WO2019226925A1 - Thioester cationic lipids - Google Patents

Thioester cationic lipids Download PDF

Info

Publication number
WO2019226925A1
WO2019226925A1 PCT/US2019/033806 US2019033806W WO2019226925A1 WO 2019226925 A1 WO2019226925 A1 WO 2019226925A1 US 2019033806 W US2019033806 W US 2019033806W WO 2019226925 A1 WO2019226925 A1 WO 2019226925A1
Authority
WO
WIPO (PCT)
Prior art keywords
cationic lipid
independently
alkyl
cationic
lipid
Prior art date
Application number
PCT/US2019/033806
Other languages
French (fr)
Inventor
Yi Zhang
Shrirang KARVE
Frank Derosa
Michael Heartlein
Original Assignee
Translate Bio, Inc.
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 Translate Bio, Inc. filed Critical Translate Bio, Inc.
Priority to JP2020565307A priority Critical patent/JP7488193B2/en
Priority to CN201980048008.6A priority patent/CN112437767B/en
Priority to CA3100254A priority patent/CA3100254A1/en
Priority to AU2019275068A priority patent/AU2019275068A1/en
Priority to CN202311296793.1A priority patent/CN117430538A/en
Priority to US17/057,929 priority patent/US20220008338A1/en
Priority to EP19731398.4A priority patent/EP3802487A1/en
Publication of WO2019226925A1 publication Critical patent/WO2019226925A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C327/00Thiocarboxylic acids
    • C07C327/20Esters of monothiocarboxylic acids
    • C07C327/30Esters of monothiocarboxylic acids having sulfur atoms of esterified thiocarboxyl groups bound to carbon atoms of hydrocarbon radicals substituted by nitrogen atoms, not being part of nitro or nitroso groups
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/7105Natural ribonucleic acids, i.e. containing only riboses attached to adenine, guanine, cytosine or uracil and having 3'-5' phosphodiester links
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/45Transferases (2)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/52Isomerases (5)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/20Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing sulfur, e.g. dimethyl sulfoxide [DMSO], docusate, sodium lauryl sulfate or aminosulfonic acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/127Liposomes
    • A61K9/1271Non-conventional liposomes, e.g. PEGylated liposomes, liposomes coated with polymers
    • A61K9/1272Non-conventional liposomes, e.g. PEGylated liposomes, liposomes coated with polymers with substantial amounts of non-phosphatidyl, i.e. non-acylglycerophosphate, surfactants as bilayer-forming substances, e.g. cationic lipids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C327/00Thiocarboxylic acids
    • C07C327/20Esters of monothiocarboxylic acids
    • C07C327/22Esters of monothiocarboxylic acids having carbon atoms of esterified thiocarboxyl groups bound to hydrogen atoms or to acyclic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D245/00Heterocyclic compounds containing rings of more than seven members having two nitrogen atoms as the only ring hetero atoms
    • C07D245/02Heterocyclic compounds containing rings of more than seven members having two nitrogen atoms as the only ring hetero atoms not condensed with other rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D257/00Heterocyclic compounds containing rings having four nitrogen atoms as the only ring hetero atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D257/00Heterocyclic compounds containing rings having four nitrogen atoms as the only ring hetero atoms
    • C07D257/02Heterocyclic compounds containing rings having four nitrogen atoms as the only ring hetero atoms not condensed with other rings

Definitions

  • mRNA messenger RNA
  • the present invention provides, among other things, cationic lipids useful in for delivery of mRNA. Delivery of mRNA provided by cationic lipids described herein can result in targeted delivery, reduce administration frequency, improve patient tolerability, and provide more potent and less toxic mRNA therapy for the treatment of a variety of diseases, including but not limited to cancer, cardiovascular, cystic fibrosis, infectious, and neurological diseases. [0004] In one aspect, the present invention provides a cationic lipid of Formula (I):
  • R 1 is hydrogen, C 6 -C 30 alkyl, C 6 -C 30 alkenyl, or C 6 -C 30 alkynyl, or Substructure Y; each a and b is an integer of 0-6;
  • each X A1 and X B1 is independently O or S;
  • each L A and L B is independently C1-C10 alkylene; C2-C10 alkenylene; or C2-C10 alkynylene;
  • X A2 is independently NH, NR A , CH2, or CHR A ;
  • X B2 is independently NH, NR B , CH 2 , or CHR B ; each R A and R B is independently C 6 -C 30 alkyl, C 6 -C 30 alkenyl, or C 6 -C 30 alkynyl; and
  • the present invention provides a cationic lipid of Formula (II):
  • R 1 is hydrogen, C6-C30 alkyl, C6-C30 alkenyl, or C6-C30 alkynyl, or Substructure Z;
  • each a and b is an integer of 0-6;
  • each X A1 and X B1 is independently O or S;
  • each L A and L B is independently C1-C10 alkylene; C2-C10 alkenylene; or C2-C10 alkynylene;
  • X A2 is independently NH, NR A , CH2, or CHR A ;
  • X B2 is independently NH, NR B , CH 2 , or CHR B ;
  • each R A and R B is independently C 6 -C 30 alkyl, C 6 -C 30 alkenyl, or C 6 -C 30 alkynyl;
  • the present invention provides a cationic lipid of Formula (III):
  • each a is an integer of 0-6;
  • each X A1 is independently O or S;
  • each L A is independently C1-C10 alkylene; C2-C10 alkenylene; or C2-C10 alkynylene;
  • X A2 is independently NH, NR A , CH 2 , or CHR A ;
  • each R A is independently C 6 -C 30 alkyl, C 6 -C 30 alkenyl, or C 6 -C 30 alkynyl.
  • the present invention provides cationic lipid having a structure according to Formula (IV),
  • R 1 is hydrogen, C1-C30 alkyl, C2-C30 alkenyl, or C2-C30 alkynyl;
  • each a and b is an integer of 0-6;
  • each X A1 and X B1 is independently O or S;
  • each L A and L B is independently C 1 -C 10 alkylene; C 2 -C 10 alkenylene; or C 2 -C 10 alkynylene; L C is independently–C(O)– or–(CH 2 ) b –;
  • X A2 is independently NH, NR A , CH 2 , or CHR A ;
  • X B2 is independently NH, NR B , CH2, or CHR B ;
  • each R A and R B is independently C6-C30 alkyl, C6-C30 alkenyl, or C6-C30 alkynyl.
  • L C is–C(O)–.
  • L C is–(CH2)b–.
  • a cationic lipid has a structure according to Formula (I-A),
  • a cationic lipid has a structure according to Formula (I-A’),
  • a cationic lipid has a structure according to Formula (IV-A):
  • each X A1 and X B1 is O, or each X A1 and X B1 is S. In embodiments, each X A1 and X B1 is O. In embodiments, each X A1 and X B1 is S.
  • each a and b is independently 0, 1, or 2.
  • each X A2 is NR A or CHR A .
  • each X B2 is NR B or CHR B .
  • a cationic lipid has a structure according to Formula (I-B),
  • a cationic lipid has a structure according to Formula (I-B’),
  • a cationic lipid has a structure according to Formula (I-B”),
  • a cationic lipid has a structure according to Formula (II-A),
  • a cationic lipid has a structure according to Formula (III-A),
  • a cationic lipid has a structure according to Formula (IV-B),
  • a cationic lipid has a structure according to Formula (IV-B’),
  • a cationic lipid has a structure according to Formula (IV-B”),
  • the present invention provides a cationic lipid has a structure according to Formula (V),
  • each L A and L B is independently C1-C10 alkylene; C2-C10 alkenylene; or C2-C10 alkynylene; and
  • each R A and R B is independently C 6 -C 30 alkyl, C 6 -C 30 alkenyl, C 6 -C 30 alkynyl, or C 1 -C 15 alkylene-C(O) 2 - C 1 -C 15 alkyl.
  • each L A is C 1 -C 10 alkylene.
  • each L B is C1-C10 alkylene.
  • each L A and L B is unsubstituted C1-C10 alkylene.
  • a cationic lipid has a structure according to Formula (I-C),
  • each c is independently an integer of 2-10.
  • a cationic lipid has a structure according to Formula (I-C’),
  • each c is independently an integer of 2-10.
  • a cationic lipid has a structure according to Formula (I-C”), wherein each c is independently an integer of 2-10, and d is independently an integer of 0-5. In embodiments, d is 0, 1, 2, 3, or 4.
  • a cationic lipid has a structure according to Formula (II-B),
  • a cationic lipid has a structure according to Formula (III-B),
  • each c is independently an integer of 2-10.
  • a cationic lipid has a structure according to Formula (III-C),
  • each c is independently an integer of 2-10.
  • a cationic lipid has a structure according to Formula (III-C’),
  • each c is independently an integer of 2-10.
  • a cationic lipid has a structure according to Formula (IV-C”),
  • the present invention provides a cationic lipid has a structure according to Formula (VI),
  • each R A and R B is independently C 1 -C 30 alkyl, C 2 -C 30 alkenyl, C 2 -C 30 alkynyl, or C 1 - C 15 alkylene-C(O) 2 - C 1 -C 15 alkyl;
  • each c is independently an integer of 2-10.
  • each c is 2, 3, or 4. In embodiments, each c is 4, 5, 6, 7, 8, 9, or 10. In
  • each c is 4. [0038] In embodiments, each R A is C6-C20 alkyl or C6-C20 alkenyl. In embodiments, each R A is
  • each R B is C 6 -C 20 alkyl or C 6 -C 20 alkenyl. In embodiments, each R B is
  • each R A and R B is C 6 -C 20 hydroxyalkyl, or each R A and R B is C 6 -C 20
  • each R A and R B is -CH 2 CH(OH)R C , and wherein R C is selected from the group consisting of:
  • the present invention also provides a cationic lipid that is any one of compounds 1-156.
  • a cationic lipid is
  • the present invention also provides a cationic lipid that is:
  • the invention features a composition comprising an mRNA encoding a
  • a cationic lipid of any of Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III-B), (IV-A), (IV-B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)-(156)).
  • a composition comprises an mRNA encoding for cystic fibrosis transmembrane conductance regulator (CFTR) protein.
  • CFTR cystic fibrosis transmembrane conductance regulator
  • a composition comprises an mRNA encoding for ornithine transcarbamylase (OTC) protein.
  • OTC ornithine transcarbamylase
  • a composition comprises an mRNA encoding for an antigen (e.g., an antigen from an infectious agent).
  • the invention features a composition comprising a nucleic acid encapsulated within a liposome, wherein the liposome comprises a cationic lipid as described herein (e.g., a cationic lipid of any of Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III-B), (IV-A), (IV-B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)-(156)).
  • a cationic lipid as described herein
  • a composition further comprises one or more lipids selected from the group consisting of one or more cationic lipids, one or more non-cationic lipids, and one or more PEG- modified lipids.
  • a nucleic acid is an mRNA encoding a peptide or polypeptide.
  • an mRNA encodes a peptide or polypeptide for use in the delivery to or
  • an mRNA encodes cystic fibrosis transmembrane conductance regulator (CFTR) protein.
  • CFTR cystic fibrosis transmembrane conductance regulator
  • an mRNA encodes a peptide or polypeptide for use in the delivery to or
  • an mRNA encodes ornithine transcarbamylase (OTC) protein.
  • OTC ornithine transcarbamylase
  • an mRNA encodes a peptide or polypeptide for use in a vaccine.
  • an mRNA encodes an antigen (e.g., an antigen from an infectious agent).
  • an antigen e.g., an antigen from an infectious agent.
  • a composition is formulated for intravenous (IV) administration.
  • a composition is formulated for intramuscular (IM) administration.
  • a composition is formulated for administration by inhalation (e.g., a composition is formulated for nebulization).
  • the present invention provides methods of treating a disease in a subject comprising administering to the subject a composition (e.g., a pharmaceutical composition) as described herein (e.g., a composition comprising a cationic lipid of any of Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III-B), (IV-A), (IV-B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)-(156)).
  • a composition e.g., a pharmaceutical composition
  • FIG.1 shows hEPO protein expression following intravenous (IV) administration of lipid
  • nanoparticle formulations comprising a cationic lipid described herein and mRNA encoding hEPO. Protein expression was determined using ELISA.
  • an amino acid has the general structure H 2 N–C(H)(R)–COOH.
  • an amino acid is a naturally occurring amino acid.
  • an amino acid is a synthetic amino acid; in some embodiments, an amino acid is a d-amino acid; in some
  • an amino acid is an l-amino acid.
  • “Standard amino acid” refers to any of the twenty standard l-amino acids commonly found in naturally occurring peptides.
  • “Nonstandard amino acid” refers to any amino acid, other than the standard amino acids, regardless of whether it is prepared synthetically or obtained from a natural source.
  • “synthetic amino acid” is a synthetic amino acid.
  • amino acids encompasses chemically modified amino acids, including but not limited to salts, amino acid derivatives (such as amides), and/or substitutions.
  • Amino acids including carboxy- and/or amino- terminal amino acids in peptides, can be modified by methylation, amidation, acetylation, protecting groups, and/or substitution with other chemical groups that can change the peptide’s circulating half-life without adversely affecting their activity.
  • Amino acids may participate in a disulfide bond.
  • Amino acids may comprise one or posttranslational modifications, such as association with one or more chemical entities (e.g., methyl groups, acetate groups, acetyl groups, phosphate groups, formyl moieties, isoprenoid groups, sulfate groups, polyethylene glycol moieties, lipid moieties, carbohydrate moieties, biotin moieties, etc.).
  • the term“amino acid” is used interchangeably with“amino acid residue,” and may refer to a free amino acid and/or to an amino acid residue of a peptide. It will be apparent from the context in which the term is used whether it refers to a free amino acid or a residue of a peptide.
  • Animal refers to any member of the animal kingdom. In some embodiments,“animal” refers to humans, at any stage of development. In some
  • “animal” refers to non-human animals, at any stage of development.
  • the non-human animal is a mammal (e.g., a rodent, a mouse, a rat, a rabbit, a monkey, a dog, a cat, a sheep, cattle, a primate, and/or a pig).
  • animals include, but are not limited to, mammals, birds, reptiles, amphibians, fish, insects, and/or worms.
  • an animal may be a transgenic animal, genetically-engineered animal, and/or a clone.
  • the term“approximately” or“about,” as applied to one or more values of interest, refers to a value that is similar to a stated reference value.
  • the term“approximately” or“about” refers to a range of values that fall within 25%, 20%, 19%, 18%, 17%, 16%, 15%, 14%, 13%, 12%, 11%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, or less in either direction (greater than or less than) of the stated reference value unless otherwise stated or otherwise evident from the context (except where such number would exceed 100% of a possible value).
  • Biologically active refers to a characteristic of any agent that has activity in a biological system, and particularly in an organism. For instance, an agent that, when administered to an organism, has a biological effect on that organism, is considered to be biologically active.
  • Delivery As used herein, the term“delivery” encompasses both local and systemic delivery.
  • delivery of mRNA encompasses situations in which an mRNA is delivered to a target tissue and the encoded protein is expressed and retained within the target tissue (also referred to as“local distribution” or“local delivery”), and situations in which an mRNA is delivered to a target tissue and the encoded protein is expressed and secreted into patient’s circulation system (e.g., serum) and systematically distributed and taken up by other tissues (also referred to as“systemic distribution” or“systemic delivery”).
  • circulation system e.g., serum
  • expression refers to translation of an mRNA into a polypeptide, assemble multiple polypeptides into an intact protein (e.g., enzyme) and/or post-translational modification of a polypeptide or fully assembled protein (e.g., enzyme).
  • the terms“expression” and“production,” and grammatical equivalent are used inter-changeably.
  • Functional As used herein, a“functional” biological molecule is a biological molecule in a form in which it exhibits a property and/or activity by which it is characterized.
  • Half-life As used herein, the term“half-life” is the time required for a quantity such as nucleic acid or protein concentration or activity to fall to half of its value as measured at the beginning of a time period.
