WO2012061259A2 - Novel low molecular weight cyclic amine containing cationic lipids for oligonucleotide delivery - Google Patents

Novel low molecular weight cyclic amine containing cationic lipids for oligonucleotide delivery Download PDF

Info

Publication number
WO2012061259A2
WO2012061259A2 PCT/US2011/058498 US2011058498W WO2012061259A2 WO 2012061259 A2 WO2012061259 A2 WO 2012061259A2 US 2011058498 W US2011058498 W US 2011058498W WO 2012061259 A2 WO2012061259 A2 WO 2012061259A2
Authority
WO
WIPO (PCT)
Prior art keywords
lipid
alkyl
cationic lipids
sirna
alkenyl
Prior art date
Application number
PCT/US2011/058498
Other languages
French (fr)
Other versions
WO2012061259A3 (en
Inventor
Matthew G. Stanton
Gregory L. Beutner
Original Assignee
Merck Sharp & Dohme Corp.
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 Merck Sharp & Dohme Corp. filed Critical Merck Sharp & Dohme Corp.
Priority to EP11838596.2A priority Critical patent/EP2635265B1/en
Priority to US13/883,487 priority patent/US9067882B2/en
Priority to DK11838596.2T priority patent/DK2635265T3/en
Publication of WO2012061259A2 publication Critical patent/WO2012061259A2/en
Publication of WO2012061259A3 publication Critical patent/WO2012061259A3/en
Priority to US14/719,513 priority patent/US9796977B2/en
Priority to US15/706,041 priority patent/US10337014B2/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • C12N15/1137Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against enzymes
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D207/00Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D207/02Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D207/04Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members
    • C07D207/10Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D207/12Oxygen or sulfur atoms
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/51Nanocapsules; Nanoparticles
    • A61K9/5107Excipients; Inactive ingredients
    • A61K9/5123Organic compounds, e.g. fats, sugars
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D211/00Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings
    • C07D211/04Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D211/06Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members
    • C07D211/36Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D211/40Oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D223/00Heterocyclic compounds containing seven-membered rings having one nitrogen atom as the only ring hetero atom
    • C07D223/02Heterocyclic compounds containing seven-membered rings having one nitrogen atom as the only ring hetero atom not condensed with other rings
    • C07D223/06Heterocyclic compounds containing seven-membered rings having one nitrogen atom as the only ring hetero atom not condensed with other rings with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D223/08Oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • 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
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/14Type of nucleic acid interfering N.A.
    • 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
    • C12N2320/00Applications; Uses
    • C12N2320/30Special therapeutic applications
    • C12N2320/32Special delivery means, e.g. tissue-specific

