WO2012031043A1 - Pegylated liposomes for delivery of immunogen-encoding rna - Google Patents

Pegylated liposomes for delivery of immunogen-encoding rna Download PDF

Info

Publication number
WO2012031043A1
WO2012031043A1 PCT/US2011/050095 US2011050095W WO2012031043A1 WO 2012031043 A1 WO2012031043 A1 WO 2012031043A1 US 2011050095 W US2011050095 W US 2011050095W WO 2012031043 A1 WO2012031043 A1 WO 2012031043A1
Authority
WO
WIPO (PCT)
Prior art keywords
rna
liposome
liposomes
peg
lipid
Prior art date
Application number
PCT/US2011/050095
Other languages
French (fr)
Inventor
Andrew Geall
Ayush Verma
Original Assignee
Novartis Ag
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=WO2012031043(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority to RS20220501A priority Critical patent/RS63315B1/en
Priority to EP21205422.5A priority patent/EP3981427B9/en
Priority to JP2013526210A priority patent/JP5911870B2/en
Priority to EP23179901.6A priority patent/EP4226941A1/en
Priority to CA2809851A priority patent/CA2809851A1/en
Priority to HRP20220695TT priority patent/HRP20220695T1/en
Priority to EP22188690.6A priority patent/EP4101462A1/en
Priority to CN2011800518918A priority patent/CN103179984A/en
Priority to EP22174931.0A priority patent/EP4066856B1/en
Priority to SI201132053T priority patent/SI2611461T1/en
Priority to DK11758014.2T priority patent/DK2611461T3/en
Priority to MX2013002336A priority patent/MX2013002336A/en
Priority to LTEPPCT/US2011/050095T priority patent/LT2611461T/en
Application filed by Novartis Ag filed Critical Novartis Ag
Priority to EP22192507.6A priority patent/EP4119155A1/en
Priority to EP21207859.6A priority patent/EP3970742B1/en
Priority to RU2013114362/10A priority patent/RU2013114362A/en
Priority to EP11758014.2A priority patent/EP2611461B1/en
Priority to EP23179897.6A priority patent/EP4226940A1/en
Priority to AU2011295935A priority patent/AU2011295935B2/en
Priority to US13/819,077 priority patent/US20130202684A1/en
Priority to BR112013004866A priority patent/BR112013004866A2/en
Priority to EP22174932.8A priority patent/EP4066857B1/en
Priority to EP22174779.3A priority patent/EP4066855B1/en
Priority to PL11758014T priority patent/PL2611461T3/en
Priority to EP22192506.8A priority patent/EP4147718A1/en
Priority to ES11758014T priority patent/ES2913791T3/en
Publication of WO2012031043A1 publication Critical patent/WO2012031043A1/en
Priority to US16/837,115 priority patent/US11759422B2/en

Links

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 lkDa and 3kDa. 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 lkDa and 3kDa.
  • 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 lkDa and 3kDa.
  • 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), l ,2-distearyloxy-N,N-dimethyl-3-aminopropane (DSDMA), l ,2-dioleyloxy-N,Ndimethyl- 3-aminopropane (DODMA), l,2-dilinoleyloxy-N,N-dimethyl-3-aminopropane (DLinDMA), 1,2- dilinolenyloxy-N,N-dimethyl-3-aminopropane (DLenDMA).
  • DOTAP dioleoyl trimethylammonium propane
  • DSDMA distearyloxy-N,N-dimethyl-3-aminopropane
  • DODMA dioleyloxy-N,Ndimethyl- 3-aminopropane
  • DLinDMA 1,2- dilinolenyloxy-N,N
  • Zwitterionic lipids include, but are not limited to, acyl zwitterionic lipids and ether zwitterionic lipids.
  • useful zwitterionic lipids are DPPC, DOPC, DSPC, dodecylphosphocholine, l,2-dioleoyl-sn-glycero-3- phosphatidylethanolamine (DOPE), and l,2-diphytanoyl-sn-glycero-3-phosphoethanolamine (DPyPE).
  • DOPE dioleoyl-sn-glycero-3- phosphatidylethanolamine
  • DPyPE l,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
  • 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 lkDa and 3kDa; 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 about 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 IkDa and 3kDa e.g. between 23 and 68, or about 45 for a 2kDa PEGylation (e.g. see FIG. 16).
  • the PEG moiety can terminate with an -O-meth l group, and so a PEGylated lipid may comprise:
  • a PEGylated lipid useful with the invention may comprise: Attorney Docket No.: PAT054248-WO-PCT
  • 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);
  • is an optionally substituted Cj.io alkylene or Ci.io heteroalkylene linker including zero, one or two of an ether (e.g., -0-), ester (e.g., -C(O)O-), succinate (e.g., -0(0)C-CH 2 -CH 2 -C(0)0-)), carbamate (e.g., -OC(0)-NR'-), carbonate (e.g., -OC(O)O-), urea (e.g., -NRC(O)NR -), amine (e.g., -NR'-), amide (e.g., -C(0)NR'-), imine (e.g., -C(NR')-), thioether (e.g., -S-), xanthate (e.g., -OC(S)S-), and phosphodiester (e.g., -OP(0) 2 0-), wherein R' is independently selected
  • Xi and X 2 are independently selected from a carbon or a heteroatom selected from -NH-, -0-, -S- or a phosphate;
  • Ai and A 2 are either independently selected from a Ce-30 alkyl, Ce-30 alkenyl, and C6.30 alkynyl, wherein A] and A 2 may be the same or different, or Ai 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 IkDa and 3kDa e.g. between 1.5-2.5kDa, between 1.7-2.3kDa, between 1.8-2.2kDa, between 1.9- 2.1 kDa, or 2kDa.
  • the PEG can be a PEG which is commonly known as "PEG 2000” or "PEG 2k", 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 Attorney Docket No.: PAT054248-WO-PCT
  • the average molecular mass of the PEG is 2kDa not lkDa.
  • 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 ⁇ 50nm, and LUVs have a diameter >50nm.
  • Liposomes of the invention are ideally LUVs with a diameter in the range of 60-180nm, and preferably in the range of 80-160nm.
  • 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 60-180nm, and preferably in the range of 80- 160nm, and/or (ii) the average diameter (by intensity e.g. Z-average) of the population is ideally in the range of 60-180nm, and preferably in the range of 80-160nm.
  • 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 600-800nm 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-repli eating 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.
  • strand transcripts can themselves be transcribed to give further copies of
  • 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 Attorney Docket No.: PAT054248-WO-PCT polymerase can be an alphavirus replicase e.g. comprising one or more of alphavirus proteins nsPl , 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.
  • dsRNA double-stranded form
  • RNA molecule useful with the invention can conveniently be prepared by in vitro transcription (rVT).
  • rV 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-tran scribed 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), mlA (1-methyladenosine); m2A (2-methyladenosine); Am (2'-0- methyladenosine); 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 one of these bacteria:
  • Neisseria meningitidis useful immunogens include, but are not limited to, membrane proteins such as adhesins, autotransporters, toxins, iron acquisition proteins, and factor H binding protein. A combination of three useful polypeptides is disclosed in reference 11.
  • Streptococcus pneumoniae useful polypeptide immunogens are disclosed in reference 12. These include, but are not limited to, the RrgB pilus subunit, the beta-N-acetyl-hexosaminidase precursor (spr0057), spr0096, General stress protein GSP-781 (spr2021, SP2216), serine/threonine kinase StkP (SP1732), and pneumococcal surface adhesin PsaA.
  • Streptococcus pyogenes include, but are not limited to, the polypeptides disclosed in references 13 and 14. Attorney Docket No.: PAT054248-WO-PCT
  • Bordetella pertussis Useful pertussis immunogens include, but are not limited to, pertussis toxin or toxoid (PT), filamentous haemagglutinin (FHA), pertactin, and agglutinogens 2 and 3.
  • Staphylococcus aureus Useful immunogens include, but are not limited to, the polypeptides disclosed in reference 15, such as a hemolysin, esxA, esxB, ferrichrome-binding protein
  • Clostridium tetani the typical immunogen is tetanus toxoid.
  • Comynebacterium diphtheriae the typical immunogen is diphtheria toxoid.
  • Haemophilus influenzae Useful immunogens include, but are not limited to, the polypeptides disclosed in references 16 and 17.
  • Streptococcus agalactiae useful immunogens include, but are not limited to, the polypeptides disclosed in reference 13.
  • Chlamydia trachomatis Useful immunogens include, but are not limited to, PepA, LcrE, ArtJ, Dnak, CT398, OmpH-like, L7/L12, OmcA, AtoS, CT547, Eno, HtrA and MurG (e.g. as disclosed in reference 18. LcrE [19] and HtrA [20] are two preferred immunogens.
  • Chlamydia pneumoniae Useful immunogens include, but are not limited to, the polypeptides disclosed in reference 21.
  • Helicobacter pylori Useful immunogens include, but are not limited to, CagA, VacA, NAP, and/or urease [22].
  • Escherichia coli Useful immunogens include, but are not limited to, immunogens derived from enterotoxigenic E. coli (ETEC), enteroaggregative E. coli (EAggEC), diffusely adhering E. coli (DAEC), enteropathogenic E. coli (EPEC), extraintestinal pathogenic E. coli (ExPEC) and/or enterohemorrhagic E. coli (EHEC).
  • ExPEC strains include uropathogenic E.coli (UPEC) and meningitis/sepsis-associated E.coli ( NEC).
  • Useful UPEC polypeptide immunogens are disclosed in references 23 and 24.
  • Useful MNEC immunogens are disclosed in reference 25.
  • a useful immunogen for several E.coli types is AcfD [26].
  • Yersinia pestis Useful immunogens include, but are not limited to, those disclosed in references 27 and 28.
  • Brucella such as B.abortus, B.canis, B.melitensis, B.neotomae, B.ovis, B.suis, B.pinnipediae.
  • Francisella such as F.novicida, F.philomiragia, F.tularensis.
  • Salmonella typhi Salmonella typhi
  • the immunogen elicits an immune response against one of these viruses: Orthomyxovirus: Useful immunogens can be from an influenza A, B or C virus, such as the hemagglutinin, neuraminidase or matrix M2 proteins. Where the immunogen is an influenza
  • a virus hemagglutinin it may be from any subtype e.g. HI, H2, H3, H4, H5, H6, H7, H8, H9, H10, HI 1, H12, H13, H14, H15 or H16.
  • Viral immunogens include, but are not limited to, those derived from Pneumoviruses (e.g. respiratory syncytial virus, RSV), Rubulaviruses (e.g. mumps virus), Paramyxoviruses (e.g. parainfluenza virus), Metapneumoviruses and Morbilliviruses (e.g. measles virus).
  • Pneumoviruses e.g. respiratory syncytial virus, RSV
  • Rubulaviruses e.g. mumps virus
  • Paramyxoviruses e.g. parainfluenza virus
  • Metapneumoviruses e.g. measles virus.
  • Viral immunogens include, but are not limited to, those derived from Orthopoxvirus such as Variola vera, including but not limited to, Variola major and Variola minor.
  • Viral immunogens include, but are not limited to, those derived from Picornaviruses, such as Enteroviruses, Rhinoviruses, Heparnavirus, Cardioviruses and
  • the enterovirus is a poliovirus e.g. a type 1 , type 2 and/or Attorney Docket No. : PAT054248- WO-PCT type 3 poliovirus.
  • the enterovirus is an EV71 enterovirus.
  • the enterovirus is a coxsackie A or B virus.
  • Viral immunogens include, but are not limited to, those derived from an Ort obunyavirus, such as California encephalitis virus, a Phlebovirus, such as Rift Valley Fever virus, or a Nairovirus, such as Crimean-Congo hemorrhagic fever virus.
  • an Ort obunyavirus such as California encephalitis virus
  • a Phlebovirus such as Rift Valley Fever virus
  • a Nairovirus such as Crimean-Congo hemorrhagic fever virus.
  • Viral immunogens include, but are not limited to, those derived from a Heparnavirus, such as hepatitis A virus (HAV).
  • HAV hepatitis A virus
  • Viral immunogens include, but are not limited to, those derived from a filovirus, such as an Ebola virus (including a Zaire, Ivory Coast, Reston or Sudan ebolavirus) or a Marburg virus.
  • a filovirus such as an Ebola virus (including a Zaire, Ivory Coast, Reston or Sudan ebolavirus) or a Marburg virus.
  • Viral immunogens include, but are not limited to, those derived from a Togavirus, such as a Rubivirus, an Alphavirus, or an Arterivirus. This includes rubella virus.
  • Flavivirus Viral immunogens include, but are not limited to, those derived from a Flavivirus, such as Tick-borne encephalitis (TBE) virus, Dengue (types 1, 2, 3 or 4) virus, Yellow Fever virus, Japanese encephalitis virus, Kyasanur Forest Virus, West Nile encephalitis virus, St. Louis encephalitis virus, Russian spring-summer encephalitis virus, Powassan encephalitis virus.
  • TBE Tick-borne encephalitis
  • Dengue types 1, 2, 3 or 4
  • Yellow Fever virus Japanese encephalitis virus
  • Kyasanur Forest Virus Japanese encephalitis virus
  • West Nile encephalitis virus West Nile encephalitis virus
  • St. Louis encephalitis virus St. Louis encephalitis virus
  • Russian spring-summer encephalitis virus Powassan encephalitis virus.
  • Viral immunogens include, but are not limited to, those derived from a Pestivirus, such as Bovine viral diarrhea (BVDV), Classical swine fever (CSFV) or Border disease (BDV).
  • BVDV Bovine viral diarrhea
  • CSFV Classical swine fever
  • BDV Border disease
  • Viral immunogens include, but are not limited to, those derived from a Hepadnavirus, such as Hepatitis B virus.
  • a composition can include hepatitis B virus surface antigen (HBsAg).
  • a composition can include an immunogen from a hepatitis C virus, delta hepatitis virus, hepatitis E virus, or hepatitis G virus.
  • Viral immunogens include, but are not limited to, those derived from a Rhabdovirus, such as a Lyssavirus (e.g. a Rabies virus) and Vesiculovirus (VSV).
  • a Lyssavirus e.g. a Rabies virus
  • VSV Vesiculovirus
  • Viral immunogens include, but are not limited to, those derived from Calciviridae, such as Norwalk virus (Norovirus), and Norwalk-like Viruses, such as Hawaii Virus and Snow Mountain Virus.
  • Viral immunogens include, but are not limited to, those derived from a SARS coronavirus, avian infectious bronchitis (IBV), Mouse hepatitis virus (MHV), and Porcine transmissible gastroenteritis virus (TGEV).
  • the coronavirus immunogen may be a spike polypeptide.
  • Retrovirus include, but are not limited to, those derived from an Oncovirus, a Lentivirus (e.g. HIV-1 or HIV-2) or a Spumavirus.
  • Viral immunogens include, but are not limited to, those derived from an Orthoreovirus, a Rotavirus, an Orbivirus, or a Coltivirus.
  • Viral immunogens include, but are not limited to, those derived from Parvovirus B 19.
  • Herpesvirus include, but are not limited to, those derived from a human herpesvirus, such as, by way of example only, Herpes Simplex Viruses (HSV) (e.g. HSV types 1 and 2), Varicella-zoster virus (VZV), Epstein-Barr virus (EBV), Cytomegalovirus (CMV), Human Herpesvirus 6 (HHV6), Human Herpesvirus 7 (HHV7), and Human
  • HSV Herpes Simplex Viruses
  • VZV Varicella-zoster virus
  • EBV Epstein-Barr virus
  • CMV Cytomegalovirus
  • HHV6 Human Herpesvirus 6
  • HHV7 Human Herpesvirus 7
  • Herpesvirus 8 (HHV8).
  • Viral immunogens include, but are not limited to, those derived from Papillomaviruses and Polyomaviruses.
  • the (human) papillomavirus may be of serotype 1 , 2, 4, 5, 6, 8, 11 , 13, 16, 18, 31, 33, 35, 39, 41, 42, 47, 51, 57, 58, 63 or 65 e.g. from one or more of serotypes 6, 1 1 , 16 and/or 18.
  • Viral immunogens include those derived from adenovirus serotype 36 (Ad-36).
  • 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 Dermatophytes, 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.
  • Cryptococcus neoformans Geotrichum clavatum, Histoplasma capsulatum, Klebsiella pneumoniae, Microsporidia, Encephalitozoon spp., Septata intestinalis and Enterocytozoon bieneusi; the less common are Brachiola spp, Microsporidium spp., Nosema spp., Pleistophora spp., Trachipleistophora spp., Vittaforma spp Paracoccidioides brasiliensis, Pneumocystis carinii, Pythiumn insidiosum, Pityrosporum ovale, Sacharomyces cerevisae, Saccharomyces boulardii, Saccharomyces pombe, Scedosporium apiosperum, Sporothrix schenckii, Trichosporon beigelii, Toxoplasma gondii, Penicillium marneffei, Malassezia s
  • 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 including 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 Attorney Docket No.: PAT054248-WO-PCT 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.
  • cancer-testis antigens
  • melanoma melanoma
  • MUM 1 associated with, e.g., melanoma
  • caspase-8 associated with, e.g. , head and neck cancer
  • CIA 0205 associated with, e.g., bladder cancer
  • HLA-A2-R1701 beta catenin (associated with, e.g., melanoma)
  • TCR associated with, e.g., T-cell non-Hodgkins lymphoma
  • BCR-abl associated with, e.g., chronic myelogenous leukemia
  • triosephosphate isomerase KIA 0205, CDC-27, and LDLR- FUT
  • over-expressed antigens for example, Galectin 4 (associated with, e.g., colorectal cancer), Galectin 9 (associated with, e.g., Hodgkin's disease), proteinase 3 (associated with, e.g.
  • chronic myelogenous leukemia WT 1 (associated with, e.g., various leukemias), carbonic anhydrase (associated with, e.g., renal cancer), aldolase A (associated with, e.g., lung cancer), PRAME (associated with, e.g., melanoma), HER-2/neu (associated with, e.g., breast, colon, lung and ovarian cancer), mammaglobin, alpha-fetoprotein (associated with, e.g., hepatoma), KSA (associated with, e.g., colorectal cancer), gastrin (associated with, e.g., pancreatic and gastric cancer), telomerase catalytic protein, MUC-1 (associated with, e.g., breast and ovarian cancer), G-250 (associated with, e.g., renal cell carcinoma), p53 (associated with, e.g.
  • melanoma-melanocyte differentiation antigens such as MART-l/Melan A, gplOO, MC 1R, melanocyte-stimulating hormone receptor, tyrosinase, tyrosinase related protein- 1/TRPl and tyrosinase related protein-2/TRP2 (associated with, e.g., melanoma);
  • prostate associated antigens such as PAP, PSA, PSMA, PSH-P1, PSM-P1, PSM-P2, associated with e.g., prostate cancer;
  • immunoglobulin idiotypes associated with myeloma and B cell lymphomas, for example).
  • tumor immunogens include, but are not limited to, pi 5, 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, pl 85erbB2, pl 80erbB-3, c-met, mn-23Hl, TAG-72-4, CA 19-9, CA 72-4, CAM 17.1 , NuMa, K-ras, pl6, TAGE, PSCA, CT7, 43-9F, 5T4, 791 Tgp72, beta-HCG, BCA225, BTAA, CA 125, CA 15-3 (CA 27.29XBCAA), CA 195, CA 242, CA-50, CAM43, CD68 ⁇ KP1, CO-0
  • 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 Attorney Docket No.: PAT054248-WO-PCT
  • TLR4 agonist e.g. an aminoalkyl glucosaminide phosphate, such as E6020
  • TLR7 agonist e.g. imiquimod
  • TLR8 agonist e.g. resiquimod
  • TLR9 agonist e.g. IC31
  • Any such agonist ideally has a molecular weight of ⁇ 2000Da.
  • such agonist(s) are also encapsulated with the RNA inside liposomes, but in other embodiments they are unencapsulated.
  • Pharmaceutical 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 5-20mM 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 ⁇ 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 -1.0ml e.g. about 0.5ml.
  • 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 ⁇ , 25 ⁇ g, 50 ⁇ g, 75 g or 100 ⁇ g), but expression can be seen at much lower levels e.g. ⁇ g/dose, ⁇ 100ng/dose, ⁇ 10ng/dose, ⁇ lng/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).
  • a mammal such as a human or a large veterinary mammal (e.g. horses, cattle, deer, goats, pigs).
  • the human is preferably a child Attorney Docket No.: PAT054248-WO-PCT
  • 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"
  • 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;
  • n is a number-averaged degree of polymerization between 10 and 200 units of Z, wherein n is optimized for different polymer types;
  • L ! is an optionally substituted C O alkylene or C M oheteroalkylene linker including zero, one or two of an ether (e.g., -0-), ester (e.g., -C(O)O-), succinate (e.g., -0(0)C-CH 2 -CH 2 -C(0)0-)), carbamate (e.g., -OC(0)-N '-), carbonate (e.g., -OC(O)O-), urea (e.g., -NRC(0)NR'-), amine (e.g., -NR'- amide (e.g., -C(0)N '-), imine (e.g., -C(NR')-), thioether (e.g., -S-), xanthate (e.g., -OC(S)S-), and phosphodiester (e.g., -OP(0) 2 0-),
  • an ether e.
  • R' is independently selected from -H, -NH-, -NH 2 , -CK -S-, a phosphate or an optionally substituted Cj.io alkylene;
  • Xi and X 2 are independently selected from a carbon or a heteroatom selected from -NH-, -0-, -S- or a phosphate;
  • a ! and A 2 are independently selected from a C6- 30 alkyl, C6-30 alkenyl, and C6.30 alkynyl, wherein Ai and A 2 may be the same or different, or Ai and A 2 together with the carbon atom to which they are attached form an optionally substituted steroid.
  • 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; Attorney Docket No.: PAT054248-WO-PCT
  • Li is an optionally substituted Q.io heteroalkylene linker containing one or two of an ether, ester, succinate, carbamate, carbonate, urea, amine, amide, imine, thioether, xanthate, and phosphodiester;
  • Xi and X 2 are oxygen
  • A! and A 2 are independently selected from a C 6 -3o alkyl, C 6 -3o alkenyl, and C6.30 alkynyl, wherein Ai and A 2 may be the same or different, or wherein Aj and A 2 together with the carbon atom to which they are attached form an optionally substituted steroid.
  • the lipids of formulae (X) and ( ⁇ '), 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-diacyl glycerol 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 ( l-[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), l ,2-dimyristoyl-sn-glycero-3-phosphoethanolamine-N-[methoxy (polyethylene glycol)-2000] (catalog #880150P from Avanti Polar Lipids).
  • PEGylated lipids are SOOl, S002, S003, S004, S005, S006, S007, S008, S009, SOlO, SOU , and CS-020SA (NOF); SOlO and S01 1 are disclosed in ref. 32 under the labels IVa and IVc, respectively. In ref. 32, a different synthesis from that reported herein is used to prepare IVa and rVc. Chemical terms and definitions
  • 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 Ci.ioalkyl, in another embodiment in another embodiment Ci-4alkyl, such as methyl, ethyl, n-propyl, i-propyl or t-butyl groups.
  • cycloalkyl includes monovalent, saturated, cyclic hydrocarbyl groups.
  • cycloalkyl is C 3 _i 0 cycloalkyl, in another embodiment C3. 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.
  • alkenyl is C 2 _ioalkenyl, 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 C3.i 0 cycloalkenyl, in another embodiment C5_iocycloalkenyl, 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 .i 0 alkynyl, in another embodiment C 2 -6alkynyl, in another embodiment C ⁇ alkyn l.
  • 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. In one embodiment cycloalkynyl is C3.] 0 cycloalkenyl, in another embodiment C5-iocycloalkynyl.
  • alkylene includes divalent, straight or branched, saturated, acyclic hydrocarbyl groups.
  • alkylene is Ci-ioalkylene, in another embodiment Ci ⁇ alkylene, in another embodiment Ci. 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 .i 0 alkenylene, in another embodiment C 2 _ 6 alkenylene, in another embodiment C . 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 Attorney Docket No.: PAT054248-WO-PCT double bonds.
  • alkynylene is C 2 -ioalkynylene, 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(0) q , N, P(0) r or Si (and preferably O, S(d) 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(0) q , N, P(0) 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(0) 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-oxazepanyl, 1 ,4-dithiepanyl, 1 ,4-thieazepanyl and 1,4-didi
  • 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(0) 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(0) 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(0) 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(0) q or N, provided at least one of the alkynyl carbon atoms remains.
  • the Attorney Docket No.: PAT054248-WO-PCT 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(0) 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(0) 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(0) 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(0) 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(0) 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 Ce-Cnaryl.
  • 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 Ce-Cnarylene.
  • 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 R N , where R N is defined below (and in one embodiment is H or alkyl (e.g. Ci_ 6 alkyl)).
  • heteroaryl groups may be monocyclic or polycyclic (e.g. bicyclic) fused ring heteroaromatic groups.
  • heteroaryl groups contain 5-13 ring members (preferably
  • a heteroaryl group may be 5, 6, 9 or 10 membered, e.g. 5-membered monocyclic,
  • 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 .
  • 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.
  • 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 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[l ,2-a]pyri
  • 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,
  • 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. Ci-ealkyl)).
  • 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 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. l ,2,3,4-tetrahydro-l ,8-naphthyridin-2-yl).
  • E is replaced by E or E (wherein E cannot be H);
  • R N is H or optionally substituted C].
  • R N is preferably H, C
  • 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 C3- 6 heteroalkyl
  • C3_ 6 heteroalkyl what is intended is a group based on C3_ 6 alkyl in which one or more of the 3-6 chain carbon atoms is replaced by O, S(0) q or N.
  • a C3_ 6 heteroalkyl group would, for example, contain less than 3-6 chain carbon atoms.
  • a pyridyl group would be classed as a Ce 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.
  • 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 1 or Sub 2 (in one embodiment Sub 2 ) wherein:
  • Z s is independently O, S or NR S ; Attorney Docket No. : PAT054248-WO-PCT
  • R s is independently H or -(Alk a ) r C 3 . 6 cycloalkyl, -(Alk a )rC 3 . 6 heterocycloalkyl, C 2 . 6 alkenyl, C 2 - 6 heteroalkenyl, -(Alk a )rC 3 - 6 cycloalkenyl, -(Alk a )rC 3 ⁇ heterocycloalkenyl, C 2-6 a]kynyl, C 2 .
  • Alk a is Ci_ 6 alkylene or C]. 6 heteroalkylene
  • R s is optionally substituted itself (in one embodiment unsubstituted) by 1 to 3 substituents Sub 2 ;
  • Z l is independently O, S, NH or N(d. 6 alkyl).
  • R s in Sub 1 can be optionally substituted by 1 to 3 substituents Sub 2 , Sub 2 is unsubstituted. However, in one embodiment, R s is unsubstituted.
  • R s is H or C
  • Sub 1 is not -R s and Sub 2 is not -Ci-ealkyl, -Ci ⁇ 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.
  • 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.
  • heteroalkyl when a group has a heteroatom, a substituent may be bonded to the heteroatom.
  • “optionally substituted heteroalkyl” includes -CH 2 -N(Sub ')-(3 ⁇ 4-, -CH(Sub')-NH-CH 2 - and -CH(Sub')-N(Sub')-CH 2 - etc.
  • 20 -heteroaryl group means that each of the four items in the list, namely the C3 -2 o-heterocycloalkyl group, the C3_ 2 o-heterocycloalkeny] group, the C3_ 2 o-heterocycloalkynyl group and the C6- 2 o-heteroaryl group, may be optionally substituted.
  • 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(0) q , N, P(0) r or Si (and preferably O, S(0) 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 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
  • RLR-2 also known as MDA5 or melanoma differentiation-associated gene 5
  • RLR-2 also has two caspase recruitment domains near its N-terminus.
  • 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: lkDa (triangles); 2kDa (circles); 3kDa (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), ⁇ g naked RNA, and l ⁇ g liposome-encapsulated RNA.
  • FIG. 8 shows anti-F IgG titers in animals receiving VRP, ⁇ ⁇ g naked RNA, and O. lg or l ⁇ g liposome-encapsulated RNA.
  • FIG. 9 shows neutralising antibody titers in animals receiving VRP or either O. lg or ⁇ 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. 1 1 shows F-specific IgG titers (2 weeks after second dose) after delivery of a replicon as naked RNA (0.01 -lpg), liposome-encapsulated RNA (0.01-l( ⁇ 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 (l g), liposome-encapsulated RNA (0.1 or lpg), 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 logio 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 (l-3kDa).
  • 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 lOkb 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 LiCl 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 2 60nm- 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 (l ,2-dilinoleyloxy-N,N-dimethyl-3-aminopropane) was synthesized using the procedure of reference 2.
  • DSPC (l ,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 (3P-[N-(N',N'-dimethylaminoethane)- carbamoyl]cholesterol hydrochloride) were from Avanti Polar Lipids.
  • 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 141nm (by intensity) or 78nm (by number).
  • fresh lipid stock solutions in ethanol were prepared.
  • 37 mg of DlinDMA, 1 1.8 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.
  • a 2 mL working solution Attorney Docket No.: PAT054248-WO-PCT of RNA was also prepared from a stock solution of - ⁇ / ⁇ . 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.
  • citrate buffer pH 6
  • Syringes containing RNA and the lipids were connected to a T mixer (PEEKTM 500 ⁇ ID junction) using FEP tubing(fluorinated ethylene-propylene; all FEP tubing used had a 2mm internal diameter and a 3mm 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.
  • 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 7mL/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 IX 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 IX 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 lOx or lOOx in I X TE buffer (from kit) before addition of the dye. Separately, liposomes were diluted lOx or lOOx in IX 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 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, 1 1.8 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 Attorney Docket No.: PAT054248-WO-PCT 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 ⁇ ⁇ g ⁇ 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 x 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 ⁇ x 105 ⁇ ) 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 l/16inches 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 3mL/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 IX PBS using the TFF system before recovering the final product. Hollow fiber filtration membranes with a 100 kDa pore size cutoff and 20cm 2 surface area were used.
  • formulations were diluted to the required RNA concentration with IX PBS.
  • liposomes prepared using the syringe/tube method with 75 ⁇ g RNA had a Z-average diameter (Zav) of 148nm and a polydispersity index (pdl) of 0.122
  • the chip mixing gave liposomes with a Zav of 97nm and a pdl 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 3.8mAU 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 y/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 12k 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
  • RNA a reporter enzyme SEAP; secreted alkaline phosphatase
  • SEAP secreted alkaline phosphatase
  • Expression levels were measured in sera diluted 1 :4 in IX 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 ⁇ 1 per t leg with O. ⁇ g or ⁇ g RNA dose. The same vector was also administered without the liposomes (in RNase free IX PBS) at l 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 lpg dose, and at day 6 expression from a 0.1 g encapsulated dose matched levels seen with ⁇ 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 lOx 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, O. ⁇ g RNA), or mixed with the liposomes after their formation (a non-encapsulated "lipoplex", O. ⁇ g RNA), or as naked RNA Ong).
  • 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 pg, the encapsulated RNA at lpg, 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 lpg liposome-encapsulated RNA was statistically insignificant (ANOVA) when compared to VRP, but the higher response seen with 10pg liposome- encapsulated RNA was statistically significant (p ⁇ 0.05) when compared to both of these groups.
  • lpg of liposome-encapsulated RNA gave much higher anti-F IgG responses ( 15 days post-second dose) than O.lpg of delivered DNA, and even was more immunogenic than 20pg plasmid DNA encoding the F antigen, delivered by electroporation (ElgenTM DNA Delivery System, Inovio).
  • RNA working solution was also prepared from a stock solution of - l ⁇ g/ ⁇ L ⁇ in 100 mM citrate buffer (pH 6). Three 20 mL glass vials (with stir bars) were rinsed with RNase Away solution (Molecular BioProducts, San Diego, CA) 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.
  • 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.
  • 4 ml of the mixture was loaded into a 5 cc syringe, which was connected to a piece of FEP tubing and in another 5 cc syringe connected to an equal length of FEP tubing, an equal amount of 100 mM citrate buffer (pH 6) was loaded.
  • the two syringes were driven at 7mL/min flow rate using the syringe, pump and the final mixture collected in a 20 mL glass vial (while stirring).
  • the mixture collected from the second mixing step were passed through a Mustang Q membrane (an anion-exchange support that binds and. removes anionic molecules, obtained from Pall Corporation, Ann Arbor, MI, USA).
  • a Mustang Q membrane an anion-exchange support that binds and. removes anionic molecules, obtained from Pall Corporation, Ann Arbor, MI, USA.
  • 4 mL of 1 M NaOH, 4 mL of 1 M NaCl and 10 mL of 100 mM citrate buffer (pH 6) were successively passed through the Mustang membrane. Liposomes were warmed for 10 min at 37°C before passing through the membrane. Next, liposomes were concentrated to 2 mL and dialyzed against 10-15 volumes of IX PBS' using TFF before recovering the final product.
  • TFF system and hollow fiber filtration membranes were purchased from Spectrum Labs and were used according to the manufacturer's guidelines.
  • Polysulfone hollow fiber filtration membranes (part number P/N: X1AB- 100-20P) with a 100 kD pore size cutoff and 8 cm 2 surface area were used.
  • formulations were diluted to the required RNA concentration with IX PBS.
  • 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 pm x 105 pm) 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 h.
  • 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 pL per leg) on days 0 and 21 with the replicon (lpg) 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% Attorney Docket No.: PAT054248-WO-PCT
  • the RV13 liposomes had 40% DOTAP, 10% DOPE, 48% cholesterol and 2% PEG- DMG. In all cases the PEG was PEG-2000 (i.e. 2kDa PEG).
  • naked plasmid DNA (20 ⁇ ) expressing the same RSV-F antigen was delivered either using electroporation or with RV01(10) liposomes (O. ⁇ g DNA).
  • Four mice were used as a naive control group.
  • 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:
  • 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: Attorney Docket No.: PAT054248-WO-PCT
  • 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 2kDa 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% Choi, 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 ⁇ 1 per leg) on days 0 & 21. Doses were 0.01, 0.03, 0.1, 0.3 or ⁇ g. F-specific serum IgG and PRNT60 titers (GMT) were as follows, 2 weeks after the first or second injection:
  • RV01 liposomes with DLinDMA as the cationic lipid and 2kDa 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 ⁇ _ per leg) on days 0, 21 and 42 with VRPs expressing gH gL (l xlO 6 IU), VRPs expressing gHsol/gL (l xlO 6 IU) and PBS as the controls.
  • Two test groups received ⁇ g of the vA160 or vA322 replicon formulated in liposomes (40% DlinDMA, 10% DSPC, 48% Choi, 2% PEG-DMG; made using method (D) but with 150 ⁇ g RNA batch size).
  • the vA160 liposomes had a Zav diameter of 168.8nm, a pdl of 0.144, and 87.4% encapsulation.
  • the vA322 liposomes had a Zav diameter of 162nm, a pdl 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.
  • vA31 1 A self-replicating RNA replicon (“vA31 1") that expresses a luciferase reporter gene (luc) was used for studying the kinetics of protein expression after injection.
  • VRP (lxlO 6 IU) expressing luciferase
  • 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 intraperitoneally 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.
  • 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 ⁇ into the hind leg, which is a relatively high volume for a mouse leg muscle.
  • a human intramuscular dose of ⁇ 0.5ml 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 :
  • Group 1 received naked replicon, 0 ⁇ g in 50 ⁇ L ⁇ per leg
  • Group 2 received naked replicon, 0.2 g in 5 ⁇ - per leg
  • Group 3 received emulsion-formulated replicon (0.2 ⁇ g, 50 ⁇ per leg)
  • Group 4 received emulsion-formulated replicon (0.2 ⁇ g, 5 ⁇ ⁇ per leg)
  • Group 5 received liposome-formulated replicon (0.2 ⁇ g, 50 ⁇ per leg)
  • Group 6 received liposome-formulated replicon (0.2 ⁇ g, 5 ⁇ L ⁇ per leg)
  • 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.
  • Cotton rats, 4 to 8 animals per group, were given intramuscular vaccinations (100 ⁇ L ⁇ in one leg) on days 0 and 21 with the four different replicons at two doses (1.0 and O.
  • lpg formulated in liposomes made using 2kDa PEG-conjugated DMG by method (D), but with a 150 g RNA batch size.
  • Control Attorney Docket No.: PAT054248-WO-PCT groups received a RSV-F subunit protein vaccine (5 ⁇ g) adjuvanted with alum (8 animals/group), VRPs expressing full-length RSV-F (lxlO 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 lpg 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 ⁇ 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 (lxlO 6 IU, 8 animals/group).
  • F-specific serum IgG titers were as follows: Attorney Docket No.: PAT054248-WO-PCT
  • 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.
  • the 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 2kDa PEG, conjugated to DMG.
  • a control group received 5pg alum-adjuvanted protein, and a naive 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:
  • RNA vaccine elicited F-specific serum IgG titers that were detected in a few calves after the second vaccination and in all calves after the third.
  • 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 66 ⁇ g of RNA.
  • liposomes were prepared using DMG to which five different PEGs were conjugated.
  • the average molecular weight of the PEG was 500Da, 750Da, lkDa, 2kDa or 3kDa.
  • Liposomes formed using the shortest PEGs were unstable or aggregated during TFF purification.
  • PEG-750 gave liposomes with a significantly higher Zaverage diameter (669nm) 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 had a small effect on liposome diameter and polydispersity index.
  • the Z-average diameter was 197nm (0.1 19 pdl) for the lkDa PEG, 142nm (0.137 pdl) for the 2kDa PEG, and 147nm (0.075 pdl) for the 3kDa 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.
  • mice 8 per group, were given bilateral intramuscular vaccinations (50 ⁇ ]_ per leg) on days 0 and ' 21 with the replicon,- either naked ( ⁇ g) or encapsulated in these liposomes (O. ⁇ 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 (2wpl):
  • the liposomes had 40% cationic lipid (RV05) and 2% PEGylated DMG (2kDa 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:
  • aGC a-galactosylceramide
  • mice 8 per group, were given bilateral intramuscular vaccinations (50 ⁇ per leg) on days 0 and 21 with the replicon, either naked ( ⁇ g) or encapsulated (O. ⁇ 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 (2wp 1 ) :

