (b) Ci_6 alkyl, (c) C6-io aryl and (d) Ci_6 alk-C6_io aryl; (18) -C(0)R , where R is selected from the group consisting of (a) Ci_2o alkyl (e.g., Ci_6 alkyl), (b) C2-20 alkenyl (e.g., C2_6 alkenyl), (c) C6-10 aryl, (d) hydrogen, (e) Ci_6 alk-C6-io aryl, (f) amino-Ci_2o alkyl, (g) polyethylene glycol of -
(CH2)s2(OCH2CH2)si(CH2)s3OR', wherein si is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R' is H or Ci_2o alkyl, and (h) amino-polyethylene glycol of - NRN1(CH2)s2(CH2CH20)si(CH2)s3NRN1, wherein si is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and each RN1 is, independently, hydrogen or optionally substituted Ci_6 alkyl; (19) -NRH C(0)R! , wherein RH is selected from the group consisting of (al) hydrogen and (bl) Ci_6 alkyl, and R1 is selected from the group consisting of (a2) Ci_2o alkyl (e.g., Ci_6 alkyl), (b2) C2-20 alkenyl (e.g., C2-e alkenyl), (c2) C6-10 aryl, (d2) hydrogen, (e2) Ci_6 alk-C6_io aryl, (f2) amino-Ci_2o alkyl, (g2) polyethylene glycol of -(CH2)S2(OCH2CH2)si(CH2)s3OR', wherein si is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R' is H or Ci_2o alkyl, and (h2) amino-polyethylene glycol of -NRN1(CH2)s2(CH2CH20)si(CH2)s3NRN1, wherein si is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and each RN1 is, independently, hydrogen or optionally substituted Ci_6 alkyl; (20) - NRJ C(0)ORK , wherein RJ is selected from the group consisting of (al) hydrogen and (bl) Ci_6 alkyl, and RK is selected from the group consisting of (a2) Ci_2o alkyl (e.g., Ci_6 alkyl), (b2) C2-20 alkenyl (e.g., C2-6 alkenyl), (c2) C6_io aryl, (d2) hydrogen, (e2) Ci_6 alk-C6_io aryl, (f2) amino-Ci_2o alkyl, (g2) polyethylene glycol of -(CH2)s2(OCH2CH2)si(CH2)s30R', wherein si is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R' is H or Ci_2o alkyl, and (h2) amino-polyethylene glycol of -NRN1(CH2)s2(CH2CH20)si(CH2)s3NRN1, wherein si is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and each RN1 is, independently, hydrogen or optionally substituted Ci_6 alkyl; and (21) amidine. In some embodiments, each of these groups can be further substituted as described herein. For example, the alkylene group of a Ci-alkaryl can be further substituted with an oxo group to afford the respective aryloyl substituent.
[00754] The term "alkylene" and the prefix "alk-," as used herein, represent a saturated divalent hydrocarbon group derived from a straight or branched chain saturated hydrocarbon by the removal of two hydrogen atoms, and is exemplified by methylene, ethylene, isopropylene, and the like. The term "Cx_y alkylene" and the prefix "Cx_y alk-" represent alkylene groups having between x and y carbons. Exemplary values for x are 1, 2, 3, 4, 5, and 6, and exemplary values for y are 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 14, 16, 18, or 20 (e.g., Ci_6, C1-10, C2-20, C2-6, C2-10, or C2-20 alkylene). In some embodiments, the alkylene can be further substituted with 1, 2, 3, or 4 substituent groups as defined herein for an alkyl group.
[00755] The term "alkylsulfmyl," as used herein, represents an alkyl group attached to the parent molecular group through an -S(O)- group. Exemplary unsubstituted alkylsulfmyl groups are from 1 to 6, from 1 to 10, or from 1 to 20 carbons. In some embodiments, the alkyl group can be further substituted with 1, 2, 3, or 4 substituent groups as defined herein.
[00756] The term "alkylsulfinylalkyl," as used herein, represents an alkyl group, as defined herein, substituted by an alkylsulfmyl group. Exemplary unsubstituted alkylsulfinylalkyl groups are from 2 to 12, from 2 to 20, or from 2 to 40 carbons. In some embodiments, each alkyl group can be further substituted with 1, 2, 3, or 4 substituent groups as defined herein.
[00757] The term "alkynyl," as used herein, represents monovalent straight or branched chain groups from 2 to 20 carbon atoms (e.g., from 2 to 4, from 2 to 6, or from 2 to 10 carbons) containing a carbon-carbon triple bond and is exemplified by ethynyl, 1-propynyl, and the like. Alkynyl groups may be optionally substituted with 1, 2, 3, or 4 substituent groups that are selected, independently, from aryl, cycloalkyl, or heterocyclyl (e.g., heteroaryl), as defined herein, or any of the exemplary alkyl substituent groups described herein.
[00758] The term "alkynyloxy" represents a chemical substituent of formula -OR, where R is a C2_ 20 alkynyl group (e.g., C2_6 or C2_io alkynyl), unless otherwise specified. Exemplary alkynyloxy groups include ethynyloxy, propynyloxy, and the like. In some embodiments, the alkynyl group can be further substituted with 1 , 2, 3, or 4 substituent groups as defined herein (e.g., a hydroxy group).
[00759] The term "amidine," as used herein, represents a -C(=NH)NH2 group.
[00760] The term "amino," as used herein, represents -N(RN1)2, wherein each RN1 is,
independently, H, OH, N02, N(RN2)2, S02ORN2, S02RN2, SORN2, an N-protecting group, alkyl, alkenyl, alkynyl, alkoxy, aryl, alkaryl, cycloalkyl, alkcycloalkyl, carboxyalkyl, sulfoalkyl, heterocyclyl (e.g., heteroaryl), or alkheterocyclyl (e.g., alkheteroaryl), wherein each of these recited
RN1
groups can be optionally substituted, as defined herein for each group; or two RN1 combine to form a heterocyclyl or an N-protecting group, and wherein each RN2 is, independently, H, alkyl, or aryl. The amino groups of the invention can be an unsubstituted amino (i.e., -NH2) or a substituted amino (i.e., -N(RN1)2). In a preferred embodiment, amino is -NH2 or -NHRN1, wherein RN1 is, independently, OH, N02, NH2, NRN2 2, S02ORN2, S02RN2, SORN2, alkyl, carboxyalkyl, sulfoalkyl, or aryl, and each RN2 can be H, Ci_2o alkyl (e.g., Ci_6 alkyl), or C6-1o aryl.
[00761] The term "amino acid," as described herein, refers to a molecule having a side chain, an amino group, and an acid group (e.g., a carboxy group of-C02H or a sulfo group of-S03H), wherein the amino acid is attached to the parent molecular group by the side chain, amino group, or acid group (e.g., the side chain). In some embodiments, the amino acid is attached to the parent molecular group by a carbonyl group, where the side chain or amino group is attached to the carbonyl group. Exemplary side chains include an optionally substituted alkyl, aryl, heterocyclyl, alkaryl, alkheterocyclyl, aminoalkyl, carbamoylalkyl, and carboxyalkyl. Exemplary amino acids include alanine, arginine, asparagine, aspartic acid, cysteine, glutamic acid, glutamine, glycine, histidine, hydroxynorvaline, isoleucine, leucine, lysine, methionine, norvaline, ornithine,
phenylalanine, proline, pyrrolysine, selenocysteine, serine, taurine, threonine, tryptophan, tyrosine, and valine. Amino acid groups may be optionally substituted with one, two, three, or, in the case of amino acid groups of two carbons or more, four substituents independently selected from the group consisting of: (1) Ci_6 alkoxy; (2) Ci_6 alkylsulfmyl; (3) amino, as defined herein (e.g., unsubstituted amino (i.e., -NH2) or a substituted amino (i.e., -N(RN1)2, where RN1 is as defined for amino); (4) C6_io aryl-Ci_6 alkoxy; (5) azido; (6) halo; (7) (C2-9 heterocyclyl)oxy; (8) hydroxy; (9) nitro; (10) oxo (e.g., carboxyaldehyde or acyl); (11) Ci_7 spirocyclyl; (12) thioalkoxy; (13) thiol; (14) -C02RA , where RA is selected from the group consisting of (a) Ci_2o alkyl (e.g., Ci_6 alkyl), (b) C2-2o alkenyl (e.g., C2_6 alkenyl), (c) C6-1o aryl, (d) hydrogen, (e) Ci_6 alk-C6-io aryl, (f) amino-Ci_2o alkyl, (g) polyethylene glycol of -(CH2)s2(OCH2CH2)si(CH2)s30R', wherein si is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R' is H or Ci_2o alkyl, and (h) amino- polyethylene glycol of -NRN1(CH2)s2(CH2CH20)si(CH2)s3NRN1, wherein si is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and each RN1 is,
independently, hydrogen or optionally substituted Ci_6 alkyl; (15) -C(0)NRB Rc , where each of RB and R is, independently, selected from the group consisting of (a) hydrogen, (b) Ci_6 alkyl, (c) C6-io aryl, and (d) Ci_6 alk-C6-io aryl; (16) -S02RD , where RD is selected from the group consisting of (a) Ci_6 alkyl, (b) C6_io aryl, (c) Ci_6 alk-C6_io aryl, and (d) hydroxy; (17) -S02NRE RF , where each of RE and RF is, independently, selected from the group consisting of (a) hydrogen, (b) Ci_6 alkyl, (c) C6-io aryl and (d) Ci_6 alk-C6-io aryl; (18) -C(0)R , where R is selected from the group consisting of (a) Ci_20 alkyl (e.g., Ci_6 alkyl), (b) C2_20 alkenyl (e.g., C2_6 alkenyl), (c) C6-10 aryl, (d) hydrogen, (e) Ci_6 alk-C6_io aryl, (f) amino-Ci_20 alkyl, (g) polyethylene glycol of -
(CH2)s2(OCH2CH2)si(CH2)s3OR', wherein si is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R' is H or Ci_2o alkyl, and (h) amino-polyethylene glycol of - NRN1(CH2)s2(CH2CH20)si(CH2)s3NRN1, wherein si is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and each RN1 is, independently, hydrogen or optionally substituted Ci_6 alkyl; (19) -NRH C(0)R! , wherein RH is selected from the group consisting of (al) hydrogen and (bl) Ci_6 alkyl, and R1 is selected from the group consisting of (a2) Ci_2o alkyl (e.g., Ci_6 alkyl), (b2) C2_20 alkenyl (e.g., C2_6 alkenyl), (c2) C6-10 aryl, (d2) hydrogen, (e2) Ci_6 alk-C6_io aryl, (f2) amino-Ci_2o alkyl, (g2) polyethylene glycol of -(CH2)s2(OCH2CH2)si(CH2)s3OR', wherein si is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R' is H or Ci_2o alkyl, and (h2) amino-polyethylene glycol of -NRN1(CH2)s2(CH2CH20)si(CH2)s3NRN1, wherein si is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and each RN1 is, independently, hydrogen or optionally substituted Ci_6 alkyl; (20) - NRJ C(0)ORK , wherein RJ is selected from the group consisting of (al) hydrogen and (bl) Ci_6 alkyl, and RK is selected from the group consisting of (a2) Ci_2o alkyl (e.g., Ci_6 alkyl), (b2) C2-2o alkenyl (e.g., C2-6 alkenyl), (c2) C6_io aryl, (d2) hydrogen, (e2) Ci_6 alk-C6_io aryl, (f2) amino-Ci_2o alkyl, (g2) polyethylene glycol of -(CH2)s2(OCH2CH2)si(CH2)s30R', wherein si is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R' is H or Ci_2o alkyl, and (h2) amino-polyethylene glycol of -NRN1(CH2)S2(CH2CH20)si(CH2)s3NRN1, wherein si is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and each RN1 is, independently, hydrogen or optionally substituted Ci_6 alkyl; and (21) amidine. In some embodiments, each of these groups can be further substituted as described herein.
[00762] The term "aminoalkoxy," as used herein, represents an alkoxy group, as defined herein, substituted by an amino group, as defined herein. The alkyl and amino each can be further substituted with 1, 2, 3, or 4 substituent groups as described herein for the respective group (e.g., C02RA , where RA is selected from the group consisting of (a) Ci_6 alkyl, (b) C6_io aryl, (c) hydrogen, and (d) Ci_6 alk-C6_io aryl, e.g., carboxy).
[00763] The term "aminoalkyl," as used herein, represents an alkyl group, as defined herein, substituted by an amino group, as defined herein. The alkyl and amino each can be further substituted with 1, 2, 3, or 4 substituent groups as described herein for the respective group (e.g., C02RA , where RA is selected from the group consisting of (a) Ci_6 alkyl, (b) C6_io aryl, (c) hydrogen, and (d) Ci_6 alk-C6-io aryl, e.g., carboxy).
[00764] The term "aryl," as used herein, represents a mono-, bicyclic, or multicyclic carbocyclic ring system having one or two aromatic rings and is exemplified by phenyl, naphthyl, 1 ,2- dihydronaphthyl, 1,2,3,4-tetrahydronaphthyl, anthracenyl, phenanthrenyl, fluorenyl, indanyl, indenyl, and the like, and may be optionally substituted with 1, 2, 3, 4, or 5 substituents
independently selected from the group consisting of: (1) Ci_7 acyl (e.g., carboxyaldehyde); (2) Ci_2o alkyl (e.g., Ci_6 alkyl, Ci_6 alkoxy-Ci_6 alkyl, Ci_6 alkylsulfmyl-Ci_6 alkyl, amino-Ci_6 alkyl, azido-Ci_ 6 alkyl, (carboxyaldehyde)-Ci_6 alkyl, halo-Ci_6 alkyl (e.g., perfluoroalkyl), hydroxy-Ci_6 alkyl, nitro- Ci_6 alkyl, or Ci_6 thioalkoxy-Ci_6 alkyl); (3) Ci_2o alkoxy (e.g., Ci_6 alkoxy, such as
perfluoroalkoxy); (4) Ci_6 alkylsulfmyl; (5) C6-10 aryl; (6) amino; (7) Ci_6 alk-C6_io aryl; (8) azido; (9) C3-8 cycloalkyl; (10) Ci_6 alk-C3-8 cycloalkyl; (11) halo; (12) C1-12 heterocyclyl (e.g., C1-12 heteroaryl); (13) (C1-12 heterocyclyl)oxy; (14) hydroxy; (15) nitro; (16) Ci_2o thioalkoxy (e.g., Ci_6 thioalkoxy); (17) -(CH2)qC02RA , where q is an integer from zero to four, and RA is selected from the group consisting of (a) Ci_6 alkyl, (b) C6_io aryl, (c) hydrogen, and (d) Ci_6 alk-C6_io aryl; (18) - (CH2)qCONR R , where q is an integer from zero to four and where R and R are independently selected from the group consisting of (a) hydrogen, (b) Ci_6 alkyl, (c) C6-1o aryl, and (d) Ci_6 alk-C6-io aryl; (19) -(CH2)qS02RD , where q is an integer from zero to four and where RD is selected from the group consisting of (a) alkyl, (b) C6_io aryl, and (c) alk-C6_io aryl; (20) -(CH2)qS02NRE RF , where q is an integer from zero to four and where each of RE and RF is, independently, selected from the group consisting of (a) hydrogen, (b) Ci_6 alkyl, (c) C6-io aryl, and (d) Ci_6 alk-C6-io aryl; (21) thiol; (22) C6-io aryloxy; (23) C3-8 cycloalkoxy; (24) C6-io aryl-Ci_6 alkoxy; (25) Ci_6 alk-Ci_i2 heterocyclyl (e.g., Ci_6 alk-Ci_i2 heteroaryl); (26) C2_20 alkenyl; and (27) C2_20 alkynyl. In some embodiments, each of these groups can be further substituted as described herein. For example, the alkylene group of a Ci-alkaryl or a Ci-alkheterocyclyl can be further substituted with an oxo group to afford the respective aryloyl and (heterocyclyl)oyl substituent group.
[00765] The term "arylalkoxy," as used herein, represents an alkaryl group, as defined herein, attached to the parent molecular group through an oxygen atom. Exemplary unsubstituted alkoxyalkyl groups include from 7 to 30 carbons (e.g., from 7 to 16 or from 7 to 20 carbons, such as C6-io aryl-Ci_6 alkoxy, C6-io aryl-Ci_io alkoxy, or C6-io aryl-Ci_2o alkoxy). In some embodiments, the arylalkoxy group can be substituted with 1, 2, 3, or 4 substituents as defined herein
[00766] The term "aryloxy" represents a chemical substituent of formula -OR', where R' is an aryl group of 6 to 18 carbons, unless otherwise specified. In some embodiments, the aryl group can be substituted with 1, 2, 3, or 4 substituents as defined herein.
[00767] The term "aryloyl," as used herein, represents an aryl group, as defined herein, that is attached to the parent molecular group through a carbonyl group. Exemplary unsubstituted aryloyl groups are of 7 to 11 carbons. In some embodiments, the aryl group can be substituted with 1, 2, 3, or 4 substituents as defined herein.
[00768] The term "azido" represents an -N3 group, which can also be represented as -N=N=N. [00769] The term "bicyclic," as used herein, refer to a structure having two rings, which may be aromatic or non-aromatic. Bicyclic structures include spirocyclyl groups, as defined herein, and two rings that share one or more bridges, where such bridges can include one atom or a chain including two, three, or more atoms. Exemplary bicyclic groups include a bicyclic carbocyclyl group, where the first and second rings are carbocyclyl groups, as defined herein; a bicyclic aryl groups, where the first and second rings are aryl groups, as defined herein; bicyclic heterocyclyl groups, where the first ring is a heterocyclyl group and the second ring is a carbocyclyl (e.g., aryl) or heterocyclyl (e.g., heteroaryl) group; and bicyclic heteroaryl groups, where the first ring is a heteroaryl group and the second ring is a carbocyclyl (e.g., aryl) or heterocyclyl (e.g., heteroaryl) group. In some
embodiments, the bicyclic group can be substituted with 1, 2, 3, or 4 substituents as defined herein for cycloalkyl, heterocyclyl, and aryl groups.
[00770] The terms "carbocyclic" and "carbocyclyl," as used herein, refer to an optionally substituted C3-12 monocyclic, bicyclic, or tricyclic structure in which the rings, which may be aromatic or non-aromatic, are formed by carbon atoms. Carbocyclic structures include cycloalkyl, cycloalkenyl, and aryl groups.
[00771] The term "carbamoyl," as used herein, represents -C(0)-N(RN1)2, where the meaning of each RN1 is found in the definition of "amino" provided herein.
[00772] The term "carbamoylalkyl," as used herein, represents an alkyl group, as defined herein, substituted by a carbamoyl group, as defined herein. The alkyl group can be further substituted with 1, 2, 3, or 4 substituent groups as described herein.
[00773] The term "carbamyl," as used herein, refers to a carbamate group having the structure -NRN1C(=0)OR or -OC(=0)N(RN1)2, where the meaning of each RN1 is found in the definition of "amino" provided herein, and R is alkyl, cycloalkyl , alkcycloalkyl, aryl, alkaryl, heterocyclyl (e.g., heteroaryl), or alkheterocyclyl (e.g., alkheteroaryl), as defined herein.
[00774] The term "carbonyl," as used herein, represents a C(O) group, which can also be represented as C=0.
[00775] The term "carboxyaldehyde" represents an acyl group having the structure -CHO.
[00776] The term "carboxy," as used herein, means -C02H.
[00777] The term "carboxy alkoxy," as used herein, represents an alkoxy group, as defined herein, substituted by a carboxy group, as defined herein. The alkoxy group can be further substituted with 1, 2, 3, or 4 substituent groups as described herein for the alkyl group. [00778] The term "carboxyalkyl," as used herein, represents an alkyl group, as defined herein, substituted by a carboxy group, as defined herein. The alkyl group can be further substituted with 1 , 2, 3, or 4 substituent groups as described herein.
[00779] The term "cyano," as used herein, represents an -CN group.
[00780] The term "cycloalkoxy" represents a chemical substituent of formula -OR, where R is a C3_ 8 cycloalkyl group, as defined herein, unless otherwise specified. The cycloalkyl group can be further substituted with 1 , 2, 3, or 4 substituent groups as described herein. . Exemplary
unsubstituted cycloalkoxy groups are from 3 to 8 carbons.
[00781] The term "cycloalkyl," as used herein represents a monovalent saturated or unsaturated non-aromatic cyclic hydrocarbon group from three to eight carbons, unless otherwise specified, and is exemplified by cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl,
bicyclo[2.2.1.]heptyl, and the like. When the cycloalkyl group includes one carbon-carbon double bond, the cycloalkyl group can be referred to as a "cycloalkenyl" group. Exemplary cycloalkenyl groups include cyclopentenyl, cyclohexenyl, and the like. The cycloalkyl groups of this invention can be optionally substituted with: (1) Ci_7 acyl (e.g., carboxy aldehyde); (2) Ci_2o alkyl (e.g., Ci_6 alkyl, Ci_6 alkoxy-Ci_6 alkyl, Ci_6 alkylsulfinyl-Ci_6 alkyl, amino-Ci_6 alkyl, azido-Ci_6 alkyl, (carboxyaldehyde)-Ci_6 alkyl, halo-Ci_6 alkyl (e.g., perfluoroalkyl), hydroxy-Ci_6 alkyl, nitro-Ci_6 alkyl, or Ci_6 thioalkoxy-Ci_6 alkyl); (3) Ci_20 alkoxy (e.g., Ci_6 alkoxy, such as perfluoroalkoxy); (4) Ci_6 alkylsulfinyl; (5) C6-10 aryl; (6) amino; (7) Ci_6 alk-C6_io aryl; (8) azido; (9) C3_8 cycloalkyl; (10) Ci_6 alk-C3_8 cycloalkyl; (1 1) halo; (12) C1-12 heterocyclyl (e.g., C1-12 heteroaryl); (13) (C1-12
heterocyclyl)oxy; (14) hydroxy; (15) nitro; (16) Ci_2o thioalkoxy (e.g., Ci_6 thioalkoxy); (17) - (CH2)qC02RA , where q is an integer from zero to four, and RA is selected from the group consisting of (a) Ci_6 alkyl, (b) C6_i0 aryl, (c) hydrogen, and (d) Ci_6 alk-C6_i0 aryl; (18) -(CH2)qCONRB'Rc', where q is an integer from zero to four and where R and R are independently selected from the group consisting of (a) hydrogen, (b) C6-1o alkyl, (c) C6-io aryl, and (d) Ci_6 alk-C6-io aryl; (19) - (CH2)qS02RD , where q is an integer from zero to four and where RD is selected from the group consisting of (a) C6_io alkyl, (b) C6_io aryl, and (c) Ci_6 alk-C6_io aryl; (20) -(CH2)qS02NRE RF , where q is an integer from zero to four and where each of RE and RF is, independently, selected from the group consisting of (a) hydrogen, (b) C6-io alkyl, (c) C6-1o aryl, and (d) Ci_6 alk-C6-io aryl; (21) thiol; (22) C6-io aryloxy; (23) C3_g cycloalkoxy; (24) C6-1o aryl-Ci_6 alkoxy; (25) Ci_6 alk-Ci_i2 heterocyclyl (e.g., Ci_6 alk-Ci_i2 heteroaryl); (26) oxo; (27) C2_20 alkenyl; and (28) C2_20 alkynyl. In some embodiments, each of these groups can be further substituted as described herein. For example, the alkylene group of a Ci-alkaryl or a Ci-alkheterocyclyl can be further substituted with an oxo group to afford the respective aryloyl and (heterocyclyl)oyl substituent group.
[00782] The term "diasteromer" means stereoisomers that are not mirror images of one another and are non-superimposable.
[00783] The term "effective amount" of an agent, as used herein, is that amount sufficient to effect beneficial or desired results, for example, clinical results, and, as such, an "effective amount" depends upon the context in which it is being applied. For example, in the context of administering an agent that treats cancer, an effective amount of an agent is, for example, an amount sufficient to achieve treatment, as defined herein, of cancer, as compared to the response obtained without administration of the agent.
[00784] The term "enantiomer," as used herein, means each individual optically active form of a compound of the invention, having an optical purity or enantiomeric excess (as determined by methods standard in the art) of at least 80% (i.e., at least 90% of one enantiomer and at most 10% of the other enantiomer), preferably at least 90% and more preferably at least 98%.
[00785] The term "halo," as used herein, represents a halogen selected from bromine, chlorine, iodine, or fluorine.
[00786] The term "haloalkoxy," as used herein, represents an alkoxy group, as defined herein, substituted by a halogen group (i.e., F, CI, Br, or I). A haloalkoxy may be substituted with one, two, three, or, in the case of alkyl groups of two carbons or more, four halogens. Haloalkoxy groups include perfiuoroalkoxys (e.g., -OCF3), -OCHF2, -OCH2F, -OCCl3, -OCH2CH2Br, - OCH2CH(CH2CH2Br)CH3, and -OCHICH3. In some embodiments, the haloalkoxy group can be further substituted with 1, 2, 3, or 4 substituent groups as described herein for alkyl groups.
[00787] The term "haloalkyl," as used herein, represents an alkyl group, as defined herein, substituted by a halogen group (i.e., F, CI, Br, or I). A haloalkyl may be substituted with one, two, three, or, in the case of alkyl groups of two carbons or more, four halogens. Haloalkyl groups include perfiuoroalkyls (e.g., -CF3), -CHF2, -CH2F, -CC13, -CH2CH2Br, -CH2CH(CH2CH2Br)CH3, and -CHICH3. In some embodiments, the haloalkyl group can be further substituted with 1, 2, 3, or 4 substituent groups as described herein for alkyl groups.
[00788] The term "heteroalkylene," as used herein, refers to an alkylene group, as defined herein, in which one or two of the constituent carbon atoms have each been replaced by nitrogen, oxygen, or sulfur. In some embodiments, the heteroalkylene group can be further substituted with 1, 2, 3, or 4 substituent groups as described herein for alkylene groups.
[00789] The term "heteroaryl," as used herein, represents that subset of heterocyclyls, as defined herein, which are aromatic: i.e., they contain 4n+2 pi electrons within the mono- or multicyclic ring system. Exemplary unsubstituted heteroaryl groups are of 1 to 12 (e.g., 1 to 11, 1 to 10, 1 to 9, 2 to 12, 2 to 11, 2 to 10, or 2 to 9) carbons. In some embodiment, the heteroaryl is substituted with 1, 2, 3, or 4 substituents groups as defined for a heterocyclyl group.
[00790] The term "heterocyclyl," as used herein represents a 5-, 6- or 7-membered ring, unless otherwise specified, containing one, two, three, or four heteroatoms independently selected from the group consisting of nitrogen, oxygen, and sulfur. The 5-membered ring has zero to two double bonds, and the 6- and 7-membered rings have zero to three double bonds. Exemplary unsubstituted heterocyclyl groups are of 1 to 12 (e.g., 1 to 11, 1 to 10, 1 to 9, 2 to 12, 2 to 11, 2 to 10, or 2 to 9) carbons. The term "heterocyclyl" also represents a heterocyclic compound having a bridged multicyclic structure in which one or more carbons and/or heteroatoms bridges two non-adjacent members of a monocyclic ring, e.g., a quinuclidinyl group. The term "heterocyclyl" includes bicyclic, tricyclic, and tetracyclic groups in which any of the above heterocyclic rings is fused to one, two, or three carbocyclic rings, e.g., an aryl ring, a cyclohexane ring, a cyclohexene ring, a cyclopentane ring, a cyclopentene ring, or another monocyclic heterocyclic ring, such as indolyl, quinolyl, isoquinolyl, tetrahydroquinolyl, benzofuryl, benzothienyl and the like. Examples of fused heterocyclyls include tropanes and l,2,3,5,8,8a-hexahydroindolizine. Heterocyclics include pyrrolyl, pyrrolinyl, pyrrolidinyl, pyrazolyl, pyrazolinyl, pyrazolidinyl, imidazolyl, imidazolinyl, imidazolidinyl, pyridyl, piperidinyl, homopiperidinyl, pyrazinyl, piperazinyl, pyrimidinyl, pyridazinyl, oxazolyl, oxazolidinyl, isoxazolyl, isoxazolidiniyl, morpholinyl, thiomorpholinyl, thiazolyl, thiazolidinyl, isothiazolyl, isothiazolidinyl, indolyl, indazolyl, quinolyl, isoquinolyl, quinoxalinyl, dihydroquinoxalinyl, quinazolinyl, cinnolinyl, phthalazinyl, benzimidazolyl, benzothiazolyl, benzoxazolyl, benzothiadiazolyl, furyl, thienyl, thiazolidinyl, isothiazolyl, triazolyl, tetrazolyl, oxadiazolyl (e.g., 1,2,3-oxadiazolyl), purinyl, thiadiazolyl (e.g., 1,2,3-thiadiazolyl), tetrahydrofuranyl, dihydrofuranyl, tetrahydrothienyl, dihydrothienyl, dihydroindolyl,
dihydroquinolyl, tetrahydroquinolyl, tetrahydroisoquinolyl, dihydroisoquinolyl, pyranyl, dihydropyranyl, dithiazolyl, benzofuranyl, isobenzofuranyl, benzothienyl, and the like, including dihydro and tetrahydro forms thereof, where one or more double bonds are reduced and replaced with hydrogens. Still other exemplary heterocyclyls include: 2,3,4,5-tetrahydro-2-oxo-oxazolyl;
2.3- dihydro-2-oxo-lH-imidazolyl; 2,3,4,5-tetrahydro-5-oxo-lH-pyrazolyl (e.g., 2,3,4,5-tetrahydro-2- phenyl-5-oxo-lH-pyrazolyl); 2,3,4,5-tetrahydro-2,4-dioxo-lH-imidazolyl (e.g., 2,3,4,5-tetrahydro-
2.4- dioxo-5-methyl-5-phenyl-lH-imidazolyl); 2,3-dihydro-2-thioxo-l,3,4-oxadiazolyl (e.g., 2,3- dihydro-2-thioxo-5 -phenyl- 1, 3, 4-oxadiazolyl); 4,5-dihydro-5-oxo-lH-triazolyl (e.g., 4,5-dihydro-3- methyl-4-amino 5-oxo-lH-triazolyl); l,2,3,4-tetrahydro-2,4-dioxopyridinyl (e.g., 1,2,3,4-tetrahydro- 2,4-dioxo-3,3-diethylpyridinyl); 2,6-dioxo-piperidinyl (e.g., 2,6-dioxo-3-ethyl-3-phenylpiperidinyl); l,6-dihydro-6-oxopyridiminyl; l,6-dihydro-4-oxopyrimidinyl (e.g., 2-(methylthio)-l,6-dihydro-4- oxo-5-methylpyrimidin-l-yl); l,2,3,4-tetrahydro-2,4-dioxopyrimidinyl (e.g., l,2,3,4-tetrahydro-2,4- dioxo-3-ethylpyrimidinyl); l,6-dihydro-6-oxo-pyridazinyl (e.g., l,6-dihydro-6-oxo-3- ethylpyridazinyl); 1 ,6-dihydro-6-oxo- 1 ,2,4-triazinyl (e.g., 1 ,6-dihydro-5-isopropyl-6-oxo- 1 ,2,4- triazinyl); 2,3-dihydro-2-oxo-lH-indolyl (e.g., 3,3-dimethyl-2,3-dihydro-2-oxo-lH-indolyl and 2,3- dihydro-2-oxo-3 ,3 ' -spiropropane- lH-indol- 1 -yl); 1 ,3 -dihydro- 1 -oxo-2H-iso-indolyl; 1 ,3-dihydro- l,3-dioxo-2H-iso-indolyl; lH-benzopyrazolyl (e.g., l-(ethoxycarbonyl)- lH-benzopyrazolyl); 2,3- dihydro-2-oxo-lH-benzimidazolyl (e.g., 3-ethyl-2,3-dihydro-2-oxo-lH-benzimidazolyl); 2,3- dihydro-2-oxo-benzoxazolyl (e.g., 5-chloro-2,3-dihydro-2-oxo-benzoxazolyl); 2,3-dihydro-2-oxo- benzoxazolyl; 2-oxo-2H-benzopyranyl; 1 ,4-benzodioxanyl; 1,3-benzodioxanyl; 2,3-dihydro-3- oxo,4H-l ,3-benzothiazinyl; 3,4-dihydro-4-oxo-3H-quinazolinyl (e.g., 2-methyl-3,4-dihydro-4-oxo- 3H-quinazolinyl); 1 ,2,3 ,4-tetrahydro-2,4-dioxo-3H-quinazolyl (e.g. , 1 -ethyl- 1 ,2,3 ,4-tetrahydro-2,4- dioxo-3H-quinazolyl); l,2,3,6-tetrahydro-2,6-dioxo-7H-purinyl (e.g., l,2,3,6-tetrahydro-l,3- dimethyl-2,6-dioxo-7 H -purinyl); l,2,3,6-tetrahydro-2,6-dioxo-l H-purinyl (e.g., 1,2,3,6- tetrahydro-3,7-dimethyl-2,6-dioxo-l H -purinyl); 2-oxobenz[c,<i]indolyl; l,l-dioxo-2H-naphth[l,8- c,d]isothiazolyl; and 1,8-naphthylenedicarboxamido. Additional heterocyclics include 3,3a,4,5,6,6a- hexahydro-pyrrolo[3,4-b]pyrrol-(2H)-yl, and 2,5-diazabicyclo[2.2.1]heptan-2-yl, homopiperazinyl (or diazepanyl), tetrahydropyranyl, dithiazolyl, benzofuranyl, benzothienyl, oxepanyl, thiepanyl, azocan l, oxecanyl, and thiocanyl. Heterocyclic groups also include groups of the formula
Figure imgf000241_0001
E' is selected from the group consisting of -N- and -CH-; F' is selected from the group consisting of -N=CH-, -NH-CH2-, -NH-C(O)-, -NH-, -CH=N-, -CH2-NH-, -C(0)-NH-, -CH=CH-, -CH2-, - CH2CH2-, -CH20-, -OCH2-, -0-, and -S-; and G' is selected from the group consisting of -CH- and - N-. Any of the heterocyclyl groups mentioned herein may be optionally substituted with one, two, three, four or five substituents independently selected from the group consisting of: (1) Ci_7 acyl (e.g., carboxyaldehyde ); (2) Ci_2o alkyl (e.g., Ci_6 alkyl, Ci_6 alkoxy-Ci_6 alkyl, Ci_6 alkylsulfmyl-Ci_ 6 alkyl, amino-Ci_6 alkyl, azido-Ci_6 alkyl, (carboxyaldehyde)-Ci_6 alkyl, halo-Ci_6 alkyl (e.g., perfluoroalkyl), hydroxy-Ci_6 alkyl, nitro-Ci_6 alkyl, or Ci_6 thioalkoxy-Ci_6 alkyl); (3) Ci_2o alkoxy (e.g., Ci_6 alkoxy, such as perfluoroalkoxy); (4) Ci_6 alkylsulfinyl; (5) C6_io aryl; (6) amino; (7) Ci_6 alk-C6-io aryl; (8) azido; (9) C3-8 cycloalkyl; (10) Ci_6 alk-C3_g cycloalkyl; (1 1) halo; (12) C1-12 heterocyclyl (e.g., C2-12 heteroaryl); (13) (C1-12 heterocyclyl)oxy; (14) hydroxy; (15) nitro; (16) Ci_2o thioalkoxy (e.g., Ci_6 thioalkoxy); (17) -(CH2)qC02RA , where q is an integer from zero to four, and RA is selected from the group consisting of (a) Ci_6 alkyl, (b) C6_io aryl, (c) hydrogen, and (d) Ci_6 alk-C6-io aryl; (18) -(CH2)qCONRB Rc , where q is an integer from zero to four and where RB and R are independently selected from the group consisting of (a) hydrogen, (b) Ci_6 alkyl, (c) C6-1o aryl, and (d) Ci_6 alk-C6-io aryl; (19) -(CH2)qS02RD , where q is an integer from zero to four and where RD is selected from the group consisting of (a) Ci_6 alkyl, (b) C6_io aryl, and (c) Ci_6 alk-C6_io aryl; (20) -(CH2)qS02NRE RF , where q is an integer from zero to four and where each of RE and RF is, independently, selected from the group consisting of (a) hydrogen, (b) Ci_6 alkyl, (c) C6-1o aryl, and (d) Ci_6 alk-C6-io aryl; (21) thiol; (22) C6-io aryloxy; (23) C3-8 cycloalkoxy; (24) arylalkoxy; (25) Ci_6 alk-Ci_i2 heterocyclyl (e.g., Ci_6 alk-Ci_i2 heteroaryl); (26) oxo; (27) (C1-12 heterocyclyl)imino; (28) C2-20 alkenyl; and (29) C2_2o alkynyl. In some embodiments, each of these groups can be further substituted as described herein. For example, the alkylene group of a Ci-alkaryl or a Ci- alkheterocyclyl can be further substituted with an oxo group to afford the respective aryloyl and (heterocyclyl)oyl substituent group.
[00791] The term "(heterocyclyl)imino," as used herein, represents a heterocyclyl group, as defined herein, attached to the parent molecular group through an imino group. In some embodiments, the heterocyclyl group can be substituted with 1, 2, 3, or 4 substituent groups as defined herein.
[00792] The term "(heterocyclyl)oxy," as used herein, represents a heterocyclyl group, as defined herein, attached to the parent molecular group through an oxygen atom. In some embodiments, the heterocyclyl group can be substituted with 1, 2, 3, or 4 substituent groups as defined herein.
[00793] The term "(heterocyclyl)oyl," as used herein, represents a heterocyclyl group, as defined herein, attached to the parent molecular group through a carbonyl group. In some embodiments, the heterocyclyl group can be substituted with 1, 2, 3, or 4 substituent groups as defined herein. [00794] The term "hydrocarbon," as used herein, represents a group consisting only of carbon and hydrogen atoms.
[00795] The term "hydroxy," as used herein, represents an -OH group.
[00796] The term "hydroxyalkenyl," as used herein, represents an alkenyl group, as defined herein, substituted by one to three hydroxy groups, with the proviso that no more than one hydroxy group may be attached to a single carbon atom of the alkyl group, and is exemplified by
dihydroxypropenyl, hydroxyisopentenyl, and the like.
[00797] The term "hydroxy alkyl," as used herein, represents an alkyl group, as defined herein, substituted by one to three hydroxy groups, with the proviso that no more than one hydroxy group may be attached to a single carbon atom of the alkyl group, and is exemplified by hydroxymethyl, dihydroxypropyl, and the like.
[00798] The term "isomer," as used herein, means any tautomer, stereoisomer, enantiomer, or diastereomer of any compound of the invention. It is recognized that the compounds of the invention can have one or more chiral centers and/or double bonds and, therefore, exist as stereoisomers, such as double-bond isomers (i.e., geometric E/Z isomers) or diastereomers (e.g., enantiomers (i.e., (+) or (-)) or cis/trans isomers). According to the invention, the chemical structures depicted herein, and therefore the compounds of the invention, encompass all of the corresponding stereoisomers, that is, both the stereomerically pure form (e.g., geometrically pure, enantiomerically pure, or diastereomerically pure) and enantiomeric and stereoisomeric mixtures, e.g., racemates. Enantiomeric and stereoisomeric mixtures of compounds of the invention can typically be resolved into their component enantiomers or stereoisomers by well-known methods, such as chiral-phase gas chromatography, chiral-phase high performance liquid chromatography, crystallizing the compound as a chiral salt complex, or crystallizing the compound in a chiral solvent. Enantiomers and stereoisomers can also be obtained from stereomerically or
enantiomerically pure intermediates, reagents, and catalysts by well-known asymmetric synthetic methods.
[00799] The term "N-protected amino," as used herein, refers to an amino group, as defined herein, to which is attached one or two N-protecting groups, as defined herein.
[00800] The term "N-protecting group," as used herein, represents those groups intended to protect an amino group against undesirable reactions during synthetic procedures. Commonly used N- protecting groups are disclosed in Greene, "Protective Groups in Organic Synthesis," 3rd Edition (John Wiley & Sons, New York, 1999), which is incorporated herein by reference. N-protecting groups include acyl, aryloyl, or carbamyl groups such as formyl, acetyl, propionyl, pivaloyl, t- butylacetyl, 2-chloroacetyl, 2-bromoacetyl, trifluoroacetyl, trichloroacetyl, phthalyl, o- nitrophenoxyacetyl, a-chlorobutyryl, benzoyl, 4-chlorobenzoyl, 4-bromobenzoyl, 4-nitrobenzoyl, and chiral auxiliaries such as protected or unprotected D, L or D, L-amino acids such as alanine, leucine, phenylalanine, and the like; sulfonyl-containing groups such as benzenesulfonyl, p- toluenesulfonyl, and the like; carbamate forming groups such as benzyloxycarbonyl, p- chlorobenzyloxycarbonyl, p-methoxybenzyloxycarbonyl, p-nitrobenzyloxycarbonyl, 2- nitrobenzyloxycarbonyl, p-bromobenzyloxycarbonyl, 3 ,4-dimethoxybenzyloxycarbonyl,
3,5-dimethoxybenzyloxycarbonyl, 2,4-dimethoxybenzyloxycarbonyl, 4-methoxybenzyloxycarbonyl, 2-nitro-4,5-dimethoxybenzyloxycarbonyl, 3,4,5-trimethoxybenzyloxycarbonyl, l-(p-biphenylyl)-l- methylethoxycarbonyl, a,a-dimethyl-3,5-dimethoxybenzyloxycarbonyl, benzhydryloxy carbonyl, t- butyloxycarbonyl, diisopropylmethoxycarbonyl, isopropyloxycarbonyl, ethoxycarbonyl,
methoxycarbonyl, allyloxycarbonyl, 2,2,2,-trichloroethoxycarbonyl, phenoxycarbonyl, 4- nitrophenoxy carbonyl, fluorenyl-9-methoxycarbonyl, cyclopentyloxycarbonyl,
adamantyloxycarbonyl, cyclohexyloxycarbonyl, phenylthiocarbonyl, and the like, alkaryl groups such as benzyl, triphenylmethyl, benzyloxymethyl, and the like and silyl groups, such as
trimethylsilyl, and the like. Preferred N-protecting groups are formyl, acetyl, benzoyl, pivaloyl, t- butylacetyl, alanyl, phenylsulfonyl, benzyl, t-butyloxycarbonyl (Boc), and benzyloxycarbonyl (Cbz).
[00801] The term "nitro," as used herein, represents an -N02 group.
[00802] The term "oxo" as used herein, represents =0.
[00803] The term "perfluoroalkyl," as used herein, represents an alkyl group, as defined herein, where each hydrogen radical bound to the alkyl group has been replaced by a fluoride radical.
Perfluoroalkyl groups are exemplified by trif uoromethyl, pentafluoroethyl, and the like.
[00804] The term "perfluoroalkoxy," as used herein, represents an alkoxy group, as defined herein, where each hydrogen radical bound to the alkoxy group has been replaced by a fluoride radical. Perfluoroalkoxy groups are exemplified by trifluoromethoxy, pentafluoroethoxy, and the like.
[00805] The term "spirocyclyl," as used herein, represents a C2_7 alkylene diradical, both ends of which are bonded to the same carbon atom of the parent group to form a spirocyclic group, and also a Ci_6 heteroalkylene diradical, both ends of which are bonded to the same atom. The heteroalkylene radical forming the spirocyclyl group can containing one, two, three, or four heteroatoms independently selected from the group consisting of nitrogen, oxygen, and sulfur. In some embodiments, the spirocyclyl group includes one to seven carbons, excluding the carbon atom to which the diradical is attached. The spirocyclyl groups of the invention may be optionally substituted with 1, 2, 3, or 4 substituents provided herein as optional substituents for cycloalkyl and/or heterocyclyl groups.
[00806] The term "stereoisomer," as used herein, refers to all possible different isomeric as well as conformational forms which a compound may possess (e.g., a compound of any formula described herein), in particular all possible stereochemically and conformationally isomeric forms, all diastereomers, enantiomers and/or conformers of the basic molecular structure. Some compounds of the present invention may exist in different tautomeric forms, all of the latter being included within the scope of the present invention.
[00807] The term "sulfoalkyl," as used herein, represents an alkyl group, as defined herein, substituted by a sulfo group of -SO3H. In some embodiments, the alkyl group can be further substituted with 1, 2, 3, or 4 substituent groups as described herein.
[00808] The term "sulfonyl," as used herein, represents an -S(0)2- group.
[00809] The term "thioalkaryl," as used herein, represents a chemical substituent of formula -SR, where R is an alkaryl group. In some embodiments, the alkaryl group can be further substituted with 1, 2, 3, or 4 substituent groups as described herein.
[00810] The term "thioalkheterocyclyl," as used herein, represents a chemical substituent of formula -SR, where R is an alkheterocyclyl group. In some embodiments, the alkheterocyclyl group can be further substituted with 1, 2, 3, or 4 substituent groups as described herein.
[00811] The term "thioalkoxy," as used herein, represents a chemical substituent of formula -SR, where R is an alkyl group, as defined herein. In some embodiments, the alkyl group can be further substituted with 1, 2, 3, or 4 substituent groups as described herein.
[00812] The term "thiol" represents an -SH group.
[00813] Compound: As used herein, the term "compound," is meant to include all stereoisomers, geometric isomers, tautomers, and isotopes of the structures depicted.
[00814] The compounds described herein can be asymmetric {e.g., having one or more
stereocenters). All stereoisomers, such as enantiomers and diastereomers, are intended unless otherwise indicated. Compounds of the present disclosure that contain asymmetrically substituted carbon atoms can be isolated in optically active or racemic forms. Methods on how to prepare optically active forms from optically active starting materials are known in the art, such as by resolution of racemic mixtures or by stereoselective synthesis. Many geometric isomers of olefins, C=N double bonds, and the like can also be present in the compounds described herein, and all such stable isomers are contemplated in the present disclosure. Cis and trans geometric isomers of the compounds of the present disclosure are described and may be isolated as a mixture of isomers or as separated isomeric forms.
[00815] Compounds of the present disclosure also include tautomeric forms. Tautomeric forms result from the swapping of a single bond with an adjacent double bond and the concomitant migration of a proton. Tautomeric forms include prototropic tautomers which are isomeric protonation states having the same empirical formula and total charge. Examples prototropic tautomers include ketone - enol pairs, amide - imidic acid pairs, lactam - lactim pairs, amide - imidic acid pairs, enamine - imine pairs, and annular forms where a proton can occupy two or more positions of a heterocyclic system, such as, 1H- and 3H-imidazole, 1H-, 2H- and 4H- 1,2,4-triazole, 1H- and 2H- isoindole, and 1H- and 2H-pyrazole. Tautomeric forms can be in equilibrium or sterically locked into one form by appropriate substitution.
[00816] Compounds of the present disclosure also include all of the isotopes of the atoms occurring in the intermediate or final compounds. "Isotopes" refers to atoms having the same atomic number but different mass numbers resulting from a different number of neutrons in the nuclei. For example, isotopes of hydrogen include tritium and deuterium.
[00817] The compounds and salts of the present disclosure can be prepared in combination with solvent or water molecules to form solvates and hydrates by routine methods.
[00818] Conserved: As used herein, the term "conserved" refers to nucleotides or amino acid residues of a polynucleotide sequence or polypeptide sequence, respectively, that are those that occur unaltered in the same position of two or more sequences being compared. Nucleotides or amino acids that are relatively conserved are those that are conserved amongst more related sequences than nucleotides or amino acids appearing elsewhere in the sequences.
[00819] In some embodiments, two or more sequences are said to be "completely conserved" if they are 100% identical to one another. In some embodiments, two or more sequences are said to be "highly conserved" if they are at least 70% identical, at least 80% identical, at least 90% identical, or at least 95% identical to one another. In some embodiments, two or more sequences are said to be "highly conserved" if they are about 70%> identical, about 80%> identical, about 90%> identical, about 95%, about 98%), or about 99% identical to one another. In some embodiments, two or more sequences are said to be "conserved" if they are at least 30%> identical, at least 40%> identical, at least 50% identical, at least 60% identical, at least 70% identical, at least 80% identical, at least 90% identical, or at least 95% identical to one another. In some embodiments, two or more sequences are said to be "conserved" if they are about 30%> identical, about 40%> identical, about 50%> identical, about 60%) identical, about 70%> identical, about 80%> identical, about 90%> identical, about 95% identical, about 98% identical, or about 99% identical to one another. Conservation of sequence may apply to the entire length of an oligonucleotide or polypeptide or may apply to a portion, region or feature thereof.
[00820] Controlled Release: As used herein, the term "controlled release" refers to a
pharmaceutical composition or compound release profile that conforms to a particular pattern of release to effect a therapeutic outcome.
[00821] Cyclic or Cyclized: As used herein, the term "cyclic" refers to the presence of a continuous loop. Cyclic molecules need not be circular, only joined to form an unbroken chain of subunits. Cyclic molecules such as the engineered RNA or mRNA of the present invention may be single units or multimers or comprise one or more components of a complex or higher order structure.
[00822] Cytostatic: As used herein, "cytostatic" refers to inhibiting, reducing, suppressing the growth, division, or multiplication of a cell {e.g., a mammalian cell {e.g., a human cell)), bacterium, virus, fungus, protozoan, parasite, prion, or a combination thereof.
[00823] Cytotoxic: As used herein, "cytotoxic" refers to killing or causing injurious, toxic, or deadly effect on a cell {e.g. , a mammalian cell {e.g. , a human cell)), bacterium, virus, fungus, protozoan, parasite, prion, or a combination thereof.
[00824] Delivery: As used herein, "delivery" refers to the act or manner of delivering a compound, substance, entity, moiety, cargo or payload.
[00825] Delivery Agent: As used herein, "delivery agent" refers to any substance which facilitates, at least in part, the in vivo delivery of a modified nucleic acid or mrnRNA to targeted cells.
[00826] Destabilized: As used herein, the term "destable," "destabilize," or "destabilizing region" means a region or molecule that is less stable than a starting, wild-type or native form of the same region or molecule.
[00827] Detectable label: As used herein, "detectable label" refers to one or more markers, signals, or moieties which are attached, incorporated or associated with another entity that is readily detected by methods known in the art including radiography, fluorescence, chemiluminescence, enzymatic activity, absorbance and the like. Detectable labels include radioisotopes, fluorophores,
chromophores, enzymes, dyes, metal ions, ligands such as biotin, avidin, streptavidin and haptens, quantum dots, and the like. Detectable labels may be located at any position in the peptides or proteins disclosed herein. They may be within the amino acids, the peptides, or proteins, or located at the N- or C- termini.
[00828] Digest: As used herein, the term "digest" means to break apart into smaller pieces or components. When referring to polypeptides or proteins, digestion results in the production of peptides.
[00829] Distal: As used herein, the term "distal" means situated away from the center or away from a point or region of interest.
[00830] Dose splitting factor (DSF)-x&t\o of PUD of dose split treatment divided by PUD of total daily dose or single unit dose. The value is derived from comparison of dosing regimens groups.
[00831] Encapsulate: As used herein, the term "encapsulate" means to enclose, surround or encase.
[00832] Engineered: As used herein, embodiments of the invention are "engineered" when they are designed to have a feature or property, whether structural or chemical, that varies from a starting point, wild type or native molecule.
[00833] Exosome: As used herein, "exosome" is a vesicle secreted by mammalian cells.
[00834] Expression: As used herein, "expression" of a nucleic acid sequence refers to one or more of the following events: (1) production of an R A template from a DNA sequence (e.g., by transcription); (2) processing of an RNA transcript (e.g., by splicing, editing, 5' cap formation, and/or 3' end processing); (3) translation of an RNA into a polypeptide or protein; and (4) post- translational modification of a polypeptide or protein.
[00835] Feature: As used herein, a "feature" refers to a characteristic, a property, or a distinctive element.
[00836] Formulation: As used herein, a "formulation" includes at least a modified nucleic acid or mmRNA and a delivery agent.
[00837] Fragment: A "fragment," as used herein, refers to a portion. For example, fragments of proteins may comprise polypeptides obtained by digesting full-length protein isolated from cultured cells. [00838] Functional: As used herein, a "functional" biological molecule is a biological molecule in a form in which it exhibits a property and/or activity by which it is characterized.
[00839] Homology: As used herein, the term "homology" refers to the overall relatedness between polymeric molecules, e.g. between nucleic acid molecules (e.g. DNA molecules and/or R A molecules) and/or between polypeptide molecules. In some embodiments, polymeric molecules are considered to be "homologous" to one another if their sequences are at least 25%, 30%>, 35%, 40%>, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 99% identical or similar. The term "homologous" necessarily refers to a comparison between at least two sequences
(polynucleotide or polypeptide sequences). In accordance with the invention, two polynucleotide sequences are considered to be homologous if the polypeptides they encode are at least about 50%>, 60%, 70%, 80%, 90%, 95%, or even 99% for at least one stretch of at least about 20 amino acids. In some embodiments, homologous polynucleotide sequences are characterized by the ability to encode a stretch of at least 4-5 uniquely specified amino acids. For polynucleotide sequences less than 60 nucleotides in length, homology is determined by the ability to encode a stretch of at least 4-5 uniquely specified amino acids. In accordance with the invention, two protein sequences are considered to be homologous if the proteins are at least about 50%>, 60%>, 70%>, 80%>, or 90%> identical for at least one stretch of at least about 20 amino acids.
[00840] Identity: As used herein, the term "identity" refers to the overall relatedness between polymeric molecules, e.g., between oligonucleotide molecules (e.g. DNA molecules and/or RNA molecules) and/or between polypeptide molecules. Calculation of the percent identity of two polynucleotide sequences, for example, can be performed by aligning the two sequences for optimal comparison purposes (e.g. , gaps can be introduced in one or both of a first and a second nucleic acid sequences for optimal alignment and non-identical sequences can be disregarded for comparison purposes). In certain embodiments, the length of a sequence aligned for comparison purposes is at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%), or 100%) of the length of the reference sequence. The nucleotides at corresponding nucleotide positions are then compared. When a position in the first sequence is occupied by the same nucleotide as the corresponding position in the second sequence, then the molecules are identical at that position. The percent identity between the two sequences is a function of the number of identical positions shared by the sequences, taking into account the number of gaps, and the length of each gap, which needs to be introduced for optimal alignment of the two sequences. The comparison of sequences and determination of percent identity between two sequences can be accomplished using a mathematical algorithm. For example, the percent identity between two nucleotide sequences can be determined using methods such as those described in Computational Molecular Biology, Lesk, A. M., ed., Oxford University Press, New York, 1988; Biocomputing: Informatics and Genome Projects, Smith, D. W., ed., Academic Press, New York, 1993; Sequence Analysis in Molecular Biology, von Heinje, G., Academic Press, 1987; Computer Analysis of Sequence Data, Part I, Griffin, A. M., and Griffin, H. G., eds., Humana Press, New Jersey, 1994; and Sequence Analysis Primer, Gribskov, M. and Devereux, J., eds., M Stockton Press, New York, 1991; each of which is incorporated herein by reference. For example, the percent identity between two nucleotide sequences can be determined using the algorithm of Meyers and Miller (CABIOS, 1989, 4: 11-17), which has been incorporated into the ALIGN program (version 2.0) using a PAM120 weight residue table, a gap length penalty of 12 and a gap penalty of 4. The percent identity between two nucleotide sequences can, alternatively, be determined using the GAP program in the GCG software package using an NWSgapdna.CMP matrix. Methods commonly employed to determine percent identity between sequences include, but are not limited to those disclosed in Carillo, H., and Lipman, D., SIAM J Applied Math., 48: 1073 (1988); incorporated herein by reference. Techniques for determining identity are codified in publicly available computer programs. Exemplary computer software to determine homology between two sequences include, but are not limited to, GCG program package, Devereux, J., et ah, Nucleic Acids Research, 12(1), 387 (1984)), BLASTP, BLASTN, and FASTA Altschul, S. F. et al, J. Molec. Biol, 215, 403 (1990)).
[00841] Inhibit expression of a gene: As used herein, the phrase "inhibit expression of a gene" means to cause a reduction in the amount of an expression product of the gene. The expression product can be an RNA transcribed from the gene {e.g. , an mRNA) or a polypeptide translated from an mRNA transcribed from the gene. Typically a reduction in the level of an mRNA results in a reduction in the level of a polypeptide translated therefrom. The level of expression may be determined using standard techniques for measuring mRNA or protein.
[00842] In vitro: As used herein, the term "in vitro" refers to events that occur in an artificial environment, e.g. , in a test tube or reaction vessel, in cell culture, in a Petri dish, etc. , rather than within an organism (e.g., animal, plant, or microbe).
[00843] In vivo: As used herein, the term "in vivo" refers to events that occur within an organism (e.g., animal, plant, or microbe or cell or tissue thereof). [00844] Isolated: As used herein, the term "isolated" refers to a substance or entity that has been separated from at least some of the components with which it was associated (whether in nature or in an experimental setting). Isolated substances may have varying levels of purity in reference to the substances from which they have been associated. Isolated substances and/or entities may be separated from at least about 10%, about 20%>, about 30%>, about 40%>, about 50%>, about 60%>, about 70%, about 80%, about 90%, or more of the other components with which they were initially associated. In some embodiments, isolated agents are more than about 80%>, about 85%, about 90%>, about 91%, about 92%, about 93%, about 94%, about 95%, about 96%, about 97%, about 98%, about 99%), or more than about 99% pure. As used herein, a substance is "pure" if it is substantially free of other components. Substantially isolated: By "substantially isolated" is meant that the compound is substantially separated from the environment in which it was formed or detected.
Partial separation can include, for example, a composition enriched in the compound of the present disclosure. Substantial separation can include compositions containing at least about 50%, at least about 60%), at least about 70%>, at least about 80%>, at least about 90%>, at least about 95%, at least about 97%), or at least about 99% by weight of the compound of the present disclosure, or salt thereof. Methods for isolating compounds and their salts are routine in the art.
[00845] Linker: As used herein, a linker refers to a group of atoms, e.g., 10-1,000 atoms, and can be comprised of the atoms or groups such as, but not limited to, carbon, amino, alkylamino, oxygen, sulfur, sulfoxide, sulfonyl, carbonyl, and imine. The linker can be attached to a modified nucleoside or nucleotide on the nucleobase or sugar moiety at a first end, and to a payload, e.g., a detectable or therapeutic agent, at a second end. The linker may be of sufficient length as to not interfere with incorporation into a nucleic acid sequence. The linker can be used for any useful purpose, such as to form mmR A multimers (e.g., through linkage of two or more modified nucleic acid molecules or mmR A molecules) or mmRNA conjugates, as well as to administer a payload, as described herein. Examples of chemical groups that can be incorporated into the linker include, but are not limited to, alkyl, alkenyl, alkynyl, amido, amino, ether, thioether, ester, alkylene, heteroalkylene, aryl, or heterocyclyl, each of which can be optionally substituted, as described herein. Examples of linkers include, but are not limited to, unsaturated alkanes, polyethylene glycols (e.g., ethylene or propylene glycol monomeric units, e.g., diethylene glycol, dipropylene glycol, triethylene glycol, tripropylene glycol, tetraethylene glycol, or tetraethylene glycol), and dextran polymers and derivatives thereof. Other examples include, but are not limited to, cleavable moieties within the linker, such as, for example, a disulfide bond (-S-S-) or an azo bond (-N=N-), which can be cleaved using a reducing agent or photolysis. Non-limiting examples of a selectively cleavable bond include an amido bond can be cleaved for example by the use of tris(2-carboxyethyl)phosphine (TCEP), or other reducing agents, and/or photolysis, as well as an ester bond can be cleaved for example by acidic or basic hydrolysis.
[00846] MicroRNA (miRNA) binding site: As used herein, a microRNA (miRNA) binding site represents a nucleotide location or region of a nucleic acid transcript to which at least the "seed" region of a miRNA binds.
[00847] Modified: As used herein "modified" refers to a changed state or structure of a molecule of the invention. Molecules may be modified in many ways including chemically, structurally, and functionally. In one embodiment, the mRNA molecules of the present invention are modified by the introduction of non-natural nucleosides and/or nucleotides.
[00848] Mucus: As used herein, "mucus" refers to a natural substance that is viscous and comprises mucin glycoproteins.
[00849] Naturally occurring: As used herein, "naturally occurring" means existing in nature without artificial aid.
[00850] Non-human vertebrate: As used herein, a "non human vertebrate" includes all vertebrates except Homo sapiens, including wild and domesticated species. Examples of non-human vertebrates include, but are not limited to, mammals, such as alpaca, banteng, bison, camel, cat, cattle, deer, dog, donkey, gayal, goat, guinea pig, horse, llama, mule, pig, rabbit, reindeer, sheep water buffalo, and yak.
[00851] Off-target: As used herein, "off target" refers to any unintended effect on any one or more target, gene, or cellular transcript.
[00852] Open reading frame: As used herein, "open reading frame" or "ORF" refers to a sequence which does not contain a stop codon in a given reading frame.
[00853] Operably linked: As used herein, the phrase "operably linked" refers to a functional connection between two or more molecules, constructs, transcripts, entities, moieties or the like.
[00854] Paratope: As used herein, a "paratope" refers to the antigen-binding site of an antibody.
[00855] Patient: As used herein, "patient" refers to a subject who may seek or be in need of treatment, requires treatment, is receiving treatment, will receive treatment, or a subject who is under care by a trained professional for a particular disease or condition. [00856] Peptide: As used herein, "peptide" is less than or equal to 50 amino acids long, e.g., about 5, 10, 15, 20, 25, 30, 35, 40, 45, or 50 amino acids long.
[00857] Pharmaceutically acceptable: The phrase "pharmaceutically acceptable" is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or
complication, commensurate with a reasonable benefit/risk ratio.
[00858] Pharmaceutically acceptable excipients: The phrase "pharmaceutically acceptable excipient," as used herein, refers any ingredient other than the compounds described herein (for example, a vehicle capable of suspending or dissolving the active compound) and having the properties of being substantially nontoxic and non-inflammatory in a patient. Excipients may include, for example: antiadherents, antioxidants, binders, coatings, compression aids, disintegrants, dyes (colors), emollients, emulsifiers, fillers (diluents), film formers or coatings, flavors, fragrances, glidants (flow enhancers), lubricants, preservatives, printing inks, sorbents, suspensing or dispersing agents, sweeteners, and waters of hydration. Exemplary excipients include, but are not limited to: butylated hydroxytoluene (BHT), calcium carbonate, calcium phosphate (dibasic), calcium stearate, croscarmellose, crosslinked polyvinyl pyrrolidone, citric acid, crospovidone, cysteine, ethylcellulose, gelatin, hydroxypropyl cellulose, hydroxypropyl methylcellulose, lactose, magnesium stearate, maltitol, mannitol, methionine, methylcellulose, methyl paraben, microcrystalline cellulose, polyethylene glycol, polyvinyl pyrrolidone, povidone, pregelatinized starch, propyl paraben, retinyl palmitate, shellac, silicon dioxide, sodium carboxymethyl cellulose, sodium citrate, sodium starch glycolate, sorbitol, starch (corn), stearic acid, sucrose, talc, titanium dioxide, vitamin A, vitamin E, vitamin C, and xylitol.
[00859] Pharmaceutically acceptable salts: The present disclosure also includes pharmaceutically acceptable salts of the compounds described herein. As used herein, "pharmaceutically acceptable salts" refers to derivatives of the disclosed compounds wherein the parent compound is modified by converting an existing acid or base moiety to its salt form (e.g., by reacting the free base group with a suitable organic acid). Examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like. Representative acid addition salts include acetate, adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, fumarate, glucoheptonate, glycerophosphate, hemisulfate, heptonate, hexanoate, hydrobromide, hydrochloride, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectinate, persulfate, 3-phenylpropionate, phosphate, picrate, pivalate, propionate, stearate, succinate, sulfate, tartrate, thiocyanate, toluenesulfonate, undecanoate, valerate salts, and the like. Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like, as well as nontoxic ammonium, quaternary ammonium, and amine cations, including, but not limited to ammonium,
tetramethylammonium, tetraethylammonium, methylamine, dimethylamine, trimethylamine, triethylamine, ethylamine, and the like. The pharmaceutically acceptable salts of the present disclosure include the conventional non-toxic salts of the parent compound formed, for example, from non-toxic inorganic or organic acids. The pharmaceutically acceptable salts of the present disclosure can be synthesized from the parent compound which contains a basic or acidic moiety by conventional chemical methods. Generally, such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two; generally, nonaqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile are preferred. Lists of suitable salts are found in Remington 's Pharmaceutical Sciences, 17th ed., Mack Publishing Company, Easton, Pa., 1985, p. 1418, Pharmaceutical Salts: Properties, Selection, and Use, P.H. Stahl and C.G. Wermuth (eds.), Wiley- VCH, 2008, and Berge et al, Journal of Pharmaceutical Science, 66, 1-19 (1977), each of which is incorporated herein by reference in its entirety.
[00860] Pharmaceutically acceptable solvate: The term "pharmaceutically acceptable solvate," as used herein, means a compound of the invention wherein molecules of a suitable solvent are incorporated in the crystal lattice. A suitable solvent is physiologically tolerable at the dosage administered. For example, solvates may be prepared by crystallization, recrystallization, or precipitation from a solution that includes organic solvents, water, or a mixture thereof. Examples of suitable solvents are ethanol, water (for example, mono-, di-, and tri-hydrates), N- methylpyrrolidinone (NMP), dimethyl sulfoxide (DMSO), N,N'-dimethylformamide (DMF), Ν,Ν'- dimethylacetamide (DMAC), l,3-dimethyl-2-imidazolidinone (DMEU), l,3-dimethyl-3,4,5,6- tetrahydro-2-(lH)-pyrimidinone (DMPU), acetonitrile (ACN), propylene glycol, ethyl acetate, benzyl alcohol, 2-pyrrolidone, benzyl benzoate, and the like. When water is the solvent, the solvate is referred to as a "hydrate."
[00861] Pharmacokinetic: As used herein, "pharmacokinetic" refers to any one or more properties of a molecule or compound as it relates to the determination of the fate of substances administered to a living organism. Pharmacokinetics is divided into several areas including the extent and rate of absorption, distribution, metabolism and excretion. This is commonly referred to as ADME where: (A) Absorption is the process of a substance entering the blood circulation; (D) Distribution is the dispersion or dissemination of substances throughout the fluids and tissues of the body; (M) Metabolism (or Biotransformation) is the irreversible transformation of parent compounds into daughter metabolites; and (E) Excretion (or Elimination) refers to the elimination of the substances from the body. In rare cases, some drugs irreversibly accumulate in body tissue.
[00862] Pharmacologic effect: As used herein, a "pharmacologic effect" is a measurable biologic phenomenon in an organism or system which occurs after the organism or system has been contacted with or exposed to an exogenous agent. Pharmacologic effects may result in therapeutically effective outcomes such as the treatment, improvement of one or more symptoms, diagnosis, prevention, and delay of onset of disease, disorder, condition or infection. Measurement of such biologic phenomena may be quantitative, qualitative or relative to another biologic phenomenon. Quantitative measurements may be statistically significant. Qualitative measurements may be by degree or kind and may be at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or more different. They may be observable as present or absent, better or worse, greater or less. Exogenous agents, when referring to pharmacologic effects are those agents which are, in whole or in part, foreign to the organism or system. For example, modifications to a wild type biomolecule, whether structural or chemical, would produce an exogenous agent. Likewise, incorporation or combination of a wild type molecule into or with a compound, molecule or substance not found naturally in the organism or system would also produce an exogenous agent. The modified mR A of the present invention, comprise exogenous agents. Examples of pharmacologic effects include, but are not limited to, alteration in cell count such as an increase or decrease in neutrophils, reticulocytes, granulocytes, erythrocytes (red blood cells), megakaryocytes, platelets, monocytes, connective tissue
macrophages, epidermal langerhans cells, osteoclasts, dendritic cells, microglial cells, neutrophils, eosinophils, basophils, mast cells, helper T cells, suppressor T cells, cytotoxic T cells, natural killer T cells, B cells, natural killer cells, or reticulocytes. Pharmacologic effects also include alterations in blood chemistry, H, hemoglobin, hematocrit, changes in levels of enzymes such as, but not limited to, liver enzymes AST and ALT, changes in lipid profiles, electrolytes, metabolic markers, hormones or other marker or profile known to those of skill in the art.
[00863] Physicochemical: As used herein, "physicochemical" means of or relating to a physical and/or chemical property.
[00864] Preventing: As used herein, the term "preventing" refers to partially or completely delaying onset of an infection, disease, disorder and/or condition; partially or completely delaying onset of one or more symptoms, features, or clinical manifestations of a particular infection, disease, disorder, and/or condition; partially or completely delaying onset of one or more symptoms, features, or manifestations of a particular infection, disease, disorder, and/or condition; partially or completely delaying progression from an infection, a particular disease, disorder and/or condition; and/or decreasing the risk of developing pathology associated with the infection, the disease, disorder, and/or condition.
[00865] Prodrug: The present disclosure also includes prodrugs of the compounds described herein. As used herein, "prodrugs" refer to any substance, molecule or entity which is in a form predicate for that substance, molecule or entity to act as a therapeutic upon chemical or physical alteration. Prodrugs may by covalently bonded or sequestered in some way and which release or are converted into the active drug moiety prior to, upon or after administered to a mammalian subject. Prodrugs can be prepared by modifying functional groups present in the compounds in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to the parent compounds.
Prodrugs include compounds wherein hydroxyl, amino, sulfhydryl, or carboxyl groups are bonded to any group that, when administered to a mammalian subject, cleaves to form a free hydroxyl, amino, sulfhydryl, or carboxyl group respectively. Preparation and use of prodrugs is discussed in T.
Higuchi and V. Stella, "Pro-drugs as Novel Delivery Systems," Vol. 14 of the A.C.S. Symposium Series, and in Bioreversible Carriers in Drug Design, ed. Edward B. Roche, American
Pharmaceutical Association and Pergamon Press, 1987, both of which are hereby incorporated by reference in their entirety.
[00866] Proliferate: As used herein, the term "proliferate" means to grow, expand or increase or cause to grow, expand or increase rapidly. "Proliferative" means having the ability to proliferate. "Anti-proliferative" means having properties counter to or inapposite to proliferative properties. [00867] Protein of interest: As used herein, the terms "proteins of interest" or "desired proteins" include those provided herein and fragments, mutants, variants, and alterations thereof.
[00868] Proximal: As used herein, the term "proximal" means situated nearer to the center or to a point or region of interest.
[00869] Pseudouridine: As used herein, pseudouridine refers to the C-glycoside isomer of the nucleoside uridine. A "pseudouridine analog" is any modification, variant, isoform or derivative of pseudouridine. For example, pseudouridine analogs include but are not limited to 1-carboxymethyl- pseudouridine, 1-propynyl-pseudouridine, 1 -taurinomethyl-pseudouridine, l-taurinomethyl-4-thio- pseudouridine, 1-methyl-pseudouridine
Figure imgf000257_0001
4-thio-l- methyl-pseudouridine, 3-methyl-pseudouridine (m3\|/), 2-thio- 1-methyl-pseudouridine, 1-methyl-l- deaza-pseudouridine, 2-thio- 1 -methyl- 1-deaza-pseudouridine, dihydropseudouridine, 2-thio- dihydropseudouridine, 2-methoxyuridine, 2-methoxy-4-thio-uridine, 4-methoxy-pseudouridine, 4- methoxy-2-thio-pseudouridine, N 1 -methyl-pseudouridine, 1 -methyl-3 -(3 -amino-3 - carboxypropyl)pseudouridine (acp3 ψ), and 2'-0-methyl-pseudouridine (\|/m).
[00870] Purified: As used herein, "purify," "purified," "purification" means to make substantially pure or clear from unwanted components, material defilement, admixture or imperfection.
[00871] Sample: As used herein, the term "sample" or "biological sample" refers to a subset of its tissues, cells or component parts (e.g. body fluids, including but not limited to blood, mucus, lymphatic fluid, synovial fluid, cerebrospinal fluid, saliva, amniotic fluid, amniotic cord blood, urine, vaginal fluid and semen). A sample further may include a homogenate, lysate or extract prepared from a whole organism or a subset of its tissues, cells or component parts, or a fraction or portion thereof, including but not limited to, for example, plasma, serum, spinal fluid, lymph fluid, the external sections of the skin, respiratory, intestinal, and genitourinary tracts, tears, saliva, milk, blood cells, tumors, organs. A sample further refers to a medium, such as a nutrient broth or gel, which may contain cellular components, such as proteins or nucleic acid molecule.
[00872] Signal Sequences: As used herein, the phrase "signal sequences" refers to a sequence which can direct the transport or localization of a protein.
[00873] Single unit dose: As used herein, a "single unit dose" is a dose of any therapeutic administed in one dose/at one time/single route/single point of contact, i.e., single administration event. [00874] Similarity: As used herein, the term "similarity" refers to the overall relatedness between polymeric molecules, e.g. between polynucleotide molecules {e.g. DNA molecules and/or R A molecules) and/or between polypeptide molecules. Calculation of percent similarity of polymeric molecules to one another can be performed in the same manner as a calculation of percent identity, except that calculation of percent similarity takes into account conservative substitutions as is understood in the art.
[00875] Split dose: As used herein, a "split dose" is the division of single unit dose or total daily dose into two or more doses.
[00876] Stable: As used herein "stable" refers to a compound that is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture, and preferably capable of formulation into an efficacious therapeutic agent.
[00877] Stabilized: As used herein, the term "stabilize", "stabilized," "stabilized region" means to make or become stable.
[00878] Subject: As used herein, the term "subject" or "patient" refers to any organism to which a composition in accordance with the invention may be administered, e.g., for experimental, diagnostic, prophylactic, and/or therapeutic purposes. Typical subjects include animals {e.g., mammals such as mice, rats, rabbits, non-human primates, and humans) and/or plants.
[00879] Substantially: As used herein, the term "substantially" refers to the qualitative condition of exhibiting total or near-total extent or degree of a characteristic or property of interest. One of ordinary skill in the biological arts will understand that biological and chemical phenomena rarely, if ever, go to completion and/or proceed to completeness or achieve or avoid an absolute result. The term "substantially" is therefore used herein to capture the potential lack of completeness inherent in many biological and chemical phenomena.
[00880] Substantially equal: As used herein as it relates to time differences between doses, the term means plus/minus 2%.
[00881] Substantially simultaneously: As used herein and as it relates to plurality of doses, the term means within 2 seconds.
[00882] Suffering from: An individual who is "suffering from" a disease, disorder, and/or condition has been diagnosed with or displays one or more symptoms of a disease, disorder, and/or condition.
[00883] Susceptible to: An individual who is "susceptible to" a disease, disorder, and/or condition has not been diagnosed with and/or may not exhibit symptoms of the disease, disorder, and/or condition but harbors a propensity to develop a disease or its symptoms. In some embodiments, an individual who is susceptible to a disease, disorder, and/or condition (for example, cancer) may be characterized by one or more of the following: (1) a genetic mutation associated with development of the disease, disorder, and/or condition; (2) a genetic polymorphism associated with development of the disease, disorder, and/or condition; (3) increased and/or decreased expression and/or activity of a protein and/or nucleic acid associated with the disease, disorder, and/or condition; (4) habits and/or lifestyles associated with development of the disease, disorder, and/or condition; (5) a family history of the disease, disorder, and/or condition; and (6) exposure to and/or infection with a microbe associated with development of the disease, disorder, and/or condition. In some embodiments, an individual who is susceptible to a disease, disorder, and/or condition will develop the disease, disorder, and/or condition. In some embodiments, an individual who is susceptible to a disease, disorder, and/or condition will not develop the disease, disorder, and/or condition.
[00884] Sustained release: As used herein, the term "sustained release" refers to a pharmaceutical composition or compound release profile that conforms to a release rate over a specific period of time.
[00885] Synthetic: The term "synthetic" means produced, prepared, and/or manufactured by the hand of man. Synthesis of polynucleotides or polypeptides or other molecules of the present invention may be chemical or enzymatic.
[00886] Targeted Cells: As used herein, "targeted cells" refers to any one or more cells of interest. The cells may be found in vitro, in vivo, in situ or in the tissue or organ of an organism. The organism may be an animal, preferably a mammal, more preferably a human and most preferably a patient.
[00887] Therapeutic Agent: The term "therapeutic agent" refers to any agent that, when
administered to a subject, has a therapeutic, diagnostic, and/or prophylactic effect and/or elicits a desired biological and/or pharmacological effect.
[00888] Therapeutically effective amount: As used herein, the term "therapeutically effective amount" means an amount of an agent to be delivered {e.g., nucleic acid, drug, therapeutic agent, diagnostic agent, prophylactic agent, etc.) that is sufficient, when administered to a subject suffering from or susceptible to an infection, disease, disorder, and/or condition, to treat, improve symptoms of, diagnose, prevent, and/or delay the onset of the infection, disease, disorder, and/or condition. [00889] Therapeutically effective outcome: As used herein, the term "therapeutically effective outcome" means an outcome that is sufficient in a subject suffering from or susceptible to an infection, disease, disorder, and/or condition, to treat, improve symptoms of, diagnose, prevent, and/or delay the onset of the infection, disease, disorder, and/or condition.
[00890] Total daily dose: As used herein, a "total daily dose" is an amount given or prescribed in 24 hr period. It may be administered as a single unit dose.
[00891] Transcription factor: As used herein, the term "transcription factor" refers to a DNA- binding protein that regulates transcription of DNA into R A, for example, by activation or repression of transcription. Some transcription factors effect regulation of transcription alone, while others act in concert with other proteins. Some transcription factor can both activate and repress transcription under certain conditions. In general, transcription factors bind a specific target sequence or sequences highly similar to a specific consensus sequence in a regulatory region of a target gene. Transcription factors may regulate transcription of a target gene alone or in a complex with other molecules.
[00892] Treating: As used herein, the term "treating" refers to partially or completely alleviating, ameliorating, improving, relieving, delaying onset of, inhibiting progression of, reducing severity of, and/or reducing incidence of one or more symptoms or features of a particular infection, disease, disorder, and/or condition. For example, "treating" cancer may refer to inhibiting survival, growth, and/or spread of a tumor. Treatment may be administered to a subject who does not exhibit signs of a disease, disorder, and/or condition and/or to a subject who exhibits only early signs of a disease, disorder, and/or condition for the purpose of decreasing the risk of developing pathology associated with the disease, disorder, and/or condition.
[00893] Unmodified: As used herein, "unmodified" refers to any substance, compound or molecule prior to being changed in any way. Unmodified may, but does not always, refer to the wild type or native form of a biomolecule. Molecules may undergo a series of modifications whereby each modified molecule may serve as the "unmodified" starting molecule for a subsequent modification.
Equivalents and Scope
[00894] Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments in accordance with the invention described herein. The scope of the present invention is not intended to be limited to the above Description, but rather is as set forth in the appended claims. [00895] In the claims, articles such as "a," "an," and "the" may mean one or more than one unless indicated to the contrary or otherwise evident from the context. Claims or descriptions that include "or" between one or more members of a group are considered satisfied if one, more than one, or all of the group members are present in, employed in, or otherwise relevant to a given product or process unless indicated to the contrary or otherwise evident from the context. The invention includes embodiments in which exactly one member of the group is present in, employed in, or otherwise relevant to a given product or process. The invention includes embodiments in which more than one, or all of the group members are present in, employed in, or otherwise relevant to a given product or process.
[00896] It is also noted that the term "comprising" is intended to be open and permits the inclusion of additional elements or steps.
[00897] Where ranges are given, endpoints are included. Furthermore, it is to be understood that unless otherwise indicated or otherwise evident from the context and understanding of one of ordinary skill in the art, values that are expressed as ranges can assume any specific value or subrange within the stated ranges in different embodiments of the invention, to the tenth of the unit of the lower limit of the range, unless the context clearly dictates otherwise.
[00898] In addition, it is to be understood that any particular embodiment of the present invention that falls within the prior art may be explicitly excluded from any one or more of the claims. Since such embodiments are deemed to be known to one of ordinary skill in the art, they may be excluded even if the exclusion is not set forth explicitly herein. Any particular embodiment of the
compositions of the invention (e.g., any nucleic acid or protein encoded thereby; any method of production; any method of use; etc.) can be excluded from any one or more claims, for any reason, whether or not related to the existence of prior art.
[00899] All cited sources, for example, references, publications, databases, database entries, and art cited herein, are incorporated into this application by reference, even if not expressly stated in the citation. In case of conflicting statements of a cited source and the instant application, the statement in the instant application shall control.
[00900] Section and table headings are not intended to be limiting.
EXAMPLES
[00901] The invention is further described in the following examples, which do not limit the scope of the invention described in the claims. Example 1. Modified mRNA Production
[00902] Modified mRNAs (mmRNA) according to the invention may be made using standard laboratory methods and materials. The open reading frame (ORF) of the gene of interest may be flanked by a 5' untranslated region (UTR) which may contain a strong Kozak translational initiation signal and/or an alpha-globin 3' UTR which may include an oligo(dT) sequence for templated addition of a poly-A tail. The modified mRNAs may be modified to reduce the cellular innate immune response. The modifications to reduce the cellular response may include pseudouridine (ψ) and 5-methyl-cytidine (5meC or m5C). (see, Kariko K et al. Immunity 23: 165-75 (2005), Kariko K et al. Mol Ther 16: 1833-40 (2008), Anderson BR et al. NAR (2010); each of which are herein incorporated by reference in their entireties).
[00903] The ORF may also include various upstream or downstream additions (such as, but not limited to, β-globin, tags, etc.) may be ordered from an optimization service such as, but limited to, DNA2.0 (Menlo Park, CA) and may contain multiple cloning sites which may have Xbal recognition. Upon receipt of the plasmid DNA, it may be reconstituted and transformed into chemically competent E. coli.
[00904] For the present invention, NEB DH5 -alpha Competent E. coli are used. Transformations are performed according to NEB instructions using 100 ng of plasmid. The protocol is as follows:
1. Thaw a tube of NEB 5-alpha Competent E. coli cells on ice for 10 minutes.
2. Add 1-5 μΐ containing 1 pg-100 ng of plasmid DNA to the cell mixture. Carefully flick the tube 4-5 times to mix cells and DNA. Do not vortex.
3. Place the mixture on ice for 30 minutes. Do not mix.
4. Heat shock at 42°C for exactly 30 seconds. Do not mix.
5. Place on ice for 5 minutes. Do not mix.
6. Pipette 950 μΐ of room temperature SOC into the mixture.
7. Place at 37°C for 60 minutes. Shake vigorously (250 rpm) or rotate.
8. Warm selection plates to 37°C.
9. Mix the cells thoroughly by flicking the tube and inverting.
[00905] Spread 50-100 μΐ of each dilution onto a selection plate and incubate overnight at 37°C. Alternatively, incubate at 30°C for 24-36 hours or 25°C for 48 hours.
[00906] A single colony is then used to inoculate 5 ml of LB growth media using the appropriate antibiotic and then allowed to grow (250 RPM, 37° C) for 5 hours. This is then used to inoculate a 200 ml culture medium and allowed to grow overnight under the same conditions. [00907] To isolate the plasmid (up to 850 μg), a maxi prep is performed using the Invitrogen PURELINK™ HiPure Maxiprep Kit (Carlsbad, CA), following the manufacturer's instructions.
[00908] In order to generate cDNA for In Vitro Transcription (IVT), the plasmid (an Example of which is shown in Figure 2) is first linearized using a restriction enzyme such as Xbal. A typical restriction digest with Xbal will comprise the following: Plasmid 1.0 μg; lOx Buffer 1.0 μΐ; Xbal 1.5 μΐ; dH20 up to 10 μΐ; incubated at 37° C for 1 hr. If performing at lab scale (< 5μg), the reaction is cleaned up using Invitrogen's PURELINK™ PCR Micro Kit (Carlsbad, CA) per manufacturer's instructions. Larger scale purifications may need to be done with a product that has a larger load capacity such as Invitrogen's standard PURELINK™ PCR Kit (Carlsbad, CA). Following the cleanup, the linearized vector is quantified using the NanoDrop and analyzed to confirm
linearization using agarose gel electrophoresis.
[00909] The methods described herein to make modified mRNA may be used to produce molecules of all sizes including long molecules. Modified mRNA using the described methods has been made for different sized molecules including glucosidase, alpha; acid (GAA) (3.2 kb), cystic fibrosis transmembrane conductance regulator (CFTR) (4.7 kb), Factor VII (7.3 kb), lysosomal acid lipase (45.4kDa), glucocerebrosidase (59.7 kDa) and iduronate 2-sulfatase (76 kDa).
[00910] As a non-limiting example, G-CSF may represent the polypeptide of interest. Sequences used in the steps outlined in Examples 1-5 are shown in Table 4. It should be noted that the start codon (ATG) has been underlined in each sequence of Table 4.
Table 4. G-CSF Sequences
Figure imgf000263_0001
cDNA having T7 polymerase site, Afel and Xba restriction site:
TAATACGACTCACTATA
GGGAAATAAGAGAGAAAAGAAGAGTAAGAAGAAATATAAGAGCCACC
ATGGCTGGACCTGCCACCCAGAGCCCCATGAAGCTGATGGCCCTGCAG
CTGCTGCTGTGGCACAGTGCACTCTGGACAGTGCAGGAAGCCACCCCC
CTGGGCCCTGCCAGCTCCCTGCCCCAGAGCTTCCTGCTCAAGTGCTTAG
AGC AAGTG AGG AAG ATC C AGGGC G ATGGCGC AGC GCTC C AGG AG AAG
CTGTGTGCCACCTACAAGCTGTGCCACCCCGAGGAGCTGGTGCTGCTC
GGACACTCTCTGGGCATCCCCTGGGCTCCCCTGAGCAGCTGCCCCAGCC
AGGCCCTGCAGCTGGCAGGCTGCTTGAGCCAACTCCATAGCGGCCTTTT
CCTCTACCAGGGGCTCCTGCAGGCCCTGGAAGGGATCTCCCCCGAGTT
GGGTCCCACCTTGGACACACTGCAGCTGGACGTCGCCGACTTTGCCAC
CACCATCTGGCAGCAGATGGAAGAACTGGGAATGGCCCCTGCCCTGCA
GCCCACCCAGGGTGCCATGCCGGCCTTCGCCTCTGCTTTCCAGCGCCGG
GCAGGAGGGGTCCTGGTTGCCTCCCATCTGCAGAGCTTCCTGGAGGTG
TCGTACCGCGTTCTACGCCACCTTGCCCAGCCCTGA
AGCGCTGCCTTCTGCGGGGCTTGCCTTCTGGCCATGCCCTTCTTCTCTCC
CTTGCACCTGTACCTCTTGGTCTTTGAATAAAGCCTGAGTAGGAAGGCG
GCCGCTCGAGCATGCATCTAGA
Optimized sequence; containing T7 polymerase site, Afel and Xba restriction site TAATACGACTCACTATA
GGGAAATAAGAGAGAAAAGAAGAGTAAGAAGAAATATAAGAGCCACC
ATGGCCGGTCCCGCGACCCAAAGCCCCATGAAACTTATGGCCCTGCAG
TTGCTGCTTTGGCACTCGGCCCTCTGGACAGTCCAAGAAGCGACTCCTC
TCGGACCTGCCTCATCGTTGCCGCAGTCATTCCTTTTGAAGTGTCTGGA
GCAGGTGCGAAAGATTCAGGGCGATGGAGCCGCACTCCAAGAGAAGC
TCTGCGCGACATACAAACTTTGCCATCCCGAGGAGCTCGTACTGCTCGG
GCACAGCTTGGGGATTCCCTGGGCTCCTCTCTCGTCCTGTCCGTCGCAG
GCTTTGCAGTTGGCAGGGTGCCTTTCCCAGCTCCACTCCGGTTTGTTCTT
GTATCAGGGACTGCTGCAAGCCCTTGAGGGAATCTCGCCAGAATTGGG
CCCGACGCTGGACACGTTGCAGCTCGACGTGGCGGATTTCGCAACAAC
CATCTGGCAGCAGATGGAGGAACTGGGGATGGCACCCGCGCTGCAGCC
CACGCAGGGGGCAATGCCGGCCTTTGCGTCCGCGTTTCAGCGCAGGGC
GGGTGGAGTCCTCGTAGCGAGCCACCTTCAATCATTTTTGGAAGTCTCG
TACCGGGTGCTGAGACATCTTGCGCAGCCGTGA
AGCGCTGCCTTCTGCGGGGCTTGCCTTCTGGCCATGCCCTTCTTCTCTCC CTTGCACCTGTACCTCTTGGTCTTTGAATAAAGCCTGAGTAGGAAGGCG GCCGCTCGAGCATGCATCTAGA mR A sequence (transcribed)
GGGAAAUAAGAGAGAAAAGAAGAGUAAGAAGAAAUAUAAGAGCCAC C
AUGGCCGGUCCCGCGACCCAAAGCCCCAUGAAACUUAUGGCCCUGCA GUUGCUGCUUUGGCACUCGGCCCUCUGGACAGUCCAAGAAGCGACUC
CUCUCGGACCUGCCUCAUCGUUGCCGCAGUCAUUCCUUUUGAAGUGU
CUGG AGC AGGUGC G AAAG AUUC AGGGC G AUGG AGC CGC ACUC C AAG
AGAAGCUCUGCGCGACAUACAAACUUUGCCAUCCCGAGGAGCUCGUA
CUGCUCGGGCACAGCUUGGGGAUUCCCUGGGCUCCUCUCUCGUCCUG
UCCGUCGCAGGCUUUGCAGUUGGCAGGGUGCCUUUCCCAGCUCCACU
CCGGUUUGUUCUUGUAUCAGGGACUGCUGCAAGCCCUUGAGGGAAU
CUC GCC AG AAUUGGGCC CG AC GCUGG AC AC GUUGC AGCUCG AC GUGG
CGGAUUUCGCAACAACCAUCUGGCAGCAGAUGGAGGAACUGGGGAU
GGCACCCGCGCUGCAGCCCACGCAGGGGGCAAUGCCGGCCUUUGCGU
CCGCGUUUCAGCGCAGGGCGGGUGGAGUCCUCGUAGCGAGCCACCUU
CAAUCAUUUUUGGAAGUCUCGUACCGGGUGCUGAGACAUCUUGCGC
AGCCGUGA
AGCGCUGCCUUCUGCGGGGCUUGCCUUCUGGCCAUGCCCUUCUUCUC
UCCCUUGCACCUGUACCUCUUGGUCUUUGAAUAAAGCCUGAGUAGGA
AG
Example 2: PCR for cDNA Production
[00911] PCR procedures for the preparation of cDNA are performed using 2x KAPA HIFI™ HotStart Ready Mix by Kapa Biosystems (Woburn, MA). This system includes 2x KAPA
ReadyMixl2.5 μΐ; Forward Primer (10 uM) 0.75 μΐ; Reverse Primer (10 uM) 0.75 μΐ; Template cDNA 100 ng; and dH20 diluted to 25.0 μΐ. The reaction conditions are at 95° C for 5 min. and 25 cycles of 98° C for 20 sec, then 58° C for 15 sec, then 72° C for 45 sec, then 72° C for 5 min. then 4° C to termination.
[00912] The reverse primer of the instant invention incorporates a poly-Ti2o for a poly-Ai2o in the mRNA. Other reverse primers with longer or shorter poly(T) tracts can be used to adjust the length of the poly(A) tail in the mRNA.
[00913] The reaction is cleaned up using Invitrogen's PURELINK™ PCR Micro Kit (Carlsbad, CA) per manufacturer's instructions (up to 5 μg). Larger reactions will require a cleanup using a product with a larger capacity. Following the cleanup, the cDNA is quantified using the
NANODROP™ and analyzed by agarose gel electrophoresis to confirm the cDNA is the expected size. The cDNA is then submitted for sequencing analysis before proceeding to the in vitro transcription reaction.
Example 3. In vitro Transcription [00914] The in vitro transcription reaction generates mRNA containing modified nucleotides or modified RNA. The input nucleotide triphosphate (NTP) mix is made in-house using natural and unnatural NTPs.
[00915] A typical in vitro transcription reaction includes the following:
1. Template cDNA 1.0 μ
2. lOx transcription buffer (400 mM Tris-HCl pH 8.0, 190 mM MgCl2, 50 mM DTT, 10 mM Spermidine) 2.0 μΐ
3. Custom NTPs (25mM each) 7.2 μΐ
4. RNase Inhibitor 20 U
5. T7 RNA polymerase 3000 U
6. d¾0 Up to 20.0 μΐ. and
7. Incubation at 37° C for 3 hr-5 hrs.
[00916] The crude IVT mix may be stored at 4° C overnight for cleanup the next day. 1 U of RNase-free DNase is then used to digest the original template. After 15 minutes of incubation at 37° C, the mRNA is purified using Ambion's MEGACLEAR™ Kit (Austin, TX) following the manufacturer's instructions. This kit can purify up to 500 μg of RNA. Following the cleanup, the RNA is quantified using the NanoDrop and analyzed by agarose gel electrophoresis to confirm the RNA is the proper size and that no degradation of the RNA has occurred.
Example 4. Enzymatic Capping of mRNA
[00917] Capping of the mRNA is performed as follows where the mixture includes: IVT RNA 60 μg-180μg and dH20 up to 72 μΐ. The mixture is incubated at 65° C for 5 minutes to denature RNA, and then is transferred immediately to ice.
[00918] The protocol then involves the mixing of lOx Capping Buffer (0.5 M Tris-HCl (pH 8.0), 60 mM KCl, 12.5 mM MgCl2) (10.0 μΐ); 20 mM GTP (5.0 μΐ); 20 mM S-Adenosyl Methionine (2.5 μΐ); RNase Inhibitor (100 U); 2'-0-Methyltransferase (400U); Vaccinia capping enzyme (Guanylyl transferase) (40 U); dH20 (Up to 28 μΐ); and incubation at 37° C for 30 minutes for 60 μg RNA or up to 2 hours for 180 μg of RNA.
[00919] The mRNA is then purified using Ambion's MEGACLEAR™ Kit (Austin, TX) following the manufacturer's instructions. Following the cleanup, the RNA is quantified using the
NANODROP™ (ThermoFisher, Waltham, MA) and analyzed by agarose gel electrophoresis to confirm the RNA is the proper size and that no degradation of the RNA has occurred. The RNA product may also be sequenced by running a reverse-transcription-PCR to generate the cDNA for sequencing.
Example 5. PolyA Tailing Reaction
[00920] Without a poly-T in the cDNA, a poly-A tailing reaction must be performed before cleaning the final product. This is done by mixing Capped IVT RNA (100 μΐ); RNase Inhibitor (20 U); lOx Tailing Buffer (0.5 M Tris-HCl (pH 8.0), 2.5 M NaCl, 100 mM MgCl2)(12.0 μΐ); 20 mM ATP (6.0 μΐ); Poly-A Polymerase (20 U); dH20 up to 123.5 μΐ and incubation at 37° C for 30 min. If the poly-A tail is already in the transcript, then the tailing reaction may be skipped and proceed directly to cleanup with Ambion's MEGACLEAR™ kit (Austin, TX) (up to 500 μg). Poly- A Polymerase is preferably a recombinant enzyme expressed in yeast.
[00921] For studies performed and described herein, the poly-A tail is encoded in the IVT template to comprise 160 nucleotides in length. However, it should be understood that the processivity or integrity of the polyA tailing reaction may not always result in exactly 160 nucleotides. Hence polyA tails of approximately 160 nucleotides, e.g, about 150-165, 155, 156, 157, 158, 159, 160, 161, 162, 163, 164 or 165 are within the scope of the invention.
Example 6. Natural 5f Caps and 5f Cap Analogues
[00922] 5 '-capping of modified RNA may be completed concomitantly during the in vitro- transcription reaction using the following chemical RNA cap analogs to generate the 5'-guanosine cap structure according to manufacturer protocols: 3 '-0-Me-m7G(5')ppp(5') G [the ARC A cap];G(5*)ppp(5*)A; G(5*)ppp(5*)G; m7G(5*)ppp(5*)A; m7G(5*)ppp(5*)G (New England BioLabs, Ipswich, MA). 5 '-capping of modified RNA may be completed post-transcriptionally using a Vaccinia Virus Capping Enzyme to generate the "Cap 0" structure: m7G(5')ppp(5')G (New England BioLabs, Ipswich, MA). Cap 1 structure may be generated using both Vaccinia Virus Capping Enzyme and a 2'-0 methyl-transferase to generate: m7G(5')ppp(5')G-2'-0-methyl. Cap 2 structure may be generated from the Cap 1 structure followed by the 2'-0-methylation of the 5'- antepenultimate nucleotide using a 2'-0 methyl-transferase. Cap 3 structure may be generated from the Cap 2 structure followed by the 2'-0-methylation of the 5'-preantepenultimate nucleotide using a 2'-0 methyl-transferase. Enzymes are preferably derived from a recombinant source.
[00923] When transfected into mammalian cells, the modified mRNAs have a stability of between 12-18 hours or more than 18 hours, e.g., 24, 36, 48, 60, 72 or greater than 72 hours.
Example 7. Capping A. Protein Expression Assay
[00924] Synthetic mRNAs encoding human G-CSF (cDNA shown in SEQ ID NO: 5; mRNA sequence fully modified with 5-methylcytosine at each cytosine and pseudouridine replacement at each uridine site shown in SEQ ID NO: 6 with a polyA tail approximately 160 nucletodies in length not shown in sequence) containing the ARCA (3' 0-Me-m7G(5')ppp(5')G) cap analog or the Capl structure can be transfected into human primary keratinocytes at equal concentrations. 6, 12, 24 and 36 hours post-transfection the amount of G-CSF secreted into the culture medium can be assayed by ELISA. Synthetic mRNAs that secrete higher levels of G-CSF into the medium would correspond to a synthetic mRNA with a higher translationally-competent Cap structure.
B. Purity Analysis Synthesis
[00925] Synthetic mRNAs encoding human G-CSF (cDNA shown in SEQ ID NO: 5; mRNA sequence fully modified with 5-methylcytosine at each cytosine and pseudouridine replacement at each uridine site shown in SEQ ID NO: 6 with a polyA tail approximately 160 nucletodies in length not shown in sequence) containing the ARCA cap analog or the Capl structure crude synthesis products can be compared for purity using denaturing Agarose-Urea gel electrophoresis or HPLC analysis. Synthetic mRNAs with a single, consolidated band by electrophoresis correspond to the higher purity product compared to a synthetic mRNA with multiple bands or streaking bands.
Synthetic mRNAs with a single HPLC peak would also correspond to a higher purity product. The capping reaction with a higher efficiency would provide a more pure mRNA population.
C. Cytokine Analysis
[00926] Synthetic mRNAs encoding human G-CSF (cDNA shown in SEQ ID NO: 5; mRNA sequence fully modified with 5-methylcytosine at each cytosine and pseudouridine replacement at each uridine site shown in SEQ ID NO: 6 with a polyA tail approximately 160 nucletodies in length not shown in sequence) containing the ARCA cap analog or the Capl structure can be transfected into human primary keratinocytes at multiple concentrations. 6, 12, 24 and 36 hours post- transfection the amount of pro-inflammatory cytokines such as TNF-alpha and IFN-beta secreted into the culture medium can be assayed by ELISA. Synthetic mRNAs that secrete higher levels of pro-inflammatory cytokines into the medium would correspond to a synthetic mRNA containing an immune-activating cap structure.
D. Capping Reaction Efficiency [00927] Synthetic mR As encoding human G-CSF (cDNA shown in SEQ ID NO: 5; mRNA sequence fully modified with 5-methylcytosine at each cytosine and pseudouridine replacement at each uridine site shown in SEQ ID NO: 6 with a polyA tail approximately 160 nucletodies in length not shown in sequence) containing the ARCA cap analog or the Capl structure can be analyzed for capping reaction efficiency by LC-MS after capped mRNA nuclease treatment. Nuclease treatment of capped mRNAs would yield a mixture of free nucleotides and the capped 5 '-5 -triphosphate cap structure detectable by LC-MS. The amount of capped product on the LC-MS spectra can be expressed as a percent of total mRNA from the reaction and would correspond to capping reaction efficiency. The cap structure with higher capping reaction efficiency would have a higher amount of capped product by LC-MS.
Example 8. Agarose Gel Electrophoresis of Modified RNA or RT PCR Products
[00928] Individual modified RNAs (200-400 ng in a 20 μΐ volume) or reverse transcribed PCR products (200-400 ng) are loaded into a well on a non-denaturing 1.2% Agarose E-Gel (Invitrogen,
Carlsbad, CA) and run for 12-15 minutes according to the manufacturer protocol.
Example 9. Formulation of Modified mRNA Using Lipidoids
[00929] Modified mRNAs (mmRNA) are formulated for in vitro experiments by mixing the mmRNA with the lipidoid at a set ratio prior to addition to cells. In vivo formulation may require the addition of extra ingredients to facilitate circulation throughout the body. To test the ability of these lipidoids to form particles suitable for in vivo work, a standard formulation process used for siRNA- lipidoid formulations was used as a starting point. Initial mmRNA-lipidoid formulations may consist of particles composed of 42% lipidoid, 48% cholesterol and 10%> PEG, with further optimization of ratios possible. After formation of the particle, mmRNA is added and allowed to integrate with the complex. The encapsulation efficiency is determined using a standard dye exclusion assays.
Materials and Methods for Examples 10-14
A. Lipid Synthesis
[00930] Six lipids, DLin-DMA, DLin-K-DMA, DLin-KC2-DMA, 98N12-5, CI 2-200 and DLin- MC3-DMA, were synthesized by methods outlined in the art in order to be formulated with modified RNA. DLin-DMA and precursorswere synthesized as described in Heyes et. al, J. Control Release, 2005, 107, 276-287. DLin-K-DMA and DLin-KC2-DMA and precursors were synthesized as described in Semple et. al, Nature Biotechnology, 2010, 28, 172-176. 98N12-5 and precursor were synthesized as described in Akinc et. al, Nature Biotechnology, 2008, 26, 561-569.
[00931] CI 2-200 and precursors were synthesized according to the method outlined in Love et. al, PNAS, 2010, 107, 1864-1869. 2-epoxydodecane (5.10 g, 27.7 mmol, 8.2 eq) was added to a vial containing Amine 200 (0.723 g, 3.36 mmol, 1 eq) and a stirring bar. The vial was sealed and warmed to 80°C. The reaction was stirred for 4 days at 80°C. Then the mixture was purified by silica gel chromatography using a gradient from pure dichloromethane (DCM) to DCM:MeOH 98:2. The target compound was further purified by RP-HPLC to afford the desired compound.
[00932] DLin-MC3-DMA and precursors were synthesized according to procedures described in WO 2010054401 herein incorporated by reference in its entirety. A mixture of dilinoleyl methanol (1.5 g, 2.8 mmol, 1 eq), Ν,Ν-dimethylaminobutyric acid (1.5 g, 2.8 mmol, leq), DIPEA (0.73 mL, 4.2 mmol, 1.5 eq) and TBTU(1.35 g, 4.2 mmol, 1.5 eq) in 10 mL of DMF was stirred for 10 h at room temperature. Then the reaction mixture was diluted in ether and washed with water. The organic layer was dried over anhydrous sodium sulfate, filtrated and concentrated under reduced pressure. The crude product was purified by silica gel chromatography using a gradient DCM to DCM:MeOH 98:2. Subsequently the target compound was subjected to an additional RP-HPLC purification which was done using a YMC - Pack C4 column to afford the target compound.
B. Formulation of Modified RNA Nanoparticles
[00933] Solutions of synthesized lipid, 1 ,2-distearoyl-3 -phosphatidylcholine (DSPC) (Avanti Polar Lipids, Alabaster, AL), cholesterol (Sigma- Aldrich, Taufkirchen, Germany), and a-[3'-(l,2- dimyristoyl-3-propanoxy)-carboxamide-propyl]-ro-methoxy-polyoxy ethylene (PEG-c-DOMG) (NOF, Bouwelven, Belgium) were prepared at concentrations of 50 mM in ethanol and stored at - 20°C. The lipids were combined to yield molar ratio of 50: 10:38.5: 1.5 (Lipid: DSPC: Cholesterol: PEG-c-DOMG) and diluted with ethanol to a final lipid concentration of 25 mM. Solutions of modified mRNA at a concentration of 1-2 mg/mL in water were diluted in 50 mM sodium citrate buffer at a pH of 3 to form a stock modified mRNA solution. Formulations of the lipid and modified mRNA were prepared by combining the synthesized lipid solution with the modified mRNA solution at total lipid to modified mRNA weight ratio of 10: 1, 15: 1, 20: 1 and 30: 1. The lipid ethanolic solution was rapidly injected into aqueous modified mRNA solution to afford a suspension containing 33% ethanol. The solutions were injected either manually (MI) or by the aid of a syringe pump (SP) (Harvard Pump 33 Dual Syringe Pump Harvard Apparatus Holliston, MA). [00934] To remove the ethanol and to achieve the buffer exhange, the formulations were dialyzed twice against phosphate buffered saline (PBS), pH 7.4 at volumes 200-times of the primary product using a Slide- A-Lyzer cassettes (Thermo Fisher Scientific Inc. Rockford, IL) with a molecular weight cutoff (MWCO) of 10 kD. The first dialysis was carried at room temperature for 3 hours and then the formulations were dialyzed overnight at 4°C. The resulting nanoparticle suspension was filtered through 0.2 μιη sterile filter (Sarstedt, Numbrecht, Germany) into glass vials and sealed with a crimp closure.
C. Characterization of formulations
[00935] A Zetasizer Nano ZS (Malvern Instruments Ltd, Malvern, Worcestershire, UK) was used to determine the particle size, the polydispersity index (PDI) and the zeta potential of the modified mRNA nanoparticles in IX PBS in determining particle size and 15 mM PBS in determining zeta potential.
[00936] Ultraviolet-visible spectroscopy was used to determine the concentration of modified mRNA nanoparticle formulation. 100 of the diluted formulation in IX PBS was added to 900 of a 4: 1 (v/v) mixture of methanol and chloroform. After mixing, the absorbance spectrum of the solution was recorded between 230 nm and 330 nm on a DU 800 spectrophotometer (Beckman Coulter, Beckman Coulter, Inc., Brea, CA). The modified RNA concentration in the nanoparicle formulation was calculated based on the extinction coefficient of the modified RNA used in the formulation and on the difference between the absorbance at a wavelength of 260 nm and the baseline value at a wavelength of 330 nm.
[00937] QUANT-IT™ RIBOGREEN® RNA assay (Invitrogen Corporation Carlsbad, CA) was used to evaluate the encapsulation of modified RNA by the nanoparticle. The samples were diluted to a concentration of approximately 5 μg/mL in TE buffer (10 mM Tris-HCl, 1 mM EDTA, pH 7.5). 50 μί of the diluted samples were transferred to a polystyrene 96 well plate, then either 50 μΐ, of TE buffer or 50 μί of a 2% Triton X-100 solution was added. The plate was incubated at a temperature of 37°C for 15 minutes. The RIBOGREEN® reagent was diluted 1 : 100 in TE buffer, 100 μΕ of this solution was added to each well. The fluorescence intensity was measured using a fluorescence plate reader (Wallac Victor 1420 Multilablel Counter; Perkin Elmer, Waltham, MA) at an excitation wavelength of -480 nm and an emission wavelength of -520 nm. The fluorescence values of the reagent blank were subtracted from that of each of the samples and the percentage of free modified RNA was determined by dividing the fluorescence intensity of the intact sample (without addition of Triton X-100) by the fluorescence value of the disrupted sample (caused by the addition of Triton X- 100).
D. In Vitro Incubation
[00938] Human embryonic kidney epithelial (HEK293) and hepatocellular carcinoma epithelial (HepG2) cells (LGC standards GmbH, Wesel, Germany) were seeded on 96-well plates (Greiner Bio-one GmbH, Frickenhausen, Germany) and plates for HEK293 cells were precoated with collagen typel . HEK293 were seeded at a density of 30,000 and HepG2 were seeded at a density of 35,000 cells per well in 100 μΐ cell culture medium. For HEK293 the cell culture medium was DMEM, 10% FCS, adding 2mM L-Glutamine, 1 mM Sodiumpyruvate and lx non-essential amino acids (Biochrom AG, Berlin, Germany) and 1.2 mg/ml Sodiumbicarbonate (Sigma- Aldrich, Munich, Germany) and for HepG2 the culture medium was MEM (Gibco Life Technologies, Darmstadt, Germany), 10% FCS adding 2mM L-Glutamine, 1 mM Sodiumpyruvate and lx non-essential amino acids (Biochrom AG, Berlin, Germany. Formulations containing mCherry mRNA (mRNA sequence shown in SEQ ID NO: 7; polyA tail of approximately 160 nucleotides not shown in sequence; 5 'cap, Capl); were added in quadruplicates directly after seeding the cells and incubated. The mCherry cDNA with the T7 promoter, 5 'untranslated region (UTR) and 3' UTR used in in vitro transcription (IVT) is given in SEQ ID NO: 8. The mCherry mRNA was modified with 5meC at each cytosine and pseudouridine replacement at each uridine site.
[00939] Cells were harvested by transferring the culture media supernatants to a 96-well Pro-Bind U-bottom plate (Beckton Dickinson GmbH, Heidelberg, Germany). Cells were trypsinized with ½ volume Trypsin/EDTA (Biochrom AG, Berlin, Germany), pooled with respective supernatants and fixed by adding one volume PBS/2%>FCS (both Biochrom AG, Berlin, Germany )/0.5 %
formaldehyde (Merck, Darmstadt, Germany). Samples then were submitted to a flow cytometer measurement with a 532nm excitation laser and the 610/20 filter for PE-Texas Red in a LSRII cytometer (Beckton Dickinson GmbH, Heidelberg, Germany). The mean fluorescence intensity (MFI) of all events and the standard deviation of four independent wells are presented in for samples analyzed.
Example 10. Purification of Nanoparticle Formulations
[00940] Nanoparticle formulations of DLin-KC2-DMA and 98N12-5 in HEK293 and HepG2 were tested to determine if the mean fluorescent intensity (MFI) was dependent on the lipid to modified RNA ratio and/or purification. Three formulations of DLin-KC2-DMA and two formulations of 98N12-5 were produced using a syringe pump to the specifications described in Table 5. Purified samples were purified by SEPHADEX™ G-25 DNA grade (GE Healthcare, Sweden). Each formulation before and after purification (aP) was tested at concentration of 250 ng modified R A per well in a 24 well plate. The percentage of cells that are positive for the marker for FL4 channel (%FL4-positive) when analyzed by the flow cytometer for each formulation and the background sample and the MFI of the marker for the FL4 channel for each formulation and the background sample are shown in Table 6. The formulations which had been purified had a slightly higher MFI than those formulations tested before purification.
Table 5. Formulations
Figure imgf000273_0001
Table 6. HEK293 and HepG2, 24-well, 250 ng Modified RNA/well
Figure imgf000273_0002
Example 11. Concentration Response Curve