Definitions

  • the present invention relates to novel cationic lipids that can be used in combination with other lipid components such as cholesterol and PEG-lipids to form lipid nanoparticles with oligonucleotides, to facilitate the cellular uptake and endosomal escape, and to knockdown target mRNA both in vitro and in vivo.
  • Cationic lipids and the use of cationic lipids in lipid nanoparticles for the delivery of oligonucleotides, in particular siRNA and miRNA have been previously disclosed.
  • Lipid nanoparticles and use of lipid nanoparticles for the delivery of oligonucleotides, in particular siRNA and miRNA has been previously disclosed.
  • Oligonucleotides (including siRNA and miRNA) and the synthesis of oligonucleotides has been previously disclosed. (See US patent applications: US 2006/0083780, US 2006/0240554, US 2008/0020058, US 2009/0263407 and US 2009/0285881 and PCT patent applications: WO 2009/086558, WO2009/127060,
  • WO2009/132131, WO2010/042877, WO2010/054384, WO2010/054401, WO2010/054405, WO2010/054406 and WO2010/105209) See also Semple S. C. et al., Rational design of cationic lipids for siRNA delivery, Nature Biotechnology, 2010, 28, 172-176.
  • Other cationic lipids are disclosed in US patent applications: US 2009/0263407, US 2009/0285881, US 2010/0055168, US 2010/0055169, US 2010/0063135, US 2010/0076055, US 2010/0099738 and US
  • the present invention employs low molecular weight cationic lipids comprising at least one short lipid chain to enhance the efficacy and tolerability of in vivo delivery of siRNA.
  • the instant invention provides for novel cationic lipids that can be used in combination with other lipid components such as cholesterol and PEG-lipids to form lipid nanoparticles with oligonucleotides. It is an object of the instant invention to provide a cationic lipid scaffold that demonstrates enhanced efficacy along with lower liver toxicity as a result of lower lipid levels in the liver.
  • the present invention employs low molecular weight cationic lipids with one short lipid chain to enhance the efficiency and tolerability of in vivo delivery of siRNA.
  • FIGURE 1 Comparison of LNP (Compound 4) efficacy in mice.
  • FIGURE 2 LNP (Compound 4) efficacy and tolerability in rats.
  • FIGURE 3 Cationic lipid (Compound 4) levels in rat liver.
  • the various aspects and embodiments of the invention are directed to the utility of novel cationic lipids useful in lipid nanoparticles to deliver oligonucleotides, in particular, siRNA and miRNA, to any target gene.
  • novel cationic lipids useful in lipid nanoparticles to deliver oligonucleotides, in particular, siRNA and miRNA, to any target gene.
  • WO2010/054384 WO2010/054401, WO2010/054405, WO2010/054406 and WO2010/105209.
  • WO2010/054384 Rational design of cationic lipids for siRNA delivery, Nature Biotechnology, published online 17 January 2010; doi:10.1038/nbt.l602.
  • the cationic lipids of the instant invention are useful components in a lipid nanoparticle for the delivery of oligonucleotides, specifically siRNA and miRNA.
  • R 1 is selected from H, (C 1 -C 6 )alkyl, heterocyclyl, polyether and polyamine, wherein said alkyl, heterocyclyl, polyether and polyamine are optionally substituted with one to three substituents selected from R';
  • n and m are independently selected from 0, 1, 2 and 3;
  • L 1 is selected from C 4 -C 24 alkyl and C 4 -C 24 alkenyl, said alkyl and alkenyl are optionally substituted with one or more substituents selected from R';
  • L 2 is selected from C 3 -C 9 alkyl and C 3 -C 9 alkenyl, said alkyl and alkenyl are optionally substituted with one or more substituents selected from R';
  • R' is independently selected from halogen, R", OR", SR", CN, CO2R" and
  • R" is independently selected from H and (C 1 -C 6 )alkyl
  • the invention features a compound having Formula A, wherein:
  • R is H or methyl
  • n and m are 1;
  • X is O
  • L 1 is selected from C 12 -C 24 alkyl and C 12 -C 24 alkenyl
  • L 2 is selected from C 3 -C 9 alkyl and C 3 -C 9 alkenyl
  • Specific cationic lipids are:
  • the cationic lipids disclosed are useful in the preparation of lipid nanoparticles.
  • the cationic lipids disclosed are useful components in a lipid nanoparticle for the delivery of oligonucleotides.
  • the cationic lipids disclosed are useful components in a lipid nanoparticle for the delivery of siRNA and miRNA.
  • the cationic lipids disclosed are useful components in a lipid nanoparticle for the delivery of siRNA.
  • the cationic lipids of the present invention may have asymmetric centers, chiral axes, and chiral planes (as described in: E.L. Eliel and S.H. Wilen, Stereochemistry of Carbon Compounds, John Wiley & Sons, New York, 1994, pages 1119-1190), and occur as racemates, racemic mixtures, and as individual diastereomers, with all possible isomers and mixtures thereof, including optical isomers, being included in the present invention.
  • the cationic lipids disclosed herein may exist as tautomers and both tautomeric forms are intended to be encompassed by the scope of the invention, even though only one tautomeric structure is depicted.
  • substituents and substitution patterns on the cationic lipids of the instant invention can be selected by one of ordinary skill in the art to provide cationic lipids that are chemically stable and that can be readily synthesized by techniques known in the art, as well as those methods set forth below, from readily available starting materials. If a substituent is itself substituted with more than one group, it is understood that these multiple groups may be on the same carbon or on different carbons, so long as a stable structure results.
  • Si atoms can be incorporated into the cationic lipids of the instant invention by one of ordinary skill in the art to provide cationic lipids that are chemically stable and that can be readily synthesized by techniques known in the art from readily available starting materials.
  • the atoms may exhibit their natural isotopic abundances, or one or more of the atoms may be artificially enriched in a particular isotope having the same atomic number, but an atomic mass or mass number different from the atomic mass or mass number predominantly found in nature.
  • the present invention is meant to include all suitable isotopic variations of the compounds of Formula A.
  • different isotopic forms of hydrogen (H) include protium ( 1 H) and deuterium ( 2 H).
  • Protium is the predominant hydrogen isotope found in nature. Enriching for deuterium may afford certain therapeutic advantages, such as increasing in vivo half-life or reducing dosage requirements, or may provide a compound useful as a standard for characterization of biological samples.
  • Isotopically-enriched compounds within Formula A can be prepared without undue experimentation by conventional techniques well known to those skilled in the art or by processes analogous to those described in the Scheme and Examples herein using appropriate isotopically-enriched reagents and/or intermediates.
  • alkyl means a straight chain, cyclic or branched saturated aliphatic hydrocarbon having the specified number of carbon atoms.
  • alkenyl means a straight chain, cyclic or branched unsaturated aliphatic hydrocarbon having the specified number of carbon atoms including but not limited to diene, triene and tetraene unsaturated aliphatic hydrocarbons.
  • heterocyclyl or “heterocycle” means a 4- to 10-membered aromatic or nonaromatic heterocycle containing from 1 to 4 heteroatoms selected from the group consisting of O, N and S, and includes bicyclic groups.
  • Heterocyclyl therefore includes, the following: benzoimidazolyl, benzofuranyl, benzofurazanyl, benzopyrazolyl, benzotriazolyl, benzothiophenyl, benzoxazolyl, carbazolyl, carbolinyl, cinnolinyl, furanyl, imidazolyl, indolinyl, indolyl, indolazinyl, indazolyl, isobenzofuranyl, isoindolyl, isoquinolyl, isothiazolyl, isoxazolyl, naphthpyridinyl, oxadiazolyl, oxazolyl, oxazoline, isoxazoline, oxetanyl, pyranyl, pyrazinyl, pyrazolyl, pyridazinyl, pyridopyridinyl, pyridazinyl, pyridyl, pyri
  • polyether means compounds having two or more ether groups. Examples include poly(ethylene) glycols.
  • polyamine means compounds having two or more amino groups. Examples include putrescine, cadaverine, spermidine, and spermine.
  • halogen means Br, Cl, F and I.
  • R 1 is selected from H and (C 1 -C 6 )alkyl, wherein said alkyl is optionally substituted with one to three substituents selected from R'.
  • R 1 is selected from H, methyl, ethyl and propyl, wherein said methyl, ethyl and propyl are optionally substituted with one to three substituents selected from R'.
  • R 1 is selected from H, methyl, ethyl and propyl. In an embodiment of Formula A, R 1 is methyl.
  • R' is is selected from H, methyl, ethyl and propyl, wherein said methyl, ethyl and propyl are optionally substituted with one or more halogen and OH.
  • n 0, 1, 2 or 3.
  • n 1 or 2.
  • n 1
  • m is 0, 1 , 2 or 3.
  • m is 1 or 2.
  • m is 1.
  • X is O.
  • L 1 is selected from C 4 -C 24 alkyl and C 4 -C 24 alkenyl, which are optionally substituted with halogen and OH. In an embodiment of Formula A, L 1 is selected from C 4 -C 24 alkyl and C 4 -C 24 alkenyl.
  • L 1 is selected from C 12 -C 24 alkyl and C 12 - C 24 alkenyl, which are optionally substituted with halogen and OH.
  • L 1 is selected from C 12 -C 24 alkyl and C 12 -
  • L 1 is selected from C 4 -C 24 alkenyl.
  • L 1 is selected from C 12 -C 24 alkenyl.
  • L 1 is C 18 alkenyl.
  • L 1 is:
  • L 2 is selected from C 3 -C 9 alkyl and C 3 -C 9 alkenyl, which are optionally substituted with halogen and OH.
  • L 2 is selected from C 5 -C 9 alkyl and C 5 -C 9 alkenyl, which are optionally substituted with halogen and OH.
  • L 2 is selected from C 7 -C 9 alkyl and C 7 -C 9 alkenyl, which are optionally substituted with halogen and OH.
  • L 2 is selected from C 3 -C 9 alkyl and C 3 -C 9 alkenyl.
  • L 2 is selected from C 5 -C 9 alkyl and C 5 -C 9 alkenyl.
  • L 2 is selected from C 7 -C 9 alkyl and C 7 -C 9 alkenyl.
  • L 2 is C 3 -C 9 alkyl.
  • L 2 is C 5 -C 9 alkyl.
  • L 2 is C 7 -C 9 alkyl.
  • L 2 is C 8 alkyl.
  • heterocyclyl is pyrolidine, piperidine, morpholine, imidazole or piperazine.
  • polyamine is putrescine, cadaverine, spermidine or spermine.
  • polyether is poly(ethylene) glycol(s).
  • alkyl is a straight chain saturated aliphatic hydrocarbon having the specified number of carbon atoms.
  • alkenyl is a straight chain unsaturated aliphatic hydrocarbon having the specified number of carbon atoms.
  • the free form of cationic lipids of Formula A includes the free form of cationic lipids of Formula A, as well as the pharmaceutically acceptable salts and stereoisomers thereof.
  • Some of the isolated specific cationic lipids exemplified herein are the protonated salts of amine cationic lipids.
  • the term “free form” refers to the amine cationic lipids in non-salt form.
  • the encompassed pharmaceutically acceptable salts not only include the isolated salts exemplified for the specific cationic lipids described herein, but also all the typical pharmaceutically acceptable salts of the free form of cationic lipids of Formula A.
  • the free form of the specific salt cationic lipids described may be isolated using techniques known in the art.
  • the free form may be regenerated by treating the salt with a suitable dilute aqueous base solution such as dilute aqueous NaOH, potassium carbonate, ammonia and sodium bicarbonate.
  • a suitable dilute aqueous base solution such as dilute aqueous NaOH, potassium carbonate, ammonia and sodium bicarbonate.
  • the free forms may differ from their respective salt forms somewhat in certain physical properties, such as solubility in polar solvents, but the acid and base salts are otherwise pharmaceutically equivalent to their respective free forms for purposes of the invention.
  • the pharmaceutically acceptable salts of the instant cationic lipids can be synthesized from the cationic lipids of this invention which contain a basic or acidic moiety by conventional chemical methods.
  • the salts of the basic cationic lipids are prepared either by ion exchange chromatography or by reacting the free base with stoichiometric amounts or with an excess of the desired salt-forming inorganic or organic acid in a suitable solvent or various combinations of solvents.
  • the salts of the acidic compounds are formed by reactions with the appropriate inorganic or organic base.
  • compositions of the cationic lipids of this invention include the conventional non-toxic salts of the cationic lipids of this invention as formed by reacting a basic instant cationic lipids with an inorganic or organic acid.
  • conventional non-toxic salts include those derived from inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, nitric and the like, as well as salts prepared from organic acids such as acetic, propionic, succinic, glycolic, stearic, lactic, malic, tartaric, citric, ascorbic, pamoic, maleic, hydroxymaleic, phenylacetic, glutamic, benzoic, salicylic, sulfanilic, 2-acetoxy-benzoic, fumaric, toluenesulfonic, methanesulfonic, ethane disulfonic, oxalic, isethionic, trifluoroacetic (
  • salts derived from inorganic bases include aluminum, ammonium, calcium, copper, ferric, ferrous, Uthium, magnesium, manganic salts, manganous, potassium, sodium, zinc and the like. Particularly preferred are the ammonium, calcium, magnesium, potassium and sodium salts. Salts derived from
  • organic non-toxic bases include salts of primary, secondary and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, such as arginine, betaine caffeine, choline, ⁇ , ⁇ 1 - dibenzylethylenediamine, diethylamin, 2-diethylaminoethanol, 2-dimethylaminoethanol, ethanolamine, ethylenediamine, N-ethylmorpholine, N-ethylpiperidine, glucamine, glucosamine, histidine, hydrabamine, isopropylainine, lysine, methylglucamine, morpholine, piperazine, piperidine, polyamine resins, procaine, purines, theobromine, triethylamine, trimethylamine tripropylamine, tromethamine and the like.
  • basic ion exchange resins such as arginine, betaine caffeine, choline
  • the cationic lipids of the present invention are potentially internal salts or zwitterions, since under physiological conditions a deprotonated acidic moiety in the compound, such as a carboxyl group, may be anionic, and this electronic charge might then be balanced off internally against the cationic charge of a protonated or alkylated basic moiety, such as a quaternary nitrogen atom.
  • Synthesis of cyclic amine containing cationic lipids is a linear process starting from cyclic dihydroxy amines i. Stepwise etherification to give ii followed by cleavage of the benzyl group to give iii and reductive amination gives products of the type iv.
  • 1-Benzyl-3,4 dihydroxy pyrroldine benzene sulfonic acid salt (1) In a 3N 5L RBF with overhead stirrer and reflux condenser, the DL-l,4-dibromo butane diol (100 g, 403 mmol), benzyl amine (39.7 ml, 363 mmol), potassium iodide (3.35 g, 20.17 mmol) and diisopropylethylamine
  • Benzyl 3,4-dihvdroxypyrrolidine-l-carboxylate (9) In a 500 mL RBF, the Cbz Pyrroline (15 g s 73.8 mmol) was dissolved in a mixture of 100 mL THF, 60 mL t-BuOH and 40 mL water for addition of the NMO (30.6 ml, 148 mmol) solution followed by the osmium tetraoxide (0.925 ml, 0.074 mmol) solution. Heated in a 70C oil bath. Solution slowly went from yellow to brown. After 2h, cooled to RT and added 50 mL 10% aqueous NaHS03.
  • Compound 12 is DLinKC2DMA as described in Nature Biotechnology, 2010, 28, 172-176, WO 2010/042877 Al, WO 2010/048536 A2, WO 2010/088537 A2, and
  • Compound 13 is MC3 as described in WO 2010/054401, and WO 2010/144740
  • lipid nanoparticle compositions of the instant invention are useful for the delivery of oligonucleotides, specifically siRNA and miRNA:
  • the Lipid Nano-Particles are prepared by an impinging jet process.
  • the particles are formed by mixing lipids dissolved in alcohol with siRNA dissolved in a citrate buffer.
  • the mixing ratio of lipids to siRNA are targeted at 45-55% lipid and 65-45% siRNA.
  • the lipid solution contains a novel cationic lipid of the instant invention, a helper lipid
  • the ratio of the lipids has a mole percent range of 25-98 for the cationic lipid with a target of 35-65, the helper lipid has a mole percent range from 0-75 with a target of 30-50, the PEG lipid has a mole percent range from 1-15 with a target of 1 -6, and the DSPC has a mole precent range of 0-15 with a target of 0-12.
  • the siRNA solution contains one or more siRNA sequences at a concentration range from 0.3 to 1 .0 mg mL with a target of 0.3 -0.9 mg/mL in a sodium citrate buffered salt solution with pH in the range of 3.5-5.
  • the two liquids are heated to a temperature in the range of 15-40°C S targeting 30-40°C, and then mixed in an impinging jet mixer instantly forming the LNP.
  • the teelD has a range from 0.25 to 1.0 mm and a total flow rate from 10 -600 mL/min.
  • the combination of flow rate and tubing ID has effect of controlling the particle size of the LNPs between 30 and 200 nm.
  • the solution is then mixed with a buffered solution at a higher pH with a mixing ratio in the range of 1 :1 to 1 :3 vol:vol but targeting 1 :2 voi:vol.
  • This buffered solution is at a temperature in the range of 15-40°C, targeting 30-40°C.
  • the mixed LNPs are held from 30 minutes to 2 hrs prior to an anion exchange filtration step.
  • the temperature during incubating is in the range of 15-40°C, targeting 30-40°C.
  • After incubating the solution is filtered through a 0.8 urn filter containing an anion exchange separation step. This process uses tubing IDs ranging from 1 mm ID to 5 mm ID and a flow rate from 10 to 2000 mL/min.
  • the LNPs are concentrated and diafiltered via an ultrafiltration process where the alcohol is removed and the citrate buffer is exchanged for the final buffer solution such as phosphate buffered saline.
  • the ultrafiltration process uses a tangential flow filtration format (TFF). This process uses a membrane nominal molecular weight cutoff range from 30 -500 D.
  • the membrane format can be hollow fiber or flat sheet cassette.
  • the TFF processes with the proper molecular weight cutoff retains the LNP in the retentate and the filtrate or permeate contains the alcohol; citrate buffer; final buffer wastes.
  • the TFF process is a multiple step process with an initial concentration to a siRNA concentration of 1 -3 mg/mL.
  • the LNPs solution is diafiltered against the final buffer for 10 -20 volumes to remove the alcohol and perform buffer exchange. The material is then concentrated an additional 1-3 fold. The final steps of the LNP process are to sterile filter the concentrated LNP solution and vial the product.
  • siRNA concentration The siRNA duplex concentrations are determined by Strong Anion-Exchange High-Performance Liquid Chromatography (SAX-HPLC) using Waters 2695 Alliance system (Water Corporation, Milford MA) with a 2996 PDA detector.
  • the LNPs otherwise referred to as RNAi Delivery Vehicles (RDVs), are treated with 0.5% Triton X- 100 to free total siRNA and analyzed by SAX separation using a Dionex BioLC DNAPac PA 200 (4 ⁇ 250 mm) column with UV detection at 254 nm.
  • Mobile phase is composed of A: 25 mM NaC10 4 , 10 mM Tris, 20% EtOH, pH 7.0 and B: 250 mM NaC10 4 , 10 mM Tris, 20% EtOH, pH 7.0 with liner gradient from 0-15 min and flow rate of 1 ml/min.
  • the siRNA amount is determined by comparing to the siRNA standard curve.
  • Fluorescence reagent S YBR Gold is employed for RNA quantitation to monitor the encapsulation rate of RDVs.
  • RDVs with or without Triton X-100 are used to determine the free siRNA and total siRNA amount.
  • the assay is performed using a SpectraMax M5e microplate spectrophotometer from Molecular Devices (Sunnyvale, CA). Samples are excited at 485 nm and fluorescence emission was measured at 530 nm. The siRNA amount is determined by comparing to the siRNA standard curve.
  • Encapsulation rate (1- free siRNA/total siRNA) xl00%
  • RDVs containing 1 g siRNA are diluted to a final volume of 3 ml with 1 ⁇ PBS.
  • the particle size and polydispersity of the samples is measured by a dynamic light scattering method using ZetaPALS instrument (Brookhaven Instruments Corporation, Holtsville, NY).
  • the scattered intensity is measured with He-Ne laser at 25°C with a scattering angle of 90°.
  • RDVs containing 1 ⁇ g siRNA are diluted to a final volume of 2 ml with 1 mM Tris buffer (pH 7.4). Electrophoretic mobility of samples is determined using ZetaPALS instrument (Brookhaven Instruments Corporation, Holtsville, NY) with electrode and He-Ne laser as a light source. The Smoluchowski limit is assumed in the calculation of zeta potentials.
  • lipid concentrations are determined by Reverse Phase High- Performance Liquid Chromatography (RP-HPLC) using Waters 2695 Alliance system (Water Corporation, Milford MA) with a Corona charged aerosol detector (CAD) (ESA Biosciences, Inc, Chelmsford, MA). Individual lipids in RDVs are analyzed using an Agilent Zorbax SB-C18 (50 x 4.6 mm, 1.8 ⁇ particle size) column with CAD at 60 °C. The mobile phase is composed of A: 0.1% TFA in H 2 0 and B: 0.1% TFA in IPA.
  • the gradient changes from 60% mobile phase A and 40% mobile phase B from time 0 to 40% mobile phase A and 60% mobile phase B at 1.00 min; 40% mobile phase A and 60% mobile phase B from 1.00 to 5.00 min; 40% mobile phase A and 60% mobile phase B from 5.00 min to 25% mobile phase A and 75% mobile phase B at 10.00 min; 25% mobile phase A and 75% mobile phase B from 10.00 min to 5% mobile phase A and 95% mobile phase B at 15.00 min; and 5% mobile phase A and 95% mobile phase B from 15.00 to 60% mobile phase A and 40% mobile phase B at 20.00 min with flow rate of 1 ml/min.
  • the individual lipid concentration is determined by comparing to the standard curve with all the lipid components in the RDVs with a quadratic curve fit. The molar percentage of each lipid is calculated based on its molecular weight.
  • Oligonucleotide synthesis is well known in the art. (See US patent applications: US 2006/0083780, US 2006/0240554, US 2008/0020058, US 2009/0263407 and US
  • WO2009/132131, WO2010/042877, WO2010/054384, WO2010/054401, WO2010/054405 and WO2010/054406) were synthesized via standard solid phase procedures.
  • the siRNA targets the mRNA transcript for the firefly (Photinus pyralis) luciferase gene (Accession # Ml 5077).
  • the primary sequence and chemical modification pattern of the luciferase siRNA is displayed above.
  • the in vivo luciferase model employs a transgenic mouse in which the firefly luciferase coding sequence is present in all cells.
  • ROSA26- LoxP-Stop-LoxP-Luc (LSL-Luc) transgenic mice licensed from the Dana Farber Cancer Institute are induced to express the Luciferase gene by first removing the LSL sequence with a recombinant Ad-Cre virus (Vector Biolabs). Due to the organo-tropic nature of the virus, expression is limited to the liver when delivered via tail vein injection. Luciferase expression levels in liver are quantitated by measuring light output, using an IVIS imager (Xenogen) following administration of the luciferin substrate (Caliper Life Sciences). Pre-dose luminescence levels are measured prior to administration of the RDVs.
  • Luciferin in PBS 15mg/mL is intraperitoneally (IP) injected in a volume of 150 ⁇ L ⁇ After a four minute incubation period mice are anesthetized with isoflurane and placed in the IVIS imager.
  • the RDVs (containing siRNA) in PBS vehicle were tail vein injected n a volume of 0.2 mL. Final dose levels ranged from 0.1 to 0.5 mg/kg siRNA.
  • PBS vehicle alone was dosed as a control.
  • Mice were imaged 48 hours post dose using the method described above. Changes in luciferin light output directly correlate with luciferase mRNA levels and represent an indirect measure of luciferase siRNA activity.
  • LNPs utilizing compounds in the nominal compositions described above were evaluated for in vivo efficacy and increases in alanine amino transferase and aspartate amino transferase in Sprague-Dawley (Crl:CD(SD) female rats (Charles River Labs).
  • the siRNA targets the mRNA transcript for the ApoB gene (Accession # NM 019287).
  • the primary sequence and chemical modification pattern of the ApoB siRNA is displayed above.
  • the RDVs (containing siRNA) in PBS vehicle were tail vein injected in a volume of 1 to 1.5 mL. Infusion rate is approximately 3 ml/min. Five rats were used in each dosing group. After LNP
  • rats are placed in cages with normal diet and water present. Six hours post dose, food is removed from the cages. Animal necropsy is performed 24 hours after LNP dosing. Rats are anesthetized under isoflurane for 5 minutes, then maintained under anesthesia by placing them in nose cones continuing the delivery of isoflurane until ex-sanguination is completed. Blood is collected from the vena cava using a 23 gauge butterfly venipuncture set and aliquoted to serum separator vacutainers for serum chemistry analysis. Punches of the excised caudate liver lobe are taken and placed in RNALater (Ambion) for mRNA analysis.
  • RNALater RNALater
  • liver tissue was homogenized and total RNA isolated using a Qiagen bead mill and the Qiagen miRNA-Easy RNA isolation kit following the manufacturer's instructions.
  • Liver ApoB mRNA levels were determined by quantitative RT-PCR. Message was amplified from purified RNA utilizing a rat ApoB commercial probe set (Applied Biosystems Cat # RN01499054_ml). The PCR reaction was performed on an ABI 7500 instrument with a 96-well Fast Block. The ApoB mRNA level is normalized to the housekeeping PPIB (NM 011149) mRNA. ⁇ mRNA levels were determined by RT-PCR using a commercial probe set (Applied Biosytems Cat. No.
  • results are expressed as a ratio of ApoB mRNA/ PPIB mRNA. All mRNA data is expressed relative to the PBS control dose.
  • Serum ALT and AST analysis were performed on the Siemens Advia 1800 Clinical Chemistry Analyzer utilizing the Siemens alanine aminotransferase (Cat# 03039631) and aspartate aminotransferase (Cat# 03039631) reagents. Modestly decreased efficacy but significantly improved tolerability was observed in rats dosed with Compound 4 containing RDV than with the RDV containing the cationic lipid
  • Liver tissue was weighed into 20-ml vials and homogenized in 9 v/w of water using a GenoGrinder 2000 (OPS Diagnostics, 1600 strokes/min, 5min). A 50 uL aliquot of each tissue homogenate was mixed with 300 L of extraction/protein precipitating solvent (50/50 acetonitrile methanol containing 500 nM internal standard) and the plate was centrifuged to sediment precipitated protein. A volume of 200 ⁇ L of each supernatant was then transferred to separate wells of a 96-well plate and 10 ⁇ L samples were directly analyzed by LC MS-MS.
  • OPS Diagnostics 1600 strokes/min, 5min.
  • a 50 uL aliquot of each tissue homogenate was mixed with 300 L of extraction/protein precipitating solvent (50/50 acetonitrile methanol containing 500 nM internal standard) and the plate was centrifuged to sediment precipitated protein. A volume of 200 ⁇ L of each supernatant was then transferred to separate well
  • Absolute quantification versus standards prepared and extracted from liver homogenate was performed using an Aria LX-2 HPLC system (Thermo Scientific) coupled to an API 4000 triple quadrupole mass spectrometer (Applied Biosystems). For each run, a total of 10 ⁇ sample was injected onto a BDS Hypersil C8 HPLC column (Thermo, 50 x 2mm, 3 ⁇ ) at ambient temperature.
  • Mobile Phase A 95% H20/5% methanol/10 mM ammonium formate/0. l%formic acid
  • Mobile Phase B 40% methanol/60% n-propanol/10 mM ammonium formate/0. l%formic acid
  • the flow rate was 0.5 mL/min and gradient elution profile was as follows: hold at 80% A for 0.25 min, linear ramp to 100% B over 1.6 min, hold at 100% B for 2.5 min, then return and hold at 80% A for 1.75 min. Total run time was 5.8 min.
  • API 4000 source parameters were CAD: 4, CUR: 15, GS1: 65, GS2: 35, IS: 4000, TEM: 550, CXP: 15, DP: 60, EP: 10.
  • liver levels were much lower than the RDV containing the cationic lipid DLinKC2DMA (Compound 12, also 0.5 mg/kg) or MC3 (Compound 13, also 0.5 mg/kg, Figure 3).