Abstract

Nucleic acid immunisation is achieved by delivering RNA encapsulated within a PEGylated liposome. The RNA encodes an immunogen of interest. The PEG has an average molecular mass of between lkDa and 3kDa. Thus 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 lkDa and 3kDa; and (ii) the aqueous core includes a RNA which encodes an immunogen. These 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.

Description

Attorney Docket No.: PAT054248-WO-PCT
PEGYLATED LIPOSOMES FOR DELIVERY OF IMMUNOGEN-ENCODING RNA
This application claims the benefit of U.S. provisional application number 61/378,826, which was filed August 31 , 2010, the complete contents of which are hereby incorporated herein by reference for all purposes. TECHNICAL FIELD
This invention is in the field of non-viral delivery of RNA for immunisation.
BACKGROUND ART
The delivery of 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.
There remains a need for further and improved nucleic acid vaccines and, in particular, for improved ways of delivering nucleic acid vaccines.
DISCLOSURE OF THE INVENTION
According to the invention, 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 lkDa and 3kDa. PEG with a lower molecular weight (e.g. 500 or 750 Da) does not form stable liposomes.
Thus 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 lkDa and 3kDa. These 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 lkDa and 3kDa. Attorney Docket No. : PAT054248- WO-PCT
The liposome
The invention utilises liposomes within which immunogen-encoding RNA is encapsulated. Thus 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.
Various amphiphilic lipids can form bilayers in an aqueous environment to encapsulate a RNA- containing aqueous core as a liposome. These lipids can have an anionic, cationic or zwitterionic hydrophilic head group. Formation of liposomes from 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), l ,2-distearyloxy-N,N-dimethyl-3-aminopropane (DSDMA), l ,2-dioleyloxy-N,Ndimethyl- 3-aminopropane (DODMA), l,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. Examples of useful zwitterionic lipids are DPPC, DOPC, DSPC, dodecylphosphocholine, l,2-dioleoyl-sn-glycero-3- phosphatidylethanolamine (DOPE), and l,2-diphytanoyl-sn-glycero-3-phosphoethanolamine (DPyPE). 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.
Thus in one embodiment 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 lkDa and 3kDa; 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. Similarly, a mixture may comprise both saturated and unsaturated lipids. For example, a mixture may comprise DSPC (zwitterionic, saturated), DlinDMA (cationic, unsaturated), and/or DMG (anionic, saturated). Where a mixture of Attorney Docket No.: PAT054248-WO-PCT lipids is used, not all of the component lipids in the mixture need to be amphophilic e.g. one or more amphiphilic lipids can be mixed with cholesterol.
Where a liposome of the invention is formed from a mixture of lipids, it is preferred that 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 about 2%. For instance, 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.
Thus 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 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.
Thus the PEGylated lipids will comprise the PEG structure:
O J n
where n provides a molecular weight for the PEG of between IkDa and 3kDa e.g. between 23 and 68, or about 45 for a 2kDa PEGylation (e.g. see FIG. 16).
The PEG moiety can terminate with an -O-meth l group, and so a PEGylated lipid may comprise:
Figure imgf000004_0001
Including attachment to a nitrogen in a lipid's head group, therefore, a PEGylated lipid useful with the invention may comprise:
Figure imgf000004_0002
Attorney Docket No.: PAT054248-WO-PCT
One suitable PEGylated lipid for use with the invention is PEG-DMG, as used in the examples. 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):
Figure imgf000005_0001
wherein:
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| is an optionally substituted Cj.io alkylene or Ci.io heteroalkylene linker including zero, one or two of an ether (e.g., -0-), ester (e.g., -C(O)O-), succinate (e.g., -0(0)C-CH2-CH2-C(0)0-)), carbamate (e.g., -OC(0)-NR'-), carbonate (e.g., -OC(O)O-), urea (e.g., -NRC(O)NR -), amine (e.g., -NR'-), amide (e.g., -C(0)NR'-), imine (e.g., -C(NR')-), thioether (e.g., -S-), xanthate (e.g., -OC(S)S-), and phosphodiester (e.g., -OP(0)20-), wherein R' is independently selected from -H, - NH-, - H2, -0-, -S-, a phosphate or an optionally substituted Ci-J0 alkylene;
Xi and X2 are independently selected from a carbon or a heteroatom selected from -NH-, -0-, -S- or a phosphate;
Ai and A2 are either independently selected from a Ce-30 alkyl, Ce-30 alkenyl, and C6.30 alkynyl, wherein A] and A2 may be the same or different, or Ai and A2 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 IkDa and 3kDa e.g. between 1.5-2.5kDa, between 1.7-2.3kDa, between 1.8-2.2kDa, between 1.9- 2.1 kDa, or 2kDa. Thus the PEG can be a PEG which is commonly known as "PEG 2000" or "PEG 2k", 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.
It is also possible for a single lipid molecule to include more than one PEG group e.g. attached to different carbon atoms in a lipid's head group (e.g. see FIG. 18). In these circumstances the reference to the molecular mass of PEG in a liposome is the molecular mass per lipid molecule rather than per Attorney Docket No.: PAT054248-WO-PCT
PEG substituent. Thus, in a liposome in which the sole PEGylated lipid has the structure shown in FIG. 18, where the boxed molecular weight is 2kDa and is made up of two chains of lkDa each, the average molecular mass of the PEG is 2kDa not lkDa.
In some embodiments 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.
In some embodiments the PEG may include copolymer groups e.g. one or more propylene monomers, to form a PEG polypropylene polymer.
As an alternative to PEGylation, a lipid may be modified by covalent attachment of a moiety different from PEG. For instance, in some embodiments a lipid may include a polyphosphazene. In some embodiments a lipid may include a poly(vinyl pyrrolidone). In some embodiments a lipid may include a poly(acryl amide). In some embodiments a lipid may include a poly(2-methyl-2-oxazoline). In some embodiments a lipid may include a poly(2-ethyl-2-oxazoline). In some embodiments a lipid may include a phosphatidyl polyglycerol. In some embodiments a lipid may include a poly[N-(2- hydroxypropyl) methacrylamide]. In some embodiments 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 <50nm, and LUVs have a diameter >50nm. Liposomes of the invention are ideally LUVs with a diameter in the range of 60-180nm, and preferably in the range of 80-160nm.
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. For 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 60-180nm, and preferably in the range of 80- 160nm, and/or (ii) the average diameter (by intensity e.g. Z-average) of the population is ideally in the range of 60-180nm, and preferably in the range of 80-160nm. 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 600-800nm and to have a high polydispersity.
Techniques for preparing suitable liposomes are well known in the art e.g. see references 4 to 6. One useful method is described in reference 7 and involves mixing (i) an ethanolic solution of the lipids (ii) an aqueous solution of the nucleic acid and (iii) buffer, followed by mixing, equilibration, dilution and purification. Preferred liposomes of the invention are obtainable by this mixing process. Attorney Docket No.: PAT054248-WO-PCT
To obtain liposomes with the desired diameter(s), 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.
The RNA
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.
The 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-repli eating 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. Thus the delivered RNA leads to the production of multiple daughter RNAs. These daughter RNAs, as well as collinear subgenomic transcripts, 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.
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. 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 Attorney Docket No.: PAT054248-WO-PCT polymerase can be an alphavirus replicase e.g. comprising one or more of alphavirus proteins nsPl , nsP2, nsP3 and nsP4.
Whereas natural alphavirus genomes encode structural virion proteins in addition to the non-structural replicase polyprotein, it is preferred that a self-replicating RNA molecule of the invention does not encode alphavirus structural proteins. Thus 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. The 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.
Thus a self-replicating 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. In some embodiments 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.
A 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 (dsRNA) 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. Attorney Docket No.: PAT054248-WO-PCT
A RNA molecule useful with the invention can conveniently be prepared by in vitro transcription (rVT). rV 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). For instance, a DNA-dependent RNA polymerase (such as the bacteriophage T7, T3 or SP6 RNA polymerases) can be used to transcribe the RNA from a DNA template. 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). These 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-tran scribed RNA can function efficiently as a substrate for its self-encoded replicase.
As discussed in reference 9, the self-replicating RNA can include (in addition to any 5' cap structure) one or more nucleotides having a modified nucleobase. Thus the RNA can comprise m5C (5- methylcytidine), m5U (5-methyluridine), m6A (N6-methyladenosine), s2U (2-thiouridine), Um (2'- O-methyluridine), mlA (1-methyladenosine); m2A (2-methyladenosine); Am (2'-0- methyladenosine); ms2m6A (2-methylthio-N6-methyladenosine); i6A (N6-isopentenyladenosine); ms2i6A (2-methylthio-N6isopentenyladenosine); io6A (N6-(cis-hydroxyisopentenyl)adenosine); ms2io6A (2-methylthio-N6-(cis-hydroxyisopentenyl) adenosine); g6A (N6- glycinylcarbamoyladenosine); t6A (N6-threonyl carbamoyladenosine); ms2t6A (2-methylthio-N6- threonyl carbamoyladenosine); m6t6A (N6-methyl-N6-threonylcarbamoyladenosine); hn6A(N6.- hydroxynorvalylcarbamoyl adenosine); ms2hn6A (2-methylthio-N6-hydroxynorvalyl carbamoyladenosine); Ar(p) (2'-0-ribosyladenosine (phosphate)); I (inosine); mi l (1- methylinosine); m'lm (l,2'-0-dimethylinosine); m3C (3-methylcytidine); Cm (2T-0-methylcytidine); s2C (2-thiocytidine); ac4C (N4-acetylcytidine); f5C (5-fonnylcytidine); m5Cm (5,2-0- dimethylcytidine); ac4Cm (N4acetyl2TOmethylcytidine); k2C (lysidine); mlG (1-methylguanosine); m2G (N2-methylguanosine); m7G (7-methylguanosine); Gm (2'-0-methylguanosine); m22G (N2,N2-dimethylguanosine); m2Gm (N2,2'-0-dimethylguanosine); - m22Gm (N2,N2,2'-0- trimethylguanosine); Gr(p) (2'-0-ribosylguanosine (phosphate)) ; yW (wybutosine); o2yW (peroxywybutosine); OHyW (hydroxy wybutosine); OHyW* (undermodified hydroxywybutosine); imG (wyosine); mimG (methylguanosine); Q (queuosine); oQ (epoxyqueuosine); galQ (galtactosyl- queuosine); manQ (mannosyl-queuosine); preQo (7-cyano-7-deazaguanosine); preQi (7- aminomethyl-7-deazaguanosine); G* (archaeosine); D (dihydrouridine); m5Um (5,2'-0- dimethyluridine); s4U (4-thiouridine); m5s2U (5-methyl-2-thiouridine); s2Um (2-thio-2'-0- methyluridine); acp3U (3-(3-amino-3-carboxypropyl)uridine); ho5U (5-hydroxyuridine); mo5U (5- methoxyuridine); cmo5U (uridine 5-oxyacetic acid); mcmo5U (uridine 5-oxyacetic acid methyl ester); chm5U (5-(carboxyhydroxymethyl)uridine)); mchm5U (5-(carboxyhydroxymethyl)uridine methyl ester); mcm5U (5-methoxycarbonyl methyluridine); mcm5Um (S-methoxycarbonylmethyl-2- O-methyluricjine); mcm5s2U (5-methoxycarbonylmethyl-2-thiouridine); nm5s2U (5-aminomethyl- 2-thiouridine); mnm5U (5-methylaminomethyluridine); mnm5s2U (5-methylaminomethyl-2- Attorney Docket No.: PAT054248-WO-PCT thiouridine); mnm5se2U (5-methylaminornethyl-2-selenouridine); ,ncm5U (5-carbamoylmethyl uridine); ncm5Um (5-carbamoylmethyl-2'-0-methyluridine); cmnm5U (5- carboxymethylaminomethyluridine); cnmm5Um (5-carboxymethylaminomethyl-2-L-0- methyl uridine); cmnm5s2U (5-carboxymethylaminomethyl-2-thiouridine); m62A (N6,N6- dimethyladenosine); Tm (2'-0-methylinosine); m4C (N4-methylcytidine); m4Cm (N4,2-0- dimethylcytidine); hm5C (5-hydroxymethylcytidine); m3U (3-methyluridine); cm5U (5- carboxymethyluridine); m6Am (N6,T-0-dimethyladenosine); rn62Am (N6,N6,0-2- trimethyladenosine); m2'7G (N2,7-dimethylguanosine); m2'2'7G (N2,N2,7-trimethylguanosine); m3Um (3,2T-0-dimethyluridine); m5D (5-methyldihydrouridine); f5Cm (5-formyl-2'-0- methylcytidine); mlGm (l,2'-0-dimethylguanosine); m'Am (1,2-O-dimethyl adenosine) irinomethyluridine); tm5s2U (S-taurinomethyl-2-thiouridine)); imG-14 (4-demethyl guanosine); imG2 (isoguanosine); or ac6A (N6-acetyladenosine), hypoxanthine, inosine, 8-oxo-adenine, 7- substituted derivatives thereof, dihydrouracil, pseudouracil, 2-thiouracil, 4-thiouracil, 5-aminouracil, 5-(Cl-C6)-alkyluracil, 5-methyluracil, 5-(C2-C6)-alkenyluracil, 5-(C2-C6)-alkynyluracil, 5- (hydroxymethyl)uracil, 5-chlorouracil, 5-fluorouracil, 5-bromouracil, 5-hydroxycytosine, 5-(Cl-C6 )-alkylcytosine, 5-methylcytosine, 5-(C2-C6)-alkenylcytosine, 5-(C2-C6)-alkynylcytosine, 5- chlorocytosine, 5-fluorocytosine, 5-bromocytosine, N2-dimethylguanine, 7-deazaguanine, 8- azaguanine, 7-deaza-7-substituted guanine, 7-deaza-7-(C2-C6)alkynylguanine, 7-deaza-8-substituted guanine, 8-hydroxyguanine, 6-thioguanine, 8-oxoguanine, 2-aminopurine, 2-amino-6-chloropurine, 2,4-diaminopurine, 2,6-diaminopurine, 8-azapurine, substituted 7-deazapurine, 7-deaza-7-substituted purine, 7-deaza-8-substituted purine, or an abasic nucleotide. For instance, a self-replicating RNA can include one or more modified pyrimidine nucleobases, such as pseudouridine and/or 5-methylcytosine residues. In some embodiments, however, 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). In other embodiments, 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.
Ideally, 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.
The amount of 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. Attorney Docket No.: PAT054248-WO-PCT
The immunogen
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.
Unlike references 1 and 10, the RNA encodes an immunogen. For the avoidance of doubt, the invention does not encompass RNA which encodes a firefly luciferase or which encodes a fusion protein of E.coli β-galactosidase or which encodes a green fluorescent protein (GFP). 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. Thus the immunogen also is not a self protein which is delivered to supplement or substitute for a defective host protein (as in gene therapy). Also, the RNA is not total mouse thymus RNA.
In some embodiments the immunogen elicits an immune response against one of these bacteria: Neisseria meningitidis: useful immunogens include, but are not limited to, membrane proteins such as adhesins, autotransporters, toxins, iron acquisition proteins, and factor H binding protein. A combination of three useful polypeptides is disclosed in reference 11.
Streptococcus pneumoniae: useful polypeptide immunogens are disclosed in reference 12. These include, but are not limited to, the RrgB pilus subunit, the beta-N-acetyl-hexosaminidase precursor (spr0057), spr0096, General stress protein GSP-781 (spr2021, SP2216), serine/threonine kinase StkP (SP1732), and pneumococcal surface adhesin PsaA.
Streptococcus pyogenes: useful immunogens include, but are not limited to, the polypeptides disclosed in references 13 and 14. Attorney Docket No.: PAT054248-WO-PCT
Moraxella catarrhalis.
Bordetella pertussis: Useful pertussis immunogens include, but are not limited to, pertussis toxin or toxoid (PT), filamentous haemagglutinin (FHA), pertactin, and agglutinogens 2 and 3.
Staphylococcus aureus: Useful immunogens include, but are not limited to, the polypeptides disclosed in reference 15, such as a hemolysin, esxA, esxB, ferrichrome-binding protein
(sta006) and/or the staOl 1 lipoprotein.
Clostridium tetani: the typical immunogen is tetanus toxoid.
Comynebacterium diphtheriae: the typical immunogen is diphtheria toxoid.
Haemophilus influenzae: Useful immunogens include, but are not limited to, the polypeptides disclosed in references 16 and 17.
Pseudomonas aeruginosa
Streptococcus agalactiae: useful immunogens include, but are not limited to, the polypeptides disclosed in reference 13.
Chlamydia trachomatis: Useful immunogens include, but are not limited to, PepA, LcrE, ArtJ, Dnak, CT398, OmpH-like, L7/L12, OmcA, AtoS, CT547, Eno, HtrA and MurG (e.g. as disclosed in reference 18. LcrE [19] and HtrA [20] are two preferred immunogens.
Chlamydia pneumoniae: Useful immunogens include, but are not limited to, the polypeptides disclosed in reference 21.
Helicobacter pylori: Useful immunogens include, but are not limited to, CagA, VacA, NAP, and/or urease [22].
Escherichia coli: Useful immunogens include, but are not limited to, immunogens derived from enterotoxigenic E. coli (ETEC), enteroaggregative E. coli (EAggEC), diffusely adhering E. coli (DAEC), enteropathogenic E. coli (EPEC), extraintestinal pathogenic E. coli (ExPEC) and/or enterohemorrhagic E. coli (EHEC). ExPEC strains include uropathogenic E.coli (UPEC) and meningitis/sepsis-associated E.coli ( NEC). Useful UPEC polypeptide immunogens are disclosed in references 23 and 24. Useful MNEC immunogens are disclosed in reference 25. A useful immunogen for several E.coli types is AcfD [26].
Bacillus anthracis
Yersinia pestis: Useful immunogens include, but are not limited to, those disclosed in references 27 and 28.
Staphylococcus epidermis
Clostridium perfringens or Clostridium botulinums
Legionella pneumophila Attorney Docket No.: PAT054248-WO-PCT
Coxiella burnetii
Brucella, such as B.abortus, B.canis, B.melitensis, B.neotomae, B.ovis, B.suis, B.pinnipediae. Francisella, such as F.novicida, F.philomiragia, F.tularensis.
Neisseria gonorrhoeae
Treponema pallidum
Haemophilus ducreyi
Enterococcus faecalis or Enterococcus faecium
Staphylococcus saprophytics
Yersinia enterocolitica
Mycobacterium tuberculosis
Rickettsia
Listeria monocytogenes
Vibrio cholerae
Salmonella typhi
Borrelia burgdorferi
Porphyromonas gingivalis
Klebsiella
In some embodiments the immunogen elicits an immune response against one of these viruses: Orthomyxovirus: Useful immunogens can be from an influenza A, B or C virus, such as the hemagglutinin, neuraminidase or matrix M2 proteins. Where the immunogen is an influenza
A virus hemagglutinin it may be from any subtype e.g. HI, H2, H3, H4, H5, H6, H7, H8, H9, H10, HI 1, H12, H13, H14, H15 or H16.
Paramyxoviridae viruses: Viral immunogens include, but are not limited to, those derived from Pneumoviruses (e.g. respiratory syncytial virus, RSV), Rubulaviruses (e.g. mumps virus), Paramyxoviruses (e.g. parainfluenza virus), Metapneumoviruses and Morbilliviruses (e.g. measles virus).
Poxviridae: Viral immunogens include, but are not limited to, those derived from Orthopoxvirus such as Variola vera, including but not limited to, Variola major and Variola minor.
Picornavirus: Viral immunogens include, but are not limited to, those derived from Picornaviruses, such as Enteroviruses, Rhinoviruses, Heparnavirus, Cardioviruses and
Aphthoviruses. In one embodiment, the enterovirus is a poliovirus e.g. a type 1 , type 2 and/or Attorney Docket No. : PAT054248- WO-PCT type 3 poliovirus. In another embodiment, the enterovirus is an EV71 enterovirus. In another embodiment, the enterovirus is a coxsackie A or B virus.
Bunyavirus: Viral immunogens include, but are not limited to, those derived from an Ort obunyavirus, such as California encephalitis virus, a Phlebovirus, such as Rift Valley Fever virus, or a Nairovirus, such as Crimean-Congo hemorrhagic fever virus.
Heparnavirus: Viral immunogens include, but are not limited to, those derived from a Heparnavirus, such as hepatitis A virus (HAV).
Filovirus: Viral immunogens include, but are not limited to, those derived from a filovirus, such as an Ebola virus (including a Zaire, Ivory Coast, Reston or Sudan ebolavirus) or a Marburg virus.
Togavirus: Viral immunogens include, but are not limited to, those derived from a Togavirus, such as a Rubivirus, an Alphavirus, or an Arterivirus. This includes rubella virus.
Flavivirus: Viral immunogens include, but are not limited to, those derived from a Flavivirus, such as Tick-borne encephalitis (TBE) virus, Dengue (types 1, 2, 3 or 4) virus, Yellow Fever virus, Japanese encephalitis virus, Kyasanur Forest Virus, West Nile encephalitis virus, St. Louis encephalitis virus, Russian spring-summer encephalitis virus, Powassan encephalitis virus.
Pestivirus: Viral immunogens include, but are not limited to, those derived from a Pestivirus, such as Bovine viral diarrhea (BVDV), Classical swine fever (CSFV) or Border disease (BDV).
Hepadnavirus: Viral immunogens include, but are not limited to, those derived from a Hepadnavirus, such as Hepatitis B virus. A composition can include hepatitis B virus surface antigen (HBsAg).
Other hepatitis viruses: A composition can include an immunogen from a hepatitis C virus, delta hepatitis virus, hepatitis E virus, or hepatitis G virus.
Rhabdovirus: Viral immunogens include, but are not limited to, those derived from a Rhabdovirus, such as a Lyssavirus (e.g. a Rabies virus) and Vesiculovirus (VSV).
Caliciviridae: Viral immunogens include, but are not limited to, those derived from Calciviridae, such as Norwalk virus (Norovirus), and Norwalk-like Viruses, such as Hawaii Virus and Snow Mountain Virus.
Coronavirus: Viral immunogens include, but are not limited to, those derived from a SARS coronavirus, avian infectious bronchitis (IBV), Mouse hepatitis virus (MHV), and Porcine transmissible gastroenteritis virus (TGEV). The coronavirus immunogen may be a spike polypeptide. Attorney Docket No.: PAT054248-WO-PCT
Retrovirus: Viral immunogens include, but are not limited to, those derived from an Oncovirus, a Lentivirus (e.g. HIV-1 or HIV-2) or a Spumavirus.
Reovirus: Viral immunogens include, but are not limited to, those derived from an Orthoreovirus, a Rotavirus, an Orbivirus, or a Coltivirus.
Parvovirus: Viral immunogens include, but are not limited to, those derived from Parvovirus B 19.
Herpesvirus: Viral immunogens include, but are not limited to, those derived from a human herpesvirus, such as, by way of example only, Herpes Simplex Viruses (HSV) (e.g. HSV types 1 and 2), Varicella-zoster virus (VZV), Epstein-Barr virus (EBV), Cytomegalovirus (CMV), Human Herpesvirus 6 (HHV6), Human Herpesvirus 7 (HHV7), and Human
Herpesvirus 8 (HHV8).
Papovaviruses: Viral immunogens include, but are not limited to, those derived from Papillomaviruses and Polyomaviruses. The (human) papillomavirus may be of serotype 1 , 2, 4, 5, 6, 8, 11 , 13, 16, 18, 31, 33, 35, 39, 41, 42, 47, 51, 57, 58, 63 or 65 e.g. from one or more of serotypes 6, 1 1 , 16 and/or 18.
Adenovirus: Viral immunogens include those derived from adenovirus serotype 36 (Ad-36).
In some embodiments, 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).
Fungal immunogens may be derived from Dermatophytes, 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. discoides, var. ochraceum, Trichophyton violaceum, and/or Trichophyton faviforme; or from Aspergillus fumigatus, Aspergillus flavus, Aspergillus niger, Aspergillus nidulans, Aspergillus terreus, Aspergillus sydowi, Aspergillus flavatus, Aspergillus glaucus, Blastoschizomyces capitatus, Candida albicans, Candida enolase, Candida tropicalis, Candida glabrata, Candida krusei, Candida parapsilosis, Candida stellatoidea, Candida kusei, Candida parakwsei, Candida lusitaniae, Candida pseudotropicalis, Candida guilliermondi, Cladosporium carrionii, Coccidioides immitis, Blastomyces dermatidis, Attorney Docket No. : PAT054248-WO-PCT
Cryptococcus neoformans, Geotrichum clavatum, Histoplasma capsulatum, Klebsiella pneumoniae, Microsporidia, Encephalitozoon spp., Septata intestinalis and Enterocytozoon bieneusi; the less common are Brachiola spp, Microsporidium spp., Nosema spp., Pleistophora spp., Trachipleistophora spp., Vittaforma spp Paracoccidioides brasiliensis, Pneumocystis carinii, Pythiumn insidiosum, Pityrosporum ovale, Sacharomyces cerevisae, Saccharomyces boulardii, Saccharomyces pombe, Scedosporium apiosperum, Sporothrix schenckii, Trichosporon beigelii, Toxoplasma gondii, Penicillium marneffei, Malassezia spp., Fonsecaea spp., Wangiella spp., Sporothrix spp., Basidiobolus spp., Conidiobolus spp., Rhizopus spp, Mucor spp, Absidia spp, Mortierella spp, Cunninghamella spp, Saksenaea spp., Alternaria spp, Curvularia spp, Helminthosporium spp, Fusarium spp, Aspergillus spp, Penicillium spp, Monolinia spp, Rhizoctonia spp, Paecilomyces spp, Pithomyces spp, and Cladosporium spp.
In some embodiments the immunogen elicits an immune response against a parasite from the Plasmodium genus, such as P '.falciparum, P.vivax, P.malariae or P. ovale. Thus the invention may be used for immunising against malaria. In some embodiments 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.
In some embodiments 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). 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. Other important inhalation allergens are those from house dust mites of the genus Dermatophagoides and Euroglyphus, storage mite e.g. Lepidoglyphys, Glycyphagus and Tyrophagus, those from cockroaches, midges and fleas e.g. Blatella, Periplaneta, Chironomus and Ctenocepphalides, and those from mammals such as cat, dog and horse, venom allergens including such originating from stinging or biting insects such as those from the taxonomic order of Hymenoptera including bees (Apidae), wasps (Vespidea), and ants (Formicoidae).
In some embodiments 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 Attorney Docket No.: PAT054248-WO-PCT 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), MUM 1 (associated with, e.g., melanoma), caspase-8 (associated with, e.g. , head and neck cancer), CIA 0205 (associated with, e.g., bladder cancer), HLA-A2-R1701 , beta catenin (associated with, e.g., melanoma), TCR (associated with, e.g., T-cell non-Hodgkins lymphoma), BCR-abl (associated with, e.g., chronic myelogenous leukemia), triosephosphate isomerase, KIA 0205, CDC-27, and LDLR- FUT; (c) over-expressed antigens, for example, Galectin 4 (associated with, e.g., colorectal cancer), Galectin 9 (associated with, e.g., Hodgkin's disease), proteinase 3 (associated with, e.g. , chronic myelogenous leukemia), WT 1 (associated with, e.g., various leukemias), carbonic anhydrase (associated with, e.g., renal cancer), aldolase A (associated with, e.g., lung cancer), PRAME (associated with, e.g., melanoma), HER-2/neu (associated with, e.g., breast, colon, lung and ovarian cancer), mammaglobin, alpha-fetoprotein (associated with, e.g., hepatoma), KSA (associated with, e.g., colorectal cancer), gastrin (associated with, e.g., pancreatic and gastric cancer), telomerase catalytic protein, MUC-1 (associated with, e.g., breast and ovarian cancer), G-250 (associated with, e.g., renal cell carcinoma), p53 (associated with, e.g. , breast, colon cancer), and carcinoembryonic antigen (associated with, e.g., breast cancer, lung cancer, and cancers of the gastrointestinal tract such as colorectal cancer); (d) shared antigens, for example, melanoma-melanocyte differentiation antigens such as MART-l/Melan A, gplOO, MC 1R, melanocyte-stimulating hormone receptor, tyrosinase, tyrosinase related protein- 1/TRPl and tyrosinase related protein-2/TRP2 (associated with, e.g., melanoma); (e) prostate associated antigens such as PAP, PSA, PSMA, PSH-P1, PSM-P1, PSM-P2, associated with e.g., prostate cancer; (f) immunoglobulin idiotypes (associated with myeloma and B cell lymphomas, for example). In certain embodiments, tumor immunogens include, but are not limited to, pi 5, 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, pl 85erbB2, pl 80erbB-3, c-met, mn-23Hl, TAG-72-4, CA 19-9, CA 72-4, CAM 17.1 , NuMa, K-ras, pl6, TAGE, PSCA, CT7, 43-9F, 5T4, 791 Tgp72, beta-HCG, BCA225, BTAA, CA 125, CA 15-3 (CA 27.29XBCAA), CA 195, CA 242, CA-50, CAM43, CD68\KP1, CO-029, FGF-5, Ga733 (EpCAM), HTgp- 175, M344, MA- 50, MG7-Ag, MOV 18, NB/70K, NY-CO-1, RCAS l , SDCCAG16, TA-90 (Mac-2 binding protein/cyclophilin C-associated protein), TAAL6, TAG72, TLP, TPS, and the like.
Pharmaceutical compositions
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. For example, the composition may include a TLR2 agonist (e.g. Pam3CSK4), a Attorney Docket No.: PAT054248-WO-PCT
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). Any such agonist ideally has a molecular weight of <2000Da. In some embodiments such agonist(s) are also encapsulated with the RNA inside liposomes, but in other embodiments they are unencapsulated. Pharmaceutical 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 5-20mM range.