  • Improve, increase, or reduce As used herein, the terms“improve,”“increase” or“reduce,” or grammatical equivalents, indicate values that are relative to a baseline measurement, such as a measurement in the same individual prior to initiation of the treatment described herein, or a measurement in a control subject (or multiple control subject) in the absence of the treatment described herein.
  • A“control subject” is a subject afflicted with the same form of disease as the subject being treated, who is about the same age as the subject being treated.
  • In Vitro As used herein, the term“in vitro” refers to events that occur in an artificial
  • the term“in vivo” refers to events that occur within a multi-cellular organism, such as a human and a non-human animal. In the context of cell-based systems, the term may be used to refer to events that occur within a living cell (as opposed to, for example, in vitro systems).
  • Isolated refers to a substance and/or entity that has been (1) separated from at least some of the components with which it was associated when initially produced (whether in nature and/or in an experimental setting), and/or (2) produced, prepared, and/or manufactured by the hand of man. Isolated substances and/or entities may be separated from about 10%, about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, about 91%, about 92%, about 93%, about 94%, about 95%, about 96%, about 97%, about 98%, about 99%, or more than about 99% of the other components with which they were initially associated.
  • isolated agents are about 80%, about 85%, about 90%, about 91%, about 92%, about 93%, about 94%, about 95%, about 96%, about 97%, about 98%, about 99%, or more than about 99% pure.
  • a substance is“pure” if it is substantially free of other components.
  • calculation of percent purity of isolated substances and/or entities should not include excipients (e.g., buffer, solvent, water, etc.).
  • mRNA as used herein encompasses both modified and unmodified RNA.
  • mRNA may contain one or more coding and non-coding regions.
  • mRNA can be purified from natural sources, produced using recombinant expression systems and optionally purified, chemically synthesized, etc. Where appropriate, e.g., in the case of chemically synthesized molecules, mRNA can comprise nucleoside analogs such as analogs having chemically modified bases or sugars, backbone modifications, etc.
  • An mRNA sequence is presented in the 5’ to 3’ direction unless otherwise indicated.
  • an mRNA is or comprises natural nucleosides (e.g., adenosine, guanosine, cytidine, uridine); nucleoside analogs (e.g., 2- aminoadenosine, 2-thiothymidine, inosine, pyrrolo-pyrimidine, 3-methyl adenosine, 5- methylcytidine, C-5 propynyl-cytidine, C-5 propynyl-uridine, 2-aminoadenosine, C5-bromouridine, C5-fluorouridine, C5-iodouridine, C5-propynyl-uridine, C5-propynyl-cytidine, C5-methylcytidine, 2- aminoadenosine, 7-deazaadenosine, 7-deazaguanosine, 8-oxoadenosine, 8-oxoguanosine, O(6)- methylguan
  • nucleic acid is a compound and/or substance that is or can be incorporated into a polynucleotide chain via a phosphodiester linkage.
  • “nucleic acid” refers to individual nucleic acid residues (e.g., nucleotides and/or nucleosides).
  • nucleic acid refers to a polynucleotide chain comprising individual nucleic acid residues.
  • “nucleic acid” encompasses RNA as well as single and/or double-stranded DNA and/or cDNA.
  • “nucleic acid” encompasses ribonucleic acids (RNA), including but not limited to any one or more of interference RNAs (RNAi), small interfering RNA (siRNA), short hairpin RNA (shRNA), antisense RNA (aRNA), messenger RNA (mRNA), modified messenger RNA (mmRNA), long non-coding RNA (lncRNA), micro-RNA (miRNA) multimeric coding nucleic acid (MCNA), polymeric coding nucleic acid (PCNA), guide RNA (gRNA) and CRISPR RNA (crRNA).
  • RNAi interference RNAs
  • siRNA small interfering RNA
  • shRNA short hairpin RNA
  • aRNA antisense RNA
  • messenger RNA messenger RNA
  • mmRNA modified messenger RNA
  • lncRNA micro-RNA
  • MCNA micro-RNA
  • PCNA polymeric coding nucleic acid
  • gRNA guide RNA
  • crRNA CRISPR RNA
  • DNA may be in the form of antisense DNA, plasmid DNA, parts of a plasmid DNA, pre-condensed DNA, a product of a polymerase chain reaction (PCR), vectors (e.g., P1, PAC, BAC, YAC, artificial chromosomes), expression cassettes, chimeric sequences, chromosomal DNA, or derivatives of these groups.
  • PCR polymerase chain reaction
  • RNA may be in the form of messenger RNA (mRNA), ribosomal RNA (rRNA), signal recognition particle RNA (7 SL RNA or SRP RNA), transfer RNA (tRNA), transfer-messenger RNA (tmRNA), small nuclear RNA (snRNA), small nucleolar RNA (snoRNA), SmY RNA, small Cajal body-specific RNA (scaRNA), guide RNA (gRNA), ribonuclease P (RNase P), Y RNA, telomerase RNA component (TERC), spliced leader RNA (SL RNA), antisense RNA (aRNA or asRNA), cis-natural antisense transcript (cis-NAT), CRISPR RNA (crRNA), long noncoding RNA (lncRNA), micro- RNA (miRNA), piwi-interacting RNA (piRNA), small interfering RNA (siRNA), transacting siRNA (tasiRNA), repeat associated siRNA (rasiRNA),
  • composition may be administered, e.g., for experimental, diagnostic, prophylactic, cosmetic, and/or therapeutic purposes.
  • Typical patients include animals (e.g., mammals such as mice, rats, rabbits, non-human primates, and/or humans).
  • a patient is a human.
  • a human includes pre- and post-natal forms.
  • pharmaceutically acceptable refers to substances that, within the scope of sound medical judgment, are suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • Pharmaceutically acceptable salt Pharmaceutically acceptable salts are well known in the art.
  • Pharmaceutically acceptable salts of the compounds of this invention include those derived from suitable inorganic and organic acids and bases.
  • suitable inorganic and organic acids and bases examples include those derived from suitable inorganic and organic acids and bases.
  • pharmaceutically acceptable, nontoxic acid addition salts are salts of an amino group formed with inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid or with organic acids such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid or rnalonic acid or by using other methods used in the art such as ion exchange.
  • Other pharmaceutically acceptable salts include adipate, alginate, ascorbate, aspartate,
  • benzenesulfonate benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, formate, fumarate, glucoheptonate, glycerophosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2- hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectinate, persulfate, 3-phenylpropionate, phosphate, picrate, pivalate
  • Salts derived from appropriate bases include alkali metal, alkaline earth metal, ammonium and N + (C1-4 alkyl)4 salts.
  • Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like.
  • Further pharmaceutically acceptable salts include, when appropriate, nontoxic ammonium. quaternary ammonium, and amine cations formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, sulfonate and aryl sulfonate.
  • Systemic distribution or delivery As used herein, the terms“systemic distribution,”“systemic delivery,” or grammatical equivalent, refer to a delivery or distribution mechanism or approach that affect the entire body or an entire organism. Typically, systemic distribution or delivery is accomplished via body’s circulation system, e.g., blood stream.
  • Subject refers to a human or any non-human animal (e.g., mouse, rat, rabbit, dog, cat, cattle, swine, sheep, horse or primate).
  • a human includes pre- and post-natal forms.
  • a subject is a human being.
  • a subject can be a patient, which refers to a human presenting to a medical provider for diagnosis or treatment of a disease.
  • the term“subject” is used herein interchangeably with“individual” or“patient.”
  • a subject can be afflicted with or is susceptible to a disease or disorder but may or may not display symptoms of the disease or disorder.
  • Target tissues refers to any tissue that is affected by a disease to be treated. In some embodiments, target tissues include those tissues that display disease-associated pathology, symptom, or feature.
  • therapeutically effective amount As used herein, the term“therapeutically effective amount” of a therapeutic agent means an amount that is sufficient, when administered to a subject suffering from or susceptible to a disease, disorder, and/or condition, to treat, diagnose, prevent, and/or delay the onset of the symptom(s) of the disease, disorder, and/or condition. It will be appreciated by those of ordinary skill in the art that a therapeutically effective amount is typically administered via a dosing regimen comprising at least one unit dose.
  • Treating refers to any method used to partially or completely alleviate, ameliorate, relieve, inhibit, prevent, delay onset of, reduce severity of and/or reduce incidence of one or more symptoms or features of a particular disease, disorder, and/or condition. Treatment may be administered to a subject who does not exhibit signs of a disease and/or exhibits only early signs of the disease for the purpose of decreasing the risk of developing pathology associated with the disease.
  • Aliphatic As used herein, the term aliphatic refers to C1-C40 hydrocarbons and includes both saturated and unsaturated hydrocarbons. An aliphatic may be linear, branched, or cyclic.
  • C 1 -C 20 aliphatics can include C 1 -C 20 alkyls (e.g., linear or branched C 1 -C 20 saturated alkyls), C 2 -C 20 alkenyls (e.g., linear or branched C 4 -C 20 dienyls, linear or branched C 6 -C 20 trienyls, and the like), and C2-C20 alkynyls (e.g., linear or branched C2-C20 alkynyls).
  • C 1 -C 20 alkyls e.g., linear or branched C 1 -C 20 saturated alkyls
  • C 2 -C 20 alkenyls e.g., linear or branched C 4 -C 20 dienyls, linear or branched C 6 -C 20 trienyls, and the like
  • C2-C20 alkynyls e.g., linear or branched C2-C20 alkyny
  • C1-C20 aliphatics can include C3-C20 cyclic aliphatics (e.g., C3-C20 cycloalkyls, C4-C20 cycloalkenyls, or C8-C20 cycloalkynyls).
  • the aliphatic may comprise one or more cyclic aliphatic and/or one or more heteroatoms such as oxygen, nitrogen, or sulfur and may optionally be substituted with one or more substituents such as alkyl, halo, alkoxyl, hydroxy, amino, aryl, ether, ester or amide.
  • An aliphatic group is unsubstituted or substituted with one or more substituent groups as described herein.
  • an aliphatic may be substituted with one or more (e.g., 1, 2, 3, 4, 5, or 6 independently selected substituents) of halogen, -COR’, -CO2H, -CO2R’, -CN, -OH, -OR’, -OCOR’, -OCO2R’, -NH2, -NHR’, -N(R’)2, -SR’ or-SO2R’, wherein each instance of R’ independently is C1-C20 aliphatic (e.g., C1- C20 alkyl, C1-C15 alkyl, C1-C10 alkyl, or C1-C3 alkyl).
  • R independently is C1-C20 aliphatic (e.g., C1- C20 alkyl, C1-C15 alkyl, C1-C10 alkyl, or C1-C3 alkyl).
  • R’ independently is an unsubstituted alkyl (e.g., unsubstituted C1-C20 alkyl, C1-C15 alkyl, C1-C10 alkyl, or C1-C3 alkyl). In embodiments, R’ independently is unsubstituted C 1 -C 3 alkyl. In embodiments, the aliphatic is unsubstituted. In embodiments, the aliphatic does not include any heteroatoms. [0083] Alkyl: As used herein, the term“alkyl” means acyclic linear and branched hydrocarbon groups, e.g.“C1-C20 alkyl” refers to alkyl groups having 1-20 carbons.
  • An alkyl group may be linear or branched.
  • alkyl groups include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, butyl, isobutyl, sec-butyl, tert-butyl, pentyl, isopentyl tert-pentylhexyl, Isohexyletc.
  • Other alkyl groups will be readily apparent to those of skill in the art given the benefit of the present disclosure.
  • An alkyl group may be unsubstituted or substituted with one or more substituent groups as described herein.
  • an alkyl group may be substituted with one or more (e.g., 1, 2, 3, 4, 5, or 6 independently selected substituents) of halogen, -COR’, -CO 2 H, -CO 2 R’, -CN, -OH, -OR’, -OCOR’, -OCO2R’, -NH2, -NHR’, -N(R’)2, -SR’ or-SO2R’, wherein each instance of R’ independently is C1- C20 aliphatic (e.g., C1-C20 alkyl, C1-C15 alkyl, C1-C10 alkyl, or C1-C3 alkyl).
  • R independently is C1- C20 aliphatic (e.g., C1-C20 alkyl, C1-C15 alkyl, C1-C10 alkyl, or C1-C3 alkyl).
  • R’ independently is an unsubstituted alkyl (e.g., unsubstituted C1-C20 alkyl, C1-C15 alkyl, C1-C10 alkyl, or C1-C3 alkyl). In embodiments, R’ independently is unsubstituted C1-C3 alkyl. In embodiments, the alkyl is substituted (e.g., with 1, 2, 3, 4, 5, or 6 substituent groups as described herein). In embodiments, an alkyl group is substituted with a–OH group and may also be referred to herein as a “hydroxyalkyl” group, where the prefix denotes the–OH group and“alkyl” is as described herein. [0084] Alkylene: The term“alkylene,” as used herein, represents a saturated divalent straight or
  • branched chain hydrocarbon group and is exemplified by methylene, ethylene, isopropylene and the like.
  • the term“alkenylene” as used herein represents an unsaturated divalent straight or branched chain hydrocarbon group having one or more unsaturated carbon-carbon double bonds that may occur in any stable point along the chain
  • the term“alkynylene” herein represents an unsaturated divalent straight or branched chain hydrocarbon group having one or more unsaturated carbon-carbon triple bonds that may occur in any stable point along the chain.
  • an alkylene, alkenylene, or alkynylene group may comprise one or more cyclic aliphatic and/or one or more heteroatoms such as oxygen, nitrogen, or sulfur and may optionally be substituted with one or more substituents such as alkyl, halo, alkoxyl, hydroxy, amino, aryl, ether, ester or amide.
  • an alkylene, alkenylene, or alkynylene may be substituted with one or more (e.g., 1, 2, 3, 4, 5, or 6 independently selected substituents) of halogen, -COR’, -CO 2 H, -CO 2 R’, -CN, -OH, -OR’, -OCOR’, -OCO 2 R’, -NH 2 , -NHR’, -N(R’) 2 , -SR’ or-SO 2 R’, wherein each instance of R’ independently is C 1 -C 20 aliphatic (e.g., C 1 -C 20 alkyl, C 1 -C 15 alkyl, C 1 -C 10 alkyl, or C 1 -C 3 alkyl).
  • R independently is C 1 -C 20 aliphatic (e.g., C 1 -C 20 alkyl, C 1 -C 15 alkyl, C 1 -C 10 alkyl, or C 1 -C 3 alkyl
  • R’ independently is an unsubstituted alkyl (e.g., unsubstituted C1-C20 alkyl, C1-C15 alkyl, C1-C10 alkyl, or C1-C3 alkyl). In embodiments, R’ independently is unsubstituted C1-C3 alkyl. In certain embodiments, an alkylene, alkenylene, or alkynylene is unsubstituted. In certain embodiments, an alkylene, alkenylene, or alkynylene does not include any heteroatoms.
  • alkenyl means any linear or branched hydrocarbon chains having one or more unsaturated carbon-carbon double bonds that may occur in any stable point along the chain, e.g.“C 2 -C 20 alkenyl” refers to an alkenyl group having 2-20 carbons.
  • an alkenyl group includes prop-2-enyl, but-2-enyl, but-3-enyl, 2-methylprop-2-enyl, hex-2-enyl, hex-5-enyl, 2,3- dimethylbut-2-enyl, and the like.
  • the alkenyl comprises 1, 2, or 3 carbon-carbon double bond.
  • the alkenyl comprises a single carbon-carbon double bond. In embodiments, multiple double bonds (e.g., 2 or 3) are conjugated.
