WO2012031046A2 - Lipids suitable for liposomal delivery of protein-coding rna - Google Patents

Lipids suitable for liposomal delivery of protein-coding rna Download PDF

Info

Publication number
WO2012031046A2
WO2012031046A2 PCT/US2011/050100 US2011050100W WO2012031046A2 WO 2012031046 A2 WO2012031046 A2 WO 2012031046A2 US 2011050100 W US2011050100 W US 2011050100W WO 2012031046 A2 WO2012031046 A2 WO 2012031046A2
Authority
WO
WIPO (PCT)
Prior art keywords
rna
liposome
optionally substituted
liposomes
group
Prior art date
Application number
PCT/US2011/050100
Other languages
French (fr)
Other versions
WO2012031046A3 (en
Inventor
Andrew Geall
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
Application filed by Novartis Ag filed Critical Novartis Ag
Priority to CA2809863A priority Critical patent/CA2809863A1/en
Priority to RU2013114330/10A priority patent/RU2577983C2/en
Priority to CN201180051927.2A priority patent/CN103384515B/en
Priority to EP11763813.0A priority patent/EP2611420B1/en
Priority to BR112013004865A priority patent/BR112013004865A2/en
Priority to US13/819,228 priority patent/US20130189351A1/en
Priority to AU2011295938A priority patent/AU2011295938B2/en
Priority to JP2013526211A priority patent/JP5908477B2/en
Priority to ES11763813T priority patent/ES2727583T3/en
Priority to MX2013002332A priority patent/MX2013002332A/en
Priority to EP19165057.1A priority patent/EP3542789A3/en
Publication of WO2012031046A2 publication Critical patent/WO2012031046A2/en
Publication of WO2012031046A3 publication Critical patent/WO2012031046A3/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
    • A61K9/1271Non-conventional liposomes, e.g. PEGylated liposomes, liposomes coated with polymers
    • A61K9/1272Non-conventional liposomes, e.g. PEGylated liposomes, liposomes coated with polymers with substantial amounts of non-phosphatidyl, i.e. non-acylglycerophosphate, surfactants as bilayer-forming substances, e.g. cationic lipids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • 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
    • 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
    • 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
    • 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
    • A61P31/04Antibacterial agents
    • 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
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • 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
    • 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
    • A61K2039/55Medicinal preparations containing antigens or antibodies characterised by the host/recipient, e.g. newborn with maternal antibodies
    • A61K2039/552Veterinary vaccine
    • 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
    • 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
    • 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