Pharmaceutical 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. 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. Thus a composition may include one or more of EDTA, EGTA, BAPTA, pentetic acid, etc.. Such chelators are typically present at between 10-500μΜ e.g. 0.1 mM. A citrate salt, such as sodium citrate, can also act as a chelator, while advantageously also providing buffering activity.
Pharmaceutical 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.
Pharmaceutical compositions of the invention may include one or more preservatives, such as thiomersal or 2-phenoxyethanol. Mercury-free compositions are preferred, and preservative-free vaccines can be prepared.
Pharmaceutical compositions of the invention are preferably sterile.
Pharmaceutical 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.
Pharmaceutical compositions of the invention are preferably gluten free.
Pharmaceutical compositions of the invention may be prepared in unit dose form. In some embodiments a unit dose may have a volume of between 0.1 -1.0ml e.g. about 0.5ml.
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. Attorney Docket No.: PAT054248-WO-PCT
Compositions comprise an immunologically effective amount of liposomes, as well as any other components, as needed. By '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. The liposome and RNA content of compositions of the invention will generally be expressed in terms of the amount of RNA per dose. A preferred dose has <100μg RNA (e.g. from 10-100μg, such as about 10μ , 25μg, 50μg, 75 g or 100μg), but expression can be seen at much lower levels e.g. <^g/dose, <100ng/dose, <10ng/dose, <lng/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. This device can be used to administer the composition to a vertebrate subject.
Liposomes of the invention do not contain ribosomes. Methods of treatment and medical uses
In contrast to the particles disclosed in reference 10, 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.
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). Where the vaccine is for prophylactic use, the human is preferably a child Attorney Docket No.: PAT054248-WO-PCT
(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. Thus 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. at an age of 6 weeks, 10 weeks and 14 weeks, as often used in the World Health Organisation's Expanded Program on Immunisation ("EPI"). In an alternative embodiment, 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. In a further embodiment, 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. Attorney Docket No.: PAT054248-WO-PCT
Formula (X)
Compounds of formula (X) contains a hydrophilic polymer head group linked to a lipid moiety. They can be described as "stealth lipids"
Figure imgf000021_0001
wherein:
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;
wherein Z is polymerized by n subunits;
n is a number-averaged degree of polymerization between 10 and 200 units of Z, wherein n is optimized for different polymer types;
L! is an optionally substituted C O alkylene or CMoheteroalkylene linker including zero, one or two of an ether (e.g., -0-), ester (e.g., -C(O)O-), succinate (e.g., -0(0)C-CH2-CH2-C(0)0-)), carbamate (e.g., -OC(0)-N '-), carbonate (e.g., -OC(O)O-), urea (e.g., -NRC(0)NR'-), amine (e.g., -NR'- amide (e.g., -C(0)N '-), imine (e.g., -C(NR')-), thioether (e.g., -S-), xanthate (e.g., -OC(S)S-), and phosphodiester (e.g., -OP(0)20-),
wherein R' is independently selected from -H, -NH-, -NH2, -CK -S-, a phosphate or an optionally substituted Cj.io alkylene;
Xi and X2 are independently selected from a carbon or a heteroatom selected from -NH-, -0-, -S- or a phosphate;
A! and A2 are independently selected from a C6-30 alkyl, C6-30 alkenyl, and C6.30 alkynyl, wherein Ai and A2 may be the same or different, or Ai and A2 together with the carbon atom to which they are attached form an optionally substituted steroid.
In one embodiment, the compou (Χ')
Figure imgf000021_0002
wherein
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; Attorney Docket No.: PAT054248-WO-PCT
Li is an optionally substituted Q.io heteroalkylene linker containing one or two of an ether, ester, succinate, carbamate, carbonate, urea, amine, amide, imine, thioether, xanthate, and phosphodiester;
Xi and X2 are oxygen;
A! and A2 are independently selected from a C6-3o alkyl, C6-3o alkenyl, and C6.30 alkynyl, wherein Ai and A2 may be the same or different, or wherein Aj and A2 together with the carbon atom to which they are attached form an optionally substituted steroid.
The lipids of formulae (X) and (Χ'), 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. In one embodiment, 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.
Suitable PEGylated lipids for use with the invention include polyethyleneglycol-diacyl glycerol 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 ( l-[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), l ,2-dimyristoyl-sn-glycero-3-phosphoethanolamine-N-[methoxy (polyethylene glycol)-2000] (catalog #880150P from Avanti Polar Lipids). Other useful PEGylated lipids are SOOl, S002, S003, S004, S005, S006, S007, S008, S009, SOlO, SOU , and CS-020SA (NOF); SOlO and S01 1 are disclosed in ref. 32 under the labels IVa and IVc, respectively. In ref. 32, a different synthesis from that reported herein is used to prepare IVa and rVc. Chemical terms and definitions
Halo
The term "halogen" (or "halo") includes fluorine, chlorine, bromine and iodine.
Alkyl. allcylene. alkenyl. alkynyl. cvcloall yl etc.
The terms "alkyl", "alkylene", "alkenyl" and "alkynyl" are used herein to refer to both straight and branched chain acyclic forms. Cyclic analogues thereof are referred to as cycloalkyl, etc. Attorney Docket No.: PAT054248-WO-PCT
The term "alkyl" includes monovalent, straight or branched, saturated, acyclic hydrocarbyl groups. In one embodiment alkyl is Ci.ioalkyl, in another embodiment
Figure imgf000023_0001
in another embodiment Ci-4alkyl, such as methyl, ethyl, n-propyl, i-propyl or t-butyl groups.
The term "cycloalkyl" includes monovalent, saturated, cyclic hydrocarbyl groups. In one embodiment cycloalkyl is C3_i0cycloalkyl, in another embodiment C3.6Cycloalkyl such as cyclopentyl and cyclohexyl.
The term "alkoxy" means alkyl-O-.
The term "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 C2_ioalkenyl, in another embodiment C2-6alkenyl, in another embodiment C2-4alkenyl.
The term "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. In one embodiment cycloalkenyl is C3.i0cycloalkenyl, in another embodiment C5_iocycloalkenyl, e.g. cyclohexenyl or benzocyclohexyl.
The term "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. In one embodiment, alkynyl is C2.i0alkynyl, in another embodiment C2-6alkynyl, in another embodiment C^alkyn l.
The term "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. In one embodiment cycloalkynyl is C3.]0cycloalkenyl, in another embodiment C5-iocycloalkynyl.
The term "alkylene" includes divalent, straight or branched, saturated, acyclic hydrocarbyl groups. In one embodiment alkylene is Ci-ioalkylene, in another embodiment Ci^alkylene, in another embodiment Ci.4alkylene, such as methylene, ethylene, n-propylene, i-propylene or t-butylene groups.
The term "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. In one embodiment alkenylene is C2.i0alkenylene, in another embodiment C2_6alkenylene, in another embodiment C .4alkenylene.
The term "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 Attorney Docket No.: PAT054248-WO-PCT double bonds. In one embodiment alkynylene is C2-ioalkynylene, in another embodiment C2.6alkynylene, in another embodiment C2-4alkynylene.
Heteroalkyl etc.
The term "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(0)q, N, P(0)r or Si (and preferably O, S(d)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(0)q, N, P(0)r or Si.
The term "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(0)q or N, provided at least one of the cycloalkyl carbon atoms remains. Examples of 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. The heterocycloalkyl 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.
The term "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(0)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(0)q or N.
The term "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(0)q or N, provided at least one of the cycloalkenyl carbon atoms remains. Examples of 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.
The term "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(0)q or N, provided at least one of the alkynyl carbon atoms remains. The Attorney Docket No.: PAT054248-WO-PCT 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(0)q or N.
The term "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(0)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.
The term "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(0)q or N, provided at least one of the alkylene carbon atoms remains.
The term "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(0)q or N, provided at least one of the alkenylene carbon atoms remains.
The term "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(0)q or N, provided at least one of the alkynylene carbon atoms remains.
Aryl
The term "aryl" includes monovalent, aromatic, cyclic hydrocarbyl groups, such as phenyl or naphthyl (e.g. 1-naphthyl or 2-naphthyl). In general, the aryl groups may be monocyclic or polycyclic fused ring aromatic groups. Preferred aryl are Ce-Cnaryl.
Other examples of 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.
The term "arylalkyl" means alkyl substituted with an aryl group, e.g. benzyl.
The term "arylene" includes divalent aromatic, cyclic hydrocarbyl groups, such as phenylene. In general, the arylene groups may be monocyclic or polycyclic fused ring aromatic groups. Preferred arylene are Ce-Cnarylene. Other examples of 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. Attorney Docket No.: PAT054248-WO-PCT
Heteroaryl
The term "heteroaryl" includes monovalent, heteroaromatic, cyclic hydrocarbyl groups additionally containing one or more heteroatoms independently selected from O, S, N and RN, where RN is defined below (and in one embodiment is H or alkyl (e.g. Ci_6alkyl)).
In general, the heteroaryl groups may be monocyclic or polycyclic (e.g. bicyclic) fused ring heteroaromatic groups. In one embodiment, 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 NRN. In one embodiment, 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 NRN.
In one embodiment, 5-membered monocyclic heteroaryl groups contain 1 ring member which is an - RN- 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.
Examples of 6-membered monocyclic heteroaryl groups are pyridinyl, pyridazinyl, pyrimidinyl and pyrazinyl.
In one embodiment, 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 NRN.
In one embodiment, 9-membered bicyclic heteroaryl groups contain 1 ring member which is an -NRN- 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).
Examples of 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[l ,2-a]pyridinyl, imidazo[l ,5-a]pyridinyl, pyrazolo[ l ,2-a]pyridinyl, pyrrolo[ l ,2-b]pyridazinyl and imidazo[l ,2-c]pyrimidinyl. Attorney Docket No.: PAT054248-WO-PCT
In one embodiment, 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).
Examples of 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-b]pyrazinyl, pyrido[3,4-b]pyrazinyl, pyrimido[5,4-d]pyrimidinyl, pyrazino[2,3-b]pyrazinyl and pyrimido[4,5-d]pyrimidinyl.
The term "heteroarylalkyl" means alkyl substituted with a heteroaryl group.
The term "heteroarylene" includes divalent heteroaromatic, cyclic hydrocarbyl groups additionally containing one or more heteroatoms independently selected from O, S, N and NRN, where RN is defined below (and in one embodiment is H or alkyl (e.g. Ci-ealkyl)). In general, the heteroarylene groups may be monocyclic or polycyclic (e.g. bicyclic) fused ring heteroaromatic groups. In one embodiment, 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 NRN. In one embodiment, a heteroarylene 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. The term "heteroarylene" includes divalent derivatives of each of the heteroaryl groups discussed above.
The terms "aryl", "aromatic", "heteroaryl" and "heteroaromatic" also include groups that are partially reduced. Thus, for example, "heteroaryl" includes fused species in which one of the rings has been reduced to a saturated ring (e.g. l ,2,3,4-tetrahydro-l ,8-naphthyridin-2-yl).
General
Unless indicated explicitly otherwise, where combinations of groups are referred to herein as one moiety, e.g. arylalkyl, the last mentioned group contains the atom by which the moiety is attached to the rest of the molecule.
Where reference is made to a carbon atom of an alkyl group or other group being replaced by O, S(0)q, N or P(0)r, what is intended is that:
H c „ p
E C E E N E E P E
E is replaced by E or E (wherein E cannot be H);
-CH= is replaced by
Figure imgf000027_0001
=C-H is replaced by≡N or≡P(0)r; or Attorney Docket No.: PAT054248-WO-PCT
-CH2- is replaced by -0-, -S(0)q- - RN— or -P(0)rRN-, where RN is H or optionally substituted C].6alkyl, Ci_6heteroalkyl, C3.6cycloalkyl, C3^heterocycloalkyl, C2-6alkenyl, C2-6heteroalkenyl, C3.6cycloalkenyl, C3_6heterocycloalkenyl, phenyl, or heteroaryl containing 5 or 6 ring members. RN is preferably H, C|.6alkyl or C3.6cycloalkyl.
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.
Where reference is made to a carbon atom being replaced by Si, what is intended is that the carbon atom is swapped for a silicon atom but that the bonds otherwise remain the same. Thus, for example, -CH2- is replaced by -SiH2-; -CH= is replaced by -SiH=; and≡C-H is replaced by≡Si-H.
By way of clarification, in relation to the above mentioned heteroatom containing groups (such as heteroalkyl etc.), where a numerical of carbon atoms is given, for instance C3-6heteroalkyl, what is intended is a group based on C3_6alkyl in which one or more of the 3-6 chain carbon atoms is replaced by O, S(0)q or N. Accordingly, a C3_6heteroalkyl group would, for example, contain less than 3-6 chain carbon atoms. As another example, a pyridyl group would be classed as a Ce heteroaryl group even though it contains 5 carbon atoms.
Substitution
Groups of the compounds of the invention (e.g. alkyl, cycloalkyl, alkoxy, alkenyl, cycloalkenyl, alkynyl, alkylene, alkenylene, heteroalkyl, heterocycloalkyl, heteroalkenyl, heterocycloalkenyl, heteroalkynyl, heteroalkylene, heteroalkenylene aryl, arylalkyl, arylheteroalkyl, heteroaryl, heteroarylalkyl or heteroarylheteroalkyl groups etc.) may be substituted or unsubstituted, in one embodiment unsubstituted. Typically, substitution involves the notional replacement of a hydrogen atom with a substituent group, or two hydrogen atoms in the case of substitution by =0.
Where substituted, 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.
In one embodiment, the substituent(s) is/are independently Sub1 or Sub2 (in one embodiment Sub2) wherein:
Sub1 is independently halogen, trihalomethyl, trihaloethyl, -N02, -CN, -N+(Rs)20", -C02H, -C02Rs, -SO3H, -SORs, -S02Rs, -S03Rs, -OC(=0)ORs, -C(=0)H, -C(=0)Rs, -OC(=0)Rs, =0, - RS 2> -C(=0)NH2, -C(=0) Rs 2, -N(Rs)C(=0)ORs, -N(Rs)C(=0)NRs 2, -OC(=0)NRs 2, -N(Rs)C(=0)Rs, -C(=S)NRS 2, -NRSC(=S)RS, -S02 Rs 2, -NRsS02Rs, -N(RS)C(=S)NRS 2, -N(Rs)S02NRs 2, -Rs or -ZSRS, wherein;
Zs is independently O, S or NRS; Attorney Docket No. : PAT054248-WO-PCT
Rs is independently H or
Figure imgf000029_0001
-(Alka)rC3.6cycloalkyl, -(Alka)rC3.6heterocycloalkyl, C2.6alkenyl, C2-6heteroalkenyl, -(Alka)rC3-6cycloalkenyl, -(Alka)rC3^heterocycloalkenyl, C2-6a]kynyl, C2.6heteroalkynyl, -(Alka)rC6-i4aryl, -(Alka)f-C6-i 4aryl or -(Alka)rheteroaryl (where heteroaryl contains 5-13 ring members), where f is O or l ;
Alka is Ci_6alkylene or C].6heteroalkylene; and
Rs is optionally substituted itself (in one embodiment unsubstituted) by 1 to 3 substituents Sub2;
Sub2 is independently halogen, trihalomethyl, trihaloethyl, -N02, -CN, -N+(Ci.6alkyl)20', -C02H, -C02C,.6alkyl, -S03H, -SOC^alkyl, -S02C,.6alkyl, -S03C,.6alkyl, -OC(=0)OC,-6alkyl,
-C(=0)H, -C(=0)C,.6alkyl, -OC(=0)C,^alkyl, =0, -N(Ci.6alkyl)2, -C(=0)NH2, -C(=0)N(C,.6alkyl)2, -N(C,.6alkyl)C(=0)0(C,^alkyl), -N(C,.6alkyl)C(=0)N(C1.6alkyl)2, -OC(=0)N(C,.6alkyl)2,
-N(C,.6alkyl)C(=0)C1^alkyl, -C(=S)N(C1.6alkyl)2, -N(C,.6alkyl)C(=S)C1.6alkyl, -S02N(d.6alkyl)2, -N(C,.6alkyl)S02C,.6alkyl, -N(C,.6alkyl)C(=S)N(C1.6alkyl)2, -N(C,.6alkyl)S02N(Ci.6alkyl)2, -Ci^alkyl, -Ci-eheteroalkyl, -C3.6Cycloalkyl, -C3.6heterocycloalkyl, -C2.6alkenyl, -C2_6heteroalkenyl, -C3.6cycloalkenyl, -C3_6heterocycloalkenyl, -C^alkynyl, -C2.6heteroalkynyl, -Cs-naryl, -C5_i3hetero. aryl, -Zl-Ci.6alkyl, -Z'-C3.6cycloalkyl, -Zl-C2.6alkenyl, -Z'-Cs^cycloalkenyl, or -Z'-C2-6alkynyl; and
Zl is independently O, S, NH or N(d.6alkyl).
While Rs in Sub1 can be optionally substituted by 1 to 3 substituents Sub2, Sub2 is unsubstituted. However, in one embodiment, Rs is unsubstituted.
In one embodiment, Rs is H or C|.6alkyl, optionally substituted by 1 to 3 substituents Sub2.
In one embodiment, Sub2 is independently halogen, trihalomethyl, trihaloethyl, -N02, -CN, -N+(C,.6alkyl)20\ -C02H, -S03H, -SOC,.6alkyl, -S02C,.6alkyl, -C(=0)H, -C(=0)C,.6alkyl, =0, -N(C,.6alkyl)2, -C(=0)NH2, -Ci-6alkyl, -C3.6cycloalkyl, -C3.6heterocycloalkyl, -Z'-C,.6alkyl or - Z'-C3.6cycloalkyl.
In one embodiment, where the substituted group is acyclic (e.g. alkyl, heteroalkyl, alkenyl etc.), Sub1 is not -Rs and Sub2 is not -Ci-ealkyl, -Ci^heteroalkyl, -C2.6alkenyl, -C2.6heteroalkenyl, -C2.6alkynyl or -C2.6heteroalkynyl.
Where a group other than Sub2 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 Sub2. In one embodiment that chain is not substituted. Thus, the Attorney Docket No.: PAT054248-WO-PCT terms optionally substituted "cycloalkyl", "cycloalkenyl", "cycloalkynyl", "heterocycloalkyl", "heterocycloalkenyl", "heterocycloalkynyl", "aryl" and "heteroaryl" include fused species. E.g. "optionally substituted cycloalkyl" includes a species in which two cycloalkyl rings are fused, and "optionally substituted heteroaryl" includes a species in which a heterocycloalkyl ring is fused to the aromatic ring (e.g. 5,6,7,8-tetrahydro-l ,8-naphthyridin-2-yl).
Where a group other than Sub2 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 Sub2. In one embodiment that chain is not substituted. Thus, the terms optionally substituted "cycloalkyl", "cycloalkenyl", "cycloalkynyl", "heterocycloalkyl", "heterocycloalkenyl", "heterocycloalkynyl", "aryl" and "heteroaryl" include spiro species.
By way of clarification, when a group has a heteroatom, a substituent may be bonded to the heteroatom. Thus, for example, "optionally substituted heteroalkyl" includes -CH2-N(Sub ')-(¾-, -CH(Sub')-NH-CH2- and -CH(Sub')-N(Sub')-CH2- etc.
Modifier terms
When a list is preceded by a modifier, it is intended that the modifier is to be understood as applying to each of the items in the list. For example, the phrase "optionally substituted C3_2o-heterocycloalkyl, C3.2o-heterocycloalkenyl, C3.2o-heterocycloalkynyl or C5.20-heteroaryl group" means that each of the four items in the list, namely the C3-2o-heterocycloalkyl group, the C3_2o-heterocycloalkeny] group, the C3_2o-heterocycloalkynyl group and the C6-2o-heteroaryl group, may be optionally substituted.
When 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. For example, if a group is described as a "C3.20-heterocycloalkynyl" (the first modifier) group and then later the same group is described as a "Cs-ie" (the subsequent modifier) group, what is meant is a C5.16 heterocycloalkynyl group.
Steroids
As used herein, the term "steroid" refers to any group comprising the following structure (which structure is referred to herein as the "steroid skeleton").
Figure imgf000030_0001
Attorney Docket No.: PAT054248-WO-PCT
Purely for the purposes of illustration, the steroid skeleton has been drawn above as fully saturated. The term steroid, however, is also intended to cover instances where there is unsaturation in the steroid skeleton. For example, the term steroid covers a group which comprises the fully unsaturated (mancude) basic skeleton, 15H-cyclopenta[a]phenanthrene:
Figure imgf000031_0001
The term steroid also covers a group which comprises a partially unsaturated steroid skeleton.
The term 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"). In one embodiment, however, such seco derivatives are not encompassed by the term "steroid". In another embodiment, such nor derivatives are not encompassed by the term "steroid". In another embodiment, such homo derivatives are not encompassed by the term "steroid". Thus in one embodiment, such seco, nor and homo derivatives are not encompassed by the term "steroid".
The term steroid also covers instances where one or more of the carbon atoms in the structure labelled steroid skeleton is replaced by a heteroatom. In one such embodiment, 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(0)q, N, P(0)r or Si (and preferably O, S(0)q or N). In one embodiment, however, 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.
Figure imgf000031_0002
The term 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. Attorney Docket No.: PAT054248-WO-PCT
Unsaturation
In accordance with standard use, 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. General
The practice of the present invention will employ, unless otherwise indicated, conventional methods of chemistry, biochemistry, molecular biology, immunology and pharmacology, within the skill of the art. Such techniques are explained fully in the literature. See, e.g., references 33-39, etc.
The term "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.
The term "about" in relation to a numerical value x is optional and means, for example, +10%.
The word "substantially" does not exclude "completely" e.g. a composition which is "substantially free" from Y may be completely free from Y. Where necessary, the word "substantially" may be omitted from the definition of the invention.
References to charge, to cations, to anions, to zwitterions, etc., are taken at pH 7.
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.
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) 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 Attorney Docket No.: PAT054248-WO-PCT
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) is the approved HGNC name for the gene encoding this enzyme, and its unique HGNC ID is HGNC:9437. The RefSeq sequence for the human PKR gene is GI:208431825.
BRIEF DESCRIPTION OF DRAWINGS
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: lkDa (triangles); 2kDa (circles); 3kDa (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. 5 shows protein expression at days 1, 3 and 6 after delivery of RNA as a virion-packaged replicon (squares), as naked RNA (diamonds), or in liposomes (+ = O. ^g, x = ^g).
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), ^g naked RNA, and lμg liposome-encapsulated RNA.
FIG. 8 shows anti-F IgG titers in animals receiving VRP, \ μg naked RNA, and O. lg or lμg liposome-encapsulated RNA.
FIG. 9 shows neutralising antibody titers in animals receiving VRP or either O. lg or ^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). Attorney Docket No.: PAT054248-WO-PCT
FIG. 1 1 shows F-specific IgG titers (2 weeks after second dose) after delivery of a replicon as naked RNA (0.01 -lpg), liposome-encapsulated RNA (0.01-l(^g), or packaged as a virion (VRP, 106 infectious units or IU).
FIG. 12 shows F-specific IgG titers (circles) and PRNT titers (squares) after delivery of a replicon as naked RNA (l g), liposome-encapsulated RNA (0.1 or lpg), or packaged as a virion (VRP, 106 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 logio 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 (l-3kDa).
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).
MODES FOR CARRYING OUT THE INVENTION RNA replicons
Various 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. The replicon is about lOkb 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. Attorney Docket No.: PAT054248-WO-PCT
Following linearization of the plasmid DNA downstream of the HDV ribozyme with a suitable restriction endonuclease, 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). The replicon RNA was precipitated with LiCl 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 OD260nm- Integrity of the in vitro transcripts was confirmed by denaturing agarose gel electrophoresis.
Liposomal encapsulation
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 (l ,2-dilinoleyloxy-N,N-dimethyl-3-aminopropane) was synthesized using the procedure of reference 2. DSPC (l ,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 (3P-[N-(N',N'-dimethylaminoethane)- carbamoyl]cholesterol hydrochloride) were from Avanti Polar Lipids.
Briefly, lipids were dissolved in ethanol (2ml), a RNA replicon was dissolved in buffer (2ml, lOOmM sodium citrate, pH 6) and these were mixed with 2ml of buffer followed by 1 hour of equilibration. The mixture was diluted with 6ml buffer then filtered. The resulting product contained liposomes, with -95% encapsulation efficiency. 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 141nm (by intensity) or 78nm (by number).
In one particular encapsulation method, fresh lipid stock solutions in ethanol were prepared. 37 mg of DlinDMA, 1 1.8 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. A 2 mL working solution Attorney Docket No.: PAT054248-WO-PCT of RNA was also prepared from a stock solution of - Ιμξ/μί. 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. Syringes containing RNA and the lipids were connected to a T mixer (PEEK™ 500 μπι ID junction) using FEP tubing(fluorinated ethylene-propylene; all FEP tubing used had a 2mm internal diameter and a 3mm 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 7mL/min flow rate using a syringe pump and the final mixture collected in a 20 mL glass vial (while stirring). Next, liposomes were concentrated to 2 mL and dialyzed against 10- 15 volumes of IX PBS using a Tangential Flow Filtration (TFF) system before recovering the final product. 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 cm2 surface area were used. For in vitro and in vivo experiments, formulations were diluted to the required RNA concentration with IX PBS.
The percentage of encapsulated 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 lOx or lOOx in I X TE buffer (from kit) before addition of the dye. Separately, liposomes were diluted lOx or lOOx in IX 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 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.
To obtain smaller liposomes 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, 1 1.8 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 Attorney Docket No.: PAT054248-WO-PCT of the stock was added to 1.773 mL ethanol to make a working lipid stock solution of 2 mL. A 4 mL working solution of RNA was also prepared from a stock solution of ~ ^g^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. Syringes containing 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 x 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 μιτι x 105 μιτι) 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. Then the mixture was loaded in a 5 cc syringe which was fitted to a piece of PTFE tubing 0.03 inches ID x l/16inches 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 3mL/min flow rate using a syringe pump and the final mixture collected in a 20 mL glass vial (while stirring). Next, liposomes were concentrated to 2 mL and dialyzed against 10-15 volumes of IX PBS using the TFF system before recovering the final product. Hollow fiber filtration membranes with a 100 kDa pore size cutoff and 20cm2 surface area were used. For in vitro and in vivo experiments, formulations were diluted to the required RNA concentration with IX PBS. Whereas liposomes prepared using the syringe/tube method with 75μg RNA had a Z-average diameter (Zav) of 148nm and a polydispersity index (pdl) of 0.122, the chip mixing gave liposomes with a Zav of 97nm and a pdl of 0.086. The proportion of encapsulated RNA decreased slightly from 90% to 87%.
Encapsulation in liposomes was shown to protect RNA from RNase digestion. Experiments used 3.8mAU 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 y/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 12k RPM. The aqueous phase (containing the RNA) was removed and used to analyze the RNA. Prior to loading (400 ng RNA per well) all the samples were incubated with formaldehyde loading dye, denatured for 10 minutes at 65°C and cooled to room temperature. Ambion Millennium markers were used to approximate the molecular weight of the RNA construct. The gel was run at 90 V. The gel was stained using 0.1 % SYBR gold according to the manufacturer's guidelines in water by rocking at room temperature for 1 hour. 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 Attorney Docket No. : PAT054248-WO-PCT liposomes are subjected to phenol extraction, undigested RNA is seen (lane 6). 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.