  • An alkenyl group may be unsubstituted or substituted with one or more substituent groups as described herein. For example, an alkenyl group may be substituted with one or more (e.g., 1, 2, 3, 4, 5, or 6
  • R’ independently selected substituents of halogen, -COR’, -CO 2 H, -CO 2 R’, -CN, -OH, -OR’, -OCOR’, - OCO2R’, -NH2, -NHR’, -N(R’)2, -SR’ or-SO2R’, wherein each instance of R’ independently is C1-C20 aliphatic (e.g., C1-C20 alkyl, C1-C15 alkyl, C1-C10 alkyl, or C1-C3 alkyl).
  • R’ independently is an unsubstituted alkyl (e.g., unsubstituted C1-C20 alkyl, C1-C15 alkyl, C1-C10 alkyl, or C1-C3 alkyl). In embodiments, R’ independently is unsubstituted C1-C3 alkyl. In embodiments, the alkenyl is unsubstituted. In embodiments, the alkenyl is substituted (e.g., with 1, 2, 3, 4, 5, or 6 substituent groups as described herein).
  • alkenyl group is substituted with a–OH group and may also be referred to herein as a“hydroxyalkenyl” group, where the prefix denotes the–OH group and“alkenyl” is as described herein.
  • Alkynyl As used herein,“alkynyl” means any hydrocarbon chain of either linear or branched configuration, having one or more carbon-carbon triple bonds occurring in any stable point along the chain, e.g.“C2-C20 alkynyl” refers to an alkynyl group having 2-20 carbons.
  • an alkynyl group examples include prop-2-ynyl, but-2-ynyl, but-3-ynyl, pent-2-ynyl, 3-methylpent-4-ynyl, hex-2-ynyl, hex- 5-ynyl, etc.
  • an alkynyl comprises one carbon-carbon triple bond.
  • An alkynyl group may be unsubstituted or substituted with one or more substituent groups as described herein.
  • an alkynyl group may be substituted with one or more (e.g., 1, 2, 3, 4, 5, or 6
  • R’ independently is C1-C20 aliphatic (e.g., C1-C20 alkyl, C1-C15 alkyl, C1-C10 alkyl, or C1-C3 alkyl).
  • R’ independently is an unsubstituted alkyl (e.g., unsubstituted C 1 -C 20 alkyl, C 1 -C 15 alkyl, C 1 -C 10 alkyl, or C 1 -C 3 alkyl). In embodiments, R’ independently is unsubstituted C 1 -C 3 alkyl. In embodiments, the alkynyl is unsubstituted. In embodiments, the alkynyl is substituted (e.g., with 1, 2, 3, 4, 5, or 6 substituent groups as described herein).
  • Cycloalkyl As used herein, the term“cycloalkyl” means a nonaromatic, saturated, cyclic group, e.g.“C3-C10 cycloalkyl.” In embodiments, a cycloalkyl is monocyclic. In embodiments, a cycloalkyl is polycyclic (e.g., bicyclic or tricyclic). In polycyclic cycloalkyl groups, individual rings can be fused, bridged, or spirocyclic.
  • cycloalkyl group examples include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, norbornanyl, bicyclo[3.2.1]octanyl, octahydro-pentalenyl, and spiro[4.5]decanyl, and the like.
  • the term“cycloalkyl” may be used interchangeably with the term“carbocycle”.
  • a cycloalkyl group may be unsubstituted or substituted with one or more substituent groups as described herein.
  • a cycloalkyl group may be substituted with one or more (e.g., 1, 2, 3, 4, 5, or 6 independently selected substituents) of halogen, -COR’, -CO2H, -CO2R’, -CN, -OH, -OR’, -OCOR’, -OCO2R’, -NH2, -NHR’, -N(R’)2, -SR’ or-SO2R’, wherein each instance of R’ independently is C1-C20 aliphatic (e.g., C 1 -C 20 alkyl, C 1 -C 15 alkyl, C 1 -C 10 alkyl, or C 1 -C 3 alkyl).
  • R independently is C1-C20 aliphatic (e.g., C 1 -C 20 alkyl, C 1 -C 15 alkyl, C 1 -C 10 alkyl, or C 1 -C 3 alkyl).
  • R’ independently is an unsubstituted alkyl (e.g., unsubstituted C 1 -C 20 alkyl, C 1 -C 15 alkyl, C 1 -C 10 alkyl, or C 1 -C 3 alkyl). In embodiments, R’ independently is unsubstituted C 1 -C 3 alkyl. In embodiments, the cycloalkyl is unsubstituted. In embodiments, the cycloalkyl is substituted (e.g., with 1, 2, 3, 4, 5, or 6 substituent groups as described herein). [0088] Halogen: As used herein, the term“halogen” means fluorine, chlorine, bromine, or iodine. Cationic Lipids
  • Liposomal-based vehicles are considered an attractive carrier for therapeutic agents and remain subject to continued development efforts. While liposomal-based vehicles that comprise a cationic lipid component have shown promising results with regards to encapsulation, stability and site localization, there remains a great need for improvement of liposomal-based delivery systems. For example, a significant drawback of liposomal delivery systems relates to the construction of liposomes that have sufficient cell culture or in vivo stability to reach desired target cells and/or intracellular compartments, and the ability of such liposomal delivery systems to efficiently release their encapsulated materials to such target cells.
  • Cationic lipids disclosed herein comprise a basic, ionizable functional group (e.g., an amine or a nitrogen-containing heteroaryl as described herein), which is present in neutral or charged form.
  • cationic lipids described herein can provide one or more desired
  • cationic lipids described herein can be characterized as having one or more properties that afford such compounds advantages relative to other similarly classified lipids.
  • cationic lipids disclosed herein can allow for the control and tailoring of the properties of liposomal compositions (e.g., lipid nanoparticles) of which they are a component.
  • cationic lipids disclosed herein can be characterized by enhanced transfection efficiencies and their ability to provoke specific biological outcomes. Such outcomes can include, for example enhanced cellular uptake, endosomal/lysosomal disruption capabilities and/or promoting the release of encapsulated materials (e.g., polynucleotides) intracellularly.
  • the present invention provides a cationic lipid of Formula (I):
  • R 1 is hydrogen, C6-C30 alkyl, C6-C30 alkenyl, or C6-C30 alkynyl, Substructure Y, or
  • each a and b is an integer of 0-6;
  • each X A1 and X B1 is independently O or S;
  • each L A and L B is independently C1-C10 alkylene; C2-C10 alkenylene; or C2-C10 alkynylene;
  • X A2 is independently NH, NR A , CH2, or CHR A ;
  • X B2 is independently NH, NR B , CH2, or CHR B ;
  • each R A and R B is independently C6-C30 alkyl, C6-C30 alkenyl, or C6-C30 alkynyl;
  • R 1 is hydrogen. In embodiments, R 1 is C6-C30 alkyl. In embodiments, R 1 is C6-C30 alkenyl. In embodiments, R 1 is C6-C30 alkynyl. In embodiments, R 1 is Substructure Y. [0096] In one aspect, the present invention provides a cationic lipid of Formula (II):
  • R 1 is hydrogen, C6-C30 alkyl, C6-C30 alkenyl, or C6-C30 alkynyl, or Substructure Z;
  • each a and b is an integer of 0-6;
  • each X A1 and X B1 is independently O or S;
  • each L A and L B is independently C1-C10 alkylene; C2-C10 alkenylene; or C2-C10 alkynylene;
  • X A2 is independently NH, NR A , CH 2 , or CHR A ;
  • X B2 is independently NH, NR B , CH 2 , or CHR B ;
  • each R A and R B is independently C 6 -C 30 alkyl, C 6 -C 30 alkenyl, or C 6 -C 30 alkynyl;
  • R 1 is hydrogen. In embodiments, R 1 is C 6 -C 30 alkyl. In embodiments, R 1 is C 6 -C 30 alkenyl. In embodiments, R 1 is C6-C30 alkynyl. In embodiments, R 1 is Substructure Z. [0098] In another aspect, the present invention provides a cationic lipid of Formula (III):
  • each a is an integer of 0-6;
  • each X A1 is independently O or S;
  • each L A is independently C 1 -C 10 alkylene; C 2 -C 10 alkenylene; or C 2 -C 10 alkynylene;
  • X A2 is independently NH, NR A , CH 2 , or CHR A ;
  • each R A is independently C 6 -C 30 alkyl, C 6 -C 30 alkenyl, or C 6 -C 30 alkynyl.
  • a cationic lipid has a structure according to Formula (IV),
  • R 1 is hydrogen, C 1 -C 30 alkyl, C 2 -C 30 alkenyl, or C 2 -C 30 alkynyl;
  • each a and b is an integer of 0-6;
  • each X A1 and X B1 is independently O or S;
  • X B2 is independently NH, NR B , CH 2 , or CHR B ;
  • each R A and R B is independently C 6 -C 30 alkyl, C 6 -C 30 alkenyl, or C 6 -C 30 alkynyl.
  • the present invention provides a cationic lipid has a structure according to Formula (V),
  • each L A and L B is independently C1-C10 alkylene; C2-C10 alkenylene; C2-C10 alkynylene; and each R A and R B is independently C 6 -C 30 alkyl, C 6 -C 30 alkenyl, C 6 -C 30 alkynyl, or , or C 1 -C 15 alkylene-C(O) 2 - C 1 -C 15 alkylene.
  • a cationic lipid has a structure according to Formula (I-A),
  • a cationic lipid has a structure according to Formula (I-A’),
  • a cationic lipid has a structure according to Formula (II-A),
  • a cationic lipid has a structure according to Formula (IV-A):
  • each X A1 and X B1 is O, or each X A1 and X B1 is S. In embodiments, each X A1 and X B1 is O. In embodiments, each X A1 and X B1 is S. [0106] In embodiments, each a and b is independently 0, 1, or 2. In embodiments, each a is 0. In
  • each a is 1. In embodiments, each a is 2. In embodiments, each b is independently 0. In embodiments, each b is 1. In embodiments, each b is 2. In embodiments, a is the same as b. In embodiments, a differs from b. In embodiments, each a and b is 0. In embodiments, each a and b is 1. In embodiments, each a and b is 2. In embodiments, each a is 0 and each b is 1. In
  • each a is 0 and each b is 2. In embodiments, each a is 1 and each b is 0. In embodiments, each a is 1 and each b is 2. In embodiments, each a is 2 and each b is 0. In embodiments, each a is 2 and each b is 1. [0107] In embodiments, each X A2 is NR A or CHR A . In embodiments, each X A2 is NR A . In embodiments, each X A2 is CHR A . [0108] In embodiments, each X B2 is NR B or CHR B . In embodiments, each X B2 is NR B . In embodiments, each X B2 is CHR B .
  • each X A2 is NR A
  • each X B2 is NR B
  • each X A2 is CHR A
  • each X B2 is CHR B
  • a cationic lipid has a structure according to Formula (I-B),
  • a cationic lipid has a structure according to Formula (I-B’),
  • a cationic lipid has a structure according to Formula (I-B”),
  • a cationic lipid has a structure according to Formula (II-A),
  • a cationic lipid has a structure according to Formula (III-A),
  • a cationic lipid has a structure according to Formula (IV-B),
  • a cationic lipid has a structure according to Formula (IV-B’),
  • a cationic lipid has a structure according to Formula (IV-B”),
  • each L A is C 1 -C 10 alkylene (e.g., C 2 -C 10 alkylene, C 4 -C 10 alkylene, or C 4 -C 8
  • each L A is unsubstituted C 1 -C 10 alkylene (e.g., unsubstituted C 2 -C 10 alkylene, unsubstituted C 4 -C 10 alkylene, or unsubstituted C 4 -C 8 alkylene).
  • each L A is substituted C1-C10 alkylene (e.g., substituted C2-C10 alkylene, substituted C4-C10 alkylene, or substituted C4-C8 alkylene).
  • each L A is -(CH2)4-.
  • each L A is -(CH2)2-.
  • each L A is -(CH2)3-.
  • each L A is -(CH2)5-.
  • each R A and R B is unsubstituted C 6 -C 30 alkenyl.
  • each R A and R B is (unsubstituted C 3 -C 15 alkylene)-C(O) 2 -(unsubstituted C 3 -C 15 alkyl).
  • each L B is C 1 -C 10 alkylene (e.g., C 2 -C 10 alkylene, C 4 -C 10 alkylene, or C 4 -C 8
  • each L B is unsubstituted C1-C10 alkylene (e.g., unsubstituted C2-C10 alkylene, unsubstituted C4-C10 alkylene, or unsubstituted C4-C8 alkylene).
  • each L B is substituted C1-C10 alkylene (e.g., substituted C2-C10 alkylene, substituted C4-C10 alkylene, or substituted C4-C8 alkylene).
  • each L B is -(CH2)4-.
  • each L B is -(CH2)2-.
  • each L B is -(CH 2 ) 3 -.
  • each L B is -(CH 2 ) 5 -.
  • each L A and L B is C 1 -C 10 alkylene. In embodiments, each L A and L B is substituted C 1 -C 10 alkylene. In embodiments, each L A and L B is unsubstituted C 1 -C 10 alkylene. In embodiments, each L A and L B is -CH2-. In embodiments, each L A and L B is -(CH2)2-. In embodiments, each L A and L B is -(CH2)3-. In embodiments, each L A and L B is -(CH2)4-. In embodiments, each L A and L B is -(CH2)5-. In embodiments, each L A and L B is -(CH2)6-. In embodiments, each L A and L B is -(CH2)7-. In
  • each L A and L B is -(CH2)8-. In embodiments, each L A and L B is -(CH2)9-. In
  • each L A and L B is -(CH 2 ) 10 -.
  • a cationic lipid has a structure according to Formula (I-C),
  • a cationic lipid has a structure according to Formula (I-C’),
  • a cationic lipid has a structure according to Formula (I-C”), wherein each c is independently an integer of 2-10, and d is independently an integer of 0-5. In embodiments, d is 0, 1, 2, 3, or 4.
  • a cationic lipid has a structure according to Formula (II-B),
  • each c is independently an integer of 2-10.
  • a cationic lipid has a structure according to Formula (III-B),
  • each c is independently an integer of 2-10.
  • a cationic lipid has a structure according to Formula (III-C),
  • a cationic lipid has a structure according to Formula (IV-C’),
  • a cationic lipid has a structure according to Formula (IV-C”),
  • the present invention provides a cationic lipid has a structure according to Formula (VI),
  • each R A and R B is independently C 6 -C 30 alkyl, C 6 -C 30 alkenyl, C 6 -C 30 alkynyl or C1-C15 alkylene-C(O)2- C1-C15 alkylene;
  • each c is independently an integer of 2-10.
  • each c is 4, 5, 6, 7, 8, 9, or 10. In embodiments, each c is 2, 3, or 4. In
  • each c is 2. In embodiments, each c is 3. In embodiments, each c is 4. In
  • each c is 5. In embodiments, each c is 6. In embodiments, each c is 7. In
  • each c is 8. In embodiments, each c is 9. In embodiments, each c is 10. [0132] In embodiments, each R A and R B is unsubstituted C6-C30 alkenyl. In embodiments, each R A and R B is (unsubstituted C3-C15 alkylene)-C(O)2-(unsubstituted C3-C15 alkyl). [0133] In embodiments, each R A is C 6 -C 30 alkyl (e.g., C 6 -C 20 alkyl) or a C 6 -C 30 alkenyl (e.g., C 6 -C 20 alkenyl).
  • each R A is unsubstituted C 6 -C 30 alkyl, C 6 -C 30 hydroxyalkyl, unsubstituted C 6 -C 30 alkenyl, or C 6 -C 30 hydroxyalkenyl. In embodiments, each R A is unsubstituted C 6 -C 20 alkyl, C 6 -C 20 hydroxyalkyl, unsubstituted C 6 -C 20 alkenyl, or C 6 -C 20 hydroxyalkenyl. [0134] In embodiments, each R A is C 6 -C 30 alkyl. In embodiments, each R A is unsubstituted C 6 -C 30 alkyl.