Definitions

  • This invention is in the field of non-viral delivery of RNA to animals.
  • nucleic acids for in vivo expression of encoded proteins is useful for both gene therapy and immunisation.
  • Various approaches to successful delivery 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.
  • RNA is delivered encapsulated within a liposome.
  • the RNA encodes a polypeptide of interest.
  • the liposome includes at least one compound selected from the group consisting of compounds of formula (I) and formula (XI). These liposomes can efficiently deliver RNA for in vivo expression.
  • the invention is particularly useful for immunisation, in which the encoded polypeptide is an immunogen.
  • the invention provides a liposome within which RNA encoding a polypeptide of interest is encapsulated, wherein the liposome includes at least one compound selected from the group consisting of compounds of formula (I) and formula (XI).
  • R 1 and R 2 together with the nitrogen atom to which they are attached form an optionally substituted C3.2o-heterocycloalkyl, C3.2o-heterocycloalkenyl, C 3 . 2 o-heterocycloalkynyl or C5.2o-heteroaryl group; a is absent or optionally substituted C M alkylene;
  • b is absent or optionally substituted Ci ⁇ alkylene
  • c is absent or optionally substituted C ⁇ alkylene
  • X 1 is O or S
  • X 2 is O or S
  • Y 1 is optionally substituted Cio oalkenyl, Cio- 3 oalkynyl, Cio- 3 oheteroalkenyl or C
  • L is absent or is wherein
  • L a is optionally substituted Q-isalkylene, Ci.i 5 alkenylene, Q.isalkynylene, Ci.i5heteroalkylene, d.isheteroalkenylene or Ci.isheteroalkynylene;
  • L b is optionally substituted C6-i 4 arylene or Cs-nheteroarylene;
  • L c is optionally substituted Ci.i 5 alkylene, Ci.i 5 alkenylene, Ci-ualkynylene, Ci.i 5 heteroalkylene, Q. ⁇ heteroalkenylene or Q. ⁇ heteroalkynylene;
  • d is 0 or 1 ;
  • e is 0 or 1 ;
  • f is 0 or 1 ;
  • Y 2 is an optionally substituted steroid.
  • R a is a N-terminal alkylamide
  • z is an integer from 2 to 10;
  • each AA is an amino acid, provided that at least one histidine and at least one cationic amino acid are present;
  • R b is -H or -Nth-
  • the invention also provides a process for preparing a RNA-containing liposome, comprising a step of mixing RNA with a compound selected from the group consisting of compounds of formula (I) and formula (XI), under conditions such that the compounds form a liposome in which the RNA is encapsulated.
  • RNA and the compound may be mixed in the presence of other compounds which also become incorporated into the liposome e.g. further lipids.
  • the invention utilises liposomes within which polypeptide-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 in the prior art include, but are not limited to, dioleoyl trimethylammonium propane (DOTAP), l,2-distearyloxy-N,N-dimethyl-3-aminopropane (DSDMA), 1 ,2-dioleyloxy- N,Ndimethyl-3-aminopropane (DODMA), 1 ,2-dilinoleyloxy-N,N-dimethyl-3-aminopropane (DlinDMA), 1 ,2-dilinolenyloxy-N,N-dimethyl-3-aminopropane (DLenDMA).
  • DOTAP dioleoyl trimethylammonium propane
  • DSDMA l,2-distearyloxy-N,N-dimethyl-3-aminopropane
  • DODMA 1 ,2-dioleyloxy- N,Ndimethyl-3-aminopropane
  • DlinDMA 1 ,
  • Zwitterionic lipids include, but are not limited to, acyl zwitterionic lipids and ether zwitterionic lipids.
  • useful zwitterionic lipids are DPPC, DOPC, DSPC, dodecylphosphocholine, 1,2-dioleoyl-sn-glycero- 3-phosphatidylethanolamine (DOPE), and l,2-diphytanoyl-sn-glycero-3-phosphoethanolamine (DPyPE).
  • DOPE 1,2-dioleoyl-sn-glycero- 3-phosphatidylethanolamine
  • DPyPE l,2-diphytanoyl-sn-glycero-3-phosphoethanolamine
  • the lipids in the liposomes of the invention 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 uns
  • Liposomes of the invention can be formed from a single lipid or, more usually, from a mixture of lipids.
  • a mixture may comprise (i) a mixture of cationic lipids (ii) a mixture of anionic lipids and cationic lipids (iii) 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. Where a mixture of lipids is used, not all of the component lipids in the mixture need to be amphiphilic e.g. one or more amphiphilic lipids can be mixed with cholesterol.
  • Liposomes of the invention comprise at least one compound of formula (I) and/or at least one compound of formula (XI).
  • Preferred liposomes of the invention include a cationic lipid of formula (I). As shown in the examples, such liposomes are particularly useful for in vivo delivery of RNA for protein expression.
  • Other preferred liposomes of the invention include a lipopeptide of formula (XI).
  • a liposome can include both a liposome of formula (I) and a lipopeptide of formula (XI), but usually includes only one of these two classes of cationic compound.
  • a liposome of the invention is formed from a mixture of lipids
  • the proportion of those lipids which have formula (I) or (XI) should be between 20-80% of the total amount of lipids e.g. between 30-70%, or between 40-60%.
  • useful liposomes are shown below in which 40% or 60% of the total lipid is a lipid of formula (I).
  • the remainder can be made of e.g. cholesterol (e.g. 35-50% cholesterol) and/or DMG (optionally PEGylated) and/or DSPC.
  • DMG optionally PEGylated
  • a liposome may include an amphiphilic lipid whose hydrophilic portion is PEGylated (i.e. modified by covalent attachment of a polyethylene glycol). This modification can increase stability and prevent non-specific adsorption of the liposomes.
  • lipids can be conjugated to PEG using techniques such as those disclosed in reference 2 and 3.
  • Useful PEGylated lipids include PEG- DMG and the lipids of reference 8's formula (XI).
  • PEG provides the liposomes with a coat which can confer favourable pharmacokinetic characteristics.
  • Various lengths of PEG can be used e.g. between 0.5-8kDa.
  • Useful mixtures of lipids, for forming liposomes of the invention comprise: a cationic lipid of formula (I); cholesterol; and a PEGylated lipid, such as PEG-DMG i.e. PEG-conjugated 1,2- dimyristoyl-sn-glycero-3-phosphoethanolamine-N-[methoxy(polyethylene glycol).
  • PEG-DMG i.e. PEG-conjugated 1,2- dimyristoyl-sn-glycero-3-phosphoethanolamine-N-[methoxy(polyethylene glycol).
  • This mixture may also include a neutral zwitterionic lipid, such as DSPC (l ,2-Diastearoyl-sn-glycero-3- phosphocholine) or DPyPE.
  • DSPC l ,2-Diastearoyl-sn-glycero-3- phosphocholine
  • DPyPE DPyPE
  • 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. Diameters in a population can be measured using dynamic light scattering. 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.
  • 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.
  • Cationic lipids of formula (I) are as follows:
  • R 1 and R 2 together with the nitrogen atom to which they are attached form an optionally substituted C 3 . 20 -heterocycloalkyl, C 3 . 20 -heterocycloalkenyl, C 3 . 20 -heterocycloalkynyl or C 5 . 2 o-heteroaryl group;
  • a is absent or optionally substituted C ⁇ alkylene
  • b is absent or optionally substituted C ⁇ alkylene
  • c is absent or optionally substituted CM alkylene
  • X 1 is O or S
  • X 2 is O or S
  • Y 1 is optionally substituted Ci 0 - 3 oalkenyl, Ci 0 - 3 oalkynyl, C 10 - 3 oheteroalkenyl or C io- 3 oheteroalkynyl ;
  • L is absent or is -(L a ) d -(L b ) e -(L c )r-, wherein
  • L a is optionally substituted Ci. ⁇ alkylene, Ci_i 5 alkenylene, Cj.isalkynylene, Ci.iiheteroalkylene, Ci.isheteroalkenylene or
  • L b is optionally substituted Ce-narylene or C5_i 3 heteroarylene
  • L c is optionally substituted Ci-isalkylene, Ci.i 5 alkenylene, Q.isalkynylene, Ci.i5heteroalkylene, C M5 heteroalkenylene or Ci-isheteroalkynylene;
  • d is 0 or 1 ;
  • Y 2 is an optionally substituted steroid.
  • R 1 and R 2 together with the nitrogen atom to which they are attached, form a cyclic "headgroup" with a tertiary amine.
  • the compounds of formula (I) have formula (II):
  • R 1 and R 2 together with the nitrogen atom to which they are attached form an optionally substituted C 3 . 2 o-heterocycloalkyl, C 3 . 2 o-heterocycloalkenyl, C 3 . 2 o-heterocycloalkynyl or C 5 . 2 o-heteroaryl group;
  • a is absent or optionally substituted CM alkylene
  • b is absent or optionally substituted C alkylene
  • c is absent or optionally substituted CM alkylene
  • X' is O or S
  • X 2 is O or S
  • Y 1 is optionally substituted Ci 0 - 3 oalkenyl, Ci 0 - 3 oalkynyl, C !0 -3oheteroalkenyl or C 10 - 3 oheteroalkynyl ;
  • L is -(L a ) d -(L b ) e -(LV, wherein
  • V is optionally substituted C M 5alkylene, Ci-i 5 alkenylene,
  • L b is optionally substituted Ce-uarylene or Cs.oheteroarylene
  • L c is optionally substituted Ci_i 5 alkylene, C M5 alkenylene, C 5 alkynylene, Cj.isheteroalkylene, Ci-isheteroalkenylene or Ci-isheteroalkynylene;
  • d is 0 or 1 ;
  • e is 0 or 1 ;
  • f is 0 or 1 ;
  • L comprises one or more heteroatoms
  • Y 2 is an optionally substituted steroid.
  • the compounds of formula (I) have formula (III):
  • R 1 and R 2 together with the nitrogen atom to which they are attached form an optionally substituted C 3 .2o-heterocycloalkyl, C 3 .2o-heterocycloalkenyl, C 3 .2o-heterocycloalkynyl or C 5 . 2 o-heteroaryl group;
  • a is methylene
  • X' is O or S
  • X 2 is O or S
  • Y 1 is optionally substituted Ci 0 - 3 oalkenyl, Ci 0 -3oalkynyl, Ci 0 - 3 oheteroalkenyl or C l o- 3 oheteroalkynyl ;
  • L is -(L a )d-(L b )e-(LV, wherein
  • V is optionally substituted Ci.
  • L b is optionally substituted C 6 -i 4 arylene or Cs-nheteroarylene;
  • L c is optionally substituted Ci.i 5 alkylene, Ci.isalkenylene, Q.isalkynylene, C).i5heteroalkylene, Ci.i 5 heteroalkenylene or Ci.isheteroalkynylene;
  • d is 0 or 1 ;
  • e is 0 or 1 ;
  • f is 0 or 1 ;
  • Y 2 is an optionally substituted steroid.
  • the compounds of formula (I) have formula (IV):
  • R and R together with the nitrogen atom to which they are attached form an optionally substituted C3-2o-heterocycloalkyl, C3.2o-heterocycloalkenyl, C3.2o-heterocycloalkynyl or Cs.2o-heteroaryl group;
  • a is methylene
  • X 1 is O or S
  • X 2 is O or S
  • Y 1 is optionally substituted Ci 0 - 3 oalkenyl, Cio oalkynyl, Ci 0 - 3 oheteroalkenyl or C 10 - 3 oheteroalkynyl;
  • L is -(L a ) d -(L b ) e -(L c ) f -, wherein
  • V is optionally substituted Ci-i 5 alkylene, Ci_i 5 alkenylene, Ci-isalkynylene, Ci.i5heteroalkylene, Ci.isheteroalkenylene or Ci.isheteroalkynylene;
  • L b is optionally substituted Ce-narylene or Cs-oheteroarylene
  • V is optionally substituted Ci. 15 alkylene, Ci.i 5 alkenylene, Ci.i 5 alkynylene, Ci.i5heteroalkylene, Ci.isheteroalkenylene or Ci. )5 heteroalkynylene;
  • d is 0 or 1 ;
  • e is 0 or 1 ;
  • f is 0 or 1 ;
  • L comprises one or more heteroatoms
  • Y 2 is an optionally substituted steroid.
  • the compounds of formula (I) have formula (V):
  • R 1 and R 2 together with the nitrogen atom to which they are attached form an optionally substituted C3. 2 o-heterocycloalkyl, C3. 2 o-heterocycloalkenyl, C3_2o-heterocycloalkynyl or C 5 . 2 o-heteroaryl group;
  • a is methylene
  • X 1 is O
  • X 2 is O
  • Y 1 is optionally substituted Ci 0 - 3 oalkenyl, Cio oalkynyl, Ci 0 - 3 oheteroalkenyl or C 10 - 3 oheteroalkynyl ;
  • L is -(L a ) d -(L b ) e -(LV, wherein
  • V is optionally substituted Ci.i 5 alkylene, Ci-isalkenylene, Ci.i 5 alkynylene, C
  • L b is optionally substituted Ce-narylene or Cs-nheteroarylene
  • L c is optionally substituted Ci-i 5 alkylene, Ci.i 5 alkenylene, Ci-i 5 alkynylene, Q.jsheteroalkylene, Ci-isheteroalkenylene or Ci.isheteroalkynylene;
  • d is 0 or 1 ;
  • e is 0 or 1 ;
  • f is 0 or 1 ;
  • L comprises one or more heteroatoms
  • Y 2 is an optionally substituted steroid.
  • the compounds of formula (I) have formula (VI):
  • R 1 and R 2 together with the nitrogen atom to which they are attached form an optionally substituted C 3 _ 2 o-heterocycloalkyl, C 3 . 2 o-heterocycloalkenyl, C 3 . 20 -heterocycloalkynyl or Cs.2o-heteroaryl group;
  • a is methylene
  • X 1 is O
  • X 2 is O
  • Y 1 is optionally substituted Ci 0 - 3 oalkenyl, Ci 0 - 3 oalkynyl, Ci 0 -3oheteroalkenyl or C i o- 3 oheteroalkynyl ;
  • L is -L c -, wherein L c is optionally substituted d.i 5 heteroalkylene, Ci. 15 heteroalkenylene or Ci_i5heteroalkynylene;
  • Y 2 is an optionally substituted steroid.
  • the compounds of formula (I) have formula (VII):
  • R 1 and R 2 together with the nitrogen atom to which they are attached form an optionally substituted C 3 . 2 o-heterocycloalkyl, C 3 _ 20 -heterocycloalkenyl, C 3 . 20 -heterocycloalkynyl or C5. 20 -heteroaryl group;
  • a is methylene
  • X 2 is O
  • Y 1 is an optionally substituted C16-22 alkenyl group
  • L is -L c -, wherein L c is optionally substituted Ci.isheteroalkylene, Ci-isheteroalkenylene or Ci.i 5 heteroalkynylene; and
  • Y 2 is an optionally substituted steroid.
  • the compounds of formula (I) have formula (VIII):
  • R and R together with the nitrogen atom to which they are attached form an optionally substituted C 3 . 2 o-heterocycloalkyl, C 3 . 2 o-heterocycloalkenyl, C 3 . 20 -heterocycloalkynyl or C 5 .2o-heteroaryl group;
  • a is methylene
  • X 1 is O
  • X 2 is O
  • Y 1 is an optionally substituted C16-22 alkenyl group
  • L is -L°- wherein L c is optionally substituted C 5 heteroalkylene, Ci_i 5 heteroalkenylene or Ci.i 5 heteroalkynylene;
  • Y 2 is cholesterol connected through the hydroxy group at the 3-position of the A steroid ring, the hydrogen atom of said hydroxy group being absent.
  • the compounds of formula (I) have formula (IX):
  • R 1 and R 2 together with the nitrogen atom to which they are attached form an optionally substituted C3.2o-heterocycloalkyl, C3.2o-heterocycloalkenyl, C 3 . 2 o-heterocycloalkynyl or C 5 _2o-heteroaryl group;
  • a is methylene
  • X 1 is O or S
  • X 2 is O or S
  • Y 1 is optionally substituted Cio- 3 oalkenyl, Cio- 3 oalkynyl, Cio- 3 oheteroalkenyl or C io- 3 oheteroalkynyl ;
  • L is -(L a ) d -(L b )e-(L c ) f _
  • L a is optionally substituted Ci.i 5 alkylene, Q-isalkynylene, Ci_i 5 heteroalkylene, Ci-isheteroalkenylene or Ci.isheteroalkynylene;
  • L b is optionally substituted C6-i 4 arylene or Cs-nheteroarylene;
  • L c is optionally substituted Ci_i 5 alkylene, Ci.i 5 alkenylene, Ci.i 5 alkynylene, Ci.i 5 heteroalkylene, Ci.i 5 heteroalkenylene or Ci.i 5 heteroalkynylene;
  • d is 0 or 1 ;
  • e is 0 or 1 ;
  • f is 0 or 1 ;
  • L comprises one or more heteroatoms
  • Y 2 is an optionally substituted steroid
  • the pKa of the compound is from about 5.9 to about 7.
  • the compounds of formula (I) have formula (X):
  • R 1 and R 2 together with the nitrogen atom to which they are attached form an optionally substituted C 3 .
  • a is methylene
  • X 1 is O or S
  • X 2 is O or S
  • Y 1 is optionally substituted Cio- 3 oalkenyl, Ci 0 - 3 oalkynyl, Ci 0 - 3 oheteroalkenyl or C
  • L is -(L a ) d -(L b ) e -(L c )r-, wherein
  • L a is optionally substituted Ci.isalkylene, Ci-isalkenylene, Ci.isalkynylene, Q.isheteroalkylene, Ci.
  • L b is optionally substituted Ce-narylene or Cs.nheteroarylene
  • L c is optionally substituted Ci.i 5 alkylene, Ci.i 5 alkenylene, Ci.i 5 alkynylene,
  • d is 0 or 1 ;
  • e is 0 or 1 ;
  • f is 0 or 1 ;
  • L comprises one or more heteroatoms
  • Y 2 is an optionally substituted steroid
  • the pKa of the compound is from about 4.5 to about 6.2.
  • a is optionally substituted C1.2 alkylene. In a further embodiment, a is optionally substituted Ci alkylene.
  • b is optionally substituted C0-2 alkylene. In a further embodiment, b is optionally substituted C ⁇ alkylene.
  • c is absent or is optionally substituted Ci alkylene. In a further embodiment, c is absent.
  • a, b and c are, if present, unsubstituted.
  • R 1 and R 2 together with the nitrogen atom to which they are attached form an optionally substituted C 3 .2o-heterocycloalkyl, C 3 . 2 o-heterocycloalkenyl, C3_2o-heterocycloalkynyl group, C 5 -heteroaryl or C 5 -heteroaryl group.
  • R 1 and R 2 together with the nitrogen atom to which they are attached form an optionally substituted C3.2o-heterocycloalkyl, C3_2o-heterocycloalkenyl or C3_2o-heterocycloalkynyl group.
  • R' and R 2 together with the nitrogen atom to which they are attached form an optionally substituted C 3 . 2 o-heterocycloalkyl group.
  • R 1 and R 2 together with the nitrogen atom to which they are attached form an optionally substituted C5.1 group. In a further embodiment, R 1 and R 2 together with the nitrogen atom to which they are attached form an optionally substituted C5.12 group. In a further embodiment, R 1 and R 2 together with the nitrogen atom to which they are attached form an optionally substituted C5 group, C6 group or C7 group. In a further embodiment, R 1 and R 2 together with the nitrogen atom to which they are attached form an optionally substituted C 5 group or C 6 group.
  • R 1 and R 2 together with the nitrogen atom to which they are attached forms a species which comprises at least one oxygen atom. .
  • R 1 and R 2 together with the nitrogen atom to which they are attached are selected from H 1 to H 48 as provided in Table 1.
  • X 1 is O. In another embodiment, X 2 is O. In a further embodiment, both X 1 and X 2 are O.
  • L comprises at least one heteroatom. This means that the chain which provides a direct link between X 2 and Y 2 has at least one heteroatom or, in other words, that any heteroatom in a substituent on L does not count for these purposes.
  • L- comprises at least one O atom.
  • L comprises at least two heteroatoms. In a further embodiment, L comprises at least two O atoms.
  • L c is optionally substituted Q.isalkylene or Ci_i 5 heteroalkylene. In one embodiment, L c is optionally substituted Cj.isalkylene or Ci.isheteroalkylene and d and e are both 0.
  • L c is selected from one of formulae L c' ' to L C"XX1 ". In one embodiment, L c is selected from one of formulae L C I to L C"X I " and d and e are both 0. L c -' -(CH 2 ) 2 0(CH 2 ) 2 -
  • L c is preferably selected from L c - ⁇ L c"v to L c" TM and L c"ix to L c" xxiii .
  • L c is optionally substituted Ci.isheteroalkylene.
  • L c is an optionally substituted Ci_n group. In a further embodiment, L is an optionally substituted Cj.g group. In a further embodiment, L c is an optionally substituted C 3 . 8 group. In a further embodiment, wherein L c is an optionally substituted C4.7 group. In a further embodiment, L c is an optionally substituted C5, C6 or C7 group.
  • Y 1 is a C 12-28 group. In a further embodiment.Y 1 is a C14.26 group. In a further embodiment, Y 1 is a C 16-24 group. In a further embodiment, Y 1 is a C16-22 group. In a further embodiment, the Y 1 chain is 18, 19, 20 or 21 atoms long.
  • Y 1 is preferably alkenyl or heteroalkenyl.
  • Y 1 has at least one alkene group. In a further embodiment, Y 1 has 1, 2 or 3 alkene groups.
  • Y' has an alkene group at the omega-3 position.
  • Y 1 has an alkene group at the omega-6 position.
  • Y 1 has an alkene group at the omega-9 position.
  • Y 1 has an alkene group at two or three of the omega-3, omega-6 and omega-9 positions.
  • Y 1 is unsaturated at the omega-6 and omega-9 positions.
  • Y 1 is unsaturated at the omega-3, omega-6 and omega-9 positions.
  • Y 1 is unsaturated at the omega-9 position.
  • Y 1 has at least one cis unsaturated alkene group. In a further embodiment, Y 1 has at least two cis unsaturated alkene groups. In a further embodiment, Y 1 has at least three cis unsaturated alkene groups. The at least o'ne cis unsaturated alkene group may be at one, two or three of the omega-3, omega-6 and omega-9 positions. Unsaturation in lipid chains is discussed in MacLachlan et ai, Journal of Controlled Release 107 (2005) 276-287.
  • Y 1 is selected from Y 1"1 to Y 1_V1 as provided in Table 2.
  • Y 2 is linked to L via an oxygen atom on the optionally substituted steroid. In a further embodiment, Y 2 is linked to L via an oxygen atom on the 3-position of the A steroid ring. In a further embodiment Y 2 is a sterol in which the hydrogen atom of the hydroxy group at the 3-position of the A steroid ring has been removed (and the connection to L is through the oxygen atom of said hydroxy group).
  • said sterol is selected from the group consisting of: annasterol; avenasterol; beta-si tosterol; brassicasterol; calciferol; campesterol; chalinosterol; chinasterol; cholestanol; cholesterol; coprostanol; cycloartol; dehydrocholesterol; desmosterol; dihydrocalciferol; dihydrocholesterol; dihydroergosterol; dinosterol; epicholesterol; ergosterol; fucosterol; hexahydrolumisterol; hexaol; hydroxycholesterol; lanosterol; lumisterol; parkeol; poriferasterol; saringosterol; sitostanol; sitosterol; stigmastanol; stigmasterol; weinbersterol; zymosterol; sterol bile acids (such as cholic acid; chenodeoxycholic acid; glycocholic acid; tauroc
  • the sterol is cholesterol.
  • the pKa of a lipid is the pH at which 50% of the lipids are charged, lying halfway between the point where the lipids are completely charged and the point where the lipids are completely uncharged. It can be measured in various ways, but is preferably measured using the method disclosed below.
  • the pKa typically should be measured for the lipid alone rather than for the lipid in the context of a mixture which also includes other lipids e.g. not as performed in reference 2, which looks at the pKa of a SNALP rather than of the individual lipids).
  • the pKa of a lipid is measured in water at standard temperature and pressure using the following technique:
  • 0.3mM solution of fluorescent probe toluene nitrosulphonic acid (TNS) in ethanol methanol 9: 1 is prepared by first making 3mM solution of TNS in methanol and then diluting to 0.3mM with ethanol.
  • An aqueous buffer containing sodium phosphate, sodium citrate sodium acetate and sodium chloride, at the concentrations 20mM, 25mM, 20mM and 150 mM, respectively, is prepared.
  • the buffer is split into eight parts and the pH adjusted either with 12N HC1 or 6N NaOH to 4.44-4.52, 5.27, 6.15-6.21 , 6.57, 7.10-7.20, 7.72-7.80, 8.27-8.33 and 10.47-1 1.12.
  • 400uL of 2mM lipid solution and 800uL of 0.3mM TNS solution are mixed.
  • • 7.5 ⁇ 1_, of probe/lipid mix are added to 242.5 ⁇ _, of buffer in a 1 mL 96 well plate. This is done with all eight buffers.
  • lOOuL of each probe/lipid/buffer mixture is transferred to a 250uL black with clear bottom 96 well plate (e.g. model COSTAR 3904, Corning).
  • Fluorescence of each probe/lipid/buffer mixture is measured (e.g. with a SpectraMax M5 spectrophotometer and SoftMax pro 5.2 software) with 322nm excitation, 431nm emission (auto cutoff at 420nm).
  • lipids having a pKa in the range of 5.0 to 7.6 have a pKa of 5.5 to 6.7 e.g. between 5.6 and 6.8, between 5.6 and 6.3, between 5.6 and 6.0, between 5.5 and 6.2, or between 5.7 and 5.9.
  • the compound may be E0024, E0014, E0052, E01 18, E0083, E001 1 , E0008, E0025, E0026, E0069, E0076, E0077, E0078, E0085 or E0088.
  • the compound may be the lipids shown below which were used in the "RV03" to "RV12" liposomes, or in the "RV15" liposomes.
  • Preferred compounds are E0026, E0069 and E0078.
  • Preferred compounds are the lipids shown below which were used in the "RV05", "RV08" and "RV09” liposomes.
  • a liposome of the invention uses compound "RV02" (structure 'shown below). Except for this substitution, all other aspects of these RV02-containing liposomes are as described elsewhere herein.
  • Compounds of formula (XI) are cationic lipopeptides which comprise a N-terminal alkylamide and from 2 to 10 amino acids.
  • Formula (XI) is:
  • R a is a N-terminal alkylamide z is an integer from 2 to 10;
  • each AA is an amino acid, provided that at least one histidine and at least one cationic amino acid are present;
  • R b is -H, -OH or -NH 2 .
  • R a moiety has formula R c -C(0)- R d - where R c is an C 2 to C 24 alkyl and R d is -H or C, to C 4 alkyl.
  • Suitable R c groups include lauryl ('Lau'; C 12 ) and palmitoyl ('Pal' ; C )6 ).
  • the amide of the R a moiety is attached to an oligopeptide chain of from 2 to 10 amino acids e.g. from 3-8 amino acids.
  • This chain includes at least one (e.g. 1 , 2, 3, 4 or 5) histidine. It also includes at least one cationic amino acid e.g. at least one arginine, lysine or ornithine residue.
  • the inclusion of at least one lysine is preferred, and ideally 2 or 3 lysines. Histidine is included because its side chain is weakly basic and predominantly un-ionized at physiological pH, but is more highly protonated in the weakly acidic environment of the endosome.
  • a cationic amino acid such as lysine or arginine, provides a unit positive charge on the lipopeptide at neutral pH.
  • Useful oligopeptides have amino acid sequence -C-Kj-H j - where: i is 1 , 2 or 3; and j is 1 , 2, 3, 4 or 5.
  • the C-terminus of the oligopeptide chain can be left as -COOH or can instead form an amide.
  • Suitable lipopeptides of formula (XI) thus include, but are not limited to: Lau-(C-K-K-H)-NH 2 , Pal- (C-K-H-H)-NH 2> Pal-(C-K-K-H-H)-NH 2 , Pal-(C-K-K-H-H-H)-NH 2 , Pal-(C-K-K-H-H-H-H)- H 2 , Pal-(C-K-K-H-H-H-H)-NH 2> Pal-(C- -K-K-H-H)-NH 2 and Pal-(C- -K- -H-H)-NH 2 .
  • These and other compounds are disclosed in reference 9, and include:
  • Liposomes of the invention include a RNA molecule which (unlike siRNA, as in reference 2) encodes a polypeptide. After in vivo administration of the particles, RNA is released from the particles and is translated inside a cell to provide the polypeptide 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 which is useful for immunisation purposes.
  • Preferred +-stranded RNAs are self-replicating.
  • a self-replicating RNA molecule (replicon) can, when delivered to a vertebrate cell even without any proteins, lead to the production of multiple daughter RNAs by transcription from itself (via an antisense copy which it generates from itself).
  • a self-replicating RNA molecule is thus typically a +-strand molecule which can be directly translated after delivery to a cell, and this translation provides a RNA-dependent RNA polymerase which then produces both antisense and sense transcripts from the delivered RNA.
  • the delivered RNA leads to the production of multiple daughter RNAs.
  • RNAs may be translated themselves to provide in situ expression of an encoded polypeptide of interest, 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 polypeptide of interest.
  • the overall result of this sequence of transcriptions is a huge amplification of the introduced replicon RNAs and so the encoded polypeptide of interest becomes a major 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 polypeptide of interest. Translation of the subgenomic transcript thus leads to in situ expression of the polypeptide 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 the
  • 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 [10].
  • 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) a polypeptide of interest.
  • the 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 polypeptide of interest, such that the subgenomic transcript encodes that polypeptide 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 a polypeptide of interest.
  • the RNA may have additional ⁇ e.g. downstream) open reading frames e.g. to encode further polypeptides of interest (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.
  • dsRNA double-stranded form
  • RNA molecule useful with the invention can conveniently be prepared by in vitro transcription (rVT).
  • rVT can use a (cDNA) template created and propagated in plasmid form in bacteria, or created synthetically (for example by gene synthesis and/or polymerase chain-reaction (PCR) engineering methods).
  • a DNA-dependent RNA polymerase such as the bacteriophage T7, T3 or SP6 RNA polymerases
  • Appropriate capping and poly-A addition reactions can be used as required (although the replicon's poly-A is usually encoded within the DNA template).
  • RNA polymerases can have stringent requirements for the transcribed 5' nucleotide(s) and in some embodiments these requirements must be matched with the requirements of the encoded replicase, to ensure that the IVT-transcribed RNA can function efficiently as a substrate for its self-encoded replicase.
  • the self-replicating RNA can include (in addition to any 5' cap structure) one or more nucleotides having a modified nucleobase.
  • the RNA can comprise m5C (5-methylcytidine), m5U (5-methyluridine), m6A (N6-methyladenosine), s2U (2-thiouridine), Urn (2'-0-methyluridine), ml A (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-hydroxyisopen
  • 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.
  • 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 of interest. After administration of the liposomes the RNA is translated in vivo and the resulting protein can exert its desired effect e.g. it can elicit an immune response in the recipient, or it can provide a function of interest, such as an enzymatic activity .
  • the RNA molecule can encode a single polypeptide of interest or multiple polypeptides. Multiple polypeptides can be presented as a single polypeptide (fusion polypeptide) or as separate polypeptides. If polypeptides 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 polypeptides may be expressed from a polyprotein that encodes individual polypeptide 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 a polypeptide with a useful in vivo function.
  • 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 but the invention concerns delivery of RNA for in vivo expression of a polypeptide which can provide a useful therapeutic or immunological response.
  • the RNA is not total mouse thymus RNA.
  • the RNA encodes 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 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 13.
  • Streptococcus pneumoniae useful polypeptide immunogens are disclosed in reference 14. 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 15 and 16.
  • 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 17, such as a hemolysin, esxA, esxB, ferrichrome-binding protein (sta006) and/or the staOl 1 lipoprotein.
  • Clostridium tetani the typical immunogen is tetanus toxoid.
  • Cornynebacterium diphtheriae the typical immunogen is diphtheria toxoid.
  • Haemophilus influenzae Useful immunogens include, but are not limited to, the polypeptides disclosed in references 18 and 19.
  • Streptococcus agalactiae useful immunogens include, but are not limited to, the polypeptides disclosed in reference 15.
  • 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 20.
  • LcrE [21] and HtrA [22] are two preferred immunogens.
  • Chlamydia pneumoniae Useful immunogens include, but are not limited to, the polypeptides disclosed in reference 23.
  • Helicobacter pylori Useful immunogens include, but are not limited to, CagA, VacA, NAP, and/or urease [24].
  • 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 (MNEC).
  • UPEC uropathogenic E.coli
  • MNEC meningitis/sepsis-associated E.coli
  • Useful UPEC polypeptide immunogens are disclosed in references 25 and 26.
  • Useful MNEC immunogens are disclosed in reference 27.
  • a useful immunogen for several E.coli types is AcfD [28].
  • Yersinia pestis Useful immunogens include, but are not limited to, those disclosed in references 29 and 30.
  • 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).
  • 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 Aphthoviruses.
  • the enterovirus is a poliovirus e.g. a type 1, type 2 and/or type 3 poliovirus.
  • the enterovirus is an EV71 enterovirus.
  • the enterovirus is a coxsackie A or B virus.
  • Bunyavirus Viral immunogens include, but are not limited to, those derived from an Orthobunyavirus, such as California encephalitis virus, a Phlebovirus, such as Rift Valley Fever virus, or a Nairovirus, such as Crimean-Congo hemorrhagic fever virus.
  • an Orthobunyavirus such as California encephalitis virus, a Phlebovirus, such as Rift Valley Fever virus, or 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.
  • 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).
  • 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 Rhabdovirus such as 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 Herpesvirus 8 (HHV8).
  • HSV Herpes Simplex Viruses
  • VZV Varicella-zoster virus
  • EBV Epstein-Barr virus
  • CMV Cytomegalovirus
  • HHV6 Human Herpesvirus 6
  • HHV7 Human Herpesvirus 7
  • HHV8 Human Herpesvirus 8
  • 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, 1 1 , 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, 11 , 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 Dermatophytres, including: Epidermophyton floccusum, Microsporum audouini, Microsporum canis, Microsporum distortum, Microsporum equinum, Microsporum gypsum, Microsporum nanum, Trichophyton concentricum, Trichophyton equinum. Trichophyton gallinae, Trichophyton gypseum, Trichophyton megnini, Trichophyton mentagrophytes, Trichophyton quinckeanum, Trichophyton rubrum, Trichophyton schoenleini, Trichophyton tonsurans, Trichophyton verrucosum, T. verrucosum var. album, var.
  • the immunogen elicits an immune response against a parasite from the Plasmodium genus, such as P. falciparum, P.vivax, P.malariae or P. ovale.
  • the invention may be used for immunising against malaria.
  • the immunogen elicits an immune response against a parasite from the Caligidae family, particularly those from the Lepeophtheirus and Caligus genera e.g. sea lice such as Lepeophtheirus salmonis or Caligus rogercresseyi.
  • the immunogen elicits an immune response against: pollen allergens (tree-, herb, weed-, and grass pollen allergens); insect or arachnid allergens (inhalant, saliva and venom allergens, e.g. mite allergens, cockroach and midges allergens, hymenopthera venom allergens); animal hair and dandruff allergens (from e.g. dog, cat, horse, rat, mouse, etc.); and food allergens ⁇ e.g. a gliadin).
  • pollen allergens tree-, herb, weed-, and grass pollen allergens
  • insect or arachnid allergens inhalant, saliva and venom allergens, e.g. mite allergens, cockroach and midges allergens, hymenopthera venom allergens
  • animal hair and dandruff allergens from e.g. dog, cat,
  • 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 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.
  • cancer-testis antigens
  • melanoma associated with, e.g., head and neck cancer
  • CIA 0205 associated with, e.g. , bladder cancer
  • HLA-A2-R1701 associated with, e.g., beta catenin