To assess in vivo expression of the RNA a reporter enzyme (SEAP; secreted alkaline phosphatase) was encoded in the replicon, rather than an immunogen. Expression levels were measured in sera diluted 1 :4 in IX 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, 50μ1 per t leg with O. ^g or ^g RNA dose. The same vector was also administered without the liposomes (in RNase free IX PBS) at l g. Virion-packaged replicons were also tested. Virion-packaged replicons used herein (referred to as "VRPs") 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.
As shown in FIG. 5, encapsulation increased SEAP levels by about ½ log at the lpg dose, and at day 6 expression from a 0.1 g encapsulated dose matched levels seen with ^g unencapsulated dose. By day 3 expression levels exceeded those achieved with VRPs (squares). Thus expressed increased when the RNA was formulated in the liposomes relative to the naked RNA control, even at a lOx 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.
To assess whether the effect seen in the liposome groups was due merely to the liposome components, or was linked to the encapsulation, the replicon was administered in encapsulated form (with two different purification protocols, O. ^g RNA), or mixed with the liposomes after their formation (a non-encapsulated "lipoplex", O. ^g RNA), or as naked RNA Ong). FIG. 10 shows that the lipoplex gave the lowest levels of expression, showing that shows encapsulation is essential for potent expression.
Further SEAP experiments showed a clear dose response in vivo, with expression seen after delivery of as little as lng RNA (FIG. 6). Further experiments comparing expression from encapsulated and naked replicons indicated that O.O^g encapsulated RNA was equivalent to \\xg of naked RNA. At a 0^g dose of RNA the encapsulated material gave a 12-fold higher expression at day 6; at a 0.1 μg dose levels were 24-fold higher at day 6.
Rather than looking at average levels in the group, individual animals were also studied. Whereas several animals were non-responders to naked replicons, encapsulation eliminated non-responders. Attorney Docket No.: PAT054248-WO-PCT
Further experiments replaced DlinDMA with DOTAP. Although the DOTAP liposomes gave better expression than naked replicon, they were inferior to the DlinDMA liposomes (2- to 3-fold difference at day 1 ).
To assess in vivo immunogenicity a replicon was constructed to express full-length F protein from respiratory syncytial virus (RSV). This was delivered naked (lpg), encapsulated in liposomes (0.1 or lpg), or packaged in virions (106 IU; "VRP") at days 0 and 21. 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 pg, the encapsulated RNA at lpg, 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.
Further experiments compared F-specific IgG titers in mice receiving VRP, 0.1 pg liposome- encapsulated RNA, or lpg liposome-encapsulated RNA. Titer ratios (VRP: liposome) at various times after the second dose were as follows:
Figure imgf000039_0001
Thus the liposome-encapsulated RNA induces essentially the same magnitude of immune response as seen with virion delivery.
Further experiments showed superior F-specific IgG responses with a 10pg dose, equivalent responses for lpg and O.lpg doses, and a lower response with a O.Olpg dose. 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 (106 IU). The response seen with lpg liposome-encapsulated RNA was statistically insignificant (ANOVA) when compared to VRP, but the higher response seen with 10pg liposome- encapsulated RNA was statistically significant (p<0.05) when compared to both of these groups. A further study confirmed that the O. lpg of liposome-encapsulated RNA gave much higher anti-F IgG responses ( 15 days post-second dose) than O.lpg of delivered DNA, and even was more immunogenic than 20pg plasmid DNA encoding the F antigen, delivered by electroporation (Elgen™ DNA Delivery System, Inovio).
Liposome manufacturing methods
In general, eight different methods have been used for preparing liposomes according to the invention. These are referred to in the text as methods (A) to (H) and they differ mainly in relation to filtration and TFF steps. Details are as follows: Attorney Docket No.: PAT054248-WO-PCT
(A) Fresh lipid stock solutions in ethanol were prepared. 37 mg of DlinDMA, 1 1.8 mg of DSPC, 27.8 mg of Cholesterol and 8.07 mg of PEG DMG 2000 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, 755 of the stock was added to 1.245 mL ethanol to make a working lipid stock solution of 2 mL. This amount of lipids was used to form liposomes with 250 μg RNA. A 2 mL working solution of RNA was also prepared from a stock solution of - lμg/μL· in 100 mM citrate buffer (pH 6). Three 20 mL glass vials (with stir bars) were rinsed with RNase Away solution (Molecular BioProducts, San Diego, CA) 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. Syringes containing RNA and the lipids were connected to a T mixer (PEEK™ 500 μπι ID junction, Idex Health Science, Oak Harbor, WA) using FEP tubing (fluorinated ethylene-propylene; al FEP tubing has a 2mm internal diameter x 3mm outer diameter, supplied by 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 FEP 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. 4 ml of the mixture was loaded into a 5 cc syringe, which was connected to a piece of FEP tubing and in another 5 cc syringe connected to an equal length of FEP tubing, an equal amount of 100 mM citrate buffer (pH 6) was loaded. The two syringes were driven at 7mL/min flow rate using the syringe, pump and the final mixture collected in a 20 mL glass vial (while stirring). Next, the mixture collected from the second mixing step (liposomes) were passed through a Mustang Q membrane (an anion-exchange support that binds and. removes anionic molecules, obtained from Pall Corporation, Ann Arbor, MI, USA). Before passing the liposomes, 4 mL of 1 M NaOH, 4 mL of 1 M NaCl and 10 mL of 100 mM citrate buffer (pH 6) were successively passed through the Mustang membrane. Liposomes were warmed for 10 min at 37°C before passing through the membrane. Next, liposomes were concentrated to 2 mL and dialyzed against 10-15 volumes of IX PBS' using TFF before recovering the final product. The TFF system and hollow fiber filtration membranes were purchased from Spectrum Labs and were used according to the manufacturer's guidelines. Polysulfone hollow fiber filtration membranes (part number P/N: X1AB- 100-20P) with a 100 kD pore size cutoff and 8 cm2 surface area were used. For in vitro and in vivo experiments, formulations were diluted to the required RNA concentration with IX PBS.
(B) As method (A) except that, after rocking, 226.7 μL· of the stock was added to 1.773 mL ethanol to make a working lipid stock solution of 2 mL, thus modifying the lipid:RNA ratio. Attorney Docket No.: PAT054248-WO-PCT
(C) As method (B) except that the Mustang filtration was omitted, so liposomes went from the 20 mL glass vial into the TFF dialysis.
(D) As method (C) except that the TFF used polyethersulfone (PES) hollow fiber membranes (part number P-Cl-lOOE-100-OlN) with a 100 kD pore size cutoff and 20 cm2 surface area.
(E) As method (D) except that a Mustang membrane was used, as in method (A).
(F) As method (A) except that the Mustang filtration was omitted, so liposomes went from the 20 mL glass vial into the TFF dialysis.
(G) As method (D) except that a 4 mL working solution of RNA was prepared from a stock solution of - lpg/pL in 100 mM citrate buffer (pH 6). Then four 20 mL glass vials were prepared in the same way. Two of them were used for the RNA working solution (2 mL in each vial) and the others for collecting the lipid and RNA mixes, as in (C). Rather than use T mixer, syringes containing 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 internal diameter x 1/16 inch outer diameter) using a 4-way edge connector (Syrris). 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 pm x 105 pm) 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 h. Then the mixture was loaded in a 5 cc syringe, which was fitted to another piece of the PTFE tubing; 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 3mL/min flow rate using a syringe pump and the final mixture collected in a 20 mL glass vial (while stirring). Next, liposomes were concentrated to 2 mL and dialyzed against 10-15 volumes of IX PBS using TFF, as in (D).
(H) As method (A) except that the 2mL working lipid stock solution was made by mixing
120.9 pL of the lipid stock with 1.879 mL ethanol. Also, after mixing in the T mixer the liposomes from the 20mL vial were loaded into Pierce Slide-A-Lyzer Dialysis Cassette (Thermo Scientific, extra strength, 0.5-3 mL capacity) and dialyzed against 400-500 mL of IX PBS overnight at 4°C in an autoclaved plastic container before recovering the final product. RSV immunogenicity
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 pL per leg) on days 0 and 21 with the replicon (lpg) 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% Attorney Docket No.: PAT054248-WO-PCT
PEG-DMG. The RV13 liposomes had 40% DOTAP, 10% DOPE, 48% cholesterol and 2% PEG- DMG. In all cases the PEG was PEG-2000 (i.e. 2kDa PEG). For comparison, naked plasmid DNA (20 μ ) expressing the same RSV-F antigen was delivered either using electroporation or with RV01(10) liposomes (O.^g DNA). Four mice were used as a naive control group.
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:
Figure imgf000042_0001
Serum was collected for antibody analysis on days 14, 36 and 49. Spleens were harvested from mice at day 49 for T cell analysis.
F-specific serum IgG titers (GMT) were as follows:
Figure imgf000042_0002
The proportion of 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: Attorney Docket No.: PAT054248-WO-PCT
Figure imgf000043_0001
Thus the liposome formulations significantly enhanced immunogenicity relative to the naked RNA controls, as determined by increased F-specific IgG titers and T cell frequencies. Plasmid DNA formulated with liposomes, or delivered naked using electroporation, was significantly less immunogenic than liposome-formulated self-replicating RNA.
Further RV01 liposomes were prepared by method (H), again using 2kDa PEG conjugated to DMG, and either encapsulating 150μg RNA (vA375 replicon encoding surface fusion glycoprotein of RSV) or encapsulating only buffer. Thus these liposomes had 40% DlinDMA, 10% DSPC, 48% Choi, and 2% PEG-DMG. Sizes and encapsulation were as follows:
Figure imgf000043_0002
The liposomes were administered to BALB/c mice (10 per group) by bilateral intramuscular injection (50μ1 per leg) on days 0 & 21. Doses were 0.01, 0.03, 0.1, 0.3 or ^g. F-specific serum IgG and PRNT60 titers (GMT) were as follows, 2 weeks after the first or second injection:
Figure imgf000043_0003
Attorney Docket No.: PAT054248-WO-PCT
Cytomegalovirus immunogenicity
RV01 liposomes with DLinDMA as the cationic lipid and 2kDa 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.
BALB/c mice, 10 per group, were given bilateral intramuscular vaccinations (50 μΙ_ per leg) on days 0, 21 and 42 with VRPs expressing gH gL (l xlO6 IU), VRPs expressing gHsol/gL (l xlO6 IU) and PBS as the controls. Two test groups received ^g of the vA160 or vA322 replicon formulated in liposomes (40% DlinDMA, 10% DSPC, 48% Choi, 2% PEG-DMG; made using method (D) but with 150μg RNA batch size).
The vA160 liposomes had a Zav diameter of 168.8nm, a pdl of 0.144, and 87.4% encapsulation. The vA322 liposomes had a Zav diameter of 162nm, a pdl of 0.131 , and 90% encapsulation.
/
The replicons were able to express two proteins from a single vector.
Sera were collected for immunological analysis on day 63 (3wp3). 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:
Figure imgf000044_0001
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.
Repeat experiments confirmed that the 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.
Expression kinetics
A self-replicating RNA replicon ("vA31 1") that expresses a luciferase reporter gene (luc) was used for studying the kinetics of protein expression after injection. BALB/c mice, 5 animals per group, received bilateral intramuscular vaccinations (50 μΐ^ per leg) on day 0 with:
Group 1 DNA expressing luciferase, delivered using electroporation (10 μg)
Group 2 self-replicating RNA (1 μg) formulated in liposomes (40% DlinDMA, 10% DSPC, 48% cholesterol, 2% PEG-2000 conjugated to DMG
Group 3 self-replicating RNA (^g) formulated with a cationic nanoemulsion (CNE17)
Group 4 self-replicating RNA (^g) formulated with a different cationic nanoemulsion Attorney Docket No.: PAT054248-WO-PCT
Group 5 VRP (lxlO6 IU) expressing luciferase
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 intraperitoneally 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.
In visual terms, luciferase-expressing cells were seen to remain primarily at the site of RNA injection, and animals imaged after removal of quads showed no signal.
In quantitative terms, luciferase expression was measured as average radiance over a period of 70 days (p/s/cm2/sr), and results were as follows for the 5 groups:
Figure imgf000045_0001
The self-replicating 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. 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.
Delivery volume
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. Attorney Docket No.: PAT054248-WO-PCT
A typical mouse delivery volume for intramuscular injection is 50 μΐ into the hind leg, which is a relatively high volume for a mouse leg muscle. In contrast, a human intramuscular dose of ~0.5ml 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 :
Group 1 received naked replicon, 0^g in 50 μL· per leg
Group 2 received naked replicon, 0.2 g in 5 μΐ- per leg
Group 3 received emulsion-formulated replicon (0.2 μg, 50 μί per leg)
Group 4 received emulsion-formulated replicon (0.2 μg, 5 μΐ^ per leg)
Group 5 received liposome-formulated replicon (0.2 μg, 50 μί per leg)
Group 6 received liposome-formulated replicon (0.2 μg, 5 μL· per leg)
The liposomes for groups 5 & 6 were 40% DlinDMA, 10% DSPC, 48% cholesterol, and 2% PEG- 2000 conjugated to DMG.
Serum was collected for antibody analysis on days 14 and 35. F-specific serum IgG GMTs were:
Figure imgf000046_0001
Thus immunogenicity of the formulated replicon did not vary according to the delivered volume, thus indicating that these RNA vaccines do not rely on hydrodynamic delivery for their efficacy.
Cotton rats
A study was performed in cotton rats (Sigmodon hispidis) instead of mice. At a \ xg dose liposome encapsulation increased F-specific IgG titers by 8.3-fold compared to naked RNA and increased PRNT titers by 9.5-fold. The magnitude of the antibody response was equivalent to that induced by 5xl06 IU VRP. Both naked and liposome-encapsulated RNA were able to protect the cotton rats from RSV challenge (lxlO5 plaque forming units), reducing lung viral load by at least 3.5 logs. Encapsulation increased the reduction by about 2-fold.
Further work in cotton rats used four different replicons: 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. Cotton rats, 4 to 8 animals per group, were given intramuscular vaccinations (100 μL· in one leg) on days 0 and 21 with the four different replicons at two doses (1.0 and O. lpg) formulated in liposomes made using 2kDa PEG-conjugated DMG by method (D), but with a 150 g RNA batch size. Control Attorney Docket No.: PAT054248-WO-PCT groups received a RSV-F subunit protein vaccine (5 μg) adjuvanted with alum (8 animals/group), VRPs expressing full-length RSV-F (lxlO6 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:
Figure imgf000047_0001
All four replicons evaluated in this study (vA317, vA318, vA142, vA140) 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 lpg replicon were 2- to 3-fold higher than after the second vaccination with 0.1 μg 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.
Further work in cotton rats again used the vA317, vA318 and vA142 replicons. Cotton rats, 2-8 animals per group, were given intramuscular vaccinations (100 μL· in one leg) on days 0 and 21 with the replicons (0.1 or ^g) encapsulated in RV01 liposomes (with PEG-2000) made by method (D) but with a 150μg RNA batch size. Control groups received the RSV-F subunit protein vaccine (5 μg) adjuvanted with alum or VRPs expressing full-length RSV-F (lxlO6 IU, 8 animals/group). All these animals received a third vaccination (day 56) with RSV-F subunit protein vaccine (5 μg) adjuvanted with alum. In addition there was a naive control (4 animals/group). In addition, an extra group was given bilateral intramuscular vaccinations (50 L per leg) on days 0 and 56 with ^g vA317 RNA in liposomes but did not receive a third vaccination with the subunit protein yaccine.
Serum was collected for antibody analysis on days 0, 21, 35, 56, 70, plus days 14, 28 & 42 for the extra group. F-specific serum IgG titers (GMT) were as follows: Attorney Docket No.: PAT054248-WO-PCT
Figure imgf000048_0001
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):
Figure imgf000048_0002
Serum titers and neutralising titers for the extra group were as follows:
Figure imgf000048_0003
Thus 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 μg replicon.
The third vaccination (protein at day 56) 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. In most cases 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.
This study also evaluated the kinetics of the antibody response to 1.0 μg vA317. F-specific serum IgG and RSV neutralization titers induced by a single vaccination reached their peak around day 21 Attorney Docket No.: PAT054248-WO-PCT 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. Further experiments involved a viral challenge. The 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. Cotton rats, 7 per group, were given intramuscular vaccinations (100 μΐ- per leg) on days 0 and 21 with vA368 in liposomes prepared by method (H), 175 μg RNA batch size, or with VRPs having the same replicon. The liposomes included 2kDa PEG, conjugated to DMG. A control group received 5pg alum-adjuvanted protein, and a naive control group was also included.
All groups received an intranasal challenge (i.n.) with l xlO6 PFU RSV four weeks after the final immunization. Serum was collected for antibody analysis on days 0, 21 , 35. Viral lung titers were measured 5 days post challenge. Results were as follows:
Figure imgf000049_0001
Thus the RNA vaccine reduced the lung viral load by over three logs, from approximately 106 PFU/g in unvaccinated control cotton rats to less than 103 PFU/g in vaccinated cotton rats.
Large mammal study
A large-animal study was performed in cattle. Calves (4-6 weeks old, -60-80 kg, 5 per group) were immunised with 66μg of replicon vA317 encoding full-length RSV F protein at days 0, 21 , 86 and 146. The replicons were formulated inside liposomes made by method (E) but with a 1.5 mg RNA batch size; they had 40% DlinDMA, 10% DSPC, 48% cholesterol, and 2% PEG-2000 conjugated to DMG. PBS alone was used as a negative control, and a licensed vaccine was used as a positive control ("Triangle 4" from Fort Dodge, containing killed virus). All calves received 15μg F protein adjuvanted with the MF59 emulsion on day 146.
The 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. Attorney Docket No.: PAT054248-WO-PCT
Calves received 2ml of each experimental vaccine, administered intramuscularly as 2 1 ml on each side of the neck. In contrast, the 'Triangle 4" vaccine was given as a single 2ml dose in the neck.
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:
Figure imgf000050_0001
RSV serum neutralizing antibody titers were as follows:
Figure imgf000050_0002
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. When assayed with complement, neutralizing antibodies were detected in all vaccinated cows. In this assay, all vaccinated calves had good neutralizing antibody titers after the second RNA vaccination Furthermore, the RNA vaccine elicited F-specific serum IgG titers that were detected in a few calves after the second vaccination and in all calves after the third.
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.
Proof of concept for 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 66μg of RNA. Attorney Docket No.: PAT054248-WO-PCT
Effect of PEG length
As mentioned above, liposomes were prepared using DMG to which five different PEGs were conjugated. The average molecular weight of the PEG was 500Da, 750Da, lkDa, 2kDa or 3kDa.
Liposomes formed using the shortest PEGs (500Da and 750Da) were unstable or aggregated during TFF purification. PEG-750 gave liposomes with a significantly higher Zaverage diameter (669nm) 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 197nm (0.1 19 pdl) for the lkDa PEG, 142nm (0.137 pdl) for the 2kDa PEG, and 147nm (0.075 pdl) for the 3kDa 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.
Different lipids and PEG lengths
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.
Physical characteristics of the liposomes, made by method (H), were:
Figure imgf000051_0001
Attorney Docket No.: PAT054248-WO-PCT
BALB/c mice, 8 per group, were given bilateral intramuscular vaccinations (50 μ]_ per leg) on days 0 and '21 with the replicon,- either naked (^g) or encapsulated in these liposomes (O.^g). Serum was collected for antibody analysis on days 14, and 35.
F-specific serum IgG titers (GMT) were as follows, 2 weeks after the two injections (2wpl):
Figure imgf000052_0001
The results show a trend, indicating that higher molecular weight PEG head groups are more immunogenic. As the length of DMG-conjugated PEG increases from lOOODa to 3000Da the 2wp2 F-specific IgG titers increase from 7412 to 15659 to 22378.
Changing the linker region from ester to ether did not impact the titers substantially. Also, at the same molecular weight of the head group (2000) there was a trend that increasing the length of the lipid tails lowers the titers (H with C14 dialkyl vs. I with C18 dialkyl). Replacing a PEG di-alkyl lipid tail with cholesterol had little impact on immunogenicity (A with DMG vs. G with cholesterol).
Similar experiments were performed with different lipids in which the 2kDa of PEG is split into 2x lkDa groups (FIG. 18, with total MW in the boxed region being 2000). The vA317 replicon was again used, with BALB/c mice, 8 per group, given bilateral intramuscular vaccinations (50μΙ_ per leg) on days 0 & 21 with ^g naked RNA or O.^g liposome-encapsulated RNA. The liposomes all had 40% cationic lipid (DlinDMA), 10% DSPC and 48% cholesterol, but the remaining 2% was varied, with different PEGylated lipids (but all with 2kDa PEG). They were made by method (H).
Physical characteristics of the liposomes were:
Figure imgf000052_0002
Attorney Docket No.: PAT054248-WO-PCT
Figure imgf000053_0001
Further liposomes were made with RV05. The liposomes had 40% cationic lipid (RV05) and 2% PEGylated DMG (2kDa 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:
Figure imgf000053_0002
aGC = a-galactosylceramide BALB/c mice, 8 per group, were given bilateral intramuscular vaccinations (50 μΙ per leg) on days 0 and 21 with the replicon, either naked (^g) or encapsulated (O.^g). Serum was collected for antibody analysis on days 14, and 35. F-specific serum IgG titers (GMT) were as follows, 2 weeks after the two injections (2wp 1 ) :
Figure imgf000053_0003
Splitting the PEG head groups thus lowered in vivo titers. Including a double bond (1 degree of instauration per alkyl tail) in the PEG lipid tails increased IgG titers, 6 fold at day 14 and 7 fold at day 35. For a cationic lipid with an asymmetrical lipid tails (alkyl + cholesterol), changing the neutral lipid from DSPC (saturated C18 lipid tail) to 18:2 or 18:3 PC (with 2 and 3 unsaturated double bonds per tail) increased total IgG titers. Comparable results were observed with replacement of DSPC with DPyPE. Attorney Docket No.: PAT054248-WO-PCT
It will be understood, that the invention has been described by way of example only and modifications may be made whilst remaining within the scope and spirit of the invention.
Attorney Docket No.: PAT054248-WO-PCT
Table 1: useful phospholipids
DDPC l ,2-Didecanoyl-sn-Glycero-3-phosphatidylcholine
DEPA l ,2-Dierucoyl-sn-Glycero-3-Phosphate
DEPC l ,2-Erucoyl-sn-Glycero-3-phosphatidylcholine
DEPE 1 ,2-Dierucoyl-sn-Glycero-3-phosphatidylethanolamine
DEPG l ,2-Dierucoyl-sn-Glycero-3[Phosphatidyl-rac-(l -glycerol...)
DLOPC l ,2-Linoleoyl-sn-Glycero-3-phosphatidylcholine
DLPA l ,2-Dilauroyl-sn-Glycero-3-Phosphate
DLPC l ,2-Dilauroyl-sn-Glycero-3-phosphatidylcholine
DLPE l ,2-Dilauroyl-sn-Glycero-3-phosphatidylethanolamine
DLPG l ,2-Dilauroyl-sn-Glycero-3[Phosphatidyl-rac-(l-glycerol...)
DLPS l ,2-Dilauroyl-sn-Glycero-3-phosphatidylserine
DMG 1 ,2-Dimyristoyl-sn-glycero-3-phosphoethanolamine
DMPA 1 ,2-Dimyristoyl-sn-Glycero-3-Phosphate
DMPC l ,2-Dimyristoyl-sn-Glycero-3-phosphatidylcholine
DMPE l ,2-Dimyristoyl-sn-Glycero-3-phosphatidylethanolamine
DMPG l ,2-Myristoyl-sn-Glycero-3[Phosphatidyl-rac-(l-glycerol...)
DMPS l ,2-Dimyristoyl-sn-Glycero-3-phosphatidylserine
DOPA l ,2-Dioleoyl-sn-Glycero-3-Phosphate
DOPC l ,2-Dioleoyl-sn-Glycero-3-phosphatidylcholine
DOPE l ,2-Dioleoyl-sn-Glycero-3-phosphatidylethanolamine
DOPG l ,2-Dioleoyl-sn-Glycero-3[Phosphatidyl-rac-(l-glycerol...)
DOPS l ,2-Dioleoyl-sn-Glycero-3-phosphatidylserine
DPPA l ,2-Dipalmitoyl-sn-Glycero-3-Phosphate
DPPC l ,2-Dipalmitoyl-sn-Glycero-3-phosphatidylcholine
DPPE l ,2-Dipalrnitoyl-sn-Glycero-3-phosphatidylethanolamine
DPPG l ,2-Dipalmitoyl-sn-Glycero-3[Phosphatidyl-rac-(l-glycerol...)
DPPS l ,2-Dipalmitoyl-sn-Glycero-3-phosphatidylserine
DPyPE l ,2-diphytanoyl-sn-glycero-3-phosphoethanolamine
DSPA l ,2-Distearoyl-sn-Glycero-3-Phosphate
DSPC l ,2-Distearoyl-sn-Glycero-3-phosphatidylcholine Attorney Docket No.: PAT054248-WO-PCT DSPE 1 )2-Οϊθ5ΐε3 >Ί-5η-Ο1>'0εΓθ-3-ρ1ιθ5ρΗαϋ<1>Ίβ11ΐ3ηοΐ3Γηϊηε
DSPG l,2-Distearoyl-sn-Glycero-3[Phosphatidyl-rac-(l -glycerol...)
DSPS 1 ,2-Distearoyl-sn-Glycero-3-phosphatidylserine
EPC Egg-PC
HEPC Hydrogenated Egg PC
HSPC High purity Hydrogenated Soy PC
HSPC Hydrogenated Soy PC
LYSOPC MYRISTIC l-Myristoyl-sn-Glycero-3-phosphatidylcholine
LYSOPC PALMITIC l-Palmitoyl-sn-Glycero-3-phosphatidylcholine
LYSOPC STEARIC l-Stearoyl-sn-Glycero-3-phosphatidylcholine
Milk Sphingomyelin MPPC l-Myristoyl,2-palmitoyl-sn-Glycero 3-phosphatidylcholine
MSPC l-Myristoyl,2-stearoyl-sn-Glycero-3-phosphatidylcholine
PMPC l-Palmitoyl,2-myristoyl-sn-Glycero-3-phosphatidylcholine
POPC l-Palmitoyl,2-oleoyl-sn-Glycero-3-phosphatidylcholine
POPE l-Palmitoyl-2-oleoyl-sn-Glycero-3-phosphatidylethanolamine
POPG 1 ,2-Dioleoyl-sn-Glycero-3 [Phosphatidyl -rac-( 1 -glycerol)...]
PSPC l-Palmitoyl,2-stearoyl-sn-Glycero-3-phosphatidylcholine
SMPC l-Stearoyl,2-myristoyl-sn-Glycero- 3-phosphatidylcholine
SOPC l-StearoyI,2-oleoyl-sn-Glycero-3-phosphatidylcholine
SPPC l-Stearoyl,2-palmitoyl-sn-Glycero-3-phosphatidylcholine
Attorney Docket No.: PAT054248-WO-PCT
REFERENCES
[I] Johanning et al. (1995) Nucleic Acids Res 23: 1495-1501.
[2] Heyes et al. (2005) J Controlled Release 107:276-87.
[3] WO2005/121348.
[4] Liposomes: Methods and Protocols, Volume 1: Pharmaceutical Nanocarriers: Methods and
Protocols, (ed. Weissig). Humana Press, 2009. ISBN 160327359X.
[5] Liposome Technology, volumes I, II & III. (ed. Gregoriadis). Informa Healthcare, 2006.
[6] Functional Polymer Colloids and Microparticles volume 4 (Microspheres, microcapsules & liposomes), (eds. Arshady & Guyot). Citus Books, 2002.
[7] Jeffs et al. (2005) Pharmaceutical Research 22 (3):362-372.
[8] WO2005/113782.
[9] WO201 1/005799.
[10] El Ouahabi et al. (1996) FEBS Letts 380: 108-12.
[I I] Giuliani et al. (2006) Proc Natl Acad Sci U SA 103(29): 10834-9.
[12] WO2009/016515. ·
[13] WO02/34771.
[14] WO2005/032582.
[15] WO2010/119343.
[16] WO2006/110413.
[17] WO2005/1 1 1066.
[18] WO2005/002619.
[19] WO2006/138004.
[20] WO2009/ 109860.
[21] WO02/02606.
[22] WO03/018054.
[23] WO2006/091517.
[24] WO2008/020330.
[25] WO2006/089264.
[26] WO2009/104092.
[27] WO2009/031043.
[28] WO2007/049155.
[29] Gennaro (2000) Remington: The Science and Practice of Pharmacy. 20th edition, ISBN: 0683306472.
[30] Romberg et al. (2008) Pharmaceutical Research 25:55-71.
[31] Hoekstra et al., Biochimica et Biophysica Acta 1660 (2004) 41-52
[32] WO2009/086558
[33] Methods In Enzymology (S. Colowick and N. Kaplan, eds., Academic Press, Inc.)
[34] Handbook of Experimental Immunology, Vols. I-IV (D.M. Weir and C.C. Blackwell, eds, 1986,
Blackwell Scientific Publications)
[35] Sambrook et al. (2001) Molecular Cloning: A Laboratory Manual, 3rd edition (Cold Spring Harbor Laboratory Press).
[36] Handbook of Surface and Colloidal Chemistry (Birdi, K.S. ed., CRC Press, 1997)
[37] Ausubel et al. (eds) (2002) Short protocols in molecular biology, 5th edition (Current
Protocols).
[38] Molecular Biology Techniques: An Intensive Laboratory Course, (Ream et al., eds., 1998, Academic Press) Attorney Docket No.: PAT054248-WO-PCT
[39] PCR (Introduction to Biolechniques Series), 2nd ed. (Newton & Graham eds., 1997, Springer Verlag)
[40] Yoneyama & Fujita (2007) Cytokine & Growth Factor Reviews 18:545-51.
[41] Maurer ei a/. (2001) Biophysical Journal, 80: 2310-2326.
[42] Perri et al. (2003) J Virol 77: 10394-10403.
[43] Boxus et al. (2007) J Virol 81 :6879-89.
[44] Taylor et al. (2005) Vaccine 23: 1242-50.