  • each R A is substituted C6-C30 alkyl. In embodiments, each R A is C6-C30 hydroxyalkyl. In embodiments, each R A is a linear C6-C30 alkyl. In embodiments, each R A is a branched C6-C30 alkyl. [0135] In embodiments, each R A is C6-C20 alkyl. In embodiments, each R A is unsubstituted C6-C20 alkyl.
  • each R A is substituted C6-C20 alkyl. In embodiments, each R A is C6-C20 hydroxyalkyl. In embodiments, each R A is a linear C 6 -C 20 alkyl. In embodiments, each R A is a branched C 6 -C 20 alkyl. [0136] In embodiments, each R A is C 6 -C 30 alkenyl. In embodiments, each R A is unsubstituted C 6 -C 30 alkenyl. In embodiments, each R A is substituted C 6 -C 30 alkenyl. In embodiments, each R A is C 6 -C 30 hydroxyalkenyl.
  • each R A is a linear C6-C30 alkenyl. In embodiments, each R A is a branched C6-C30 alkenyl. In embodiments, a C6-C30 alkenyl is a monoalkenyl, a dienyl, or a trienyl. [0137] In embodiments, each R A is C6-C20 alkenyl. In embodiments, each R A is unsubstituted C6-C20 alkenyl. In embodiments, each R A is substituted C6-C20 alkenyl. In embodiments, each R A is C6-C20 hydroxyalkenyl. In embodiments, each R A is a linear C 6 -C 20 alkenyl.
  • each R A is a branched C 6 -C 20 alkenyl. In embodiments, a C 6 -C 20 alkenyl is a monoalkenyl, a dienyl, or a trienyl. [0138] In embodiments, each R A is C 4 -C 10 alkylene-C(O) 2 - C 4 -C 10 alkyl. In embodiments, each R A is unsubstituted C4-C10 alkylene-C(O)2- C4-C10 alkyl. In embodiments, each R A is substituted C4-C10 alkylene-C(O)2- C4-C10 alkyl.
  • each R A is C4-C10 alkylene-C(O)2- C4-C10 hydroxyalkyl.
  • each R B is C6-C30 alkyl (e.g., C6-C20 alkyl) or a C6-C30 alkenyl (e.g., C6-C20 alkenyl).
  • each R B is unsubstituted C6-C30 alkyl, C6-C30 hydroxyalkyl, unsubstituted C6-C30 alkenyl, or C 6 -C 30 hydroxyalkenyl. In embodiments, each R B is unsubstituted C 6 -C 20 alkyl, C 6 -C 20 hydroxyalkyl, unsubstituted C 6 -C 20 alkenyl, or C 6 -C 20 hydroxyalkenyl. [0140] In embodiments, each R B is C 6 -C 20 alkyl. In embodiments, each R B is unsubstituted C 6 -C 20 alkyl.
  • each R B is substituted C6-C20 alkyl. In embodiments, each R B is C6-C20 hydroxyalkyl. In embodiments, each R B is a linear C6-C20 alkyl. In embodiments, each R B is a branched C6-C20 alkyl. [0141] In embodiments, each R B is C6-C30 alkenyl. In embodiments, each R B is unsubstituted C6-C30 alkenyl. In embodiments, each R B is substituted C6-C30 alkenyl. In embodiments, each R B is C6-C30 hydroxyalkenyl. In embodiments, each R B is a linear C 6 -C 30 alkenyl.
  • each R B is a branched C 6 -C 30 alkenyl. In embodiments, a C 6 -C 30 alkenyl is a monoalkenyl, a dienyl, or a trienyl. [0142] In embodiments, each R B is C 6 -C 20 alkenyl. In embodiments, each R B is unsubstituted C 6 -C 20 alkenyl. In embodiments, each R B is substituted C 6 -C 20 alkenyl. In embodiments, each R B is C 6 -C 20 hydroxyalkenyl. In embodiments, each R B is a linear C 6 -C 20 alkenyl.
  • each R B is a branched C6-C20 alkenyl.
  • a C6-C20 alkenyl is a monoalkenyl, a dienyl, or a trienyl.
  • each R B is C4-C10 alkylene-C(O)2- C4-C10 alkyl.
  • each R B is
  • each R B is substituted C4-C10 alkylene-C(O)2- C4-C10 alkyl. In embodiments, each R B is C4-C10 alkylene-C(O)2- C4-C10 hydroxyalkyl. [0144] In embodiments, R A is the same as R B . In embodiments R A differs from R B . [0145] In embodiments, each R A and R B is C 6 -C 30 alkyl. In embodiments, each R A and R B is unsubstituted C 6 -C 30 alkyl.
  • each R A and R B is substituted C 6 -C 30 alkyl. In embodiments, each R A and R B is C6-C30 hydroxyalkyl. In embodiments, each R A and R B is a linear C6-C30 alkyl.
  • each R A and R B is a branched C6-C30 alkyl.
  • each R A and R B is C6-C20 alkyl.
  • each R A and R B is unsubstituted C6-C20 alkyl.
  • each R A and R B is substituted C6-C20 alkyl.
  • each R A and R B is C 6 -C 20 hydroxyalkyl.
  • each R A and R B is a linear C 6 -C 20 alkyl.
  • each R A and R B is a branched C 6 -C 20 alkyl. [0147] In embodiments, each R A and R B is C 6 -C 30 alkenyl. In embodiments, each R A and R B is
  • each R A and R B is substituted C6-C30 alkenyl. In embodiments, each R A and R B is C6-C30 hydroxyalkenyl. In embodiments, each R A and R B is a linear C6-C30 alkenyl. In embodiments, each R A and R B is a branched C6-C30 alkenyl. In embodiments, a C6- C30 alkenyl is a monoalkenyl, a dienyl, or a trienyl. [0148] In embodiments, each R A and R B is C 6 -C 20 alkenyl. In embodiments, each R A and R B is
  • each R A and R B is substituted C 6 -C 20 alkenyl.
  • each R A and R B is C 6 -C 20 hydroxyalkenyl.
  • each R A and R B is a linear C6-C20 alkenyl.
  • each R A and R B is a branched C6-C20 alkenyl.
  • a C6- C20 alkenyl is a monoalkenyl, a dienyl, or a trienyl.
  • each R A and R B is C6-C30 hydroxyalkyl, or each R A and R B is C6-C30
  • each R A and R B is C6-C20 hydroxyalkyl, or each R A and R B is C6-C20 hydroxyalkenyl.
  • each R A and R B is C 4 -C 10 alkylene-C(O) 2 - C 4 -C 10 alkyl.
  • each R A and R B is unsubstituted C 4 -C 10 alkylene-C(O) 2 - C 4 -C 10 alkyl.
  • each R A and R B is substituted C 4 -C 10 alkylene-C(O) 2 - C 4 -C 10 alkyl.
  • each R A and R B is C 4 -C 10 alkylene- C(O) 2 - C 4 -C 10 hydroxyalkyl. [0151] In embodiments, each R A and R B is C 6 -C 30 hydroxyalkyl. In embodiments, each R A and R B is C 6 -C 20 hydroxyalkyl. In embodiments, each R A and R B is -CH2CH(OH)C10H21. In embodiments, each R A and R B is -CH2CH(OH)C8H17. In embodiments, each R A and R B is -CH2CH(OH)C12H25. In embodiments, each R A and R B is -CH2CH(OH)C14H29.
  • each R A and R B is C4-C10 alkylene-C(O)2- C4-C10 hydroxyalkyl.
  • each R A and R B is -CH2CH(OH)(CH2)7C(O)2C7H15.
  • each R A and R B is -CH2CH(OH)R C , and wherein R C is selected from the group consisting of:
  • a C6-C30 alkyl (e.g., each R A and/or each R B ) is a C8-26 alkyl. In embodiments, a C6-C30 alkyl (e.g., each R A and/or each R B ) is a straight-chain C8-26 alkyl.
  • a C 6 -C 30 alkyl (e.g., each R A and/or each R B ) is CH 3 (CH 2 ) 6 CH 2 -, CH 3 (CH 2 ) 7 CH 2 -, CH 3 (CH 2 ) 8 CH 2 -, CH 3 (CH 2 ) 9 CH 2 -, CH 3 (CH 2 ) 10 CH 2 -, CH 3 (CH 2 ) 11 CH 2 -, CH 3 (CH 2 ) 12 CH 2 -, CH 3 (CH 2 ) 13 CH 2 -, CH 3 (CH 2 ) 14 CH 2 -, CH 3 (CH 2 ) 15 CH 2 -, CH 3 (CH 2 ) 16 CH 2 -, CH 3 (CH 2 ) 17 CH 2 -, CH 3 (CH 2 ) 18 CH 2 -, CH 3 (CH 2 ) 19 CH 2 -, CH3(CH2)20CH2-, CH3(CH2)21CH2-, CH3(CH2)22CH2-, CH3(CH2)23CH2- or
  • a C6-C30 alkyl (e.g., each R A and/or each R B ) is CH3(CH2)13CH2-, CH3(CH2)14CH2-, CH3(CH2)15CH2-, CH3(CH2)16CH2-, CH3(CH2)17CH2- or CH3(CH2)18CH2-.
  • a C6-C30 alkyl (e.g., each R A and/or each R B ) is CH3(CH2)14CH2-, CH3(CH2)15CH2- or CH 3 (CH 2 ) 16 CH 2 -.
  • a C 6 -C 30 alkenyl (e.g., each R A and/or each R B ) is a C 8-26 alkenyl having one or two carbon-carbon double bonds.
  • a C6-C30 alkenyl (e.g., each R A and/or each R B ) is C8-26 aliphatic having three, four, five or six carbon-carbon double bonds.
  • a C6-C30 alkenyl (e.g., each R A and/or each R B ) is
  • a C 6 -C 30 alkenyl (e.g., each R A and/or each R B ) is
  • a C6-C30 alkenyl (e.g., each R A and/or each R B ) is
  • each R A and/or each R B independently is an aliphatic chain of a saturated or unsaturated fatty acid, i.e., R'-(CH 2 )- for a fatty acid R'-C(O)-.
  • each R A and/or each R B independently is the aliphatic chain of caprylic, pelargonic, capric, undecylic, lauric, tridecyclic, myristic, pentadecylic, margaric, stearic, nonadecylic, arachidic, heneicosylic, behenic, triosylic, lignoceric, oleic, linoleic, pentacosylic or cerotic acid.
  • each R A and/or each R B is the aliphatic chain of caprylic, pelargonic, capric, undecylic, lauric, tridecyclic, myristic, pentadecylic, or margaric acid.
  • each R A and/or each R B is the aliphatic chain of lauric, tridecyclic, myristic, or pentadecylic acid. In some embodiments, each R A and/or each R B is the aliphatic chain of lauric or myristic acid. In some embodiments, each R A and/or each R B is the aliphatic chain of stearic, nonadecylic, arachidic, heneicosylic, behenic, triosylic, lignoceric, oleic, linoleic, pentacosylic or cerotic acid.
  • each R A and/or each R B is the aliphatic chain of lignoceric, oleic, linoleic, pentacosylic or cerotic acid. In some embodiments, each R A and/or each R B is the aliphatic chain of oleic, linoleic or pentacosylic acid. In some embodiments, each R A and/or each R B is the aliphatic chain of oleic or linoleic acid. In some embodiments, each R A and/or each R B is the aliphatic chain of oleic acid. In some embodiments, each R A and/or each R B is the aliphatic chain of linoleic acid. Exemplary Cationic Lipids
  • Exemplary cationic lipids include cationic lipids such as Cationic Lipid (1), (2) (3), (4), (5), and (6),
  • a cationic lipid is Compound (1). In embodiments, a cationic lipid is
  • a cationic lipid is Compound (3).
  • a cationic lipid is Compound (4).
  • a cationic lipid is Compound (5).
  • a cationic lipid is Compound (6).
  • the present invention also provides a cationic lipid that is:
  • a cationic lipid is Compound (7). In embodiments, a cationic lipid is
  • a cationic lipid is Compound (9). In embodiments, a cationic lipid is Compound (10). In embodiments, a cationic lipid is Compound (11). In embodiments, a cationic lipid is Compound (12). In embodiments, a cationic lipid is Compound (13). In embodiments, a cationic lipid is Compound (14). In embodiments, a cationic lipid is Compound (15). In embodiments, a cationic lipid is Compound (16). [0173] In embodiments, a cationic lipid described herein can be obtained from combination of any of the diacids in Table A and the thiols of Table B, including of the following compounds as described herein. [0174] In embodiments, a cationic lipid is Compound (17). In embodiments, a cationic lipid is Compound (17). In embodiments, a cationic lipid is Compound (17). In embodiments, a cationic lipid is Compound (17). In embodiments,
  • a cationic lipid is Compound (19). In embodiments, a cationic lipid is Compound (20). In embodiments, a cationic lipid is Compound (21). In embodiments, a cationic lipid is Compound (22). In embodiments, a cationic lipid is Compound (23). In embodiments, a cationic lipid is Compound (19). In embodiments, a cationic lipid is Compound (19). In embodiments, a cationic lipid is Compound (20). In embodiments, a cationic lipid is Compound (21). In embodiments, a cationic lipid is Compound (22). In embodiments, a cationic lipid is Compound (23). In
  • a cationic lipid is Compound (24). In embodiments, a cationic lipid is Compound (25). In embodiments, a cationic lipid is Compound (26). In embodiments, a cationic lipid is Compound (27). In embodiments, a cationic lipid is Compound (28). In embodiments, a cationic lipid is Compound (29). In embodiments, a cationic lipid is Compound (30). In embodiments, a cationic lipid is Compound (31). In embodiments, a cationic lipid is Compound (32). In embodiments, a cationic lipid is Compound (33). In embodiments, a cationic lipid is Compound (34). In
  • a cationic lipid is Compound (35). In embodiments, a cationic lipid is Compound (36). In embodiments, a cationic lipid is Compound (37). In embodiments, a cationic lipid is Compound (38). In embodiments, a cationic lipid is Compound (39). In embodiments, a cationic lipid is Compound (40). In embodiments, a cationic lipid is Compound (41). In embodiments, a cationic lipid is Compound (42). In embodiments, a cationic lipid is Compound (43). In embodiments, a cationic lipid is Compound (44). In embodiments, a cationic lipid is Compound (45). In
  • a cationic lipid is Compound (46). [0175] In embodiments, a cationic lipid is Compound (7). In embodiments, a cationic lipid is
  • a cationic lipid is Compound (9). In embodiments, a cationic lipid is Compound (10). In embodiments, a cationic lipid is Compound (11). In embodiments, a cationic lipid is Compound (12). In embodiments, a cationic lipid is Compound (13). In embodiments, a cationic lipid is Compound (14). In embodiments, a cationic lipid is Compound (15). In embodiments, a cationic lipid is Compound (9). In embodiments, a cationic lipid is Compound (10). In embodiments, a cationic lipid is Compound (11). In embodiments, a cationic lipid is Compound (12). In embodiments, a cationic lipid is Compound (13). In embodiments, a cationic lipid is Compound (14). In embodiments, a cationic lipid is Compound (15). In
  • a cationic lipid is Compound (16). In embodiments, a cationic lipid is Compound (47). In embodiments, a cationic lipid is Compound (48). In embodiments, a cationic lipid is Compound (49). In embodiments, a cationic lipid is Compound (50). In embodiments, a cationic lipid is Compound (51). In embodiments, a cationic lipid is Compound (52). In embodiments, a cationic lipid is Compound (53). In embodiments, a cationic lipid is Compound (54). In embodiments, a cationic lipid is Compound (55). In embodiments, a cationic lipid is Compound (56). In
  • a cationic lipid is Compound (57). In embodiments, a cationic lipid is Compound (58). In embodiments, a cationic lipid is Compound (59). In embodiments, a cationic lipid is Compound (60). In embodiments, a cationic lipid is Compound (61). In embodiments, a cationic lipid is Compound (62). In embodiments, a cationic lipid is Compound (63). In embodiments, a cationic lipid is Compound (64). In embodiments, a cationic lipid is Compound (65). In embodiments, a cationic lipid is Compound (66). [0176] In embodiments, a cationic lipid is Compound (67). In embodiments, a cationic lipid is Compound (67). In embodiments, a cationic lipid is a cationic lipid is
  • a cationic lipid is Compound (69). In embodiments, a cationic lipid is Compound (70). In embodiments, a cationic lipid is Compound (71). In embodiments, a cationic lipid is Compound (72). In embodiments, a cationic lipid is Compound (73). In
  • a cationic lipid is Compound (74). In embodiments, a cationic lipid is Compound (75). In embodiments, a cationic lipid is Compound (76). In embodiments, a cationic lipid is Compound (77). In embodiments, a cationic lipid is Compound (78). In embodiments, a cationic lipid is Compound (79). In embodiments, a cationic lipid is Compound (80). In embodiments, a cationic lipid is Compound (81). In embodiments, a cationic lipid is Compound (82). In embodiments, a cationic lipid is Compound (83). In embodiments, a cationic lipid is Compound (84). In
  • a cationic lipid is Compound (85). In embodiments, a cationic lipid is Compound (86). In embodiments, a cationic lipid is Compound (87). In embodiments, a cationic lipid is Compound (88). In embodiments, a cationic lipid is Compound (89). In embodiments, a cationic lipid is Compound (90). In embodiments, a cationic lipid is Compound (91). In embodiments, a cationic lipid is Compound (92). In embodiments, a cationic lipid is Compound (93). In embodiments, a cationic lipid is Compound (94). In embodiments, a cationic lipid is Compound (95). In
  • a cationic lipid is Compound (96). [0177] In embodiments, a cationic lipid is Compound (97). In embodiments, a cationic lipid is
  • a cationic lipid is Compound (99). In embodiments, a cationic lipid is Compound (100). In embodiments, a cationic lipid is Compound (101). In embodiments, a cationic lipid is Compound (102). In embodiments, a cationic lipid is Compound (103). In embodiments, a cationic lipid is Compound (104). In embodiments, a cationic lipid is Compound (105). In embodiments, a cationic lipid is Compound (106). In embodiments, a cationic lipid is Compound (107). In embodiments, a cationic lipid is Compound (108). In embodiments, a cationic lipid is Compound (109).
