US20130202684A1 - Pegylated liposomes for delivery of immunogen encoding rna - Google Patents

Pegylated liposomes for delivery of immunogen encoding rna Download PDF

Info

Publication number
US20130202684A1
US20130202684A1 US13/819,077 US201113819077A US2013202684A1 US 20130202684 A1 US20130202684 A1 US 20130202684A1 US 201113819077 A US201113819077 A US 201113819077A US 2013202684 A1 US2013202684 A1 US 2013202684A1
Authority
US
United States
Prior art keywords
rna
liposome
lipid
liposomes
peg
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US13/819,077
Other languages
English (en)
Inventor
Andrew Geall
Ayush Verma
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
GlaxoSmithKline Biologicals SA
Lichtstrasse
Original Assignee
Lichtstrasse
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=44653554&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20130202684(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Lichtstrasse filed Critical Lichtstrasse
Priority to US13/819,077 priority Critical patent/US20130202684A1/en
Assigned to NOVARTIS VACCINES AND DIAGNOSTICS, INC. reassignment NOVARTIS VACCINES AND DIAGNOSTICS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: VERMA, AYUSH, GEALL, ANDREW
Assigned to NOVARTIS AG reassignment NOVARTIS AG ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: NOVARTIS VACCINES AND DIAGNOSTICS, INC.
Publication of US20130202684A1 publication Critical patent/US20130202684A1/en
Assigned to GLAXOSMITHKLINE BIOLOGICALS SA reassignment GLAXOSMITHKLINE BIOLOGICALS SA ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: NOVARTIS AG
Assigned to GLAXOSMITHKLINE BIOLOGICALS SA reassignment GLAXOSMITHKLINE BIOLOGICALS SA CORRECTIVE ASSIGNMENT TO CORRECT THE INCORRECT APPL. NO. 13/819,007 PREVIOUSLY RECORDED AT REEL: 038982 FRAME: 0135. ASSIGNOR(S) HEREBY CONFIRMS THE ASSIGNMENT. Assignors: NOVARTIS AG
Abandoned legal-status Critical Current