[00941] Nanoparticle formulations of 98N12-5 (NPA-005) and DLin-KC2-DMA (NPA-003) were tested at varying concentrations to determine the MFI of FL4 or mCherry (mRNA sequence shown in SEQ ID NO: 7; polyA tail of approximately 160 nucleotides not shown in sequence; 5 'cap, Capl; fully modified with 5-methylcytosine and pseudouridine) over a range of doses. The formulations tested are outlined in Table 7. To determine the optimal concentration of nanoparticle formulations of 98N12-5, varying concentrations of formulated modified RNA (100 ng, 10 ng, 1.0 ng, 0.1 ng and 0.01 ng per well) were tested in a 24-well plate of HEK293, and the results of the FL4 MFI of each dose are shown in Table 8. Likewise, to determine the optimal concentration of nanoparticle formulations of DLin-KC2-DMA, varying concentrations of formulated modified RNA (250 ng 100 ng, 10 ng, 1.0 ng, 0.1 ng and 0.01 ng per well) were tested in a 24-well plate of HEK293, and the results of the FL4 MFI of each dose are shown in Table 9. Nanoparticle formulations of DLin-KC2- DMA were also tested at varying concentrations of formulated modified RNA (250 ng, 100 ng and 30 ng per well) in a 24 well plate of HEK293, and the results of the FL4 MFI of each dose are shown in Table 10. A dose of 1 ng/well for 98N12-5 and a dose of 10 ng/well for DLin-KC2-DMA were found to resemble the FL4 MFI of the background.
[00942] To determine how close the concentrations resembled the background, we utilized a flow cytometer with optimized filter sets for detection of mCherry expression, and were able to obtain results with increased sensitivity relative to background levels. Doses of 25 ng/well, 0.25 ng/well, 0.025 ng/well and 0.0025 ng/well were analyzed for 98N12-5 (NPA-005) and DLin-KC2-DMA (NPA-003) to determine the MFI of mCherry. As shown in Table 11, the concentration of 0.025 ng/well and lesser concentrations are similar to the background MFI level of mCherry which is about 386.125.
Table 7. Formulations
Figure imgf000274_0001
Table 8. HEK293, NPA-005, 24-well, n=4
Formulation FL4 MFI
Figure imgf000275_0001
Table 9. HEK293, NPA-003, 24-well, n=4
Figure imgf000275_0002
Table 10. HEK293, NPA-003, 24-well, n=4
Figure imgf000275_0003
Table 11. Concentration and MFI
Figure imgf000275_0004
Example 12. Manual Injection and Syringe Pump Formulations
[00943] Two formulations of DLin-KC2-DMA and 98N12-5 were prepared by manual injection (MI) and syringe pump injection (SP) and analyzed along with a background sample to compare the MFI of mCherry (m NA sequence shown in SEQ ID NO: 7; polyA tail of approximately 160 nucleotides not shown in sequence; 5 'cap, Capl; fully modified with 5-methylcytosine and pseudouridine) of the different formulations. Table 12 shows that the syringe pump formulations had a higher MFI as compared to the manual injection formulations of the same lipid and lipid/RNA ratio.
Table 12. Formulations and MFI Lipid/RNA Mean size Method of
Formulation # Lipid MFI
wt/wt (nm) formulation
Untreated
N/A N/A N/A N/A 674.67
Control
DLin-KC2- 140 nm
NPA-002 15 MI 10318.25
DMA PDI: 0.11
DLin-KC2- 105 nm
NPA-002-2 15 SP 37054.75
DMA PDI: 0.04
DLin-KC2- 114 nm
NPA-003 20 MI 22037.5
DMA PDI: 0.08
DLin-KC2- 95 nm
NPA-003-2 20 SP 37868.75
DMA PDI: 0.02
127 nm
NPA-005 98N12-5 15 MI 11504.75
PDI: 0.12
106 nm
NPA-005-2 98N12-5 15 SP 9343.75
PDI: 0.07
126 nm
NPA-006 98N12-5 20 MI 11182.25
PDI: 0.08
93 nm
NPA-006-2 98N12-5 20 SP 5167
PDI: 0.08
Example 13. LNP Formulations
[00944] Formulations of DLin-DMA, DLin-K-DMA, DLin-KC2-DMA, 98N12-5, CI 2-200 and DLin-MC3-DMA were incubated at a concentration of 60 ng/well or 62.5 ng/well in a plate of HEK293 and 62.5 ng/well in a plate of HepG2 cells for 24 hours to determine the MFI of mCherry (mR A sequence shown in SEQ ID NO: 7; polyA tail of approximately 160 nucleotides not shown in sequence; 5 'cap, Capl; fully modified with 5-methylcytosine and pseudouridine) for each formulation. The formulations tested are outlined in Table 13 below. As shown in Table 14 for the 60 ng/well and Tables 15, 16, 17 and 18 for the 62.5 ng/well,the formulation of NPA-003 and NPA- 018 have the highest mCherry MFI and the formulations of NPA-008, NPA-010 and NPA-013 are most the similar to the background sample mCherry MFI value.
Table 13. Formulations
Figure imgf000276_0001
NPA-005 98N12-5 15 127 nm
PDI: 0.12
NPA-006 98N12-5 20 126 nm
PDI: 0.08
NPA-007 DLin-DMA 15 148 nm
PDI: 0.09
NPA-008 DLin-K-DMA 15 121 nm
PDI: 0.08
NPA-009 C12-200 15 138 nm
PDI: 0.15
NPA-010 DLin-MC3-DMA 15 126 nm
PDI: 0.09
NPA-012 DLin-DMA 20 86 nm
PDI: 0.08
NPA-013 DLin-K-DMA 20 104 nm
PDI: 0.03
NPA-014 C12-200 20 101 nm
PDI: 0.06
NPA-015 DLin-MC3-DMA 20 109 nm
PDI: 0.07
Table 14. HEK293, 96-well, 60 ng Modified RNA well
Figure imgf000277_0001
NPA-012 1 1466
NPA-013 2098.25
NPA-014 3194.25
NPA-015 14524
Table 16. HEK293, 62.5 ng /well
Formulation MFI mCherry
Untreated 295
NPA-007 3504
NPA-012 8286
NPA-017 6128
NPA-003-2 17528
NPA-018 34142
NPA-010 1095
NPA-015 5859
NPA-019 3229
Table 17. HepG2, 62.5 ng /well
Formulation MFI mCherry
Untreated 649.94
NPA-001 6006.25
NPA-002 8705
NPA-002-2 15860.25
NPA-003 15059.25
NPA-003-2 28881
NPA-005 1676
NPA-006 1473
NPA-007 15678
NPA-008 2976.25
NPA-009 961.75
NPA-010 3301.75
NPA-012 18333.25
NPA-013 5853
NPA-014 2257
NPA-015 16225.75
Table 18. HepG2, 62.5 ng /well
Formulation MFI mCherry
Untreated control 656
NPA-007 16798
NPA-012 21993
NPA-017 20377
NPA-003-2 35651
NPA-018 40154
NPA-010 2496
NPA-015 19741
NPA-019 16373 Example 14. In vivo formulation studies
[00945] Rodents (n=5) are administered intravenously, subcutaneously or intramuscularly a single dose of a formulation containing at least one modified mRNA and a lipid. The modified mRNA administered to the rodents is selected from G-CSF (mRNA sequence shown in SEQ ID NO: 6; polyA tail of approximately 160 nucleotides not shown in sequence; 5 'cap, Capl), erythropoietin (EPO) (mRNA sequence shown in SEQ ID NO: 9; polyA tail of approximately 160 nucleotides not shown in sequence; 5 'cap, Capl), Factor IX (mRNA shown in SEQ ID NO: 10; polyA tail of approximately 160 nucleotides not shown in sequence; 5'cap, Capl) or mCherry (mRNA sequence shown in SEQ ID NO: 7; polyA tail of approximately 160 nucleotides not shown in sequence; 5'cap, Capl). The erythropoietin cDNA with the T7 promoter, 5 'untranslated region (UTR) and 3' UTR used in in vitro transcription (IVT) is given in SEQ ID NO: 11 and SEQ ID NO: 12.
[00946] Each formulation also contains a lipid which is selected from one of DLin-DMA, DLin-K- DMA, DLin-KC2-DMA, 98N12-5, C 12-200, DLin-MC3 -DM A, reLNP, ATUPLEX®, DACC, and DBTC. The rodents are injected with lOOug, 10 ug or 1 ug of the formulated modified mRNA and samples are collected at specified time intervals.
[00947] Serum from the rodents administered formulations containing human G-CSF modified mRNA are measured by specific G-CSF ELISA and serum from mice administered human factor IX modified RNA is analyzed by specific factor IX ELISA or chromogenic assay. The liver and spleen from the mice administered with mCherry modified mRNA are analyzed by immunohistochemistry (IHC) or fluorescence-activated cell sorting (FACS). As a control, a group of mice are not injected with any formulation and their serum and tissue are collected analyzed by ELISA, FACS and/or IHC.
A. Time Course
[00948] The rodents are administered formulations containing at least one modified mRNA to study the time course of protein expression for the administered formulation. The rodents are bled at specified time intervals prior to and after administration of the modified mRNA formulations to determine protein expression and complete blood count. Samples are also collected from the site of administration of rodents administered modified mRNA formulations subcutaneously and intramuscularly to determine the protein expression in the tissue.
B. Dose Response [00949] The rodents are administered formulations containing at least one modified mRNA to determine dose response of each formulation. The rodents are bled at specified time intervals prior to and after administration of the modified mRNA formulations to determine protein expression and complete blood count. The rodents are also sacrified to analyze the effect of the modified mRNA formulation on the internal tissue. Samples are also collected from the site of administration of rodents administered modified mRNA formulations subcutaneously and intramuscularly to determine the protein expression in the tissue.
C. Toxicity
[00950] The rodents are administered formulations containing at least one modified mRNA to study toxicity of each formulation. The rodents are bled at specified time intervals prior to and after administration of the modified mRNA formulations to determine protein expression and complete blood count. The rodents are also sacrificed to analyze the effect of the modified mRNA
formulation on the internal tissue. Samples are also collected from the site of administration of rodents administered modified mRNA formulations subcutaneously and intramuscularly to determine the protein expression in the tissue.
Example 15. PLGA Microsphere Formulations
[00951] Optimization of parameters used in the formulation of PLGA microspheres may allow for tunable release rates and high encapsulation efficiencies while maintaining the integrity of the modified RNA encapsulated in the microspheres. Parameters such as, but not limited to, particle size, recovery rates and encapsulation efficiency may be optimized to achieve the optimal formulation.
A. Synthesis of PLGA microspheres
[00952] Polylacticglycolic acid (PLGA) microspheres were synthesized using the water/oil/water double emulsification methods known in the art using PLGA (Lactel, Cat# B6010-2, inherent viscosity 0.55-0.75, 50:50 LA:GA), polyvinylalcohol (PVA) (Sigma, Cat# 348406-25G, MW 13- 23k) dichloromethane and water. Briefly, 0.1 ml of water (Wl) was added to 2 ml of PLGA dissolved in dichloromethane (DCM) (01) at concentrations ranging from 50 - 200 mg/ml of PLGA. The Wl/Ol emulsion was homogenized (IKA Ultra-Turrax Homogenizer, Tl 8) for 30 seconds at speed 4 (-15,000 rpm). The Wl/Ol emulsion was then added to 100 to 200 ml of 0.3 to 1% PVA (W2) and homogenized for 1 minute at varied speeds. Formulations were left to stir for 3 hours and then washed by centrifugation (20-25 min, 4,000 rpm, 4°C). The supernatant was discarded and the PLGA pellets were resuspended in 5-10 ml of water, which was repeated 2x. Average particle size (represents 20 -30 particles) for each formulation was determined by microscopy after washing. Table 19 shows that an increase in the PLGA concentration led to larger sized microspheres. A PLGA concentration of 200 mg/mL gave an average particle size of 14.8 μιη, 100 mg/mL was 8.7 μιη, and 50 mg/mL of PLGA gave an average particle size of 4.0 μιη.
Table 19. Varied PLGA Concentration
Figure imgf000281_0001
[00953] Table 20 shows that decreasing the homogenization speed from 5 (-20,000 rpm) to speed 4 (-15,000 rpm) led to an increase in particle size from 14.8 μιη to 29.7 μιη.
Table 20. Varied Homogenization Speed
Figure imgf000281_0002
[00954] Table 21 shows that increasing the W2 volume (i.e. increasing the ratio of W2:01 from 50: 1 to 100: 1), decreased average particle size slightly. Altering the PVA concentration from 0.3 to 1 wt% had little impact on PLGA microsphere size.
Table 21. Varied W2 Volume and Concentration
Figure imgf000281_0003
B. Encapsulation of modified mRNA
[00955] Modified G-CSF mRNA (SEQ ID NO: 6; polyA tail of approximately 160 nucleotides not shown in sequence; 5'cap, Capl; fully modified with 5-methylcytosine and pseudouridine) was dissolved in water at a concentration of 2 mg/ml (W3). Three batches of PLGA microsphere formulations were made as described above with the following parameters: 0.1 ml of W3 at 2 mg/ml, 1.6 ml of 01 at 200 mg/ml, 160 ml of W2 at 1%, and homogenized at a speed of 4 for the first emulsion (W3/01) and homogenized at a speed of 5 for the second emulstion (W3/01/W2). After washing by centrifugation, the formulations were frozen in liquid nitrogen and then lyophilized for 3 days. To test the encapsulation efficiency of the formulations, the lyophilized material was deformulated in DCM for 6 hours followed by an overnight extraction in water. The modified RNA concentration in the samples was then determined by OD260. Encapsulation efficiency was calculated by taking the actual amount of modified RNA and dividing by the starting amount of modified RNA. In the three batches tested, there was an encapsulation efficiency of 59.2, 49.8 and 61.3.
C. Integrity of modified mRNA encapsulated in PLGA microspheres
[00956] Modified Factor IX mRNA (SEQ ID NO: 10; polyA tail of approximately 160 nucleotides not shown in sequence; 5'cap, Capl; fully modified with 5-methylcytosine and pseudouridine) was dissolved in water at varied concentrations (W4) to vary the weight percent loading in the
formulation (mg modified RNA/mg PLGA * 100) and to determine encapsulation efficiency. The parameters in Table 22 were used to make four different batches of PLGA microsphere formulations with a homogenization speed of 4 for the first emulstion (W4/01) and a homogenization speed of 5 for the second emulsion (W4/01/W2).
Table 22. Factor IX PLGA Microsphere Formulation Parameters
Figure imgf000282_0001
[00957] After lyophilization, PLGA microspheres were weighed out in 2 ml eppendorf tubes to correspond to ~ 10 ug of modified RNA. Lyophilization was found to not destroy the overall structure of the PLGA microspheres. To increase weight percent loading (wt%) for the PLGA microspheres, increasing amounts of modified RNA were added to the samples. PLGA microspheres were deformulated by adding 1.0 ml of DCM to each tube and then shaking the samples for 6 hours. For modified RNA extraction, 0.5 ml of water was added to each sample and the samples were shaken overnight before the concentration of modified RNA in the samples was determined by OD260. To determine the recovery of the extraction process, unformulated Factor IX modified RNA (SEQ ID NO: 10; polyA tail of approximately 160 nucleotides not shown in sequence; 5'cap, Capl; fully modified with 5-methylcytosine and pseudouridine) (de formulation control) was spiked into DCM and was subjected to the deformulation process. Table 23 shows the loading and encapsulation efficiency for the samples. All encapsulation efficiency samples were normalized to the deformulation control.
Table 23. Weight Percent Loading and Encapsulation Efficiency
Figure imgf000283_0001
D. Release study of modified mRNA encapsulated in PLGA microspheres
[00958] PLGA microspheres formulated with Factor IX modified RNA (SEQ ID NO: 10) were deformulated as described above and the integrity of the extracted modified RNA was determined by automated electrophoresis (Bio-Rad Experion). The extracted modified mRNA was compared against unformulated modified mRNA and the deformulation control in order to test the integrity of the encapsulated modified mRNA. As shown in Figure 3, the majority of modRNA was intact for batch ID A, B, C and D, for the deformulated control (Deform control) and the unformulated control (Unform control).
E. Protein Expression of modified mRNA encapsulated in PLGA microspheres
[00959] PLGA microspheres formulated with Factor IX modified RNA (SEQ ID NO: 10; polyA tail of approximately 160 nucleotides not shown in sequence; 5'cap, Capl; fully modified with 5- methylcytosine and pseudouridine) were deformulated as described above and the protein expression of the extracted modified RNA was determined by an in vitro transfection assay. HEK293 cells were reverse trans fected with 250 ng of Factor IX modified RNA complexed with RNAiMAX (Invitrogen) in triplicate. [00960] Factor IX modified RNA was diluted in nuc lease-free water to a concentration of 25 ng/μΐ and RNAiMAX was diluted 13.3x in serum- free EMEM. Equal volumes of diluted modified RNA and diluted RNAiMAX were mixed together and were allowed to stand for 20 to 30 minutes at room temperature. Subsequently, 20 μΐ of the transfection mix containing 250 ng of Factor IX modified RNA was added to 80 μΐ of a cell suspension containing 30,000 cells. Cells were then incubated for 16h in a humidified 37°C/5% C02 cell culture incubator before harvesting the cell culture supernatant. Factor IX protein expression in the cell supernatant was analyzed by an ELISA kit specific for Factor IX (Molecular Innovations, Cat # HFIXKT-TOT) and the protein expression is shown in Table 24. In all PLGA microsphere batches tested, Factor IX modified RNA remained active and expressed Factor IX protein after formulation in PLGA microspheres and subsequent deformulation.
Table 24. Protein Expression
Figure imgf000284_0001
F. Release study of modified mRNA encapsulated in PLGA microspheres
[00961] PLGA micropsheres formulated with Factor IX modified RNA (SEQ ID NO: 10; polyA tail of approximately 160 nucleotides not shown in sequence; 5 'cap, Capl; fully modified with 5- methylcytosine and pseudouridine) were resuspended in water to a PLGA microsphere concentration of 24 mg/ml. After resuspension, 150 ul of the PLGA microsphere suspension was aliquoted into eppendorf tubes. Samples were kept incubating and shaking at 37°C during the course of the study. Triplicate samples were pulled at 0.2, 1, 2, 8, 14, and 21 days. To determine the amount of modified RNA released from the PLGA microspheres, samples were centrifuged, the supernatant was removed, and the modified RNA concentration in the supernatant was determined by OD 260. The percent release, shown in Table 25, was calculated based on the total amount of modified RNA in each sample. After 31 days, 96% of the Factor IX modified RNA was released from the PLGA microsphere formulations.
Table 25. Percent Release Time (days) % Release
0 0.0
0.2 27.0
1 37.7
2 45.3
4 50.9
8 57.0
14 61.8
21 75.5
31 96.4
G. Particle size reproducibility of PLGA microspheres
[00962] Three batches of Factor IX modified RNA (SEQ ID NO: 10 polyA tail of approximately 160 nucleotides not shown in sequence; 5 'cap, Capl ; fully modified with 5-methylcytosine and pseudouridine) PLGA microspheres were made using the same conditions described for Batch D, shown in Table 22, (0.4 ml of W4 at 4 mg/ml, 2.0 ml of 01 at 200 mg/ml, 200 ml of W2 at 1%, and homogenized at a speed of 5 for the W4/01/W2 emulsion). To improve the homogeneity of the PLGA microsphere suspension, filtration was incorporated prior to centrifugation. After stirring for 3 hours and before centrifuging, all formulated material was passed through a 100 μιη nylon mesh strainer (Fisherbrand Cell Strainer, Cat # 22-363-549) to remove larger aggregates. After washing and resuspension with water, 100-200 μΐ of a PLGA microspheres sample was used to measure particle size of the formulations by laser diffraction (Malvern Mastersizer2000). The particle size of the samples is shown in Table 26.
Table 26. Particle Size Summary
Figure imgf000285_0001
[00963] Results of the 3 PLGA microsphere batches using filtration were compared to a PLGA microsphere batch made under the same conditions without filtration. The inclusion of a filtration step before washing reduced the mean particle size and demonstrated a consistent particle size distribution between 3 PLGA microsphere batches.
H. Serum Stability of Factor IX PLGA Microspheres
[00964] Factor IX mRNA RNA (SEQ ID NO: 10 polyA tail of approximately 160 nucleotides not shown in sequence; 5'cap, Capl; fully modified with 5 -methylcytosme and pseudouridine) in buffer (TE) or 90% serum (Se), or Factor IX mRNA in PLGA in buffer, 90% serum or 1% serum was incubated in buffer, 90% serum or 1% serum at an mRNA concentration of 50 ng/ul in a total volume of 70 ul. The samples were removed at 0, 30, 60 or 120 minutes. RNases were inactivated with proteinase K digestion for 20 minutes at 55°C by adding 25 ul of 4x proteinase K buffer (0.4 ml 1M TRIS-HCl pH 7.5, 0.1 ml 0.5M EDTA, 0.12 ml 5M NaCl, and 0.4ml 10% SDS) and 8 ul of proteinase K at 20 mg/ml. The Factor IX mRNA was precipitated (add 250 ul 95% ethanol for 1 hour, centrifuge for 10 min at 13 k rpm and remove supernatant, add 200 ul 70% ethanol to the pellet, centrifuge again for 5 min at 13 k rpm and remove supernatant and resuspend the pellet in 70 ul water) or extracted from PLGA microspheres (centrifuge 5 min at 13k rpm and remove supernatant, wash pellet with 1 ml water, centrifuge 5 min at 13k rpm and remove supernatant, add 280 ul dichloromethane to the pellet and shake for 15 minutes, add 70 ul water and then shake for 2 hours and remove the aqueous phase) before being analyzed by bioanalyzer. PLGA microspheres protect Factor IX modified mRNA from degradation in 90% and 1% serum over 2 hours. Factor IX modified mRNA completely degrades in 90% serum at the initial time point.
Example 16. Lipid nanoparticle in vivo studies
[00965] G-CSF (cDNA with the T7 promoter, 5 ' Untranslated region (UTR) and 3 'UTR used in in vitro transcription is given in SEQ ID NO: 5. mRNA sequence shown in SEQ ID NO: 6; polyA tail of approximately 160 nucleotides not shown in sequence; 5'cap, Cap 1; fully modified with 5- methylcytosine and pseudouridine) and Factor IX (cDNA with the T7 promoter, 5 ' UTR and 3 'UTR used in in vitro transcription is given in SEQ ID NO: 13. mRNA sequence shown in SEQ ID NO: 10; polyA tail of approximately 160 nucleotides not shown in sequence; 5'cap, Cap 1; fully modified with 5 -methylcytosme and pseudouridine) modified mRNA were formulated as lipid nanoparticles (LNPs) using the syringe pump method. The LNPs were formulated at a 20: 1 weight ratio of total lipid to modified mRNA with a final lipid molar ratio of 50: 10:38.5: 1.5 (DLin-KC2-DMA: DSPC: Cholesterol: PEG-c-DOMG). Formulations, listed in Table 27, were characterized by particle size, zeta potential, and encapsulation. Table 27. Formulations
Figure imgf000287_0001
[00966] LNP formulations were administered to mice (n=5) intravenously at a modified mRNA dose of 100, 10, or 1 ug. Mice were sacrificed at 8 hrs after dosing. Serum was collected by cardiac puncture from mice that were administered with G-CSF or Factor IX modified mRNA formulations. Protein expression was determined by ELISA.
[00967] There was no significant body weight loss (<5%) in the G-CSF or Factor IX dose groups. Protein expression for G-CSF or Factor IX dose groups was determined by ELISA from a standard curve. Serum samples were diluted (about 20-2500x for G-CSF and about 10-250x for Factor IX) to ensure samples were within the linear range of the standard curve. As shown in Table 28, G-CSF protein expression determined by ELISA was approximately 17, 1200, and 4700 ng /ml for the 1, 10, and 100 ug dose groups, respectively. As shown in Table 29, Factor IX protein expression determined by ELISA was approximately 36, 380, and 3000-11000 ng/ml for the 1, 10, and 100 ug dose groups, respectively.
Table 28. G-CSF Protein Expression
Figure imgf000287_0002
Table 29. Factor IX Protein Expression
Figure imgf000287_0003
Figure imgf000288_0001
[00968] As shown in Table 30, the LNP formulations described above have about a 10,000- 100,000-fold increase in protein production compared to an administration of an intravenous (IV)- lipoplex formulation for the same dosage of modified mRNA and intramuscular (IM) or subcutaneous (SC) administration of the same dose of modified mRNA in saline. As used in Table 30, the symbol "~" means about.
Table 30. Protein Production
Figure imgf000288_0002
Materials and Methods for Examples 17-22
[00969] G-CSF (mRNA sequence shown in SEQ ID NO: 6; polyA tail of approximately 160 nulceotides not shown in sequence; 5 'cap, Cap 1; fully modified with 5-methylcytosine and pseudouridine) and EPO (mRNA sequence shown in SEQ ID NO: 9; polyA tail of approximately 160 nulceotides not shown in sequence; 5'cap, Cap 1; fully modified with 5-methylcytosine and pseudouridine) modified mRNA were formulated as lipid nanoparticles (LNPs) using the syringe pump method. The LNPs were formulated at a 20: 1 weight ratio of total lipid to modified mRNA with a final lipid molar ratio of 50: 10:38.5: 1.5 (DLin-KC2-DMA: DSPC: Cholesterol: PEG-c- DOMG). Formulations, listed in Table 31, were characterized by particle size, zeta potential, and encapsulation.
Table 31. Formulations
Figure imgf000288_0003
PDI: 0.04 PDI: 0.03
Zeta at pH 7.4 0.8 mV 1.5 mV
Encapsulation
95% 98%
(RiboGreen)
Example 17. Lipid nanoparticle in vivo studies with modified mRNA
[00970] LNP formulations, shown in Table 31 (above), were administered to rats (n=5)
intravenously (IV), intramuscularly (IM) or subcutaneously (SC) at a single modified mRNA dose of 0.05 mg/kg. A control group of rats (n=4) was untreated. The rats were bled at 2 hours, 8 hours, 24 hours, 48 hours and 96 hours and after they were administered with G-CSF or EPO modified mRNA formulations to determine protein expression using ELISA. The rats administered EPO modified mRNA intravenously were also bled at 7 days.
[00971] As shown in Table 32, EPO protein expression in the rats intravenously administered modified EPO mRNA was detectable out to 5 days. G-CSF in the rats intravenously administered modified G-CSF mRNA was detectable to 7 days. Subcutaneous and intramuscular administration of EPO modified mRNA was detectable to at least 24 hours and G-CSF modified mRNA was detectable to at least 8 hours. In Table 32, "OSC" refers to values that were outside the standard curve and "NT" means not tested.
Table 32. G-CSF and EPO Protein Expression
Figure imgf000289_0001
SC 24 hours 544.2 OSC
SC 48 hours NT OSC
SC 5 days NT OSC
Untreated All bleeds 0 0
Example 18. Time course in vivo study
[00972] LNP formulations, shown in Table 31 (above), were administered to mice (n=5) intravenously (IV) at a single modified mRNA dose of 0.5, 0.05 or 0.005 mg/kg. The mice were bled at 8 hours, 24 hours, 72 hours and 6 days after they were administered with G-CSF or EPO modified mRNA formulations to determine protein expression using ELISA.
[00973] As shown in Table 33, EPO and G-CSF protein expression in the mice administered with the modified mRNA intravenously was detectable out to 72 hours for the mice dosed with 0.005 mg/kg and 0.05 mg/kg of modified mRNA and out to 6 days for the mice administered the EPO modified mRNA. In Table 33, ">" means greater than and "ND" means not detected.
Table 33. Protein Expression
Figure imgf000290_0001
Example 19. LNP formulations in vivo study in rodents
A. LNP Formulations in Mice
[00974] LNP formulations, shown in Table 31 (above), were administered to mice (n=4) intravenously (IV) at a single modified mRNA dose 0.05 mg/kg or 0.005 mg/kg. There was also 3 control groups of mice (n=4) that were untreated. The mice were bled at 2 hours, 8 hours, 24 hours, 48 hours and 72 hours after they were administered with G-CSF or EPO modified mRNA formulations to determine the protein expression. Protein expression of G-CSF and EPO were determined using ELISA.
[00975] As shown in Table 34, EPO and G-CSF protein expression in the mice was detectable at least out to 48 hours for the mice that received a dose of 0.005 mg/kg modified RNA and 72 hours for the mice that received a dose of 0.05 mg/kg modified RNA. In Table 34, "OSC" refers to values that were outside the standard curve and "NT" means not tested.
Table 34. Protein Expression in Mice
Figure imgf000291_0001
B. LNP Formulations in Rats
[00976] LNP formulations, shown in Table 31 (above), are administered to rats (n=4) intravenously (IV) at a single modified mRNA dose 0.05 mg/kg. There is also a control group of rats (n=4) that are untreated. The rats are bled at 2 hours, 8 hours, 24 hours, 48 hours, 72 hours, 7 days and 14 days after they were administered with G-CSF or EPO modified mRNA formulations to determine the protein expression. Protein expression of G-CSF and EPO are determined using ELISA.
Example 20. Early Time Course Study of LNPs
[00977] LNP formulations, shown in Table 31 (above), are administered to mammals intravenously (IV), intramuscularly (IM) or subcutaneously (SC) at a single modified mRNA dose of 0.5 mg/kg, 0.05 mg/kg or 0.005 mg/kg. A control group of mammals are not treated. The mammals are bled at 5 minutes, 10 minutes, 20 minutes, 30 minutes, 45 minutes, 1 hour, 1.5 hours and/or 2 hours after they are administered with the modified mRNA LNP formulations to determine protein expression using ELISA. The mammals are also bled to determine the complete blood count such as the granulocyte levels and red blood cell count.
Example 21. Non-human primate in vivo study [00978] LNP formulations, shown in Table 31 (above), were administered to non-human primates (NHP) (cynomolgus monkey) (n=2) as a bolus intravenous injection (IV) over approximately 30 seconds using a hypodermic needle, which may be attached to a syringe/abbocath or butterfly if needed. The NHP were administered a single modified mRNA IV dose of 0.05mg/kg of EPO or G- CSF or 0.005 mg/kg of EPO in a dose volume of 0.5 mL/kg. The NHPs were bled 5-6 days before dosing with the modified mRNA LNP formulations to determine protein expression in the serum and a baseline complete blood count. After administration with the modified mRNA formulation the NHP were bled at 8, 24, 48 and 72 hours to determined protein expression. At 24 and 72 hours after administration the complete blood count of the NHP was also determined. Protein expression of G- CSF and EPO was determined by ELISA. Urine from the NHPs was collected over the course of the entire experiment and analyzed to evaluate clinical safety. Samples were collected from the NHPs after they were administered with G-CSF or EPO modified mRNA formulations to determine protein expression using ELISA. Clinical chemistry, hematology, urinalysis and cytokines of the non- human primates were also analyzed.
[00979] As shown in Table 35, EPO protein expression in the NHPs administered 0.05 mg/kg is detectable out to 72 hours and the 0.005 mg/kg dosing of the EPO formulation is detectable out to 48 hours. In Table 35, the "<" means less than a given value. G-CSF protein expression was seen out to 24 hours after administration with the modified mRNA formulation. Preliminarily, there was an increase in granulocytes and reticulocytes levels seen in the NHP after administration with the modified mRNA formulations.
Table 35. Protein Expression in Non-Human Primates
Figure imgf000292_0001
8 hours 19,858 7,072 13,465
24 hours 18,178 4,913 1 1,546
48 hours 5,291 498 2,895
72 hours 744 60 402
Pre-bleed 0 0 0
8 hours 523 250 387
EPO 0.005 24 hours 302 1 13 208
48 hours <7.8 <7.8 <7.8
72 hours 0 0 0
Example 22. Non-human primate in vivo study for G-CSF and EPO
[00980] LNP formulations, shown in Table 31 (above), were administered to non-human primates (NHP) (cynomolgus monkey) (n=2) as intravenous injection (IV). The NHP were administered a single modified mRNA IV dose of 0.5 mg/kg, 0.05mg/kg or 0.005 mg/kg of G-CSF or EPO in a dose volume of 0.5 mL/kg. The NHPs were bled before dosing with the modified mRNA LNP formulations to determine protein expression in the serum and a baseline complete blood count. After administration with the G-CSF modified mRNA formulation the NHP were bled at 8, 24, 48 and 72 hours to determined protein expression. After administration with the EPO modified mRNA formulation the NHP were bled at 8, 24, 48, 72 hours and 7 days to determined protein expression.
[00981] Samples collected from the NHPs after they were administered with G-CSF or EPO modified mRNA formulations were analyzed by ELISA to determine protein expression. Neutrophil and reticulocyte count was also determined pre-dose, 24 hours, 3 days, 7 days, 14 days and 18 days after administration of the modified G-CSF or EPO formulation.
[00982] As shown in Table 36, G-CSF protein expression was not detected beyond 72 hours. In Table 36, "<39" refers to a value below the lower limit of detection of 39 pg/ml.
Table 36. G-CSF Protein Expression
Figure imgf000293_0001
24 hours 1 1,374 3,628
48 hours 1,100 833
72 hours <39 306
Pre-bleed <39 <39
8 hours 3,289 1,722
G-CSF 0.05 24 hours 722 307
48 hours <39 <39
72 hours <39 <39
Pre-bleed <39 <39
8 hours 559 700
G-CSF 0.005 24 hours 155 <39
48 hours <39 <39
72 hours <39 <39 983] As shown in Table 37, EPO protein expression was not detected beyond 7 days. In Table, "<7.8" refers to a value below the lower limit of detection of 7.8 pg/ml.
Table 37. EPO Protein Expression
Figure imgf000294_0001
Pre-bleed <7.8 <7.8
8 hours 523 250
24 hours 302 1 13
EPO 0.005
48 hours 1 1 29
72 hours <7.8 <7.8
7 days <7.8 <7.8
[00984] As shown in Table 38, there was an increase in neutrophils in all G-CSF groups relative to pre-dose levels.
Table 38. Pharmacologic Effect of G-CSF mRNA in NHP
Figure imgf000295_0001
[00985] As shown in Table 39, there was an increase in reticulocytes in all EPO groups 3 days to 14/18 days after dosing relative to reticulocyte levels 24 hours after dosing. Table 39. Pharmacologic Effect of EPO mRNA on Neutrophil Count
Figure imgf000296_0001
[00986] As shown in Tables 40-42, the administration of EPO modified RNA had an effect on other erythropoetic parameters including hemoglobin (HGB), hematocrit (HCT) and red blood cell (RBC) count.
Figure imgf000296_0002
24 hours 122 117 123 116
3 days 126 115 116 120
7 days 126 116 126 121
14/18 days 134 123 133 129
Pre-dose 128 129 132 136
24 hours 117 127 122 128
0.005 3 days 116 127 125 130
7 days 116 129 119 127
14/18 days 118 129 128 129
Table 41. 1 'harmacologic Effect of EPO mRNA on Hematocrit
Dose Male NHP (G- Female NHP (G- Male NHP (EPO) Female NHP
Time
(mg/kg) CSF) HCT (L/L) CSF) HCT (L/L) HCT (L/L) (EPO) HCT (L/L)
Pre-dose 0.46 0.43 0.44 0.4
24 hours 0.37 0.38 0.4 0.36
0.5 3 days 0.39 0.38 0.41 0.39
7 days 0.39 0.38 0.45 0.45
14/18 days 0.34 0.37 0.48 0.46
Pre-dose 0.44 0.44 0.45 0.43
24 hours 0.39 0.4 0.43 0.39
0.05 3 days 0.41 0.39 0.38 0.4
7 days 0.42 0.4 0.45 0.41
14/18 days 0.44 0.4 0.46 0.43
Pre-dose 0.42 0.42 0.48 0.45
24 hours 0.4 0.42 0.42 0.43
0.005 3 days 0.4 0.41 0.44 0.42
7 days 0.39 0.42 0.41 0.42
14/18 days 0.41 0.42 0.42 0.42
Table 42. Pharmacologic Effect of EPO mRNA on Red Blood Cells
Male NHP (G- Female NHP (G- Female NHP
Dose Male NHP (EPO)
Time CSF) RBC CSF) RBC (EPO) RBC (mg/kg) RBC (1012/L)
(1012/L) (1012/L) (1012/L)
Pre-dose 5.57 5.57 5.43 5.26
0.5 24 hours 4.66 4.96 5.12 4.69
3 days 4.91 4.97 5.13 5.15 7 days 4.8 5.04 5.55 5.68
14/18 days 4.21 4.92 5.83 5.72
Pre-dose 5.68 5.64 5.57 5.84
24 hours 4.96 5.08 5.25 5.18
0.05 3 days 5.13 5.04 4.81 5.16
7 days 5.17 5.05 5.37 5.31
14/18 days 5.43 5.26 5.57 5.57
Pre-dose 5.67 5.36 6.15 5.72
24 hours 5.34 5.35 5.63 5.35
0.005 3 days 5.32 5.24 5.77 5.42
7 days 5.25 5.34 5.49 5.35
14/18 days 5.37 5.34 5.67 5.36
[00987] As shown in Tables 43 and 44, the administration of modified RNA had an effect on serum chemistry parameters including alanine transaminase (ALT) and aspartate transaminase (AST).
Table 43. Pharmacolo ic Effect of EPO mRNA on Alanine Transaminase
Figure imgf000298_0001
Table 44. Pharmacolo ic Effect of EPO mRNA on As artate Transaminase
Figure imgf000299_0001
[00988] As shown in Table 45, the administration of modified RNA cause an increase in cytokines, interferon-alpha (IFN-alpha) after administration of modified mRNA.
Table 45. Pharmacolo ic Effect of EPO mRNA on Alanine Transaminase
Figure imgf000299_0002
Example 23. Study of Intramuscular and/or Subcutaneous Administration in Non-Human Primates
[00989] Formulations containing modified EPO mRNA (SEQ ID NO: 9; polyA tail of
approximately 160 nucleotides not shown in sequence; 5 'cap, Capl; fully modified with 5- methylcytosine and pseudouridine) or G-CSF mRNA (SEQ ID NO: 6; polyA tail of approximately 160 nucleotides not shown in sequence; 5'cap, Capl; fully modified with 5-methylcytosine and pseudouridine) in saline were administered to non-human primates (Cynomolgus monkey) (NHP) intramuscularly (IM) or subcutaneously (SC). The single modified mRNA dose of 0.05mg/kg or 0.005 mg/kg was in a dose volume of 0.5 mL/kg. The non-human primates are bled 5-6 days prior to dosing to determine serum protein concentration and a baseline complete blood count. After administration with the modified mRNA formulation the NHP are bled at 8 hours, 24 hours, 48 hours, 72 hours, 7 days and 14 days to determined protein expression. Protein expression of G-CSF and EPO is determined by ELISA.At 24 hours, 72 hours, 7 days and 14 days after administration the complete blood count of the NHP is also determined. Urine from the NHPs is collected over the course of the entire experiment and analyzed to evaluate clinical safety. Tissue near the injection site is also collected and analyzed to determine protein expression.
Example 24. Modified mRNA Trafficking
[00990] In order to determine localization and/or trafficking of the modified mRNA, studies may be performed as follows.
[00991] LNP formulations of siRNA and modified mRNA are formulated according to methods known in the art and/or described herein. The LNP formulations may include at least one modified mRNA which may encode a protein such as G-CSF, EPO, Factor VII, and/or any protein described herein. The formulations may be administered locally into muscle of mammals using intramuscular or subcutaneous injection. The dose of modified mRNA and the size of the LNP may be varied to determine the effect on trafficking in the body of the mammal and/or to assess the impact on a biologic reaction such as, but not limited to, inflammation. The mammal may be bled at different time points to determine the expression of protein encoded by the modified mRNA administered present in the serum and/or to determine the complete blood count in the mammal.
[00992] For example, modified mRNA encoding Factor VII, expressed in the liver and secreted into the serum, may be administered intramuscularly and/or subcutaneously. Coincident or prior to modified mRNA administration, siRNA is administered to knock out endogenous Factor VII. Factor VII arising from the intramuscular and/or subcutaneous injection of modified mRNA is administered is measured in the blood. Also, the levels of Factor VII is measured in the tissues near the injection site. If Factor VII is expressed in blood then there is trafficking of the modified mRNA. If Factor VII is expressed in tissue and not in the blood than there is only local expression of Factor VII.
Example 25. Formulations of Multiple Modified mRNA
[00993] LNP formulations of modified mRNA are formulated according to methods known in the art and/or described herein. The LNP formulations may include at least one modified mRNA which may encode a protein such as G-CSF, EPO, thrombopoietin and/or any protein described herein or known in the art. The at least one modified mRNA may include 1, 2, 3, 4 or 5 modified mRNA molecules. The formulations containing at least one modified mRNA may be administered intravenously, intramuscularly or subcutaneously in a single or multiple dosing regimens. Biological samples such as, but not limited to, blood and/or serum may be collected and analyzed at different time points before and/or after administration of the at least one modified mRNA formulation. An expression of a protein in a biological sample of 50-200 pg/ml after the mammal has been administered a formulation containing at least one modified mRNA encoding said protein would be considered biologically effective.
Example 26. Polyethylene Glycol Ratio Studies
A. Formulation and Characterization of PEG LNPs
[00994] Lipid nanoparticles (LNPs) were formulated using the syringe pump method. The LNPs were formulated at a 20: 1 weight ratio of total lipid to modified G-CSF mRNA (SEQ ID NO: 6; polyA tail of approximately 160 nucleotides not shown in sequence; 5 'cap, Capl; fully modified with 5-methylcytosine and pseudouridine). The molar ratio ranges of the formulations are shown in Table 46.
Table 46. Molar Ratios
Figure imgf000301_0001
[00995] Two types of PEG lipid, 1 ,2-Dimyristoyl-sn-glycerol, methoxypolyethylene Glycol (PEG- DMG, NOF Cat # SUNBRIGHT® GM-020) and 1 ,2-Distearoyl-sn-glycerol, methoxypolyethylene Glycol (PEG-DSG, NOF Cat # SUNBRIGHT® GS-020), were tested at 1.5 or 3.0 mol%. After the formation of the LNPs and the encapsulation of the modified G-CSF mRNA, the LNP formulations were characterized by particle size, zeta potential and encapsulation percentage and the results are shown in Table 47.
Table 47. Characterization of LNP Formulations
Figure imgf000302_0001
B. In Vivo Screening of PEG LNPs
[00996] Formulations of the PEG LNPs described in Table 40 were administered to mice (n=5) intravenously at a dose of 0.5 mg/kg. Serum was collected from the mice at 2 hours, 8 hours, 24 hours, 48 hours, 72 hours and 8 days after administration of the formulation. The serum was analyzed by ELISA to determine the protein expression of G-CSF and the expression levels are shown in Table 48. LNP formulations using PEG-DMG gave substantially higher levels of protein expression than LNP formulations with PEG-DSA.
Table 48. Protein Expression
Figure imgf000302_0002
8 hours 6,162
24 hours 446
48 hours 197
72 hours 124
8 days 5
2 hours 259
8 hours 567
24 hours 258
PEG-DSA, 3% NPA-074-1
48 hours 160
72 hours 328
8 days 33
Example 27. Cationic Lipid Formulation Studies
A. Formulation and Characterization of Cationic Lipid Nanoparticles
[00997] Lipid nanoparticles (LNPs) were formulated using the syringe pump method. The LNPs were formulated at a 20: 1 weight ratio of total lipid to modified mRNA. The final lipid molar ratio ranges of cationic lipid, DSPC, cholesterol and PEG-c-DOMG are outlined in Table 49.
Table 49. Molar Ratios
Figure imgf000303_0001
[00998] A 25 mM lipid solution in ethanol and modified RNA in 50mM citrate at a pH of 3 were mixed to create spontaneous vesicle formation. The vesicles were stabilized in ethanol before the ethanol was removed and there was a buffer exchange by dialysis. The LNPs were then
characterized by particle size, zeta potential, and encapsulation percentage. Table 50 describes the characterization of LNPs encapsulating EPO modified mRNA (SEQ ID NO: 9 polyA tail of approximately 160 nucleotides not shown in sequence; 5 'cap, Capl; fully modified with 5- methylcytosine and pseudouridine) or G-CSF modified mRNA (SEQ ID NO: 6; polyA tail of approximately 160 nucleotdies not shown in sequence; 5 'cap, Capl; fully modified with 5- methylcytosine and pseudouridine) using DLin-MC3-DMA, DLin-DMA or CI 2-200 as the cationic lipid.
Table 50. Characterization of Cationic Lipid Formulations
Figure imgf000303_0002
No. 071-1 072-1 073-1 074-1 075-1 076-1
Lipid DLin- DLin- DLin- DLin- C12-200 C12-200
MC3- MC3- DMA DMA
DMA DMA
Modified
EPO G-CSF EPO G-CSF EPO G-CSF
RNA
Mean Size 96 nm 70 nm 73 nm 97 nm 103 nm
89 nm
PDI: PDI: PDI: PDI: PDI:
PDI: 0.07
0.08 0.04 0.06 0.05 0.09
Zeta at pH 7.4 -1.1 mV - 1.4 mV -1.6 mV -0.4 mV 1.4 mV 0.9 mV
Encapsulation
100% 100% 99% 100% 88% 98%
(RiboGreen)
B. In Vivo Screening of Cationic LNP Formulations
[00999] Formulations of the cationic lipid formulations described in Table 42 were administered to mice (n=5) intravenously at a dose of 0.5 mg/kg. Serum was collected from the mice at 2 hours, 24 hours, 72 hours and/or 7 days after administration of the formulation. The serum was analyzed by ELISA to determine the protein expression of EPO or G-CSF and the expression levels are shown in Table 51.
Table 51. Protein Expression
Figure imgf000304_0001
[001000] Toxcity was seen in the mice administered the LNPs formulations with the cationic lipid CI 2-200 (NPA-075-1 and NPA-076-1) and they were sacrificed at 24 hours because they showed symptoms such as scrubby fur, cowering behavior and weight loss of greater than 10%. CI 2-200 was expected to be more toxic but also had a high level of expression over a short period. The cationic lipid DLin-DMA (NPA-073-1 and NPA-074-1) had the lowest expression out of the three cationic lipids tested. DLin-MC3-DMA (NPA-071-1 and NPA-072-1) showed good expression up to day three and was above the background sample out to day 7 for EPO formulations.
Example 28. Method of Screening for Protein Expression
A. Electrospray Ionization
[001001] A biological sample which may contain proteins encoded by modified RNA administered to the subject is prepared and analyzed according to the manufacturer protocol for electrospray ionization (ESI) using 1, 2, 3 or 4 mass analyzers. A biologic sample may also be analyzed using a tandem ESI mass spectrometry system.
[001002] Patterns of protein fragments, or whole proteins, are compared to known controls for a given protein and identity is determined by comparison.
B. Matrix- Assisted Laser Desorption/Ionization
[001003] A biological sample which may contain proteins encoded by modified RNA administered to the subject is prepared and analyzed according to the manufacturer protocol for matrix-assisted laser desorption/ionization (MALDI).
[001004] Patterns of protein fragments, or whole proteins, are compared to known controls for a given protein and identity is determined by comparison.
C. Liquid Chromatography-Mass spectrometry-Mass spectrometry
[001005] A biological sample, which may contain proteins encoded by modified RNA, may be treated with a trypsin enzyme to digest the proteins contained within. The resulting peptides are analyzed by liquid chromatography-mass spectrometry-mass spectrometry (LC/MS/MS). The peptides are fragmented in the mass spectrometer to yield diagnostic patterns that can be matched to protein sequence databases via computer algorithms. The digested sample may be diluted to achieve 1 ng or less starting material for a given protein. Biological samples containing a simple buffer background (e.g. water or volatile salts) are amenable to direct in-solution digest; more complex backgrounds (e.g. detergent, non-volatile salts, glycerol) require an additional clean-up step to facilitate the sample analysis.
[001006] Patterns of protein fragments, or whole proteins, are compared to known controls for a given protein and identity is determined by comparison.
Example 29. LNP in vivo studies
[001007] mCherry mRNA (SEQ ID NO: 14; polyA tail of approximately 160 nucleotides not shown in sequence; 5'cap, Capl; fully modified with 5-methylcytosine and pseudouridine) was formulated as a lipid nanoparticle (LNP) using the syringe pump method. The LNP was formulated at a 20: 1 weight ratio of total lipid to modified mRNA with a final lipid molar ratio of 50:10:38.5: 1.5 (DLin- KC2-DMA: DSPC: Cholesterol: PEG-c-DOMG). The mCherry formulation, listed in Table 52, was characterized by particle size, zeta potential, and encapsulation.
Table 52. mCherry Formulation
Figure imgf000306_0001
[001008] The LNP formulation was administered to mice (n=5) intravenously at a modified mRNA dose of 100 ug. Mice were sacrificed at 24 hrs after dosing. The liver and spleen from the mice administered with mCherry modified mRNA formulations were analyzed by immunohistochemistry (IHC), western blot, or fluorescence-activated cell sorting (FACS).
[001009] Histology of the liver showed uniform mCherry expression throughout the section, while untreated animals did not express mCherry. Western blots were also used to confirm mCherry expression in the treated animals, whereas mCherry was not detected in the untreated animals.
Tubulin was used as a control marker and was detected in both treated and untreated mice, indicating that normal protein expression in hepatocytes was unaffected.
[001010] FACS and IHC were also performed on the spleens of mCherry and untreated mice. All leukocyte cell populations were negative for mCherry expression by FACS analysis. By IHC, there were also no observable differences in the spleen in the spleen between mCherry treated and untreated mice.
Example 30. Syringe Pump In Vivo studies
[001011] mCherry modified mRNA is formulated as a lipid nanoparticle (LNP) using the syringe pump method. The LNP is formulated at a 20: 1 weight ratio of total lipid to modified mRNA with a final lipid molar ratio of 50: 10:38.5: 1.5 (DLin-KC2-DMA: DSPC: Cholesterol: PEG-c-DOMG). The mCherry formulation is characterized by particle size, zeta potential, and encapsulation.
[001012] The LNP formulation is administered to mice (n=5) intravenously at a modified mRNA dose of 10 or 100 ug. Mice are sacrificed at 24 hrs after dosing. The liver and spleen from the mice administered with mCherry modified mR A formulations are analyzed by immunohistochemistry (IHC), western blot, and/or fluorescence-activated cell sorting (FACS).
Example 31. In vitro and in vivo expression
A. In vitro Expression in Human Cells Using Lipidoid Formulations
[001013] The ratio of mmRNA to lipidoid used to test for in vitro transfection is tested empirically at different lipidoid:mmRNA ratios. Previous work using siRNA and lipidoids have utilized 2.5: 1, 5: 1, 10: 1, and 15:1 lipidoid: siRNA wtwt ratios. Given the longer length of mmRNA relative to siRNA, a lower wtwt ratio of lipidoid to mmRNA may be effective. In addition, for comparison mmRNA were also formulated using RNAIMAX™ (Invitrogen, Carlsbad, CA) or TRANSIT-mRNA (Minis Bio, Madison, WI) cationic lipid delivery vehicles. The ability of lipidoid- formulated Luciferase (IVT cDNA sequence as shown in SEQ ID NO: 15; mRNA sequence shown in SEQ ID NO: 16, polyA tail of approximately 160 nucleotides not shown in sequence, 5'cap, Capl, fully modified with 5-methylcytosine at each cytosine and pseudouridine replacement at each uridine site), green fluorescent protein (GFP) (IVT cDNA wild-type sequence is shown in SEQ ID NO: 17; mRNA sequence shown in SEQ ID NO: 18, polyA tail of approximately 160 nucleotides not shown in sequence, 5'cap, Capl), G-CSF (mRNA sequence shown in SEQ ID NO: 6; polyA tail of approximately 160 nucleotides not shown in sequence; 5'cap, Capl), and EPO mRNA (mRNA sequence shown in SEQ ID NO: 9; polyA tail of approximately 160 nucleotides not shown in sequence; 5'cap, Capl) to express the desired protein product can be confirmed by luminescence for luciferase expression, flow cytometry for GFP expression, and by ELISA for G-CSF and
Erythropoietin (EPO) secretion.
B. In vivo Expression Following Intravenous Injection
[001014] Systemic intravenous administration of the formulations are created using various different lipidoids including, but not limited to, 98N12-5, CI 2-200, and MD1.
[001015] Lipidoid formulations containing mmRNA are injected intravenously into animals. The expression of the modified mRNA (mmRNA)-encoded proteins are assessed in blood and/or other organs samples such as, but not limited to, the liver and spleen collected from the animal.
Conducting single dose intravenous studies will also allow an assessment of the magnitude, dose responsiveness, and longevity of expression of the desired product.
[001016] In one embodiment, lipidoid based formulations of 98N12-5, C12-200, MD1 and other lipidoids, are used to deliver luciferase, green fluorescent protein (GFP), mCherry fluorescent protein, secreted alkaline phosphatase (sAP), human G-CSF, human Factor IX, or human Erythropoietin (EPO) mmRNA into the animal. After formulating mmRNA with a lipid, as described previously, animals are divided into groups to receive either a saline formulation, or a lipidoid-formulation which contains one of a different mmRNA selected from luciferase, GFP, mCherry, sAP, human G-CSF, human Factor IX, and human EPO. Prior to injection into the animal, mmRNA-containing lipidoid formulations are diluted in PBS. Animals are then
administered a single dose of formulated mmRNA ranging from a dose of 10 mg/kg to doses as low as 1 ng/kg, with a preferred range to be 10 mg/kg to 100 ng/kg, where the dose of mmRNA depends on the animal body weight such as a 20 gram mouse receiving a maximum formulation of 0.2 ml (dosing is based no mmRNA per kg body weight). After the administration of the mmRNA-lipidoid formulation, serum, tissues, and/or tissue lysates are obtained and the level of the mmRNA-encoded product is determined at a single and/or a range of time intervals. The ability of lipidoid- formulated Luciferase, GFP, mCherry, sAP, G-CSF, Factor IX, and EPO mmRNA to express the desired protein product is confirmed by luminescence for the expression of Luciferase, flow cytometry for the expression of GFP and mCherry expression, by enzymatic activity for sAP, or by ELISA for the section of G-CSF, Factor IX and/or EPO.
[001017] Further studies for a multi-dose regimen are also performed to determine the maximal expression of mmRNA, to evaluate the saturability of the mmRNA-driven expression (by giving a control and active mmRNA formulation in parallel or in sequence), and to determine the feasibility of repeat drug administration (by giving mmRNA in doses separated by weeks or months and then determining whether expression level is affected by factors such as immunogenicity). An assessment of the physiological function of proteins such as G-CSF and EPO are also determined through analyzing samples from the animal tested and detecting increases in granulocyte and red blood cell counts, respectively. . Activity of an expressed protein product such as Factor IX, in animals can also be assessed through analysis of Factor IX enzymatic activity (such as an activated partial thromboplastin time assay) and effect of clotting times.
C. In vitro Expression Following Intramuscular and/or Subcutaneous Injection
[001018] The use of lipidoid formulations to deliver oligonucleotides, including mRNA, via an intramuscular route or a subcutaneous route of injection needs to be evaluated as it has not been previously reported. Intramuscular and/or subcutaneous injection of mmRNA are evaluated to determine if mmRNA-containing lipidoid formulations are capabable to produce both localized and systemic expression of a desired portiens. [001019] Lipidoid formulations of 98N12-5, C12-200, and MD1 containing mmRNA selected from luciferase, green fluorescent protein (GFP), mCherry fluorescent protein, secreted alkaline phosphatase (sAP), human G-CSF, human factor IX, or human Erythropoietin (EPO) mmRNA are injected intramuscularly and/or subcutaneously into animals. The expression of mmRNA-encoded proteins are assessed both within the muscle or subcutaneous tissue and systemically in blood and other organs such as the liver and spleen. Single dose studies allow an assessment of the magnitude, dose responsiveness, and longevity of expression of the desired product.
[001020] Animals are divided into groups to receive either a saline formulation or a formulation containing modified mRNA. Prior to injection mmRNA-containing lipidoid formulations are diluted in PBS. Animals are administered a single intramuscular dose of formulated mmRNA ranging from 50 mg/kg to doses as low as 1 ng/kg with a preferred range to be 10 mg/kg to 100 ng/kg. A maximum dose for intramuscular administration, for a mouse, is roughly 1 mg mmRNA or as low as 0.02 ng mmRNA for an intramuscular injection into the hind limb of the mouse. For subcutaneous administration, the animals are administered a single subcutaneous dose of formulated mmRNA ranging from 400 mg/kg to doses as low as 1 ng/kg with a preferred range to be 80 mg/kg to 100 ng/kg. A maximum dose for subcutaneous administration, for a mouse, is roughly 8 mg mmRNA or as low as 0.02 ng mmRNA.
[001021] For a 20 gram mouse the volume of a single intramuscular injection is maximally 0.025 ml and a single subcutaneous injection is maximally 0.2 ml. The optimal dose of mmRNA
administered is calculated from the body weight of the animal. At various points in time points following the administration of the mmRNA-lipidoid, serum, tissues, and tissue lysates is obtained and the level of the mmRNA-encoded product is determined. The ability of lipidoid- formulated luciferase, green fluorescent protein (GFP), mCherry fluorescent protein, secreted alkaline phosphatase (sAP), human G-CSF, human factor IX, or human Erythropoietin (EPO) mmRNA to express the desired protein product is confirmed by luminescence for luciferase expression, flow cytometry for GFP and mCherry expression, by enzymatic activity for sAP, and by ELISA for G- CSF, Factor IX and Erythropoietin (EPO) secretion.
[001022] Additional studies for a multi-dose regimen are also performed to determine the maximal expression using mmRNA, to evaluate the saturability of the mmRNA-driven expression (achieved by giving a control and active mmRNA formulation in parallel or in sequence), and to determine the feasibility of repeat drug administration (by giving mmRNA in doses separated by weeks or months and then determining whether expression level is affected by factors such as immunogenicity).
Studies utilizing multiple subcutaneous or intramuscular injection sites at one time point, are also utilized to further increase mmRNA drug exposure and improve protein production. An assessment of the physiological function of proteins, such as GFP, mCherry, sAP, human G-CSF, human factor IX, and human EPO, are determined through analyzing samples from the tested animals and detecting a change in granulocyte and/or red blood cell counts. Activity of an expressed protein product such as Factor IX, in animals can also be assessed through analysis of Factor IX enzymatic activity (such as an activated partial thromboplastin time assay) and effect of clotting times.
Example 32: In Vivo Delivery Using Lipoplexes
A. Human EPO Modified RNA Lipoplex
[001023] A formulation containing 100 μg of modified human erythropoietin (EPO) mRNA (mRNA sequence shown in SEQ ID NO: 9; polyA tail of approximately 160 nucleotides not shown in sequence; 5'cap, Capl) (EPO; fully modified 5-methylcytosine; Nl-methylpseudouridine) was lipoplexed with 30% by volume of RNAIMAX™ (Lipoplex-h-Epo-46; Generation 2 or Gen2) in 50- 70 uL delivered intramuscularly to four C57/BL6 mice. Other groups consisted of mice receiving an injection of the lipoplexed modified luciferase mRNA (Lipoplex-luc) (IVT cDNA sequence shown in SEQ ID NO: 15; mRNA sequence shown in SEQ ID NO: 16, polyA tail of approximately 160 nucleotides not shown in sequence, 5'cap, Capl, fully modified with 5-methylcytosine at each cytosine and pseudouridine replacement at each uridine site) which served as a control containing 100 μg of modified luciferase mRNA was lipoplexed with 30% by volume of RNAiMAX™ or mice receiving an injection of the formulation buffer as negative control at a dose volume of 65ul. 13 hours after the intramuscular injection, serum was collected from each mouse to measure the amount of human EPO protein in the mouse serum by human EPO ELISA and the results are shown in Table 53.
Table 53. Human EPO Production (IM Injection Route)
Figure imgf000310_0001
B. Human G-CSF Modified RNA Lipoplex
[001024] A formulation containing 100 μg of one of two versions of modified human G-CSF mRNA (mRNA sequence shown in SEQ ID NO: 6; polyA tail of approximately 160 nucleotides not shown in sequence; 5'cap, Capl) (G-CSF fully modified with 5-methylcytosine and pseudouridine (G- CSF) or G-CSF fully modified with 5-methylcytosine and Nl-methyl-pseudouridine (G-CSF-N1) lipoplexed with 30% by volume of R AIMAX™ and delivered in 150 uL intramuscularly (l.M), in 150 uL subcutaneously (S.C) and in 225uL intravenously (I.V) to C57/BL6 mice.
[001025] Three control groups were administered either 100 μg of modified luciferase mRNA (IVT cDNA sequence shown in SEQ ID NO: 15; mRNA sequence shown in SEQ ID NO: 16, polyA tail of approximately 160 nucleotides not shown in sequence, 5'cap, Capl, fully modified with 5- methylcytosine at each cytosine and pseudouridine replacement at each uridine site) intramuscularly (Luc-unsp l.M.) or 150 μg of modified luciferase mRNA intravenously (Luc-unsp I.V.) or 150 uL of the formulation buffer intramuscularly (Buffer I.M.). 6 hours after administration of a formulation, serum was collected from each mouse to measure the amount of human G-CSF protein in the mouse serum by human G-CSF ELISA and the results are shown in Table 54.
[001026] These results demonstrate that both 5-methylcytosine/pseudouridine and 5- methylcytosine/Nl-methylpseudouridine modified human G-CSF mRNA can result in specific human G-CSF protein expression in serum when delivered via I.V. or l.M. route of administration in a lipoplex formulation.
Table 54. Human G-CSF in Serum (I.M., I.V., S.C. Injection Route)
Figure imgf000311_0001
C. Human G-CSF Modified RNA Lipoplex Comparison
[001027] A formulation containing 100 μg of either modified human G-CSF mRNA lipoplexed with 30% by volume of RNAIMAX™ with a 5-methylcytosine (5mc) and a pseudouridine (ψ) modification (G-CSF-Genl -Lipoplex), modified human G-CSF mRNA with a 5mc and ψ modification in saline (G-CSF-Genl -Saline), modified human G-CSF mRNA with a Nl-5- methylcytosine (Nl-5mc) and a ψ modification lipoplexed with 30% by volume of RNAIMAX™ (G-CSF-Gen2-Lipoplex), modified human G-CSF mRNA with a Nl-5mc and ψ modification in saline (G-CSF-Gen2-Saline), modified luciferase with a 5mc and ψ modification lipoplexed with 30% by volume of RNAIMAX™ (Luc-Lipoplex), or modified luciferase mRNA with a a 5mc and ψ modification in saline (Luc-Saline) was delivered intramuscularly (I.M.) or subcutaneously (S.C.) and a control group for each method of administration was giving a dose of 80uL of the formulation buffer (F. Buffer) to C57/BL6 mice. 13 hours post injection serum and tissue from the site of injection were collected from each mouse and analyzed by G-CSF ELISA to compare human G-CSF protein levels. The results of the human G-CSF protein in mouse serum from the intramuscular administration and the subcutaneous administration results are shown in Table 55.
[001028] These results demonstrate that 5-methylcytosine/pseudouridine and 5-methylcytosine/Nl- methylpseudouridine modified human G-CSF mRNA can result in specific human G-CSF protein expression in serum when delivered via I.M. or S.C. route of administration whether in a saline formulation or in a lipoplex formulation. As shown in Table 55, 5-methylcytosine/Nl- methylpseudouridine modified human G-CSF mRNA generally demonstrates increased human G- CSF protein production relative to 5-methylcytosine/pseudouridine modified human G-CSF mRNA.
Table 55. Human G-CSF Protein in Mouse Serum
Figure imgf000312_0001
D. mCherry Modified RNA Lipoplex Comparison
Intramuscular and Subcutaneous Administration
[001029] A formulation containing 100 μg of either modified mCherry mRNA (mRNA sequence shown in SEQ ID NO: 7; polyA tail of approximately 160 nucleotides not shown in sequence; 5 'cap, Capl) lipoplexed with 30% by volume of RNAIMAX™ or modified mCherry mRNA in saline is delivered intramuscularly and subcutaneously to mice. A formulation buffer is also administered to a control group of mice either intramuscularly or subcutaneously. The site of injection on the mice may be collected 17 hours post injection for sectioning to determine the cell type(s) responsible for producing protein.
Intravitreal Administration [001030] A formulation containing 10 μg of either modified mCherry mRNA lipoplexed with RNAIMAX™, modified mCherry mRNA in a formulation buffer, modified luciferase mRNA lipoplexed with RNAMAX™, modified luciferase mRNA in a formulation buffer can be
administered by intravitreal injection (IVT) in rats in a dose volume of 5 μΐ/eye. A formulation buffer is also administrered by IVT to a control group of rats in a dose volume of 5 μΐ/eye. Eyes from treated rats can be collected after 18 hours post injection for sectioning and lysating to determine whether mrnRNA can be effectively delivered in vivo to the eye and result in protein production, and to also determine the cell type(s) responsible for producing protein in vivo.
Intranasal Administration
[001031] A formulation containing 100 μg of either modified mCherry mRNA lipoplexed with 30% by volume of RNAIMAX™, modified mCherry mRNA in saline, modified luciferase mRNA lipoplexed with 30% by volume of RNAIMAX™ or modified luciferase mRNA in saline is delivered intranasally. A formulation buffer is also administered to a control group intranasally. Lungs may be collected about 13 hours post instillation for sectioning (for those receiving mCherry mRNA) or homogenization (for those receiving luciferase mRNA). These samples will be used to determine whether mrnRNA can be effectively delivered in vivo to the lungs and result in protein production, and to also determine the cell type(s) responsible for producing protein in vivo.
Example 33: In Vivo Delivery Using Varying Lipid Ratios
[001032] Modified mRNA was delivered to C57/BL6 mice to evaluate varying lipid ratios and the resulting protein expression. Formulations of 100μg modified human EPO mRNA (mRNA sequence shown in SEQ ID NO: 9; polyA tail of approximately 160 nucleotides not shown in sequence; 5 'cap, Capl; fully modified with 5-methylcytosine and pseudouridine) lipoplexed with 10%, 30% or 50% RNAIMAX™, 100μg modified luciferase mRNA (IVT cDNA sequence shown in SEQ ID NO: 15; mRNA sequence shown in SEQ ID NO: 16, polyA tail of approximately 160 nucleotides not shown in sequence, 5'cap, Capl, fully modified with 5-methylcytosine at each cytosine and pseudouridine replacement at each uridine site) lipoplexed with 10%>, 30%> or 50%> RNAIMAX™ or a formulation buffer were administered intramuscularly to mice in a single 70 μΐ dose. Serum was collected 13 hours post injection to undergo a human EPO ELISA to determine the human EPO protein level in each mouse. The results of the human EPO ELISA, shown in Table 56, show that modified human EPO expressed in the muscle is secreted into the serum for each of the different percentage of RNAIMAX™. Table 56. Human EPO Protein in Mouse Serum (IM Injection Route)
Figure imgf000314_0001
Example 34: Intramuscular and Subcutaneous In Vivo Delivery in Mammals
[001033] Modified human EPO mRNA (mR A sequence shown in SEQ ID NO: 9; polyA tail of approximately 160 nucleotides not shown in sequence; 5'cap, Capl; fully modified with 5- methylcytosine and pseudouridine) formulated in formulation buffer was delivered to either C57/BL6 mice or Sprague-Dawley rats to evaluate the dose dependency on human EPO production. Rats were intramuscularlly injected with 50 μΐ of the modified human EPO mRNA (h-EPO), modified luciferase mRNA (Luc) (IVT cDNA sequence shown in SEQ ID NO: 15; mRNA sequence shown in SEQ ID NO: 16, polyA tail of approximately 160 nucleotides not shown in sequence, 5'cap, Capl, fully modified with 5-methylcytosine at each cytosine and pseudouridine replacement at each uridine site) or the formulation buffer (F. Buffer) as described in the dosing chart Table 57.
[001034] Mice were intramuscularly or subcutaneously injected with 50 μΐ of the modified human EPO mRNA (h-EPO), modified luciferase mRNA (Luc) or the formulation buffer (F. Buffer) as described in the dosing chart Table 58. 13 hours post injection blood was collected and serum was analyzed to determine the amount human EPO for each mouse or rat. The average and geometric mean in pg/ml for the rat study are also shown in Table 57.
Table 57. Rat Study
Figure imgf000314_0002
Table 58. Mouse Study
Figure imgf000315_0001
Example 35: Duration of Activity after Intramuscular In Vivo Delivery
[001035] Modified human EPO mRNA (mRNA sequence shown in SEQ ID NO: 9; polyA tail of approximately 160 nucleotides not shown in sequence; 5 'cap, Capl; fully modified with 5- methylcytosine and pseudouridine) formulated in formulation buffer was delivered to Sprague- Dawley rats to determine the duration of the dose response. Rats were intramuscularly injected with 50 μΐ of the modified human EPO mRNA (h-EPO), modified luciferase mRNA (IVT cDNA sequence shown in SEQ ID NO: 15; mRNA sequence shown in SEQ ID NO: 16, polyA tail of approximately 160 nucleotides not shown in sequence, 5'cap, Capl, fully modified with 5- methylcytosine at each cytosine and pseudouridine replacement at each uridine site) (Luc) or the formulation buffer (F.Buffer) as described in the dosing chart Table 59. The rats were bled 2, 6, 12, 24, 48 and 72 hours after the intramuscular injection to determine the concentration of human EPO in serum at a given time. The average and geometric mean in pg/ml for this study are also shown in Table 59.
Table 59. Dosing Chart
Figure imgf000315_0002
h-EPO 72 hour 100 80.1 75.8
Luc 24, 48 100 37.2 29.2
and 72
hour
F.Buffer 24, 48 48.9 10.4
and 72
hour
Example 36: Routes of Administration
[001036] Studies were performed to investigate split dosing using different routes of administration. Studies utilizing multiple subcutaneous or intramuscular injection sites at one time point were designed and performed to investigate ways to increase mmRNA drug exposure and improve protein production. In addition to detection of the expressed protein product, an assessment of the physiological function of proteins was also determined through analyzing samples from the animal tested.
[001037] Surprisingly, it has been determined that split dosing of mmRNA produces greater protein production and phenotypic responses than those produced by single unit dosing or multi-dosing schemes.
[001038] The design of a split dose experiment involved using human erythropoietin (EPO) mmRNA (mRNA sequence shown in SEQ ID NO: 9; polyA tail of approximately 160 nucleotides not shown in sequence; 5 'cap, Capl) or luciferase mmRNA (mRNA sequence shown in SEQ ID NO: 16; polyA tail of approximately 160 nucleotides not shown in sequence; 5 'cap, Capl) administered in buffer alone or formulated with 30% lipoplex (RNAIMAX™). The dosing vehicle (formulation buffer) consisted of 150mM NaCl, 2 mM CaCl2, 2 mM Na+-phosphate (1.4mM monobasic sodium phosphate; 0.6mM dibasic sodium phosphate), and 0.5 mM EDTA, pH 6.5. The pH was adjusted using sodium hydroxide and the final solution was filter sterilized. The mmRNA was modified with 5meC at each cytosine and pseudouridine replacement at each uridine site.
[001039] 4 mice per group were dosed intramuscularly (I.M.), intravenously (I.V.) or subcutaneously (S.C.) by the dosing chart outlined in Table 60. Serum was collected 13 hours post injection from all mice, tissue was collected from the site of injection from the intramuscular and subcutaneous group and the spleen, liver and kidneys were collected from the intravenous group. The results from the intramuscular group and the subcutaneous group results are shown in Table 61.
Table 60. Dosing Chart Group Treatment Route Dose of mmRNA Total Dosing
Dose Vehicle
1 Lipoplex-human EPO mmRNA I.M. 4 x 100 ug + 30% 4x70 ul Lipoplex
Lipoplex
2 Lipoplex-human EPO mmRNA I.M. 4 x lOO ug 4x70 ul Buffer
3 Lipoplex-human EPO mmRNA S.C. 4 x 100 ug + 30% 4x70 ul Lipoplex
Lipoplex
4 Lipoplex-human EPO mmRNA S.C. 4 x 100 ug 4x70 ul Buffer
5 Lipoplex-human EPO mmRNA I.V. 200 ug + 30% Lipoplex 140 ul Lipoplex
6 Lipoplexed-Luciferase mmRNA I.M. 100 ug + 30% Lipoplex 4x70 ul Lipoplex
7 Lipoplexed-Luciferase mmRNA I.M. 100 ug 4x70 ul Buffer
8 Lipoplexed-Luciferase mmRNA S.C. 100 ug + 30% Lipoplex 4x70 ul Lipoplex
9 Lipoplexed-Luciferase mmRNA S.C. 100 ug 4x70 ul Buffer
10 Lipoplexed-human EPO mmRNA I.V. 200 ug + 30% Lipoplex 140 ul Lipoplex
11 Formulation Buffer I.M. 4x multi dosing 4x70 ul Buffer
Table 61. Human EPO Protein in Mouse Serum (I.M. Injection Route)
Figure imgf000317_0001
Example 37. Rapidly eliminated Lipid Nanoparticle (reLNP) Studies
A. Formulation of Modified RNA reLNPs
[001040] Solutions of synthesized lipid, 1 ,2-distearoyl-3 -phosphatidylcholine (DSPC) (Avanti Polar Lipids, Alabaster, AL), cholesterol (Sigma- Aldrich, Taufkirchen, Germany), and a-[3'-(l,2- dimyristoyl-3-propanoxy)-carboxamide-propyl]-ro-methoxy-polyoxy ethylene (PEG-c-DOMG) (NOF, Bouwelven, Belgium) are prepared and stored at -20°C. The synthesized lipid is selected from DLin-DMA with an internal ester, DLin-DMA with a terminal ester, DLin-MC3-DMA-internal ester, and DLin-MC3-DMA with a terminal ester. The reLNPs are combined to yield a molar ratio of 50: 10:38.5:1.5 (reLNP: DSPC: Cholesterol: PEG-c-DOMG). Formulations of the reLNPs and modified mRNA are prepared by combining the lipid solution with the modified mRNA solution at total lipid to modified mRNA weight ratio of 10 : 1 , 15: 1, 20: 1 and 30: 1.
B. Characterization of formulations
[001041] A Zetasizer Nano ZS (Malvern Instruments Ltd, Malvern, Worcestershire, UK) is used to determine the particle size, the polydispersity index (PDI) and the zeta potential of the modified mRNA nanoparticles in IX PBS in determining particle size and 15 mM PBS in determining zeta potential.
[001042] Ultraviolet-visible spectroscopy is used to determine the concentration of modified mRNA nanoparticle formulation. After mixing, the absorbance spectrum of the solution is recorded between 230 nm and 330 nm on a DU 800 spectrophotometer (Beckman Coulter, Beckman Coulter, Inc., Brea, CA). The modified RNA concentration in the nanoparicle formulation is calculated based on the extinction coefficient of the modified RNA used in the formulation and on the difference between the absorbance at a wavelength of 260 nm and the baseline value at a wavelength of 330 nm.
[001043] QUANT-IT™ RIBOGREEN® RNA assay (Invitrogen Corporation Carlsbad, CA) is used to evaluate the encapsulation of modified RNA by the nanoparticle. The samples are diluted, transferred to a polystyrene 96 well plate, then either a TE buffer or a 2% Triton X-100 solution is added. The plate is incubated and the RIBOGREEN® reagent is diluted in TE buffer, and of this solution is added to each well. The fluorescence intensity is measured using a fluorescence plate reader (Wallac Victor 1420 Multilablel Counter; Perkin Elmer, Waltham, MA) The fluorescence values of the reagent blank are subtracted from each of the samples and the percentage of free modified RNA is determined by dividing the fluorescence intensity of the intact sample by the fluorescence value of the disrupted sample.
C. In Vitro Incubation
[001044] Human embryonic kidney epithelial (HEK293) and hepatocellular carcinoma epithelial (HepG2) cells (LGC standards GmbH, Wesel, Germany) are seeded on 96-well plates (Greiner Bio- one GmbH, Frickenhausen, Germany) and plates for HEK293 cells are precoated with collagen typel . HEK293 are seeded at a density of about 30,000 and HepG2 are seeded at a density of about 35,000 cells per well in 100 μΐ cell culture medium. Formulations containing mCherry mRNA (mRNA sequence shown in SEQ ID NO: 7; polyA tail of approximately 160 nucleotides not shown in sequence; 5 'cap, Capl) are added directly after seeding the cells and incubated. The mCherry cDNA with the T7 promoter, 5 'untranslated region (UTR) and 3' UTR used in in vitro transcription (IVT) is given in SEQ ID NO: 8.
[001045] Cells are harvested by transferring the culture media supematants to a 96-well Pro-Bind U- bottom plate (Beckton Dickinson GmbH, Heidelberg, Germany). Cells are trypsinized with ½ volume Trypsin/EDTA (Biochrom AG, Berlin, Germany), pooled with respective supematants and fixed by adding one volume PBS/2%FCS (both Biochrom AG, Berlin, Germany)/0.5% formaldehyde (Merck, Darmstadt, Germany). Samples are then submitted to a flow cytometer measurement with an excitation laser and a filter for PE-Texas Red in a LSRII cytometer (Beckton Dickinson GmbH, Heidelberg, Germany). The mean fluorescence intensity (MFI) of all events and the standard deviation of four independent wells are presented in for samples analyzed.
D. In Vivo Formulation Studies
[001046] Mice are administered intravenously a single dose of a formulation containing a modified mRNA and a reLNP. The modified mRNA administered to the mice is selected from G-CSF (mRNA sequence shown in SEQ ID NO: 6; polyA tail of approximately 160 nucleotides not shown in sequence; 5 'cap, Capl), Factor IX (mRNA shown in SEQ ID NO: 10; polyA tail of approximately 160 nucleotides not shown in sequence; 5 'cap, Capl) or mCherry (mRNA sequence shown in SEQ ID NO: 7; polyA tail of approximately 160 nucleotides not shown in sequence; 5 'cap, Capl).
[001047] The mice are injected with lOOug, 10 ug or 1 ug of the formulated modified mRNA and are sacrificed 8 hours after they are administered the formulation. Serum from the mice administered formulations containing human G-CSF modified mRNA are measured by specific G-CSF ELISA and serum from mice administered human Factor IX modified RNA is analyzed by specific factor IX ELISA or chromogenic assay. The liver and spleen from the mice administered with mCherry modified mRNA are analyzed by immunohistochemistry (IHC) or fluorescence-activated cell sorting (FACS). As a control, a group of mice are not injected with any formulation and their serum and tissue are collected analyzed by ELISA, FACS and/or IHC.
Example 38. In Vitro Transfection of VEGF-A
[001048] Human vascular endothelial growth factor-isoform A (VEGF-A) modified mRNA (mRNA sequence shown in SEQ ID NO: 19; polyA tail of approximately 160 nucleotides not shown in sequence; 5'cap, Capl) was transfected via reverse transfection in Human Keratinocyte cells in 24 multi-well plates. The VEGF-A cDNA with the T7 promoter, 5' untranslated region (UTR) and 3' UTR used in in vitro transcription (IVT) is given in SEQ ID NO: 20. Human Keratinocytes cells were grown in EPILIFE® medium with Supplement S7 from Invitrogen (Carlsbad, CA) until they reached a confluence of 50-70%. The cells were transfected with 0, 46.875, 93.75, 187.5, 375, 750, and 1500 ng of modified mRNA (mmRNA) encoding VEGF-A which had been complexed with RNAIMAX™ from Invitrogen (Carlsbad, CA). The RNA : RN AIM AX™ complex was formed by first incubating the RNA with Supplement- free EPILIFE® media in a 5X volumetric dilution for 10 minutes at room temperature. In a second vial, RNAIMAX reagent was incubated with Supplement-free EPILIFE® Media in a 10X volumetric dilution for 10 minutes at room temperature. The RNA vial was then mixed with the RNAIMAX™ vial and incubated for 20-30 minutes at room temperature before being added to the cells in a drop-wise fashion.
[001049] The fully optimized mRNA encoding VEGF-A (mRNA sequence shown in SEQ ID NO: 19; polyA tail of approximately 160 nucleotides not shown in sequence; 5 'cap, Capl) transfected with the Human Keratinocyte cells included modifications during translation such as natural nucleoside triphosphates (NTP), pseudouridine at each uridine site and 5-methylcytosine at each cytosine site (pseudo-U/5mC), and Nl-methyl-pseudouridine at each uridine site and 5- methylcytosine at each cytosine site (Nl-methyl-Pseudo-U/5mC). Cells were transfected with the mmRNA encoding VEGF-A and secreted VEGF-A concentration (pg/ml) in the culture medium was measured at 6, 12, 24, and 48 hours post-transfection for each of the concentrations using an ELISA kit from Invitrogen (Carlsbad, CA) following the manufacturers recommended instructions. These data, shown in Table 62, show that modified mRNA encoding VEGF-A is capable of being translated in Human Keratinocyte cells and that VEGF-A is transported out of the cells and released into the extracellular environment.
Table 62. VEGF-A Dosing and Protein Secretion
Figure imgf000320_0001
93.75 12.37 46.38 121.23 167.56
187.5 104.55 365.71 1025.41 1056.91
375 605.89 1201.23 1653.63 1889.23
750 445.41 1036.45 1522.86 1954.81
1500 261.61 714.68 1053.12 1513.39
Example 39. In vivo studies of Factor IX
[001050] Human Factor IX mmRNA (Genl; fully modified 5-methycytosine and pseudouridine) formulated in formulation buffer was delivered to mice via intramuscular injection. The results demonstrate that Factor IX protein was elevated in serum as measured 13 hours after administration.
[001051] In this study, mice (N=5 for Factor IX, N=3 for Luciferase or Buffer controls) were intramuscularly injected with 50 μΐ of the Factor IX mmRNA (mRNA sequence shown in SEQ ID NO: 10; polyA tail of approximately 160 nucleotides not shown in sequence; 5 'cap, Capl),
Luciferase (cDNA sequence for IVT shown in SEQ ID NO: 15; mRNA sequence shown in SEQ ID NO: 16, polyA tail of approximately 160 nucleotides not shown in sequence, 5 'cap, Capl, fully modified with 5-methylcytosine at each cytosine and pseudouridine replacement at each uridine site) or the formulation buffer (F. Buffer) at 2x 100 ug/mouse. The mice were bled at 13 hours after the intramuscular injection to determine the concentration of human the polypeptide in serum in pg/mL. The results revealed that administration of Factor IX mmRNA resulted in levels of 1600 pg/mL at 13 hours as compared to less than 100 pg/mL of Factor IX for either Luciferase or buffer control administration.
Example 40. Multi-site administration: Intramuscular and Subcutaneous
[001052] Human G-CSF modified mRNA (mRNA sequence shown in SEQ ID NO: 6; polyA tail of approximately 160 nucleotides not shown in sequence; 5'cap, Capl) modified as either Genl or
Gen2 (5-methylcytosine (5mc) and a pseudouridine (ψ) modification, G-CSF-Genl; or Nl-5- methylcytosine (Nl-5mc) and a ψ modification, G-CSF-Gen2) and formulated in formulation buffer were delivered to mice via intramuscular (IM) or subcutaneous (SC) injection. Injection of four doses or 2x 50ug (two sites) daily for three days (24 hrs interval) was performed. The fourth dose was administered 6 hrs before blood collection and CBC analysis. Controls included Luciferase
(cDNA sequence for IVT shown in SEQ ID NO: 15; mRNA sequence shown in SEQ ID NO: 16, polyA tail of approximately 160 nucleotides not shown in sequence, 5'cap, Capl, fully modified with 5-methylcytosine at each cytosine and pseudouridine replacement at each uridine site) or the formulation buffer (F.Buffer). The mice were bled at 72 hours after the first mRNA injection (6 hours after the last mRNA dose) to determine the effect of mRNA-encoded human G-CSF on the neutrophil count. The dosing regimen is shown in Table 63 as are the resulting neutrophil counts (thousands/uL). In Table 63, an asterisks(*) indicate statistical significance at p<0.05.
[001053] For intramuscular administration, the data reveal a four fold increase in neutrophil count above control at day 3 for the Genl G-CSF mRNA and a two fold increase for the Gen2 G-CSF mmRNA. For subcutaneous administration, the data reveal a two fold increase in neutrophil count above control at day 3 for the Gen2 G-CSF mRNA.
[001054] These data demonstrate that both 5-methylcytidine/pseudouridine and 5- methylcytidine/Nl-methylpseudouridine-modified mRNA can be biologically active, as evidenced by specific increases in blood neutrophil counts.
Table 63. Dosing Regimen
Figure imgf000322_0001
Example 41. Intravenous administration
[001055] Human G-CSF modified mRNA (mRNA sequence shown in SEQ ID NO: 6; polyA tail of approximately 160 nucleotides not shown in sequence; 5'cap, Capl) modified with 5-methylcytosine (5mc) and a pseudouridine (ψ) modification (Genl); or having no modifications and formulated in 10% lipoplex (RNAiMax) were delivered to mice at a dose of 50 ug RNA and in a volume of 100 ul via intravenous (IV) injection at days 0, 2 and 4. Neutrophils were measured at days 1, 5 and 8. Controls included non-specific mammalian RNA or the formulation buffer alone (F.Buffer). The mice were bled at days 1, 5 and 8 to determine the effect of niRNA-encoded human G-CSF to increase neutrophil count. The dosing regimen is shown in Table 64 as are the resulting neutrophil counts (thousands/uL; K/uL). [001056] For intravenous administration, the data reveal a four to five fold increase in neutrophil count above control at day 5 with G-CSF modified mRNA but not with unmodified G-CSF mRNA or non-specific controls. Blood count returned to baseline four days after the final injection. No other changes in leukocyte populations were observed.
[001057] In Table 64, an asterisk^) indicates statistical significance at p<0.001 compared to buffer.
[001058] These data demonstrate that lipoplex-formulated 5-methylcytidine/pseudouridine -modified mRNA can be biologically active, when delivered through an I.V. route of administration as evidenced by specific increases in blood neutrophil counts. No other cell subsets were significantly altered. Unmodified G-CSF mRNA similarly administered showed no pharmacologic effect on neutrophil counts.
Table 64. Dosing Regimen
Figure imgf000323_0001
Example 42. Saline formulation: Intramuscular Administration
A. Protein Expression
[001059] Human G-CSF modified mRNA (mRNA sequence shown in SEQ ID NO: 6; polyA tail of approximately 160 nucleotides not shown in sequence; 5'cap, Capl) and human EPO mmRNA (mRNA sequence shown in SEQ ID NO: 9; polyA tail of approximately 160 nucleotides not shown in sequence; 5'cap, Capl); G-CSF modified mRNA (modified with 5-methylcytosine (5mc) and pseudouridine (ψ)) and EPO modified mRNA (modified with Nl -5-methylcytosine (Nl-5mc) and ψ modification), were formulated in formulation buffer (150 mM sodium chloride, 2mM calcium chloride, 2mM phosphate, 0.5 mM EDTA at a pH of 6.5) and delivered to mice via intramuscular (IM) injection at a dose of 100 ug. [001060] Controls included Luciferase (cDNA sequence for IVT, SEQ ID NO: 15; mRNA sequence shown in SEQ ID NO: 16, polyA tail of approximately 160 nucleotides not shown in sequence, 5'cap, Capl, fully modified with 5 -methylcytosme at each cytosine and pseudouridine replacement at each uridine site) or the formulation buffer (F.Buffer). The mice were bled at 13 hours after the injection to determine the concentration of the human polypeptide in serum in pg/mL (G-CSF groups measured human G-CSF in mouse serum and EPO groups measured human EPO in mouse serum). The data are shown in Table 65.
Table 65. Dosing Regimen
Figure imgf000324_0001
B. Dose Response
[001061] Human EPO modified mRNA (mRNA sequence shown in SEQ ID NO: 9; polyA tail of approximately 160 nucleotides not shown in sequence; 5'cap, Capl; fully modified with 5- methylcytosine and pseudouridine) were formulated in formulation buffer and delivered to mice via intramuscular (IM) injection.
[001062] Controls included Luciferase (mRNA sequence shown in SEQ ID NO: 16, polyA tail of approximately 160 nucleotides not shown in sequence, 5'cap, Capl, fully modified with 5- methylcytosine and pseudouridine) or the formulation buffer (F.Buffer). The mice were bled at 13 hours after the injection to determine the concentration of the human polypeptide in serum in pg/mL. The dose and expression are shown in Table 66.
Table 66. Dosing Regimen and Expression
Figure imgf000324_0002
Figure imgf000325_0001
Example 43. Muti-dose/Multi-administration
[001063] Studies utilizing multiple intramuscular injection sites at one time point were designed and performed.
[001064] The design of a single multi-dose experiment involved using human erythropoietin (EPO) mmR A (mR A sequence shown in SEQ ID NO: 9; polyA tail of approximately 160 nucleotides not shown in sequence; 5 'cap, Capl) or G-CSF (mRNA sequence shown in SEQ ID NO: 6; polyA tail of approximately 160 nucleotides not shown in sequence; 5 'cap, Capl) administered in formulation buffer. The dosing vehicle (F. buffer) was used as a control. The EPO and G-CSF mmRNA were modified with 5-methylcytosine at each cytosine and pseudouridine replacement at each uridine site.
[001065] Animals (n=5), Sprague-Dawley rats, were injected IM (intramuscular) for the single unit dose of 100 ug (delivered to one thigh). For multi-dosing 6 doses of 100 ug (delivered to two thighs) were used for both EPO and G-CSF mmRNA. Control dosing involved use of buffer at a single dose. Human EPO blood levels were evaluated 13 hrs post injection.
[001066] Human EPO protein was measured in rat serum 13h post I.M. Five groups of rats were treated and evaluated. The results are shown in Table 67.
Table 67. Multi-dose study
Figure imgf000325_0002
Example 44. Signal Sequence Exchange Study
[001067] Several variants of mmRNAs encoding human Granulocyte colony stimulating factor (G- CSF) (mRNA sequence shown in SEQ ID NO: 6; polyA tail of approximately 160 nucleotides not shown in sequence; 5'cap, Capl) were synthesized using modified nucleotides pseudouridine and 5- methylcytosine (pseudo-U/5mC). These variants included the G-CSF constructs encoding either the wild-type N terminal secretory signal peptide sequence (MAGPATQSPMKLMALQLLLWHSALWTVQEA; SEQ ID NO: 21), no secretory signal peptide sequence, or secretory signal peptide sequences taken from other mRNAs. These included sequences where the wild type GCSF signal peptide sequence was replaced with the signal peptide sequence of either: human α-1-anti trypsin (AAT) (MMPSSVSWGILLLAGLCCLVPVSLA; SEQ ID NO: 22), human Factor IX (FIX)
(MQRVNMIMAESPSLITICLLGYLLSAECTVFLDHENANKILNRPKR; SEQ ID NO: 23), human Prolactin (Prolac) (MKGSLLLLLVSNLLLCQSVAP; SEQ ID NO: 24), or human Albumin (Alb) (MKWVTFISLLFLFSSAYSPvGVFRR; SEQ ID NO: 25).
[001068] 250ng of modified mRNA encoding each G-CSF variant was transfected into HEK293A (293 A in the table), mouse myoblast (MM in the table) (C2C12, CRL-1772, ATCC) and rat myoblast (RM in the table) (L6 line, CRL-1458, ATCC) cell lines in a 24 well plate using 1 ul of Lipofectamine 2000 (Life Technologies), each well containing 300,000 cells. The supematants were harvested after 24 hrs and the secreted G-CSF protein was analyzed by ELISA using the Human G- CSF ELISA kit (Life Technologies). The data shown in Table 68 reveal that cells transfected with G-CSF mmRNA encoding the Albumin signal peptide secrete at least 12 fold more G-CSF protein than its wild type counterpart.
Table 68. Signal Peptide Exchange
Figure imgf000326_0001
Example 45. Cytokine Study: PBMC
[001069] PBMC isolation and Culture: 50 mL of human blood from two donors was received from Research Blood Components (lots KP30928 and KP30931) in sodium heparin tubes. For each donor, the blood was pooled and diluted to 70 mL with DPBS (SAFC Bioscience 59331C, lot 071M8408) and split evenly between two 50 mL conical tubes. 10 mL of Ficoll Paque (GE Healthcare 17-5442- 03, lot 10074400) was gently dispensed below the blood layer. The tubes were centrifuged at 2000 rpm for 30 minutes with low acceleration and braking. The tubes were removed and the buffy coat PBMC layers were gently transferred to a fresh 50 mL conical and washed with DPBS. The tubes were centrifuged at 1450 rpm for 10 minutes.
[001070] The supernatant was aspirated and the PBMC pellets were resuspended and washed in 50 mL of DPBS. The tubes were centrifuged at 1250 rpm for 10 minutes. This wash step was repeated, and the PBMC pellets were resuspended in 19 mL of Optimem I (Gibco 11058, lot 1072088) and counted. The cell suspensions were adjusted to a concentration of 3.0 x 10A6 cells / mL live cells.
[001071] These cells were then plated on five 96 well tissue culture treated round bottom plates (Costar 3799) per donor at 50 uL per well. Within 30 minutes, transfection mixtures were added to each well at a volume of 50 uL per well. After 4 hours post transfection, the media was
supplemented with 10 uL of Fetal Bovine Serum (Gibco 10082, lot 1012368)
[0010721 Transfection Preparation: mmRNA encoding human G-CSF (mR A sequence shown in SEQ ID NO: 6; polyA tail of approximately 160 nucleotides not shown in sequence; 5'cap, Capl) (containing either (1) natural NTPs, (2) 100% substitution with 5-methyl cytidine and pseudouridine, or (3) 100%) substitution with 5-methyl cytidine and Nl-methyl pseudouridine; mmRNA encoding luciferase (IVT cDNA sequence shown in SEQ ID NO: 15; mRNA sequence shown in SEQ ID NO: 16, polyA tail of approximately 160 nucleotides not shown in sequence, 5'cap, Capl, fully modified with 5-methylcytosine at each cytosine and pseudouridine replacement at each uridine site)
(containing either (1) natural NTPs or (2) 100% substitution with 5-methyl cytidine and
pseudouridine) and TLR agonist R848 (Invivogen tlrl-r848) were diluted to 38.4 ng / uL in a final volume of 2500 uL Optimem I.
[001073] Separately, 432 uL of Lipofectamine 2000 (Invitrogen 11668-027, lot 1070962) was diluted with 13.1 mL Optimem I. In a 96 well plate nine aliquots of 135 uL of each mmRNA, positive control (R-848) or negative control (Optimem I) was added to 135 uL of the diluted
Lipofectamine 2000. The plate containing the material to be trans fected was incubated for 20 minutes. The transfection mixtures were then transferred to each of the human PBMC plates at 50 uL per well. The plates were then incubated at 37 C. At 2, 4, 8, 20, and 44 hours each plate was removed from the incubator, and the supernatants were frozen.
[001074] After the last plate was removed, the supernatants were assayed using a human G-CSF ELISA kit (Invitrogen KHC2032) and human IFN-alpha ELISA kit (Thermo Scientific 41105-2). Each condition was done in duplicate. [0010751 Results: The ability of unmodified and modified mRNA (mmRNAs) to produce the encoded protein was assessed (G-CSF production) over time as was the ability of the mRNA to trigger innate immune recognition as measured by interferon-alpha production. Use of in vitro PBMC cultures is an accepted way to measure the immunostimulatory potential of oligonucleotides (Robbins et al, Oligonucleotides 2009 19:89-102).
[001076] Results were interpolated against the standard curve of each ELISA plate using a four parameter logistic curve fit. Shown in Tables 69 and 70 are the average from 2 separate PBMC donors of the G-CSF and IFN-alpha production over time as measured by specific ELISA.
[001077] In the G-CSF ELISA, background signal from the Lipofectamine 2000 untreated condition was subtracted at each timepoint. The data demonstrated specific production of human G-CSF protein by human peripheral blood mononuclear is seen with G-CSF mRNA containing natural NTPs, 100% substitution with 5-methyl cytidine and pseudouridine, or 100% substitution with 5- methyl cytidine and Nl -methyl pseudouridine. Production of G-CSF was significantly increased through the use of modified mRNA relative to unmodified mRNA, with the 5-methyl cytidine and Nl-methyl pseudouridine containing G-CSF mmRNA showing the highest level of G-CSF production. With regards to innate immune recognition, unmodified mRNA resulted in substantial IFN-alpha production, while the modified mRNA largely prevented interferon-alpha production. G- CSF mRNA fully modified with 5-methyl cytidine and Nl-methylpseudouridine did not significantly increase cytokines whereas G-CSF mRNA fully modified with 5-methyl cytidine and pseudouridine induced IFN-alpha, TNF-alpha and IP 10. Many other cytokines were not affected by either modification.
Table 69. G-CSF Signal
Figure imgf000328_0001
Table 70. IFN-alpha signal
Figure imgf000328_0002
pseudouridine)
G-CSF(Natural) 0.0 2.1 23.3 74.9 119.7
Luciferase (5mC/pseudouridine) 0.4 0.4 4.7 1.0 2.4
R-848 39.1 151.3 278.4 362.2 208.1
Lpf. 2000 control 0.8 17.2 16.5 0.7 3.1
Example 46. Chemical modification ranges of modified mRNA
[001078] Modified nucleotides such as, but not limited to, the chemical modifications 5- methylcytosine and pseudouridine have been shown to lower the innate immune response and increase expression of R A in mammalian cells. Surprisingly, and not previously known, the effects manifested by the chemical modifications can be titrated when the amount of chemical modification is less than 100%. Previously, it was believed that full modification was necessary and sufficient to elicit the beneficial effects of the chemical modifications and that less than 100% modification of an mRNA had little effect. However, it has now been shown that the benefits of chemical modification can be derived using less than complete modification and that the effects are target, concentration and modification dependent.
A. Modified RNA transfected in PBMC
[001079] 960 ng of G-CSF mRNA modified with 5-methylcytosine (5mC) and pseudouridine (pseudoU) or unmodified G-CSF mRNA was transfected with 0.8 uL of Lipofectamine 2000 into peripheral blood mononuclear cells (PBMC) from three normal blood donors (Dl , D2, D3). The G- CSF mRNA (SEQ ID NO: 6; polyA tail of approximately 160 nucleotides not shown in sequence; 5 'cap, Capl) was completely modified with 5mC and pseudoU (100% modification), not modified with 5mC and pseudoU (0%> modification) or was partially modified with 5mC and pseudoU so the mRNA would contain 50% modification, 25% modification, 10% modification, %5 modification, 1% modification or 0.1% modification. A control sample of Luciferase (mRNA sequence shown in SEQ ID NO: 16; polyA tail of approximately 160 nucleotides not shown in sequence; 5 'cap, Capl ; fully modified 5meC and pseudoU) was also analyzed for G-CSF expression. For TNF-alpha and IFN-alpha control samples of Lipofectamine2000, LPS, R-848, Luciferase (mRNA sequence shown in SEQ ID NO: 16; polyA tail of approximately 160 nucleotides not shown in sequence; 5 'cap, Capl ; fully modified 5mC and pseudo), and P(I)P(C) were also analyzed. The supernatant was harvested and run by ELISA 22 hours after transfection to determine the protein expression. The expression of G-CSF is shown in Table 71 and the expression of IFN-alpha and TNF-alpha is shown in Table 72. The expression of IFN-alpha and TNF-alpha may be a secondary effect from the transfection of the G-CSF mRNA. Tables and shows that the amount of chemical modification of G- CSF, IFN-alpha and TNF-alpha is titratable when the mRNA is not fully modified and the titratable trend is not the same for each target.
Table 71. G-CSF Expression
Figure imgf000330_0001
Table 72. IFN-alpha and TNF-alpha Expression
Figure imgf000330_0002
B. Modified RNA transfected in HEK293
[001080] Human embryonic kidney epithelial (HEK293) cells were seeded on 96-well plates at a density of 30,000 cells per well in 100 ul cell culture medium. 250 ng of modified G-CSF mRNA (SEQ ID NO: 6; polyA tail of approximately 160 nucleotides not shown in sequence; 5'cap, Capl) formulated with RNAiMAX™ (Invitrogen, Carlsbad, CA) was added to a well. The G-CSF was completely modified with 5mC and pseudoU (100% modification), not modified with 5mC and pseudoU (0%> modification) or was partially modified with 5mC and pseudoU so the mRNA would contain 75% modification, 50% modification or 25% modification. Control samples (AK 5/2, mCherry (SEQ ID NO: 7; polyA tail of approximately 160 nucleotides not shown in sequence; 5 'cap, Capl; fully modified 5mC and pseudoU) and untreated) were also analyzed. The half-life of G-CSF mRNA fully modified with 5-methylcytosine and pseudouridine is approximately 8-10 hours. The supernatants were harvested after 16 hours and the secreted G-CSF protein was analyzed by ELISA. Table 73 shows that the amount of chemical modification of G-CSF is titratable when the mRNA is not fully modified.
Table 73. G-CSF Expression
Figure imgf000331_0001
Example 47: In Vivo Delivery of Modified mRNA (mniRNA)
[001081] Modified RNA was delivered to C57/BL6 mice intramuscularly, subcutaneously, or intravenously to evaluate the bio-distribution of modified RNA using luciferase. A formulation buffer used with all delivery methods contained 150mM sodium chloride, 2mM calcium chloride, 2 mM Na+-phosphate which included 1.4mM monobasic sodium phosphate and 0.6 mM of dibasic sodium phosphate, and 0.5 mM ethylenediaminetetraacetic acid (EDTA) was adjusted using sodium hydroxide to reach a final pH of 6.5 before being filtered and sterilized. A IX concentration was used as the delivery buffer. To create the lipoplexed solution delivered to the mice, in one vial 50 μg of RNA was equilibrated for 10 minutes at room temperature in the delivery buffer and in a second vial 10 μΐ RNAiMAX™ was equilibrated for 10 minutes at room temperature in the delivery buffer. After equilibrium, the vials were combined and delivery buffer was added to reach a final volume of 100 μΐ which was then incubated for 20 minutes at room temperature. Luciferin was administered by intraperitoneal injection (IP) at 150 mg/kg to each mouse prior to imaging during the plateau phase of the luciferin exposure curve which was between 15 and 30 minutes. To create luciferin, 1 g of D-luciferin potassium or sodium salt was dissolved in 66.6 ml of distilled phosphate buffer solution (DPBS), not containing Mg2+ or Ca2+, to make a 15 mg/ml solution. The solution was gently mixed and passed through a 0.2 μιη syringe filter, before being purged with nitrogen, aliquoted and frozen at -80°C while being protected from light as much as possible. The solution was thawed using a waterbath if luciferin was not dissolved, gently mixed and kept on ice on the day of dosing.
[001082] Whole body images were taken of each mouse 2, 8 and 24 hours after dosing. Tissue images and serum was collected from each mouse 24 hours after dosing. Mice administered doses intravenously had their liver, spleen, kidneys, lungs, heart, peri-renal adipose tissue and thymus imaged. Mice administered doses intramuscularly or subcutaneously had their liver, spleen, kidneys, lungs, peri-renal adipose tissue, and muscle at the injection site. From the whole body images the bioluminescence was measured in photon per second for each route of administration and dosing regimen.
A. Intramuscular Administration
[001083] Mice were intramuscularly (I.M.) administered either modified luciferase m NA fully modified with 5-methylcytosine and pseudouridine (Naked-Luc), lipoplexed modified luciferase mRNA fully modified with 5-methylcytosine and pseudouridine (Lipoplex-luc) (IVT cDNA sequence shown in SEQ ID NO: 15; mRNA sequence shown in SEQ ID NO: 16, polyA tail of approximately 160 nucleotides not shown in sequence, 5'cap, Capl, fully modified with 5- methylcytosine at each cytosine and pseudouridine replacement at each uridine site), lipoplexed modified granulocyte colony-stimulating factor (G-CSF) mRNA (mRNA sequence shown in SEQ ID NO: 6; polyA tail of approximately 160 nucleotides not shown in sequence; 5'cap, Capl; fully modified with 5-methylcytosine and pseudouridine) (Lipoplex-Cytokine) or the formation buffer at a single dose of 50 μg of modified RNA in an injection volume of 50μ1 for each formulation in the right hind limb and a single dose of 5 μg of modified RNA in an injection volume of 50 μΐ in the left hind limb. The bioluminescence average for the luciferase expression signals for each group at 2, 8 and 24 hours after dosing are shown in Table 74. The bioluminescence showed a positive signal at the injection site of the 5 μg and 50 μg modified RNA formulations containing and not containing lipoplex.
Table 74. In vivo Biophotoic Imaging (I.M. Injection Route)
Figure imgf000332_0001
Formulation Buffer 50 59,300 69,200 63,600
B. Subcutaneous Administration
[001084] Mice were subcutaneously (S.C.) administered either modified luciferase mRNA (Naked- Luc), lipoplexed modified luciferase mRNA (Lipoplex-luc), lipoplexed modified G-CSF mRNA (Lipoplex-G-CSF) or the formation buffer at a single dose of 50 μg of modified mRNA in an injection volume of 100 μΐ for each formulation. The bio luminescence average for the luciferase expression signals for each group at 2, 8 and 24 hours after dosing are shown in Table 75. The bio luminescence showed a positive signal at the injection site of the 50 μg modified mRNA formulations containing and not containing lipoplex.
Table 75. In vivo Biophotoic Imaging (S.C. Injection Route)
Figure imgf000333_0001
C. Intravenous Administration
[001085] Mice were intravenously (I.V.) administered either modified luciferase mRNA (Naked- Luc), lipoplexed modified luciferase mRNA (Lipoplex-luc), lipoplexed modified G-CSF mRNA (Lipoplex-G-CSF) or the formation buffer at a single dose of 50 μg of modified mRNA in an injection volume of 100 μΐ for each formulation. The bioluminescence average for the luciferase expression signal in the spleen from each group at 2 hours after dosing is shown in Table 76. The bioluminescence showed a positive signal in the spleen of the 50 μg modified mRNA formulations containing lipoplex.
Table 76 In vivo Biophotoic Imaging (I.V. Injection Route)
Figure imgf000333_0002
Example 48. Split dose studies [001086] Studies utilizing multiple subcutaneous or intramuscular injection sites at one time point were designed and performed to investigate ways to increase mmRNA drug exposure and improve protein production. In addition to detection of the expressed protein product, an assessment of the physiological function of proteins was also determined through analyzing samples from the animal tested.
[001087] Surprisingly, it has been determined that split dosing of mmRNA produces greater protein production and phenotypic responses than those produced by single unit dosing or multi-dosing schemes.
[001088] The design of a single unit dose, multi-dose and split dose experiment involved using human erythropoietin (EPO) modified mRNA (mRNA shown in SEQ ID NO: 9; polyA tail of approximately 160 nucleotides not shown in sequence; 5'cap, Capl) administered in buffer alone. The dosing vehicle (F. buffer) consisted of 150mM NaCl, 2 mM CaCl2, 2 mM Na+-phosphate (1.4mM monobasic sodium phosphate; 0.6mM dibasic sodium phosphate), and 0.5 mM EDTA, pH 6.5. The pH was adjusted using sodium hydroxide and the final solution was filter sterilized. The mmRNA was modified with 5meC at each cytosine and pseudouridine replacement at each uridine site.
[001089] Animals (n=5) were injected IM (intramuscular) for the single unit dose of 100 ug. For multi-dosing, two schedules were used, 3 doses of 100 ug and 6 doses of 100 ug. For the split dosing scheme, two schedules were used, 3 doses at 33.3 ug and 6 doses of 16.5 ug mmRNA. Control dosing involved use of buffer only at 6 doses. Control mmRNA involved the use of luciferase mmRNA (IVT cDNA sequence shown in SEQ ID NO: 15; mRNA sequence shown in SEQ ID NO: 16; polyA tail of approximately 160 nucleotides not shown in sequence; 5'cap, Capl; fully modified 5meC at each cytosine and pseudouridine replacement at each uridine site) dosed 6 times at 100 ug. Blood and muscle tissue were evaluated 13 hrs post injection.
[001090] Human EPO protein was measured in mouse serum 13h post I.M. single, multi- or split dosing of the EPO mmRNA in buffer. Seven groups of mice (n=5 mice per group) were treated and evaluated. The results are shown in Table 77.
Table 77. Split dose study
Figure imgf000334_0001
Figure imgf000335_0001
7 Buffer Alone — — 0 — —
[001091] The splitting factor is defined as the product per unit drug divided by the single dose product per unit drug (PUD). For example for treatment group 2 the value .28 or product (EPO) per unit drug (mmRNA) is divided by the single dose product per unit drug of 0.14. The result is 2. Likewise, for treatment group 4, the value 1.1 or product (EPO) per unit drug (mmRNA) is divided by the single dose product per unit drug of 0.14. The result is 7.9. Consequently, the dose splitting factor (DSF) may be used as an indicator of the efficacy of a split dose regimen. For any single administration of a total daily dose, the DSF should be equal to 1. Therefore any DSF greater than this value in a split dose regimen is an indication of increased efficacy.
[001092] To determine the dose response trends, impact of injection site and impact of injection timing, studies are performed. In these studies, varied doses of lug, 5ug, 10 ug, 25 ug, 50 ug, and values in between are used to determine dose response outcomes. Split dosing for a 100 ug total dose includes three or six doses of 1.6 ug, 4.2 ug, 8.3 ug, 16.6 ug, or values and total doses equal to administration of the total dose selected.
[001093] Injection sites are chosen from the limbs or any body surface presenting enough area suitable for injection. This may also include a selection of injection depth to target the dermis (Intradermal), epidermis (Epidermal), subcutaneous tissue (SC) or muscle (IM). Injection angle will vary based on targeted delivery site with injections targeting the intradermal site to be 10-15 degree angles from the plane of the surface of the skin, between 20-45 degrees from the plane of the surface of the skin for subcutaneous injections and angles of between 60-90 degrees for injections substantially into the muscle.
Example 49. Quantification in Exosomes
[001094] The quantity and localization of the mmRNA of the present invention can be determined by measuring the amounts (initial, timecourse, or residual basis) in isolated exosomes. In this study, since the mmRNA are typically codon-optimized and distinct in sequence from endogenous mRNA, the levels of mmRNA are quantitated as compared to endogenous levels of native or wild type mRNA by using the methods of Gibbings, PCT/IB2009/005878, the contents of which are incorporated herein by reference in their entirety.
[001095] In these studies, the method is performed by first isolating exosomes or vesicles preferably from a bodily fluid of a patient previously treated with a modified mmRNA of the invention, then measuring, in said exosomes, the modified mmRNA levels by one of mRNA microarray, qRT-PCR, or other means for measuring RNA in the art including by suitable antibody or
immunohistochemical methods.