  • a cationic lipid is Compound (110). In embodiments, a cationic lipid is Compound (111). In embodiments, a cationic lipid is Compound (112). In embodiments, a cationic lipid is Compound (113). In embodiments, a cationic lipid is Compound (114). In embodiments, a cationic lipid is Compound (115). In embodiments, a cationic lipid is Compound (116). In embodiments, a cationic lipid is Compound (117). In embodiments, a cationic lipid is Compound (118). In embodiments, a cationic lipid is Compound (119). In embodiments, a cationic lipid is Compound (120).
  • a cationic lipid is Compound (121). In embodiments, a cationic lipid is Compound (122). In embodiments, a cationic lipid is Compound (123). In embodiments, a cationic lipid is Compound (124). In embodiments, a cationic lipid is Compound (125). In embodiments, a cationic lipid is Compound (126). [0178] In embodiments, a cationic lipid is Compound (127). In embodiments, a cationic lipid is Compound (128). In embodiments, a cationic lipid is Compound (129). In embodiments, a cationic lipid is Compound (130). In embodiments, a cationic lipid is Compound (131).
  • a cationic lipid is Compound (132). In embodiments, a cationic lipid is Compound (133). In embodiments, a cationic lipid is Compound (134). In embodiments, a cationic lipid is Compound (135). In embodiments, a cationic lipid is Compound (136). In embodiments, a cationic lipid is Compound (137). In embodiments, a cationic lipid is Compound (138). In embodiments, a cationic lipid is Compound (139). In embodiments, a cationic lipid is Compound (140). In embodiments, a cationic lipid is Compound (141). In embodiments, a cationic lipid is Compound (142).
  • a cationic lipid is Compound (143). In embodiments, a cationic lipid is Compound (144). In embodiments, a cationic lipid is Compound (145). In embodiments, a cationic lipid is Compound (146). In embodiments, a cationic lipid is Compound (147). In embodiments, a cationic lipid is Compound (148). In embodiments, a cationic lipid is Compound (149). In embodiments, a cationic lipid is Compound (150). In embodiments, a cationic lipid is Compound (151). In embodiments, a cationic lipid is Compound (152). In embodiments, a cationic lipid is Compound (153). In embodiments, a cationic lipid is Compound (154). In embodiments, a cationic lipid is Compound (155). In embodiments, a cationic lipid is Compound (156).
  • Cationic lipids described herein e.g., a cationic lipid of any of Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III-B), (IV-A), (IV-B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)-(156)).
  • cationic lipids of the present invention may be prepared via thioesterification of a starting carboxylic acid followed by deprotection.
  • the cationic lipids described e.g., Cationic Lipid (1) herein may be prepared according to Scheme 1.
  • an alkylating agent such as Compound A can be treated with a thiol reagent such as benzyl mercaptan to afford a bis-protected difunctional intermediate such as Compound B.
  • a thiol reagent such as benzyl mercaptan
  • Deprotection of the phthalimide moiety of Compound B affords the nucleophilic Compound C, which in turn can be treated with another electrophile such as an epoxide (Compound D) to provide a tertiary amine (Compound E).
  • the hydroxyl groups of Compound E can be protected using protecting groups and conditions known in the art (e.g., TBSCl/imidazole) to afford the
  • Cationic lipids described herein e.g., a cationic lipid of any of Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III-B), (IV-A), (IV-B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)-(156)) can be used to prepare compositions useful for the delivery of nucleic acids. Synthesis of Nucleic Acids
  • Nucleic acids according to the present invention may be synthesized according to any known methods.
  • mRNAs according to the present invention may be synthesized via in vitro transcription (IVT).
  • IVT in vitro transcription
  • a linear or circular DNA template containing a promoter, a pool of ribonucleotide triphosphates, a buffer system that may include DTT and magnesium ions, and an appropriate RNA polymerase (e.g., T3, T7, mutated T7 or SP6 RNA polymerase), DNAse I, pyrophosphatase, and/or RNAse inhibitor.
  • RNA polymerase e.g., T3, T7, mutated T7 or SP6 RNA polymerase
  • a DNA template is transcribed in vitro.
  • a suitable DNA template typically has a promoter, for example a T3, T7, mutated T7 or SP6 promoter, for in vitro transcription, followed by desired nucleotide sequence for desired mRNA and a termination signal.
  • Desired mRNA sequence(s) according to the invention may be determined and incorporated into a DNA template using standard methods. For example, starting from a desired amino acid sequence (e.g., an enzyme sequence), a virtual reverse translation is carried out based on the degenerated genetic code. Optimization algorithms may then be used for selection of suitable codons.
  • the G/C content can be optimized to achieve the highest possible G/C content on one hand, taking into the best possible account the frequency of the tRNAs according to codon usage on the other hand.
  • the optimized RNA sequence can be established and displayed, for example, with the aid of an appropriate display device and compared with the original (wild-type) sequence.
  • a secondary structure can also be analyzed to calculate stabilizing and destabilizing properties or, respectively, regions of the RNA.
  • DNA may be in the form of antisense DNA, plasmid DNA, parts of a plasmid DNA, pre-condensed DNA, a product of a polymerase chain reaction (PCR), vectors (e.g., P1, PAC, BAC, YAC, artificial chromosomes), expression cassettes, chimeric sequences, chromosomal DNA, or derivatives of these groups.
  • PCR polymerase chain reaction
  • RNA may be in the form of messenger RNA (mRNA), ribosomal RNA (rRNA), signal recognition particle RNA (7 SL RNA or SRP RNA), transfer RNA (tRNA), transfer-messenger RNA (tmRNA), small nuclear RNA (snRNA), small nucleolar RNA (snoRNA), SmY RNA, small Cajal body-specific RNA (scaRNA), guide RNA (gRNA), ribonuclease P (RNase P), Y RNA, telomerase RNA component (TERC), spliced leader RNA (SL RNA), antisense RNA (aRNA or asRNA), cis-natural antisense transcript (cis-NAT), CRISPR RNA (crRNA), long noncoding RNA (lncRNA), microRNA (miRNA), piwi-interacting RNA (piRNA), small interfering RNA (siRNA), transacting siRNA (tasiRNA), repeat associated siRNA (rasiRNA), 73K RNA
  • mRNAs according to the present invention may be synthesized according to any of a variety of known methods.
  • mRNAs according to the present invention may be synthesized via in vitro transcription (IVT).
  • IVT in vitro transcription
  • IVT is typically performed with a linear or circular DNA template containing a promoter, a pool of ribonucleotide triphosphates, a buffer system that may include DTT and magnesium ions, and an appropriate RNA polymerase (e.g., T3, T7 or SP6 RNA polymerase), DNAse I, pyrophosphatase, and/or RNAse inhibitor.
  • RNA polymerase e.g., T3, T7 or SP6 RNA polymerase
  • the in vitro transcribing occurs in a single batch.
  • a DNA template is transcribed in vitro.
  • a suitable DNA template typically has a promoter, for example a T3, T7 or SP6 promoter, for in vitro transcription, followed by desired nucleotide sequence for desired mRNA and a termination signal.
  • Desired mRNA sequence(s) according to the invention may be determined and incorporated into a DNA template using standard methods.
  • RNA sequence For example, starting from a desired amino acid sequence (e.g., an enzyme sequence), a virtual reverse translation is carried out based on the degenerated genetic code. Optimization algorithms may then be used for selection of suitable codons. Typically, the G/C content can be optimized to achieve the highest possible G/C content on one hand, taking into the best possible account the frequency of the tRNAs according to codon usage on the other hand.
  • the optimized RNA sequence can be established and displayed, for example, with the aid of an appropriate display device and compared with the original (wild-type) sequence.
  • a secondary structure can also be analyzed to calculate stabilizing and destabilizing properties or, respectively, regions of the RNA. Modified mRNA
  • mRNA according to the present invention may be synthesized as
  • Modified mRNA comprise nucleotide modifications in the RNA.
  • a modified mRNA according to the invention can thus include nucleotide modification that are, for example, backbone modifications, sugar modifications or base modifications.
  • mRNAs may be synthesized from naturally occurring nucleotides and/or nucleotide analogues (modified nucleotides) including, but not limited to, purines (adenine (A), guanine (G)) or pyrimidines (thymine (T), cytosine (C), uracil (U)), and as modified nucleotides analogues or derivatives of purines and pyrimidines, such as e.g.1-methyl-adenine, 2-methyl-adenine, 2- methylthio-N-6-isopentenyl-adenine, N6-methyl-adenine, N6-isopentenyl-adenine, 2-thio-cytosine, 3-methyl-cytosine, 4-acetyl-cytosine, 5-methyl-cytosine, 2,6-diaminopurine, 1-methyl-guanine, 2- methyl-guanine, 2,2-dimethyl-guanine, 7-methyl-guan
  • mRNAs may contain RNA backbone modifications.
  • a backbone modification is a modification in which the phosphates of the backbone of the nucleotides contained in the RNA are modified chemically.
  • Exemplary backbone modifications typically include, but are not limited to, modifications from the group consisting of methylphosphonates,
  • mRNAs may contain sugar modifications.
  • a typical sugar modification is a chemical modification of the sugar of the nucleotides it contains including, but not limited to, sugar modifications chosen from the group consisting of 4'-thio-ribonucleotide (see, e.g., US Patent Application Publication No.
  • mRNAs may contain modifications of the bases of the nucleotides (base modifications).
  • a modified nucleotide which contains a base modification is also called a base- modified nucleotide.
  • base-modified nucleotides include, but are not limited to, 2- amino-6-chloropurine riboside 5'-triphosphate, 2-aminoadenosine 5'-triphosphate, 2-thiocytidine 5'- triphosphate, 2-thiouridine 5'-triphosphate, 4-thiouridine 5'-triphosphate, 5-aminoallylcytidine 5'- triphosphate, 5-aminoallyluridine 5'-triphosphate, 5-bromocytidine 5'-triphosphate, 5-bromouridine 5'-triphosphate, 5-iodocytidine 5'-triphosphate, 5-iodouridine 5'-triphosphate, 5-methylcytidine 5'- triphosphate, 5-methyluridine 5'--
  • mRNA synthesis includes the addition of a“cap” on the N-terminal (5’) end, and a “tail” on the C-terminal (3’) end.
  • the presence of the cap is important in providing resistance to nucleases found in most eukaryotic cells.
  • the presence of a“tail” serves to protect the mRNA from exonuclease degradation.
  • mRNAs include a 5’ cap structure.
  • a 5’ cap is typically added as follows: first, an RNA terminal phosphatase removes one of the terminal phosphate groups from the 5’ nucleotide, leaving two terminal phosphates; guanosine triphosphate (GTP) is then added to the terminal phosphates via a guanylyl transferase, producing a 5’5’5 triphosphate linkage; and the 7- nitrogen of guanine is then methylated by a methyltransferase.
  • Examples of cap structures include, but are not limited to, m7G(5')ppp (5'(A,G(5')ppp(5')A and G(5')ppp(5')G.
  • mRNAs include a 3’ poly(A) tail structure. A poly-A tail on the 3'
  • terminus of mRNA typically includes about 10 to 300 adenosine nucleotides (e.g., about 10 to 200 adenosine nucleotides, about 10 to 150 adenosine nucleotides, about 10 to 100 adenosine nucleotides, about 20 to 70 adenosine nucleotides, or about 20 to 60 adenosine nucleotides).
  • mRNAs include a 3’ poly(C) tail structure.
  • a suitable poly-C tail on the 3' terminus of mRNA typically include about 10 to 200 cytosine nucleotides (e.g., about 10 to 150 cytosine nucleotides, about 10 to 100 cytosine nucleotides, about 20 to 70 cytosine nucleotides, about 20 to 60 cytosine nucleotides, or about 10 to 40 cytosine nucleotides).
  • the poly-C tail may be added to the poly-A tail or may substitute the poly-A tail.
  • mRNAs include a 5’ and/or 3’ untranslated region.
  • a 5’ untranslated region includes one or more elements that affect an mRNA’s stability or translation, for example, an iron responsive element. In some embodiments, a 5’ untranslated region may be between about 50 and 500 nucleotides in length. [0199] In some embodiments, a 3’ untranslated region includes one or more of a polyadenylation signal, a binding site for proteins that affect an mRNA’s stability of location in a cell, or one or more binding sites for miRNAs. In some embodiments, a 3’ untranslated region may be between 50 and 500 nucleotides in length or longer.
  • mRNAs include a 5’ cap structure.
  • a 5’ cap is typically added as follows: first, an RNA terminal phosphatase removes one of the terminal phosphate groups from the 5’ nucleotide, leaving two terminal phosphates; guanosine triphosphate (GTP) is then added to the terminal phosphates via a guanylyl transferase, producing a 5’5’5 triphosphate linkage; and the 7- nitrogen of guanine is then methylated by a methyltransferase.
  • GTP guanosine triphosphate
  • cap structures include, but are not limited to, m7G(5')ppp (5'(A,G(5')ppp(5')A and G(5')ppp(5')G. [0201] Naturally occurring cap structures comprise a 7-methyl guanosine that is linked via a
  • the cap is added enzymatically.
  • the cap is added in the nucleus and is catalyzed by the enzyme guanylyl transferase.
  • the addition of the cap to the 5' terminal end of RNA occurs immediately after initiation of transcription.
  • the terminal nucleoside is typically a guanosine, and is in the reverse orientation to all the other nucleotides, i.e., G(5')ppp(5')GpNpNp.
  • a common cap for mRNA produced by in vitro transcription is m 7 G(5')ppp(5')G, which has been used as the dinucleotide cap in transcription with T7 or SP6 RNA polymerase in vitro to obtain RNAs having a cap structure in their 5'-termini.