Links

Images

Classifications

    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/39Medicinal preparations containing antigens or antibodies characterised by the immunostimulating additives, e.g. chemical adjuvants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • A61K39/155Paramyxoviridae, e.g. parainfluenza virus
    • 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
    • A61K9/10Dispersions; Emulsions
    • 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/10Antimycotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic 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
    • A61P37/04Immunostimulants
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/53DNA (RNA) vaccination
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55505Inorganic adjuvants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55555Liposomes; Vesicles, e.g. nanoparticles; Spheres, e.g. nanospheres; Polymers
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/16011Herpesviridae
    • C12N2710/16111Cytomegalovirus, e.g. human herpesvirus 5
    • C12N2710/16134Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein
    • 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
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/18011Paramyxoviridae
    • C12N2760/18511Pneumovirus, e.g. human respiratory syncytial virus
    • C12N2760/18534Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein
    • 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
    • C12N2770/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses positive-sense
    • C12N2770/00011Details
    • C12N2770/36011Togaviridae
    • C12N2770/36111Alphavirus, e.g. Sindbis virus, VEE, EEE, WEE, Semliki
    • C12N2770/36141Use of virus, viral particle or viral elements as a vector
    • C12N2770/36143Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2770/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses positive-sense
    • C12N2770/00011Details
    • C12N2770/36011Togaviridae
    • C12N2770/36111Alphavirus, e.g. Sindbis virus, VEE, EEE, WEE, Semliki
    • C12N2770/36171Demonstrated in vivo effect
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • This invention is in the field of non-viral delivery of RNA for immunisation.
  • nucleic acids for immunising animals has been a goal for several years.
  • Various approaches have been tested, including the use of DNA or RNA, of viral or non-viral delivery vehicles (or even no delivery vehicle, in a “naked” vaccine), of replicating or non-replicating vectors, or of viral or non-viral vectors.
  • nucleic acid vaccines There remains a need for further and improved nucleic acid vaccines and, in particular, for improved ways of delivering nucleic acid vaccines.
  • nucleic acid immunisation is achieved by delivering RNA encapsulated within a liposome.
  • the RNA encodes an immunogen of interest.
  • the liposome includes a PEGylated lipid i.e. the lipid is modified by covalent attachment of a polyethylene glycol.
  • PEG provides the liposomes with a coat which can confer favourable pharmacokinetic characteristics e.g. it can increase stability and prevent non-specific adsorption of the liposomes.
  • the inventors have found that the length of the PEG can affect in vivo expression of encapsulated RNA and so the invention uses liposomes which comprise PEG which has an average molecular mass of between 1 kDa and 3 kDa. PEG with a lower molecular weight (e.g. 500 or 750 Da) does not form stable liposomes.
  • the invention provides a liposome within which RNA encoding an immunogen of interest is encapsulated, wherein the liposome comprises at least one lipid which includes a polyethylene glycol moiety, such that polyethylene glycol is present on the liposome's exterior, wherein the average molecular mass of the polyethylene glycol is between 1 kDa and 3 kDa.
  • the liposomes are suitable for in vivo delivery of the RNA to a vertebrate cell and so they are useful as components in pharmaceutical compositions for immunising subjects against various diseases.
  • the invention also provides a process for preparing a RNA-containing liposome, comprising a step of mixing RNA with one or more lipids, under conditions such that the lipids form a liposome in which the RNA is encapsulated, wherein at least one lipid includes a polyethylene glycol moiety which becomes located on the liposome's exterior during the process, and wherein the average molecular mass of the polyethylene glycol is between 1 kDa and 3 kDa.
  • the liposome The liposome
  • the invention utilises liposomes within which immunogen-encoding RNA is encapsulated.
  • the RNA is (as in a natural virus) separated from any external medium. Encapsulation within the liposome has been found to protect RNA from RNase digestion.
  • the liposomes can include some external RNA (e.g. on their surface), but at least half of the RNA (and ideally all of it) is encapsulated in the liposome's core. Encapsulation within liposomes is distinct from, for instance, the lipid/RNA complexes disclosed in reference 1, where RNA is mixed with pre-formed liposomes.
  • RNA-containing aqueous core can have an anionic, cationic or zwitterionic hydrophilic head group.
  • anionic phospholipids dates back to the 1960s, and cationic liposome-forming lipids have been studied since the 1990s.
  • Some phospholipids are anionic whereas other are zwitterionic and others are cationic.
  • Suitable classes of phospholipid include, but are not limited to, phosphatidylethanolamines, phosphatidylcholines, phosphatidylserines, and phosphatidyl-glycerols, and some useful phospholipids are listed in Table 1.
  • Useful cationic lipids include, but are not limited to, dioleoyl trimethylammonium propane (DOTAP), 1,2-distearyloxy-N,N-dimethyl-3-aminopropane (DSDMA), 1,2-dioleyloxy-N,Ndimethyl-3-aminopropane (DODMA), 1,2-dilinoleyloxy-N,N-dimethyl-3-aminopropane (DLinDMA), 1,2-dilinolenyloxy-N,N-dimethyl-3-aminopropane (DLenDMA).
  • Zwitterionic lipids include, but are not limited to, acyl zwitterionic lipids and ether zwitterionic lipids.
  • lipids can be saturated or unsaturated.
  • DOPE 1,2-dioleoyl-sn-glycero-3-phosphatidylethanolamine
  • DPyPE 1,2-diphytanoyl-sn-glycero-3-phosphoethanolamine
  • the lipids can be saturated or unsaturated. The use of at least one unsaturated lipid for preparing liposomes is preferred. If an unsaturated lipid has two tails, both tails can be unsaturated, or it can have one saturated tail and one unsaturated tail.
  • a lipid can include a steroid group in one tail e.g. as in RV05.
  • the invention provides a liposome having a lipid bilayer encapsulating an aqueous core, wherein: (i) the lipid bilayer comprises at least one lipid which includes a polyethylene glycol moiety, such that polyethylene glycol is present on the liposome's exterior, wherein the average molecular mass of the polyethylene glycol is between 1 kDa and 3 kDa; and (ii) the aqueous core includes a RNA which encodes an immunogen.
  • Liposomes of the invention can be formed from a single lipid or from a mixture of lipids.
  • a mixture may comprise (i) a mixture of anionic lipids (ii) a mixture of cationic lipids (iii) a mixture of zwitterionic lipids (iv) a mixture of anionic lipids and cationic lipids (v) a mixture of anionic lipids and zwitterionic lipids (vi) a mixture of zwitterionic lipids and cationic lipids or (vii) a mixture of anionic lipids, cationic lipids and zwitterionic lipids.
  • a mixture may comprise both saturated and unsaturated lipids.
  • a mixture may comprise DSPC (zwitterionic, saturated), DlinDMA (cationic, unsaturated), and/or DMG (anionic, saturated).
  • DSPC zwitterionic, saturated
  • DlinDMA cationic, unsaturated
  • DMG anionic, saturated
  • a liposome of the invention is formed from a mixture of lipids
  • the proportion of those lipids which are PEGylated as described herein is less than 10% of the total amount of lipids e.g. between 0.5-5%, between 1-4%, or abbut 2%.
  • useful liposomes are shown below in which 2% of the total lipid is a PEG-DMG.
  • the remainder can be made of e.g. cholesterol (e.g. 35-50% cholesterol) and/or cationic lipid (e.g. 30-70%) and/or DSPC (e.g. 5-15%).
  • Such mixtures are used below. These percentage values are mole percentages.
  • a liposome can be formed from a cationic lipid (e.g. DlinDMA, RV05), a zwitterionic lipid (e.g. DSPC, DPyPE), a cholesterol, and a PEGylated lipid.
  • a cationic lipid e.g. DlinDMA, RV05
  • a zwitterionic lipid e.g. DSPC, DPyPE
  • cholesterol e.g. a zwitterionic lipid
  • PEGylated lipid e.g. a mixture of DSPC, DlinDMA, PEG-DMG and cholesterol is used in the examples, as well as several further mixtures.
  • At least one lipid within the liposome includes a polyethylene glycol moiety.
  • Liposomes which include these PEGylated lipids will have PEG oriented so that it is present on at least the exterior of the liposome (but some PEG may also be exposed to the liposome's interior i.e. to the aqueous core). This orientation can be achieved by attaching the PEG to an appropriate part of the lipid. For instance, in an amphiphilic lipid the PEG would be attached to the hydrophilic head, as it is this head which orients itself to the lipid bilayer's aqueous-facing exterior. PEGylation in this way can be achieved by covalent attachment of a PEG to a lipid e.g. using techniques such as those disclosed in reference 2 and 3.
  • PEGylated lipids will comprise the PEG structure:
  • n provides a molecular weight for the PEG of between 1 kDa and 3 kDa e.g. between 23 and 68, or about 45 for a 2 kDa PEGylation (e.g. see FIG. 16 ).
  • the PEG moiety can terminate with an —O-methyl group, and so a PEGylated lipid may comprise:
  • a PEGylated lipid useful with the invention may comprise:
  • PEG-DMG PEG-DMG
  • FIGS. 17A to 17E show further useful PEGylated lipids.
  • PEGylated cholesterol can also be used.
  • Other PEGylated lipids can be used e.g. lipids of Formula (X):
  • Z is a hydrophilic head group component selected from PEG and polymers based on poly(oxazoline), poly(ethylene oxide), poly(vinyl alcohol), poly(glycerol), poly(N-vinylpyrrolidone), poly[N-(2-hydroxypropyl)methacrylamide] and poly(amino acid)s, wherein the polymer may be linear or branched, and wherein the polymer may be optionally substituted;
  • Z is polymerized by n subunits
  • n is a number-averaged degree of polymerization between 10 and 200 units of Z (and can be optimized for different Z groups);
  • L 1 is an optionally substituted C 1-10 alkylene or C 1-10 heteroalkylene linker including zero, one or two of an ether (e.g., —C(O)O—), ester (e.g., —C(O)O—), succinate (e.g., —O(O)C—CH 2 —CH 2 —C(O)O—)), carbamate (e.g., —OC(O)—NR′—), carbonate (e.g., —OC(O)O—), urea (e.g., —NRC(O)NR′—), amine (e.g., —NR′—), amide (e.g., —C(O)NR′—), imine (e.g., —C(NR′)—), thioether (e.g., —S—), xanthate (e.g., —OC(S)S—), and phosphodiester (e.g., —OP(
  • X 1 and X 2 are independently selected from a carbon or a heteroatom selected from —NH—, —O—, —S— or a phosphate;
  • a 1 and A 2 are either independently selected from a C 6-30 alkyl, C 6 - 30 alkenyl, and C 6-30 alkynyl, wherein A 1 and A 2 may be the same or different, or A 1 and A 2 together with the carbon atom to which they are attached form an optionally substituted steroid.
  • a liposome of the invention will typically include a large number of PEG moieties, which may be the same or different.
  • the average molecular mass of the PEG in a liposome of the invention is between 1 kDa and 3 kDa e.g. between 1.5-2.5 kDa, between 1.7-2.3 kDa, between 1.8-2.2 kDa, between 1.9-2.1 kDa, or 2 kDa.
  • the PEG can be a PEG which is commonly known as “PEG 2000” or “PEG 2 k”, although the shorter “PEG 1000” and longer “PEG 3000” can also be used.
  • the PEG will usually comprise linear polymer chains but, in some embodiments, the PEG may comprise branched polymer chains.
  • a single lipid molecule may include more than one PEG group e.g. attached to different carbon atoms in a lipid's head group (e.g. see FIG. 18 ).
  • the reference to the molecular mass of PEG in a liposome is the molecular mass per lipid molecule rather than per PEG substituent.
  • the average molecular mass of the PEG is 2 kDa not 1 kDa.
  • the PEG may be a substituted PEG e.g. in which one or more carbon atoms in the polymer is substituted by one or more alkyl, alkoxy, acyl or aryl groups.
  • the PEG may include copolymer groups e.g. one or more propylene monomers, to form a PEG polypropylene polymer.
  • a lipid may be modified by covalent attachment of a moiety different from PEG.
  • a lipid may include a polyphosphazene.
  • a lipid may include a poly(vinyl pyrrolidone).
  • a lipid may include a poly(acryl amide);
  • a lipid may include a poly(2-methyl-2-oxazoline).
  • a lipid may include a poly(2-ethyl-2-oxazoline).
  • a lipid may include a phosphatidyl polyglycerol.
  • a lipid may include a poly[N-(2-hydroxypropyl)methacrylamide].
  • a lipid may include a polyalkylene ether polymer, other than PEG.
  • Liposomes are usually divided into three groups: multilamellar vesicles (MLV); small unilamellar vesicles (SUV); and large unilamellar vesicles (LUV).
  • MLVs have multiple bilayers in each vesicle, forming several separate aqueous compartments.
  • SUVs and LUVs have a single bilayer encapsulating an aqueous core; SUVs typically have a diameter ⁇ 50 nm, and LUVs have a diameter >50 nm.
  • Liposomes of the invention are ideally LUVs with a diameter in the range of 60-180 nm, and preferably in the range of 80-160 nm.
  • a liposome of the invention can be part of a composition comprising a plurality of liposomes, and the liposomes within the plurality can have a range of diameters.
  • a composition comprising a population of liposomes with different diameters (i) at least 80% by number of the liposomes should have diameters in the range of 60-180 nm, and preferably in the range of 80-160 nm, and/or (ii) the average diameter (by intensity e.g. Z-average) of the population is ideally in the range of 60-180 nm, and preferably in the range of 80-160 nm.
  • the diameters within the plurality should ideally have a polydispersity index ⁇ 0.2.
  • the liposome/RNA complexes of reference 1 are expected to have a diameter in the range of 600-800 nm and to have a high polydispersity.
  • mixing can be performed using a process in which two feed streams of aqueous RNA solution are combined in a single mixing zone with one stream of an ethanolic lipid solution, all at the same flow rate e.g. in a microfluidic channel as described below.
  • Liposomes of the invention include a RNA molecule which (unlike siRNA, as in reference 2) encodes an immunogen. After in vivo administration of the particles, RNA is released from the particles and is translated inside a cell to provide the immunogen in situ.
  • RNA is +-stranded, and so it can be translated by cells without needing any intervening replication steps such as reverse transcription. It can also bind to TLR7 receptors expressed by immune cells, thereby initiating an adjuvant effect.
  • Preferred +-stranded RNAs are self-replicating.
  • a self-replicating RNA molecule (replicon) can, when delivered to a vertebrate cell even without any proteins, lead to the production of multiple daughter RNAs by transcription from itself (via an antisense copy which it generates from itself).
  • a self-replicating RNA molecule is thus typically a +-strand molecule which can be directly translated after delivery to a cell, and this translation provides a RNA-dependent RNA polymerase which then produces both antisense and sense transcripts from the delivered RNA.
  • the delivered RNA leads to the production of multiple daughter RNAs.
  • RNAs may be translated themselves to provide in situ expression of an encoded immunogen, or may be transcribed to provide further transcripts with the same sense as the delivered RNA which are translated to provide in situ expression of the immunogen.
  • the overall result of this sequence of transcriptions is a huge amplification in the number of the introduced replicon RNAs and so the encoded immunogen becomes a major polypeptide product of the cells.
  • RNA replicon One suitable system for achieving self-replication is to use an alphavirus-based RNA replicon. These +-stranded replicons are translated after delivery to a cell to give of a replicase (or replicase-transcriptase). The replicase is translated as a polyprotein which auto-cleaves to provide a replication complex which creates genomic ⁇ strand copies of the +-strand delivered RNA. These ⁇ -strand transcripts can themselves be transcribed to give further copies of the +-stranded parent RNA and also to give a subgenomic transcript which encodes the immunogen. Translation of the subgenomic transcript thus leads to in situ expression of the immunogen by the infected cell.
  • a replicase or replicase-transcriptase
  • the replicase is translated as a polyprotein which auto-cleaves to provide a replication complex which creates genomic ⁇ strand copies of the +-strand delivered RNA.
  • These ⁇ -strand transcripts can themselves be transcribed to give
  • Suitable alphavirus replicons can use a replicase from a Sindbis virus, a Semliki forest virus, an eastern equine encephalitis virus, a Venezuelan equine encephalitis virus, etc.
  • Mutant or wild-type viruses sequences can be used e.g. the attenuated TC83 mutant of VEEV has been used in replicons [8].
  • a preferred self-replicating RNA molecule thus encodes (i) a RNA-dependent RNA polymerase which can transcribe RNA from the self-replicating RNA molecule and (ii) an immunogen.
  • the polymerase can be an alphavirus replicase e.g. comprising one or more of alphavirus proteins nsP1, nsP2, nsP3 and nsP4.
  • RNA molecule of the invention does not encode alphavirus structural proteins.
  • a preferred self-replicating RNA can lead to the production of genomic RNA copies of itself in a cell, but not to the production of RNA-containing virions.
  • the inability to produce these virions means that, unlike a wild-type alphavirus, the self-replicating RNA molecule cannot perpetuate itself in infectious form.
  • alphavirus structural proteins which are necessary for perpetuation in wild-type viruses are absent from self-replicating RNAs of the invention and their place is taken by gene(s) encoding the immunogen of interest, such that the subgenomic transcript encodes the immunogen rather than the structural alphavirus virion proteins.
  • RNA molecule useful with the invention may have two open reading frames.
  • the first (5′) open reading frame encodes a replicase; the second (3′) open reading frame encodes an immunogen.
  • the RNA may have additional (e.g. downstream) open reading frames e.g. to encode further immunogens (see below) or to encode accessory polypeptides.
  • a self-replicating RNA molecule can have a 5′ sequence which is compatible with the encoded replicase.
  • Self-replicating RNA molecules can have various lengths but they are typically 5000-25000 nucleotides long e.g. 8000-15000 nucleotides, or 9000-12000 nucleotides. Thus the RNA is longer than seen in siRNA delivery.
  • a RNA molecule useful with the invention may have a 5′ cap (e.g. a 7-methylguanosine). This cap can enhance in vivo translation of the RNA.
  • the 5′ nucleotide of a RNA molecule useful with the invention may have a 5′ triphosphate group. In a capped RNA this may be linked to a 7-methylguanosine via a 5′-to-5′ bridge.
  • a 5′ triphosphate can enhance RIG-I binding and thus promote adjuvant effects.
  • a RNA molecule may have a 3′ poly-A tail. It may also include a poly-A polymerase recognition sequence (e.g. AAUAAA) near its 3′ end.
  • AAUAAA poly-A polymerase recognition sequence
  • RNA molecule useful with the invention will typically be single-stranded.
  • Single-stranded RNAs can generally initiate an adjuvant effect by binding to TLR7, TLR8, RNA helicases and/or PKR.
  • RNA delivered in double-stranded form can bind to TLR3, and this receptor can also be triggered by dsRNA which is formed either during replication of a single-stranded RNA or within the secondary structure of a single-stranded RNA.
  • RNA molecule useful with the invention can conveniently be prepared by in vitro transcription (IVT).
  • IVT in vitro transcription
  • NT can use a (cDNA) template created and propagated in plasmid form in bacteria, or created synthetically (for example by gene synthesis and/or polymerase chain-reaction (PCR) engineering methods).
  • a DNA-dependent RNA polymerase such as the bacteriophage T7, T3 or SP6 RNA polymerases
  • Appropriate capping and poly-A addition reactions can be used as required (although the replicon's poly-A is usually encoded within the DNA template).
  • RNA polymerases can have stringent requirements for the transcribed 5′ nucleotide(s) and in some embodiments these requirements must be matched with the requirements of the encoded replicase, to ensure that the IVT-transcribed RNA can function efficiently as a substrate for its self-encoded replicase.
  • the self-replicating RNA can include (in addition to any 5′ cap structure) one or more nucleotides having a modified nucleobase.
  • the RNA can comprise m5C (5-methylcytidine), m5U (5-methyluridine), m6A (N6-methyladenosine), s2U (2-thiouridine), Um (2′-O-methyluridine), m1A (1-methyladenosine); m2A (2-methyladenosine); Am (2′-O-methyl adenosine); ms2m6A (2-methylthio-N6-methyladenosine); i6A (N6-isopentenyladenosine); ms2i6A (2-methylthio-N6isopentenyladenosine); io6A (N6-(cis-hydroxyisopentenyl)adenosine); ms2io6A (2-methylthio-N6-(cis-hydroxyis
  • a self-replicating RNA can include one or more modified pyrimidine nucleobases, such as pseudouridine and/or 5-methylcytosine residues.
  • the RNA includes no modified nucleobases, and may include no modified nucleotides i.e. all of the nucleotides in the RNA are standard A, C, G and U ribonucleotides (except for any 5′ cap structure, which may include a 7′-methylguanosine).
  • the RNA may include a 5′ cap comprising a 7′-methylguanosine, and the first 1, 2 or 3 5′ ribonucleotides may be methylated at the 2′ position of the ribose.
  • a RNA used with the invention ideally includes only phosphodiester linkages between nucleosides, but in some embodiments it can contain phosphoramidate, phosphorothioate, and/or methylphosphonate linkages.
  • a liposome includes fewer than 10 different species of RNA e.g. 5, 4, 3, or 2 different species; most preferably, a liposome includes a single RNA species i.e. all RNA molecules in the liposome have the same sequence and same length.
  • RNA per liposome can vary.
  • the number of individual self-replicating RNA molecules per liposome is typically ⁇ 50 e.g. ⁇ 20, ⁇ 10, ⁇ 5, or 1-4 per liposome.
  • RNA molecules used with the invention encode a polypeptide immunogen. After administration of the liposomes the RNA is translated in vivo and the immunogen can elicit an immune response in the recipient.
  • the immunogen may elicit an immune response against a bacterium, a virus, a fungus or a parasite (or, in some embodiments, against an allergen; and in other embodiments, against a tumor antigen).
  • the immune response may comprise an antibody response (usually including IgG) and/or a cell-mediated immune response.
  • the polypeptide immunogen will typically elicit an immune response which recognises the corresponding bacterial, viral, fungal or parasite (or allergen or tumour) polypeptide, but in some embodiments the polypeptide may act as a mimotope to elicit an immune response which recognises a bacterial, viral, fungal or parasite saccharide.
  • the immunogen will typically be a surface polypeptide e.g. an adhesin, a hemagglutinin, an envelope glycoprotein, a spike glycoprotein, etc.
  • the RNA molecule can encode a single polypeptide immunogen or multiple polypeptides. Multiple immunogens can be presented as a single polypeptide immunogen (fusion polypeptide) or as separate polypeptides. If immunogens are expressed as separate polypeptides from a replicon then one or more of these may be provided with an upstream IRES or an additional viral promoter element. Alternatively, multiple immunogens may be expressed from a polyprotein that encodes individual immunogens fused to a short autocatalytic protease (e.g. foot-and-mouth disease virus 2A protein), or as inteins.
  • a short autocatalytic protease e.g. foot-and-mouth disease virus 2A protein
  • the RNA encodes an immunogen.
  • the invention does not encompass RNA which encodes a firefly luciferase or which encodes a fusion protein of E. coli ⁇ -galactosidase or which encodes a green fluorescent protein (GFP).
  • GFP green fluorescent protein
  • Such polypeptides may be useful as markers, or even in a gene therapy context, but the invention concerns delivery of RNA for eliciting an immunological response system.
  • the immunogen also is not a self protein which is delivered to supplement or substitute for a defective host protein (as in gene therapy).
  • the RNA is not total mouse thymus RNA.
  • the immunogen elicits an immune response against a virus which infects fish, such as: infectious salmon anemia virus (ISAV), salmon pancreatic disease virus (SPDV), infectious pancreatic necrosis virus (IPNV), channel catfish virus (CCV), fish lymphocystis disease virus (FLDV), infectious hematopoietic necrosis virus (IHNV), koi herpesvirus, salmon picorna-like virus (also known as picorna-like virus of atlantic salmon), landlocked salmon virus (LSV), atlantic salmon rotavirus (ASR), trout strawberry disease virus (TSD), coho salmon tumor virus (CSTV), or viral hemorrhagic septicemia virus (VHSV).
  • infectious salmon anemia virus ISAV
  • SPDV salmon pancreatic disease virus
  • IPNV infectious pancreatic necrosis virus
  • CCV channel catfish virus
  • FLDV fish lymphocystis disease virus
  • IHNV infectious hematopoietic necrosis virus
  • Fungal immunogens may be derived from Dermatophytres, including: Epidermophyton floccusum, Microsporum audouini, Microsporum canis, Microsporum distortum, Microsporum equinum, Microsporum gypsum, Microsporum nanum, Trichophyton concentricum, Trichophyton equinum, Trichophyton gallinae, Trichophyton gypseum, Trichophyton megnini, Trichophyton mentagrophytes, Trichophyton quinckeanum, Trichophyton rubrum, Trichophyton schoenleini, Trichophyton tonsurans, Trichophyton verrucosum, T. verrucosum var. album, var.
  • the immunogen elicits an immune response against a parasite from the Plasmodium genus, such as P.falciparum, P.vivax, P.malariae or P.ovale .
  • the invention may be used for immunising against malaria.
  • the immunogen elicits an immune response against a parasite from the Caligidae family, particularly those from the Lepeophtheirus and Caligus genera e.g. sea lice such as Lepeophtheirus salmonis or Caligus rogercresseyi.
  • the immunogen elicits an immune response against: pollen allergens (tree-, herb, weed-, and grass pollen allergens); insect or arachnid allergens (inhalant, saliva and venom allergens, e.g. mite allergens, cockroach and midges allergens, hymenopthera venom allergens); animal hair and dandruff allergens (from e.g. dog, cat, horse, rat, mouse, etc.); and food allergens (e.g. a gliadin).
  • pollen allergens tree-, herb, weed-, and grass pollen allergens
  • insect or arachnid allergens inhalant, saliva and venom allergens, e.g. mite allergens, cockroach and midges allergens, hymenopthera venom allergens
  • animal hair and dandruff allergens from e.g. dog, cat, horse
  • Important pollen allergens from trees, grasses and herbs are such originating from the taxonomic orders of Fagales, Oleales, Pinales and platanaceae including, but not limited to, birch ( Betula ), alder ( Alnus ), hazel ( Corylus ), hornbeam ( Carpinus ) and olive ( Olea ), cedar ( Cryptomeria and Juniperus ), plane tree ( Platanus ), the order of Poales including grasses of the genera Lolium, Phleum, Poa, Cynodon, Dactylis, Holcus, Phalaris, Secale , and Sorghum , the orders of Asterales and Urticales including herbs of the genera Ambrosia, Artemisia , and Parietaria .
  • venom allergens including such originating from stinging or biting insects such as those from the taxonomic order of Hymenoptera inclikling bees (Apidae), wasps (Vespidea), and ants (Formicoidae).
  • the immunogen is a tumor antigen selected from: (a) cancer-testis antigens such as NY-ESO-1, SSX2, SCP1 as well as RAGE, BAGE, GAGE and MAGE family polypeptides, for example, GAGE-1, GAGE-2, MAGE-1, MAGE-2, MAGE-3, MAGE-4, MAGE-5, MAGE-6, and MAGE-12 (which can be used, for example, to address melanoma, lung, head and neck, NSCLC, breast, gastrointestinal, and bladder tumors; (b) mutated antigens, for example, p53 (associated with various solid tumors, e.g., colorectal, lung, head and neck cancer), p21/Ras (associated with, e.g., melanoma, pancreatic cancer and colorectal cancer), CDK4 (associated with, e.g., melanoma), MUM1 (associated with, e.g., melanoma), caspase-8
  • tumor immunogens include, but are not limited to, p15, Hom/Mel-40, H-Ras, E2A-PRL, H4-RET, IGH-IGK, MYL-RAR, Epstein Barr virus antigens, EBNA, human papillomavirus (HPV) antigens, including E6 and E7, hepatitis B and C virus antigens, human T-cell lymphotropic virus antigens, TSP-180, p185erbB2, p180erbB-3, c-met, mn-23H1, TAG-72-4, CA 19-9, CA 72-4, CAM 17.1, NuMa, K-ras, p16, TAGE, PSCA, CT7, 43-9F, 5T4, 791 Tgp72, beta-HCG, BCA225, BTAA, CA 125, CA 15-3 (CA 27.29 ⁇ BCAA), CA 195, CA 242, CA-50, CAM43, CD68 ⁇ KP1,
  • Liposomes of the invention are useful as components in pharmaceutical compositions for immunising subjects against various diseases. These compositions will typically include a pharmaceutically acceptable carrier in addition to the liposomes. A thorough discussion of pharmaceutically acceptable carriers is available in reference 29.
  • a pharmaceutical composition of the invention may include one or more small molecule immunopotentiators.
  • the composition may include a TLR2 agonist (e.g. Pam3CSK4), a TLR4 agonist (e.g. an aminoalkyl glucosaminide phosphate, such as E6020), a TLR7 agonist (e.g. imiquimod), a TLR8 agonist (e.g. resiquimod) and/or a TLR9 agonist (e.g. IC31).
  • a TLR2 agonist e.g. Pam3CSK4
  • TLR4 agonist e.g. an aminoalkyl glucosaminide phosphate, such as E6020
  • TLR7 agonist e.g. imiquimod
  • a TLR8 agonist e.g. resiquimod
  • TLR9 agonist e.g. IC31
  • Any such agonist ideally has a molecular weight of ⁇ 2000 Da
  • compositions of the invention may include the liposomes in plain water (e.g. w.f.i.) or in a buffer e.g. a phosphate buffer, a Tris buffer, a borate buffer, a succinate buffer, a histidine buffer, or a citrate buffer.
  • Buffer salts will typically be included in the 5-20 mM range.
  • compositions of the invention may have a pH between 5.0 and 9.5 e.g. between 6.0 and 8.0.
  • compositions of the invention may include, sodium salts (e.g. sodium chloride) to give tonicity.
  • sodium salts e.g. sodium chloride
  • a concentration of 10 ⁇ 2 mg/ml NaCl is typical e.g. about 9 mg/ml.
  • compositions of the invention may include metal ion chelators. These can prolong RNA stability by removing ions which can accelerate phosphodiester hydrolysis.
  • a composition may include one or more of EDTA, EGTA, BAPTA, pentetic acid, etc.
  • chelators are typically present at between 10-500 mM e.g. 0.1 mM.
  • a citrate salt such as sodium citrate, can also act as a chelator, while advantageously also providing buffering activity.
  • compositions of the invention may have an osmolality of between 200 mOsm/kg and 400 mOsm/kg, e.g. between 240-360 mOsm/kg, or between 290-310 mOsm/kg.
  • compositions of the invention may include one or more preservatives, such as thiomersal or 2-phenoxyethanol.
  • preservatives such as thiomersal or 2-phenoxyethanol.
  • Mercury-free compositions are preferred, and preservative-free vaccines can be prepared.
  • compositions of the invention are preferably sterile.
  • compositions of the invention are preferably non-pyrogenic e.g. containing ⁇ 1 EU (endotoxin unit, a standard measure) per dose, and preferably ⁇ 0.1 EU per dose.
  • ⁇ 1 EU endotoxin unit, a standard measure
  • compositions of the invention are preferably gluten free.
  • compositions of the invention may be prepared in unit dose form.
  • a unit dose may have a volume of between 0.1-1.0 ml e.g. about 0.5 ml.
  • the compositions may be prepared as injectables, either as solutions or suspensions.
  • the composition may be prepared for pulmonary administration e.g. by an inhaler, using a fine spray.
  • the composition may be prepared for nasal, aural or ocular administration e.g. as spray or drops. Injectables for intramuscular administration are typical.
  • compositions comprise an immunologically effective amount of liposomes, as well as any other components, as needed.
  • immunologically effective amount it is meant that the administration of that amount to an individual, either in a single dose or as part of a series, is effective for treatment or prevention. This amount varies depending upon the health and physical condition of the individual to be treated, age, the taxonomic group of individual to be treated (e.g. non-human primate, primate, etc.), the capacity of the individual's immune system to synthesise antibodies, the degree of protection desired, the formulation of the vaccine, the treating doctor's assessment of the medical situation, and other relevant factors. It is expected that the amount will fall in a relatively broad range that can be determined through routine trials.
  • compositions of the invention will generally be expressed in terms of the amount of RNA per dose.
  • a preferred dose has ⁇ 100 ⁇ g RNA (e.g. from 10-100 ⁇ g, such as about 10 ⁇ g, 25 ⁇ g, 50 ⁇ g, 75 ⁇ g or 100 ⁇ g), but expression can be seen at much lower levels e.g. ⁇ 1 ⁇ g/dose, ⁇ 100 ng/dose, ⁇ 10 ng/dose, ⁇ 1 ng/dose, etc
  • the invention also provides a delivery device (e.g. syringe, nebuliser, sprayer, inhaler, dermal patch, etc.) containing a pharmaceutical composition of the invention.
  • a delivery device e.g. syringe, nebuliser, sprayer, inhaler, dermal patch, etc.
  • This device can be used to administer the composition to a vertebrate subject.
  • Liposomes of the invention do not contain ribosomes.
  • liposomes and pharmaceutical compositions of the invention are for in vivo use for eliciting an immune response against an immunogen of interest.
  • the invention provides a method for raising an immune response in a vertebrate comprising the step of administering an effective amount of a liposome or pharmaceutical composition of the invention.
  • the immune response is preferably protective and preferably involves antibodies and/or cell-mediated immunity.
  • the method may raise a booster response.
  • the invention also provides a liposome or pharmaceutical composition of the invention for use in a method for raising an immune response in a vertebrate.
  • the invention also provides the use of a liposome of the invention in the manufacture of a medicament for raising an immune response in a vertebrate.
  • the vertebrate By raising an immune response in the vertebrate by these uses and methods, the vertebrate can be protected against various diseases and/or infections e.g. against bacterial and/or viral diseases as discussed above.
  • the liposomes and compositions are immunogenic, and are more preferably vaccine compositions.
  • Vaccines according to the invention may either be prophylactic (i.e. to prevent infection) or therapeutic (i.e. to treat infection), but will typically be prophylactic.
  • the vertebrate is preferably a mammal, such as a human or a large veterinary mammal (e.g. horses, cattle, deer, goats, pigs).
  • the human is preferably a child (e.g. a toddler or infant) or a teenager; where the vaccine is for therapeutic use, the human is preferably a teenager or an adult.
  • a vaccine intended for children may also be administered to adults e.g. to assess safety, dosage, immunogenicity, etc.
  • Vaccines prepared according to the invention may be used to treat both children and adults.
  • a human patient may be less than 1 year old, less than 5 years old, 1-5 years old, 5-15 years old, 15-55 years old, or at least 55 years old.
  • Preferred patients for receiving the vaccines are the elderly (e.g. ⁇ 50 years old, ⁇ 60 years old, and preferably ⁇ 65 years), the young (e.g. ⁇ 5 years old), hospitalised patients, healthcare workers, armed service and military personnel, pregnant women, the chronically ill, or immunodeficient patients.
  • the vaccines are not suitable solely for these groups, however, and may be used more generally in a population.
  • compositions of the invention will generally be administered directly to a patient.
  • Direct delivery may be accomplished by parenteral injection (e.g. subcutaneously, intraperitoneally, intravenously, intramuscularly, intradermally, or to the interstitial space of a tissue; unlike reference 1, intraglossal injection is not typically used with the present invention).
  • Alternative delivery routes include rectal, oral (e.g. tablet, spray), buccal, sublingual, vaginal, topical, transdermal or transcutaneous, intranasal, ocular, aural, pulmonary or other mucosal administration.
  • Intradermal and intramuscular administration are two preferred routes. Injection may be via a needle (e.g. a hypodermic needle), but needle-free injection may alternatively be used.
  • a typical intramuscular dose is 0.5 ml.
  • the invention may be used to elicit systemic and/or mucosal immunity, preferably to elicit an enhanced systemic and/or mucosal immunity.
  • Dosage can be by a single dose schedule or a multiple dose schedule. Multiple doses may be used in a primary immunisation schedule and/or in a booster immunisation schedule. In a multiple dose schedule the various doses may be given by the same or different routes e.g. a parenteral prime and mucosal boost, a mucosal prime and parenteral boost, etc. Multiple doses will typically be administered at least 1 week apart (e.g. about 2 weeks, about 3 weeks, about 4 weeks, about 6 weeks, about 8 weeks, about 10 weeks, about 12 weeks, about 16 weeks, etc.). In one embodiment, multiple doses may be administered approximately 6 weeks, 10 weeks and 14 weeks after birth, e.g.
  • two primary doses are administered about two months apart, e.g. about 7, 8 or 9 weeks apart, followed by one or more booster doses about 6 months to 1 year after the second primary dose, e.g. about 6, 8, 10 or 12 months after the second primary dose.
  • three primary doses are administered about two months apart, e.g. about 7, 8 or 9 weeks apart, followed by one or more booster doses about 6 months to 1 year after the third primary dose, e.g. about 6, 8, 10, or 12 months after the third primary dose.
  • Compounds of formula (X) contains a hydrophilic polymer head group linked to a lipid moiety. They can be described as “stealth lipids” and they have formula:
  • Z is a hydrophilic head group component selected from PEG and polymers based on poly(oxazoline), poly(ethylene oxide), poly(vinyl alcohol), poly(glycerol), poly(N-vinylpyrrolidone), poly[N-(2-hydroxypropyl)methacrylamide] and poly(amino acids, wherein the polymer may be linear or branched, and wherein the polymer may be optionally substituted;
  • n is a number-averaged degree of polymerization between 10 and 200 units of Z, wherein n is optimized for different polymer types;
  • L 1 is an optionally substituted C 1-10 alkylene or C 1-10 heteroalkylene linker including zero, one or two of an ether (e.g., —O—), ester (e.g., —C(O)O—), succinate (e.g., —O(O)C—CH 2 —CH 2 —C(O)O—)), carbamate (e.g., —OC(O)—NR′—), carbonate (e.g., —OC(O)O—), urea (e.g., —NRC(O)NR′—), amine (e.g., —NR′—), amide (e.g., —C(O)NR′—), imine (e.g., —C(NR′)—), thioether (e.g., —S—), xanthate (e.g., —OC(S)S—), and phosphodiester (e.g., —OP(O) 2 O
  • R′ is independently selected from —H, —NH—, —NH 2 , —O—; —S—, a phosphate or an optionally substituted C 1-10 alkylene;
  • X 1 and X 2 are independently selected from a carbon or a heteroatom selected from —NH—, —O—, —S— or a phosphate;
  • a 1 and A 2 are independently selected from a C 6-30 alkyl, C 6-30 alkenyl, and C 6-30 alkynyl, wherein A 1 and A 2 may be the same or different, or A 1 and A 2 together with the carbon atom to which they are attached form an optionally substituted steroid.
  • the compound of formula (X) has formula (X)
  • PEG is a poly(ethylene glycol) subunit, wherein the PEG may be linear or branched;
  • n is a number-averaged degree of polymerization between 10 and 200 units of PEG, preferably around 45 units;
  • L 1 is an optionally substituted C 1-10 heteroalkylene linker containing one or two of an ether, ester, succinate, carbamate, carbonate, urea, amine, amide, imine, thioether, xanthate, and phosphodiester;
  • X 1 and X 2 are oxygen
  • a 1 and A 2 are independently selected from a C 6-30 alkyl, C 6-30 alkenyl, and C 6-30 alkynyl, wherein A 1 and A 2 may be the same or different, or wherein A 1 and A 2 together with the carbon atom to which they are attached form an optionally substituted steroid.
  • the lipids of formulae (X) and (X′), when formulated with cationic lipids to form liposomes, can increase the length of time for which a liposome can exist in vivo (e.g. in the blood). They can shield the surface of a liposome surface and thereby reduce opsonisation by blood proteins and uptake by macrophages. Further details are in references 30 and 31.
  • the lipid comprises a group selected from PEG (sometimes referred to as poly(ethylene oxide)) and polymers based on poly(oxazoline), poly(vinyl alcohol), poly(glycerol), poly(N-vinylpyrrolidone), poly[N-(2-hydroxypropyl)methacrylamide] and poly(amino acid)s.
  • PEG sometimes referred to as poly(ethylene oxide)
  • polymers based on poly(oxazoline), poly(vinyl alcohol), poly(glycerol), poly(N-vinylpyrrolidone), poly[N-(2-hydroxypropyl)methacrylamide] and poly(amino acid)s PEG (sometimes referred to as poly(ethylene oxide)) and polymers based on poly(oxazoline), poly(vinyl alcohol), poly(glycerol), poly(N-vinylpyrrolidone), poly[N-(2-hydroxypropyl)methacrylamide] and poly(amino acid)s.
  • Suitable PEGylated lipids for use with the invention include polyethyleneglycol-diacylglycerol or polyethyleneglycol-diacylglycamide (PEG-DAG) conjugates including those comprising a dialkylglycerol or dialkylglycamide group having alkyl chain length independently comprising from about C4 to about C40 saturated or unsaturated carbon atoms.
  • the dialkylglycerol or dialkylglycamide group can further comprise one or more substituted alkyl groups.
  • the PEGyltaed lipid can be selected from PEG-dilaurylglycerol, PEG-dimyristylglycerol (catalog #GM-020 from NOF), PEG-dipalmitoylglycerol, PEG-disterylglycerol, PEG-dilaurylglycamide, PEG-dimyristylglycamide, PEG-dipalmitoyl-glycamide, and PEG-disterylglycamide, PEG-cholesterol (1-[8′-(Cholest-5-en-3 [beta]-oxy)carboxamido-3′,6′-dioxaoctanyl]carbamoyl-[omega]-methyl-poly(ethylene glycol), PEG-DMB (3,4-Ditetradecoxylbenzyl-[omega]-methyl-poly(ethylene glycol)ether), 1,2-dimyristoyl-s
  • S010 and S011 are disclosed in ref. 32 under the labels IVa and IVc, respectively.
  • ref. 32 a different synthesis from that reported herein is used to prepare IVa and IVc.
  • halogen includes fluorine, chlorine, bromine and iodine.
  • alkyl alkylene
  • alkenyl alkynyl
  • alkynyl alkynyl
  • alkyl includes monovalent, straight or branched, saturated, acyclic hydrocarbyl groups.
  • alkyl is C 1-10 alkyl, in another embodiment C 1-6 alkyl, in another embodiment C 1-4 alkyl, such as methyl, ethyl, n-propyl, i-propyl or t-butyl groups.
  • cycloalkyl includes monovalent, saturated, cyclic hydrocarbyl groups.
  • cycloalkyl is C 3-10 cycloalkyl, in another embodiment C 3-6 cycloalkyl such as cyclopentyl and cyclohexyl.
  • alkoxy means alkyl-O—.
  • alkenyl includes monovalent, straight or branched, unsaturated, acyclic hydrocarbyl groups having at least one carbon-carbon double bond and, in one embodiment, no carbon-carbon triple bonds. In one embodiment alkenyl is C 2-10 alkenyl, in another embodiment C 2-6 alkenyl, in another embodiment C 2-4 alkenyl.
  • cycloalkenyl includes monovalent, partially unsaturated, cyclic hydrocarbyl groups having at least one carbon-carbon double bond and, in one embodiment, no carbon-carbon triple bonds.
  • cycloalkenyl is C 3-10 cycloalkenyl, in another embodiment C 5-10 cycloalkenyl, e.g. cyclohexenyl or benzocyclohexyl.
  • alkynyl includes monovalent, straight or branched, unsaturated, acyclic hydrocarbyl groups having at least one carbon-carbon triple bond and, in one embodiment, no carbon-carbon double bonds.
  • alkynyl is C 2-10 alkynyl, in another embodiment C 2-6 alkynyl, in another embodiment C 2-4 alkynyl.
  • cycloalkynyl includes monovalent, partially unsaturated, cyclic hydrocarbyl groups having at least one carbon-carbon triple bond and, in one embodiment, no carbon-carbon double bonds.
  • cycloalkynyl is C 3-10 cycloalkenyl, in another embodiment C 5-10 cycloalkynyl
  • alkylene includes divalent, straight or branched, saturated, acyclic hydrocarbyl groups.
  • alkylene is C 1-10 alkylene, in another embodiment C 1-6 alkylene, in another embodiment C 1-4 alkylene, such as methylene, ethylene, n-propylene, i-propylene or t-butylene groups.
  • alkenylene includes divalent, straight or branched, unsaturated, acyclic hydrocarbyl groups having at least one carbon-carbon double bond and, in one embodiment, no carbon-carbon triple bonds.
  • alkenylene is C 2-10 alkenylene, in another embodiment C 2-6 alkenylene, in another embodiment C 2-4 alkenylene.
  • alkynylene includes divalent, straight or branched, unsaturated, acyclic hydrocarbyl groups having at least one carbon-carbon triple bond and, in one embodiment, no carbon-carbon double bonds.
  • alkynylene is C 2-10 alkynylene, in another embodiment C 2-6 alkynylene, in another embodiment C 2-4 alkynylene.
  • heteroalkyl includes alkyl groups in which up to six carbon atoms, in one embodiment up to five carbon atoms, in another embodiment up to four carbon atoms, in another embodiment up to three carbon atoms, in another embodiment up to two carbon atoms, in another embodiment one carbon atom, are each replaced independently by O, S(O) q , N, P(O) s or Si (and preferably O, S(O) q or N), provided at least one of the alkyl carbon atoms remains.
  • the heteroalkyl group may be C-linked or hetero-linked, i.e. it may be linked to the remainder of the molecule through a carbon atom or through O, S(O) q , N, P(O) r or Si.
  • heterocycloalkyl includes cycloalkyl groups in which up to six carbon atoms, in one embodiment up to five carbon atoms, in another embodiment up to four carbon atoms, in another embodiment up to three carbon atoms, in another embodiment up to two carbon atoms, in another embodiment one carbon atom, are each replaced independently by O, S(O) q or N, provided at least one of the cycloalkyl carbon atoms remains.
  • heterocycloalkyl groups include oxiranyl, thiaranyl, aziridinyl, oxetanyl, thiatanyl, azetidinyl, tetrahydrofuranyl, tetrahydrothiophenyl, pyrrolidinyl, tetrahydropyranyl, tetrahydrothiopyranyl, piperidinyl, 1,4-dioxanyl, 1,4-oxathianyl, morpholinyl, 1,4-dithianyl, piperazinyl, 1,4-azathianyl, oxepanyl, thiepanyl, azepanyl, 1,4-dioxepanyl, 1,4-oxathiepanyl, 1,4-oxaazepanyl, 1,4-dithiepanyl, 1,4-thieazepanyl and 1,4-diazepanyl.
  • heteroalkenyl includes alkenyl groups in which up to three carbon atoms, in one embodiment up to two carbon atoms, in another embodiment one carbon atom, are each replaced independently by O, S(O) q or N, provided at least one of the alkenyl carbon atoms remains.
  • the heteroalkenyl group may be C-linked or hetero-linked, i.e. it may be linked to the remainder of the molecule through a carbon atom or through O, S(O) q or N.
  • heterocycloalkenyl includes cycloalkenyl groups in which up to three carbon atoms, in one embodiment up to two carbon atoms, in another embodiment one carbon atom, are each replaced independently by O, S(O) q or N, provided at least one of the cycloalkenyl carbon atoms remains.
  • heterocycloalkenyl groups include 3,4-dihydro-2H-pyranyl, 5-6-dihydro-2H-pyranyl, 2H-pyranyl, 1,2,3,4-tetrahydropyridinyl and 1,2,5,6-tetrahydropyridinyl.
  • the heterocycloalkenyl group may be C-linked or N-linked, i.e. it may be linked to the remainder of the molecule through a carbon atom or through a nitrogen atom.
  • heteroalkynyl includes alkynyl groups in which up to three carbon atoms, in one embodiment up to two carbon atoms, in another embodiment one carbon atom, are each replaced independently by O, S(O) q or N, provided at least one of the alkynyl carbon atoms remains.
  • the heteroalkynyl group may be C-linked or hetero-linked, i.e. it may be linked to the remainder of the molecule through a carbon atom or through O, S(O) q or N.
  • heterocycloalkynyl includes cycloalkynyl groups in which up to three carbon atoms, in one embodiment up to two carbon atoms, in another embodiment one carbon atom, are each replaced independently by O, S(O) q or N, provided at least one of the cycloalkynyl carbon atoms remains.
  • the heterocycloalkenyl group may be C-linked or N-linked, i.e. it may be linked to the remainder of the molecule through a carbon atom or through a nitrogen atom.
  • heteroalkylene includes alkylene groups in which up to three carbon atoms, in one embodiment up to two carbon atoms, in another embodiment one carbon atom, are each replaced independently by O, S(O) q or N, provided at least one of the alkylene carbon atoms remains.
  • heteroalkenylene includes alkenylene groups in which up to three carbon atoms, in one embodiment up to two carbon atoms, in another embodiment one carbon atom, are each replaced independently by O, S(O) q or N, provided at least one of the alkenylene carbon atoms remains.