Example 50. Modified mRNA Transfection
A. Reverse Transfection
[001096] For experiments performed in a 24-well collagen-coated tissue culture plate, Keratinocytes are seeded at a cell density of 1 x 105. For experiments performed in a 96-well collagen-coated tissue culture plate, Keratinocytes are seeded at a cell density of 0.5 x 105. For each modified mRNA (mmRNA) to be transfected, modified mRNA: RNAIMAX™ is prepared as described and mixed with the cells in the multi-well plate within a period of time, e.g., 6 hours, of cell seeding before cells had adhered to the tissue culture plate.
B. Forward Transfection
[001097] In a 24-well collagen-coated tissue culture plate, Keratinocytes are seeded at a cell density of 0.7 x 105. For experiments performed in a 96-well collagen-coated tissue culture plate,
Keratinocytes are seeded at a cell density of 0.3 x 105. Keratinocytes are grown to a confluency of >70% for over 24 hours. For each modified mRNA (mmRNA) to be transfected, modified mRNA: RNAIMAX™ is prepared as described and transfected onto the cells in the multi-well plate over 24 hours after cell seeding and adherence to the tissue culture plate.
C. Modified mRNA Translation Screen: G-CSF ELISA
[001098] Keratinocytes are grown in EPILIFE medium with Supplement S7 from Invitrogen
(Carlsbad, CA) at a confluence of >70%. One set of keratinocytes were reverse transfected with 300 ng of the chemically modified mRNA (mmRNA) complexed with RNAIMAX™ from Invitrogen. Another set of keratinocytes are forward transfected with 300 ng modified mRNA complexed with RNAIMAX™ from Invitrogen. The modified mRNA: RNAIMAX™ complex is formed by first incubating the RNA with Supplement- free EPILIFE® media in a 5X volumetric dilution for 10 minutes at room temperature. [001099] In a second vial, RNAIMAX reagent was incubated with Supplement-free EPILIFE® Media in 10X volumetric dilution for 10 minutes at room temperature. The R A vial was then mixed with the RNAIMAX™ vial and incubated for 20-30 minutes at room temperature before being added to the cells in a drop-wise fashion. Secreted human Granulocyte-Colony Stimulating Factor (G-CSF) concentration in the culture medium is measured at 18 hours post-transfection for each of the chemically modified mRNA in triplicate.
[001100] Secretion of Human G-CSF from trans fected human keratinocytes is quantified using an
ELISA kit from Invitrogen or R&D Systems (Minneapolis, MN) following the manufacturers recommended instructions.
D. Modified mRNA Dose and Duration: G-CSF ELISA
[001101] Keratinocytes are grown in EPILIFE® medium with Supplement S7 from Invitrogen at a confluence of >70%. Keratinocytes are reverse transfected with either Ong, 46.875ng, 93.75ng,
187.5ng, 375ng, 750ng, or 1500ng modified mRNA complexed with the RNAIMAX™ from
Invitrogen (Carlsbad, CA). The modified mRNA: RNAIMAX™ complex is formed as described.
Secreted human G-CSF concentration in the culture medium is measured at 0, 6, 12, 24, and 48 hours post-transfection for each concentration of each modified mRNA in triplicate. Secretion of human G-CSF from transfected human keratinocytes is quantified using an ELISA kit from
Invitrogen or R&D Systems following the manufacturers recommended instructions.
Example 51. Detection of a Cellular Innate Immune Response to Modified mRNA Using an ELISA Assay
[001102] An enzyme-linked immunosorbent assay (ELISA) for Human Tumor Necrosis Factor-a (TNF-a), Human Interferon-β (IFN-β) and Human Granulocyte-Colony Stimulating Factor (G-CSF) secreted from in vzYro-transfected Human Keratinocyte cells is tested for the detection of a cellular innate immune response. Keratinocytes are grown in EPILIFE® medium with Human Keratinocyte Growth Supplement in the absence of hydrocortisone from Invitrogen (Carlsbad, CA) at a confluence of >70%. Secreted TNF-a keratinocytes are reverse transfected with Ong, 93.75ng, 1 87.5ng, 375ng, 750ng, 1500ng or 3000ng of the chemically modified mRNA (mmRNA) complexed with RNAIMAX™ from Invitrogen as described in triplicate. Secreted TNF-a in the culture medium is measured 24 hours post-transfection for each of the chemically modified mRNA using an ELISA kit from Invitrogen according to the manufacturer protocols.
[001103] Secreted IFN-β in the same culture medium is measured 24 hours post-transfection for each of the chemically modified mRNA using an ELISA kit from Invitrogen according to the manufacturer protocols. Secreted human G-CSF concentration in the same culture medium is measured at 24 hours post-transfection for each of the chemically modified mRNA. Secretion of human G-CSF from transfected human keratinocytes is quantified using an ELISA kit from
Invitrogen or R&D Systems (Minneapolis, MN) following the manufacturers recommended instructions. These data indicate which modified mRNA (mmRNA) are capable eliciting a reduced cellular innate immune response in comparison to natural and other chemically modified
polynucleotides or reference compounds by measuring exemplary type 1 cytokines TNF-a and IFN- β·
Example 52. Human Granulocyte -Colony Stimulating Factor (G-CSF) Modified mRNA- induced Cell Proliferation Assay
[001104] Human keratinocytes are grown in EPILIFE® medium with Supplement S7 from
Invitrogen at a confluence of >70% in a 24-well collagen-coated TRANSWELL® (Coming, Lowell, MA) co-culture tissue culture plate. Keratinocytes are reverse transfected with 750ng of the indicated chemically modified mRNA (mmRNA) complexed with RNAIMAX from Invitrogen as described in triplicate. The modified mRNA:RNAIMAX complex is formed as described.
Keratinocyte media is exchanged 6-8 hours post-transfection. 42-hours post-transfection, the 24-well TRANSWELL® plate insert with a 0.4 μιη-pore semi-permeable polyester membrane is placed into the human GCSF modified mRNA-transfected keratinocyte containing culture plate
[001105] Human myeloblast cells, Kasumi-1 cells or KG- 1 (0.2 x 105 cells), are seeded into the insert well and cell proliferation is quantified 42 hours post-co-culture initiation using the CyQuant Direct Cell Proliferation Assay (Invitrogen, Carlsbad, CA) in a 100-120μ1 volume in a 96-well plate. Modified mRNA-encoding human G-CSF-induced myeloblast cell proliferation is expressed as a percent cell proliferation normalized to untransfected keratinocyte/myeloblast co-culture control wells. Secreted human G-CSF concentration in both the keratinocyte and myeloblast insert co- culture wells is measured at 42 hours post-co-culture initiation for each modified mRNA in duplicate. Secretion of human G-CSF is quantified using an ELISA kit from Invitrogen following the manufacturer recommended instructions.
[001106] Transfected human G-CSF modified mRNA in human keratinocyte feeder cells and untransfected human myeloblast cells are detected by RT -PCR. Total RNA from sample cells is extracted and lysed using RNEASY® kit (Qiagen, Valencia, CA) according to the manufacturer instructions. Extracted total RNA is submitted to RT-PCR for specific amplification of modified mRNA-G-CSF using PROTOSCRIPT® M-MuLV Tag RT-PCR kit (New England BioLabs, Ipswich, MA) according to the manufacturer instructions with human G-CSF-specific primers. RT- PCR products are visualized by 1.2% agarose gel electrophoresis.
Example 53. Buffer Formulation Studies
[001107] G-CSF modified mRNA (SEQ ID NO: 6; polyA tail of approximately 160 nucleotides not shown in sequence; 5 'cap, Capl ; fully modified with Nl-pseudouridine and 5-methylcytosine) or Factor IX modified mRNA (SEQ ID NO: 10; polyA tail of approximately 160 nucleotides not shown in sequence; 5 'cap, Capl ; fully modified with Nl-pseudouridine and 5-methylcytosine) in a buffer solution is administered intramuscularly to rats in an injection volume of 50 μΐ (n=5) at a modified mRNA dose of 200 ug per rat as described in Table 78. The modified mRNA is lyophilized in water for 1-2 days. It is then reconstituted in the buffers listed below to a target concentration of 6 mg/ml. Concentration is determined by OD 260. Samples are diluted to 4 mg/ml in the appropriate buffer before dosing.
[001108] To precipitate the modified mRNA, 3M sodium acetate, pH 5.5 and pure ethanol are added at 1/10th the total volume and 4 times the total volume of modified mRNA, respectively. The material is placed at -80C for a minimum of 1 hour. The material is then centrifuged for 30 minutes at 4000 rpm, 4C. The supernatant is removed and the pellet is centrifuged and washed 3x with 75% ethanol. Finally, the pellet is reconstituted with buffer to a target concentration of 6 mg/ml.
Concentration is determined by OD 260. Samples are diluted to 4 mg/ml in the appropriate buffer before dosing. All samples are prepared by lyophilization unless noted below.
Table 78. Buffer Dosing Groups
Figure imgf000339_0001
Factor IX 0.9% saline (precipitation) 200
[001109] Serum samples are collected from the rats at various time intervals and analyzed for G-CSF or Factor IX protein expression using G-CSF or Factor IX ELISA.
Example 54. Multi-Dose Study
[001110] Sprague-Dawley rats (n=8; 4 female, 4 male) are injected intravenously eight times (twice a week) over 28 days. The rats are injected with 0.5 mg/kg, 0.05 mg/kg, 0.005 mg/kg or 0.0005 mg/kg of human G-CSF modified mRNA of luciferase modified mRNA formulated in a lipid nanoparticle, 0.5 mg/kg of human G-CSF modified mRNA in saline, 0.2 mg/kg of the human G-CSF protein Neupogen or non-translatable human G-CSF modified mRNA formulated in a lipid nanoparticle. Serum is collected during pre-determined time intervals to evaluate G-CSF protein expression (8, 24 and 72 hours after the first dose of the week), complete blood count and white blood count (24 and 72 hours after the first dose of the week) and clinical chemistry (24 and 72 hours after the first dose of the week). The rats are sacrificed at day 29, 4 days after the final dosing, to determine the complete blood count, white blood count, clinical chemistry, protein expression and to evaluate the effect on the major organs by histopathology and necropsy. Further, an antibody assay is performed on the rats on day 29.
Example 55. Luciferase LNP in vivo study
[001111] Luciferase modified mRNA (SEQ ID NO: 16; polyA tail of approximately 160 nucleotides not shown in sequence, 5' cap, Capl; fully modified with 5-methylcytosine and pseudouridine was formulated as a lipid nanoparticle (LNP) using the syringe pump method. The LNP was formulated at a 20: 1 weight ratio of total lipid to modified mRNA with a final lipid molar ratio of 50: 10:38.5: 1.5 (DLin-KC2-DMA: DSPC: Cholesterol: PEG-DMG). As shown in Table 79, the luciferase LNP formulation was characterized by particle size, zeta potential, and encapsulation.
Table 79. Luciferase Formulation
Figure imgf000340_0001
[001112] As outlined in Table 80, the luciferase LNP formulation was administered to Balb-C mice (n=3) intramuscularly, intravenously and subcutaneously and a luciferase modified RNA formulated in PBS was administered to mice intravenously.
Table 80. Luciferase Formulations
Figure imgf000341_0001
[001113] The mice administered the luciferase LNP formulation intravenously and intramuscularly were imaged at 2, 8, 24, 48, 120 and 192 hours and the mice administered the luciferase LNP formulation subcutaneously were imaged at 2, 8, 24, 48 and 120 hours to determine the luciferase expression as shown in Table 81. In Table 81 , "NT" means not tested. Twenty minutes prior to imaging, mice were injected intraperitoneally with a D-luciferin solution at 150 mg/kg. Animals were then anesthetized and images were acquired with an IVIS Lumina II imaging system (Perkin Elmer). Bioluminescence was measured as total flux (photons/second) of the entire mouse.
Table 81. Luciferase Expression
Figure imgf000341_0002
[001114] One mouse administered the LNP formulation intravenously was sacrificed at 8 hours to determine the luciferase expression in the liver and spleen. Also, one mouse administered the LNP formulation intramuscular was sacrificed at 8 hours to determine the luciferase expression of the muscle around the injection site and in the liver and spleen. As shown in Table 82, expression was seen in the both the liver and spleen after intravenous and intramuscular administration and in the muscle around the intramuscular injection site.
Table 82. Luciferase Expression in Tissue
Figure imgf000341_0003
Administration (photon/second)
Muscle around the
3.688E+07
injection site
Liver 1.507E+08
Spleen 1.096E+07
Example 56. In Vitro PBMC Studies: Percent modification
[001115] 480 ng of G-CSF mRNA modified with 5-methylcytosine (5mC) and pseudouridine (pseudoU) or unmodified G-CSF mRNA was transfected with 0.4 uL of Lipofectamine 2000 into peripheral blood mononuclear cells (PBMC) from three normal blood donors (Dl, D2, and D3). The G-CSF mRNA (SEQ ID NO: 6; polyA tail of approximately 160 nucleotides not shown in sequence; 5'cap, Capl) was completely modified with 5mC and pseudoU (100% modification), not modified with 5mC and pseudoU (0% modification) or was partially modified with 5mC and pseudoU so the mRNA would contain 75% modification, 50% modification or 25% modification. A control sample of Luciferase (mRNA sequence shown in SEQ ID NO: 16; polyA tail of approximately 160 nucleotides not shown in sequence; 5'cap, Capl; fully modified 5meC and pseudoU) was also analyzed for G-CSF expression. For TNF-alpha and IFN-alpha control samples of
Lipofectamine2000, LPS, R-848, Luciferase (mRNA sequence shown in SEQ ID NO: 16; polyA tail of approximately 160 nucleotides not shown in sequence; 5'cap, Capl; fully modified 5mC and pseudo), and P(I)P(C) were also analyzed. The supernatant was harvested and run by ELISA 22 hours after transfection to determine the protein expression. The expression of G-CSF is shown in Table 83 and the expression of IFN-alpha and TNF-alpha is shown in Table 84. The expression of IFN-alpha and TNF-alpha may be a secondary effect from the transfection of the G-CSF mRNA. Tables 83 and 84 show that the amount of chemical modification of G-CSF, interferon alpha (IFN- alpha) and tumor necrosis factor-alpha (TNF-alpha) is titratable when the mRNA is not fully modified and the titratable trend is not the same for each target.
[001116] As mentioned above, using PBMC as an in vitro assay system it is possible to establish a correlation between translation (in this case G-CSF protein production) and cytokine production (in this case exemplified by IFN-alpha protein production). Better protein production is correlated with lower induction of innate immune activation pathway, and the percentage modification of a chemistry can be judged favorably based on this ratio (Table 85). As calculated from Tables 83 and 84 and shown in Table 85, full modification with 5-methylcytidine and pseudouridine shows a much better ratio of protein/cytokine production than without any modification (natural G-CSF mRNA) (100-fold for IFN-alpha and 27-fold for TNF-alpha). Partial modification shows a linear relationship with increasingly less modification resulting in a lower protein/cytokine ratio.
Table 83. G-CSF Expression
Figure imgf000343_0001
Table 84. IFN-alpha and TNF-alpha Expression
Figure imgf000343_0002
Table 85. PC Ratio and Effect of Percentage of Modification
Figure imgf000343_0003
Example 57. Modified RNA transfected in PBMC
[001117] 500 ng of G-CSF mRNA modified with 5-methylcytosine (5mC) and pseudouridine (pseudoU) or unmodified G-CSF mRNA was transfected with 0.4 uL of Lipofectamine 2000 into peripheral blood mononuclear cells (PBMC) from three normal blood donors (Dl, D2, and D3). The G-CSF mRNA (SEQ ID NO: 6; polyA tail of approximately 160 nucleotides not shown in sequence; 5'cap, Capl) was completely modified with 5mC and pseudoU (100% modification), not modified with 5mC and pseudoU (0% modification) or was partially modified with 5mC and pseudoU so the mR A would contain 50% modification, 25% modification, 10% modification, %5 modification, 1% modification or 0.1% modification. A control sample of mCherry (mRNA sequence shown in SEQ ID NO: 7; polyA tail of approximately 160 nucleotides not shown in sequence; 5 'cap, Capl ; fully modified 5meC and pseudouridine) and G-CSF fully modified with 5-methylcytosine and pseudouridine (Control G-CSF) was also analyzed for G-CSF expression. For tumor necrosis factor- alpha (TNF-alpha) and interferon-alpha (IFN-alpha) control samples of Lipofectamine2000, LPS, R- 848, Luciferase (mRNA sequence shown in SEQ ID NO: 16; polyA tail of approximately 160 nucleotides not shown in sequence; 5 'cap, Capl ; fully modified 5mC and pseudo), and P(I)P(C) were also analyzed. The supernatant was harvested 6 hours and 18 hours after transfection and run by ELISA to determine the protein expression. The expression of G-CSF, IFN-alpha, and TNF-alpha for Donor 1 is shown in Table 86, Donor 2 is shown in Table 87 and Donor 3 is shown in Table 88.
[001118] Full 100% modification with 5-methylcytidine and pseudouridine resulted in the most protein translation (G-CSF) and the least amount of cytokine produced across all three human PBMC donors. Decreasing amounts of modification results in more cytokine production (IFN-alpha and TNF-alpha), thus further highlighting the importance of fully modification to reduce cytokines and to improve protein translation (as evidenced here by G-CSF production).
Table 86. Donor 1
Figure imgf000344_0001
Table 87. Donor 2
Figure imgf000344_0002
10% Mod 203 223 25 700 22 39
1% Mod 288 275 27 962 51 66
0.1% Mod 318 288 33 635 28 5
0 % Mod 389 413 26 748 1 253
Control G-CSF 1461 1634 1 59 481 814
mCherry 0 7 0 1 0 0
Untreated N/A N/A 1 0 0 0
Table 88. Donor 3
G-CSF (pg/mL) IFN-alpha (pg/mL) TNF-alpha (pg/mL)
6 hours 18 hours 6 hours 18 hours 6 hours 18 hours
100% Mod 6086 7549 7 658 11 11
75% Mod 2479 2378 23 752 4 35
50% Mod 667 774 24 896 22 18
25% Mod 480 541 57 1557 43 115
10% Mod 838 956 159 2755 144 123
1% Mod 1 108 1 197 235 3415 88 270
0.1% Mod 1338 1 177 191 2873 37 363
0 % Mod 1463 1666 215 3793 74 429
Control G-CSF 3272 3603 16 1557 731 9066
mCherry 0 0 2 645 0 0
Untreated N/A N/A 1 1 0 8
Example 58. Innate Immune Response Study in BJ Fibroblasts
A. Single Transfection
[001119] Human primary foreskin fibroblasts (BJ fibroblasts) were obtained from American Type Culture Collection (ATCC) (catalog # CRL-2522) and grown in Eagle's Minimum Essential Medium (ATCC, catalog # 30-2003) supplemented with 10% fetal bovine serum at 37°C, under 5% C02. BJ fibroblasts were seeded on a 24-well plate at a density of 300,000 cells per well in 0.5 ml of culture medium. 250 ng of modified G-CSF mRNA (mRNA sequence shown in SEQ ID NO: 6; polyA tail of approximately 140 nucleotides not shown in sequence; 5 'cap, Capl) fully modified with 5-methylcytosine and pseudouridine (Genl) or fully modified with 5-methylcytosine and Nl- methylpseudouridine (Gen2) having CapO, Capl or no cap was transfected using Lipofectamine 2000 (Invitrogen, catalog # 1 1668-019), following manufacturer's protocol. Control samples of poly I:C (PIC), Lipofectamine 2000 (Lipo), natural luciferase mRNA (mRNA sequence shown in SEQ ID NO: 16; polyA tail of approximately 160 nucleotides not shown in sequence; 5 'cap, Capl) and natural G-CSF mRNA were also transfected. The cells were harvested after 18 hours, the total RNA was isolated and DNASE® treated using the RNeasy micro kit (catalog #74004) following the manufacturer's protocol. 100 ng of total RNA was used for cDNA synthesis using High Capacity cDNA Reverse Transcription kit (catalog #4368814) following the manufacturer's protocol. The cDNA was then analyzed for the expression of innate immune response genes by quantitative real time PCR using SybrGreen in a Biorad CFX 384 instrument following manufacturer's protocol. Table 89 shows the expression level of innate immune response transcripts relative to house-keeping gene HPRT (hypoxanthine phosphoribosytransferase) and is expressed as fold-induction relative to HPRT. In the table, the panel of standard metrics includes: RIG-I is retinoic acid inducible gene 1, IL6 is interleukin-6, OAS-1 is oligoadenylate synthetase 1, IFNb is interferon-beta, AIM2 is absent in melanoma-2, IFIT-1 is interferon-induced protein with tetratricopeptide repeats 1, PKR is protein kinase R, TNFa is tumor necrosis factor alpha and IFNa is interferon alpha.
Table 89. Innate Immune Response Transcript Levels
Figure imgf000346_0001
B. Repeat Transfection
[001120] Human primary foreskin fibroblasts (BJ fibroblasts) were obtained from American Type Culture Collection (ATCC) (catalog # CRL-2522) and grown in Eagle's Minimum Essential Medium (ATCC, catalog # 30-2003) supplemented with 10% fetal bovine serum at 37°C, under 5% C02. BJ fibroblasts were seeded on a 24-well plate at a density of 300,000 cells per well in 0.5 ml of culture medium. 250 ng of modified G-CSF mRNA (mRNA sequence shown in SEQ ID NO: 6; polyA tail of approximately 140 nucleotides not shown in sequence; 5'cap, Capl) unmodified, fully modified with 5-methylcytosine and pseudouridine (Genl) or fully modified with 5-methylcytosine and Nl-methylpseudouridine (Gen2) was transfected daily for 5 days following manufacturer's protocol. Control samples of Lipofectamine 2000 (L2000) and mCherry mRNA (mRNA sequence shown in SEQ ID NO: 7; polyA tail of approximately 160 nucleotides not shown in sequence; 5 'cap, Capl; fully modified with 5-methylcytidine and pseudouridine) were also transfected daily for 5 days. The results are shown in Table 90.
[001121] Unmodified mRNA showed a cytokine response in interferon-beta (IFN-beta) and interleukin-6 (IL-6) after one day. mRNA modified with at least pseudouridine showed a cytokine response after 2-3 days whereas mRNA modified with 5-methylcytosine and Nl- methylpseudouridine showed a reduced response after 3-5 days.
Table 90. Cytokine Response
Figure imgf000347_0001
Example 59. In vivo detection of Innate Immune Response Study [001122] Female BALB/C mice (n=5) were injected intramuscularly with G-CSF mRNA (GCSF mRNA unmod) (mRNA sequence shown in SEQ ID NO: 6; polyA tail of approximately 160 nucleotides not shown in sequence;) with a 5 'cap of Capl , G-CSF mRNA fully modified with 5- methylcytosine and pseudouridine (GCSF mRNA 5mc/pU), G-CSF mRNA fully modified with 5- methylcytosine and Nl-methylpseudouridine with (GCSF mRNA 5mc/NlpU) or without a 5' cap (GCSF mRNA 5mc/Nl pU no cap) or a control of either R848 or 5% sucrose as described in Table 91. Blood is collected at 8 hours after dosing and using ELISA the protein levels of G-CSF and interferon-alpha (IFN-alpha) is determined by ELISA and are shown in Table 81.
[001123] As shown in Table 91 , unmodified, 5mc/pU, and 5mc/N lpU modified G-CSF mRNA resulted in human G-CSF expression in mouse serum. The uncapped 5mC/NlpU modified G-CSF mRNA showed no human G-CSF expression in serum, highlighting the importance of having a 5' cap structure for protein translation.
[001124] As expected, no human G-CSF protein was expressed in the R848, 5% sucrose only, and untreated groups. Importantly, significant differences were seen in cytokine production as measured by mouse IFN-alpha in the serum. As expected, unmodified G-CSF mRNA demonstrated a robust cytokine response in vivo (greater than the R848 positive control). The 5mc/pU modified G-CSF mRNA did show a low but detectable cytokine response in vivo, while the 5mc/NlpU modified mRNA showed no detectable IFN-alpha in the serum (and same as vehicle or untreated animals).
[001125] Also, the response of 5mc/NlpU modified mRNA was the same regardless of whether it was capped or not. These in vivo results reinforce the conclusion that 1) that unmodified mRNA produce a robust innate immune response, 2) that this is reduced, but not abolished, through 100% incorporation of 5mc/pU modification, and 3) that incorporation of 5mc/NlpU modifications results in no detectable cytokine response.
[001126] Lastly, given that these injections are in 5% sucrose (which has no effect by itself), these result should accurately reflect the immunostimulatory potential of these modifications.
[001127] From the data it is evident that NlpU modified molecules produce more protein while concomitantly having little or no effect on IFN-alpha expression. It is also evident that capping is required for protein production for this chemical modification. The Protein: Cytokine Ratio of 748 as compared to the PC Ratio for the unmodified mRNA (PC=9) means that this chemical modification is far superior as related to the effects or biological implications associated with IFN-alpha.
Table 91. Human G-CSF and Mouse IFN-alpha in serum
Figure imgf000349_0001
Example 60: In Vivo Delivery of Modified RNA
[001128] Protein production of modified mRNA was evaluated by delivering modified G-CSF mRNA or modified Factor IX mRNA to female Sprague Dawley rats (n=6). Rats were injected with 400 ug in 100 ul of G-CSF mRNA (mRNA sequence shown in SEQ ID NO: 6; polyA tail of approximately 160 nucleotides not shown in sequence; 5'cap, Capl) fully modified with 5- methylcytosine and pseudouridine (G-CSF Genl), G-CSF mRNA fully modified with 5- methylcytosine and Nl-methylpseudouridine (G-CSF Gen2) or Factor IX mRNA (mRNA sequence shown in SEQ ID NO: 10; polyA tail of approximately 160 nucleotides not shown in sequence; 5'cap, Capl) fully modified with 5-methylcytosine and pseudouridine (Factor IX Genl)
reconstituted from the lyophilized form in 5% sucrose. Blood was collected 8 hours after injection and the G-CSF protein level in serum was measured by ELISA. Table 92 shows the G-CSF protein levels in serum after 8 hours.
[001129] These results demonstrate that both G-CSF Gen 1 and G-CSF Gen 2 modified mRNA can produce human G-CSF protein in a rat following a single intramuscular injection, and that human G- CSF protein production is improved when using Gen 2 chemistry over Gen 1 chemistry.
Table 92. G-CSF Protein in Rat Serum (I.M. Injection Route)
Figure imgf000349_0002
Example 61. Stability of Modified RNA
[001130] Stability experiments were conducted to obtain a better understanding of storage conditions to retain the integrity of modified RNA. Unmodified G-CSF mRNA (mRNA sequence shown in SEQ ID NO: 6; polyA tail of approximately 160 nucleotides not shown in sequence; 5'cap, Capl), G-CSF mRNA (mRNA sequence shown in SEQ ID NO: 6; polyA tail of approximately 160 nucleotides not shown in sequence; 5'cap, Capl) fully modified with 5-methylcytosine and pseudouridine and G-CSF mRNA fully modified with 5-methylcytosine and pseudouridine lipoplexed with 0.75% by volume of RNAIMAX™ was stored at 50°C, 40°C, 37°C, 25°C, 4°C or - 20°C. After the mRNA had been stored for 0 hours, 2 hours, 6 hours, 24 hours, 48 hours, 5 days and 14 days, the mRNA was analyzed by gel electrophoresis using a Bio-Rad EXPERION™ system. The modified, unmodified and lipoplexed G-CSF mRNA was also stored in RNASTABLE® (Biomatrica, Inc. San Diego, CA) at 40°C or water at -80 °C or 40°C for 35 days before being analyzed by gel electrophoresis.
[001131] All mRNA samples without stabilizer were stable after 2 weeks after storage at 4°C or -20°C. Modified G-CSF mRNA, with or without lipoplex, was more stable than unmodified G- CSF when stored at 25°C (stable out to 5 days versus 48 hours), 37°C (stable out to 24 hours versus 6 hours) and 50°C (stable out to 6 hours versus 2 hours). Unmodified G-CSF mRNA, modified G- CSF mRNA with or without lipoplex tolerated 12 freeze/thaw cycles.
[001132] mRNA samples stored in stabilizer at 40°C showed similar stability to the mRNA samples stored in water at -80°C after 35 days whereas the mRNA stored in water at 40°C showed heavy degradation after 18 days.
[001133] mRNA samples stored at 4°C, 25 °C and 37 °C were stored in lx TE buffer or the formulation buffer (150 mM sodium chloride, 2mM calcium chloride, 2mM phosphate, 0.5 mM EDTA at a pH of 6.5). The mRNA stored at 4°C was stable to at least 60 days in both the TE and formulation buffer. At 25°C the mRNA in formulation buffer was stable out to 14 days and the TE buffer was stable out to at least 6 days. Storage of mRNA in the formulation buffer at 37°C was stable to 6 days compared to the TE buffer which was stable only until 4 days.
Example 62. Effects of Chemical Modifications on Expression of Formulated mRNA
[001134] Luciferase mRNA (SEQ ID NO: 16; polyA tail of approximately 140 nucleotides not shown in sequence; 5'cap, Capl) fully modified with 5-methylcytosine and 2'Fluorouridine is formulated in saline or DLin-MC3-DMA and administered intravenously, intramuscularly or subcutaneously to rodents at a dose of 0.5 mg/kg, 0.05 mg/kg, 0.005 mg/kg and/or 0.0005 mg/kg. Luciferase mRNA (SEQ ID NO: 16; polyA tail of approximately 160 nucleotides not shown in sequence; 5'cap, Capl) fully modified with 5-methylcytosine and pseudouridine is formulated in DLin-MC3-DMA and administered intramuscularly or subcutaneously to rodents at a dose of 0.5 mg/kg, 0.05 mg/kg, 0.005 mg/kg and/or 0.0005 mg/kg. The DLin-MC 3 -DMA formulations are analyzed prior to administration to determine the mean size and zeta potential. The rodents are imaged at 2 hours, 8 hours, 24 hours, 72 hours, 96 hours, 144 hours and 168 hours after dosing and the bioluminescence is measured in photon per second for each route of administration and formulation.
Example 63. Expression of PLGA Formulated mRNA
A. Synthesis and Characterization of Luciferase PLGA Microspheres
[001135] Luciferase mRNA (mRNA sequence shown in SEQ ID NO: 16; polyA tail of
approximately 140 nucleotides not shown in sequence; 5'cap, Capl) fully modified with 5- methylcytosine and Nl-methyl pseudouridine, modified with 25% of uridine replaced with 2- thiouridine and 25% of cytosine replaced with 5-methylcytosine, fully modified with Nl-methyl pseudouridine, or fully modified with pseudouridine was reconstituted in lx TE buffer and then formulated in PLGA microspheres. PLGA microspheres were synthesized using the water/oil/water double emulsification methods known in the art using PLGA-ester cap (Lactel, Cat# B6010-2, inherent viscosity 0.55-0.75, 50:50 LA:GA), polyvinylalcohol (PVA) (Sigma, Cat# 348406-25G, MW 13 -23k) dichloromethane and water. Briefly, 0.4 ml of mRNA in TE buffer at 4 mg/ml (Wl) was added to 2 ml of PLGA dissolved in dichloromethane (DCM) (01) at a concentration of 200 mg/ml of PLGA. The Wl/Ol emulsion was homogenized (IKA Ultra-Turrax Homogenizer, T18) for 30 seconds at speed 5 (-19,000 rpm). The Wl/Ol emulsion was then added to 250 ml 1% PVA (W2) and homogenized for 1 minute at speed 5 (-19,000 rpm). Formulations were left to stir for 3 hours, then passed through a 100 μιη nylon mesh strainer (Fisherbrand Cell Strainer, Cat # 22-363- 549) to remove larger aggregates, and finally washed by centrifugation (10 min, 9,250 rpm, 4°C). The supernatant was discarded and the PLGA pellets were resuspended in 5-10 ml of water, which was repeated 2x. After washing and resuspension with water, 100-200 μΐ of a PLGA microspheres sample was used to measure particle size of the formulations by laser diffraction (Malvern
Mastersizer2000). The washed formulations were frozen in liquid nitrogen and then lyophilized for 2-3 days.
[001136] After lyophilization, -10 mg of PLGA MS were weighed out in 2 ml eppendorf tubes and deformulated by adding 1 ml of DCM and letting the samples shake for 2-6 hrs. The mRNA was extracted from the deformulated PLGA micropsheres by adding 0.5 ml of water and shaking the sample overnight. Unformulated luciferase mRNA in TE buffer (unformulated control) was spiked into DCM and went through the deformulation process (deformulation control) to be used as controls in the transfection assay. The encapsulation efficiency, weight percent loading and particle size are shown in Table 93. Encapsulation efficiency was calculated as mg of mRNA from deformulation of PLGA microspheres divided by the initial amount of mRNA added to the formulation. Weight percent loading in the formulation was calculated as mg of mRNA from deformulation of PLGA microspheres divided by the initial amount of PLGA added to the formulation.
Table 93. PLGA Characteristics
Figure imgf000352_0001
B. Protein Expression of modified mRNA Encapsulated in PLGA Microspheres
[001137] The day before transfection, 20,000 HeLa cells (ATCC no. CCL-2; Manassas, VA) were harvested by treatment with Trypsin-EDTA solution (LifeTechnologies, Grand Island, NY) and seeded in a total volume of lOOul EMEM medium (supplemented with 10%FCS and lx Glutamax) per well in a 96-well cell culture plate (Corning, Manassas, VA). The cells were grown at 37oC in a 5% C02 atmosphere overnight. The next day, 83 ng of the deformulated luciferase mRNA PLGA microsphere samples, deformulated luciferase mRNA control (Deform control), or unformulated luciferase mRNA control (Unfomul control) was diluted in a lOul final volume of OPTI-MEM (LifeTechnologies, Grand Island, NY). Lipofectamine 2000 (LifeTechnologies, Grand Island, NY) was used as a transfection reagent and 0.2ul was diluted in a lOul final volume of OPTI-MEM. After 5min of incubation at room temperature, both solutions were combined and incubated an additional 15min at room temperature. Then 20ul of the combined solution was added to lOOul of cell culture medium containing the HeLa cells. The plates were then incubated as described before. [001138] After a 18 to 22 hour incubation, cells expressing luciferase were lysed with lOOul Passive Lysis Buffer (Promega, Madison, WI) according to manufacturer instructions. Aliquots of the lysates were transferred to white opaque polystyrene 96-well plates (Corning, Manassas, VA) and combined with lOOul complete luciferase assay solution (Promega, Madison, WI). The background signal of the plates without reagent was about 200 relative light units per well. The plate reader was a BioTek Synergy HI (BioTek, Winooski, VT).
[001139] Cells were harvested and the bioluminescence (in relative light units, RLU) for each sample is shown in Table 94. Transfection of these samples confirmed that the varied chemistries of luciferase mR A is still able to express luciferase protein after PLGA microsphere formulation.
Table 94. Chemical Modifications
Figure imgf000353_0001
Example 64. In vitro studies of Factor IX
A. Serum-Free Media
[001140] Human Factor IX mRNA (mRNA sequence shown in SEQ ID NO: 10; polyA tail of approximately 160 nucleotides not shown in sequence; 5 'cap, Capl; fully modified with 5- methylcytosine and pseudouridine) was transfected in serum-free media. The cell culture supernatant was collected and subjected to trypsin digestion before undergoing 2-dimensional HPLC separation of the peptides. Matrix-assisted laser desorption/ionization was used to detect the peptides. 8 peptides were detected and 7 of the detected peptides are unique to Factor IX. These results indicate that the mR A transfected in the serum-free media was able to express full-length Factor IX protein.
B. Human Embryonic Kidney (HEK) 293A Cells
[001141] 250 ng of codon optimized Human Factor IX mRNA (mRNA sequence shown in SEQ ID NO: 10; fully modified with 5-methylcytosine and pseudouridine; polyA tail of approximately 160 nucleotides not shown in sequence; 5'cap, Capl) was transfected into HEK 293A cells (150, 000 cells/well) using Lipofectamine 2000 in DMEM in presence of 10% FBS. The transfection complexes were removed 3 hours after transfection. Cells were harvested at 3, 6, 9, 12, 24, 48 and 72 hours after transfection. Total RNA was isolated and used for cDNA synthesis. The cDNA was subjected to analysis by quantitative Real-Time PCR using codon optimized Factor IX specific primer set. Human hypoxanthine phosphoribosyltransfersase 1 (HPRT) level was used for normalization. The data is plotted as a percent of detectable mRNA considering the mRNA level as 100% at the 3 hour time point. The half-life of Factor IX modified mRNA fully modified with 5- methylcytosine and pseudouridine in human embryonic kidney 293 (HEK293) cells is about 8-10 hours.
Example 65. Saline formulation: Subcutaneous Administration
[001142] Human G-CSF modified mRNA (mRNA sequence shown in SEQ ID NO: 6; polyA tail of approximately 160 nucleotides not shown in sequence; 5'cap, Capl; fully modified with 5- methylcytosine and pseudouridine) and human EPO modified mRNA (mRNA sequence shown in SEQ ID NO: 9; polyA tail of approximately 160 nucleotides not shown in sequence; 5'cap, Capl; fully modified with 5-methylcytosine and pseudouridine), were formulated in saline and delivered to mice via intramuscular (IM) injection at a dose of 100 ug.
[001143] Controls included Luciferase (mRNA sequence shown in SEQ ID NO: 16; polyA tail of approximately 160 nucleotides not shown in sequence; 5'cap, Capl; fully modified with 5- methylcytosine and pseudouridine)) or the formulation buffer (F. Buffer). The mice were bled at 13 hours after the injection to determine the concentration of the human polypeptide in serum in pg/mL. (G-CSF groups measured human G-CSF in mouse serum and EPO groups measured human EPO in mouse serum). The data are shown in Table 95.
[001144] mPvNA degrades rapidly in serum in the absence of formulation suggesting the best method to deliver m NA to last longer in the system is by formulating the mR A. As shown in Table 95, mR A can be delivered subcutaneously using only a buffer formulation.
Table 95. Dosing Regimen
Figure imgf000355_0001
Example 66. Stability of Nanoparticle of Formulations
[001145] Formulations of DLin-KC2-DMA, Teta-5-Lap, DLin-DMA, DLin-K-DMA, C 12-200, DLin-MC3-DMA at a lipid:mRNA ratio of 20: 1 were evaluated for particle size, polydispersity index and encapsulation efficiency for stability at room temperature. Most nanoparticles are stable at room temperature for at least one month as shown in Tables 96 and 97.
Table 96. Particle Size and Polydispersity Index
Figure imgf000355_0002
Table 97. Encapsulation Efficiency
Figure imgf000356_0001
Example 67. Intravitreal Delivery
[001146] mCherry modified mRNA (mRNA sequence shown in SEQ ID NO: 7; polyA tail of approximately 160 nucleotides not shown in sequence; 5 'cap, Capl ; fully modified with 5- methylcytosine and pseudouridine) and luciferase modified mRNA (mRNA sequence shown in SEQ ID NO: 16; polyA tail of approximately 160 nucleotides not shown in sequence; 5 'cap, Capl ; fully modified with 5 -methylcytosme and pseudouridine) formulated in saline was delivered intravitreally in rats as described in Table 98. The sample was compared against a control of saline only delivered intravitreally.
Table 98. Dosing Chart
Figure imgf000356_0002
[001147] The formulation will be administered to the left or right eye of each animal on day 1 while the animal is anesthetized. On the day prior to administration gentamicin ophthalmic ointment or solution was applied to both eyes twice. The gentamicin ophthalmic ointment or solution was also applied immediately following the injection and on the day following the injection.Prior to dosing, mydriatic drops (1% tropicamide and/or 2.5% phenylephrine) are applied to each eye.
[001148] 18 hours post dosing the eyes receiving the dose of mCherry and delivery buffer are enucleated and each eye was separately placed in a tube containing 10 mL 4% paraformaldehyde at room temperature for overnight tissue fixation. The following day, eyes will be separately transferred to tubes containing 10 mL of 30% sucurose and stored at 21°C until they were processed and sectioned. The slides prepared from different sections were evaluated under F-microscopy. Postive expression was seen in the slides prepared with the eyes administered mCherry modified mR A and the control showed no expression.
Example 68. In Vivo Cytokine Expression Study
[001149] Mice were injected intramuscularly with 200 ug of G-CSF modified mRNA (mRNA sequence shown in SEQ ID NO: 6; polyA tail of approximately 160 nucleotides not shown in sequence) which was unmodified with a 5'cap, Capl (unmodified), fully modified with 5- methylcytosine and pseudouridine and a 5'cap, Capl (Genl) or fully modified with 5- methylcytosine and Nl-methylpseudouridine and a 5'cap, Capl (Gen2 cap) or no cap (Gen2 uncapped). Controls of R-848, 5% sucrose and untreated mice were also analyzed. After 8 hours serum was collected from the mice and analyzed for interferon-alpha (IFN-alpha) expression. The results are shown in Table 99.
Table 99. IFN-alpha Expression
Figure imgf000357_0001
Example 69. In vitro expression of VEGF modified mRNA
[001150] HEK293 cells were transfected with modified mRNA (mniRNA) VEGF-A (mRNA sequence shown in SEQ ID NO: 19; polyA tail of approximately 160 nucleotides not shown in sequence; 5'cap, Capl; fully modified with 5-methylcytosine and pseudouridine) which had been complexed with Lipofectamine2000 from Invitrogen (Carlsbad, CA) at the concentration shown in Table 100. The protein expression was detected by ELISA and the protein (pg/ml) is shown in Table
Table 100. Protein Expression
Figure imgf000357_0002
Protein
10495 10038 2321.23 189.6 0 0 0 0
(pg/ml)
Example 70. In vitro Screening in HeLa Cells of GFP
[001151] The day before transfection, 20,000 HeLa cells (ATCC no. CCL-2; Manassas, VA) were harvested by treatment with Trypsin-EDTA solution (LifeTechnologies, Grand Island, NY) and seeded in a total volume of lOOul EMEM medium (supplemented with 10%FCS and lx Glutamax) per well in a 96-well cell culture plate (Corning, Manassas, VA). The cells were grown at 37°C in 5% C02 atmosphere overnight. Next day, 37.5 ng or 75 ng of Green Fluroescent protein (GFP) modified RNA (mRNA sequence shown in SEQ ID NO: 18; polyA tail of approximately 160 nucleotides not shown in sequence; 5 'cap, Capl) with the chemical modification described in Table 101, were diluted in lOul final volume of OPTI-MEM (LifeTechnologies, Grand Island, NY). Lipofectamine 2000 (LifeTechnologies, Grand Island, NY) was used as transfection reagent and 0.2 ul were diluted in 10 ul final volume of OPTI-MEM. After 5 minutes of incubation at room temperature, both solutions were combined and incubated an additional 15 minute at room temperature. Then the 20ul combined solution was added to the lOOul cell culture medium containing the HeLa cells and incubated at room temperature.
[001152] After a 18 to 22 hour incubation cells expressing luciferase were lysed with 100 ul of Passive Lysis Buffer (Promega, Madison, WI) according to manufacturer instructions. Aliquots of the lysates were transferred to white opaque polystyrene 96-well plates (Corning, Manassas, VA) and combined with 100 ul complete luciferase assay solution (Promega, Madison, WI). The median fluorescence intensity (MFI) was determined for each chemistry and is shown in Table 101.
[001153] These results demonstrate that GFP fully modified with Nl-methylpseudouridine and 5- methylcytosine produces more protein in HeLa cells compared to the other chemistry. Additionally the higher dose of GFP administered to the cells resulted in the highest MFI value.
Table 101. Mean Fluorescence Intensity
Figure imgf000358_0001
Example 71. Toxicity Studies
A. Study Design [001154] Sprague-Dawley rats (n=8, 4 male, 4 female) were administered by injection modified luciferase mRNA (mRNA sequence shown in SEQ ID NO: 16; polyA tail of approximately 160 nucleotides not shown in sequence; 5 'cap, Capl; fully modified with 5-methylcytosine and pseudouridine) as outlined in the dosing chart in Table 102. A control group were administered the formulation buffer (F. Buffer). After 7 days the rats were sacrificed.
Table 102. Dosins Chart
Figure imgf000359_0001
B. Weight Gain and Food Consumption
[001155] The rats were weighed before the administration of mRNA and 7 days after administration. Table 103 shows the mean weight gain and weight gain percent per group tested separated by gender. All animals continued to gain weight and behave normally. Each group analyzed consumed about the same amount of food over the course of the study.
Table 103. Weight Gain
Figure imgf000359_0002
C. Electrolytes
[001156] After 7 days the rats were sacrificed and samples were taken to determine electrolytes. The calcium, bicarbonate, potassium, phosphorus, chloride and sodium levels in each group were analyzed. The results are shown in Table 104. There was no change in electrolytes seen in the rats after 7 days.
Table 104. Electrolytes
Figure imgf000359_0003
(mg/dL) (mEg/L) (mEg/L) (mg/dL) (mEg/L) (mEg/L)
100 ug 9.8 19.9 4.7 8.3 101.0 139.6
300 ug 9.8 23.3 4.4 8.2 100.5 139.6
1000 ug 10.6 22.5 5.2 9.1 101.0 138.8
3 x 1000 ug 10.2 22.6 4.6 8.11 100.4 138.8
F. Buffer 9.6 20.1 5.4 9.2 99.5 139.9
D. Hematology
[001157] After 7 days the rats were sacrificed and samples were taken to determine hematology levels. The red blood cell (RBC), hematocrit (HGT), mean corpuscular volume (MCV), hemoglobin (HGB), mean corpuscular hemoglobin (MCH) and mean corpuscular hemoglobin concentration (MCHC) was determined for each group. The results are shown in Table 105. There was no change in blood count or blood clotting factors 7 days after administration.
Table 105. Hematology
Figure imgf000360_0001
E. White Blood Cells
[001158] After 7 days the rats were sacrificed and samples were taken to determine white blood cell count. Neutrophils (percent segmented neutrophils), monocytes, basophils, lymphocytes, eosinophil and white blood cell (WBC) was determined for each group. The results are shown in Table 106. In Table 106, "NT" means not tested. 7 days after administration there was no increase in white blood cells which suggests there was no inflammation.
Table 106. White Blood Cell
Figure imgf000360_0002
F. Serum Chemistry [001159] After 7 days the rats were sacrificed and samples were taken to determine serum chemistry. The alkaline phosphatase (ALP), aspartate transaminase (AST), alanine transaminase (ALT) and creatine phosphokinase (CPK) was determined for each group. The results are shown in Table 107.
Table 107. Serum Chemistry
Figure imgf000361_0001
G. Liver Proteins
[001160] After 7 days the rats were sacrificed and samples were taken to determine liver protein levels. The level of albumin, globulin and total protein was determined for each group. The results are shown in Table 108. There was no change seen in liver enzyme or liver protein production 7 days after administration with the modified mRNA.
Table 108. Hematology
Figure imgf000361_0002
H. Conclusions
[001161] From the analysis of the rats 7 days after administration with the modified mRNA, administration of high doses of mRNA do not result in adverse effects. Doses as high as 30 times the effective dose appear to be safe from this analysis. Histopathology showed only minimal inflammation at the site of injection and the site of injection showed only changes consistent with injection and nothing to suggest dose related issues. Additionally there was no chance in muscle enzymes to suggest there was muscle damage.
Example 72. Storage Conditions for Modified RNA
A. Organics
[001162] To evaluate the ability of mRNA to withstand an organic environment, luciferase mRNA (mRNA sequence shown in SEQ ID NO: 16; polyA tail of approximately 160 nucleotides not shown in sequence; 5'cap, Capl; fully modified with 5-methylcytosine and pseudouridine) was stored at room temperature in solutions of ethanol, methanol or dichloromethane at a concentration of 1 mg/ml. Samples were collected at 1 hour, 6 hours and 1 day. The sample was diluted with water to 200 ng/ul and incubated overnight at room temperature in a fume hood to evaporate off the organic solvent. Control samples were completed in parallel with mRNA in water (Water control, organic). The mRNA was stable at room temperature for 1 day in each of the three solutions as determined by running samples on a bioanalyzer.
B. Aqueous Solvent
[001163] Luciferase mRNA (mRNA sequence shown in SEQ ID NO: 16; polyA tail of
approximately 160 nucleotides not shown in sequence; 5 'cap, Capl; fully modified with
pseudouridine and 5-methylcytosine)was added to 3 different buffers and water to evaluate the effect of aqueous solvents on mRNA stability. mRNA was added to citrate buffer (pH3, lOOmM citric acid), phosphate buffered saline (PBS) buffer (pH 7.4, 6.7 mM phosphate and 154 mM sodium chloride), TE buffer (pH 8, 10 mM Tris-hydrochloric acid and 1 mM ethylenediaminetetraacetic acid) or water (pH 5.5, water for injection (WFI)) at 1 mg/ml. Samples were collected at 1 hour, 6 hours and 1 day and diluted with water to a concentration of 200 ng/ul. Control samples were completed in parallel with mRNA in water (Water control, aqueous). The incubation of mRNA in the PBS buffer, TE buffer and water did not affect the mRNA integrity after 1 day. Samples incubated in citrate were not detectable by bioanalyzer.
[001164] In additional studies to evaluate the citrate buffer, citrate buffer at a pH of 2, 3 and 4 each having 10 mM citrate and lmg/ml of luciferase mRNA (mRNA sequence shown in SEQ ID NO: 16; polyA tail of approximately 160 nucleotides not shown in sequence; 5 'cap, Capl; fully modified with pseudouridine and 5-methylcytosine)were evaluated. At a pH of 2 precipitation was visually detected and mRNA was not detected by bioanalyzer below a pH of 4. When compared against phosphate buffer, mRNA was not detected in samples with low pH and precipitation was visible in phosphate buffer samples having a pH of 2.
C. pH
[001165] In order to study the effects of pH on the stability of mRNA, luciferase mRNA (mRNA sequence shown in SEQ ID NO: 16; polyA tail of approximately 160 nucleotides not shown in sequence; 5 'cap, Capl; fully modified with pseudouridine and 5-methylcytosine) was stored at room temperature in aqueous buffers having a pH of 5.8, 6.5 or 7.2. Samples were collected at 1 hour, 1 day and 1 week after the mRNA was added to the pH sample. After collection the samples were incubated at 1 mg/ml concentration and then diluted to 200 ng/ul with water before freezing and characterizing by bioanalyzer. The mRNA was stable after 1 week of storage at room temperature in the pH range of 5.8-7.2 evaluated.
D. Freeze/thaw and Lyophilization
[001166] To evaluate the effect of freeze/thaw cycles on mRNA stability, luciferase mRNA (mRNA sequence shown in SEQ ID NO: 16; polyA tail of approximately 160 nucleotides not shown in sequence; 5 'cap, Capl; fully modified with 5-methylcytosine and pseudouridine) in formulation buffer was subjected to numerous freeze/thaw cycles. mRNA was found to be stable for at least 18 cycles.
[001167] In addition, luciferase mRNA (mRNA SEQ ID NO: 16; polyA tail of approximately 160 nucleotides not shown in sequence; 5 'cap, Capl; fully modified with 5-methylcytosine and pseudouridine) was subjected to 3 rounds of lyophilization to test the stability of the mRNA. mRNA was added to water and samples were collected after each of the 3 rounds of lyophilization. The dried mRNA was diluted with water to reach a concentration of 1 mg/ml. The samples were stored frozen until bioanalyzer characterization at 200 ng/ul. Control samples were completed in parallel with mRNA and water formulations and followed the same freezing and thawing cycles. The mRNA was found to be stable after 3 cycles of lyophilization when analyzed by bioanalyzer characterization at 200 ng/ul.
E. Centrifugation
[001168] To evaluate the effects of centrifugation on mRNA integrity, luciferase mRNA (mRNA sequence shown in SEQ ID NO: 16; polyA tail of approximately 160 nucleotides not shown in sequence; 5 'cap, Capl; 5-methylcytosine and pseudouridine) in water at lmg/ml was exposed to 10 cycles of 10k RPM (13.3k xg) for 10 minutes at 4°C. mRNA and water samples were stored at 4°C as a control during centrifugation . After 10 cycles of centrifugation the mRNA was still stable when analyzed by bioanalyzer characterization at 200 ng/ul.
F. In Vitro Transfection After Storage
[001169] The day before transfection, 20,000 HeLa cells (ATCC no. CCL-2; Manassas, VA) were harvested by treatment with Trypsin-EDTA solution (LifeTechnologies, Grand Island, NY) and seeded in a total volume of lOOul EMEM medium (supplemented with 10%FCS and lx Glutamax) per well in a 96-well cell culture plate (Corning, Manassas, VA). The cells were grown at 37oC in a 5% C02 atmosphere overnight. The next day, 250 ng of luciferase mRNA from the formulations of the lyophilized, centrifuged, organic and aqueous solvent samples were diluted in a lOul final volume of OPTI-MEM (LifeTechnologies, Grand Island, NY). Lipofectamine 2000
(LifeTechnologies, Grand Island, NY) was used as a transfection reagent and 0.2ul was diluted in a lOul final volume of OPTI-MEM. After 5min of incubation at room temperature, both solutions were combined and incubated an additional 15min at room temperature. Then 20ul of the combined solution was added to lOOul of cell culture medium containing the HeLa cells. The plates were then incubated as described before.
[001170] After 18h to 22h incubation, cells expressing luciferase were lysed with lOOul Passive Lysis Buffer (Promega, Madison, WI) according to manufacturer instructions. Aliquots of the lysates were transferred to white opaque polystyrene 96-well plates (Corning, Manassas, VA) and combined with lOOul complete luciferase assay solution (Promega, Madison, WI). The background signal of the plates without reagent was about 200 relative light units per well. The plate reader was a BioTek Synergy HI (BioTek, Winooski, VT).
[001171] Controls of mock transfection (transfection reagent alone), luciferase mRNA control in water, and untreated were also evaluated. Cells were harvested and the bioluminescence average (in relative light units, RLU) for each signal is shown in Table 109. Transfection of these samples confirmed that lyophilization, centrifugation, organic solvents and aqueous solvents except citrate buffer did not impact the activity of luciferase mRNA. Citrate buffer showed a reduced activity after transfection.
Table 109. Bioluminescence
Figure imgf000364_0001
Water control, aqueous, 1 hour 3394000
Citrate buffer, 1 day 269790
PBS buffer, 1 day 4084330
TE buffer, 1 day 5344400
Water control, aqueous, 1 day 3579270
Untreated 5580
Mock Transfection 7560
Luciferase mRNA control 4950090
Example 73. Homogenization
[001172] Different luciferase mRNA solutions (as described in Table 110 where "X" refers to the solution containing that component) (mRNA sequence shown in SEQ ID NO: 16; polyA tail of approximately 160 nucleotides not shown in sequence; 5 'cap, Capl; fully modified with 5- methylcytosine and pseudouridine) were evaluated to test the percent yield of the different solutions, the integrity of the mRNA by bioanalyzer, and the protein expression of the mRNA by in vitro transfection. The mRNA solutions were prepared in water, lx TE buffer at 4 mg/ml as indicated in Table 110, and added to either dichloromethane (DCM) or DCM containing 200 mg/ml of poly(lactic-co-glycolic acid) (PLGA) (Lactel, Cat# B6010-2, inherent viscosity 0.55-0.75, 50:50 LA:GA) to achieve a final mRNA concentration of 0.8 mg/ml. The solutions requiring
homogenization were homogenized for 30 seconds at speed 5 (approximately 19,000 rpm) (IKA Ultra-Turrax Homogenizer, T18). The mRNA samples in water, dicloromethane and poly(lactic-co- glycolic acid) (PLGA) were not recoverable (NR). All samples, except the NR samples, maintained integrity of the mRNA as determined by bioanalyzer (Bio-rad Experion).
[001173] The day before transfection, 20,000 HeLa cells (ATCC no. CCL-2; Manassas, VA) were harvested by treatment with Trypsin-EDTA solution (LifeTechnologies, Grand Island, NY) and seeded in a total volume of lOOul EMEM medium (supplemented with 10%FCS and lx Glutamax) per well in a 96-well cell culture plate (Corning, Manassas, VA). The cells were grown at 37oC in a 5% C02 atmosphere overnight. The next day, 250 ng of luciferase mRNA from the recoverable samples was diluted in a lOul final volume of OPTI-MEM (LifeTechnologies, Grand Island, NY). Lipofectamine 2000 (LifeTechnologies, Grand Island, NY) was used as a transfection reagent and 0.2ul was diluted in a lOul final volume of OPTI-MEM. After 5 minutes of incubation at room temperature, both solutions were combined and incubated an additional 15 minutes at room temperature. Then 20ul of the combined solution was added to lOOul of cell culture medium containing the HeLa cells. The plates were then incubated as described before. Controls luciferase mR A (luciferase mR A formulated in saline) (Control) and untreated cells (Untreat.) were also evaluated. Cells were harvested and the bio luminescence average (in photons/second) (biolum. (p/s)) for each signal is also shown in Table 110. The recoverable samples all showed activity of luciferase mRNA when analyzed.
[001174] After 18 to 22 hour incubation, cells expressing luciferase were lysed with lOOul Passive Lysis Buffer (Promega, Madison, WI) according to manufacturer instructions. Aliquots of the lysates were transferred to white opaque polystyrene 96-well plates (Corning, Manassas, VA) and combined with lOOul complete luciferase assay solution (Promega, Madison, WI). The background signal of the plates without reagent was about 200 relative light units per well. The plate reader was a BioTek Synergy HI (BioTek, Winooski, VT).
[001175] Cells were harvested and the bioluminescence average (in relative light units, RLU) (biolum. (RLU)) for each signal is also shown in Table 110. The recoverable samples all showed activity of luciferase mRNA when analyzed.
Table 110. Solutions
Figure imgf000366_0001
Example 74. TE Buffer and Water Evaluation
[001176] Luciferase mRNA (mRNA sequence shown in SEQ ID NO: 16; polyA tail of
approximately 160 nucleotides not shown in sequence; 5 'cap, Capl; fully modified with 5- methylcytosine and pseudouridine) was reconstituted in water or TE buffer as outlined in Table 111 and then formulated in PLGA microspheres. PLGA microspheres were synthesized using the water/oil/water double emulsification methods known in the art using PLGA (Lactel, Cat# B6010-2, inherent viscosity 0.55-0.75, 50:50 LA:GA), polyvinylalcohol (PVA) (Sigma, Cat# 348406-25G, MW 13-23k) dichloromethane and water. Briefly, 0.2 to 0.6 ml of mRNA in water or TE buffer at a concentration of 2 to 6 mg/ml (Wl) was added to 2 ml of PLGA dissolved in dichloromethane (DCM) (01) at a concentration of 100 mg/ml of PLGA. The Wl/Ol emulsion was homogenized (IKA Ultra-Turrax Homogenizer, T18) for 30 seconds at speed 5 (-19,000 rpm). The Wl/Ol emulsion was then added to 250 ml 1% PVA (W2) and homogenized for 1 minute at speed 5 (-19,000 rpm). Formulations were left to stir for 3 hours, then passed through a 100 μιη nylon mesh strainer (Fisherbrand Cell Strainer, Cat # 22-363-549) to remove larger aggregates, and finally washed by centrifugation (10 min, 9,250 rpm, 4°C). The supernatant was discarded and the PLGA pellets were resuspended in 5-10 ml of water, which was repeated 2x. The washed formulations were frozen in liquid nitrogen and then lyophilized for 2-3 days. After lyophilization, -10 mg of PLGA MS were weighed out in 2 ml eppendorf tubes and deformulated by adding 1 ml of DCM and letting the samples shake for 2-6 hrs. mRNA was extracted from the deformulated PLGA micropsheres by adding 0.5 ml of water and shaking the sample overnight. Unformulated luciferase mRNA in water or TE buffer (deformulation controls) was spiked into DCM and went through the deformulation process to be used as controls in the transfection assay.
[001177] The day before transfection, 20,000 HeLa cells (ATCC no. CCL-2; Manassas, VA) were harvested by treatment with Trypsin-EDTA solution (LifeTechnologies, Grand Island, NY) and seeded in a total volume of lOOul EMEM medium (supplemented with 10%FCS and lx Glutamax) per well in a 96-well cell culture plate (Corning, Manassas, VA). The cells were grown at 37oC in a 5% C02 atmosphere overnight. The next day, 100 ng of the deformulated luciferase mRNA samples was diluted in a lOul final volume of OPTI-MEM (LifeTechnologies, Grand Island, NY).
Lipofectamine 2000 (LifeTechnologies, Grand Island, NY) was used as a transfection reagent and 0.2ul was diluted in a lOul final volume of OPTI-MEM. After 5 minutes of incubation at room temperature, both solutions were combined and incubated an additional 15 minutes at room temperature. Then 20ul of the combined solution was added to lOOul of cell culture medium containing the HeLa cells. The plates were then incubated as described before.
[001178] After 18 to 22 hour incubation, cells expressing luciferase were lysed with lOOul Passive Lysis Buffer (Promega, Madison, WI) according to manufacturer instructions. Aliquots of the lysates were transferred to white opaque polystyrene 96-well plates (Corning, Manassas, VA) and combined with lOOul complete luciferase assay solution (Promega, Madison, WI). The background signal of the plates without reagent was about 200 relative light units per well. The plate reader was a BioTek Synergy HI (BioTek, Winooski, VT). To determine the activity of the luciferase mRNA from each formulation, the relative light units (RLU) for each formulation was divided by the RLU of the appropriate mRNA deformulation control (mRNA in water or TE buffer). Table 111 shows the activity of the luciferase mRNA. The activity of the luciferase mRNA in the PLGA microsphere formulations (Form.) was substantially improved by formulating in TE buffer versus water.
Table 111. Formulations
Figure imgf000368_0001
Example 75. Chemical Modifications on mRNA
[001179] The day before transfection, 20,000 HeLa cells (ATCC no. CCL-2; Manassas, VA) were harvested by treatment with Trypsin-EDTA solution (Life Technologies, Grand Island, NY) and seeded in a total volume of lOOul EMEM medium (supplemented with 10%FCS and lx Glutamax) per well in a 96-well cell culture plate (Corning, Manassas, VA). The cells were grown at 37°C in 5% C02 atmosphere overnight. The next day, 83 ng of Luciferase modified RNA (mRNA sequence shown SEQ ID NO: 16; polyA tail of approximately 140 nucleotides not shown in sequence; 5'cap, Capl) with the chemical modification described in Table 112, were diluted in lOul final volume of OPTI-MEM (LifeTechnologies, Grand Island, NY). Lipofectamine 2000 (LifeTechnologies, Grand Island, NY) was used as transfection reagent and 0.2 ul were diluted in 10 ul final volume of OPTI- MEM. After 5 minutes of incubation at room temperature, both solutions were combined and incubated an additional 15 minute at room temperature. Then the 20ul combined solution was added to the lOOul cell culture medium containing the HeLa cells and incubated at room temperature.
[001180] After 18 to 22 hours of incubation cells expressing luciferase were lysed with 100 ul of Passive Lysis Buffer (Promega, Madison, WI) according to manufacturer instructions. Aliquots of the lysates were transferred to white opaque polystyrene 96-well plates (Corning, Manassas, VA) and combined with 100 ul complete luciferase assay solution (Promega, Madison, WI). The lysate volumes were adjusted or diluted until no more than 2 mio relative light units (RLU) per well were detected for the strongest signal producing samples and the RLUs for each chemistry tested are shown in Table 112. The plate reader was a BioTek Synergy HI (BioTek, Winooski, VT).The background signal of the plates without reagent was about 200 relative light units per well.
Table 112. Chemical Modifications
Figure imgf000369_0001
Example 76. Intramuscular and Subcutaneous Administration of Modified mRNA
[001181] Luciferase modified mRNA (mRNA sequence shown in SEQ ID NO: 16; polyA tail of approximately 140 nucleotides not shown in sequence; 5'cap, Capl) fully modified with 5- methylcytosine and pseudouridine (5mC/pU), fully modified with 5-methylcytosine and Nl- methylpseudouridine (5mC/NlmpU), fully modified with pseudouridine (pU), fully modified with Nl-methylpseudouridine (NlmpU) or modified where 25% of the cytosines replaced with 5- methylcytosine and 25% of the uridines replaced with 2-thiouridine (5mC/s2U) formulated in PBS (pH 7.4) was administered to Balb-C mice intramuscularly or subcutaneously at a dose of 2.5 mg/kg. The mice were imaged at 2 hours, 8 hours, 24 hours, 48 hours, 72 hours, 96 hours, 120 hours and 144 hours for intramuscular delivery and 2 hours, 8 hours, 24 hours, 48 hours, 72 hours, 96 hours and 120 hours for subcutaneous delivery. Twenty minutes prior to imaging, mice were injected intraperitoneally with a D-luciferin solution at 150 mg/kg. Animals were then anesthetized and images were acquired with an IVIS Lumina II imaging system (Perkin Elmer). Bio luminescence was measured as total flux (photons/second) of the entire mouse. The average total flux
(photons/second) for intramuscular administration is shown in Table 113 and the average total flux (photons/second) for subcutaneous administration is shown in Table 114. The background signal was 3.79E+05 (p/s). The peak expression for intramuscular administration was seen between 24 and 48 hours for all chemistry and expression was still detected at 144 hours. For subcutaneous delivery the peak expression was seen at 2-8 hours and expression was detected at 72 hours.
Table 113. Intramuscular Administration
Figure imgf000370_0001
Example 77. Osmotic Pump Study
[001182] Prior to implantation, an osmotic pump (ALZET® Osmotic Pump 2001D, DURECT Corp. Cupertino, CA) is loaded with the 0.2 ml of IX PBS (pH 7.4) (PBS loaded pump) or 0.2 ml of luciferase modified mRNA (mRNA sequence shown in SEQ ID NO: 16; polyA tail of approximately 140 nucleotides not shown in sequence; 5'cap, Capl; fully modified with 5-methylcytosine and Nl- methylpseudouridine) at 1 mg/ml in lx PBS (pH 7.4) (Luciferase loaded pump) and incubated overnight in lx PBS (pH 7.4) at 37°C.
[001183] Balb-C mice (n=3) are implanted subcutaneously with either the PBS loaded pump or the luciferase loaded pump and imaged at 2 hours, 8 hours and 24 hours. As a control a PBS loaded pump is implanted subcutaneously and the mice are injected subcutaneously with luciferase modified mR A in lx PBS (PBS loaded pump; SC Luciferase) or an osmotic pump is not implanted and the mice are injected subcutaneously with luciferase modified mRNA in lx PBS (SC
Luciferase). The luciferase formulations are outlined in Table 115
Table 115. Luciferase Formulations
Figure imgf000371_0001
Example 78. External Osmotic Pump Study
[001184] An external osmotic pump (ALZET® Osmotic Pump 200 ID, DURECT Corp. Cupertino, CA) is loaded with the 0.2 ml of IX PBS (pH 7.4) (PBS loaded pump) or 0.2 ml of luciferase modified mRNA (mRNA sequence shown in SEQ ID NO: 16; polyA tail of approximately 140 nucleotides not shown in sequence; 5'cap, Capl; fully modified with 5-methylcytosine and Nl- methylpseudouridine) at 1 mg/ml in lx PBS (pH 7.4) (luciferase loaded pump) and incubated overnight in lx PBS (pH 7.4) at 37°C.
[001185] Using a catheter connected to the external PBS loaded pump or the luciferase loaded pump Balb-C mice (n=3) are administered the formulation. The mice are imaged at 2 hours, 8 hours and 24 hours. As a control an external PBS loaded pump is used and the mice are injected
subcutaneously with luciferase modified mRNA in lx PBS (PBS loaded pump; SC Luciferase) or the external pump is not used and the mice are only injected subcutaneously with luciferase modified mRNA in lx PBS (SC Luciferase). Twenty minutes prior to imaging, mice are injected
intraperitoneally with a D-luciferin solution at 150 mg/kg. Animals are then anesthetized and images are acquired with an IVIS Lumina II imaging system (Perkin Elmer). Bioluminescence is measured as total flux (photons/second) of the entire mouse. The luciferase formulations are outlined in Table 116 and the average total flux (photons/second).
Table 116. Luciferase Formulations
Example 79. Fibrin Sealant Study
[001186] Fibrin sealant, such as Tisseel (Baxter Healthcare Corp., Deerfield, IL), is composed of fibrinogen and thrombin in a dual-barreled syringe. Upon mixing, fibrinogen is converted to fibrin to form a fibrin clot in about 10 to 30 seconds. This clot can mimic the natural clotting mechanism of the body. Additionally a fibrin hydrogel is a three dimensional structure that can potentially be used in sustained release delivery. Currently, fibrin sealant is approved for application in hemostasis and sealing to replace conventional surgical techniques such as suture, ligature and cautery.
[001187] The thrombin and fibrinogen components were loaded separately into a dual barreled syringe. Balb-C mice (n=3) were injected subcutaneously with 50 ul of fibrinogen, 50 ul of thrombin and they were also injected at the same site with modified luciferase mRNA (mRNA sequence shown in SEQ ID NO: 16; polyA tail of approximately 140 nucleotides not shown in sequence; 5'cap, Capl; fully modified with 5-methylcytosine and Nl-methylpseudouridine) (Tisseel+Luciferase), 50 ul of fibrinogen and 50 ul thrombin (Tisseel) or modified luciferase mRNA (Luciferase). The injection of fibrinogen and thrombin was done simultaneously using the dual- barreled syringe. The subcutaneous injection of luciferase was done 15 minutes after the
fibrinogen/thrombin injection to allow the fibrin hydrogel to polymerize (Tisseel + Luciferase group). A control group of untreated mice were also evaluated. The mice were imaged at 5 hours and 24 hours. Twenty minutes prior to imaging, mice were injected intraperitoneally with a D- luciferin solution at 150 mg/kg. Animals were then anesthetized and images were acquired with an IVIS Lumina II imaging system (Perkin Elmer). Bioluminescence was measured as total flux (photons/second) of the entire mouse. The luciferase formulations are outlined in Table 117 and the average total flux (photons/second) is shown in Table 118. The fibrin sealant was found to not interfere with imaging and the injection of luciferase and Tisseel showed expression of luciferase. Table 117. Luciferase Formulations
Figure imgf000373_0001
Table 118. Total Flux
Figure imgf000373_0002
Example 80. Fibrin Containing mRNA Sealant Study
A. Modified mRNA and Calcium Chloride
[001188] Prior to reconstitution, luciferase mRNA (mRNA sequence shown in SEQ ID NO: 16; polyA tail of approximately 140 nucleotides not shown in sequence; 5 'cap, Capl) fully modified with 5-methylcytosine and Nl-methylpseudouridine or fully modified with Nl-methylpseudouridine is added to calcium chloride. The calcium chloride is then used to reconstitute thrombin. Fibrinogen is reconstituted with fibrinolysis inhibitor solution per the manufacturer's instructions. The reconstituted thrombin containing modified mRNA and fibrinogen is loaded into a dual barreled syringe. Mice are injected subcutaneously with 50 ul of fibrinogen and50 ul of thrombin containing modified mRNA or they were injected with 50 ul of PBS containing an equivalent dose of modified luciferase mRNA. A control group of untreated mice is also evaluated. The mice are imaged at predetermined intervals to determine the average total flux (photons/second).
B. Lipid Nanoparticle Formulated Modified mRNA and Calcium Chloride
[001189] Prior to reconstitution, luciferase mRNA (mRNA sequence shown in SEQ ID NO: 16; polyA tail of approximately 140 nucleotides not shown in sequence; 5 'cap, Capl) fully modified with 5-methylcytosine and Nl-methylpseudouridine or fully modified with Nl-methylpseudouridine is formulated in a lipid nanoparticle is added to calcium chloride. The calcium chloride is then used to reconstitute thrombin. Fibrinogen is reconstituted with fibrinolysis inhibitor solution per the manufacturer's instructions. The reconstituted thrombin containing modified mRNA and fibrinogen is loaded into a dual barreled syringe. Mice are injected subcutaneously with 50 ul of fibrinogen and50 ul of thrombin containing modified mRNA or they were injected with 50 ul of PBS containing an equivalent dose of modified luciferase mRNA. A control group of untreated mice is also evaluated. The mice are imaged at predetermined intervals to determine the average total flux (photons/second).
C. Modified mRNA and Fibrinogen
[001190] Prior to reconstitution, luciferase mRNA (mRNA sequence shown in SEQ ID NO: 16; polyA tail of approximately 140 nucleotides not shown in sequence; 5 'cap, Capl) fully modified with 5-methylcytosine and Nl-methylpseudouridine or fully modified with Nl-methylpseudouridine is added to the fibrinolysis inhibitor solution. The fibrinolysis inhibitor solution is then used to reconstitute fibrinogen. Thrombin is reconstituted with the calcium chloride solution per the manufacturer's instructions. The reconstituted fibrinogen containing modified mRNA and thrombin is loaded into a dual barreled syringe. Mice are injected subcutaneously with 50 ul of thrombin and 50 ul of fibrinogen containing modified mRNA or they were injected with 50 ul of PBS containing an equivalent dose of modified luciferase mRNA. A control group of untreated mice is also evaluated. The mice are imaged at predetermined intervals to determine the average total flux (photons/second).
D. Lipid Nanoparticle Formualted Modified mRNA and Fibrinogen
[001191] Prior to reconstitution, luciferase mRNA (mRNA sequence shown in SEQ ID NO: 16; polyA tail of approximately 140 nucleotides not shown in sequence; 5 'cap, Capl) fully modified with 5-methylcytosine and Nl-methylpseudouridine or fully modified with Nl-methylpseudouridine is formulated in a lipid nanoparticle is added to the fibrinolysis inhibitor solution. The fibrinolysis inhibitor solution is then used to reconstitute fibrinogen. Thrombin is reconstituted with the calcium chloride solution per the manufacturer's instructions. The reconstituted fibrinogen containing modified mRNA and thrombin is loaded into a dual barreled syringe. Mice are injected
subcutaneously with 50 ul of thrombin and 50 ul of fibrinogen containing modified mRNA or they were injected with 50 ul of PBS containing an equivalent dose of modified luciferase mRNA. A control group of untreated mice is also evaluated. The mice are imaged at predetermined intervals to determine the average total flux (photons/second).
E. Modified mRNA and Thrombin [001192] Prior to reconstitution, luciferase mR A (mR A sequence shown in SEQ ID NO: 16; polyA tail of approximately 140 nucleotides not shown in sequence; 5'cap, Capl) fully modified with 5-methylcytosine and Nl-methylpseudouridine or fully modified with Nl-methylpseudouridine is added to the reconstituted thrombin after it is reconstituted with the calcium chloride per the manufactuer's instructions. The fibrinolysis inhibitor solution is then used to reconstitute fibrinogen per the manufacturer's instructions. The reconstituted fibrinogen and thrombin containing modified mRNA is loaded into a dual barreled syringe. Mice are injected subcutaneously with 50 ul of thrombin containing modified mRNA and 50 ul of fibrinogen or they were injected with 50 ul of PBS containing an equivalent dose of modified luciferase mRNA. A control group of untreated mice is also evaluated. The mice are imaged at predetermined intervals to determine the average total flux (photons/second).
F. Lipid Nanoparticle Formualted Modified mRNA and Thrombin
[001193] Prior to reconstitution, luciferase mRNA (mRNA sequence shown in SEQ ID NO: 16; polyA tail of approximately 140 nucleotides not shown in sequence; 5'cap, Capl) fully modified with 5-methylcytosine and Nl-methylpseudouridine or fully modified with Nl-methylpseudouridine is formulated in a lipid nanoparticle is added to the reconstituted thrombin after it is reconstituted with the calcium chloride per the manufactuer's instructions. The fibrinolysis inhibitor solution is then used to reconstitute fibrinogen per the manufacturer's instructions. The reconstituted fibrinogen and thrombin containing modified mRNA is loaded into a dual barreled syringe. Mice are injected subcutaneously with 50 ul of thrombin containing modified mRNA and 50 ul of fibrinogen or they were injected with 50 ul of PBS containing an equivalent dose of modified luciferase mRNA. A control group of untreated mice is also evaluated. The mice are imaged at predetermined intervals to determine the average total flux (photons/second).
Example 81. Cationic Lipid Formulation of 5-Methylcytosine and Nl-Methylpseudouridine Modified mRNA
[001194] Luciferase mRNA (SEQ ID NO: 16; polyA tail of approximately 140 nucleotides not shown in sequence; 5'cap, Capl) fully modified with 5-methylcytosine and Nl-methylpseudouridine was formulated in the cationic lipids described in Table 119. The formulations were administered intravenously (I.V.), intramuscularly (I.M.) or subcutaneously (S.C.) to Balb-C mice at a dose of 0.05 mg/kg.
Table 119. Cationic Lipid Formulations Formulation NPA- 126-1 NPA- 127-1 NPA- 128-1 NPA- 129-1 111612-B
Lipid DLin-MC3- DLin-KC2- C12-200 DLinDMA DODMA
DMA DMA
Lipid/mR A 20: 1 20:1 20: 1 20: 1 20: 1
ratio (wt/wt)
Mean Size 122 nm 114 nm 153 nm 137 nm 223.2 nm
PDI: 0.13 PDI: 0.10 PDI: 0.17 PDI: 0.09 PDI: 0.142
Zeta at pH 7.4 -1.4 mV -0.5 mV -1.4 mV 2.0 mV -3.09 mV
Encaps. 95% 77% 69% 80% 64%
(RiboGr)
[001195] Twenty minutes prior to imaging, mice were injected intraperitoneally with a D-luciferin solution at 150 mg/kg. Animals were then anesthetized and images were acquired with an IVIS Lumina II imaging system (Perkin Elmer). Bio luminescence was measured as total flux
(photons/second) of the entire mouse. The mice were imaged at 2 hours, 8 hours and 24 hours after dosing and the average total flux (photons/second) was measured for each route of administration and cationic lipid formulation. The background flux was about 4.17E+05 p/s. The results of the imaging are shown in Table 120. In Table 120, "NT" means not tested.
Table 120. Flux
Figure imgf000376_0001
Example 82. Lipid Nanoparticle Intravenous Study
[001196] Luciferase mRNA (SEQ ID NO: 16; polyA tail of approximately 160 nucleotides not shown in sequence; 5'cap, Capl; fully modified with 5-methylcytosine and pseudouridine) was formulated in a lipid nanoparticle containing 50%> DLin-MC3-DMA OR DLin-KC2-DMA as described in Table 121, 38.5% cholesterol, 10% DSPC and 1.5% PEG. The formulation was administered intravenously (I.V.) to Balb-C mice at a dose of 0.5 mg/kg, 0.05 mg/kg, 0.005 mg/kg or 0.0005 mg/kg. Twenty minutes prior to imaging, mice were injected intraperitoneally with a D- luciferin solution at 150 mg/kg. Animals were then anesthetized and images were acquired with an IVIS Lumina II imaging system (Perkin Elmer). Bioluminescence was measured as total flux (photons/second) of the entire mouse.
Table 121. Formulations
Figure imgf000377_0001
[001197] For DLin-KC2-DMA the mice were imaged at 2 hours, 8 hours, 24 hours, 72 hours, 96 hours and 168 hours after dosing and the average total flux (photons/second) was measured for each route of administration and cationic lipid formulation. The background flux was about 3.66E+05 p/s. The results of the imaging are shown in Table 122. Organs were imaged at 8 hours and the average total flux (photons/second) was measured for the liver, spleen, lung and kidney. A control for each organ was also analyzed. The results are shown in Table 123. The peak signal for all dose levels was at 8 hours after administration. Also, distribution to the various organs (liver, spleen, lung, and kidney) may be able to be controlled by increasing or decreasing the LNP dose.
Table 122. Flux
Figure imgf000377_0002
[001198] For DLin-MC 3 -DMA the mice were imaged at 2 hours, 8 hours, 24 hours, 48 hours, 72 hours and 144 hours after dosing and the average total flux (photons/second) was measured for each route of administration and cationic lipid formulation. The background flux was about 4.51E+05 p/s. The results of the imaging are shown in Table 124. Organs were imaged at 8 hours and the average total flux (photons/second) was measured for the liver, spleen, lung and kidney. A control for each organ was also analyzed. The results are shown in Table 125. The peak signal for all dose levels was at 8 hours after administration. Also, distribution to the various organs (liver, spleen, lung, and kidney) may be able to be controlled by increasing or decreasing the LNP dose.
Table 124. Flux
Figure imgf000378_0001
Example 83. Lipid Nanoparticle Subcutaneous Study
[001199] Luciferase mR A (SEQ ID NO: 16; polyA tail of approximately 160 nucleotides not shown in sequence; 5'cap, Capl; fully modified with 5-methylcytosine and pseudouridine) was formulated in a lipid nanoparticle containing 50% DLin-KC2-DMA as described in Table 126, 385% cholesterol, 10% DSPC and 1.5% PEG. The formulation was administered subcutaneously (S.C.) to Balb-C mice at a dose of 0.5 mg/kg, 0.05 mg/kg or 0.005 mg/kg.
Table 126. DLin-KC 2-DM A Formulation
Figure imgf000378_0002
Figure imgf000379_0001
[001200] Twenty minutes prior to imaging, mice were injected intraperitoneally with a D-luciferin solution at 150 mg/kg. Animals were then anesthetized and images were acquired with an IVIS Lumina II imaging system (Perkin Elmer). Bio luminescence was measured as total flux
(photons/second) of the entire mouse. The mice were imaged at 2 hours, 8 hours, 24 hours, 48 hours, 72 hours and 144 hours after dosing and the average total flux (photons/second) was measured for each route of administration and cationic lipid formulation. The lower limit of detection was about 3E+05 p/s. The results of the imaging are shown in Table 127. Organs were imaged at 8 hours and the average total flux (photons/second) was measured for the liver, spleen, lung and kidney. A control for each organ was also analyzed. The results are shown in Table 128. The peak signal for all dose levels was at 8 hours after administration. Also, distribution to the various organs (liver, spleen, lung, and kidney) may be able to be controlled by increasing or decreasing the LNP dose. At high doses, the LNP formulations migrates outside of the subcutaneous injection site, as high levels of luciferase expression are detected in the liver, spleen, lung, and kidney.
Table 127. Flux
Figure imgf000379_0002
Example 84. Catioinic Lipid Nanoparticle Subcutaneous Study [001201] Luciferase mR A (SEQ ID NO: 16; polyA tail of approximately 160 nucleotides not shown in sequence; 5'cap, Capl; fully modified with 5-methylcytosine and pseudouridine) is formulated in a lipid nanoparticle containing 50% DLin-MC3-DMA, 38.5% cholesterol, 10% DSPC and 1.5% PEG. The formulation is administered subcutaneously (S.C.) to Balb-C mice at a dose of 0.5 mg/kg, 0.05 mg/kg or 0.005 mg/kg.
[001202] The mice are imaged at 2 hours, 8 hours, 24 hours, 48 hours, 72 hours and 144 hours after dosing and the average total flux (photons/second) was measured for each route of administration and cationic lipid formulation. Organs are imaged at 8 hours and the average total flux
(photons/second) is measured for the liver, spleen, lung and kidney. A control for each organ is also analyzed.
Example 85. Lipoplex Study
[001203] Lipoplexed luciferase mRNA (SEQ ID NO: 16; polyA tail of approximately 140 nucleotides not shown in sequence; 5'cap, Capl) fully modified with 5-methylcytosine and pseudouridine (5mC/pU), fully modified with 5-methylcytosine and Nl-methylpseudouridine (5mC/NlmpU) or modified where 25% of the cytosines replaced with 5-methylcytosine and 25% of the uridines replaced with 2-thiouridine (5mC/s2U). The formulation was administered
intravenously (I.V.), intramuscularly (I.M.) or subcutaneously (S.C.) to Balb-C mice at a dose of 0.10 mg/kg.
[001204] Twenty minutes prior to imaging, mice were injected intraperitoneally with a D-luciferin solution at 150 mg/kg. Animals were then anesthetized and images were acquired with an IVIS Lumina II imaging system (Perkin Elmer). Bio luminescence was measured as total flux
(photons/second) of the entire mouse. The mice were imaged at 8 hours, 24 hours and 48 hours after dosing and the average total flux (photons/second) was measured for each route of
administration and chemical modification. The background signal was about 3.91E+05 p/s. The results of the imaging are shown in Table 129. Organs were imaged at 6 hours and the average total flux (photons/second) was measured for the liver, spleen, lung and kidney. A control for each organ was also analyzed. The results are shown in Table 130.
Table 129. Flux
Figure imgf000380_0001
I.M. 8 hrs 1.90E+06 2.53E+06 1.29E+06
I.M. 24 hrs 9.33E+05 7.84E+05 6.48E+05
I.M. 48 hrs 8.51E+05 6.59E+05 5.49E+05
S.C. 8 hrs 2.85E+06 6.48E+06 1.14E+06
s.c. 24 hrs 6.66E+05 7.15E+06 3.93E+05
s.c. 48 hrs 3.24E+05 3.20E+06 5.45E+05
Table 130. Organ Flux
Figure imgf000381_0001
Example 86. Cationic Lipid Formulation of Modified mRNA
[001205] Luciferase mRNA (SEQ ID NO: 16; polyA tail of approximately 140 nucleotides not shown in sequence; 5'cap, Capl) modified where 25% of the cytosines replaced with 5- methylcytosine and 25% of the uridines replaced with 2-thiouridine (5mC/s2U) was formulated in the cationic lipids described in Table 131. The formulations were administered intravenously (I.V.), intramuscularly (I.M.) or subcutaneously (S.C.) to Balb-C mice at a dose of 0.05 mg/kg.
Table 131. Cationic Lipid Formulations
Figure imgf000381_0002
[001206] Twenty minutes prior to imaging, mice were injected intraperitoneally with a D-luciferin solution at 150 mg/kg. Animals were then anesthetized and images were acquired with an IVIS Lumina II imaging system (Perkin Elmer). Bio luminescence was measured as total flux (photons/second) of the entire mouse. The mice were imaged at 2 hours, 8 hours and 24 hours after dosing and the average total flux (photons/second) was measured for each route of administration and cationic lipid formulation. The background flux was about 3.31E+05 p/s. The results of the imaging are shown in Table 132. In Table 132, "NT" means not tested. Untreated mice showed an average flux of 3.14E+05 at 2 hours, 3.33E+05 at 8 hours and 3.46E+05 at 24 hours. Peak expression was seen for all three routes tested at 8 hours. DLin-KC2-DMA has better expression than DLin-MC3-DMA and DODMA showed expression for all routes evaluated.
Table 132. Flux
Figure imgf000382_0001
Example 87. Formulation of 5-Methylcytosine and Nl-Methylpseudouridine Modified mRNA
[001207] Luciferase mRNA (SEQ ID NO: 16; polyA tail of approximately 140 nucleotides not shown in sequence; 5'cap, Capl) fully modified with 5-methylcytosine and Nl-methylpseudouridine was formulated in PBS (pH of 7.4). The formulations were administered intramuscularly (I.M.) or subcutaneously (S.C.) to Balb-C mice at a dose of 2.5 mg/kg.
[001208] Twenty minutes prior to imaging, mice were injected intraperitoneally with a D-luciferin solution at 150 mg/kg. Animals were then anesthetized and images were acquired with an IVIS Lumina II imaging system (Perkin Elmer). Bio luminescence was measured as total flux
(photons/second) of the entire mouse. The mice were imaged at 5 minutes, 30 minutes, 60 minutes and 120 minutes after dosing and the average total flux (photons/second) was measured for each route of administration and cationic lipid formulation. The background flux was about 3.78E+05 p/s. The results of the imaging are shown in Table 133. Expression of luciferase was already seen at 30 minutes with both routes of delivery. Peak expression from subcutaneous administration appears between 30 to 60 minutes. Intramuscular expression was still increasing at 120 minutes. Table 133. Flux
Figure imgf000383_0001
Example 88. Intramuscular and Subcutaneous Administration of Chemically Modified mRNA
[001209] Luciferase modified mRNA (mRNA sequence shown in SEQ ID NO: 16; polyA tail of approximately 140 nucleotides not shown in sequence; 5'cap, Capl) fully modified with N4- acetylcytidine, fully modified with 5-methoxyuridine, fully modified with N4-acetylcytidine and Nl-methylpseudouridine or fully modified 5-methylcytosine and 5-methoxyuridine formulated in PBS (pH 7.4) was administered to Balb-C mice intramuscularly or subcutaneously at a dose of 2.5 mg/kg. Twenty minutes prior to imaging, mice were injected intraperitoneally with a D-luciferin solution at 150 mg/kg. Animals were then anesthetized and images were acquired with an IVIS Lumina II imaging system (Perkin Elmer). Bio luminescence was measured as total flux
(photons/second) of the entire mouse. The mice were imaged at 2 hours, 8 hours and 24 hours. The average total flux (photons/second) for intramuscular administration is shown in Table 134_and the average total flux (photons/second) for subcutaneous administration is shown in Table 135. The background signal was 3.84E+05 (p/s). The peak expression for intramuscular administration was seen between 24 and 48 hours for all chemistry and expression was still detected at 120 hours. For subcutaneous delivery the peak expression was seen at 2-8 hours and expression was detected at 72 hours.
Table 134. Intramuscular Administration
Figure imgf000383_0002
Table 135. Subcutaneous Administration
Figure imgf000384_0001
Example 89. In vivo study
[001210] Luciferase modified mRNA containing at least one chemical modification is formulated as a lipid nanoparticle (LNP) using the syringe pump method and characterized by particle size, zeta potential, and encapsulation.
[001211] As outlined in Table 136, the luciferase LNP formulation is administered to Balb-C mice intramuscularly (I.M.), intravenously (I.V.) and subcutaneously (S.C.). As a control luciferase modified RNA formulated in PBS is administered intravenously to mice.
Table 136. Luciferase Formulations
Figure imgf000384_0002
[001212] The mice are imaged at 2, 8, 24, 48, 120 and 192 hours to determine the bioluminescence (measured as total flux (photons/second) of the entire mouse). At 8 hours or 192 hours the liver, spleen, kidney and injection site for subcutaneous and intramuscular administration are imaged to determine the bioluminescence.
Example 90. Cationic Lipid Formulation Studies of Chemically Modified mRNA [001213] Luciferase mR A (SEQ ID NO: 16; polyA tail of approximately 140 nucleotides not shown in sequence; 5 'cap, Capl) fully modified with 5-methylcytosine and pseudouridine
(5mC/pU), pseudouridine (pU) or Nl-methylpseudouridine (NlmpU) was formulated in the cationic lipids described in Table 137. The formulations were administered intravenously (I.V.), intramuscularly (I.M.) or subcutaneously (S.C.) to Balb-C mice at a dose of 0.05 mg/kg.
Table 137. Cationic Lipid Formulations
Figure imgf000385_0001
[001214] Twenty minutes prior to imaging, mice were injected intraperitoneally with a D-luciferin solution at 150 mg/kg. Animals were then anesthetized and images were acquired with an IVIS Lumina II imaging system (Perkin Elmer). Bio luminescence was measured as total flux
(photons/second) of the entire mouse. The mice were imaged at 2 hours, 8 hours and 24 hours after dosing and the average total flux (photons/second) was measured for each route of administration and cationic lipid formulation. The background flux was about 4.1 1E+05 p/s. The results of the imaging are shown in Table 138. Peak expression was seen for all three routes tested at 8 hours.
Table 138. Flux
Figure imgf000385_0002
Example 91. Studies of Chemical Modified mRNA
[001215] Luciferase mRNA (SEQ ID NO: 16; polyA tail of approximately 140 nucleotides not shown in sequence; 5 'cap, Capl) fully modified with N4-acetylcytidine (N4-acetyl), fully modified with 5-methoxyuridine (5meth), fully modified with N4-acetylcytidine and Nl-methylpseudouridine (N4-acetyl/NlmpU) or fully modified with 5-methylcytosine and 5-methoxyuridine (5mC/5-meth) was formulated in DLin-MC3-DMA as described in Table 139. The formulations were administered intravenously (I.V.), intramuscularly (I.M.) or subcutaneously (S.C.) to Balb-C mice at a dose of 0.05 mg/kg.
Table 139. Cationic Lipid Formulations
Figure imgf000386_0001
[001216] Twenty minutes prior to imaging, mice were injected intraperitoneally with a D-luciferin solution at 150 mg/kg. Animals were then anesthetized and images were acquired with an IVIS Lumina II imaging system (Perkin Elmer). Bio luminescence was measured as total flux
(photons/second) of the entire mouse. The mice were imaged at 2 hours, 6 hours and 24 hours after dosing and the average total flux (photons/second) was measured for each route of administration and cationic lipid formulation. The background flux was about 2.70E+05 p/s. The results of the imaging are shown in Table 140.
Table 140. Flux
Figure imgf000386_0002
I.M. 2 hrs 8.59E+06 7.86E+05 5.30E+06 5.1 1E+05
I.M. 6 hrs 1.27E+08 8.88E+06 3.82E+07 3.17E+06
I.M. 24 hrs 4.46E+07 1.38E+06 2.00E+07 1.39E+06
S.C. 2 hrs 1.83E+07 9.67E+05 4.45E+06 1.01E+06
s.c. 6 hrs 2.89E+08 1.78E+07 8.91E+07 1.29E+07
S.C. 24 hrs 6.09E+07 6.40E+06 2.08E+08 6.63E+06
Example 92. PLGA Microspheres
A. Synthesis of PLGA microspheres
[001217] Polylacticglycolic acid (PLGA) microspheres were synthesized using the water/oil/water double emulsification methods known in the art using PLGA-ester cap (Lactel, Cat# B6010-2, inherent viscosity 0.55-0.75, 50:50 LA:GA) or PLGA-acid cap (Lactel, Cat#B6013-2, inherent viscosity 0.55-0.75, 50:50 LA:GA), polyvinylalcohol (PVA) (Sigma, Cat# 348406-25G, MW 13- 23k) dichloromethane and water. Briefly, 0.4 ml of mR A in water (Wl) at 4 mg/ml was added to 2 ml of PLGA dissolved in dichloromethane (DCM) (01) at concentrations ranging from 50 - 200 mg/ml of PLGA. The Wl/Ol emulsion was homogenized (IKA Ultra-Turrax Homogenizer, T18) for 30 seconds at speed 4 (-15,000 rpm). The Wl/Ol emulsion was then added to 250 ml of 1% PVA (W2) and homogenized for 1 minute at speed 5 (-19,000 rpm). Formulations were left to stir for 3 hours, then passed through a 100 μιη nylon mesh strainer (Fisherbrand Cell Strainer, Cat # 22- 363-549) to remove larger aggregates, and finally washed by centrifugation (10 min, 9,250 rpm, 4°C). The supernatant was discarded and the PLGA pellets were resuspended in 5-10 ml of water, which was repeated 2x. The washed formulations were frozen in liquid nitrogen and then lyophilized for 2-3 days.
B. Decreasing homogenization speed or PLGA conentration
[001218] PLGA luciferase microspheres (luciferase mRNA shown in SEQ ID NO: 16; polyA tail of approximately 160 nucleotides not shown in sequence; 5 'cap, Capl; fully modified with 5- methylcysotine and pseudouridine) were made using the conditions described above with ester- capped PLGA. After washing and resuspension with water, 100-200 μΐ of a PLGA microspheres sample was used to measure particle size of the formulations by laser diffraction (Malvern
Mastersizer2000). The particle size of the microspheres made by decreasing homogenization speed during the addition of the first emulsion to the second emulsion with a PLGA concentration of 200 mg/ml is shown in Table 141 and the particle size of the microspheres made by decreasing PLGA concentration in dichloromethane (DCM) is shown in Table 142 with a homogenization speed of 5 during the addition of the first emulsion to the second emulsion. Table 141. Decreasing Homogenization Speed
Figure imgf000388_0001
Table 142. Decreasing PLGA Concentration in DCM
Figure imgf000388_0002
[001219] PLGA with an inherent viscosity of 0.55 - 0.75 either acid or ester-capped was used to make microspheres shown in Table 143. The particle size of the microspheres and the release kinetics were also determined and are shown in Table 143.
Table 143. Decreasing PLGA Concentration in DCM
Figure imgf000388_0003
C. Release study of modified mRNA encapsulated in PLGA microspheres
[001220] PLGA microspheres formulated with Luciferase modified RNA (SEQ ID NO: 16; polyA tail of approximately 160 nucleotides not shown in sequence; 5 'cap, Capl; fully modified with 5-methylcytosine and pseudouridine) were de formulated and the integrity of the extracted modified RNA was determined by automated electrophoresis (Bio-Rad Experion). After lyophilization, -10 mg of PLGA MS were weighed out in 2 ml eppendorf tubes and deformulated by adding 1 ml of DCM and letting the samples shake for 2-6 hrs. mRNA was extracted from the deformulated PLGA micropsheres by adding 0.5 ml of water and shaking the sample overnight. Unformulated luciferase mRNA in water (Deform control) was spiked into DCM and went through the deformulation process to be used as a control. The extracted modified mRNA was compared against unformulated modified mRNA and the deformulation control in order to test the integrity of the encapsulated modified mRNA. The majority of modified RNA was intact for batch ID A, B, C, D, E, as compared to the deformulated control (Deform control) and the unformulated control (Unform control).
D. Release study of modified mRNA encapsulated in PLGA microspheres
[001221] PLGA micropsheres formulated with Luciferase modified RNA (SEQ ID NO: 16; polyA tail of approximately 160 nucleotides not shown in sequence; 5'cap, Capl; fully modified with 5- methylcytosine and pseudouridine) were resuspended in TE buffer to a PLGA microsphere concentration of 80 mg/ml in duplicate or triplicate. After resuspension, samples were kept incubating and shaking at 37°C during the course of the study. For each time point (0.04, 0.25, 1.2, 4, and 7 days), the tubes were centrifuged, the supernantant was removed and the pellet was resupsneded in 0.25 ml of fresh TE buffer. To determine the amount of modified RNA released from the PLGA microspheres, the modified RNA concentration in the supernatant was determined by OD 260. The percent release, shown in Table 144, was calculated based on the total amount of modified RNA in each sample. The release rate of mRNA formulations can be tailored by altering the particle size, the PLGA concentration, and the acid versus ester-end cap.
Table 144. Percent Release
Figure imgf000389_0001
E. Luciferase PLGA Microspheres In Vivo Study
[001222] PLGA microspheres containing luciferase mRNA (SEQ ID NO: 16; polyA tail of approximately 140 nucleotides not shown in sequence; 5'cap, Capl) fully modified with 5- methylcytosine and Nl-methylpseudouridine or fully modified with Nl-methylpseudouridine are formulated as described in Table 145 and injected subcutaneously into mice. Twenty minutes prior to imaging, mice are injected intraperitoneally with a D-luciferin solution at 150 mg/kg. Animals are then anesthetized and images were acquired with an IVIS Lumina II imaging system (Perkin Elmer). Bioluminescence was measured as total flux (photons/second) of the entire mouse.
Table 145. Formulation
Figure imgf000390_0001
Example 93. Buffer Formulations
[001223] Modified mRNA may be formulated in water based buffers. Buffers which are similar to biological systems are traditionally isotonic. Such buffers and buffer solutions may be prepared according to the following guidelines. Example components are given in Table 146.
[001224] In some embodiments, calcium ions may be added to a buffer solution for formulations.
Table 146. Buffers
Figure imgf000390_0002
Example 94. Lipid Nanoparticle Containing A Plurality of Modified mRNAs [001225] EPO mRNA (SEQ ID NO: 9; polyA tail of approximately 140 nucleotides not shown in sequence; 5'cap, Capl; fully modified with 5-methylcytosine and Nl-methylpseudouridine), G-CSF mRNA (SEQ ID NO: 6; polyA tail of approximately 140 nucleotides not shown in sequence; 5'cap, Capl; fully modified with 5-methylcytosine and Nl-methylpseudouridine) and Factor IX mRNA (SEQ ID NO: 10; polyA tail of approximately 140 nucleotides not shown in sequence; 5'cap, Capl; fully modified with 5-methylcytosine and Nl-methylpseudouridine), is formulated in DLin-MC3- DMA as described in Table 147. The formulations are administered intravenously (I.V.), intramuscularly (I.M.) or subcutaneously (S.C.) to Balb-C mice at a dose of 0.05 mg/kg. Control LNP formulations containing only one mRNA are also admininistered at an equivalent dose.
Table 147. DLin-MC3-DMA Formulation
Figure imgf000391_0001
[001226] Serum is collected from the mice at 8 hours, 24 hours, 72 hours and/or 7 days after administration of the formulation. The serum is analyzed by ELISA to determine the protein expression of EPO, G-CSF, and Factor IX.
Example 95. Cationic Lipid Formulation Studies of of 5-Methylcytosine and Nl- Methylpseudouridine Modified mRNA
[001227] EPO mRNA (SEQ ID NO: 9; polyA tail of approximately 140 nucleotides not shown in sequence; 5'cap, Capl; fully modified with 5-methylcytosine and Nl-methylpseudouridine) or G- CSF mRNA (SEQ ID NO: 6; polyA tail of approximately 140 nucleotides not shown in sequence; 5'cap, Capl; fully modified with 5-methylcytosine and Nl-methylpseudouridine) is formulated in DLin-MC3-DMA and DLin-KC2-DMA as described in Table 148. The formulations are administered intravenously (I.V), intramuscularly (I.M.) or subcutaneously (S.C.) to Balb-C mice at a dose of 0.05 mg/kg.
Table 148. DLin-MC3-DMA and DLin-KC2-DMA Formulations
Figure imgf000391_0002
mR A EPO EPO G-CSF G-CSF
DLin-MC3- DLin-KC2- DLin-MC3- DLin-KC2-
Lipid
DMA DMA DMA DMA
Lipid/mR A ratio
20: 1 20: 1 20: 1 20: 1
(wt/wt)
1 17 nm 82 nm 1 19 nm 88 nm
Mean Size
PDI: 0.14 PDI: 0.08 PDI: 0.13 PDI: 0.08
Zeta at pH 7.4 -1.7 mV 0.6 mV 3.6 mV 2.2 mV
Encaps. (RiboGr) 100% 96% 100% 100%
[001228] Serum is collected from the mice at 8 hours, 24 hours, 72 hours and/or 7 days after administration of the formulation. The serum is analyzed by ELISA to determine the protein expression of EPO and G-CSF.
Example 96. Directed SAR of Pseudouridine and Nl-methyl PseudoUridine
[001229] With the recent focus on the pyrimidine nucleoside pseudouridine, a series of structure- activity studies were designed to investigate mRNA containing modifications to pseudouridine or N 1 -methyl-pseudourdine.
[001230] The study was designed to explore the effect of chain length, increased lipophilicity, presence of ring structures, and alteration of hydrophobic or hydrophilic interactions when modifications were made at the Nl position, C6 position, the 2-position, the 4-position and on the phosphate backbone. Stability is also investigated.
[001231] To this end, modifications involving alkylation, cycloalkylation, alkyl-cycloalkylation, arylation, alkyl-arylation, alkylation moieties with amino groups, alkylation moieties with carboxylic acid groups, and alkylation moieties containing amino acid charged moieties are investigated. The degree of alkylation is generally Ci-C6. Examples of the chemistry modifications include those listed in Table 149 and Table 150.
Table 149. Pseudouridine and Nl-methyl Pseudo Uridine SAR
Figure imgf000392_0001
Figure imgf000393_0001
- e-a p a-t o-pseu o-
Table 150. Pseudouridine and Nl-methyl Pseudo Uridine SAR
Compound Naturally
Chemistry Modification
# occuring
N 1 -Methyl-pseudo-UTP 1 Y
Nl-Butyl-pseudo-UTP 2 N
N 1 -tert-Butyl-pseudo-UTP 3 N
N 1 -Pentyl-pseudo-UTP 4 N
Nl-Hexyl-pseudo-UTP 5 N
N 1 -Trifluoromethyl-pseudo-UTP 6 Y
N 1 -Cyclobutyl-pseudo-UTP 7 N
N 1 -Cyclopentyl-pseudo-UTP 8 N
N 1 -Cyclohexyl-pseudo-UTP 9 N
N 1 -Cycloheptyl-pseudo-UTP 10 N
N 1 -Cyclooctyl-pseudo-UTP 11 N
N 1 -Cyclobutylmethyl-pseudo-UTP 12 N
N 1 -Cyclopentylmethyl-pseudo-UTP 13 N
N 1 -Cyclohexylmethyl-pseudo-UTP 14 N
N 1 -Cycloheptylmethyl-pseudo-UTP 15 N N 1 -Cyclooctylmethyl-pseudo-UTP 16 N
Nl-p-tolyl-pseudo-UTP 17 N
N 1 -(2,4,6-Trimethyl-phenyl)pseudo-UTP 18 N
N 1 -(4-Methoxy-phenyl)pseudo-UTP 19 N
N 1 -(4- Amino-phenyl)pseudo-UTP 20 N
N 1 (4-Nitro-phenyl)pseudo-UTP 21 N
Pseudo-UTP-N l-/?-benzoic acid 22 N
N 1 -(4-Methyl-benzyl)pseudo-UTP 24 N
N 1 -(2,4,6-Trimethyl-benzyl)pseudo-UTP 23 N
N 1 -(4-Methoxy-benzyl)pseudo-UTP 25 N
N 1 -(4- Amino-benzyl)pseudo-UTP 26 N
N 1 -(4-Nitro-benzyl)pseudo-UTP 27 N
Pseudo-UTP-N 1 -methyl-p-benzoic acid 28 N
N 1 -(2- Amino-ethyl)pseudo-UTP 29 N
N 1 -(3 - Amino-propyl)pseudo-UTP 30 N
N 1 -(4- Amino-butyl)pseudo-UTP 31 N
N 1 -(5 - Amino-pentyl)pseudo-UTP 32 N
N 1 -(6- Amino-hexyl)pseudo-UTP 33 N
Pseudo-UTP-N 1-3 -propionic acid 34 N
Pseudo-UTP-N 1-4-butanoic acid 35 N
Pseudo-UTP-N 1 -5 -pentanoic acid 36 N
Pseudo-UTP-N 1-6-hexanoic acid 37 N
Pseudo-UTP-N 1-7-heptanoic acid 38 N
N 1 -(2- Amino-2-carboxyethyl)pseudo-UTP 39 N
N 1 -(4- Amino-4-carboxybutyl)pseudo-UTP 40 N
N3-Alkyl-pseudo-UTP 41 N
6-Ethyl-pseudo-UTP 42 N
6-Propyl-pseudo-UTP 43 N
6-iso-Propyl-pseudo-UTP 44 N
6-Butyl-pseudo-UTP 45 N
6-tert-Butyl-pseudo-UTP 46 N
6-(2,2,2-Trifluoroethyl)-pseudo-UTP 47 N
6-Ethoxy-pseudo-UTP 48 N
6-Trifluoromethoxy-pseudo-UTP 49 N
6-Phenyl-pseudo-UTP 50 N
6-(Substituted-Phenyl)-pseudo-UTP 51 N
6-Cyano-pseudo-UTP 52 N
6-Azido-pseudo-UTP 53 N
6-Amino-pseudo-UTP 54 N
6-Ethylcarboxylate-pseudo-UTP 54b N
6-Hydroxy-pseudo-UTP 55 N
6-Methylamino-pseudo-UTP 55b N
6-Dimethylamino-pseudo-UTP 57 N 6-Hydroxyamino-pseudo-UTP 59 N
6-Formyl-pseudo-UTP 60 N
6-(4-Morpholino)-pseudo-UTP 61 N
6-(4-Thiomorpholino)-pseudo-UTP 62 N
N 1 -Me-4-thio-pseudo-UTP 63 N
N 1 -Me-2-thio-pseudo-UTP 64 N
1 ,6-Dimethyl-pseudo-UTP 65 N l-Methyl-6-trifluoromethyl-pseudo-UTP 66 N
1 -Methyl-6-ethyl-pseudo-UTP 67 N
1 -Methyl-6-propyl-pseudo-UTP 68 N l-Methyl-6-iso-propyl-pseudo-UTP 69 N
1 -Methyl-6-butyl-pseudo-UTP 70 N
1 -Methyl-6-tert-butyl-pseudo-UTP 71 N l-Methyl-6-(2,2,2-Trifluoroethyl)pseudo-UTP 72 N
1 -Methyl-6-iodo-pseudo-UTP 73 N
1 -Methyl-6-bromo-pseudo-UTP 74 N
1 -Methyl-6-chloro-pseudo-UTP 75 N
1 -Methyl-6-fluoro-pseudo-UTP 76 N
1 -Methyl-6-methoxy-pseudo-UTP 77 N
1 -Methyl-6-ethoxy-pseudo-UTP 78 N l-Methyl-6-trifluoromethoxy-pseudo-UTP 79 N
1 -Methyl-6-phenyl-pseudo-UTP 80 N
1 -Methyl-6-(substituted phenyl)pseudo-UTP 81 N
1 -Methyl-6-cyano-pseudo-UTP 82 N
1 -Methyl-6-azido-pseudo-UTP 83 N
1 -Methyl-6-amino-pseudo-UTP 84 N l-Methyl-6-ethylcarboxylate-pseudo-UTP 85 N
1 -Methyl-6-hydroxy-pseudo-UTP 86 N l-Methyl-6-methylamino-pseudo-UTP 87 N l-Methyl-6-dimethylamino-pseudo-UTP 88 N l-Methyl-6-hydroxyamino-pseudo-UTP 89 N
1 -Methyl-6-formyl-pseudo-UTP 90 N l-Methyl-6-(4-morpholino)-pseudo-UTP 91 N l-Methyl-6-(4-thiomorpholino)-pseudo-UTP 92 N
1 - Alkyl-6-vinyl-pseudo-UTP 93 N
1 - Alkyl-6-allyl-pseudo-UTP 94 N
1 -Alkyl-6-homoallyl-pseudo-UTP 95 N
1 - Alkyl-6-ethynyl-pseudo-UTP 96 N l-Alkyl-6-(2-propynyl)-pseudo-UTP 97 N
1 - Alkyl-6-( 1 -propynyl)-pseudo-UTP 98 N
Example 97. Incorporation of naturally and non-naturally occuring nucleosides [001232] Naturally and non-naturally occurring nucleosides are incorporated into mRNA encoding a polypeptide of interest. Examples of these are given in Tables 151 and 152. Certain commercially available nucleoside triphosphates (NTPs) are investigated in the polynucleotides of the invention. A selection of these are given in Table 152. The resultant mRNA are then examined for their ability to produce protein, induce cytokines, and/or produce a therapeutic outcome.
Table 151. Naturally and non-naturally occurring nucleosides
Figure imgf000396_0001
Table 152. Non-naturally occurring nucleoside triphosphates
Figure imgf000396_0002
Figure imgf000397_0001
2-Amino-riboside-TP 19 N
Example 98. Incorporation of modifications to the nucleobase and carbohydrate (sugar)
[001233] Naturally and non-naturally occurring nucleosides are incorporated into mRNA encoding a polypeptide of interest. Commercially available nucleosides and NTPs having modifications to both the nucleobase and carbohydrate (sugar) are examined for their ability to be incorporated into mRNA and to produce protein, induce cytokines, and/or produce a therapeutic outcome. Examples of these nucleosides are given in Tables 153 and 154.
Table 153. Combination modifications
Figure imgf000397_0002
Table 154. Naturally occuring combinations
Figure imgf000398_0001
[001234] In the tables "UTP" stands for uridine triphosphate, "GTP" stands for guanosine triphosphate, "ATP" stands for adenosine triphosphate, "CTP" stands for cytosine triphosphate, "TP" stands for triphosphate and "Bz" stands for benzyl.
[001235] It is to be understood that the words which have been used are words of description rather than limitation, and that changes may be made within the purview of the appended claims without departing from the true scope and spirit of the invention in its broader aspects.
[001236] While the present invention has been described at some length and with some particularity with respect to the several described embodiments, it is not intended that it should be limited to any such particulars or embodiments or any particular embodiment, but it is to be construed with references to the appended claims so as to provide the broadest possible interpretation of such claims in view of the prior art and, therefore, to effectively encompass the intended scope of the invention.
[001237] All publications, patent applications, patents, and other references mentioned herein are incorporated by reference in their entirety. In case of conflict, the present specification, including definitions, will control. In addition, section headings, the materials, methods, and examples are illustrative only and not intended to be limiting.