  • the prevailing method for the in vitro synthesis of caPPEd mRNA employs a pre-formed dinucleotide of the form m 7 G(5')ppp(5')G (“m 7 GpppG”) as an initiator of transcription.
  • a usual form of a synthetic dinucleotide cap used in in vitro translation experiments is the Anti-Reverse Cap Analog (“ARCA”) or modified ARCA, which is generally a modified cap analog in which the 2' or 3' OH group is replaced with -OCH3.
  • Additional cap analogs include, but are not limited to, a chemical structures selected from the group consisting of m 7 GpppG, m 7 GpppA, m 7 GpppC; unmethylated cap analogs (e.g., GpppG);
  • dimethylated cap analog e.g., m 2,7 GpppG
  • trimethylated cap analog e.g., m 2,2,7 GpppG
  • dimethylated symmetrical cap analogs e.g., m 7 Gpppm 7 G
  • anti reverse cap analogs e.g., ARCA; m 7 , 2'Ome GpppG, m 72'd GpppG, m 7,3'Ome GpppG, m 7,3'd GpppG and their tetraphosphate derivatives
  • a suitable cap is a 7-methyl guanylate (“m 7 G”) linked via a triphosphate bridge to the 5'-end of the first transcribed nucleotide, resulting in m 7 G(5')ppp(5')N, where N is any nucleoside.
  • a preferred embodiment of a m 7 G cap utilized in embodiments of the invention is m 7 G(5')ppp(5')G.
  • the cap is a Cap0 structure. Cap0 structures lack a 2'-O-methyl residue of the ribose attached to bases 1 and 2.
  • the cap is a Cap1 structure.
  • Cap1 structures have a 2'-O-methyl residue at base 2.
  • the cap is a Cap2 structure.
  • Cap2 structures have a 2'-O-methyl residue attached to both bases 2 and 3.
  • a variety of m 7 G cap analogs are known in the art, many of which are commercially available.
  • cap analogs for use in embodiments of the invention include N7-benzylated dinucleoside tetraphosphate analogs (described in Grudzien, E. et al., RNA, 10: 1479-1487 (2004)), phosphorothioate cap analogs (described in Grudzien-Nogalska, E., et al., RNA, 13: 1745-1755 (2007)), and cap analogs (including biotinylated cap analogs) described in U.S. Patent Nos.8,093,367 and 8,304,529, incorporated by reference herein.
  • a“tail” serves to protect the mRNA from exonuclease degradation.
  • the poly A tail is thought to stabilize natural messengers and synthetic sense RNA. Therefore, in certain embodiments a long poly A tail can be added to an mRNA molecule thus rendering the RNA more stable.
  • Poly A tails can be added using a variety of art-recognized techniques. For example, long poly A tails can be added to synthetic or in vitro transcribed RNA using poly A polymerase (Yokoe, et al. Nature Biotechnology.1996; 14: 1252-1256).
  • a transcription vector can also encode long poly A tails.
  • poly A tails can be added by transcription directly from PCR products.
  • Poly A may also be ligated to the 3' end of a sense RNA with RNA ligase (see, e.g., Molecular Cloning A Laboratory Manual, 2nd Ed., ed. by Sambrook, Fritsch and Maniatis (Cold Spring Harbor
  • mRNAs include a 3’ poly(A) tail structure.
  • the length of the poly A tail can be at least about 10, 50, 100, 200, 300, 400 at least 500 nucleotides.
  • a poly-A tail on the 3' terminus of mRNA typically includes about 10 to 300 adenosine nucleotides (e.g., about 10 to 200 adenosine nucleotides, about 10 to 150 adenosine nucleotides, about 10 to 100 adenosine nucleotides, about 20 to 70 adenosine nucleotides, or about 20 to 60 adenosine nucleotides).
  • mRNAs include a 3’ poly(C) tail structure.
  • a suitable poly-C tail on the 3' terminus of mRNA typically include about 10 to 200 cytosine nucleotides (e.g., about 10 to 150 cytosine nucleotides, about 10 to 100 cytosine nucleotides, about 20 to 70 cytosine nucleotides, about 20 to 60 cytosine nucleotides, or about 10 to 40 cytosine nucleotides).
  • the poly- C tail may be added to the poly-A tail or may substitute the poly-A tail.
  • the length of the poly A or poly C tail is adjusted to control the stability of a modified sense mRNA molecule of the invention and, thus, the transcription of protein.
  • the length of the poly A tail can influence the half-life of a sense mRNA molecule, the length of the poly A tail can be adjusted to modify the level of resistance of the mRNA to nucleases and thereby control the time course of polynucleotide expression and/or polypeptide production in a target cell. 5’ and 3’ Untranslated Region
  • mRNAs include a 5’ and/or 3’ untranslated region.
  • a 5’ untranslated region includes one or more elements that affect an mRNA’s stability or translation, for example, an iron responsive element. In some embodiments, a 5’ untranslated region may be between about 50 and 500 nucleotides in length.
  • a 3’ untranslated region includes one or more of a polyadenylation signal, a binding site for proteins that affect an mRNA’s stability of location in a cell, or one or more binding sites for miRNAs. In some embodiments, a 3’ untranslated region may be between 50 and 500 nucleotides in length or longer.
  • Exemplary 3' and/or 5' UTR sequences can be derived from mRNA molecules which are stable (e.g., globin, actin, GAPDH, tubulin, histone, or citric acid cycle enzymes) to increase the stability of the sense mRNA molecule.
  • a 5’ UTR sequence may include a partial sequence of a CMV immediate-early 1 (IE1) gene, or a fragment thereof to improve the nuclease resistance and/or improve the half-life of the polynucleotide.
  • IE1 immediate-early 1
  • hGH human growth hormone
  • modifications improve the stability and/or pharmacokinetic properties (e.g., half-life) of the polynucleotide relative to their unmodified counterparts, and include, for example modifications made to improve such polynucleotides’ resistance to in vivo nuclease digestion.
  • cationic lipids described herein described e.g., a cationic lipid of any of Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III- B), (IV-A), (IV-B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)-(156)), as well as pharmaceutical and liposomal compositions comprising such lipids, can be used in formulations to facilitate the delivery of encapsulated materials (e.g., one or more polynucleotides such as mRNA) to, and subsequent transfection of one or more target cells.
  • encapsulated materials e.g., one or more polynucleotides such as mRNA
  • cationic lipids described herein are characterized as resulting in one or more of receptor-mediated endocytosis, clathrin- mediated and caveolae-mediated endocytosis, phagocytosis and macropinocytosis, fusogenicity, endosomal or lysosomal disruption and/or releasable properties that afford such compounds advantages relative other similarly classified lipids.
  • a nucleic acid e.g., mRNA encoding a protein (e.g., a full length, fragment or portion of a protein) as described herein may be delivered via a delivery vehicle comprising a cationic lipid as described (e.g., a cationic lipid of any of Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III-B), (IV-A), (IV-B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)-(156)).
  • a delivery vehicle comprising a cationic lipid as described (e.g., a cationic lipid of any of Formulas (I)-(VI) such as any of Formulas (I
  • the present invention provides a composition (e.g., a pharmaceutical composition) comprising a cationic lipid described herein (e.g., a cationic lipid of any of Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III-B), (IV-A), (IV-B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)-(156)) and one or more polynucleotides.
  • a composition e.g., a pharmaceutical composition
  • a cationic lipid described herein e.g., a cationic lipid of any of Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I
  • a composition may further comprise one or more cationic lipids, one or more non-cationic lipids, one or more cholesterol-based lipids and/or one or more PEG-modified lipids.
  • a composition exhibits an enhanced (e.g., increased) ability to transfect one or more target cells. Accordingly, also provided herein are methods of transfecting one or more target cells.
  • Such methods generally comprise the step of contacting the one or more target cells with the cationic lipids and/or pharmaceutical compositions disclosed herein (e.g., a liposomal formulation comprising a cationic lipid described herein (e.g., a cationic lipid of any of Formulas (I)- (VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III-B), (IV-A), (IV- B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)-(156)) encapsulating one or more polynucleotides) such that the one or more target cells are transfected with the materials encapsulated therein (e.g., one or more polynucleotides).
  • transfect or “transfection” refer to the intracellular introduction of one or more encapsulated materials (e.g., nucleic acids and/or polynucleotides) into a cell, or preferably into a target cell.
  • the introduced polynucleotide may be stably or transiently maintained in the target cell.
  • transfection efficiency refers to the relative amount of such encapsulated material (e.g., polynucleotides) up- taken by, introduced into and/or expressed by the target cell which is subject to transfection. In practice, transfection efficiency may be estimated by the amount of a reporter polynucleotide product produced by the target cells following transfection.
  • the compounds and pharmaceutical compositions described herein demonstrate high transfection efficiencies thereby improving the likelihood that appropriate dosages of the encapsulated materials (e.g., one or more polynucleotides) will be delivered to the site of pathology and subsequently expressed, while at the same time minimizing potential systemic adverse effects or toxicity associated with the compound or their encapsulated contents.
  • the polynucleotides e.g., one or more polynucleotides
  • the production of the product (e.g., a polypeptide or protein) encoded by such polynucleotide may be preferably stimulated and the capability of such target cells to express the polynucleotide and produce, for example, a polypeptide or protein of interest is enhanced.
  • transfection of a target cell by one or more compounds or pharmaceutical compositions encapsulating mRNA will enhance (i.e., increase) the production of the protein or enzyme encoded by such mRNA.
  • delivery vehicles described herein may be prepared to preferentially distribute to other target tissues, cells or organs, such as the heart, lungs, kidneys, spleen.
  • the lipid nanoparticles of the present invention may be prepared to achieve enhanced delivery to the target cells and tissues.
  • polynucleotides e.g., mRNA
  • encapsulated in one or more of the compounds or pharmaceutical and liposomal compositions described herein can be delivered to and/or transfect targeted cells or tissues.
  • the encapsulated polynucleotides are capable of being expressed and functional polypeptide products produced (and in some instances excreted) by the target cell, thereby conferring a beneficial property to, for example the target cells or tissues.
  • encapsulated polynucleotides may encode, for example, a hormone, enzyme, receptor, polypeptide, peptide or other protein of interest.
  • a composition is a suitable delivery vehicle.
  • a suitable delivery vehicle In embodiments, a
  • composition is a liposomal delivery vehicle, e.g., a lipid nanoparticle.
  • liposomal delivery vehicle e.g., a lipid nanoparticle.
  • Enriching liposomal compositions with one or more of the cationic lipids disclosed herein may be used as a means of improving (e.g., reducing) the toxicity or otherwise conferring one or more desired properties to such enriched liposomal composition (e.g., improved delivery of the encapsulated polynucleotides to one or more target cells and/or reduced in vivo toxicity of a liposomal composition).
  • compositions and in particular liposomal compositions, that comprise one or more of the cationic lipids disclosed herein.
  • the compounds described herein e.g., a cationic lipid of any of Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III- B), (IV-A), (IV-B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)-(156)) are cationic lipids that may be used as a component of a liposomal composition to facilitate or enhance the delivery and release of encapsulated materials (e.g., one or more therapeutic agents) to one or more target cells (e.g
  • liposomal delivery vehicles e.g., lipid nanoparticles
  • lipid nanoparticles are usually characterized as microscopic vesicles having an interior aqua space sequestered from an outer medium by a membrane of one or more bilayers.
  • Bilayer membranes of liposomes are typically formed by amphiphilic molecules, such as lipids of synthetic or natural origin that comprise spatially separated hydrophilic and hydrophobic domains (Lasic, Trends Biotechnol., 16: 307-321, 1998).
  • Bilayer membranes of the liposomes can also be formed by amphophilic polymers and surfactants (e.g., polymerosomes, niosomes, etc.).
  • a liposomal delivery vehicle typically serves to transport a desired mRNA to a target cell or tissue.
  • compositions e.g., liposomal compositions
  • a composition (e.g., a pharmaceutical composition) comprises an mRNA
  • a liposome comprises one or more cationic lipids, one or more non-cationic lipids, one or more cholesterol-based lipids and one or more PEG-modified lipids, and at least one cationic lipid is a cationic lipid as described herein (e.g., a cationic lipid of any of Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III-B), (IV-A), (IV-B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)-(156)).
  • a cationic lipid of any of Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’
  • a composition comprises an mRNA encoding for a protein (e.g., any protein described herein).
  • a composition comprises an mRNA encoding for cystic fibrosis transmembrane conductance regulator (CFTR) protein.
  • CFTR cystic fibrosis transmembrane conductance regulator
  • a composition comprises an mRNA encoding for ornithine transcarbamylase (OTC) protein.
  • an mRNA encodes for an antigen (e.g., an antigen from an infectious agent.
  • a composition (e.g., a pharmaceutical composition) comprises a nucleic acid encapsulated within a liposome, wherein the liposome comprises any cationic lipid (e.g., a cationic lipid of any of Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II- B), (III-A), (III-B), (IV-A), (IV-B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)- (156)) as described herein.
  • a nucleic acid is an mRNA encoding a peptide or polypeptide.
  • an mRNA encodes a peptide or polypeptide for use in the delivery to or treatment of the lung of a subject or a lung cell (e.g., an mRNA encodes cystic fibrosis transmembrane conductance regulator (CFTR) protein).
  • an mRNA encodes a peptide or polypeptide for use in the delivery to or treatment of the liver of a subject or a liver cell (e.g., an mRNA encodes ornithine transcarbamylase (OTC) protein).
  • OTC ornithine transcarbamylase
  • a liposomal delivery vehicle e.g., a lipid nanoparticle
  • a liposomal delivery vehicle e.g., a lipid nanoparticle
  • a liposomal delivery vehicle can have a net negative charge.
  • a liposomal delivery vehicle e.g., a lipid nanoparticle
  • a net neutral charge e.g., a lipid nanoparticle
  • a lipid nanoparticle that encapsulates a nucleic acid comprises one or more cationic lipids described herein (e.g., a cationic lipid of any of Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III-B), (IV-A), (IV-B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)-(156)).
  • a cationic lipids described herein such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (I-A), (I-B), (III-A), (III-B), (IV-A), (IV
  • wt% percentage of the combined dry weight of all lipids of a composition
  • a cationic lipid of any of Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III-B), (IV-A), (IV-B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV- C”), or any of Compounds (1)-(156)) is present in an amount that is about 0.5 wt% to about 30 wt% (e.g., about 0.5 wt% to about 20 wt%) of the combined dry weight of all lipids present in a composition (e.g., a liposomal composition).
  • a composition e.g., a liposomal composition
  • a cationic lipid as described herein e.g., a cationic lipid of any of Formulas (I)- (VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III-B), (IV-A), (IV- B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)-(156)) is present in an amount that is about 1 wt% to about 30 wt%, about 1 wt% to about 20 wt%, about 1 wt% to about 15 wt%, about 1 wt% to about 10 wt%, or about 5 wt% to about 25 wt% of the combined dry weight of all lipids present in a composition (e.
  • a cationic lipid as described herein e.g., a cationic lipid of any of Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III-B), (IV-A), (IV-B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV- C”), or any of Compounds (1)-(156)) is present in an amount that is about 0.5 wt% to about 5 wt%, about 1 wt% to about 10 wt%, about 5 wt% to about 20 wt%, or about 10 wt% to about 20 wt% of the combined molar amounts of all lipids present in a composition such as a liposomal delivery vehicle.
  • the amount of a cationic lipid as described herein is present in an amount that is at least about 5 wt%, about 10 wt%, about 15 wt%, about 20 wt%, about 25 wt%, about 30 wt%, about 35 wt%, about 40 wt%, about 45 wt%, about 50 wt%, about 55 wt%, about 60 wt%
  • the amount of a cationic lipid as described herein is present in an amount that is no more than about 5 wt%, about 10 wt%, about 15 wt%, about 20 wt%, about 25 wt%, about 30 wt%, about 35 wt%, about 40 wt%, about 45 wt%, about 50 wt%, about 55 wt%, about 60 wt
  • a composition e.g., a liposomal delivery vehicle such as a lipid nanoparticle
  • a cationic lipid described herein e.g., a cationic lipid of any of Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III-B), (IV-A), (IV-B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV- C”), or any of Compounds (1)-(156)).