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Genetics & Genomics (AREA)
  • Biomedical Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Wood Science & Technology (AREA)
  • General Engineering & Computer Science (AREA)
  • Biotechnology (AREA)
  • Molecular Biology (AREA)
  • Zoology (AREA)
  • General Health & Medical Sciences (AREA)
  • Physics & Mathematics (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Microbiology (AREA)
  • Plant Pathology (AREA)
  • Epidemiology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Medicinal Chemistry (AREA)
  • Optics & Photonics (AREA)
  • Animal Behavior & Ethology (AREA)
  • Virology (AREA)
  • Nanotechnology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

The instant invention provides for novel cationic lipids that can be used in combination with other lipid components such as cholesterol and PEG-lipids to form lipid nanoparticles with oligonucleotides. It is an object of the instant invention to provide a cationic lipid scaffold that demonstrates enhanced efficacy along with lower liver toxicity as a result of lower lipid levels in the liver. The present invention employs low molecular weight cationic lipids comprising at least one short lipid chain to enhance the efficiency and tolerability of in vivo delivery of siRNA.

Description

TITLE OF THE INVENTION
NOVEL LOW MOLECULAR WEIGHT CYCLIC AMINE CONTAINING CATIONIC LIPIDS FOR OLIGONUCLEOTIDE DELIVERY BACKGROUND OF THE INVENTION
The present invention relates to novel cationic lipids that can be used in combination with other lipid components such as cholesterol and PEG-lipids to form lipid nanoparticles with oligonucleotides, to facilitate the cellular uptake and endosomal escape, and to knockdown target mRNA both in vitro and in vivo.
Cationic lipids and the use of cationic lipids in lipid nanoparticles for the delivery of oligonucleotides, in particular siRNA and miRNA, have been previously disclosed. Lipid nanoparticles and use of lipid nanoparticles for the delivery of oligonucleotides, in particular siRNA and miRNA, has been previously disclosed. Oligonucleotides (including siRNA and miRNA) and the synthesis of oligonucleotides has been previously disclosed. (See US patent applications: US 2006/0083780, US 2006/0240554, US 2008/0020058, US 2009/0263407 and US 2009/0285881 and PCT patent applications: WO 2009/086558, WO2009/127060,
WO2009/132131, WO2010/042877, WO2010/054384, WO2010/054401, WO2010/054405, WO2010/054406 and WO2010/105209). See also Semple S. C. et al., Rational design of cationic lipids for siRNA delivery, Nature Biotechnology, 2010, 28, 172-176. Other cationic lipids are disclosed in US patent applications: US 2009/0263407, US 2009/0285881, US 2010/0055168, US 2010/0055169, US 2010/0063135, US 2010/0076055, US 2010/0099738 and US
2010/0104629.
Further, the specific cationic lipids cis and trans l-methyl-3,4-bis-[((9Z,12Z)- octadeca-9,12-dienyl)oxy] -pyrrolidine are disclosed in WO2010/054384.
It is an object of the instant invention to provide a cationic lipid scaffold that demonstrates enhanced efficacy along with lower liver toxicity. The present invention employs low molecular weight cationic lipids comprising at least one short lipid chain to enhance the efficacy and tolerability of in vivo delivery of siRNA. SUMMARY OF THE INVENTION
The instant invention provides for novel cationic lipids that can be used in combination with other lipid components such as cholesterol and PEG-lipids to form lipid nanoparticles with oligonucleotides. It is an object of the instant invention to provide a cationic lipid scaffold that demonstrates enhanced efficacy along with lower liver toxicity as a result of lower lipid levels in the liver. The present invention employs low molecular weight cationic lipids with one short lipid chain to enhance the efficiency and tolerability of in vivo delivery of siRNA. BRIEF DESCRIPTION OF THE FIGURES
FIGURE 1 : Comparison of LNP (Compound 4) efficacy in mice.
FIGURE 2: LNP (Compound 4) efficacy and tolerability in rats.
FIGURE 3: Cationic lipid (Compound 4) levels in rat liver.
DETAILED DESCRIPTION OF THE INVENTION
The various aspects and embodiments of the invention are directed to the utility of novel cationic lipids useful in lipid nanoparticles to deliver oligonucleotides, in particular, siRNA and miRNA, to any target gene. (See US patent applications: US 2006/0083780, US 2006/0240554, US 2008/0020058, US 2009/0263407 and US 2009/0285881 and PCT patent applications: WO 2009/086558, WO2009/ 127060, WO2009/132131, WO2010/042877,
WO2010/054384, WO2010/054401, WO2010/054405, WO2010/054406 and WO2010/105209). See also Semple S. C. et al., Rational design of cationic lipids for siRNA delivery, Nature Biotechnology, published online 17 January 2010; doi:10.1038/nbt.l602.
The cationic lipids of the instant invention are useful components in a lipid nanoparticle for the delivery of oligonucleotides, specifically siRNA and miRNA.
In a first embodiment of this invention, the cationic lipids are illustrated by the
Formula A:
Figure imgf000004_0001
wherein:
R1 is selected from H, (C1-C6)alkyl, heterocyclyl, polyether and polyamine, wherein said alkyl, heterocyclyl, polyether and polyamine are optionally substituted with one to three substituents selected from R';
n and m are independently selected from 0, 1, 2 and 3;
X is independently selected from a bond, O, NR", (C=O)O, O(C=O), (C=O)NR",
NR"(C=O), O(C=O)O, NR"(C=O)NR", O(C=O)NR", and NR"(C=O)O;
L1 is selected from C4-C24 alkyl and C4-C24 alkenyl, said alkyl and alkenyl are optionally substituted with one or more substituents selected from R'; and
L2 is selected from C3-C9 alkyl and C3-C9 alkenyl, said alkyl and alkenyl are optionally substituted with one or more substituents selected from R';
R' is independently selected from halogen, R", OR", SR", CN, CO2R" and
CON(R")2;
R" is independently selected from H and (C1-C6)alkyl;
or any pharmaceutically acceptable salt or stereoisomer thereof. In a second embodiment, the invention features a compound having Formula A, wherein:
R is H or methyl;
n and m are 1;
X is O;
L1 is selected from C12-C24 alkyl and C12-C24 alkenyl; and
L2 is selected from C3-C9 alkyl and C3-C9 alkenyl;
or any pharmaceutically acceptable salt or stereoisomer thereof.
Specific cationic lipids are:
trans- l-Memyl-3-[((9Z,12Z)-octadeca-9,12-dienyl)oxy]-4-octyloxy-pyrrolidine
(Compound 4);
trans-1 -merayl-3-[(9Z)-octadec-9-en- 1 -yloxy]-4-(octyloxy)pyrrolidine
(Compound 5);
trans-1 -methyl-3-[(12Z)-octadec-l 2-en- 1 -yloxy]-4-(octyloxy)pyrrolidine (Compound 6);
trans-3-[(3,7-dimethyloctyl)oxy]-l-methyl-4-[(9Z,12Z)-octadeca-9,12-dien-l- yloxy]pyrrolidine (Compound 7); and
cis- 1 -methyl-3-[(9Z, 12Z)-octadeca-9, 12-dien- 1 -yloxy]-4-(octyloxy)pyrrolidine (Compound 11)
or any pharmaceutically acceptable salt or stereoisomer thereof.
In another embodiment, the cationic lipids disclosed are useful in the preparation of lipid nanoparticles.
In another embodiment, the cationic lipids disclosed are useful components in a lipid nanoparticle for the delivery of oligonucleotides.
In another embodiment, the cationic lipids disclosed are useful components in a lipid nanoparticle for the delivery of siRNA and miRNA.
In another embodiment, the cationic lipids disclosed are useful components in a lipid nanoparticle for the delivery of siRNA.
The cationic lipids of the present invention may have asymmetric centers, chiral axes, and chiral planes (as described in: E.L. Eliel and S.H. Wilen, Stereochemistry of Carbon Compounds, John Wiley & Sons, New York, 1994, pages 1119-1190), and occur as racemates, racemic mixtures, and as individual diastereomers, with all possible isomers and mixtures thereof, including optical isomers, being included in the present invention. In addition, the cationic lipids disclosed herein may exist as tautomers and both tautomeric forms are intended to be encompassed by the scope of the invention, even though only one tautomeric structure is depicted.
It is understood that substituents and substitution patterns on the cationic lipids of the instant invention can be selected by one of ordinary skill in the art to provide cationic lipids that are chemically stable and that can be readily synthesized by techniques known in the art, as well as those methods set forth below, from readily available starting materials. If a substituent is itself substituted with more than one group, it is understood that these multiple groups may be on the same carbon or on different carbons, so long as a stable structure results.
It is understood that one or more Si atoms can be incorporated into the cationic lipids of the instant invention by one of ordinary skill in the art to provide cationic lipids that are chemically stable and that can be readily synthesized by techniques known in the art from readily available starting materials.
In the compounds of Formula A, the atoms may exhibit their natural isotopic abundances, or one or more of the atoms may be artificially enriched in a particular isotope having the same atomic number, but an atomic mass or mass number different from the atomic mass or mass number predominantly found in nature. The present invention is meant to include all suitable isotopic variations of the compounds of Formula A. For example, different isotopic forms of hydrogen (H) include protium (1H) and deuterium (2H). Protium is the predominant hydrogen isotope found in nature. Enriching for deuterium may afford certain therapeutic advantages, such as increasing in vivo half-life or reducing dosage requirements, or may provide a compound useful as a standard for characterization of biological samples. Isotopically- enriched compounds within Formula A can be prepared without undue experimentation by conventional techniques well known to those skilled in the art or by processes analogous to those described in the Scheme and Examples herein using appropriate isotopically-enriched reagents and/or intermediates.
As used herein, "alkyl" means a straight chain, cyclic or branched saturated aliphatic hydrocarbon having the specified number of carbon atoms.
As used herein, "alkenyl" means a straight chain, cyclic or branched unsaturated aliphatic hydrocarbon having the specified number of carbon atoms including but not limited to diene, triene and tetraene unsaturated aliphatic hydrocarbons.
Examples of a cyclic "alkyl" or "alkenyl are:
Figure imgf000006_0001
As used herein, "heterocyclyl" or "heterocycle" means a 4- to 10-membered aromatic or nonaromatic heterocycle containing from 1 to 4 heteroatoms selected from the group consisting of O, N and S, and includes bicyclic groups. "Heterocyclyl" therefore includes, the following: benzoimidazolyl, benzofuranyl, benzofurazanyl, benzopyrazolyl, benzotriazolyl, benzothiophenyl, benzoxazolyl, carbazolyl, carbolinyl, cinnolinyl, furanyl, imidazolyl, indolinyl, indolyl, indolazinyl, indazolyl, isobenzofuranyl, isoindolyl, isoquinolyl, isothiazolyl, isoxazolyl, naphthpyridinyl, oxadiazolyl, oxazolyl, oxazoline, isoxazoline, oxetanyl, pyranyl, pyrazinyl, pyrazolyl, pyridazinyl, pyridopyridinyl, pyridazinyl, pyridyl, pyrimidyl, pyrrolyl, quinazolinyl, quinolyl, quinoxalinyl, tetrahydropyranyl, tetrazolyl, tetrazolopyridyl, thiadiazolyl, thiazolyl, thienyl, triazolyl, azetidinyl, 1,4-dioxanyl, hexahydroazepinyl, piperazinyl, piperidinyl, pyrrol idinyl, morpholinyl, thiomorpholinyl, dihydrobenzoirnidazolyl, dihydrobenzofuranyl, dihydrobenzothiophenyl, dihydrobenzoxazolyl, dihydrofuranyl, dihydroimidazolyl,
dihydroindolyl, dihydroisooxazolyl, dihydroisothiazolyl, dihydrooxadiazolyl, dihydrooxazoiyl, dihydropyrazinyl, dihydropyrazolyl, dihydropyridinyl, dihydropyrirnidinyl, dihydropyrrolyl, dihydroquinolinyl, dihydrotetrazolyl, dihydrothiadiazolyl, dihydrothiazolyl, dihydrothienyl, dihydrotriazolyl, dihydroazetidinyl, methylenedioxybenzoyl, tetrahydrofuranyl, and
tetrahydrothienyl, and N-oxides thereof all of which are optionally substituted with one to three substituents selected from R".
As used herein, "polyether" means compounds having two or more ether groups. Examples include poly(ethylene) glycols.
As used herein, "polyamine" means compounds having two or more amino groups. Examples include putrescine, cadaverine, spermidine, and spermine.
As used herein, "halogen" means Br, Cl, F and I.
In an embodiment of Formula A, R1 is selected from H and (C1-C6)alkyl, wherein said alkyl is optionally substituted with one to three substituents selected from R'.
In an embodiment of Formula A, R1 is selected from H, methyl, ethyl and propyl, wherein said methyl, ethyl and propyl are optionally substituted with one to three substituents selected from R'.
In an embodiment of Formula A, R1 is selected from H, methyl, ethyl and propyl. In an embodiment of Formula A, R1 is methyl.
In an embodiment of Formula A, R' is is selected from H, methyl, ethyl and propyl, wherein said methyl, ethyl and propyl are optionally substituted with one or more halogen and OH.
In an embodiment of Formula A, n is 0, 1, 2 or 3.
In an embodiment of Formula A, n is 1 or 2.
In an embodiment of Formula A, n is 1.
In an embodiment of Formula A, m is 0, 1 , 2 or 3.
In an embodiment of Formula A, m is 1 or 2.
In an embodiment of Formula A, m is 1.
In an embodiment of Formula A, X is a bond, O, NR", (C=O)O, NR"(C-0), 0(C-0)O, NR"(C=O)NR", O(C=O)NR", or NR"(C=O)O.
In an embodiment of Formula A, X is (C=O)O or O.
In an embodiment of Formula A, X is O.
In an embodiment of Formula A, L1 is selected from C4-C24 alkyl and C4-C24 alkenyl, which are optionally substituted with halogen and OH. In an embodiment of Formula A, L1 is selected from C4-C24 alkyl and C4-C24 alkenyl.
In an embodiment of Formula A, L1 is selected from C12-C24 alkyl and C12- C24 alkenyl, which are optionally substituted with halogen and OH.
In an embodiment of Formula A, L1 is selected from C12-C24 alkyl and C12-
C24 alkenyl.
In an embodiment of Formula A, L1 is selected from C4-C24 alkenyl.
In an embodiment of Formula A, L1 is selected from C12-C24 alkenyl.
In an embodiment of Formula A, L1 is C18 alkenyl.
In an embodiment of Formula A, L1 is:
Figure imgf000008_0001
In an embodiment of Formula A, L2 is selected from C3-C9 alkyl and C3-C9 alkenyl, which are optionally substituted with halogen and OH.
In an embodiment of Formula A, L2 is selected from C5-C9 alkyl and C5-C9 alkenyl, which are optionally substituted with halogen and OH.
In an embodiment of Formula A, L2 is selected from C7-C9 alkyl and C7-C9 alkenyl, which are optionally substituted with halogen and OH.
In an embodiment of Formula A, L2 is selected from C3-C9 alkyl and C3-C9 alkenyl.
In an embodiment of Formula A, L2 is selected from C5-C9 alkyl and C5-C9 alkenyl.
In an embodiment of Formula A, L2 is selected from C7-C9 alkyl and C7-C9 alkenyl.