Claims

1. 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 lkDa and 3kDa.
2. The liposome of claim 1, comprising PEG-DMG and/or a lipid of formula (X).
3. The liposome of any preceding claim, wherein the liposome has a diameter in the range of 80-160nm.
4. The liposome of any preceding claim, wherein the liposome comprises a lipid with a cationic head group.
5. The liposome of any preceding claim, wherein the liposome comprises a lipid with a zwitterionic head group.
6. The liposome of any preceding claim, wherein the RNA is a self-replicating RNA.
7. The liposome of claim 6, wherein the self-replicating RNA molecule encodes (i) a RNA- dependent RNA polymerase which can transcribe RNA from the self-replicating RNA molecule and (ii) an immunogen.
8. The liposome of claim 7, wherein the RNA molecule has two open reading frames, the first of which encodes an alphavirus replicase and the second of which encodes the immunogen.
9. The liposome of any preceding claim, wherein the RNA molecule is 9000-12000 nucleotides long.
10. The liposome of any preceding claim, wherein the immunogen can elicit an immune response in vivo against a bacterium, a virus, a fungus or a parasite.
11. A pharmaceutical composition comprising a liposome of any preceding claim.
12. A method for raising a protective immune response in a vertebrate, comprising the step of administering to the vertebrate an effective amount of the liposome of claims 1-10, or the pharmaceutical composition of claim 11.
PCT/US2011/050095 2010-08-31 2011-08-31 Pegylated liposomes for delivery of immunogen-encoding rna WO2012031043A1 (en)

Priority Applications (27)

Application Number Priority Date Filing Date Title
ES11758014T ES2913791T3 (en) 2010-08-31 2011-08-31 Pegylated liposomes for delivery of RNA encoding immunogen
EP21207859.6A EP3970742B1 (en) 2010-08-31 2011-08-31 Pegylated liposomes for delivery of immunogen-encoding rna
EP22192507.6A EP4119155A1 (en) 2010-08-31 2011-08-31 Pegylated liposomes for delivery of immunogen-encoding rna
EP23179901.6A EP4226941A1 (en) 2010-08-31 2011-08-31 Pegylated liposomes for delivery of immunogen-encoding rna
EP21205422.5A EP3981427B9 (en) 2010-08-31 2011-08-31 Pegylated liposomes for delivery of immunogen-encoding rna
HRP20220695TT HRP20220695T1 (en) 2010-08-31 2011-08-31 Pegylated liposomes for delivery of immunogen-encoding rna
EP22188690.6A EP4101462A1 (en) 2010-08-31 2011-08-31 Pegylated liposomes for delivery of immunogen-encoding rna
CN2011800518918A CN103179984A (en) 2010-08-31 2011-08-31 Pegylated liposomes for delivery of immunogen-encoding RNA
EP22174931.0A EP4066856B1 (en) 2010-08-31 2011-08-31 Pegylated liposomes for delivery of immunogen-encoding rna
SI201132053T SI2611461T1 (en) 2010-08-31 2011-08-31 Pegylated liposomes for delivery of immunogen-encoding rna
DK11758014.2T DK2611461T3 (en) 2010-08-31 2011-08-31 Pegylated liposomes to release RNA encoding immunogen
RU2013114362/10A RU2013114362A (en) 2010-08-31 2011-08-31 PEGILATED LIPOSOMES FOR THE DELIVERY OF ENCODING IMMUNOGEN RNA
LTEPPCT/US2011/050095T LT2611461T (en) 2010-08-31 2011-08-31 Pegylated liposomes for delivery of immunogen-encoding rna
RS20220501A RS63315B1 (en) 2010-08-31 2011-08-31 Pegylated liposomes for delivery of immunogen-encoding rna
JP2013526210A JP5911870B2 (en) 2010-08-31 2011-08-31 PEGylated liposomes for delivery of RNA encoding immunogens
CA2809851A CA2809851A1 (en) 2010-08-31 2011-08-31 Pegylated liposomes for delivery of immunogen-encoding rna
MX2013002336A MX2013002336A (en) 2010-08-31 2011-08-31 Pegylated liposomes for delivery of immunogen-encoding rna.
EP11758014.2A EP2611461B1 (en) 2010-08-31 2011-08-31 Pegylated liposomes for delivery of immunogen-encoding rna
EP23179897.6A EP4226940A1 (en) 2010-08-31 2011-08-31 Pegylated liposomes for delivery of immunogen-encoding rna
AU2011295935A AU2011295935B2 (en) 2010-08-31 2011-08-31 Pegylated liposomes for delivery of immunogen-encoding RNA
US13/819,077 US20130202684A1 (en) 2010-08-31 2011-08-31 Pegylated liposomes for delivery of immunogen encoding rna
BR112013004866A BR112013004866A2 (en) 2010-08-31 2011-08-31 pegylated liposomes for encoded immunogen RNA delivery
EP22174932.8A EP4066857B1 (en) 2010-08-31 2011-08-31 Pegylated liposomes for delivery of immunogen-encoding rna
EP22174779.3A EP4066855B1 (en) 2010-08-31 2011-08-31 Pegylated liposomes for delivery of immunogen-encoding rna
PL11758014T PL2611461T3 (en) 2010-08-31 2011-08-31 Pegylated liposomes for delivery of immunogen-encoding rna
EP22192506.8A EP4147718A1 (en) 2010-08-31 2011-08-31 Pegylated liposomes for delivery of immunogen-encoding rna
US16/837,115 US11759422B2 (en) 2010-08-31 2020-04-01 Pegylated liposomes for delivery of immunogen-encoding RNA

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US37882610P 2010-08-31 2010-08-31
US61/378,826 2010-08-31

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US13/819,077 A-371-Of-International US20130202684A1 (en) 2010-08-31 2011-08-31 Pegylated liposomes for delivery of immunogen encoding rna
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
WO2012031043A1 true WO2012031043A1 (en) 2012-03-08

Family

ID=44653554

Family Applications (1)

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

Country Status (20)

Country Link
US (2) US20130202684A1 (en)
EP (11) EP4066855B1 (en)
JP (1) JP5911870B2 (en)
CN (1) CN103179984A (en)
AU (1) AU2011295935B2 (en)
BR (1) BR112013004866A2 (en)
CA (1) CA2809851A1 (en)
DK (6) DK4066857T3 (en)
ES (6) ES2913791T3 (en)
FI (2) FI4066855T3 (en)
HR (6) HRP20221533T1 (en)
HU (6) HUE058361T2 (en)
LT (6) LT4066855T (en)
MX (1) MX2013002336A (en)
PL (6) PL4066857T3 (en)
PT (6) PT3981427T (en)
RS (6) RS63983B1 (en)
RU (1) RU2013114362A (en)
SI (6) SI3981427T1 (en)
WO (1) WO2012031043A1 (en)