  • heteroalkynylene includes alkynylene groups in which up to three carbon atoms, in one embodiment up to two carbon atoms, in another embodiment one carbon atom, are each replaced independently by O, S(O) q or N, provided at least one of the alkynylene carbon atoms remains.
  • aryl includes monovalent, aromatic, cyclic hydrocarbyl groups, such as phenyl or naphthyl (e.g. 1-naphthyl or 2-naphthyl).
  • the aryl groups may be monocyclic or polycyclic fused ring aromatic groups.
  • Preferred aryl are C 6 -C 10 aryl.
  • aryl groups are monovalent derivatives of aceanthrylene, acenaphthylene, acephenanthrylene, anthracene, azulene, chrysene, coronene, fluoranthene, fluorene, as-indacene, s-indacene, indene, naphthalene, ovalene, perylene, phenalene, phenanthrene, picene, pleiadene, pyrene, pyranthrene and rubicene.
  • arylalkyl means alkyl substituted with an aryl group, e.g. benzyl.
  • arylene includes divalent aromatic, cyclic hydrocarbyl groups, such as phenylene.
  • the arylene groups may be monocyclic or polycyclic fused ring aromatic groups.
  • Preferred arylene are C 6 -C 10 arylene.
  • arylene groups are divalent derivatives of aceanthrylene, acenaphthylene, acephenanthrylene, anthracene, azulene, chrysene, coronene, fluoranthene, fluorene, as-indacene, s-indacene, indene, naphthalene, ovalene, perylene, phenalene, phenanthrene, picene, pleiadene, pyrene, pyranthrene and rubicene.
  • heteroaryl includes monovalent, heteroaromatic, cyclic hydrocarbyl groups additionally containing one or more heteroatoms independently selected from O, S, N and NR N , where R N is defined below (and in one embodiment is H or alkyl (e.g. C 1-6 alkyl)).
  • the heteroaryl groups may be monocyclic or polycyclic (e.g. bicyclic) fused ring heteroaromatic groups.
  • heteroaryl groups contain 5-13 ring members (preferably 5-10 members) and 1, 2, 3 or 4 ring heteroatoms independently selected from O, S, N and NR N .
  • a heteroaryl group may be 5, 6, 9 or 10 membered, e.g. 5-membered monocyclic, 6-membered monocyclic, 9-membered fused-ring bicyclic or 10-membered fused-ring bicyclic.
  • Monocyclic heteroaromatic groups include heteroaromatic groups containing 5-6 ring members and 1, 2, 3 or 4 heteroatoms selected from O, S, N or NR N .
  • 5-membered monocyclic heteroaryl groups contain 1 ring member which is an —NR N — group, an —O— atom or an —S— atom and, optionally, 1-3 ring members (e.g. 1 or 2 ring members) which are ⁇ N— atoms (where the remainder of the 5 ring members are carbon atoms).
  • Examples of 5-membered monocyclic heteroaryl groups are pyrrolyl, furanyl, thiophenyl, pyrazolyl, imidazolyl, isoxazolyl, oxazolyl, isothiazolyl, thiazolyl, 1,2,3 triazolyl, 1,2,4 triazolyl, 1,2,3 oxadiazolyl, 1,2,4 oxadiazolyl, 1,2,5 oxadiazolyl, 1,3,4 oxadiazolyl, 1,3,4 thiadiazolyl, pyridyl, pyrimidinyl, pyridazinyl, pyrazinyl, 1,3,5 triazinyl, 1,2,4 triazinyl, 1,2,3 triazinyl and tetrazolyl.
  • 6-membered monocyclic heteroaryl groups are pyridinyl, pyridazinyl, pyrimidinyl and pyrazinyl.
  • 6-membered monocyclic heteroaryl groups contain 1 or 2 ring members which are ⁇ N— atoms (where the remainder of the 6 ring members are carbon atoms).
  • Bicyclic heteroaromatic groups include fused-ring heteroaromatic groups containing 9-13 ring members and 1, 2, 3, 4 or more heteroatoms selected from O, S, N or NR N .
  • 9-membered bicyclic heteroaryl groups contain 1 ring member which is an —NR N — group, an —O— atom or an —S— atom and, optionally, 1-3 ring members (e.g. 1 or 2 ring members) which are ⁇ N— atoms (where the remainder of the 9 ring members are carbon atoms).
  • 9-membered fused-ring bicyclic heteroaryl groups are benzofuranyl, benzothiophenyl, indolyl, benzimidazolyl, indazolyl, benzotriazolyl, pyrrolo[2,3-b]pyridinyl, pyrrolo[2,3-c]pyridinyl, pyrrolo[3,2-c]pyridinyl, pyrrolo[3,2-b]pyridinyl, imidazo[4,5-b]pyridinyl, imidazo[4,5-c]pyridinyl, pyrazolo[4,3-d]pyridinyl, pyrazolo[4,3-c]pyridinyl, pyrazolo[3,4-c]pyridinyl, pyrazolo[3,4-b]pyridinyl, isoindolyl, indazolyl, purinyl, indolininyl, imidazo[1,2-a]pyridiny
  • 10-membered bicyclic heteroaryl groups contain 1-3 ring members which are ⁇ N— atoms (Where the remainder of the 10 ring members are carbon atoms).
  • 10-membered fused-ring bicyclic heteroaryl groups are quinolinyl, isoquinolinyl, cinnolinyl, quinazolinyl, quinoxalinyl, phthalazinyl, 1,6-naphthyridinyl, 1,7-naphthyridinyl, 1,8-naphthyridinyl, 1,5-naphthyridinyl, 2,6-naphthyridinyl, 2,7-naphthyridinyl, pyrido [3 ,2-d]pyrimidinyl, pyrido[4,3-d]pyrimidinyl, pyrido[3,4-d]pyrimidinyl, pyrido [2,3-d]pyrimidinyl, pyrido[2,3-d]pyrimidinyl, pyrido[2,3-b]pyrazinyl, pyrido[3,4
  • heteroarylalkyl means alkyl substituted with a heteroaryl group.
  • heteroarylene includes divalent heteroaromatic, cyclic hydrocarbyl groups additionally containing one or more heteroatoms independently selected from O, S, N and NR N , where R N is defined below (and in one embodiment is H or alkyl (e.g. C 1-6 alkyl)).
  • R N is defined below (and in one embodiment is H or alkyl (e.g. C 1-6 alkyl)).
  • the heteroarylene groups may be monocyclic or polycyclic (e.g. bicyclic) fused ring heteroaromatic groups.
  • heteroarylene groups contain 5-13 ring members (preferably 5-10 members) and 1, 2, 3 or 4 ring heteroatoms independently selected from O, S, N and NR N .
  • a heteroarylene group may be 5, 6, 9 or 10 membered, e.g.
  • heteroarylene includes divalent derivatives of each of the heteroaryl groups discussed above.
  • aryl aromatic, aromatic and heteroaryl and “heteroaromatic” also include groups that are partially reduced.
  • heteroaryl includes fused species in which one of the rings has been reduced to a saturated ring (e.g. 1,2,3,4-tetrahydro-1,8-naphthyridin-2-yl).
  • ⁇ C—H is replaced by ⁇ N or ⁇ P(O) r ;
  • R N is H or optionally substituted C 1-6 alkyl, C 1-6 heteroalkyl, C 3-6 cycloalkyl, C 3-6 heterocycloalkyl, C 2-6 alkenyl, C 2-6 heteroalkenyl, C 3-6 cycloalkenyl, C 3-6 heterocycloalkenyl, phenyl, or heteroaryl containing 5 or 6 ring members.
  • R N is preferably H, C 1-6 alkyl or C 3-6 cycloalkyl.
  • q is independently 0, 1 or 2. In one embodiment, q is 0.
  • r is independently 0 or 1. In one embodiment, r is 0.
  • heteroatom containing groups such as heteroalkyl etc.
  • a numerical of carbon atoms is given, for instance C 3-6 heteroalkyl
  • a C 3-6 heteroalkyl group would, for example, contain less than 3-6 chain carbon atoms.
  • a pyridyl group would be classed as a C 6 heteroaryl group even though it contains 5 carbon atoms.
  • Groups of the compounds of the invention may be substituted or unsubstituted, in one embodiment unsubstituted.
  • substitution involves the notional replacement of a hydrogen atom with a substituent group, or two hydrogen atoms in the case of substitution by ⁇ O.
  • substituents on each group there will generally be 1 to 5 substituents on each group, in one embodiment 1 to 3 substituents, in one embodiment 1 or 2 substituents, in one embodiment 1 substituent.
  • One embodiment includes more than one substituent on the same atom, e.g. an acetal group.
  • the substituent(s) is/are independently Sub' or Sub 2 (in one embodiment Sub 2 ) wherein:
  • R s in Sub 1 can be optionally substituted by 1 to 3 substituents Sub 2 , Sub 2 is unsubstituted.
  • R s is unsubstituted.
  • Rs is H or C 1-6 alkyl, optionally substituted by 1 to 3 substituents Sub 2 .
  • Sub 2 is independently halogen, trihalomethyl, trihaloethyl, —NO 2 , —CN, —N + (C 1-6 alkyl) 2 O ⁇ ,—CO 2 H, —SO 3 H, —SOC 1-6 alkyl, —SO 2 C 1-6 alkyl, —C( ⁇ O)H, —C( ⁇ O)C 1-6 alkyl, ⁇ O, —N(C 1-6 alkyl) 2 , —C( ⁇ O)NH 2 , —C 1-6 alkyl, —C 3-6 cycloalkyl, —C 3-6 heterocycloalkyl, —Z t —C 1-6 alkyl or —Z t —C 3-6 cycloalkyl.
  • Sub 1 is not —R s and Sub 2 is not —C 1-6 alkyl, —C 1-6 heteroalkyl, —C 2-6 alkenyl, —C 2-6 heteroalkenyl, —C 2-6 alkynyl or —C 2-6 heteroalkynyl.
  • a group other than Sub 2 has at least 2 positions which may be substituted
  • the group may be substituted by both ends of an alkylene, alkenylene, alkynylene, heteroalkylene, heteroalkenylene or heteroalkynylene chain (in one embodiment containing 1 to 6 atoms, in a further embodiment 3 to 6 atoms, and in a further embodiment 3 or 4 atoms) to form a cyclic moiety.
  • That chain is optionally substituted by 1 to 3 substituents Sub 2 . In one embodiment that chain is not substituted.
  • cycloalkyl cycloalkenyl
  • cycloalkynyl cycloalkynyl
  • heterocycloalkyl cycloalkenyl
  • heterocycloalkynyl cycloalkynyl
  • aryl cycloaryl
  • heteroaryl includes a species in which a heterocycloalkyl ring is fused to the aromatic ring (e.g. 5 ,6,7,8-tetrahydro- 1 ,8-naphthyridin-2-yl).
  • a group other than Sub 2 has an atom which may be substituted twice, that atom may be substituted by both ends of an alkylene, alkenylene, alkynylene, heteroalkylene, heteroalkenylene or heteroalkynylene chain (in one embodiment containing 2 to 8 atoms, in a further embodiment 3 to 6 atoms, and in a further embodiment 4 or 5 atoms) to form a cyclic moiety. That chain is optionally substituted by 1 to 3 substituents Sub 2 . In one embodiment that chain is not substituted.
  • cycloalkyl optionally substituted “cycloalkyl”, “cycloalkenyl”, “cycloalkynyl”, “heterocycloalkyl”, “heterocycloalkenyl”, “heterocycloalkynyl”, “aryl” and “heteroaryl” include spiro species.
  • “optionally substituted heteroalkyl” includes —CH 2 —N(Sub 1 )—CH 2 —, —CH(Sub 1 )—NH—CH 2 — and —CH(Sub 1 )—N(Sub 1 )—CH 2 — etc.
  • the modifier is to be understood as applying to each of the items in the list.
  • 20 -heteroaryl group means that each of the four items in the list, namely the C 3-20 -heterocycloalkyl group, the C 3-20 -heterocycloalkenyl group, the C 3-20 -heterocycloalkynyl group and the C 6-20 -heteroaryl group, may be optionally substituted.
  • a group is characterised by a first modifier and then, later on, the same group is characterised by a subsequent modifier, what is meant is that the group is characterised by both modifiers simultaneously.
  • a group is described as a “C 3-20 -heterocycloalkynyl” (the first modifier) group and then later the same group is described as a “C 5-16 ” (the subsequent modifier) group, what is meant is a C 5-16 heterocycloalkynyl group.
  • steroid refers to any group comprising the following structure (which structure is referred to herein as the “steroid skeleton”).
  • the steroid skeleton has been drawn above as fully saturated.
  • the term steroid is also intended to cover instances where there is unsaturation in the steroid skeleton.
  • the term steroid covers a group which comprises the fully unsaturated (mancude) basic skeleton, 15H-cyclopenta[a]phenanthrene:
  • steroid also covers a group which comprises a partially unsaturated steroid skeleton.
  • steroid also covers “seco” derivatives of the steroid skeleton, i.e. groups in which ring cleavage has been effected; “nor” and “homo” derivatives of the steroid skeleton which involve ring contraction and expansion, respectively (see Systemic Nomenclature of Organic Chemistry, by D. Hellwinkel, published by Springer, 2001, ISBN: 3-540-41138-0, page 203 for “seco” and page 204 for “nor” and “homo”).
  • seco derivatives are not encompassed by the term “steroid”.
  • nor derivatives are not encompassed by the term “steroid”.
  • such homo derivatives are not encompassed by the term “steroid”.
  • seco, nor and homo derivatives are not encompassed by the term “steroid”.
  • steroid also covers instances where one or more of the carbon atoms in the structure labelled steroid skeleton is replaced by a heteroatom.
  • up to six carbon atoms in one embodiment up to five carbon atoms, in another embodiment up to four carbon atoms, in another embodiment up to three carbon atoms, in another embodiment up to two carbon atoms, in another embodiment one carbon atom, are each replaced independently by O, S(O) q , N, P(O) r or Si (and preferably O, S(O) q or N).
  • the term “steroid” comprises species in which the “steroid basic skeleton” contains no heteroatoms.
  • a steroid ring system is numbered according to the convention set out below.
  • steroid encompasses sterols, steroid hormones, bile acids and salts of bile acids.
  • a sterol is any steroid with a hydroxyl group at the 3-position of the A-ring.
  • the omega-3 position refers to the third bond from the (methyl) terminal of the chain; the omega-6 position refers to the sixth bond from the (methyl) terminal of the chain and the omega-9 position refers to the ninth bond from the (methyl) terminal of the chain.
  • composition “comprising” encompasses “including” as well as “consisting” e.g. a composition “comprising” X may consist exclusively of X or may include something additional e.g. X+Y.
  • TLR3 is the Toll-like receptor 3. It is a single membrane-spanning receptor which plays a key role in the innate immune system.
  • Known TLR3 agonists include poly(I:C).
  • TLR3 is the approved HGNC name for the gene encoding this receptor, and its unique HGNC ID is HGNC:11849.
  • the RefSeq sequence for the human TLR3 gene is GI:2459625.
  • TLR7 is the Toll-like receptor 7. It is a single membrane-spanning receptor which plays a key role in the innate immune system.
  • Known TLR7 agonists include e.g. imiquimod.
  • TLR7 is the approved HGNC name for the gene encoding this receptor, and its unique HGNC ID is HGNC:15631.
  • the RefSeq sequence for the human TLR7 gene is GI:67944638.
  • TLR8 is the Toll-like receptor 8. It is a single membrane-spanning receptor which plays a key role in the innate immune system.
  • Known TLR8 agonists include e.g. resiquimod.
  • TLR8 is the approved HGNC name for the gene encoding this receptor, and its unique HGNC ID is HGNC:15632.
  • the RefSeq sequence for the human TLR8 gene is GI:20302165.
  • RLR-1 The RIG-I-like receptor (“RLR”) family includes various RNA helicases which play key roles in the innate immune system[40].
  • RLR-1 also known as RIG-I or retinoic acid inducible gene I
  • RLR-1 helicase has two caspase recruitment domains near its N-terminus.
  • the approved HGNC name for the gene encoding the RLR-1 helicase is “DDX58” (for DEAD (Asp-Glu-Ala-Asp) box polypeptide 58) and the unique HGNC ID is HGNC:19102.
  • the RefSeq sequence for the human RLR-1 gene is GI:77732514.
  • RLR-2 (also known as MDA5 or melanoma differentiation-associated gene 5) also has two caspase recruitment domains near its N-terminus.
  • the approved HGNC name for the gene encoding the RLR-2 helicase is “IFIH1” (for interferon induced with helicase C domain 1) and the unique HGNC ID is HGNC:18873.
  • the RefSeq sequence for the human RLR-2 gene is GI: 27886567.
  • RLR-3 (also known as LGP2 or laboratory of genetics and physiology 2) has no caspase recruitment domains.
  • the approved HGNC name for the gene encoding the RLR-3 helicase is “DHX58” (for DEXH (Asp-Glu-X-His) box polypeptide 58) and the unique HGNC ID is HGNC:29517.
  • the RefSeq sequence for the human RLR-3 gene is GI:149408121.
  • PKR is a double-stranded RNA-dependent protein kinase. It plays a key role in the innate immune system.
  • EIF2AK2 for eukaryotic translation initiation factor 2-alpha kinase 2
  • HGNC HGNC:9437
  • the RefSeq sequence for the human PKR gene is GI:208431825.
  • FIG. 1 shows a gel with stained RNA. Lanes show (1) markers (2) naked replicon (3) replicon after RNase treatment (4) replicon encapsulated in liposome (5) liposome after RNase treatment (6) liposome treated with RNase then subjected to phenol/chloroform extraction.
  • FIG. 2 is an electron micrograph of liposomes.
  • FIG. 3 shows protein expression (as relative light units, RLU) at days 1, 3 and 6 after delivery of RNA in liposomes With PEGs of different lengths: 1 kDa (triangles); 2 kDa (circles); 3 kDa (squares).
  • FIG. 4 shows a gel with stained RNA. Lanes show (1) markers (2) naked replicon (3 ) replicon encapsulated in liposome (4) liposome treated with RNase then subjected to phenol/chloroform extraction.
  • FIG. 6 shows protein expression at days 1, 3 and 6 after delivery of four different doses of liposome-encapsulated RNA.
  • FIG. 7 shows anti-F IgG titers in animals receiving virion-packaged replicon (VRP or VSRP), naked RNA, and 1 ⁇ g liposome-encapsulated RNA.
  • FIG. 8 shows anti-F IgG titers in animals receiving VRP, 1 ⁇ g naked RNA, and 0.1 g or liposome-encapsulated RNA.
  • FIG. 9 shows neutralising antibody titers in animals receiving VRP or either 0.1 g or 1 ⁇ g liposome-encapsulated RNA.
  • FIG. 10 shows expression levels after delivery of a replicon as naked RNA (circles), liposome-encapsulated RNA (triangle & square), or as a lipoplex (inverted triangle).
  • FIG. 11 shows F-specific IgG titers (2 weeks after second dose) after delivery of a replicon as naked RNA (0.01-1 ⁇ g), liposome-encapsulated RNA (0.01-10 ⁇ g), or packaged as a virion (VRP, 10 6 infectious units or IU).
  • FIG. 12 shows F-specific IgG titers (circles) and PRNT titers (squares) after delivery of a replicon as naked RNA (1 ⁇ g), liposome-encapsulated RNA (0.1 or 1 ⁇ g), or packaged as a virion (VRP, 10 6 IU). Titers in naive mice are also shown. Solid lines show geometric means.
  • FIG. 13 shows intracellular cytokine production after restimulation with synthetic peptides representing the major epitopes in the F protein, 4 weeks after a second dose.
  • the y-axis shows the % cytokine+ of CD8+CD4 ⁇ .
  • FIG. 14 shows the structure of lipid “RV05”.
  • FIG. 15 shows F-specific IgG titers (mean log 10 titers ⁇ std dev) over 210 days after immunisation of calves.
  • the three lines are easily distinguished at day 63 and are, from bottom to top: PBS negative control; liposome-delivered RNA; and the “Triangle 4” product.
  • FIG. 16 shows structures of three PEG-conjugated DMG lipids (1-3 kDa).
  • FIGS. 17A to 17E show structures of various PEG-conjugated lipids, where R is PEG of a desired length.
  • FIG. 18 shows the structure of a useful “split” PEG-conjugated lipid.
  • the box shows the total MW of PEG in the lipid (which, in the specific example below, was 2000).
  • replicons are used below. In general these are based on a hybrid alphavirus genome with non-structural proteins from venezuelan equine encephalitis virus (VEEV), a packaging signal from VEEV, and a 3′ UTR from Sindbis virus or a VEEV mutant.
  • VEEV venezuelan equine encephalitis virus
  • the replicon is about 10 kb long and has a poly-A tail.
  • Plasmid DNA encoding alphavirus replicons (named: pT7-mVEEV-FL.RSVF or A317; pT7-mVEEV-SEAP or A306; pSP6-VCR-GFP or A50) served as a template for synthesis of RNA in vitro.
  • the replicons contain the alphavirus genetic elements required for RNA replication but lack those encoding gene products necessary for particle assembly; the structural proteins are instead replaced by a protein of interest (either a reporter, such as SEAP or GFP, or an immunogen, such as full-length RSV F protein) and so the replicons are incapable of inducing the generation of infectious particles.
  • a bacteriophage (T7 or SP6) promoter upstream of the alphavirus cDNA facilitates the synthesis of the replicon RNA in vitro and a hepatitis delta virus (HDV) ribozyme immediately downstream of the poly(A)-tail generates the correct 3′-end through its self-cleaving activity.
  • HDV hepatitis delta virus
  • run-off transcripts were synthesized in vitro using T7 or SP6 bacteriophage derived DNA-dependent RNA polymerase. Transcriptions were performed for 2 hours at 37° C. in the presence of 7.5 mM (T7 RNA polymerase) or 5 mM (SP6 RNA polymerase) of each of the nucleoside triphosphates (ATP, CTP, GTP and UTP) following the instructions provided by the manufacturer (Ambion). Following transcription the template DNA was digested with TURBO DNase (Ambion).
  • RNA was precipitated with LiCI and reconstituted in nuclease-free water.
  • Uncapped RNA was capped post-transcriptionally with Vaccinia Capping Enzyme (VCE) using the ScriptCap m7G Capping System (Epicentre Biotechnologies) as outlined in the user manual; replicons capped in this way are given the “v” prefix e.g. vA317 is the A317 replicon capped by VCE.
  • Post-transcriptionally capped RNA was precipitated with LiCl and reconstituted in nuclease-free water. The concentration of the RNA samples was determined by measuring OD 260 nm . Integrity of the in vitro transcripts was confirmed by denaturing agarose gel electrophoresis.
  • RNA was encapsulated in liposomes made essentially by the method of references 7 and 41.
  • the liposomes were made of 10% DSPC (zwitterionic), 40% DlinDMA (cationic), 48% cholesterol and 2% PEG-conjugated DMG. These proportions refer to the % moles in the total liposome.
  • DlinDMA (1,2-dilinoleyloxy-N,N-dimethyl-3-aminopropane) was synthesized using the procedure of reference 2.
  • DSPC (1,2-Diastearoyl-sn-glycero-3-phosphocholine) was purchased from Genzyme. Cholesterol was obtained from Sigma-Aldrich.
  • PEG-conjugated DMG (1,2-dimyristoyl-sn-glycero-3-phosphoethanolamine-N-[methoxy(polyethylene glycol), ammonium salt), DOTAP (1,2-dioleoyl-3-trimethylammonium-propane, chloride salt) and DC-chol (3 ⁇ -[N-(N′,N′-dimethylaminoethane)-carbamoylicholesterol hydrochloride) were from Avanti Polar Lipids.
  • lipids were dissolved in ethanol (2 ml), a RNA replicon was dissolved in buffer (2 ml, 100 mM sodium citrate, pH 6) and these were mixed with 2 ml of buffer followed by 1 hour of equilibration.
  • FIG. 2 shows an example electron micrograph of liposomes prepared by these methods. These liposomes contain encapsulated RNA encoding full-length RSV F antigen. Dynamic light scattering of one batch showed an average diameter of 141 nm (by intensity) or 78 nm (by number).
  • fresh lipid stock solutions in ethanol were prepared.
  • 37 mg of DlinDMA, 11.8 mg of DSPC, 27.8 mg of Cholesterol and 8.07 mg of PEG-conjugated DMG were weighed and dissolved in 7.55 mL of ethanol.
  • Five different conjugated PEGs were used: PEG-500, PEG-750, PEG-1000, PEG-2000 or PEG-3000.
  • the freshly prepared lipid stock solution was gently rocked at 37° C. for about 15 min to form a homogenous mixture. Then, 226.7 ⁇ L of the stock was added to 1.773 mL ethanol to make a working lipid stock solution of 2 mL.
  • RNA was also prepared from a stock solution of ⁇ 1 ⁇ g/ ⁇ L in 100 mM citrate buffer (pH 6). Three 20 mL glass vials (with stir bars) were rinsed with RNase Away solution and washed with plenty of MilliQ water before use to decontaminate the vials of RNAses. One of the vials was used for the RNA working solution and the others for collecting the lipid and RNA mixes (as described later). The working lipid and RNA solutions were heated at 37° C. for 10 min before being loaded into 3cc luer-lok syringes. 2 mL of citrate buffer (pH 6) was loaded in another 3 cc syringe.
  • RNA and the lipids were connected to a T mixer (PEEKTM 500 ⁇ m ID junction) using FEP tubing(fluorinated ethylene-propylene; all FEP tubing used had a 2 mm internal diameter and a 3 mm outer diameter; obtained from Idex Health Science).
  • the outlet from the T mixer was also FEP tubing.
  • the third syringe containing the citrate buffer was connected to a separate piece of tubing. All syringes were then driven at a flow rate of 7 mL/min using a syringe pump. The tube outlets were positioned to collect the mixtures in a 20 mL glass vial (while stirring).
  • the stir bar was taken out and the ethanol/aqueous solution was allowed to equilibrate to room temperature for 1 hour. Then the mixture was loaded in a 5 cc syringe, which was fitted to a piece of FEP tubing and in another 5 cc syringe with equal length of FEP tubing, an equal volume of 100 mM citrate buffer (pH 6) was loaded. The two syringes were driven at 7 mL/min flow rate using a syringe pump and the final mixture collected in a 20 mL glass vial (while stirring).
  • liposomes were concentrated to 2 mL and dialyzed against 10-15 volumes of 1 ⁇ PBS using a Tangential Flow Filtration (TFF) system before recovering the final product.
  • TFF Tangential Flow Filtration
  • the TFF system and hollow fiber filtration membranes were purchased from Spectrum Labs and were used according to the manufacturer's guidelines. Hollow fiber filtration membranes with a 100 kD pore size cutoff and 20 cm 2 surface area were used.
  • formulations were diluted to the required RNA concentration with 1 ⁇ PBS.
  • RNA and RNA concentration were determined by Quant-iT RiboGreen RNA reagent kit (Invitrogen), following manufacturer's instructions.
  • the ribosomal RNA standard provided in the kit was used to generate a standard curve.
  • Liposomes were diluted 10 ⁇ or 100 ⁇ in 1 ⁇ TE buffer (from kit) before addition of the dye. Separately, liposomes were diluted 10 ⁇ or 100 ⁇ in 1 ⁇ TE buffer containing 0.5% Triton X before addition of the dye (to disrupt the liposomes and thus to assay total RNA). Thereafter an equal amount of dye was added to each solution and then ⁇ 180 ⁇ L of each solution after dye addition was loaded in duplicate into a 96 well tissue culture plate. The fluorescence (Ex 485 nm, Em 528 nm) was read on a microplate reader. All liposome formulations were dosed in vivo based on the encapsulated amount of RNA.
  • the syringe/tube method was replaced by a method in which the lipid and RNA solutions are mixed in channels on a microfluidic chip.
  • Fresh lipid stock solutions in ethanol were prepared. 37 mg of DlinDMA, 11.8 mg of DSPC, 27.8 mg of cholesterol and 8.07 mg of PEG-DMG were weighed and dissolved in 7.55 mL of ethanol. The freshly prepared lipid stock solution was gently rocked at 37° C. for about 15 min to form a homogenous mixture. Then, 226.7 ⁇ L of the stock was added to 1.773 mL ethanol to make a working lipid stock solution of 2 mL.
  • RNA working solution was also prepared from a stock solution of ⁇ 1 ⁇ / ⁇ L in 100 mM citrate buffer (pH 6).
  • Four 20 mL glass vials (with stir bars) were rinsed with RNase Away solution and washed with plenty of MilliQ water before use to decontaminate the vials of RNAses.
  • Two of the vials were used for the RNA working solution (2 mL in each vial) and the others for collecting the lipid and RNA mixes.
  • the working lipid and RNA solutions were heated at 37° C. for 10 min before being loaded into 3cc luer-lok syringes.
  • RNA and the lipids were connected to a Mitos Droplet junction Chip (a glass microfluidic device obtained from Syrris, Part no. 3000158) using PTFE tubing 0.03 inches ID ⁇ 1/16 inch OD, (Syrris) using a 4-way edge connector.
  • Two RNA streams and one lipid stream were driven by syringe pumps and the mixing of the ethanol and aqueous phase was done at the X junction (100 ⁇ m ⁇ 105 ⁇ m) of the chip.
  • the flow rate of all three streams was kept at 1.5 ml/min, hence the ratio of total aqueous to ethanolic flow rate was 2:1.
  • the tube outlet was positioned to collect the mixtures in a 20 mL glass vial (while stirring).
  • the stir bar was taken out and the ethanol/aqueous solution was allowed to equilibrate to room temperature for 1 hour.
  • the mixture was loaded in a 5 cc syringe which was fitted to a piece of PTFE tubing 0.03 inches ID x 1/16 inches OD and in another 5 cc syringe with equal length of PTFE tubing, an equal volume of 100 mM citrate buffer (pH 6) was loaded.
  • the two syringes were driven at 3 mllmin flow rate using a syringe pump and the final mixture collected in a 20 mL glass vial (while stirring).
  • liposomes were concentrated to 2 mL and dialyzed against 10-15 volumes of 1 ⁇ PBS using the TFF system before recovering the final product. Hollow fiber filtration membranes with a 100 kDa pore size cutoff and 20 cm 2 surface area were used.
  • formulations were diluted to the required RNA concentration with 1 ⁇ PBS.
  • liposomes prepared using the syringe/tube method with 75 ⁇ g RNA had a Z-average diameter (Zav) of 148 nm and a polydispersity index (pdI) of 0.122
  • the chip mixing gave liposomes with a Zav of 97 nm and a pdI of 0.086.
  • the proportion of encapsulated RNA decreased slightly from 90% to 87%.
  • RNA from liposomes was shown to protect RNA from RNase digestion. Experiments used 3.8 mAU of RNase A per microgram of RNA, incubated for 30 minutes at room temperature. RNase was inactivated with Proteinase K at 55° C. for 10 minutes. A 1:1 v/v mixture of sample to 25:24:1 v/v/v, phenol:chloroform:isoamyl alcohol was then added to extract the RNA from the lipids into the aqueous phase. Samples were mixed by vortexing for a few seconds and then placed on a centrifuge for 15 minutes at 12 k RPM. The aqueous phase (containing the RNA) was removed and used to analyze the RNA.
  • FIG. 1 shows that RNase completely digests RNA in the absence of encapsulation (lane 3). RNA is undetectable after encapsulation (lane 4), and no change is seen if these liposomes are treated with RNase (lane 4). After RNase-treated liposomes are subjected to phenol extraction, undigested RNA is seen (lane 6).
  • RNA Even after 1 week at 4° C. the RNA could be seen without any fragmentation ( FIG. 4 , arrow). Protein expression in vivo was unchanged after 6 weeks at 4° C. and one freeze-thaw cycle. Thus liposome-encapsulated RNA is stable.
  • RNA a reporter enzyme SEAP; secreted alkaline phosphatase
  • SEAP secreted alkaline phosphatase
  • Expression levels were measured in sera diluted 1:4 in 1 ⁇ Phospha-Light dilution buffer using a chemiluminescent alkaline phosphate substrate. 8-10 week old BALB/c mice (5/group) were injected intramuscularly on day 0, 50 ⁇ l per leg with 0.1 ⁇ g or 1 ⁇ g RNA dose. The same vector was also administered without the liposomes (in RNase free 1X PBS) at 1 ⁇ g. Virion-packaged replicons were also tested.
  • Virion-packaged replicons used herein were obtained by the methods of reference 42, where the alphavirus replicon is derived from the mutant VEEV or a chimera derived from the genome of VEEV engineered to contain the 3′ UTR of Sindbis virus and a Sindbis virus packaging signal (PS), packaged by co-electroporating them into BHK cells with defective helper RNAs encoding the Sindbis virus capsid and glycoprotein genes.
  • PS Sindbis virus packaging signal
  • encapsulation increased SEAP levels by about 1 ⁇ 2 log at the 1 ⁇ g dose, and at day 6 expression from a 0.1 ⁇ g encapsulated dose matched levels seen with 1 ⁇ g unencapsulated dose.
  • day 3 expression levels exceeded those achieved with VRPs (squares).
  • VRPs squares
  • RNA was formulated in the liposomes relative to the naked RNA control, even at a 10 ⁇ lower dose. Expression was also higher relative to the VRP control, but the kinetics of expression were very different (see FIG. 5 ). Delivery of the RNA with electroporation resulted in increased expression relative to the naked RNA control, but these levels were lower than with liposomes.
  • the replicon was administered in encapsulated form (with two different purification protocols, 0.1 ⁇ g RNA), or mixed with the liposomes after their formation (a non-encapsulated “lipoplex”, 0.1 ⁇ g RNA), or as naked RNA (1 ⁇ g).
  • FIG. 10 shows that the lipoplex gave the lowest levels of expression, showing that shows encapsulation is essential for potent expression.
  • FIG. 7 shows anti-F IgG titers 2 weeks after the second dose, and the liposomes clearly enhance immunogenicity.
  • FIG. 8 shows titers 2 weeks later, by which point there was no statistical difference between the encapsulated RNA at 0.1 ⁇ g, the encapsulated RNA at 1 ⁇ g, or the VRP group.
  • Neutralisation titers (measured as 60% plaque reduction, “PRNT60”) were not significantly different in these three groups 2 weeks after the second dose ( FIG. 9 ).
  • FIG. 12 shows both IgG and PRNT titers 4 weeks after the second dose.
  • FIG. 13 confirms that the RNA elicits a robust CD8 T cell response.
  • VRP liposome
  • liposome-encapsulated RNA induces essentially the same magnitude of immune response as seen with virion delivery.
  • FIG. 11 shows IgG titers in mice receiving the replicon in naked form at 3 different doses, in liposomes at 4 different doses, or as VRP (10 6 IU).
  • the response seen with 1 ⁇ g liposome-encapsulated RNA was statistically insignificant (ANOVA) when compared to VRP, but the higher response seen with 10 ⁇ g liposome-encapsulated RNA was statistically significant (p ⁇ 0.05) when compared to both of these groups.
  • IgG responses (15 days post-second dose) than 0.1 ⁇ g of delivered DNA, and even was more immunogenic than 20 ⁇ g plasmid DNA encoding the F antigen, delivered by electroporation (ElgenTM DNA Delivery System, Inovio).
  • the vA317 self-replicating replicon encoding RSV F protein was administered to BALB/c mice, 4 or 8 animals per group, by bilateral intramuscular vaccinations (50 ⁇ L per leg) on days 0 and 21 with the replicon (1 ⁇ g) alone or formulated as liposomes with DlinDMA (“RV01”) or DOTAP (“RV13”) or the lipid shown in FIG. 14 (“RV05”).
  • the RV01 liposomes had 40% DlinDMA, 10% DSPC, 48% cholesterol and 2% PEG-DMG, but with differing amounts of RNA.
  • the RV05 liposomes had either 40% RV05, 10% DSPC, 48% cholesterol and 2% PEG-DMG or 60% RV05, 38% cholesterol and 2% PEG-DMG.
  • the RV13 liposomes had 40% DOTAP, 10% DOPE, 48% cholesterol and 2% PEG-DMG.
  • the PEG was PEG-2000 (i.e. 2 kDa PEG).
  • naked plasmid DNA (20 ⁇ g) expressing the same RSV-F antigen was delivered either using electroporation or with RV01(10) liposomes (0.1 ⁇ g DNA).
  • RV01(10) liposomes 0.1 ⁇ g DNA.
  • Liposomes were prepared by method (A) or method (B). For some liposomes made by method (A) a double or half amount of RNA was used. Z average particle diameter and polydispersity index were:
  • F-specific serum IgG titers were as follows:
  • RV Day 14 Day 36 Naked DNA plasmid 439 6712 Naked A317 RNA 78 2291 RV01 (10) 3020 26170 RV01 (08) 2326 9720 RV01 (05) 5352 54907 RV01 (09) 4428 51316 RV05 (01) 1356 5346 RV05 (02) 961 6915 RV01 (10) DNA 5 13 RV13 (02) 644 3616
  • T cells which are cytokine-positive and specific for RSV F51-66 peptide are as follows, showing only figures which are statistically significantly above zero:
  • liposome formulations significantly enhanced immunogenicity relative to the naked RNA controls, as determined by increased F-specific IgG titers and T cell frequencies.
  • RV01 liposomes were prepared by method (H), again using 2 kDa PEG conjugated to DMG, and either encapsulating 150 ⁇ g RNA (vA375 replicon encoding surface fusion glycoprotein of RSV) or encapsulating only buffer. Thus these liposomes had 40% DlinDMA, 10% DSPC, 48% Chol, and 2% PEG-DMG. Sizes and encapsulation were as follows:
  • the liposomes were administered to BALB/c mice (10 per group) by bilateral intramuscular injection (50 ⁇ l per leg) on days 0 & 21. Doses were 0.01, 0.03, 0.1, 0.3 or 1 ⁇ g. F-specific serum IgG and PRNT60 titers (GMT) were as follows, 2 weeks after the first or second injection:
  • RV RNA ( ⁇ g) 2wp1 2wp2 PRNT60 (2wp2) Buffer control 0 — — 10 RV01 (36) 0 — — 10 RV01 (36) 0.01 3399 50691 37 RV01 (36) 0.03 3446 53463 83 RV01 (36) 0.1 8262 76808 238 RV01 (36) 0.3 5913 82599 512 RV01 (36) 1 8213 85138 441
  • RV01 liposomes with DLinDMA as the cationic lipid and 2 kDa PEG were used to deliver RNA replicons encoding CMV glycoproteins.
  • the “vA160” replicon encodes full-length glycoproteins H and L (gH/gL), whereas the “vA322” replicon encodes a soluble form (gHsol/gL).
  • the two proteins are under the control of separate subgenomic promoters in a single replicon; co-administration of two separate vectors, one encoding gH and one encoding gL, did not give good results.
  • mice 10 per group, were given bilateral intramuscular vaccinations (50 ⁇ L per leg) on days 0, 21 and 42 with VRPs expressing gH/gL (1 ⁇ 10 6 IU), VRPs expressing gHsol/gL (1 ⁇ 10 6 IU) and PBS as the controls.
  • Two test groups received 1 ⁇ g of the vA160 or vA322 replicon formulated in liposomes (40% DlinDMA, 10% DSPC, 48% Chol, 2% PEG-DMG; made using method (D) but with 150 ⁇ g RNA batch size).
  • the vA160 liposomes had a Zav diameter of 168.8 nm, a pdI of 0.144, and 87.4% encapsulation.
  • the vA322 liposomes had a Zav diameter of 162 nm, a pdI of 0.131, and 90% encapsulation.
  • the replicons were able to express two proteins from a single vector.
  • CMV neutralization titers (the reciprocal of the serum dilution producing a 50% reduction in number of positive virus foci per well, relative to controls) were as follows:
  • RNA expressing either a full-length or a soluble form of the CMV gH/gL complex thus elicited high titers of neutralizing antibodies, as assayed on epithelial cells.
  • the average titers elicited by the liposome-encapsulated RNAs were at least as high as for the corresponding VRPs.
  • RNA replicon was able to express two proteins from a single vector.
  • the RNA replicon gave a 3wp3 titer of 11457, compared to 5516 with VRPs.
  • vA311 A self-replicating RNA replicon (“vA311”) that expresses a luciferase reporter gene (luc) was used for studying the kinetics of protein expression after injection.
  • mice Prior to vaccination mice were depilated. Mice were anesthetized (2% isoflurane in oxygen), hair was first removed with an electric razor and then chemical Nair. Bioluminescence data was then acquired using a Xenogen IVIS 200 imaging system (Caliper Life Sciences) on days 3, 7, 14, 21, 28, 35, 42, 49, 63 and 70. Five minutes prior to imaging mice were injected intraperitbneally with 8 mg/kg of luciferin solution. Animals were then anesthetized and transferred to the imaging system. Image acquisition times were kept constant as bioluminescence signal was measured with a cooled CCD camera.
  • luciferase-expressing cells were seen to remain primarily at the site of RNA injection, and animals imaged after removal of quads showed no signal.
  • luciferase expression was measured as average radiance over a period of 70 days (p/s/cm 2 /sr), and results were as follows for the 5 groups:
  • RNA formulated with cationic nanoemulsions showed measurable bioluminescence at day 3, which peaked at day 7 and then reduced to background levels by days 28 , to 35.
  • the RNA When formulated in liposomes the RNA showed measurable bioluminescence at day 3, which peaked at day 7 and reduced to background levels by day 63.
  • RNA delivered using VRPs showed enhanced bioluminescence at day 21 when compared to the formulated RNA, but expression had reduced to background levels by day 28.
  • Electroporated DNA showed the highest level of bioluminescence at all time points measured and levels of bioluminescence did not reduce to background levels within the 70 days of the experiment.
  • Hydrodynamic delivery employs the force generated by the rapid injection of a large volume of solution to overcome the physical barriers of cell membranes which prevent large and membrane-impermeable compounds from entering cells. This phenomenon has previously been shown to be useful for the intracellular delivery of DNA vaccines.
  • a typical mouse delivery volume for intramuscular injection is 50 ⁇ l into the hind leg, which is a relatively high volume for a mouse leg muscle.
  • a human intramuscular dose of ⁇ 0.5 ml is relatively small. If immunogenicity in mice would be volume-dependent then the replicon vaccines' efficacy might be due, at least in part, on hydrodynamic forces, which would not be encouraging for use of the same vaccines in humans and larger animals.
  • the vA317 replicon was delivered to BALB/c mice, 10 per group, by bilateral intramuscular vaccinations (5 or 50 per leg) on day 0 and 21:
  • the liposomes for groups 5 & 6 were 40% DlinDMA, 10% DSPC, 48% cholesterol, and 2% PEG-2000 conjugated to DMG.
  • F-specific serum IgG GMTs were:
  • vA317 expresses full-length RSV-F
  • vA318 expresses truncated (transmembrane and cytoplasmic tail removed) RSV-F
  • vA142 expresses RSV-F with its fusion peptide deleted
  • vA140 expresses the truncated RSV-F also without its peptide.
  • Control groups received a RSV-F subunit protein vaccine (5 ⁇ g) adjuvanted with alum (8 animals/group), VRPs expressing full-length RSV-F (1 ⁇ 10 6 IU, 8 animals/group), or naive control (4 animals/group). Serum was collected for antibody analysis on days 0, 21 and 34.
  • F-specific serum IgG titers and RSV serum neutralizing antibody titers on day 21 and 34 were:
  • All four replicons evaluated in this study were immunogenic in cotton rats when delivered by liposome, although serum neutralization titers were at least ten-fold lower than those induced by adjuvanted protein vaccines or by VRPs.
  • the liposome/RNA vaccines elicited serum F-specific IgG and RSV neutralizing antibodies after the first vaccination, and a second vaccination boosted the response effectively.
  • F-specific IgG titers after the second vaccination with 1 ⁇ g replicon were 2- to 3-fold higher than after the second vaccination with 0.1 ⁇ g replicon.
  • the four replicons elicited comparable antibody titers, suggesting that full length and truncated RSV-F, each with or without the fusion peptide, are similarly immunogenic in cotton rats.
  • Cotton rats again used the vA317, vA318 and vA142 replicons.
  • Cotton rats, 2-8 animals per group were given intramuscular vaccinations (100 ⁇ L in one leg) on days 0 and 21 with the replicons (0.1 or 1 ⁇ g) encapsulated in RV01 liposomes (with PEG-2000) made by method (D) but with a 150 ⁇ g RNA batch size.
  • Control groups received the RSV-F subunit protein vaccine (5 ⁇ g) adjuvanted with alum or VRPs expressing full-length RSV-F (1 ⁇ 10 6 IU, 8 animals/group).
  • F-specific serum IgG titers were as follows:
  • Serum neutralisation titers were as follows (60% RSV neutralization titers for 2 pools of 3-4 animals per group, GMT of these 2 pools per group):
  • Serum titers and neutralising titers for the extra group were as follows:
  • the replicons are confirmed as immunogenic in cotton rats, eliciting serum F-specific IgG and RSV neutralizing antibodies after the first vaccination.
  • a second vaccination boosted the responses effectively.
  • F-specific IgG titers after the second vaccination with 1.0 ⁇ g replicon were 1.5 to 4-fold higher than after the second vaccination with 0.1 ⁇ g replicon.
  • the third vaccination did not boost titers in cotton rats previously vaccinated with F trimer subunit+alum, but it did provide a large boost to titers in cotton rats previously vaccinated with replicon.
  • the RSV serum neutralization titers after two replicon vaccinations followed by protein boost were equal to or greater than titers induced by two or three sequential protein vaccinations.
  • F-specific serum IgG and RSV neutralization titers induced by a single vaccination reached their peak around day 21 and were maintained through at least day 56 (50-70% drop in F-specific IgG titer, little change in RSV neutralization titer).
  • a homologous second vaccination was given to these animals on day 56, and boosted antibody titers to a level at least equal to that achieved when the second vaccination was administered on day 21.
  • vA368 replicon encodes the full-length wild type surface fusion glycoprotein of RSV with the fusion peptide deleted, with expression driven by the EV71 IRES.
  • the liposomes included 2 kDa PEG, conjugated to DMG.
  • a control group received 5 ⁇ g alum-adjuvanted protein, and a na ⁇ ve control group was also included.
  • RNA vaccine reduced the lung viral load by over three logs, from approximately 10 6 PFU/g in unvaccinated control cotton rats to less than 10 3 PFU/g in vaccinated cotton rats.
  • RNA vaccines encoded human RSV F whereas the “Triangle 4” vaccine contains bovine RSV F, but the RSV F protein is highly conserved between BRSV and HRSV.
  • Serum was collected for antibody analysis on days 0, 14, 21, 35, 42, 56, 63, 86, 100, 107, 114, 121, 128, 135, 146, 160, 167, 174, 181, 188, 195, and 202. If an individual animal had a titer below the limit of detection it was assigned a titer of 5.
  • FIG. 15 shows F-specific IgG titers over 210 days. Over the first 63 days the RNA replicon was immunogenic in the cows via liposomes, although it gave lower titers than the licensed vaccine. All vaccinated cows showed F-specific antibodies after the second dose, and titers were very stable from the period of 2 to 6 weeks after the second dose (and were particularly stable for the RNA vaccines). Titres up to day 202 were as follows:
  • RSV serum neutralizing antibody titers were as follows:
  • the material used for the second liposome dose was not freshly prepared, and the same lot of RNA showed a decrease in potency in a mouse immunogenicity study. Therefore it is possible that the vaccine would have been more immunogenic if fresh material had been used for all vaccinations.
  • MF59-adjuvanted RSV-F was able to boost the IgG response in all previously vaccinated calves, and to boost complement-independent neutralization titers of calves previously vaccinated with RNA.
  • RNA vaccines in large animals is particularly important in light of the loss in potency observed previously with DNA-based vaccines when moving from small animal models to larger animals and humans.
  • a typical dose for a cow DNA vaccine would be 0.5-1 mg [43, 44] and so it is very encouraging that immune responses were induced with only 66ps of RNA.
  • liposomes were prepared using DMG to which five different PEGs were conjugated.
  • the average molecular weight of the PEG was 500 Da, 750 Da, 1 kDa, 2 kDa or 3 kDa.
  • Liposomes formed using the shortest PEGs were unstable or aggregated during TFF purification.
  • PEG-750 gave liposomes with a significantly higher Zaverage diameter (669 nm) and polydispersity index (0.21), with 77% encapsulation.
  • the PEG-500 liposomes visibly aggregated in solution during the TFF process and the experiment was terminated. Thus these short PEG liposomes were unstable, but the longer PEGs formed stable liposomes.
  • the different PEG lengths ( FIG. 16 ) had a small effect on liposome diameter and polydispersity index.
  • the Z-average diameter was 197 nm (0.119 pdI) for the 1 kDa PEG, 142 nm (0.137 pdI) for the 2 kDa PEG, and 147 nm (0.075 pdI) for the 3 kDa PEG.
  • RNA encapsulation increased gradually as the PEG length increased, from 81.7% to 85.9% to 91.5% (although this relationship was not always seen in subsequent experiments).
  • the liposomes were administered to mice by intramuscular injection on day 0. Serum SEAP levels were measured at days 1, 3 and 6 by chemiluminescent assay. As shown in FIG. 3 , the three PEG lengths were all effective, but varying the length of the PEG had some effect on serum SEAP levels, with PEG 2000 giving the highest expression.
  • the vA317 replicon was administered in liposomes having a variety of different lipids with different PEG lengths.
  • the liposomes all had 40% DlinDMA, 10% DSPC and 48% cholesterol, but the remaining 2% was varied, with different PEGylated lipids (e.g. FIGS. 17A to 17E ) and different PEG lengths.
  • FIG. 17D 2000 122.2 0.122 77.84
  • FIG. 17E 2000 129.8 0.125 82.57
  • FIG. 17B 2000 113.4 0.091 89.12
  • mice 8 per group, were given bilateral intramuscular vaccinations (50 ⁇ L per leg) on days 0 and 21 with the replicon,. either naked (1 ⁇ g) or encapsulated in these liposomes (0.1 ⁇ g). Serum was collected for antibody analysis on days 14, and 35.
  • F-specific serum IgG titers were as follows, 2 weeks after the two injections (2wp1):
  • RV 2wp1 2wp2 Naked RNA 216 1356 A 3271 15659 B 3860 22378 C 1691 7412 D 1025 1767 E 1618 9536 F 2684 11221 G 3514 10566 H 4142 22810 I 952 10410
  • the liposomes had 40% cationic lipid (RV05) and 2% PEGylated DMG (2 kDa PEG), while the remaining components varied (but cholesterol was always included).
  • the liposomes were made by method (H) but with pH 5. Physical characteristics were:
  • mice 8 per group, were given bilateral intramuscular vaccinations (50 ⁇ L per leg) on days 0 and 21 with the replicon, either naked (1 ⁇ g) or encapsulated (0.1 ⁇ g). Serum was collected for antibody analysis on days 14, and 35.
  • F-specific serum IgG titers were as follows, 2 weeks after the two injections (2wp1):
  • RV 2wp1 2wp2 Naked RNA 321 915 A 2761 17040 B 866 3657 C 1734 5209 D 426 2079 E 2696 15794 F 551 955 G 342 2531 H 1127 3881 I 364 1741 J 567 5679 K 1251 5303
  • DLOPC 1,2-Linoleoyl-sn-Glycero-3-phosphatidylcholine
  • DLPA 1,2-Dilauroyl-sn-Glycero-3-Phosphate
  • DLPC 1,2-Dilauroyl-sn-Glycero-3-phosphatidylcholine
  • DLPE 1,2-Dilauroyl-sn-Glycero-3-phosphatidylethanolamine
  • DLPG 1,2-Dilauroyl-sn-Glycero-3[Phosphatidyl-rac-(1- glycerol . .
  • DLPS 1,2-Dilauroyl-sn-Glycero-3-phosphatidylserine
  • DMG 1,2-Dimyristoyl-sn-glycero-3-phosphoethanolamine
  • DMPA 1,2-Dimyristoyl-sn-Glycero-3-Phosphate
  • DMPC 1,2-Dimyristoyl-sn-Glycero-3-phosphatidylcholine
  • DMPE 1,2-Dimyristoyl-sn-Glycero-3-phosphatidylethanolamine
  • DMPG 1,2-Myristoyl-sn-Glycero-3[Phosphatidyl-rac-(1- glycerol . .
  • DMPS 1,2-Dimyristoyl-sn-Glycero-3-phosphatidylserine
  • DOPA 1,2-Dioleoyl-sn-Glycero-3-Phosphate
  • DOPC 1,2-Dioleoyl-sn-Glycero-3-phosphatidylcholine
  • DOPE 1,2-Dioleoyl-sn-Glycero-3-phosphatidylethanolamine
  • DOPG 1,2-Dioleoyl-sn-Glycero-3[Phosphatidyl-rac-(1- glycerol . .
  • DPPS 1,2-Dipalmitoyl-sn-Glycero-3-phosphatidylserine
  • DPyPE 1,2-diphytanoyl-sn-glycero-3-phosphoethanolamine
  • DSPA 1,2-Distearoyl-sn-Glycero-3-Phosphate
  • DSPC 1,2-Distearoyl-sn-Glycero-3-phosphatidylcholine
  • DSPE 1,2-Diostearpyl-sn-Glycero-3-phosphatidylethanolamine
  • DSPG 1,2-Distearoyl-sn-Glycero-3[Phosphatidyl-rac-(1- glycerol . .
  • PSPC 1-Palmitoyl,2-stearoyl-sn-Glycero-3-phosphatidylcholine
  • SMPC 1-Stearoyl,2-myristoyl-sn-Glycero-3-phosphatidylcholine
  • SOPC 1-Stearoyl,2-oleoyl-sn-Glycero-3-phosphatidylcholine
  • SPPC 1-Stearoyl,2-palmitoyl-sn-Glycero-3-phosphatidylcholine