Claims

WHAT IS CLAIMED IS:
1. A method of producing a polypeptide of interest in a mammalian cell or tissue, the method comprising, contacting said mammalian cell or tissue with a formulation comprising a modified mR A encoding the polypeptide of interest, wherein the formulation is selected from the group consisting of nanoparticles, poly(lactic-co-glycolic acid) (PLGA)
microspheres, lipidoid, lipoplex, liposome, polymers, carbohydrates (including simple sugars), cationic lipids, fibrin gel, fibrin hydrogel, fibrin glue, fibrin sealant, fibrinogen, thrombin, rapidly eliminated lipid nanoparticles (reLNPs) and combinations thereof.
2. The method of claim 1, wherein the modified mRNA comprises a purified IVT transcript.
3. The method of claim 1, wherein the formulation comprising the modified mRNA is a
nanoparticle and wherein said nanoparticle comprises at least one lipid.
4. The method of claim 3, wherein the lipid is selected from the group consisting of DLin- DMA, DLin-K-DMA, 98N12-5, C12-200, DLin-MC3 -DMA, DLin-KC2-DMA, DODMA, PLGA, PEG, PEG-DMG and PEGylated lipids.
5. The method of claim 3, wherein the lipid is cationic lipid.
6. The method of claim 5, wherein the cationic lipid is selected from the group consisting of DLin-DMA, DLin-K-DMA, DLin-MC3-DMA, DLin-KC2-DMA and DODMA.
7. The method of claim 3, wherein the lipid to modified mRNA weight ratio is between 10: 1, and 30: 1.
8. The method of claim 7, wherein the mean size of the nanoparticle formulation comprising the modified mRNA is between 60-225 nm.
9. The method of claim 8, wherein the PDI of the nanoparticle formulation comprising the modified mRNA is between 0.03 and 0.15.
10. The method of claim 3, wherein the zeta potential of the lipid is from -10 to +10 at a pH of
11. The method of claim 7, wherein the nanoparticle formulation comprising the modified mRNA further comprises a fusogenic lipid, cholesterol, and a PEG lipid.
12. The method of claim 11, wherein the nanoparticle formulation comprising the modified mRNA has a molar ratio of 50: 10:38.5: 1.5-3.0 (Cationic Lipid: fusogenic lipid: Cholesterol: PEG lipid).
13. The method of claim 12, wherein the PEG lipid is selected from PEG-c-DOMG and PEG- DMG and the fusogenic lipid is DSPC.
14. The method of claim 1, wherein contacting is through the use of a device selected from the group consisting of syringe pump, internal osmotic pump and external osmotic pump.
15. The method of claim 1, wherein the formulation comprising the modified mRNA is a
poly(lactic-co-glycolic acid) (PLGA) microsphere.
16. The method of claim 15, wherein microspheres of the PLGA microsphere formulation
comprising the modified mRNA are between 4 and 20 μιη in size.
17. The method of claim 15, wherein the PLGA microsphere formulation comprising the
modified mRNA release less than 50% of the modified mRNA in a 48 hour time period.
18. The method of claim 15, wherein the PLGA microsphere formulation comprising the
modified mRNA is stable in serum.
19. The method of claim 18, wherein the stability is determined relative to unformulated
modified mRNA in 90% serum.
20. The method of claim 15, wherein the loading weight percent is at least 0.05%, at least 0.1%, at least 0.2%, at least 0.3%, or at least 0.4%.
21. The method of claim 15, wherein the encapsulation efficiency of the modified mRNA in the PLGA microspheres is at least 50%>.
22. The method of claim 15, wherein the encapsulation efficiency of the modified mRNA in the PLGA microspheres is at least 70%>. The method of claim 15, wherein the encapsulation efficiency of the modified mRNA in the PLGA microspheres is at least 90%.
The method of claim 15, wherein the encapsulation efficiency of the modified mRNA in the PLGA microspheres is at least 97%.
The method of claim 11 , wherein contacting said mammalian cells or tissues occurs via a route of administration selected from the group consisting of intraveneous, intramuscular, intra vitreal, intrathecal, intratumoral, pulmonary, and subcutaneous.
The method of claim 25, wherein the polypeptide of interest is detectable in the serum for up to 72 hours after contacting at levels higher than the levels prior to contacting.
The method of claim 26, wherein the polypeptide of interest is detectable in the serum of female subjects at levels greater than in the serum of male subjects.
The method of claim 1 , wherein the formulation further comprises a second modified mRNA.
The method of claim 28, wherein the formulation further comprises a third modified mRNA.
The method of claim 1 , wherein the formulation comprising the modified mRNA comprises a rapidly eliminated lipid nanoparticle.
The method of claim 30, wherein the rapidly eliminated lipid nanoparticle comprises an reLNP lipid, fusogenic lipid, cholesterol and a PEG lipid at a molar ratio of
50: 10:38.5: 1.5(reLNP Lipid: Fusogenic lipid: Cholesterol: PEG lipid).
The method of claim 31, wherein the fusogenic lipid is DSPC and the PEG lipid is PEG-c- DOMG.
The method of claim 31 wherein the reLNP lipid is selected from the group consisting of DLin-DMA with an internal ester, DLin-DMA with a terminal ester, DLin-MC3-DMA-with an internal ester, and DLin-MC3-DMA with a terminal ester.
34. The method of claim 30, wherein in the total lipid to modified mRNA weight ratio is between 10: 1 and 30: 1.
35. The method of claim 1, wherein contacting occurs via injection using a split dosing schedule.
36. The method of claim 35, wherein the injection is made to the tissue selected from the group consisting of intradermal space, epidermis, subcutaneous tissue, and muscle.
37. The method of claim 1, wherein the formulation comprising the modified mRNA comprises a fibrin sealant.
38. The method of claim 1, wherein the formulation comprising the modified mRNA comprises a lipidoid and wherein the lipid is selected from the group consisting of CI 2-200 and 98N12-5.
39. The method of claim 1, wherein the formulation comprising the modified mRNA is a
polymer and said polymer is coated, covered, surrounded, enclosed or comprises a layer of hydrogel or surgical sealant.
40. The method of claim 39, wherein the polymer is selected from the group consisting of
PLGA, ethylene vinyl acetate, poloxamer and GELSITE®.
41. The method of claim 40, further comprising an additional layer of polymer, hydrogel or
surgical sealant.
42. The method of claim 2, wherein the modified mRNA comprises at least one 5' terminal cap selected from the group consisting of CapO, Capl, ARC A, inosine, Nl-methyl-guanosine, 2'fluoro-guanosine, 7-deaza-guanosine, 8-oxo-guanosine, 2-amino-guanosine, LNA- guanosine, and 2-azido-guanosine.
43. The method of claim 42, wherein the 5 ' terminal cap is Cap 1.
44. The method of claim 43, wherein the modified mRNA comprises at least two modifications.
45. The method of claim 44, wherein the at least two modifications are independently selected from the group consisting of 5-methylcytidine, pseudouridine, and 1-methyl-pseudouridine.
46. A method of producing a polypeptide of interest in a mammalian cell or tissue, the method comprising, contacting said mammalian cell or tissue with a buffer formulation comprising a modified mR A encoding the polypeptide of interest.
47. The method of claim 46, wherein the buffer formulation is selected from the group consisting of saline, phosphate buffered saline, and Ringer's lactate.
48. The method of claim 46, wherein the buffer formulation comprises a calcium concentration of between 1-10 mM.
49. The method of claim 46, wherein the modified mRNA comprises a purified IVT transcript.
50. The method of claim 46, wherein contacting said mammalian cells or tissues occurs via a route of administration selected from the group consisting of intraveneous, intramuscular, intra vitreal, intrathecal, intratumoral, pulmonary, and subcutaneous.
51. The method of claim 25 or 50, wherein said polypeptide of interest is produced in said cell or tissue in a location systemic from the location of contacting.
52. The method of claim 51 wherein the route of administration is either via intramuscular or subcutaneous.
53. The method of claim 3, wherein the lipid nanoparticle formulation is further formulated in a sealant.
54. The method of claim 53, wherein said sealant is a fibrin sealant.
55. A method of producing a pharmacologic effect in a primate comprising contacting said
primate with a composition comprising a formulated modified mRNA encoding a polypeptide of interest.
56. The method of claim 55, wherein the modified mRNA comprises a purified IVT transcript.
57. The method of claim 56, wherein the formulation is selected from the group consisting of nanoparticles, poly(lactic-co-glycolic acid) (PLGA) microspheres, lipidoid, lipoplex, liposome, polymers, carbohydrates (including simple sugars), cationic lipids, fibrin gel, fibrin hydrogel, fibrin glue, fibrin sealant, fibrinogen, thrombin, rapidly eliminated lipid nanoparticles (reLNPs) and combinations thereof.
58. The method of claim 57, wherein the pharmacologic effect is greater than the pharmacologic effect associated with a therapeutic agent known to produce said pharmacologic effect.
59. The method of claim 57, wherein the pharmacologic effect is greater than the pharmacologic effect produced by a composition comprising an unformulated modified mR A encoding the polypeptide of interest.
60. The method of claim 57, wherein the pharmacologic effect is greater than the pharmacologic effect produced by a composition comprising a formulated unmodified mRNA encoding the polypeptide of interest.
61. The method of claim 57, wherein the pharmacologic effect results in a therapeutically
effective outcome of a disease, disorder, condition or infection.
62. The method of claim 61, wherein the pharmacologic effect is selected from the group
consisting of a change in cell count, alteration in serum chemistry, alteration of enzyme activity, increase in hemoglobin, and increase in hematocrit.
63. The method of claim 62, wherein the therapeutically effective outcome is selected from the group consisting of treatment, improvement of one or more symptoms, diagnosis, prevention, and delay of onset.
PCT/US2012/069610 2011-12-16 2012-12-14 Modified nucleoside, nucleotide, and nucleic acid compositions WO2013090648A1 (en)