  • a cationic lipid described herein e.g., a cationic lipid of any of Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I
  • a delivery vehicle (e.g., a liposomal delivery vehicle such as a lipid nanoparticle) comprises about 0.5 wt%, about 1 wt%, about 3 wt%, about 5 wt%, or about 10 wt% a cationic lipid described herein (e.g., a cationic lipid of any of Formulas (I)- (VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III-B), (IV-A), (IV- B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)-(156)).
  • a cationic lipid described herein such as any of Formulas (I-A)-(I-C), (I-A’)-(I
  • a delivery vehicle (e.g., a liposomal delivery vehicle such as a lipid nanoparticle) comprises up to about 0.5 wt%, about 1 wt%, about 3 wt%, about 5 wt%, about 10 wt%, about 15 wt%, or about 20 wt% of a cationic lipid described herein (e.g., a cationic lipid of any of Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III-B), (IV-A), (IV-B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)-(156)).
  • a cationic lipid described herein e.g., a
  • the percentage results in an improved beneficial effect (e.g., improved delivery to targeted tissues such as the liver or the lung).
  • a cationic lipid as described herein e.g., a cationic lipid of any of Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III-B), (IV-A), (IV-B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)-(156)) is present in an amount that is about 0.5 mol% to about30 mol% (e.g., about 0.5 mol% to about20 mol%) of the combined molar amounts of all lipids present in a composition such as a liposomal delivery vehicle.
  • a cationic lipid as described herein e.g., a cationic lipid of any of Formulas (I)- (VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III-B), (IV-A), (IV- B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)-(156)) is present in an amount that is about 0.5 mol% to about 5 mol%, about 1 mol% to about 10 mol%, about 5 mol% to about 20 mol%, or about 10 mol% to about 20 mol% of the combined molar amounts of all lipids present in a composition such as a liposomal delivery vehicle.
  • a cationic lipid as described herein e.g., a cationic lipid of any of Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III-B), (IV-A), (IV-B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)-(156)) is present in an amount that is about 1 mol% to about 30 mol%, about 1 mol% to about 20 mol%, about 1 mol% to about 15 mol%, about 1 mol% to about 10 mol%, or about 5 mol% to about 25 mol% of the combined dry weight of all lipids present in a composition such as a liposomal delivery vehicle [0243] In certain embodiment
  • Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III- B), (IV-A), (IV-B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)-(156)) can comprise from about 0.1 mol% to about 50 mol%, or from 0.5 mol% to about 50 mol%, or from about 1 mol% to about 25 mol%, or from about 1 mol% to about 10 mol% of the total amount of lipids in a composition (e.g., a liposomal delivery vehicle).
  • a cationic lipid as described herein e.g., a cationic lipid of any of
  • Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III- B), (IV-A), (IV-B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)-(156)) can comprise greater than about 0.1 mol%, or greater than about 0.5 mol%, or greater than about 1 mol%, or greater than about 5 mol% of the total amount of lipids in the lipid nanoparticle.
  • a cationic lipid as described herein e.g., a cationic lipid of any of Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III- B), (IV-A), (IV-B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)-(156)) can comprise less than about 25 mol%, or less than about 10 mol%, or less than about 5 mol%, or less than about 1 mol% of the total amount of lipids in a composition (e.g., a liposomal delivery vehicle).
  • a composition e.g., a liposomal delivery vehicle
  • the amount of a cationic lipid as described herein is present in an amount that is at least about 5 mol%, about 10 mol%, about 15 mol%, about 20 mol%, about 25 mol%, about 30 mol%, about 35 mol%, about 40 mol%, about 45 mol%, about 50 mol%, about 55 mol%, about 60 mol%, about 65 mol%, about 70 mol%
  • the amount of a cationic lipid as described herein is present in an amount that is no more than about 5 mol%, about 10 mol%, about 15 mol%, about 20 mol%, about 25 mol%, about 30 mol%, about 35 mol%, about 40 mol%, about 45 mol%, about 50 mol%, about 55 mol%, about 60 mol%, about 65 mol%, about 70 mol
  • a composition further comprises one more lipids (e.g., one more lipids
  • such pharmaceutical (e.g., liposomal) compositions comprise one or more of a PEG-modified lipid, a non-cationic lipid and a cholesterol lipid.
  • such pharmaceutical (e.g., liposomal) compositions comprise: one or more PEG-modified lipids; one or more non-cationic lipids; and one or more cholesterol lipids.
  • such pharmaceutical (e.g., liposomal) compositions comprise: one or more PEG-modified lipids and one or more cholesterol lipids.
  • a composition that encapsulates a nucleic acid (e.g., mRNA encoding a peptide or polypeptide) comprises one or more cationic lipids as described herein (e.g., a cationic lipid of any of Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III-B), (IV-A), (IV-B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)-(156)) and one or more lipids selected from the group consisting of a cationic lipid, a non-cationic lipid, and a PEGylated lipid.
  • a composition that encapsulates a nucleic acid (e.g., mRNA encoding a peptide or polypeptide) comprises one or more cationic lipids as described herein (e.g., a cationic lipid of any of Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III-B), (IV-A), (IV-B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)-(156)); one or more lipids selected from the group consisting of a cationic lipid, a non-cationic lipid, and a PEGylated lipid; and further comprises
  • a lipid nanoparticle that encapsulates a nucleic acid comprises one or more cationic lipids as described herein (e.g., a cationic lipid of any of Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II- B), (III-A), (III-B), (IV-A), (IV-B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)- (156)), as well as one or more lipids selected from the group consisting of a cationic lipid, a non- cationic lipid, a PEGylated lipid, and a cholesterol-based lipid.
  • a cationic lipids as described herein
  • a lipid nanoparticle comprises one or more cationic lipids described herein (e.g., a cationic lipid of any of Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III-B), (IV-A), (IV-B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)-(156)), a non-cationic lipid (e.g., DOPE), a cationic lipid of any of Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (I-A), (I-B), (III-A),
  • the selection of cationic lipids, non-cationic lipids and/or PEG-modified lipids which comprise the lipid nanoparticle, as well as the relative molar ratio of such lipids to each other, is based upon the characteristics of the selected lipid(s), the nature of the intended target cells, the characteristics of the mRNA to be delivered. Additional considerations include, for example, the saturation of the alkyl chain, as well as the size, charge, pH, pKa, fusogenicity and toxicity of the selected lipid(s). Thus, the molar ratios may be adjusted accordingly. Further Cationic Lipids
  • cationic lipids as described herein e.g., a cationic lipid of any of
  • a composition may comprise one or more further cationic lipids.
  • liposomes may comprise one or more further cationic lipids.
  • the phrase“cationic lipid” refers to any of a number of lipid species that have a net positive charge at a selected pH, such as physiological pH. Several cationic lipids have been described in the literature, many of which are commercially available. [0258] In some embodiments, liposomes may comprise one or more additional cationic lipids. As used herein, the phrase“cationic lipid” refers to any of a number of lipid species that have a net positive charge at a selected pH, such as physiological pH. Several cationic lipids have been described in the literature, many of which are commercially available.
  • Suitable additional cationic lipids for use in the compositions include the cationic lipids as described in International Patent Publication WO 2010/144740, which is incorporated herein by reference.
  • the compositions include a cationic lipid, (6Z,9Z,28Z,31Z)- heptatriaconta-6,9,28,31-tetraen-19-yl 4-(dimethylamino) butanoate, having a compound structure of:
  • compositions include ionizable cationic lipids as described in International Patent Publication WO 2013/149140, which is incorporated herein by reference.
  • compositions include a cationic lipid of one of the following formulas:
  • R1 and R2 are each independently selected from the group consisting of hydrogen, an optionally substituted, variably saturated or unsaturated C 1 -C 20 alkyl and an optionally substituted, variably saturated or unsaturated C 6 -C 20 acyl; wherein L 1 and L 2 are each independently selected from the group consisting of hydrogen, an optionally substituted C 1 -C 30 alkyl, an optionally substituted variably unsaturated C 1 -C 30 alkenyl, and an optionally substituted C1-C30 alkynyl; wherein m and o are each independently selected from the group consisting of zero and any positive integer (e.g., where m is three); and wherein n is zero or any positive integer (e.g., where n is one).
  • compositions include the cationic lipid (15Z, 18Z)-N,N-dimethyl-6-(9Z,12Z)-octadeca-9,12-dien-l -yl) tetracosa- 15,18-dien-1- amine (“HGT5000”), having a compound structure of:
  • compositions include the cationic lipid (15Z, 18Z)-N,N-dimethyl-6-((9Z,12Z)-octadeca-9,12-dien-1-yl) tetracosa-4,15,18- trien-l -amine (“HGT5001”), having a compound structure of:
  • the include the cationic lipid and (15Z,18Z)-N,N-dimethyl-6-((9Z,12Z)-octadeca-9,12-dien-1-yl) tetracosa-5,15,18-trien- 1 - amine (“HGT5002”), having a compound structure of:
  • compositions include cationic lipids
  • compositions include a cationic lipid having a compound structure of:
  • compositions include the cationic lipids as described in International Patent Publication WO 2016/118725, which is incorporated herein by reference.
  • compositions include a cationic lipid having a compound structure of:
  • compositions include the cationic lipids as described in International Patent Publication WO 2016/118724, which is incorporated herein by reference.
  • the compositions include a cationic lipid having a compound structure of:
  • compositions include a cationic lipid having the formula of 14,25-ditridecyl 15,18,21,24-tetraaza-octatriacontane, and pharmaceutically acceptable salts thereof.
  • additional cationic lipids for use in the compositions include the cationic lipids as described in International Patent Publications WO 2013/063468 and WO 2016/205691, each of which are incorporated herein by reference.
  • the compositions include a cationic lipid of the following formula:
  • compositions include a cationic lipid having a compound structure of:
  • compositions include a cationic lipid having a compound structure of:
  • compositions include a cationic lipid having a compound structure of:
  • compositions include a cationic lipid having a compound structure of:
  • compositions include the cationic lipids as described in International Patent Publication WO 2015/184256, which is incorporated herein by reference.
  • compositions include a cationic lipid of the following formula:
  • the compositions include
  • compositions include the cationic lipids as described in International Patent Publication WO 2016/004202, which is incorporated herein by reference.
  • compositions include a cationic lipid having the compound structure:
  • compositions include a cationic lipid having the compound structure:
  • compositions include a cationic lipid having the compound structure:
  • Suitable additional cationic lipids for use in the compositions include the cationic lipids as described in J. McClellan, M. C. King, Cell 2010, 141, 210-217 and in Whitehead et al., Nature Communications (2014) 5:4277, which is incorporated herein by reference.
  • cationic lipids as described in J. McClellan, M. C. King, Cell 2010, 141, 210-217 and in Whitehead et al., Nature Communications (2014) 5:4277, which is incorporated herein by reference.
  • the cationic lipids of the compositions include a cationic lipid having a compound structure of:
  • compositions include the cationic lipids as described in International Patent Publication WO 2015/199952, which is incorporated herein by reference.
  • compositions include a cationic lipid having the compound structure:
  • compositions include a cationic lipid having the compound structure:
  • compositions include a cationic lipid having the compound structure:
  • compositions include a cationic lipid having the compound structure:
  • compositions include a cationic lipid having the compound structure:
  • compositions include a cationic lipid having the compound structure:
  • compositions include a cationic lipid having the compound structure:
  • compositions include a cationic lipid having the compound structure:
  • compositions include a cationic lipid having the compound structure:
  • compositions include a cationic lipid having the compound structure:
  • compositions include a cationic lipid having the compound structure:
  • compositions include a cationic lipid having the compound structure:
  • compositions include a cationic lipid having the compound
  • compositions include a cationic lipid having the compound structure:
  • compositions include a cationic lipid having the compound structure:
  • compositions include a cationic lipid having the compound
  • compositions include a cationic lipid having the compound
  • compositions include a cationic lipid having the compound structure:
  • compositions include a cationic lipid having the compound
  • compositions include a cationic lipid having the compound structure:
  • compositions include a cationic lipid having the compound structure:
  • compositions include a cationic lipid having the compound structure:
  • compositions include a cationic lipid having the compound structure:
  • compositions include a cationic lipid having the compound
  • compositions include a cationic lipid having the compound structure:
  • compositions include a cationic lipid having the compound structure:
  • compositions include a cationic lipid having the compound
  • compositions include a cationic lipid having the compound structure:
  • compositions include a cationic lipid having the compound
  • compositions include a cationic lipid having the compound
  • compositions include the cationic lipids as described in International Patent Publication WO 2017/075531, which is incorporated herein by reference.
  • compositions include a cationic lipid of the following formula:
  • compositions include the cationic lipids as described in International Patent Publication WO 2017/117528, which is incorporated herein by reference.
  • compositions include a cationic lipid having the compound structure:
  • compositions include a cationic lipid having the compound structure:
  • compositions include a cationic lipid having the compound structure:
  • Suitable additional cationic lipids for use in the compositions include the cationic lipids as described in International Patent Publication WO 2017/049245, which is incorporated herein by reference.
  • the cationic lipids of the compositions and methods of the present invention include a compound of one of the following formulas:
  • R4 is independently selected from -(CH 2 ) n Q and -(CH 2 ) n CHQR;
  • Q is selected from the group consisting of - OR, -OH, -O(CH 2 ) n N(R) 2 , -OC(O)R, -CX 3 , -CN, -N(R)C(O)R, -N(H)C(O)R, -N(R)S(O) 2 R, -N(H)S(O) 2 R, - N(R)C(O)N(R) 2 , -N(H)C(O)N(R) 2 , -N(H)C(O)N(R), -N(R)C(S)N(R) 2 , -N(H)C(S)N(R) 2 , -N(H)C(S)N(H)(R), and a
  • compositions include a cationic lipid having a compound structure of:
  • compositions include a cationic lipid having a compound structure of:
  • compositions include a cationic lipid having a compound structure of:
  • compositions include a cationic lipid having a compound structure of:
  • compositions include a cationic lipid having a compound structure of:
  • compositions include a cationic lipid having a compound structure of:
  • compositions include a cationic lipid having a compound structure of:
  • compositions include cholesterol-based cationic lipids.
  • compositions include imidazole cholesterol ester or “ICE”, having a compound structure of:
  • compositions include cleavable cationic lipids as described in International Patent Publication WO 2012/170889, which is incorporated herein by reference.
  • compositions include a cationic lipid of the following formula: ,
  • R 1 is selected from the group consisting of imidazole, guanidinium, amino, imine, enamine, an optionally-substituted alkyl amino (e.g., an alkyl amino such as dimethylamino) and pyridyl; wherein R2 is selected from the group consisting of one of the following two formulas:
  • R 3 and R 4 are each independently selected from the group consisting of an optionally substituted, variably saturated or unsaturated C 6 -C 20 alkyl and an optionally substituted, variably saturated or unsaturated C 6 -C 20 acyl; and wherein n is zero or any positive integer (e.g., one, two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, fourteen, fifteen, sixteen, seventeen, eighteen, nineteen, twenty or more).
  • compositions include a cationic lipid,“HGT4001”, having a
  • compositions include a cationic lipid,“HGT4002”, having a
  • compositions include a cationic lipid,“HGT4003”, having a
  • compositions include a cationic lipid,“HGT4004”, having a compound structure of: and pharmaceutically acceptable salts thereof.
  • compositions include a cationic lipid“HGT4005”, having a
  • the compositions include the cationic lipid, N-[l-(2,3-dioleyloxy)propyl]- N,N,N-trimethylammonium chloride (“DOTMA”).