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Immunology (AREA)
  • Organic Chemistry (AREA)
  • Microbiology (AREA)
  • Genetics & Genomics (AREA)
  • Virology (AREA)
  • Engineering & Computer Science (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Mycology (AREA)
  • Dispersion Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Engineering & Computer Science (AREA)
  • Wood Science & Technology (AREA)
  • Biomedical Technology (AREA)
  • Zoology (AREA)
  • Biotechnology (AREA)
  • Oncology (AREA)
  • Communicable Diseases (AREA)
  • Pulmonology (AREA)
  • Plant Pathology (AREA)
  • Physics & Mathematics (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • Dermatology (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Medicinal Preparation (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
US13/819,077 2010-08-31 2011-08-31 Pegylated liposomes for delivery of immunogen encoding rna Abandoned US20130202684A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US13/819,077 US20130202684A1 (en) 2010-08-31 2011-08-31 Pegylated liposomes for delivery of immunogen encoding rna

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US37882610P 2010-08-31 2010-08-31
US13/819,077 US20130202684A1 (en) 2010-08-31 2011-08-31 Pegylated liposomes for delivery of immunogen encoding rna
PCT/US2011/050095 WO2012031043A1 (en) 2010-08-31 2011-08-31 Pegylated liposomes for delivery of immunogen-encoding rna

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2011/050095 A-371-Of-International WO2012031043A1 (en) 2010-08-31 2011-08-31 Pegylated liposomes for delivery of immunogen-encoding rna

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US16/837,115 Continuation US11759422B2 (en) 2010-08-31 2020-04-01 Pegylated liposomes for delivery of immunogen-encoding RNA

Publications (1)

Publication Number Publication Date
US20130202684A1 true US20130202684A1 (en) 2013-08-08

Family

ID=44653554

Family Applications (2)

Application Number Title Priority Date Filing Date
US13/819,077 Abandoned US20130202684A1 (en) 2010-08-31 2011-08-31 Pegylated liposomes for delivery of immunogen encoding rna
US16/837,115 Active 2031-12-31 US11759422B2 (en) 2010-08-31 2020-04-01 Pegylated liposomes for delivery of immunogen-encoding RNA