Cited By (170)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013006825A1 (en) * 2011-07-06 2013-01-10 Novartis Ag Liposomes having useful n:p ratio for delivery of rna molecules
US20130195968A1 (en) * 2010-07-06 2013-08-01 Novartis Ag Virion-like delivery particles for self-replicating rna molecules
WO2013151666A2 (en) 2012-04-02 2013-10-10 modeRNA Therapeutics Modified polynucleotides for the production of biologics and proteins associated with human disease
WO2013151736A2 (en) 2012-04-02 2013-10-10 modeRNA Therapeutics In vivo production of proteins
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
WO2014108515A1 (en) 2013-01-10 2014-07-17 Novartis Ag Influenza virus immunogenic compositions and uses thereof
WO2014113089A2 (en) 2013-01-17 2014-07-24 Moderna Therapeutics, Inc. Signal-sensor polynucleotides for the alteration of cellular phenotypes
US8822663B2 (en) 2010-08-06 2014-09-02 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
WO2014136086A1 (en) 2013-03-08 2014-09-12 Novartis Ag Lipids and lipid compositions for the delivery of active agents
WO2014140211A1 (en) 2013-03-15 2014-09-18 Novartis Ag Rna purification methods
WO2014152211A1 (en) 2013-03-14 2014-09-25 Moderna Therapeutics, Inc. Formulation and delivery of modified nucleoside, nucleotide, and nucleic acid compositions
WO2014159813A1 (en) 2013-03-13 2014-10-02 Moderna Therapeutics, Inc. Long-lived polynucleotide molecules
WO2015034925A1 (en) 2013-09-03 2015-03-12 Moderna Therapeutics, Inc. Circular polynucleotides
WO2015034928A1 (en) 2013-09-03 2015-03-12 Moderna Therapeutics, Inc. Chimeric polynucleotides
US8980864B2 (en) 2013-03-15 2015-03-17 Moderna Therapeutics, Inc. Compositions and methods of altering cholesterol levels
WO2015051214A1 (en) 2013-10-03 2015-04-09 Moderna Therapeutics, Inc. Polynucleotides encoding low density lipoprotein receptor
US9107886B2 (en) 2012-04-02 2015-08-18 Moderna Therapeutics, Inc. Modified polynucleotides encoding basic helix-loop-helix family member E41
WO2015177312A1 (en) 2014-05-22 2015-11-26 Glaxosmithkline Biologicals Sa Rsvf trimerization domains
WO2016014846A1 (en) 2014-07-23 2016-01-28 Moderna Therapeutics, Inc. Modified polynucleotides for the production of intrabodies
US9254265B2 (en) 2010-08-31 2016-02-09 Novartis Ag Small liposomes for delivery of immunogen encoding RNA
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
EP3061826A1 (en) 2015-02-27 2016-08-31 Novartis AG Flavivirus replicons
US9464124B2 (en) 2011-09-12 2016-10-11 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
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
WO2017162461A1 (en) 2016-03-21 2017-09-28 Biontech Rna Pharmaceuticals Gmbh Trans-replicating rna
WO2017200957A1 (en) * 2016-05-16 2017-11-23 Infectious Disease Research Institute Pegylated liposomes and methods of use
WO2017208191A1 (en) 2016-06-02 2017-12-07 Glaxosmithkline Biologicals Sa Zika viral antigen constructs
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
WO2018060288A1 (en) 2016-09-29 2018-04-05 Glaxosmithkline Biologicals S.A. Compositions and methods of treatment of persistent hpv infection
WO2018065931A1 (en) 2016-10-05 2018-04-12 Glaxosmithkline Biologicals Sa Vaccine
WO2018091540A1 (en) 2016-11-17 2018-05-24 Glaxosmithkline Biologicals Sa Zika viral antigen constructs
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
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
US10106490B2 (en) 2014-06-25 2018-10-23 Acuitas Therapeutics, Inc. Lipids and lipid nanoparticle formulations for delivery of nucleic acids
EP2750707B1 (en) 2011-08-31 2018-10-24 GlaxoSmithKline Biologicals SA Pegylated liposomes for delivery of immunogen-encoding rna
WO2018213731A1 (en) 2017-05-18 2018-11-22 Modernatx, Inc. Polynucleotides encoding tethered interleukin-12 (il12) polypeptides and uses thereof
WO2018213789A1 (en) 2017-05-18 2018-11-22 Modernatx, Inc. Modified messenger rna comprising functional rna elements
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
US10221127B2 (en) 2015-06-29 2019-03-05 Acuitas Therapeutics, Inc. Lipids and lipid nanoparticle formulations for delivery of nucleic acids
WO2019053012A1 (en) 2017-09-13 2019-03-21 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 an artificial t cell receptor
EP3461497A1 (en) 2017-09-27 2019-04-03 GlaxoSmithKline Biologicals S.A. Viral antigens
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 Novel artificial nucleic acid molecules
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
WO2019104160A2 (en) 2017-11-22 2019-05-31 Modernatx, Inc. Polynucleotides encoding phenylalanine hydroxylase for the treatment of phenylketonuria
WO2019104152A1 (en) 2017-11-22 2019-05-31 Modernatx, Inc. Polynucleotides encoding ornithine transcarbamylase for the treatment of urea cycle disorders
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
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
WO2020035609A2 (en) 2018-08-17 2020-02-20 Glaxosmithkline Biologicals Sa Immunogenic compositions and uses thereof
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
WO2020144295A1 (en) 2019-01-10 2020-07-16 Biontech Rna Pharmaceuticals Gmbh Localized administration of rna molecules for therapy
US10730924B2 (en) 2016-05-18 2020-08-04 Modernatx, Inc. Polynucleotides encoding relaxin
EP3701959A1 (en) 2016-03-21 2020-09-02 BioNTech RNA Pharmaceuticals GmbH Rna replicon for versatile and efficient gene expression
WO2020180752A1 (en) 2019-03-01 2020-09-10 Flagship Pioneering Innovations Vi, Llc Polyribonucleotides and cosmetic uses thereof
WO2020180751A1 (en) 2019-03-01 2020-09-10 Flagship Pioneering Innovations Vi, Llc Compositions, methods, and kits for delivery of polyribonucleotides
EP3718565A1 (en) * 2015-10-22 2020-10-07 ModernaTX, Inc. Respiratory virus vaccines
US10815291B2 (en) 2013-09-30 2020-10-27 Modernatx, Inc. Polynucleotides encoding immune modulating polypeptides
AU2014287009B2 (en) * 2013-07-11 2020-10-29 Modernatx, Inc. Compositions comprising synthetic polynucleotides encoding CRISPR related proteins and synthetic sgRNAs and methods of use
WO2020227642A1 (en) 2019-05-08 2020-11-12 Modernatx, Inc. Compositions for skin and wounds and methods of use thereof
US10842859B2 (en) 2014-03-25 2020-11-24 Yale University Uses of parasite macrophage migration inhibitory factors
US10857105B2 (en) 2017-03-15 2020-12-08 MordernaTX, Inc. Compounds and compositions for intracellular delivery of therapeutic agents
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
WO2021013798A1 (en) 2019-07-21 2021-01-28 Glaxosmithkline Biologicals Sa Therapeutic viral vaccine
WO2021061815A1 (en) 2019-09-23 2021-04-01 Omega Therapeutics, Inc. COMPOSITIONS AND METHODS FOR MODULATING HEPATOCYTE NUCLEAR FACTOR 4-ALPHA (HNF4α) GENE EXPRESSION
WO2021061707A1 (en) 2019-09-23 2021-04-01 Omega Therapeutics, Inc. Compositions and methods for modulating apolipoprotein b (apob) gene expression
EP3819377A1 (en) 2019-11-08 2021-05-12 Justus-Liebig-Universität Gießen Circular rna and uses thereof for inhibiting rna-binding proteins
US11066355B2 (en) 2019-09-19 2021-07-20 Modernatx, Inc. Branched tail lipid compounds and compositions for intracellular delivery of therapeutic agents
WO2021183720A1 (en) 2020-03-11 2021-09-16 Omega Therapeutics, Inc. Compositions and methods for modulating forkhead box p3 (foxp3) gene expression
WO2021195214A1 (en) 2020-03-24 2021-09-30 Generation Bio Co. Non-viral dna vectors and uses thereof for expressing factor ix therapeutics
WO2021195218A1 (en) 2020-03-24 2021-09-30 Generation Bio Co. Non-viral dna vectors and uses thereof for expressing gaucher therapeutics
WO2021209970A1 (en) 2020-04-16 2021-10-21 Glaxosmithkline Biologicals Sa Sars cov-2 spike protein construct
WO2021236930A1 (en) 2020-05-20 2021-11-25 Flagship Pioneering Innovations Vi, Llc Immunogenic compositions and uses thereof
WO2021245611A1 (en) 2020-06-05 2021-12-09 Glaxosmithkline Biologicals Sa Modified betacoronavirus spike proteins
WO2021247507A1 (en) 2020-06-01 2021-12-09 Modernatx, Inc. Phenylalanine hydroxylase variants and uses thereof
WO2021245090A1 (en) 2020-06-04 2021-12-09 BioNTech SE Rna replicon for versatile and efficient gene expression
US11203569B2 (en) 2017-03-15 2021-12-21 Modernatx, Inc. Crystal forms of amino lipids
WO2022002783A1 (en) 2020-06-29 2022-01-06 Glaxosmithkline Biologicals Sa Adjuvants
WO2022023284A1 (en) 2020-07-27 2022-02-03 Anjarium Biosciences Ag Compositions of dna molecules, methods of making therefor, and methods of use thereof
EP2611461B1 (en) 2010-08-31 2022-03-09 GlaxoSmithKline Biologicals SA Pegylated liposomes for delivery of immunogen-encoding rna
WO2022051629A1 (en) 2020-09-03 2022-03-10 Flagship Pioneering Innovations Vi, Llc Immunogenic compositions and uses thereof
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
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
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
US11453639B2 (en) 2019-01-11 2022-09-27 Acuitas Therapeutics, Inc. Lipids for lipid nanoparticle delivery of active agents
WO2022204390A1 (en) 2021-03-24 2022-09-29 Modernatx, Inc. Lipid nanoparticles containing polynucleotides encoding phenylalanine hydroxylase and uses thereof
WO2022204371A1 (en) 2021-03-24 2022-09-29 Modernatx, Inc. Lipid nanoparticles containing polynucleotides encoding glucose-6-phosphatase 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
WO2022204369A1 (en) 2021-03-24 2022-09-29 Modernatx, Inc. Polynucleotides encoding methylmalonyl-coa mutase for the treatment of methylmalonic acidemia
WO2022200575A1 (en) 2021-03-26 2022-09-29 Glaxosmithkline Biologicals Sa Immunogenic compositions
US11471525B2 (en) 2020-02-04 2022-10-18 Curevac Ag Coronavirus vaccine
WO2022223556A1 (en) 2021-04-20 2022-10-27 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
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
WO2022248353A1 (en) 2021-05-24 2022-12-01 Glaxosmithkline Biologicals Sa Adjuvants
US11524023B2 (en) 2021-02-19 2022-12-13 Modernatx, Inc. Lipid nanoparticle compositions and methods of formulating the same
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
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
US11547764B2 (en) 2011-06-08 2023-01-10 Translate Bio, Inc. Lipid nanoparticle compositions and methods for MRNA delivery
WO2023283359A2 (en) 2021-07-07 2023-01-12 Omega Therapeutics, Inc. Compositions and methods for modulating secreted frizzled receptor protein 1 (sfrp1) gene expression
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
WO2023020994A1 (en) 2021-08-16 2023-02-23 Glaxosmithkline Biologicals Sa Novel methods
WO2023020992A1 (en) 2021-08-16 2023-02-23 Glaxosmithkline Biologicals Sa Novel methods
WO2023021427A1 (en) 2021-08-16 2023-02-23 Glaxosmithkline Biologicals Sa Freeze-drying of lipid nanoparticles (lnps) encapsulating rna and formulations thereof
WO2023021421A1 (en) 2021-08-16 2023-02-23 Glaxosmithkline Biologicals Sa Low-dose lyophilized rna vaccines and methods for preparing and using the same
WO2023020993A1 (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
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
WO2023066875A1 (en) 2021-10-18 2023-04-27 BioNTech SE Modified replicable rna and related compositions and their use
WO2023066874A1 (en) 2021-10-18 2023-04-27 BioNTech SE Methods for determining mutations for increasing modified replicable rna function and related compositions and their use
US11639370B2 (en) 2010-10-11 2023-05-02 Glaxosmithkline Biologicals Sa Antigen delivery platforms
WO2023081526A1 (en) 2021-11-08 2023-05-11 Orna Therapeutics, Inc. Lipid nanoparticle compositions for delivering circular polynucleotides
US11648324B2 (en) 2015-10-28 2023-05-16 Acuitas Therapeutics, Inc. Lipids and lipid nanoparticle formulations for delivery of nucleic acids
US11655475B2 (en) 2010-07-06 2023-05-23 Glaxosmithkline Biologicals Sa Immunisation of large mammals with low doses of RNA
RU2796539C2 (en) * 2016-05-16 2023-05-25 Инфекшес Дизис Рисёрч Инститьют Pegylated liposomes and their application methods
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
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
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
US11744801B2 (en) 2017-08-31 2023-09-05 Modernatx, Inc. Methods of making lipid nanoparticles
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
US11771769B2 (en) 2017-11-10 2023-10-03 Cocoon Biotech Inc. Ocular applications of silk-based products
US11786607B2 (en) 2017-06-15 2023-10-17 Modernatx, Inc. RNA formulations
US11793843B2 (en) 2019-01-10 2023-10-24 Janssen Biotech, Inc. Prostate neoantigens and their uses
WO2023213783A1 (en) 2022-05-02 2023-11-09 BioNTech SE Replicon compositions and methods of using same for the treatment of diseases
WO2023239756A1 (en) 2022-06-07 2023-12-14 Generation Bio Co. Lipid nanoparticle compositions and uses thereof
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
US11872280B2 (en) 2020-12-22 2024-01-16 CureVac SE RNA vaccine against SARS-CoV-2 variants
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
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
US11896636B2 (en) 2011-07-06 2024-02-13 Glaxosmithkline Biologicals Sa Immunogenic combination compositions and uses thereof
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
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

Families Citing this family (35)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9512456B2 (en) 2012-08-14 2016-12-06 Modernatx, Inc. Enzymes and polymerases for the synthesis of RNA
KR20160102069A (en) * 2013-12-27 2016-08-26 텔로리젠, 인크. Compositions and methods for providing active telomerase to cells in vivo
EP3096742A4 (en) 2014-01-20 2017-09-27 University of Utah Research Foundation Compositions and methods for modifying the surface of cells and methods of use
WO2016165825A1 (en) 2015-04-13 2016-10-20 Curevac Ag Method for producing rna compositions
US11364292B2 (en) 2015-07-21 2022-06-21 Modernatx, Inc. CHIKV RNA vaccines
TW201718638A (en) 2015-07-21 2017-06-01 現代治療公司 Infectious disease vaccines
WO2017031232A1 (en) 2015-08-17 2017-02-23 Modernatx, Inc. Methods for preparing particles and related compositions
AU2016336344A1 (en) 2015-10-05 2018-04-19 Modernatx, Inc. Methods for therapeutic administration of messenger ribonucleic acid drugs
WO2017070624A1 (en) 2015-10-22 2017-04-27 Modernatx, Inc. Tropical disease vaccines
AU2016342045A1 (en) 2015-10-22 2018-06-07 Modernatx, Inc. Human cytomegalovirus vaccine
JP2018531290A (en) 2015-10-22 2018-10-25 モデルナティーエックス, インコーポレイテッド Sexually transmitted disease vaccine
EP3364980A4 (en) 2015-10-22 2019-07-10 ModernaTX, Inc. Nucleic acid vaccines for varicella zoster virus (vzv)
MA46584A (en) 2016-10-21 2019-08-28 Modernatx Inc HUMAN CYTOMEGALOVIRUS VACCINE
WO2018107088A2 (en) 2016-12-08 2018-06-14 Modernatx, Inc. Respiratory virus nucleic acid vaccines
BR112019015797A2 (en) * 2017-02-01 2020-03-17 Modernatx, Inc. IMMUNOMODULATORY THERAPEUTIC MRNA COMPOSITIONS THAT CODE ACTIVATING ONCOGEN MUTATION PEPTIDES
MA47515A (en) 2017-02-16 2019-12-25 Modernatx Inc VERY POWERFUL IMMUNOGENIC COMPOSITIONS
WO2018170260A1 (en) 2017-03-15 2018-09-20 Modernatx, Inc. Respiratory syncytial virus vaccine
US11045540B2 (en) 2017-03-15 2021-06-29 Modernatx, Inc. Varicella zoster virus (VZV) vaccine
WO2018170256A1 (en) 2017-03-15 2018-09-20 Modernatx, Inc. Herpes simplex virus vaccine
EP3595676A4 (en) 2017-03-17 2021-05-05 Modernatx, Inc. Zoonotic disease rna vaccines
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
SG11201910101SA (en) 2017-05-08 2019-11-28 Gritstone Oncology Inc Alphavirus neoantigen vectors
WO2018220553A1 (en) * 2017-05-30 2018-12-06 Glaxosmithkline Biologicals Sa Methods for manufacturing a liposome encapsulated rna
US11639329B2 (en) 2017-08-16 2023-05-02 Acuitas Therapeutics, Inc. Lipids for use in lipid nanoparticle formulations
MA50253A (en) 2017-09-14 2020-07-22 Modernatx Inc ZIKA VIRUS RNA VACCINES
CA3087537A1 (en) 2018-01-04 2019-07-11 Jan-willem THEUNISSEN Anti-tissue factor antibodies, antibody-drug conjugates, and related methods
MA54676A (en) 2018-01-29 2021-11-17 Modernatx Inc RSV RNA VACCINES
US11351242B1 (en) 2019-02-12 2022-06-07 Modernatx, Inc. HMPV/hPIV3 mRNA vaccine composition
BR122024002387A2 (en) 2019-05-30 2024-03-12 Gritstone Bio, Inc. ADENOVIRUS VECTORS, PHARMACEUTICAL COMPOSITION, ISOLATED NUCLEOTIDE SEQUENCE, ISOLATED CELL, VECTOR, KIT, USES OF A VECTOR, METHOD FOR MAKING THE VECTOR, METHODS FOR PRODUCING A VIRUS AND VIRAL VECTOR
TW202200605A (en) 2020-03-09 2022-01-01 美商亞克圖羅斯醫療公司 Coronavirus vaccine compositions and methods
KR20230046313A (en) 2020-08-06 2023-04-05 그릿스톤 바이오, 인코포레이티드 Multi-epitope vaccine cassette
US11406703B2 (en) 2020-08-25 2022-08-09 Modernatx, Inc. Human cytomegalovirus vaccine
CA3206128A1 (en) * 2021-02-09 2022-08-18 J. Milton Harris Polyoxazoline-lipid conjugates and lipid nanoparticles and pharmaceutical compositions including same
CN113230394A (en) * 2021-04-30 2021-08-10 广州源博医药科技有限公司 RNA vaccine for bovine viral diarrhea and construction method thereof
WO2024002985A1 (en) 2022-06-26 2024-01-04 BioNTech SE Coronavirus vaccine

Citations (34)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5820873A (en) 1994-09-30 1998-10-13 The University Of British Columbia Polyethylene glycol modified ceramide lipids and liposome uses thereof
WO1999052503A2 (en) 1998-04-16 1999-10-21 John Wayne Cancer Institute Rna cancer vaccine and methods for its use
WO2002002606A2 (en) 2000-07-03 2002-01-10 Chiron S.P.A. Immunisation against chlamydia pneumoniae
WO2002034771A2 (en) 2000-10-27 2002-05-02 Chiron Srl Nucleic acids and proteins from streptococcus groups a & b
WO2003018054A1 (en) 2001-08-31 2003-03-06 Chiron Srl. Helicobacter pylori vaccination
EP0910576B1 (en) 1996-04-11 2004-08-11 University Of British Columbia Fusogenic liposomes
WO2005002619A2 (en) 2003-06-26 2005-01-13 Chiron Corporation Immunogenic compositions for chlamydia trachomatis
US6858224B2 (en) 1995-06-07 2005-02-22 Inex Pharmaceuticals Corporation Method of preventing aggregation of a lipid:nucleic acid complex
WO2005026372A1 (en) 2003-09-15 2005-03-24 Protiva Biotherapeutics, Inc. Polyethyleneglycol-modified lipid compounds and uses thereof
WO2005032582A2 (en) 2003-07-31 2005-04-14 Chiron Corporation Immunogenic compositions for streptococcus pyogenes
WO2005089148A2 (en) 2004-03-12 2005-09-29 The Regents Of The University Of California Delivery of genes encoding short hairpin rna using receptor-specific nanocontainers
WO2005111066A2 (en) 2004-05-14 2005-11-24 Chiron Srl Polypeptides from non-typeable haemophilus influenzae
WO2005113782A1 (en) 2004-05-18 2005-12-01 Alphavax, Inc. Tc-83-derived alphavirus vectors, particles and methods
WO2005120152A2 (en) 2004-06-07 2005-12-22 Protiva Biotherapeutics, Inc. Cationic lipids and methods of use
WO2005121348A1 (en) 2004-06-07 2005-12-22 Protiva Biotherapeutics, Inc. Lipid encapsulated interfering rna
WO2006089264A2 (en) 2005-02-18 2006-08-24 Novartis Vaccines And Diagnostics Inc. Proteins and nucleic acids from meningitis/sepsis-associated escherichia coli
WO2006091517A2 (en) 2005-02-18 2006-08-31 Novartis Vaccines And Diagnostics Inc. Immunogens from uropathogenic escherichia coli
WO2006110413A2 (en) 2005-03-30 2006-10-19 Novartis Vaccines And Diagnostics Inc. Haemophilus influenzae type b
WO2006138004A2 (en) 2005-05-12 2006-12-28 Novartis Vaccines And Diagnostics, Inc. Immunogenic compositions for chlamydia trachomatis
WO2007049155A2 (en) 2005-10-25 2007-05-03 Novartis Vaccines And Diagnostics Srl Compositions comprising yersinia pestis antigens
WO2008020330A2 (en) 2006-08-16 2008-02-21 Novartis Ag Immunogens from uropathogenic escherichia coli
WO2008103276A2 (en) * 2007-02-16 2008-08-28 Merck & Co., Inc. Compositions and methods for potentiated activity of biologicaly active molecules
WO2009016515A2 (en) 2007-08-01 2009-02-05 Novartis Ag Compositions comprising pneumococcal antigens
WO2009031043A2 (en) 2007-09-04 2009-03-12 Novartis Ag Compositions comprising yersinia pestis antigens
WO2009086558A1 (en) 2008-01-02 2009-07-09 Tekmira Pharmaceuticals Corporation Improved compositions and methods for the delivery of nucleic acids
WO2009104092A2 (en) 2008-02-22 2009-08-27 Novartis Ag Escherichia coli immunogens with improved solubility
WO2009111088A2 (en) * 2008-01-02 2009-09-11 The Johns Hopkins University Antitumor immunization by liposomal delivery of vaccine to the spleen
WO2009109860A2 (en) 2008-03-06 2009-09-11 Novartis Ag Mutant forms of chlamydia htra
WO2010119343A2 (en) 2009-04-14 2010-10-21 Novartis Ag Compositions for immunising against staphylococcus aureus
WO2011005799A2 (en) 2009-07-06 2011-01-13 Novartis Ag Self replicating rna molecules and uses thereof
WO2012006378A1 (en) 2010-07-06 2012-01-12 Novartis Ag Liposomes with lipids having an advantageous pka- value for rna delivery
WO2012006376A2 (en) 2010-07-06 2012-01-12 Novartis Ag Virion-like delivery particles for self-replicating rna molecules
WO2012006372A1 (en) 2010-07-06 2012-01-12 Novartis Ag Delivery of rna to different cell types
WO2012006369A2 (en) 2010-07-06 2012-01-12 Novartis Ag Immunisation of large mammals with low doses of rna

Family Cites Families (218)