Priority Applications (62)

Application Number Priority Date Filing Date Title
AU2012352180A AU2012352180A1 (en) 2011-12-16 2012-12-14 Modified nucleoside, nucleotide, and nucleic acid compositions
DE12858350.7T DE12858350T1 (en) 2011-12-16 2012-12-14 MODIFIED MRNA COMPOSITIONS
ES12858350T ES2923757T3 (en) 2011-12-16 2012-12-14 Modified mRNA compositions
EP12858350.7A EP2791160B1 (en) 2011-12-16 2012-12-14 Modified mrna compositions
SG11201402666WA SG11201402666WA (en) 2011-12-16 2012-12-14 Modified nucleoside, nucleotide, and nucleic acid compositions
LTEPPCT/US2012/069610T LT2791160T (en) 2011-12-16 2012-12-14 Modified mrna compositions
DK12858350.7T DK2791160T3 (en) 2011-12-16 2012-12-14 MODIFIED MRNA COMPOSITIONS
KR1020147019601A KR20140102759A (en) 2011-12-16 2012-12-14 Modified nucleoside, nucleotide, and nucleic acid compositions
MX2014007233A MX2014007233A (en) 2011-12-16 2012-12-14 Modified nucleoside, nucleotide, and nucleic acid compositions.
EP22159380.9A EP4144378A1 (en) 2011-12-16 2012-12-14 Modified nucleoside, nucleotide, and nucleic acid compositions
HRP20220717TT HRP20220717T1 (en) 2011-12-16 2012-12-14 Modified mrna compositions
CN201280069609.3A CN104114572A (en) 2011-12-16 2012-12-14 Modified nucleoside, nucleotide, and nucleic acid compositions
RS20220503A RS63244B1 (en) 2011-12-16 2012-12-14 Modified mrna compositions
JP2014547454A JP2015501844A (en) 2011-12-16 2012-12-14 Modified nucleosides, nucleotides and nucleic acid compositions
PL12858350T PL2791160T3 (en) 2011-12-16 2012-12-14 Modified mrna compositions
CA2859387A CA2859387A1 (en) 2011-12-16 2012-12-14 Modified nucleoside, nucleotide, and nucleic acid compositions
RU2014129004A RU2649364C2 (en) 2011-12-16 2012-12-14 Modified nucleoside, nucleotide and nucleic acid compositions
SI201232001T SI2791160T1 (en) 2011-12-16 2012-12-14 Modified mrna compositions
US14/103,188 US9220792B2 (en) 2012-04-02 2013-12-11 Modified polynucleotides encoding aquaporin-5
US14/104,568 US9095552B2 (en) 2012-04-02 2013-12-12 Modified polynucleotides encoding copper metabolism (MURR1) domain containing 1
US14/104,591 US9233141B2 (en) 2012-04-02 2013-12-12 Modified polynucleotides for the production of proteins associated with blood and lymphatic disorders
US14/104,585 US20140193482A1 (en) 2012-04-02 2013-12-12 Modified polynucleotides encoding v-myc avian myelocytomatosis viral oncogene homolog
US14/104,556 US9114113B2 (en) 2012-04-02 2013-12-12 Modified polynucleotides encoding citeD4
US14/104,531 US9107886B2 (en) 2012-04-02 2013-12-12 Modified polynucleotides encoding basic helix-loop-helix family member E41
US14/105,214 US20140171485A1 (en) 2012-04-02 2013-12-13 Modified polynucleotides encoding cd28 molecule
US14/105,210 US20150044277A1 (en) 2012-04-02 2013-12-13 Modified polynucleotides encoding caspase 3
US14/105,217 US20140255467A1 (en) 2012-04-02 2013-12-13 Modified polynucleotides encoding cytotoxic t-lymphocyte-associated protein 4
US14/105,221 US20140255468A1 (en) 2012-04-02 2013-12-13 Modified polynucleotides encoding programmed cell death 1
US14/105,224 US9220755B2 (en) 2012-04-02 2013-12-13 Modified polynucleotides for the production of proteins associated with blood and lymphatic disorders
US14/106,957 US9050297B2 (en) 2012-04-02 2013-12-16 Modified polynucleotides encoding aryl hydrocarbon receptor nuclear translocator
US14/107,029 US20140113959A1 (en) 2012-04-02 2013-12-16 Modified polynucleotides encoding caspase 6
US14/106,988 US9301993B2 (en) 2012-04-02 2013-12-16 Modified polynucleotides encoding apoptosis inducing factor 1
US14/107,053 US9216205B2 (en) 2012-04-02 2013-12-16 Modified polynucleotides encoding granulysin
US14/107,079 US9149506B2 (en) 2012-04-02 2013-12-16 Modified polynucleotides encoding septin-4
US14/107,105 US9255129B2 (en) 2012-04-02 2013-12-16 Modified polynucleotides encoding SIAH E3 ubiquitin protein ligase 1
US14/170,744 US20140148502A1 (en) 2012-04-02 2014-02-03 Modified polynucleotides for treating protein deficiency
US14/170,910 US20140155474A1 (en) 2012-04-02 2014-02-03 Modified polynucleotides for treating cystic fibrosis
US14/170,903 US20140155473A1 (en) 2012-04-02 2014-02-03 Modified glucosidase, beta, acid polynucleotides for treating protein deficiency
US14/171,119 US20140179771A1 (en) 2012-04-02 2014-02-03 Modified polynucleotides for treating dolichyl-phosphate (udp-n-acetylglucosamine) n-acetylglucosaminephosphotransferase 1 (glcnac-1-p transferase) protein deficiency
US14/170,751 US20140200263A1 (en) 2012-04-02 2014-02-03 Modified polynucleotides for treating galactosidase, alpha protein deficiency
US14/170,914 US20140155475A1 (en) 2012-04-02 2014-02-03 Modified polynucleotides for treating lipase a, lysosomal acid, cholesterol esterase protein deficiency
US14/170,897 US20140155472A1 (en) 2012-04-02 2014-02-03 Modified polynucleotides for treating argininosuccinate synthase 1 protein deficiency
US14/170,747 US20140194494A1 (en) 2012-04-02 2014-02-03 Modified polynucleotides for treating protein deficiency
US14/171,226 US9061059B2 (en) 2012-04-02 2014-02-03 Modified polynucleotides for treating protein deficiency
US14/171,242 US20140200264A1 (en) 2012-04-02 2014-02-03 Modified polynucleotides for treating carboxypeptidase n, polypeptide 1 protein deficiency
US14/171,249 US9089604B2 (en) 2012-04-02 2014-02-03 Modified polynucleotides for treating galactosylceramidase protein deficiency
US14/171,235 US20140206755A1 (en) 2012-04-02 2014-02-03 Modified polynucleotides for treating arylsulfatase a protein deficiency
US14/280,867 US20140275229A1 (en) 2012-04-02 2014-05-19 Modified polynucleotides encoding udp glucuronosyltransferase 1 family, polypeptide a1
IL232749A IL232749A0 (en) 2011-12-16 2014-05-22 Modified nucleoside, nucleotide, and nucleic acid compositions
ZA2014/03783A ZA201403783B (en) 2011-12-16 2014-05-23 Modified nucleoside, nucleotide, and nucleic acid compositions
HK15103589.4A HK1203077A1 (en) 2011-12-16 2015-04-13 Modified nucleoside, nucleotide, and nucleic acid compositions
US14/694,357 US9283287B2 (en) 2012-04-02 2015-04-23 Modified polynucleotides for the production of nuclear proteins
US14/750,004 US9828416B2 (en) 2012-04-02 2015-06-25 Modified polynucleotides for the production of secreted proteins
US14/975,141 US9504734B2 (en) 2012-04-02 2015-12-18 Modified polynucleotides for the production of nuclear proteins
US15/015,684 US9587003B2 (en) 2012-04-02 2016-02-04 Modified polynucleotides for the production of oncology-related proteins and peptides
US15/060,707 US9782462B2 (en) 2012-04-02 2016-03-04 Modified polynucleotides for the production of proteins associated with human disease
US15/174,219 US20160289674A1 (en) 2012-04-02 2016-06-06 Modified polynucleotides for the production of membrane proteins
US15/425,813 US10501513B2 (en) 2012-04-02 2017-02-06 Modified polynucleotides for the production of oncology-related proteins and peptides
US15/440,523 US20180086807A1 (en) 2012-04-02 2017-02-23 Modified polynucleotides for the production of proteins associated with human disease
US16/665,985 US20210115101A1 (en) 2012-04-02 2019-10-28 Modified polynucleotides for the production of proteins associated with human disease
US16/707,998 US20200247861A1 (en) 2012-04-02 2019-12-09 Modified polynucleotides for the production of oncology-related proteins and peptides
US16/860,121 US20220273555A1 (en) 2012-04-02 2020-04-28 Modified polynucleotides for the production of membrane proteins

Applications Claiming Priority (16)

Application Number Priority Date Filing Date Title
US201161576705P 2011-12-16 2011-12-16
US61/576,705 2011-12-16
US201261618957P 2012-04-02 2012-04-02
US61/618,957 2012-04-02
US201261648244P 2012-05-17 2012-05-17
US61/648,244 2012-05-17
US201261681712P 2012-08-10 2012-08-10
US61/681,712 2012-08-10
US201261696381P 2012-09-04 2012-09-04
US61/696,381 2012-09-04
US201261709303P 2012-10-03 2012-10-03
USPCT/US2012/058519 2012-10-03
US61/709,303 2012-10-03
PCT/US2012/058519 WO2013052523A1 (en) 2011-10-03 2012-10-03 Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US201261712490P 2012-10-11 2012-10-11
US61/712,490 2012-10-11

Publications (1)

Publication Number Publication Date
WO2013090648A1 true WO2013090648A1 (en) 2013-06-20

Family

ID=48610363

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2012/069610 WO2013090648A1 (en) 2011-12-16 2012-12-14 Modified nucleoside, nucleotide, and nucleic acid compositions

Country Status (24)

Country Link
US (17) US20130156849A1 (en)
EP (2) EP4144378A1 (en)
JP (2) JP2015501844A (en)
KR (1) KR20140102759A (en)
CN (2) CN110201187A (en)
AU (3) AU2012352180A1 (en)
CA (2) CA3018046A1 (en)
DE (1) DE12858350T1 (en)
DK (1) DK2791160T3 (en)
ES (1) ES2923757T3 (en)
HK (1) HK1203077A1 (en)
HR (1) HRP20220717T1 (en)
HU (1) HUE059110T2 (en)
IL (1) IL232749A0 (en)
LT (1) LT2791160T (en)
MX (1) MX2014007233A (en)
PL (1) PL2791160T3 (en)
PT (1) PT2791160T (en)
RS (1) RS63244B1 (en)
RU (1) RU2649364C2 (en)
SG (2) SG11201402666WA (en)
SI (1) SI2791160T1 (en)
WO (1) WO2013090648A1 (en)
ZA (1) ZA201403783B (en)

Cited By (124)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013151665A3 (en) * 2012-04-02 2014-02-20 modeRNA Therapeutics Modified polynucleotides for the production of proteins associated with human disease
WO2014081507A1 (en) 2012-11-26 2014-05-30 Moderna Therapeutics, Inc. Terminally modified rna
WO2014113089A2 (en) 2013-01-17 2014-07-24 Moderna Therapeutics, Inc. Signal-sensor polynucleotides for the alteration of cellular phenotypes
US8822663B2 (en) 2010-08-06 2014-09-02 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
WO2015006747A2 (en) 2013-07-11 2015-01-15 Moderna Therapeutics, Inc. Compositions comprising synthetic polynucleotides encoding crispr related proteins and synthetic sgrnas and methods of use.
WO2015034925A1 (en) 2013-09-03 2015-03-12 Moderna Therapeutics, Inc. Circular polynucleotides
WO2015034928A1 (en) 2013-09-03 2015-03-12 Moderna Therapeutics, Inc. Chimeric polynucleotides
US8980864B2 (en) 2013-03-15 2015-03-17 Moderna Therapeutics, Inc. Compositions and methods of altering cholesterol levels
WO2015051214A1 (en) 2013-10-03 2015-04-09 Moderna Therapeutics, Inc. Polynucleotides encoding low density lipoprotein receptor
WO2015075557A2 (en) 2013-11-22 2015-05-28 Mina Alpha Limited C/ebp alpha compositions and methods of use
US9107886B2 (en) 2012-04-02 2015-08-18 Moderna Therapeutics, Inc. Modified polynucleotides encoding basic helix-loop-helix family member E41
US9186372B2 (en) 2011-12-16 2015-11-17 Moderna Therapeutics, Inc. Split dose administration
WO2016014846A1 (en) 2014-07-23 2016-01-28 Moderna Therapeutics, Inc. Modified polynucleotides for the production of intrabodies
US9283287B2 (en) 2012-04-02 2016-03-15 Moderna Therapeutics, Inc. Modified polynucleotides for the production of nuclear proteins
EP2931319A4 (en) * 2012-12-13 2016-05-04 Moderna Therapeutics Inc Modified nucleic acid molecules and uses thereof
US9334328B2 (en) 2010-10-01 2016-05-10 Moderna Therapeutics, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
EP3053585A1 (en) * 2013-12-13 2016-08-10 Moderna Therapeutics, Inc. Alternative nucleic acid molecules and uses thereof
US9428535B2 (en) 2011-10-03 2016-08-30 Moderna Therapeutics, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US9464124B2 (en) 2011-09-12 2016-10-11 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
CN106047879A (en) * 2016-08-18 2016-10-26 广州市锐博生物科技有限公司 Oligonucleotide molecule used for inhibiting expression of mRNA of target gene and composition set thereof
US9533047B2 (en) 2011-03-31 2017-01-03 Modernatx, Inc. Delivery and formulation of engineered nucleic acids
US9572897B2 (en) 2012-04-02 2017-02-21 Modernatx, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
WO2017062513A1 (en) 2015-10-05 2017-04-13 Modernatx, Inc. Methods for therapeutic administration of messenger ribonucleic acid drugs
US9670261B2 (en) 2012-12-21 2017-06-06 Sanofi Functionalized exendin-4 derivatives
WO2017112943A1 (en) 2015-12-23 2017-06-29 Modernatx, Inc. Methods of using ox40 ligand encoding polynucleotides
US9694053B2 (en) 2013-12-13 2017-07-04 Sanofi Dual GLP-1/glucagon receptor agonists
WO2017120612A1 (en) 2016-01-10 2017-07-13 Modernatx, Inc. Therapeutic mrnas encoding anti ctla-4 antibodies
US9750788B2 (en) 2013-12-13 2017-09-05 Sanofi Non-acylated exendin-4 peptide analogues
US9751926B2 (en) 2013-12-13 2017-09-05 Sanofi Dual GLP-1/GIP receptor agonists
US9758561B2 (en) 2014-04-07 2017-09-12 Sanofi Dual GLP-1/glucagon receptor agonists derived from exendin-4
US9771406B2 (en) 2014-04-07 2017-09-26 Sanofi Peptidic dual GLP-1/glucagon receptor agonists derived from exendin-4
US9775904B2 (en) 2014-04-07 2017-10-03 Sanofi Exendin-4 derivatives as peptidic dual GLP-1/glucagon receptor agonists
US9789165B2 (en) 2013-12-13 2017-10-17 Sanofi Exendin-4 peptide analogues as dual GLP-1/GIP receptor agonists
WO2018009838A1 (en) 2016-07-07 2018-01-11 Rubius Therapeutics, Inc. Compositions and methods related to therapeutic cell systems expressing exogenous rna
US9932381B2 (en) 2014-06-18 2018-04-03 Sanofi Exendin-4 derivatives as selective glucagon receptor agonists
EP3177732A4 (en) * 2014-08-08 2018-04-25 ModernaTX, Inc. Compositions and methods for the treatment of ophthalmic diseases and conditions
US9982029B2 (en) 2015-07-10 2018-05-29 Sanofi Exendin-4 derivatives as selective peptidic dual GLP-1/glucagon receptor agonists
US10077439B2 (en) 2013-03-15 2018-09-18 Modernatx, Inc. Removal of DNA fragments in mRNA production process
US10106800B2 (en) 2005-09-28 2018-10-23 Biontech Ag Modification of RNA, producing an increased transcript stability and translation efficiency
EP3122878B1 (en) 2014-03-24 2018-10-24 Translate Bio, Inc. Mrna therapy for the treatment of ocular diseases
WO2018213731A1 (en) 2017-05-18 2018-11-22 Modernatx, Inc. Polynucleotides encoding tethered interleukin-12 (il12) polypeptides and uses thereof
US10138507B2 (en) 2013-03-15 2018-11-27 Modernatx, Inc. Manufacturing methods for production of RNA transcripts
US10155031B2 (en) 2012-11-28 2018-12-18 Biontech Rna Pharmaceuticals Gmbh Individualized vaccines for cancer
WO2019048632A1 (en) 2017-09-08 2019-03-14 Mina Therapeutics Limited Stabilized hnf4a sarna compositions and methods of use
CN109562153A (en) * 2016-08-07 2019-04-02 诺华股份有限公司 The immunization method that mRNA is mediated
US10258698B2 (en) 2013-03-14 2019-04-16 Modernatx, Inc. Formulation and delivery of modified nucleoside, nucleotide, and nucleic acid compositions
EP3364982A4 (en) * 2015-10-22 2019-04-17 ModernaTX, Inc. Sexually transmitted disease vaccines
US10286086B2 (en) 2014-06-19 2019-05-14 Modernatx, Inc. Alternative nucleic acid molecules and uses thereof
EP3369816A4 (en) * 2015-10-30 2019-06-26 Bonac Corporation Composition stably containing single-stranded nucleic acid molecule that suppresses expression of tgf-beta1 gene
EP3365007A4 (en) * 2015-10-22 2019-07-03 ModernaTX, Inc. Broad spectrum influenza virus vaccine
EP3365009A4 (en) * 2015-10-22 2019-07-03 ModernaTX, Inc. Herpes simplex virus vaccine
EP3364980A4 (en) * 2015-10-22 2019-07-10 ModernaTX, Inc. Nucleic acid vaccines for varicella zoster virus (vzv)
EP3364981A4 (en) * 2015-10-22 2019-08-07 ModernaTX, Inc. Human cytomegalovirus vaccine
US10385088B2 (en) 2013-10-02 2019-08-20 Modernatx, Inc. Polynucleotide molecules and uses thereof
WO2019158720A1 (en) 2018-02-16 2019-08-22 Mina Therapeutics Limited C/ebp alpha sarna compositions and methods of use
US10407683B2 (en) 2014-07-16 2019-09-10 Modernatx, Inc. Circular polynucleotides
US10413598B2 (en) 2014-11-12 2019-09-17 Ucl Business Plc Factor IX gene therapy
WO2019197845A1 (en) 2018-04-12 2019-10-17 Mina Therapeutics Limited Sirt1-sarna compositions and methods of use
EP3364950A4 (en) * 2015-10-22 2019-10-23 ModernaTX, Inc. Tropical disease vaccines
EP3364983A4 (en) * 2015-10-22 2019-10-23 ModernaTX, Inc. Respiratory virus vaccines
US10485884B2 (en) 2012-03-26 2019-11-26 Biontech Rna Pharmaceuticals Gmbh RNA formulation for immunotherapy
US10501513B2 (en) 2012-04-02 2019-12-10 Modernatx, Inc. Modified polynucleotides for the production of oncology-related proteins and peptides
WO2019239144A1 (en) 2018-06-15 2019-12-19 Mina Therapeutics Limited Combination therapies comprising c/ebp alpha sarna
US10525075B2 (en) 2013-02-22 2020-01-07 The Board Of Trustees Of The Leland Stanford Junior University Compounds, compositions, methods, and kits relating to telomere extension
RU2714404C2 (en) * 2014-01-31 2020-02-14 Фэктор Байосайенс Инк. Methods and products for producing and delivering nucleic acids
US10590161B2 (en) 2013-03-15 2020-03-17 Modernatx, Inc. Ion exchange purification of mRNA
WO2020056155A3 (en) * 2018-09-13 2020-04-16 Modernatx, Inc. Polynucleotides encoding branched-chain alpha-ketoacid dehydrogenase complex e1-alpha, e1-beta, and e2 subunits for the treatment of maple syrup urine disease
US10709779B2 (en) 2014-04-23 2020-07-14 Modernatx, Inc. Nucleic acid vaccines
US10724052B2 (en) 2018-09-07 2020-07-28 Crispr Therapeutics Ag Universal donor cells
US10738355B2 (en) 2011-05-24 2020-08-11 Tron-Translationale Onkologie An Der Universitätsmedizin Der Johannes Gutenberg-Universität Mainz Ggmbh Individualized vaccines for cancer
US10758592B2 (en) 2012-10-09 2020-09-01 Sanofi Exendin-4 derivatives as dual GLP1/glucagon agonists
WO2020208361A1 (en) 2019-04-12 2020-10-15 Mina Therapeutics Limited Sirt1-sarna compositions and methods of use
US10806797B2 (en) 2015-06-05 2020-10-20 Sanofi Prodrugs comprising an GLP-1/glucagon dual agonist linker hyaluronic acid conjugate
US10842885B2 (en) 2018-08-20 2020-11-24 Ucl Business Ltd Factor IX encoding nucleotides
WO2021007515A1 (en) 2019-07-11 2021-01-14 Tenaya Therapeutics, Inc. Cardiac cell reprogramming with micrornas and other factors
WO2021032777A1 (en) 2019-08-19 2021-02-25 Mina Therapeutics Limited Oligonucleotide conjugate compositions and methods of use
US11015211B2 (en) 2018-08-30 2021-05-25 Tenaya Therapeutics, Inc. Cardiac cell reprogramming with myocardin and ASCL1
US11104918B2 (en) 2019-09-05 2021-08-31 Crispr Therapeutics Ag Universal donor cells
WO2021178246A1 (en) 2020-03-02 2021-09-10 Tenaya Therapeutics, Inc. Gene vector control by cardiomyocyte-expressed micrornas
US11116797B2 (en) 2019-09-05 2021-09-14 Crispr Therapeutics Ag Universal donor cells
US11141378B2 (en) 2008-04-15 2021-10-12 Arbutus Biopharma Corporation Lipid formulations for nucleic acid delivery
US11156617B2 (en) 2015-02-12 2021-10-26 BioNTech RNA Pharmaceuticals GbmH Predicting T cell epitopes useful for vaccination
US11173120B2 (en) 2014-09-25 2021-11-16 Biontech Rna Pharmaceuticals Gmbh Stable formulations of lipids and liposomes
US11207398B2 (en) 2017-09-14 2021-12-28 Modernatx, Inc. Zika virus mRNA vaccines
US11222711B2 (en) 2013-05-10 2022-01-11 BioNTech SE Predicting immunogenicity of T cell epitopes
US11298426B2 (en) 2003-10-14 2022-04-12 BioNTech SE Recombinant vaccines and use thereof
US20220125723A1 (en) 2010-07-06 2022-04-28 Glaxosmithkline Biologicals Sa Lipid formulations with viral immunogens
WO2022113056A1 (en) 2020-11-30 2022-06-02 Crispr Therapeutics Ag Gene-edited natural killer cells
US11364292B2 (en) 2015-07-21 2022-06-21 Modernatx, Inc. CHIKV RNA vaccines
US11377470B2 (en) 2013-03-15 2022-07-05 Modernatx, Inc. Ribonucleic acid purification
US11407997B2 (en) 2017-02-22 2022-08-09 Crispr Therapeutics Ag Materials and methods for treatment of primary hyperoxaluria type 1 (PH1) and other alanine-glyoxylate aminotransferase (AGXT) gene related conditions or disorders
US11434486B2 (en) 2015-09-17 2022-09-06 Modernatx, Inc. Polynucleotides containing a morpholino linker
US11446383B2 (en) 2009-07-01 2022-09-20 Arbutus Biopharma Corporation Lipid formulations for delivery of therapeutic agents
WO2022200810A1 (en) 2021-03-26 2022-09-29 Mina Therapeutics Limited Tmem173 sarna compositions and methods of use
EP3324979B1 (en) 2015-07-21 2022-10-12 ModernaTX, Inc. Infectious disease vaccines
WO2022229644A1 (en) 2021-04-28 2022-11-03 Mina Therapeutics Limited Combination therapies comprising c/ebp alpha sarna
US11492628B2 (en) 2015-10-07 2022-11-08 BioNTech SE 3′-UTR sequences for stabilization of RNA
US11547764B2 (en) 2011-06-08 2023-01-10 Translate Bio, Inc. Lipid nanoparticle compositions and methods for MRNA delivery
US11547673B1 (en) 2020-04-22 2023-01-10 BioNTech SE Coronavirus vaccine
US11559588B2 (en) 2017-02-22 2023-01-24 Crispr Therapeutics Ag Materials and methods for treatment of Spinocerebellar Ataxia Type 1 (SCA1) and other Spinocerebellar Ataxia Type 1 Protein (ATXN1) gene related conditions or disorders
US11566230B2 (en) 2020-12-31 2023-01-31 Crispr Therapeutics Ag Universal donor cells
US11564997B2 (en) 2016-06-29 2023-01-31 Crispr Therapeutics Ag Materials and methods for treatment of friedreich ataxia and other related disorders
US11596645B2 (en) 2010-07-06 2023-03-07 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
WO2023031394A1 (en) 2021-09-03 2023-03-09 CureVac SE Novel lipid nanoparticles for delivery of nucleic acids
EP4159741A1 (en) 2014-07-16 2023-04-05 ModernaTX, Inc. Method for producing a chimeric polynucleotide encoding a polypeptide having a triazole-containing internucleotide linkage
EP4023755B1 (en) 2014-12-12 2023-04-26 CureVac SE Artificial nucleic acid molecules for improved protein expression
WO2023069498A1 (en) 2021-10-22 2023-04-27 Senda Biosciences, Inc. Mrna vaccine composition
US11639370B2 (en) 2010-10-11 2023-05-02 Glaxosmithkline Biologicals Sa Antigen delivery platforms
WO2023073228A1 (en) 2021-10-29 2023-05-04 CureVac SE Improved circular rna for expressing therapeutic proteins
US11655475B2 (en) 2010-07-06 2023-05-23 Glaxosmithkline Biologicals Sa Immunisation of large mammals with low doses of RNA
WO2023096858A1 (en) 2021-11-23 2023-06-01 Senda Biosciences, Inc. A bacteria-derived lipid composition and use thereof
WO2023099884A1 (en) 2021-12-01 2023-06-08 Mina Therapeutics Limited Pax6 sarna compositions and methods of use
WO2023122080A1 (en) 2021-12-20 2023-06-29 Senda Biosciences, Inc. Compositions comprising mrna and lipid reconstructed plant messenger packs
EP4219715A2 (en) 2017-09-08 2023-08-02 MiNA Therapeutics Limited Stabilized cebpa sarna compositions and methods of use
WO2023144330A1 (en) 2022-01-28 2023-08-03 CureVac SE Nucleic acid encoded transcription factor inhibitors
US11718852B2 (en) 2010-06-30 2023-08-08 Arbutus Biopharma Corporation Non-liposomal systems for nucleic acid delivery
WO2023170435A1 (en) 2022-03-07 2023-09-14 Mina Therapeutics Limited Il10 sarna compositions and methods of use
US11759422B2 (en) 2010-08-31 2023-09-19 Glaxosmithkline Biologicals Sa Pegylated liposomes for delivery of immunogen-encoding RNA
WO2023227608A1 (en) 2022-05-25 2023-11-30 Glaxosmithkline Biologicals Sa Nucleic acid based vaccine encoding an escherichia coli fimh antigenic polypeptide
US11878055B1 (en) 2022-06-26 2024-01-23 BioNTech SE Coronavirus vaccine
US11896636B2 (en) 2011-07-06 2024-02-13 Glaxosmithkline Biologicals Sa Immunogenic combination compositions and uses thereof
US11920148B2 (en) 2017-02-22 2024-03-05 Crispr Therapeutics Ag Compositions and methods for gene editing
US11926817B2 (en) 2019-08-09 2024-03-12 Nutcracker Therapeutics, Inc. Microfluidic apparatus and methods of use thereof
US11951180B2 (en) 2023-04-03 2024-04-09 Translate Bio, Inc. Lipid nanoparticle compositions and methods for MRNA delivery

Families Citing this family (309)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101083907B (en) * 2004-03-03 2012-10-10 雷文斯治疗公司 Compositions and methods for topical diagnostic and therapeutic transport
US9211248B2 (en) 2004-03-03 2015-12-15 Revance Therapeutics, Inc. Compositions and methods for topical application and transdermal delivery of botulinum toxins
EP1861112A4 (en) * 2005-03-03 2009-07-22 Revance Therapeutics Inc Compositions and methods for topical application and transdermal delivery of botulinum toxins
US8153435B1 (en) 2005-03-30 2012-04-10 Tracer Detection Technology Corp. Methods and articles for identifying objects using encapsulated perfluorocarbon tracers
NZ733634A (en) 2009-12-01 2022-10-28 Translate Bio Inc Delivery of mrna for the augmentation of proteins and enzymes in human genetic diseases
CA2796464C (en) 2010-04-16 2021-08-03 Immune Disease Institute, Inc. Sustained polypeptide expression from synthetic, modified rnas and uses thereof
AU2011248108B2 (en) 2010-05-04 2016-05-26 Corium Pharma Solutions, Inc. Method and device for transdermal delivery of parathyroid hormone using a microprojection array
CA2816420C (en) * 2010-10-29 2018-01-16 Najib Babul Compositions of (-)-17-(cyclobutylmethyl)morphinan-3,14-diol
US8853377B2 (en) 2010-11-30 2014-10-07 Shire Human Genetic Therapies, Inc. mRNA for use in treatment of human genetic diseases
EP2734621B1 (en) 2011-07-22 2019-09-04 President and Fellows of Harvard College Evaluation and improvement of nuclease cleavage specificity
DK3260140T3 (en) 2011-12-05 2021-04-19 Factor Bioscience Inc METHODS AND CELL TRANSFER PROCEDURES
ES2921724T1 (en) 2011-12-07 2022-08-31 Alnylam Pharmaceuticals Inc Biodegradable lipids for the administration of active agents
CN104519915A (en) 2012-06-08 2015-04-15 夏尔人类遗传性治疗公司 Pulmonary delivery of mRNA to non-lung target cells
EP3536787A1 (en) 2012-06-08 2019-09-11 Translate Bio, Inc. Nuclease resistant polynucleotides and uses thereof
EP2885419A4 (en) 2012-08-14 2016-05-25 Moderna Therapeutics Inc Enzymes and polymerases for the synthesis of rna
US9095625B2 (en) * 2012-08-31 2015-08-04 University Of Massachusetts Graft-copolymer stabilized metal nanoparticles
US20150246138A1 (en) * 2012-09-04 2015-09-03 Lauren Sciences Llc Bolaamphiphilic compounds, compositions and uses thereof
US20140284270A1 (en) * 2012-09-21 2014-09-25 George Fox RNA pores and methods and compositions for making and using same
RU2711249C2 (en) 2012-11-01 2020-01-15 Фэктор Байосайенс Инк. Methods and products for expression of proteins in cells
US20140179770A1 (en) 2012-12-12 2014-06-26 Massachusetts Institute Of Technology Delivery, engineering and optimization of systems, methods and compositions for sequence manipulation and therapeutic applications
RU2698095C2 (en) * 2012-12-21 2019-08-22 Кориум Интернэшнл, Инк. Microarray for therapeutic agent delivery and methods of using
US20160024181A1 (en) 2013-03-13 2016-01-28 Moderna Therapeutics, Inc. Long-lived polynucleotide molecules
WO2014152774A1 (en) 2013-03-14 2014-09-25 Shire Human Genetic Therapies, Inc. Methods and compositions for delivering mrna coded antibodies
DK2970955T3 (en) 2013-03-14 2019-02-11 Translate Bio Inc METHODS FOR CLEANING MESSENGER RNA
KR20210122917A (en) 2013-03-14 2021-10-12 샤이어 휴먼 지네틱 테라피즈 인크. Cftr mrna compositions and related methods and uses
ES2795249T3 (en) * 2013-03-15 2020-11-23 Translate Bio Inc Synergistic enhancement of nucleic acid delivery through mixed formulations
JP6700170B2 (en) 2013-03-15 2020-05-27 コリウム, インコーポレイテッド Microarrays for delivery of therapeutic agents and methods of use
EP4286517A3 (en) 2013-04-04 2024-03-13 President and Fellows of Harvard College Therapeutic uses of genome editing with crispr/cas systems
MX2015017311A (en) 2013-06-17 2017-04-10 Broad Inst Inc Delivery, engineering and optimization of systems, methods and compositions for targeting and modeling diseases and disorders of post mitotic cells.
CN114015726A (en) 2013-06-17 2022-02-08 布罗德研究所有限公司 Delivery, use and therapeutic applications of CRISPR-CAS systems and compositions for targeting disorders and diseases with viral components
BR112015031608A2 (en) 2013-06-17 2017-08-22 Massachusetts Inst Technology APPLICATION AND USE OF CRISPR-CAS SYSTEMS, VECTORS AND COMPOSITIONS FOR LIVER TARGETING AND THERAPY
WO2015006498A2 (en) 2013-07-09 2015-01-15 President And Fellows Of Harvard College Therapeutic uses of genome editing with crispr/cas systems
CA2919226A1 (en) * 2013-07-23 2015-01-29 Protiva Biotherapeutics, Inc. Compositions and methods for delivering messenger rna
US9163284B2 (en) 2013-08-09 2015-10-20 President And Fellows Of Harvard College Methods for identifying a target site of a Cas9 nuclease
US9359599B2 (en) 2013-08-22 2016-06-07 President And Fellows Of Harvard College Engineered transcription activator-like effector (TALE) domains and uses thereof
US9322037B2 (en) 2013-09-06 2016-04-26 President And Fellows Of Harvard College Cas9-FokI fusion proteins and uses thereof
US9737604B2 (en) 2013-09-06 2017-08-22 President And Fellows Of Harvard College Use of cationic lipids to deliver CAS9
US9340799B2 (en) 2013-09-06 2016-05-17 President And Fellows Of Harvard College MRNA-sensing switchable gRNAs
WO2015048744A2 (en) 2013-09-30 2015-04-02 Moderna Therapeutics, Inc. Polynucleotides encoding immune modulating polypeptides
CN106413811A (en) 2013-10-22 2017-02-15 夏尔人类遗传性治疗公司 Mrna therapy for argininosuccinate synthetase deficiency
CA2928040A1 (en) 2013-10-22 2015-04-30 Shire Human Genetic Therapies, Inc. Cns delivery of mrna and uses thereof
AU2014340083B2 (en) 2013-10-22 2019-08-15 Translate Bio, Inc. mRNA therapy for phenylketonuria
CN105813656B (en) 2013-10-22 2021-01-15 夏尔人类遗传性治疗公司 Lipid formulations for delivery of messenger RNA
ES2670983T3 (en) 2013-11-07 2018-06-04 Editas Medicine, Inc. Methods and compositions related to CRISPR with rRNA guiding
US10098982B2 (en) 2013-11-20 2018-10-16 Drexel University Compositions and methods for macrophage conversion
WO2015089486A2 (en) 2013-12-12 2015-06-18 The Broad Institute Inc. Systems, methods and compositions for sequence manipulation with optimized functional crispr-cas systems
BR112016013547A2 (en) 2013-12-12 2017-10-03 Broad Inst Inc COMPOSITIONS AND METHODS OF USE OF CRISPR-CAS SYSTEMS IN NUCLEOTIDE REPEAT DISORDERS
AU2014361781B2 (en) 2013-12-12 2021-04-01 Massachusetts Institute Of Technology Delivery, use and therapeutic applications of the CRISPR -Cas systems and compositions for genome editing
WO2015089473A1 (en) 2013-12-12 2015-06-18 The Broad Institute Inc. Engineering of systems, methods and optimized guide compositions with new architectures for sequence manipulation
KR20160089530A (en) 2013-12-12 2016-07-27 더 브로드 인스티튜트, 인코퍼레이티드 Delivery, use and therapeutic applications of the crispr-cas systems and compositions for hbv and viral diseases and disorders
US11053481B2 (en) 2013-12-12 2021-07-06 President And Fellows Of Harvard College Fusions of Cas9 domains and nucleic acid-editing domains
WO2015089419A2 (en) 2013-12-12 2015-06-18 The Broad Institute Inc. Delivery, use and therapeutic applications of the crispr-cas systems and compositions for targeting disorders and diseases using particle delivery components
ES2895941T3 (en) 2014-02-10 2022-02-23 Univ Leland Stanford Junior Activation of innate immunity for enhanced nuclear reprogramming of somatic cells with mRNA
WO2015151048A1 (en) * 2014-04-01 2015-10-08 Institut National De La Sante Et De La Recherche Medicale (Inserm) Capped and uncapped rna molecules and block copolymers for intracellular delivery of rna
EP3134506B1 (en) 2014-04-25 2019-08-07 Translate Bio, Inc. Methods for purification of messenger rna
WO2015175748A1 (en) * 2014-05-14 2015-11-19 Evorx Technologies, Inc. Methods and compositions for controlling gene expression and treating cancer
BR112016027705A2 (en) 2014-05-30 2018-01-30 Shire Human Genetic Therapies biodegradable lipids for nucleic acid delivery
SG11201610670WA (en) 2014-06-24 2017-01-27 Shire Human Genetic Therapies Stereochemically enriched compositions for delivery of nucleic acids
CA2953341C (en) 2014-06-25 2023-01-24 Acuitas Therapeutics Inc. Lipids and lipid nanoparticle formulations for delivery of nucleic acids
JP2017520549A (en) * 2014-06-26 2017-07-27 ラモット アット テル アビブ ユニバーシティ, リミテッド Liposome formulation for delivery of nucleic acids
CN114344275A (en) 2014-07-02 2022-04-15 川斯勒佰尔公司 Encapsulation of messenger RNA
FR3023483A1 (en) * 2014-07-09 2016-01-15 Jerome Lemoine PROCESS FOR PREPARING MESSENGER RNA NANOPARTICLES AND HYPOTONIC AQUEOUS COMPOSITION COMPRISING THE ARNM NANOPARTICLES
EP4079847A1 (en) 2014-07-30 2022-10-26 President And Fellows Of Harvard College Cas9 proteins including ligand-dependent inteins
EP3686279B1 (en) 2014-08-17 2023-01-04 The Broad Institute, Inc. Genome editing using cas9 nickases
WO2016049163A2 (en) 2014-09-24 2016-03-31 The Broad Institute Inc. Use and production of chd8+/- transgenic animals with behavioral phenotypes characteristic of autism spectrum disorder
WO2016049258A2 (en) 2014-09-25 2016-03-31 The Broad Institute Inc. Functional screening with optimized functional crispr-cas systems
KR101776368B1 (en) * 2014-10-02 2017-09-07 서울시립대학교 산학협력단 mRNA nanoparticles and manufacturing method thereof
US9816080B2 (en) 2014-10-31 2017-11-14 President And Fellows Of Harvard College Delivery of CAS9 via ARRDC1-mediated microvesicles (ARMMs)
US20170362627A1 (en) 2014-11-10 2017-12-21 Modernatx, Inc. Multiparametric nucleic acid optimization
EP3884964A1 (en) 2014-12-05 2021-09-29 Translate Bio, Inc. Messenger rna therapy for treatment of articular disease
WO2016094872A1 (en) 2014-12-12 2016-06-16 The Broad Institute Inc. Dead guides for crispr transcription factors
WO2016094867A1 (en) 2014-12-12 2016-06-16 The Broad Institute Inc. Protected guide rnas (pgrnas)
WO2016094880A1 (en) 2014-12-12 2016-06-16 The Broad Institute Inc. Delivery, use and therapeutic applications of crispr systems and compositions for genome editing as to hematopoietic stem cells (hscs)
WO2016094874A1 (en) 2014-12-12 2016-06-16 The Broad Institute Inc. Escorted and functionalized guides for crispr-cas systems
EP3234192B1 (en) 2014-12-19 2021-07-14 The Broad Institute, Inc. Unbiased identification of double-strand breaks and genomic rearrangement by genome-wide insert capture sequencing
WO2016106236A1 (en) 2014-12-23 2016-06-30 The Broad Institute Inc. Rna-targeting system
EP3237615B2 (en) 2014-12-24 2023-07-26 The Broad Institute, Inc. Crispr having or associated with destabilization domains
AU2015374296B2 (en) 2014-12-29 2021-09-02 Novartis Ag Methods of making chimeric antigen receptor-expressing cells
WO2016118724A1 (en) * 2015-01-21 2016-07-28 Moderna Therapeutics, Inc. Lipid nanoparticle compositions
CN107530436B (en) 2015-01-21 2022-03-29 菲泽尔克斯公司 Methods, compositions and systems for delivering therapeutic and diagnostic agents into cells
US20180085474A1 (en) * 2015-01-23 2018-03-29 Moderna Therapeutics, Inc. Lipid nanoparticle compositions
JP7199809B2 (en) 2015-02-13 2023-01-06 ファクター バイオサイエンス インコーポレイテッド Nucleic acid product and its administration method
JP6895892B2 (en) 2015-03-19 2021-06-30 トランスレイト バイオ, インコーポレイテッド MRNA treatment for Pompe disease
WO2016168595A1 (en) 2015-04-17 2016-10-20 Barrett David Maxwell Methods for improving the efficacy and expansion of chimeric antigen receptor-expressing cells
US10682401B2 (en) 2015-05-19 2020-06-16 Morphogenesis, Inc. Multi-indication mRNA cancer immunotherapy
WO2016189532A1 (en) * 2015-05-26 2016-12-01 Ramot At Tel-Aviv University Ltd. Targeted lipid particles for systemic delivery of nucleic acid molecules to leukocytes
EP3929287A3 (en) 2015-06-18 2022-04-13 The Broad Institute, Inc. Crispr enzyme mutations reducing off-target effects
US9790490B2 (en) 2015-06-18 2017-10-17 The Broad Institute Inc. CRISPR enzymes and systems
WO2016205745A2 (en) 2015-06-18 2016-12-22 The Broad Institute Inc. Cell sorting
WO2016205749A1 (en) 2015-06-18 2016-12-22 The Broad Institute Inc. Novel crispr enzymes and systems
EP3430134B1 (en) 2015-06-18 2022-09-21 The Broad Institute, Inc. Novel crispr enzymes and systems
US10857093B2 (en) 2015-06-29 2020-12-08 Corium, Inc. Microarray for delivery of therapeutic agent, methods of use, and methods of making
IL283545B2 (en) 2015-06-29 2023-09-01 Acuitas Therapeutics Inc Lipids and lipid nanoparticle formulations for delivery of nucleic acids
EP3325504A1 (en) 2015-07-21 2018-05-30 Novartis AG Methods for improving the efficacy and expansion of immune cells
JP2018521084A (en) * 2015-07-24 2018-08-02 フィブラリン コーポレイションFibralign Corp. Compositions for targeted delivery of nucleic acid based therapeutics
MA42543A (en) * 2015-07-30 2018-06-06 Modernatx Inc CONCATEMERIC PEPTIDIC EPITOPE RNA
WO2017031232A1 (en) 2015-08-17 2017-02-23 Modernatx, Inc. Methods for preparing particles and related compositions
US11214800B2 (en) 2015-08-18 2022-01-04 The Broad Institute, Inc. Methods and compositions for altering function and structure of chromatin loops and/or domains
PT3350157T (en) 2015-09-17 2022-03-18 Modernatx Inc Compounds and compositions for intracellular delivery of therapeutic agents
EP3349730B1 (en) 2015-09-18 2020-12-02 Dnarx Systems for nucleic acid expression in vivo
US10792374B2 (en) 2015-10-09 2020-10-06 Case Western Reserve University Compositions and methods for the delivery of nucleic acids
WO2017069958A2 (en) 2015-10-09 2017-04-27 The Brigham And Women's Hospital, Inc. Modulation of novel immune checkpoint targets
EP3362555B1 (en) 2015-10-14 2021-01-13 Translate Bio, Inc. Modification of rna-related enzymes for enhanced production
US20170211142A1 (en) 2015-10-22 2017-07-27 The Broad Institute, Inc. Novel crispr enzymes and systems
CN114404581A (en) * 2015-10-22 2022-04-29 摩登纳特斯有限公司 Cancer vaccine
AU2016341311B2 (en) 2015-10-22 2023-11-16 Modernatx, Inc. Respiratory syncytial virus vaccine
CN108513575A (en) 2015-10-23 2018-09-07 哈佛大学的校长及成员们 Nucleobase editing machine and application thereof
WO2017075038A1 (en) * 2015-10-26 2017-05-04 Rana Therapeutics, Inc. Nanoparticle formulations for delivery of nucleic acid complexes
WO2017074788A1 (en) 2015-10-27 2017-05-04 The Broad Institute Inc. Compositions and methods for targeting cancer-specific sequence variations
WO2017075451A1 (en) 2015-10-28 2017-05-04 The Broad Institute Inc. Compositions and methods for evaluating and modulating immune responses by detecting and targeting pou2af1
WO2017075465A1 (en) 2015-10-28 2017-05-04 The Broad Institute Inc. Compositions and methods for evaluating and modulating immune responses by detecting and targeting gata3
EP3368689B1 (en) 2015-10-28 2020-06-17 The Broad Institute, Inc. Composition for modulating immune responses by use of immune cell gene signature
WO2018081480A1 (en) 2016-10-26 2018-05-03 Acuitas Therapeutics, Inc. Lipid nanoparticle formulations
RS63986B1 (en) 2015-10-28 2023-03-31 Acuitas Therapeutics Inc Novel lipids and lipid nanoparticle formulations for delivery of nucleic acids
US11413346B2 (en) 2015-11-09 2022-08-16 Curevac Ag Rotavirus vaccines
US20190307857A1 (en) 2015-12-09 2019-10-10 Modernatx, Inc. MODIFIED mRNA ENCODING A URIDINE DIPHOPSPHATE GLUCURONOSYL TRANSFERASE AND USES THEREOF
WO2017100551A1 (en) 2015-12-09 2017-06-15 Alexion Pharmaceuticals, Inc. HETEROLOGOUS UTR SEQUENCES FOR ENHANCED mRNA EXPRESSION
LT3386484T (en) 2015-12-10 2022-06-10 Modernatx, Inc. Compositions and methods for delivery of therapeutic agents
US20190233814A1 (en) 2015-12-18 2019-08-01 The Broad Institute, Inc. Novel crispr enzymes and systems
US10799463B2 (en) 2015-12-22 2020-10-13 Modernatx, Inc. Compounds and compositions for intracellular delivery of agents
US10465190B1 (en) 2015-12-23 2019-11-05 Modernatx, Inc. In vitro transcription methods and constructs
CN108700535B (en) * 2015-12-23 2021-10-12 加利福尼亚大学董事会 Nanosensor for nucleic acid detection and identification
CA3009852A1 (en) 2015-12-28 2017-07-06 Novartis Ag Methods of making chimeric antigen receptor-expressing cells
EP3405579A1 (en) 2016-01-22 2018-11-28 Modernatx, Inc. Messenger ribonucleic acids for the production of intracellular binding polypeptides and methods of use thereof
US11780934B2 (en) 2016-02-05 2023-10-10 Institut Pasteur Use of inhibitors of ADAM12 as adjuvants in tumor therapies
US20210189062A1 (en) 2016-03-01 2021-06-24 Alexion Pharmaceuticals, Inc. Biodegradable activated polymers for therapeutic delivery
WO2017177169A1 (en) 2016-04-08 2017-10-12 Rana Therapeutics, Inc. Multimeric coding nucleic acid and uses thereof
WO2017180917A2 (en) 2016-04-13 2017-10-19 Modernatx, Inc. Lipid compositions and their uses for intratumoral polynucleotide delivery
US20200263190A1 (en) 2016-04-19 2020-08-20 The Broad Institute, Inc. Novel crispr enzymes and systems
WO2017184786A1 (en) 2016-04-19 2017-10-26 The Broad Institute Inc. Cpf1 complexes with reduced indel activity
AU2017253089B2 (en) 2016-04-19 2023-07-20 Massachusetts Institute Of Technology Novel CRISPR enzymes and systems
US10751423B2 (en) 2016-05-02 2020-08-25 Massachusetts Institute Of Technology Nanoparticle conjugates of highly potent toxins and intraperitoneal administration of nanoparticles for treating or imaging cancer
ES2871537T3 (en) * 2016-05-09 2021-10-29 Astrazeneca Ab Lipid nanoparticles comprising lipophilic anti-inflammatory agents and methods of using the same
EP3458034A4 (en) 2016-05-18 2020-01-01 ModernaTX, Inc. Polynucleotides encoding relaxin
RS62556B1 (en) 2016-06-07 2021-12-31 Modernatx Inc Modified rna encoding vegf-a polypeptides, formulations, and uses relating thereto
US10835583B2 (en) 2016-06-13 2020-11-17 Translate Bio, Inc. Messenger RNA therapy for the treatment of ornithine transcarbamylase deficiency
JP2019525901A (en) * 2016-06-14 2019-09-12 モデルナティエックス インコーポレイテッドModernaTX,Inc. Stabilized preparation of lipid nanoparticles
KR20230156150A (en) 2016-06-17 2023-11-13 더 브로드 인스티튜트, 인코퍼레이티드 Type vi crispr orthologs and systems
US20210222164A1 (en) 2016-06-29 2021-07-22 The Broad Institute, Inc. Crispr-cas systems having destabilization domain
JP7086870B2 (en) 2016-06-30 2022-06-20 アルブータス・バイオファーマー・コーポレイション Compositions and Methods for Delivering Messenger RNA
KR20230095129A (en) 2016-08-03 2023-06-28 프레지던트 앤드 펠로우즈 오브 하바드 칼리지 Adenosine nucleobase editors and uses thereof
EP3497214B1 (en) 2016-08-09 2023-06-28 President and Fellows of Harvard College Programmable cas9-recombinase fusion proteins and uses thereof
WO2018035388A1 (en) 2016-08-17 2018-02-22 The Broad Institute, Inc. Novel crispr enzymes and systems
CN110312799A (en) 2016-08-17 2019-10-08 博德研究所 Novel C RISPR enzyme and system
IL308824A (en) 2016-08-17 2024-01-01 Factor Bioscience Inc Nucleic acid products and methods of administration thereof
WO2018039438A1 (en) 2016-08-24 2018-03-01 President And Fellows Of Harvard College Incorporation of unnatural amino acids into proteins using base editing
WO2018049025A2 (en) 2016-09-07 2018-03-15 The Broad Institute Inc. Compositions and methods for evaluating and modulating immune responses
JP6983455B2 (en) 2016-09-14 2021-12-17 モデルナティーエックス, インコーポレイテッド High-purity RNA composition and methods for its preparation
US11730823B2 (en) 2016-10-03 2023-08-22 President And Fellows Of Harvard College Delivery of therapeutic RNAs via ARRDC1-mediated microvesicles
WO2018067991A1 (en) 2016-10-07 2018-04-12 The Brigham And Women's Hospital, Inc. Modulation of novel immune checkpoint targets
KR101896615B1 (en) * 2016-10-13 2018-09-07 한국과학기술연구원 A method for packing polynucleotide
SG11201903089RA (en) 2016-10-14 2019-05-30 Harvard College Aav delivery of nucleobase editors
CA3041307A1 (en) 2016-10-21 2018-04-26 Giuseppe Ciaramella Human cytomegalovirus vaccine
US20190274968A1 (en) * 2016-10-27 2019-09-12 The Trustees Of The University Of Pennsylvania Nucleoside-modified rna for inducing an adaptive immune response
WO2018089540A1 (en) 2016-11-08 2018-05-17 Modernatx, Inc. Stabilized formulations of lipid nanoparticles
AU2017356190A1 (en) * 2016-11-10 2019-05-16 Translate Bio, Inc. Subcutaneous delivery of messenger RNA
WO2018089851A2 (en) 2016-11-11 2018-05-17 Modernatx, Inc. Influenza vaccine
EP3973955A3 (en) * 2016-11-23 2022-06-15 Mayo Foundation for Medical Education and Research Particle-mediated delivery of inhibitory rna
MA50335A (en) 2016-12-08 2020-08-19 Modernatx Inc NUCLEIC ACID VACCINES AGAINST RESPIRATORY VIRUSES
CA3045122A1 (en) * 2016-12-09 2018-06-14 Sangamo Therapeutics, Inc. Delivery of target specific nucleases
WO2018111967A1 (en) 2016-12-13 2018-06-21 Modernatx, Inc. Rna affinity purification
CN110494549A (en) * 2016-12-22 2019-11-22 阿维塔斯有限公司 By invertibity infiltration to the carrier-free Intracellular delivery of non-adherent cell
WO2018119359A1 (en) 2016-12-23 2018-06-28 President And Fellows Of Harvard College Editing of ccr5 receptor gene to protect against hiv infection
WO2018144775A1 (en) * 2017-02-01 2018-08-09 Modernatx, Inc. Immunomodulatory therapeutic mrna compositions encoding activating oncogene mutation peptides
MA47515A (en) 2017-02-16 2019-12-25 Modernatx Inc VERY POWERFUL IMMUNOGENIC COMPOSITIONS
EA201991747A1 (en) 2017-02-27 2020-06-04 Транслейт Био, Инк. NEW CODON-OPTIMIZED CFTR mRNA
US11898179B2 (en) 2017-03-09 2024-02-13 President And Fellows Of Harvard College Suppression of pain by gene editing
JP2020510439A (en) 2017-03-10 2020-04-09 プレジデント アンド フェローズ オブ ハーバード カレッジ Base-editing factor from cytosine to guanine
US11576961B2 (en) 2017-03-15 2023-02-14 Modernatx, Inc. Broad spectrum influenza virus vaccine
US11203569B2 (en) 2017-03-15 2021-12-21 Modernatx, Inc. Crystal forms of amino lipids
WO2018170306A1 (en) 2017-03-15 2018-09-20 Modernatx, Inc. Compounds and compositions for intracellular delivery of therapeutic agents
MA47787A (en) 2017-03-15 2020-01-22 Modernatx Inc RESPIRATORY SYNCYTIAL VIRUS VACCINE
WO2018170270A1 (en) 2017-03-15 2018-09-20 Modernatx, Inc. Varicella zoster virus (vzv) vaccine
US11752206B2 (en) 2017-03-15 2023-09-12 Modernatx, Inc. Herpes simplex virus vaccine
EP3596207B1 (en) 2017-03-15 2023-12-20 The Broad Institute, Inc. Novel cas13b orthologues crispr enzymes and systems
US20200030432A1 (en) * 2017-03-17 2020-01-30 Modernatx, Inc. Zoonotic disease rna vaccines
US11268082B2 (en) 2017-03-23 2022-03-08 President And Fellows Of Harvard College Nucleobase editors comprising nucleic acid programmable DNA binding proteins
WO2018187590A1 (en) 2017-04-05 2018-10-11 Modernatx, Inc. Reduction or elimination of immune responses to non-intravenous, e.g., subcutaneously administered therapeutic proteins
US11840711B2 (en) 2017-04-12 2023-12-12 The Broad Institute, Inc. Type VI CRISPR orthologs and systems
WO2018191719A1 (en) 2017-04-13 2018-10-18 Acuitas Therapeutics, Inc. Lipid delivery of therapeutic agents to adipose tissue
WO2018191657A1 (en) 2017-04-13 2018-10-18 Acuitas Therapeutics, Inc. Lipids for delivery of active agents
CN110536694A (en) 2017-04-20 2019-12-03 Atyr 医药公司 For treating pulmonary inflammatory composition and method
WO2018200975A1 (en) 2017-04-27 2018-11-01 Vanderbilt University Hepatitis c virus gene sequences and methods of use therefor
CN110799492B (en) 2017-04-28 2023-06-27 爱康泰生治疗公司 Novel carbonyl lipid and lipid nanoparticle formulations for delivery of nucleic acids
US11591601B2 (en) 2017-05-05 2023-02-28 The Broad Institute, Inc. Methods for identification and modification of lncRNA associated with target genotypes and phenotypes
WO2018209320A1 (en) 2017-05-12 2018-11-15 President And Fellows Of Harvard College Aptazyme-embedded guide rnas for use with crispr-cas9 in genome editing and transcriptional activation
WO2018213476A1 (en) 2017-05-16 2018-11-22 Translate Bio, Inc. Treatment of cystic fibrosis by delivery of codon-optimized mrna encoding cftr
NZ759034A (en) * 2017-05-22 2023-07-28 Takeda Pharmaceuticals Co Viral vectors encoding recombinant fix with increased expression for gene therapy of hemophilia b
EP3638215A4 (en) 2017-06-15 2021-03-24 Modernatx, Inc. Rna formulations
US10034951B1 (en) 2017-06-21 2018-07-31 New England Biolabs, Inc. Use of thermostable RNA polymerases to produce RNAs having reduced immunogenicity
MA49634A (en) 2017-07-21 2020-05-27 Modernatx Inc MODIFIED RNA CODING FOR A PROPIONYL-COA-CARBOXYLASE AND ASSOCIATED USES
MA49684A (en) 2017-07-24 2020-06-03 Modernatx Inc GLUCOSE-6-PHOSPHATASE MODIFIED RNA AND ASSOCIATED USES
US11732274B2 (en) 2017-07-28 2023-08-22 President And Fellows Of Harvard College Methods and compositions for evolving base editors using phage-assisted continuous evolution (PACE)
JP7355731B2 (en) 2017-08-16 2023-10-03 アクイタス セラピューティクス インコーポレイテッド Lipids for use in lipid nanoparticle formulations
US11542225B2 (en) 2017-08-17 2023-01-03 Acuitas Therapeutics, Inc. Lipids for use in lipid nanoparticle formulations
WO2019036030A1 (en) 2017-08-17 2019-02-21 Acuitas Therapeutics, Inc. Lipids for use in lipid nanoparticle formulations
MA49914A (en) 2017-08-18 2021-04-21 Modernatx Inc HPLC ANALYTICAL PROCESSES
MA49922A (en) 2017-08-18 2021-06-02 Modernatx Inc PROCESSES FOR HPLC ANALYSIS
JP7408098B2 (en) 2017-08-18 2024-01-05 モデルナティエックス インコーポレイテッド RNA polymerase variants
WO2019139645A2 (en) 2017-08-30 2019-07-18 President And Fellows Of Harvard College High efficiency base editors comprising gam
US11744801B2 (en) 2017-08-31 2023-09-05 Modernatx, Inc. Methods of making lipid nanoparticles
GB201714430D0 (en) * 2017-09-07 2017-10-25 Micol Romain Compositions and processes for targeted delivery and expression and modulation of therapeutic components in tissue
CA3073848A1 (en) 2017-09-21 2019-03-28 The Broad Institute, Inc. Systems, methods, and compositions for targeted nucleic acid editing
CA3077255A1 (en) * 2017-09-29 2019-04-04 Intellia Therapeutics, Inc. Polynucleotides, compositions, and methods for genome editing
US11795443B2 (en) 2017-10-16 2023-10-24 The Broad Institute, Inc. Uses of adenosine base editors
JP2021500894A (en) 2017-10-25 2021-01-14 ノバルティス アーゲー Method for producing chimeric antigen receptor-expressing cells
WO2019089828A1 (en) 2017-10-31 2019-05-09 Acuitas Therapeutics, Inc. Lamellar lipid nanoparticles
WO2019089803A1 (en) 2017-10-31 2019-05-09 The Broad Institute, Inc. Methods and compositions for studying cell evolution
SI3703658T1 (en) * 2017-10-31 2022-09-30 Astrazeneca Ab Lipid nanoparticles for delivering modified rna encoding a vegf-a polypeptide
WO2019094983A1 (en) 2017-11-13 2019-05-16 The Broad Institute, Inc. Methods and compositions for treating cancer by targeting the clec2d-klrb1 pathway
US11167043B2 (en) 2017-12-20 2021-11-09 Translate Bio, Inc. Composition and methods for treatment of ornithine transcarbamylase deficiency
AU2018397910A1 (en) * 2017-12-27 2020-07-16 Takeda Pharmaceutical Company Limited Nucleic acid-containing lipid nano-particle and use thereof
CN109988777A (en) * 2017-12-29 2019-07-09 南京金斯瑞生物科技有限公司 Glutamine synthetase gene and application
EP3746090A4 (en) 2018-01-29 2021-11-17 ModernaTX, Inc. Rsv rna vaccines
AU2019221826A1 (en) * 2018-02-19 2020-09-17 Combined Therapeutics, Inc. Compositions and methods for organ-protective expression and modulation of coding ribonucleic acids
US20190292596A1 (en) * 2018-03-21 2019-09-26 Roche Molecular Systems, Inc. Modified nucleoside phosphates with high thermal stability
KR102114787B1 (en) * 2018-04-02 2020-05-26 중앙대학교 산학협력단 Preparing method for 3 dimension corneal endothelial graft comprising ribonuclease 5 overexepressing corneal endothelial cells
US10968257B2 (en) 2018-04-03 2021-04-06 The Broad Institute, Inc. Target recognition motifs and uses thereof
JP2021519595A (en) * 2018-04-17 2021-08-12 キュアバック アーゲー New RSV RNA molecule and vaccination composition
CA3098382A1 (en) * 2018-04-24 2019-10-31 Ligandal, Inc. Methods and compositions for genome editing
AU2019258679A1 (en) * 2018-04-25 2020-10-15 Ethris Gmbh Cryoprotective agents for particulate formulations
JP7241782B2 (en) * 2018-06-22 2023-03-17 ザ トラスティーズ オブ コロンビア ユニバーシティ イン ザ シティ オブ ニューヨーク Sustained Release Compositions and Methods for Treatment of Temporomandibular Joint Degenerative Diseases
US20210315819A1 (en) * 2018-06-28 2021-10-14 Astrazeneca Ab Exosome extracellular vesicles and methods of use
CN112673100A (en) 2018-07-09 2021-04-16 格罗生物科学公司 Compositions comprising non-standard amino acids and uses thereof
CN112513270A (en) 2018-07-13 2021-03-16 加利福尼亚大学董事会 Retrotransposon-based delivery vehicles and methods of use thereof
US20210315820A1 (en) 2018-07-30 2021-10-14 Trucode Gene Repair, Inc. Lipid nanoparticle formulations comprising nucleic acid mimics
JP2021533098A (en) * 2018-08-03 2021-12-02 サンガモ セラピューティクス, インコーポレイテッド Improved clinical parameters due to factor VIII expression
CN113286884A (en) 2018-08-07 2021-08-20 博德研究所 Novel CAS12B enzymes and systems
US20210317429A1 (en) 2018-08-20 2021-10-14 The Broad Institute, Inc. Methods and compositions for optochemical control of crispr-cas9
US11174500B2 (en) 2018-08-24 2021-11-16 Translate Bio, Inc. Methods for purification of messenger RNA
JP2021534783A (en) 2018-08-31 2021-12-16 ノバルティス アーゲー Method for producing chimeric antigen receptor-expressing cells
EP3844265A2 (en) 2018-08-31 2021-07-07 Novartis AG Methods of making chimeric antigen receptor-expressing cells
CA3113449A1 (en) 2018-09-21 2020-03-26 Acuitas Therapeutics, Inc. Systems and methods for manufacturing lipid nanoparticles and liposomes
US11072808B2 (en) 2018-10-04 2021-07-27 New England Biolabs, Inc. Methods and compositions for increasing capping efficiency of transcribed RNA
EP3861108A1 (en) 2018-10-04 2021-08-11 New England Biolabs, Inc. Methods and compositions for increasing capping efficiency of transcribed rna
CA3114699A1 (en) 2018-10-09 2020-04-16 The University Of British Columbia Compositions and systems comprising transfection-competent vesicles free of organic-solvents and detergents and methods related thereto
TW202022112A (en) * 2018-10-18 2020-06-16 日商武田藥品工業股份有限公司 Method for activation/proliferation of t cells
US20210395721A1 (en) 2018-10-24 2021-12-23 Codiak Biosciences, Inc. Methods to improve potency of electroporation
MX2021008358A (en) 2019-01-11 2021-09-30 Acuitas Therapeutics Inc Lipids for lipid nanoparticle delivery of active agents.
CN109810184B (en) * 2019-01-17 2022-07-12 武汉明德生物科技股份有限公司 Human NF155 antigen, human NF155 antibody detection kit, preparation method and application thereof
US11351242B1 (en) 2019-02-12 2022-06-07 Modernatx, Inc. HMPV/hPIV3 mRNA vaccine composition
CA3129532A1 (en) 2019-02-15 2020-08-20 Crispr Therapeutics Ag Gene editing for hemophilia a with improved factor viii expression
KR20210142636A (en) 2019-02-20 2021-11-25 모더나티엑스, 인크. RNA polymerase variants for co-transcriptional capping
US11851694B1 (en) 2019-02-20 2023-12-26 Modernatx, Inc. High fidelity in vitro transcription
JP2022523222A (en) * 2019-03-01 2022-04-21 フラッグシップ パイオニアリング イノベーションズ シックス,エルエルシー Compositions, Methods, and Kits for Delivery of Polyribonucleotides
GB2595606B (en) 2019-03-07 2022-09-21 Univ California CRISPR-Cas effector polypeptides and methods of use thereof
WO2020191102A1 (en) 2019-03-18 2020-09-24 The Broad Institute, Inc. Type vii crispr proteins and systems
WO2020191249A1 (en) 2019-03-19 2020-09-24 The Broad Institute, Inc. Methods and compositions for editing nucleotide sequences
EP3947646A1 (en) * 2019-04-05 2022-02-09 Precision BioSciences, Inc. Methods of preparing populations of genetically-modified immune cells
US20220168389A1 (en) 2019-04-12 2022-06-02 Novartis Ag Methods of making chimeric antigen receptor-expressing cells
US11490453B2 (en) * 2019-05-16 2022-11-01 Apple Inc. Self-organizing device
WO2020236972A2 (en) 2019-05-20 2020-11-26 The Broad Institute, Inc. Non-class i multi-component nucleic acid targeting systems
RU2721562C1 (en) * 2019-06-04 2020-05-20 Автономная некоммерческая образовательная организация высшего образования "Сколковский институт науки и технологий" Method of loading inorganic nanoparticles or organic molecules into porous particles of micron or submicron size
WO2020247382A1 (en) * 2019-06-05 2020-12-10 Guide Therapeutics, Inc. Analysis of materials for tissue delivery
US10501404B1 (en) 2019-07-30 2019-12-10 Factor Bioscience Inc. Cationic lipids and transfection methods
DE112020003843T5 (en) 2019-08-14 2022-05-19 Acuitas Therapeutics, Inc. Improved lipid nanoparticles for delivery of nucleic acids
EP4031524A1 (en) 2019-09-19 2022-07-27 ModernaTX, Inc. Branched tail lipid compounds and compositions for intracellular delivery of therapeutic agents
US20220397570A1 (en) * 2019-09-27 2022-12-15 Merck Sharp & Dohme Corp. Method for measuring nucleic acid content in lipid nanoparticles using ultraviolet spectrometry
JP7389134B2 (en) 2019-11-15 2023-11-29 富士フイルム株式会社 lipid composition
MX2022006391A (en) 2019-11-26 2022-06-24 Novartis Ag Chimeric antigen receptors binding bcma and cd19 and uses thereof.
CN111041025B (en) 2019-12-17 2021-06-18 深圳市瑞吉生物科技有限公司 mRNA targeting molecule based on combination of N-acetylgalactosamine polypeptide and preparation method thereof
CN111217867B (en) * 2020-02-18 2024-03-22 福建瑞博奥科技有限公司 Method for preparing floxuridine
CN112121180B (en) * 2020-02-19 2023-08-25 深圳厚存纳米药业有限公司 Composite nanoparticles of poloxamer and or poloxamer and PEG lipids
CN111944157B (en) * 2020-02-21 2022-02-11 武汉中科先进技术研究院有限公司 Nano magnetic bead and application thereof in 2019 novel coronavirus nucleic acid extraction, amplification and detection
EP4110377A2 (en) 2020-02-27 2023-01-04 Novartis AG Methods of making chimeric antigen receptor-expressing cells
MX2022012105A (en) 2020-03-30 2022-10-18 BioNTech SE Rna compositions targeting claudin-18.2.
EP4132478A1 (en) 2020-04-09 2023-02-15 Finncure Oy Mimetic nanoparticles for preventing the spreading and lowering the infection rate of novel coronaviruses
DE112021002672T5 (en) 2020-05-08 2023-04-13 President And Fellows Of Harvard College METHODS AND COMPOSITIONS FOR EDIT BOTH STRANDS SIMULTANEOUSLY OF A DOUBLE STRANDED NUCLEOTIDE TARGET SEQUENCE
KR20230035043A (en) 2020-06-15 2023-03-10 더 리서치 인스티튜트 앳 네이션와이드 칠드런스 하스피탈 Adeno-associated viral vector delivery for muscular dystrophy
CN111744019B (en) 2020-07-01 2023-08-04 深圳瑞吉生物科技有限公司 Mannose-based mRNA targeted delivery system and application thereof
AU2021326420A1 (en) 2020-07-31 2023-03-09 Combined Therapeutics, Inc. Compositions and methods for improved vaccination
US11406703B2 (en) 2020-08-25 2022-08-09 Modernatx, Inc. Human cytomegalovirus vaccine
CN112076149B (en) * 2020-09-09 2022-03-01 上海交通大学 Coumarin targeted controlled-release nanogel and preparation method thereof
EP4212162A4 (en) 2020-09-14 2024-02-28 Fujifilm Corp Lipid composition
KR20230164648A (en) 2020-12-22 2023-12-04 큐어백 에스이 RNA vaccines against SARS-CoV-2 variants
US11524023B2 (en) 2021-02-19 2022-12-13 Modernatx, Inc. Lipid nanoparticle compositions and methods of formulating the same
EP4319803A1 (en) 2021-04-08 2024-02-14 Vaxthera SAS Coronavirus vaccine comprising a mosaic protein
AU2022273530A1 (en) 2021-05-12 2023-11-23 Massachusetts Institute Of Technology Modified mrna, modified non-coding rna, and uses thereof
KR20230008627A (en) 2021-07-07 2023-01-16 주식회사 제넥신 Hyaluronic acid-lipid derivatives, lipid nanoparticles comprising the same, and uses thereof
WO2023003826A2 (en) * 2021-07-20 2023-01-26 The Regents Of The University Of California Single- and multi-epitope peptide and mrna vaccines to generate tolerogenic effects for allergic and autoimmune disease by targeting liver sinusoidal endothelial cells
WO2023023055A1 (en) 2021-08-16 2023-02-23 Renagade Therapeutics Management Inc. Compositions and methods for optimizing tropism of delivery systems for rna
CA3231523A1 (en) 2021-09-14 2023-03-23 Renagade Therapeutics Management Inc. Acyclic lipids and methods of use thereof
TW202328067A (en) 2021-09-14 2023-07-16 美商雷納嘉德醫療管理公司 Cyclic lipids and methods of use thereof
WO2023064469A1 (en) 2021-10-13 2023-04-20 Modernatx, Inc. Compositions of mrna-encoded il15 fusion proteins and methods of use thereof
WO2023081756A1 (en) 2021-11-03 2023-05-11 The J. David Gladstone Institutes, A Testamentary Trust Established Under The Will Of J. David Gladstone Precise genome editing using retrons
WO2023091490A1 (en) 2021-11-16 2023-05-25 Senda Biosciences, Inc. Novel ionizable lipids and lipid nanoparticles and methods of using the same
WO2023091787A1 (en) 2021-11-22 2023-05-25 Senda Biosciences, Inc. Novel ionizable lipids and lipid nanoparticles and methods of using the same
WO2023122752A1 (en) 2021-12-23 2023-06-29 Renagade Therapeutics Management Inc. Constrained lipids and methods of use thereof
WO2023131254A1 (en) * 2022-01-06 2023-07-13 上海吉量医药工程有限公司 Pseudouridine modified at n1 position and use thereof in mrna synthesis
WO2023141602A2 (en) 2022-01-21 2023-07-27 Renagade Therapeutics Management Inc. Engineered retrons and methods of use
WO2023147090A1 (en) 2022-01-27 2023-08-03 BioNTech SE Pharmaceutical compositions for delivery of herpes simplex virus antigens and related methods
WO2023149775A1 (en) * 2022-02-07 2023-08-10 한국과학기술원 Oligonucleotide for gene delivery and lipid nanoparticle for gene delivery comprising same
WO2023172747A1 (en) * 2022-03-11 2023-09-14 W. L. Gore & Associates, Inc. Bioabsorbable particles and method of use
WO2023183616A1 (en) 2022-03-25 2023-09-28 Senda Biosciences, Inc. Novel ionizable lipids and lipid nanoparticles and methods of using the same
WO2023196818A1 (en) 2022-04-04 2023-10-12 The Regents Of The University Of California Genetic complementation compositions and methods
WO2023196931A1 (en) 2022-04-07 2023-10-12 Renagade Therapeutics Management Inc. Cyclic lipids and lipid nanoparticles (lnp) for the delivery of nucleic acids or peptides for use in vaccinating against infectious agents
WO2023218431A1 (en) 2022-05-13 2023-11-16 BioNTech SE Rna compositions targeting hiv
WO2023230295A1 (en) 2022-05-25 2023-11-30 BioNTech SE Rna compositions for delivery of monkeypox antigens and related methods
WO2023232747A1 (en) 2022-05-30 2023-12-07 BioNTech SE Complexes for delivery of nucleic acids
WO2023239593A1 (en) * 2022-06-08 2023-12-14 Merck Sharp & Dohme Llc Method for sustained delivery of mrna vaccines
WO2024042236A1 (en) 2022-08-26 2024-02-29 Ethris Gmbh Stable lipid or lipidoid nanoparticle suspensions
EP4327829A1 (en) 2022-08-26 2024-02-28 Ethris GmbH Stabilization of lipid or lipidoid nanoparticle suspensions
WO2024049979A2 (en) 2022-08-31 2024-03-07 Senda Biosciences, Inc. Novel ionizable lipids and lipid nanoparticles and methods of using the same
WO2024056809A1 (en) 2022-09-15 2024-03-21 Novartis Ag Treatment of autoimmune disorders using chimeric antigen receptor therapy
WO2024064934A1 (en) 2022-09-23 2024-03-28 BioNTech SE Compositions for delivery of plasmodium csp antigens and related methods
WO2024063789A1 (en) 2022-09-23 2024-03-28 BioNTech SE Compositions for delivery of malaria antigens and related methods
WO2024064931A1 (en) 2022-09-23 2024-03-28 BioNTech SE Compositions for delivery of liver stage antigens and related methods
WO2024063788A1 (en) 2022-09-23 2024-03-28 BioNTech SE Compositions for delivery of malaria antigens and related methods
CN115671045B (en) * 2022-12-30 2023-04-07 华南理工大学 Non-liver-targeting nucleic acid nano preparation and preparation method and application thereof

Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6303378B1 (en) * 1997-10-24 2001-10-16 Valentis, Inc. Methods for preparing polynucleotide transfection complexes
US20020064517A1 (en) * 1998-04-30 2002-05-30 Stewart A. Cederholm-Williams Fibrin sealant as a transfection/transformation vehicle for gene therapy
US20020130430A1 (en) * 2000-12-29 2002-09-19 Castor Trevor Percival Methods for making polymer microspheres/nanospheres and encapsulating therapeutic proteins and other products
US20060018971A1 (en) * 2004-05-12 2006-01-26 Terrence Scott Nucleic acid microspheres, production and delivery thereof
US20080274463A1 (en) * 2007-05-04 2008-11-06 Ventana Medical Systems, Inc. Method for quantifying biomolecules conjugated to a nanoparticle
US20090264511A1 (en) * 2007-12-10 2009-10-22 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of factor vii gene
US20090286852A1 (en) 2005-08-23 2009-11-19 Katalin Kariko RNA containing modified nucleosides and methods of use thereof
US20100015232A1 (en) * 2006-07-07 2010-01-21 Aarhus Universitet Nanoparticles for nucleic acid delivery
US20110244026A1 (en) * 2009-12-01 2011-10-06 Braydon Charles Guild Delivery of mrna for the augmentation of proteins and enzymes in human genetic diseases
WO2011130624A2 (en) * 2010-04-16 2011-10-20 Immune Disease Institute, Inc. Sustained polypeptide expression from synthetic, modified rnas and uses thereof