Family Applications After (1)

Application Number Title Priority Date Filing Date
US16/837,115 Active 2031-12-31 US11759422B2 (en) 2010-08-31 2020-04-01 Pegylated liposomes for delivery of immunogen-encoding RNA

Country Status (20)

Country Link
US (2) US20130202684A1 (pl)
EP (11) EP4066856B1 (pl)
JP (1) JP5911870B2 (pl)
CN (1) CN103179984A (pl)
AU (1) AU2011295935B2 (pl)
BR (1) BR112013004866A2 (pl)
CA (1) CA2809851A1 (pl)
DK (6) DK3981427T3 (pl)
ES (6) ES2938866T3 (pl)
FI (2) FI4066857T3 (pl)
HR (6) HRP20230185T8 (pl)
HU (6) HUE061043T2 (pl)
LT (6) LT3981427T (pl)
MX (1) MX2013002336A (pl)
PL (6) PL3981427T3 (pl)
PT (6) PT2611461T (pl)
RS (6) RS63367B1 (pl)
RU (1) RU2013114362A (pl)
SI (6) SI4066856T1 (pl)
WO (1) WO2012031043A1 (pl)

Cited By (138)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20130195969A1 (en) * 2010-08-31 2013-08-01 Novartis Ag Small liposomes for delivery of immunogen encoding rna
US8664194B2 (en) 2011-12-16 2014-03-04 Moderna Therapeutics, Inc. Method for producing a protein of interest in a primate
US8710200B2 (en) 2011-03-31 2014-04-29 Moderna Therapeutics, Inc. Engineered nucleic acids encoding a modified erythropoietin and their expression
US20140193484A1 (en) * 2013-01-10 2014-07-10 Sylvie Carine Bertholet Girardin Influenza virus immunogenic compositions and uses thereof
US20140227346A1 (en) * 2011-07-06 2014-08-14 Andrew Geall Immunogenic combination compositions and uses thereof
US8822663B2 (en) 2010-08-06 2014-09-02 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
WO2014152211A1 (en) 2013-03-14 2014-09-25 Moderna Therapeutics, Inc. Formulation and delivery of modified nucleoside, nucleotide, and nucleic acid compositions
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
US8980864B2 (en) 2013-03-15 2015-03-17 Moderna Therapeutics, Inc. Compositions and methods of altering cholesterol levels
US8999380B2 (en) 2012-04-02 2015-04-07 Moderna Therapeutics, Inc. Modified polynucleotides for the production of biologics and proteins associated with human disease
WO2015051214A1 (en) 2013-10-03 2015-04-09 Moderna Therapeutics, Inc. Polynucleotides encoding low density lipoprotein receptor
WO2015109325A1 (en) * 2014-01-20 2015-07-23 University Of Utah Research Foundation Compositions and methods for modifying the surface of cells 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
WO2015100269A3 (en) * 2013-12-27 2015-11-12 Double Helix Corporation Compositions and methods for providing active telomerase to cells in vivo
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
US9334328B2 (en) 2010-10-01 2016-05-10 Moderna Therapeutics, 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
US9464124B2 (en) 2011-09-12 2016-10-11 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US9512456B2 (en) 2012-08-14 2016-12-06 Modernatx, Inc. Enzymes and polymerases for the synthesis of RNA
US9572897B2 (en) 2012-04-02 2017-02-21 Modernatx, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
US9597380B2 (en) 2012-11-26 2017-03-21 Modernatx, Inc. Terminally modified RNA
WO2017112943A1 (en) 2015-12-23 2017-06-29 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
US9872900B2 (en) 2014-04-23 2018-01-23 Modernatx, Inc. Nucleic acid vaccines
WO2018033254A2 (en) 2016-08-19 2018-02-22 Curevac Ag Rna for cancer therapy
WO2018104538A1 (en) 2016-12-08 2018-06-14 Curevac Ag Rna for treatment or prophylaxis of a liver disease
WO2018104540A1 (en) 2016-12-08 2018-06-14 Curevac Ag Rnas for wound healing
WO2018115525A1 (en) 2016-12-23 2018-06-28 Curevac Ag Lassa virus vaccine
WO2018115527A2 (en) 2016-12-23 2018-06-28 Curevac Ag Mers coronavirus vaccine
US10023626B2 (en) 2013-09-30 2018-07-17 Modernatx, Inc. Polynucleotides encoding immune modulating polypeptides
US10064934B2 (en) 2015-10-22 2018-09-04 Modernatx, Inc. Combination PIV3/hMPV RNA vaccines
US10064935B2 (en) 2015-10-22 2018-09-04 Modernatx, Inc. Human cytomegalovirus RNA vaccines
WO2018167320A1 (en) 2017-03-17 2018-09-20 Curevac Ag Rna vaccine and immune checkpoint inhibitors for combined anticancer therapy
WO2018172556A1 (en) 2017-03-24 2018-09-27 Curevac Ag Nucleic acids encoding crispr-associated proteins and uses thereof
US10124055B2 (en) 2015-10-22 2018-11-13 Modernatx, Inc. Zika virus RNA vaccines
WO2018213789A1 (en) 2017-05-18 2018-11-22 Modernatx, Inc. Modified messenger rna comprising functional rna elements
WO2018213731A1 (en) 2017-05-18 2018-11-22 Modernatx, Inc. Polynucleotides encoding tethered interleukin-12 (il12) polypeptides and uses thereof
WO2018222890A1 (en) 2017-05-31 2018-12-06 Arcturus Therapeutics, Inc. Synthesis and structure of high potency rna therapeutics
WO2018232006A1 (en) 2017-06-14 2018-12-20 Modernatx, Inc. Polynucleotides encoding coagulation factor viii
EP3424524A2 (en) 2017-07-04 2019-01-09 CureVac AG Cancer rna-vaccine
US10195156B2 (en) 2015-12-22 2019-02-05 Modernatx, Inc. Compounds and compositions for intracellular delivery of agents
US10207010B2 (en) 2015-12-10 2019-02-19 Modernatx, Inc. Compositions and methods for delivery of agents
US10266485B2 (en) 2015-09-17 2019-04-23 Modernatx, Inc. Compounds and compositions for intracellular delivery of therapeutic agents
WO2019077001A1 (en) 2017-10-19 2019-04-25 Curevac Ag NEW ARTIFICIAL NUCLEIC ACID MOLECULES
US10273269B2 (en) 2017-02-16 2019-04-30 Modernatx, Inc. High potency immunogenic zika virus compositions
WO2019104152A1 (en) 2017-11-22 2019-05-31 Modernatx, Inc. Polynucleotides encoding ornithine transcarbamylase for the treatment of urea cycle disorders
WO2019104160A2 (en) 2017-11-22 2019-05-31 Modernatx, Inc. Polynucleotides encoding phenylalanine hydroxylase for the treatment of phenylketonuria
WO2019104195A1 (en) 2017-11-22 2019-05-31 Modernatx, Inc. Polynucleotides encoding propionyl-coa carboxylase alpha and beta subunits for the treatment of propionic acidemia
WO2019136241A1 (en) 2018-01-05 2019-07-11 Modernatx, Inc. Polynucleotides encoding anti-chikungunya virus antibodies
WO2019200171A1 (en) 2018-04-11 2019-10-17 Modernatx, Inc. Messenger rna comprising functional rna elements
US10449244B2 (en) 2015-07-21 2019-10-22 Modernatx, Inc. Zika RNA vaccines
WO2019226650A1 (en) 2018-05-23 2019-11-28 Modernatx, Inc. Delivery of dna
US10493143B2 (en) 2015-10-22 2019-12-03 Modernatx, Inc. Sexually transmitted disease vaccines
WO2020023390A1 (en) 2018-07-25 2020-01-30 Modernatx, Inc. Mrna based enzyme replacement therapy combined with a pharmacological chaperone for the treatment of lysosomal storage disorders
WO2020047201A1 (en) 2018-09-02 2020-03-05 Modernatx, Inc. Polynucleotides encoding very long-chain acyl-coa dehydrogenase for the treatment of very long-chain acyl-coa dehydrogenase deficiency
WO2020056155A2 (en) 2018-09-13 2020-03-19 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
WO2020056239A1 (en) 2018-09-14 2020-03-19 Modernatx, Inc. Polynucleotides encoding uridine diphosphate glycosyltransferase 1 family, polypeptide a1 for the treatment of crigler-najjar syndrome
WO2020056147A2 (en) 2018-09-13 2020-03-19 Modernatx, Inc. Polynucleotides encoding glucose-6-phosphatase for the treatment of glycogen storage disease
WO2020069169A1 (en) 2018-09-27 2020-04-02 Modernatx, Inc. Polynucleotides encoding arginase 1 for the treatment of arginase deficiency
WO2020097409A2 (en) 2018-11-08 2020-05-14 Modernatx, Inc. Use of mrna encoding ox40l to treat cancer in human patients
US10653767B2 (en) 2017-09-14 2020-05-19 Modernatx, Inc. Zika virus MRNA vaccines
US10695419B2 (en) 2016-10-21 2020-06-30 Modernatx, Inc. Human cytomegalovirus vaccine
WO2020227642A1 (en) 2019-05-08 2020-11-12 Modernatx, Inc. Compositions for skin and wounds and methods of use thereof
US10849920B2 (en) 2015-10-05 2020-12-01 Modernatx, Inc. Methods for therapeutic administration of messenger ribonucleic acid drugs
US10857105B2 (en) 2017-03-15 2020-12-08 MordernaTX, Inc. Compounds and compositions for intracellular delivery of therapeutic agents
WO2020263985A1 (en) 2019-06-24 2020-12-30 Modernatx, Inc. Messenger rna comprising functional rna elements and uses thereof
WO2020263883A1 (en) 2019-06-24 2020-12-30 Modernatx, Inc. Endonuclease-resistant messenger rna and uses thereof
WO2021009336A1 (en) 2019-07-18 2021-01-21 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for inducing full ablation of hematopoiesis
WO2021061707A1 (en) 2019-09-23 2021-04-01 Omega Therapeutics, Inc. Compositions and methods for modulating apolipoprotein b (apob) gene expression
WO2021061815A1 (en) 2019-09-23 2021-04-01 Omega Therapeutics, Inc. COMPOSITIONS AND METHODS FOR MODULATING HEPATOCYTE NUCLEAR FACTOR 4-ALPHA (HNF4α) GENE EXPRESSION
US20210128474A1 (en) * 2017-05-30 2021-05-06 Glaxosmithkline Biologicals S.A. Methods for manufacturing a liposome encapsulated rna
US11045540B2 (en) 2017-03-15 2021-06-29 Modernatx, Inc. Varicella zoster virus (VZV) vaccine
US11066355B2 (en) 2019-09-19 2021-07-20 Modernatx, Inc. Branched tail lipid compounds and compositions for intracellular delivery of therapeutic agents
US11103578B2 (en) 2016-12-08 2021-08-31 Modernatx, Inc. Respiratory virus nucleic acid vaccines
WO2021183720A1 (en) 2020-03-11 2021-09-16 Omega Therapeutics, Inc. Compositions and methods for modulating forkhead box p3 (foxp3) gene expression
WO2021195218A1 (en) 2020-03-24 2021-09-30 Generation Bio Co. Non-viral dna vectors and uses thereof for expressing gaucher therapeutics
WO2021195214A1 (en) 2020-03-24 2021-09-30 Generation Bio Co. Non-viral dna vectors and uses thereof for expressing factor ix therapeutics
WO2021247507A1 (en) 2020-06-01 2021-12-09 Modernatx, Inc. Phenylalanine hydroxylase variants and uses thereof
US11203569B2 (en) 2017-03-15 2021-12-21 Modernatx, Inc. Crystal forms of amino lipids
US11291635B2 (en) 2010-07-06 2022-04-05 Glaxosmithkline Biological Sa Virion-like delivery particles for self-replicating RNA molecules
US11291682B2 (en) 2010-07-06 2022-04-05 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
WO2022104131A1 (en) 2020-11-13 2022-05-19 Modernatx, Inc. Polynucleotides encoding cystic fibrosis transmembrane conductance regulator for the treatment of cystic fibrosis
US11351242B1 (en) 2019-02-12 2022-06-07 Modernatx, Inc. HMPV/hPIV3 mRNA vaccine composition
US11364292B2 (en) 2015-07-21 2022-06-21 Modernatx, Inc. CHIKV RNA vaccines
US11406703B2 (en) 2020-08-25 2022-08-09 Modernatx, Inc. Human cytomegalovirus vaccine
US11447566B2 (en) 2018-01-04 2022-09-20 Iconic Therapeutics, Inc. Anti-tissue factor antibodies, antibody-drug conjugates, and related methods
WO2022204369A1 (en) 2021-03-24 2022-09-29 Modernatx, Inc. Polynucleotides encoding methylmalonyl-coa mutase for the treatment of methylmalonic acidemia
WO2022204390A1 (en) 2021-03-24 2022-09-29 Modernatx, Inc. Lipid nanoparticles containing polynucleotides encoding phenylalanine hydroxylase and uses thereof
WO2022204380A1 (en) 2021-03-24 2022-09-29 Modernatx, Inc. Lipid nanoparticles containing polynucleotides encoding propionyl-coa carboxylase alpha and beta subunits and uses thereof
WO2022204370A1 (en) 2021-03-24 2022-09-29 Modernatx, Inc. Lipid nanoparticles and polynucleotides encoding ornithine transcarbamylase for the treatment of ornithine transcarbamylase deficiency
WO2022204371A1 (en) 2021-03-24 2022-09-29 Modernatx, Inc. Lipid nanoparticles containing polynucleotides encoding glucose-6-phosphatase and uses thereof
US11464848B2 (en) 2017-03-15 2022-10-11 Modernatx, Inc. Respiratory syncytial virus vaccine
WO2022232286A1 (en) 2021-04-27 2022-11-03 Generation Bio Co. Non-viral dna vectors expressing anti-coronavirus antibodies and uses thereof
WO2022232289A1 (en) 2021-04-27 2022-11-03 Generation Bio Co. Non-viral dna vectors expressing therapeutic antibodies and uses thereof
US11497807B2 (en) 2017-03-17 2022-11-15 Modernatx, Inc. Zoonotic disease RNA vaccines
US11504421B2 (en) 2017-05-08 2022-11-22 Gritstone Bio, Inc. Alphavirus neoantigen vectors
US11524023B2 (en) 2021-02-19 2022-12-13 Modernatx, Inc. Lipid nanoparticle compositions and methods of formulating the same
WO2022266083A2 (en) 2021-06-15 2022-12-22 Modernatx, Inc. Engineered polynucleotides for cell-type or microenvironment-specific expression
WO2022271776A1 (en) 2021-06-22 2022-12-29 Modernatx, Inc. Polynucleotides encoding uridine diphosphate glycosyltransferase 1 family, polypeptide a1 for the treatment of crigler-najjar syndrome
US11564893B2 (en) 2015-08-17 2023-01-31 Modernatx, Inc. Methods for preparing particles and related compositions
WO2023006999A2 (en) 2021-07-30 2023-02-02 CureVac SE Mrnas for treatment or prophylaxis of liver diseases
US11583504B2 (en) 2016-11-08 2023-02-21 Modernatx, Inc. Stabilized formulations of lipid nanoparticles
US11591619B2 (en) 2019-05-30 2023-02-28 Gritstone Bio, Inc. Modified adenoviruses
US11603399B2 (en) 2013-03-13 2023-03-14 Modernatx, Inc. Long-lived polynucleotide molecules
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
WO2023056044A1 (en) 2021-10-01 2023-04-06 Modernatx, Inc. Polynucleotides encoding relaxin for the treatment of fibrosis and/or cardiovascular disease
US11639329B2 (en) 2017-08-16 2023-05-02 Acuitas Therapeutics, Inc. Lipids for use in lipid nanoparticle formulations
US11639370B2 (en) 2010-10-11 2023-05-02 Glaxosmithkline Biologicals Sa Antigen delivery platforms
US11643441B1 (en) 2015-10-22 2023-05-09 Modernatx, Inc. Nucleic acid vaccines for varicella zoster virus (VZV)
WO2023081526A1 (en) 2021-11-08 2023-05-11 Orna Therapeutics, Inc. Lipid nanoparticle compositions for delivering circular polynucleotides
US11655475B2 (en) 2010-07-06 2023-05-23 Glaxosmithkline Biologicals Sa Immunisation of large mammals with low doses of RNA
WO2023135298A1 (en) 2022-01-17 2023-07-20 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods of inducing cell death of a population of solid tumor cells
WO2023144193A1 (en) 2022-01-25 2023-08-03 CureVac SE Mrnas for treatment of hereditary tyrosinemia type i
WO2023152365A1 (en) 2022-02-14 2023-08-17 INSERM (Institut National de la Santé et de la Recherche Médicale) Use of the 15-lipoxygenase for the treatment of lymphedema
WO2023161350A1 (en) 2022-02-24 2023-08-31 Io Biotech Aps Nucleotide delivery of cancer therapy
US11744886B2 (en) 2015-04-13 2023-09-05 CureVac Manufacturing GmbH Method for producing RNA compositions
US11744801B2 (en) 2017-08-31 2023-09-05 Modernatx, Inc. Methods of making lipid nanoparticles
US11752206B2 (en) 2017-03-15 2023-09-12 Modernatx, Inc. Herpes simplex virus vaccine
US11759422B2 (en) 2010-08-31 2023-09-19 Glaxosmithkline Biologicals Sa Pegylated liposomes for delivery of immunogen-encoding RNA
WO2023177655A1 (en) 2022-03-14 2023-09-21 Generation Bio Co. Heterologous prime boost vaccine compositions and methods of use
WO2023183909A2 (en) 2022-03-25 2023-09-28 Modernatx, Inc. Polynucleotides encoding fanconi anemia, complementation group proteins for the treatment of fanconi anemia
US11771747B2 (en) 2020-08-06 2023-10-03 Gritstone Bio, Inc. Multiepitope vaccine cassettes
US11786607B2 (en) 2017-06-15 2023-10-17 Modernatx, Inc. RNA formulations
WO2023239756A1 (en) 2022-06-07 2023-12-14 Generation Bio Co. Lipid nanoparticle compositions and uses thereof
WO2024023034A1 (en) 2022-07-25 2024-02-01 Institut National de la Santé et de la Recherche Médicale Use of apelin for the treatment of lymphedema
WO2024026254A1 (en) 2022-07-26 2024-02-01 Modernatx, Inc. Engineered polynucleotides for temporal control of expression
US11905525B2 (en) 2017-04-05 2024-02-20 Modernatx, Inc. Reduction of elimination of immune responses to non-intravenous, e.g., subcutaneously administered therapeutic proteins
WO2024040222A1 (en) 2022-08-19 2024-02-22 Generation Bio Co. Cleavable closed-ended dna (cedna) and methods of use thereof
US11911453B2 (en) 2018-01-29 2024-02-27 Modernatx, Inc. RSV RNA vaccines
WO2024044147A1 (en) 2022-08-23 2024-02-29 Modernatx, Inc. Methods for purification of ionizable lipids
WO2024047247A1 (en) 2022-09-02 2024-03-07 Institut National de la Santé et de la Recherche Médicale Base editing approaches for the treatment of amyotrophic lateral sclerosis
US11969506B2 (en) 2017-03-15 2024-04-30 Modernatx, Inc. Lipid nanoparticle formulation
WO2024102762A1 (en) 2022-11-08 2024-05-16 Orna Therapeutics, Inc. Lipids and lipid nanoparticle compositions for delivering polynucleotides
WO2024102730A1 (en) 2022-11-08 2024-05-16 Orna Therapeutics, Inc. Lipids and nanoparticle compositions for delivering polynucleotides
WO2024102677A1 (en) 2022-11-08 2024-05-16 Orna Therapeutics, Inc. Circular rna compositions

Families Citing this family (71)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103906527B (zh) 2011-06-08 2020-07-10 川斯勒佰尔公司 Mrna递送的脂质纳米颗粒组合物和方法
SG10201605537XA (en) * 2011-07-06 2016-09-29 Novartis Ag Liposomes having useful n:p ratio for delivery of rna molecules
TR201900264T4 (tr) 2011-08-31 2019-02-21 Glaxosmithkline Biologicals Sa İmmünojen şifreleyici rna'nın verilmesi için pegile edilmiş lipozomlar.
CA2868391A1 (en) 2012-04-02 2013-10-10 Stephane Bancel Polynucleotides comprising n1-methyl-pseudouridine and methods for preparing the same
JP2016504050A (ja) 2013-01-17 2016-02-12 モデルナ セラピューティクス インコーポレイテッドModerna Therapeutics,Inc. 細胞表現型の改変のためのシグナルセンサーポリヌクレオチド
MX2015011955A (es) 2013-03-08 2016-04-07 Novartis Ag Lipidos y composiciones de lipidos para el suministro de agentes activos.
ES2923018T3 (es) 2013-03-15 2022-09-22 Glaxosmithkline Biologicals Sa Métodos de purificación de ARN
JP7019233B2 (ja) 2013-07-11 2022-02-15 モデルナティエックス インコーポレイテッド CRISPR関連タンパク質をコードする合成ポリヌクレオチドおよび合成sgRNAを含む組成物ならびに使用方法
EP3122774B1 (en) 2014-03-25 2020-11-04 Yale University Uses of parasite macrophage migration inhibitory factors
EP2974739A1 (en) 2014-07-15 2016-01-20 Novartis AG RSVF trimerization domains
JP6594421B2 (ja) 2014-06-25 2019-10-23 アクイタス セラピューティクス インコーポレイテッド 核酸の送達のための新規脂質および脂質ナノ粒子製剤
EP3061826A1 (en) 2015-02-27 2016-08-31 Novartis AG Flavivirus replicons
WO2017004143A1 (en) 2015-06-29 2017-01-05 Acuitas Therapeutics Inc. Lipids and lipid nanoparticle formulations for delivery of nucleic acids
WO2017075531A1 (en) 2015-10-28 2017-05-04 Acuitas Therapeutics, Inc. Novel lipids and lipid nanoparticle formulations for delivery of nucleic acids
WO2017162266A1 (en) 2016-03-21 2017-09-28 Biontech Rna Pharmaceuticals Gmbh Rna replicon for versatile and efficient gene expression
WO2017162265A1 (en) 2016-03-21 2017-09-28 Biontech Rna Pharmaceuticals Gmbh Trans-replicating rna
JP7195147B2 (ja) 2016-05-16 2022-12-23 アクセス ツー アドバンスト ヘルス インスティチュート Peg化リポソームおよび使用方法
BR112018073683A2 (pt) 2016-05-18 2019-02-26 Modernatx, Inc. polinucleotídeos codificadores de relaxina
WO2017208191A1 (en) 2016-06-02 2017-12-07 Glaxosmithkline Biologicals Sa Zika viral antigen constructs
WO2018060288A1 (en) 2016-09-29 2018-04-05 Glaxosmithkline Biologicals S.A. Compositions and methods of treatment of persistent hpv infection
GB201616904D0 (en) 2016-10-05 2016-11-16 Glaxosmithkline Biologicals Sa Vaccine
EP3541416A1 (en) 2016-11-17 2019-09-25 GlaxoSmithKline Biologicals SA Zika viral antigen constructs
BR112019015797A2 (pt) * 2017-02-01 2020-03-17 Modernatx, Inc. Composições de mrna terapêuticas imunomoduladoras que codificam peptídeos de mutação de oncogene de ativação
EP3681993A1 (en) 2017-09-13 2020-07-22 BioNTech RNA Pharmaceuticals GmbH Rna replicon for reprogramming somatic cells
WO2019053056A1 (en) 2017-09-13 2019-03-21 Biontech Cell & Gene Therapies Gmbh RNA REPLICON FOR EXPRESSING A T CELL RECEPTOR OR ARTIFICIAL LYMPHOCYTIC T CELL RECEPTOR
EP3461497A1 (en) 2017-09-27 2019-04-03 GlaxoSmithKline Biologicals S.A. Viral antigens
WO2019094702A1 (en) 2017-11-10 2019-05-16 Cocoon Biotech Inc. Ocular applications of silk-based products
EP3836963A2 (en) 2018-08-17 2021-06-23 GlaxoSmithKline Biologicals S.A. Immunogenic compositions and uses thereof
EP3908328A1 (en) 2019-01-10 2021-11-17 BioNTech RNA Pharmaceuticals GmbH Localized administration of rna molecules for therapy
TW202043256A (zh) 2019-01-10 2020-12-01 美商健生生物科技公司 前列腺新抗原及其用途
AU2020205717A1 (en) 2019-01-11 2021-08-12 Acuitas Therapeutics, Inc. Lipids for lipid nanoparticle delivery of active agents
IL311628A (en) 2019-03-01 2024-05-01 Flagship Pioneering Innovations Vi Llc Polyribonucleotides and their cosmetic uses
MA55082A (fr) 2019-03-01 2022-01-05 Flagship Pioneering Innovations Vi Llc Compositions, procédés, et kits pour l'administration de polyribonucléotides
CN114206909A (zh) 2019-07-21 2022-03-18 葛兰素史克生物有限公司 治疗性病毒疫苗
EP3819377A1 (en) 2019-11-08 2021-05-12 Justus-Liebig-Universität Gießen Circular rna and uses thereof for inhibiting rna-binding proteins
US11576966B2 (en) 2020-02-04 2023-02-14 CureVac SE Coronavirus vaccine
JP2023519173A (ja) 2020-03-09 2023-05-10 アークトゥラス・セラピューティクス・インコーポレイテッド 免疫応答を誘導するための組成物及び方法
US20230364219A1 (en) 2020-04-16 2023-11-16 Glaxosmithkline Biologicals Sa Sars cov-2 spike protein construct
EP4153223A1 (en) 2020-05-20 2023-03-29 Flagship Pioneering Innovations VI, LLC Immunogenic compositions and uses thereof
AU2021286169A1 (en) 2020-06-04 2023-01-19 BioNTech SE RNA replicon for versatile and efficient gene expression
US20230234992A1 (en) 2020-06-05 2023-07-27 Glaxosmithkline Biologicals Sa Modified betacoronavirus spike proteins
US20230256090A1 (en) 2020-06-29 2023-08-17 Glaxosmithkline Biologicals Sa Adjuvants
AU2021308681A1 (en) 2020-07-16 2023-03-09 Acuitas Therapeutics, Inc. Cationic lipids for use in lipid nanoparticles
CN116234917A (zh) 2020-07-27 2023-06-06 安杰瑞姆生物科学公司 Dna分子组合物及其制备方法和使用方法
AU2021336976A1 (en) 2020-09-03 2023-03-23 Flagship Pioneering Innovations Vi, Llc Immunogenic compositions and uses thereof
EP4008785A1 (en) 2020-12-03 2022-06-08 Justus-Liebig-Universität Gießen Circular nucleic acids and uses thereof for interfering with genome expression and proliferation of coronaviruses
MX2023007574A (es) 2020-12-22 2023-09-29 CureVac SE "vacuna de arn contra variantes de sars-cov-2.
WO2022137128A2 (en) 2020-12-23 2022-06-30 Glaxosmithkline Biologicals Sa Self-amplifying messenger rna
EP4032546A1 (en) 2021-01-20 2022-07-27 GlaxoSmithKline Biologicals S.A. Therapeutic viral vaccine
CN117098558A (zh) * 2021-02-09 2023-11-21 塞瑞纳治疗公司 聚噁唑啉-脂质缀合物及包含其的脂质纳米颗粒和药物组合物
JP2024511206A (ja) 2021-03-26 2024-03-12 グラクソスミスクライン バイオロジカルズ ソシエテ アノニム 免疫原性組成物
EP4326860A1 (en) 2021-04-20 2024-02-28 Anjarium Biosciences AG Compositions of dna molecules encoding amylo-alpha-1, 6-glucosidase, 4-alpha-glucanotransferase, methods of making thereof, and methods of use thereof
CN113230394B (zh) * 2021-04-30 2024-04-30 广州源博医药科技有限公司 一种用于牛病毒性腹泻的rna疫苗及其构建方法
WO2022248353A1 (en) 2021-05-24 2022-12-01 Glaxosmithkline Biologicals Sa Adjuvants
WO2022259191A1 (en) 2021-06-09 2022-12-15 Glaxosmithkline Biologicals Sa Release assay for determining potency of self-amplifying rna drug product and methods for using
EP4367242A2 (en) 2021-07-07 2024-05-15 Omega Therapeutics, Inc. Compositions and methods for modulating secreted frizzled receptor protein 1 (sfrp1) gene expression
WO2023020993A1 (en) 2021-08-16 2023-02-23 Glaxosmithkline Biologicals Sa Novel methods
WO2023021421A1 (en) 2021-08-16 2023-02-23 Glaxosmithkline Biologicals Sa Low-dose lyophilized rna vaccines and methods for preparing and using the same
WO2023021427A1 (en) 2021-08-16 2023-02-23 Glaxosmithkline Biologicals Sa Freeze-drying of lipid nanoparticles (lnps) encapsulating rna and formulations thereof
WO2023020992A1 (en) 2021-08-16 2023-02-23 Glaxosmithkline Biologicals Sa Novel methods
WO2023020994A1 (en) 2021-08-16 2023-02-23 Glaxosmithkline Biologicals Sa Novel methods
WO2023031855A1 (en) 2021-09-03 2023-03-09 Glaxosmithkline Biologicals Sa Substitution of nucleotide bases in self-amplifying messenger ribonucleic acids
AU2022369342A1 (en) 2021-10-18 2024-03-14 BioNTech SE Modified replicable rna and related compositions and their use
AU2022372325A1 (en) 2021-10-18 2024-05-02 BioNTech SE Methods for determining mutations for increasing modified replicable rna function and related compositions and their use
WO2023135273A2 (en) 2022-01-14 2023-07-20 Anjarium Biosciences Ag Compositions of dna molecules encoding factor viii, methods of making thereof, and methods of use thereof
WO2023213378A1 (en) 2022-05-02 2023-11-09 BioNTech SE Replicon compositions and methods of using same for the treatment of diseases
WO2023242817A2 (en) 2022-06-18 2023-12-21 Glaxosmithkline Biologicals Sa Recombinant rna molecules comprising untranslated regions or segments encoding spike protein from the omicron strain of severe acute respiratory coronavirus-2
US11878055B1 (en) 2022-06-26 2024-01-23 BioNTech SE Coronavirus vaccine
WO2024017479A1 (en) 2022-07-21 2024-01-25 BioNTech SE Multifunctional cells transiently expressing an immune receptor and one or more cytokines, their use and methods for their production
WO2024056856A1 (en) 2022-09-15 2024-03-21 BioNTech SE Systems and compositions comprising trans-amplifying rna vectors with mirna
WO2024068545A1 (en) 2022-09-26 2024-04-04 Glaxosmithkline Biologicals Sa Influenza virus vaccines