  • TCR associated with, e.g., T-cell non-Hodgkins lymphoma
  • BCR-abl associated with, e.g.
  • 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 associated with, e.g., hepatoma
  • KSA associated with, e.g., colorectal cancer
  • gastrin associated with, e.g., pancreatic and gastric cancer
  • telomerase catalytic protein 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;
  • tumor immunogens include, but are not limited to, pl5, 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, pl85erbB2, 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 27.29VBCAA), CA 195, CA 242, CA-50, CAM43, CD68 ⁇ KP1, CO-029,
  • the RNA encodes a polypeptide which is useful in a gene therapy context.
  • This encoded protein is provided in addition to any polypeptides which are encoded for a RNA's ability to self-replicate.
  • the RNA may encode an enzyme (for example, an enzyme which does not bind to RNA), a cytokine, a transmembrane receptor, an ion channel, a hormone, a blood protein, or an antibody.
  • the RNA preferably encodes a human polypeptide in these categories.
  • Enzymes of interest include, but are not limited to: DNA polymerase alpha, DNA polymerase delta,
  • Cytokines of interest include, but are not limited to: interleukin 1; interleukin 2; interleukin 4; interleukin 6; interleukin 7; interleukin 12; interleukin 17; GM-CSF; G-CSF; TNF-alpha; interferon alpha; interferon beta; interferon gamma; and secretoneurin.
  • Receptors of interest include, but are not limited to: the leptin receptor; the low-density lipoprotein receptor; the bone morphogenetic protein type 2 receptor; the TNF receptor; the gonadotropin- releasing hormone receptor; the dopamine receptor; the somatostatin receptor; the vitamin D receptor; the urokinase plasminogen activator receptor; the transferrin receptor; etc.
  • Ion channels of interest include, but are not limited to: HCN2; HCN4; CFTR; the a-subunit of the Maxi-K channel; KCNQ2; KCNQ3; and Kvl .5.
  • Hormones of interest include, but are not limited to: chorionic gonadotropin; corticotrophin; erythropoietin; glucagons; IGF-1 ; oxytocin; platelet-derived growth factor; calcitonin; follicle- stimulating hormone; luteinizing hormone; thyroid-stimulating hormone; insulin; gonadotropin- releasing hormone; vasopressin; somatostatin; prolactin; adrenocorticotropic hormone; antidiuretic hormone; thyrotropin-releasing hormone; octreotide; human growth hormone; relaxin; growth hormone-releasing hormone; parathyroid hormone; calcitrol; calciferol; atrial-natriuretic peptide; gastrin; secretin; cholecystokinin; leptin; neuropeptide Y; ghrelin; angiotensinogen; dopamine; and thrombopoietin.
  • Blood proteins of interest include, but are not limited to: haemoglobin; fibrinogen; factor VII; factor Vila; factor VIII; factor IX; fibrinogen; thrombin; von Willebrand factor.
  • 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 31.
  • a pharmaceutical composition of the invention may include one or more small molecule immunopotentiators.
  • the composition may include a TLR2 agonist ⁇ e.g. Pam3CSK4), a TLR4 agonist ⁇ e.g. an aminoalkyl glucosaminide phosphate, such as E6020), a TLR7 agonist ⁇ e.g. imiquimod), a TLR8 agonist ⁇ e.g. resiquimod) and/or a TLR9 agonist ⁇ e.g. IC31).
  • 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.
  • 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.
  • 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. O.lmM.
  • 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.
  • 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.
  • 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 ⁇ RNA ⁇ e.g. from 10-100 ⁇ g, such as about 10pg, 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, or for gene therapy.
  • 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 a liposome or pharmaceutical composition of the invention for use in a method of gene therapy in a vertebrate.
  • the invention also provides the use of a liposome of the invention in the manufacture of a medicament for raising an immune response in a vertebrate.
  • the vertebrate By raising an immune response in the vertebrate by these uses and methods, the vertebrate can be protected against various diseases and/or infections e.g. against bacterial and/or viral diseases as discussed above.
  • the liposomes and compositions are immunogenic, and are more preferably vaccine compositions.
  • Vaccines according to the invention may either be prophylactic (i.e. to prevent infection) or therapeutic (i.e. to treat infection), but will typically be prophylactic.
  • the vertebrate is preferably a mammal, such as a human or a large veterinary mammal (e.g. horses, cattle, deer, goats, pigs).
  • the human is preferably a child (e.g. a toddler or infant) or a teenager; where the vaccine is for therapeutic use, the human is preferably a teenager or an adult.
  • a vaccine intended for children may also be administered to adults e.g. to assess safety, dosage, immunogenicity, etc.
  • Vaccines prepared according to the invention may be used to treat both children and adults.
  • a human patient may be less than 1 year old, less than 5 years old, 1-5 years old, 5-15 years old, 15-55 years old, or at least 55 years old.
  • Preferred patients for receiving the vaccines are the elderly (e.g. >50 years old, >60 years old, and preferably >65 years), the young (e.g. ⁇ 5 years old), hospitalised patients, healthcare workers, armed service and military personnel, pregnant women, the chronically ill, or immunodeficient patients.
  • the vaccines are not suitable solely for these groups, however, and may be used more generally in a population.
  • compositions of the invention will generally be administered directly to a patient.
  • Direct delivery may be accomplished by parenteral injection (e.g. subcutaneously, intraperitoneally, intravenously, intramuscularly, intradermally, or to the interstitial space of a tissue; unlike reference 1 , intraglossal injection is not typically used with the present invention).
  • Alternative delivery routes include rectal, oral (e.g. tablet, spray), buccal, sublingual, vaginal, topical, transdermal or transcutaneous, intranasal, ocular, aural, pulmonary or other mucosal administration.
  • Intradermal and intramuscular administration are two preferred routes. Injection may be via a needle (e.g. a hypodermic needle), but needle-free injection may alternatively be used.
  • a typical intramuscular dose is 0.5 ml.
  • the invention may be used to elicit systemic and/or mucosal immunity, preferably to elicit an enhanced systemic and/or mucosal immunity.
  • Dosage can be by a single dose schedule or a multiple dose schedule. Multiple doses may be used in a primary immunisation schedule and/or in a booster immunisation schedule. In a multiple dose schedule the various doses may be given by the same or different routes e.g. a parenteral prime and mucosal boost, a mucosal prime and parenteral boost, etc. Multiple doses will typically be administered at least 1 week apart (e.g. about 2 weeks, about 3 weeks, about 4 weeks, about 6 weeks, about 8 weeks, about 10 weeks, about 12 weeks, about 16 weeks, etc.).
  • 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 (' ⁇ ').
  • 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.
  • halogen includes fluorine, chlorine, bromine and iodine.
  • Alkyl alkylene, alkenyl, alkvnyl, cvcloalkyl etc.
  • 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 ⁇ alkyl, such as methyl, ethyl, n-propyl, i-propyl or t-butyl groups.
  • cycloalkyl includes monovalent, saturated, cyclic hydrocarbyl groups.
  • cycloalkyl is C 3 .iocycloalkyl, 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 C2-ioalkenyl, in another embodiment C2- 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.i 0 cycloalkenyl, e.g. cyclohexenyl or benzocyclohexyl.
  • alkynyl includes monovalent, straight or branched, unsaturated, acyclic hydrocarbyl groups having at least one carbon-carbon triple bond and, in one embodiment, no carbon-carbon double bonds.
  • alkynyl is C 2 _ioalkynyl, in another embodiment C2- 6 alkynyl, in another embodiment C 2-4 alkynyl.
  • cycloalkynyl includes monovalent, partially unsaturated, cyclic hydrocarbyl groups having at least one carbon-carbon triple bond and, in one embodiment, no carbon-carbon double bonds.
  • cycloalkynyl is C3_iocycloalkenyl, in another embodiment Cj.iocycloalkynyl.
  • alkylene includes divalent, straight or branched, saturated, acyclic hydrocarbyl groups.
  • alkylene is Ci.i 0 alkylene, in another embodiment Ci-ealkylene, in another embodiment Ci ⁇ 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 .ioalkenylene, in another embodiment C 2 - 6 alkenylene, in another embodiment C 2 - 4 alkenylene.
  • alkynylene includes divalent, straight or branched, unsaturated, acyclic hydrocarbyl groups having at least one carbon-carbon triple bond and, in one embodiment, no carbon-carbon double bonds.
  • alkynylene is C 2 _ioalkynylene, in another embodiment C 2 _ 6 alkynylene, in another embodiment C 2 ⁇ 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(0) 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) confuse 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-oxathiany], morpholinyl, 1 ,4-dithianyl, piperazinyl, 1,4-azathianyl, oxepanyl, thiepanyl, azepanyl, 1,4-dioxepanyl, 1 ,4-oxathiepanyl, 1 ,4-oxazepanyl, 1,4-di thiepanyl, 1,4-thieazepanyl and 1,4-di a
  • 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-tetrahydropyridinyi 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 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 C6-Ci 4 aryl.
  • aryl groups are monovalent derivatives of aceanthrylene, acenaphthylene, acephenanthrylene, anthracene, azulene, chrysene, coronene, fluoranthene, fluorene, as-indacene, s-indacene, indene, naphthalene, ovalene, perylene, phenalene, phenanthrene, picene, pleiadene, pyrene, pyranthrene and rubicene.
  • arylalkyl means alkyl substituted with an aryl group, e.g. benzyl.
  • arylene includes divalent aromatic, cyclic hydrocarbyl groups, such as phenylene.
  • the arylene groups may be monocyclic or polycyclic fused ring aromatic groups.
  • Preferred arylene are C 6 -Ci 4 arylene.
  • arylene groups are divalent derivatives of aceanthrylene, acenaphthylene, acephenanthrylene, anthracene, azulene, chrysene, coronene, fluoranthene, fluorene, as-indacene, s-indacene, indene, naphthalene, ovalene, perylene, phenalene, phenanthrene, picene, pleiadene, pyrene, pyranthrene and rubicene.
  • heteroaryl includes monovalent, heteroaromatic, cyclic hydrocarbyl groups additionally containing one or more heteroatoms independently selected from O, S, N and NR N , where R N is defined below (and in one embodiment is H or alkyl (e.g. Ci_ 6 alkyl)).
  • the heteroaryl groups may be monocyclic or polycyclic (e.g. bicyclic) fused ring heteroaromatic groups.
  • heteroaryl groups contain 5-13 ring members (preferably 5-10 members) and 1 , 2, 3 or 4 ring heteroatoms independently selected from O, S, N and NR N .
  • a heteroaryl group may be 5, 6, 9 or 10 membered, e.g. 5-membered monocyclic, 6-membered monocyclic, 9-membered fused-ring bicyclic or 10- membered fused-ring bicyclic.
  • Monocyclic heteroaromatic groups include heteroaromatic groups containing 5-6 ring members and 1 , 2, 3 or 4 heteroatoms selected from O, S, N or NR N .
  • 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, pyridazin
  • 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. 6 alkyl)).
  • the heteroarylene groups may be monocyclic or polycyclic (e.g. bicyclic) fused ring heteroaromatic groups.
  • heteroarylene groups contain 5- 13 ring members (preferably 5-10 members) and 1, 2, 3 or 4 ring heteroatoms independently selected from O, S, N and NR .
  • 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).
  • group a, b or c in formula (I) is "absent", what is meant is that a single bond is present instead, i.e. that the two groups either side of group a, b or c are directly bonded to each other.
  • E is replaced by E or E (wherein E cannot be H);
  • ⁇ C-H is replaced by ⁇ N or ⁇ P(0) r ;
  • R N is H or optionally substituted Ci. 6 alkyl, d. 6 heteroalkyl, C 3 . 6 cycloalkyl, C 3 . 6 heterocycloalkyl, C 2 - 6 alkenyl, C 2 - 6 heteroalkenyl, C 3 . 6 cycloalkenyl, C3. 6 heterocycloalkenyl, phenyl, or heteroaryl containing 5 or 6 ring members.
  • R N is preferably H, Ci ⁇ alkyl or C3_ 6 cycloalkyl.
  • q is independently 0, 1 or 2. In one embodiment, q is 0.
  • r is independently 0 or 1. In one embodiment, r is 0.
  • heteroatom containing groups such as heteroalkyl etc.
  • a numerical of carbon atoms is given, for instance C 3 _ 6 heteroalkyl
  • a C3. 6 heteroalkyl group would, for example, contain less than 3-6 chain carbon atoms.
  • a pyridyl group would be classed as a C3 ⁇ 4 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 R S ;
  • R s is independently H or C ⁇ alkyl, Cj ⁇ heteroalkyl, -(Alk a ) r C 3 . 6 cycloalkyl, -(Alk a )rC 3 . 6 heterocycloalkyl, C 2 _ 6 alkenyl, C 2 . 6 heteroalkenyl, -(Alk a ) r C 3 . 6 cycloalkenyl, -(Alk a )rC 3 ⁇ heterocycloalkenyl, C ⁇ alkynyl, C 2 . 6 heteroalkynyl, -(Alk a )rC6-i4aryl,
  • Alk a is or Ci_ 6 heteroalkylene
  • R s is optionally substituted itself (in one embodiment unsubstituted) by 1 to 3 substituents Sub 2 ;
  • Sub 2 is independently halogen, trihalomethyl, trihaloethyl, -N0 2 , -CN, -N + (Ci_ 6 alkyl) 2 0 " , -C0 2 H, -C0 2 Ci. 6 alkyl, -S0 3 H,
  • Z 1 is independently O, S, NH or N(Ci. 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 Ci_ 6 alkyl, optionally substituted by 1 to 3 substituents Sub 2 .
  • Sub 1 is not -R s and Sub 2 is not -C 2 . 6 alkenyl, -C 2 - 6 heteroalkenyl, -C 2-6 alkynyl or -C 2-6 heteroalky nyl .
  • a group other than Sub 2 has at least 2 positions which may be substituted
  • the group may be substituted by both ends of an alkylene, alkenylene, alkynylene, heteroalkylene, heteroalkenylene or heteroalkynylene chain (in one embodiment containing 1 to 6 atoms, in a further embodiment 3 to 6 atoms, and in a further embodiment 3 or 4 atoms) to form a cyclic moiety.
  • That chain is optionally substituted by 1 to 3 substituents Sub 2 . In one embodiment that chain is not substituted.
  • cycloalkyl cycloalkenyl
  • cycloalkynyl cycloalkenyl
  • heterocycloalkyl cycloalkenyl
  • heterocycloalkynyl cycloalkynyl
  • aryl cycloaryl
  • heteroaryl includes a species in which a heterocycloalkyl ring is fused to the aromatic ring (e.g. 5,6,7,8-tetrahydro-l ,8-naphthyridin-2-yl).
  • a group other than Sub 2 has an atom which may be substituted twice, that atom may be substituted by both ends of an alkylene, alkenylene, alkynylene, heteroalkylene, heteroalkenylene or heteroalkynylene chain (in one embodiment containing 2 to 8 atoms, in a further embodiment 3 to 6 atoms, and in a further embodiment 4 or 5 atoms) to form a cyclic moiety. That chain is optionally substituted by 1 to 3 substituents Sub 2 . In one embodiment that chain is not substituted.
  • cycloalkyl examples include spiro species.
  • cycloalkenyl when a group has a heteroatom, a substituent may be bonded to the heteroatom.
  • “optionally substituted heteroalkyl” includes -CH 2 -N(Sub')-CH 2 -, -CH(Sub')-NH-CH 2 - and -CH(Sub')-N(Sub')-CH 2 - etc.
  • the phrase "optionally substituted C3_2o-heterocycloalkyl, C3-2o-heterocycloalkenyl, C3.2 ⁇ rheterocycloalkynyl or C 5 . 2 o-heteroaryl group” means that each of the four items in the list, namely the C3_2o-heterocycloalkyl group, the C3.2o-heterocycloalkenyl group, the C3_2o-heterocycloalkynyl group and the C6-2o-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, 15//-cyclopenta[a]phenanthrene:
  • 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”.
  • such 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.
  • the term "about” in relation to a numerical value x is optional and means, for example, ⁇ 10 ⁇ .
  • 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: 1 1849.
  • 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[39].
  • RLR-1 also known as RIG-I or retinoic acid inducible gene I
  • RLR-1 has two caspase recruitment domains near its N-terminus.
  • the approved HGNC name for the gene encoding the RLR- 1 helicase is "DDX58" (for DEAD (Asp-Glu-Ala-Asp) box polypeptide 58) and the unique HGNC ID is HGNC: 19102.
  • the RefSeq sequence for the human RLR-1 gene is GI:77732514.
  • RLR-2 (also known as MDA5 or melanoma differentiation-associated gene 5) also has two caspase recruitment domains near its N-terminus.
  • the approved HGNC name for the gene encoding the RLR-2 helicase is "IFIH1" (for interferon induced with helicase C domain 1) and the unique HGNC ID is HGNC: 18873.
  • the RefSeq sequence for the human RLR-2 gene is GI: 27886567.
  • RLR-3 (also known as LGP2 or laboratory of genetics and physiology 2) has no caspase recruitment domains.
  • the approved HGNC name for the gene encoding the RLR-3 helicase is "DHX58" (for DEXH (Asp-Glu- X-His) box polypeptide 58) and the unique HGNC ID is HGNC:29517.
  • the RefSeq sequence for the human RLR-3 gene is GI: 149408121.
  • PKR is a double-stranded RNA-dependent protein kinase. It plays a key role in the innate immune system.
  • EIF2AK2 for eukaryotic translation initiation factor 2-alpha kinase 2
  • HGNC HGNC:9437
  • the RefSeq sequence for the human PKR gene is GI:208431825.
  • FIG. 1 shows a gel with stained RNA. Lanes show (1) markers (2) naked replicon (3) replicon after RNase treatment (4) replicon encapsulated in liposome (5) liposome after RNase treatment (6) liposome treated with RNase then subjected to phenol/chloroform extraction.
  • FIG. 2 is an electron micrograph of liposomes.
  • FIG. 3 shows protein expression (as relative light units, RLU) at day 6 after delivery of RNA in liposomes with various cationic lipids.
  • 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), l ⁇ g naked RNA, and lpg liposome-encapsulated RNA.
  • FIG. 8 shows anti-F IgG titers in animals receiving VRP, l ⁇ g naked RNA, and O. lg or ⁇ 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- ⁇ g), liposome-encapsulated RNA (0.01- ⁇ 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 ⁇ g), or packaged as a virion (VRP, 10 6 IU). Titers in naive mice are also shown. Solid lines show geometric means.
  • FIG. 13 shows intracellular cytokine production after restimulation with synthetic peptides representing the major epitopes in the F protein, 4 weeks after a second dose.
  • the y-axis shows the % cytokine+ of CD8+CD4-.
  • FIG. 14 shows F-specific IgG titers (mean logio titers ⁇ std dev) over 63 days after immunisation of cows at days 0 & 21.
  • 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 Sindbis virus, 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 40.
  • the liposomes were made of 10% DSPC (zwitterionic), 40% DlinDMA (cationic), 48% cholesterol and 2% PEG-conjugated DMG (2kDa PEG). These proportions refer to the % moles in the total liposome.
  • DlinDMA l ,2-dilinoleyloxy-N,N-dimethyl-3-arninopropane
  • DSPC l ,2-Diastearoyl-sn-glycero-3-phosphocholine
  • Cholesterol was obtained from Sigma-Aldrich.
  • PEG-conjugated DMG (1 ,2-dimyristoyl-sn-glycero- 3-phosphoethanolamine-N-[methoxy(polyethylene glycol), ammonium salt), DOTAP (1 ,2-dioleoyl- 3-trimethylammonium-propane, chloride salt) and DC-chol (3 -[N-(N',N'-dimethylaminoethane)- 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 (Zav by intensity) or 78nm (by number).
  • fresh lipid stock solutions in ethanol were prepared.
  • 37 mg of DlinDMA, 11.8 mg of DSPC, 27.8 mg of Cholesterol and 8.07 mg of PEG-conjugated DMG were . weighed and dissolved in 7.55 mL of ethanol.
  • Three different conjugated PEGs were used: PEG- 1000, PEG-2000 or PEG-3000.
  • the freshly prepared lipid stock solution was gently rocked at 37°C for about 15 min to form a homogenous mixture. Then, 226.7 ⁇ L ⁇ of the stock was added to 1.773 mL ethanol to make a working lipid stock solution of 2 mL.
  • RNA was also prepared from a stock solution of ⁇ ⁇ ⁇ ⁇ in 100 mM citrate buffer (pH 6).
  • Three 20 mL glass vials (with stir bars) were rinsed with RNase Away solution and washed with plenty of MilliQ water before use to decontaminate the vials of RNAses.
  • One of the vials was used for the RNA working solution and the others for collecting the lipid and RNA mixes (as described later).
  • the working lipid and RNA solutions were heated at 37°C for 10 min before being loaded into 3cc luer- lok syringes.
  • 2 mL of citrate buffer (pH 6) was loaded in another 3 cc syringe.
  • RNA and the lipids were connected to a T mixer (PEEKTM 500 ⁇ 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. 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).
  • a Mustang Q membrane an anion-exchange support that binds and removes anionic molecules, obtained from Pall Corporation.
  • 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 it. Liposomes were warmed for 10 min at 37°C before passing through the membrane.
  • liposomes were concentrated to 2 mL and dialyzed against 10-15 volumes of IX PBS using by tangential flow filtration before recovering the final product.
  • TFF system and hollow fiber filtration membranes were purchased from Spectrum Labs (Rancho Dominguez) and were used according to the manufacturer's guidelines. Polysulfone hollow fiber filtration membranes with a 100 kD pore size cutoff and 8 cm 2 surface area were used. For in vitro and in vivo experiments 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 IX 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).
  • 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 ⁇ L ⁇ of the stock was added to 1.773 mL ethanol to make a working lipid stock solution of 2 mL.
  • RNA working solution was also prepared from a stock solution of - 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 of 148nm and a polydispersity index of 0.122
  • the chip mixing gave liposomes with a Z average diameter of 97nm and a polydispersity index 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 v/v mixture of sample to 25:24: 1 v/v/v, phenol:chloroform:isoamyl alcohol was then added to extract the RNA from the lipids into the aqueous phase. Samples were mixed by vortexing for a few seconds and then placed on a centrifuge for 15 minutes at 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 4), and no change is seen if these liposomes are treated with RNase (lane 4). After RNase-treated liposomes are subjected to phenol extraction, undigested RNA is seen (lane 6).
  • RNA Even after 1 week at 4°C the RNA could be seen without any fragmentation (FIG. 4, arrow). Protein expression in vivo was unchanged after 6 weeks at 4 °C and one freeze-thaw cycle. Thus liposome-encapsulated RNA is stable.
  • RNA a reporter enzyme SEAP; secreted alkaline phosphatase
  • SEAP secreted alkaline phosphatase
  • Expression levels were measured in sera diluted 1 :4 in 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 leg with O. ⁇ g or ⁇ g RNA dose. The same vector was also administered without the liposomes (in RNase free IX PBS) at ⁇ . Virion-packaged replicons were also tested.
  • Virion-packaged replicons used herein were obtained by the methods of reference 41, 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 ⁇ g dose, and at day 6 expression from a O. ⁇ g encapsulated dose matched levels seen with l ⁇ 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", 0 ⁇ g RNA), or as naked RNA ( ⁇ g).
  • FIG. 10 shows that the lipoplex gave the lowest levels of expression, showing that shows encapsulation is essential for potent expression.
  • FIG. 7 shows anti-F IgG titers 2 weeks after the second dose, and the liposomes clearly enhance immunogenicity.
  • FIG. 8 shows titers 2 weeks later, by which point there was no statistical difference between the encapsulated RNA at O. ⁇ g, the encapsulated RNA at ⁇ g, or the VRP group.
  • Neutralisation titers (measured as 60% plaque reduction, "PRNT60") were not significantly different in these three groups 2 weeks after the second dose (FIG. 9).
  • FIG. 12 shows both IgG and PRNT titers 4 weeks after the second dose.
  • FIG. 13 confirms that the RNA elicits a robust CD8 T cell response.
  • Further experiments compared F-specific IgG titers in mice receiving VRP, O. ⁇ g liposome- encapsulated RNA, or ⁇ g liposome-encapsulated RNA. Titer ratios (VRP: liposome) at various times after the second dose were as follows:
  • liposome-encapsulated RNA induces essentially the same magnitude of immune response as seen with virion delivery.
  • FIG. 1 1 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 l g liposome-encapsulated RNA was statistically insignificant (ANOVA) when compared to VRP, but the higher response seen with 10 ⁇ g liposome- encapsulated RNA was statistically significant (p ⁇ 0.05) when compared to both of these groups.
  • FIG. 14 shows F-specific IgG titers over a 63 day period starting from the first immunisation.
  • the RNA replicon was immunogenic in the cows, 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 vaccine).
  • the cationic lipids of reference 8 are used. These lipids can be synthesised as disclosed in reference 8.
  • the liposomes formed above using DlinDMA are referred to hereafter as the "RV01" series.
  • the DlinDMA was replaced with various cationic lipids in series “RV02" to “RV12” as described below.
  • Two different types of each liposome were formed, using 2% PEG2000-DMG with either (01) 40% of the cationic lipid, 10% DSPC, and 48% cholesterol, or (02) 60% of the cationic lipid and 38% cholesterol.
  • a comparison of the (01) and (02) liposomes shows the effect of the neutral zwitterionic lipid.
  • RV02 liposomes were made using the following cationic lipid:
  • RV03 liposomes were made using the following cationic lipid:
  • RV04 liposomes were made using the following cationic lipid:
  • RV07 liposomes were made using the following cationic lipid:
  • RV08 liposomes were made using the following cationic lipid:
  • RV10 liposomes were made using the following cationic lipid:
  • RV13 liposomes were made using the following cationic lipid (DOTAP, for comparative purposes):
  • RV14 liposomes were made using the following cationic lipid (DC-cholesterol, for comparison):
  • RV15 liposomes were made using the following cationic lipid:
  • liposomes were tested with the SEAP reporter described above.
  • the following table shows the size of the liposomes (Z average and polydispersity index), the % of RNA encapsulation in each liposome, together with the SEAP activity detected at days 1 and 6 after injection.
  • SEAP activity is relative to "RV01(02)" liposomes made from DlinDMA, cholesterol and PEG-DMG:
  • FIG. 3 illustrates the SEAP expression levels seen at day 6. The best results were seen with RV04, RV05, RV07, RV08, RV09, and RV1 1.
  • Liposomes with different lipids were incubated with BHK cells overnight and assessed for protein expression potency. From a baseline with RV05 lipid, expression could be increased 18x by adding 10% l ,2-diphytanoyl-sn-glycero-3-phosphoethanolamine (DPyPE) to the liposome or lOx by adding 10% 18:2 (cis) phosphatidylcholine.
  • DPyPE diphytanoyl-sn-glycero-3-phosphoethanolamine
  • lOx 10% 18:2 (cis) phosphatidylcholine.
  • unsaturated lipid tails tend to enhance IgG titers raised against encoded antigens.
  • the vA317 self-replicating replicon encoding RSV F protein was administered to BALB/c mice, 4 or 8 animals per group, by bilateral intramuscular vaccinations (50 ⁇ L ⁇ per leg) on days 0 and 21 with the replicon ( ⁇ g) alone or formulated as liposomes with RV05 or (for comparison) with RV01 or RV13.
  • the RV01 liposomes had 40% DlinDMA, 10% DSPC, 48% cholesterol and 2% PEG-DMG, but with differing amounts of RNA.
  • the RV05 liposomes had either 40% RV05, 10% DSPC, 48% cholesterol and 2% PEG-DMG or 60% RV05, 38% cholesterol and 2% PEG-DMG.
  • the RV13 liposomes had 40% DOTAP, 10% DOPE, 48% cholesterol and 2% PEG-DMG.
  • the liposomes were prepared using various techniques.
  • naked plasmid DNA (20 ⁇ g) expressing the same RSV-F antigen was delivered either using electroporation or with RV01(10) liposomes (O. ⁇ g DNA).
  • RV01(10) liposomes O. ⁇ g DNA
  • 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:
  • liposome formulations significantly enhanced immunogenicity relative to the naked RNA controls, as determined by increased F-specific IgG titers and T cell frequencies.
  • vA142 encodes the full-length wild type surface fusion (F) glycoprotein of RSV but with the fusion peptide deleted, and the 3' end is formed by ribozyme-mediated cleavage. It was tested in three different mouse strains.
  • mice were given bilateral intramuscular vaccinations (50 ⁇ - per leg) on days 0 and 22. Animals were divided into 8 test groups (5 animals per group) and a naive control (2 animals):
  • Group 1 were given naked replicon (1 ⁇ g).
  • Group 2 were given ⁇ g replicon delivered in liposomes "RV01(37)" with 40% DlinDMA, 10% DSPC, 48% Choi, 2% PEG-conjugated DMG.
  • Group 3 were given the same as group 2, but at O. ⁇ g RNA.
  • Group 4 were ⁇ g replicon in "RV05(11)" liposomes (40% RV05 lipid, 30% 18:2 PE (DLoPE, 28% cholesterol, 2% PEG-DMG).
  • Group 5 were given 5 ⁇ g RSV-F subunit protein adjuvanted with aluminium hydroxide.
  • Group 6 were a naive control (2 animals)
  • F-specific serum IgG GMTs were:
  • F-specific IgGl and IgG2a titers were as follows:
  • RSV serum neutralizing antibody titers at days 35 and 49 were as follows (data are 60% plaque reduction neutralization titers of pools of 2-5 mice, 1 pool per group):
  • VRPs lxlO 6 IU
  • F-specific IgG titers were:
  • F-specific IgGl and IgG2a titers were as follows:
  • RSV serum neutralizing antibody titers at days 35 and 49 were as follows (data are 60% plaque reduction neutralization titers of pools of 2-5 mice, 1 pool per group):
  • F-specific IgG titers were:
  • F-specific IgGl and IgG2a titers were as follows:
  • RSV serum neutralizing antibody titers at days 35 and 49 were as follows: Thus the different lipids (RVOl & RV05; pKa 5.8 & 5.85) were tested in three different inbred mouse strains. For BALB/c and C3H strains RV05 was less effective than RVOl , but it was more effective in B6 strain. In all cases, however, the liposomes were more effective than two cationic nanoemulsions which were tested in parallel.
  • vA142 replicon was also tested in cotton rats using liposomes formed from:
  • VRPs (lxlO 6 IU) expressing the full-length wild type surface F glycoprotein of RSV
  • F-specific serum IgG titers were as follows:
  • RSV serum neutralizing antibody titers were as follows:
  • the protein vaccination at day 49 did not boost antibody titers in cotton rats previously vaccinated with protein, but it provided a large boost to titers in cotton rats previously vaccinated with RNA.
  • the titers (total IgG and neutralization) were higher at day 64 using RV05 than when using RV01.
  • Different cationic lipids with vA317 RSV replicon were higher at day 64 using RV05 than when using RV01.
  • HT, SUV and MLV liposomes were also made with RV01 , using the same components at the same proportions, but with manufacturing methods which are non- scalable (but are quicker). Briefly, an ethanol stock solution was created containing 37mg/ml DLinDMA, 12mg/ml DSPC, 28mg/ml cholesterol, and 8 mg/ml of PEG DMG 2000. ⁇ of the stock solution was diluted in a total of 1 ml of ethanol. Liposomes were prepared by evaporating the ethanol solution using a rotary evaporator at 150 milliTorr, pressure for 30 minutes at 50°C.
  • lipid film was hydrated and dispersed by adding 1.0 mL of filtered deionized water and placed at 50°C to ensure full suspension of the lipids into MLVs. An aliquot was removed from the MLVs and sonicated with a probe sonicator with a 1 second pulse for 5 minutes at 100% power fo form the SUVs. Both of the resulting solutions were complexed with replicon RNA.
  • the HT liposomes were made using an ethanol stock solution containing 37mg/ml DLinDMA, 12mg/ml DSPC, 28mg/ml cholesterol, and 8 mg/ml of PEG DMG 2000.
  • ⁇ of the stock solution was diluted to 400 ⁇ 1 with ethanol.
  • the resulting ethanol solution was added drop wise to 600 ⁇ 1 of lOmM citrate buffer at pH 6.5 containing 4( ⁇ g of RNA under constant stirring.
  • the resulting solution was dialyzed overnight against 4L of PBS buffer using a 10,000 MWCO dialysis membrane.
  • mice 8 per group, were given bilateral intramuscular vaccinations (50 ⁇ L ⁇ per leg) on days 0 and 21 with naked replicon ( ⁇ g) or O. ⁇ g encapsulated RNA.
  • F-specific serum IgG titers (GMT) 2 weeks after these two injections were as follows:
  • Liposome N (with DC-cholesterol) performed poorly, even below the naked RNA control. In contrast, the remaining cationic lipids gave useful results. Liposome O was prepared by a different mixing method (microfluidic chip) from liposome G and this smaller liposome gave better results with approximately the same encapsulation.
  • the RV10 lipid in liposome Q has a pKa of 7.86 which seems too high to be useful in vivo. Even inside the useful pKa range of 5.0 to 7.6, however, although results were good, none of the lipids with one alkyl tail and one steroid-containing tail gave results as good as RV01.
  • aGC a-galactosylceramide
  • a liposome was made with 40% RV05, 10% 18:2 PC, 40% DPyPE, 8% cholesterol and 2% PEG DMG 2000. These liposomes had a Zav diameter of 124.7nm, a pdl of 0.17 and a RNA encapsulation of 61.5%. They were used to vaccinate BALB/c mice as before (O. ⁇ g RNA dose), in comparison with naked RNA ( ⁇ g) or with RV01 -based liposomes (40% DlinDMA, 10% DPSC, 48% cholesterol, 2% PEG DMG 2000).
  • F-specific serum IgG titers were as follows:
  • RV05 liposomes were more immunogenic than naked RNA, but less immunogenic than RV01 liposomes.
  • RV05 gave better results than RV01.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Veterinary Medicine (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Epidemiology (AREA)
  • Virology (AREA)
  • Immunology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Oncology (AREA)
  • Communicable Diseases (AREA)
  • Dispersion Chemistry (AREA)
  • Molecular Biology (AREA)
  • Biophysics (AREA)
  • Pulmonology (AREA)
  • Dermatology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Engineering & Computer Science (AREA)
  • Medicinal Preparation (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Steroid Compounds (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Peptides Or Proteins (AREA)