In an embodiment of Formula A, L2 is C3-C9 alkyl.
In an embodiment of Formula A, L2 is C5-C9 alkyl.
In an embodiment of Formula A, L2 is C7-C9 alkyl.
In an embodiment of Formula A, L2 is C8 alkyl.
In an embodiment of Formula A, "heterocyclyl" is pyrolidine, piperidine, morpholine, imidazole or piperazine.
In an embodiment of Formula A, "polyamine" is putrescine, cadaverine, spermidine or spermine.
In an embodiment of Formula A, "polyether" is poly(ethylene) glycol(s).
In an embodiment, "alkyl" is a straight chain saturated aliphatic hydrocarbon having the specified number of carbon atoms.
In an embodiment, "alkenyl" is a straight chain unsaturated aliphatic hydrocarbon having the specified number of carbon atoms. Included in the instant invention is the free form of cationic lipids of Formula A, as well as the pharmaceutically acceptable salts and stereoisomers thereof. Some of the isolated specific cationic lipids exemplified herein are the protonated salts of amine cationic lipids. The term "free form" refers to the amine cationic lipids in non-salt form. The encompassed pharmaceutically acceptable salts not only include the isolated salts exemplified for the specific cationic lipids described herein, but also all the typical pharmaceutically acceptable salts of the free form of cationic lipids of Formula A. The free form of the specific salt cationic lipids described may be isolated using techniques known in the art. For example, the free form may be regenerated by treating the salt with a suitable dilute aqueous base solution such as dilute aqueous NaOH, potassium carbonate, ammonia and sodium bicarbonate. The free forms may differ from their respective salt forms somewhat in certain physical properties, such as solubility in polar solvents, but the acid and base salts are otherwise pharmaceutically equivalent to their respective free forms for purposes of the invention.
The pharmaceutically acceptable salts of the instant cationic lipids can be synthesized from the cationic lipids of this invention which contain a basic or acidic moiety by conventional chemical methods. Generally, the salts of the basic cationic lipids are prepared either by ion exchange chromatography or by reacting the free base with stoichiometric amounts or with an excess of the desired salt-forming inorganic or organic acid in a suitable solvent or various combinations of solvents. Similarly, the salts of the acidic compounds are formed by reactions with the appropriate inorganic or organic base.
Thus, pharmaceutically acceptable sails of the cationic lipids of this invention include the conventional non-toxic salts of the cationic lipids of this invention as formed by reacting a basic instant cationic lipids with an inorganic or organic acid. For example, conventional non-toxic salts include those derived from inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, nitric and the like, as well as salts prepared from organic acids such as acetic, propionic, succinic, glycolic, stearic, lactic, malic, tartaric, citric, ascorbic, pamoic, maleic, hydroxymaleic, phenylacetic, glutamic, benzoic, salicylic, sulfanilic, 2-acetoxy-benzoic, fumaric, toluenesulfonic, methanesulfonic, ethane disulfonic, oxalic, isethionic, trifluoroacetic (TFA) and the like.
When the cationic lipids of the present invention are acidic, suitable
"pharmaceutically acceptable salts" refers to salts prepared form pharmaceutically acceptable non-toxic bases including inorganic bases and organic bases. Salts derived from inorganic bases include aluminum, ammonium, calcium, copper, ferric, ferrous, Uthium, magnesium, manganic salts, manganous, potassium, sodium, zinc and the like. Particularly preferred are the ammonium, calcium, magnesium, potassium and sodium salts. Salts derived from
pharmaceutically acceptable organic non-toxic bases include salts of primary, secondary and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, such as arginine, betaine caffeine, choline, Ν,Ν1- dibenzylethylenediamine, diethylamin, 2-diethylaminoethanol, 2-dimethylaminoethanol, ethanolamine, ethylenediamine, N-ethylmorpholine, N-ethylpiperidine, glucamine, glucosamine, histidine, hydrabamine, isopropylainine, lysine, methylglucamine, morpholine, piperazine, piperidine, polyamine resins, procaine, purines, theobromine, triethylamine, trimethylamine tripropylamine, tromethamine and the like.
The preparation of the pharmaceutically acceptable salts described above and other typical pharmaceutically acceptable salts is more fully described by Berg et al. ,
"Pharmaceutical Salts," J. Pharm. Set, 1977:66:1-19.
It will also be noted that the cationic lipids of the present invention are potentially internal salts or zwitterions, since under physiological conditions a deprotonated acidic moiety in the compound, such as a carboxyl group, may be anionic, and this electronic charge might then be balanced off internally against the cationic charge of a protonated or alkylated basic moiety, such as a quaternary nitrogen atom.
EXAMPLES
Examples provided are intended to assist in a further understanding of the invention. Particular materials employed, species and conditions are intended to be further illustrative of the invention and not limitative of the reasonable scope thereof. The reagents utilized in synthesizing cationic lipids are either commercially available or are readily prepared by one of ordinary skill in the art.
Synthesis of cyclic amine containing cationic lipids is a linear process starting from cyclic dihydroxy amines i. Stepwise etherification to give ii followed by cleavage of the benzyl group to give iii and reductive amination gives products of the type iv.
Figure imgf000010_0001
EXAMPLE 1
1 -Methyl-3-[((9Z.12Z)-octadeca-9, 12-dienyl)oxyl-4-octyloxy-pyrrolidine (4):
Figure imgf000011_0001
1-Benzyl-3,4 dihydroxy pyrroldine benzene sulfonic acid salt (1): In a 3N 5L RBF with overhead stirrer and reflux condenser, the DL-l,4-dibromo butane diol (100 g, 403 mmol), benzyl amine (39.7 ml, 363 mmol), potassium iodide (3.35 g, 20.17 mmol) and diisopropylethylamine
(DIPEA, 70.3 ml, 403 mmol) were suspended in 500 mL dioxane and heated to 110°C overnight. After 17h cooled at 80°C, added 750 mL 1M aqueous K2CO3 and 750 mL ethtyl acetate
(EtOAc). Allowed to cool to 23°C and separated layers. Extracted with 500 mL EtOAc and then dried combined organics over Na2SO4. Filtered and concentrated to 400 mL solution. Added benzene sulfonic acid (38.3 g, 242 mmol) in 200 mL EtOAc slowly over lh. After 16h at 23°C, cooled to 0°C and filtered. Washed cake with 200 mL methyl tert-butyl ether (MTBE) and then dried under vacuum at 40°C for 6h to give 73.8 g of 1 as an off-white solid (52% yield). 1H NMR (400 MHz, CD3OD) δ 7.85 (m, 2H), 7.65-7.41 (m, 8H), 4.42 (m, 2H), 4.28 (m, 2H), 3.79 (dd, J= 12.7, 4.2 Hz, 1H), 3.51 (dd, J= 12.2, 3.4 Hz, 1H), 3.38 (d, J= 12.2 Hz, 1H), 3.25 (d, J= 12.7 Hz, 1H). l-Benzyl-3-[((9Z,12Z)-octadeca-9,12-dienyl)oxy]-4-octyloxy-pyrrolidine (2): In 500 mL 3N RBF, compound 1 (18 g, 51.2 mmol) and triethylamine (0.714 ml, 5.12 mmol) were suspended in 180 mL toluene for addition of the sodium hydride (4.30 g, 108 mmol). Heated suspension to 50°C for lh. Cooled back to 23°C and then added the «-octyl mesylate (10.67 g, 51.2 mmol). Heated slurry up to 80°C for 18h. Cooled to 23°C and poured into 200 mL 1M K2CO3. Extracted with methyl tert-butyl ether (MTBE) and then dried combiend layers over Na2SO4. Filtered, concentrated and purified oil by silica gel chromatogtaphy to obtain 7.22 g of a slightly yellow oil. In 500 mL 3N RBF, 4.04 g of the aforementioned oil (13.23 mmol) and triethylamine (0.184 ml, 1.323 mmol) was dissolved in 80 mL toluene for addition of the sodium hydride (0.582 g, 14.55 mmol). Heated suspension to 50°C for 1h. Cooled back to 23°C and then added the linoleoyl mesylate (5.01 g, 14.55 mmol). Heated slurry up to 80°C for 21h or until complete dissappearance of the starting material was observed by HPLC. Cooled to 23°C, diluted with 100 mL hexane and washed with 100 mL 1M K2CO3. Extracted aquoues layer with 100 mL hexane and then washed combined organic layers with 100 mL brine and dried over MgSCv Filtered and concentrated. Purified by silica gel chromatography to obtain 5.02 g of 2 as a clear oil (33% yield over 2 steps). 1H NMR (400 MHz, CDC13) δ 7.28 (m, 5H), 5.40 (m , 4H), 3.85 (Ψt, J= 4.8 Hz, 2H), 3.61 (m, 2H), 3.42 (m, 4H), 2.88 (dd, J= 9.7, 6.1 Hz, 2H), 2.79 (Ψt, J= 6.4 Hz, 2H). 2.51 (dd, J= 10.1, 4.2 Hz, 2H), 2.06 (q, J= 6.7 Hz, 2H), 1.56 (q, 4H), 1.41-1.29 (m, 26H), 0.9 (m, 6H). 3-r((9Z,12ZVoctade(a-9,12-dienvnoxy1-4-octyloxy-pyrrolidine (3 : In a 500 mL 3N RBF, compound 2 (6.32 g, 11.41 mmol) was dissolved in 60 mL dichloromethane (DCM) at 0°C for addition of the DIPEA (5.96 ml, 34.2 mmol) and chloroethyl chloroformate (3.73 ml, 34.2 mmol). Aged at for 3h at 23°C or until complete dissappearance of 2 is observed by HPLC. Concentrated down and then added 60 mL methanol (MeOH). Continued aging at 23°C. After an additional 18h, concentrated to a thick oil and then partitioned between 200 mL hexane and 200 mL 1M K2CO3. Extracted with hexane and then dried over Na2SO4. Filtered and concentrated before purification by silica gel chromatography to obtain 2.70 g of 3 as a yellow oil (51% yield). 1H NMR (400 MHz, CDC13) δ 5.38 (m, 4H). 3.78 (m, 2H), 3.45 (m, 4H), 3.11 (dd, J= 12.4, 5.2 Hz, 2H), 2.84 (dd, J= 12.5, 2.6 Hz, 2H), 2.79 (Ψt, J= 6.6 Hz, 2H), 2.07 (q, J= 6.7 Hz, 4H), 1.55 (q, J= 6.6 Hz, 4H), 1.41 - 1.16 (m, 26H), 0.90 (m, 6H). l-Methyl-3-rfr9Z,12ZVoctadeca-9,12-dienyl)oxyl-4-octyloxy-pyrrolidine (4 : In a 500 mL RBF, compound 3 (4.24 g, 9.14 mmol) was dissolved in 80 mL tetrahydrofurn (THF) for addition of 20 mL glacial acetic acid. To this was added the paraformaldehyde (2.75 g, 91 mmol) followed by the pyridine-borane complex (1.829 ml, 18.28 mmol). Aged at 23°C for 20h or until complete dissappearance of 3 was observed by HPLC. Added 100 mL hexane and 100 mL 1M K2CO3 and stirred until bubbling had subsided. Separated layers, extracted with 100 mL hexane and washed combined organics with 100 mL brine. Dried over Na2SO4, filtered and concentrated before purification by silica gel chromatography to obtain 1.75 g of 4 as a clear oil (41% yield). !H NMR (400 MHz, CDC13) δ 5.37 (m, 4H), 3.85 (Ψt, J= 4.4 Hz, 2H), 3.44 (m, 4H), 2.93 (dd, J= 10.2, 6.2 Hz, 2H), 2.79 (Ψt, J= 6.5 Hz, 2H), 2.6 (m, 2H), 2.39 (s, 3H), 2.06 (q, J= 7.6 Hz, 4H), 1.58 (m, 4H), 1.41 - 1.31 (m, 26H), 0.90 (m, 6H).
Compounds 5-7 were prepared in a manner analogous to that described for Compound 4.
1 -methyl-3-[(9Z)-octadec-9-en-1 -yloxyl-4-(octyloxy)pyrrolidine (5): LC/MS (M+H) = 480.6.
Figure imgf000013_0003
l-methyl-3-rn2Z) >ctadec-12-en -vIoxy1^-focl loxypmoiidine(6'): H NMR (400 MHz, CDC ) δ 5.37 (m, 2H), 3.85 (Ψΐ, J= 4.4 Hz, 2H), 3.48 (m54H)S 2.85 (dd, J= 10.2, 6.2 Hz, 2H), 2.48 (m, 2H)S 2.34 (s, 3H), 2.06 (m, 4H), 1.58 (m, 4H), 1.41 - 1.31 (m, 30H), 0.90 (m, 6H).
Figure imgf000013_0001
3 n.7-dimethyloc toxyH-met ^
LC/MS (M+H) = 506.6.
Figure imgf000013_0002
EXAMPLE 2
Ci5-l-methyl-3 (9Z,12ZVocta^^
Figure imgf000014_0001
Benzyl 3,4-dihvdroxypyrrolidine-l-carboxylate (9): In a 500 mL RBF, the Cbz Pyrroline (15 gs 73.8 mmol) was dissolved in a mixture of 100 mL THF, 60 mL t-BuOH and 40 mL water for addition of the NMO (30.6 ml, 148 mmol) solution followed by the osmium tetraoxide (0.925 ml, 0.074 mmol) solution. Heated in a 70C oil bath. Solution slowly went from yellow to brown. After 2h, cooled to RT and added 50 mL 10% aqueous NaHS03. Concentrated and partitioned between 100 mL brine and 100 mL EtOAc. Separated layers and extracted with 100 mL EtOAc. Dried over MgS04, filtered and concentrated to 22.3 g of thick yellow oil. Flash column chromatography from 50-100% EtOAc in hexane provided compound 9. !H NMR (400 MHz, CDC13) 5 7.35 (m, 5H), 5.22 (s, 2H), 4.42 (m, 4H), 3.63 (m, 2H), 3.42 (m, 3H), 3.20 (m, IH).
Benzyl 3-[f9Z,122^-octadeca-9.12-dien-l-Yloxy]^-foctyloxy)pmolidine-l-carboxylate (10 : In 2500 mL RBF, compound 9 (9.7 g, 40.0 mmol) and triethylamine (0.558 ml, 4.0 mmol) were suspended in 100 mL toluene for addition of the sodium hydride (1.76 g, 44 mmol). Heated suspension to 50°C for Ih. Cooled back to 23°C and then added the linoleyl mesylate (13.8 g, 40.0 mmol). Heated slurry up to 80°C for 18h. Cooled to 23°C and poured into 200 mL 1M
K2CO3. Extracted with methyl tert-butyl ether (MTBE) and then dried combiend layers over Na2S0 . Filtered, concentrated and purified oil by silica gel chromatogtaphy to obtain
monoalkylated product as a slightly yellow oil In 250 mL RBF, 8g of the aforementioned oil (16.47 mmol) and triethylamine (0.23 mi, 1.65 mmol) was dissolved in 100 mL toluene for addition of the sodium hydride (0.0.725 g, 18.12 mmol). Heated suspension to 50°C for Ih.
Cooled back to 23°C and then added the w-octyl mesylate (3.77 g, 18.12 mmol). Heated slurry up to 80°C for 21h or until complete dissappearance of the starting material was observed by HPLC. Cooled to 23°C, diluted with 100 mL hexane and washed with 100 mL 1M K2C03. Extracted aquoues layer with 100 mL hexane and then washed combined organic layers with 100 mL brine and dried over MgS04. Filtered and concentrated. Purified by silica gel chromatography to obtain 8.4 g of 10 as a clear oil. Product contains some octyl mesylate but was carried into next reaction.
Cis-l-memyl-3-r(9Z,12Z -ocfadeca-9,12^ In a 100 mL RJBF fitted with reflux condenser, compound 10 (4 g, 6.69 mmol) was dissolved in 26 mL THF for addition of the lithium aluminum hydride (13.38 ml, 13.38 mmol). Heated to reflux in a 85C oil bath. After 2h, HPLC shows complete dissappearance of the SM. Cooled to RT and poured into 200 mL 1M K2C03 and 200 mL TBE. Stirred for 30 min, separated layers and then extracted with 100 mL MTBE. Dried combined organics over Na2S04, filtered through Celite and concentrated. Flash chromatography (10-30% DP A in DCM) provided compound 11 as a clear oil. 1H NMR (400 MHz, CDC13) δ 5.39 (m, 4H), 3.93 (m, 2H), 3.50 (m, 4H), 3.04 (m, 2H), 2.79 (m, 2H), 2.48 (m, 2H), 2.39 (s, 3H), 2.08 (m, 4H), 1.62 (m, 4H), 1.40-1.25 (m, 26H), 0.91 (m, 6H).
Compound 12 is DLinKC2DMA as described in Nature Biotechnology, 2010, 28, 172-176, WO 2010/042877 Al, WO 2010/048536 A2, WO 2010/088537 A2, and
WO 2009/127060 Al.
Figure imgf000015_0001
(12)
Compound 13 is MC3 as described in WO 2010/054401, and WO 2010/144740
Figure imgf000015_0002
LNP COMPOSITIONS
The following lipid nanoparticle compositions (LNPs) of the instant invention are useful for the delivery of oligonucleotides, specifically siRNA and miRNA:
Cationic Lipid / Cholesterol / PEG-DMG 56.6/38/5.4;
Cationic Lipid / Cholesterol / PEG-DMG 60/38/2;
Cationic Lipid/ Cholesterol / PEG-DMG 67.3/29/3.7;
Cationic Lipid / Cholesterol / PEG-DMG 49.3/47/3.7;