Citations (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1989000812A1 (en) * 1987-07-28 1989-02-09 Micro-Pak, Inc. Method of manufacturing unilamellar lipid vesicles
US5013556A (en) * 1989-10-20 1991-05-07 Liposome Technology, Inc. Liposomes with enhanced circulation time
WO1996008235A1 (en) * 1994-09-13 1996-03-21 Depotech Corporation Preparation of multivesicular liposomes for controlled release of active agents
WO2001093836A2 (en) * 2000-06-09 2001-12-13 Teni Boulikas Encapsulation of polynucleotides and drugs into targeted liposomes
US20030124134A1 (en) * 1999-11-19 2003-07-03 Harbor-Ucla Research And Education Institute Pharmaceutical compositions and methods to vaccinate against candidiasis
US6790449B2 (en) * 1995-09-27 2004-09-14 The United States Of America As Represented By The Department Of Health And Human Services Methods for producing self-replicating infectious RSV particles comprising recombinant RSV genomes or antigenomes and the N, P, L, and M2 proteins
US20050042230A1 (en) * 2003-07-15 2005-02-24 Id Biomedical Corporation Of Quebec Subunit vaccine against respiratory syncytial virus infection
US20050064026A1 (en) * 2003-09-05 2005-03-24 Boehringer Ingelheim Pharma Gmbh & Co. Kg Method for preparing homogenous liposomes and lipoplexes
US20060051405A1 (en) * 2004-07-19 2006-03-09 Protiva Biotherapeutics, Inc. Compositions for the delivery of therapeutic agents and uses thereof
US20060177819A1 (en) * 2001-09-06 2006-08-10 Alphavax, Inc. Alphavirus replicon vector systems
US20060251620A1 (en) * 2002-08-22 2006-11-09 Lidia Ivanova Inducible alphaviral/orip based gene expression system
WO2007014754A1 (en) * 2005-08-02 2007-02-08 I.D.M. Immuno-Designed Molecules Process for the preparation of liposomal formulations
WO2009146867A1 (en) * 2008-06-04 2009-12-10 Institut Für Viruskrankheiten Und Immunprophylaxe Pestivirus replicons providing an rna-based viral vector system
US20100173980A1 (en) * 2002-09-13 2010-07-08 Andrew Vaillant Antiviral oligonucleotides targeting rsv
WO2010088537A2 (en) * 2009-01-29 2010-08-05 Alnylam Pharmaceuticals, Inc. Improved lipid formulation

Family Cites Families (237)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6090406A (en) 1984-04-12 2000-07-18 The Liposome Company, Inc. Potentiation of immune responses with liposomal adjuvants
EP0465529B1 (en) 1989-03-21 1998-04-29 Vical, Inc. Expression of exogenous polynucleotide sequences in a vertebrate
US6867195B1 (en) 1989-03-21 2005-03-15 Vical Incorporated Lipid-mediated polynucleotide administration to reduce likelihood of subject's becoming infected
US5279833A (en) 1990-04-04 1994-01-18 Yale University Liposomal transfection of nucleic acids into animal cells
US5264618A (en) 1990-04-19 1993-11-23 Vical, Inc. Cationic lipids for intracellular delivery of biologically active molecules
FR2676072B1 (fr) 1991-05-03 1994-11-18 Transgene Sa Vecteur de delivrance d'arn.
US5750390A (en) 1992-08-26 1998-05-12 Ribozyme Pharmaceuticals, Inc. Method and reagent for treatment of diseases caused by expression of the bcl-2 gene
US5693535A (en) 1992-05-14 1997-12-02 Ribozyme Pharmaceuticals, Inc. HIV targeted ribozymes
CA2134773A1 (en) 1992-06-04 1993-12-09 Robert J. Debs Methods and compositions for in vivo gene therapy
AU4769893A (en) 1992-07-17 1994-02-14 Ribozyme Pharmaceuticals, Inc. Method and reagent for treatment of animal diseases
US5474914A (en) 1992-07-29 1995-12-12 Chiron Corporation Method of producing secreted CMV glycoprotein H
US20020102273A1 (en) 1995-08-08 2002-08-01 Robert B. Grieve Use of alphavirus expression vectors to produce parasite anitgens
EP1624068A1 (en) 1993-06-01 2006-02-08 Life Technologies Inc. Genetic immunization with cationic lipids
US6015686A (en) 1993-09-15 2000-01-18 Chiron Viagene, Inc. Eukaryotic layered vector initiation systems
AU2215995A (en) 1994-04-07 1995-10-30 Akzo Nobel N.V. Freeze-dried compositions comprising rna
US5820873A (en) 1994-09-30 1998-10-13 The University Of British Columbia Polyethylene glycol modified ceramide lipids and liposome uses thereof
US5885613A (en) 1994-09-30 1999-03-23 The University Of British Columbia Bilayer stabilizing components and their use in forming programmable fusogenic liposomes
AU4594996A (en) 1994-11-30 1996-06-19 Chiron Viagene, Inc. Recombinant alphavirus vectors
US5965434A (en) 1994-12-29 1999-10-12 Wolff; Jon A. Amphipathic PH sensitive compounds and delivery systems for delivering biologically active compounds
US5792462A (en) 1995-05-23 1998-08-11 University Of North Carolina At Chapel Hill Alphavirus RNA replicon systems
US5981501A (en) 1995-06-07 1999-11-09 Inex Pharmaceuticals Corp. Methods for encapsulating plasmids in lipid bilayers
US7422902B1 (en) 1995-06-07 2008-09-09 The University Of British Columbia Lipid-nucleic acid particles prepared via a hydrophobic lipid-nucleic acid complex intermediate and use for gene transfer
ATE225668T1 (de) 1996-02-12 2002-10-15 Cobra Therapeutics Ltd Neue methode zur impfung sowie impfstoffe dafür, die eine epitopkodierende nukleinsäure und ein epitopenthaltende peptid beinhalten
DE19605548A1 (de) 1996-02-15 1997-09-04 Boehringer Ingelheim Int Zusammensetzung für die Transfektion höherer eukaryotischer Zellen
US6451592B1 (en) 1996-04-05 2002-09-17 Chiron Corporation Recombinant alphavirus-based vectors with reduced inhibition of cellular macromolecular synthesis
AU2284697A (en) 1996-04-11 1997-10-29 University Of British Columbia, The Fusogenic liposomes
EP0910343A1 (en) 1996-07-03 1999-04-28 University Of Pittsburgh Emulsion formulations for hydrophilic active agents
EP1254657B1 (en) 1996-09-13 2008-05-21 Lipoxen Technologies Limited Liposomes
US7384923B2 (en) 1999-05-14 2008-06-10 Lipoxen Technologies Limited Liposomes
US6395302B1 (en) 1996-11-19 2002-05-28 Octoplus B.V. Method for the preparation of microspheres which contain colloidal systems
CA2289702C (en) 1997-05-14 2008-02-19 Inex Pharmaceuticals Corp. High efficiency encapsulation of charged therapeutic agents in lipid vesicles
US6048546A (en) 1997-07-31 2000-04-11 Sandia Corporation Immobilized lipid-bilayer materials
US6060308A (en) 1997-09-04 2000-05-09 Connaught Laboratories Limited RNA respiratory syncytial virus vaccines
JP2002500003A (ja) 1997-11-28 2002-01-08 ザ・クラウン・イン・ザ・ライト・オヴ・ザ・クイーンズランド・デパートメント・オヴ・ヘルス フラビウイルスの発現および送達のシステム
US6009406A (en) 1997-12-05 1999-12-28 Square D Company Methodology and computer-based tools for re-engineering a custom-engineered product line
GB9726555D0 (en) 1997-12-16 1998-02-11 Smithkline Beecham Plc Vaccine
US6432925B1 (en) 1998-04-16 2002-08-13 John Wayne Cancer Institute RNA cancer vaccine and methods for its use
EP2311436A1 (en) 1998-04-27 2011-04-20 Altus Pharmaceuticals Inc. Stabilized protein crystals, formulations containing them and methods of making them
WO2000000616A2 (en) 1998-06-29 2000-01-06 U.S. Medical Research Institute Of Infectious Diseases Marburg virus vaccines
WO2000003683A2 (en) 1998-07-20 2000-01-27 Inex Pharmaceuticals Corporation Liposomal encapsulated nucleic acid-complexes
JP2002533124A (ja) 1998-12-31 2002-10-08 カイロン コーポレイション Hivポリペプチドの改善された発現およびウイルス様粒子の生成
ATE289630T1 (de) 1999-09-09 2005-03-15 Curevac Gmbh Transfer von mrnas unter verwendung von polykationischen verbindungen
EP1222289B1 (en) 1999-10-20 2008-04-16 The Johns Hopkins University School Of Medicine Chimeric immunogenic compositions and nucleic acids encoding them
US20030212022A1 (en) 2001-03-23 2003-11-13 Jean-Marie Vogel Compositions and methods for gene therapy
AU2001253323A1 (en) 2000-04-18 2001-10-30 Human Genome Sciences, Inc. Extracellular matrix polynucleotides, polypeptides, and antibodies
EP1297005B1 (en) 2000-07-03 2009-08-26 Novartis Vaccines and Diagnostics S.r.l. Immunisation against chlamydia pneumoniae
US7318928B2 (en) 2000-08-01 2008-01-15 The Johns Hopkins University Molecular vaccine linking intercellular spreading protein to an antigen
US20040142474A1 (en) 2000-09-14 2004-07-22 Expression Genetics, Inc. Novel cationic lipopolymer as a biocompatible gene delivery agent
ATE320792T1 (de) 2000-09-28 2006-04-15 Chiron Corp Mikropartikel zur verabreichung von heterologen nukleinsäure
MXPA03003690A (es) 2000-10-27 2004-05-05 Chiron Spa Acidos nucleicos y proteinas de los grupos a y b de estreptococos.
WO2002079239A2 (en) 2001-01-31 2002-10-10 U.S. Army Medical Research Institute Of Infectious Diseases Chimeric filovirus glycoprotein
EP1363660A4 (en) 2001-02-01 2006-06-21 Univ Johns Hopkins HIGHER MOLECULAR VACCINE BASED ON AUTOMPLICATIVE RNA, SUICIDE DNA OR UNDNA DNA VECTOR, WHICH LINKS ANTIGEN WITH A POLYPEPTIDE WHICH PROMOTES THE PRESENTATION OF THE ANTIGEN
WO2002072027A2 (en) 2001-03-14 2002-09-19 University Of Alabama Research Foundation Oncolytic rna replicons
WO2002074920A2 (en) 2001-03-16 2002-09-26 Johns Hopkins University A replication-defective alphavirus vaccine linking antigen with an immunogenicity-potentiating polypeptide and a method of delivery the same
CA2445947A1 (en) 2001-04-30 2002-11-07 Targeted Genetics Corporation Lipid-comprising drug delivery complexes and methods for their production
US20030077251A1 (en) 2001-05-23 2003-04-24 Nicolas Escriou Replicons derived from positive strand RNA virus genomes useful for the production of heterologous proteins
EP1604688B1 (de) 2001-06-05 2010-02-03 CureVac GmbH Stabilisierte Tumorantigen-mRNA mit erhöhtem G/C-Gehalt
US20050175629A1 (en) 2001-08-31 2005-08-11 Giuseppe Del Giudice Helicobacter pylori vaccination
AU2003211103A1 (en) 2002-02-13 2003-09-04 Northeastern University Intracellular delivery of therapeutic agents
DE10207177A1 (de) 2002-02-19 2003-09-04 Novosom Ag Fakultativ kationische Lipide
CA2491164C (en) 2002-06-28 2012-05-08 Cory Giesbrecht Method and apparatus for producing liposomes
RU2311911C2 (ru) 2002-07-05 2007-12-10 Липоксен Текнолоджиз Лимитед Способ усиления иммунного ответа при вакцинации нуклеиновой кислотой
ES2242462B1 (es) 2002-07-18 2007-01-01 Marti Industria Metalurgica, S.L Pulverizador giratorio seleccionable.
WO2004055166A2 (en) 2002-12-13 2004-07-01 Alphavax, Inc. Multi-antigenic alphavirus replicon particles and methods
ES2429338T3 (es) 2002-12-23 2013-11-14 Vical Incorporated Vacuna basada en polinucleótidos optimizados por codones contra infección por citomegalovirus humano
US8338583B2 (en) 2003-02-04 2012-12-25 Bar-Ilan University Snornai-small nucleolar RNA degradation by RNA interference in trypanosomatids
WO2004076645A2 (en) 2003-02-27 2004-09-10 University Of Massachusetts Compositions and methods for cytomegalovirus treatment
PL1608762T3 (pl) 2003-03-20 2014-06-30 Alphavax Inc Udoskonalone replikony alfawirusowe i konstrukty plazmidów pomocniczych
US7731967B2 (en) 2003-04-30 2010-06-08 Novartis Vaccines And Diagnostics, Inc. Compositions for inducing immune responses
CA2527301A1 (en) 2003-05-30 2004-12-09 Nippon Shinyaku Co., Ltd. Oligonucleic acid-bearing composite and pharmaceutical composition containing the composite
EP1635865A2 (en) 2003-06-26 2006-03-22 Chiron Corporation Immunogenic compositions for chlamydia trachomatis
WO2005007689A1 (en) 2003-07-11 2005-01-27 Alphavax, Inc. Alphavirus-based cytomegalovirus vaccines
CA2532228C (en) 2003-07-16 2017-02-14 Protiva Biotherapeutics, Inc. Lipid encapsulated interfering rna
ES2505695T3 (es) 2003-07-31 2014-10-10 Novartis Vaccines And Diagnostics, Inc. Composiciones inmunógenas para Streptococcus pyogenes
NZ592917A (en) 2003-09-15 2012-12-21 Protiva Biotherapeutics Inc Stable polyethyleneglycol (PEG) dialkyloxypropyl (DAA) lipid conjugates
WO2005046621A2 (en) 2003-11-12 2005-05-26 The United States Of America As Represented By The Secretary Of The Navy Enhancement of vaccine-induced immune responses and protection by heterologous boosting with alphavirus replicon vaccines
US20050202075A1 (en) 2004-03-12 2005-09-15 Pardridge William M. Delivery of genes encoding short hairpin RNA using receptor-specific nanocontainers
US7303881B2 (en) 2004-04-30 2007-12-04 Pds Biotechnology Corporation Antigen delivery compositions and methods of use
GB0410866D0 (en) 2004-05-14 2004-06-16 Chiron Srl Haemophilius influenzae
MXPA06013124A (es) 2004-05-18 2007-05-23 Alphavax Inc Vectores de alfavirus derivados de tc-83, particulas y metodos.
US20060024670A1 (en) 2004-05-18 2006-02-02 Luke Catherine J Influenza virus vaccine composition and methods of use
WO2006078294A2 (en) 2004-05-21 2006-07-27 Novartis Vaccines And Diagnostics Inc. Alphavirus vectors for respiratory pathogen vaccines
GB0411428D0 (en) 2004-05-21 2004-06-23 Got A Gene Ab Vectors
CA2609260A1 (en) * 2004-06-02 2005-12-22 Sourcepharm, Inc. Rna-containing microvesicles and methods therefor
JP4796062B2 (ja) 2004-06-07 2011-10-19 プロチバ バイオセラピューティクス インコーポレイティッド 脂質封入干渉rna
EP1781593B1 (en) 2004-06-07 2011-12-14 Protiva Biotherapeutics Inc. Cationic lipids and methods of use
CA2572921C (en) 2004-07-09 2017-01-03 The University Of North Carolina At Chapel Hill Replication-competent and propagation-defective venezuelan equine encephalitis (vee) viral adjuvants
JP5086082B2 (ja) 2004-10-01 2012-11-28 ノバルティス ヴァクシンズ アンド ダイアグノスティクス エスアールエル C型肝炎ウイルス複製系
EP1811960A2 (en) 2004-11-19 2007-08-01 Novosom AG Improvements in or relating to pharmaceutical compositions for local administration
GB2421025A (en) 2004-12-09 2006-06-14 Oxxon Therapeutics Ltd HSV vaccination vectors
EP1922300A2 (en) 2005-02-14 2008-05-21 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
NZ560929A (en) 2005-02-18 2009-12-24 Novartis Vaccines & Diagnostic Immunogens from uropathogenic escherichia coli
HUE027400T2 (en) 2005-02-18 2016-10-28 Glaxosmithkline Biologicals Sa Proteins and nucleic acids from meningitis / sepsis with Escherichia coli
DE602006008278D1 (de) 2005-03-02 2009-09-17 Pharmazeutische zusammensetzung
GB0504436D0 (en) 2005-03-03 2005-04-06 Glaxosmithkline Biolog Sa Vaccine
NZ562381A (en) 2005-03-30 2011-06-30 Novartis Vaccines & Diagnostic Haemophilus influenzae type B
US7618393B2 (en) 2005-05-03 2009-11-17 Pharmajet, Inc. Needle-less injector and method of fluid delivery
CN101238146A (zh) 2005-05-12 2008-08-06 诺华疫苗和诊断有限公司 砂眼衣原体的免疫原性组合物
US8703095B2 (en) 2005-07-07 2014-04-22 Sanofi Pasteur S.A. Immuno-adjuvant emulsion
US7951384B2 (en) 2005-08-05 2011-05-31 University Of Massachusetts Virus-like particles as vaccines for paramyxovirus
ES2735531T3 (es) 2005-08-23 2019-12-19 Univ Pennsylvania ARN que contiene nucleósidos modificados y métodos de uso del mismo
EP1764089A1 (en) 2005-09-15 2007-03-21 Novosom AG Serum stable liposomes comprising amphoter II lipid mixtures
DE102005046490A1 (de) 2005-09-28 2007-03-29 Johannes-Gutenberg-Universität Mainz Modifikationen von RNA, die zu einer erhöhten Transkriptstabilität und Translationseffizienz führen
DE602006019296D1 (de) 2005-09-29 2011-02-10 Elan Pharm Inc Pyrimidinylamidverbindungen, die die durch vla-4 vermittelte leukozytenadhäsion inhibieren
WO2007047749A1 (en) 2005-10-18 2007-04-26 Novartis Vaccines And Diagnostics Inc. Mucosal and systemic immunizations with alphavirus replicon particles
JP2007112768A (ja) 2005-10-24 2007-05-10 Kyoto Univ 肝指向性リポソーム組成物
US7776336B2 (en) 2005-10-25 2010-08-17 Novartis Vaccines And Diagnostics Srl Compositions comprising Yersinia pestis antigens
CA2630220C (en) 2005-11-22 2020-10-13 Doris Coit Norovirus and sapovirus antigens
US9393215B2 (en) 2005-12-02 2016-07-19 Novartis Ag Nanoparticles for use in immunogenic compositions
EP2004141A2 (en) 2006-03-17 2008-12-24 Novosom AG An efficient method for loading amphoteric liposomes with nucleic acid active substances
EP2010659B1 (en) 2006-04-14 2014-06-18 CellScript, Inc. Kits and methods for generating 5' capped RNA
EP2037959B1 (en) 2006-06-07 2016-01-27 The Trustees Of Princeton University Cytomegalovirus surface protein complex for use in vaccines and as a drug target
US7915399B2 (en) 2006-06-09 2011-03-29 Protiva Biotherapeutics, Inc. Modified siRNA molecules and uses thereof
PL2035581T3 (pl) 2006-06-21 2013-01-31 Scripps Research Inst Kompozycja DNA przeciwko FAP - antygenowi zrębowemu nowotworu i sposoby jej stosowania
JP2009544754A (ja) 2006-07-28 2009-12-17 アプライド バイオシステムズ, エルエルシー ジヌクレオチドmrnaキャップアナログ
EP2064230A2 (en) 2006-08-16 2009-06-03 Novartis AG Immunogens from uropathogenic escherichia coli
US20090022760A1 (en) 2006-09-12 2009-01-22 Alphavax Alphavirus Replicon Particles Matched to Protein Antigens as Immunological Adjuvants
DE102007001370A1 (de) 2007-01-09 2008-07-10 Curevac Gmbh RNA-kodierte Antikörper
US20100015218A1 (en) * 2007-02-16 2010-01-21 Vasant Jadhav Compositions and methods for potentiated activity of biologically active molecules
US20100196492A1 (en) 2007-03-08 2010-08-05 Green Jordan J Electrostatic coating of particles for drug delivery
US8877206B2 (en) 2007-03-22 2014-11-04 Pds Biotechnology Corporation Stimulation of an immune response by cationic lipids
AU2008232891B2 (en) * 2007-03-29 2012-01-12 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of a gene from the Ebola
US8748591B2 (en) 2007-04-17 2014-06-10 The Board Of Regents Of The University Of Texas System Chimeric sindbis-western equine encephalitis virus and uses thereof
CN101674853B (zh) 2007-05-04 2013-03-27 玛瑞纳生物技术有限公司 氨基酸脂质及其用途
JP2010528591A (ja) 2007-05-23 2010-08-26 マンカインド コーポレイション マルチシストロン性ベクター及びそれらの設計方法
DE102007029471A1 (de) 2007-06-20 2008-12-24 Novosom Ag Neue fakultativ kationische Sterole
AU2008266807B2 (en) 2007-06-21 2012-08-16 Alphavax, Inc. Promoterless cassettes for expression of alphavirus structural proteins
EP2173771A1 (en) 2007-07-04 2010-04-14 Ribovax Biotechnologies SA Antibodies against human cytomegalovirus (hcmv)
GB0714963D0 (en) 2007-08-01 2007-09-12 Novartis Ag Compositions comprising antigens
US20110177155A1 (en) 2007-08-21 2011-07-21 Immune Disease Institute, Inc. Methods of delivery of agents to leukocytes and endothelial cells
GB0717187D0 (en) 2007-09-04 2007-10-17 Novartis Ag Compositions comprising yersinia pestis antigens
US20090162395A1 (en) 2007-09-26 2009-06-25 Crowe Jr James E Vaccine for rsv and mpv
EP2042193A1 (en) 2007-09-28 2009-04-01 Biomay AG RNA Vaccines
JP5627464B2 (ja) 2007-11-26 2014-11-19 ノバルティス アーゲー アルファウイルス粒子を生成する方法
EP2067749A1 (en) 2007-11-29 2009-06-10 Total Petrochemicals France Process for purification of an aqueous phase containing polyaromatics
WO2009074861A2 (en) 2007-12-10 2009-06-18 Powderject Research Limited Improved vaccine
WO2009111088A2 (en) * 2008-01-02 2009-09-11 The Johns Hopkins University Antitumor immunization by liposomal delivery of vaccine to the spleen
CA3044134A1 (en) 2008-01-02 2009-07-09 Arbutus Biopharma Corporation Improved compositions and methods for the delivery of nucleic acids
ITMI20081249A1 (it) 2008-07-09 2010-01-09 Novartis Vaccines & Diagnostic Immunogeni di escherichia coli con solubilità migliorata.
WO2009109860A2 (en) 2008-03-06 2009-09-11 Novartis Ag Mutant forms of chlamydia htra
DK2279254T3 (en) 2008-04-15 2017-09-18 Protiva Biotherapeutics Inc PRESENT UNKNOWN LIPID FORMS FOR NUCLEIC ACID ADMINISTRATION
WO2009127230A1 (en) 2008-04-16 2009-10-22 Curevac Gmbh MODIFIED (m)RNA FOR SUPPRESSING OR AVOIDING AN IMMUNOSTIMULATORY RESPONSE AND IMMUNOSUPPRESSIVE COMPOSITION
WO2009132131A1 (en) 2008-04-22 2009-10-29 Alnylam Pharmaceuticals, Inc. Amino lipid based improved lipid formulation
WO2009132206A1 (en) 2008-04-25 2009-10-29 Liquidia Technologies, Inc. Compositions and methods for intracellular delivery and release of cargo
US20100040650A1 (en) 2008-05-30 2010-02-18 Crowe Jr James E Virus-Like paramyxovirus particles and vaccines
WO2009156155A1 (en) 2008-06-25 2009-12-30 Probiogen Ag Cell line for propagation of highly attenuated alphaviruses
US9421251B2 (en) 2008-06-25 2016-08-23 Novartis Ag Rapid responses to delayed booster immunisations
MX2011000543A (es) 2008-07-16 2011-04-27 Inst Research In Biomedicine Anticuerpos neutralizantes de citomegalovirus humano y uso de los mismos.
NZ591055A (en) 2008-07-16 2012-08-31 Inst Research In Biomedicine Human cytomegalovirus neutralising antibodies and use thereof
JP2010025644A (ja) 2008-07-16 2010-02-04 Kochi Univ Of Technology 硝酸イオンの呈色試薬並びにこれを用いた硝酸イオンの検出及び定量方法
JP5667566B2 (ja) 2008-08-06 2015-02-12 ノバルティス アーゲー 免疫原性組成物における使用のための微粒子
CL2008002322A1 (es) 2008-08-07 2009-06-05 Univ Concepcion Formulacion farmaceutica veterinaria que comprende un sistema vectorial viral constituido por una particula recombinante de arn que codifica una cu/zn superoxido dismutasa de la bacteria patogena de bovinos brucella abortus, y al menos un alfavirus arn perteneciente a la familia del virus semliki forest (sfv), util como vacuna.
JP5848127B2 (ja) 2008-08-13 2016-01-27 カリフォルニア インスティテュート オブ テクノロジー 担体ナノ粒子ならびに関連する組成物、方法およびシステム
WO2010036948A2 (en) 2008-09-26 2010-04-01 The United States Of America, As Represented By The Secretary, Department Of Health & Human Services Dna prime/inactivated vaccine boost immunization to influenza virus
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
CA2984026C (en) 2008-10-09 2020-02-11 Arbutus Biopharma Corporation Improved amino lipids and methods for the delivery of nucleic acids
MX353900B (es) 2008-11-07 2018-02-01 Massachusetts Inst Technology Lipidoides de aminoalcohol y usos de los mismos.
MX359674B (es) 2008-11-10 2018-10-05 Alnylam Pharmaceuticals Inc Lipidos y composiciones novedosas para el suministro de terapeuticos.
AU2009316680B2 (en) 2008-11-18 2016-03-24 Takeda Vaccines, Inc. RSV F VLPs and methods of manufacture and use thereof
EP2510947B1 (en) 2009-04-14 2016-02-10 GlaxoSmithKline Biologicals SA Compositions for immunising against Staphylococcus aureus
WO2010129687A1 (en) 2009-05-05 2010-11-11 Alnylam Pharmaceuticals, Inc Methods of delivering oligonucleotides to immune cells
KR102205886B1 (ko) 2009-06-10 2021-01-21 알닐람 파마슈티칼스 인코포레이티드 향상된 지질 조성물
EP2449114B9 (en) 2009-07-01 2017-04-19 Protiva Biotherapeutics Inc. Novel lipid formulations for delivery of therapeutic agents to solid tumors
WO2011001780A1 (ja) 2009-07-02 2011-01-06 コニカミノルタホールディングス株式会社 特定分散剤を含有する外水相を利用する二段階乳化法によるリポソームの製造方法、ならびに当該リポソームの製造方法を用いるリポソーム分散液またはその乾燥粉末の製造方法およびそれにより製造されるリポソーム分散液またはその乾燥粉末
CA2766907A1 (en) 2009-07-06 2011-01-13 Novartis Ag Self replicating rna molecules and uses thereof
PL3178490T3 (pl) 2009-07-15 2022-08-01 Glaxosmithkline Biologicals S.A. Kompozycje białka f rsv i sposoby ich wytwarzania
US9642903B2 (en) 2009-07-16 2017-05-09 Lior Carmon Antigen specific multi epitope-based anti-infective vaccines
WO2011012316A2 (de) 2009-07-31 2011-02-03 Ludwig-Maximilians-Universität Rna mit einer kombination aus unmodifizierten und modifizierten nucleotiden zur proteinexpression
MX2012002723A (es) 2009-09-02 2012-04-11 Novartis Ag Composiciones inmunogenicas que incluyen moduladores de la actividad de receptores tipo toll.
JO3257B1 (ar) 2009-09-02 2018-09-16 Novartis Ag مركبات وتركيبات كمعدلات لفاعلية tlr
US20110070260A1 (en) 2009-09-09 2011-03-24 Baric Ralph S Multivalent Immunogenic Compositions Against Noroviruses and Methods of Use
RU2573409C2 (ru) 2009-11-04 2016-01-20 Дзе Юниверсити Оф Бритиш Коламбиа Содержащие нуклеиновые кислоты липидные частицы и относящиеся к ним способы
US20110112353A1 (en) 2009-11-09 2011-05-12 Circulite, Inc. Bifurcated outflow cannulae
TR201901311T4 (tr) 2009-12-01 2019-02-21 Translate Bio Inc İnsan genetik hastalıklarında mRNA'nın teslimi için steroid türevi.
KR102505097B1 (ko) 2009-12-07 2023-03-02 더 트러스티스 오브 더 유니버시티 오브 펜실베니아 세포 리프로그래밍을 위한 정제된 변형 rna를 포함하는 rna 제제
EP3296398A1 (en) 2009-12-07 2018-03-21 Arbutus Biopharma Corporation Compositions for nucleic acid delivery
US20130017223A1 (en) 2009-12-18 2013-01-17 The University Of British Columbia Methods and compositions for delivery of nucleic acids
EP2516010A2 (en) 2009-12-23 2012-10-31 Novartis AG Lipids, lipid compositions, and methods of using them
WO2012082165A1 (en) 2010-01-24 2012-06-21 Novartis Ag Irradiated biodegradable polymer microparticles
EP2544693B1 (en) 2010-03-09 2017-09-13 Biomedical Research Models, Inc. A novel mucosal vaccination approach for herpes simplex virus type-2
EP2556151A1 (en) 2010-04-07 2013-02-13 Novartis AG Method for generating a parvovirus b19 virus-like particle
EP2591097A1 (en) 2010-07-06 2013-05-15 Novartis AG Norovirus derived immunogenic compositions and methods
CN103052400B (zh) 2010-07-06 2016-11-16 诺华股份有限公司 自我复制rna分子的病毒样递送颗粒
PL3243526T3 (pl) 2010-07-06 2020-05-18 Glaxosmithkline Biologicals S.A. Dostarczanie rna w celu wyzwolenia wielu szlaków immunologicznych
MX2013000164A (es) 2010-07-06 2013-03-05 Novartis Ag Liposomas con lipidos que tienen valor de pka ventajoso para suministro de arn.
BR112013000391B8 (pt) 2010-07-06 2022-10-04 Novartis Ag Composição de emulsão catiônica de óleo em água e seu uso
US10487332B2 (en) 2010-07-06 2019-11-26 Glaxosmithkline Biologicals Sa Immunisation of large mammals with low doses of RNA
US9770463B2 (en) 2010-07-06 2017-09-26 Glaxosmithkline Biologicals Sa Delivery of RNA to different cell types
US9192661B2 (en) 2010-07-06 2015-11-24 Novartis Ag Delivery of self-replicating RNA using biodegradable polymer particles
US8898852B2 (en) 2010-08-04 2014-12-02 Honeywell International Inc. Air burst to clear detection window
WO2012019168A2 (en) 2010-08-06 2012-02-09 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
ES2727583T3 (es) 2010-08-31 2019-10-17 Glaxosmithkline Biologicals Sa Lípidos adecuados para la administración liposómica de ARN que codifica proteínas
FI4043040T3 (fi) 2010-08-31 2023-04-04 Glaxosmithkline Biologicals Sa Pieniä liposomeja immunogeeniä koodaavan rna:n toimittamiseksi
DK3981427T3 (da) 2010-08-31 2022-07-11 Glaxosmithkline Biologicals Sa Pegylerede liposomer til afgivelse af immunogen-kodende RNA
WO2012034025A2 (en) 2010-09-09 2012-03-15 Virginia Commonwealth University Human cytomegalovirus vaccine
DE19177059T1 (de) 2010-10-01 2021-10-07 Modernatx, Inc. N1-methyl-pseudouracile enthältendes ribonucleinsäuren sowie ihre verwendungen
EP4098325A1 (en) 2010-10-11 2022-12-07 GlaxoSmithKline Biologicals S.A. Antigen delivery platforms
BR112013009946B1 (pt) 2010-10-25 2019-04-30 Stepan Company Composição de sulfobetaina, betaina ou amônio quaternário, derivado; formulação de glifosato, composição de herbicida solúvel em água ou composição antimicrobiana, limpador de superfície áspera, formulação de detergente para vestuário sujo, xampu ou condicionador de cabelo ou produto de limpeza pessoal ou sabonete, inibidor de corrosão, dispersante de parafina, espumante de poço de gás, espumante, aditivo de espuma ou dispersante e emulsificante aniônico para composições agrícolas
EP4144368A1 (en) 2011-01-26 2023-03-08 GlaxoSmithKline Biologicals S.A. Rsv immunization regimen
US9243041B2 (en) 2011-01-31 2016-01-26 The Trustees Of The University Of Pennsylvania Nucleic acid molecules encoding novel herpes antigens, vaccine comprising the same, and methods of use thereof
WO2012116714A1 (en) 2011-03-02 2012-09-07 Curevac Gmbh Vaccination in elderly patients
DE12722942T1 (de) 2011-03-31 2021-09-30 Modernatx, Inc. Freisetzung und formulierung von manipulierten nukleinsäuren
WO2012158613A1 (en) 2011-05-13 2012-11-22 Novartis Ag Pre-fusion rsv f antigens
AU2012255913A1 (en) 2011-05-17 2013-11-21 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof for non-human vertebrates
ES2795110T3 (es) 2011-06-08 2020-11-20 Translate Bio Inc Lípidos escindibles
BR112014000227A8 (pt) 2011-07-06 2018-03-06 Novartis Ag emulsões óleo-em-água que contêm ácidos nucleicos
CA2841047A1 (en) 2011-07-06 2013-01-10 Novartis Ag Immunogenic compositions and uses thereof
SG10201605537XA (en) 2011-07-06 2016-09-29 Novartis Ag Liposomes having useful n:p ratio for delivery of rna molecules
RU2649133C2 (ru) 2011-07-06 2018-03-29 Новартис Аг Катионные эмульсии масло-в-воде
ES2656050T3 (es) 2011-07-06 2018-02-22 Glaxosmithkline Biologicals Sa Composiciones de combinación inmunogénica y usos de las mismas
TR201900264T4 (tr) 2011-08-31 2019-02-21 Glaxosmithkline Biologicals Sa İmmünojen şifreleyici rna'nın verilmesi için pegile edilmiş lipozomlar.
WO2013039861A2 (en) 2011-09-12 2013-03-21 modeRNA Therapeutics Engineered nucleic acids and methods of use thereof
JP6113737B2 (ja) 2011-10-03 2017-04-12 モデルナティエックス インコーポレイテッドModernaTX,Inc. 修飾型のヌクレオシド、ヌクレオチドおよび核酸、ならびにそれらの使用方法
WO2013055905A1 (en) 2011-10-11 2013-04-18 Novartis Ag Recombinant self-replicating polycistronic rna molecules
WO2013054199A2 (en) 2011-10-12 2013-04-18 Novartis Ag Cmv antigens and uses thereof
US20140378538A1 (en) 2011-12-14 2014-12-25 Moderma Therapeutics, Inc. Methods of responding to a biothreat
MX2014007233A (es) 2011-12-16 2015-02-04 Moderna Therapeutics Inc Composiciones de nucleosidos, nucleotidos y acidos nucleicos modificados.
CN104968354A (zh) 2011-12-21 2015-10-07 现代治疗公司 增加器官或器官外植体的活力或寿命的方法
CA2868391A1 (en) 2012-04-02 2013-10-10 Stephane Bancel Polynucleotides comprising n1-methyl-pseudouridine and methods for preparing the same
EP2834259A4 (en) 2012-04-02 2016-08-24 Moderna Therapeutics Inc MODIFIED POLYNUCLEOTIDES
CA2892529C (en) 2012-11-26 2023-04-25 Moderna Therapeutics, Inc. Terminally modified rna
EP2943221A1 (en) 2013-01-10 2015-11-18 Novartis AG Influenza virus immunogenic compositions and uses thereof
US9504747B2 (en) 2013-03-08 2016-11-29 Novartis Ag Lipids and lipid compositions for the delivery of active agents
US10258698B2 (en) 2013-03-14 2019-04-16 Modernatx, Inc. Formulation and delivery of modified nucleoside, nucleotide, and nucleic acid compositions
US20160032316A1 (en) 2013-03-14 2016-02-04 The Trustees Of The University Of Pennsylvania Purification and Purity Assessment of RNA Molecules Synthesized with Modified Nucleosides
US8980864B2 (en) 2013-03-15 2015-03-17 Moderna Therapeutics, Inc. Compositions and methods of altering cholesterol levels
EP3083556B1 (en) 2013-12-19 2019-12-25 Novartis AG Lipids and lipid compositions for the delivery of active agents
RS63030B1 (sr) 2015-09-17 2022-04-29 Modernatx Inc Jedinjenja i kompozicije za intracelularno isporučivanje terapeutskih sredstava
WO2017075531A1 (en) 2015-10-28 2017-05-04 Acuitas Therapeutics, Inc. Novel lipids and lipid nanoparticle formulations for delivery of nucleic acids
WO2018075980A1 (en) 2016-10-21 2018-04-26 Modernatx, Inc. Human cytomegalovirus vaccine
WO2018089790A1 (en) 2016-11-10 2018-05-17 Translate Bio, Inc. Improved ice-based lipid nanoparticle formulation for delivery of mrna
US11045540B2 (en) 2017-03-15 2021-06-29 Modernatx, Inc. Varicella zoster virus (VZV) vaccine
AU2019384557A1 (en) 2018-11-21 2021-06-10 Translate Bio, Inc. Treatment of cystic fibrosis by delivery of nebulized mRNA encoding CFTR
US20230172858A1 (en) 2019-08-30 2023-06-08 Glaxosmithkline Biologicals Sa Jet mixing lipid nanoparticle manufacturing process
WO2022137133A1 (en) 2020-12-22 2022-06-30 Curevac Ag Rna vaccine against sars-cov-2 variants
US20230000970A1 (en) 2021-01-11 2023-01-05 Modernatx, Inc. Seasonal rna influenza virus vaccines