Family Cites Families (1391)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US618862A (en) 1899-02-07 Automatic stoker
US533554A (en) 1895-02-05 Henry d
US618953A (en) 1899-02-07 Apparatus for preventing pounding or water-hammering in circulating-pipes of heating
US533537A (en) 1895-02-05 Watchmakers calipers
US618873A (en) 1899-02-07 Oyster-tongs
US618885A (en) 1899-02-07 Railway-car brake
US576705A (en) 1897-02-09 Vegetable cutter and grater
US618957A (en) 1899-02-07 Inda may kelly
US618896A (en) 1899-02-07 Potato-bug gatherer
US681712A (en) 1898-03-26 1901-09-03 American Railway Electric Light Co Means for controlling electric currents.
US648244A (en) 1899-04-28 1900-04-24 Orrin A Dever Tank-mold.
US696381A (en) 1900-03-01 1902-03-25 Hugo Loewenbach Paper-feeding machine.
US709303A (en) 1901-11-19 1902-09-16 Edward Chester & Company Ltd Construction of tanks.
US712490A (en) 1902-01-13 1902-11-04 Francis Blossom Economizer system.
US2008526A (en) 1932-11-03 1935-07-16 Wappler Frederick Charles Method and means for treating living tissue
US2588623A (en) 1948-05-10 1952-03-11 Eliscu Frank Surgical instrument for intradermal injection of fluids
US3467096A (en) 1966-04-12 1969-09-16 Ferrell S Horn Multiple hypodermic syringe arrangement
US3572336A (en) 1968-04-30 1971-03-23 Daniel R Hershberg Syringe
BE757653A (en) 1969-10-21 1971-04-16 Ugine Kuhlmann NEW DRUGS DERIVED FROM NUCLEIC ACIDS AND METHODS FOR THEIR PREPARATION
BE786542A (en) 1971-07-22 1973-01-22 Dow Corning SUCTION DEVICE ALLOWING TO OBTAIN CELL SAMPLES
US3906092A (en) 1971-11-26 1975-09-16 Merck & Co Inc Stimulation of antibody response
US4270537A (en) 1979-11-19 1981-06-02 Romaine Richard A Automatic hypodermic syringe
US4399216A (en) 1980-02-25 1983-08-16 The Trustees Of Columbia University Processes for inserting DNA into eucaryotic cells and for producing proteinaceous materials
US4500707A (en) 1980-02-29 1985-02-19 University Patents, Inc. Nucleosides useful in the preparation of polynucleotides
US5132418A (en) 1980-02-29 1992-07-21 University Patents, Inc. Process for preparing polynucleotides
US4458066A (en) 1980-02-29 1984-07-03 University Patents, Inc. Process for preparing polynucleotides
US4411657A (en) 1980-05-19 1983-10-25 Anibal Galindo Hypodermic needle
US4668777A (en) 1981-03-27 1987-05-26 University Patents, Inc. Phosphoramidite nucleoside compounds
US4973679A (en) 1981-03-27 1990-11-27 University Patents, Inc. Process for oligonucleo tide synthesis using phosphormidite intermediates
US4415732A (en) 1981-03-27 1983-11-15 University Patents, Inc. Phosphoramidite compounds and processes
US4373071A (en) 1981-04-30 1983-02-08 City Of Hope Research Institute Solid-phase synthesis of polynucleotides
US4401796A (en) 1981-04-30 1983-08-30 City Of Hope Research Institute Solid-phase synthesis of polynucleotides
US4474569A (en) 1982-06-28 1984-10-02 Denver Surgical Developments, Inc. Antenatal shunt
JPS5927900A (en) 1982-08-09 1984-02-14 Wakunaga Seiyaku Kk Oligonucleotide derivative and its preparation
US4588585A (en) 1982-10-19 1986-05-13 Cetus Corporation Human recombinant cysteine depleted interferon-β muteins
US4737462A (en) 1982-10-19 1988-04-12 Cetus Corporation Structural genes, plasmids and transformed cells for producing cysteine depleted muteins of interferon-β
FR2540122B1 (en) 1983-01-27 1985-11-29 Centre Nat Rech Scient NOVEL COMPOUNDS COMPRISING A SEQUENCE OF OLIGONUCLEOTIDE LINKED TO AN INTERCALATION AGENT, THEIR SYNTHESIS PROCESS AND THEIR APPLICATION
US4605735A (en) 1983-02-14 1986-08-12 Wakunaga Seiyaku Kabushiki Kaisha Oligonucleotide derivatives
US4948882A (en) 1983-02-22 1990-08-14 Syngene, Inc. Single-stranded labelled oligonucleotides, reactive monomers and methods of synthesis
US4824941A (en) 1983-03-10 1989-04-25 Julian Gordon Specific antibody to the native form of 2'5'-oligonucleotides, the method of preparation and the use as reagents in immunoassays or for binding 2'5'-oligonucleotides in biological systems
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US4587044A (en) 1983-09-01 1986-05-06 The Johns Hopkins University Linkage of proteins to nucleic acids
US5118800A (en) 1983-12-20 1992-06-02 California Institute Of Technology Oligonucleotides possessing a primary amino group in the terminal nucleotide
US5118802A (en) 1983-12-20 1992-06-02 California Institute Of Technology DNA-reporter conjugates linked via the 2' or 5'-primary amino group of the 5'-terminal nucleoside
US4579849A (en) 1984-04-06 1986-04-01 Merck & Co., Inc. N-alkylguanine acyclonucleosides as antiviral agents
US4957735A (en) 1984-06-12 1990-09-18 The University Of Tennessee Research Corporation Target-sensitive immunoliposomes- preparation and characterization
FR2567892B1 (en) 1984-07-19 1989-02-17 Centre Nat Rech Scient NOVEL OLIGONUCLEOTIDES, THEIR PREPARATION PROCESS AND THEIR APPLICATIONS AS MEDIATORS IN DEVELOPING THE EFFECTS OF INTERFERONS
US5430136A (en) 1984-10-16 1995-07-04 Chiron Corporation Oligonucleotides having selectably cleavable and/or abasic sites
US5258506A (en) 1984-10-16 1993-11-02 Chiron Corporation Photolabile reagents for incorporation into oligonucleotide chains
US4828979A (en) 1984-11-08 1989-05-09 Life Technologies, Inc. Nucleotide analogs for nucleic acid labeling and detection
US4959314A (en) 1984-11-09 1990-09-25 Cetus Corporation Cysteine-depleted muteins of biologically active proteins
US5036006A (en) 1984-11-13 1991-07-30 Cornell Research Foundation, Inc. Method for transporting substances into living cells and tissues and apparatus therefor
US5116943A (en) 1985-01-18 1992-05-26 Cetus Corporation Oxidation-resistant muteins of Il-2 and other protein
CA1288073C (en) 1985-03-07 1991-08-27 Paul G. Ahlquist Rna transformation vector
US5034506A (en) 1985-03-15 1991-07-23 Anti-Gene Development Group Uncharged morpholino-based polymers having achiral intersubunit linkages
US4596556A (en) 1985-03-25 1986-06-24 Bioject, Inc. Hypodermic injection apparatus
EP0204401A1 (en) 1985-04-09 1986-12-10 Biogen, Inc. Method of improving the yield of polypeptides produced in a host cell by stabilizing mRNA
US4762779A (en) 1985-06-13 1988-08-09 Amgen Inc. Compositions and methods for functionalizing nucleic acids
US5017691A (en) 1986-07-03 1991-05-21 Schering Corporation Mammalian interleukin-4
US5317098A (en) 1986-03-17 1994-05-31 Hiroaki Shizuya Non-radioisotope tagging of fragments
US5153319A (en) 1986-03-31 1992-10-06 University Patents, Inc. Process for preparing polynucleotides
US4695273A (en) 1986-04-08 1987-09-22 I-Flow Corporation Multiple needle holder and subcutaneous multiple channel infusion port
US4879111A (en) 1986-04-17 1989-11-07 Cetus Corporation Treatment of infections with lymphokines
JPS638396A (en) 1986-06-30 1988-01-14 Wakunaga Pharmaceut Co Ltd Poly-labeled oligonucleotide derivative
US4886499A (en) 1986-12-18 1989-12-12 Hoffmann-La Roche Inc. Portable injection appliance
GB8704027D0 (en) 1987-02-20 1987-03-25 Owen Mumford Ltd Syringe needle combination
US4904582A (en) 1987-06-11 1990-02-27 Synthetic Genetics Novel amphiphilic nucleic acid conjugates
US4790824A (en) 1987-06-19 1988-12-13 Bioject, Inc. Non-invasive hypodermic injection device
US4941880A (en) 1987-06-19 1990-07-17 Bioject, Inc. Pre-filled ampule and non-invasive hypodermic injection device assembly
US4940460A (en) 1987-06-19 1990-07-10 Bioject, Inc. Patient-fillable and non-invasive hypodermic injection device assembly
IE72468B1 (en) 1987-07-31 1997-04-09 Univ Leland Stanford Junior Selective amplification of target polynucleotide sequences
US6090591A (en) 1987-07-31 2000-07-18 The Board Of Trustees Of The Leland Stanford Junior University Selective amplification of target polynucleotide sequences
US5585481A (en) 1987-09-21 1996-12-17 Gen-Probe Incorporated Linking reagents for nucleotide probes
US5525465A (en) 1987-10-28 1996-06-11 Howard Florey Institute Of Experimental Physiology And Medicine Oligonucleotide-polyamide conjugates and methods of production and applications of the same
DE3738460A1 (en) 1987-11-12 1989-05-24 Max Planck Gesellschaft MODIFIED OLIGONUCLEOTIDS
DE68908054T2 (en) 1988-01-21 1994-03-10 Genentech Inc REINFORCEMENT AND DETECTION OF NUCLEIC ACID SEQUENCES.
CA1340807C (en) 1988-02-24 1999-11-02 Lawrence T. Malek Nucleic acid amplification process
JP2650159B2 (en) 1988-02-24 1997-09-03 アクゾ・ノベル・エヌ・ベー Nucleic acid amplification method
US5082830A (en) 1988-02-26 1992-01-21 Enzo Biochem, Inc. End labeled nucleotide probe
ATE123652T1 (en) 1988-03-04 1995-06-15 Cancer Res Campaign Tech ANTIGENS.
US5339163A (en) 1988-03-16 1994-08-16 Canon Kabushiki Kaisha Automatic exposure control device using plural image plane detection areas
AU631545B2 (en) 1988-04-15 1992-12-03 Protein Design Labs, Inc. Il-2 receptor-specific chimeric antibodies
US5109124A (en) 1988-06-01 1992-04-28 Biogen, Inc. Nucleic acid probe linked to a label having a terminal cysteine
US5021335A (en) 1988-06-17 1991-06-04 The Board Of Trustees Of The Leland Stanford Junior University In situ transcription in cells and tissues
US5168038A (en) 1988-06-17 1992-12-01 The Board Of Trustees Of The Leland Stanford Junior University In situ transcription in cells and tissues
US5130238A (en) 1988-06-24 1992-07-14 Cangene Corporation Enhanced nucleic acid amplification process
US5262536A (en) 1988-09-15 1993-11-16 E. I. Du Pont De Nemours And Company Reagents for the preparation of 5'-tagged oligonucleotides
US5759802A (en) 1988-10-26 1998-06-02 Tonen Corporation Production of human serum alubumin A
FR2638359A1 (en) 1988-11-03 1990-05-04 Tino Dalto SYRINGE GUIDE WITH ADJUSTMENT OF DEPTH DEPTH OF NEEDLE IN SKIN
US5512439A (en) 1988-11-21 1996-04-30 Dynal As Oligonucleotide-linked magnetic particles and uses thereof
US5262530A (en) 1988-12-21 1993-11-16 Applied Biosystems, Inc. Automated system for polynucleotide synthesis and purification
US5047524A (en) 1988-12-21 1991-09-10 Applied Biosystems, Inc. Automated system for polynucleotide synthesis and purification
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
US5457183A (en) 1989-03-06 1995-10-10 Board Of Regents, The University Of Texas System Hydroxylated texaphyrins
US5599923A (en) 1989-03-06 1997-02-04 Board Of Regents, University Of Tx Texaphyrin metal complexes having improved functionalization
CA2489769A1 (en) 1989-03-21 1990-10-04 Philip L. Felgner Expression of exogenous polynucleotide sequences in a vertebrate
US5703055A (en) 1989-03-21 1997-12-30 Wisconsin Alumni Research Foundation Generation of antibodies through lipid mediated DNA delivery
US6214804B1 (en) 1989-03-21 2001-04-10 Vical Incorporated Induction of a protective immune response in a mammal by injecting a DNA sequence
US5693622A (en) 1989-03-21 1997-12-02 Vical Incorporated Expression of exogenous polynucleotide sequences cardiac muscle of a mammal
US6867195B1 (en) 1989-03-21 2005-03-15 Vical Incorporated Lipid-mediated polynucleotide administration to reduce likelihood of subject's becoming infected
US6673776B1 (en) 1989-03-21 2004-01-06 Vical Incorporated Expression of exogenous polynucleotide sequences in a vertebrate, mammal, fish, bird or human
US5012818A (en) 1989-05-04 1991-05-07 Joishy Suresh K Two in one bone marrow surgical needle
IE66597B1 (en) 1989-05-10 1996-01-24 Akzo Nv Method for the synthesis of ribonucleic acid (RNA)
US5332671A (en) 1989-05-12 1994-07-26 Genetech, Inc. Production of vascular endothelial cell growth factor and DNA encoding same
US5240855A (en) 1989-05-12 1993-08-31 Pioneer Hi-Bred International, Inc. Particle gun
US5391723A (en) 1989-05-31 1995-02-21 Neorx Corporation Oligonucleotide conjugates
US4958013A (en) 1989-06-06 1990-09-18 Northwestern University Cholesteryl modified oligonucleotides
CA2020958C (en) 1989-07-11 2005-01-11 Daniel L. Kacian Nucleic acid sequence amplification methods
US5451463A (en) 1989-08-28 1995-09-19 Clontech Laboratories, Inc. Non-nucleoside 1,3-diol reagents for labeling synthetic oligonucleotides
US5254469A (en) 1989-09-12 1993-10-19 Eastman Kodak Company Oligonucleotide-enzyme conjugate that can be used as a probe in hybridization assays and polymerase chain reaction procedures
US5591722A (en) 1989-09-15 1997-01-07 Southern Research Institute 2'-deoxy-4'-thioribonucleosides and their antiviral activity
US5545522A (en) 1989-09-22 1996-08-13 Van Gelder; Russell N. Process for amplifying a target polynucleotide sequence using a single primer-promoter complex
ATE190981T1 (en) 1989-10-24 2000-04-15 Isis Pharmaceuticals Inc 2'-MODIFIED NUCLEOTIDES
US5208020A (en) 1989-10-25 1993-05-04 Immunogen Inc. Cytotoxic agents comprising maytansinoids and their therapeutic use
US5064413A (en) 1989-11-09 1991-11-12 Bioject, Inc. Needleless hypodermic injection device
US5215899A (en) 1989-11-09 1993-06-01 Miles Inc. Nucleic acid amplification employing ligatable hairpin probe and transcription
NO904633L (en) 1989-11-09 1991-05-10 Molecular Diagnostics Inc AMPLIFICATION OF NUCLEIC ACIDS BY TRANSCRIPABLE HAIRNEL PROBE.
US5312335A (en) 1989-11-09 1994-05-17 Bioject Inc. Needleless hypodermic injection device
US5292873A (en) 1989-11-29 1994-03-08 The Research Foundation Of State University Of New York Nucleic acids labeled with naphthoquinone probe
US5633076A (en) 1989-12-01 1997-05-27 Pharming Bv Method of producing a transgenic bovine or transgenic bovine embryo
US5697901A (en) 1989-12-14 1997-12-16 Elof Eriksson Gene delivery by microneedle injection
US5486603A (en) 1990-01-08 1996-01-23 Gilead Sciences, Inc. Oligonucleotide having enhanced binding affinity
US7037646B1 (en) 1990-01-11 2006-05-02 Isis Pharmaceuticals, Inc. Amine-derivatized nucleosides and oligonucleosides
US5670633A (en) 1990-01-11 1997-09-23 Isis Pharmaceuticals, Inc. Sugar modified oligonucleotides that detect and modulate gene expression
US5646265A (en) 1990-01-11 1997-07-08 Isis Pharmceuticals, Inc. Process for the preparation of 2'-O-alkyl purine phosphoramidites
US6783931B1 (en) 1990-01-11 2004-08-31 Isis Pharmaceuticals, Inc. Amine-derivatized nucleosides and oligonucleosides
US5578718A (en) 1990-01-11 1996-11-26 Isis Pharmaceuticals, Inc. Thiol-derivatized nucleosides
US5214136A (en) 1990-02-20 1993-05-25 Gilead Sciences, Inc. Anthraquinone-derivatives oligonucleotides
AU7579991A (en) 1990-02-20 1991-09-18 Gilead Sciences, Inc. Pseudonucleosides and pseudonucleotides and their polymers
US5264618A (en) * 1990-04-19 1993-11-23 Vical, Inc. Cationic lipids for intracellular delivery of biologically active molecules
GB9009980D0 (en) 1990-05-03 1990-06-27 Amersham Int Plc Phosphoramidite derivatives,their preparation and the use thereof in the incorporation of reporter groups on synthetic oligonucleotides
ATE167523T1 (en) 1990-05-11 1998-07-15 Microprobe Corp IMMERSIBLE TEST STRIPS FOR NUCLEIC ACID HYBRIDIZATION ASSAY AND METHOD FOR COVALENT IMMOBILIZATION OF OLIGONUCLEOTIDES
US5194370A (en) 1990-05-16 1993-03-16 Life Technologies, Inc. Promoter ligation activated transcription amplification of nucleic acid sequences
WO1992001813A1 (en) 1990-07-25 1992-02-06 Syngene, Inc. Circular extension for generating multiple nucleic acid complements
US5608046A (en) 1990-07-27 1997-03-04 Isis Pharmaceuticals, Inc. Conjugated 4'-desmethyl nucleoside analog compounds
US5218105A (en) 1990-07-27 1993-06-08 Isis Pharmaceuticals Polyamine conjugated oligonucleotides
US5138045A (en) 1990-07-27 1992-08-11 Isis Pharmaceuticals Polyamine conjugated oligonucleotides
US5602240A (en) 1990-07-27 1997-02-11 Ciba Geigy Ag. Backbone modified oligonucleotide analogs
US5688941A (en) 1990-07-27 1997-11-18 Isis Pharmaceuticals, Inc. Methods of making conjugated 4' desmethyl nucleoside analog compounds
US5489677A (en) 1990-07-27 1996-02-06 Isis Pharmaceuticals, Inc. Oligonucleoside linkages containing adjacent oxygen and nitrogen atoms
US5245022A (en) 1990-08-03 1993-09-14 Sterling Drug, Inc. Exonuclease resistant terminally substituted oligonucleotides
US5190521A (en) 1990-08-22 1993-03-02 Tecnol Medical Products, Inc. Apparatus and method for raising a skin wheal and anesthetizing skin
US5512667A (en) 1990-08-28 1996-04-30 Reed; Michael W. Trifunctional intermediates for preparing 3'-tailed oligonucleotides
US6140496A (en) 1990-10-09 2000-10-31 Benner; Steven Albert Precursors for deoxyribonucleotides containing non-standard nucleosides
KR930702373A (en) 1990-11-08 1993-09-08 안토니 제이. 페이네 Addition of Multiple Reporter Groups to Synthetic Oligonucleotides
US5527288A (en) 1990-12-13 1996-06-18 Elan Medical Technologies Limited Intradermal drug delivery device and method for intradermal delivery of drugs
US6100024A (en) 1991-02-08 2000-08-08 Promega Corporation Methods and compositions for nucleic acid detection by target extension and probe amplification
DE10199067I2 (en) 1991-03-18 2006-03-16 Univ New York Human Tumor Necrosis Factor Specific Monoclonal and Chimeric Antibodies
US5426180A (en) 1991-03-27 1995-06-20 Research Corporation Technologies, Inc. Methods of making single-stranded circular oligonucleotides
WO1992019759A1 (en) 1991-04-25 1992-11-12 Chugai Seiyaku Kabushiki Kaisha Reconstituted human antibody against human interleukin 6 receptor
US5714331A (en) 1991-05-24 1998-02-03 Buchardt, Deceased; Ole Peptide nucleic acids having enhanced binding affinity, sequence specificity and solubility
US5539082A (en) 1993-04-26 1996-07-23 Nielsen; Peter E. Peptide nucleic acids
US5719262A (en) 1993-11-22 1998-02-17 Buchardt, Deceased; Ole Peptide nucleic acids having amino acid side chains
US5169766A (en) 1991-06-14 1992-12-08 Life Technologies, Inc. Amplification of nucleic acid molecules
US5371241A (en) 1991-07-19 1994-12-06 Pharmacia P-L Biochemicals Inc. Fluorescein labelled phosphoramidites
US5199441A (en) 1991-08-20 1993-04-06 Hogle Hugh H Fine needle aspiration biopsy apparatus and method
GB9118204D0 (en) 1991-08-23 1991-10-09 Weston Terence E Needle-less injector
SE9102652D0 (en) 1991-09-13 1991-09-13 Kabi Pharmacia Ab INJECTION NEEDLE ARRANGEMENT
DE59208572D1 (en) 1991-10-17 1997-07-10 Ciba Geigy Ag Bicyclic nucleosides, oligonucleotides, processes for their preparation and intermediates
US5298422A (en) 1991-11-06 1994-03-29 Baylor College Of Medicine Myogenic vector systems
US5359044A (en) 1991-12-13 1994-10-25 Isis Pharmaceuticals Cyclobutyl oligonucleotide surrogates
US5824307A (en) 1991-12-23 1998-10-20 Medimmune, Inc. Human-murine chimeric antibodies against respiratory syncytial virus
US5565552A (en) 1992-01-21 1996-10-15 Pharmacyclics, Inc. Method of expanded porphyrin-oligonucleotide conjugate synthesis
US5595726A (en) 1992-01-21 1997-01-21 Pharmacyclics, Inc. Chromophore probe for detection of nucleic acid
CA2128616A1 (en) 1992-01-23 1993-08-05 Gary H. Rhodes Ex vivo gene transfer
FR2687679B1 (en) 1992-02-05 1994-10-28 Centre Nat Rech Scient OLIGOTHIONUCLEOTIDES.
US5328483A (en) 1992-02-27 1994-07-12 Jacoby Richard M Intradermal injection device with medication and needle guard
JP3368603B2 (en) 1992-02-28 2003-01-20 オリンパス光学工業株式会社 Gene therapy treatment device
CA2076465C (en) 1992-03-25 2002-11-26 Ravi V. J. Chari Cell binding agent conjugates of analogues and derivatives of cc-1065
US6174666B1 (en) 1992-03-27 2001-01-16 The United States Of America As Represented By The Department Of Health And Human Services Method of eliminating inhibitory/instability regions from mRNA
US6132419A (en) 1992-05-22 2000-10-17 Genetronics, Inc. Electroporetic gene and drug therapy
US5514545A (en) 1992-06-11 1996-05-07 Trustees Of The University Of Pennsylvania Method for characterizing single cells based on RNA amplification for diagnostics and therapeutics
EP0577558A2 (en) 1992-07-01 1994-01-05 Ciba-Geigy Ag Carbocyclic nucleosides having bicyclic rings, oligonucleotides therefrom, process for their preparation, their use and intermediates
US5272250A (en) 1992-07-10 1993-12-21 Spielvogel Bernard F Boronated phosphoramidate compounds
US6670178B1 (en) 1992-07-10 2003-12-30 Transkaryotic Therapies, Inc. In Vivo production and delivery of insulinotropin for gene therapy
US5383851A (en) 1992-07-24 1995-01-24 Bioject Inc. Needleless hypodermic injection device
DE69310179T2 (en) 1992-07-31 1997-07-31 Behringwerke Ag METHOD FOR INTRODUCING DEFINED SEQUENCES AT THE 3 'END OF POLYNUCLEOTIDES
US5273525A (en) 1992-08-13 1993-12-28 Btx Inc. Injection and electroporation apparatus for drug and gene delivery
US5240885A (en) 1992-09-21 1993-08-31 Corning Incorporated Rare earth-doped, stabilized cadmium halide glasses
US5569189A (en) 1992-09-28 1996-10-29 Equidyne Systems, Inc. hypodermic jet injector
US5334144A (en) 1992-10-30 1994-08-02 Becton, Dickinson And Company Single use disposable needleless injector
AU5665694A (en) 1992-11-04 1994-05-24 Denver Biomaterials Inc. Apparatus for removal of pleural effusion fluid
PL174721B1 (en) 1992-11-13 1998-09-30 Idec Pharma Corp Monoclonal antibody anty-cd2
US5736137A (en) 1992-11-13 1998-04-07 Idec Pharmaceuticals Corporation Therapeutic application of chimeric and radiolabeled antibodies to human B lymphocyte restricted differentiation antigen for treatment of B cell lymphoma
US5574142A (en) 1992-12-15 1996-11-12 Microprobe Corporation Peptide linkers for improved oligonucleotide delivery
CA2153692C (en) 1993-01-12 2011-11-08 Roy R. Lobb Recombinant anti-vla4 antibody molecules
FR2703253B1 (en) 1993-03-30 1995-06-23 Centre Nat Rech Scient APPLICATOR OF ELECTRIC PULSES FOR TREATING BIOLOGICAL TISSUES.
CA2159631A1 (en) 1993-03-30 1994-10-13 Sanofi Acyclic nucleoside analogs and oligonucleotide sequences containing them
DE4311944A1 (en) 1993-04-10 1994-10-13 Degussa Coated sodium percarbonate particles, process for their preparation and detergent, cleaning and bleaching compositions containing them
US7135312B2 (en) 1993-04-15 2006-11-14 University Of Rochester Circular DNA vectors for synthesis of RNA and DNA
US5773244A (en) 1993-05-19 1998-06-30 Regents Of The University Of California Methods of making circular RNA
US5851829A (en) 1993-07-16 1998-12-22 Dana-Farber Cancer Institute Method of intracellular binding of target molecules
US6294664B1 (en) 1993-07-29 2001-09-25 Isis Pharmaceuticals, Inc. Synthesis of oligonucleotides
US5672491A (en) 1993-09-20 1997-09-30 The Leland Stanford Junior University Recombinant production of novel polyketides
US6432711B1 (en) 1993-11-03 2002-08-13 Diacrin, Inc. Embryonic stem cells capable of differentiating into desired cell lines
US6096503A (en) 1993-11-12 2000-08-01 The Scripps Research Institute Method for simultaneous identification of differentially expresses mRNAs and measurement of relative concentrations
US5446137B1 (en) 1993-12-09 1998-10-06 Behringwerke Ag Oligonucleotides containing 4'-substituted nucleotides
US5519134A (en) 1994-01-11 1996-05-21 Isis Pharmaceuticals, Inc. Pyrrolidine-containing monomers and oligomers
US7435802B2 (en) 1994-01-25 2008-10-14 Elan Pharaceuticals, Inc. Humanized anti-VLA4 immunoglobulins
US5840299A (en) 1994-01-25 1998-11-24 Athena Neurosciences, Inc. Humanized antibodies against leukocyte adhesion molecule VLA-4
DK0668350T4 (en) 1994-02-16 2009-02-23 Us Gov Health & Human Serv Melanoma-associated antigen, epitopes thereof and melanoma vaccines
WO1995022316A1 (en) * 1994-02-17 1995-08-24 New York Blood Center, Inc. Biologic bioadhesive compositions containing fibrin glue and liposomes, methods of preparation and use
IL112820A0 (en) 1994-03-07 1995-05-26 Merck & Co Inc Coordinate in vivo gene expression
WO1995024176A1 (en) 1994-03-07 1995-09-14 Bioject, Inc. Ampule filling device
US5466220A (en) 1994-03-08 1995-11-14 Bioject, Inc. Drug vial mixing and transfer device
JP3482209B2 (en) 1994-03-18 2003-12-22 ジェロン・コーポレーション Oligonucleotides N3 '→ P5' phosphoramidate: synthesis and compounds; hybridization and nuclease resistance properties
US5457041A (en) 1994-03-25 1995-10-10 Science Applications International Corporation Needle array and method of introducing biological substances into living cells using the needle array
WO1995026204A1 (en) 1994-03-25 1995-10-05 Isis Pharmaceuticals, Inc. Immune stimulation by phosphorothioate oligonucleotide analogs
US5627053A (en) 1994-03-29 1997-05-06 Ribozyme Pharmaceuticals, Inc. 2'deoxy-2'-alkylnucleotide containing nucleic acid
US5464395A (en) 1994-04-05 1995-11-07 Faxon; David P. Catheter for delivering therapeutic and/or diagnostic agents to the tissue surrounding a bodily passageway
US6074642A (en) 1994-05-02 2000-06-13 Alexion Pharmaceuticals, Inc. Use of antibodies specific to human complement component C5 for the treatment of glomerulonephritis
JP3555086B2 (en) 1994-05-18 2004-08-18 プランテック バイオテクノロジスク ゲーエムベーハー フォーシュング アンド エンテゥウィックラング DNA sequence encoding an enzyme capable of promoting the synthesis of linear α-1,4 glucan in plants, fungi and microorganisms
EP0802980A1 (en) 1994-06-02 1997-10-29 Chiron Corporation Nucleic acid immunization using a virus-based infection/transfection system
GB9412230D0 (en) 1994-06-17 1994-08-10 Celltech Ltd Interleukin-5 specific recombiant antibodies
US6239116B1 (en) 1994-07-15 2001-05-29 University Of Iowa Research Foundation Immunostimulatory nucleic acid molecules
US5597696A (en) 1994-07-18 1997-01-28 Becton Dickinson And Company Covalent cyanine dye oligonucleotide conjugates
ES2251723T3 (en) 1994-08-12 2006-05-01 Immunomedics, Inc. SPECIFIC HUMANIZED ANTIBODIES AND ANTIBODIES FOR B-cell LYMPHOMA AND LEUKEMIA CELLS.
US5580731A (en) 1994-08-25 1996-12-03 Chiron Corporation N-4 modified pyrimidine deoxynucleotides and oligonucleotide probes synthesized therewith
US5597909A (en) 1994-08-25 1997-01-28 Chiron Corporation Polynucleotide reagents containing modified deoxyribose moieties, and associated methods of synthesis and use
US5665545A (en) 1994-11-28 1997-09-09 Akzo Nobel N.V. Terminal repeat amplification method
US5641665A (en) 1994-11-28 1997-06-24 Vical Incorporated Plasmids suitable for IL-2 expression
US5588960A (en) 1994-12-01 1996-12-31 Vidamed, Inc. Transurethral needle delivery device with cystoscope and method for treatment of urinary incontinence
US5807718A (en) 1994-12-02 1998-09-15 The Scripps Research Institute Enzymatic DNA molecules
US5585108A (en) 1994-12-30 1996-12-17 Nanosystems L.L.C. Formulations of oral gastrointestinal therapeutic agents in combination with pharmaceutically acceptable clays
ATE373094T1 (en) 1995-01-06 2007-09-15 Plant Res Int Bv DNA SEQUENCES CODING FOR CARBOHYDRATE POLYMERS-FORMING ENZYMES AND METHOD FOR PRODUCING TRANSGENIC PLANTS
US5599302A (en) 1995-01-09 1997-02-04 Medi-Ject Corporation Medical injection system and method, gas spring thereof and launching device using gas spring
US5795587A (en) 1995-01-23 1998-08-18 University Of Pittsburgh Stable lipid-comprising drug delivery complexes and methods for their production
US5824497A (en) 1995-02-10 1998-10-20 Mcmaster University High efficiency translation of mRNA molecules
DE69629326D1 (en) 1995-02-15 2003-09-11 Joseph Eldor Spinal needle with several holes
DE69621507T2 (en) 1995-03-28 2003-01-09 Japan Science & Tech Corp Method for molecular indexing of genes using restriction enzymes
US5869230A (en) 1995-03-30 1999-02-09 Beth Israel Hospital Association Gene transfer into the kidney
US5986054A (en) 1995-04-28 1999-11-16 The Hospital For Sick Children, Hsc Research And Development Limited Partnership Genetic sequences and proteins related to alzheimer's disease
FR2733762B1 (en) 1995-05-02 1997-08-01 Genset Sa METHOD FOR THE SPECIFIC COUPLING OF THE HAIR OF THE 5 'END OF A RNAM FRAGMENT AND PREPARATION OF RNAM AND COMPLETE DNA
US5700642A (en) 1995-05-22 1997-12-23 Sri International Oligonucleotide sizing using immobilized cleavable primers
US5730723A (en) 1995-10-10 1998-03-24 Visionary Medical Products Corporation, Inc. Gas pressured needle-less injection device and method
US6111095A (en) 1995-06-07 2000-08-29 Merck & Co., Inc. Capped synthetic RNA, analogs, and aptamers
US6051429A (en) 1995-06-07 2000-04-18 Life Technologies, Inc. Peptide-enhanced cationic lipid transfections
US5889136A (en) 1995-06-09 1999-03-30 The Regents Of The University Of Colorado Orthoester protecting groups in RNA synthesis
US5713863A (en) 1996-01-11 1998-02-03 Interventional Technologies Inc. Catheter with fluid medication injectors
US5766903A (en) 1995-08-23 1998-06-16 University Technology Corporation Circular RNA and uses thereof
US6265389B1 (en) 1995-08-31 2001-07-24 Alkermes Controlled Therapeutics, Inc. Microencapsulation and sustained release of oligonucleotides
AU7073096A (en) 1995-09-19 1997-04-09 University Of Massachusetts Inhibited biological degradation of oligodeoxynucleotides
US5830879A (en) 1995-10-02 1998-11-03 St. Elizabeth's Medical Center Of Boston, Inc. Treatment of vascular injury using vascular endothelial growth factor
US6265387B1 (en) 1995-10-11 2001-07-24 Mirus, Inc. Process of delivering naked DNA into a hepatocyte via bile duct
US6132988A (en) 1995-10-27 2000-10-17 Takeda Chemical Industries, Ltd. DNA encoding a neuronal cell-specific receptor protein
CU22584A1 (en) 1995-11-17 1999-11-03 Centro Inmunologia Molecular PHARMACEUTICAL COMPOSITIONS CONTAINING A MONOCLONAL ANTIBODY THAT RECOGNIZES THE CD6 HUMAN LEUKOCYTARY DIFFERENTIATION ANTIGEN AND ITS USES FOR THE DIAGNOSIS AND TREATMENT OF PSORIASIS
US6090382A (en) 1996-02-09 2000-07-18 Basf Aktiengesellschaft Human antibodies that bind human TNFα
US5962271A (en) 1996-01-03 1999-10-05 Cloutech Laboratories, Inc. Methods and compositions for generating full-length cDNA having arbitrary nucleotide sequence at the 3'-end
US5893397A (en) 1996-01-12 1999-04-13 Bioject Inc. Medication vial/syringe liquid-transfer apparatus
US6261584B1 (en) 1996-02-02 2001-07-17 Alza Corporation Sustained delivery of an active agent using an implantable system
US6395292B2 (en) 1996-02-02 2002-05-28 Alza Corporation Sustained delivery of an active agent using an implantable system
AU1874397A (en) 1996-02-16 1997-09-02 Stichting Rega Vzw Hexitol containing oligonucleotides and their use in antisense strategies
US6534312B1 (en) 1996-02-22 2003-03-18 Merck & Co., Inc. Vaccines comprising synthetic genes
US6090391A (en) 1996-02-23 2000-07-18 Aviron Recombinant tryptophan mutants of influenza
SE9601245D0 (en) 1996-03-29 1996-03-29 Pharmacia Ab Chimeric superantigens and their use
US6300487B1 (en) 1996-03-19 2001-10-09 Cell Therapuetics, Inc. Mammalian lysophosphatidic acid acyltransferase
TW517061B (en) 1996-03-29 2003-01-11 Pharmacia & Amp Upjohn Ab Modified/chimeric superantigens and their use
GB9607549D0 (en) 1996-04-11 1996-06-12 Weston Medical Ltd Spring-powered dispensing device
US20030073908A1 (en) 1996-04-26 2003-04-17 2000 Injectx, Inc. Method and apparatus for delivery of genes, enzymes and biological agents to tissue cells
US5712127A (en) 1996-04-29 1998-01-27 Genescape Inc. Subtractive amplification
US5853719A (en) 1996-04-30 1998-12-29 Duke University Methods for treating cancers and pathogen infections using antigen-presenting cells loaded with RNA
US7329741B2 (en) 1996-06-05 2008-02-12 Chiron Corporation Polynucleotides that hybridize to DP-75 and their use
US6300484B1 (en) 1996-06-05 2001-10-09 Chiron Corporation DNA encoding DP. 75 and a process for its use
EP0912607A2 (en) 1996-06-21 1999-05-06 Merck & Co., Inc. Vaccines comprising synthetic genes
JP2002515786A (en) 1996-06-28 2002-05-28 ソントラ メディカル,エル.ピー. Ultrasound enhancement of transdermal delivery
US5939262A (en) 1996-07-03 1999-08-17 Ambion, Inc. Ribonuclease resistant RNA preparation and utilization
US5677124A (en) 1996-07-03 1997-10-14 Ambion, Inc. Ribonuclease resistant viral RNA standards
US7288266B2 (en) 1996-08-19 2007-10-30 United States Of America As Represented By The Secretary, Department Of Health And Human Services Liposome complexes for increased systemic delivery
US5849546A (en) 1996-09-13 1998-12-15 Epicentre Technologies Corporation Methods for using mutant RNA polymerases with reduced discrimination between non-canonical and canonical nucleoside triphosphates
US6114148C1 (en) 1996-09-20 2012-05-01 Gen Hospital Corp High level expression of proteins
CA2266325C (en) 1996-09-24 2012-07-17 Lxr Biotechnology, Inc. A family of genes encoding apoptosis-related peptides, peptides encoded thereby and methods of use thereof
US6214966B1 (en) 1996-09-26 2001-04-10 Shearwater Corporation Soluble, degradable poly(ethylene glycol) derivatives for controllable release of bound molecules into solution
ES2241042T3 (en) 1996-10-11 2005-10-16 The Regents Of The University Of California IMMUNO STIMULATOR POLINUCLEOTIDE CONJUGATES / IMMUNOMODULATOR MOLECULA.
EP0839912A1 (en) 1996-10-30 1998-05-06 Instituut Voor Dierhouderij En Diergezondheid (Id-Dlo) Infectious clones of RNA viruses and vaccines and diagnostic assays derived thereof
GB9623051D0 (en) 1996-11-06 1997-01-08 Schacht Etienne H Delivery of DNA to target cells in biological systems
US5980887A (en) 1996-11-08 1999-11-09 St. Elizabeth's Medical Center Of Boston Methods for enhancing angiogenesis with endothelial progenitor cells
US5759179A (en) 1996-12-31 1998-06-02 Johnson & Johnson Medical, Inc. Needle and valve assembly for use with a catheter
WO1998031700A1 (en) 1997-01-21 1998-07-23 The General Hospital Corporation Selection of proteins using rna-protein fusions
EP0855184A1 (en) 1997-01-23 1998-07-29 Grayson B. Dr. Lipford Pharmaceutical composition comprising a polynucleotide and an antigen especially for vaccination
SK106699A3 (en) 1997-02-07 2000-06-12 Merck & Co Inc Synthetic polynucleotide, immunogenic composition and pharmaceutical composition containing the same and its use
US6251665B1 (en) 1997-02-07 2001-06-26 Cem Cezayirli Directed maturation of stem cells and production of programmable antigen presenting dentritic cells therefrom
US6228640B1 (en) 1997-02-07 2001-05-08 Cem Cezayirli Programmable antigen presenting cell of CD34 lineage
US6696291B2 (en) 1997-02-07 2004-02-24 Merck & Co., Inc. Synthetic HIV gag genes
US6576752B1 (en) 1997-02-14 2003-06-10 Isis Pharmaceuticals, Inc. Aminooxy functionalized oligomers
JP3756313B2 (en) 1997-03-07 2006-03-15 武 今西 Novel bicyclonucleosides and oligonucleotide analogues
US6406705B1 (en) 1997-03-10 2002-06-18 University Of Iowa Research Foundation Use of nucleic acids containing unmethylated CpG dinucleotide as an adjuvant
US6306393B1 (en) 1997-03-24 2001-10-23 Immunomedics, Inc. Immunotherapy of B-cell malignancies using anti-CD22 antibodies
US6261281B1 (en) 1997-04-03 2001-07-17 Electrofect As Method for genetic immunization and introduction of molecules into skeletal muscle and immune cells
US5914269A (en) 1997-04-04 1999-06-22 Isis Pharmaceuticals, Inc. Oligonucleotide inhibition of epidermal growth factor receptor expression
WO1998047913A2 (en) 1997-04-18 1998-10-29 The University Of Medicine And Dentistry Of New Jersey Inhibition of hiv-1 replication by a tat rna-binding domain peptide analog
US5958688A (en) 1997-04-28 1999-09-28 The Trustees Of The University Of Pennsylvania Characterization of mRNA patterns in neurites and single cells for medical diagnosis and therapeutics
US6235883B1 (en) 1997-05-05 2001-05-22 Abgenix, Inc. Human monoclonal antibodies to epidermal growth factor receptor
US5989911A (en) 1997-05-09 1999-11-23 University Of Massachusetts Site-specific synthesis of pseudouridine in RNA
US6761726B1 (en) 1998-05-15 2004-07-13 Pyng Medical Corp. Method and apparatus for the intraosseous introduction of a device such as an infusion tube
US5993412A (en) 1997-05-19 1999-11-30 Bioject, Inc. Injection apparatus
US6124091A (en) 1997-05-30 2000-09-26 Research Corporation Technologies, Inc. Cell growth-controlling oligonucleotides
US6589940B1 (en) 1997-06-06 2003-07-08 Dynavax Technologies Corporation Immunostimulatory oligonucleotides, compositions thereof and methods of use thereof
EP2085090A3 (en) 1997-06-06 2012-05-02 The Regents of the University of California Inhibitors of DNA immunostimulatory sequence activity
EP1012331B1 (en) 1997-07-01 2006-03-29 Isis Pharmaceuticals, Inc. Compositions and methods for the delivery of oligonucleotides via the alimentary canal
US5994511A (en) 1997-07-02 1999-11-30 Genentech, Inc. Anti-IgE antibodies and methods of improving polypeptides
DE69837706T2 (en) 1997-07-21 2008-01-10 Active Biotech Ab CYTOLYSIS OF APPROPRIATE TARGET CELLS WITH SUPERANTIC CONJUGATES USING THE INDUCTION OF T-CELL ACTIVATION
US20030073640A1 (en) 1997-07-23 2003-04-17 Ribozyme Pharmaceuticals, Inc. Novel compositions for the delivery of negatively charged molecules
WO1999006073A1 (en) 1997-07-31 1999-02-11 St. Elizabeth's Medical Center Of Boston, Inc. Method for the treatment of grafts
US6794499B2 (en) 1997-09-12 2004-09-21 Exiqon A/S Oligonucleotide analogues
US7151172B1 (en) 1997-09-18 2006-12-19 The Trustees Of The University Of Pennsylvania Attenuated vif DNA immunization cassettes for genetic vaccines
JP2002508299A (en) 1997-09-19 2002-03-19 セクイター, インク. Sense mRNA therapy
US6004573A (en) 1997-10-03 1999-12-21 Macromed, Inc. Biodegradable low molecular weight triblock poly(lactide-co-glycolide) polyethylene glycol copolymers having reverse thermal gelation properties
AU750106B2 (en) 1997-10-07 2002-07-11 University Of Maryland Biotechnology Institute Method for introducing and expressing RNA in animal cells
CA2306796A1 (en) 1997-10-20 1999-04-29 Genzyme Transgenics Corporation Novel modified msp-1 nucleic acid sequences and methods for increasing mrna levels and protein expression in cell systems
US6019747A (en) 1997-10-21 2000-02-01 I-Flow Corporation Spring-actuated infusion syringe
US6077251A (en) 1997-10-30 2000-06-20 Ting; Windsor Medicinal agent administration system
EP1987845B1 (en) 1997-11-20 2012-03-21 Vical Incorporated Treatment of cancer using cytokine-expressing polynucleotides and compositions therefor.
US7655777B2 (en) 1997-11-24 2010-02-02 Monsanto Technology Llc Nucleic acid molecules associated with the tocopherol pathway
US6517869B1 (en) 1997-12-12 2003-02-11 Expression Genetics, Inc. Positively charged poly(alpha-(omega-aminoalkyl)lycolic acid) for the delivery of a bioactive agent via tissue and cellular uptake
WO1999029758A1 (en) 1997-12-12 1999-06-17 Samyang Corporation Positively-charged poly[alpha-(omega-aminoalkyl)glycolic acid] for the delivery of a bioactive agent via tissue and cellular uptake
WO1999033982A2 (en) 1997-12-23 1999-07-08 Chiron Corporation Human genes and gene expression products i
US6320017B1 (en) 1997-12-23 2001-11-20 Inex Pharmaceuticals Corp. Polyamide oligomers
US6383811B2 (en) 1997-12-30 2002-05-07 Mirus Corporation Polyampholytes for delivering polyions to a cell
WO1999034831A1 (en) 1998-01-05 1999-07-15 University Of Washington Enhanced transport using membrane disruptive agents
IT1298087B1 (en) 1998-01-08 1999-12-20 Fiderm S R L DEVICE FOR CHECKING THE PENETRATION DEPTH OF A NEEDLE, IN PARTICULAR APPLICABLE TO A SYRINGE FOR INJECTIONS
EP1045714A1 (en) 1998-01-08 2000-10-25 Sontra Medical, L.P. Sonophoretic enhanced transdermal transport
US8287483B2 (en) 1998-01-08 2012-10-16 Echo Therapeutics, Inc. Method and apparatus for enhancement of transdermal transport
US6365346B1 (en) 1998-02-18 2002-04-02 Dade Behring Inc. Quantitative determination of nucleic acid amplification products
US5955310A (en) 1998-02-26 1999-09-21 Novo Nordisk Biotech, Inc. Methods for producing a polypeptide in a bacillus cell
US6432925B1 (en) 1998-04-16 2002-08-13 John Wayne Cancer Institute RNA cancer vaccine and methods for its use
US6429301B1 (en) 1998-04-17 2002-08-06 Whitehead Institute For Biomedical Research Use of a ribozyme to join nucleic acids and peptides
GB9808327D0 (en) 1998-04-20 1998-06-17 Chiron Spa Antidiotypic compounds
US6395253B2 (en) * 1998-04-23 2002-05-28 The Regents Of The University Of Michigan Microspheres containing condensed polyanionic bioactive agents and methods for their production
CN1177931C (en) 1998-04-23 2004-12-01 宝生物工程株式会社 Method for synthesizing DNA
US20090208418A1 (en) 2005-04-29 2009-08-20 Innexus Biotechnology Internaltional Ltd. Superantibody synthesis and use in detection, prevention and treatment of disease
DE69939740D1 (en) 1998-05-20 2008-11-27 Expression Genetics Inc LACTOSE OR GALACTOSE-POLYETHYLENE GLYCOL-PREPARED POLY-L-LYSINE AS A GM CARRIER
US6503231B1 (en) 1998-06-10 2003-01-07 Georgia Tech Research Corporation Microneedle device for transport of molecules across tissue
US7091192B1 (en) 1998-07-01 2006-08-15 California Institute Of Technology Linear cyclodextrin copolymers
CA2337652C (en) 1998-07-13 2013-03-26 Genetronics, Inc. Skin and muscle-targeted gene therapy by pulsed electrical field
AU5100899A (en) 1998-07-13 2000-02-01 Expression Genetics, Inc. Polyester analogue of poly-l-lysine as a soluble, biodegradable gene delivery carrier
US6222030B1 (en) 1998-08-03 2001-04-24 Agilent Technologies, Inc. Solid phase synthesis of oligonucleotides using carbonate protecting groups and alpha-effect nucleophile deprotection
EP1946775A3 (en) 1998-08-11 2008-08-06 Biogen Idec Inc. Combination therapies for B-cell lymphomas comprising administration of anti-CD20 antibody
GB9817662D0 (en) 1998-08-13 1998-10-07 Crocker Peter J Substance delivery
US20020065236A1 (en) 1998-09-09 2002-05-30 Yew Nelson S. CpG reduced plasmids and viral vectors
US6924365B1 (en) 1998-09-29 2005-08-02 Transkaryotic Therapies, Inc. Optimized messenger RNA
US20090042283A1 (en) 1998-09-29 2009-02-12 Shire Human Genetic Therapies, Inc., A Delaware Corporation Optimized messenger rna
WO2000026226A1 (en) 1998-11-03 2000-05-11 Yale University Multidomain polynucleotide molecular sensors
MY155913A (en) 1998-11-09 2015-12-15 Biogen Inc Chimeric anti-cd20 antibody treatment of patients receiving bmt or pbsc transpants
KR20010103655A (en) 1998-11-09 2001-11-23 케네쓰 제이. 울코트 Treatment of hematologic malignancies associated with circulating tumor cells using chimeric anti-cd20 antibody
WO2000027340A2 (en) 1998-11-12 2000-05-18 The Children's Medical Center Corporation USE OF t-RNA AND FRAGMENTS FOR INHIBITING ANGIOGENESIS AND COMPOSITIONS THEREOF
EP1829856A3 (en) 1998-11-12 2009-02-25 Invitrogen Corporation Transfection reagents
US6210931B1 (en) 1998-11-30 2001-04-03 The United States Of America As Represented By The Secretary Of Agriculture Ribozyme-mediated synthesis of circular RNA
US20040171980A1 (en) 1998-12-18 2004-09-02 Sontra Medical, Inc. Method and apparatus for enhancement of transdermal transport
EP1141376A4 (en) 1998-12-23 2002-03-13 Human Genome Sciences Peptidoglycan recognition proteins
GB9902000D0 (en) 1999-01-30 1999-03-17 Delta Biotechnology Ltd Process
CA2364921A1 (en) 1999-02-22 2000-08-31 European Molecular Biology Laboratory Translation system
US6255476B1 (en) 1999-02-22 2001-07-03 Pe Corporation (Ny) Methods and compositions for synthesis of labelled oligonucleotides and analogs on solid-supports
US7629311B2 (en) 1999-02-24 2009-12-08 Edward Lewis Tobinick Methods to facilitate transmission of large molecules across the blood-brain, blood-eye, and blood-nerve barriers
WO2000050006A2 (en) 1999-02-26 2000-08-31 Chiron Corporation Microemulsions with adsorbed macromoelecules and microparticles
JP2003535617A (en) 1999-03-01 2003-12-02 プロ ダクト ヘルス インコーポレーティッド Apparatus, method, and kit for delivering a substance to multiple ducts simultaneously
US7084125B2 (en) 1999-03-18 2006-08-01 Exiqon A/S Xylo-LNA analogues
EP1165798A2 (en) 1999-03-29 2002-01-02 Statens Serum Institut Nucleotide construct with optimised codons for an hiv genetic vaccine based on a primary, early hiv isolate and synthetic envelope
CN1311005C (en) 1999-04-09 2007-04-18 迪纳尔生物技术公司 Process for the preparation of monodisperse polymer particles
DK1178999T3 (en) 1999-05-04 2007-08-06 Santaris Pharma As L-ribo-LNA analogues
EP1642596A3 (en) 1999-05-07 2006-04-12 Genentech, Inc. Treatment of autoimmune diseases with antagonists which bind to B cell surface markers
KR20020011985A (en) 1999-05-07 2002-02-09 파르마솔 게엠베하 Lipid particles on the basis of mixtures of liquid and solid lipids and method for producing same
US7171264B1 (en) 1999-05-10 2007-01-30 Genetronics, Inc. Intradermal delivery of active agents by needle-free injection and electroporation
US6346382B1 (en) 1999-06-01 2002-02-12 Vanderbilt University Human carbamyl phosphate synthetase I polymorphism and diagnostic methods related thereto
WO2000075356A1 (en) 1999-06-04 2000-12-14 Lin Shi Lung Rna polymerase chain reaction
US6743211B1 (en) 1999-11-23 2004-06-01 Georgia Tech Research Corporation Devices and methods for enhanced microneedle penetration of biological barriers
US6611707B1 (en) 1999-06-04 2003-08-26 Georgia Tech Research Corporation Microneedle drug delivery device
US6303573B1 (en) 1999-06-07 2001-10-16 The Burnham Institute Heart homing peptides and methods of using same
NZ515957A (en) 1999-06-08 2003-08-29 Aventis Pasteur Immunostimulant oligonucleotide
DE60042785D1 (en) 1999-06-09 2009-10-01 Immunomedics Inc IMMUNOTHERAPY OF AUTOIMMUNE DISEASES THROUGH THE USE OF B-CELL SPECIFIC ANTIBODIES
US6949245B1 (en) 1999-06-25 2005-09-27 Genentech, Inc. Humanized anti-ErbB2 antibodies and treatment with anti-ErbB2 antibodies
CN1362965A (en) 1999-06-30 2002-08-07 先进细胞技术公司 cytoplasmic transfer to de-differentiate recipient cells
US6514948B1 (en) 1999-07-02 2003-02-04 The Regents Of The University Of California Method for enhancing an immune response
BR0012325A (en) 1999-07-09 2002-05-21 American Home Prod Methods and compositions for preventing the formation of abnormal RNA during the transcription of a plasmid sequence
US8557244B1 (en) 1999-08-11 2013-10-15 Biogen Idec Inc. Treatment of aggressive non-Hodgkins lymphoma with anti-CD20 antibody
ATE354655T1 (en) 1999-08-24 2007-03-15 Medarex Inc HUMAN ANTIBODIES TO CTLA-4 AND THEIR USES
US20050112141A1 (en) 2000-08-30 2005-05-26 Terman David S. Compositions and methods for treatment of neoplastic disease
US6551338B1 (en) 1999-09-01 2003-04-22 Mcgill University Method and device for myogenesis and angiogenesis of the heart
US20040106567A1 (en) 1999-09-07 2004-06-03 Hagstrom James E. Intravascular delivery of non-viral nucleic acid
ATE492644T1 (en) 1999-09-09 2011-01-15 Curevac Gmbh TRANSFER OF MRNA USING POLYCATIONIC COMPOUNDS
AU7398200A (en) 1999-09-17 2001-04-24 Aventis Pasteur Limited Chlamydia antigens and corresponding dna fragments and uses thereof
US6623457B1 (en) 1999-09-22 2003-09-23 Becton, Dickinson And Company Method and apparatus for the transdermal administration of a substance
WO2002064799A2 (en) 1999-09-28 2002-08-22 Transkaryotic Therapies, Inc. Optimized messenger rna
AU7863200A (en) 1999-10-06 2001-05-10 Quark Biotech, Inc. Method for enrichment of natural antisense messenger rna
US7060291B1 (en) 1999-11-24 2006-06-13 Transave, Inc. Modular targeted liposomal delivery system
US6613026B1 (en) 1999-12-08 2003-09-02 Scimed Life Systems, Inc. Lateral needle-less injection apparatus and method
US6277974B1 (en) 1999-12-14 2001-08-21 Cogent Neuroscience, Inc. Compositions and methods for diagnosing and treating conditions, disorders, or diseases involving cell death
US6245929B1 (en) 1999-12-20 2001-06-12 General Electric Company Catalyst composition and method for producing diaryl carbonates, using bisphosphines
EP1157045B1 (en) 1999-12-22 2004-10-20 Basell Poliolefine Italia S.p.A. Alpha-olefin polymerization catalyst system which contains an aromatic silane compound
SE515932C2 (en) 1999-12-23 2001-10-29 Prostalund Operations Ab Method and apparatus for the treatment of prostate
AU2764801A (en) 2000-01-07 2001-07-24 University Of Washington Enhanced transport of agents using membrane disruptive agents
AU2001232485A1 (en) 2000-01-13 2001-07-24 Amsterdam Support Diagnostics B.V. A universal nucleic acid amplification system for nucleic acids in a sample
AU2001231245A1 (en) 2000-01-31 2001-08-07 The Regents Of The University Of California Immunomodulatory polynucleotides in treatment of an infection by an intracellular pathogen
WO2001055306A2 (en) 2000-01-31 2001-08-02 Human Genome Sciences, Inc. Nucleic acids, proteins, and antibodies
US7833992B2 (en) 2001-05-18 2010-11-16 Merck Sharpe & Dohme Conjugates and compositions for cellular delivery
US7491805B2 (en) 2001-05-18 2009-02-17 Sirna Therapeutics, Inc. Conjugates and compositions for cellular delivery
WO2001062827A2 (en) 2000-02-22 2001-08-30 Shearwater Corporation N-maleimidyl polymer derivatives
CN1426423B (en) 2000-02-24 2010-05-12 华盛顿大学 Humanized antibodies that sequester amyloid beta peptide
BR0108962A (en) 2000-03-03 2002-12-24 Valentis Inc Nucleic acid formulations for gene distribution and methods of use
IL151865A0 (en) 2000-03-31 2003-04-10 Genentech Inc Compositions and methods for detecting and quantifying gene expression
JP2004500412A (en) 2000-03-31 2004-01-08 アイデック ファーマスーティカルズ コーポレイション Combination of anti-cytokine antibody or antagonist and anti-CD20 for treatment of B-cell lymphoma
US6565572B2 (en) 2000-04-10 2003-05-20 Sdgi Holdings, Inc. Fenestrated surgical screw and method
EP1832599A3 (en) 2000-04-12 2007-11-21 Human Genome Sciences, Inc. Albumin fusion proteins
US6368801B1 (en) 2000-04-12 2002-04-09 Molecular Staging, Inc. Detection and amplification of RNA using target-mediated ligation of DNA by RNA ligase
US20010046496A1 (en) 2000-04-14 2001-11-29 Brettman Lee R. Method of administering an antibody
US6468247B1 (en) 2000-04-21 2002-10-22 Mark Zamoyski Perfusion device for localized drug delivery
US6375972B1 (en) 2000-04-26 2002-04-23 Control Delivery Systems, Inc. Sustained release drug delivery devices, methods of use, and methods of manufacturing thereof
US7871598B1 (en) 2000-05-10 2011-01-18 Novartis Ag Stable metal ion-lipid powdered pharmaceutical compositions for drug delivery and methods of use
US20040229271A1 (en) 2000-05-19 2004-11-18 Williams Richard B. Compositions and methods for the identification and selection of nucleic acids and polypeptides
WO2001092523A2 (en) 2000-05-30 2001-12-06 Curagen Corporation Human polynucleotides and polypeptides encoded thereby
US6537242B1 (en) 2000-06-06 2003-03-25 Becton, Dickinson And Company Method and apparatus for enhancing penetration of a member for the intradermal sampling or administration of a substance
US20040052763A1 (en) 2000-06-07 2004-03-18 Mond James J. Immunostimulatory RNA/DNA hybrid molecules
US6607513B1 (en) 2000-06-08 2003-08-19 Becton, Dickinson And Company Device for withdrawing or administering a substance and method of manufacturing a device
ATE343589T1 (en) 2000-06-23 2006-11-15 Wyeth Corp MODOFIED MORBILLIVIRUS V PROTEINS
US20050181033A1 (en) 2000-06-29 2005-08-18 Dekker John P.Iii Method for delivering interferons to the intradermal compartment
AU2001276619A1 (en) 2000-07-03 2002-01-14 Chiron S.P.A. Immunisation against chlamydia pneumoniae
US6440096B1 (en) 2000-07-14 2002-08-27 Becton, Dickinson And Co. Microdevice and method of manufacturing a microdevice
DK1301614T3 (en) 2000-07-21 2007-04-02 Glaxo Group Ltd Codon-optimized papillomavirus sequences
US6902734B2 (en) 2000-08-07 2005-06-07 Centocor, Inc. Anti-IL-12 antibodies and compositions thereof
US20040142474A1 (en) 2000-09-14 2004-07-22 Expression Genetics, Inc. Novel cationic lipopolymer as a biocompatible gene delivery agent
US6696038B1 (en) 2000-09-14 2004-02-24 Expression Genetics, Inc. Cationic lipopolymer as biocompatible gene delivery agent
UA83458C2 (en) * 2000-09-18 2008-07-25 Байоджен Айдек Ма Інк. The isolated polypeptide baff-r (the receptor of the factor of activation of b-cells of the family tnf)
WO2002024873A1 (en) 2000-09-20 2002-03-28 Christopher Ralph Franks Stem cell therapy
WO2002028165A2 (en) 2000-10-04 2002-04-11 The Trustees Of The University Of Pennsylvania Compositions and methods of using capsid protein from flaviviruses and pestiviruses
ATE325806T1 (en) 2000-10-04 2006-06-15 Santaris Pharma As IMPROVED SYNTHESIS OF PURINE-BLOCKED NUCLEIC ACID ANALOGS
US6998115B2 (en) 2000-10-10 2006-02-14 Massachusetts Institute Of Technology Biodegradable poly(β-amino esters) and uses thereof
US6649138B2 (en) 2000-10-13 2003-11-18 Quantum Dot Corporation Surface-modified semiconductive and metallic nanoparticles having enhanced dispersibility in aqueous media
US7202226B2 (en) 2000-10-23 2007-04-10 Detroit R & D Augmentation of wound healing by elF-4E mRNA and EGF mRNA
US20030077604A1 (en) 2000-10-27 2003-04-24 Yongming Sun Compositions and methods relating to breast specific genes and proteins
ES2382636T3 (en) * 2000-10-31 2012-06-12 Surmodics Pharmaceuticals, Inc. Method for producing compositions for improved administration of bioactive molecules
US20020132788A1 (en) 2000-11-06 2002-09-19 David Lewis Inhibition of gene expression by delivery of small interfering RNA to post-embryonic animal cells in vivo
AU2002226930A1 (en) 2000-11-17 2002-05-27 The Government Of The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Reduction of the nonspecific animal toxicity of immunotoxins by mutating the framework regions of the fv to lower the isoelectric point
CA2430379A1 (en) 2000-12-07 2002-06-13 Chiron Corporation Endogenous retroviruses up-regulated in prostate cancer
US7708915B2 (en) 2004-05-06 2010-05-04 Castor Trevor P Polymer microspheres/nanospheres and encapsulating therapeutic proteins therein
US7628780B2 (en) 2001-01-13 2009-12-08 Medtronic, Inc. Devices and methods for interstitial injection of biologic agents into tissue
EP1351981B1 (en) 2001-01-19 2012-08-08 Vironovative B.V. A virus causing respiratory tract illness in susceptible mammals
EP1224943A1 (en) 2001-01-19 2002-07-24 Crucell Holland B.V. Fibronectin as a tumor marker detected by phage antibodies
US20040110191A1 (en) 2001-01-31 2004-06-10 Winkler Matthew M. Comparative analysis of nucleic acids using population tagging
US20030186237A1 (en) 2001-02-14 2003-10-02 Baylor College Of Medicine Methods and compositions of amplifying RNA
US6652886B2 (en) 2001-02-16 2003-11-25 Expression Genetics Biodegradable cationic copolymers of poly (alkylenimine) and poly (ethylene glycol) for the delivery of bioactive agents
DE10109897A1 (en) 2001-02-21 2002-11-07 Novosom Ag Optional cationic liposomes and their use
US7232425B2 (en) 2001-03-02 2007-06-19 Sorenson Development, Inc. Apparatus and method for specific interstitial or subcutaneous diffusion and dispersion of medication
DE02708018T1 (en) 2001-03-09 2004-09-30 Gene Stream Pty. Ltd. NEW EXPRESSION VECTORS
JP2002262882A (en) 2001-03-12 2002-09-17 Nisshinbo Ind Inc Method for amplifying rna
FR2822164B1 (en) 2001-03-19 2004-06-18 Centre Nat Rech Scient POLYPEPTIDES DERIVED FROM POLYMERASE RNAS, AND USES THEREOF
US6520949B2 (en) 2001-04-02 2003-02-18 Martin St. Germain Method and apparatus for administering fluid to animals subcutaneously
US6625486B2 (en) 2001-04-11 2003-09-23 Advanced Cardiovascular Systems, Inc. Method and apparatus for intracellular delivery of an agent
DE10119005A1 (en) 2001-04-18 2002-10-24 Roche Diagnostics Gmbh Process for protein expression starting from stabilized linear short DNA in cell-free in vitro transcription / translation systems with exonuclease-containing lysates or in a cellular system containing exonucleases
US20030171253A1 (en) 2001-04-19 2003-09-11 Averil Ma Methods and compositions relating to modulation of A20
ES2295351T3 (en) 2001-04-21 2008-04-16 Curevac Gmbh DEVICE FOR INJECTION OF RNAM.
US7332332B2 (en) 2001-04-23 2008-02-19 Amaxa Ag Buffer solution for electroporation and a method comprising the use of the same
US7560424B2 (en) 2001-04-30 2009-07-14 Zystor Therapeutics, Inc. Targeted therapeutic proteins
US6777187B2 (en) 2001-05-02 2004-08-17 Rubicon Genomics, Inc. Genome walking by selective amplification of nick-translate DNA library and amplification from complex mixtures of templates
EP1392587A4 (en) 2001-05-08 2009-03-18 Magnatech International L P Electronic length control wire pay-off system and method
US20050137155A1 (en) 2001-05-18 2005-06-23 Sirna Therapeutics, Inc. RNA interference mediated treatment of Parkinson disease using short interfering nucleic acid (siNA)
US8137911B2 (en) 2001-05-22 2012-03-20 Cellscript, Inc. Preparation and use of single-stranded transcription substrates for synthesis of transcription products corresponding to target sequences
AU2002314855B2 (en) 2001-05-30 2007-08-09 Board Of Trustees Of The Leland Stanford, Jr., University Delivery system for nucleic acids
EP1832603B1 (en) 2001-06-05 2010-02-03 CureVac GmbH Stabilised mRNA with increased G/C-content encoding a bacterial antigen and its use
EP1402035A2 (en) 2001-06-18 2004-03-31 Novartis AG G-protein coupled receptors and dna sequences thereof
EP1270732A1 (en) 2001-06-21 2003-01-02 Schuler, Gerold Improved transfection of eukaryontic cells with linear polynucleotides by electroporation
US7547551B2 (en) 2001-06-21 2009-06-16 University Of Antwerp. Transfection of eukaryontic cells with linear polynucleotides by electroporation
US7785610B2 (en) 2001-06-21 2010-08-31 Dynavax Technologies Corporation Chimeric immunomodulatory compounds and methods of using the same—III
WO2003002604A2 (en) 2001-06-26 2003-01-09 Novartis Ag G protein coupled receptors and dna sequences thereof
SE0102327D0 (en) 2001-06-28 2001-06-28 Active Biotech Ab A novel engineered superantigen for human therapy
BR0210628A (en) 2001-06-29 2004-08-10 Becton Dickinson Co Intradermal release of vaccines and genetic therapeutic agents via microcannula
AU2002359236A1 (en) 2001-07-13 2003-04-14 Advanced Research And Technology Institute Peptidoglycan recognition protein encoding nucleic acids and methods of use thereof
US6586524B2 (en) 2001-07-19 2003-07-01 Expression Genetics, Inc. Cellular targeting poly(ethylene glycol)-grafted polymeric gene carrier
US7169750B2 (en) 2001-07-31 2007-01-30 Anormed, Inc. Methods to mobilize progenitor/stem cells
AU2002312543A1 (en) 2001-08-01 2003-02-17 University Of Utah, Technology Transfer Office Isoform-selective inhibitors and activators of pde3 cyclic
US20050106659A1 (en) 2001-08-27 2005-05-19 Klemens Kaupmann Novel g-protein coupled receptor and dna sequences thereof
US20040142325A1 (en) 2001-09-14 2004-07-22 Liat Mintz Methods and systems for annotating biomolecular sequences
EP2390330B1 (en) 2001-09-28 2018-04-25 Max-Planck-Gesellschaft zur Förderung der Wissenschaften e.V. MicroRNA molecules
AR045702A1 (en) 2001-10-03 2005-11-09 Chiron Corp COMPOSITIONS OF ASSISTANTS.
EP1443905A4 (en) 2001-10-03 2010-06-23 Univ Johns Hopkins Compositions for oral gene therapy and methods of using same
DE10148886A1 (en) 2001-10-04 2003-04-30 Avontec Gmbh Inhibition of STAT-1
US7276489B2 (en) 2002-10-24 2007-10-02 Idera Pharmaceuticals, Inc. Modulation of immunostimulatory properties of oligonucleotide-based compounds by optimal presentation of 5′ ends
US7429258B2 (en) 2001-10-26 2008-09-30 Massachusetts Institute Of Technology Microneedle transport device
US7384739B2 (en) 2001-11-14 2008-06-10 Toyo Boseki Kabushiki Kaisha Compositions for enhancing DNA synthesis, DNA polymerase-related factors and utilization thereof
AU2002361642A1 (en) 2001-11-16 2003-06-10 The University Of Tennessee Research Corporation Recombinant antibody fusion proteins and methods for detection of apoptotic cells
WO2003046578A2 (en) 2001-11-29 2003-06-05 Novartis Ag Method for the assessment and prognosis of sarcoidosis
CA2409775C (en) 2001-12-03 2010-07-13 F. Hoffmann-La Roche Ag Reversibly modified thermostable enzymes for dna synthesis and amplification in vitro
US20060275747A1 (en) 2001-12-07 2006-12-07 Hardy Stephen F Endogenous retrovirus up-regulated in prostate cancer
DE60235413D1 (en) 2001-12-07 2010-04-01 Novartis Vaccines & Diagnostic IN PROSTATE CANCER HIGHLY REGULATED ENDOGENIC RETROVIRUS
CA2469049A1 (en) 2001-12-07 2003-06-19 Chiron Corporation Endogenous retrovirus polypeptides linked to oncogenic transformation
US20050107589A1 (en) 2001-12-17 2005-05-19 Gung-Wei Chirn Novel g-protein coupled receptors and dna sequences thereof
DE10162480A1 (en) 2001-12-19 2003-08-07 Ingmar Hoerr The application of mRNA for use as a therapeutic agent against tumor diseases
CA2467763C (en) 2001-12-21 2011-09-13 Alcon, Inc. Use of synthetic inorganic nanoparticles as carriers for ophthalmic and otic drugs
WO2003059381A2 (en) 2002-01-18 2003-07-24 Curevac Gmbh Immunogenic preparations and vaccines on the basis of mrna
CA2474709A1 (en) 2002-02-04 2003-08-14 Biomira, Inc. Immunostimulatory, covalently lipidated oligonucleotides
US6870034B2 (en) 2002-02-05 2005-03-22 Genentech, Inc. Protein purification
FR2835749B1 (en) 2002-02-08 2006-04-14 Inst Nat Sante Rech Med PHARMACEUTICAL COMPOSITION IMPROVING IN VIVO GENE TRANSFER
DE10207178A1 (en) 2002-02-19 2003-09-04 Novosom Ag Components for the production of amphoteric liposomes
US20050222064A1 (en) 2002-02-20 2005-10-06 Sirna Therapeutics, Inc. Polycationic compositions for cellular delivery of polynucleotides
AR038568A1 (en) 2002-02-20 2005-01-19 Hoffmann La Roche ANTI-A BETA ANTIBODIES AND ITS USE
US7354742B2 (en) 2002-02-22 2008-04-08 Ortho-Mcneil Pharmaceutical, Inc. Method for generating amplified RNA
AU2003267943C1 (en) 2002-02-26 2009-05-21 Altravax, Inc. Novel flavivirus antigens
SI1487856T1 (en) 2002-03-04 2010-12-31 Imclone Llc Human antibodies specific to kdr and uses thereof
AU2003221497A1 (en) 2002-03-13 2003-09-22 Novartis Ag Pharmaceutical microparticles
US7074596B2 (en) 2002-03-25 2006-07-11 Board Of Supervisors Of Louisiana State University And Agricultural And Mechanical College Synthesis and use of anti-reverse mRNA cap analogues
ES2543710T3 (en) 2002-04-04 2015-08-21 Zoetis Belgium S.A. Immunostimulatory oligonucleotides containing G and U
JP2005529590A (en) 2002-04-17 2005-10-06 ノバルティス アクチエンゲゼルシャフト Method for identifying inhibitor of binding of ARE-containing mRNA and HuR protein
GB0209539D0 (en) 2002-04-26 2002-06-05 Avecia Ltd Monomer Polymer and process
EP1361277A1 (en) 2002-04-30 2003-11-12 Centre National De La Recherche Scientifique (Cnrs) Optimization of transgene expression in mammalian cells
EP2371392B1 (en) 2002-05-02 2015-07-08 Wyeth Holdings LLC Calicheamicin derivative-carrier conjugates
AU2003217531A1 (en) 2002-05-02 2003-11-17 Massachusetts Eye And Ear Infirmary Ocular drug delivery systems and use thereof
EP1501567B1 (en) 2002-05-06 2018-02-21 Becton, Dickinson and Company Device for controlling drug pharmacokinetics
EP1503716A4 (en) 2002-05-08 2007-07-04 Univ California System and method for treating cardiac arrhythmias with fibroblast cells
US7374930B2 (en) 2002-05-21 2008-05-20 Expression Genetics, Inc. GLP-1 gene delivery for the treatment of type 2 diabetes
US20040018525A1 (en) 2002-05-21 2004-01-29 Bayer Aktiengesellschaft Methods and compositions for the prediction, diagnosis, prognosis, prevention and treatment of malignant neoplasma
DE10224200C1 (en) 2002-05-31 2003-08-21 Artus Ges Fuer Molekularbiolog Replicating RNA, useful, after reverse transcription, for analysis on microarrays, comprises conversion to cDNA then reverse transcription of this to form antisense sequences
AU2003237367A1 (en) 2002-06-03 2003-12-19 Chiron Corporation Use of nrg4, or inhibitors thereof, in the treatment of colon and pancreatic cancer
SE0201907D0 (en) 2002-06-19 2002-06-19 Atos Medical Ab Patches for tracheostoma valves
AU2003245160B2 (en) 2002-06-28 2009-09-24 Arbutus Biopharma Corporation Method and apparatus for producing liposomes
PL374580A1 (en) 2002-07-01 2005-10-31 The Kenneth S.Warren Institute, Inc. Recombinant tissue protective cytokines and encoding nucleic acids thereof for protection, restoration, and enhancement of responsive cells, tissues, and organs
DE10229872A1 (en) 2002-07-03 2004-01-29 Curevac Gmbh Immune stimulation through chemically modified RNA
GB0215509D0 (en) 2002-07-04 2002-08-14 Novartis Ag Marker genes
US20040009180A1 (en) 2002-07-11 2004-01-15 Allergan, Inc. Transdermal botulinum toxin compositions
TW200403338A (en) 2002-07-16 2004-03-01 Advisys Inc Codon optimized synthetic plasmids
US7927791B2 (en) 2002-07-24 2011-04-19 Ptc Therapeutics, Inc. Methods for identifying small molecules that modulate premature translation termination and nonsense mediated mRNA decay
EP1393745A1 (en) 2002-07-29 2004-03-03 Hybridon, Inc. Modulation of immunostimulatory properties of oligonucleotide-based compounds by optimal presentation of 5'ends
US6653468B1 (en) 2002-07-31 2003-11-25 Isis Pharmaceuticals, Inc. Universal support media for synthesis of oligomeric compounds
EP1386925A1 (en) 2002-07-31 2004-02-04 Girindus AG Method for preparing oligonucleotides
US7972358B2 (en) 2002-08-01 2011-07-05 Abbott Laboratories Vascular Enterprises Limited Apparatus for sealing a puncture by causing a reduction in the circumference of the puncture
EP1873180B1 (en) 2002-08-14 2014-05-07 Novartis AG Ophthalmic device made from a radiation-curable prepolymer
BR0313890A (en) 2002-08-30 2005-07-26 Becton Dickinson Co Pharmacokinetic control method of immunomodulatory compounds
BRPI0314042B8 (en) 2002-09-06 2021-05-25 Calando Pharmaceuticals Inc cyclodextrin-based polymers for the delivery of covalently linked therapeutic agents
JP4959133B2 (en) 2002-09-09 2012-06-20 ネクター セラピューティックス Method for preparing water-soluble polymer derivative having terminal carboxylic acid or ester thereof
WO2004024067A2 (en) 2002-09-10 2004-03-25 Vical Incorporated Codon-optimized polynucleotide-based vaccines against bacillus anthracis infection
US7534872B2 (en) 2002-09-27 2009-05-19 Syngen, Inc. Compositions and methods for the use of FMOC derivatives in DNA/RNA synthesis
EA021644B1 (en) 2002-10-17 2015-08-31 Генмаб А/С Human monoclonal antibody against cd20 and use thereof
AU2003301576A1 (en) 2002-10-22 2004-05-13 Eisai Co., Ltd. Gene expressed specifically in dopamine-producing neuron precursor cells after termination of division
US6896666B2 (en) 2002-11-08 2005-05-24 Kochamba Family Trust Cutaneous injection delivery under suction
AU2003297557B2 (en) 2002-11-21 2009-02-26 Cellscript, Inc. Methods for using primers that encode one strand of a double-stranded promoter
US7491234B2 (en) 2002-12-03 2009-02-17 Boston Scientific Scimed, Inc. Medical devices for delivery of therapeutic agents
AU2003297832A1 (en) 2002-12-09 2004-06-30 Medtronic Vascular Modular stent having polymer bridges at modular unit contact sites
DK1572744T3 (en) 2002-12-16 2010-09-20 Genentech Inc Immunoglobulin variants and their applications
EP1625140A4 (en) 2002-12-23 2008-06-18 Dynavax Tech Corp Branched immunomodulatory compounds and methods of using the same
US7169892B2 (en) 2003-01-10 2007-01-30 Astellas Pharma Inc. Lipid-peptide-polymer conjugates for long blood circulation and tumor specific drug delivery systems
US20080227085A1 (en) 2003-01-17 2008-09-18 Pellegrini Matthew C Methods and Systems for the Identification of Rna Regulatory Sequences and Compounds that Modulate their Function
US8426194B2 (en) 2003-01-21 2013-04-23 Ptc Therapeutics, Inc. Methods and agents for screening for compounds capable of modulating VEGF expression
US9068234B2 (en) 2003-01-21 2015-06-30 Ptc Therapeutics, Inc. Methods and agents for screening for compounds capable of modulating gene expression
US8460864B2 (en) 2003-01-21 2013-06-11 Ptc Therapeutics, Inc. Methods for identifying compounds that modulate untranslated region-dependent gene expression and methods of using same
US20040147027A1 (en) 2003-01-28 2004-07-29 Troy Carol M. Complex for facilitating delivery of dsRNA into a cell and uses thereof
ES2819011T3 (en) 2003-02-10 2021-04-14 Biogen Ma Inc Immunoglobulin formulation and procedure for its preparation
US20040167090A1 (en) 2003-02-21 2004-08-26 Monahan Sean D. Covalent modification of RNA for in vitro and in vivo delivery
CA2450289A1 (en) 2003-03-20 2005-05-19 Imclone Systems Incorporated Method of producing an antibody to epidermal growth factor receptor
US7320961B2 (en) 2003-03-24 2008-01-22 Abbott Laboratories Method for treating a disease, disorder or adverse effect caused by an elevated serum concentration of an UGT1A1 substrate
WO2004087868A2 (en) 2003-03-25 2004-10-14 Stratagene Dna polymerase fusions and uses thereof
US20040242502A1 (en) 2003-04-08 2004-12-02 Galenica Pharmaceuticals, Inc. Semi-synthetic saponin analogs with carrier and immune stimulatory activities for DNA and RNA vaccines
ZA200507805B (en) 2003-04-09 2006-12-27 Genentech Inc Therapy of autoimmune disease in a patient with an inadequate response to a TNF-alpha inhibitor
EP1620140B1 (en) 2003-05-05 2013-10-09 Ben-Gurion University Of The Negev Research And Development Authority Injectable cross-linked polymeric preparations and uses thereof
HUE026384T2 (en) 2003-05-06 2016-06-28 Biogen Hemophilia Inc Clotting factor chimeric proteins for treatment of a hemostatic disorder
US7348004B2 (en) 2003-05-06 2008-03-25 Syntonix Pharmaceuticals, Inc. Immunoglobulin chimeric monomer-dimer hybrids
TWI353991B (en) 2003-05-06 2011-12-11 Syntonix Pharmaceuticals Inc Immunoglobulin chimeric monomer-dimer hybrids
US20040226556A1 (en) 2003-05-13 2004-11-18 Deem Mark E. Apparatus for treating asthma using neurotoxin
US9567591B2 (en) 2003-05-15 2017-02-14 Mello Biotechnology, Inc. Generation of human embryonic stem-like cells using intronic RNA
GB0313132D0 (en) 2003-06-06 2003-07-09 Ich Productions Ltd Peptide ligands
EP1636385A4 (en) 2003-06-24 2010-06-02 Mirus Bio Corp Inhibition of gene function by delivery of polynucleotide-based gene expression inhibitors to mammalian cells in vivo
GB0316089D0 (en) 2003-07-09 2003-08-13 Xo Bioscience Ltd Differentiation method
US8592197B2 (en) 2003-07-11 2013-11-26 Novavax, Inc. Functional influenza virus-like particles (VLPs)
US7575572B2 (en) 2003-07-15 2009-08-18 Spinal Generations, Llc Method and device for delivering medicine to bone
US20050013870A1 (en) 2003-07-17 2005-01-20 Toby Freyman Decellularized extracellular matrix of conditioned body tissues and uses thereof
DK1648998T3 (en) 2003-07-18 2015-01-05 Amgen Inc Specific binding agents for hepatocyte growth factor
CA2533701A1 (en) 2003-07-31 2005-02-17 Isis Pharmaceuticals, Inc. Oligomeric compounds and compositions for use in modulation of small non-coding rnas
DE10335833A1 (en) 2003-08-05 2005-03-03 Curevac Gmbh Transfection of blood cells with mRNA for immune stimulation and gene therapy
US8668926B1 (en) 2003-09-15 2014-03-11 Shaker A. Mousa Nanoparticle and polymer formulations for thyroid hormone analogs, antagonists, and formulations thereof
US7135010B2 (en) 2003-09-30 2006-11-14 Damage Control Surgical Technologies, Inc. Method and apparatus for rapid deployment chest drainage
JP2007507460A (en) 2003-10-06 2007-03-29 ノバルティス アクチエンゲゼルシャフト Use of genetic polymorphisms associated with therapeutic efficacy in inflammatory diseases
DE10347710B4 (en) 2003-10-14 2006-03-30 Johannes-Gutenberg-Universität Mainz Recombinant vaccines and their use
US20050130201A1 (en) 2003-10-14 2005-06-16 Dharmacon, Inc. Splint-assisted enzymatic synthesis of polyribounucleotides
KR101364902B1 (en) 2003-11-05 2014-02-21 로슈 글리카트 아게 Cd20 antibodies with increased fc receptor binding affinity and effector function
WO2005047536A2 (en) 2003-11-13 2005-05-26 Novartis Ag Detection of genomic amplification and deletion in cancer
US20070054278A1 (en) 2003-11-18 2007-03-08 Applera Corporation Polymorphisms in nucleic acid molecules encoding human enzyme proteins, methods of detection and uses thereof
US7998119B2 (en) 2003-11-18 2011-08-16 Nano Pass Technologies Ltd. System and method for delivering fluid into flexible biological barrier
US7699852B2 (en) 2003-11-19 2010-04-20 Zimmer Spine, Inc. Fenestrated bone tap and method
US20050153333A1 (en) 2003-12-02 2005-07-14 Sooknanan Roy R. Selective terminal tagging of nucleic acids
EP1691746B1 (en) 2003-12-08 2015-05-27 Gel-Del Technologies, Inc. Mucoadhesive drug delivery devices and methods of making and using thereof
US7674884B2 (en) 2003-12-10 2010-03-09 Novimmune S.A. Neutralizing antibodies and methods of use thereof
US7927873B2 (en) 2003-12-19 2011-04-19 University Of Cincinnati Polyamides for nucleic acid delivery
PL3718564T3 (en) 2003-12-23 2024-03-18 Genentech, Inc. Novel anti-il 13 antibodies and uses thereof
US20050143336A1 (en) 2003-12-30 2005-06-30 Board Of Regents, The University Of Texas System Methods and compositions for improved non-viral gene therapy
US7150726B2 (en) 2004-01-23 2006-12-19 Norfolk Medical Device for subcutaneous infusion of fluids
US8957034B2 (en) 2004-01-28 2015-02-17 Johns Hopkins University Drugs and gene carrier particles that rapidly move through mucous barriers
DK1716233T3 (en) 2004-01-30 2009-10-26 Maxygen Holdings Ltd Regulated stop codon readout
US7309487B2 (en) 2004-02-09 2007-12-18 George Inana Methods and compositions for detecting and treating retinal diseases
EP3101034A1 (en) 2004-02-12 2016-12-07 Morphotek, Inc. Monoclonal antibodies that specifically bind to folate receptor alpha
US20070265220A1 (en) 2004-03-15 2007-11-15 City Of Hope Methods and compositions for the specific inhibition of gene expression by double-stranded RNA
EP1728801A4 (en) 2004-03-24 2009-10-21 Chugai Pharmaceutical Co Ltd Subtype of humanized antibody against interleukin-6 receptor
WO2005098433A2 (en) 2004-04-01 2005-10-20 Novartis Ag Diagnostic assays for alzheimer’s disease
JP4981660B2 (en) 2004-04-12 2012-07-25 アラーガン、インコーポレイテッド Multi-site injection system
CA2563533C (en) 2004-04-15 2013-10-01 Shmuel A. Ben-Sasson Compositions capable of facilitating penetration across a biological barrier
US7850962B2 (en) 2004-04-20 2010-12-14 Genmab A/S Human monoclonal antibodies against CD20
ES2246694B1 (en) 2004-04-29 2007-05-01 Instituto Cientifico Y Tecnologico De Navarra, S.A. PEGILATED NANOPARTICLES.
WO2005108411A2 (en) 2004-05-05 2005-11-17 Isis Pharmaceuticals, Inc. Substituted pixyl protecting groups for oligonucleotide synthesis
CN1980641A (en) * 2004-05-12 2007-06-13 巴克斯特国际公司 Oligonucleotide-containing microspheres, their use for the manufacture of a medicament for treating diabetes type 1
US20080103053A1 (en) 2005-11-22 2008-05-01 Helicos Biosciences Corporation Methods and compositions for sequencing a nucleic acid
US20060020329A1 (en) 2004-05-26 2006-01-26 Medtronic Vascular, Inc. Semi-directional drug delivering stents
US8012747B2 (en) 2004-06-01 2011-09-06 San Diego State University Foundation Expression system
ATE537263T1 (en) 2004-06-07 2011-12-15 Protiva Biotherapeutics Inc CATIONIC LIPIDS AND METHODS OF USE
EP1766035B1 (en) 2004-06-07 2011-12-07 Protiva Biotherapeutics Inc. Lipid encapsulated interfering rna
WO2005123114A2 (en) 2004-06-11 2005-12-29 Trustees Of Tufts College Silk-based drug delivery system
US8338648B2 (en) 2004-06-12 2012-12-25 Signum Biosciences, Inc. Topical compositions and methods for epithelial-related conditions
WO2006046978A2 (en) 2004-06-28 2006-05-04 Argos Therapeutics, Inc. Cationic peptide-mediated transformation
EP1765411B2 (en) 2004-06-30 2017-10-11 Nektar Therapeutics Polymer-factor ix moiety conjugates
EP1768742A4 (en) 2004-07-06 2007-10-17 Transpharma Medical Ltd Delivery system for transdermal immunization
CA2574088C (en) 2004-07-21 2013-09-17 Alnylam Pharmaceuticals, Inc. Oligonucleotides comprising a modified or non-natural nucleobase
DE102004035227A1 (en) 2004-07-21 2006-02-16 Curevac Gmbh mRNA mixture for vaccination against tumor diseases
US7603349B1 (en) 2004-07-29 2009-10-13 Yahoo! Inc. User interfaces for search systems using in-line contextual queries
GB0417487D0 (en) 2004-08-05 2004-09-08 Novartis Ag Organic compound
SE0402025D0 (en) 2004-08-13 2004-08-13 Active Biotech Ab Treatment of hyperproliferative disease with superantigens in combination with another anticancer agent
US7291208B2 (en) 2004-08-13 2007-11-06 Gore Enterprise Holdings, Inc. Grooved active and passive adsorbent filters
CA2478458A1 (en) 2004-08-20 2006-02-20 Michael Panzara Treatment of pediatric multiple sclerosis
US8691963B2 (en) 2004-08-26 2014-04-08 Engeneic Molecular Delivery Pty. Ltd. Delivering functional nucleic acids to mammalian cells via bacterially-derived, intact minicells
DE102004042546A1 (en) 2004-09-02 2006-03-09 Curevac Gmbh Combination therapy for immune stimulation
US7501486B2 (en) 2004-09-07 2009-03-10 Burnham Institute For Medical Research Peptides that selectively home to heart vasculature and related conjugates and methods
US8396548B2 (en) 2008-11-14 2013-03-12 Vessix Vascular, Inc. Selective drug delivery in a lumen
US8663599B1 (en) 2004-10-05 2014-03-04 Gp Medical, Inc. Pharmaceutical composition of nanoparticles
WO2006041088A1 (en) 2004-10-12 2006-04-20 Tissue Targeting Japan Inc. Brain disposition marrow progenitor
WO2006044503A2 (en) 2004-10-13 2006-04-27 Ptc Therapeutics, Inc. Compounds for nonsense suppression, use of these compounds for the manufacture of a medicament for treating somatic mutation-related diseases
US8057821B2 (en) 2004-11-03 2011-11-15 Egen, Inc. Biodegradable cross-linked cationic multi-block copolymers for gene delivery and methods of making thereof
EP1812569A2 (en) 2004-11-08 2007-08-01 K.U. Leuven Research and Development Modified nucleosides for rna interference
CA2587216C (en) 2004-11-18 2014-11-18 Nanopass Technologies Ltd. System and method for delivering fluid into flexible biological barrier
WO2006058088A2 (en) 2004-11-23 2006-06-01 Ptc Therapeutics, Inc. Carbazole, carboline and indole derivatives useful in the inhibition of vegf production
US7964571B2 (en) 2004-12-09 2011-06-21 Egen, Inc. Combination of immuno gene therapy and chemotherapy for treatment of cancer and hyperproliferative diseases
EP1856179B1 (en) 2004-12-10 2013-05-15 Kala Pharmaceuticals, Inc. Functionalized poly (ether-anhydride) block copolymers
WO2006071903A2 (en) 2004-12-28 2006-07-06 Ptc Therapeutics, Inc. Cell based methods and systems for the identification of rna regulatory sequences and compounds that modulate their functions
US8187570B1 (en) 2005-01-04 2012-05-29 Gp Medical, Inc. Nanoparticles for protein drug delivery
US8192718B1 (en) 2005-01-04 2012-06-05 Gp Medical, Inc. Pharmaceutical composition of nanoparticles
US8535702B2 (en) 2005-02-01 2013-09-17 Boston Scientific Scimed, Inc. Medical devices having porous polymeric regions for controlled drug delivery and regulated biocompatibility
WO2007086883A2 (en) 2005-02-14 2007-08-02 Sirna Therapeutics, Inc. Cationic lipids and formulated molecular compositions containing them
US7404969B2 (en) 2005-02-14 2008-07-29 Sirna Therapeutics, Inc. Lipid nanoparticle based compositions and methods for the delivery of biologically active molecules
US20060263338A1 (en) 2005-03-04 2006-11-23 Jacoby Douglas B Catheter-based delivery of Skeletal Myoblasts to the Myocardium of Damaged Hearts
WO2006095259A2 (en) 2005-03-11 2006-09-14 Novartis Ag Biomarkers for cardiovascular side-effects induced by cox-2 inhibitory compounds
JP4793806B2 (en) 2005-03-22 2011-10-12 Tti・エルビュー株式会社 Iontophoresis device
US8415325B2 (en) 2005-03-31 2013-04-09 University Of Delaware Cell-mediated delivery and targeted erosion of noncovalently crosslinked hydrogels
CA2603853C (en) 2005-04-01 2013-11-19 Intezyne Technologies, Incorporated Polymeric micelles for drug delivery
WO2006110585A2 (en) 2005-04-07 2006-10-19 Novartis Vaccines And Diagnostics Inc. Cancer-related genes (prlr)
EP1865981A2 (en) 2005-04-07 2007-12-19 Chiron Corporation Cacna1e in cancer diagnosis, detection and treatment
EP1885403B1 (en) 2005-04-12 2013-05-08 Nektar Therapeutics Poly(ethyleneglycol) conjugates of Lysostaphin
US20060241076A1 (en) 2005-04-26 2006-10-26 Coley Pharmaceutical Gmbh Modified oligoribonucleotide analogs with enhanced immunostimulatory activity
US7850656B2 (en) 2005-04-29 2010-12-14 Warsaw Orthopedic, Inc. Devices and methods for delivering medical agents
CA2607147C (en) 2005-05-09 2018-07-17 Ono Pharmaceutical Co., Ltd. Human monoclonal antibodies to programmed death 1 (pd-1) and methods for treating cancer using anti-pd-1 antibodies alone or in combination with other immunotherapeutics
US8246995B2 (en) 2005-05-10 2012-08-21 The Board Of Trustees Of The Leland Stanford Junior University Hydrophobic nanotubes and nanoparticles as transporters for the delivery of drugs into cells
US20070072175A1 (en) 2005-05-13 2007-03-29 Biogen Idec Ma Inc. Nucleotide array containing polynucleotide probes complementary to, or fragments of, cynomolgus monkey genes and the use thereof
US20060265771A1 (en) 2005-05-17 2006-11-23 Lewis David L Monitoring microrna expression and function
DE102005023170A1 (en) 2005-05-19 2006-11-23 Curevac Gmbh Optimized formulation for mRNA
JP2008541770A (en) 2005-06-03 2008-11-27 ジェネンテック・インコーポレーテッド Methods for producing antibodies having altered fucosylation levels
US7550264B2 (en) 2005-06-10 2009-06-23 Datascope Investment Corporation Methods and kits for sense RNA synthesis
US7691086B2 (en) 2005-06-14 2010-04-06 Tengiz Tkebuchava Catheter for introduction of medications to the tissues of a heart or other organ
RS53968B1 (en) 2005-06-16 2015-08-31 Nektar Therapeutics Conjugates having a degradable linkage and polymeric reagents useful in preparing such conjugates
US8202835B2 (en) 2005-06-17 2012-06-19 Yitzchak Hillman Disease treatment via antimicrobial peptides or their inhibitors
US20110182805A1 (en) 2005-06-17 2011-07-28 Desimone Joseph M Nanoparticle fabrication methods, systems, and materials
US8101385B2 (en) 2005-06-30 2012-01-24 Archemix Corp. Materials and methods for the generation of transcripts comprising modified nucleotides
KR20080025181A (en) 2005-06-30 2008-03-19 아케믹스 코포레이션 Materials and methods for generation of fully 2'-modified nucleic acid transcripts
US20080220471A1 (en) 2005-07-27 2008-09-11 Genentech, Inc. Vectors and Methods Using Same
US9012219B2 (en) 2005-08-23 2015-04-21 The Trustees Of The University Of Pennsylvania RNA preparations comprising purified modified RNA for reprogramming cells
US20070048741A1 (en) 2005-08-24 2007-03-01 Getts Robert C Methods and kits for sense RNA synthesis
CN101287488B (en) 2005-09-01 2013-01-30 诺华疫苗和诊断有限两合公司 Multiple vaccination including serogroup C meningococcus
DE602006020300D1 (en) 2005-09-01 2011-04-07 Celgene Corp IMMUNOLOGICAL USES OF IMMUNOMODULATIVE COMPOUNDS FOR A VACCINATE AND THERAPY AGAINST INFECTIOUS DISEASES
CA2621055A1 (en) * 2005-09-02 2007-03-15 Takeda Pharmaceutical Company Limited Sustained-release microsphere containing short chain deoxyribonucleic acid or short chain ribonucleic acid and method of producing the same
US8420605B2 (en) 2005-09-07 2013-04-16 The University Of Strathclyde Hydrogel compositions
US20120021042A1 (en) 2005-09-15 2012-01-26 Steffen Panzner Efficient Method For Loading Amphoteric Liposomes With Nucleic Acid Active Substances
US20070185432A1 (en) 2005-09-19 2007-08-09 Transport Pharmaceuticals, Inc. Electrokinetic system and method for delivering methotrexate
DE102005046490A1 (en) 2005-09-28 2007-03-29 Johannes-Gutenberg-Universität Mainz New nucleic acid molecule comprising promoter, a transcriptable nucleic acid sequence, a first and second nucleic acid sequence for producing modified RNA with transcriptional stability and translational efficiency
US20070087437A1 (en) 2005-10-14 2007-04-19 Jifan Hu Methods for rejuvenating cells in vitro and in vivo
US8012096B2 (en) 2005-10-17 2011-09-06 Cardiogenesis Corporation Surgical device and method for performing combination revascularization and therapeutic substance delivery to tissue
DK1951299T3 (en) 2005-11-04 2012-04-02 Novartis Vaccines & Diagnostic Influenza vaccines containing combinations of particulate adjuvants and immune enhancers
US20070105124A1 (en) 2005-11-08 2007-05-10 Getts Robert C Methods and kits for nucleic acid amplification
AU2006314757A1 (en) 2005-11-18 2007-05-24 Bioline Limited A method for enhancing enzymatic DNA polymerase reactions
BRPI0619056A2 (en) 2005-11-28 2011-09-20 Genmab As monovalent antibody, method for preparing and producing a monovalent antibody, nucleic acid construct, host cell, immunoconjugate, use of a monovalent antibody, and pharmaceutical composition
AU2005338632B2 (en) 2005-11-30 2010-05-20 Epicentre Technologies Corporation Selective terminal tagging of nucleic acids
TWI389709B (en) 2005-12-01 2013-03-21 Novartis Ag Transdermal therapeutic system
US8603457B2 (en) 2005-12-02 2013-12-10 University Of Rochester Nonsense suppression and genetic codon alteration by targeted modification
US9393215B2 (en) 2005-12-02 2016-07-19 Novartis Ag Nanoparticles for use in immunogenic compositions
US7579318B2 (en) 2005-12-06 2009-08-25 Centre De La Recherche De La Scientifique Cell penetrating peptides for intracellular delivery of molecules
WO2007069090A2 (en) 2005-12-06 2007-06-21 Centre National De La Recherche Scientifique Cell penetrating peptides for intracellular delivery of molecules
EP1960547A2 (en) 2005-12-08 2008-08-27 Novartis AG Effects of inhibitors of fgfr3 on gene transcription
US8048999B2 (en) 2005-12-13 2011-11-01 Kyoto University Nuclear reprogramming factor
WO2007069068A2 (en) 2005-12-16 2007-06-21 Diatos Cell penetrating peptide conjugates for delivering nucleic acids into cells
WO2007082305A2 (en) 2006-01-12 2007-07-19 Massachusetts Institute Of Technology Biodegradable elastomers
AU2007208452B2 (en) 2006-01-13 2012-07-05 The Trustees Of The University Of Pennsylvania Vaccines and immunotherapeutics using codon optimized IL-15 and methods for using the same
DE602007009487D1 (en) 2006-01-27 2010-11-11 Isis Pharmaceutical Inc 6-MODIFIED BICYCLIC NUCLEIC ACID ANALOGUE
US8669345B2 (en) 2006-01-27 2014-03-11 Biogen Idec Ma Inc. Nogo receptor antagonists
US20070178103A1 (en) 2006-01-30 2007-08-02 Fey Georg H CD19-specific immunotoxin and treatment method
US9458444B2 (en) 2006-02-03 2016-10-04 Opko Biologics Ltd. Long-acting coagulation factors and methods of producing same
US8476234B2 (en) 2006-02-03 2013-07-02 Prolor Biotech Inc. Long-acting coagulation factors and methods of producing same
US8946155B2 (en) 2006-02-03 2015-02-03 Opko Biologics Ltd. Long-acting polypeptides and methods of producing and administering same
DE102006007433A1 (en) 2006-02-17 2007-08-23 Curevac Gmbh Immunostimulant adjuvant useful in vaccines against cancer or infectious diseases comprises a lipid-modified nucleic acid
WO2007097561A1 (en) 2006-02-20 2007-08-30 Ewha University - Industry Collaboration Foundation Peptide having cell membrane penetrating activity
CA2642905C (en) 2006-02-21 2015-04-28 Nektar Therapeutics Al, Corporation Segmented degradable polymers and conjugates made therefrom
US20080038278A1 (en) 2006-02-24 2008-02-14 Jingsong Cao GPAT3 encodes a mammalian, microsomal acyl-coa:glycerol 3- phosphate acyltransferase
CA2643322C (en) 2006-02-24 2015-07-21 Novartis Ag Microparticles containing biodegradable polymer and cationic polysaccharide for use in immunogenic compositions
US7910152B2 (en) 2006-02-28 2011-03-22 Advanced Cardiovascular Systems, Inc. Poly(ester amide)-based drug delivery systems with controlled release rate and morphology
JP2009528359A (en) 2006-02-28 2009-08-06 エラン ファーマシューティカルズ,インコーポレイテッド Methods of treating inflammatory and autoimmune diseases with natalizumab
GB0605217D0 (en) 2006-03-15 2006-04-26 Novartis Ag Method and compositions for assessing acute rejection
WO2007109244A2 (en) 2006-03-21 2007-09-27 Morehouse School Of Medicine Novel nanoparticles for delivery of active agents
ES2776100T3 (en) 2006-03-31 2020-07-29 Massachusetts Inst Technology System for targeted delivery of therapeutic agents
US8257685B2 (en) 2006-04-04 2012-09-04 Stc.Unm Swellable particles for drug delivery
US20070281336A1 (en) 2006-04-14 2007-12-06 Epicentre Technologies Kits and methods for generating 5' capped RNA
EP1852127A1 (en) 2006-05-02 2007-11-07 Charité - Universitätsmedizin Berlin Use of a B-cell-depleting antibody for treatment of polyoma virus infections
US8367113B2 (en) 2006-05-15 2013-02-05 Massachusetts Institute Of Technology Polymers for functional particles
WO2007135172A2 (en) 2006-05-24 2007-11-29 Laboratoires Serono S.A. Cladribine regimen for treating multiple sclerosis
EP2030015B1 (en) 2006-06-02 2016-02-17 President and Fellows of Harvard College Protein surface remodeling
EP2046383B1 (en) 2006-07-04 2014-11-19 Genmab A/S Cd20 binding molecules for the treatment of copd
AU2007272558B2 (en) 2006-07-12 2010-12-09 Novartis Ag Actinically crosslinkable copolymers for manufacturing contact lenses
MX2009000622A (en) 2006-07-20 2009-01-29 Novartis Ag Amigo-2 inhibitors for treating, diagnosing or detecting cancer.
US8728527B2 (en) 2006-07-24 2014-05-20 Luminus Biosciences, Inc. Solid nanoparticle formulation of water insoluble pharmaceutical substances with reduced ostwald ripening
EP2049665A2 (en) 2006-07-28 2009-04-22 Applera Corporation Dinucleotide mrna cap analogs
DE102006035618A1 (en) 2006-07-31 2008-02-07 Curevac Gmbh New nucleic acid useful as immuno-stimulating adjuvant for manufacture of a composition for treatment of cancer diseases e.g. colon carcinomas and infectious diseases e.g. influenza and malaria
AU2007280690C1 (en) 2006-07-31 2012-08-23 Curevac Gmbh Nucleic acid of formula (I): GIXmGn, or (II): CIXmCn, in particular as an immune-stimulating agent/adjuvant
WO2008019371A1 (en) 2006-08-07 2008-02-14 Genzyme Corporation Combination therapy
US20080076701A1 (en) 2006-08-18 2008-03-27 Nastech Pharmaceutical Company Inc. Dicer substrate rna peptide conjugates and methods for rna therapeutics
US8658211B2 (en) 2006-08-18 2014-02-25 Arrowhead Madison Inc. Polyconjugates for in vivo delivery of polynucleotides
CA2661634C (en) 2006-09-06 2017-03-28 The Regents Of The University Of California Selectively targeted antimicrobial peptides and the use thereof
CA2663388C (en) 2006-09-07 2017-01-17 Crucell Holland B.V. Human binding molecules capable of neutralizing influenza virus h5n1 and uses thereof
US20080140061A1 (en) 2006-09-08 2008-06-12 Arbel Medical Ltd. Method And Device For Combined Treatment
JP2010502713A (en) 2006-09-08 2010-01-28 ザ・ジョンズ・ホプキンス・ユニバーシティー Compositions and methods for enhancing transport through mucus
US8454948B2 (en) 2006-09-14 2013-06-04 Medgenics Medical Israel Ltd. Long lasting drug formulations
GB0619182D0 (en) 2006-09-29 2006-11-08 Leuven K U Res & Dev Oligonucleotide arrays
CA2927045A1 (en) 2006-10-03 2008-04-10 Muthiah Manoharan Lipid containing formulations
MX2009003680A (en) 2006-10-05 2009-07-17 Univ Johns Hopkins Water-dispersible oral, parenteral, and topical formulations for poorly water soluble drugs using smart polymeric nanoparticles.
DE102006051516A1 (en) * 2006-10-31 2008-05-08 Curevac Gmbh (Base) modified RNA to increase the expression of a protein
US8414927B2 (en) 2006-11-03 2013-04-09 Boston Scientific Scimed, Inc. Cross-linked polymer particles
US7999087B2 (en) 2006-11-15 2011-08-16 Agilent Technologies, Inc. 2′-silyl containing thiocarbonate protecting groups for RNA synthesis
US8242258B2 (en) 2006-12-03 2012-08-14 Agilent Technologies, Inc. Protecting groups for RNA synthesis
US8399007B2 (en) 2006-12-05 2013-03-19 Landec Corporation Method for formulating a controlled-release pharmaceutical formulation
JP2011506264A (en) 2006-12-06 2011-03-03 ノバルティス アーゲー Vaccines containing antigens from four strains of influenza virus
US9034348B2 (en) 2006-12-11 2015-05-19 Chi2Gel Ltd. Injectable chitosan mixtures forming hydrogels
EP4026553A1 (en) 2006-12-18 2022-07-13 Acceleron Pharma, Inc. Activin-actrii antagonists and uses for increasing red blood cell levels
AU2007339280B2 (en) 2006-12-21 2013-12-05 Stryker Corporation Sustained-release formulations comprising crystals, macromolecular gels, and particulate suspensions of biologic agents
DK2104739T3 (en) 2006-12-21 2013-10-07 Novozymes Inc Modified messenger RNA stabilization sequences for expression of genes in bacterial cells
DE102006061015A1 (en) 2006-12-22 2008-06-26 Curevac Gmbh Process for the purification of RNA on a preparative scale by HPLC
CA2673029C (en) 2006-12-22 2017-03-28 Archemix Corp. Materials and methods for the generation of transcripts comprising modified nucleotides
US7699803B2 (en) 2007-01-03 2010-04-20 Medtronic Vascular, Inc. Devices and methods for injection of multiple-component therapies
US8338166B2 (en) 2007-01-04 2012-12-25 Lawrence Livermore National Security, Llc Sorting, amplification, detection, and identification of nucleic acid subsequences in a complex mixture
DE102007001370A1 (en) 2007-01-09 2008-07-10 Curevac Gmbh RNA-encoded antibodies
WO2008091799A2 (en) 2007-01-22 2008-07-31 The Trustees Of Columbia University In The City Of New York Cell-based methods for identifying inhibitors of parkinson's disease-associated lrrk2 mutants
CA3001783C (en) 2007-01-30 2020-09-08 Epivax, Inc. Regulatory t cell epitopes, compositions and uses thereof
WO2008097926A2 (en) 2007-02-02 2008-08-14 Yale University Transient transfection with rna
TWI432449B (en) 2007-02-02 2014-04-01 Acceleron Pharma Inc Variants derived from actriib and uses therefor
WO2008096370A2 (en) 2007-02-05 2008-08-14 Natco Pharma Limited An efficient and novel purification method of recombinant hg-csf
US8333799B2 (en) 2007-02-12 2012-12-18 C. R. Bard, Inc. Highly flexible stent and method of manufacture
CA2689042A1 (en) 2007-02-16 2008-08-28 Merck & Co., Inc. Compositions and methods for potentiated activity of biologicaly active molecules
US8242087B2 (en) 2007-02-27 2012-08-14 K.U.Leuven Research & Development Phosphate modified nucleosides useful as substrates for polymerases and as antiviral agents
EP1964922A1 (en) 2007-03-02 2008-09-03 Boehringer Ingelheim Pharma GmbH &amp; Co. KG Improvement of protein production
MY148472A (en) 2007-03-02 2013-04-30 Boehringer Ingelheim Pharma Improvement of protein production
AU2008222822A1 (en) 2007-03-05 2008-09-12 Washington University Nanoparticle delivery systems for membrane-integrating peptides
US8029496B2 (en) 2007-03-05 2011-10-04 Ebrahim Versi Method and device for delivering drug to the trigone of the bladder
WO2008115504A2 (en) 2007-03-20 2008-09-25 Millennium Pharmaceuticals, Inc. Nucleic acids encoding humanized immunoglobulin that binds a4b7 integrin
WO2008121949A1 (en) 2007-03-30 2008-10-09 Bind Biosciences, Inc. Cancer cell targeting using nanoparticles
AU2008245585B2 (en) 2007-04-27 2011-10-06 Echo Therapeutics, Inc. Skin permeation device for analyte sensing or transdermal drug delivery
EP2152290B1 (en) 2007-04-30 2014-06-04 GlaxoSmithKline LLC Methods for administering anti-il-5 antibodies
US8703204B2 (en) 2007-05-03 2014-04-22 Bend Research, Inc. Nanoparticles comprising a cholesteryl ester transfer protein inhibitor and anon-ionizable polymer
EP2157982B1 (en) 2007-05-04 2014-12-17 Marina Biotech, Inc. Amino acid lipids and uses thereof
WO2009023311A2 (en) 2007-05-07 2009-02-19 Alba Therapeutics Corporation Transcutaneous delivery of therapeutic agents
JP5296328B2 (en) 2007-05-09 2013-09-25 独立行政法人理化学研究所 Single-stranded circular RNA and method for producing the same
US8202983B2 (en) 2007-05-10 2012-06-19 Agilent Technologies, Inc. Thiocarbon-protecting groups for RNA synthesis
HUE036885T2 (en) 2007-05-14 2018-08-28 Astrazeneca Ab Methods of reducing basophil levels
WO2008144365A2 (en) 2007-05-17 2008-11-27 Novartis Ag Method for making dry powder compositions containing ds-rna based on supercritical fluid technology
CA2687746A1 (en) 2007-05-22 2008-12-18 Novartis Ag Methods of treating, diagnosing and detecting fgf21-associated disorders
EP2160464B1 (en) 2007-05-30 2014-05-21 Northwestern University Nucleic acid functionalized nanoparticles for therapeutic applications
CN101802172A (en) 2007-05-30 2010-08-11 通用医疗公司 methods of generating pluripotent cells from somatic cells
CN104072561B (en) 2007-06-19 2017-12-22 路易斯安那州州立大学及农业机械学院管理委员会 The synthesis of the anti-reverse phosphorothioate analogs of mRNA cap and purposes
WO2009006438A2 (en) 2007-06-29 2009-01-08 Epicentre Technologies Corporation Copy dna and sense rna
US8367318B2 (en) 2007-07-23 2013-02-05 Dharmacon, Inc. Screening of micro-RNA cluster inhibitor pools
US20090042825A1 (en) 2007-08-06 2009-02-12 Majed Matar Composition, method of preparation & application of concentrated formulations of condensed nucleic acids with a cationic lipopolymer
US9144546B2 (en) 2007-08-06 2015-09-29 Clsn Laboratories, Inc. Nucleic acid-lipopolymer compositions
SG183726A1 (en) 2007-08-14 2012-09-27 Hutchinson Fred Cancer Res Needle array assembly and method for delivering therapeutic agents
US20100161032A1 (en) 2007-08-15 2010-06-24 Francisco Avellanet Biologically engineered stent
EP2183390A1 (en) 2007-08-23 2010-05-12 Novartis Ag Methods for detecting oligonucleotides
WO2009030254A1 (en) 2007-09-04 2009-03-12 Curevac Gmbh Complexes of rna and cationic peptides for transfection and for immunostimulation
JP5209723B2 (en) 2007-09-05 2013-06-12 エフ.ホフマン−ラ ロシュ アーゲー Combination therapy with type I and type II anti-CD20 antibodies
US8506928B2 (en) 2007-09-07 2013-08-13 The Regents Of The University Of California Methods and compounds for targeting tissues
US20110086904A1 (en) 2007-09-17 2011-04-14 The Trustees Of The University Of Pennsylvania GENERATION OF HYPERSTABLE mRNAs
EP2535419A3 (en) 2007-09-26 2013-05-29 Intrexon Corporation Synthetic 5'UTRs, expression vectors, and methods for increasing transgene expression
US8394763B2 (en) 2007-09-26 2013-03-12 Oregon Health & Science University Cyclic undecapeptides and derivatives as multiple sclerosis therapies
EP2042193A1 (en) 2007-09-28 2009-04-01 Biomay AG RNA Vaccines
WO2009046388A1 (en) 2007-10-03 2009-04-09 United States Medical Research & Material Command Cr-2 binding peptide p28 as molecular adjuvant for dna vaccines
WO2009046738A1 (en) 2007-10-09 2009-04-16 Curevac Gmbh Composition for treating lung cancer, particularly of non-small lung cancers (nsclc)
WO2009046739A1 (en) 2007-10-09 2009-04-16 Curevac Gmbh Composition for treating prostate cancer (pca)
JP2011500569A (en) 2007-10-12 2011-01-06 マサチューセッツ インスティテュート オブ テクノロジー Vaccine nanotechnology
US20090098118A1 (en) 2007-10-15 2009-04-16 Thomas Friess Combination therapy of a type ii anti-cd20 antibody with an anti-bcl-2 active agent
JP5336500B2 (en) * 2007-10-17 2013-11-06 韓国科学技術院 Low density lipoprotein-like (LDL-like) cationic nanoparticles for nucleic acid transfer, method for producing the same, and method for transferring nucleic acid using the same
WO2009052830A1 (en) 2007-10-22 2009-04-30 Genmab A/S Novel antibody therapies
CN101965409A (en) 2007-11-01 2011-02-02 罗切斯特大学 Recombinant factor VIII with stability of increase
US20110002934A1 (en) 2007-11-09 2011-01-06 Novartis Ag Uses of anti-cd40 antibodies
EP2223934B1 (en) 2007-11-14 2012-10-03 Institute Of Microbiology, Chinese Academy Of Sciences Polypeptides for inhibiting influenza virus infection
US20090137945A1 (en) 2007-11-28 2009-05-28 Claire Marquez Electro Collagen Induction Therapy Device
EP2222709B1 (en) 2007-11-30 2016-11-23 Glaxo Group Limited Antigen-binding constructs
EP2610341B1 (en) 2007-12-11 2014-09-17 The Scripps Research Institute Compositions and methods related to mRNA translational enhancer elements
AU2008334948B2 (en) 2007-12-13 2014-11-20 Alnylam Pharmaceuticals, Inc. Methods and compositions for prevention or treatment of RSV infection
EP2072618A1 (en) 2007-12-14 2009-06-24 Johannes Gutenberg-Universität Mainz Use of RNA for reprogramming somatic cells
WO2009086072A2 (en) 2007-12-21 2009-07-09 Genentech, Inc. Therapy of rituximab-refractory rheumatoid arthritis patients
BRPI0906795A2 (en) 2008-01-23 2015-08-18 Ajinomoto Kk Method to Produce an L-Amino Acid
US20100297750A1 (en) 2008-01-24 2010-11-25 Toru Natsume Polynucleotide or analogue thereof, and gene expression regulation method using the polynucleotide or the analogue thereof
AU2009210266B2 (en) 2008-01-31 2015-01-29 CureVac SE Nucleic acids comprising formula (NuGlXmGmGnNv)a and derivatives thereof as an immunostimulating agents/adjuvants
EP2250252A2 (en) 2008-02-11 2010-11-17 Cambridge Enterprise Limited Improved reprogramming of mammalian cells, and the cells obtained
CA2726861C (en) 2008-02-13 2014-05-27 Intarcia Therapeutics, Inc. Devices, formulations, and methods for delivery of multiple beneficial agents
DE102008009920A1 (en) 2008-02-15 2009-08-20 Aj Innuscreen Gmbh Mobile device for nucleic acid isolation
US20120027813A1 (en) 2008-02-22 2012-02-02 Novartis Vaccines And Diagnostics Srl Adjuvanted influenza vaccines for pediatric use
US8506966B2 (en) 2008-02-22 2013-08-13 Novartis Ag Adjuvanted influenza vaccines for pediatric use
US20100004313A1 (en) 2008-02-29 2010-01-07 Tbd Modified Poloxamers for Gene Expression and Associated Methods
CN102027043A (en) 2008-03-14 2011-04-20 艾根股份有限公司 Biodegradable cross-linked branched poly (alkylene imines)
MY159517A (en) 2008-03-14 2017-01-13 Biocon Ltd A monoclonal antibody and a method thereof
AU2009228163B2 (en) 2008-03-28 2012-08-30 Glaxosmithkline Llc Methods of treatment
NZ588583A (en) * 2008-04-15 2012-08-31 Protiva Biotherapeutics Inc Novel lipid formulations for nucleic acid delivery
WO2009127230A1 (en) * 2008-04-16 2009-10-22 Curevac Gmbh MODIFIED (m)RNA FOR SUPPRESSING OR AVOIDING AN IMMUNOSTIMULATORY RESPONSE AND IMMUNOSUPPRESSIVE COMPOSITION
DK2288336T3 (en) 2008-04-25 2017-03-13 Univ Northwestern NANOSTRUCTURES SUITABLE FOR COMPLEXATION OF CHOLESTEROL
WO2009134808A2 (en) 2008-04-28 2009-11-05 President And Fellows Of Harvard College Supercharged proteins for cell penetration
US8715689B2 (en) 2008-04-30 2014-05-06 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services, Centers For Disease Control And Prevention Chimeric west nile/dengue viruses
US9394538B2 (en) 2008-05-07 2016-07-19 Shi-Lung Lin Development of universal cancer drugs and vaccines
CN102202708B (en) 2008-05-08 2015-01-21 迷你泵有限责任公司 Drug-delivery pumps and methods of manufacture
US8697098B2 (en) 2011-02-25 2014-04-15 South Dakota State University Polymer conjugated protein micelles
CN102083878B (en) 2008-05-13 2013-07-10 华盛顿大学 Diblock copolymers and polynucleotide complexes thereof for delivery into cells
WO2009141146A1 (en) 2008-05-21 2009-11-26 Gunther Hartmann 5' triphosphate oligonucleotide with blunt end and uses thereof
ES2872377T3 (en) 2008-05-26 2021-11-02 Univ Zuerich Protamine / RNA Nanoparticles for Immunostimulation
US20120264686A9 (en) 2008-05-29 2012-10-18 Hanall Biopharma Co. Ltd Modified erythropoietin (epo) polypeptides that exhibit increased protease resistance and pharmaceutical compositions thereof
FR2931824B1 (en) 2008-05-29 2014-11-28 Centre Nat Rech Scient PROCESS FOR RNA SYNTHESIS THROUGH CHEMICAL.
US20100086922A1 (en) 2008-05-30 2010-04-08 Millennium Pharmaceuticals, Inc. Assessment of chromosomal alterations to predict clinical outcome of bortezomib treatment
EP2433664B1 (en) 2008-06-06 2016-08-10 Wockhardt Limited A device for delivery of biological material
PL215513B1 (en) 2008-06-06 2013-12-31 Univ Warszawski New borane phosphate analogs of dinucleotides, their application, RNA particle, method of obtaining RNA and method of obtaining peptides or protein
TWI451876B (en) 2008-06-13 2014-09-11 Lilly Co Eli Pegylated insulin lispro compounds
US8613951B2 (en) 2008-06-16 2013-12-24 Bind Therapeutics, Inc. Therapeutic polymeric nanoparticles with mTor inhibitors and methods of making and using same
EP2309991B1 (en) 2008-06-16 2019-03-06 Pfizer Inc Therapeutic polymeric nanoparticles comprising vinca alkaloids and methods of making and using same
EP2285350B1 (en) 2008-06-16 2017-11-15 Pfizer Inc Methods for the preparation of targeting agent functionalized diblock copolymers for use in fabrication of therapeutic nanoparticles
SI2774608T1 (en) 2008-06-16 2020-02-28 Pfizer Inc. Drug loaded polymeric nanoparticles and methods of making and using same
US7799016B2 (en) 2008-06-20 2010-09-21 Pharmaco-Kinesis Corporation Magnetic breather pump and a method for treating a brain tumor using the same
US8996104B2 (en) 2008-06-25 2015-03-31 Fe3 Medical, Inc. Patches and method for the transdermal delivery of a therapeutically effective amount of iron
US20100009424A1 (en) 2008-07-14 2010-01-14 Natasha Forde Sonoporation systems and methods
EP2313085A2 (en) 2008-07-15 2011-04-27 Novartis AG Immunogenic amphipathic peptide compositions
WO2010009065A2 (en) 2008-07-15 2010-01-21 Novartis Ag Amphipathic peptide compositions
WO2010021865A1 (en) 2008-08-18 2010-02-25 Merck Sharp & Dohme Corp. Novel lipid nanoparticles and novel components for delivery of nucleic acids
WO2010024871A1 (en) 2008-08-26 2010-03-04 Med Institute, Inc. Balloon catheters having a plurality of needles for the injection of one or more therapeutic agents
EP2331561A4 (en) 2008-09-03 2013-02-27 Xenome Ltd Libraries of peptide conjugates and methods for making them
CA2735251C (en) 2008-09-06 2017-07-11 Chemgenes Corporation Rna synthesis - phosphoramidites for synthetic rna in the reverse direction, and application in convenient introduction of ligands, chromophores and modifications of synthetic rna at the 3' - end
US20120100558A1 (en) 2008-09-08 2012-04-26 Hanash Samir M Lung cancer diagnosis
WO2010030763A2 (en) 2008-09-10 2010-03-18 Bind Biosciences, Inc. High throughput fabrication of nanoparticles
AR073295A1 (en) 2008-09-16 2010-10-28 Genentech Inc METHODS TO TREAT PROGRESSIVE MULTIPLE SCLEROSIS. MANUFACTURING ARTICLE.
US20120021519A1 (en) 2008-09-19 2012-01-26 Presidents And Fellows Of Harvard College Efficient induction of pluripotent stem cells using small molecule compounds
WO2010037408A1 (en) 2008-09-30 2010-04-08 Curevac Gmbh Composition comprising a complexed (m)rna and a naked mrna for providing or enhancing an immunostimulatory response in a mammal and uses thereof
WO2010042490A1 (en) 2008-10-06 2010-04-15 Boston Medical Center Corporation A single lentiviral vector system for induced pluripotent (ips) stem cells derivation
CA2740000C (en) 2008-10-09 2017-12-12 Tekmira Pharmaceuticals Corporation Improved amino lipids and methods for the delivery of nucleic acids
WO2010040559A2 (en) 2008-10-10 2010-04-15 Milux Holding Sa Infusion of drugs
US8535655B2 (en) 2008-10-10 2013-09-17 Polyactiva Pty Ltd. Biodegradable polymer—bioactive moiety conjugates
US8343498B2 (en) 2008-10-12 2013-01-01 Massachusetts Institute Of Technology Adjuvant incorporation in immunonanotherapeutics
ITVI20080239A1 (en) 2008-10-14 2010-04-15 Antoine Assaf MEDICAL APPARATUS FOR MULTIPLE INJECTIONS.
WO2010047765A2 (en) 2008-10-20 2010-04-29 Massachussetts Institute Of Technology Nanostructures for drug delivery
US20120015899A1 (en) 2008-10-25 2012-01-19 Plant Bioscience, Limited Modified plant virus particles and uses therefor
EP3269395A1 (en) 2008-11-07 2018-01-17 Massachusetts Institute Of Technology Aminoalcohol lipidoids and uses thereof
AU2009313201B2 (en) 2008-11-10 2016-06-16 Arbutus Biopharma Corporation Novel lipids and compositions for the delivery of therapeutics
WO2010057203A2 (en) 2008-11-17 2010-05-20 The Board Of Regents Of The University Of Texas System Hdl particles for delivery of nucleic acids
US20110223257A1 (en) 2008-11-17 2011-09-15 Enzon Pharmaceuticals, Inc. Releasable fusogenic lipids for nucleic acids delivery systems
KR101073875B1 (en) 2008-11-28 2011-10-14 한국생명공학연구원 Diagnostic kit of colon cancer using colon cancer related marker, and Diagnostic method therof
EP2191840A1 (en) 2008-11-28 2010-06-02 Sanofi-Aventis Antitumor combinations containing antibodies recognizing specifically CD38 and melphalan
JP5855462B2 (en) 2008-12-10 2016-02-09 アルナイラム ファーマシューティカルズ, インコーポレイテッドAlnylam Pharmaceuticals, Inc. DsRNA compositions targeting GNAQ and methods for inhibiting expression
EP2196476A1 (en) 2008-12-10 2010-06-16 Novartis Ag Antibody formulation
US8563041B2 (en) 2008-12-12 2013-10-22 Bind Therapeutics, Inc. Therapeutic particles suitable for parenteral administration and methods of making and using same
US8512964B2 (en) 2008-12-12 2013-08-20 The Regents Of The University Of California Targets for treatment of hypercholesterolemia
EA201100765A1 (en) 2008-12-15 2012-04-30 Бинд Биосаиэнсис Long-term circulation nanoparticles
US20110250262A1 (en) * 2008-12-24 2011-10-13 Biomedcore, Inc. Method for producing liposome and method for dissolving cholesterol
WO2010080724A1 (en) 2009-01-12 2010-07-15 Merck Sharp & Dohme Corp. Novel lipid nanoparticles and novel components for delivery of nucleic acids
MX2011007589A (en) 2009-01-16 2011-08-17 Glaxosmithkline Llc Treatment of a cancer using a combination of bendamustine and an anti-cd20 antibody.
WO2010084371A1 (en) 2009-01-26 2010-07-29 Mitoprod Novel circular interfering rna molecules
CA3036963A1 (en) 2009-01-29 2010-08-05 Arbutus Biopharma Corporation Lipid formulations comprising cationic lipid and a targeting lipid comprising n-acetyl galactosamine for delivery of nucleic acid
US8669085B2 (en) 2009-02-05 2014-03-11 Ut-Battelle, Llc Transformation of gram positive bacteria by sonoporation
WO2010088927A1 (en) 2009-02-09 2010-08-12 Curevac Gmbh Use of pei for the improvement of endosomal release and expression of transfected nucleic acids, complexed with cationic or polycationic compounds
US20140141089A1 (en) 2009-02-11 2014-05-22 Colorado School Of Mines Nanoparticles, Compositions Thereof, and Methods of Use, and Methods of Making the Same
SG174150A1 (en) 2009-02-24 2011-10-28 Scripps Research Inst Reengineering mrna primary structure for enhanced protein production
WO2010141135A2 (en) 2009-03-05 2010-12-09 Trustees Of Boston University Bacteriophages expressing antimicrobial peptides and uses thereof
US8685458B2 (en) 2009-03-05 2014-04-01 Bend Research, Inc. Pharmaceutical compositions of dextran polymer derivatives
WO2010105277A1 (en) 2009-03-13 2010-09-16 Egen, Inc. Compositions and methods for the delivery of biologically active rnas
WO2010108108A2 (en) 2009-03-20 2010-09-23 Egen, Inc. Polyamine derivatives
WO2010111290A1 (en) 2009-03-23 2010-09-30 University Of Utah Research Foundation Methods and compositions related to modified guanine bases for controlling off-target effects in rna interference
JP5622254B2 (en) 2009-03-31 2014-11-12 国立大学法人東京大学 Double-stranded ribonucleic acid polyion complex
AU2010271116B2 (en) 2009-04-03 2015-08-13 University Of Chicago Compositions and methods related to Protein A (SpA) variants
JP5658230B2 (en) 2009-04-13 2015-01-21 インサーム(インスティテュート ナショナル デ ラ セント エ ドゥ ラ ルシェルシュ メディカル) HPV particles and uses thereof
EP2419137A4 (en) 2009-04-17 2013-01-09 Biogen Idec Inc Compositions and methods to treat acute myelogenous leukemia
KR20120022984A (en) 2009-04-21 2012-03-12 셀렉타 바이오사이언시즈, 인크. Immunonanotherapeutics providing a th1-biased response
US20100273220A1 (en) 2009-04-22 2010-10-28 Massachusetts Institute Of Technology Innate immune suppression enables repeated delivery of long rna molecules
NO2424895T3 (en) 2009-04-27 2018-02-03
US8715736B2 (en) 2009-04-30 2014-05-06 Florida Agricultural And Mechanical University Nanoparticle formulations for skin delivery
US8287910B2 (en) 2009-04-30 2012-10-16 Intezyne Technologies, Inc. Polymeric micelles for polynucleotide encapsulation
CA2760706C (en) * 2009-05-05 2019-08-06 Alnylam Pharmaceuticals, Inc. Methods of delivering oligonucleotides to immune cells
KR20220150411A (en) 2009-05-05 2022-11-10 알닐람 파마슈티칼스 인코포레이티드 Lipid compositions
DE202009007116U1 (en) 2009-05-18 2010-10-14 Amoena Medizin-Orthopädie-Technik GmbH Anti decubitus cushions
BRPI1012036A2 (en) 2009-05-27 2017-10-10 Selecta Biosciences Inc nanocarriers that have components with different release rates
US8574835B2 (en) 2009-05-29 2013-11-05 Life Technologies Corporation Scaffolded nucleic acid polymer particles and methods of making and using
EP2440556A1 (en) 2009-06-10 2012-04-18 Vertex Pharmaceuticals Incorporated Inhibitors of phosphatidylinositol 3-kinase
PL3431076T3 (en) 2009-06-10 2022-01-31 Arbutus Biopharma Corporation Improved lipid formulation
WO2010148013A2 (en) 2009-06-15 2010-12-23 Alnylam Pharmaceuticals, Inc. Lipid formulated dsrna targeting the pcsk9 gene
US20110097329A1 (en) 2009-06-26 2011-04-28 Massachusetts Institute Of Technology Compositions and methods for treating cancer and modulating stress granule formation
CA2767127A1 (en) 2009-07-01 2011-01-06 Protiva Biotherapeutics, Inc. Novel lipid formulations for delivery of therapeutic agents to solid tumors
WO2011000106A1 (en) 2009-07-01 2011-01-06 Protiva Biotherapeutics, Inc. Improved cationic lipids and methods for the delivery of therapeutic agents
WO2011005799A2 (en) 2009-07-06 2011-01-13 Novartis Ag Self replicating rna molecules and uses thereof
ES2731273T3 (en) 2009-07-31 2019-11-14 Ethris Gmbh RNA with a combination of unmodified and modified nucleotides for protein expression
EP2281579A1 (en) 2009-08-05 2011-02-09 BioNTech AG Vaccine composition comprising 5'-Cap modified RNA
EP2467357B1 (en) 2009-08-20 2016-03-30 Sirna Therapeutics, Inc. Novel cationic lipids with various head groups for oligonucleotide delivery
IN2012DN02589A (en) 2009-08-26 2015-08-28 Selecta Biosciences Inc
US20110053829A1 (en) 2009-09-03 2011-03-03 Curevac Gmbh Disulfide-linked polyethyleneglycol/peptide conjugates for the transfection of nucleic acids
US20110070227A1 (en) 2009-09-18 2011-03-24 Anna-Marie Novotney-Barry Treatment of Autoimmune and Inflammatory Diseases
WO2011043913A2 (en) 2009-10-08 2011-04-14 Merck Sharp & Dohme Corp. Novel cationic lipids with short lipid chains for oligonucleotide delivery
US8859284B2 (en) 2009-10-22 2014-10-14 The United States Of America, As Represented By The Secretary Of The Navy Delivery of nanoparticles to neurons
CA2816925C (en) 2009-11-04 2023-01-10 The University Of British Columbia Nucleic acid-containing lipid particles and related methods
US8449916B1 (en) 2009-11-06 2013-05-28 Iowa State University Research Foundation, Inc. Antimicrobial compositions and methods
WO2011060250A1 (en) 2009-11-13 2011-05-19 Bend Research, Inc. Cationic dextran polymer derivatives
US9415113B2 (en) 2009-11-18 2016-08-16 University Of Washington Targeting monomers and polymers having targeting blocks
US8530429B2 (en) 2009-11-24 2013-09-10 Arch Cancer Therapeutics, Inc. Brain tumor targeting peptides and methods
US20110245756A1 (en) 2009-12-03 2011-10-06 Rishi Arora Devices for material delivery, electroporation, sonoporation, and/or monitoring electrophysiological activity
DE102009056884B4 (en) 2009-12-03 2021-03-18 Novartis Ag Vaccine Adjuvants and Improved Methods for Making Same
PT3326643T (en) 2009-12-06 2021-07-12 Bioverativ Therapeutics Inc Factor viii-fc chimeric and hybrid polypeptides, and methods of use thereof
EP3296398A1 (en) 2009-12-07 2018-03-21 Arbutus Biopharma Corporation Compositions for nucleic acid delivery
US20130189741A1 (en) 2009-12-07 2013-07-25 Cellscript, Inc. Compositions and methods for reprogramming mammalian cells
WO2011071936A2 (en) 2009-12-07 2011-06-16 Gary Dahl Compositions and methods for reprogramming eukaryotic cells
WO2011069528A1 (en) 2009-12-09 2011-06-16 Curevac Gmbh Lyophilization of nucleic acids in lactate-containing solutions
WO2011069529A1 (en) 2009-12-09 2011-06-16 Curevac Gmbh Mannose-containing solution for lyophilization, transfection and/or injection of nucleic acids
EA036522B1 (en) 2009-12-11 2020-11-19 Пфайзер Инк. Pharmaceutical composition suitable for lyophilization comprising a plurality of therapeutic particles
JP5898627B2 (en) 2009-12-15 2016-04-06 バインド セラピューティックス インコーポレイテッド Therapeutic polymer nanoparticles containing epothilone and methods of making and using the same
EA201290499A1 (en) 2009-12-15 2013-01-30 Байнд Байосайенсиз, Инк. COMPOSITIONS OF THERAPEUTIC POLYMER NANOPARTICLES WITH HIGH GLASSING TEMPERATURE AND HIGH-MOLECULAR CO-POLYMERS
WO2011084518A2 (en) 2009-12-15 2011-07-14 Bind Biosciences, Inc. Therapeutic polymeric nanoparticles comprising corticosteroids and methods of making and using same
DE102009058769A1 (en) 2009-12-16 2011-06-22 MagForce Nanotechnologies AG, 10589 Temperature-dependent activation of catalytic nucleic acids for controlled drug release
US20130017265A1 (en) 2009-12-16 2013-01-17 Massachusetts Institute Of Technology Particles for multiple agent delivery
CA2784568A1 (en) 2009-12-18 2011-06-23 Martin A. Maier Lipid particles for delivery of nucleic acids
EP2338520A1 (en) 2009-12-21 2011-06-29 Ludwig Maximilians Universität Conjugate with targeting ligand and use of same
US20110200582A1 (en) 2009-12-23 2011-08-18 Novartis Ag Lipids, lipid compositions, and methods of using them
US20110171248A1 (en) 2010-01-08 2011-07-14 Selecta Biosciences, Inc. Synthetic virus-like particles conjugated to human papillomavirus capsid peptides for use as vaccines
WO2011088309A1 (en) 2010-01-14 2011-07-21 Regulus Therapeutics Inc. Microrna compositions and methods
EP2525781A1 (en) 2010-01-22 2012-11-28 Schering Corporation Novel cationic lipids for oligonucleotide delivery
US20130101609A1 (en) 2010-01-24 2013-04-25 Novartis Ag Irradiated biodegradable polymer microparticles
PL2539451T3 (en) 2010-02-24 2016-08-31 Arrowhead Res Corporation Compositions for targeted delivery of sirna
US8889193B2 (en) 2010-02-25 2014-11-18 The Johns Hopkins University Sustained delivery of therapeutic agents to an eye compartment
WO2011112608A1 (en) 2010-03-08 2011-09-15 University Of Rochester Synthesis of nanoparticles using reducing gases
WO2011116072A1 (en) 2010-03-16 2011-09-22 Escape Therapeutics, Inc. Hybrid hydrogel scaffold compositions and methods of use
JP2013521805A (en) 2010-03-16 2013-06-13 ユニバーシティ・オブ・ユタ・リサーチ・ファウンデイション Cleavageable modifications to reducing poly (amidoethyleneimine) to enhance nucleotide delivery
US20110230816A1 (en) 2010-03-18 2011-09-22 Tyco Healthcare Group Lp Gels for Transdermal Delivery
WO2011115862A1 (en) 2010-03-18 2011-09-22 Merck Sharp & Dohme Corp. Endosomolytic poly(amidoamine) disulfide polymers for the delivery of oligonucleotides
US9149432B2 (en) 2010-03-19 2015-10-06 Massachusetts Institute Of Technology Lipid vesicle compositions and methods of use
CA2794189C (en) * 2010-03-24 2022-01-11 Rxi Pharmaceuticals Corporation Rna interference in dermal and fibrotic indications
GB201005005D0 (en) 2010-03-25 2010-05-12 Angeletti P Ist Richerche Bio New vaccine
EP2553019A1 (en) 2010-03-26 2013-02-06 Mersana Therapeutics, Inc. Modified polymers for delivery of polynucleotides, method of manufacture, and methods of use thereof
WO2011119262A1 (en) 2010-03-26 2011-09-29 Cerulean Pharma Inc. Methods and systems for generating nanoparticles
US20110247090A1 (en) 2010-04-02 2011-10-06 Intrexon Corporation Synthetic 5'UTRs, Expression Vectors, and Methods for Increasing Transgene Expression
JP2013523818A (en) 2010-04-05 2013-06-17 ザ・ユニバーシティー・オブ・シカゴ Compositions and methods relating to protein A (SpA) antibodies as enhancers of immune responses
EP2556151A1 (en) 2010-04-07 2013-02-13 Novartis AG Method for generating a parvovirus b19 virus-like particle
US20130037977A1 (en) 2010-04-08 2013-02-14 Paul A. Burke Preparation of Lipid Nanoparticles
WO2011125469A1 (en) 2010-04-09 2011-10-13 国立大学法人東京大学 Micro-rna-regulated recombinant vaccinia virus and utilization thereof
CN104959087B (en) 2010-04-09 2017-08-15 帕西拉制药有限公司 For preparing the method that major diameter synthesizes membrane vesicle
US20110262491A1 (en) 2010-04-12 2011-10-27 Selecta Biosciences, Inc. Emulsions and methods of making nanocarriers
KR101196667B1 (en) 2010-04-15 2012-11-02 포항공과대학교 산학협력단 A DELEVERY SYSTEM OF ANTI-CANCER AGENT USING pH SENSITIVE METAL NANOPARTICLE
CA2832672A1 (en) 2010-04-16 2011-10-20 Nuevolution A/S Bi-functional complexes and methods for making and using such complexes
EP2377938A1 (en) 2010-04-16 2011-10-19 Eukarys Capping-prone RNA polymerase enzymes and their applications
WO2011133868A2 (en) 2010-04-22 2011-10-27 Alnylam Pharmaceuticals, Inc. Conformationally restricted dinucleotide monomers and oligonucleotides
US9629979B2 (en) 2010-04-28 2017-04-25 Sanovas, Inc. Pressure/Vacuum actuated catheter drug delivery probe
AU2011311255B2 (en) 2010-04-28 2015-10-08 Sorrento Therapeutics, Inc. Method for increasing permeability of an epithelial barrier
WO2011139911A2 (en) 2010-04-29 2011-11-10 Isis Pharmaceuticals, Inc. Lipid formulated single stranded rna
MA34263B1 (en) 2010-04-30 2013-05-02 Novartis Ag PREDICTION MARKERS USEFUL IN TREATING FRAGILE X (FXS) SYNDROME
WO2011143230A1 (en) 2010-05-10 2011-11-17 Alnylam Pharmaceuticals Methods and compositions for delivery of active agents
WO2011141704A1 (en) 2010-05-12 2011-11-17 Protiva Biotherapeutics, Inc Novel cyclic cationic lipids and methods of use
CA2799091A1 (en) 2010-05-12 2011-11-17 Protiva Biotherapeutics, Inc. Cationic lipids and methods of use thereof
EP2387999A1 (en) 2010-05-21 2011-11-23 CureVac GmbH Histidine-containing solution for transfection and/or injection of nucleic acids and uses thereof
US8802863B2 (en) 2010-05-24 2014-08-12 Sirna Therapeutics, Inc. Amino alcohol cationic lipids for oligonucleotide delivery
JP6367554B2 (en) 2010-05-26 2018-08-01 セレクタ バイオサイエンシーズ インコーポレーテッドSelecta Biosciences,Inc. Dosage selection of adjuvanted synthetic nanocarriers
DK2575767T3 (en) 2010-06-04 2017-03-13 Sirna Therapeutics Inc HOWEVER UNKNOWN LOW MOLECULAR CATIONIC LIPIDS TO PROCESS OIGONUCLEOTIDES
WO2011157715A1 (en) 2010-06-14 2011-12-22 F. Hoffmann-La Roche Ag Cell-penetrating peptides and uses therof
WO2011163121A1 (en) 2010-06-21 2011-12-29 Alnylam Pharmaceuticals, Inc. Multifunctional copolymers for nucleic acid delivery
WO2011163264A2 (en) 2010-06-21 2011-12-29 Candela Corporation Driving microneedle arrays into skin and delivering rf energy
AU2011268507B2 (en) 2010-06-25 2014-08-14 Novartis Ag Combinations of meningococcal factor H binding proteins
CN103079592B (en) 2010-07-01 2015-10-21 浦项工科大学校产学协力团 Use from the microbubble treatment of cell and the method for cancer diagnosis
US8821894B2 (en) 2010-07-02 2014-09-02 The University Of Chicago Compositions and methods related to protein A (SpA) variants
KR101130137B1 (en) 2010-07-02 2012-03-28 연세대학교 산학협력단 Led module
US8353871B2 (en) 2010-07-05 2013-01-15 Roller Jet Ltd. Drug delivery device with needles and roller
US10487332B2 (en) 2010-07-06 2019-11-26 Glaxosmithkline Biologicals Sa Immunisation of large mammals with low doses of RNA
HUE047796T2 (en) 2010-07-06 2020-05-28 Glaxosmithkline Biologicals Sa Delivery of rna to trigger multiple immune pathways
US20130171241A1 (en) 2010-07-06 2013-07-04 Novartis Ag Liposomes with lipids having an advantageous pka-value for rna delivery
ES2600892T3 (en) 2010-07-06 2017-02-13 Glaxosmithkline Biologicals Sa Virion-like administration particles for self-replicating RNA molecules
US9192661B2 (en) 2010-07-06 2015-11-24 Novartis Ag Delivery of self-replicating RNA using biodegradable polymer particles
US9770463B2 (en) 2010-07-06 2017-09-26 Glaxosmithkline Biologicals Sa Delivery of RNA to different cell types
AU2011276236B2 (en) 2010-07-06 2016-03-10 Glaxosmithkline Biologicals S.A. Cationic oil-in-water emulsions
BR112013000650A8 (en) 2010-07-09 2017-10-17 Biogen Idec Hemophilia Inc factor ix polypeptides and methods of use thereof.
EP2591101B1 (en) 2010-07-09 2018-11-07 Bioverativ Therapeutics Inc. Systems for factor viii processing and methods thereof
WO2012009406A2 (en) 2010-07-13 2012-01-19 University Of Utah Research Foundation Gold particles and methods of making and using the same in cancer treatment
GB201012410D0 (en) 2010-07-23 2010-09-08 Medical Res Council Intracellular immunity
DE102010032758B4 (en) 2010-07-29 2012-02-23 Fujitsu Technology Solutions Intellectual Property Gmbh Computer system, method of programming a real time clock and computer program product
DK2449113T3 (en) 2010-07-30 2016-01-11 Curevac Ag Complex formation of nucleic acids with the disulfide cross-linked cationic components for transfection and immunostimulation
US20130190626A1 (en) 2010-08-02 2013-07-25 Curtin University Of Technology Determining location of, and imaging, a subsurface boundary
US8822663B2 (en) 2010-08-06 2014-09-02 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US9121065B2 (en) 2010-08-09 2015-09-01 The Trustees Of The University Of Pennsylvania Nanoparticle-oligonucleotide hybrid structures and methods of use thereof
WO2012019630A1 (en) 2010-08-13 2012-02-16 Curevac Gmbh Nucleic acid comprising or coding for a histone stem-loop and a poly(a) sequence or a polyadenylation signal for increasing the expression of an encoded protein
BR112013003825A2 (en) 2010-08-20 2019-09-24 Cerulean Pharma Inc related conjugates, particles, compositions and methods
US20130142868A1 (en) 2010-08-20 2013-06-06 University Of Washington Circumferential Aerosol Device for Delivering Drugs to Olfactory Epithelium and Brain
WO2012024632A2 (en) 2010-08-20 2012-02-23 Selecta Biosciences, Inc. Synthetic nanocarrier vaccines comprising peptides obtained or derived from human influenza a virus m2e
EP2605792B1 (en) 2010-08-20 2014-12-10 Novartis AG Soluble needle arrays for delivery of influenza vaccines
AU2011293508A1 (en) 2010-08-23 2013-01-24 Selecta Biosciences, Inc. Targeted multi-epitope dosage forms for induction of an immune response to antigens
CN103080582B (en) 2010-08-24 2015-10-14 Gkn动力传动系统国际有限责任公司 For the cover with transition region of universal joint especially for constant velocity universal joint
RU2577983C2 (en) 2010-08-31 2016-03-20 Новартис Аг Lipids suitable for liposomal delivery of rna encoding protein
WO2012030683A2 (en) 2010-08-31 2012-03-08 Merck Sharp & Dohme Corp. Novel single chemical entities and methods for delivery of oligonucleotides
EP3556396B1 (en) 2010-08-31 2022-04-20 Theraclone Sciences, Inc. Human immunodeficiency virus (hiv)-neutralizing antibodies
US9254265B2 (en) 2010-08-31 2016-02-09 Novartis Ag Small liposomes for delivery of immunogen encoding RNA
JP6165629B2 (en) 2010-08-31 2017-07-19 キヤノン ユー.エス. ライフ サイエンシズ, インコーポレイテッドCanon U.S. Life Sciences, Inc. Method, device and system for fluid mixing and chip interface
SI3970742T1 (en) 2010-08-31 2022-08-31 Glaxosmithkline Biologicals S.A. Pegylated liposomes for delivery of immunogen-encoding rna
US9549901B2 (en) 2010-09-03 2017-01-24 The Brigham And Women's Hospital, Inc. Lipid-polymer hybrid particles
EP2614074A1 (en) 2010-09-09 2013-07-17 The University of Chicago Methods and compositions involving protective staphylococcal antigens
WO2012034077A2 (en) 2010-09-09 2012-03-15 The University Of Chicago Compositions and methods related to attenuated staphylococcal strains
WO2012039979A2 (en) 2010-09-10 2012-03-29 The Johns Hopkins University Rapid diffusion of large polymeric nanoparticles in the mammalian brain
US8466122B2 (en) 2010-09-17 2013-06-18 Protiva Biotherapeutics, Inc. Trialkyl cationic lipids and methods of use thereof
CN103167866B (en) 2010-09-20 2015-09-23 瑟纳治疗公司 For the novel low molecular weight amount cation lipid of oligonucleotide delivery
WO2012038448A1 (en) 2010-09-21 2012-03-29 Riboxx Gmbh Method for synthesizing rna using dna template
BR112013008697A2 (en) 2010-09-24 2016-06-21 Massachusetts Inst Technology nanostructured gels capable of controlled release of encapsulated agents
WO2012050975A2 (en) 2010-09-29 2012-04-19 The University Of North Carolina At Chapel Hill Novel circular mammalian rna molecules and uses thereof
US9029604B2 (en) 2010-09-30 2015-05-12 Sirna Therapeutics, Inc. Low molecular weight cationic lipids for oligonucleotide delivery
NZ608972A (en) 2010-10-01 2015-09-25 Moderna Therapeutics Inc Engineered nucleic acids and methods of use thereof
US10078075B2 (en) 2011-12-09 2018-09-18 Vanderbilt University Integrated organ-on-chip systems and applications of the same
MX363307B (en) 2010-10-11 2019-03-20 Novartis Ag Star Antigen delivery platforms.
US9782342B2 (en) 2010-10-11 2017-10-10 Wichita State University Composite magnetic nanoparticle drug delivery system
EP3485913A1 (en) 2010-10-21 2019-05-22 Sirna Therapeutics, Inc. Low molecular weight cationic lipids for oligonucleotide delivery
WO2012054923A2 (en) 2010-10-22 2012-04-26 Bind Biosciences, Inc. Therapeutic nanoparticles with high molecular weight copolymers
WO2012154202A1 (en) 2010-10-29 2012-11-15 Merck Sharp & Dohme Corp. Recombinant subunit dengue virus vaccine
US9067882B2 (en) 2010-11-05 2015-06-30 Sirna Therapeutics, Inc. Low molecular weight cyclic amine containing cationic lipids for oligonucleotide delivery
EP2635254B1 (en) 2010-11-05 2019-05-15 The John Hopkins University Compositions and methods relating to reduced mucoadhesion
WO2012061717A1 (en) 2010-11-05 2012-05-10 Selecta Biosciences, Inc. Modified nicotinic compounds and related methods
JP2014500867A (en) 2010-11-09 2014-01-16 ザ リージェンツ オブ ザ ユニバーシティ オブ カリフォルニア Skin penetrating and cell invasive (SPACE) peptides and uses thereof
CN106434676B (en) 2010-11-12 2020-10-23 宾夕法尼亚大学托管会 Consensus prostate antigens, nucleic acid molecules encoding said antigens, vaccines comprising said nucleic acid molecules and uses thereof
EP2640426B1 (en) 2010-11-16 2016-02-03 Selecta Biosciences, Inc. Immunostimulatory oligonucleotides
LT2640842T (en) 2010-11-17 2018-09-10 Aduro Biotech, Inc. Methods and compositions for inducing an immune response to egfrviii
EP2640400A4 (en) 2010-11-19 2016-01-20 Sirna Therapeutics Inc Poly(amide) polymers for the delivery of oligonucleotides
ES2697606T3 (en) 2010-11-19 2019-01-25 Idera Pharmaceuticals Inc Immunoregulatory oligonucleotide (IRO) compounds to modulate the immune response based on a Toll-like receptor
US8853377B2 (en) 2010-11-30 2014-10-07 Shire Human Genetic Therapies, Inc. mRNA for use in treatment of human genetic diseases
WO2012072096A1 (en) 2010-12-03 2012-06-07 Biontech Ag Method for cellular rna expression
EP2651445A2 (en) 2010-12-16 2013-10-23 SPRNA GmbH Pharmaceutical composition consisting of rna having alkali metal as counter ion and formulated with dications
WO2012082574A1 (en) 2010-12-17 2012-06-21 Merck Sharp & Dohme Corp. Membrane lytic poly(amido amine) polymers for the delivery of oligonucleotides
US8501930B2 (en) 2010-12-17 2013-08-06 Arrowhead Madison Inc. Peptide-based in vivo siRNA delivery system
US9394537B2 (en) 2010-12-22 2016-07-19 President And Fellows Of Harvard College Continuous directed evolution
WO2012089225A1 (en) 2010-12-29 2012-07-05 Curevac Gmbh Combination of vaccination and inhibition of mhc class i restricted antigen presentation
BR112013016761B1 (en) 2010-12-29 2021-04-06 F. Hoffmann-La Roche Ag USE OF A COMPOUND, COMPOUND AND PHARMACEUTICAL COMPOSITION
US20120171229A1 (en) 2010-12-30 2012-07-05 Selecta Biosciences, Inc. Synthetic nanocarriers with reactive groups that release biologically active agents
WO2012092569A2 (en) 2010-12-31 2012-07-05 Selecta Biosciences, Inc. Compositions comprising immunostimulatory nucleic acids and related methods
US10364440B2 (en) 2011-01-04 2019-07-30 Brown University Nanotubes as carriers of nucleic acids into cells
WO2012094574A2 (en) 2011-01-06 2012-07-12 The Johns Hopkins University Stabilized polyribonucleotide nanoparticles
US20140080766A1 (en) 2011-01-07 2014-03-20 Massachusetts Institute Of Technology Compositions and methods for macromolecular drug delivery
AU2012207606B2 (en) 2011-01-11 2017-02-23 Alnylam Pharmaceuticals, Inc. Pegylated lipids and their use for drug delivery
DE102011082231A1 (en) 2011-01-12 2012-07-12 Robert Bosch Gmbh Ignition coil, especially for small engines
WO2012099805A2 (en) 2011-01-19 2012-07-26 Ocean Nanotech, Llc Nanoparticle based immunological stimulation
CA2825023A1 (en) 2011-01-26 2012-08-02 Cenix Bioscience Gmbh Delivery system and conjugates for compound delivery via naturally occurring intracellular transport routes
US10363309B2 (en) 2011-02-04 2019-07-30 Case Western Reserve University Targeted nanoparticle conjugates
WO2012109121A1 (en) 2011-02-07 2012-08-16 Purdue Research Foundation Carbohydrate nanoparticles for prolonged efficacy of antimicrobial peptide
WO2012116715A1 (en) 2011-03-02 2012-09-07 Curevac Gmbh Vaccination in newborns and infants
US20120207840A1 (en) 2011-02-10 2012-08-16 Aura Biosciences, Inc. Virion Derived Protein Nanoparticles For Delivering Diagnostic Or Therapeutic Agents For The Treatment Of Non-Melanoma Skin Cancer
US20140038185A1 (en) 2011-02-14 2014-02-06 Swift Biosciences, Inc. Polynucleotide primers and probes
CA2827118A1 (en) 2011-02-15 2012-08-23 Merrimack Pharmaceuticals, Inc. Compositions and methods for delivering nucleic acid to a cell
US20140081012A1 (en) 2011-02-15 2014-03-20 The University Of North Carolina At Chapel Hill Nanoparticle, liposomes, polymers, agents and proteins modified with reversible linkers
EP2489371A1 (en) 2011-02-18 2012-08-22 Instituto Nacional de Investigacion y Tecnologia Agraria y Alimentaria Carrier peptides for drug delivery
WO2012113413A1 (en) 2011-02-21 2012-08-30 Curevac Gmbh Vaccine composition comprising complexed immunostimulatory nucleic acids and antigens packaged with disulfide-linked polyethyleneglycol/peptide conjugates
JP6091435B2 (en) 2011-02-22 2017-03-08 カリフォルニア インスティチュート オブ テクノロジー Protein delivery using adeno-associated virus (AAV) vectors
US8696637B2 (en) 2011-02-28 2014-04-15 Kimberly-Clark Worldwide Transdermal patch containing microneedles
CN103533954B (en) 2011-03-02 2015-09-09 诺华股份有限公司 Contain compared with the antigen of low dosage and/or the combined vaccine of adjuvant
WO2012116714A1 (en) 2011-03-02 2012-09-07 Curevac Gmbh Vaccination in elderly patients
EP2683812A4 (en) 2011-03-07 2014-12-03 Massachusetts Inst Technology Methods for transfecting cells with nucleic acids
US20120237565A1 (en) 2011-03-14 2012-09-20 Intezyne Technologies, Incorporated Pegylated polyplexes containing two or more different polymers for polynucleotide delivery
WO2012125680A1 (en) 2011-03-16 2012-09-20 Novartis Ag Methods of treating vasculitis using an il-17 binding molecule
US20140212503A1 (en) 2011-03-17 2014-07-31 Hyukjin Lee Delivery system
WO2012125812A1 (en) 2011-03-17 2012-09-20 Novartis Ag Fgfr and ligands thereof as biomarkers for breast cancer in hr positive subjects
WO2012129483A1 (en) 2011-03-24 2012-09-27 Novartis Ag Adjuvant nanoemulsions with phospholipids
BR112013024655A2 (en) 2011-03-25 2016-12-20 Selecta Biosciences Inc synthetic nanotransporters for osmotic mediated release
JP2014510533A (en) 2011-03-28 2014-05-01 ノバルティス アーゲー Markers associated with cyclin-dependent kinase inhibitors
EP2691443B1 (en) 2011-03-28 2021-02-17 Massachusetts Institute of Technology Conjugated lipomers and uses thereof
EP2691078A1 (en) 2011-03-31 2014-02-05 Ingell Technologies Holding B.V. Biodegradable compositions suitable for controlled release
WO2012131104A2 (en) 2011-03-31 2012-10-04 Ingell Technologies Holding B.V. Biodegradable compositions suitable for controlled release
AU2012236099A1 (en) 2011-03-31 2013-10-03 Moderna Therapeutics, Inc. Delivery and formulation of engineered nucleic acids
WO2012138453A1 (en) 2011-04-03 2012-10-11 The General Hospital Corporation Efficient protein expression in vivo using modified rna (mod-rna)
ES2587512T3 (en) 2011-04-04 2016-10-25 The U.S.A. As Represented By The Secretary, Department Of Health And Human Services 2'-O-aminooxymethyl nucleoside derivatives for use in the synthesis and modification of nucleosides, nucleotides and oligonucleotides
WO2012142132A1 (en) 2011-04-11 2012-10-18 Life Technologies Corporation Polymer particles and methods of making and using same
US11135174B2 (en) 2011-04-13 2021-10-05 The Trustees Of The University Of Pennsylvania Coated mesoporous nanoparticles
WO2013158127A1 (en) 2012-04-16 2013-10-24 Molecular Transfer, Inc. Agents for improved delivery of nucleic acids to eukaryotic cells
US20140178894A1 (en) 2011-04-20 2014-06-26 Novartis Forschungsstiftung, Zweigniederlassung Culture medium suitable for the culture of undifferentiated cells
US20140287022A1 (en) 2011-04-26 2014-09-25 Molecular Express, Inc. Liposomal formulations
WO2012148684A1 (en) 2011-04-27 2012-11-01 President And Fellows Of Harvard College Cell-friendly inverse opal hydrogels for cell encapsulation, drug and protein delivery, and functional nanoparticle encapsulation
JP2014512409A (en) 2011-04-28 2014-05-22 エスティーシー. ユーエヌエム Lipid bilayer (protocell) supported on porous nanoparticles for targeted delivery and method of use thereof
US20160272697A2 (en) 2011-04-28 2016-09-22 The Henry M. Jackson Foundation For The Advancement Of Military Medicine, Inc. Neutralizing Antibodies to Nipah and Hendra Virus
CA2834571A1 (en) 2011-04-29 2012-11-01 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers for inducing regulatory b cells
US20120283503A1 (en) 2011-04-29 2012-11-08 The Johns Hopkins University Nanoparticle loaded stem cells and their use in mri guided hyperthermia
EP2702408A1 (en) 2011-04-29 2014-03-05 Novartis AG Methods of treating squamous cell carcinoma related applications
EP2705143B1 (en) 2011-05-02 2021-02-17 Wayne State University A protein-induced pluripotent cell technology uses thereof
UA116189C2 (en) 2011-05-02 2018-02-26 Мілленніум Фармасьютікалз, Інк. FORMULATION FOR ANTI-α4β7 ANTIBODY
EP3409112B1 (en) 2011-05-04 2022-07-27 The University of Nottingham Novel polymers which resist bacterial attachment
US9327029B2 (en) 2011-05-05 2016-05-03 Celacare Technologies, Llc Antimicrobial silver hydrogel composition for the treatment of burns and wounds
US8945588B2 (en) 2011-05-06 2015-02-03 The University Of Chicago Methods and compositions involving protective staphylococcal antigens, such as EBH polypeptides
US9650745B2 (en) 2011-05-10 2017-05-16 Basf Se Oil-in-water emulsions
US9283279B2 (en) 2011-05-11 2016-03-15 Ramot At Tel-Aviv University Ltd. Targeted polymeric conjugates and uses thereof
CA2835492A1 (en) 2011-05-12 2012-11-15 Helmut Vockner Novel pharmaceutical formulation
ES2762224T3 (en) 2011-05-12 2020-05-22 Yissum Res Dev Co Of Hebrew Univ Jerusalem Ltd Liposomes Comprising Polymer Conjugated Lipids and Related Uses
HUE048777T2 (en) 2011-05-13 2020-08-28 Glaxosmithkline Biologicals Sa Pre-fusion rsv f antigens
US8691750B2 (en) 2011-05-17 2014-04-08 Axolabs Gmbh Lipids and compositions for intracellular delivery of biologically active compounds
JP2014520084A (en) 2011-05-17 2014-08-21 モデルナ セラピューティクス インコーポレイテッド Modified nucleic acids for non-human vertebrates and methods of use thereof
CN103052757B (en) 2011-05-20 2015-02-25 科勒公司 Toilet installation system and method
SG193553A1 (en) 2011-05-24 2013-10-30 Biontech Ag Individualized vaccines for cancer
US20140227372A1 (en) 2011-05-25 2014-08-14 Novartis Ag Biomarkers for lung cancer
US20120302940A1 (en) 2011-05-26 2012-11-29 Jackson State University Popcorn Shape Gold Nanoparticle For Targeted Diagnosis, Photothermal Treatment and In-Situ Monitoring Therapy Response for Cancer and Multiple Drug Resistance Bacteria
WO2012166923A2 (en) 2011-05-31 2012-12-06 Bind Biosciences Drug loaded polymeric nanoparticles and methods of making and using same
MX349011B (en) 2011-06-02 2017-07-05 Novartis Ag Biomarkers for hedgehog inhibitor therapy.
JP6100762B2 (en) 2011-06-02 2017-03-22 ザ リージェンツ オブ ザ ユニバーシティ オブ カリフォルニア Nanoparticles encapsulated in membranes and methods of use
EP2717911A1 (en) 2011-06-06 2014-04-16 Novartis Forschungsstiftung, Zweigniederlassung Protein tyrosine phosphatase, non-receptor type 11 (ptpn11) and triple-negative breast cancer
DK3336082T3 (en) 2011-06-08 2020-04-27 Translate Bio Inc SPLITLY LIPIDS
RS59037B1 (en) 2011-06-08 2019-08-30 Translate Bio Inc Lipid nanoparticle compositions and methods for mrna delivery
WO2012170607A2 (en) 2011-06-10 2012-12-13 Novartis Ag Use of pcsk9 antagonists
WO2012168491A1 (en) 2011-06-10 2012-12-13 Novartis Ag Pharmaceutical formulations of pcsk9 antagonists
US8636696B2 (en) 2011-06-10 2014-01-28 Kimberly-Clark Worldwide, Inc. Transdermal device containing microneedles
WO2012170753A2 (en) 2011-06-10 2012-12-13 Novartis Ag Bovine vaccines and methods
US8916696B2 (en) 2011-06-12 2014-12-23 City Of Hope Aptamer-mRNA conjugates for targeted protein or peptide expression and methods for their use
WO2012172495A1 (en) 2011-06-14 2012-12-20 Novartis Ag Compositions and methods for antibodies targeting tem8
WO2012172424A1 (en) 2011-06-15 2012-12-20 Crontech Pharma Ab Injection needle and device
CA2838478A1 (en) 2011-06-16 2012-12-20 Novartis Ag Soluble proteins for use as therapeutics
AU2012273039B2 (en) 2011-06-20 2016-12-01 University Of Pittsburgh - Of The Commonwealth System Of Higher Education Computationally optimized broadly reactive antigens for H1N1 influenza
US9862926B2 (en) 2011-06-27 2018-01-09 Cellscript, Llc. Inhibition of innate immune response
JP2014524796A (en) 2011-06-28 2014-09-25 イノビオ ファーマシューティカルズ,インコーポレイティド Minimally invasive epidermal electroporation device
PL2726099T3 (en) 2011-07-01 2018-12-31 Novartis Ag Method for treating metabolic disorders
AU2012278910A1 (en) 2011-07-04 2014-01-16 Commonwealth Scientific And Industrial Research Organisation Nucleic acid complex
SG10201605500TA (en) 2011-07-06 2016-08-30 Novartis Ag Cationic oil-in-water emulsions
CA2840989A1 (en) 2011-07-06 2013-01-10 Novartis Ag Immunogenic combination compositions and uses thereof
WO2013006842A2 (en) 2011-07-06 2013-01-10 Novartis Ag Immunogenic compositions and uses thereof
EP4115875A1 (en) 2011-07-06 2023-01-11 GlaxoSmithKline Biologicals S.A. Liposomes having useful n:p ratio for delivery of rna molecules
MX350198B (en) 2011-07-06 2017-08-30 Novartis Ag Oil-in-water emulsions that contain nucleic acids.
US8975302B2 (en) 2011-07-07 2015-03-10 Life Technologies Corporation Polymer particles, nucleic acid polymer particles and methods of making and using the same
US20130012566A1 (en) 2011-07-10 2013-01-10 Aura Biosciences, Inc. Virion Derived Protein Nanoparticles For Delivering Diagnostic Or Therapeutic Agents For The Treatment of Alopecia
WO2013009717A1 (en) 2011-07-10 2013-01-17 Elisabet De Los Pinos Virion derived protein nanoparticles for delivering diagnostic or therapeutic agents for the treatment of skin-related diseases
WO2013009736A2 (en) 2011-07-10 2013-01-17 President And Fellows Of Harvard College Compositions and methods for self-assembly of polymers with complementary macroscopic and microscopic scale units
GB2492999A (en) 2011-07-20 2013-01-23 Univ Central Lancashire Neutron detector
US20140148503A1 (en) 2011-07-20 2014-05-29 University Of Iowa Research Foundation Nucleic acid aptamers
US10376461B2 (en) 2011-07-21 2019-08-13 Croda International Plc Branched polyether-polyamide block copolymers and methods of making and using the same
US9493549B2 (en) 2011-07-25 2016-11-15 The Rockefeller University Antibodies directed toward the HIV-1 GP120 CD4 binding site with increased potency and breadth
WO2013016460A1 (en) 2011-07-25 2013-01-31 Novartis Ag Compositions and methods for assessing functional immunogenicity of parvovirus vaccines
CN103702687A (en) 2011-07-29 2014-04-02 西莱克塔生物科技公司 Synthetic nanocarriers that generate humoral and cytotoxic T lymphocyte (CTL) immune responses
US9556281B2 (en) 2011-08-15 2017-01-31 The University Of Chicago Compositions and methods related to antibodies to staphylococcal protein A
JP2014531476A (en) 2011-08-26 2014-11-27 アローヘッド リサーチ コーポレイション Poly (vinyl ester) polymers for in vivo nucleic acid delivery
US9126966B2 (en) 2011-08-31 2015-09-08 Protiva Biotherapeutics, Inc. Cationic lipids and methods of use thereof
US8889657B2 (en) 2011-08-31 2014-11-18 Mallinckrodt Llc Nanoparticle PEG modification with H-phosphonates
BR112014004607A2 (en) 2011-08-31 2017-03-21 Novartis Ag pegylated liposomes for immunogenic encoded RNA delivery
US20140206682A1 (en) 2011-09-01 2014-07-24 Novartis Pharmaceuticals Uk Limited Compounds and compositions as pdgfr kinase inhibitors
EP3521432A1 (en) 2011-09-02 2019-08-07 Arrowhead Pharmaceuticals, Inc. Organic compositions to treat hsf1-related diseases
EP2755986A4 (en) * 2011-09-12 2015-05-20 Moderna Therapeutics Inc Engineered nucleic acids and methods of use thereof
WO2013039861A2 (en) * 2011-09-12 2013-03-21 modeRNA Therapeutics Engineered nucleic acids and methods of use thereof
US9464124B2 (en) 2011-09-12 2016-10-11 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
TR201909110T4 (en) 2011-09-14 2019-07-22 Glaxosmithkline Biologicals Sa Methods for making saccharide-protein glycoconjugates.
CA2848410A1 (en) 2011-09-16 2013-03-21 The Trustees Of The University Of Pennsylvania Rna engineered t cells for the treatment of cancer
US20150017245A1 (en) 2011-09-22 2015-01-15 Bind Therapeutics, Inc. Methods of treating cancers with therapeutic nanoparticles
US9375388B2 (en) 2011-09-23 2016-06-28 Indian Institute Of Technology, Bombay Nanoparticle based cosmetic composition
JO3476B1 (en) 2011-09-26 2020-07-05 Novartis Ag Fusion proteins for treating metabolic disorders
US9458214B2 (en) 2011-09-26 2016-10-04 Novartis Ag Dual function fibroblast growth factor 21 proteins
EP2760477B1 (en) 2011-09-27 2018-08-08 Alnylam Pharmaceuticals, Inc. Di-aliphatic substituted pegylated lipids
WO2013045505A1 (en) 2011-09-28 2013-04-04 Novartis Ag Biomarkers for raas combination therapy
DE19216461T1 (en) 2011-10-03 2021-10-07 Modernatx, Inc. MODIFIED NUCLEOSIDES, NUCLEOTIDES AND NUCLEIC ACIDS AND USES THEREOF
RU2014118727A (en) 2011-10-11 2015-11-20 Новартис Аг RECOMBINANT SELF-REPLICING POLYCISTRON RNA MOLECULES
WO2013055971A1 (en) 2011-10-11 2013-04-18 Arizona Board Of Regents For And On Behalf Of Arizona State University Polymers for delivering a substance into a cell
WO2013055331A1 (en) 2011-10-12 2013-04-18 The Curators Of The University Of Missouri Pentablock polymers
US20190209690A9 (en) 2011-10-12 2019-07-11 The Johns Hopkins University Bioreducible Poly (Beta-Amino Ester)s For siRNA Delivery
CN109111523B (en) 2011-10-14 2022-06-07 诺华股份有限公司 Antibodies and methods for Wnt pathway related diseases
US20150272885A1 (en) 2011-10-14 2015-10-01 Stc.Unm Porous nanoparticle-supported lipid bilayers (protocells) for targeted delivery including transdermal delivery of cargo and methods thereof
PT3597644T (en) 2011-10-18 2021-11-03 Dicerna Pharmaceuticals Inc Amine cationic lipids and uses thereof
AU2012326010B2 (en) 2011-10-18 2017-05-11 Micell Technologies, Inc. Drug delivery medical device
JP2014532620A (en) 2011-10-20 2014-12-08 ノバルティス アーゲー Adjuvant-added influenza B virus vaccine for primary immunization in children
EA034964B1 (en) 2011-10-20 2020-04-13 Новартис Аг Biomarkers predictive of responsiveness to alpha 7 nicotinic acetylcholine receptor activator treatment
EP3915545A1 (en) 2011-10-25 2021-12-01 The University of British Columbia Limit size lipid nanoparticles and related methods
US20130110043A1 (en) 2011-10-26 2013-05-02 Nanopass Technologies Ltd. Microneedle Intradermal Drug Delivery Device with Auto-Disable Functionality
EP3574950B1 (en) 2011-10-27 2021-02-17 Sorrento Therapeutics, Inc. Transdermal delivery of high viscosity bioactive agents
CA3119789A1 (en) 2011-10-27 2013-05-02 Massachusetts Institute Of Technology Amino acid derivatives functionalized on the n-terminal capable of forming drug encapsulating microspheres
JP2014534864A (en) 2011-10-28 2014-12-25 プレサージュ バイオサイエンシズ,インコーポレイテッド Drug delivery method
WO2013062140A1 (en) 2011-10-28 2013-05-02 Kyoto University Method for efficiently inducing differentiation of pluripotent stem cells into hepatic lineage cells
KR101759694B1 (en) 2011-10-28 2017-07-19 인테그리티 바이오, 아이엔씨. Protein formulations containing amino acids
LT3091029T (en) 2011-10-31 2023-02-27 F. Hoffmann-La Roche Ag Anti-il13 antibody formulations
JP6143767B2 (en) 2011-10-31 2017-06-07 マリンクロッド エルエルシー Combination liposome composition for cancer treatment
US9579338B2 (en) 2011-11-04 2017-02-28 Nitto Denko Corporation Method of producing lipid nanoparticles for drug delivery
CN104039830A (en) 2011-11-04 2014-09-10 诺华股份有限公司 Low density lipoprotein-related protein 6 (lrp6) - half life extender constructs
JP6133883B2 (en) 2011-11-04 2017-05-24 日東電工株式会社 Method for producing lipid nanoparticles for drug delivery
US9872870B2 (en) 2011-11-04 2018-01-23 University Of Notre Dame Du Lac Multifunctional micellar nanoparticle-based drug and targeting agent system
SG11201401964TA (en) 2011-11-04 2014-05-29 Agency Science Tech & Res Self-assembled composite ultrasmall peptide-polymer hydrogels
US20130116408A1 (en) 2011-11-05 2013-05-09 Aura Biosciences, Inc. Virion Derived Protein Nanoparticles For Delivering Radioisotopes For The Diagnosis And Treatment Of Malignant And Systemic Disease And The Monitoring Of Therapy
US20130115247A1 (en) 2011-11-05 2013-05-09 Aura Biosciences, Inc. Virion Derived Protein Nanoparticles For Delivering Radioisotopes For The Diagnosis And Treatment Of Malignant And Systemic Disease And The Monitoring Of Therapy
EP2776022A1 (en) 2011-11-08 2014-09-17 Novartis Forschungsstiftung, Zweigniederlassung Friedrich Miescher Institute For Biomedical Research New treatment for neurodegenerative diseases
EP2776013B8 (en) 2011-11-08 2023-08-30 The Board of Trustees of the University of Arkansas Methods and compositions for x-ray induced release from ph sensitive liposomes
EP2776838A1 (en) 2011-11-08 2014-09-17 Novartis Forschungsstiftung, Zweigniederlassung Friedrich Miescher Institute For Biomedical Research Early diagnostic of neurodegenerative diseases
EP2776459A1 (en) 2011-11-08 2014-09-17 Novartis Forschungsstiftung, Zweigniederlassung Friedrich Miescher Institute For Biomedical Research Rod cell-specific promoter
WO2013070653A1 (en) 2011-11-09 2013-05-16 Board Of Trustees Michigan State University Metallic nanoparticle synthesis with carbohydrate capping agent
WO2013071047A1 (en) 2011-11-11 2013-05-16 Children's Medical Center Corporation Compositions and methods for in vitro transcription of rna
JP6232544B2 (en) 2011-11-11 2017-11-22 ヴァリエーション バイオテクノロジーズ インコーポレイテッド Compositions and methods for the treatment of cytomegalovirus
MX2014005548A (en) 2011-11-14 2014-08-21 Novartis Ag Immunogenic complexes of polyanionic carbomers and env polypeptides and methods of manufacture and use thereof.
EP2780017A1 (en) 2011-11-15 2014-09-24 Novartis AG Combination of a phosphoinositide 3-kinase inhibitor and a modulator of the janus kinase 2-signal transducer and activator of transcription 5 pathway
EP2790681B9 (en) 2011-11-18 2023-07-26 Regeneron Pharmaceuticals, Inc. Method of manufacturing an extended release pharmaceutical formulation comprising polymer coated protein microparticles using spray-drying
JP2015504430A (en) 2011-11-21 2015-02-12 ノバルティス アーゲー Methods of treating psoriatic arthritis (PSA) using IL-7 antagonists and PSA-responsive or non-responsive alleles
WO2013078199A2 (en) 2011-11-23 2013-05-30 Children's Medical Center Corporation Methods for enhanced in vivo delivery of synthetic, modified rnas
TR201111743A2 (en) 2011-11-28 2012-04-24 Nesl�Han G�Rsoy Reyhan Oil based nanocarrier systems for use in cancer treatment.
US20150037428A1 (en) 2011-11-29 2015-02-05 The University Of North Carolina At Chapel Hill Geometrically engineered particles and methods for modulating macrophage or immune responses
KR102143435B1 (en) 2011-11-30 2020-08-11 쓰리엠 이노베이티브 프로퍼티즈 컴파니 Microneedle device including a peptide therapeutic agent and an amino acid and methods of making and using the same
US9364549B2 (en) 2011-11-30 2016-06-14 Andreas Voigt Hydrophobic drug-delivery material, method for manufacturing thereof and methods for delivery of a drug-delivery composition
WO2013082529A1 (en) 2011-12-02 2013-06-06 Yale University Enzymatic synthesis of poly(amine-co-esters) and methods of use thereof for gene delivery
EP2785333A4 (en) 2011-12-02 2015-04-01 Pegasus Lab Inc Amphipathic lipid-based sustained release compositions
WO2013082590A1 (en) 2011-12-02 2013-06-06 Invivo Therapeutics Corporation Peg based hydrogel for peripheral nerve injury applications and compositions and method of use of synthetic hydrogel sealants
EP2788439A4 (en) 2011-12-05 2015-09-02 Nano Prec Medical Inc Device having titania nanotube membrane for drug delivery
US8497124B2 (en) 2011-12-05 2013-07-30 Factor Bioscience Inc. Methods and products for reprogramming cells to a less differentiated state
DK3260140T3 (en) 2011-12-05 2021-04-19 Factor Bioscience Inc METHODS AND CELL TRANSFER PROCEDURES
WO2013086373A1 (en) 2011-12-07 2013-06-13 Alnylam Pharmaceuticals, Inc. Lipids for the delivery of active agents
EP2788316B1 (en) 2011-12-07 2019-04-24 Alnylam Pharmaceuticals, Inc. Branched alkyl and cycloalkyl terminated biodegradable lipids for the delivery of active agents
ES2921724T1 (en) 2011-12-07 2022-08-31 Alnylam Pharmaceuticals Inc Biodegradable lipids for the administration of active agents
GB201121070D0 (en) 2011-12-07 2012-01-18 Isis Innovation composition for delivery of biotherapeutics
EP2787977A4 (en) 2011-12-09 2015-05-06 Univ California Liposomal drug encapsulation
WO2013086486A1 (en) 2011-12-09 2013-06-13 President And Fellows Of Harvard College Integrated human organ-on-chip microphysiological systems
WO2013086502A1 (en) 2011-12-09 2013-06-13 President And Fellows Of Harvard College Organ chips and uses thereof
US9839616B2 (en) 2011-12-12 2017-12-12 Kyowa Hakko Kirin Co., Ltd. Lipid nano particles comprising cationic lipid for drug delivery system
AU2012353463B2 (en) 2011-12-12 2017-08-24 Kyowa Hakko Kirin Co., Ltd. Lipid nanoparticles containing combinations of cationic lipids
CA2858884A1 (en) 2011-12-12 2013-06-20 The Trustees Of The University Of Pennsylvania Proteins comprising mrsa pbp2a and fragments thereof, nucleic acids encoding the same, and compositions and their use to prevent and treat mrsa infections
EP2604253A1 (en) 2011-12-13 2013-06-19 Otto Glatter Water-in-oil emulsions and methods for their preparation
CA2858315C (en) 2011-12-13 2020-05-12 Engeneic Molecular Delivery Pty Ltd Bacterially derived, intact minicells for delivery of therapeutic agents to brain tumors
US20150000936A1 (en) 2011-12-13 2015-01-01 Schlumberger Technology Corporation Energization of an element with a thermally expandable material
US20140378538A1 (en) 2011-12-14 2014-12-25 Moderma Therapeutics, Inc. Methods of responding to a biothreat
US20140343129A1 (en) 2011-12-14 2014-11-20 Moderna Therapeutics, Inc. Modified nucleic acids, and acute care uses thereof
US9636414B2 (en) 2011-12-15 2017-05-02 Biontech Ag Particles comprising single stranded RNA and double stranded RNA for immunomodulation
WO2013090897A1 (en) 2011-12-15 2013-06-20 The Trustees Of The University Of Pennsylvania Using adaptive immunity to detect drug resistance
WO2013090648A1 (en) 2011-12-16 2013-06-20 modeRNA Therapeutics Modified nucleoside, nucleotide, and nucleic acid compositions
KR20140103939A (en) 2011-12-16 2014-08-27 노파르티스 아게 Aerosolization apparatus for inhalation profile-independent drug delivery
RU2014129268A (en) 2011-12-16 2016-02-10 Аллерган, Инк. OPHTHALMIC COMPOSITIONS THAT CONTAIN GRAVITY POLYVINYL POLYVINYL PROCALT-POLYVINYL ACETATE-POLYETHYLENE Glycol copolymers
EP2791171A1 (en) 2011-12-16 2014-10-22 Synthon Biopharmaceuticals B.V. EXPRESSION OF SECRETORY IgA ANTIBODIES IN DUCKWEED
EP2791210A1 (en) 2011-12-16 2014-10-22 Massachusetts Institute of Technology Alpha-aminoamidine polymers and uses thereof
WO2013090601A2 (en) 2011-12-16 2013-06-20 Massachusetts Institute Of Technology Compact nanoparticles for biological applications
EP2794701B1 (en) 2011-12-19 2017-03-08 The University Of Sydney A peptide-hydrogel composite
US9241829B2 (en) 2011-12-20 2016-01-26 Abbott Medical Optics Inc. Implantable intraocular drug delivery apparatus, system and method
CA2859691A1 (en) 2011-12-21 2013-06-27 Moderna Therapeutics, Inc. Methods of increasing the viability or longevity of an organ or organ explant
WO2013096812A1 (en) 2011-12-23 2013-06-27 Genentech, Inc. Articles of manufacture and methods for co-administration of antibodies
US10814115B2 (en) 2011-12-27 2020-10-27 Massachusetts Institute Of Technology Microneedle devices and uses thereof
JP2015505309A (en) 2011-12-29 2015-02-19 ノバルティス アーゲー Adjuvanted combination of meningococcal factor H binding protein
WO2013101690A1 (en) 2011-12-29 2013-07-04 modeRNA Therapeutics Modified mrnas encoding cell-penetrating polypeptides
LT3421601T (en) 2011-12-30 2020-01-10 Cellscript, Llc MAKING AND USING IN VITRO-SYNTHESIZED ssRNA FOR INTRODUCING INTO MAMMALIAN CELLS TO INDUCE A BIOLOGICAL OR BIOCHEMICAL EFFECT
WO2013103842A1 (en) 2012-01-06 2013-07-11 Michigan Life Therapeutics, Llc Methods of reducing risk of cardiovascular disease
US20150030576A1 (en) 2012-01-10 2015-01-29 Moderna Therapeutics, Inc. Methods and compositions for targeting agents into and across the blood-brain barrier
KR20140129054A (en) 2012-01-26 2014-11-06 라이프 테크놀로지스 코포레이션 Methods for increasing the infectivity of viruses
KR20140128966A (en) 2012-01-26 2014-11-06 라이프 테크놀로지스 코포레이션 Methods for increasing the infectivity of viruses
WO2013113326A1 (en) 2012-01-31 2013-08-08 Curevac Gmbh Pharmaceutical composition comprising a polymeric carrier cargo complex and at least one protein or peptide antigen
EP2623121A1 (en) 2012-01-31 2013-08-07 Bayer Innovation GmbH Pharmaceutical composition comprising a polymeric carrier cargo complex and an antigen
WO2013113325A1 (en) 2012-01-31 2013-08-08 Curevac Gmbh Negatively charged nucleic acid comprising complexes for immunostimulation
KR102019297B1 (en) 2012-02-09 2019-09-06 라이프 테크놀로지스 코포레이션 Hydrophilic polymeric particles and methods for making same
US20140037573A1 (en) 2012-02-22 2014-02-06 Cerulean Pharma Inc. Conjugates, particles, compositions, and related methods
ES2745208T3 (en) 2012-02-22 2020-02-28 Univ Northwestern Nanostructures to treat cancers
US20130243867A1 (en) 2012-02-23 2013-09-19 University Of South Florida (A Florida Non-Profit Corporation) Micelle compositions and methods for their use
EP2819652A1 (en) 2012-02-27 2015-01-07 Epitarget As Use of a particulate immunomodulator in cancer therapy
WO2013129936A1 (en) 2012-02-27 2013-09-06 Epitarget As Use of an antibody and a particulate immunomodulator in therapy
WO2013130535A1 (en) 2012-02-27 2013-09-06 Newgen Biopharma Corporation Topical delivery of hormonal and non hormonal nano formulations, methods of making and using the same
CA2864177C (en) 2012-03-01 2019-11-26 Amgen Research (Munich) Gmbh Prolonged half-life albumin-binding protein fused bispecific antibodies
US20130236504A1 (en) 2012-03-06 2013-09-12 Medical University Of South Carolina Delivery System for Enhancing Drug Efficacy
DE112013001457T5 (en) 2012-03-13 2014-12-04 University Of Kwazulu-Natal Transdermal application system
US10322089B2 (en) 2012-03-14 2019-06-18 The Board Of Trustees Of The Leland Stanford Junior University Nanoparticles, nanoparticle delivery methods, and systems of delivery
CA2867381C (en) 2012-03-16 2016-09-20 The Johns Hopkins University Controlled release formulations for the delivery of hif-1 inhibitors
SG10201610401XA (en) 2012-03-16 2017-01-27 Merck Patent Gmbh Targeting aminoacid lipids
AU2013232300B2 (en) 2012-03-16 2015-12-17 The Johns Hopkins University Non-linear multiblock copolymer-drug conjugates for the delivery of active agents
WO2013142349A1 (en) 2012-03-23 2013-09-26 University Of Chicago Compositions and methods related to staphylococcal sbi
US9610346B2 (en) 2012-03-23 2017-04-04 International Aids Vaccine Initiative Recombinant viral vectors
WO2013148186A1 (en) 2012-03-26 2013-10-03 President And Fellows Of Harvard College Lipid-coated nucleic acid nanostructures of defined shape
WO2013143555A1 (en) 2012-03-26 2013-10-03 Biontech Ag Rna formulation for immunotherapy
AU2013242403B2 (en) 2012-03-27 2018-10-18 Curevac Ag Artificial nucleic acid molecules
WO2013143700A2 (en) 2012-03-27 2013-10-03 Curevac Gmbh Artificial nucleic acid molecules comprising a 5'top utr
DK2831241T3 (en) 2012-03-27 2017-11-06 Curevac Ag ARTIFICIAL NUCLEIC ACID MOLECULES FOR IMPROVED PROTEIN OR PEPTIDE EXPRESSION.
US9446132B2 (en) 2012-03-27 2016-09-20 Sima Therapeutics, Inc. Diether based biodegradable cationic lipids for siRNA delivery
CA2868030C (en) 2012-03-29 2021-05-25 Shire Human Genetic Therapies, Inc. Lipid-derived neutral nanoparticles
CN108949772A (en) 2012-04-02 2018-12-07 现代泰克斯公司 For generating the modification polynucleotides of biological agent relevant to human diseases and protein
US20140275229A1 (en) 2012-04-02 2014-09-18 Moderna Therapeutics, Inc. Modified polynucleotides encoding udp glucuronosyltransferase 1 family, polypeptide a1
AU2013243948A1 (en) 2012-04-02 2014-10-30 Moderna Therapeutics, Inc. Modified polynucleotides for the production of proteins associated with human disease
US20150050354A1 (en) 2012-04-02 2015-02-19 Moderna Therapeutics, Inc. Modified polynucleotides for the treatment of otic diseases and conditions
US9878056B2 (en) 2012-04-02 2018-01-30 Modernatx, Inc. Modified polynucleotides for the production of cosmetic proteins and peptides
US20150064115A1 (en) 2012-04-05 2015-03-05 University Of Florida Research Foundation, Inc. Neurophilic nanoparticles
JP6375289B2 (en) 2012-04-05 2018-08-15 マサチューセッツ インスティテュート オブ テクノロジー Immune stimulating composition and method of use thereof
WO2013152351A2 (en) 2012-04-06 2013-10-10 The Trustees Of Columbia University In The City Of New York Fusion polypeptides and methods of use thereof
ES2821098T3 (en) 2012-04-08 2021-04-23 Urogen Pharma Ltd Reversible thermal hydrogel preparations for use in the treatment of urothelial disorders
KR101607422B1 (en) 2012-04-11 2016-03-29 인터자인 테크놀로지스, 인코포레이티드 Block copolymers for stable micelles
EP2836234B1 (en) 2012-04-12 2019-06-12 Yale University Vehicles for controlled delivery of different pharmaceutical agents
WO2013154766A1 (en) 2012-04-13 2013-10-17 New York University Microrna control of ldl receptor pathway
US11001797B2 (en) 2012-04-13 2021-05-11 President And Fellows Of Harvard College Devices and methods for in vitro aerosol delivery
AU2012377385A1 (en) 2012-04-18 2014-01-23 Arrowhead Research Corporation Poly(acrylate) polymers for in vivo nucleic acid delivery
CA2870941C (en) 2012-04-19 2021-05-25 Sirna Therapeutics, Inc. Diester and triester based low molecular weight, biodegradable cationic lipids for oligonucleotide delivery
EP2841056A4 (en) 2012-04-23 2015-09-16 Massachusetts Inst Technology Stable layer-by-layer coated particles
AR090825A1 (en) 2012-04-25 2014-12-10 Regulus Therapeutics Inc MicroRNA COMPOSITE AND METHODS OF MODULATION OF miR-21 ACTIVITY
EP4008355A1 (en) 2012-05-03 2022-06-08 Kala Pharmaceuticals, Inc. Pharmaceutical nanoparticles showing improved mucosal transport
JP6392209B2 (en) 2012-05-04 2018-09-19 ザ・ジョンズ・ホプキンス・ユニバーシティー Lipid-based drug carriers for rapid permeation through the mucus lining
WO2013173657A1 (en) 2012-05-16 2013-11-21 Micell Technologies, Inc. Low burst sustained release lipophilic and biologic agent compositions
WO2013173582A1 (en) 2012-05-17 2013-11-21 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Hepatitis c virus neutralizing antibody
WO2013173693A1 (en) 2012-05-18 2013-11-21 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Nanoparticles with enhanced entry into cancer cells
WO2013177419A2 (en) 2012-05-23 2013-11-28 The Ohio State University Lipid nanoparticle compositions and methods of making and methods of using the same
US20150306249A1 (en) 2012-05-25 2015-10-29 Curevac Gmbh Reversible immobilization and/or controlled release of nucleic acid containing nanoparticles by (biodegradable) polymer coatings
ES2811798T3 (en) 2012-06-06 2021-03-15 Loma Vista Medical Inc Inflatable medical devices
BR112014029807A2 (en) 2012-06-08 2017-06-27 Ethris Gmbh pulmonary administration of messenger rna
CN104519915A (en) 2012-06-08 2015-04-15 夏尔人类遗传性治疗公司 Pulmonary delivery of mRNA to non-lung target cells
RS62288B9 (en) 2012-06-08 2021-12-31 Nitto Denko Corp Lipids for therapeutic agent delivery formulations
JP6275707B2 (en) 2012-06-20 2018-02-07 フランク・グー Mucoadhesive nanoparticle delivery system
WO2014014613A2 (en) 2012-06-20 2014-01-23 President And Fellows Of Harvard College Self-assembling peptides, peptide nanostructures and uses thereof
US9803010B2 (en) 2012-06-27 2017-10-31 Merck Sharp & Dohme Corp. Crystalline anti-human IL-23p19 antibodies
US9150841B2 (en) 2012-06-29 2015-10-06 Shire Human Genetic Therapies, Inc. Cells for producing recombinant iduronate-2-sulfatase
US9956291B2 (en) 2012-07-10 2018-05-01 Shaker A. Mousa Nanoformulation and methods of use of thyroid receptor beta1 agonists for liver targeting
WO2014014890A1 (en) 2012-07-16 2014-01-23 Nanoderm Sciences, Inc. Targeted therapeutic nanoparticles
EP2687251A1 (en) 2012-07-17 2014-01-22 Sanofi-Aventis Deutschland GmbH Drug delivery device
EP2687252A1 (en) 2012-07-17 2014-01-22 Sanofi-Aventis Deutschland GmbH Drug delivery device
CN112587658A (en) 2012-07-18 2021-04-02 博笛生物科技有限公司 Targeted immunotherapy for cancer
WO2014015334A1 (en) 2012-07-20 2014-01-23 Brown University System and methods for nanostructure protected delivery of treatment agent and selective release thereof
JP6596336B2 (en) 2012-07-24 2019-10-23 プレジデント アンド フェローズ オブ ハーバード カレッジ Self-assembly of nucleic acid nanostructures
WO2014015422A1 (en) 2012-07-27 2014-01-30 Ontario Institute For Cancer Research Cellulose-based nanoparticles for drug delivery
GB201213624D0 (en) 2012-07-27 2012-09-12 Univ Ulster The Method and system for production of conjugated nanoparticles
WO2014024193A1 (en) 2012-08-07 2014-02-13 Prodel Pharma Ltd. Compositions and methods for rapid transmucosal delivery of pharmaceutical ingredients
WO2014025795A1 (en) 2012-08-07 2014-02-13 Northeastern University Compositions for the delivery of rna and drugs into cells
US20140155861A1 (en) 2012-08-08 2014-06-05 Presage Biosciences, Inc. Extrusion methods and devices for drug delivery
CN104582747B (en) 2012-08-08 2016-12-21 南洋理工大学 For manufacturing the method for the hydrogel fines with living cells and for manufacturing the compositions of tissue engineering bracket
CN104736181A (en) 2012-08-10 2015-06-24 北德克萨斯大学健康科学中心 Drug delivery vehicle comprising conjugates between targeting polyamino acids and fatty acids
WO2014027006A1 (en) 2012-08-13 2014-02-20 Edko Pazarlama Tanitim Ticaret Limited Sirketi Bioadhesive formulations for use in drug delivery
CA2884870C (en) 2012-08-13 2022-03-29 Massachusetts Institute Of Technology Amine-containing lipidoids and uses thereof
EP2885419A4 (en) 2012-08-14 2016-05-25 Moderna Therapeutics Inc Enzymes and polymerases for the synthesis of rna
US9827321B2 (en) 2012-08-14 2017-11-28 The Trustees Of The University Of Pennsylvania Stabilizing shear-thinning hydrogels
WO2014026284A1 (en) 2012-08-14 2014-02-20 Froese Aaron Internal structured self assembling liposomes
CA2882248A1 (en) 2012-08-15 2014-02-20 The University Of Chicago Exosome-based therapeutics against neurodegenerative disorders
US10179134B2 (en) 2012-09-05 2019-01-15 Creighton University Polymeric nanoparticles in a thermosensitive gel for coital-independent vaginal prophylaxis of HIV
US8703197B2 (en) 2012-09-13 2014-04-22 International Business Machines Corporation Branched polyamines for delivery of biologically active materials
JP6356678B2 (en) 2012-09-17 2018-07-11 ファイザー・インク Method for producing therapeutic nanoparticles
WO2014047649A1 (en) 2012-09-24 2014-03-27 The Regents Of The University Of California Methods for arranging and packing nucleic acids for unusual resistance to nucleases and targeted delivery for gene therapy
CA2925687A1 (en) 2012-09-27 2014-04-03 The University Of North Carolina At Chapel Hill Lipid coated nanoparticles containing agents having low aqueous and lipid solubilities and methods thereof
WO2014053882A1 (en) 2012-10-04 2014-04-10 Centre National De La Recherche Scientifique Cell penetrating peptides for intracellular delivery of molecules
US20140100178A1 (en) 2012-10-04 2014-04-10 Aslam Ansari Composition and methods for site-specific drug delivery to treat malaria and other liver diseases
EP2716655A1 (en) 2012-10-04 2014-04-09 Institut Pasteur Neutralizing antibodies directed against Hepatitis C virus ectodomain glycoprotein E2
WO2014053879A1 (en) 2012-10-04 2014-04-10 Centre National De La Recherche Scientifique Cell penetrating peptides for intracellular delivery of molecules
WO2014053881A1 (en) 2012-10-04 2014-04-10 Centre National De La Recherche Scientifique Cell penetrating peptides for intracellular delivery of molecules
WO2014053880A1 (en) 2012-10-04 2014-04-10 Centre National De La Recherche Scientifique Cell penetrating peptides for intracellular delivery of molecules
CN104812372A (en) 2012-10-04 2015-07-29 约翰内斯堡金山大学 Liposomal medicine delivery system
EP2716689A1 (en) 2012-10-05 2014-04-09 National University of Ireland, Galway Polymer comprising a plurality of branches having at least one disulfide group and/or at least one vinyl group
WO2014064534A2 (en) 2012-10-05 2014-05-01 Chrontech Pharma Ab Injection needle, device, immunogenic compositions and method of use
US9931410B2 (en) 2012-10-09 2018-04-03 The Brigham And Women's Hospital, Inc. Nanoparticles for targeted delivery of multiple therapeutic agents and methods of use
US20140106260A1 (en) 2012-10-11 2014-04-17 The Trustees Of The University Of Pennsylvania Core-shell nanoparticulate compositions and methods
WO2014062697A2 (en) 2012-10-16 2014-04-24 Endocyte, Inc. Drug delivery conjugates containing unnatural amino acids and methods for using
WO2014063059A1 (en) 2012-10-18 2014-04-24 Rockefeller University (The) Broadly-neutralizing anti-hiv antibodies
EP2908864B1 (en) 2012-10-22 2019-12-11 Sabag-Rfa Ltd. Phosphate compounds for delivering therapeutic agents into living cells and cells nuclei
EP2912182B1 (en) * 2012-10-23 2021-12-08 Caris Science, Inc. Aptamers and uses thereof
WO2014066898A1 (en) 2012-10-26 2014-05-01 The Johns Hopkins University A layer-by-layer approach to co-deliver dna and sirna via aunps: a potential platform for modifying release kinetics
CA2889608A1 (en) 2012-10-26 2014-05-01 Nlife Therapeutics, S.L. Compositions and methods for selective delivery of oligonucleotide molecules to cell types
WO2014066912A1 (en) 2012-10-26 2014-05-01 Vanderbilt University Polymeric nanoparticles
WO2014067551A1 (en) 2012-10-29 2014-05-08 Technische Universität Dortmund T7 rna polymerase variants and methods of using the same
RU2711249C2 (en) 2012-11-01 2020-01-15 Фэктор Байосайенс Инк. Methods and products for expression of proteins in cells
WO2014071072A2 (en) 2012-11-02 2014-05-08 Pungente Michael D Novel cationic carotenoid-based lipids for cellular nucleic acid uptake
US10017767B2 (en) 2012-11-05 2018-07-10 Fondazione Centro San Raffaele Targets in multiple myeloma and other disorders
US9975916B2 (en) 2012-11-06 2018-05-22 President And Fellows Of Harvard College Compositions and methods relating to complex nucleic acid nanostructures
NZ707855A (en) 2012-11-06 2018-11-30 Rochal Ind Llc Delivery of biologically-active agents using volatile, hydrophobic solvents
EP2916874B1 (en) 2012-11-07 2018-08-29 Council of Scientific and Industrial Research Nanocomplex containing cationic peptide for biomolecule delivery
EP2916873B1 (en) 2012-11-07 2017-07-26 Council of Scientific & Industrial Research Nanocomplex containing amphipathic peptide useful for efficient transfection of biomolecules
TW201428101A (en) 2012-11-08 2014-07-16 Inviragen Inc Compositions, methods and uses for dengue virus serotype-4 constructs
KR20150082422A (en) 2012-11-08 2015-07-15 노보자임스 바이오파마 디케이 에이/에스 Albumin variants
CA2890471C (en) 2012-11-08 2021-07-27 Clearside Biomedical, Inc. Methods and devices for the treatment of ocular diseases in human subjects
JP6480338B2 (en) 2012-11-08 2019-03-06 セセン バイオ, インコーポレイテッド IL-6 antagonists and uses thereof
WO2014072468A1 (en) 2012-11-09 2014-05-15 Velin-Pharma A/S Compositions for pulmonary delivery
WO2014071963A1 (en) 2012-11-09 2014-05-15 Biontech Ag Method for cellular rna expression
AU2013343864B2 (en) 2012-11-09 2019-04-04 BioNTech SE Method for cellular RNA expression
US9200119B2 (en) 2012-11-09 2015-12-01 Momentive Performance Materials Inc. Silicon-containing zwitterionic linear copolymer composition
AU2013341711A1 (en) 2012-11-12 2015-05-21 Redwood Bioscience, Inc. Compounds and methods for producing a conjugate
US9833502B2 (en) 2012-11-12 2017-12-05 Genvec, Inc. Malaria antigens and methods of use
GB201220354D0 (en) 2012-11-12 2012-12-26 Medpharm Ltd Dermal compositions
WO2014078399A1 (en) 2012-11-13 2014-05-22 Baylor College Of Medicine Multi-arm biodegradable polymers for nucleic acid delivery
WO2014078636A1 (en) 2012-11-16 2014-05-22 President And Fellows Of Harvard College Nucleic acid hydrogel self-assembly
US9310374B2 (en) 2012-11-16 2016-04-12 Redwood Bioscience, Inc. Hydrazinyl-indole compounds and methods for producing a conjugate
EP2732825B1 (en) 2012-11-19 2015-07-01 Invivogen Conjugates of a TLR7 and/or TLR8 agonist and a TLR2 agonist
EP2919760A4 (en) 2012-11-19 2016-08-03 Technion Res & Dev Foundation Liposomes for in-vivo delivery
US20140141037A1 (en) 2012-11-20 2014-05-22 Novartis Ag Rsv f prefusion trimers
WO2014081849A1 (en) 2012-11-20 2014-05-30 Phasebio Pharmaceuticals, Inc. Formulations of active agents for sustained release
WO2014081299A1 (en) 2012-11-22 2014-05-30 Tagworks Pharmaceuticals B.V. Activatable liposomes
WO2014081300A1 (en) 2012-11-22 2014-05-30 Tagworks Pharmaceuticals B.V. Channel protein activatable liposomes
FI2922574T3 (en) 2012-11-22 2023-08-11 Tagworks Pharmaceuticals B V Chemically cleavable group
ES2921623T3 (en) * 2012-11-26 2022-08-30 Modernatx Inc terminally modified RNA
EP2931914A4 (en) 2012-12-13 2016-08-17 Moderna Therapeutics Inc Modified polynucleotides for altering cell phenotype
EP2931319B1 (en) 2012-12-13 2019-08-21 ModernaTX, Inc. Modified nucleic acid molecules and uses thereof
CA2897752A1 (en) 2013-01-10 2014-07-17 Novartis Ag Influenza virus immunogenic compositions and uses thereof
WO2014113089A2 (en) 2013-01-17 2014-07-24 Moderna Therapeutics, Inc. Signal-sensor polynucleotides for the alteration of cellular phenotypes
EP2964234A4 (en) 2013-03-09 2016-12-07 Moderna Therapeutics Inc Heterologous untranslated regions for mrna
EP2968397A4 (en) 2013-03-12 2016-12-28 Moderna Therapeutics Inc Diagnosis and treatment of fibrosis
US20160024181A1 (en) 2013-03-13 2016-01-28 Moderna Therapeutics, Inc. Long-lived polynucleotide molecules
EP2971010B1 (en) 2013-03-14 2020-06-10 ModernaTX, Inc. Formulation and delivery of modified nucleoside, nucleotide, and nucleic acid compositions
WO2014152027A1 (en) 2013-03-15 2014-09-25 Moderna Therapeutics, Inc. Manufacturing methods for production of rna transcripts
US20160032273A1 (en) 2013-03-15 2016-02-04 Moderna Therapeutics, Inc. Characterization of mrna molecules
EP2971161B1 (en) 2013-03-15 2018-12-26 ModernaTX, Inc. Ribonucleic acid purification
US8980864B2 (en) 2013-03-15 2015-03-17 Moderna Therapeutics, Inc. Compositions and methods of altering cholesterol levels
EP2971165A4 (en) 2013-03-15 2016-11-23 Moderna Therapeutics Inc Removal of dna fragments in mrna production process
US20160017313A1 (en) 2013-03-15 2016-01-21 Moderna Therapeutics, Inc. Analysis of mrna heterogeneity and stability
WO2014144767A1 (en) 2013-03-15 2014-09-18 Moderna Therapeutics, Inc. Ion exchange purification of mrna
HUE056760T2 (en) 2013-07-11 2022-03-28 Modernatx Inc Compositions comprising synthetic polynucleotides encoding crispr related proteins and synthetic sgrnas and methods of use
WO2015034925A1 (en) 2013-09-03 2015-03-12 Moderna Therapeutics, Inc. Circular polynucleotides
AU2014315287A1 (en) 2013-09-03 2015-03-12 Moderna Therapeutics, Inc. Chimeric polynucleotides
US9925277B2 (en) 2013-09-13 2018-03-27 Modernatx, Inc. Polynucleotide compositions containing amino acids
WO2015048744A2 (en) 2013-09-30 2015-04-02 Moderna Therapeutics, Inc. Polynucleotides encoding immune modulating polypeptides
EA201690675A1 (en) 2013-10-03 2016-08-31 Модерна Терапьютикс, Инк. POLYNUCLEOTES ENCODING THE RECEPTOR OF LOW DENSITY LIPOPROTEINS
WO2015058069A1 (en) 2013-10-18 2015-04-23 Moderna Therapeutics, Inc. Compositions and methods for tolerizing cellular systems
WO2015105926A1 (en) 2014-01-08 2015-07-16 Moderna Therapeutics, Inc. Polynucleotides for the in vivo production of antibodies
EP3159407A1 (en) * 2015-10-23 2017-04-26 Silence Therapeutics (London) Ltd Guide rnas, methods and uses
WO2017075038A1 (en) * 2015-10-26 2017-05-04 Rana Therapeutics, Inc. Nanoparticle formulations for delivery of nucleic acid complexes
BR112019025224A2 (en) 2017-05-31 2020-12-08 Ultragenyx Pharmaceutical Inc. THERAPEUTIC FOR TYPE III GLYCOGEN STORAGE DISEASE
SG11201911430PA (en) * 2017-07-04 2020-01-30 Curevac Ag Novel nucleic acid molecules
KR20220004648A (en) * 2019-03-28 2022-01-11 인텔리아 테라퓨틱스, 인크. Compositions and methods comprising a polynucleotide encoding a TTR guide RNA, and an RNA-guide DNA binding agent
CN116710079A (en) * 2020-07-24 2023-09-05 斯特兰德生物科技公司 Lipid nanoparticles comprising modified nucleotides