Abstract

RNA is encapsulated within a liposome for in vivo delivery. The RNA encodes a polypeptide of interest, such as an immunogen for immunisation purposes. The liposome includes at least one compound selected from the group consisting of compounds of formula (I) and formula (XI).

Description

LIPIDS SUITABLE FOR LIPOSOMAL DELIVERY OF PROTEIN-CODING RNA
This application claims the benefit of U.S. provisional application number 61/378,833, 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 to animals.
BACKGROUND ART
The delivery of nucleic acids for in vivo expression of encoded proteins is useful for both gene therapy and immunisation. Various approaches to successful delivery 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 ways of delivering nucleic acids to animals for in vivo expression of their encoded proteins.
DISCLOSURE OF THE INVENTION
According to the invention, RNA is delivered encapsulated within a liposome. The RNA encodes a polypeptide of interest. The liposome includes at least one compound selected from the group consisting of compounds of formula (I) and formula (XI). These liposomes can efficiently deliver RNA for in vivo expression. The invention is particularly useful for immunisation, in which the encoded polypeptide is an immunogen.
Thus the invention provides a liposome within which RNA encoding a polypeptide of interest is encapsulated, wherein the liposome includes at least one compound selected from the group consisting of compounds of formula (I) and formula (XI).
Formula (I) is:
Figure imgf000002_0001
wherein:
R1 and R2 together with the nitrogen atom to which they are attached form an optionally substituted C3.2o-heterocycloalkyl, C3.2o-heterocycloalkenyl, C3.2o-heterocycloalkynyl or C5.2o-heteroaryl group; a is absent or optionally substituted CM alkylene;
b is absent or optionally substituted Ci^ alkylene;
c is absent or optionally substituted C^ alkylene;
X1 is O or S;
X2 is O or S;
Y1 is optionally substituted Cio oalkenyl, Cio-3oalkynyl, Cio-3oheteroalkenyl or C|o-3oheteroalkynyl; '
L is absent or is
Figure imgf000003_0001
wherein
La is optionally substituted Q-isalkylene, Ci.i5alkenylene, Q.isalkynylene, Ci.i5heteroalkylene, d.isheteroalkenylene or Ci.isheteroalkynylene;
Lb is optionally substituted C6-i4arylene or Cs-nheteroarylene;
Lc is optionally substituted Ci.i5alkylene, Ci.i5alkenylene, Ci-ualkynylene, Ci.i5heteroalkylene, Q.^heteroalkenylene or Q.^heteroalkynylene;
d is 0 or 1 ;
e is 0 or 1 ; and
f is 0 or 1 ; and
Y2 is an optionally substituted steroid.
Formula (XI) is:
Ra-(AA)Z-Rb
wherein
Ra is a N-terminal alkylamide;
z is an integer from 2 to 10;
each AA is an amino acid, provided that at least one histidine and at least one cationic amino acid are present;
Rb is -H or -Nth-
The invention also provides a process for preparing a RNA-containing liposome, comprising a step of mixing RNA with a compound selected from the group consisting of compounds of formula (I) and formula (XI), under conditions such that the compounds form a liposome in which the RNA is encapsulated. The RNA and the compound may be mixed in the presence of other compounds which also become incorporated into the liposome e.g. further lipids. The liposome
The invention utilises liposomes within which polypeptide-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 in the prior art include, but are not limited to, dioleoyl trimethylammonium propane (DOTAP), l,2-distearyloxy-N,N-dimethyl-3-aminopropane (DSDMA), 1 ,2-dioleyloxy- N,Ndimethyl-3-aminopropane (DODMA), 1 ,2-dilinoleyloxy-N,N-dimethyl-3-aminopropane (DlinDMA), 1 ,2-dilinolenyloxy-N,N-dimethyl-3-aminopropane (DLenDMA). Zwitterionic lipids include, but are not limited to, acyl zwitterionic lipids and ether zwitterionic lipids. Examples of useful zwitterionic lipids are DPPC, DOPC, DSPC, dodecylphosphocholine, 1,2-dioleoyl-sn-glycero- 3-phosphatidylethanolamine (DOPE), and l,2-diphytanoyl-sn-glycero-3-phosphoethanolamine (DPyPE). The lipids in the liposomes of the invention 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.
Liposomes of the invention can be formed from a single lipid or, more usually, from a mixture of lipids. A mixture may comprise (i) a mixture of cationic lipids (ii) a mixture of anionic lipids and cationic lipids (iii) 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. Where a mixture of lipids is used, not all of the component lipids in the mixture need to be amphiphilic e.g. one or more amphiphilic lipids can be mixed with cholesterol.
Liposomes of the invention comprise at least one compound of formula (I) and/or at least one compound of formula (XI). Preferred liposomes of the invention include a cationic lipid of formula (I). As shown in the examples, such liposomes are particularly useful for in vivo delivery of RNA for protein expression. Other preferred liposomes of the invention include a lipopeptide of formula (XI). A liposome can include both a liposome of formula (I) and a lipopeptide of formula (XI), but usually includes only one of these two classes of cationic compound.
Where a liposome of the invention is formed from a mixture of lipids, it is preferred that the proportion of those lipids which have formula (I) or (XI) should be between 20-80% of the total amount of lipids e.g. between 30-70%, or between 40-60%. For instance, useful liposomes are shown below in which 40% or 60% of the total lipid is a lipid of formula (I). The remainder can be made of e.g. cholesterol (e.g. 35-50% cholesterol) and/or DMG (optionally PEGylated) and/or DSPC. Such mixtures are used below. These percentage values are mole percentages.
A liposome may include an amphiphilic lipid whose hydrophilic portion is PEGylated (i.e. modified by covalent attachment of a polyethylene glycol). This modification can increase stability and prevent non-specific adsorption of the liposomes. For instance, lipids can be conjugated to PEG using techniques such as those disclosed in reference 2 and 3. Useful PEGylated lipids include PEG- DMG and the lipids of reference 8's formula (XI). PEG provides the liposomes with a coat which can confer favourable pharmacokinetic characteristics. Various lengths of PEG can be used e.g. between 0.5-8kDa.
Useful mixtures of lipids, for forming liposomes of the invention, comprise: a cationic lipid of formula (I); cholesterol; and a PEGylated lipid, such as PEG-DMG i.e. PEG-conjugated 1,2- dimyristoyl-sn-glycero-3-phosphoethanolamine-N-[methoxy(polyethylene glycol). This mixture may also include a neutral zwitterionic lipid, such as DSPC (l ,2-Diastearoyl-sn-glycero-3- phosphocholine) or DPyPE. These (and other) mixtures are used in the examples below.
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. Diameters in a population can be measured using dynamic light scattering. 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.
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.
Formula (I)
Cationic lipids of formula (I) are as follows:
Figure imgf000006_0001
wherein:
R1 and R2 together with the nitrogen atom to which they are attached form an optionally substituted C3.20-heterocycloalkyl, C3.20-heterocycloalkenyl, C3.20-heterocycloalkynyl or C5.2o-heteroaryl group;
a is absent or optionally substituted C^ alkylene;
b is absent or optionally substituted C^ alkylene;
c is absent or optionally substituted CM alkylene;
X1 is O or S;
X2 is O or S;
Y1 is optionally substituted Ci0-3oalkenyl, Ci0-3oalkynyl, C10-3oheteroalkenyl or C io-3oheteroalkynyl ;
L is absent or is -(La)d-(Lb)e-(Lc)r-, wherein
La is optionally substituted Ci.^alkylene, Ci_i5alkenylene, Cj.isalkynylene, Ci.iiheteroalkylene, Ci.isheteroalkenylene or
Figure imgf000006_0002
Lb is optionally substituted Ce-narylene or C5_i3heteroarylene;
Lc is optionally substituted Ci-isalkylene, Ci.i5alkenylene, Q.isalkynylene, Ci.i5heteroalkylene, CM5heteroalkenylene or Ci-isheteroalkynylene;
d is 0 or 1 ;
e is 0 or 1 ; and f is 0 or 1 ; and
Y2 is an optionally substituted steroid.
Thus R1 and R2, together with the nitrogen atom to which they are attached, form a cyclic "headgroup" with a tertiary amine. These compounds are described in more detail in reference 8, the complete contents of which are incorporated herein by reference.
In some embodiments the compounds of formula (I) have formula (II):
Figure imgf000007_0001
wherein:
R1 and R2 together with the nitrogen atom to which they are attached form an optionally substituted C3.2o-heterocycloalkyl, C3.2o-heterocycloalkenyl, C3.2o-heterocycloalkynyl or C5.2o-heteroaryl group;
a is absent or optionally substituted CM alkylene;
b is absent or optionally substituted C alkylene;
c is absent or optionally substituted CM alkylene;
X' is O or S;
X2 is O or S;
Y1 is optionally substituted Ci0-3oalkenyl, Ci0-3oalkynyl, C!0-3oheteroalkenyl or C 10-3oheteroalkynyl ;
L is -(La)d-(Lb)e-(LV, wherein
V is optionally substituted CM5alkylene, Ci-i5alkenylene,
Figure imgf000007_0002
Ci_i5heteroalkylene,
Figure imgf000007_0003
or Ci-isheteroalkynylene;
Lb is optionally substituted Ce-uarylene or Cs.oheteroarylene;
Lc is optionally substituted Ci_i5alkylene, CM5alkenylene, C 5alkynylene, Cj.isheteroalkylene, Ci-isheteroalkenylene or Ci-isheteroalkynylene;
d is 0 or 1 ;
e is 0 or 1 ; and
f is 0 or 1 ;
provided that L comprises one or more heteroatoms, and Y2 is an optionally substituted steroid.
In some embodiments the compounds of formula (I) have formula (III):
Figure imgf000008_0001
wherein:
R1 and R2 together with the nitrogen atom to which they are attached form an optionally substituted C3.2o-heterocycloalkyl, C3.2o-heterocycloalkenyl, C3.2o-heterocycloalkynyl or C5.2o-heteroaryl group;
a is methylene;
b is methylene;
c is absent;
X' is O or S;
X2 is O or S;
Y1 is optionally substituted Ci0-3oalkenyl, Ci0-3oalkynyl, Ci0-3oheteroalkenyl or C l o-3oheteroalkynyl ;
L is -(La)d-(Lb)e-(LV, wherein
V is optionally substituted Ci.|5alkylene, Ci.isalkenylene, CM5alkynylene, Ci.i5heteroalkylene, Ci.isheteroalkenylene or Ci.isheteroalkynylene;
Lb is optionally substituted C6-i4arylene or Cs-nheteroarylene;
Lc is optionally substituted Ci.i5alkylene, Ci.isalkenylene, Q.isalkynylene, C).i5heteroalkylene, Ci.i5heteroalkenylene or Ci.isheteroalkynylene;
d is 0 or 1 ;
e is 0 or 1 ; and
f is 0 or 1 ; and
Y2 is an optionally substituted steroid.
In some embodiments the compounds of formula (I) have formula (IV):
Figure imgf000009_0001
wherein:
R and R together with the nitrogen atom to which they are attached form an optionally substituted C3-2o-heterocycloalkyl, C3.2o-heterocycloalkenyl, C3.2o-heterocycloalkynyl or Cs.2o-heteroaryl group;
a is methylene;
b is methylene;
c is absent;
X1 is O or S;
X2 is O or S;
Y1 is optionally substituted Ci0-3oalkenyl, Cio oalkynyl, Ci0-3oheteroalkenyl or C10-3oheteroalkynyl;
L is -(La)d-(Lb)e-(Lc)f-, wherein
V is optionally substituted Ci-i5alkylene, Ci_i5alkenylene, Ci-isalkynylene, Ci.i5heteroalkylene, Ci.isheteroalkenylene or Ci.isheteroalkynylene;
Lb is optionally substituted Ce-narylene or Cs-oheteroarylene;
V is optionally substituted Ci.15alkylene, Ci.i5alkenylene, Ci.i5alkynylene, Ci.i5heteroalkylene, Ci.isheteroalkenylene or Ci.)5heteroalkynylene;
d is 0 or 1 ;
e is 0 or 1 ; and
f is 0 or 1 ;
provided that L comprises one or more heteroatoms, and
Y2 is an optionally substituted steroid.
In some embodiments the compounds of formula (I) have formula (V):
Figure imgf000010_0001
wherein:
R1 and R2 together with the nitrogen atom to which they are attached form an optionally substituted C3.2o-heterocycloalkyl, C3.2o-heterocycloalkenyl, C3_2o-heterocycloalkynyl or C5.2o-heteroaryl group;
a is methylene;
b is methylene;
c is absent;
X1 is O;
X2 is O;
Y1 is optionally substituted Ci0-3oalkenyl, Cio oalkynyl, Ci0-3oheteroalkenyl or C 10-3oheteroalkynyl ;
L is -(La)d-(Lb)e-(LV, wherein
V is optionally substituted Ci.i5alkylene, Ci-isalkenylene, Ci.i5alkynylene, C|.i5heteroalkylene, Ci.isheteroalkenylene or Ci-uheteroalkynylene;
Lb is optionally substituted Ce-narylene or Cs-nheteroarylene;
Lc is optionally substituted Ci-i5alkylene, Ci.i5alkenylene, Ci-i5alkynylene, Q.jsheteroalkylene, Ci-isheteroalkenylene or Ci.isheteroalkynylene;
d is 0 or 1 ;
e is 0 or 1 ; and
f is 0 or 1 ;
provided that L comprises one or more heteroatoms, and
Y2 is an optionally substituted steroid.
In some embodiments the compounds of formula (I) have formula (VI):
Figure imgf000011_0001
wherein:
R1 and R2 together with the nitrogen atom to which they are attached form an optionally substituted C3_2o-heterocycloalkyl, C3.2o-heterocycloalkenyl, C3.20-heterocycloalkynyl or Cs.2o-heteroaryl group;
a is methylene;
b is methylene;
c is absent;
X1 is O;
X2 is O;
Y1 is optionally substituted Ci0-3oalkenyl, Ci0-3oalkynyl, Ci0-3oheteroalkenyl or C i o-3oheteroalkynyl ;
L is -Lc-, wherein Lc is optionally substituted d.i5heteroalkylene, Ci.15heteroalkenylene or Ci_i5heteroalkynylene; and
Y2 is an optionally substituted steroid.
In some embodiments the compounds of formula (I) have formula (VII):
Figure imgf000011_0002
wherein:
R1 and R2 together with the nitrogen atom to which they are attached form an optionally substituted C3.2o-heterocycloalkyl, C3_20-heterocycloalkenyl, C3.20-heterocycloalkynyl or C5.20-heteroaryl group;
a is methylene;
b is methylene;
c is absent; X1 is O;
X2 is O;
Y1 is an optionally substituted C16-22 alkenyl group;
L is -Lc-, wherein Lc is optionally substituted Ci.isheteroalkylene, Ci-isheteroalkenylene or Ci.i5heteroalkynylene; and
Y2 is an optionally substituted steroid.
In some embodiments the compounds of formula (I) have formula (VIII):
Figure imgf000012_0001
wherein:
R and R together with the nitrogen atom to which they are attached form an optionally substituted C3.2o-heterocycloalkyl, C3.2o-heterocycloalkenyl, C3.20-heterocycloalkynyl or C5.2o-heteroaryl group;
a is methylene;
b is methylene;
c is absent;
X1 is O;
X2 is O;
Y1 is an optionally substituted C16-22 alkenyl group;
L is -L°- wherein Lc is optionally substituted C 5heteroalkylene, Ci_i5heteroalkenylene or Ci.i5heteroalkynylene; and
Y2 is cholesterol connected through the hydroxy group at the 3-position of the A steroid ring, the hydrogen atom of said hydroxy group being absent.
In some embodiments the compounds of formula (I) have formula (IX):
Figure imgf000012_0002
(IX) wherein:
R1 and R2 together with the nitrogen atom to which they are attached form an optionally substituted C3.2o-heterocycloalkyl, C3.2o-heterocycloalkenyl, C3.2o-heterocycloalkynyl or C5_2o-heteroaryl group;
a is methylene;
b is methylene;
c is absent;
X1 is O or S;
X2 is O or S;
Y1 is optionally substituted Cio-3oalkenyl, Cio-3oalkynyl, Cio-3oheteroalkenyl or C io-3oheteroalkynyl ;
L is -(La)d-(Lb)e-(Lc)f_ wherein
La is optionally substituted Ci.i5alkylene,
Figure imgf000013_0001
Q-isalkynylene, Ci_i5heteroalkylene, Ci-isheteroalkenylene or Ci.isheteroalkynylene;
Lb is optionally substituted C6-i4arylene or Cs-nheteroarylene;
Lc is optionally substituted Ci_i5alkylene, Ci.i5alkenylene, Ci.i5alkynylene, Ci.i5heteroalkylene, Ci.i5heteroalkenylene or Ci.i5heteroalkynylene;
d is 0 or 1 ;
e is 0 or 1 ; and
f is 0 or 1 ;
provided that L comprises one or more heteroatoms,
Y2 is an optionally substituted steroid; and
the pKa of the compound is from about 5.9 to about 7.
In some embodiments the compounds of formula (I) have formula (X):
Figure imgf000013_0002
wherein: R1 and R2 together with the nitrogen atom to which they are attached form an optionally substituted C3.2o-heterocycloalkyl, C3.2o-heterocycloalkenyl, C3_2o-heterocycloalkynyl or C5.2o-heteroaryl group;
a is methylene;
b is methylene;
c is absent;
X1 is O or S;
X2 is O or S;
Y1 is optionally substituted Cio-3oalkenyl, Ci0-3oalkynyl, Ci0-3oheteroalkenyl or C|0-3oheteroalkynyl;
L is -(La)d-(Lb)e-(Lc)r-, wherein
La is optionally substituted Ci.isalkylene, Ci-isalkenylene, Ci.isalkynylene, Q.isheteroalkylene, Ci.|5heteroalkenylene or Ci.i5heteroalkynylene;
Lb is optionally substituted Ce-narylene or Cs.nheteroarylene;
Lc is optionally substituted Ci.i5alkylene, Ci.i5alkenylene, Ci.i5alkynylene,
Ci.i5heteroalkylene, Q-isheteroalkenylene or Ci.isheteroalkynylene;
d is 0 or 1 ;
e is 0 or 1 ; and
f is 0 or 1 ;
provided that L comprises one or more heteroatoms,
Y2 is an optionally substituted steroid; and
the pKa of the compound is from about 4.5 to about 6.2.
a. b and c
In one embodiment, a is optionally substituted C1.2 alkylene. In a further embodiment, a is optionally substituted Ci alkylene.
In one embodiment, b is optionally substituted C0-2 alkylene. In a further embodiment, b is optionally substituted C\ alkylene.
In one embodiment, c is absent or is optionally substituted Ci alkylene. In a further embodiment, c is absent.
In one embodiment, a, b and c are, if present, unsubstituted.
The headgroup
In one embodiment, R1 and R2 together with the nitrogen atom to which they are attached form an optionally substituted C3.2o-heterocycloalkyl, C3.2o-heterocycloalkenyl, C3_2o-heterocycloalkynyl group, C5-heteroaryl or C5-heteroaryl group. In one embodiment, R1 and R2 together with the nitrogen atom to which they are attached form an optionally substituted C3.2o-heterocycloalkyl, C3_2o-heterocycloalkenyl or C3_2o-heterocycloalkynyl group. In a further embodiment, R' and R2 together with the nitrogen atom to which they are attached form an optionally substituted C3.2o-heterocycloalkyl group.
In one embodiment, R1 and R2 together with the nitrogen atom to which they are attached form an optionally substituted C5.1 group. In a further embodiment, R1 and R2 together with the nitrogen atom to which they are attached form an optionally substituted C5.12 group. In a further embodiment, R1 and R2 together with the nitrogen atom to which they are attached form an optionally substituted C5 group, C6 group or C7 group. In a further embodiment, R1 and R2 together with the nitrogen atom to which they are attached form an optionally substituted C5 group or C6 group.
In one of the preferred embodiments of this invention, R1 and R2 together with the nitrogen atom to which they are attached forms a species which comprises at least one oxygen atom. .
In one embodiment, R1 and R2 together with the nitrogen atom to which they are attached are selected from H1 to H48 as provided in Table 1.
Table 1— Moieties named H1 to H 1
Figure imgf000015_0001
Figure imgf000016_0001
-15-
Figure imgf000017_0001
X' and X2
In one embodiment, X1 is O. In another embodiment, X2 is O. In a further embodiment, both X1 and X2 are O.
Linker
In a preferred embodiment, L comprises at least one heteroatom. This means that the chain which provides a direct link between X2 and Y2 has at least one heteroatom or, in other words, that any heteroatom in a substituent on L does not count for these purposes. In a further embodiment, L- comprises at least one O atom.
In one embodiment, L comprises at least two heteroatoms. In a further embodiment, L comprises at least two O atoms.
In one embodiment, Lc is optionally substituted Q.isalkylene or Ci_i5heteroalkylene. In one embodiment, Lc is optionally substituted Cj.isalkylene or Ci.isheteroalkylene and d and e are both 0.
In one embodiment, Lc is selected from one of formulae Lc'' to LC"XX1". In one embodiment, Lc is selected from one of formulae LC I to LC"X I" and d and e are both 0. Lc-' -(CH2)20(CH2)2-
Lc-" -(CH2)4-
Lc iii -CO(CH2)2CO-
Lc iv -CO-
Lc v -COCH2OCH2CO-
Lc"vi -(CH2)20(CH2)2 HCO-
Lc"vii -(CH2)30(CH2)3-
Lc"viii -(CH2)2-
Lc ix -(CH2)20(CH2)20(CH2)20(CH2)2- c x
Figure imgf000018_0001
Lc xv -(CH2)20(CH2)2OCH(CH3)-
Lc xvi -(CH2)20(CH2)2OC(=0)(CH2)2CO-
Lc-xvii -(CH2)2OC(=0)(CH2)2CO-
Lc-xvi>i -(CH2)20(CH2)2OCO-
Lc-xix -(CH2)2NHC(=0)CH2OCH2C(=0)-
Lc-xx -(CH2)2 HC(=0)(CH2)2C(=0)-
Lc-xxi -(CH2)2NHC(=0)-
Lc-xxii -(CH2)2NHC(=0)CH2NHC(=0)-
Lc-xxi,i .(CH2)2NHC(=0)CH(side-chain-l)NHC(=0)-, wherein side-chain- 1 represents the
\ group N the dashed line representing the bond to the rest of the molecule. Since groups in which L comprises at least one heteroatom are preferred, Lc is preferably selected from Lc-\ Lc"v to Lc"™ and Lc"ix to Lc" xxiii.
In one embodiment, Lc is optionally substituted Ci.isheteroalkylene.
In one embodiment, Lc is an optionally substituted Ci_n group. In a further embodiment, L is an optionally substituted Cj.g group. In a further embodiment, Lc is an optionally substituted C3.8 group. In a further embodiment, wherein Lc is an optionally substituted C4.7 group. In a further embodiment, Lc is an optionally substituted C5, C6 or C7 group.
In a preferred embodiment, d is 0; e is 0, and f is 1. In a preferred embodiment, d is 0; e is 0, and f is 1 and Lc is, within the chain lengths set out above, heteroalkylene.
Ϋ_
In one embodiment, Y1 is a C 12-28 group. In a further embodiment.Y1 is a C14.26 group. In a further embodiment, Y1 is a C 16-24 group. In a further embodiment, Y1 is a C16-22 group. In a further embodiment, the Y1 chain is 18, 19, 20 or 21 atoms long.
Within the carbon ranges set out above, Y1 is preferably alkenyl or heteroalkenyl.
In one embodiment, Y1 has at least one alkene group. In a further embodiment, Y1 has 1, 2 or 3 alkene groups.
In one embodiment, Y' has an alkene group at the omega-3 position. In another embodiment, Y1 has an alkene group at the omega-6 position. In another embodiment, Y1 has an alkene group at the omega-9 position. In a further embodiment, Y1 has an alkene group at two or three of the omega-3, omega-6 and omega-9 positions. In one embodiment, Y1 is unsaturated at the omega-6 and omega-9 positions. In another embodiment, Y1 is unsaturated at the omega-3, omega-6 and omega-9 positions. In one embodiment, Y1 is unsaturated at the omega-9 position.
In one embodiment, Y1 has at least one cis unsaturated alkene group. In a further embodiment, Y1 has at least two cis unsaturated alkene groups. In a further embodiment, Y1 has at least three cis unsaturated alkene groups. The at least o'ne cis unsaturated alkene group may be at one, two or three of the omega-3, omega-6 and omega-9 positions. Unsaturation in lipid chains is discussed in MacLachlan et ai, Journal of Controlled Release 107 (2005) 276-287.
In one embodiment Y1 is selected from Y1"1 to Y1_V1 as provided in Table 2.
Table 2. Yl related Moieties named Y > to Y'~" Name Structure Name Structure
Figure imgf000020_0001
Name Structure Name Structure
Figure imgf000020_0002
Name Structure Name Structure
Figure imgf000020_0003
In one embodiment, Y2 is linked to L via an oxygen atom on the optionally substituted steroid. In a further embodiment, Y2 is linked to L via an oxygen atom on the 3-position of the A steroid ring. In a further embodiment Y2 is a sterol in which the hydrogen atom of the hydroxy group at the 3-position of the A steroid ring has been removed (and the connection to L is through the oxygen atom of said hydroxy group).
In one embodiment said sterol is selected from the group consisting of: annasterol; avenasterol; beta-si tosterol; brassicasterol; calciferol; campesterol; chalinosterol; chinasterol; cholestanol; cholesterol; coprostanol; cycloartenol; dehydrocholesterol; desmosterol; dihydrocalciferol; dihydrocholesterol; dihydroergosterol; dinosterol; epicholesterol; ergosterol; fucosterol; hexahydrolumisterol; hexaol; hydroxycholesterol; lanosterol; lumisterol; parkeol; poriferasterol; saringosterol; sitostanol; sitosterol; stigmastanol; stigmasterol; weinbersterol; zymosterol; sterol bile acids (such as cholic acid; chenodeoxycholic acid; glycocholic acid; taurocholic acid; deoxycholic acid, and lithocholic acid); and salts thereof.