Patent Citations (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1989000812A1 (en) * 1987-07-28 1989-02-09 Micro-Pak, Inc. Method of manufacturing unilamellar lipid vesicles
US5013556A (en) * 1989-10-20 1991-05-07 Liposome Technology, Inc. Liposomes with enhanced circulation time
WO1996008235A1 (en) * 1994-09-13 1996-03-21 Depotech Corporation Preparation of multivesicular liposomes for controlled release of active agents
US6790449B2 (en) * 1995-09-27 2004-09-14 The United States Of America As Represented By The Department Of Health And Human Services Methods for producing self-replicating infectious RSV particles comprising recombinant RSV genomes or antigenomes and the N, P, L, and M2 proteins
US20030124134A1 (en) * 1999-11-19 2003-07-03 Harbor-Ucla Research And Education Institute Pharmaceutical compositions and methods to vaccinate against candidiasis
WO2001093836A2 (en) * 2000-06-09 2001-12-13 Teni Boulikas Encapsulation of polynucleotides and drugs into targeted liposomes
US20060177819A1 (en) * 2001-09-06 2006-08-10 Alphavax, Inc. Alphavirus replicon vector systems
US20060251620A1 (en) * 2002-08-22 2006-11-09 Lidia Ivanova Inducible alphaviral/orip based gene expression system
US20100173980A1 (en) * 2002-09-13 2010-07-08 Andrew Vaillant Antiviral oligonucleotides targeting rsv
US20050042230A1 (en) * 2003-07-15 2005-02-24 Id Biomedical Corporation Of Quebec Subunit vaccine against respiratory syncytial virus infection
US20050064026A1 (en) * 2003-09-05 2005-03-24 Boehringer Ingelheim Pharma Gmbh & Co. Kg Method for preparing homogenous liposomes and lipoplexes
US20060051405A1 (en) * 2004-07-19 2006-03-09 Protiva Biotherapeutics, Inc. Compositions for the delivery of therapeutic agents and uses thereof
WO2007014754A1 (en) * 2005-08-02 2007-02-08 I.D.M. Immuno-Designed Molecules Process for the preparation of liposomal formulations
WO2009146867A1 (en) * 2008-06-04 2009-12-10 Institut Für Viruskrankheiten Und Immunprophylaxe Pestivirus replicons providing an rna-based viral vector system
WO2010088537A2 (en) * 2009-01-29 2010-08-05 Alnylam Pharmaceuticals, Inc. Improved lipid formulation