* 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
US4853228A (en) * 1987-07-28 1989-08-01 Micro-Pak, Inc. Method of manufacturing unilamellar lipid vesicles
US6867195B1 (en) 1989-03-21 2005-03-15 Vical Incorporated Lipid-mediated polynucleotide administration to reduce likelihood of subject's becoming infected
DE69032284T2 (en) 1989-03-21 1998-10-08 Vical Inc EXPRESSION OF EXOGENOUS POLYNUCLEOTIDE SEQUENCES IN VERTEBLE
US5013556A (en) * 1989-10-20 1991-05-07 Liposome Technology, Inc. Liposomes with enhanced circulation time
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 (en) 1991-05-03 1994-11-18 Transgene Sa RNA DELIVERY VECTOR.
US5693535A (en) 1992-05-14 1997-12-02 Ribozyme Pharmaceuticals, Inc. HIV targeted ribozymes
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
CA2134773A1 (en) 1992-06-04 1993-12-09 Robert J. Debs Methods and compositions for in vivo gene therapy
EP0654077A4 (en) 1992-07-17 1996-03-13 Ribozyme Pharm 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
US5993850A (en) * 1994-09-13 1999-11-30 Skyepharma Inc. Preparation of multivesicular liposomes for controlled release of encapsulated biologically active substances
US5885613A (en) 1994-09-30 1999-03-23 The University Of British Columbia Bilayer stabilizing components and their use in forming programmable fusogenic liposomes
WO1996017072A2 (en) 1994-11-30 1996-06-06 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
WO1997012032A1 (en) * 1995-09-27 1997-04-03 The Government Of The United States Of America, As Represented By The Department Of Health And Human Services Production of infectious respiratory syncytial virus from cloned nucleotide sequences
DK0880360T3 (en) 1996-02-12 2003-01-27 Ml Lab Plc New methods of vaccination and vaccines thereto, which include a nucleic acid encoding a first epitope and a peptide containing a secondary epitope
DE19605548A1 (en) 1996-02-15 1997-09-04 Boehringer Ingelheim Int Composition for transfection of higher eukaryotic cells
US6451592B1 (en) 1996-04-05 2002-09-17 Chiron Corporation Recombinant alphavirus-based vectors with reduced inhibition of cellular macromolecular synthesis
JP2001503735A (en) 1996-07-03 2001-03-21 ユニバーシティ オブ ピッツバーグ Emulsion formulation for hydrophilic active reagent
KR100507660B1 (en) 1996-09-13 2005-08-10 리폭센 테크놀로지즈 리미티드 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
US6287591B1 (en) 1997-05-14 2001-09-11 Inex Pharmaceuticals Corp. Charged therapeutic agents encapsulated in lipid particles containing four lipid components
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 (en) 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 Flavivirus expression and delivery systems
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
EP2311436A1 (en) 1998-04-27 2011-04-20 Altus Pharmaceuticals Inc. Stabilized protein crystals, formulations containing them and methods of making them
EP1092031A2 (en) 1998-06-29 2001-04-18 U.S. Medical Research Institute of Infectious Diseases Marburg virus vaccines
CA2335393C (en) 1998-07-20 2008-09-23 Inex Pharmaceuticals Corporation Liposomal encapsulated nucleic acid-complexes
US6602705B1 (en) 1998-12-31 2003-08-05 Chiron Corporation Expression of HIV polypeptides and production of virus-like particles
ATE492644T1 (en) 1999-09-09 2011-01-15 Curevac Gmbh TRANSFER OF MRNA USING POLYCATIONIC COMPOUNDS
AU784605B2 (en) 1999-10-20 2006-05-11 Johns Hopkins University School Of Medicine, The Chimeric immunogenic compositions and nucleic acids encoding them
US8541008B2 (en) * 1999-11-19 2013-09-24 Los Angeles Biomedical Research Institute At Harbor-Ucla Medical Center Pharmaceutical compositions and methods to vaccinate against candidiasis
US20030212022A1 (en) 2001-03-23 2003-11-13 Jean-Marie Vogel Compositions and methods for gene therapy
CA2403508A1 (en) 2000-04-18 2001-10-25 Human Genome Sciences, Inc. Extracellular matrix polynucleotides, polypeptides, and antibodies
CA2411542A1 (en) * 2000-06-09 2001-12-13 Teni Boulikas Encapsulation of polynucleotides and drugs into targeted liposomes
AU2001290520A1 (en) 2000-08-01 2002-02-13 The Johns Hokpins University Intercellular transport protein linked to an antigen as a molecular vaccine
US20040142474A1 (en) 2000-09-14 2004-07-22 Expression Genetics, Inc. Novel cationic lipopolymer as a biocompatible gene delivery agent
ATE320792T1 (en) 2000-09-28 2006-04-15 Chiron Corp MICROPARTICLES FOR ADMINISTRATION OF HETEROLOGUE NUCLEIC ACID
WO2002079239A2 (en) 2001-01-31 2002-10-10 U.S. Army Medical Research Institute Of Infectious Diseases Chimeric filovirus glycoprotein
AU2002242016A1 (en) 2001-02-01 2002-08-12 The Johns Hopkins University Nucleic acid derived vaccine that encodes an antigen linked to a polypeptide that promotes antigen presentation
AU2002306709A1 (en) 2001-03-14 2002-09-24 Replicon Technologies, Inc. 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
AU2002256398A2 (en) 2001-04-30 2002-11-11 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
EP2305699B1 (en) 2001-06-05 2014-08-13 CureVac GmbH Stabilised mRNA with increased G/C content which is optimised for translation in its coded areas for the vaccination against sleeping sickness, leishmaniosis and toxoplasmosis
US7045335B2 (en) * 2001-09-06 2006-05-16 Alphavax, Inc. Alphavirus replicon vector systems
WO2003068190A1 (en) 2002-02-13 2003-08-21 Northeastern University Intracellular delivery of therapeutic agents
DE10207177A1 (en) 2002-02-19 2003-09-04 Novosom Ag Optionally cationic lipids
ES2354607T3 (en) 2002-06-28 2011-03-16 Protiva Biotherapeutics Inc. PROCEDURE AND APPLIANCE TO PRODUCE LIPOSOMES.
ATE348633T1 (en) 2002-07-05 2007-01-15 Lipoxen Technologies Ltd METHOD FOR BOOSTING AN IMMUNE RESPONSE BY NUCLEIC ACID VACCINATIONS
ES2242462B1 (en) 2002-07-18 2007-01-01 Marti Industria Metalurgica, S.L SELECTABLE ROTATING SPRAYER.
US20060251620A1 (en) * 2002-08-22 2006-11-09 Lidia Ivanova Inducible alphaviral/orip based gene expression system
CN1694959B (en) * 2002-09-13 2013-09-18 雷普利瑟公司 Non-sequence complementary antiviral oligonucleotides
AU2003297039B2 (en) 2002-12-13 2009-03-05 Alphavax, Inc. Multi-antigenic alphavirus replicon particles and methods
PT2311848E (en) 2002-12-23 2013-10-03 Vical Inc Codon-optimized polynucleotide-based vaccines against human cytomegalovirus infection
US8338583B2 (en) 2003-02-04 2012-12-25 Bar-Ilan University Snornai-small nucleolar RNA degradation by RNA interference in trypanosomatids
US20040228842A1 (en) 2003-02-27 2004-11-18 Shan Lu Compositions and methods for cytomegalovirus treatment
KR101454842B1 (en) 2003-03-20 2014-11-04 알파벡스, 인크. Improved alphavirus replicons and helper constructs
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
ES2327643T3 (en) 2003-07-11 2009-11-02 Alphavax, Inc. VACCINES AGAINST CITOMEGALOVIRUS BASED ON ALFAVIRUS.
US7368537B2 (en) * 2003-07-15 2008-05-06 Id Biomedical Corporation Of Quebec Subunit vaccine against respiratory syncytial virus infection
CN101291653B (en) 2003-07-16 2012-06-27 普洛体维生物治疗公司 Lipid encapsulated interfering rna
EP1512393A1 (en) * 2003-09-08 2005-03-09 BOEHRINGER INGELHEIM PHARMA GMBH &amp; CO. KG Process for the production of homogeneous liposomes and lipoplexes
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
US7303881B2 (en) 2004-04-30 2007-12-04 Pds Biotechnology Corporation Antigen delivery compositions and methods of use
EP1766094A4 (en) 2004-05-18 2009-11-25 Vical Inc Influenza virus vaccine composition and method of use
US20090104226A1 (en) 2004-05-21 2009-04-23 Novartis Vaccines And Diagnostics Inc. Alphavirus Vectors for Respiratory Pathogen Vaccines
GB0411428D0 (en) 2004-05-21 2004-06-23 Got A Gene Ab Vectors
US20080268429A1 (en) * 2004-06-02 2008-10-30 Sourcepharm, Inc. Rna - Containing Microvesicles and Methods Therefor
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
WO2006007712A1 (en) * 2004-07-19 2006-01-26 Protiva Biotherapeutics, Inc. Methods comprising polyethylene glycol-lipid conjugates for delivery of therapeutic agents
WO2006038129A2 (en) 2004-10-01 2006-04-13 Novartis Vaccines And Diagnostics Srl Hepatitis c virus replication system
JP2008520600A (en) 2004-11-19 2008-06-19 ノヴォソム アクチェンゲゼルシャフト Improvements in or relating to pharmaceutical compositions for topical administration
GB2421025A (en) 2004-12-09 2006-06-14 Oxxon Therapeutics Ltd HSV vaccination vectors
WO2007086883A2 (en) 2005-02-14 2007-08-02 Sirna Therapeutics, Inc. Cationic lipids and formulated molecular compositions containing them
US7404969B2 (en) 2005-02-14 2008-07-29 Sirna Therapeutics, Inc. Lipid nanoparticle based compositions and methods for the delivery of biologically active molecules
JP2008531672A (en) 2005-03-02 2008-08-14 イギリス国 Pharmaceutical composition
GB0504436D0 (en) 2005-03-03 2005-04-06 Glaxosmithkline Biolog Sa Vaccine
US7618393B2 (en) 2005-05-03 2009-11-17 Pharmajet, Inc. Needle-less injector and method of fluid delivery
US8703095B2 (en) 2005-07-07 2014-04-22 Sanofi Pasteur S.A. Immuno-adjuvant emulsion
EP1909758A1 (en) * 2005-08-02 2008-04-16 I.D.M. Immuno-Designed Molecules Process for the preparation of liposomal formulations
US7951384B2 (en) 2005-08-05 2011-05-31 University Of Massachusetts Virus-like particles as vaccines for paramyxovirus
HUE043492T2 (en) 2005-08-23 2019-08-28 Univ Pennsylvania Rna containing modified nucleosides and methods of use thereof
EP1764089A1 (en) 2005-09-15 2007-03-21 Novosom AG Serum stable liposomes comprising amphoter II lipid mixtures
DE102005046490A1 (en) 2005-09-28 2007-03-29 Johannes-Gutenberg-Universität Mainz New nucleic acid molecule comprising promoter, a transcriptable nucleic acid sequence, a first and second nucleic acid sequence for producing modified RNA with transcriptional stability and translational efficiency
BRPI0616687A2 (en) 2005-09-29 2011-06-28 Elan Pharm Inc pyrimidinyl amide compounds that inhibit vla-4 mediated leukocyte adhesion
EP1945247A1 (en) 2005-10-18 2008-07-23 Novartis Vaccines and Diagnostics, Inc. Mucosal and systemic immunizations with alphavirus replicon particles
JP2007112768A (en) 2005-10-24 2007-05-10 Kyoto Univ Liver-directed liposome composition
ES2514316T3 (en) 2005-11-22 2014-10-28 Novartis Vaccines And Diagnostics, Inc. Norovirus and Sapovirus virus-like particles (VLPs)
WO2008051245A2 (en) 2005-12-02 2008-05-02 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
US20070281336A1 (en) 2006-04-14 2007-12-06 Epicentre Technologies Kits and methods for generating 5' capped RNA
US7704510B2 (en) 2006-06-07 2010-04-27 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
DK2035581T3 (en) 2006-06-21 2012-10-08 Scripps Research Inst DNA COMPOSITION AGAINST TUMOR STREAM ANTI-FAP AND PROCEDURES FOR USING IT
CA2659301A1 (en) 2006-07-28 2008-02-07 Applera Corporation Dinucleotide mrna cap analogs
EP2061508B1 (en) 2006-09-12 2014-10-15 Alphavax, Inc. Alphavirus replicon particles matched to protein antigens as immunological adjuvants
DE102007001370A1 (en) 2007-01-09 2008-07-10 Curevac Gmbh RNA-encoded antibodies
WO2008109806A2 (en) 2007-03-08 2008-09-12 Massachusetts Institute Of Technology 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
JP5475643B2 (en) 2007-05-04 2014-04-16 マリーナ バイオテック,インコーポレイテッド Amino acid lipids and uses thereof
US20090131355A1 (en) 2007-05-23 2009-05-21 Adrian Ion Bot Multicistronic vectors and methods for their design
DE102007029471A1 (en) 2007-06-20 2008-12-24 Novosom Ag New optional cationic sterols
ES2670813T3 (en) 2007-06-21 2018-06-01 Alphavax, Inc. Alphavirus Replicon Particles for Vaccination Use
US8202518B2 (en) 2007-07-04 2012-06-19 Ribovax Biotechnologies S.A. Antibodies against human cytomegalovirus (HCMV)
WO2009026328A2 (en) 2007-08-21 2009-02-26 Immune Disease Institute, Inc. Methods of delivery of agents to leukocytes and endothelial cells
WO2009042794A2 (en) 2007-09-26 2009-04-02 Vanderbilt University Venezuelan equine encephalitis replicons expressing paramyxovirus glycoproteins as vaccine
EP2042193A1 (en) 2007-09-28 2009-04-01 Biomay AG RNA Vaccines
WO2009079185A2 (en) 2007-11-26 2009-06-25 Novartis Vaccines And Diagnostics, Inc. Methods of generating alphavirus particles
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
JP5475753B2 (en) 2008-04-15 2014-04-16 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 Lipid formulations for nucleic acid delivery
WO2009127230A1 (en) 2008-04-16 2009-10-22 Curevac Gmbh MODIFIED (m)RNA FOR SUPPRESSING OR AVOIDING AN IMMUNOSTIMULATORY RESPONSE AND IMMUNOSUPPRESSIVE COMPOSITION
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
EP2130912A1 (en) * 2008-06-04 2009-12-09 Institut für Viruskrankeiten und Immunprophylaxe Pestivirus replicons providing an RNA-based viral vector system
US20110229969A1 (en) 2008-06-25 2011-09-22 Volker Sandig Cell Line for Propagation of Highly Attenuated AlphaViruses
JP5712126B2 (en) 2008-06-25 2015-05-07 ノバルティス アーゲー Rapid response to delayed boost immunization
EP3009449B1 (en) 2008-07-16 2018-06-06 Institute for Research in Biomedicine Human cytomegalovirus neutralizing antibodies and use thereof
JP2010025644A (en) 2008-07-16 2010-02-04 Kochi Univ Of Technology Coloration reagent of nitrate ions and method for detecting and quantifying nitrate ions using it
EA201170204A1 (en) 2008-07-16 2011-08-30 ХЬЮМЭБС ЭлЭлСи ANTIBODIES, NEUTRALIZING HUMAN CYTOMEGALOVIRUS AND THEIR APPLICATION
CA2733147A1 (en) 2008-08-06 2010-02-11 Novartis Ag Microparticles for use in immunogenic compositions
CL2008002322A1 (en) 2008-08-07 2009-06-05 Univ Concepcion Veterinary pharmaceutical formulation comprising a viral vector system consisting of a recombinant RNA particle encoding a cu / zn superoxide dismutase from the pathogenic bacterium of bovine brucella abortus, and at least one arn alphavirus belonging to the family of the semliki forest virus (sfv) , useful as a vaccine.
ES2915692T3 (en) 2008-08-13 2022-06-24 California Inst Of Techn Carrier nanoparticles and related compositions, methods and systems
US20110177122A1 (en) 2008-09-26 2011-07-21 The United States Of America, As Represented By The Secretary, Dept. Of Health & Human Services Dna prime/activated 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
EP2743265B1 (en) 2008-10-09 2017-03-15 Arbutus Biopharma Corporation Improved amino lipids and methods for the delivery of nucleic acids
WO2010053572A2 (en) 2008-11-07 2010-05-14 Massachusetts Institute Of Technology Aminoalcohol lipidoids and uses thereof
EP3207944B1 (en) 2008-11-10 2020-01-15 Arbutus Biopharma Corporation Novel lipids and compositions for the delivery of therapeutics
EP2367844A4 (en) 2008-11-18 2012-08-01 Ligocyte Pharmaceuticals Inc Rsv f vlps and methods of manufacture and use thereof
WO2010088537A2 (en) * 2009-01-29 2010-08-05 Alnylam Pharmaceuticals, Inc. Improved lipid formulation
AU2010245933B2 (en) 2009-05-05 2016-06-16 Arbutus Biopharma Corporation Methods of delivering oligonucleotides to immune cells
HUE038796T2 (en) 2009-06-10 2018-11-28 Arbutus Biopharma Corp Improved lipid formulation
ES2613498T3 (en) 2009-07-01 2017-05-24 Protiva Biotherapeutics Inc. New lipid formulations for the delivery of therapeutic agents to solid tumors
JP4900536B2 (en) 2009-07-02 2012-03-21 コニカミノルタホールディングス株式会社 Method for producing single cell liposomes by a two-stage emulsification method using an external aqueous phase containing a specific dispersant, and a method for producing a single cell liposome dispersion or a dry powder thereof using the method for producing single cell liposomes
SG178026A1 (en) 2009-07-15 2012-03-29 Novartis Ag Rsv f protein compositions and methods for making same
WO2011007359A2 (en) 2009-07-16 2011-01-20 Vaxil Biotherapeutics Ltd. Antigen specific multi epitope -based anti-infective vaccines
EP3581197A1 (en) 2009-07-31 2019-12-18 ethris GmbH Rna with a combination of unmodified and modified nucleotides for protein expression
CN102844047B (en) 2009-09-02 2017-04-05 诺华股份有限公司 Immunogenic composition containing TLR active regulators
TWI445708B (en) 2009-09-02 2014-07-21 Irm Llc Compounds and compositions as tlr activity modulators
US20110070260A1 (en) 2009-09-09 2011-03-24 Baric Ralph S Multivalent Immunogenic Compositions Against Noroviruses and Methods of Use
EP2496700B1 (en) 2009-11-04 2017-03-01 The University Of British Columbia Nucleic acid-containing lipid particles and related methods
US20110112353A1 (en) 2009-11-09 2011-05-12 Circulite, Inc. Bifurcated outflow cannulae
AU2010326132B9 (en) 2009-12-01 2014-10-02 Translate Bio, Inc. Delivery of mRNA for the augmentation of proteins and enzymes in human genetic diseases
CA3170391A1 (en) 2009-12-07 2011-06-16 The Trustees Of The University Of Pennsylvania Rna preparations comprising purified modified rna for reprogramming cells
WO2011071860A2 (en) 2009-12-07 2011-06-16 Alnylam Pharmaceuticals, Inc. 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
MX348474B (en) 2009-12-23 2017-06-14 Novartis Ag * Lipids, lipid compositions, and methods of using them.
US20130101609A1 (en) 2010-01-24 2013-04-25 Novartis Ag Irradiated biodegradable polymer microparticles
ES2651005T3 (en) 2010-03-09 2018-01-23 Biomedical Research Models, Inc. A new mucosal vaccination strategy 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
US9192661B2 (en) 2010-07-06 2015-11-24 Novartis Ag Delivery of self-replicating RNA using biodegradable polymer particles
NZ606591A (en) 2010-07-06 2015-02-27 Novartis Ag Cationic oil-in-water emulsions
PT3243526T (en) 2010-07-06 2020-03-04 Glaxosmithkline Biologicals Sa Delivery of rna to trigger multiple immune pathways
EP3153578A1 (en) 2010-07-06 2017-04-12 Novartis Ag Norovirus derived immunogenic compositions and methods
US8898852B2 (en) 2010-08-04 2014-12-02 Honeywell International Inc. Air burst to clear detection window
CA2807552A1 (en) 2010-08-06 2012-02-09 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
JP5908477B2 (en) 2010-08-31 2016-04-26 ノバルティス アーゲー Lipids suitable for liposome delivery of protein-encoding RNA
HUE059214T2 (en) 2010-08-31 2022-10-28 Glaxosmithkline Biologicals Sa Small liposomes for delivery of immunogen-encoding rna
MX2013002336A (en) 2010-08-31 2013-03-18 Novartis Ag Pegylated liposomes for delivery of immunogen-encoding rna.
WO2012034025A2 (en) 2010-09-09 2012-03-15 Virginia Commonwealth University Human cytomegalovirus vaccine
EP4108671A1 (en) 2010-10-01 2022-12-28 ModernaTX, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
BR112013008700B8 (en) 2010-10-11 2022-10-04 Novartis Ag SELF-REPLICATING RNA MOLECULE, ALPHAVIRUS REPLICON PARTICLE, COMPOSITION, RECOMBINANT DNA MOLECULE, USE OF SELF-REPLICATING RNA MOLECULE
BR112013009946B1 (en) 2010-10-25 2019-04-30 Stepan Company COMPOSITION OF SULFOBETAIN, BETAIN OR QUARTERNARY AMMONIUM, DERIVED; GLYPHOSATE FORMULATION, WATER SOLUBLE HERBICIDE COMPOSITION OR ANTIMICROBIAN COMPOSITION, Rough Surface Cleaner, FORMULATION FOR DIRTY CLOTHING, Shampoo OR CONDITIONER OF HAIR SPRAYING, PERSONAL PURPOSE CLEANING , SPARKLING, FOAM ADDITIVE OR DISPERSING AND ANIONIC EMULSIFICANT FOR AGRICULTURAL COMPOSITIONS
AU2012211278B2 (en) 2011-01-26 2016-11-10 Glaxosmithkline Biologicals Sa RSV immunization regimen
JP6099573B2 (en) 2011-01-31 2017-03-22 ザ トラスティーズ オブ ザ ユニバーシティ オブ ペンシルバニア Nucleic acid molecules encoding novel herpes antigens, vaccines containing them and methods of use thereof
WO2012116714A1 (en) 2011-03-02 2012-09-07 Curevac Gmbh Vaccination in elderly patients
AU2012236099A1 (en) 2011-03-31 2013-10-03 Moderna Therapeutics, Inc. Delivery and formulation of engineered nucleic acids
LT2707385T (en) 2011-05-13 2017-12-11 Glaxosmithkline Biologicals Sa Pre-fusion rsv f antigens
WO2012158736A1 (en) 2011-05-17 2012-11-22 modeRNA Therapeutics Engineered nucleic acids and methods of use thereof for non-human vertebrates
ES2663360T3 (en) 2011-06-08 2018-04-12 Translate Bio, Inc. Cleavable lipids
EP2729125B1 (en) 2011-07-06 2017-12-13 GlaxoSmithKline Biologicals SA Oil-in-water emulsions that contain nucleic acids
US20140141070A1 (en) 2011-07-06 2014-05-22 Andrew Geall Liposomes having useful n:p ratio for delivery of rna molecules
EP3332802A1 (en) 2011-07-06 2018-06-13 GlaxoSmithKline Biologicals SA Immunogenic combination compositions and uses thereof
EP2729124B1 (en) 2011-07-06 2018-10-24 GlaxoSmithKline Biologicals SA Cationic oil-in-water emulsions
EP3508219A1 (en) 2011-07-06 2019-07-10 GlaxoSmithKline Biologicals S.A. Self-replicating rna prime - protein boost vaccines
SG10201602456WA (en) 2011-08-31 2016-04-28 Novartis Ag Pegylated liposomes for delivery of immunogen-encoding rna
EP2755693A4 (en) 2011-09-12 2015-05-20 Moderna Therapeutics Inc Engineered nucleic acids and methods of use thereof
CN103974724B (en) 2011-10-03 2019-08-30 现代泰克斯公司 Nucleosides, nucleotide and nucleic acid of modification and application thereof
CA2872033A1 (en) 2011-10-11 2013-04-18 Novartis Ag Recombinant self-replicating polycistronic rna molecules
US20140348863A1 (en) 2011-10-12 2014-11-27 Alessia Bianchi Cmv antigens and uses thereof
WO2013130161A1 (en) 2011-12-14 2013-09-06 modeRNA Therapeutics Methods of responding to a biothreat
CA2859387A1 (en) 2011-12-16 2013-06-20 Moderna Therapeutics, Inc. Modified nucleoside, nucleotide, and nucleic acid compositions
JP2015510495A (en) 2011-12-21 2015-04-09 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. Methods for extending the viability or longevity of an organ or organ graft
AU2013243951A1 (en) 2012-04-02 2014-10-30 Moderna Therapeutics, Inc. Modified polynucleotides for the production of secreted proteins
CN104411338A (en) 2012-04-02 2015-03-11 现代治疗公司 Modified polynucleotides for the production of biologics and proteins associated with human disease
JP6144355B2 (en) 2012-11-26 2017-06-07 モデルナティエックス インコーポレイテッドModernaTX,Inc. Chemically modified mRNA
CA2897752A1 (en) 2013-01-10 2014-07-17 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
WO2014160243A1 (en) 2013-03-14 2014-10-02 The Trustees Of The University Of Pennsylvania Purification and purity assessment of rna molecules synthesized with modified nucleosides
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
PL3083556T3 (en) 2013-12-19 2020-06-29 Novartis Ag Lipids and lipid compositions for the delivery of active agents
LT3350157T (en) 2015-09-17 2022-02-25 Modernatx, Inc. Compounds and compositions for intracellular delivery of therapeutic agents
CN113636947A (en) 2015-10-28 2021-11-12 爱康泰生治疗公司 Novel lipid and lipid nanoparticle formulations for delivery of nucleic acids
MA46584A (en) 2016-10-21 2019-08-28 Modernatx Inc HUMAN CYTOMEGALOVIRUS VACCINE
CN110114058B (en) 2016-11-10 2023-05-26 川斯勒佰尔公司 Improved ICE-based lipid nanoparticle formulations for delivery of MRNA
US11045540B2 (en) 2017-03-15 2021-06-29 Modernatx, Inc. Varicella zoster virus (VZV) vaccine
EP3883592A1 (en) 2018-11-21 2021-09-29 Translate Bio, Inc. Treatment of cystic fibrosis by delivery of nebulized mrna encoding cftr
WO2021038508A1 (en) 2019-08-30 2021-03-04 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
WO2022150717A1 (en) 2021-01-11 2022-07-14 Modernatx, Inc. Seasonal rna influenza virus vaccines

Patent Citations (34)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5820873A (en) 1994-09-30 1998-10-13 The University Of British Columbia Polyethylene glycol modified ceramide lipids and liposome uses thereof
US6858224B2 (en) 1995-06-07 2005-02-22 Inex Pharmaceuticals Corporation Method of preventing aggregation of a lipid:nucleic acid complex
EP0910576B1 (en) 1996-04-11 2004-08-11 University Of British Columbia Fusogenic liposomes
WO1999052503A2 (en) 1998-04-16 1999-10-21 John Wayne Cancer Institute Rna cancer vaccine and methods for its use
WO2002002606A2 (en) 2000-07-03 2002-01-10 Chiron S.P.A. Immunisation against chlamydia pneumoniae
WO2002034771A2 (en) 2000-10-27 2002-05-02 Chiron Srl Nucleic acids and proteins from streptococcus groups a & b
WO2003018054A1 (en) 2001-08-31 2003-03-06 Chiron Srl. Helicobacter pylori vaccination
WO2005002619A2 (en) 2003-06-26 2005-01-13 Chiron Corporation Immunogenic compositions for chlamydia trachomatis
WO2005032582A2 (en) 2003-07-31 2005-04-14 Chiron Corporation Immunogenic compositions for streptococcus pyogenes
WO2005026372A1 (en) 2003-09-15 2005-03-24 Protiva Biotherapeutics, Inc. Polyethyleneglycol-modified lipid compounds and uses thereof
WO2005089148A2 (en) 2004-03-12 2005-09-29 The Regents Of The University Of California Delivery of genes encoding short hairpin rna using receptor-specific nanocontainers
WO2005111066A2 (en) 2004-05-14 2005-11-24 Chiron Srl Polypeptides from non-typeable haemophilus influenzae
WO2005113782A1 (en) 2004-05-18 2005-12-01 Alphavax, Inc. Tc-83-derived alphavirus vectors, particles and methods
WO2005120152A2 (en) 2004-06-07 2005-12-22 Protiva Biotherapeutics, Inc. Cationic lipids and methods of use
WO2005121348A1 (en) 2004-06-07 2005-12-22 Protiva Biotherapeutics, Inc. Lipid encapsulated interfering rna
WO2006091517A2 (en) 2005-02-18 2006-08-31 Novartis Vaccines And Diagnostics Inc. Immunogens from uropathogenic escherichia coli
WO2006089264A2 (en) 2005-02-18 2006-08-24 Novartis Vaccines And Diagnostics Inc. Proteins and nucleic acids from meningitis/sepsis-associated escherichia coli
WO2006110413A2 (en) 2005-03-30 2006-10-19 Novartis Vaccines And Diagnostics Inc. Haemophilus influenzae type b
WO2006138004A2 (en) 2005-05-12 2006-12-28 Novartis Vaccines And Diagnostics, Inc. Immunogenic compositions for chlamydia trachomatis
WO2007049155A2 (en) 2005-10-25 2007-05-03 Novartis Vaccines And Diagnostics Srl Compositions comprising yersinia pestis antigens
WO2008020330A2 (en) 2006-08-16 2008-02-21 Novartis Ag Immunogens from uropathogenic escherichia coli
WO2008103276A2 (en) * 2007-02-16 2008-08-28 Merck & Co., Inc. Compositions and methods for potentiated activity of biologicaly active molecules
WO2009016515A2 (en) 2007-08-01 2009-02-05 Novartis Ag Compositions comprising pneumococcal antigens
WO2009031043A2 (en) 2007-09-04 2009-03-12 Novartis Ag Compositions comprising yersinia pestis antigens
WO2009086558A1 (en) 2008-01-02 2009-07-09 Tekmira Pharmaceuticals Corporation Improved compositions and methods for the delivery of nucleic acids
WO2009111088A2 (en) * 2008-01-02 2009-09-11 The Johns Hopkins University Antitumor immunization by liposomal delivery of vaccine to the spleen
WO2009104092A2 (en) 2008-02-22 2009-08-27 Novartis Ag Escherichia coli immunogens with improved solubility
WO2009109860A2 (en) 2008-03-06 2009-09-11 Novartis Ag Mutant forms of chlamydia htra
WO2010119343A2 (en) 2009-04-14 2010-10-21 Novartis Ag Compositions for immunising against staphylococcus aureus
WO2011005799A2 (en) 2009-07-06 2011-01-13 Novartis Ag Self replicating rna molecules and uses thereof
WO2012006378A1 (en) 2010-07-06 2012-01-12 Novartis Ag Liposomes with lipids having an advantageous pka- value for rna delivery
WO2012006376A2 (en) 2010-07-06 2012-01-12 Novartis Ag Virion-like delivery particles for self-replicating rna molecules
WO2012006372A1 (en) 2010-07-06 2012-01-12 Novartis Ag Delivery of rna to different cell types
WO2012006369A2 (en) 2010-07-06 2012-01-12 Novartis Ag Immunisation of large mammals with low doses of rna

Non-Patent Citations (26)

* Cited by examiner, † Cited by third party
Title
"Functional Polymer Colloids and Microparticles volume 4 (Microspheres, microcapsules & liposomes", vol. 4, 2002, CITUS BOOKS
"Handbook of Experimental Immunolog", vol. I-IV, 1986, BLACKWELL SCIENTIFIC PUBLICATIONS
"Handbook of Surface and Colloidal Chemistry", 1997, CRC PRESS
"Liposome Technology", vol. I, II, I, 2006, INFORMA HEALTHCARE
"Liposomes: Methods and Protocols, Volume 1: Pharmaceutical Nanocarriers: Methods and Protocols.", vol. 1, 2009, HUMANA PRESS
"Methods In Enzymology", ACADEMIC PRESS, INC.
"Molecular Biology Techniques: An Intensive Laboratory Course", 1998, ACADEMIC PRESS
"PCR (Introduction to Biotechniques Series", 1997, SPRINGER VERLAG
"Short protocols in molecular biology", 2002, CURRENT PROTOCOLS
BOXUS ET AL., J VIROL, vol. 81, 2007, pages 6879 - 89
CHEN, Y. ET AL.: "Nanoparticles Targeted With NGR Motif Deliver c-myc siRNA and Doxorubicin for Anticancer Therapy", MOLECULAR THERAPY, vol. 18, no. 4, 12 January 2010 (2010-01-12), pages 828 - 834, XP055363965
EL OUAHABI ET AL., FEBS LETTS, vol. 380, 1996, pages 108 - 12
GENNARO: "Remington: The Science and Practice of Pharmacy", 2000
GIULIANI ET AL., PROC NATL ACAD SCI USA, vol. 103, no. 29, 2006, pages 10834 - 9
HEYES ET AL., J CONTROLLED RELEASE, vol. 107, 2005, pages 276 - 87
HOEKSTRA ET AL., BIOCHIMICA ET BIOPHYSICA ACTA, vol. 1660, 2004, pages 41 - 52
JEFFS ET AL., PHARMACEUTICAL RESEARCH, vol. 22, no. 3, 2005, pages 362 - 372
JOHANNING ET AL., NUCLEIC ACIDS RES, vol. 23, 1995, pages 1495 - 1501
MAURER ET AL., BIOPHYSICAL JOUMAL, vol. 80, 2001, pages 2310 - 2326
MOCKEY M ET AL: "mRNA-based cancer vaccine: prevention of B16 melanoma progression and metastasis by systemic injection of MART1 mRNA histidylated lipopolyplexes", CANCER GENE THERAPY, vol. 14, no. 9, September 2007 (2007-09-01), pages 802 - 814, XP055007324, ISSN: 0929-1903 *
MOCKEY, M. ET AL.: "mRNA-based cancer vaccine: prevention of B16 melanoma progression and metastasis by systemic injection of MART1 mRNA histidylated lipopolyplexes", CANCER GENE THERAPY, vol. 14, 22 June 2007 (2007-06-22), pages 802 - 814, XP002666288
PERRI ET AL., J VIROL, vol. 77, 2003, pages 10394 - 10403
ROMBERG ET AL., PHARMACEUTICAL RESEARCH, vol. 25, 2008, pages 55 - 71
SAMBROOK ET AL.: "Molecular Cloning: A Laboratory Manual", 2001, COLD SPRING HARBOR LABORATORY PRESS
TAYLOR ET AL., VACCINE, vol. 23, 2005, pages 1242 - 50
YONEYAMA, FUJITA, CYTOKINE & GROWTH FACTOR REVIEWS, vol. 18, 2007, pages 545 - 51