Cationic Lipid / Cholesterol / PEG-DMG 50.3/44.3/5.4;
Cationic Lipid / Cholesterol / PEG-C-DMA / DSPC 40/48/2/10;
Cationic Lipid / Cholesterol / PEG-DMG / DSPC 40/48/2/10; and Cationic Lipid / Cholesterol / PEG-DMG / DSPC 58/30/2/10.
LNP process description:
The Lipid Nano-Particles (LNP) are prepared by an impinging jet process. The particles are formed by mixing lipids dissolved in alcohol with siRNA dissolved in a citrate buffer. The mixing ratio of lipids to siRNA are targeted at 45-55% lipid and 65-45% siRNA. The lipid solution contains a novel cationic lipid of the instant invention, a helper lipid
(cholesterol) , PEG (e.g. PEG-C-DMA, PEG-DMG) lipid, and DSPC at a concentration of 5-15 mg/mL with a target of 9-12 mg/mL in an alcohol (for example ethanol). The ratio of the lipids has a mole percent range of 25-98 for the cationic lipid with a target of 35-65, the helper lipid has a mole percent range from 0-75 with a target of 30-50, the PEG lipid has a mole percent range from 1-15 with a target of 1 -6, and the DSPC has a mole precent range of 0-15 with a target of 0-12. The siRNA solution contains one or more siRNA sequences at a concentration range from 0.3 to 1 .0 mg mL with a target of 0.3 -0.9 mg/mL in a sodium citrate buffered salt solution with pH in the range of 3.5-5. The two liquids are heated to a temperature in the range of 15-40°CS targeting 30-40°C, and then mixed in an impinging jet mixer instantly forming the LNP. The teelD has a range from 0.25 to 1.0 mm and a total flow rate from 10 -600 mL/min. The combination of flow rate and tubing ID has effect of controlling the particle size of the LNPs between 30 and 200 nm. The solution is then mixed with a buffered solution at a higher pH with a mixing ratio in the range of 1 :1 to 1 :3 vol:vol but targeting 1 :2 voi:vol. This buffered solution is at a temperature in the range of 15-40°C, targeting 30-40°C. The mixed LNPs are held from 30 minutes to 2 hrs prior to an anion exchange filtration step. The temperature during incubating is in the range of 15-40°C, targeting 30-40°C. After incubating the solution is filtered through a 0.8 urn filter containing an anion exchange separation step. This process uses tubing IDs ranging from 1 mm ID to 5 mm ID and a flow rate from 10 to 2000 mL/min. The LNPs are concentrated and diafiltered via an ultrafiltration process where the alcohol is removed and the citrate buffer is exchanged for the final buffer solution such as phosphate buffered saline. The ultrafiltration process uses a tangential flow filtration format (TFF). This process uses a membrane nominal molecular weight cutoff range from 30 -500 D. The membrane format can be hollow fiber or flat sheet cassette. The TFF processes with the proper molecular weight cutoff retains the LNP in the retentate and the filtrate or permeate contains the alcohol; citrate buffer; final buffer wastes. The TFF process is a multiple step process with an initial concentration to a siRNA concentration of 1 -3 mg/mL. Following concentration, the LNPs solution is diafiltered against the final buffer for 10 -20 volumes to remove the alcohol and perform buffer exchange. The material is then concentrated an additional 1-3 fold. The final steps of the LNP process are to sterile filter the concentrated LNP solution and vial the product.
Analytical Procedure:
1) siRNA concentration The siRNA duplex concentrations are determined by Strong Anion-Exchange High-Performance Liquid Chromatography (SAX-HPLC) using Waters 2695 Alliance system (Water Corporation, Milford MA) with a 2996 PDA detector. The LNPs, otherwise referred to as RNAi Delivery Vehicles (RDVs), are treated with 0.5% Triton X- 100 to free total siRNA and analyzed by SAX separation using a Dionex BioLC DNAPac PA 200 (4 χ 250 mm) column with UV detection at 254 nm. Mobile phase is composed of A: 25 mM NaC104, 10 mM Tris, 20% EtOH, pH 7.0 and B: 250 mM NaC104, 10 mM Tris, 20% EtOH, pH 7.0 with liner gradient from 0-15 min and flow rate of 1 ml/min. The siRNA amount is determined by comparing to the siRNA standard curve.
2) Encapsulation rate
Fluorescence reagent S YBR Gold is employed for RNA quantitation to monitor the encapsulation rate of RDVs. RDVs with or without Triton X-100 are used to determine the free siRNA and total siRNA amount. The assay is performed using a SpectraMax M5e microplate spectrophotometer from Molecular Devices (Sunnyvale, CA). Samples are excited at 485 nm and fluorescence emission was measured at 530 nm. The siRNA amount is determined by comparing to the siRNA standard curve.
Encapsulation rate = (1- free siRNA/total siRNA) xl00%
3 Particle size and polvdispersitv
RDVs containing 1 g siRNA are diluted to a final volume of 3 ml with 1 χ PBS. The particle size and polydispersity of the samples is measured by a dynamic light scattering method using ZetaPALS instrument (Brookhaven Instruments Corporation, Holtsville, NY). The scattered intensity is measured with He-Ne laser at 25°C with a scattering angle of 90°.
4) Zeta Potential analysis
RDVs containing 1 μg siRNA are diluted to a final volume of 2 ml with 1 mM Tris buffer (pH 7.4). Electrophoretic mobility of samples is determined using ZetaPALS instrument (Brookhaven Instruments Corporation, Holtsville, NY) with electrode and He-Ne laser as a light source. The Smoluchowski limit is assumed in the calculation of zeta potentials.
5) Lipid analysis
Individual lipid concentrations are determined by Reverse Phase High- Performance Liquid Chromatography (RP-HPLC) using Waters 2695 Alliance system (Water Corporation, Milford MA) with a Corona charged aerosol detector (CAD) (ESA Biosciences, Inc, Chelmsford, MA). Individual lipids in RDVs are analyzed using an Agilent Zorbax SB-C18 (50 x 4.6 mm, 1.8 μπι particle size) column with CAD at 60 °C. The mobile phase is composed of A: 0.1% TFA in H20 and B: 0.1% TFA in IPA. The gradient changes from 60% mobile phase A and 40% mobile phase B from time 0 to 40% mobile phase A and 60% mobile phase B at 1.00 min; 40% mobile phase A and 60% mobile phase B from 1.00 to 5.00 min; 40% mobile phase A and 60% mobile phase B from 5.00 min to 25% mobile phase A and 75% mobile phase B at 10.00 min; 25% mobile phase A and 75% mobile phase B from 10.00 min to 5% mobile phase A and 95% mobile phase B at 15.00 min; and 5% mobile phase A and 95% mobile phase B from 15.00 to 60% mobile phase A and 40% mobile phase B at 20.00 min with flow rate of 1 ml/min. The individual lipid concentration is determined by comparing to the standard curve with all the lipid components in the RDVs with a quadratic curve fit. The molar percentage of each lipid is calculated based on its molecular weight.
Utilizing the above described LNP process, specific LNPs with the following ratios were identified:
Nominal composition:
Cationic Lipid / Cholesterol / PEG-DMG 60/38/2; and
Cationic Lipid / Cholesterol / PEG-DMG / DSPC 58/30/2/10.
Oligonucleotide synthesis is well known in the art. (See US patent applications: US 2006/0083780, US 2006/0240554, US 2008/0020058, US 2009/0263407 and US
2009/0285881 and PCT patent applications: WO 2009/086558, WO2009/127060,
WO2009/132131, WO2010/042877, WO2010/054384, WO2010/054401, WO2010/054405 and WO2010/054406). The Luc siRNA incorporated in the LNPs disclosed and utilized in the Examples were synthesized via standard solid phase procedures.
Luc siRNA
S'-iB-AC/AAGGCi/Al/GAAGAGAi/ATT-iB 3' (SEQ.ID.NO.:l) y-WJUAUUCCGAUACUUCUCUAO-S1 (SEQ.ID.NO. :2)
AUGC - Ribose
iB ~- Inverted deoxy abasic
UC- T Fluoro
AGT - 2' Deoxy
AGU - 2' OCH3
Nominal composition
Cationic Lipid /Cholesterol/PEG-DMG 60/38/2
Cationic Lipid / Cholesterol / PEG-DMG / DSPC 40/48/2/10
Cationic Lipid / Cholesterol / PEG-DMG / DSPC 58/30/2/10
ApoB siRNA
S'-iB-CUUU^C^UUCCUG^UTsT-iB-S' (SEQ ID NO.:3) 3'AJsVGAMVUGUVMGGACVsmJsA-5' (SEQ ID NO.:4)
AUGC - Ribose
iB - Inverted deoxy abasic
UC- 2' Fluoro
AGT - 2' Deoxy
AGU - 2' OCH3
UsA - phophorothioate linkage EXAMPLE 1
Mouse In Vivo Evaluation of Efficacy
LNPs utilizing Compound 4, in the nominal compositions described immediately above, were evaluated for in vivo efficacy. The siRNA targets the mRNA transcript for the firefly (Photinus pyralis) luciferase gene (Accession # Ml 5077). The primary sequence and chemical modification pattern of the luciferase siRNA is displayed above. The in vivo luciferase model employs a transgenic mouse in which the firefly luciferase coding sequence is present in all cells. ROSA26- LoxP-Stop-LoxP-Luc (LSL-Luc) transgenic mice licensed from the Dana Farber Cancer Institute are induced to express the Luciferase gene by first removing the LSL sequence with a recombinant Ad-Cre virus (Vector Biolabs). Due to the organo-tropic nature of the virus, expression is limited to the liver when delivered via tail vein injection. Luciferase expression levels in liver are quantitated by measuring light output, using an IVIS imager (Xenogen) following administration of the luciferin substrate (Caliper Life Sciences). Pre-dose luminescence levels are measured prior to administration of the RDVs. Luciferin in PBS (15mg/mL) is intraperitoneally (IP) injected in a volume of 150 μL· After a four minute incubation period mice are anesthetized with isoflurane and placed in the IVIS imager. The RDVs (containing siRNA) in PBS vehicle were tail vein injected n a volume of 0.2 mL. Final dose levels ranged from 0.1 to 0.5 mg/kg siRNA. PBS vehicle alone was dosed as a control. Mice were imaged 48 hours post dose using the method described above. Changes in luciferin light output directly correlate with luciferase mRNA levels and represent an indirect measure of luciferase siRNA activity. In vivo efficacy results are expressed as % inhibition of luminescence relative to pre-dose luminescence levels. Systemic administration of the luciferase siRNA RDVs decreased luciferase expression in a dose dependant manner. Greater efficacy was observed in mice dosed with Compound 4 containing RDVs than with the RDV containing the trans 1 - methyl-3,4-bis-[((9Z}12Z)-octadeca-9,12-dienyl)oxy]-pyrrolidine (Compound A) as disclosed in WO2010/054384.
EXAMPLE 2
Rat In Vivo Evaluation of Efficacy and Toxicity
LNPs utilizing compounds in the nominal compositions described above, were evaluated for in vivo efficacy and increases in alanine amino transferase and aspartate amino transferase in Sprague-Dawley (Crl:CD(SD) female rats (Charles River Labs). The siRNA targets the mRNA transcript for the ApoB gene (Accession # NM 019287). The primary sequence and chemical modification pattern of the ApoB siRNA is displayed above. The RDVs (containing siRNA) in PBS vehicle were tail vein injected in a volume of 1 to 1.5 mL. Infusion rate is approximately 3 ml/min. Five rats were used in each dosing group. After LNP
administration, rats are placed in cages with normal diet and water present. Six hours post dose, food is removed from the cages. Animal necropsy is performed 24 hours after LNP dosing. Rats are anesthetized under isoflurane for 5 minutes, then maintained under anesthesia by placing them in nose cones continuing the delivery of isoflurane until ex-sanguination is completed. Blood is collected from the vena cava using a 23 gauge butterfly venipuncture set and aliquoted to serum separator vacutainers for serum chemistry analysis. Punches of the excised caudate liver lobe are taken and placed in RNALater (Ambion) for mRNA analysis. Preserved liver tissue was homogenized and total RNA isolated using a Qiagen bead mill and the Qiagen miRNA-Easy RNA isolation kit following the manufacturer's instructions. Liver ApoB mRNA levels were determined by quantitative RT-PCR. Message was amplified from purified RNA utilizing a rat ApoB commercial probe set (Applied Biosystems Cat # RN01499054_ml). The PCR reaction was performed on an ABI 7500 instrument with a 96-well Fast Block. The ApoB mRNA level is normalized to the housekeeping PPIB (NM 011149) mRNA. ΡΡΪΒ mRNA levels were determined by RT-PCR using a commercial probe set (Applied Biosytems Cat. No.
Mm00478295jml). Results are expressed as a ratio of ApoB mRNA/ PPIB mRNA. All mRNA data is expressed relative to the PBS control dose. Serum ALT and AST analysis were performed on the Siemens Advia 1800 Clinical Chemistry Analyzer utilizing the Siemens alanine aminotransferase (Cat# 03039631) and aspartate aminotransferase (Cat# 03039631) reagents. Modestly decreased efficacy but significantly improved tolerability was observed in rats dosed with Compound 4 containing RDV than with the RDV containing the cationic lipid
DLinKC2DMA (Compound 12, Figure 2).
EXAMPLE 3
Determination of Cationic Lipid Levels in Rat Liver
Liver tissue was weighed into 20-ml vials and homogenized in 9 v/w of water using a GenoGrinder 2000 (OPS Diagnostics, 1600 strokes/min, 5min). A 50 uL aliquot of each tissue homogenate was mixed with 300 L of extraction/protein precipitating solvent (50/50 acetonitrile methanol containing 500 nM internal standard) and the plate was centrifuged to sediment precipitated protein. A volume of 200 μL of each supernatant was then transferred to separate wells of a 96-well plate and 10 μL samples were directly analyzed by LC MS-MS.
Standards were prepared by spiking known amounts of a methanol stock solution of compound into untreated rat liver homogenate (9 vol water/weight liver). Aliquots (50 μL) each standard/liver homogenate was mixed with 300 μL of extraction/protein precipitating solvent (50/50 acetonitrile/methanol containing 500 nM internal standard) and the plate was centrifuged to sediment precipitated protein. A volume of 200 μL of each supernatant was transferred to separate wells of a 96-well plate and 10 μΐ of each standard was directly analyzed by LC/MS-MS.
Absolute quantification versus standards prepared and extracted from liver homogenate was performed using an Aria LX-2 HPLC system (Thermo Scientific) coupled to an API 4000 triple quadrupole mass spectrometer (Applied Biosystems). For each run, a total of 10 μϋ sample was injected onto a BDS Hypersil C8 HPLC column (Thermo, 50 x 2mm, 3 μιη) at ambient temperature.
Mobile Phase A: 95% H20/5% methanol/10 mM ammonium formate/0. l%formic acid Mobile Phase B: 40% methanol/60% n-propanol/10 mM ammonium formate/0. l%formic acid The flow rate was 0.5 mL/min and gradient elution profile was as follows: hold at 80% A for 0.25 min, linear ramp to 100% B over 1.6 min, hold at 100% B for 2.5 min, then return and hold at 80% A for 1.75 min. Total run time was 5.8 min. API 4000 source parameters were CAD: 4, CUR: 15, GS1: 65, GS2: 35, IS: 4000, TEM: 550, CXP: 15, DP: 60, EP: 10.
In rats dosed with Compound 4 (0.5 mg/kg) containing RDV, liver levels were much lower than the RDV containing the cationic lipid DLinKC2DMA (Compound 12, also 0.5 mg/kg) or MC3 (Compound 13, also 0.5 mg/kg, Figure 3).