In a further embodiment, the sterol is cholesterol.
pKa
The pKa of a lipid is the pH at which 50% of the lipids are charged, lying halfway between the point where the lipids are completely charged and the point where the lipids are completely uncharged. It can be measured in various ways, but is preferably measured using the method disclosed below. The pKa typically should be measured for the lipid alone rather than for the lipid in the context of a mixture which also includes other lipids e.g. not as performed in reference 2, which looks at the pKa of a SNALP rather than of the individual lipids).
The pKa of a lipid is measured in water at standard temperature and pressure using the following technique:
• 2mM solution of lipid in ethanol is prepared by weighing the lipid and dissolving in ethanol.
0.3mM solution of fluorescent probe toluene nitrosulphonic acid (TNS) in ethanol: methanol 9: 1 is prepared by first making 3mM solution of TNS in methanol and then diluting to 0.3mM with ethanol.
• An aqueous buffer containing sodium phosphate, sodium citrate sodium acetate and sodium chloride, at the concentrations 20mM, 25mM, 20mM and 150 mM, respectively, is prepared. The buffer is split into eight parts and the pH adjusted either with 12N HC1 or 6N NaOH to 4.44-4.52, 5.27, 6.15-6.21 , 6.57, 7.10-7.20, 7.72-7.80, 8.27-8.33 and 10.47-1 1.12. 400uL of 2mM lipid solution and 800uL of 0.3mM TNS solution are mixed. • 7.5μ1_, of probe/lipid mix are added to 242.5μΙ_, of buffer in a 1 mL 96 well plate. This is done with all eight buffers. After mixing, lOOuL of each probe/lipid/buffer mixture is transferred to a 250uL black with clear bottom 96 well plate (e.g. model COSTAR 3904, Corning).
• Fluorescence of each probe/lipid/buffer mixture is measured (e.g. with a SpectraMax M5 spectrophotometer and SoftMax pro 5.2 software) with 322nm excitation, 431nm emission (auto cutoff at 420nm).
• After the measurement, the background fluorescence value of an empty well on the 96 well plate is subtracted from each probe/lipid/buffer mixture. The fluorescence intensity values are then normalized to the value at lowest pH. The normalized fluorescence intensity is then plotted against pH and a line of best fit is provided.
• The point on the line of best fit at which the normalized fluorescence intensity is equal to 0.5 is found. The pH corresponding to normalized fluorescence intensity equal to 0.5 is found and is considered the pKa of the lipid.
The best immunological results are seen with lipids having a pKa in the range of 5.0 to 7.6. Within this pKa range, preferred lipids have a pKa of 5.5 to 6.7 e.g. between 5.6 and 6.8, between 5.6 and 6.3, between 5.6 and 6.0, between 5.5 and 6.2, or between 5.7 and 5.9.
Specific compounds of formula (I)
Specific compounds of formula (I) which are useful with the invention are disclosed in reference 8. For instance, the compound may be E0024, E0014, E0052, E01 18, E0083, E001 1 , E0008, E0025, E0026, E0069, E0076, E0077, E0078, E0085 or E0088. The compound may be the lipids shown below which were used in the "RV03" to "RV12" liposomes, or in the "RV15" liposomes. Preferred compounds are E0026, E0069 and E0078. Preferred compounds are the lipids shown below which were used in the "RV05", "RV08" and "RV09" liposomes.
In an alternative embodiment, rather than use a compound of formula (I), a liposome of the invention uses compound "RV02" (structure 'shown below). Except for this substitution, all other aspects of these RV02-containing liposomes are as described elsewhere herein.
Formula (XI)
Compounds of formula (XI) are cationic lipopeptides which comprise a N-terminal alkylamide and from 2 to 10 amino acids. Formula (XI) is:
Ra-(AA)Z-Rb
wherein
Ra is a N-terminal alkylamide z is an integer from 2 to 10;
each AA is an amino acid, provided that at least one histidine and at least one cationic amino acid are present;
Rb is -H, -OH or -NH2.
The Ra moiety has formula Rc-C(0)- Rd- where Rc is an C2 to C24 alkyl and Rd is -H or C, to C4 alkyl. Suitable Rc groups include lauryl ('Lau'; C12) and palmitoyl ('Pal' ; C)6).
The amide of the Ra moiety is attached to an oligopeptide chain of from 2 to 10 amino acids e.g. from 3-8 amino acids. This chain includes at least one (e.g. 1 , 2, 3, 4 or 5) histidine. It also includes at least one cationic amino acid e.g. at least one arginine, lysine or ornithine residue. The inclusion of at least one lysine is preferred, and ideally 2 or 3 lysines. Histidine is included because its side chain is weakly basic and predominantly un-ionized at physiological pH, but is more highly protonated in the weakly acidic environment of the endosome. A cationic amino acid, such as lysine or arginine, provides a unit positive charge on the lipopeptide at neutral pH. Useful oligopeptides have amino acid sequence -C-Kj-Hj- where: i is 1 , 2 or 3; and j is 1 , 2, 3, 4 or 5.
The C-terminus of the oligopeptide chain can be left as -COOH or can instead form an amide.
Compounds of formula (XI) can be described in terms of their alkyl chain, their amino acid sequence, and their C-terminal group. For instance, the lipopeptide "Lau-(C-K-H-H)-NH2" has a N-terminus lauryl chain, then a cysteine, then lysine, then two histidines, and a C-terminus amine. Suitable lipopeptides of formula (XI) thus include, but are not limited to: Lau-(C-K-K-H)-NH2, Pal- (C-K-H-H)-NH2> Pal-(C-K-K-H-H)-NH2, Pal-(C-K-K-H-H-H)-NH2, Pal-(C-K-K-H-H-H-H)- H2, Pal-(C-K-K-H-H-H-H-H)-NH2> Pal-(C- -K-K-H-H)-NH2 and Pal-(C- -K- -H-H-H)-NH2. These and other compounds are disclosed in reference 9, and include:
Figure imgf000023_0001
H3+ NH3+
The RNA
Liposomes of the invention include a RNA molecule which (unlike siRNA, as in reference 2) encodes a polypeptide. After in vivo administration of the particles, RNA is released from the particles and is translated inside a cell to provide the polypeptide 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 which is useful for immunisation purposes.
Preferred +-stranded RNAs are self-replicating. A self-replicating RNA molecule (replicon) can, when delivered to a vertebrate cell even without any proteins, lead to the production of multiple daughter RNAs by transcription from itself (via an antisense copy which it generates from itself). A self-replicating RNA molecule is thus typically a +-strand molecule which can be directly translated after delivery to a cell, and this translation provides a RNA-dependent RNA polymerase which then produces both antisense and sense transcripts from the delivered RNA. 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 polypeptide of interest, 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 polypeptide of interest. The overall result of this sequence of transcriptions is a huge amplification of the introduced replicon RNAs and so the encoded polypeptide of interest becomes a major 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 polypeptide of interest. Translation of the subgenomic transcript thus leads to in situ expression of the polypeptide 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 [10].
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) a polypeptide of interest. The 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 polypeptide of interest, such that the subgenomic transcript encodes that polypeptide 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 a polypeptide of interest. In some embodiments the RNA may have additional {e.g. downstream) open reading frames e.g. to encode further polypeptides of interest (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.
A RNA molecule useful with the invention can conveniently be prepared by in vitro transcription (rVT). rVT 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-transcribed RNA can function efficiently as a substrate for its self-encoded replicase.
As discussed in reference 11 , 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), Urn (2'-0-methyluridine), ml A (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- trimefhylguanosine); 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-methylaminomethyl uridine); mnm5s2U (5-methylaminomethyl-2- thiouridine); mnm5se2U (5-methylaminomethyl-2-selenouridine); ncm5U (5-carbamoylmethyl uridine); ncm5Um (5-carbamoylmethyl-2'-0-methyluridine); cmnm5U (5- carboxymethylaminomethyluridine); cnmm5Um (5-carboxymethylaminomethyl-2-L- Omethyluridine); 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.
The encoded polypeptide of interest
RNA molecules used with the invention encode a polypeptide of interest. After administration of the liposomes the RNA is translated in vivo and the resulting protein can exert its desired effect e.g. it can elicit an immune response in the recipient, or it can provide a function of interest, such as an enzymatic activity .
The RNA molecule can encode a single polypeptide of interest or multiple polypeptides. Multiple polypeptides can be presented as a single polypeptide (fusion polypeptide) or as separate polypeptides. If polypeptides 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 polypeptides may be expressed from a polyprotein that encodes individual polypeptide fused to a short autocatalytic protease (e.g. foot-and-mouth disease virus 2A protein), or as inteins.
Unlike references 1 and 12, the RNA encodes a polypeptide with a useful in vivo function. 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 but the invention concerns delivery of RNA for in vivo expression of a polypeptide which can provide a useful therapeutic or immunological response. Also, the RNA is not total mouse thymus RNA.
Immunosens
In some embodiments the RNA encodes 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. 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 13.
Streptococcus pneumoniae: useful polypeptide immunogens are disclosed in reference 14. 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 15 and 16.
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 17, such as a hemolysin, esxA, esxB, ferrichrome-binding protein (sta006) and/or the staOl 1 lipoprotein.
Clostridium tetani: the typical immunogen is tetanus toxoid.
Cornynebacterium diphtheriae: the typical immunogen is diphtheria toxoid.
Haemophilus influenzae: Useful immunogens include, but are not limited to, the polypeptides disclosed in references 18 and 19.
Pseudomonas aeruginosa
Streptococcus agalactiae: useful immunogens include, but are not limited to, the polypeptides disclosed in reference 15.
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 20. LcrE [21] and HtrA [22] are two preferred immunogens.
Chlamydia pneumoniae: Useful immunogens include, but are not limited to, the polypeptides disclosed in reference 23.
Helicobacter pylori: Useful immunogens include, but are not limited to, CagA, VacA, NAP, and/or urease [24].
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 (MNEC). Useful UPEC polypeptide immunogens are disclosed in references 25 and 26. Useful MNEC immunogens are disclosed in reference 27. A useful immunogen for several E.coli types is AcfD [28].
Bacillus anthracis
Yersinia pestis: Useful immunogens include, but are not limited to, those disclosed in references 29 and 30.
Staphylococcus epidermis
Clostridium perfringens or Clostridium botulinums
Legionella pneumophila
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 saprophyticus
Yersinia enterocolitica
Mycobacterium tuberculosis
Rickettsia
Listeria monocytogenes
Vibrio cholerae
Salmonella typhi
Borrelia burgdorferi
Porphyromonas gingivalis
Klebsiella
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 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 Orthobunyavirus, 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.
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, 1 1 , 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, 11 , 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 Dermatophytres, including: Epidermophyton floccusum, Microsporum audouini, Microsporum canis, Microsporum distortum, Microsporum equinum, Microsporum gypsum, Microsporum nanum, Trichophyton concentricum, Trichophyton equinum. Trichophyton gallinae, Trichophyton gypseum, Trichophyton megnini, Trichophyton mentagrophytes, Trichophyton quinckeanum, Trichophyton rubrum, Trichophyton schoenleini, Trichophyton tonsurans, Trichophyton verrucosum, T. verrucosum var. album, var. 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, 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 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, pl5, 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, pl85erbB2, 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 27.29VBCAA), CA 195, CA 242, CA-50, CAM43, CD68\KP1, CO-029, FGF-5, Ga733 (EpCAM), HTgp-175, M344, MA- 50, MG7-Ag, MOV18, NB/70K, NY-CO-1, RCAS1, SDCCAG16, TA-90 (Mac-2 binding protein/cyclophilin C-associated protein), TAAL6, TAG72, TLP, TPS, and the like.
Gene therapy
In some embodiments the RNA encodes a polypeptide which is useful in a gene therapy context. This encoded protein is provided in addition to any polypeptides which are encoded for a RNA's ability to self-replicate. Thus the RNA may encode an enzyme (for example, an enzyme which does not bind to RNA), a cytokine, a transmembrane receptor, an ion channel, a hormone, a blood protein, or an antibody. The RNA preferably encodes a human polypeptide in these categories.
Enzymes of interest include, but are not limited to: DNA polymerase alpha, DNA polymerase delta,
Cytokines of interest include, but are not limited to: interleukin 1; interleukin 2; interleukin 4; interleukin 6; interleukin 7; interleukin 12; interleukin 17; GM-CSF; G-CSF; TNF-alpha; interferon alpha; interferon beta; interferon gamma; and secretoneurin.
Receptors of interest include, but are not limited to: the leptin receptor; the low-density lipoprotein receptor; the bone morphogenetic protein type 2 receptor; the TNF receptor; the gonadotropin- releasing hormone receptor; the dopamine receptor; the somatostatin receptor; the vitamin D receptor; the urokinase plasminogen activator receptor; the transferrin receptor; etc.
Ion channels of interest include, but are not limited to: HCN2; HCN4; CFTR; the a-subunit of the Maxi-K channel; KCNQ2; KCNQ3; and Kvl .5.
Hormones of interest include, but are not limited to: chorionic gonadotropin; corticotrophin; erythropoietin; glucagons; IGF-1 ; oxytocin; platelet-derived growth factor; calcitonin; follicle- stimulating hormone; luteinizing hormone; thyroid-stimulating hormone; insulin; gonadotropin- releasing hormone; vasopressin; somatostatin; prolactin; adrenocorticotropic hormone; antidiuretic hormone; thyrotropin-releasing hormone; octreotide; human growth hormone; relaxin; growth hormone-releasing hormone; parathyroid hormone; calcitrol; calciferol; atrial-natriuretic peptide; gastrin; secretin; cholecystokinin; leptin; neuropeptide Y; ghrelin; angiotensinogen; dopamine; and thrombopoietin. Where a hormone requires multiple polypeptide subunits for activity, the RNA may encode one or more of such subunits e.g. the RNA may encode the alpha subunit and/or the beta subunit of follicle-stimulating hormone.
Blood proteins of interest include, but are not limited to: haemoglobin; fibrinogen; factor VII; factor Vila; factor VIII; factor IX; fibrinogen; thrombin; von Willebrand factor.
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 31.
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 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. O.lmM. 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.
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 <100μ RNA {e.g. from 10-100μg, such as about 10pg, 25μg, 50μg, 75μg 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 12, liposomes and pharmaceutical compositions of the invention are for in vivo use for eliciting an immune response against an immunogen of interest, or for gene therapy.
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 a liposome or pharmaceutical composition of the invention for use in a method of gene therapy 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 (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 ('ΈΡΓ'). 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.
Chemical terms and definitions
Halo
The term "halogen" (or "halo") includes fluorine, chlorine, bromine and iodine.
Alkyl. alkylene, alkenyl, alkvnyl, cvcloalkyl 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.
The term "alkyl" includes monovalent, straight or branched, saturated, acyclic hydrocarbyl groups. In one embodiment alkyl is Ci-ioalkyl, in another embodiment
Figure imgf000039_0001
in another embodiment Ci^alkyl, 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.iocycloalkyl, 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.i0cycloalkenyl, 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_ioalkynyl, in another embodiment C2-6alkynyl, in another embodiment C2-4alkynyl.
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_iocycloalkenyl, in another embodiment Cj.iocycloalkynyl.
The term "alkylene" includes divalent, straight or branched, saturated, acyclic hydrocarbyl groups. In one embodiment alkylene is Ci.i0alkylene, in another embodiment Ci-ealkylene, in another embodiment Ci^alkylene, 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.ioalkenylene, in another embodiment C2-6alkenylene, in another embodiment C2-4alkenylene.
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 double bonds. In one embodiment alkynylene is C2_ioalkynylene, in another embodiment C2_6alkynylene, in another embodiment C2^alkynylene.
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(0)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)„ 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-oxathiany], morpholinyl, 1 ,4-dithianyl, piperazinyl, 1,4-azathianyl, oxepanyl, thiepanyl, azepanyl, 1,4-dioxepanyl, 1 ,4-oxathiepanyl, 1 ,4-oxaazepanyl, 1,4-di thiepanyl, 1,4-thieazepanyl and 1,4-di azepanyl. 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-tetrahydropyridinyi 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 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 C6-Ci4aryl.
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 C6-Ci4arylene. 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.
Heteroaryl
The term "heteroaryl" includes monovalent, 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_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 -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 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.
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.6alkyl)). 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 NR . 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).
Absent groups
When group a, b or c in formula (I) is "absent", what is meant is that a single bond is present instead, i.e. that the two groups either side of group a, b or c are directly bonded to each other.
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:
E C E E N E E p
E is replaced by E or E ( wherein E cannot be H);
-CH= is replaced by -N= or -P(0)r=;
≡C-H is replaced by≡N or≡P(0)r; or
-CH2- is replaced by -0-, -S(0)q- -NRN- or -P(0)rRN- where RN is H or optionally substituted Ci.6alkyl, d.6heteroalkyl, C3.6cycloalkyl, C3.6heterocycloalkyl, C2-6alkenyl, C2-6heteroalkenyl, C3.6cycloalkenyl, C3.6heterocycloalkenyl, phenyl, or heteroaryl containing 5 or 6 ring members. RN is preferably H, Ci^alkyl 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 C¾ 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, -NRS 2, -C(=0)NH2, -C(=0)NRs 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, -S02NRs 2, -NRsS02Rs, -N(RS)C(=S)NRS 2, -N(Rs)S02NRs 2, -Rs or -ZSRS, wherein;
Zs is independently O, S or RS;
Rs is independently H or C^alkyl, Cj ^heteroalkyl, -(Alka)rC3.6cycloalkyl, -(Alka)rC3.6heterocycloalkyl, C2_6alkenyl, C2.6heteroalkenyl, -(Alka)rC3.6cycloalkenyl, -(Alka)rC3^heterocycloalkenyl, C^alkynyl, C2.6heteroalkynyl, -(Alka)rC6-i4aryl,
-(Alka)rC6-i4aryl or -(Alka)f-heteroaryl (where heteroaryl contains 5-13 ring members), where f is 0 or 1 ;
Alka is
Figure imgf000045_0001
or Ci_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, -C02Ci.6alkyl, -S03H,
Figure imgf000045_0002
-C(=0)H, -C(=0)C,.6alkyl, -OC(=0)C,^alkyl, =0, -N(C,.6alkyl)2, -C(=0)NH2, -C(=0)N(C,.6alkyl)2) -N(C,.6alkyl)C(=0)0(C1.6alkyl), -N(C,.6alkyl)C(=0)N(C1.6alkyl)2; -OC(=0)N(C,.6alkyl)2,
-N(C1.6alkyl)C(=0)C,.6alkyl, -C(=S)N(C1.6alkyl)2, -N(C1.6alkyl)C(=S)C,.6alkyl, -S02N(C1-6alkyl)2, -N(C , .6alkyl)S02C , -6alkyl, -N(C, .6alkyl)C(=S)N(C ,.6alkyl)2, -N(C i .6alkyl)S02N(C,.6alkyl)2,
-Ci_6alkyl, -Ci.6heteroalkyl, -C3_6cycloalkyl, -C3.6heterocycloalkyl, -C2.6alkenyl, -C2-6heteroalkenyl, -C3.6cycloalkenyl, -C3_6heterocycloalkenyl, -C2.6alkynyl, -C2.6heteroalkynyl, -C6.i4aryl, -C5_i3hetero. aryl, -Z'-Ci.6alkyl, -Z'-C3.6cycloalkyl, -Zt-C2.6alkenyl, -Z'-C3.6cycloalkenyl, or -Z'-C^alkynyl; and
Z1 is independently O, S, NH or N(Ci.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 Ci_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^alkyl 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
Figure imgf000046_0001
-C2.6alkenyl, -C2-6heteroalkenyl, -C2-6alkynyl or -C2-6heteroalky nyl .
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 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')-CH2-, -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, C3.2<rheterocycloalkynyl or C5.2o-heteroaryl group" means that each of the four items in the list, namely the C3_2o-heterocycloalkyl group, the C3.2o-heterocycloalkenyl 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_2o-heterocycloalkynyl" (the first modifier) group and then later the same group is described as a "C5.16" (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 imgf000047_0001
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, 15//-cyclopenta[a]phenanthrene:
Figure imgf000047_0002
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 imgf000048_0001
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.
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 32-38, 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, χ±10Ψο.
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: 1 1849. 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[39]. 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 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 day 6 after delivery of RNA in liposomes with various cationic lipids.
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 = lMg).
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), lμg naked RNA, and lpg liposome-encapsulated RNA.
FIG. 8 shows anti-F IgG titers in animals receiving VRP, lμg naked RNA, and O. lg or ^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-^g), liposome-encapsulated RNA (0.01-^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 ^g), 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 F-specific IgG titers (mean logio titers ± std dev) over 63 days after immunisation of cows at days 0 & 21.
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 sindbis virus, 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.
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.
Encapsulation in DlinDMA-based liposomes