Patent Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6303378B1 (en) * 1997-10-24 2001-10-16 Valentis, Inc. Methods for preparing polynucleotide transfection complexes
US20020064517A1 (en) * 1998-04-30 2002-05-30 Stewart A. Cederholm-Williams Fibrin sealant as a transfection/transformation vehicle for gene therapy
US20020130430A1 (en) * 2000-12-29 2002-09-19 Castor Trevor Percival Methods for making polymer microspheres/nanospheres and encapsulating therapeutic proteins and other products
US20060018971A1 (en) * 2004-05-12 2006-01-26 Terrence Scott Nucleic acid microspheres, production and delivery thereof
US20090286852A1 (en) 2005-08-23 2009-11-19 Katalin Kariko RNA containing modified nucleosides and methods of use thereof
US20100015232A1 (en) * 2006-07-07 2010-01-21 Aarhus Universitet Nanoparticles for nucleic acid delivery
US20080274463A1 (en) * 2007-05-04 2008-11-06 Ventana Medical Systems, Inc. Method for quantifying biomolecules conjugated to a nanoparticle
US20090264511A1 (en) * 2007-12-10 2009-10-22 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of factor vii gene
US20110244026A1 (en) * 2009-12-01 2011-10-06 Braydon Charles Guild Delivery of mrna for the augmentation of proteins and enzymes in human genetic diseases
WO2011130624A2 (en) * 2010-04-16 2011-10-20 Immune Disease Institute, Inc. Sustained polypeptide expression from synthetic, modified rnas and uses thereof

Non-Patent Citations (8)

* Cited by examiner, † Cited by third party
Title
AKINC, NATURE BIOTECHNOLOGY, vol. 26, 2008, pages 561 - 569
BASHA ET AL.: "Infuence of Cationic lipid Composition on Gene Silencing Properties of Lipid Nanoparticle Formulations of siRNA in Antigen-Presenting Cells.", MOLECULAR THERAPY., vol. 19, no. 12, October 2011 (2011-10-01), pages 2186 - 2200, XP055024764 *
GREENE ET AL.: "Protective Groups in Organic Synthesis,", 1991, WILEY & SONS
GREENE, PROTECTIVE GROUPS IN ORGANIC SYNTHESIS
HEYES, J. CONTROL RELEASE, vol. 107, 2005, pages 276 - 287
LOVE, PNAS, vol. 107, 2010, pages 1864 - 1869
SEMPLE ET AL.: "Rational design of cationic lipids for siRNA delivery.", NATURE BIOTECHNOLOGY, vol. 28, no. 2, February 2010 (2010-02-01), pages 172 - 176+2PP, XP002633693 *
SEMPLE, NATURE BIOTECHNOLOGY, vol. 28, 2010, pages 172 - 176

Cited By (277)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11298426B2 (en) 2003-10-14 2022-04-12 BioNTech SE Recombinant vaccines and use thereof
US10106800B2 (en) 2005-09-28 2018-10-23 Biontech Ag Modification of RNA, producing an increased transcript stability and translation efficiency
US11141378B2 (en) 2008-04-15 2021-10-12 Arbutus Biopharma Corporation Lipid formulations for nucleic acid delivery
US11446383B2 (en) 2009-07-01 2022-09-20 Arbutus Biopharma Corporation Lipid formulations for delivery of therapeutic agents
US11786598B2 (en) 2009-07-01 2023-10-17 Arbutus Biopharma Corporation Lipid formulations for delivery of therapeutic agents
US11718852B2 (en) 2010-06-30 2023-08-08 Arbutus Biopharma Corporation Non-liposomal systems for nucleic acid delivery
US11690862B1 (en) 2010-07-06 2023-07-04 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11717529B2 (en) 2010-07-06 2023-08-08 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US20220125723A1 (en) 2010-07-06 2022-04-28 Glaxosmithkline Biologicals Sa Lipid formulations with viral immunogens
US11739334B2 (en) 2010-07-06 2023-08-29 Glaxosmithkline Biologicals Sa Immunisation of large mammals with low doses of RNA
US11730754B2 (en) 2010-07-06 2023-08-22 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11638693B2 (en) 2010-07-06 2023-05-02 Glaxosmithkline Biologicals Sa Vaccine for eliciting immune response comprising RNA encoding an immunogen and lipid formulations comprising mole percentage of lipids
US11638694B2 (en) 2010-07-06 2023-05-02 Glaxosmithkline Biologicals Sa Vaccine for eliciting immune response comprising lipid formulations and RNA encoding multiple immunogens
US11596645B2 (en) 2010-07-06 2023-03-07 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11655475B2 (en) 2010-07-06 2023-05-23 Glaxosmithkline Biologicals Sa Immunisation of large mammals with low doses of RNA
US11883534B2 (en) 2010-07-06 2024-01-30 Glaxosmithkline Biologicals Sa Immunisation with lipid formulations with RNA encoding immunogens
US11759475B2 (en) 2010-07-06 2023-09-19 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11766401B2 (en) 2010-07-06 2023-09-26 Glaxosmithkline Biologicals Sa Methods of administering lipid formulations with immunogens
US11773395B1 (en) 2010-07-06 2023-10-03 Glaxosmithkline Biologicals Sa Immunization of large mammals with low doses of RNA
US11905514B2 (en) 2010-07-06 2024-02-20 Glaxosmithkline Biological Sa Immunisation of large mammals with low doses of RNA
US11666534B2 (en) 2010-07-06 2023-06-06 Glaxosmithkline Biologicals Sa Methods of administering lipid formulations with viral immunogens
US11857562B2 (en) 2010-07-06 2024-01-02 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11891608B2 (en) 2010-07-06 2024-02-06 Glaxosmithkline Biologicals Sa Immunization of large mammals with low doses of RNA
US11707482B2 (en) 2010-07-06 2023-07-25 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11786467B2 (en) 2010-07-06 2023-10-17 Glaxosmithkline Biologicals Sa Lipid formulations with immunogens
US11839686B2 (en) 2010-07-06 2023-12-12 Glaxosmithkline Biologicals Sa Lipid formulations with viral immunogens
US11845925B2 (en) 2010-07-06 2023-12-19 Glaxosmithkline Biologicals Sa Immunisation of large mammals with low doses of RNA
US11690864B2 (en) 2010-07-06 2023-07-04 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11696923B2 (en) 2010-07-06 2023-07-11 Glaxosmithkline Biologicals, Sa Delivery of RNA to trigger multiple immune pathways
US11851660B2 (en) 2010-07-06 2023-12-26 Glaxosmithkline Biologicals Sa Immunisation of large mammals with low doses of RNA
US11690863B2 (en) 2010-07-06 2023-07-04 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11690865B2 (en) 2010-07-06 2023-07-04 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11865080B2 (en) 2010-07-06 2024-01-09 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11690861B2 (en) 2010-07-06 2023-07-04 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11850305B2 (en) 2010-07-06 2023-12-26 Glaxosmithkline Biologicals Sa Method of making lipid formulations with RNA encoding immunogens
US11857681B2 (en) 2010-07-06 2024-01-02 Glaxosmithkline Biologicals Sa Lipid formulations with RNA encoding immunogens
US11913001B2 (en) 2010-07-06 2024-02-27 Glaxosmithkline Biologicals Sa Immunisation of large mammals with low doses of RNA
US8822663B2 (en) 2010-08-06 2014-09-02 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US9181319B2 (en) 2010-08-06 2015-11-10 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US9447164B2 (en) 2010-08-06 2016-09-20 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US9937233B2 (en) 2010-08-06 2018-04-10 Modernatx, Inc. Engineered nucleic acids and methods of use thereof
US11759422B2 (en) 2010-08-31 2023-09-19 Glaxosmithkline Biologicals Sa Pegylated liposomes for delivery of immunogen-encoding RNA
US9334328B2 (en) 2010-10-01 2016-05-10 Moderna Therapeutics, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US9657295B2 (en) 2010-10-01 2017-05-23 Modernatx, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US10064959B2 (en) 2010-10-01 2018-09-04 Modernatx, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US11639370B2 (en) 2010-10-11 2023-05-02 Glaxosmithkline Biologicals Sa Antigen delivery platforms
US9950068B2 (en) 2011-03-31 2018-04-24 Modernatx, Inc. Delivery and formulation of engineered nucleic acids
US9533047B2 (en) 2011-03-31 2017-01-03 Modernatx, Inc. Delivery and formulation of engineered nucleic acids
US10898574B2 (en) 2011-03-31 2021-01-26 Modernatx, Inc. Delivery and formulation of engineered nucleic acids
US11911474B2 (en) 2011-03-31 2024-02-27 Modernatx, Inc. Delivery and formulation of engineered nucleic acids
US11248264B2 (en) 2011-05-24 2022-02-15 Tron-Translationale Onkologie An Der Universitätsmedizin Der Johannes Gutenberg-Universität Mainz Ggmbh Individualized vaccines for cancer
US10738355B2 (en) 2011-05-24 2020-08-11 Tron-Translationale Onkologie An Der Universitätsmedizin Der Johannes Gutenberg-Universität Mainz Ggmbh Individualized vaccines for cancer
US11547764B2 (en) 2011-06-08 2023-01-10 Translate Bio, Inc. Lipid nanoparticle compositions and methods for MRNA delivery
US11730825B2 (en) 2011-06-08 2023-08-22 Translate Bio, Inc. Lipid nanoparticle compositions and methods for mRNA delivery
US11896636B2 (en) 2011-07-06 2024-02-13 Glaxosmithkline Biologicals Sa Immunogenic combination compositions and uses thereof
US10022425B2 (en) 2011-09-12 2018-07-17 Modernatx, Inc. Engineered nucleic acids and methods of use thereof
US9464124B2 (en) 2011-09-12 2016-10-11 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US10751386B2 (en) 2011-09-12 2020-08-25 Modernatx, Inc. Engineered nucleic acids and methods of use thereof
EP3682905B1 (en) 2011-10-03 2021-12-01 ModernaTX, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US9428535B2 (en) 2011-10-03 2016-08-30 Moderna Therapeutics, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US9271996B2 (en) 2011-12-16 2016-03-01 Moderna Therapeutics, Inc. Formulation and delivery of PLGA microspheres
US9186372B2 (en) 2011-12-16 2015-11-17 Moderna Therapeutics, Inc. Split dose administration
EP2791160B1 (en) 2011-12-16 2022-03-02 ModernaTX, Inc. Modified mrna compositions
US9295689B2 (en) 2011-12-16 2016-03-29 Moderna Therapeutics, Inc. Formulation and delivery of PLGA microspheres
US11559587B2 (en) 2012-03-26 2023-01-24 Tron-Translationale Onkologie An Der Universitätsmedizin Der Johannes Gutenberg-Universität Mainz Ggmbh RNA formulation for immunotherapy
US10485884B2 (en) 2012-03-26 2019-11-26 Biontech Rna Pharmaceuticals Gmbh RNA formulation for immunotherapy
US9878056B2 (en) 2012-04-02 2018-01-30 Modernatx, Inc. Modified polynucleotides for the production of cosmetic proteins and peptides
US9107886B2 (en) 2012-04-02 2015-08-18 Moderna Therapeutics, Inc. Modified polynucleotides encoding basic helix-loop-helix family member E41
US8999380B2 (en) 2012-04-02 2015-04-07 Moderna Therapeutics, Inc. Modified polynucleotides for the production of biologics and proteins associated with human disease
US9814760B2 (en) 2012-04-02 2017-11-14 Modernatx, Inc. Modified polynucleotides for the production of biologics and proteins associated with human disease
US9827332B2 (en) 2012-04-02 2017-11-28 Modernatx, Inc. Modified polynucleotides for the production of proteins
US9782462B2 (en) 2012-04-02 2017-10-10 Modernatx, Inc. Modified polynucleotides for the production of proteins associated with human disease
EP2833921A2 (en) * 2012-04-02 2015-02-11 Moderna Therapeutics, Inc. Modified polynucleotides for the production of secreted proteins
WO2013151665A3 (en) * 2012-04-02 2014-02-20 modeRNA Therapeutics Modified polynucleotides for the production of proteins associated with human disease
US11564998B2 (en) 2012-04-02 2023-01-31 Modernatx, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
US10703789B2 (en) 2012-04-02 2020-07-07 Modernatx, Inc. Modified polynucleotides for the production of secreted proteins
US9675668B2 (en) 2012-04-02 2017-06-13 Moderna Therapeutics, Inc. Modified polynucleotides encoding hepatitis A virus cellular receptor 2
EP3978030A1 (en) * 2012-04-02 2022-04-06 ModernaTX, Inc. Modified polynucleotides for the production of proteins associated with human disease
US9050297B2 (en) 2012-04-02 2015-06-09 Moderna Therapeutics, Inc. Modified polynucleotides encoding aryl hydrocarbon receptor nuclear translocator
US10583203B2 (en) 2012-04-02 2020-03-10 Modernatx, Inc. In vivo production of proteins
US9587003B2 (en) 2012-04-02 2017-03-07 Modernatx, Inc. Modified polynucleotides for the production of oncology-related proteins and peptides
US10577403B2 (en) 2012-04-02 2020-03-03 Modernatx, Inc. Modified polynucleotides for the production of secreted proteins
US9061059B2 (en) 2012-04-02 2015-06-23 Moderna Therapeutics, Inc. Modified polynucleotides for treating protein deficiency
US9089604B2 (en) 2012-04-02 2015-07-28 Moderna Therapeutics, Inc. Modified polynucleotides for treating galactosylceramidase protein deficiency
US9572897B2 (en) 2012-04-02 2017-02-21 Modernatx, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
US10501513B2 (en) 2012-04-02 2019-12-10 Modernatx, Inc. Modified polynucleotides for the production of oncology-related proteins and peptides
US9572896B2 (en) 2012-04-02 2017-02-21 Modernatx, Inc. In vivo production of proteins
US10501512B2 (en) 2012-04-02 2019-12-10 Modernatx, Inc. Modified polynucleotides
US9828416B2 (en) 2012-04-02 2017-11-28 Modernatx, Inc. Modified polynucleotides for the production of secreted proteins
US10493167B2 (en) 2012-04-02 2019-12-03 Modernatx, Inc. In vivo production of proteins
US9301993B2 (en) 2012-04-02 2016-04-05 Moderna Therapeutics, Inc. Modified polynucleotides encoding apoptosis inducing factor 1
AU2017232121B2 (en) * 2012-04-02 2019-05-09 Modernatx, Inc. In vivo production of proteins
US9303079B2 (en) 2012-04-02 2016-04-05 Moderna Therapeutics, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
US9283287B2 (en) 2012-04-02 2016-03-15 Moderna Therapeutics, Inc. Modified polynucleotides for the production of nuclear proteins
US9255129B2 (en) 2012-04-02 2016-02-09 Moderna Therapeutics, Inc. Modified polynucleotides encoding SIAH E3 ubiquitin protein ligase 1
EP3501550A1 (en) * 2012-04-02 2019-06-26 Moderna Therapeutics, Inc. Modified polynucleotides for the production of proteins associated with human disease
EP3505176A1 (en) * 2012-04-02 2019-07-03 Moderna Therapeutics, Inc. Modified polynucleotides for the production of secreted proteins
US9254311B2 (en) 2012-04-02 2016-02-09 Moderna Therapeutics, Inc. Modified polynucleotides for the production of proteins
US9233141B2 (en) 2012-04-02 2016-01-12 Moderna Therapeutics, Inc. Modified polynucleotides for the production of proteins associated with blood and lymphatic disorders
US9220755B2 (en) 2012-04-02 2015-12-29 Moderna Therapeutics, Inc. Modified polynucleotides for the production of proteins associated with blood and lymphatic disorders
US9220792B2 (en) 2012-04-02 2015-12-29 Moderna Therapeutics, Inc. Modified polynucleotides encoding aquaporin-5
EP3520821A1 (en) * 2012-04-02 2019-08-07 Moderna Therapeutics, Inc. Modified polynucleotides for the production of biologics and proteins associated with human disease
US9221891B2 (en) 2012-04-02 2015-12-29 Moderna Therapeutics, Inc. In vivo production of proteins
US10385106B2 (en) 2012-04-02 2019-08-20 Modernatx, Inc. Modified polynucleotides for the production of secreted proteins
US9216205B2 (en) 2012-04-02 2015-12-22 Moderna Therapeutics, Inc. Modified polynucleotides encoding granulysin
US9192651B2 (en) 2012-04-02 2015-11-24 Moderna Therapeutics, Inc. Modified polynucleotides for the production of secreted proteins
US10772975B2 (en) 2012-04-02 2020-09-15 Modernatx, Inc. Modified Polynucleotides for the production of biologics and proteins associated with human disease
US9149506B2 (en) 2012-04-02 2015-10-06 Moderna Therapeutics, Inc. Modified polynucleotides encoding septin-4
US9114113B2 (en) 2012-04-02 2015-08-25 Moderna Therapeutics, Inc. Modified polynucleotides encoding citeD4
US9095552B2 (en) 2012-04-02 2015-08-04 Moderna Therapeutics, Inc. Modified polynucleotides encoding copper metabolism (MURR1) domain containing 1
US10463751B2 (en) 2012-04-02 2019-11-05 Modernatx, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
US10758592B2 (en) 2012-10-09 2020-09-01 Sanofi Exendin-4 derivatives as dual GLP1/glucagon agonists
WO2014081507A1 (en) 2012-11-26 2014-05-30 Moderna Therapeutics, Inc. Terminally modified rna
EP2922554B1 (en) 2012-11-26 2022-02-23 ModernaTX, Inc. Terminally modified rna
AU2017202228B2 (en) * 2012-11-26 2019-03-14 Modernatx, Inc. Terminally modified RNA
US10925935B2 (en) 2012-11-26 2021-02-23 Modernatx, Inc. Terminally Modified RNA
US10155029B2 (en) 2012-11-26 2018-12-18 Modernatx, Inc. Terminally modified RNA
US9597380B2 (en) 2012-11-26 2017-03-21 Modernatx, Inc. Terminally modified RNA
EP4074834A1 (en) 2012-11-26 2022-10-19 ModernaTX, Inc. Terminally modified rna
US11504419B2 (en) 2012-11-28 2022-11-22 BioNTech SE Individualized vaccines for cancer
US10155031B2 (en) 2012-11-28 2018-12-18 Biontech Rna Pharmaceuticals Gmbh Individualized vaccines for cancer
EP2931319A4 (en) * 2012-12-13 2016-05-04 Moderna Therapeutics Inc Modified nucleic acid molecules and uses thereof
US9670261B2 (en) 2012-12-21 2017-06-06 Sanofi Functionalized exendin-4 derivatives
US9745360B2 (en) 2012-12-21 2017-08-29 Sanofi Dual GLP1/GIP or trigonal GLP1/GIP/glucagon agonists
US10253079B2 (en) 2012-12-21 2019-04-09 Sanofi Functionalized Exendin-4 derivatives
US11708396B2 (en) 2013-01-17 2023-07-25 Modernatx, Inc. Signal-sensor polynucleotides for the alteration of cellular phenotypes
WO2014113089A2 (en) 2013-01-17 2014-07-24 Moderna Therapeutics, Inc. Signal-sensor polynucleotides for the alteration of cellular phenotypes
US10525075B2 (en) 2013-02-22 2020-01-07 The Board Of Trustees Of The Leland Stanford Junior University Compounds, compositions, methods, and kits relating to telomere extension
US11007210B2 (en) 2013-02-22 2021-05-18 The Board Of Trustees Of The Leland Stanford Junior University Compounds, compositions, methods, and kits relating to telomere extension
US11872243B2 (en) 2013-02-22 2024-01-16 The Board Of Trustees Of The Leland Stanford Junior University Compounds, compositions, methods, and kits relating to telomere extension
US10258698B2 (en) 2013-03-14 2019-04-16 Modernatx, Inc. Formulation and delivery of modified nucleoside, nucleotide, and nucleic acid compositions
US11845772B2 (en) 2013-03-15 2023-12-19 Modernatx, Inc. Ribonucleic acid purification
US8980864B2 (en) 2013-03-15 2015-03-17 Moderna Therapeutics, Inc. Compositions and methods of altering cholesterol levels
US10138507B2 (en) 2013-03-15 2018-11-27 Modernatx, Inc. Manufacturing methods for production of RNA transcripts
US11377470B2 (en) 2013-03-15 2022-07-05 Modernatx, Inc. Ribonucleic acid purification
US10590161B2 (en) 2013-03-15 2020-03-17 Modernatx, Inc. Ion exchange purification of mRNA
US10077439B2 (en) 2013-03-15 2018-09-18 Modernatx, Inc. Removal of DNA fragments in mRNA production process
US10858647B2 (en) 2013-03-15 2020-12-08 Modernatx, Inc. Removal of DNA fragments in mRNA production process
US11222711B2 (en) 2013-05-10 2022-01-11 BioNTech SE Predicting immunogenicity of T cell epitopes
EP3971287A1 (en) 2013-07-11 2022-03-23 ModernaTX, Inc. Compositions comprising synthetic polynucleotides encoding crispr related proteins and synthetic sgrnas and methods of use
EP3019619B1 (en) 2013-07-11 2021-08-25 ModernaTX, Inc. Compositions comprising synthetic polynucleotides encoding crispr related proteins and synthetic sgrnas and methods of use
US11027025B2 (en) 2013-07-11 2021-06-08 Modernatx, Inc. Compositions comprising synthetic polynucleotides encoding CRISPR related proteins and synthetic sgRNAs and methods of use
JP7136826B2 (en) 2013-07-11 2022-09-13 モデルナティエックス インコーポレイテッド Compositions Comprising Synthetic Polynucleotides Encoding CRISPR-Related Proteins and Synthetic sgRNAs and Methods of Use
WO2015006747A2 (en) 2013-07-11 2015-01-15 Moderna Therapeutics, Inc. Compositions comprising synthetic polynucleotides encoding crispr related proteins and synthetic sgrnas and methods of use.
JP2020108379A (en) * 2013-07-11 2020-07-16 モデルナティエックス インコーポレイテッドModernaTX,Inc. COMPOSITIONS COMPRISING SYNTHETIC POLYNUCLEOTIDES ENCODING CRISPR RELATED PROTEINS AND SYNTHETIC sgRNAS, AND METHODS OF USE
WO2015034928A1 (en) 2013-09-03 2015-03-12 Moderna Therapeutics, Inc. Chimeric polynucleotides
WO2015034925A1 (en) 2013-09-03 2015-03-12 Moderna Therapeutics, Inc. Circular polynucleotides
US10385088B2 (en) 2013-10-02 2019-08-20 Modernatx, Inc. Polynucleotide molecules and uses thereof
US10323076B2 (en) 2013-10-03 2019-06-18 Modernatx, Inc. Polynucleotides encoding low density lipoprotein receptor
WO2015051214A1 (en) 2013-10-03 2015-04-09 Moderna Therapeutics, Inc. Polynucleotides encoding low density lipoprotein receptor
US10202601B2 (en) 2013-11-22 2019-02-12 Mina Therapeutics Limited C/EBPα short activating RNA compositions and methods of use
WO2015075557A2 (en) 2013-11-22 2015-05-28 Mina Alpha Limited C/ebp alpha compositions and methods of use
EP3594348A1 (en) 2013-11-22 2020-01-15 Mina Therapeutics Limited C/ebp alpha short activating rna compositions and methods of use
EP3985118A1 (en) 2013-11-22 2022-04-20 MiNA Therapeutics Limited C/ebp alpha short activating rna compositions and methods of use
US10633659B2 (en) 2013-11-22 2020-04-28 Mina Therapeutics Limited C/EBPα short activating RNA compositions and methods of use
EP3053585A1 (en) * 2013-12-13 2016-08-10 Moderna Therapeutics, Inc. Alternative nucleic acid molecules and uses thereof
EP3080289A4 (en) * 2013-12-13 2017-11-15 Moderna Therapeutics, Inc. Modified nucleic acid molecules and uses thereof
US9694053B2 (en) 2013-12-13 2017-07-04 Sanofi Dual GLP-1/glucagon receptor agonists
US9750788B2 (en) 2013-12-13 2017-09-05 Sanofi Non-acylated exendin-4 peptide analogues
US9789165B2 (en) 2013-12-13 2017-10-17 Sanofi Exendin-4 peptide analogues as dual GLP-1/GIP receptor agonists
US9751926B2 (en) 2013-12-13 2017-09-05 Sanofi Dual GLP-1/GIP receptor agonists
RU2714404C2 (en) * 2014-01-31 2020-02-14 Фэктор Байосайенс Инк. Methods and products for producing and delivering nucleic acids
EP3699274A1 (en) * 2014-03-24 2020-08-26 Translate Bio, Inc. Mrna therapy for the treatment of ocular diseases
EP3122878B1 (en) 2014-03-24 2018-10-24 Translate Bio, Inc. Mrna therapy for the treatment of ocular diseases
US9771406B2 (en) 2014-04-07 2017-09-26 Sanofi Peptidic dual GLP-1/glucagon receptor agonists derived from exendin-4
US9758561B2 (en) 2014-04-07 2017-09-12 Sanofi Dual GLP-1/glucagon receptor agonists derived from exendin-4
US9775904B2 (en) 2014-04-07 2017-10-03 Sanofi Exendin-4 derivatives as peptidic dual GLP-1/glucagon receptor agonists
EP3134131B1 (en) 2014-04-23 2021-12-22 ModernaTX, Inc. Nucleic acid vaccines
US10709779B2 (en) 2014-04-23 2020-07-14 Modernatx, Inc. Nucleic acid vaccines
US9932381B2 (en) 2014-06-18 2018-04-03 Sanofi Exendin-4 derivatives as selective glucagon receptor agonists
US10286086B2 (en) 2014-06-19 2019-05-14 Modernatx, Inc. Alternative nucleic acid molecules and uses thereof
US10407683B2 (en) 2014-07-16 2019-09-10 Modernatx, Inc. Circular polynucleotides
EP4159741A1 (en) 2014-07-16 2023-04-05 ModernaTX, Inc. Method for producing a chimeric polynucleotide encoding a polypeptide having a triazole-containing internucleotide linkage
WO2016014846A1 (en) 2014-07-23 2016-01-28 Moderna Therapeutics, Inc. Modified polynucleotides for the production of intrabodies
EP3177732A4 (en) * 2014-08-08 2018-04-25 ModernaTX, Inc. Compositions and methods for the treatment of ophthalmic diseases and conditions
US11173120B2 (en) 2014-09-25 2021-11-16 Biontech Rna Pharmaceuticals Gmbh Stable formulations of lipids and liposomes
US10413598B2 (en) 2014-11-12 2019-09-17 Ucl Business Plc Factor IX gene therapy
US11344608B2 (en) 2014-11-12 2022-05-31 Ucl Business Ltd Factor IX gene therapy
EP4023755B1 (en) 2014-12-12 2023-04-26 CureVac SE Artificial nucleic acid molecules for improved protein expression
US11761009B2 (en) 2014-12-12 2023-09-19 CureVac SE Artificial nucleic acid molecules for improved protein expression
US11156617B2 (en) 2015-02-12 2021-10-26 BioNTech RNA Pharmaceuticals GbmH Predicting T cell epitopes useful for vaccination
US10806797B2 (en) 2015-06-05 2020-10-20 Sanofi Prodrugs comprising an GLP-1/glucagon dual agonist linker hyaluronic acid conjugate
US9982029B2 (en) 2015-07-10 2018-05-29 Sanofi Exendin-4 derivatives as selective peptidic dual GLP-1/glucagon receptor agonists
US11364292B2 (en) 2015-07-21 2022-06-21 Modernatx, Inc. CHIKV RNA vaccines
EP3324979B1 (en) 2015-07-21 2022-10-12 ModernaTX, Inc. Infectious disease vaccines
US11434486B2 (en) 2015-09-17 2022-09-06 Modernatx, Inc. Polynucleotides containing a morpholino linker
US11590157B2 (en) * 2015-10-05 2023-02-28 Modernatx, Inc. Methods for therapeutic administration of messenger ribonucleic acid drugs
WO2017062513A1 (en) 2015-10-05 2017-04-13 Modernatx, Inc. Methods for therapeutic administration of messenger ribonucleic acid drugs
US20230338410A1 (en) * 2015-10-05 2023-10-26 Modernatx, Inc. Methods for therapeutic administration of messenger ribonucleic acid drugs
US20180256628A1 (en) * 2015-10-05 2018-09-13 Modernatx, Inc. Methods for therapeutic administration of messenger ribonucleic acid drugs
EP3359670B1 (en) 2015-10-05 2020-05-13 Modernatx, Inc. Methods for therapeutic administration of messenger ribonucleic acid drugs
US10849920B2 (en) 2015-10-05 2020-12-01 Modernatx, Inc. Methods for therapeutic administration of messenger ribonucleic acid drugs
US11492628B2 (en) 2015-10-07 2022-11-08 BioNTech SE 3′-UTR sequences for stabilization of RNA
US11643441B1 (en) 2015-10-22 2023-05-09 Modernatx, Inc. Nucleic acid vaccines for varicella zoster virus (VZV)
EP3365007A4 (en) * 2015-10-22 2019-07-03 ModernaTX, Inc. Broad spectrum influenza virus vaccine
EP3718565A1 (en) * 2015-10-22 2020-10-07 ModernaTX, Inc. Respiratory virus vaccines
EP3365009A4 (en) * 2015-10-22 2019-07-03 ModernaTX, Inc. Herpes simplex virus vaccine
EP3364983A4 (en) * 2015-10-22 2019-10-23 ModernaTX, Inc. Respiratory virus vaccines
US11278611B2 (en) 2015-10-22 2022-03-22 Modernatx, Inc. Zika virus RNA vaccines
US10675342B2 (en) 2015-10-22 2020-06-09 Modernatx, Inc. Chikungunya virus RNA vaccines
US11235052B2 (en) 2015-10-22 2022-02-01 Modernatx, Inc. Chikungunya virus RNA vaccines
EP3364981A4 (en) * 2015-10-22 2019-08-07 ModernaTX, Inc. Human cytomegalovirus vaccine
EP4011451A1 (en) * 2015-10-22 2022-06-15 ModernaTX, Inc. Metapneumovirus mrna vaccines
EP3718565B1 (en) 2015-10-22 2022-04-27 ModernaTX, Inc. Respiratory virus vaccines
EP3364980A4 (en) * 2015-10-22 2019-07-10 ModernaTX, Inc. Nucleic acid vaccines for varicella zoster virus (vzv)
EP3364950A4 (en) * 2015-10-22 2019-10-23 ModernaTX, Inc. Tropical disease vaccines
US11872278B2 (en) 2015-10-22 2024-01-16 Modernatx, Inc. Combination HMPV/RSV RNA vaccines
EP3364982A4 (en) * 2015-10-22 2019-04-17 ModernaTX, Inc. Sexually transmitted disease vaccines
US10933127B2 (en) 2015-10-22 2021-03-02 Modernatx, Inc. Betacoronavirus mRNA vaccine
EP3369816A4 (en) * 2015-10-30 2019-06-26 Bonac Corporation Composition stably containing single-stranded nucleic acid molecule that suppresses expression of tgf-beta1 gene
US10751426B2 (en) 2015-10-30 2020-08-25 Bonac Corporation Composition stably containing single-stranded nucleic acid molecule that suppresses expression of TGF-β1 gene
WO2017112943A1 (en) 2015-12-23 2017-06-29 Modernatx, Inc. Methods of using ox40 ligand encoding polynucleotides
EP4039699A1 (en) 2015-12-23 2022-08-10 ModernaTX, Inc. Methods of using ox40 ligand encoding polynucleotides
WO2017120612A1 (en) 2016-01-10 2017-07-13 Modernatx, Inc. Therapeutic mrnas encoding anti ctla-4 antibodies
US11564997B2 (en) 2016-06-29 2023-01-31 Crispr Therapeutics Ag Materials and methods for treatment of friedreich ataxia and other related disorders
WO2018009838A1 (en) 2016-07-07 2018-01-11 Rubius Therapeutics, Inc. Compositions and methods related to therapeutic cell systems expressing exogenous rna
US11878060B2 (en) 2016-08-07 2024-01-23 Novartis Ag mRNA-mediated immunization methods
CN109562153A (en) * 2016-08-07 2019-04-02 诺华股份有限公司 The immunization method that mRNA is mediated
CN106047879A (en) * 2016-08-18 2016-10-26 广州市锐博生物科技有限公司 Oligonucleotide molecule used for inhibiting expression of mRNA of target gene and composition set thereof
US11559588B2 (en) 2017-02-22 2023-01-24 Crispr Therapeutics Ag Materials and methods for treatment of Spinocerebellar Ataxia Type 1 (SCA1) and other Spinocerebellar Ataxia Type 1 Protein (ATXN1) gene related conditions or disorders
US11920148B2 (en) 2017-02-22 2024-03-05 Crispr Therapeutics Ag Compositions and methods for gene editing
US11407997B2 (en) 2017-02-22 2022-08-09 Crispr Therapeutics Ag Materials and methods for treatment of primary hyperoxaluria type 1 (PH1) and other alanine-glyoxylate aminotransferase (AGXT) gene related conditions or disorders
WO2018213731A1 (en) 2017-05-18 2018-11-22 Modernatx, Inc. Polynucleotides encoding tethered interleukin-12 (il12) polypeptides and uses thereof
EP4219715A2 (en) 2017-09-08 2023-08-02 MiNA Therapeutics Limited Stabilized cebpa sarna compositions and methods of use
EP4183882A1 (en) 2017-09-08 2023-05-24 MiNA Therapeutics Limited Stabilized hnf4a sarna compositions and methods of use
EP4233880A2 (en) 2017-09-08 2023-08-30 MiNA Therapeutics Limited Hnf4a sarna compositions and methods of use
WO2019048631A1 (en) 2017-09-08 2019-03-14 Mina Therapeutics Limited Hnf4a sarna compositions and methods of use
WO2019048632A1 (en) 2017-09-08 2019-03-14 Mina Therapeutics Limited Stabilized hnf4a sarna compositions and methods of use
US11207398B2 (en) 2017-09-14 2021-12-28 Modernatx, Inc. Zika virus mRNA vaccines
WO2019158720A1 (en) 2018-02-16 2019-08-22 Mina Therapeutics Limited C/ebp alpha sarna compositions and methods of use
EP4242307A2 (en) 2018-04-12 2023-09-13 MiNA Therapeutics Limited Sirt1-sarna compositions and methods of use
WO2019197845A1 (en) 2018-04-12 2019-10-17 Mina Therapeutics Limited Sirt1-sarna compositions and methods of use
WO2019239144A1 (en) 2018-06-15 2019-12-19 Mina Therapeutics Limited Combination therapies comprising c/ebp alpha sarna
US10842885B2 (en) 2018-08-20 2020-11-24 Ucl Business Ltd Factor IX encoding nucleotides
US11517631B2 (en) 2018-08-20 2022-12-06 Ucl Business Ltd Factor IX encoding nucleotides
US11913012B2 (en) 2018-08-30 2024-02-27 Tenaya Therapeutics, Inc. Cardiac cell reprogramming with myocardin and ASCL1
US11015211B2 (en) 2018-08-30 2021-05-25 Tenaya Therapeutics, Inc. Cardiac cell reprogramming with myocardin and ASCL1
US11008587B2 (en) 2018-09-07 2021-05-18 Crispr Therapeutics Ag Universal donor cells
US10865424B2 (en) 2018-09-07 2020-12-15 Crispr Therapeutics Ag Universal donor cells
US11008586B2 (en) 2018-09-07 2021-05-18 Crispr Therapeutics Ag Universal donor cells
US10724052B2 (en) 2018-09-07 2020-07-28 Crispr Therapeutics Ag Universal donor cells
US11180776B1 (en) 2018-09-07 2021-11-23 Crispr Therapeutics Ag Universal donor cells
WO2020056155A3 (en) * 2018-09-13 2020-04-16 Modernatx, Inc. Polynucleotides encoding branched-chain alpha-ketoacid dehydrogenase complex e1-alpha, e1-beta, and e2 subunits for the treatment of maple syrup urine disease
WO2020208361A1 (en) 2019-04-12 2020-10-15 Mina Therapeutics Limited Sirt1-sarna compositions and methods of use
WO2021007515A1 (en) 2019-07-11 2021-01-14 Tenaya Therapeutics, Inc. Cardiac cell reprogramming with micrornas and other factors
US11926817B2 (en) 2019-08-09 2024-03-12 Nutcracker Therapeutics, Inc. Microfluidic apparatus and methods of use thereof
WO2021032777A1 (en) 2019-08-19 2021-02-25 Mina Therapeutics Limited Oligonucleotide conjugate compositions and methods of use
US11433103B2 (en) 2019-09-05 2022-09-06 Crispr Therapeutics Ag Universal donor cells
US11116797B2 (en) 2019-09-05 2021-09-14 Crispr Therapeutics Ag Universal donor cells
US11434505B2 (en) 2019-09-05 2022-09-06 Crispr Therapeutics Ag Universal donor cells
US11116798B2 (en) 2019-09-05 2021-09-14 Crispr Therapeutics Ag Universal donor cells
US11104918B2 (en) 2019-09-05 2021-08-31 Crispr Therapeutics Ag Universal donor cells
US11118196B2 (en) 2019-09-05 2021-09-14 Crispr Therapeutics Ag Universal donor cells
US11118195B2 (en) 2019-09-05 2021-09-14 Crispr Therapeutics Ag Universal donor cells
WO2021178246A1 (en) 2020-03-02 2021-09-10 Tenaya Therapeutics, Inc. Gene vector control by cardiomyocyte-expressed micrornas
US11779659B2 (en) 2020-04-22 2023-10-10 BioNTech SE RNA constructs and uses thereof
US11951185B2 (en) 2020-04-22 2024-04-09 BioNTech SE RNA constructs and uses thereof
US11925694B2 (en) 2020-04-22 2024-03-12 BioNTech SE Coronavirus vaccine
US11547673B1 (en) 2020-04-22 2023-01-10 BioNTech SE Coronavirus vaccine
WO2022113056A1 (en) 2020-11-30 2022-06-02 Crispr Therapeutics Ag Gene-edited natural killer cells
US11578309B2 (en) 2020-12-31 2023-02-14 Crispr Therapeutics Ag Universal donor cells
US11566230B2 (en) 2020-12-31 2023-01-31 Crispr Therapeutics Ag Universal donor cells
WO2022200810A1 (en) 2021-03-26 2022-09-29 Mina Therapeutics Limited Tmem173 sarna compositions and methods of use
WO2022229644A1 (en) 2021-04-28 2022-11-03 Mina Therapeutics Limited Combination therapies comprising c/ebp alpha sarna
WO2023031394A1 (en) 2021-09-03 2023-03-09 CureVac SE Novel lipid nanoparticles for delivery of nucleic acids
WO2023069498A1 (en) 2021-10-22 2023-04-27 Senda Biosciences, Inc. Mrna vaccine composition
WO2023073228A1 (en) 2021-10-29 2023-05-04 CureVac SE Improved circular rna for expressing therapeutic proteins
WO2023096858A1 (en) 2021-11-23 2023-06-01 Senda Biosciences, Inc. A bacteria-derived lipid composition and use thereof
WO2023099884A1 (en) 2021-12-01 2023-06-08 Mina Therapeutics Limited Pax6 sarna compositions and methods of use
WO2023122080A1 (en) 2021-12-20 2023-06-29 Senda Biosciences, Inc. Compositions comprising mrna and lipid reconstructed plant messenger packs
WO2023144330A1 (en) 2022-01-28 2023-08-03 CureVac SE Nucleic acid encoded transcription factor inhibitors
WO2023170435A1 (en) 2022-03-07 2023-09-14 Mina Therapeutics Limited Il10 sarna compositions and methods of use
WO2023227608A1 (en) 2022-05-25 2023-11-30 Glaxosmithkline Biologicals Sa Nucleic acid based vaccine encoding an escherichia coli fimh antigenic polypeptide
US11878055B1 (en) 2022-06-26 2024-01-23 BioNTech SE Coronavirus vaccine
US11951180B2 (en) 2023-04-03 2024-04-09 Translate Bio, Inc. Lipid nanoparticle compositions and methods for MRNA delivery
US11951181B2 (en) 2023-04-03 2024-04-09 Translate Bio, Inc. Lipid nanoparticle compositions and methods for mRNA delivery
US11951179B2 (en) 2023-04-03 2024-04-09 Translate Bio, Inc. Lipid nanoparticle compositions and methods for MRNA delivery

Also Published As

Publication number Publication date
ZA201403783B (en) 2015-09-30
RS63244B1 (en) 2022-06-30
US20130245106A1 (en) 2013-09-19
SI2791160T1 (en) 2022-07-29
IL232749A0 (en) 2014-08-03
CA2859387A1 (en) 2013-06-20
US20130237592A1 (en) 2013-09-12
EP2791160B1 (en) 2022-03-02
ES2923757T3 (en) 2022-09-30
EP2791160A4 (en) 2016-01-06
JP2015501844A (en) 2015-01-19
US9295689B2 (en) 2016-03-29
AU2018207584A1 (en) 2018-08-09
SG11201402666WA (en) 2014-10-30
JP2019059793A (en) 2019-04-18
US8680069B2 (en) 2014-03-25
AU2012352180A1 (en) 2014-07-31
SG10201604896TA (en) 2016-08-30
EP4144378A1 (en) 2023-03-08
US9271996B2 (en) 2016-03-01
KR20140102759A (en) 2014-08-22
US20130237593A1 (en) 2013-09-12
US20130245107A1 (en) 2013-09-19
US20130245105A1 (en) 2013-09-19
PL2791160T3 (en) 2022-06-20
US20130244278A1 (en) 2013-09-19
RU2014129004A (en) 2016-02-10
US9186372B2 (en) 2015-11-17
US8754062B2 (en) 2014-06-17
EP2791160A1 (en) 2014-10-22
DK2791160T3 (en) 2022-05-30
CN104114572A (en) 2014-10-22
AU2016231503A1 (en) 2016-10-06
US20130237594A1 (en) 2013-09-12
HUE059110T2 (en) 2022-10-28
US20200164038A1 (en) 2020-05-28
HK1203077A1 (en) 2015-10-16
US20130253043A1 (en) 2013-09-26
LT2791160T (en) 2022-06-10
CN110201187A (en) 2019-09-06
US20180125937A1 (en) 2018-05-10
US20130244279A1 (en) 2013-09-19
US20130236974A1 (en) 2013-09-12
US20130266640A1 (en) 2013-10-10
RU2649364C2 (en) 2018-04-02
US20130156849A1 (en) 2013-06-20
US20130252281A1 (en) 2013-09-26
CA3018046A1 (en) 2013-06-20
PT2791160T (en) 2022-07-04
MX2014007233A (en) 2015-02-04
US20160193299A1 (en) 2016-07-07
US8664194B2 (en) 2014-03-04
DE12858350T1 (en) 2021-10-07
HRP20220717T1 (en) 2022-07-22
US20130245104A1 (en) 2013-09-19

Similar Documents

Publication Publication Date Title
US20200164038A1 (en) Modified nucleoside, nucleotide, and nucleic acid compositions
AU2016202985A1 (en) Methods of increasing the viability or longevity of an organ or organ explant
WO2013106496A1 (en) Methods and compositions for targeting agents into and across the blood-brain barrier
US20140378538A1 (en) Methods of responding to a biothreat
WO2013090186A1 (en) Modified nucleic acids, and acute care uses thereof

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 12858350

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 232749

Country of ref document: IL

ENP Entry into the national phase

Ref document number: 2859387

Country of ref document: CA

Ref document number: 2014547454

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: MX/A/2014/007233

Country of ref document: MX

ENP Entry into the national phase

Ref document number: 20147019601

Country of ref document: KR

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2014129004

Country of ref document: RU

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2012352180

Country of ref document: AU

Date of ref document: 20121214

Kind code of ref document: A