Claims

WHAT IS CLAIMED IS:
1. A cationic lipid of Formula A:
Figure imgf000022_0001
wherein:
R1 is selected from H, (C1-C6)alkyl, heterocyclyl, poly ether and polyamine, wherein said alkyl, heterocyclyl, polyether and polyamine are optionally substituted with one to three substituents selected from R'; n and m are independently selected from 0, 1, 2 and 3;
X is independently selected from a bond, O, NR", (C=O)O, NR"(C=O), 0(CO)0,
NR"(C=O)NR"i 0(C=O)NR", and NR"(C=O)0;
L1 is selected from C4-C24 alkyl and C4-C24 alkenyl, said alkyl and alkenyl are optionally substituted with one or more substituents selected from R1; and L2 is selected from C3-C9 alkyl and C3-C9 alkenyl, said alkyl and alkenyl are optionally substituted with one or more substituents selected from R';
R' is independently selected from halogen, R", OR", SR", CN, CO2R" and CON(R")2; R" is independently selected from H and (C1-C6)alkyl; or any pharmaceutically acceptable salt or stereoisomer thereof.
2. A cationic lipid of Formula A according to Claim 1, wherein: R1 is H or methyl; n and m are 1;
X is O; Li is selected from C12-C24 alkyl and C12-C24 alkenyl; and L2 is selected from C3-C9 alkyl and C3-C9 alkenyl; or any pharmaceutically acceptable salt or stereoisomer thereof.
3. A cationic lipid which is: trans- 1 -Methyl-3 - [((9Z , 12Z)-octadeca-9, 12-dienyl)oxy] -4-octyloxy-pyrrolidine (Compound 4) ; trans-l -methyl-3-[(9Z)-octadec-9-en-l-yloxy]-4-(octyloxy)pyrrolidine (Compound 5);
trans- 1 -methyl-3-[( 12Z)-octadec- 12-en- 1 -yloxy]-4-(octyloxy)pyrrolidine (Compound 6);
trans-3 - [(3 }7-dimethy loctyl)oxy] - 1 -methyl-4~[(9Z, 12Z)-octadeca-9512-dien- 1 -yloxyjpyrrolidine (Compound 7);
cis- 1 -methyl-3 - [(9Z, 12Z)-octadeca-9, 12-dien- 1 -yloxy] -4-(octyloxy)pyrrolidine (Compound 11 ) or any pharmaceutically acceptable salt or stereoisomer thereof.
4. The use of a cationic lipid according to Claim 1 for the preparation of lipid nanoparticles.
5. The use of a cationic lipid according to Claim 1 as a component in a lipid nanoparticle for the delivery of oligonucleotides.
6. The use according to Claim 5 wherein the oligonucleotides are siRNA or miRNA.
7. The use according to Claim 5 wherein the oligonucleotides are siRNA.
PCT/US2011/058498 2010-11-05 2011-10-31 Novel low molecular weight cyclic amine containing cationic lipids for oligonucleotide delivery WO2012061259A2 (en)

Priority Applications (5)

Application Number Priority Date Filing Date Title
EP11838596.2A EP2635265B1 (en) 2010-11-05 2011-10-31 Novel low molecular weight cyclic amine containing cationic lipids for oligonucleotide delivery
US13/883,487 US9067882B2 (en) 2010-11-05 2011-10-31 Low molecular weight cyclic amine containing cationic lipids for oligonucleotide delivery
DK11838596.2T DK2635265T3 (en) 2010-11-05 2011-10-31 New low molecular weight cyclic amine-containing cationic lipids for oligonucleotide delivery
US14/719,513 US9796977B2 (en) 2010-11-05 2015-05-22 Low molecular weight cyclic amine containing cationic lipids for oligonucleotide delivery
US15/706,041 US10337014B2 (en) 2010-11-05 2017-09-15 Low molecular weight cyclic amine containing cationic lipids for oligonucleotide delivery

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US41047210P 2010-11-05 2010-11-05
US61/410,472 2010-11-05

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US13/883,487 A-371-Of-International US9067882B2 (en) 2010-11-05 2011-10-31 Low molecular weight cyclic amine containing cationic lipids for oligonucleotide delivery
US14/719,513 Continuation US9796977B2 (en) 2010-11-05 2015-05-22 Low molecular weight cyclic amine containing cationic lipids for oligonucleotide delivery

Publications (2)

Publication Number Publication Date
WO2012061259A2 true WO2012061259A2 (en) 2012-05-10
WO2012061259A3 WO2012061259A3 (en) 2012-06-28

Family

ID=46025039

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2011/058498 WO2012061259A2 (en) 2010-11-05 2011-10-31 Novel low molecular weight cyclic amine containing cationic lipids for oligonucleotide delivery

Country Status (4)

Country Link
US (3) US9067882B2 (en)
EP (1) EP2635265B1 (en)
DK (1) DK2635265T3 (en)
WO (1) WO2012061259A2 (en)