RNA was encapsulated in liposomes made essentially by the method of references 7 and 40. The liposomes were made of 10% DSPC (zwitterionic), 40% DlinDMA (cationic), 48% cholesterol and 2% PEG-conjugated DMG (2kDa PEG). These proportions refer to the % moles in the total liposome. DlinDMA (l ,2-dilinoleyloxy-N,N-dimethyl-3-arninopropane) 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 (3 -[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 (Zav by intensity) or 78nm (by number).
In one particular encapsulation method, fresh lipid stock solutions in ethanol were prepared. 37 mg of DlinDMA, 11.8 mg of DSPC, 27.8 mg of Cholesterol and 8.07 mg of PEG-conjugated DMG were . weighed and dissolved in 7.55 mL of ethanol. Three different conjugated PEGs were used: 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 μL· 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 of RNA was also prepared from a stock solution of ~ Ιμ μΙ^ 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. 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). Before using this membrane for 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 it. 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 by tangential flow filtration before recovering the final product. The TFF system and hollow fiber filtration membranes were purchased from Spectrum Labs (Rancho Dominguez) and were used according to the manufacturer's guidelines. Polysulfone hollow fiber filtration membranes 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.
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 IX 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 μΐ^ 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 μL· 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 -
Figure imgf000053_0001
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 μιη x 105 μπι) 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 of 148nm and a polydispersity index of 0.122, the chip mixing gave liposomes with a Z average diameter of 97nm and a polydispersity index 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 v/v mixture of sample to 25:24: 1 v/v/v, phenol:chloroform:isoamyl alcohol was then added to extract the RNA from the lipids into the aqueous phase. Samples were mixed by vortexing for a few seconds and then placed on a centrifuge for 15 minutes at 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 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 leg with O. ^g or ^g RNA dose. The same vector was also administered without the liposomes (in RNase free IX PBS) at Ιμξ. Virion-packaged replicons were also tested. Virion-packaged replicons used herein (referred to as "VRPs") were obtained by the methods of reference 41, 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 ^g dose, and at day 6 expression from a O. ^g encapsulated dose matched levels seen with lμg 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", 0Λμg RNA), or as naked RNA (^g). 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 l ng RNA (FIG. 6). Further experiments comparing expression from encapsulated and naked replicons indicated that O.O^g encapsulated RNA was equivalent to ^g of naked RNA. At a 0^g dose of RNA the encapsulated material gave a 12-fold higher expression at day 6; at a O. ^g 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.
Further experiments replaced DlinDMA with DOTAP ("RV13"). 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 (Ιμξ), encapsulated in liposomes (0.1 or ^g), 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 O. ^g, the encapsulated RNA at ^g, or the VRP group. Neutralisation titers (measured as 60% plaque reduction, "PRNT60") were not significantly different in these three groups 2 weeks after the second dose (FIG. 9). FIG. 12 shows both IgG and PRNT titers 4 weeks after the second dose.
FIG. 13 confirms that the RNA elicits a robust CD8 T cell response. Further experiments compared F-specific IgG titers in mice receiving VRP, O. ^g liposome- encapsulated RNA, or ^g liposome-encapsulated RNA. Titer ratios (VRP: liposome) at various times after the second dose were as follows:
Figure imgf000056_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 0μg dose, equivalent responses for ^g and O. ^g doses, and a lower response with a O.O^g dose. FIG. 1 1 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 l g liposome-encapsulated RNA was statistically insignificant (ANOVA) when compared to VRP, but the higher response seen with 10μg liposome- encapsulated RNA was statistically significant (p<0.05) when compared to both of these groups.
A further study confirmed that the O. ^g of liposome-encapsulated RNA gave much higher anti-F IgG responses ( 15 days post-second dose) than O. ^g of delivered DNA, and even was more immunogenic than 20μg plasmid DNA encoding the F antigen, delivered by electroporation (Elgen™ DNA Delivery System, Inovio).
A further study was performed in cotton rats (Sigmodon hispidis) instead of mice. At a dose liposome encapsulation increased F-specific IgG titers by 8.3-fold compared to naked RNA and increased neutralisation titers (measured as PR T60) 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.
A large-animal study was performed in cattle. Cows were immunised with 66μg of replicon encoding full-length RSV F protein at days 0 and 21 , formulated inside liposomes. 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). FIG. 14 shows F-specific IgG titers over a 63 day period starting from the first immunisation. The RNA replicon was immunogenic in the cows, 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 vaccine).
Encapsulation in liposomes using alternative cationic lipids
As an alternative to using DlinDMA, the cationic lipids of reference 8 are used. These lipids can be synthesised as disclosed in reference 8. The liposomes formed above using DlinDMA are referred to hereafter as the "RV01" series. The DlinDMA was replaced with various cationic lipids in series "RV02" to "RV12" as described below. Two different types of each liposome were formed, using 2% PEG2000-DMG with either (01) 40% of the cationic lipid, 10% DSPC, and 48% cholesterol, or (02) 60% of the cationic lipid and 38% cholesterol. Thus a comparison of the (01) and (02) liposomes shows the effect of the neutral zwitterionic lipid.
RV02 liposomes were made using the following cationic lipid:
Figure imgf000057_0001
RV03 liposomes were made using the following cationic lipid:
Figure imgf000057_0002
RV04 liposomes were made using the following cationic lipid:
Figure imgf000058_0001
RV07 liposomes were made using the following cationic lipid:
Figure imgf000059_0001
RV08 liposomes were made using the following cationic lipid:
Figure imgf000059_0002
Figure imgf000059_0003
RV10 liposomes were made using the following cationic lipid:
Figure imgf000060_0001
Figure imgf000060_0002
Figure imgf000060_0003
RV13 liposomes were made using the following cationic lipid (DOTAP, for comparative purposes):
Figure imgf000061_0001
RV14 liposomes were made using the following cationic lipid (DC-cholesterol, for comparison):
Figure imgf000061_0002
RV15 liposomes were made using the following cationic lipid:
Figure imgf000061_0003
These liposomes were tested with the SEAP reporter described above. The following table shows the size of the liposomes (Z average and polydispersity index), the % of RNA encapsulation in each liposome, together with the SEAP activity detected at days 1 and 6 after injection. SEAP activity is relative to "RV01(02)" liposomes made from DlinDMA, cholesterol and PEG-DMG:
Figure imgf000061_0004
RV03 (01) 158.3 (0.212) 62.0 65.7 44.9
RV03 (02) 164.2 (0.145) 86 62.2 39.7
RV04 (01) 131.0 (0.145) 74.0 91 154.8
RV04 (02) 134.6 (0.117) 81.5 90.4 142.6
RV05 (01 ) 164.0 (0.162) 76.0 76.9 329.8
RV05 (02) 177.8 (0.1 17) 72.8 67.1 227.9
RV06 (01 ) 1 16.0 (0.180) 79.8 25.5 12.4
RV06 (02) 136.3 (0.164) 74.9 24.8 23.1
RV07 (01) 140.6 (0.184) 77 26.5 163.3
RV07 (02) 138.6 (0.122) 87 29.7 74.8
RV 08 (01) 176.7 (0.185) 50 76.5 187
RV08 (02) 199.5 (0.191) 46.3 82.4 329.8
RV09 (01 ) 165.3 (0.169) 72.2 65.1 453.9
RV09 (02) 179.5 (0.157) 65 68.5 658.2
RV10 (01 ) 129.7 (0.184) 78.4 1 13.4 47.8
RV10 (02) 147.6 (0.131) 80.9 78.2 10.4
RV1 1 (01) 129.2 (0.186) 71 1 13.6 242.2
RVU (02) 139 (0198) 75.2 71.8 187.2
RV12 (01) 135.7 (0.161) 78.8 65 10
RV12 (02) 158.3 (0.287) 69.4 78.8 8.2
FIG. 3 illustrates the SEAP expression levels seen at day 6. The best results were seen with RV04, RV05, RV07, RV08, RV09, and RV1 1.
Various of these liposomes were also used to deliver a replicon encoding full-length RSV F protein. One study compared RV01, RV05 and RV13; the highest F-specific serum IgG titers were seen with RV01 and the lowest with RV13. Another study compared RV01 , RV02, RV04 and RV07; the best results were again seen with RV01 ,with RV07 performing poorly. Another study compared RV01, RV03, RV08, RV09 and RV14; the best results were again seen with RV01 , with RV03 and RV14 performing poorly. Another study compared RV01, RV10, RV1 1 and RV15; the best results were again seen with RV01. Overall, the best results were seen with RV01 , RV05, RV08 and RV09, whereas RV 13 (DOTAP) and RV 14 (DC-cholesterol) were poor.
Thus not all of the liposomes were effective for eliciting immune responses. In general, though, it was observed that the best immunological efficacy was seen when the cationic lipid in the liposomes had a pKa in the range of 5.0 to 7.6, and particularly in the range 5.5 to 6.7, between 5.6 and 6.3, between 5.6 and 6.0, or between 5.7 and 5.9.
BHK expression
Liposomes with different lipids were incubated with BHK cells overnight and assessed for protein expression potency. From a baseline with RV05 lipid, expression could be increased 18x by adding 10% l ,2-diphytanoyl-sn-glycero-3-phosphoethanolamine (DPyPE) to the liposome or lOx by adding 10% 18:2 (cis) phosphatidylcholine. In general, in vivo studies showed that unsaturated lipid tails tend to enhance IgG titers raised against encoded antigens.
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 μL· per leg) on days 0 and 21 with the replicon (^g) alone or formulated as liposomes with RV05 or (for comparison) with RV01 or RV13. The RV01 liposomes had 40% DlinDMA, 10% DSPC, 48% cholesterol and 2% PEG-DMG, but with differing amounts of RNA. The RV05 liposomes had either 40% RV05, 10% DSPC, 48% cholesterol and 2% PEG-DMG or 60% RV05, 38% cholesterol and 2% PEG-DMG. The RV13 liposomes had 40% DOTAP, 10% DOPE, 48% cholesterol and 2% PEG-DMG. The liposomes were prepared using various techniques. For comparison, naked plasmid DNA (20 μg) 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.
Z average particle diameter and polydispersity index were:
Figure imgf000063_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 imgf000063_0002
RV05 (02) 961 6915
RV01 (lO) DNA 5 13
RV13 (02) 644 3616
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:
Figure imgf000064_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.
RSV immunogenicity in different mouse strains
Replicon "vA142" encodes the full-length wild type surface fusion (F) glycoprotein of RSV but with the fusion peptide deleted, and the 3' end is formed by ribozyme-mediated cleavage. It was tested in three different mouse strains.
BALB/c mice were given bilateral intramuscular vaccinations (50 μΐ- per leg) on days 0 and 22. Animals were divided into 8 test groups (5 animals per group) and a naive control (2 animals):
Group 1 were given naked replicon (1 μg).
Group 2 were given μg replicon delivered in liposomes "RV01(37)" with 40% DlinDMA, 10% DSPC, 48% Choi, 2% PEG-conjugated DMG.
Group 3 were given the same as group 2, but at O. ^g RNA.
Group 4 were ^g replicon in "RV05(11)" liposomes (40% RV05 lipid, 30% 18:2 PE (DLoPE, 28% cholesterol, 2% PEG-DMG). Group 5 were given 5μg RSV-F subunit protein adjuvanted with aluminium hydroxide.
Group 6 were a naive control (2 animals)
Sera were collected for antibody analysis on days 14, 35 and 49. F-specific serum IgG GMTs were:
Figure imgf000065_0001
At day 35 F-specific IgGl and IgG2a titers (GMT) were as follows:
Figure imgf000065_0002
RSV serum neutralizing antibody titers at days 35 and 49 were as follows (data are 60% plaque reduction neutralization titers of pools of 2-5 mice, 1 pool per group):
Figure imgf000065_0003
Spleens were harvested at day 49 for T cell analysis. Average net F-specific cytokine-positive T cell frequencies (CD4+ or CD8+) were as follows, showing only figures which were statistically significantly above zero (specific for RSV peptides F51-66, F164-178, F309-323 for CD4+, or for peptides F85-93 and F249-258 for CD8+):
Figure imgf000065_0004
C57BL 6 mice were immunised in the same way, but a 7th group received VRPs (lxlO6 IU) expressing the full-length wild-type surface fusion glycoprotein of RSV (fusion peptide deletion).
Sera were collected for antibody analysis on days 14, 35 & 49. F-specific IgG titers (GMT) were:
Day 1 2 3 4 5 6 7
14 1 140 2133 1026 3045 2975 5 1101
35 1721 5532 3184 9525 39251 5 12139 At day 35 F-specific IgGl and IgG2a titers (GMT) were as follows:
Figure imgf000066_0001
RSV serum neutralizing antibody titers at days 35 and 49 were as follows (data are 60% plaque reduction neutralization titers of pools of 2-5 mice, 1 pool per group):
Figure imgf000066_0002
Spleens were harvested at day 49 for T cell analysis. Average net F-specific cytokine-positive T cell frequencies (CD8+) were as follows, showing only figures which were statistically significantly above zero (specific for RSV peptides F85-93 and F249-258):
Figure imgf000066_0003
Nine groups of C3H/HeN mice were immunised in the same way. F-specific IgG titers (GMT) were:
Figure imgf000066_0004
At day 35 F-specific IgGl and IgG2a titers (GMT) were as follows:
Figure imgf000066_0005
RSV serum neutralizing antibody titers at days 35 and 49 were as follows:
Figure imgf000066_0006
Thus the different lipids (RVOl & RV05; pKa 5.8 & 5.85) were tested in three different inbred mouse strains. For BALB/c and C3H strains RV05 was less effective than RVOl , but it was more effective in B6 strain. In all cases, however, the liposomes were more effective than two cationic nanoemulsions which were tested in parallel.
Cotton rats
The vA142 replicon was also tested in cotton rats using liposomes formed from:
(a) 40% DlinDMA, 10% DPSC, 48% cholesterol and 2% PEG DMG 2000.
(b) 40% RV05, 30% DLoPE (18:2 PE), 28% cholesterol and 2% PEG DMG 2000.
Cotton rats, 4-8 animals per group, were given intramuscular vaccinations (100 μί in one leg) on days 0 and 21 with:
Group 1 self-replicating RNA (vA142, 0.1 μg, RSV-F) formulated in liposomes (a)
Group 2 self-replicating RNA (vA142, 0.1 μg, RSV-F) formulated in liposomes (b)
Group 3 self-replicating RNA (vA142, 1 μg, RSV-F) formulated in liposomes (a)
Group 4 self-replicating RNA (vA142, 1 μg, RSV-F) formulated in liposomes (b)
Group 5 VRPs (lxlO6 IU) expressing the full-length wild type surface F glycoprotein of RSV
Group 6 RSV-F subunit protein vaccine (5 μg) adjuvanted with aluminium hydroxide
Group 7 a naive control (3 animals)
All cotton rats (except group 7) were vaccinated with 5 μg F subunit + aluminium hydroxide on day 49 (four weeks after the second vaccination).
Serum was collected for antibody analysis on days 0, 21, 35, 49, 64.
F-specific serum IgG titers (GMT) were as follows:
Figure imgf000067_0001
RSV serum neutralizing antibody titers were as follows:
Group Day 21 Day 35 Day 49 Day 64
1 26 162 58 1772
2 27 371 163 2449
3 66 788 306 161 4 75 448 201 5733
5 137 2879 1029 1920
6 307 2570 1124 2897
7 10 - - 10
Thus cotton rats were vaccinated with vA142 replicon formulated with RV01 or RV05, and the replicon was given at two doses (1.0 and 0.1 μg). After the first replicon vaccination F-specific serum IgG titers were higher with RV01 than RV05, but neutralization titers were approximately equal. Titers in all groups were boosted by a homologous second vaccination given on day 21. After the second replicon vaccination, F-specific serum IgG titers were again higher with RV01 than with RV05, and RSV neutralization titers generally followed this same trend.
The protein vaccination at day 49 did not boost antibody titers in cotton rats previously vaccinated with protein, but it provided a large boost to titers in cotton rats previously vaccinated with RNA. The titers (total IgG and neutralization) were higher at day 64 using RV05 than when using RV01. Different cationic lipids with vA317 RSV replicon
Further experiments compared four different cationic lipids (RV01 , RV02, RV04 & RV07). All liposomes contained 2% PEG-DMG 2000 but remaining lipid compositions varied. The compositions and physical characteristics were as follows:
Figure imgf000068_0001
For comparison of immunogenicity, HT, SUV and MLV liposomes were also made with RV01 , using the same components at the same proportions, but with manufacturing methods which are non- scalable (but are quicker). Briefly, an ethanol stock solution was created containing 37mg/ml DLinDMA, 12mg/ml DSPC, 28mg/ml cholesterol, and 8 mg/ml of PEG DMG 2000. ΙΟΟμΙ of the stock solution was diluted in a total of 1 ml of ethanol. Liposomes were prepared by evaporating the ethanol solution using a rotary evaporator at 150 milliTorr, pressure for 30 minutes at 50°C. Residual ethanol evaporation was insured by placing the samples overnight under vacuum in a freeze dryer. The lipid film was hydrated and dispersed by adding 1.0 mL of filtered deionized water and placed at 50°C to ensure full suspension of the lipids into MLVs. An aliquot was removed from the MLVs and sonicated with a probe sonicator with a 1 second pulse for 5 minutes at 100% power fo form the SUVs. Both of the resulting solutions were complexed with replicon RNA. The HT liposomes were made using an ethanol stock solution containing 37mg/ml DLinDMA, 12mg/ml DSPC, 28mg/ml cholesterol, and 8 mg/ml of PEG DMG 2000. ΙΟΟμΙ of the stock solution was diluted to 400μ1 with ethanol. The resulting ethanol solution was added drop wise to 600μ1 of lOmM citrate buffer at pH 6.5 containing 4(^g of RNA under constant stirring. The resulting solution was dialyzed overnight against 4L of PBS buffer using a 10,000 MWCO dialysis membrane.
BALB/c mice, 8 per group, were given bilateral intramuscular vaccinations (50 μL· per leg) on days 0 and 21 with naked replicon (^g) or O.^g encapsulated RNA. F-specific serum IgG titers (GMT) 2 weeks after these two injections were as follows:
Figure imgf000069_0001
For RV07 the absence of DSPC caused a large decrease in immunogenicity.
Further lipids (RV01, RV03, RV08, RV09, RV14) were tested in the same way:
Figure imgf000069_0002
Liposomes Day 14 Day 35
Naked A317 RNA 35 457
G 2421 10757
H 15 52 J 991 1921
K 7 610
L 1082 1421
M 146 286
N 27 212
0 4695 19773
Liposome N (with DC-cholesterol) performed poorly, even below the naked RNA control. In contrast, the remaining cationic lipids gave useful results. Liposome O was prepared by a different mixing method (microfluidic chip) from liposome G and this smaller liposome gave better results with approximately the same encapsulation.
Further lipids (RV01, RV10, RV1 1, RV15) were tested in the same way:
Figure imgf000070_0001
Figure imgf000070_0002
Except for liposome Q each of these liposomes performed better than the control. The RV10 lipid in liposome Q has a pKa of 7.86 which seems too high to be useful in vivo. Even inside the useful pKa range of 5.0 to 7.6, however, although results were good, none of the lipids with one alkyl tail and one steroid-containing tail gave results as good as RV01.
Further liposomes were made with RV05. The liposomes all had 40% RV05 and 2% PEGylated lipid, but the remaining components varied (although cholesterol was always included). Physical characteristics were:
Figure imgf000070_0003
V Cholesterol 10% DSPC, 46% chol, 2% aGC 103.7 0.107 72.58 w DMG 10% DPyPE, 48% chol 99.6 0.115 78.34
X DMG 10% 18:3 PC, 48% chol 130 0.14 87.92
Y DMG 10% 18:2 PC, 48% chol 101.1 0.133 76.64 z DMG 30% 18:2 PC, 28% chol 134.3 0.158 57.76
aGC = a-galactosylceramide
BALB/c mice were tested as before:
Figure imgf000071_0001
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 by replacing DSPC with DPyPE.
In a final experiment with the RV05 lipid a liposome was made with 40% RV05, 10% 18:2 PC, 40% DPyPE, 8% cholesterol and 2% PEG DMG 2000. These liposomes had a Zav diameter of 124.7nm, a pdl of 0.17 and a RNA encapsulation of 61.5%. They were used to vaccinate BALB/c mice as before (O.^g RNA dose), in comparison with naked RNA (^g) or with RV01 -based liposomes (40% DlinDMA, 10% DPSC, 48% cholesterol, 2% PEG DMG 2000). F-specific serum IgG titers (GMT) were as follows:
Figure imgf000071_0002
Thus the RV05 liposomes were more immunogenic than naked RNA, but less immunogenic than RV01 liposomes.
Spleens were harvested at day 49 for T cell analysis. Average net F-specific cytokine-positive T cell frequencies (CD4+ or CD8+) were as follows, showing only figures which were statistically significantly above zero (specific for RSV peptides F51-66, F164-178, F309-323 for CD4+, or for peptides F85-93 and F249-258 for CD8+):
Figure imgf000072_0001
In terms of T cell responses, therefore, RV05 gave better results than RV01.
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.
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 l ,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 l ,2-Dimyristoyl-sn-glycero-3-phosphoethanolamine
DMPA l ,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 1 ,2-Dioleoyl-sn-Glycero-3 [Phosphatidyl-rac-( 1 -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 1 ,2-Dipalmitoyl-sn-Gl ycero-3-phosphatidylethanolamine
DPPG 1 ,2-Dipalmitoyl-sn-Glycero-3 [Phosphatidyl-rac-( 1 -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 DSPE 1 ,2-Diostearpyl-sn-Glycero-3-phosphatidylethanolamine
DSPG 1 ,2-Distearoyl-sn-Glycero-3 [Phosphatidyl-rac-( 1 -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-Stearoyl,2-oleoyl-sn-Glycero-3-phosphatidylcholine
SPPC l-Stearoyl,2-palmitoyl-sn-Glycero-3-phosphatidylcholine
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] WO201 1/076807.
[9] Tarwadi et al. (2008) Bioconjugate Chem. 19:940-950.
[10] WO2005/113782.
[I I] WO2011/005799.
[12] El Ouahabi et al. (1996) FEBS Letts 380: 108-12.
[13] Giuliani et al. (2006) Proc Natl Acad Sci U S A 103(29): 10834-9.
[14] WO2009/016515.
[15] WO02/34771.
[16] WO2005/032582.
[17] WO2010/119343.
[18] WO2006/110413.
[19] WO2005/111066.
[20] WO2005/002619.
[21] WO2006/138004.
[22] WO2009/109860.
[23] WO02/02606.
[24] WO03/018054.
[25] WO2006/091517.
[26] WO2008/020330.
[27] WO2006/089264.
[28] WO2009/104092.
[29] WO2009/031043.
[30] WO2007/049155.
[31] Gennaro (2000) Remington: The Science and Practice of Pharmacy. 20th edition, ISBN: 0683306472.
[32] Methods In Enzymology (S. Colowick and N. Kaplan, eds., Academic Press, Inc.)
[33] Handbook of Experimental Immunology, Vols. I-IV (D.M. Weir and C.C. Blackwell, eds, 1986, Blackwell Scientific Publications) [34] Sambrook et al. (2001) Molecular Cloning: A Laboratory Manual, 3rd edition (Cold Spring Harbor Laboratory Press).
[35] Handbook of Surface and Colloidal Chemistry (Birdi, K.S. ed., CRC Press, 1997)
[36] Ausubel et al. (eds) (2002) Short protocols in molecular biology, 5th edition (Current Protocols).
[37] Molecular Biology Techniques: An Intensive Laboratory Course, (Ream et al, eds., 1998, Academic Press)
[38] PCR (Introduction to Biotechniques Series), 2nd ed. (Newton & Graham eds., 1997, Springer Verlag)
[39] Yoneyama & Fujita (2007) Cytokine & Growth Factor Reviews 18:545-51.
[40] Maurer et al. (2001) Biophysical Journal, 80: 2310-2326.
[41] Perri et al. (2003) J Virol 77: 10394-10403.