Cited By (251)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11850305B2 (en) 2010-07-06 2023-12-26 Glaxosmithkline Biologicals Sa Method of making lipid formulations with RNA encoding immunogens
US11291635B2 (en) 2010-07-06 2022-04-05 Glaxosmithkline Biological Sa Virion-like delivery particles for self-replicating RNA molecules
US11291682B2 (en) 2010-07-06 2022-04-05 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
US11324770B2 (en) 2010-07-06 2022-05-10 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11596645B2 (en) 2010-07-06 2023-03-07 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11638694B2 (en) 2010-07-06 2023-05-02 Glaxosmithkline Biologicals Sa Vaccine for eliciting immune response comprising lipid formulations and RNA encoding multiple immunogens
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
US11913001B2 (en) 2010-07-06 2024-02-27 Glaxosmithkline Biologicals Sa Immunisation 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
US11655475B2 (en) 2010-07-06 2023-05-23 Glaxosmithkline Biologicals Sa Immunisation of large mammals with low doses of RNA
US11891608B2 (en) 2010-07-06 2024-02-06 Glaxosmithkline Biologicals Sa Immunization 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
US11883534B2 (en) 2010-07-06 2024-01-30 Glaxosmithkline Biologicals Sa Immunisation with lipid formulations with RNA encoding immunogens
US11690865B2 (en) 2010-07-06 2023-07-04 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11690863B2 (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
US11690864B2 (en) 2010-07-06 2023-07-04 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11690862B1 (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
US11707482B2 (en) 2010-07-06 2023-07-25 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
US11857681B2 (en) 2010-07-06 2024-01-02 Glaxosmithkline Biologicals Sa Lipid formulations with RNA encoding immunogens
US11730754B2 (en) 2010-07-06 2023-08-22 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11739334B2 (en) 2010-07-06 2023-08-29 Glaxosmithkline Biologicals Sa Immunisation of large mammals with low doses of RNA
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
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
US11857562B2 (en) 2010-07-06 2024-01-02 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11845925B2 (en) 2010-07-06 2023-12-19 Glaxosmithkline Biologicals Sa Immunisation of large mammals with low doses of RNA
US11851660B2 (en) 2010-07-06 2023-12-26 Glaxosmithkline Biologicals Sa Immunisation of large mammals with low doses of RNA
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
US8822663B2 (en) 2010-08-06 2014-09-02 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
US20130195969A1 (en) * 2010-08-31 2013-08-01 Novartis Ag Small liposomes for delivery of immunogen encoding rna
US11759422B2 (en) 2010-08-31 2023-09-19 Glaxosmithkline Biologicals Sa Pegylated liposomes for delivery of immunogen-encoding RNA
US9254265B2 (en) * 2010-08-31 2016-02-09 Novartis Ag Small 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
US10064959B2 (en) 2010-10-01 2018-09-04 Modernatx, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US9701965B2 (en) 2010-10-01 2017-07-11 Modernatx, Inc. Engineered nucleic acids and methods of use thereof
US9657295B2 (en) 2010-10-01 2017-05-23 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
US8710200B2 (en) 2011-03-31 2014-04-29 Moderna Therapeutics, Inc. Engineered nucleic acids encoding a modified erythropoietin and their expression
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
US11896636B2 (en) * 2011-07-06 2024-02-13 Glaxosmithkline Biologicals Sa Immunogenic combination compositions and uses thereof
US20140227346A1 (en) * 2011-07-06 2014-08-14 Andrew Geall Immunogenic combination compositions and uses thereof
US10751386B2 (en) 2011-09-12 2020-08-25 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
US10022425B2 (en) 2011-09-12 2018-07-17 Modernatx, Inc. Engineered nucleic acids and methods of use thereof
US9428535B2 (en) 2011-10-03 2016-08-30 Moderna Therapeutics, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US9295689B2 (en) 2011-12-16 2016-03-29 Moderna Therapeutics, Inc. Formulation and delivery of PLGA microspheres
US8680069B2 (en) 2011-12-16 2014-03-25 Moderna Therapeutics, Inc. Modified polynucleotides for the production of G-CSF
US8664194B2 (en) 2011-12-16 2014-03-04 Moderna Therapeutics, Inc. Method for producing a protein of interest in a primate
US9186372B2 (en) 2011-12-16 2015-11-17 Moderna Therapeutics, Inc. Split dose administration
US9271996B2 (en) 2011-12-16 2016-03-01 Moderna Therapeutics, Inc. Formulation and delivery of PLGA microspheres
US8754062B2 (en) 2011-12-16 2014-06-17 Moderna Therapeutics, Inc. DLIN-KC2-DMA lipid nanoparticle delivery of modified polynucleotides
US9221891B2 (en) 2012-04-02 2015-12-29 Moderna Therapeutics, Inc. In vivo production of proteins
US9095552B2 (en) 2012-04-02 2015-08-04 Moderna Therapeutics, Inc. Modified polynucleotides encoding copper metabolism (MURR1) domain containing 1
US9878056B2 (en) 2012-04-02 2018-01-30 Modernatx, Inc. Modified polynucleotides for the production of cosmetic proteins and peptides
US9220792B2 (en) 2012-04-02 2015-12-29 Moderna Therapeutics, Inc. Modified polynucleotides encoding aquaporin-5
US9220755B2 (en) 2012-04-02 2015-12-29 Moderna Therapeutics, Inc. Modified polynucleotides for the production of proteins associated with blood and lymphatic disorders
US9255129B2 (en) 2012-04-02 2016-02-09 Moderna Therapeutics, Inc. Modified polynucleotides encoding SIAH E3 ubiquitin protein ligase 1
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
US9827332B2 (en) 2012-04-02 2017-11-28 Modernatx, Inc. Modified polynucleotides for the production of proteins
US9828416B2 (en) 2012-04-02 2017-11-28 Modernatx, Inc. Modified polynucleotides for the production of secreted proteins
US10501512B2 (en) 2012-04-02 2019-12-10 Modernatx, Inc. Modified polynucleotides
US9814760B2 (en) 2012-04-02 2017-11-14 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
US9107886B2 (en) 2012-04-02 2015-08-18 Moderna Therapeutics, Inc. Modified polynucleotides encoding basic helix-loop-helix family member E41
US9233141B2 (en) 2012-04-02 2016-01-12 Moderna Therapeutics, Inc. Modified polynucleotides for the production of proteins associated with blood and lymphatic disorders
US9089604B2 (en) 2012-04-02 2015-07-28 Moderna Therapeutics, Inc. Modified polynucleotides for treating galactosylceramidase protein deficiency
US9061059B2 (en) 2012-04-02 2015-06-23 Moderna Therapeutics, Inc. Modified polynucleotides for treating protein deficiency
US9050297B2 (en) 2012-04-02 2015-06-09 Moderna Therapeutics, Inc. Modified polynucleotides encoding aryl hydrocarbon receptor nuclear translocator
US8999380B2 (en) 2012-04-02 2015-04-07 Moderna Therapeutics, Inc. Modified polynucleotides for the production of biologics and proteins associated with human disease
US9782462B2 (en) 2012-04-02 2017-10-10 Modernatx, Inc. Modified polynucleotides for the production of proteins associated with human disease
US9254311B2 (en) 2012-04-02 2016-02-09 Moderna Therapeutics, Inc. Modified polynucleotides for the production of proteins
US9675668B2 (en) 2012-04-02 2017-06-13 Moderna Therapeutics, Inc. Modified polynucleotides encoding hepatitis A virus cellular receptor 2
US9283287B2 (en) 2012-04-02 2016-03-15 Moderna Therapeutics, Inc. Modified polynucleotides for the production of nuclear proteins
US9587003B2 (en) 2012-04-02 2017-03-07 Modernatx, Inc. Modified polynucleotides for the production of oncology-related proteins and peptides
US9303079B2 (en) 2012-04-02 2016-04-05 Moderna Therapeutics, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
US9572897B2 (en) 2012-04-02 2017-02-21 Modernatx, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
US9301993B2 (en) 2012-04-02 2016-04-05 Moderna Therapeutics, Inc. Modified polynucleotides encoding apoptosis inducing factor 1
US9512456B2 (en) 2012-08-14 2016-12-06 Modernatx, Inc. Enzymes and polymerases for the synthesis of RNA
US9597380B2 (en) 2012-11-26 2017-03-21 Modernatx, Inc. Terminally modified RNA
US20140193484A1 (en) * 2013-01-10 2014-07-10 Sylvie Carine Bertholet Girardin Influenza virus immunogenic compositions and uses thereof
US11603399B2 (en) 2013-03-13 2023-03-14 Modernatx, Inc. Long-lived polynucleotide molecules
WO2014152211A1 (en) 2013-03-14 2014-09-25 Moderna Therapeutics, Inc. Formulation and delivery of modified nucleoside, nucleotide, and nucleic acid compositions
US10258698B2 (en) 2013-03-14 2019-04-16 Modernatx, Inc. Formulation and delivery of modified nucleoside, nucleotide, and nucleic acid compositions
US8980864B2 (en) 2013-03-15 2015-03-17 Moderna Therapeutics, Inc. Compositions and methods of altering cholesterol levels
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
US10815291B2 (en) 2013-09-30 2020-10-27 Modernatx, Inc. Polynucleotides encoding immune modulating polypeptides
US10023626B2 (en) 2013-09-30 2018-07-17 Modernatx, Inc. Polynucleotides encoding immune modulating polypeptides
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
WO2015100269A3 (en) * 2013-12-27 2015-11-12 Double Helix Corporation Compositions and methods for providing active telomerase to cells in vivo
US10869932B2 (en) 2014-01-20 2020-12-22 University Of Utah Research Foundation Compositions and methods for modifying the surface of cells and methods of use
US10279047B2 (en) 2014-01-20 2019-05-07 University Of Utah Research Foundation Compositions and methods for modifying the surface of cells and methods of use
WO2015109325A1 (en) * 2014-01-20 2015-07-23 University Of Utah Research Foundation Compositions and methods for modifying the surface of cells and methods of use
US9872900B2 (en) 2014-04-23 2018-01-23 Modernatx, Inc. Nucleic acid vaccines
US10709779B2 (en) 2014-04-23 2020-07-14 Modernatx, Inc. Nucleic acid vaccines
US10022435B2 (en) 2014-04-23 2018-07-17 Modernatx, Inc. Nucleic acid vaccines
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
US11744886B2 (en) 2015-04-13 2023-09-05 CureVac Manufacturing GmbH Method for producing RNA compositions
US11364292B2 (en) 2015-07-21 2022-06-21 Modernatx, Inc. CHIKV RNA vaccines
US10449244B2 (en) 2015-07-21 2019-10-22 Modernatx, Inc. Zika RNA vaccines
US11007260B2 (en) 2015-07-21 2021-05-18 Modernatx, Inc. Infectious disease vaccines
US10702597B2 (en) 2015-07-21 2020-07-07 Modernatx, Inc. CHIKV RNA vaccines
US11564893B2 (en) 2015-08-17 2023-01-31 Modernatx, Inc. Methods for preparing particles and related compositions
US11220476B2 (en) 2015-09-17 2022-01-11 Modernatx, Inc. Compounds and compositions for intracellular delivery of therapeutic agents
US10266485B2 (en) 2015-09-17 2019-04-23 Modernatx, Inc. Compounds and compositions for intracellular delivery of therapeutic agents
US10442756B2 (en) 2015-09-17 2019-10-15 Modernatx, Inc. Compounds and compositions for intracellular delivery of therapeutic agents
US10392341B2 (en) 2015-09-17 2019-08-27 Modernatx, Inc. Compounds and compositions for intracellular delivery of therapeutic agents
US11590157B2 (en) 2015-10-05 2023-02-28 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
US11235052B2 (en) 2015-10-22 2022-02-01 Modernatx, Inc. Chikungunya virus RNA vaccines
US10675342B2 (en) 2015-10-22 2020-06-09 Modernatx, Inc. Chikungunya virus RNA vaccines
US11643441B1 (en) 2015-10-22 2023-05-09 Modernatx, Inc. Nucleic acid vaccines for varicella zoster virus (VZV)
US10064935B2 (en) 2015-10-22 2018-09-04 Modernatx, Inc. Human cytomegalovirus RNA vaccines
US10716846B2 (en) 2015-10-22 2020-07-21 Modernatx, Inc. Human cytomegalovirus RNA vaccines
US10272150B2 (en) 2015-10-22 2019-04-30 Modernatx, Inc. Combination PIV3/hMPV RNA vaccines
US10702600B1 (en) 2015-10-22 2020-07-07 Modernatx, Inc. Betacoronavirus mRNA vaccine
US11484590B2 (en) 2015-10-22 2022-11-01 Modernatx, Inc. Human cytomegalovirus RNA vaccines
US10124055B2 (en) 2015-10-22 2018-11-13 Modernatx, Inc. Zika virus RNA vaccines
US10543269B2 (en) 2015-10-22 2020-01-28 Modernatx, Inc. hMPV RNA vaccines
US10933127B2 (en) 2015-10-22 2021-03-02 Modernatx, Inc. Betacoronavirus mRNA vaccine
US10702599B2 (en) 2015-10-22 2020-07-07 Modernatx, Inc. HPIV3 RNA vaccines
US11278611B2 (en) 2015-10-22 2022-03-22 Modernatx, Inc. Zika virus RNA vaccines
US10064934B2 (en) 2015-10-22 2018-09-04 Modernatx, Inc. Combination PIV3/hMPV RNA vaccines
US10238731B2 (en) 2015-10-22 2019-03-26 Modernatx, Inc. Chikagunya virus RNA vaccines
US10383937B2 (en) 2015-10-22 2019-08-20 Modernatx, Inc. Human cytomegalovirus RNA vaccines
US10493143B2 (en) 2015-10-22 2019-12-03 Modernatx, Inc. Sexually transmitted disease vaccines
US11872278B2 (en) 2015-10-22 2024-01-16 Modernatx, Inc. Combination HMPV/RSV RNA vaccines
US10517940B2 (en) 2015-10-22 2019-12-31 Modernatx, Inc. Zika virus RNA vaccines
US10556018B2 (en) 2015-12-10 2020-02-11 Modernatx, Inc. Compositions and methods for delivery of agents
US10485885B2 (en) 2015-12-10 2019-11-26 Modernatx, Inc. Compositions and methods for delivery of agents
US11285222B2 (en) 2015-12-10 2022-03-29 Modernatx, Inc. Compositions and methods for delivery of agents
US10207010B2 (en) 2015-12-10 2019-02-19 Modernatx, Inc. Compositions and methods for delivery of agents
US10799463B2 (en) 2015-12-22 2020-10-13 Modernatx, Inc. Compounds and compositions for intracellular delivery of agents
US10195156B2 (en) 2015-12-22 2019-02-05 Modernatx, Inc. Compounds and compositions for intracellular delivery of agents
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
WO2018033254A2 (en) 2016-08-19 2018-02-22 Curevac Ag Rna for cancer therapy
US10695419B2 (en) 2016-10-21 2020-06-30 Modernatx, Inc. Human cytomegalovirus vaccine
US11197927B2 (en) 2016-10-21 2021-12-14 Modernatx, Inc. Human cytomegalovirus vaccine
US11541113B2 (en) 2016-10-21 2023-01-03 Modernatx, Inc. Human cytomegalovirus vaccine
US11583504B2 (en) 2016-11-08 2023-02-21 Modernatx, Inc. Stabilized formulations of lipid nanoparticles
EP3808380A1 (en) 2016-12-08 2021-04-21 CureVac AG Rna for treatment or prophylaxis of a liver disease
US11103578B2 (en) 2016-12-08 2021-08-31 Modernatx, Inc. Respiratory virus nucleic acid vaccines
WO2018104540A1 (en) 2016-12-08 2018-06-14 Curevac Ag Rnas for wound healing
WO2018104538A1 (en) 2016-12-08 2018-06-14 Curevac Ag Rna for treatment or prophylaxis of a liver disease
WO2018115525A1 (en) 2016-12-23 2018-06-28 Curevac Ag Lassa virus vaccine
WO2018115527A2 (en) 2016-12-23 2018-06-28 Curevac Ag Mers coronavirus vaccine
US10273269B2 (en) 2017-02-16 2019-04-30 Modernatx, Inc. High potency immunogenic zika virus compositions
US11045540B2 (en) 2017-03-15 2021-06-29 Modernatx, Inc. Varicella zoster virus (VZV) vaccine
US11752206B2 (en) 2017-03-15 2023-09-12 Modernatx, Inc. Herpes simplex virus vaccine
US11203569B2 (en) 2017-03-15 2021-12-21 Modernatx, Inc. Crystal forms of amino lipids
US11918644B2 (en) 2017-03-15 2024-03-05 Modernatx, Inc. Varicella zoster virus (VZV) vaccine
US11464848B2 (en) 2017-03-15 2022-10-11 Modernatx, Inc. Respiratory syncytial virus vaccine
US11969506B2 (en) 2017-03-15 2024-04-30 Modernatx, Inc. Lipid nanoparticle formulation
US10857105B2 (en) 2017-03-15 2020-12-08 MordernaTX, Inc. Compounds and compositions for intracellular delivery of therapeutic agents
WO2018167320A1 (en) 2017-03-17 2018-09-20 Curevac Ag Rna vaccine and immune checkpoint inhibitors for combined anticancer therapy
US11497807B2 (en) 2017-03-17 2022-11-15 Modernatx, Inc. Zoonotic disease RNA vaccines
WO2018172556A1 (en) 2017-03-24 2018-09-27 Curevac Ag Nucleic acids encoding crispr-associated proteins and uses thereof
US11905525B2 (en) 2017-04-05 2024-02-20 Modernatx, Inc. Reduction of elimination of immune responses to non-intravenous, e.g., subcutaneously administered therapeutic proteins
US11504421B2 (en) 2017-05-08 2022-11-22 Gritstone Bio, Inc. Alphavirus neoantigen vectors
US11510973B2 (en) 2017-05-08 2022-11-29 Gritstone Bio, Inc. Alphavirus antigen vectors
WO2018213789A1 (en) 2017-05-18 2018-11-22 Modernatx, Inc. Modified messenger rna comprising functional rna elements
EP4253544A2 (en) 2017-05-18 2023-10-04 ModernaTX, Inc. Modified messenger rna comprising functional rna elements
WO2018213731A1 (en) 2017-05-18 2018-11-22 Modernatx, Inc. Polynucleotides encoding tethered interleukin-12 (il12) polypeptides and uses thereof
US20210128474A1 (en) * 2017-05-30 2021-05-06 Glaxosmithkline Biologicals S.A. Methods for manufacturing a liposome encapsulated rna
WO2018222890A1 (en) 2017-05-31 2018-12-06 Arcturus Therapeutics, Inc. Synthesis and structure of high potency rna therapeutics
WO2018232006A1 (en) 2017-06-14 2018-12-20 Modernatx, Inc. Polynucleotides encoding coagulation factor viii
US11786607B2 (en) 2017-06-15 2023-10-17 Modernatx, Inc. RNA formulations
WO2019008001A1 (en) 2017-07-04 2019-01-10 Curevac Ag NEW NUCLEIC ACID MOLECULES
EP3424524A2 (en) 2017-07-04 2019-01-09 CureVac AG Cancer rna-vaccine
US11639329B2 (en) 2017-08-16 2023-05-02 Acuitas Therapeutics, Inc. Lipids for use in lipid nanoparticle formulations
US11744801B2 (en) 2017-08-31 2023-09-05 Modernatx, Inc. Methods of making lipid nanoparticles
US11207398B2 (en) 2017-09-14 2021-12-28 Modernatx, Inc. Zika virus mRNA vaccines
US10653767B2 (en) 2017-09-14 2020-05-19 Modernatx, Inc. Zika virus MRNA vaccines
WO2019077001A1 (en) 2017-10-19 2019-04-25 Curevac Ag NEW ARTIFICIAL NUCLEIC ACID MOLECULES
WO2019104160A2 (en) 2017-11-22 2019-05-31 Modernatx, Inc. Polynucleotides encoding phenylalanine hydroxylase for the treatment of phenylketonuria
WO2019104195A1 (en) 2017-11-22 2019-05-31 Modernatx, Inc. Polynucleotides encoding propionyl-coa carboxylase alpha and beta subunits for the treatment of propionic acidemia
WO2019104152A1 (en) 2017-11-22 2019-05-31 Modernatx, Inc. Polynucleotides encoding ornithine transcarbamylase for the treatment of urea cycle disorders
US11447566B2 (en) 2018-01-04 2022-09-20 Iconic Therapeutics, Inc. Anti-tissue factor antibodies, antibody-drug conjugates, and related methods
WO2019136241A1 (en) 2018-01-05 2019-07-11 Modernatx, Inc. Polynucleotides encoding anti-chikungunya virus antibodies
US11911453B2 (en) 2018-01-29 2024-02-27 Modernatx, Inc. RSV RNA vaccines
WO2019200171A1 (en) 2018-04-11 2019-10-17 Modernatx, Inc. Messenger rna comprising functional rna elements
WO2019226650A1 (en) 2018-05-23 2019-11-28 Modernatx, Inc. Delivery of dna
WO2020023390A1 (en) 2018-07-25 2020-01-30 Modernatx, Inc. Mrna based enzyme replacement therapy combined with a pharmacological chaperone for the treatment of lysosomal storage disorders
WO2020047201A1 (en) 2018-09-02 2020-03-05 Modernatx, Inc. Polynucleotides encoding very long-chain acyl-coa dehydrogenase for the treatment of very long-chain acyl-coa dehydrogenase deficiency
WO2020056155A2 (en) 2018-09-13 2020-03-19 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
WO2020056147A2 (en) 2018-09-13 2020-03-19 Modernatx, Inc. Polynucleotides encoding glucose-6-phosphatase for the treatment of glycogen storage disease
WO2020056239A1 (en) 2018-09-14 2020-03-19 Modernatx, Inc. Polynucleotides encoding uridine diphosphate glycosyltransferase 1 family, polypeptide a1 for the treatment of crigler-najjar syndrome
WO2020069169A1 (en) 2018-09-27 2020-04-02 Modernatx, Inc. Polynucleotides encoding arginase 1 for the treatment of arginase deficiency
WO2020097409A2 (en) 2018-11-08 2020-05-14 Modernatx, Inc. Use of mrna encoding ox40l to treat cancer in human patients
US11351242B1 (en) 2019-02-12 2022-06-07 Modernatx, Inc. HMPV/hPIV3 mRNA vaccine composition
WO2020227642A1 (en) 2019-05-08 2020-11-12 Modernatx, Inc. Compositions for skin and wounds and methods of use thereof
US11591619B2 (en) 2019-05-30 2023-02-28 Gritstone Bio, Inc. Modified adenoviruses
WO2020263883A1 (en) 2019-06-24 2020-12-30 Modernatx, Inc. Endonuclease-resistant messenger rna and uses thereof
WO2020263985A1 (en) 2019-06-24 2020-12-30 Modernatx, Inc. Messenger rna comprising functional rna elements and uses thereof
WO2021009336A1 (en) 2019-07-18 2021-01-21 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for inducing full ablation of hematopoiesis
US11066355B2 (en) 2019-09-19 2021-07-20 Modernatx, Inc. Branched tail lipid compounds and compositions for intracellular delivery of therapeutic agents
US11597698B2 (en) 2019-09-19 2023-03-07 Modernatx, Inc. Branched tail lipid compounds and compositions for intracellular delivery of therapeutic agents
WO2021061707A1 (en) 2019-09-23 2021-04-01 Omega Therapeutics, Inc. Compositions and methods for modulating apolipoprotein b (apob) gene expression
WO2021061815A1 (en) 2019-09-23 2021-04-01 Omega Therapeutics, Inc. COMPOSITIONS AND METHODS FOR MODULATING HEPATOCYTE NUCLEAR FACTOR 4-ALPHA (HNF4α) GENE EXPRESSION
WO2021183720A1 (en) 2020-03-11 2021-09-16 Omega Therapeutics, Inc. Compositions and methods for modulating forkhead box p3 (foxp3) gene expression
WO2021195218A1 (en) 2020-03-24 2021-09-30 Generation Bio Co. Non-viral dna vectors and uses thereof for expressing gaucher therapeutics
WO2021195214A1 (en) 2020-03-24 2021-09-30 Generation Bio Co. Non-viral dna vectors and uses thereof for expressing factor ix therapeutics
WO2021247507A1 (en) 2020-06-01 2021-12-09 Modernatx, Inc. Phenylalanine hydroxylase variants and uses thereof
US11771747B2 (en) 2020-08-06 2023-10-03 Gritstone Bio, Inc. Multiepitope vaccine cassettes
US11406703B2 (en) 2020-08-25 2022-08-09 Modernatx, Inc. Human cytomegalovirus vaccine
WO2022104131A1 (en) 2020-11-13 2022-05-19 Modernatx, Inc. Polynucleotides encoding cystic fibrosis transmembrane conductance regulator for the treatment of cystic fibrosis
US11622972B2 (en) 2021-02-19 2023-04-11 Modernatx, Inc. Lipid nanoparticle compositions and methods of formulating the same
US11524023B2 (en) 2021-02-19 2022-12-13 Modernatx, Inc. Lipid nanoparticle compositions and methods of formulating the same
WO2022204380A1 (en) 2021-03-24 2022-09-29 Modernatx, Inc. Lipid nanoparticles containing polynucleotides encoding propionyl-coa carboxylase alpha and beta subunits and uses thereof
WO2022204371A1 (en) 2021-03-24 2022-09-29 Modernatx, Inc. Lipid nanoparticles containing polynucleotides encoding glucose-6-phosphatase and uses thereof
WO2022204370A1 (en) 2021-03-24 2022-09-29 Modernatx, Inc. Lipid nanoparticles and polynucleotides encoding ornithine transcarbamylase for the treatment of ornithine transcarbamylase deficiency
WO2022204390A1 (en) 2021-03-24 2022-09-29 Modernatx, Inc. Lipid nanoparticles containing polynucleotides encoding phenylalanine hydroxylase and uses thereof
WO2022204369A1 (en) 2021-03-24 2022-09-29 Modernatx, Inc. Polynucleotides encoding methylmalonyl-coa mutase for the treatment of methylmalonic acidemia
WO2022232286A1 (en) 2021-04-27 2022-11-03 Generation Bio Co. Non-viral dna vectors expressing anti-coronavirus antibodies and uses thereof
WO2022232289A1 (en) 2021-04-27 2022-11-03 Generation Bio Co. Non-viral dna vectors expressing therapeutic antibodies and uses thereof
WO2022266083A2 (en) 2021-06-15 2022-12-22 Modernatx, Inc. Engineered polynucleotides for cell-type or microenvironment-specific expression
WO2022271776A1 (en) 2021-06-22 2022-12-29 Modernatx, Inc. Polynucleotides encoding uridine diphosphate glycosyltransferase 1 family, polypeptide a1 for the treatment of crigler-najjar syndrome
WO2023006999A2 (en) 2021-07-30 2023-02-02 CureVac SE Mrnas for treatment or prophylaxis of liver diseases
WO2023056044A1 (en) 2021-10-01 2023-04-06 Modernatx, Inc. Polynucleotides encoding relaxin for the treatment of fibrosis and/or cardiovascular disease
WO2023081526A1 (en) 2021-11-08 2023-05-11 Orna Therapeutics, Inc. Lipid nanoparticle compositions for delivering circular polynucleotides
WO2023135298A1 (en) 2022-01-17 2023-07-20 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods of inducing cell death of a population of solid tumor cells
WO2023144193A1 (en) 2022-01-25 2023-08-03 CureVac SE Mrnas for treatment of hereditary tyrosinemia type i
WO2023152365A1 (en) 2022-02-14 2023-08-17 INSERM (Institut National de la Santé et de la Recherche Médicale) Use of the 15-lipoxygenase for the treatment of lymphedema
WO2023161350A1 (en) 2022-02-24 2023-08-31 Io Biotech Aps Nucleotide delivery of cancer therapy
WO2023177655A1 (en) 2022-03-14 2023-09-21 Generation Bio Co. Heterologous prime boost vaccine compositions and methods of use
WO2023183909A2 (en) 2022-03-25 2023-09-28 Modernatx, Inc. Polynucleotides encoding fanconi anemia, complementation group proteins for the treatment of fanconi anemia
WO2023239756A1 (en) 2022-06-07 2023-12-14 Generation Bio Co. Lipid nanoparticle compositions and uses thereof
WO2024023034A1 (en) 2022-07-25 2024-02-01 Institut National de la Santé et de la Recherche Médicale Use of apelin for the treatment of lymphedema
WO2024026254A1 (en) 2022-07-26 2024-02-01 Modernatx, Inc. Engineered polynucleotides for temporal control of expression
WO2024040222A1 (en) 2022-08-19 2024-02-22 Generation Bio Co. Cleavable closed-ended dna (cedna) and methods of use thereof
WO2024044147A1 (en) 2022-08-23 2024-02-29 Modernatx, Inc. Methods for purification of ionizable lipids
WO2024047247A1 (en) 2022-09-02 2024-03-07 Institut National de la Santé et de la Recherche Médicale Base editing approaches for the treatment of amyotrophic lateral sclerosis
WO2024102762A1 (en) 2022-11-08 2024-05-16 Orna Therapeutics, Inc. Lipids and lipid nanoparticle compositions for delivering polynucleotides
WO2024102730A1 (en) 2022-11-08 2024-05-16 Orna Therapeutics, Inc. Lipids and nanoparticle compositions for delivering polynucleotides
WO2024102677A1 (en) 2022-11-08 2024-05-16 Orna Therapeutics, Inc. Circular rna compositions

Also Published As

Publication number Publication date
EP4066855A1 (en) 2022-10-05
SI4066856T1 (sl) 2023-03-31
LT2611461T (lt) 2022-08-10
DK4066856T3 (da) 2022-12-19
PT4066856T (pt) 2023-01-02
ES2918649T3 (es) 2022-07-19
SI2611461T1 (sl) 2022-08-31
FI4066855T3 (fi) 2023-03-23
RS63367B1 (sr) 2022-07-29
DK4066855T3 (da) 2023-02-20
HUE058666T2 (hu) 2022-09-28
EP3970742A1 (en) 2022-03-23
PT3981427T (pt) 2022-06-27
PL4066855T3 (pl) 2023-05-08
ES2938327T3 (es) 2023-04-10
EP4066856A1 (en) 2022-10-05
EP4147718A1 (en) 2023-03-15
EP4066855B1 (en) 2022-12-28
HUE061068T2 (hu) 2023-05-28
HRP20220835T1 (hr) 2022-10-14
HRP20221023T1 (hr) 2022-11-11
PL3981427T3 (pl) 2022-09-19
LT4066857T (lt) 2023-02-27
SI4066855T1 (sl) 2023-04-28
CA2809851A1 (en) 2012-03-08
CN103179984A (zh) 2013-06-26
HRP20230185T1 (hr) 2023-03-31
DK3970742T3 (da) 2022-07-04
BR112013004866A2 (pt) 2016-06-07
RU2013114362A (ru) 2014-10-10
AU2011295935B2 (en) 2016-02-11
RS63329B1 (sr) 2022-07-29
HRP20230226T1 (hr) 2023-04-14
EP4226941A1 (en) 2023-08-16
PL4066856T3 (pl) 2023-02-27
HRP20220695T1 (hr) 2022-07-08
HRP20230185T8 (hr) 2023-07-21
PT2611461T (pt) 2022-05-26
HUE058667T2 (hu) 2022-09-28
SI4066857T1 (sl) 2023-04-28
LT3970742T (lt) 2022-08-10
ES2913791T3 (es) 2022-06-06
US11759422B2 (en) 2023-09-19
RS63983B1 (sr) 2023-03-31
MX2013002336A (es) 2013-03-18
ES2935009T3 (es) 2023-03-01
EP2611461A1 (en) 2013-07-10
RS63949B1 (sr) 2023-02-28
PL4066857T3 (pl) 2023-03-20
EP4066856B1 (en) 2022-12-07
LT3981427T (lt) 2022-08-10
SI3981427T1 (sl) 2022-08-31
FI4066857T3 (fi) 2023-03-20
HUE060524T2 (hu) 2023-03-28
EP3981427B9 (en) 2022-07-27
EP3970742B1 (en) 2022-05-25
RS63816B1 (sr) 2023-01-31
DK2611461T3 (da) 2022-05-16
EP4119155A1 (en) 2023-01-18
DK4066857T3 (da) 2023-02-20
ES2918192T3 (es) 2022-07-14
WO2012031043A1 (en) 2012-03-08
EP4066857B1 (en) 2022-12-21
EP4226940A1 (en) 2023-08-16
HRP20220835T8 (hr) 2023-01-06
JP5911870B2 (ja) 2016-04-27
PT4066855T (pt) 2023-02-28
LT4066856T (lt) 2023-01-25
US20200230058A1 (en) 2020-07-23
HUE061043T2 (hu) 2023-05-28
LT4066855T (lt) 2023-03-10
EP4066857A1 (en) 2022-10-05
EP3981427B1 (en) 2022-05-25
AU2011295935A1 (en) 2013-04-04
PL3970742T3 (pl) 2022-09-19
EP2611461B1 (en) 2022-03-09
EP4101462A1 (en) 2022-12-14
ES2938866T3 (es) 2023-04-17
PT3970742T (pt) 2022-06-27
HUE058361T2 (hu) 2022-07-28
JP2013536803A (ja) 2013-09-26
EP3981427A1 (en) 2022-04-13
DK3981427T3 (da) 2022-07-11
HRP20221533T1 (hr) 2023-02-17
RS63315B1 (sr) 2022-07-29
PT4066857T (pt) 2023-01-31
SI3970742T1 (sl) 2022-08-31
PL2611461T3 (pl) 2022-07-04

Similar Documents

Publication Publication Date Title
US11759422B2 (en) Pegylated liposomes for delivery of immunogen-encoding RNA
US11883534B2 (en) Immunisation with lipid formulations with RNA encoding immunogens
US9254265B2 (en) Small liposomes for delivery of immunogen encoding RNA
EP2611420B1 (en) Lipids suitable for liposomal delivery of protein-coding rna

Legal Events

Date Code Title Description
AS Assignment

Owner name: NOVARTIS VACCINES AND DIAGNOSTICS, INC., CALIFORNI

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:GEALL, ANDREW;VERMA, AYUSH;SIGNING DATES FROM 20130201 TO 20130208;REEL/FRAME:029928/0231

Owner name: NOVARTIS AG, SWITZERLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:NOVARTIS VACCINES AND DIAGNOSTICS, INC.;REEL/FRAME:029928/0258

Effective date: 20130301

AS Assignment

Owner name: GLAXOSMITHKLINE BIOLOGICALS SA, BELGIUM

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:NOVARTIS AG;REEL/FRAME:038982/0135

Effective date: 20160615

AS Assignment

Owner name: GLAXOSMITHKLINE BIOLOGICALS SA, BELGIUM

Free format text: CORRECTIVE ASSIGNMENT TO CORRECT THE INCORRECT APPL. NO. 13/819,007 PREVIOUSLY RECORDED AT REEL: 038982 FRAME: 0135. ASSIGNOR(S) HEREBY CONFIRMS THE ASSIGNMENT;ASSIGNOR:NOVARTIS AG;REEL/FRAME:042683/0280

Effective date: 20160615

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

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

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

Free format text: FINAL REJECTION MAILED

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

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

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

Free format text: FINAL REJECTION MAILED

STCB Information on status: application discontinuation

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