Cited By (78)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013151666A2 (en) 2012-04-02 2013-10-10 modeRNA Therapeutics Modified polynucleotides for the production of biologics and proteins associated with human disease
WO2013151736A2 (en) 2012-04-02 2013-10-10 modeRNA Therapeutics In vivo production of proteins
WO2014152540A1 (en) 2013-03-15 2014-09-25 Moderna Therapeutics, Inc. Compositions and methods of altering cholesterol levels
WO2014152211A1 (en) 2013-03-14 2014-09-25 Moderna Therapeutics, Inc. Formulation and delivery of modified nucleoside, nucleotide, and nucleic acid compositions
WO2015006747A2 (en) 2013-07-11 2015-01-15 Moderna Therapeutics, Inc. Compositions comprising synthetic polynucleotides encoding crispr related proteins and synthetic sgrnas and methods of use.
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
WO2015051214A1 (en) 2013-10-03 2015-04-09 Moderna Therapeutics, Inc. Polynucleotides encoding low density lipoprotein receptor
WO2015075557A2 (en) 2013-11-22 2015-05-28 Mina Alpha Limited C/ebp alpha compositions and methods of use
WO2015110957A2 (en) 2014-01-21 2015-07-30 De Beer Joel Hybridosomes, compositions comprising the same, processes for their production and uses thereof
WO2016014846A1 (en) 2014-07-23 2016-01-28 Moderna Therapeutics, Inc. Modified polynucleotides for the production of intrabodies
WO2016065349A2 (en) 2014-10-24 2016-04-28 University Of Maryland, Baltimore Short non-coding protein regulatory rnas (sprrnas) and methods of use
WO2017070613A1 (en) 2015-10-22 2017-04-27 Modernatx, Inc. Human cytomegalovirus vaccine
CN106631956A (en) * 2016-12-29 2017-05-10 常州市阳光药业有限公司 Preparation method of N-substituent-3-hydroxytetrahydropyrrole
WO2017112943A1 (en) 2015-12-23 2017-06-29 Modernatx, Inc. Methods of using ox40 ligand encoding polynucleotides
WO2017120612A1 (en) 2016-01-10 2017-07-13 Modernatx, Inc. Therapeutic mrnas encoding anti ctla-4 antibodies
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
WO2018232006A1 (en) 2017-06-14 2018-12-20 Modernatx, Inc. Polynucleotides encoding coagulation factor viii
US10221127B2 (en) 2015-06-29 2019-03-05 Acuitas Therapeutics, Inc. Lipids and lipid nanoparticle formulations for delivery of nucleic acids
WO2019048631A1 (en) 2017-09-08 2019-03-14 Mina Therapeutics Limited Hnf4a sarna compositions and methods of use
WO2019048645A1 (en) 2017-09-08 2019-03-14 Mina Therapeutics Limited Stabilized cebpa sarna compositions and methods of use
WO2019131580A1 (en) * 2017-12-27 2019-07-04 エーザイ・アール・アンド・ディー・マネジメント株式会社 Cationic lipid
WO2019197845A1 (en) 2018-04-12 2019-10-17 Mina Therapeutics Limited Sirt1-sarna compositions and methods of use
WO2019217964A1 (en) 2018-05-11 2019-11-14 Lupagen, Inc. Systems and methods for closed loop, real-time modifications of patient cells
US10501416B2 (en) 2016-06-24 2019-12-10 Eisai R&D Management Co., Ltd. Cationic lipid
WO2020033791A1 (en) 2018-08-09 2020-02-13 Verseau Therapeutics, Inc. Oligonucleotide compositions for targeting ccr2 and csf1r and uses thereof
WO2020061284A1 (en) 2018-09-19 2020-03-26 Modernatx, Inc. Peg lipids and uses thereof
WO2020061295A1 (en) 2018-09-19 2020-03-26 Modernatx, Inc. High-purity peg lipids and uses thereof
WO2020128031A2 (en) 2018-12-21 2020-06-25 Curevac Ag Rna for malaria vaccines
WO2020161342A1 (en) 2019-02-08 2020-08-13 Curevac Ag Coding rna administered into the suprachoroidal space in the treatment of ophtalmic diseases
WO2020208361A1 (en) 2019-04-12 2020-10-15 Mina Therapeutics Limited Sirt1-sarna compositions and methods of use
WO2020254535A1 (en) 2019-06-18 2020-12-24 Curevac Ag Rotavirus mrna vaccine
WO2021026310A1 (en) 2019-08-06 2021-02-11 L.E.A.F. Holdings Group Llc Processes of preparing polyglutamated antifolates and uses of their compositions
WO2021028439A1 (en) 2019-08-14 2021-02-18 Curevac Ag Rna combinations and compositions with decreased immunostimulatory properties
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
WO2021156267A1 (en) 2020-02-04 2021-08-12 Curevac Ag Coronavirus vaccine
WO2021183720A1 (en) 2020-03-11 2021-09-16 Omega Therapeutics, Inc. Compositions and methods for modulating forkhead box p3 (foxp3) gene expression
WO2021239880A1 (en) 2020-05-29 2021-12-02 Curevac Ag Nucleic acid based combination vaccines
WO2022023559A1 (en) 2020-07-31 2022-02-03 Curevac Ag Nucleic acid encoded antibody mixtures
WO2022032154A2 (en) 2020-08-06 2022-02-10 Modernatx, Inc. Compositions for the delivery of payload molecules to airway epithelium
WO2022043551A2 (en) 2020-08-31 2022-03-03 Curevac Ag Multivalent nucleic acid based coronavirus vaccines
WO2022122872A1 (en) 2020-12-09 2022-06-16 Ucl Business Ltd Therapeutics for the treatment of neurodegenerative disorders
WO2022137133A1 (en) 2020-12-22 2022-06-30 Curevac Ag Rna vaccine against sars-cov-2 variants
WO2022135993A2 (en) 2020-12-22 2022-06-30 Curevac Ag Pharmaceutical composition comprising lipid-based carriers encapsulating rna for multidose administration
WO2022162027A2 (en) 2021-01-27 2022-08-04 Curevac Ag Method of reducing the immunostimulatory properties of in vitro transcribed rna
US11453639B2 (en) 2019-01-11 2022-09-27 Acuitas Therapeutics, Inc. Lipids for lipid nanoparticle delivery of active agents
WO2022200575A1 (en) 2021-03-26 2022-09-29 Glaxosmithkline Biologicals Sa Immunogenic compositions
WO2022200810A1 (en) 2021-03-26 2022-09-29 Mina Therapeutics Limited Tmem173 sarna compositions and methods of use
WO2022207862A2 (en) 2021-03-31 2022-10-06 Curevac Ag Syringes containing pharmaceutical compositions comprising rna
EP4074834A1 (en) 2012-11-26 2022-10-19 ModernaTX, Inc. Terminally modified rna
WO2022233880A1 (en) 2021-05-03 2022-11-10 Curevac Ag Improved nucleic acid sequence for cell type specific expression
WO2022261394A1 (en) 2021-06-11 2022-12-15 LifeEDIT Therapeutics, Inc. Rna polymerase iii promoters and methods of use
WO2023283359A2 (en) 2021-07-07 2023-01-12 Omega Therapeutics, Inc. Compositions and methods for modulating secreted frizzled receptor protein 1 (sfrp1) gene expression
WO2023014974A1 (en) 2021-08-06 2023-02-09 University Of Iowa Research Foundation Double stranded mrna vaccines
US11590229B2 (en) 2011-12-07 2023-02-28 Alnylam Pharmaceuticals, Inc. Biodegradable lipids for the delivery of active agents
EP4144378A1 (en) 2011-12-16 2023-03-08 ModernaTX, Inc. Modified nucleoside, nucleotide, and nucleic acid compositions
WO2023031392A2 (en) 2021-09-03 2023-03-09 CureVac SE Novel lipid nanoparticles for delivery of nucleic acids comprising phosphatidylserine
WO2023031394A1 (en) 2021-09-03 2023-03-09 CureVac SE Novel lipid nanoparticles for delivery of nucleic acids
EP4159741A1 (en) 2014-07-16 2023-04-05 ModernaTX, Inc. Method for producing a chimeric polynucleotide encoding a polypeptide having a triazole-containing internucleotide linkage
WO2023073228A1 (en) 2021-10-29 2023-05-04 CureVac SE Improved circular rna for expressing therapeutic proteins
US11648324B2 (en) 2015-10-28 2023-05-16 Acuitas Therapeutics, Inc. Lipids and lipid nanoparticle formulations for delivery of nucleic acids
WO2023086465A1 (en) 2021-11-12 2023-05-19 Modernatx, Inc. Compositions for the delivery of payload molecules to airway epithelium
WO2023099884A1 (en) 2021-12-01 2023-06-08 Mina Therapeutics Limited Pax6 sarna compositions and methods of use
WO2023104964A1 (en) 2021-12-09 2023-06-15 Ucl Business Ltd Therapeutics for the treatment of neurodegenerative disorders
WO2023144330A1 (en) 2022-01-28 2023-08-03 CureVac SE Nucleic acid encoded transcription factor inhibitors
WO2023154818A1 (en) 2022-02-09 2023-08-17 Modernatx, Inc. Mucosal administration methods and formulations
WO2023161350A1 (en) 2022-02-24 2023-08-31 Io Biotech Aps Nucleotide delivery of cancer therapy
WO2023170435A1 (en) 2022-03-07 2023-09-14 Mina Therapeutics Limited Il10 sarna compositions and methods of use
WO2023183616A1 (en) * 2022-03-25 2023-09-28 Senda Biosciences, Inc. Novel ionizable lipids and lipid nanoparticles and methods of using the same
US11820728B2 (en) 2017-04-28 2023-11-21 Acuitas Therapeutics, Inc. Carbonyl lipids and lipid nanoparticle formulations for delivery of nucleic acids
WO2023227608A1 (en) 2022-05-25 2023-11-30 Glaxosmithkline Biologicals Sa Nucleic acid based vaccine encoding an escherichia coli fimh antigenic polypeptide
WO2024033901A1 (en) 2022-08-12 2024-02-15 LifeEDIT Therapeutics, Inc. Rna-guided nucleases and active fragments and variants thereof and methods of use
DE202023106198U1 (en) 2022-10-28 2024-03-21 CureVac SE Nucleic acid-based vaccine
EP4342460A1 (en) 2022-09-21 2024-03-27 NovoArc GmbH Lipid nanoparticle with nucleic acid cargo
WO2024068545A1 (en) 2022-09-26 2024-04-04 Glaxosmithkline Biologicals Sa Influenza virus vaccines
US11976019B2 (en) 2020-07-16 2024-05-07 Acuitas Therapeutics, Inc. Cationic lipids for use in lipid nanoparticles

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2934551B1 (en) * 2012-12-20 2017-11-29 Sirna Therapeutics, Inc. Post-synthetic orthogonal amidation plus metal catalyzed azide-alkyne cycloaddition click chemistry on si-rna
JP7156154B2 (en) * 2019-04-18 2022-10-19 株式会社島津製作所 Medium processing system and medium processing method
WO2023031855A1 (en) 2021-09-03 2023-03-09 Glaxosmithkline Biologicals Sa Substitution of nucleotide bases in self-amplifying messenger ribonucleic acids
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

Citations (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060083780A1 (en) 2004-06-07 2006-04-20 Protiva Biotherapeutics, Inc. Cationic lipids and methods of use
US20060240554A1 (en) 2005-02-14 2006-10-26 Sirna Therapeutics, Inc. Lipid nanoparticle based compositions and methods for the delivery of biologically active molecules
US20080020058A1 (en) 2005-02-14 2008-01-24 Sirna Therapeutics, Inc. Lipid nanoparticle based compositions and methods for the delivery of biologically active molecules
WO2009086558A1 (en) 2008-01-02 2009-07-09 Tekmira Pharmaceuticals Corporation Improved compositions and methods for the delivery of nucleic acids
WO2009127060A1 (en) 2008-04-15 2009-10-22 Protiva Biotherapeutics, Inc. Novel lipid formulations for nucleic acid delivery
US20090263407A1 (en) 2008-04-16 2009-10-22 Abbott Laboratories Cationic Lipids and Uses Thereof
WO2009132131A1 (en) 2008-04-22 2009-10-29 Alnylam Pharmaceuticals, Inc. Amino lipid based improved lipid formulation
US20090285881A1 (en) 2008-04-16 2009-11-19 Abbott Laboratories Cationic lipids and uses thereof
US20100055169A1 (en) 2008-04-16 2010-03-04 Abbott Laboratories Cationic lipids and uses thereof
US20100055168A1 (en) 2008-04-16 2010-03-04 Abbott Laboratories Cationic lipids and uses thereof
US20100063135A1 (en) 2008-09-10 2010-03-11 Abbott Laboratories Polyethylene glycol lipid conjugates and uses thereof
US20100076055A1 (en) 2008-04-16 2010-03-25 Abbott Laboratories Cationic Lipids and Uses Thereof
WO2010042877A1 (en) 2008-10-09 2010-04-15 Tekmira Pharmaceuticals Corporation Improved amino lipids and methods for the delivery of nucleic acids
US20100099738A1 (en) 2008-09-10 2010-04-22 Abbott Laboratories Polyethylene glycol lipid conjugates and uses thereof
US20100104629A1 (en) 2008-04-16 2010-04-29 Abbott Laboratories Cationic lipids and uses thereof
WO2010048536A2 (en) 2008-10-23 2010-04-29 Alnylam Pharmaceuticals, Inc. Processes for preparing lipids
WO2010054384A1 (en) 2008-11-10 2010-05-14 Alnylam Pharmaceuticals, Inc. Lipids and compositions for the delivery of therapeutics
WO2010054401A1 (en) 2008-11-10 2010-05-14 Alnylam Pharmaceuticals, Inc. Novel lipids and compositions for the delivery of therapeutics
WO2010088537A2 (en) 2009-01-29 2010-08-05 Alnylam Pharmaceuticals, Inc. Improved lipid formulation
WO2010105209A1 (en) 2009-03-12 2010-09-16 Alnylam Pharmaceuticals, Inc. LIPID FORMULATED COMPOSITIONS AND METHODS FOR INHIBITING EXPRESSION OF Eg5 AND VEGF GENES
WO2010144740A1 (en) 2009-06-10 2010-12-16 Alnylam Pharmaceuticals, Inc. Improved lipid formulation

Patent Citations (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060083780A1 (en) 2004-06-07 2006-04-20 Protiva Biotherapeutics, Inc. Cationic lipids and methods of use
US20060240554A1 (en) 2005-02-14 2006-10-26 Sirna Therapeutics, Inc. Lipid nanoparticle based compositions and methods for the delivery of biologically active molecules
US20080020058A1 (en) 2005-02-14 2008-01-24 Sirna Therapeutics, Inc. Lipid nanoparticle based compositions and methods for the delivery of biologically active molecules
WO2009086558A1 (en) 2008-01-02 2009-07-09 Tekmira Pharmaceuticals Corporation Improved compositions and methods for the delivery of nucleic acids
WO2009127060A1 (en) 2008-04-15 2009-10-22 Protiva Biotherapeutics, Inc. Novel lipid formulations for nucleic acid delivery
US20100104629A1 (en) 2008-04-16 2010-04-29 Abbott Laboratories Cationic lipids and uses thereof
US20090263407A1 (en) 2008-04-16 2009-10-22 Abbott Laboratories Cationic Lipids and Uses Thereof
US20090285881A1 (en) 2008-04-16 2009-11-19 Abbott Laboratories Cationic lipids and uses thereof
US20100055169A1 (en) 2008-04-16 2010-03-04 Abbott Laboratories Cationic lipids and uses thereof
US20100055168A1 (en) 2008-04-16 2010-03-04 Abbott Laboratories Cationic lipids and uses thereof
US20100076055A1 (en) 2008-04-16 2010-03-25 Abbott Laboratories Cationic Lipids and Uses Thereof
WO2009132131A1 (en) 2008-04-22 2009-10-29 Alnylam Pharmaceuticals, Inc. Amino lipid based improved lipid formulation
US20100063135A1 (en) 2008-09-10 2010-03-11 Abbott Laboratories Polyethylene glycol lipid conjugates and uses thereof
US20100099738A1 (en) 2008-09-10 2010-04-22 Abbott Laboratories Polyethylene glycol lipid conjugates and uses thereof
WO2010042877A1 (en) 2008-10-09 2010-04-15 Tekmira Pharmaceuticals Corporation Improved amino lipids and methods for the delivery of nucleic acids
WO2010048536A2 (en) 2008-10-23 2010-04-29 Alnylam Pharmaceuticals, Inc. Processes for preparing lipids
WO2010054384A1 (en) 2008-11-10 2010-05-14 Alnylam Pharmaceuticals, Inc. Lipids and compositions for the delivery of therapeutics
WO2010054401A1 (en) 2008-11-10 2010-05-14 Alnylam Pharmaceuticals, Inc. Novel lipids and compositions for the delivery of therapeutics
WO2010054406A1 (en) 2008-11-10 2010-05-14 Alnylam Pharmaceuticals, Inc. Novel lipids and compositions for the delivery of therapeutics
WO2010054405A1 (en) 2008-11-10 2010-05-14 Alnylam Pharmaceuticals, Inc. Novel lipids and compositions for the delivery of therapeutics
WO2010088537A2 (en) 2009-01-29 2010-08-05 Alnylam Pharmaceuticals, Inc. Improved lipid formulation
WO2010105209A1 (en) 2009-03-12 2010-09-16 Alnylam Pharmaceuticals, Inc. LIPID FORMULATED COMPOSITIONS AND METHODS FOR INHIBITING EXPRESSION OF Eg5 AND VEGF GENES
WO2010144740A1 (en) 2009-06-10 2010-12-16 Alnylam Pharmaceuticals, Inc. Improved lipid formulation

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
BERG ET AL.: "Pharmaceutical Salts", J. PHARM. SCI., vol. 66, 1977, pages 1 - 19
E.L. ELIEL; S.H. WILEN: "Stereochemistry of Carbon Compounds", 1994, JOHN WILEY & SONS, pages: 1119 - 1190
NATURE BIOTECHNOLOGY, vol. 28, 2010, pages 172 - 176
See also references of EP2635265A4
SEMPLE S. C. ET AL.: "Rational design of cationic lipids for siRNA delivery", NATURE BIOTECHNOLOGY, 17 January 2010 (2010-01-17)
SEMPLE S. C. ET AL.: "Rational design of cationic lipids for siRNA delivery", NATURE BIOTECHNOLOGY, vol. 28, 2010, pages 172 - 176