Claims

1. A liposome within which RNA encoding a polypeptide of interest is encapsulated, wherein the liposome includes at least one compound selected from the group consisting of compounds of formula (I) and formula (XI), where
Formula (I) is:
Figure imgf000077_0001
wherein:
R1 and R2 together with the nitrogen atom to which they are attached form an optionally substituted C3_2o-heterocycloalkyl, C3_2o-heterocycloalkenyl, C3_2o-heterocycloalkynyl or C5-2o-heteroaryl group;
a is absent or optionally substituted C1.4 alkylene;
b is absent or optionally substituted C1-4 alkylene;
c is absent or optionally substituted C1.4 alkylene;
X1 is O or S;
X2 is O or S;
Y1 is optionally substituted Cio-3oalkenyl, Cio-3oalkynyl, Cio-3oheteroalkenyl or Cio-3oheteroalkynyl;
L is absent or is -(La)d-(Lb)e-(Lc)f-, wherein
La is optionally substituted Ci-isalkylene, Ci-isalkenylene, Ci-isalkynylene, Ci-i5heteroalkylene, Ci-isheteroalkenylene or Ci-isheteroalkynylene;
Lb is optionally substituted C6-i4arylene or C5_i3heteroarylene;
If is optionally substituted Ci-isalkylene, Ci-isalkenylene, Ci-isalkynylene, Ci_i5heteroalkylene, Ci_i5heteroalkenylene or Ci_i5heteroalkynylene;
d is 0 or 1 ;
e is 0 or 1 ; and
f is 0 or 1 ; and
Y2 is an optionally substituted steroid.
Formula (XI) is: Ra-(AA)z-k
wherein
Ra is a N- terminal alkylamide;
z is an integer from 2 to 10;
each AA is an amino acid, provided that at least one histidine is present and at least one cationic amino acid is present;
Rb is -H or -NH2.
2. The liposome of claim 1, wherein the liposome has a diameter in the range of 80-160nm.
3. The liposome of any preceding claim, wherein the RNA is a self -replicating RNA.
4. The liposome of claim 3, 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) the polypeptide of interest.
5. The liposome of claim 4, wherein the RNA molecule has two open reading frames, the first of which encodes an alphavirus replicase and the second of which encodes the polypeptide of interest.
6. The liposome of any preceding claim, wherein the RNA molecule is 9000-12000 nucleotides long.
7. The liposome of any preceding claim, wherein the polypeptide of interest is an immunogen.
8. The liposome of claim 7, wherein the immunogen can elicit an immune response in vivo against a bacterium, a virus, a fungus or a parasite.
9. The liposome of claim 8, wherein the immunogen can elicit an immune response in vivo against respiratory syncytial virus glycoprotein F.
10. A pharmaceutical composition comprising a liposome of any preceding claim.
11. 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 1-9, or the pharmaceutical composition of claim 10.
PCT/US2011/050100 2010-08-31 2011-08-31 Lipids suitable for liposomal delivery of protein-coding rna WO2012031046A2 (en)

Priority Applications (11)

Application Number Priority Date Filing Date Title
CA2809863A CA2809863A1 (en) 2010-08-31 2011-08-31 Lipids suitable for liposomal delivery of protein-coding rna
RU2013114330/10A RU2577983C2 (en) 2010-08-31 2011-08-31 Lipids suitable for liposomal delivery of rna encoding protein
CN201180051927.2A CN103384515B (en) 2010-08-31 2011-08-31 Lipids suitable for liposomal delivery of protein-coding RNA
EP11763813.0A EP2611420B1 (en) 2010-08-31 2011-08-31 Lipids suitable for liposomal delivery of protein-coding rna
BR112013004865A BR112013004865A2 (en) 2010-08-31 2011-08-31 lipids suitable for liposomal delivery of rna protein coders
US13/819,228 US20130189351A1 (en) 2010-08-31 2011-08-31 Lipids suitable for liposomal delivery of protein coding rna
AU2011295938A AU2011295938B2 (en) 2010-08-31 2011-08-31 Lipids suitable for liposomal delivery of protein-coding RNA
JP2013526211A JP5908477B2 (en) 2010-08-31 2011-08-31 Lipids suitable for liposome delivery of protein-encoding RNA
ES11763813T ES2727583T3 (en) 2010-08-31 2011-08-31 Lipids suitable for liposomal administration of RNA encoding proteins
MX2013002332A MX2013002332A (en) 2010-08-31 2011-08-31 Lipids suitable for liposomal delivery of protein-coding rna.
EP19165057.1A EP3542789A3 (en) 2010-08-31 2011-08-31 Lipids suitable for liposomal delivery of protein-coding rna

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US37883310P 2010-08-31 2010-08-31
US61/378,833 2010-08-31

Publications (2)

Publication Number Publication Date
WO2012031046A2 true WO2012031046A2 (en) 2012-03-08
WO2012031046A3 WO2012031046A3 (en) 2014-04-03

Family

ID=44720124

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2011/050100 WO2012031046A2 (en) 2010-08-31 2011-08-31 Lipids suitable for liposomal delivery of protein-coding rna

Country Status (12)

Country Link
US (1) US20130189351A1 (en)
EP (2) EP2611420B1 (en)
JP (2) JP5908477B2 (en)
CN (1) CN103384515B (en)
AU (3) AU2011295938B2 (en)
BR (1) BR112013004865A2 (en)
CA (1) CA2809863A1 (en)
ES (1) ES2727583T3 (en)
MX (1) MX2013002332A (en)
RU (1) RU2577983C2 (en)
TR (1) TR201908635T4 (en)
WO (1) WO2012031046A2 (en)

Cited By (153)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013151736A2 (en) 2012-04-02 2013-10-10 modeRNA Therapeutics In vivo production of proteins
WO2013151666A2 (en) 2012-04-02 2013-10-10 modeRNA Therapeutics Modified polynucleotides for the production of biologics and proteins associated with human disease
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
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
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
WO2017115268A1 (en) 2015-12-28 2017-07-06 Novartis Ag Compositions and methods for the treatment of hemoglobinopathies
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
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
WO2018104540A1 (en) 2016-12-08 2018-06-14 Curevac Ag Rnas for wound healing
WO2018104538A1 (en) 2016-12-08 2018-06-14 Curevac Ag Rna for treatment or prophylaxis of a liver disease
WO2018115525A1 (en) 2016-12-23 2018-06-28 Curevac Ag Lassa virus vaccine
WO2018115527A2 (en) 2016-12-23 2018-06-28 Curevac Ag Mers coronavirus vaccine
WO2018142364A1 (en) 2017-02-06 2018-08-09 Novartis Ag Compositions and methods for the treatment of hemoglobinopathies
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
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
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
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
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
US10501513B2 (en) 2012-04-02 2019-12-10 Modernatx, Inc. Modified polynucleotides for the production of oncology-related proteins and peptides
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
EP3701959A1 (en) 2016-03-21 2020-09-02 BioNTech RNA Pharmaceuticals GmbH Rna replicon for versatile and efficient gene expression
US10815291B2 (en) 2013-09-30 2020-10-27 Modernatx, Inc. Polynucleotides encoding immune modulating polypeptides
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
WO2020263985A1 (en) 2019-06-24 2020-12-30 Modernatx, Inc. Messenger rna comprising functional rna elements and uses thereof
WO2020263883A1 (en) 2019-06-24 2020-12-30 Modernatx, Inc. Endonuclease-resistant messenger rna and uses thereof
WO2021009336A1 (en) 2019-07-18 2021-01-21 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for inducing full ablation of hematopoiesis
WO2021013798A1 (en) 2019-07-21 2021-01-28 Glaxosmithkline Biologicals Sa Therapeutic viral vaccine
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
WO2021123920A1 (en) 2019-12-18 2021-06-24 Novartis Ag Compositions and methods for the treatment of hemoglobinopathies
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
WO2021209970A1 (en) 2020-04-16 2021-10-21 Glaxosmithkline Biologicals Sa Sars cov-2 spike protein construct
WO2021245090A1 (en) 2020-06-04 2021-12-09 BioNTech SE Rna replicon for versatile and efficient gene expression
WO2021247507A1 (en) 2020-06-01 2021-12-09 Modernatx, Inc. Phenylalanine hydroxylase variants and uses thereof
WO2021245611A1 (en) 2020-06-05 2021-12-09 Glaxosmithkline Biologicals Sa Modified betacoronavirus spike proteins
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
US11291682B2 (en) 2010-07-06 2022-04-05 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11291635B2 (en) 2010-07-06 2022-04-05 Glaxosmithkline Biological Sa Virion-like delivery particles for self-replicating RNA molecules
EP3718565B1 (en) 2015-10-22 2022-04-27 ModernaTX, Inc. Respiratory virus vaccines
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
WO2022204369A1 (en) 2021-03-24 2022-09-29 Modernatx, Inc. Polynucleotides encoding methylmalonyl-coa mutase for the treatment of methylmalonic acidemia
WO2022204390A1 (en) 2021-03-24 2022-09-29 Modernatx, Inc. Lipid nanoparticles containing polynucleotides encoding phenylalanine hydroxylase and uses thereof
WO2022200575A1 (en) 2021-03-26 2022-09-29 Glaxosmithkline Biologicals Sa Immunogenic compositions
WO2022204380A1 (en) 2021-03-24 2022-09-29 Modernatx, Inc. Lipid nanoparticles containing polynucleotides encoding propionyl-coa carboxylase alpha and beta subunits and uses thereof
WO2022204370A1 (en) 2021-03-24 2022-09-29 Modernatx, Inc. Lipid nanoparticles and polynucleotides encoding ornithine transcarbamylase for the treatment of ornithine transcarbamylase deficiency
WO2022204371A1 (en) 2021-03-24 2022-09-29 Modernatx, Inc. Lipid nanoparticles containing polynucleotides encoding glucose-6-phosphatase and uses thereof
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
WO2022269518A2 (en) 2021-06-23 2022-12-29 Novartis Ag Compositions and methods for the treatment of hemoglobinopathies
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
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
WO2023020994A1 (en) 2021-08-16 2023-02-23 Glaxosmithkline Biologicals Sa Novel methods
WO2023020993A1 (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
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
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
US11639370B2 (en) 2010-10-11 2023-05-02 Glaxosmithkline Biologicals Sa Antigen delivery platforms
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
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
US11744801B2 (en) 2017-08-31 2023-09-05 Modernatx, Inc. Methods of making lipid nanoparticles
US11759422B2 (en) 2010-08-31 2023-09-19 Glaxosmithkline Biologicals Sa Pegylated liposomes for delivery of immunogen-encoding RNA
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
WO2023213378A1 (en) 2022-05-02 2023-11-09 BioNTech SE Replicon compositions and methods of using same for the treatment of diseases
WO2023242817A2 (en) 2022-06-18 2023-12-21 Glaxosmithkline Biologicals Sa Recombinant rna molecules comprising untranslated regions or segments encoding spike protein from the omicron strain of severe acute respiratory coronavirus-2
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
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
US11964011B2 (en) 2020-02-04 2024-04-23 CureVac SE Coronavirus vaccine
US11969506B2 (en) 2017-03-15 2024-04-30 Modernatx, Inc. Lipid nanoparticle formulation
US11976019B2 (en) 2020-07-16 2024-05-07 Acuitas Therapeutics, Inc. Cationic lipids for use in lipid nanoparticles

Families Citing this family (52)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014028429A2 (en) 2012-08-14 2014-02-20 Moderna Therapeutics, Inc. Enzymes and polymerases for the synthesis of rna
US8980864B2 (en) 2013-03-15 2015-03-17 Moderna Therapeutics, Inc. Compositions and methods of altering cholesterol levels
EP3324979B1 (en) 2015-07-21 2022-10-12 ModernaTX, Inc. Infectious disease vaccines
US11364292B2 (en) 2015-07-21 2022-06-21 Modernatx, Inc. CHIKV RNA vaccines
EP3359670B2 (en) 2015-10-05 2024-02-14 ModernaTX, Inc. Methods for therapeutic administration of messenger ribonucleic acid drugs
JP6925688B2 (en) 2015-10-22 2021-08-25 モデルナティーエックス, インコーポレイテッド Nucleic acid vaccine for varicella-zoster virus (VZV)
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
WO2017070624A1 (en) 2015-10-22 2017-04-27 Modernatx, Inc. Tropical disease vaccines
KR20230074598A (en) 2016-05-18 2023-05-30 모더나티엑스, 인크. Polynucleotides encoding relaxin
US10925958B2 (en) 2016-11-11 2021-02-23 Modernatx, Inc. Influenza vaccine
US11103578B2 (en) 2016-12-08 2021-08-31 Modernatx, Inc. Respiratory virus nucleic acid vaccines
EP3609534A4 (en) 2017-03-15 2021-01-13 ModernaTX, Inc. Broad spectrum influenza virus vaccine
WO2018170260A1 (en) 2017-03-15 2018-09-20 Modernatx, Inc. Respiratory syncytial virus vaccine
WO2018170256A1 (en) 2017-03-15 2018-09-20 Modernatx, Inc. Herpes simplex virus vaccine
WO2018170270A1 (en) 2017-03-15 2018-09-20 Modernatx, Inc. Varicella zoster virus (vzv) vaccine
EP3634449A4 (en) 2017-05-08 2021-03-17 Gritstone Oncology, Inc. Alphavirus neoantigen vectors
AU2018286475A1 (en) * 2017-06-13 2019-12-19 Monash University Non-viral gene delivery agent comprising lipopeptide (LP) compounds
JP7355731B2 (en) 2017-08-16 2023-10-03 アクイタス セラピューティクス インコーポレイテッド Lipids for use in lipid nanoparticle formulations
EP3681514A4 (en) 2017-09-14 2021-07-14 ModernaTX, Inc. Zika virus rna vaccines
AU2019205330A1 (en) 2018-01-04 2020-08-27 Iconic Therapeutics Llc 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
AU2020282369A1 (en) 2019-05-30 2022-01-20 Gritstone Bio, Inc. Modified adenoviruses
JP2022542839A (en) 2019-07-19 2022-10-07 フラッグシップ パイオニアリング イノベーションズ シックス,エルエルシー Recombinase compositions and methods of use
CA3179420A1 (en) 2020-05-20 2021-11-25 Avak Kahvejian Coronavirus antigen compositions and their uses
CA3179444A1 (en) 2020-05-20 2021-11-25 Avak Kahvejian Immunogenic compositions and uses thereof
EP4158032A2 (en) 2020-05-29 2023-04-05 Flagship Pioneering Innovations VI, LLC Trem compositions and methods relating thereto
CN116018405A (en) 2020-05-29 2023-04-25 旗舰先锋创新Vi有限责任公司 TREM compositions and related methods
WO2022032196A2 (en) 2020-08-06 2022-02-10 Gritstone Bio, Inc. Multiepitope vaccine cassettes
CN116157148A (en) 2020-09-03 2023-05-23 旗舰创业创新第六有限责任公司 Immunogenic compositions and uses thereof
IL303886A (en) 2020-12-23 2023-08-01 Flagship Pioneering Inc Compositions of modified trems and uses thereof
US20220325287A1 (en) 2021-03-31 2022-10-13 Flagship Pioneering Innovations V, Inc. Thanotransmission polypeptides and their use in treating cancer
WO2023009547A1 (en) 2021-07-26 2023-02-02 Flagship Pioneering Innovations Vi, Llc Trem compositions and uses thereof
AR127073A1 (en) 2021-09-17 2023-12-13 Flagship Pioneering Innovations Vi Llc COMPOSITIONS AND METHODS FOR PRODUCING CIRCULAR POLYRIBONUCLEOTIDES
AU2022370530A1 (en) 2021-10-18 2024-05-02 Flagship Pioneering Innovations Vi, Llc Compositions and methods for purifying polyribonucleotides
WO2023096990A1 (en) 2021-11-24 2023-06-01 Flagship Pioneering Innovation Vi, Llc Coronavirus immunogen compositions and their uses
WO2023096963A1 (en) 2021-11-24 2023-06-01 Flagship Pioneering Innovations Vi, Llc Varicella-zoster virus immunogen compositions and their uses
WO2023097003A2 (en) 2021-11-24 2023-06-01 Flagship Pioneering Innovations Vi, Llc Immunogenic compositions and their uses
AR128002A1 (en) 2021-12-17 2024-03-20 Flagship Pioneering Innovations Vi Llc CIRCULAR RNA ENRICHMENT METHODS UNDER DENATURALING CONDITIONS
TW202340461A (en) 2021-12-22 2023-10-16 美商旗艦先鋒創新有限責任公司 Compositions and methods for purifying polyribonucleotides
WO2023122789A1 (en) 2021-12-23 2023-06-29 Flagship Pioneering Innovations Vi, Llc Circular polyribonucleotides encoding antifusogenic polypeptides
WO2023183616A1 (en) 2022-03-25 2023-09-28 Senda Biosciences, Inc. Novel ionizable lipids and lipid nanoparticles and methods of using the same
WO2023196634A2 (en) 2022-04-08 2023-10-12 Flagship Pioneering Innovations Vii, Llc Vaccines and related methods
WO2023220083A1 (en) 2022-05-09 2023-11-16 Flagship Pioneering Innovations Vi, Llc Trem compositions and methods of use for treating proliferative disorders
WO2023220729A2 (en) 2022-05-13 2023-11-16 Flagship Pioneering Innovations Vii, Llc Double stranded dna compositions and related methods
WO2023250112A1 (en) 2022-06-22 2023-12-28 Flagship Pioneering Innovations Vi, Llc Compositions of modified trems and uses thereof
US20240042021A1 (en) 2022-08-01 2024-02-08 Flagship Pioneering Innovations Vii, Llc Immunomodulatory proteins and related methods
WO2024035952A1 (en) 2022-08-12 2024-02-15 Remix Therapeutics Inc. Methods and compositions for modulating splicing at alternative splice sites
WO2024049979A2 (en) 2022-08-31 2024-03-07 Senda Biosciences, Inc. Novel ionizable lipids and lipid nanoparticles and methods of using the same
WO2024077191A1 (en) 2022-10-05 2024-04-11 Flagship Pioneering Innovations V, Inc. Nucleic acid molecules encoding trif and additionalpolypeptides and their use in treating cancer
WO2024097664A1 (en) 2022-10-31 2024-05-10 Flagship Pioneering Innovations Vi, Llc Compositions and methods for purifying polyribonucleotides
WO2024102799A1 (en) 2022-11-08 2024-05-16 Flagship Pioneering Innovations Vi, Llc Compositions and methods for producing circular polyribonucleotides

Citations (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
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
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
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
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
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
WO2011076807A2 (en) 2009-12-23 2011-06-30 Novartis Ag Lipids, lipid compositions, and methods of using them

Family Cites Families (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6656498B1 (en) * 1998-11-25 2003-12-02 Vanderbilt University Cationic liposomes for gene transfer
AU2001233246A1 (en) * 2000-02-04 2001-08-14 California Pacific Medical Center Research Institute Novel use of ribozymes to block gene expression
WO2004018506A2 (en) * 2002-08-22 2004-03-04 Cytos Biotechnology Ag Inducible alphaviral/orip based gene expression system
US7745651B2 (en) * 2004-06-07 2010-06-29 Protiva Biotherapeutics, Inc. Cationic lipids and methods of use
US20080207544A1 (en) * 2005-01-25 2008-08-28 Sullivan Sean M Compositions And Methods For Delivery Of Agents Into Cells
EP1922300A2 (en) * 2005-02-14 2008-05-21 Sirna Therapeutics Inc. Cationic lipids and formulated molecular compositions containing them
US7404969B2 (en) * 2005-02-14 2008-07-29 Sirna Therapeutics, Inc. Lipid nanoparticle based compositions and methods for the delivery of biologically active molecules
CN101421293B (en) * 2006-03-01 2013-09-25 福冈县政府 Peptide lipid-containing carrier and method for introducing compound into cells using same
WO2008103276A2 (en) * 2007-02-16 2008-08-28 Merck & Co., Inc. Compositions and methods for potentiated activity of biologicaly active molecules
EP2197495A2 (en) * 2007-10-02 2010-06-23 MDRNA, Inc. Lipopeptides for delivery of nucleic acids
EP2130912A1 (en) * 2008-06-04 2009-12-09 Institut für Viruskrankeiten und Immunprophylaxe Pestivirus replicons providing an RNA-based viral vector system
WO2010003276A1 (en) * 2008-07-11 2010-01-14 桂林吉星电子等平衡动力有限公司 Four-wheel-drive power train and operating control method of hev
EP3072881A1 (en) * 2009-08-20 2016-09-28 Sirna Therapeutics, Inc. Novel cationic lipids with various head groups for oligonucleotide delivery
WO2011043913A2 (en) * 2009-10-08 2011-04-14 Merck Sharp & Dohme Corp. Novel cationic lipids with short lipid chains for oligonucleotide delivery
JP2013531634A (en) * 2010-05-24 2013-08-08 メルク・シャープ・エンド・ドーム・コーポレイション Novel aminoalcohol cationic lipids for oligonucleotide delivery