Cited By (296)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11666534B2 (en) 2010-07-06 2023-06-06 Glaxosmithkline Biologicals Sa Methods of administering lipid formulations with viral immunogens
US11707482B2 (en) 2010-07-06 2023-07-25 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11324770B2 (en) 2010-07-06 2022-05-10 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
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
US11883534B2 (en) 2010-07-06 2024-01-30 Glaxosmithkline Biologicals Sa Immunisation with lipid formulations with RNA encoding immunogens
US11913001B2 (en) 2010-07-06 2024-02-27 Glaxosmithkline Biologicals Sa Immunisation of large mammals with low doses of RNA
EP4005592B1 (en) 2010-07-06 2022-10-12 GlaxoSmithKline Biologicals S.A. Virion-like delivery particles for self-replicating rna molecules
US11905514B2 (en) 2010-07-06 2024-02-20 Glaxosmithkline Biological Sa Immunisation of large mammals with low doses of RNA
US11773395B1 (en) 2010-07-06 2023-10-03 Glaxosmithkline Biologicals Sa Immunization of large mammals with low doses of RNA
US20130195968A1 (en) * 2010-07-06 2013-08-01 Novartis Ag Virion-like delivery particles for self-replicating rna molecules
US11865080B2 (en) 2010-07-06 2024-01-09 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
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
US11857562B2 (en) 2010-07-06 2024-01-02 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
US11655475B2 (en) 2010-07-06 2023-05-23 Glaxosmithkline Biologicals Sa Immunisation of large mammals with low doses of RNA
US11857681B2 (en) 2010-07-06 2024-01-02 Glaxosmithkline Biologicals Sa Lipid formulations with RNA encoding immunogens
US11690863B2 (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
US11690865B2 (en) 2010-07-06 2023-07-04 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
US11696923B2 (en) 2010-07-06 2023-07-11 Glaxosmithkline Biologicals, Sa Delivery of RNA to trigger multiple immune pathways
US11850305B2 (en) 2010-07-06 2023-12-26 Glaxosmithkline Biologicals Sa Method of making lipid formulations with RNA encoding immunogens
US11717529B2 (en) 2010-07-06 2023-08-08 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11851660B2 (en) 2010-07-06 2023-12-26 Glaxosmithkline Biologicals Sa Immunisation of large mammals with low doses of RNA
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
US11845925B2 (en) 2010-07-06 2023-12-19 Glaxosmithkline Biologicals Sa Immunisation of large mammals with low doses of RNA
US11766401B2 (en) 2010-07-06 2023-09-26 Glaxosmithkline Biologicals Sa Methods of administering lipid formulations with immunogens
US11891608B2 (en) 2010-07-06 2024-02-06 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
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
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
EP3970742B1 (en) 2010-08-31 2022-05-25 GlaxoSmithKline Biologicals S.A. Pegylated liposomes for delivery of immunogen-encoding rna
EP4066855B1 (en) 2010-08-31 2022-12-28 GlaxoSmithKline Biologicals SA Pegylated liposomes for delivery of immunogen-encoding rna
EP4066857B1 (en) 2010-08-31 2022-12-21 GlaxoSmithKline Biologicals SA Pegylated 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
EP4066856B1 (en) 2010-08-31 2022-12-07 GlaxoSmithKline Biologicals SA Pegylated liposomes for delivery of immunogen-encoding rna
EP2611461B1 (en) 2010-08-31 2022-03-09 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
EP3981427B1 (en) 2010-08-31 2022-05-25 GlaxoSmithKline Biologicals S.A. Pegylated liposomes for delivery of immunogen-encoding rna
US9334328B2 (en) 2010-10-01 2016-05-10 Moderna Therapeutics, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US10064959B2 (en) 2010-10-01 2018-09-04 Modernatx, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US9657295B2 (en) 2010-10-01 2017-05-23 Modernatx, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US11639370B2 (en) 2010-10-11 2023-05-02 Glaxosmithkline Biologicals Sa Antigen delivery platforms
US9533047B2 (en) 2011-03-31 2017-01-03 Modernatx, Inc. Delivery and formulation of engineered nucleic acids
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
US11547764B2 (en) 2011-06-08 2023-01-10 Translate Bio, Inc. Lipid nanoparticle compositions and methods for MRNA delivery
US11951180B2 (en) 2011-06-08 2024-04-09 Translate Bio, Inc. Lipid nanoparticle compositions and methods for MRNA delivery
US11951181B2 (en) 2011-06-08 2024-04-09 Translate Bio, Inc. Lipid nanoparticle compositions and methods for mRNA delivery
US11730825B2 (en) 2011-06-08 2023-08-22 Translate Bio, Inc. Lipid nanoparticle compositions and methods for mRNA delivery
US11951179B2 (en) 2011-06-08 2024-04-09 Translate Bio, Inc. Lipid nanoparticle compositions and methods for MRNA delivery
EP4014966A1 (en) * 2011-07-06 2022-06-22 GlaxoSmithKline Biologicals S.A. Liposomes having useful n:p ratio for delivery of rna molecules
EP4115875A1 (en) * 2011-07-06 2023-01-11 GlaxoSmithKline Biologicals S.A. Liposomes having useful n:p ratio for delivery of rna molecules
EP4115876A1 (en) * 2011-07-06 2023-01-11 GlaxoSmithKline Biologicals S.A. Liposomes having useful n:p ratio for delivery of rna molecules
EP3821879A1 (en) * 2011-07-06 2021-05-19 GlaxoSmithKline Biologicals S.A. Liposomes having useful n:p ratio for delivery of rna molecules
WO2013006825A1 (en) * 2011-07-06 2013-01-10 Novartis Ag Liposomes having useful n:p ratio for delivery of rna molecules
US11896636B2 (en) 2011-07-06 2024-02-13 Glaxosmithkline Biologicals Sa Immunogenic combination compositions and uses thereof
EP2750707B1 (en) 2011-08-31 2018-10-24 GlaxoSmithKline Biologicals SA Pegylated liposomes for delivery of immunogen-encoding rna
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
US10751386B2 (en) 2011-09-12 2020-08-25 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
US9271996B2 (en) 2011-12-16 2016-03-01 Moderna Therapeutics, Inc. Formulation and delivery of PLGA microspheres
US9295689B2 (en) 2011-12-16 2016-03-29 Moderna Therapeutics, Inc. Formulation and delivery of PLGA microspheres
US9186372B2 (en) 2011-12-16 2015-11-17 Moderna Therapeutics, Inc. Split dose administration
EP4144378A1 (en) 2011-12-16 2023-03-08 ModernaTX, Inc. Modified nucleoside, nucleotide, and nucleic acid compositions
US8754062B2 (en) 2011-12-16 2014-06-17 Moderna Therapeutics, Inc. DLIN-KC2-DMA lipid nanoparticle delivery of modified polynucleotides
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
US9192651B2 (en) 2012-04-02 2015-11-24 Moderna Therapeutics, Inc. Modified polynucleotides for the production of secreted proteins
US9782462B2 (en) 2012-04-02 2017-10-10 Modernatx, Inc. Modified polynucleotides for the production of proteins associated with human disease
WO2013151666A2 (en) 2012-04-02 2013-10-10 modeRNA Therapeutics Modified polynucleotides for the production of biologics and proteins associated with human disease
WO2013151736A2 (en) 2012-04-02 2013-10-10 modeRNA Therapeutics In vivo production of proteins
US9149506B2 (en) 2012-04-02 2015-10-06 Moderna Therapeutics, Inc. Modified polynucleotides encoding septin-4
US9572897B2 (en) 2012-04-02 2017-02-21 Modernatx, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
US9675668B2 (en) 2012-04-02 2017-06-13 Moderna Therapeutics, Inc. Modified polynucleotides encoding hepatitis A virus cellular receptor 2
US9114113B2 (en) 2012-04-02 2015-08-25 Moderna Therapeutics, Inc. Modified polynucleotides encoding citeD4
US9301993B2 (en) 2012-04-02 2016-04-05 Moderna Therapeutics, Inc. Modified polynucleotides encoding apoptosis inducing factor 1
US9303079B2 (en) 2012-04-02 2016-04-05 Moderna Therapeutics, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
US9220755B2 (en) 2012-04-02 2015-12-29 Moderna Therapeutics, Inc. Modified polynucleotides for the production of proteins associated with blood and lymphatic disorders
US10501512B2 (en) 2012-04-02 2019-12-10 Modernatx, Inc. Modified polynucleotides
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
US9107886B2 (en) 2012-04-02 2015-08-18 Moderna Therapeutics, Inc. Modified polynucleotides encoding basic helix-loop-helix family member E41
US9095552B2 (en) 2012-04-02 2015-08-04 Moderna Therapeutics, Inc. Modified polynucleotides encoding copper metabolism (MURR1) domain containing 1
US9827332B2 (en) 2012-04-02 2017-11-28 Modernatx, Inc. Modified polynucleotides for the production of proteins
US9878056B2 (en) 2012-04-02 2018-01-30 Modernatx, Inc. Modified polynucleotides for the production of cosmetic proteins and peptides
US9254311B2 (en) 2012-04-02 2016-02-09 Moderna Therapeutics, 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
US9255129B2 (en) 2012-04-02 2016-02-09 Moderna Therapeutics, Inc. Modified polynucleotides encoding SIAH E3 ubiquitin protein ligase 1
US9814760B2 (en) 2012-04-02 2017-11-14 Modernatx, Inc. Modified polynucleotides for the production of biologics and proteins associated with human disease
US8999380B2 (en) 2012-04-02 2015-04-07 Moderna Therapeutics, Inc. Modified polynucleotides for the production of biologics and proteins associated with human disease
US9233141B2 (en) 2012-04-02 2016-01-12 Moderna Therapeutics, Inc. Modified polynucleotides for the production of proteins associated with blood and lymphatic disorders
US9220792B2 (en) 2012-04-02 2015-12-29 Moderna Therapeutics, Inc. Modified polynucleotides encoding aquaporin-5
US9050297B2 (en) 2012-04-02 2015-06-09 Moderna Therapeutics, Inc. Modified polynucleotides encoding aryl hydrocarbon receptor nuclear translocator
US9221891B2 (en) 2012-04-02 2015-12-29 Moderna Therapeutics, Inc. In vivo production of proteins
US9061059B2 (en) 2012-04-02 2015-06-23 Moderna Therapeutics, Inc. Modified polynucleotides for treating protein deficiency
US9216205B2 (en) 2012-04-02 2015-12-22 Moderna Therapeutics, Inc. Modified polynucleotides encoding granulysin
US9089604B2 (en) 2012-04-02 2015-07-28 Moderna Therapeutics, Inc. Modified polynucleotides for treating galactosylceramidase protein deficiency
EP4074834A1 (en) 2012-11-26 2022-10-19 ModernaTX, Inc. Terminally modified rna
US9597380B2 (en) 2012-11-26 2017-03-21 Modernatx, Inc. Terminally modified RNA
WO2014108515A1 (en) 2013-01-10 2014-07-17 Novartis Ag Influenza virus immunogenic compositions and uses thereof
WO2014113089A2 (en) 2013-01-17 2014-07-24 Moderna Therapeutics, Inc. Signal-sensor polynucleotides for the alteration of cellular phenotypes
EP3434774A1 (en) 2013-01-17 2019-01-30 ModernaTX, Inc. Signal-sensor polynucleotides for the alteration of cellular phenotypes
EP3608308A1 (en) 2013-03-08 2020-02-12 Novartis AG Lipids and lipid compositions for the delivery of active agents
WO2014136086A1 (en) 2013-03-08 2014-09-12 Novartis Ag Lipids and lipid compositions for the delivery of active agents
WO2014159813A1 (en) 2013-03-13 2014-10-02 Moderna Therapeutics, 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
US8980864B2 (en) 2013-03-15 2015-03-17 Moderna Therapeutics, Inc. Compositions and methods of altering cholesterol levels
EP3521429A1 (en) 2013-03-15 2019-08-07 GlaxoSmithKline Biologicals SA Rna purification methods
WO2014140211A1 (en) 2013-03-15 2014-09-18 Novartis Ag Rna purification methods
AU2014287009B2 (en) * 2013-07-11 2020-10-29 Modernatx, Inc. Compositions comprising synthetic polynucleotides encoding CRISPR related proteins and synthetic sgRNAs and methods of use
US11027025B2 (en) 2013-07-11 2021-06-08 Modernatx, Inc. Compositions comprising synthetic polynucleotides encoding CRISPR related proteins and synthetic sgRNAs and methods of use
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
US10815291B2 (en) 2013-09-30 2020-10-27 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
US10842859B2 (en) 2014-03-25 2020-11-24 Yale University Uses of parasite macrophage migration inhibitory factors
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
US9872900B2 (en) 2014-04-23 2018-01-23 Modernatx, Inc. Nucleic acid vaccines
WO2015177312A1 (en) 2014-05-22 2015-11-26 Glaxosmithkline Biologicals Sa Rsvf trimerization domains
US10106490B2 (en) 2014-06-25 2018-10-23 Acuitas Therapeutics, Inc. Lipids and lipid nanoparticle formulations for delivery of nucleic acids
EP4159741A1 (en) 2014-07-16 2023-04-05 ModernaTX, Inc. Method for producing a chimeric polynucleotide encoding a polypeptide having a triazole-containing internucleotide linkage
WO2016014846A1 (en) 2014-07-23 2016-01-28 Moderna Therapeutics, Inc. Modified polynucleotides for the production of intrabodies
WO2016135675A1 (en) 2015-02-27 2016-09-01 Novartis Ag Flavivirus replicons
EP3061826A1 (en) 2015-02-27 2016-08-31 Novartis AG Flavivirus replicons
US11168051B2 (en) 2015-06-29 2021-11-09 Acuitas Therapeutics, Inc. Lipids and lipid nanoparticle formulations for delivery of nucleic acids
US10221127B2 (en) 2015-06-29 2019-03-05 Acuitas Therapeutics, Inc. Lipids and lipid nanoparticle formulations for delivery of nucleic acids
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
US10933127B2 (en) 2015-10-22 2021-03-02 Modernatx, Inc. Betacoronavirus mRNA vaccine
US11872278B2 (en) 2015-10-22 2024-01-16 Modernatx, Inc. Combination HMPV/RSV RNA vaccines
EP3718565A1 (en) * 2015-10-22 2020-10-07 ModernaTX, Inc. Respiratory virus vaccines
EP4011451A1 (en) * 2015-10-22 2022-06-15 ModernaTX, Inc. Metapneumovirus mrna vaccines
US11648324B2 (en) 2015-10-28 2023-05-16 Acuitas Therapeutics, Inc. Lipids and lipid nanoparticle formulations for delivery of nucleic acids
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
US10195156B2 (en) 2015-12-22 2019-02-05 Modernatx, Inc. Compounds and compositions for intracellular delivery of agents
US10799463B2 (en) 2015-12-22 2020-10-13 Modernatx, Inc. Compounds and compositions for intracellular delivery of agents
EP4039699A1 (en) 2015-12-23 2022-08-10 ModernaTX, Inc. Methods of using ox40 ligand encoding polynucleotides
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
WO2017162461A1 (en) 2016-03-21 2017-09-28 Biontech Rna Pharmaceuticals Gmbh Trans-replicating rna
EP3964584A1 (en) 2016-03-21 2022-03-09 BioNTech SE Trans-replicating rna
EP3701959A1 (en) 2016-03-21 2020-09-02 BioNTech RNA Pharmaceuticals GmbH Rna replicon for versatile and efficient gene expression
WO2017200957A1 (en) * 2016-05-16 2017-11-23 Infectious Disease Research Institute Pegylated liposomes and methods of use
US11266602B2 (en) 2016-05-16 2022-03-08 Infectious Disease Research Institute PEGylated liposomes and methods of use
RU2796539C2 (en) * 2016-05-16 2023-05-25 Инфекшес Дизис Рисёрч Инститьют Pegylated liposomes and their application methods
CN109562057A (en) * 2016-05-16 2019-04-02 传染病研究所 Pegylated liposomal and application method
AU2017268175B2 (en) * 2016-05-16 2023-02-23 Access To Advanced Health Institute PEGylated liposomes and methods of use
US10730924B2 (en) 2016-05-18 2020-08-04 Modernatx, Inc. Polynucleotides encoding relaxin
WO2017208191A1 (en) 2016-06-02 2017-12-07 Glaxosmithkline Biologicals Sa Zika viral antigen constructs
WO2018033254A2 (en) 2016-08-19 2018-02-22 Curevac Ag Rna for cancer therapy
WO2018060288A1 (en) 2016-09-29 2018-04-05 Glaxosmithkline Biologicals S.A. Compositions and methods of treatment of persistent hpv infection
WO2018065931A1 (en) 2016-10-05 2018-04-12 Glaxosmithkline Biologicals Sa Vaccine
US11583504B2 (en) 2016-11-08 2023-02-21 Modernatx, Inc. Stabilized formulations of lipid nanoparticles
EP4043031A1 (en) 2016-11-17 2022-08-17 GlaxoSmithKline Biologicals SA Zika viral antigen constructs
WO2018091540A1 (en) 2016-11-17 2018-05-24 Glaxosmithkline Biologicals Sa Zika viral antigen constructs
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
EP3808380A1 (en) 2016-12-08 2021-04-21 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
US11203569B2 (en) 2017-03-15 2021-12-21 Modernatx, Inc. Crystal forms of amino lipids
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
WO2018172556A1 (en) 2017-03-24 2018-09-27 Curevac Ag Nucleic acids encoding crispr-associated proteins and uses thereof
WO2018213731A1 (en) 2017-05-18 2018-11-22 Modernatx, Inc. Polynucleotides encoding tethered interleukin-12 (il12) polypeptides and uses thereof
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
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
EP3424524A2 (en) 2017-07-04 2019-01-09 CureVac AG Cancer rna-vaccine
US10988754B2 (en) 2017-07-04 2021-04-27 Cure Vac AG Nucleic acid molecules
WO2019008001A1 (en) 2017-07-04 2019-01-10 Curevac Ag Novel nucleic acid molecules
US11744801B2 (en) 2017-08-31 2023-09-05 Modernatx, Inc. Methods of making lipid nanoparticles
WO2019053056A1 (en) 2017-09-13 2019-03-21 Biontech Cell & Gene Therapies Gmbh Rna replicon for expressing a t cell receptor or an artificial t cell receptor
WO2019053012A1 (en) 2017-09-13 2019-03-21 Biontech Rna Pharmaceuticals Gmbh Rna replicon for reprogramming somatic cells
EP3461497A1 (en) 2017-09-27 2019-04-03 GlaxoSmithKline Biologicals S.A. Viral antigens
WO2019063565A1 (en) 2017-09-27 2019-04-04 Glaxosmithkline Biologicals Sa Viral antigens
WO2019077001A1 (en) 2017-10-19 2019-04-25 Curevac Ag Novel artificial nucleic acid molecules
US11771769B2 (en) 2017-11-10 2023-10-03 Cocoon Biotech Inc. Ocular applications of silk-based products
WO2019104160A2 (en) 2017-11-22 2019-05-31 Modernatx, Inc. Polynucleotides encoding phenylalanine hydroxylase for the treatment of phenylketonuria
WO2019104152A1 (en) 2017-11-22 2019-05-31 Modernatx, Inc. Polynucleotides encoding ornithine transcarbamylase for the treatment of urea cycle disorders
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
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
WO2020035609A2 (en) 2018-08-17 2020-02-20 Glaxosmithkline Biologicals Sa Immunogenic compositions and uses thereof
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
WO2020144295A1 (en) 2019-01-10 2020-07-16 Biontech Rna Pharmaceuticals Gmbh Localized administration of rna molecules for therapy
US11793843B2 (en) 2019-01-10 2023-10-24 Janssen Biotech, Inc. Prostate neoantigens and their uses
US11453639B2 (en) 2019-01-11 2022-09-27 Acuitas Therapeutics, Inc. Lipids for lipid nanoparticle delivery of active agents
WO2020180752A1 (en) 2019-03-01 2020-09-10 Flagship Pioneering Innovations Vi, Llc Polyribonucleotides and cosmetic uses thereof
WO2020180751A1 (en) 2019-03-01 2020-09-10 Flagship Pioneering Innovations Vi, Llc Compositions, methods, and kits for delivery of polyribonucleotides
WO2020227642A1 (en) 2019-05-08 2020-11-12 Modernatx, Inc. Compositions for skin and wounds and methods of use thereof
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
WO2021013798A1 (en) 2019-07-21 2021-01-28 Glaxosmithkline Biologicals Sa Therapeutic viral vaccine
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
EP3819377A1 (en) 2019-11-08 2021-05-12 Justus-Liebig-Universität Gießen Circular rna and uses thereof for inhibiting rna-binding proteins
US11471525B2 (en) 2020-02-04 2022-10-18 Curevac Ag Coronavirus vaccine
US11576966B2 (en) 2020-02-04 2023-02-14 CureVac SE Coronavirus vaccine
US11596686B2 (en) 2020-02-04 2023-03-07 CureVac SE Coronavirus vaccine
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
WO2021209970A1 (en) 2020-04-16 2021-10-21 Glaxosmithkline Biologicals Sa Sars cov-2 spike protein construct
WO2021236930A1 (en) 2020-05-20 2021-11-25 Flagship Pioneering Innovations Vi, Llc Immunogenic compositions and uses thereof
WO2021247507A1 (en) 2020-06-01 2021-12-09 Modernatx, Inc. Phenylalanine hydroxylase variants and uses thereof
WO2021245090A1 (en) 2020-06-04 2021-12-09 BioNTech SE Rna replicon for versatile and efficient gene expression
WO2021245611A1 (en) 2020-06-05 2021-12-09 Glaxosmithkline Biologicals Sa Modified betacoronavirus spike proteins
WO2022002783A1 (en) 2020-06-29 2022-01-06 Glaxosmithkline Biologicals Sa Adjuvants
WO2022023284A1 (en) 2020-07-27 2022-02-03 Anjarium Biosciences Ag Compositions of dna molecules, methods of making therefor, and methods of use thereof
WO2022051629A1 (en) 2020-09-03 2022-03-10 Flagship Pioneering Innovations Vi, Llc Immunogenic compositions and uses thereof
WO2022104131A1 (en) 2020-11-13 2022-05-19 Modernatx, Inc. Polynucleotides encoding cystic fibrosis transmembrane conductance regulator for the treatment of cystic fibrosis
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
US11872280B2 (en) 2020-12-22 2024-01-16 CureVac SE RNA vaccine against SARS-CoV-2 variants
US11918643B2 (en) 2020-12-22 2024-03-05 CureVac SE RNA vaccine against SARS-CoV-2 variants
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
WO2022157153A2 (en) 2021-01-20 2022-07-28 Glaxosmithkline Biologicals Sa Therapeutic viral vaccine
US11524023B2 (en) 2021-02-19 2022-12-13 Modernatx, Inc. Lipid nanoparticle compositions and methods of formulating the same
US11622972B2 (en) 2021-02-19 2023-04-11 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
WO2022204390A1 (en) 2021-03-24 2022-09-29 Modernatx, Inc. Lipid nanoparticles containing polynucleotides encoding phenylalanine hydroxylase 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
WO2022204369A1 (en) 2021-03-24 2022-09-29 Modernatx, Inc. Polynucleotides encoding methylmalonyl-coa mutase for the treatment of methylmalonic acidemia
WO2022200574A1 (en) 2021-03-26 2022-09-29 Glaxosmithkline Biologicals Sa Immunogenic compositions
WO2022200575A1 (en) 2021-03-26 2022-09-29 Glaxosmithkline Biologicals Sa Immunogenic compositions
WO2022223556A1 (en) 2021-04-20 2022-10-27 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
WO2022232289A1 (en) 2021-04-27 2022-11-03 Generation Bio Co. Non-viral dna vectors expressing therapeutic antibodies and uses thereof
WO2022232286A1 (en) 2021-04-27 2022-11-03 Generation Bio Co. Non-viral dna vectors expressing anti-coronavirus antibodies and uses thereof
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
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
WO2023283359A2 (en) 2021-07-07 2023-01-12 Omega Therapeutics, Inc. Compositions and methods for modulating secreted frizzled receptor protein 1 (sfrp1) gene expression
WO2023006999A2 (en) 2021-07-30 2023-02-02 CureVac SE Mrnas for treatment or prophylaxis of liver diseases
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
WO2023056044A1 (en) 2021-10-01 2023-04-06 Modernatx, Inc. Polynucleotides encoding relaxin for the treatment of fibrosis and/or cardiovascular disease
WO2023066874A1 (en) 2021-10-18 2023-04-27 BioNTech SE Methods for determining mutations for increasing modified replicable rna function and related compositions and their use
WO2023066875A1 (en) 2021-10-18 2023-04-27 BioNTech SE Modified replicable rna and related compositions and their use
WO2023081526A1 (en) 2021-11-08 2023-05-11 Orna Therapeutics, Inc. Lipid nanoparticle compositions for delivering circular polynucleotides
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
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
WO2023213378A1 (en) 2022-05-02 2023-11-09 BioNTech SE Replicon compositions and methods of using same for the treatment of diseases
WO2023213783A1 (en) 2022-05-02 2023-11-09 BioNTech SE Replicon compositions and methods of using same for the treatment of diseases
WO2023239756A1 (en) 2022-06-07 2023-12-14 Generation Bio Co. Lipid nanoparticle compositions and uses thereof
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
WO2024018035A1 (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
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
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
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

Also Published As

Publication number Publication date
BR112013004866A2 (en) 2016-06-07
HRP20230185T8 (en) 2023-07-21
EP2611461B1 (en) 2022-03-09
US11759422B2 (en) 2023-09-19
DK2611461T3 (en) 2022-05-16
SI3981427T1 (en) 2022-08-31
EP3970742B1 (en) 2022-05-25
LT3981427T (en) 2022-08-10
DK4066856T3 (en) 2022-12-19
DK3970742T3 (en) 2022-07-04
EP2611461A1 (en) 2013-07-10
HRP20230226T1 (en) 2023-04-14
CN103179984A (en) 2013-06-26
ES2918192T3 (en) 2022-07-14
SI4066856T1 (en) 2023-03-31
DK4066857T3 (en) 2023-02-20
LT4066856T (en) 2023-01-25
LT3970742T (en) 2022-08-10
EP3970742A1 (en) 2022-03-23
EP4066855B1 (en) 2022-12-28
PT4066857T (en) 2023-01-31
PT4066855T (en) 2023-02-28
PT2611461T (en) 2022-05-26
HRP20220835T1 (en) 2022-10-14
SI2611461T1 (en) 2022-08-31
RS63983B1 (en) 2023-03-31
FI4066857T3 (en) 2023-03-20
AU2011295935B2 (en) 2016-02-11
HRP20221023T1 (en) 2022-11-11
EP3981427A1 (en) 2022-04-13
EP4101462A1 (en) 2022-12-14
LT4066855T (en) 2023-03-10
US20130202684A1 (en) 2013-08-08
FI4066855T3 (en) 2023-03-23
RS63367B1 (en) 2022-07-29
RS63949B1 (en) 2023-02-28
EP4066856A1 (en) 2022-10-05
PL2611461T3 (en) 2022-07-04
EP4066857A1 (en) 2022-10-05
EP4066856B1 (en) 2022-12-07
PL4066857T3 (en) 2023-03-20
ES2918649T3 (en) 2022-07-19
HRP20220835T8 (en) 2023-01-06
SI4066855T1 (en) 2023-04-28
PL3981427T3 (en) 2022-09-19
HUE061068T2 (en) 2023-05-28
EP4119155A1 (en) 2023-01-18
HUE058667T2 (en) 2022-09-28
ES2913791T3 (en) 2022-06-06
RS63816B1 (en) 2023-01-31
EP4226940A1 (en) 2023-08-16
LT2611461T (en) 2022-08-10
ES2935009T3 (en) 2023-03-01
PT3970742T (en) 2022-06-27
HUE061043T2 (en) 2023-05-28
RS63329B1 (en) 2022-07-29
JP2013536803A (en) 2013-09-26
ES2938866T3 (en) 2023-04-17
PL4066856T3 (en) 2023-02-27
AU2011295935A1 (en) 2013-04-04
HRP20230185T1 (en) 2023-03-31
EP4147718A1 (en) 2023-03-15
HRP20220695T1 (en) 2022-07-08
EP4226941A1 (en) 2023-08-16
SI4066857T1 (en) 2023-04-28
EP4066855A1 (en) 2022-10-05
PT3981427T (en) 2022-06-27
ES2938327T3 (en) 2023-04-10
US20200230058A1 (en) 2020-07-23
HRP20221533T1 (en) 2023-02-17
EP3981427B1 (en) 2022-05-25
EP4066857B1 (en) 2022-12-21
PL4066855T3 (en) 2023-05-08
DK4066855T3 (en) 2023-02-20
LT4066857T (en) 2023-02-27
MX2013002336A (en) 2013-03-18
JP5911870B2 (en) 2016-04-27
HUE060524T2 (en) 2023-03-28
RU2013114362A (en) 2014-10-10
PL3970742T3 (en) 2022-09-19
CA2809851A1 (en) 2012-03-08
SI3970742T1 (en) 2022-08-31
PT4066856T (en) 2023-01-02
RS63315B1 (en) 2022-07-29
HUE058666T2 (en) 2022-09-28
DK3981427T3 (en) 2022-07-11
HUE058361T2 (en) 2022-07-28
EP3981427B9 (en) 2022-07-27

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
AU2018204178B2 (en) Small liposomes for delivery of immunogen-encoding RNA
EP2611420B1 (en) Lipids suitable for liposomal delivery of protein-coding rna
EP2750707A1 (en) Pegylated liposomes for delivery of immunogen-encoding rna

Legal Events

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

Ref document number: 11758014

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2013526210

Country of ref document: JP

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2809851

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: MX/A/2013/002336

Country of ref document: MX

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2011758014

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2013114362

Country of ref document: RU

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2011295935

Country of ref document: AU

Date of ref document: 20110831

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 13819077

Country of ref document: US

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112013004866

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 112013004866

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20130228