Cited By (103)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11679158B2 (en) 2011-12-07 2023-06-20 Alnylam Pharmaceuticals, Inc. Biodegradable lipids for the delivery of active agents
US11590229B2 (en) 2011-12-07 2023-02-28 Alnylam Pharmaceuticals, Inc. Biodegradable lipids for the delivery of active agents
US11612657B2 (en) 2011-12-07 2023-03-28 Alnylam Pharmaceuticals, Inc. Biodegradable lipids for the delivery of active agents
US11633480B2 (en) 2011-12-07 2023-04-25 Alnylam Pharmaceuticals, Inc. Biodegradable lipids for the delivery of active agents
US11633479B2 (en) 2011-12-07 2023-04-25 Alnylam Pharmaceuticals, Inc. Biodegradable lipids for the delivery of active agents
EP4144378A1 (en) 2011-12-16 2023-03-08 ModernaTX, Inc. Modified nucleoside, nucleotide, and nucleic acid compositions
WO2013151666A2 (en) 2012-04-02 2013-10-10 modeRNA Therapeutics Modified polynucleotides for the production of biologics and proteins associated with human disease
WO2013151736A2 (en) 2012-04-02 2013-10-10 modeRNA Therapeutics In vivo production of proteins
EP4074834A1 (en) 2012-11-26 2022-10-19 ModernaTX, Inc. Terminally modified rna
WO2014152211A1 (en) 2013-03-14 2014-09-25 Moderna Therapeutics, Inc. Formulation and delivery of modified nucleoside, nucleotide, and nucleic acid compositions
WO2014152540A1 (en) 2013-03-15 2014-09-25 Moderna Therapeutics, Inc. Compositions and methods of altering cholesterol levels
EP3971287A1 (en) 2013-07-11 2022-03-23 ModernaTX, Inc. Compositions comprising synthetic polynucleotides encoding crispr related proteins and synthetic sgrnas and methods of use
WO2015006747A2 (en) 2013-07-11 2015-01-15 Moderna Therapeutics, Inc. Compositions comprising synthetic polynucleotides encoding crispr related proteins and synthetic sgrnas and methods of use.
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
WO2015051214A1 (en) 2013-10-03 2015-04-09 Moderna Therapeutics, Inc. Polynucleotides encoding low density lipoprotein receptor
EP3985118A1 (en) 2013-11-22 2022-04-20 MiNA Therapeutics Limited C/ebp alpha short activating rna compositions and methods of use
WO2015075557A2 (en) 2013-11-22 2015-05-28 Mina Alpha Limited C/ebp alpha compositions and methods of use
EP3594348A1 (en) 2013-11-22 2020-01-15 Mina Therapeutics Limited C/ebp alpha short activating rna compositions and methods of use
EP3791863A1 (en) 2014-01-21 2021-03-17 Anjarium Biosciences AG Process for the production of hybridosomes
WO2015110957A2 (en) 2014-01-21 2015-07-30 De Beer Joel Hybridosomes, compositions comprising the same, processes for their production and uses thereof
US11944706B2 (en) 2014-01-21 2024-04-02 Anjarium Biosciences Ag Hybridosomes, compositions comprising the same, processes for their production and uses thereof
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
WO2016065349A2 (en) 2014-10-24 2016-04-28 University Of Maryland, Baltimore Short non-coding protein regulatory rnas (sprrnas) and methods of use
US10221127B2 (en) 2015-06-29 2019-03-05 Acuitas Therapeutics, Inc. Lipids and lipid nanoparticle formulations for delivery of nucleic acids
US11168051B2 (en) 2015-06-29 2021-11-09 Acuitas Therapeutics, Inc. Lipids and lipid nanoparticle formulations for delivery of nucleic acids
WO2017070613A1 (en) 2015-10-22 2017-04-27 Modernatx, Inc. Human cytomegalovirus vaccine
US11648324B2 (en) 2015-10-28 2023-05-16 Acuitas Therapeutics, Inc. Lipids and lipid nanoparticle formulations for delivery of nucleic acids
EP4039699A1 (en) 2015-12-23 2022-08-10 ModernaTX, Inc. Methods of using ox40 ligand encoding polynucleotides
WO2017112943A1 (en) 2015-12-23 2017-06-29 Modernatx, Inc. Methods of using ox40 ligand encoding polynucleotides
WO2017120612A1 (en) 2016-01-10 2017-07-13 Modernatx, Inc. Therapeutic mrnas encoding anti ctla-4 antibodies
US10501416B2 (en) 2016-06-24 2019-12-10 Eisai R&D Management Co., Ltd. Cationic lipid
CN106631956A (en) * 2016-12-29 2017-05-10 常州市阳光药业有限公司 Preparation method of N-substituent-3-hydroxytetrahydropyrrole
US11820728B2 (en) 2017-04-28 2023-11-21 Acuitas Therapeutics, Inc. Carbonyl 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
WO2018232006A1 (en) 2017-06-14 2018-12-20 Modernatx, Inc. Polynucleotides encoding coagulation factor viii
EP4183882A1 (en) 2017-09-08 2023-05-24 MiNA Therapeutics Limited Stabilized hnf4a sarna compositions and methods of use
WO2019048631A1 (en) 2017-09-08 2019-03-14 Mina Therapeutics Limited Hnf4a sarna compositions and methods of use
EP4219715A2 (en) 2017-09-08 2023-08-02 MiNA Therapeutics Limited Stabilized cebpa sarna compositions and methods of use
EP4233880A2 (en) 2017-09-08 2023-08-30 MiNA Therapeutics Limited Hnf4a sarna compositions and methods of use
WO2019048632A1 (en) 2017-09-08 2019-03-14 Mina Therapeutics Limited Stabilized hnf4a sarna compositions and methods of use
WO2019048645A1 (en) 2017-09-08 2019-03-14 Mina Therapeutics Limited Stabilized cebpa sarna compositions and methods of use
US10947193B2 (en) 2017-12-27 2021-03-16 Eisai R&D Management Co., Ltd. Cationic lipid
WO2019131580A1 (en) * 2017-12-27 2019-07-04 エーザイ・アール・アンド・ディー・マネジメント株式会社 Cationic lipid
JPWO2019131580A1 (en) * 2017-12-27 2020-12-24 エーザイ・アール・アンド・ディー・マネジメント株式会社 Cationic lipid
JP7164547B2 (en) 2017-12-27 2022-11-01 エーザイ・アール・アンド・ディー・マネジメント株式会社 cationic lipid
WO2019197845A1 (en) 2018-04-12 2019-10-17 Mina Therapeutics Limited Sirt1-sarna compositions and methods of use
EP4242307A2 (en) 2018-04-12 2023-09-13 MiNA Therapeutics Limited Sirt1-sarna compositions and methods of use
WO2019217964A1 (en) 2018-05-11 2019-11-14 Lupagen, Inc. Systems and methods for closed loop, real-time modifications of patient cells
WO2020033791A1 (en) 2018-08-09 2020-02-13 Verseau Therapeutics, Inc. Oligonucleotide compositions for targeting ccr2 and csf1r and uses thereof
WO2020061295A1 (en) 2018-09-19 2020-03-26 Modernatx, Inc. High-purity peg lipids and uses thereof
WO2020061284A1 (en) 2018-09-19 2020-03-26 Modernatx, Inc. Peg lipids and uses thereof
WO2020128031A2 (en) 2018-12-21 2020-06-25 Curevac Ag Rna for malaria vaccines
US11453639B2 (en) 2019-01-11 2022-09-27 Acuitas Therapeutics, Inc. Lipids for lipid nanoparticle delivery of active agents
WO2020161342A1 (en) 2019-02-08 2020-08-13 Curevac Ag Coding rna administered into the suprachoroidal space in the treatment of ophtalmic diseases
WO2020208361A1 (en) 2019-04-12 2020-10-15 Mina Therapeutics Limited Sirt1-sarna compositions and methods of use
WO2020254535A1 (en) 2019-06-18 2020-12-24 Curevac Ag Rotavirus mrna vaccine
WO2021026310A1 (en) 2019-08-06 2021-02-11 L.E.A.F. Holdings Group Llc Processes of preparing polyglutamated antifolates and uses of their compositions
WO2021028439A1 (en) 2019-08-14 2021-02-18 Curevac Ag Rna combinations and compositions with decreased immunostimulatory properties
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
DE202021004130U1 (en) 2020-02-04 2022-10-26 Curevac Ag Coronavirus Vaccine
EP4147717A1 (en) 2020-02-04 2023-03-15 CureVac SE Coronavirus vaccine
DE202021003575U1 (en) 2020-02-04 2022-01-17 Curevac Ag Coronavirus Vaccine
WO2021156267A1 (en) 2020-02-04 2021-08-12 Curevac Ag Coronavirus vaccine
DE202021004123U1 (en) 2020-02-04 2022-10-26 Curevac Ag Coronavirus Vaccine
DE112021000012T5 (en) 2020-02-04 2021-11-18 Curevac Ag Coronavirus vaccine
WO2021183720A1 (en) 2020-03-11 2021-09-16 Omega Therapeutics, Inc. Compositions and methods for modulating forkhead box p3 (foxp3) gene expression
WO2021239880A1 (en) 2020-05-29 2021-12-02 Curevac Ag Nucleic acid based combination vaccines
US11976019B2 (en) 2020-07-16 2024-05-07 Acuitas Therapeutics, Inc. Cationic lipids for use in lipid nanoparticles
WO2022023559A1 (en) 2020-07-31 2022-02-03 Curevac Ag Nucleic acid encoded antibody mixtures
WO2022032154A2 (en) 2020-08-06 2022-02-10 Modernatx, Inc. Compositions for the delivery of payload molecules to airway epithelium
WO2022043551A2 (en) 2020-08-31 2022-03-03 Curevac Ag Multivalent nucleic acid based coronavirus vaccines
WO2022122872A1 (en) 2020-12-09 2022-06-16 Ucl Business Ltd Therapeutics for the treatment of neurodegenerative disorders
WO2022137133A1 (en) 2020-12-22 2022-06-30 Curevac Ag Rna vaccine against sars-cov-2 variants
WO2022135993A2 (en) 2020-12-22 2022-06-30 Curevac Ag Pharmaceutical composition comprising lipid-based carriers encapsulating rna for multidose administration
WO2022162027A2 (en) 2021-01-27 2022-08-04 Curevac Ag Method of reducing the immunostimulatory properties of in vitro transcribed rna
WO2022200575A1 (en) 2021-03-26 2022-09-29 Glaxosmithkline Biologicals Sa Immunogenic compositions
WO2022200810A1 (en) 2021-03-26 2022-09-29 Mina Therapeutics Limited Tmem173 sarna compositions and methods of use
WO2022207862A2 (en) 2021-03-31 2022-10-06 Curevac Ag Syringes containing pharmaceutical compositions comprising rna
WO2022233880A1 (en) 2021-05-03 2022-11-10 Curevac Ag Improved nucleic acid sequence for cell type specific expression
WO2022261394A1 (en) 2021-06-11 2022-12-15 LifeEDIT Therapeutics, Inc. Rna polymerase iii promoters and methods of use
WO2023283359A2 (en) 2021-07-07 2023-01-12 Omega Therapeutics, Inc. Compositions and methods for modulating secreted frizzled receptor protein 1 (sfrp1) gene expression
WO2023014974A1 (en) 2021-08-06 2023-02-09 University Of Iowa Research Foundation Double stranded mrna vaccines
WO2023031392A2 (en) 2021-09-03 2023-03-09 CureVac SE Novel lipid nanoparticles for delivery of nucleic acids comprising phosphatidylserine
WO2023031394A1 (en) 2021-09-03 2023-03-09 CureVac SE Novel lipid nanoparticles for delivery of nucleic acids
WO2023073228A1 (en) 2021-10-29 2023-05-04 CureVac SE Improved circular rna for expressing therapeutic proteins
WO2023086465A1 (en) 2021-11-12 2023-05-19 Modernatx, Inc. Compositions for the delivery of payload molecules to airway epithelium
WO2023099884A1 (en) 2021-12-01 2023-06-08 Mina Therapeutics Limited Pax6 sarna compositions and methods of use
WO2023104964A1 (en) 2021-12-09 2023-06-15 Ucl Business Ltd Therapeutics for the treatment of neurodegenerative disorders
WO2023144330A1 (en) 2022-01-28 2023-08-03 CureVac SE Nucleic acid encoded transcription factor inhibitors
WO2023154818A1 (en) 2022-02-09 2023-08-17 Modernatx, Inc. Mucosal administration methods and formulations
WO2023161350A1 (en) 2022-02-24 2023-08-31 Io Biotech Aps Nucleotide delivery of cancer therapy
WO2023170435A1 (en) 2022-03-07 2023-09-14 Mina Therapeutics Limited Il10 sarna compositions and methods of use
WO2023183616A1 (en) * 2022-03-25 2023-09-28 Senda Biosciences, Inc. Novel ionizable lipids and lipid nanoparticles and methods of using the same
WO2023227608A1 (en) 2022-05-25 2023-11-30 Glaxosmithkline Biologicals Sa Nucleic acid based vaccine encoding an escherichia coli fimh antigenic polypeptide
WO2024033901A1 (en) 2022-08-12 2024-02-15 LifeEDIT Therapeutics, Inc. Rna-guided nucleases and active fragments and variants thereof and methods of use
EP4342460A1 (en) 2022-09-21 2024-03-27 NovoArc GmbH Lipid nanoparticle with nucleic acid cargo
WO2024062001A1 (en) 2022-09-21 2024-03-28 Novoarc Gmbh Lipid nanoparticle with nucleic acid cargo
WO2024068545A1 (en) 2022-09-26 2024-04-04 Glaxosmithkline Biologicals Sa Influenza virus vaccines
DE202023106198U1 (en) 2022-10-28 2024-03-21 CureVac SE Nucleic acid-based vaccine

Also Published As

Publication number Publication date
EP2635265B1 (en) 2018-04-04
US20180037893A1 (en) 2018-02-08
WO2012061259A3 (en) 2012-06-28
US9067882B2 (en) 2015-06-30
EP2635265A2 (en) 2013-09-11
DK2635265T3 (en) 2018-07-16
US9796977B2 (en) 2017-10-24
US20130225836A1 (en) 2013-08-29
EP2635265A4 (en) 2014-06-04
US10337014B2 (en) 2019-07-02
US20150361434A1 (en) 2015-12-17

Similar Documents

Publication Publication Date Title
US10337014B2 (en) Low molecular weight cyclic amine containing cationic lipids for oligonucleotide delivery
US20180208545A1 (en) Novel low molecular weight cationic lipids for oligonucleotide delivery
EP2575767B1 (en) Novel low molecular weight cationic lipids for oligonucleotide delivery
EP2467357B1 (en) Novel cationic lipids with various head groups for oligonucleotide delivery
EP2621480B1 (en) Low molecular weight cationic lipids for oligonucleotide delivery
US9044512B2 (en) Amino alcohol cationic lipids for oligonucleotide delivery
WO2013016058A1 (en) Novel bis-nitrogen containing cationic lipids for oligonucleotide delivery
WO2011043913A2 (en) Novel cationic lipids with short lipid chains for oligonucleotide delivery
KR20180083440A (en) Novel low molecular weight cationic lipids for oligonucleotide delivery

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: 11838596

Country of ref document: EP

Kind code of ref document: A2

WWE Wipo information: entry into national phase

Ref document number: 13883487

Country of ref document: US

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2011838596

Country of ref document: EP