Patent Citations (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
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
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
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
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
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
WO2011076807A2 (en) 2009-12-23 2011-06-30 Novartis Ag Lipids, lipid compositions, and methods of using them

Non-Patent Citations (22)

* 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 Immunology", 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 Biolechniques Series", 1997, SPRINGER VERLAG
"Short protocols in molecular biology", 2002, CURRENT PROTOCOLS
D. HELLWINKEL: "Systemic Nomenclature of Organic Chemistry", 2001, SPRINGER, pages: 203
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
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
MACLACHLAN ET AL., JOURNAL OF CONTROLLED RELEASE, vol. 107, 2005, pages 276 - 287
MAURER ET AL., BIOPHYSICAL JOURNAL, vol. 80, 2001, pages 2310 - 2326
PERRI ET AL., J VIROL, vol. 77, 2003, pages 10394 - 10403
SAMBROOK ET AL.: "Molecular Cloning: A Laboratory Manual", 2001, COLD SPRING HARBOR LABORATORY PRESS
TARWADI ET AL., BIOCONJUGATE CHEM, vol. 19, 2008, pages 940 - 950
YONEYAMA, FUJITA, CYTOKINE & GROWTH FACTOR REVIEWS, vol. 18, 2007, pages 545 - 51

Cited By (269)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11857562B2 (en) 2010-07-06 2024-01-02 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11857681B2 (en) 2010-07-06 2024-01-02 Glaxosmithkline Biologicals Sa Lipid formulations with RNA encoding immunogens
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
US11739334B2 (en) 2010-07-06 2023-08-29 Glaxosmithkline Biologicals Sa Immunisation of large mammals with low doses of RNA
US11291682B2 (en) 2010-07-06 2022-04-05 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
EP4005592B1 (en) 2010-07-06 2022-10-12 GlaxoSmithKline Biologicals S.A. Virion-like delivery particles for self-replicating rna molecules
US11913001B2 (en) 2010-07-06 2024-02-27 Glaxosmithkline Biologicals Sa Immunisation of large mammals with low doses of RNA
US11596645B2 (en) 2010-07-06 2023-03-07 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11638694B2 (en) 2010-07-06 2023-05-02 Glaxosmithkline Biologicals Sa Vaccine for eliciting immune response comprising lipid formulations and RNA encoding multiple immunogens
US11905514B2 (en) 2010-07-06 2024-02-20 Glaxosmithkline Biological Sa Immunisation of large mammals with low doses of RNA
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
US11655475B2 (en) 2010-07-06 2023-05-23 Glaxosmithkline Biologicals Sa Immunisation of large mammals with low doses of RNA
US11666534B2 (en) 2010-07-06 2023-06-06 Glaxosmithkline Biologicals Sa Methods of administering lipid formulations with viral immunogens
US11730754B2 (en) 2010-07-06 2023-08-22 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
US11690863B2 (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
US11690865B2 (en) 2010-07-06 2023-07-04 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11696923B2 (en) 2010-07-06 2023-07-11 Glaxosmithkline Biologicals, Sa Delivery of RNA to trigger multiple immune pathways
US11707482B2 (en) 2010-07-06 2023-07-25 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11717529B2 (en) 2010-07-06 2023-08-08 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11690862B1 (en) 2010-07-06 2023-07-04 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11891608B2 (en) 2010-07-06 2024-02-06 Glaxosmithkline Biologicals Sa Immunization of large mammals with low doses of RNA
US11851660B2 (en) 2010-07-06 2023-12-26 Glaxosmithkline Biologicals Sa Immunisation of large mammals with low doses of RNA
US11883534B2 (en) 2010-07-06 2024-01-30 Glaxosmithkline Biologicals Sa Immunisation with lipid formulations with RNA encoding immunogens
US11759475B2 (en) 2010-07-06 2023-09-19 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11766401B2 (en) 2010-07-06 2023-09-26 Glaxosmithkline Biologicals Sa Methods of administering lipid formulations with immunogens
US11773395B1 (en) 2010-07-06 2023-10-03 Glaxosmithkline Biologicals Sa Immunization of large mammals with low doses of RNA
US11786467B2 (en) 2010-07-06 2023-10-17 Glaxosmithkline Biologicals Sa Lipid formulations with immunogens
US11839686B2 (en) 2010-07-06 2023-12-12 Glaxosmithkline Biologicals Sa Lipid formulations with viral immunogens
US11845925B2 (en) 2010-07-06 2023-12-19 Glaxosmithkline Biologicals Sa Immunisation of large mammals with low doses of RNA
US11865080B2 (en) 2010-07-06 2024-01-09 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
US11324770B2 (en) 2010-07-06 2022-05-10 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
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
US8822663B2 (en) 2010-08-06 2014-09-02 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
US11759422B2 (en) 2010-08-31 2023-09-19 Glaxosmithkline Biologicals Sa Pegylated liposomes for delivery of immunogen-encoding RNA
US9254265B2 (en) 2010-08-31 2016-02-09 Novartis Ag Small liposomes for delivery of immunogen encoding RNA
US9657295B2 (en) 2010-10-01 2017-05-23 Modernatx, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
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
US11639370B2 (en) 2010-10-11 2023-05-02 Glaxosmithkline Biologicals Sa Antigen delivery platforms
US8710200B2 (en) 2011-03-31 2014-04-29 Moderna Therapeutics, Inc. Engineered nucleic acids encoding a modified erythropoietin and their expression
US9950068B2 (en) 2011-03-31 2018-04-24 Modernatx, Inc. Delivery and formulation of engineered nucleic acids
US9533047B2 (en) 2011-03-31 2017-01-03 Modernatx, Inc. Delivery and formulation of engineered nucleic acids
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
US11951180B2 (en) 2011-06-08 2024-04-09 Translate Bio, Inc. Lipid nanoparticle compositions and methods for MRNA delivery
US11547764B2 (en) 2011-06-08 2023-01-10 Translate Bio, Inc. Lipid nanoparticle compositions and methods for MRNA delivery
US11896636B2 (en) 2011-07-06 2024-02-13 Glaxosmithkline Biologicals Sa Immunogenic combination compositions and uses thereof
US10751386B2 (en) 2011-09-12 2020-08-25 Modernatx, 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
US9464124B2 (en) 2011-09-12 2016-10-11 Moderna Therapeutics, 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
EP4144378A1 (en) 2011-12-16 2023-03-08 ModernaTX, Inc. Modified nucleoside, nucleotide, and nucleic acid compositions
US9186372B2 (en) 2011-12-16 2015-11-17 Moderna Therapeutics, Inc. Split dose administration
US9271996B2 (en) 2011-12-16 2016-03-01 Moderna Therapeutics, Inc. Formulation and delivery of PLGA microspheres
US9295689B2 (en) 2011-12-16 2016-03-29 Moderna Therapeutics, Inc. Formulation and delivery of PLGA microspheres
US8680069B2 (en) 2011-12-16 2014-03-25 Moderna Therapeutics, Inc. Modified polynucleotides for the production of G-CSF
US8664194B2 (en) 2011-12-16 2014-03-04 Moderna Therapeutics, Inc. Method for producing a protein of interest in a primate
US8999380B2 (en) 2012-04-02 2015-04-07 Moderna Therapeutics, Inc. Modified polynucleotides for the production of biologics and proteins associated with human disease
US9221891B2 (en) 2012-04-02 2015-12-29 Moderna Therapeutics, Inc. In vivo production of proteins
WO2013151666A2 (en) 2012-04-02 2013-10-10 modeRNA Therapeutics Modified polynucleotides for the production of biologics and proteins associated with human disease
US9050297B2 (en) 2012-04-02 2015-06-09 Moderna Therapeutics, Inc. Modified polynucleotides encoding aryl hydrocarbon receptor nuclear translocator
US9587003B2 (en) 2012-04-02 2017-03-07 Modernatx, Inc. Modified polynucleotides for the production of oncology-related proteins and peptides
WO2013151736A2 (en) 2012-04-02 2013-10-10 modeRNA Therapeutics In vivo production of proteins
US9061059B2 (en) 2012-04-02 2015-06-23 Moderna Therapeutics, Inc. Modified polynucleotides for treating protein deficiency
US9572897B2 (en) 2012-04-02 2017-02-21 Modernatx, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
US10772975B2 (en) 2012-04-02 2020-09-15 Modernatx, Inc. Modified Polynucleotides for the production of biologics and proteins associated with human disease
JP2015518705A (en) * 2012-04-02 2015-07-06 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. Modified polynucleotides for the production of biologics and proteins associated with human diseases
US10703789B2 (en) 2012-04-02 2020-07-07 Modernatx, Inc. Modified polynucleotides for the production of secreted proteins
US9878056B2 (en) 2012-04-02 2018-01-30 Modernatx, Inc. Modified polynucleotides for the production of cosmetic proteins and peptides
JP2015519881A (en) * 2012-04-02 2015-07-16 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. Modified polynucleotides for the production of nucleoproteins
US9089604B2 (en) 2012-04-02 2015-07-28 Moderna Therapeutics, Inc. Modified polynucleotides for treating galactosylceramidase protein deficiency
US9095552B2 (en) 2012-04-02 2015-08-04 Moderna Therapeutics, Inc. Modified polynucleotides encoding copper metabolism (MURR1) domain containing 1
US9107886B2 (en) 2012-04-02 2015-08-18 Moderna Therapeutics, Inc. Modified polynucleotides encoding basic helix-loop-helix family member E41
US9828416B2 (en) 2012-04-02 2017-11-28 Modernatx, Inc. Modified polynucleotides for the production of secreted proteins
US10583203B2 (en) 2012-04-02 2020-03-10 Modernatx, Inc. In vivo production of proteins
US9675668B2 (en) 2012-04-02 2017-06-13 Moderna Therapeutics, Inc. Modified polynucleotides encoding hepatitis A virus cellular receptor 2
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
US9827332B2 (en) 2012-04-02 2017-11-28 Modernatx, Inc. Modified polynucleotides for the production of proteins
US9814760B2 (en) 2012-04-02 2017-11-14 Modernatx, Inc. Modified polynucleotides for the production of biologics and proteins associated with human disease
US9283287B2 (en) 2012-04-02 2016-03-15 Moderna Therapeutics, Inc. Modified polynucleotides for the production of nuclear proteins
US9114113B2 (en) 2012-04-02 2015-08-25 Moderna Therapeutics, Inc. Modified polynucleotides encoding citeD4
US11564998B2 (en) 2012-04-02 2023-01-31 Modernatx, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
US10577403B2 (en) 2012-04-02 2020-03-03 Modernatx, Inc. Modified polynucleotides for the production of secreted proteins
US9149506B2 (en) 2012-04-02 2015-10-06 Moderna Therapeutics, Inc. Modified polynucleotides encoding septin-4
US9782462B2 (en) 2012-04-02 2017-10-10 Modernatx, Inc. Modified polynucleotides for the production of proteins associated with human disease
US9254311B2 (en) 2012-04-02 2016-02-09 Moderna Therapeutics, Inc. Modified polynucleotides for the production of proteins
US9255129B2 (en) 2012-04-02 2016-02-09 Moderna Therapeutics, Inc. Modified polynucleotides encoding SIAH E3 ubiquitin protein ligase 1
US10501513B2 (en) 2012-04-02 2019-12-10 Modernatx, Inc. Modified polynucleotides for the production of oncology-related proteins and peptides
US10501512B2 (en) 2012-04-02 2019-12-10 Modernatx, Inc. Modified polynucleotides
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
US10493167B2 (en) 2012-04-02 2019-12-03 Modernatx, Inc. In vivo production of 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
US9192651B2 (en) 2012-04-02 2015-11-24 Moderna Therapeutics, Inc. Modified polynucleotides for the production of secreted proteins
US9216205B2 (en) 2012-04-02 2015-12-22 Moderna Therapeutics, Inc. Modified polynucleotides encoding granulysin
US10463751B2 (en) 2012-04-02 2019-11-05 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
EP4074834A1 (en) 2012-11-26 2022-10-19 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
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
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
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
US10022435B2 (en) 2014-04-23 2018-07-17 Modernatx, Inc. Nucleic acid vaccines
US10709779B2 (en) 2014-04-23 2020-07-14 Modernatx, Inc. Nucleic acid vaccines
US9872900B2 (en) 2014-04-23 2018-01-23 Modernatx, Inc. Nucleic acid vaccines
EP3134131B1 (en) 2014-04-23 2021-12-22 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
EP3061826A1 (en) 2015-02-27 2016-08-31 Novartis AG Flavivirus replicons
WO2016135675A1 (en) 2015-02-27 2016-09-01 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
US10266485B2 (en) 2015-09-17 2019-04-23 Modernatx, Inc. Compounds and compositions for intracellular delivery of therapeutic agents
US11220476B2 (en) 2015-09-17 2022-01-11 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
US10442756B2 (en) 2015-09-17 2019-10-15 Modernatx, Inc. Compounds and compositions for intracellular delivery of therapeutic agents
EP3718565B1 (en) 2015-10-22 2022-04-27 ModernaTX, Inc. Respiratory virus vaccines
US11648324B2 (en) 2015-10-28 2023-05-16 Acuitas Therapeutics, Inc. Lipids and lipid nanoparticle formulations for delivery of nucleic acids
US10485885B2 (en) 2015-12-10 2019-11-26 Modernatx, Inc. Compositions and methods for delivery of agents
US10556018B2 (en) 2015-12-10 2020-02-11 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
US11285222B2 (en) 2015-12-10 2022-03-29 Modernatx, Inc. Compositions and methods for delivery of agents
US10799463B2 (en) 2015-12-22 2020-10-13 Modernatx, Inc. Compounds and compositions for intracellular delivery of agents
US10195156B2 (en) 2015-12-22 2019-02-05 Modernatx, Inc. Compounds and compositions for intracellular delivery of agents
WO2017112943A1 (en) 2015-12-23 2017-06-29 Modernatx, Inc. Methods of using ox40 ligand encoding polynucleotides
EP4039699A1 (en) 2015-12-23 2022-08-10 ModernaTX, Inc. Methods of using ox40 ligand encoding polynucleotides
WO2017115268A1 (en) 2015-12-28 2017-07-06 Novartis Ag Compositions and methods for the treatment of hemoglobinopathies
EP4053277A1 (en) 2015-12-28 2022-09-07 Novartis AG Compositions and methods for the treatment of hemoglobinopathies
WO2017120612A1 (en) 2016-01-10 2017-07-13 Modernatx, Inc. Therapeutic mrnas encoding anti ctla-4 antibodies
EP3701959A1 (en) 2016-03-21 2020-09-02 BioNTech RNA Pharmaceuticals GmbH Rna replicon for versatile and efficient gene expression
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
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
WO2018091540A1 (en) 2016-11-17 2018-05-24 Glaxosmithkline Biologicals Sa Zika viral antigen constructs
EP4043031A1 (en) 2016-11-17 2022-08-17 GlaxoSmithKline Biologicals SA Zika viral antigen constructs
EP3808380A1 (en) 2016-12-08 2021-04-21 CureVac AG Rna for treatment or prophylaxis of a liver disease
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
WO2018142364A1 (en) 2017-02-06 2018-08-09 Novartis Ag Compositions and methods for the treatment of hemoglobinopathies
US11466271B2 (en) 2017-02-06 2022-10-11 Novartis Ag Compositions and methods for the treatment of hemoglobinopathies
US10857105B2 (en) 2017-03-15 2020-12-08 MordernaTX, Inc. Compounds and compositions for intracellular delivery of therapeutic agents
US11203569B2 (en) 2017-03-15 2021-12-21 Modernatx, Inc. Crystal forms of amino lipids
US11969506B2 (en) 2017-03-15 2024-04-30 Modernatx, Inc. Lipid nanoparticle formulation
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
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
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
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
WO2019104195A1 (en) 2017-11-22 2019-05-31 Modernatx, Inc. Polynucleotides encoding propionyl-coa carboxylase alpha and beta subunits for the treatment of propionic acidemia
WO2019104152A1 (en) 2017-11-22 2019-05-31 Modernatx, Inc. Polynucleotides encoding ornithine transcarbamylase for the treatment of urea cycle disorders
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
US11453639B2 (en) 2019-01-11 2022-09-27 Acuitas Therapeutics, Inc. Lipids for lipid nanoparticle delivery of active agents
WO2020227642A1 (en) 2019-05-08 2020-11-12 Modernatx, Inc. Compositions for skin and wounds and methods of use thereof
WO2020263985A1 (en) 2019-06-24 2020-12-30 Modernatx, Inc. Messenger rna comprising functional rna elements and uses thereof
WO2020263883A1 (en) 2019-06-24 2020-12-30 Modernatx, Inc. Endonuclease-resistant messenger rna and uses thereof
WO2021009336A1 (en) 2019-07-18 2021-01-21 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for inducing full ablation of hematopoiesis
WO2021013798A1 (en) 2019-07-21 2021-01-28 Glaxosmithkline Biologicals Sa Therapeutic viral vaccine
US11597698B2 (en) 2019-09-19 2023-03-07 Modernatx, Inc. Branched tail lipid compounds and compositions for intracellular delivery of therapeutic agents
US11066355B2 (en) 2019-09-19 2021-07-20 Modernatx, Inc. Branched tail lipid compounds and compositions for intracellular delivery of therapeutic agents
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
WO2021123920A1 (en) 2019-12-18 2021-06-24 Novartis Ag Compositions and methods for the treatment of hemoglobinopathies
US11964011B2 (en) 2020-02-04 2024-04-23 CureVac SE Coronavirus vaccine
US11964012B2 (en) 2020-02-04 2024-04-23 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
WO2021209970A1 (en) 2020-04-16 2021-10-21 Glaxosmithkline Biologicals Sa Sars cov-2 spike protein construct
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
US11976019B2 (en) 2020-07-16 2024-05-07 Acuitas Therapeutics, Inc. Cationic lipids for use in lipid nanoparticles
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
US11918643B2 (en) 2020-12-22 2024-03-05 CureVac SE RNA vaccine against SARS-CoV-2 variants
US11872280B2 (en) 2020-12-22 2024-01-16 CureVac SE RNA vaccine against SARS-CoV-2 variants
WO2022137128A2 (en) 2020-12-23 2022-06-30 Glaxosmithkline Biologicals Sa Self-amplifying messenger rna
WO2022157153A2 (en) 2021-01-20 2022-07-28 Glaxosmithkline Biologicals Sa Therapeutic viral vaccine
EP4032546A1 (en) 2021-01-20 2022-07-27 GlaxoSmithKline Biologicals S.A. Therapeutic viral vaccine
US11622972B2 (en) 2021-02-19 2023-04-11 Modernatx, Inc. Lipid nanoparticle compositions and methods of formulating the same
US11524023B2 (en) 2021-02-19 2022-12-13 Modernatx, Inc. Lipid nanoparticle compositions and methods of formulating the same
WO2022204390A1 (en) 2021-03-24 2022-09-29 Modernatx, Inc. Lipid nanoparticles containing polynucleotides encoding phenylalanine hydroxylase and uses thereof
WO2022204370A1 (en) 2021-03-24 2022-09-29 Modernatx, Inc. Lipid nanoparticles and polynucleotides encoding ornithine transcarbamylase for the treatment of ornithine transcarbamylase deficiency
WO2022204371A1 (en) 2021-03-24 2022-09-29 Modernatx, Inc. Lipid nanoparticles containing polynucleotides encoding glucose-6-phosphatase and uses thereof
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
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
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
WO2022269518A2 (en) 2021-06-23 2022-12-29 Novartis Ag Compositions and methods for the treatment of hemoglobinopathies
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
WO2023021427A1 (en) 2021-08-16 2023-02-23 Glaxosmithkline Biologicals Sa Freeze-drying of lipid nanoparticles (lnps) encapsulating rna and formulations thereof
WO2023020993A1 (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
WO2023020992A1 (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
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
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
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
WO2023183909A2 (en) 2022-03-25 2023-09-28 Modernatx, Inc. Polynucleotides encoding fanconi anemia, complementation group proteins for the treatment of fanconi anemia
WO2023213783A1 (en) 2022-05-02 2023-11-09 BioNTech SE Replicon compositions and methods of using same for the treatment of diseases
WO2023213378A1 (en) 2022-05-02 2023-11-09 BioNTech SE Replicon compositions and methods of using same for the treatment of diseases
WO2023242817A2 (en) 2022-06-18 2023-12-21 Glaxosmithkline Biologicals Sa Recombinant rna molecules comprising untranslated regions or segments encoding spike protein from the omicron strain of severe acute respiratory coronavirus-2
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
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
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
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
AU2011295938B2 (en) 2016-01-14
CN103384515B (en) 2017-02-15
CA2809863A1 (en) 2012-03-08
TR201908635T4 (en) 2019-07-22
AU2016201063A1 (en) 2016-03-10
MX2013002332A (en) 2013-03-18
JP2016006128A (en) 2016-01-14
ES2727583T3 (en) 2019-10-17
AU2018203680A1 (en) 2018-06-14
EP3542789A2 (en) 2019-09-25
BR112013004865A2 (en) 2016-06-07
RU2577983C2 (en) 2016-03-20
EP3542789A3 (en) 2020-01-01
JP5908477B2 (en) 2016-04-26
US20130189351A1 (en) 2013-07-25
EP2611420B1 (en) 2019-03-27
CN103384515A (en) 2013-11-06
RU2013114330A (en) 2014-10-10
WO2012031046A3 (en) 2014-04-03
JP2014505007A (en) 2014-02-27
EP2611420A2 (en) 2013-07-10
AU2011295938A1 (en) 2013-04-04

Similar Documents

Publication Publication Date Title
US11759422B2 (en) Pegylated liposomes for delivery of immunogen-encoding RNA
US11883534B2 (en) Immunisation with lipid formulations with RNA encoding immunogens
EP2611420B1 (en) Lipids suitable for liposomal delivery of protein-coding rna
EP2611467A1 (en) Small 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: 11763813

Country of ref document: EP

Kind code of ref document: A2

ENP Entry into the national phase

Ref document number: 2013526211

Country of ref document: JP

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2809863

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: MX/A/2013/002332

Country of ref document: MX

WWE Wipo information: entry into national phase

Ref document number: 2011763813

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2013114330

Country of ref document: RU

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2011295938

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: 13819228

Country of ref document: US

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112013004865

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 112013004865

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20130228