  • DOTMA N-[l-(2,3-dioleyloxy)propyl]- N,N,N-trimethylammonium chloride
  • DOTMA can be formulated alone or can be combined with a neutral lipid (e.g., dioleoylphosphatidyl-ethanolamine or“DOPE”) or still other cationic or non-cationic lipids into a liposomal transfer vehicle or a lipid nanoparticle, and such liposomes can be used to enhance the delivery of nucleic acids into target cells.
  • a neutral lipid e.g., dioleoylphosphatidyl-ethanolamine or“DOPE”
  • DOPE dioleoylphosphatidyl-ethanolamine
  • cationic lipids suitable for the compositions include, for example, 5- carboxyspermylglycinedioctadecylamide (“DOGS”); 2,3-dioleyloxy-N-[2(spermine- carboxamido)ethyl]-N,N-dimethyl-l-propanaminium (“DOSPA”) (Behr et al. Proc. Nat.'l Acad. Sci.86, 6982 (1989), U.S. Pat. No.5,171,678; U.S. Pat.
  • DOGS 5- carboxyspermylglycinedioctadecylamide
  • DOSPA 2,3-dioleyloxy-N-[2(spermine- carboxamido)ethyl]-N,N-dimethyl-l-propanaminium
  • Additional exemplary cationic lipids suitable for the compositions also include: l,2-distearyloxy- N,N-dimethyl-3-aminopropane (“DSDMA”); 1,2-dioleyloxy-N,N-dimethyl-3-aminopropane
  • DODMA 1 ,2-dilinoleyloxy-N,N-dimethyl-3-aminopropane
  • DLenDMA l,2-dilinolenyloxy-N,N- dimethyl-3-aminopropane
  • DODAC N-dioleyl-N,N-dimethylammonium chloride
  • DDAB N,N-distearyl-N,N-dimethylarnrnonium bromide
  • DMRIE N-(l,2-dimyristyloxyprop-3-yl)-N,N- dimethyl-N-hydroxyethyl ammonium bromide
  • DMRIE 3-dimethylamino-2-(cholest-5-en-3-beta- oxybutan-4-oxy)-l-(cis,cis-9,12-octadecadienoxy)propane
  • CLinDMA 2-[5'-(cholest-5-en-3-beta
  • one or more of the cationic lipids comprise at least one of an imidazole, dialkylamino, or guanidinium moiety.
  • one or more cationic lipids suitable for the compositions include 2,2- Dilinoley1-4-dimethylaminoethy1-[1,3]-dioxolane (“XTC”); (3aR,5s,6aS)-N,N-dimethyl-2,2- di((9Z,12Z)-octadeca-9,12-dienyl)tetrahydro-3aH-cyclopenta[d] [1 ,3]dioxol-5-amine (“ALNY-100”) and/or 4,7,13-tris(3-oxo-3-(undecylamino)propyl)-N1,N16-diundecyl-4,7,10,13-tetraazahexadecane- 1,16-diamide (“NC98-5”).
  • XTC 2,2- Dilinoley1-4-dimethylaminoethy1-[1,3]-dioxolane
  • XTC 2,2- Dil
  • the percentage of total cationic lipids in a composition may be no more than 10%, no more than 20%, no more than 30%, no more than 40%, no more than 50%, no more than 60%, no more than 70%, no more than 80%, no more than 90%, or no more than 95% of total lipids as measured by molar ratios (mol%) or by weight (wt%).
  • the percentage of total cationic lipids in a composition may be greater than 10%, greater than 20%, greater than 30%, greater than 40%, greater than 50%, greater than 60%, greater than 70%, greater than 80%, greater than 90%, or greater than 95% of total lipids as measured by molar ratios (mol%) or by weight (wt%).
  • total cationic lipid(s) constitute(s) about 30-50 % (e.g., about 30-45%, about 30-40%, about 35-50%, about 35-45%, or about 35-40%) of the liposome by weight.
  • the cationic lipid constitutes about 30%, about 35%, about 40 %, about 45%, or about 50% of a composition (e.g., a liposomal composition) by molar ratio.
  • total cationic lipid(s) constitute(s) about 30-50 % (e.g., about 30-45%, about 30-40%, about 35-50%, about 35-45%, or about 35-40%) of the liposome by weight.
  • the cationic lipid constitutes about 30%, about 35%, about 40 %, about 45%, or about 50% of a composition (e.g., a liposomal composition) by weight.
  • the compositions include one or more cationic lipids that constitute at least about 5%, 10%, 20%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, or 70%, measured by weight, of the total lipid content in the composition, e.g., a lipid nanoparticle.
  • the compositions include one or more cationic lipids that constitute at least about 5%, 10%, 20%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, or 70%, measured as a mol%, of the total lipid content in the composition, e.g., a lipid nanoparticle.
  • the compositions include one or more cationic lipids that constitute about 30-70 % (e.g., about 30-65%, about 30-60%, about 30- 55%, about 30-50%, about 30-45%, about 30-40%, about 35-50%, about 35-45%, or about 35-40%), measured by weight, of the total lipid content in the composition, e.g., a lipid nanoparticle.
  • the compositions include one or more cationic lipids that constitute about 30-70 % (e.g., about 30-65%, about 30-60%, about 30-55%, about 30-50%, about 30-45%, about 30-40%, about 35-50%, about 35-45%, or about 35-40%), measured as mol %, of the total lipid content in the composition, e.g., a lipid nanoparticle.
  • compositions may also comprise one or more non-cationic molecules
  • non-cationic lipid refers to any neutral, zwitterionic or anionic lipid.
  • anionic lipid refers to any of a number of lipid species that carry a net negative charge at a selected pH, such as physiological pH.
  • Non-cationic lipids include, but are not limited to, distearoylphosphatidylcholine (DSPC), dioleoylphosphatidylcholine (DOPC), dipalmitoylphosphatidylcholine (DPPC), dioleoylphosphatidylglycerol (DOPG),
  • DPPG dipalmitoylphosphatidylglycerol
  • DOPE dioleoylphosphatidylethanolamine
  • palmitoyloleoylphosphatidylcholine POPC
  • palmitoyloleoyl-phosphatidylethanolamine POPE
  • dioleoyl-phosphatidylethanolamine 4-(N-maleimidomethyl)-cyclohexane-l-carboxylate DOPE-mal
  • dipalmitoyl phosphatidyl ethanolamine DPPE
  • dimyristoylphosphoethanolamine DMPE
  • distearoyl-phosphatidyl-ethanolamine DSPE
  • 16-O-monomethyl PE 16-O-dimethyl PE
  • 18-1-trans PE l-stearoyl-2-oleoyl-phosphatidyethanolamine
  • SOPE l-stearoyl-2-oleoyl-phosphatidyethanolamine
  • a non-cationic or helper lipid is dioleoylphosphatidylethanolamine (DOPE).
  • DOPE dioleoylphosphatidylethanolamine
  • a non-cationic lipid is a neutral lipid, i.e., a lipid that does not carry a net charge in the conditions under which the composition is formulated and/or administered.
  • a non-cationic lipid may be present in a molar ratio (mol%) of about 5% to about 90%, about 5% to about 70%, about 5% to about 50%, about 5% to about 40%, about 5% to about 30%, about 10 % to about 70%, about 10% to about 50%, or about 10% to about 40% of the total lipids present in a composition.
  • total non-cationic lipids may be present in a molar ratio (mol%) of about 5% to about 90%, about 5% to about 70%, about 5% to about 50%, about 5% to about 40%, about 5% to about 30%, about 10 % to about 70%, about 10% to about 50%, or about 10% to about 40% of the total lipids present in a composition.
  • the percentage of non-cationic lipid in a liposome may be greater than about 5 mol%, greater than about 10 mol%, greater than about 20 mol%, greater than about 30 mol%, or greater than about 40 mol%.
  • the percentage total non-cationic lipids in a liposome may be greater than about 5 mol%, greater than about 10 mol%, greater than about 20 mol%, greater than about 30 mol%, or greater than about 40 mol%. In some embodiments, the percentage of non-cationic lipid in a liposome is no more than about 5 mol%, no more than about 10 mol%, no more than about 20 mol%, no more than about 30 mol%, or no more than about 40 mol%.
  • the percentage total non-cationic lipids in a liposome may be no more than about 5 mol%, no more than about 10 mol%, no more than about 20 mol%, no more than about 30 mol%, or no more than about 40 mol%.
  • a non-cationic lipid may be present in a weight ratio (wt%) of about 5% to about 90%, about 5% to about 70%, about 5% to about 50%, about 5% to about 40%, about 5% to about 30%, about 10 % to about 70%, about 10% to about 50%, or about 10% to about 40% of the total lipids present in a composition.
  • total non-cationic lipids may be present in a weight ratio (wt%) of about 5% to about 90%, about 5% to about 70%, about 5% to about 50%, about 5% to about 40%, about 5% to about 30%, about 10 % to about 70%, about 10% to about 50%, or about 10% to about 40% of the total lipids present in a composition.
  • the percentage of non-cationic lipid in a liposome may be greater than about 5 wt%, greater than about 10 wt%, greater than about 20 wt%, greater than about 30 wt%, or greater than about 40 wt%.
  • the percentage total non-cationic lipids in a liposome may be greater than about 5 wt%, greater than about 10 wt%, greater than about 20 wt%, greater than about 30 wt%, or greater than about 40 wt%. In some embodiments, the percentage of non- cationic lipid in a liposome is no more than about 5 wt%, no more than about 10 wt%, no more than about 20 wt%, no more than about 30 wt%, or no more than about 40 wt%.
  • the percentage total non-cationic lipids in a liposome may be no more than about 5 wt%, no more than about 10 wt%, no more than about 20 wt%, no more than about 30 wt%, or no more than about 40 wt%.
  • a composition (e.g., a liposomal composition) comprises one or more cholesterol-based lipids.
  • suitable cholesterol-based lipids include cholesterol and , for example, DC-Chol (N,N-dimethyl-N-ethylcarboxamidocholesterol), 1,4-bis(3-N-oleylamino- propyl)piperazine (Gao, et al. Biochem. Biophys. Res. Comm.179, 280 (1991); Wolf et al.
  • a cholesterol-based lipid is cholesterol.
  • a cholesterol-based lipid may be present in a molar ratio (mol%) of about 1% to about 30%, or about 5% to about 20% of the total lipids present in a liposome.
  • the percentage of cholesterol-based lipid in the lipid nanoparticle may be greater than about 5 mol%, greater than about 10 mol%, greater than about 20 mol%, greater than about 30 mol%, or greater than about 40 mol%.
  • the percentage of cholesterol- based lipid in the lipid nanoparticle may be no more than about 5 mol%, no more than about 10 mol%, no more than about 20 mol%, no more than about 30 mol%, or no more than about 40 mol%.
  • a cholesterol-based lipid may be present in a weight ratio (wt%) of
  • the percentage of cholesterol-based lipid in the lipid nanoparticle may be greater than about 5 wt%, greater than about 10 wt%, greater than about 20 wt%, greater than about 30 wt%, or greater than about 40 wt%. In some embodiments, the percentage of cholesterol-based lipid in the lipid nanoparticle may be no more than about 5 wt%, no more than about 10 wt%, no more than about 20 wt%, no more than about 30 wt%, or no more than about 40 wt%.
  • a composition (e.g., a liposomal composition) comprises one or more PEGylated lipids.
  • PEG polyethylene glycol
  • derivatized lipids such as derivatized ceramides (PEG-CER), including N-octanoyl-sphingosine-1-[succinyl(methoxy polyethylene glycol)-2000] (C8 PEG-2000 ceramide) is also contemplated by the present invention in combination with one or more of the cationic and, in some embodiments, other lipids together which comprise the liposome.
  • particularly useful exchangeable lipids are PEG-ceramides having shorter acyl chains (e.g., C 14 or C 18 ).
  • Contemplated PEG-modified lipids include, but are not limited to, a polyethylene glycol chain of up to 5 kDa in length covalently attached to a lipid with alkyl chain(s) of C6-C20 length.
  • a PEG-modified or PEGylated lipid is PEGylated cholesterol or PEG-2K.
  • a PEG-modified lipid is 1,2-dimyristoyl-sn-glycerol, methoxypolyethylene glycol (DMG-PEG2000).
  • a PEG-modified phospholipid and derivatized lipids of the present invention may be present in a molar ratio (mol%) from about 0% to about 15%, about 0.5% to about 15%, about 1% to about 15%, about 4% to about 10%, or about 2% of the total lipid present in the composition (e.g., a liposomal composition).
  • a PEG-modified phospholipid and derivatized lipids of the present invention may be present in a weight ratio (wt%) from about 0% to about 15%, about 0.5% to about 15%, about 1% to about 15%, about 4% to about 10%, or about 2% of the total lipid present in the composition (e.g., a liposomal composition).
  • Cationic lipids described herein e.g., a cationic lipid of any of Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III-B), (IV-A), (IV-B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)-(156)) may be used in the preparation of compositions (e.g., to construct liposomal compositions) that facilitate or enhance the delivery and release of encapsulated materials (e.g., one or more therapeutic polynucleotides) to one or more target cells (e.g., by permeating or fusing with the lipid membranes of such target cells).
  • encapsulated materials e.g., one or more therapeutic
  • a liposomal composition e.g., a lipid nanoparticle
  • the phase transition in the lipid bilayer of the one or more target cells may facilitate the delivery of the encapsulated materials (e.g., one or more therapeutic polynucleotides encapsulated in a lipid nanoparticle) into the one or more target cells.
  • the encapsulated materials e.g., one or more therapeutic polynucleotides encapsulated in a lipid nanoparticle
  • cationic lipids described herein e.g., a cationic lipid of any of Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III- B), (IV-A), (IV-B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)-(156)) may be used to prepare liposomal vehicles that are characterized by their reduced toxicity in vivo.
  • the reduced toxicity is a function of the high transfection efficiencies associated with the compositions disclosed herein, such that a reduced quantity of such composition may administered to the subject to achieve a desired therapeutic response or outcome.
  • pharmaceutical formulations comprising a cationic lipid described herein (e.g., a cationic lipid of any of Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II- B), (III-A), (III-B), (IV-A), (IV-B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)- (156)) and nucleic acids provided by the present invention may be used for various therapeutic purposes.
  • a cationic lipid described herein e.g., a cationic lipid of any of Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III-B), (IV-A), (IV-B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)-(156)) and nucleic acids can be formulated in combination with one or more additional pharmaceutical carriers, targeting ligands or stabilizing reagents.
  • additional pharmaceutical carriers targeting ligands or stabilizing reagents.
  • a cationic lipid described herein e.g., a cationic lipid of any of Formulas (I)-(VI) such as any of Formulas (I-A)-(I- C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III-B), (IV-A), (IV-B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)-(156)) can be formulated via pre-mixed lipid solution.
  • a composition comprising a cationic lipid described herein (e.g., a cationic lipid of any of Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III-B), (IV-A), (IV-B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)-(156)) can be formulated using post-insertion techniques into the lipid membrane of the nanoparticles.
  • a cationic lipid described herein e.g., a cationic lipid of any of Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C
  • Suitable routes of administration include, for example, oral, rectal, vaginal, transmucosal,
  • intramuscular administration is to a muscle selected from the group consisting of skeletal muscle, smooth muscle and cardiac muscle.
  • the administration results in delivery of the nucleic acids to a muscle cell.
  • the administration results in delivery of the nucleic acids to a hepatocyte (i.e., liver cell).
  • administration is intramuscular.
  • administration is intravenous.
  • compositions of the invention may be administered in a local rather than systemic manner, for example, via injection of the pharmaceutical formulation directly into a targeted tissue, preferably in a sustained release formulation.
  • Local delivery can be affected in various ways, depending on the tissue to be targeted.
  • Exemplary tissues in which delivered mRNA may be delivered and/or expressed include, but are not limited to the liver, kidney, heart, spleen, serum, brain, skeletal muscle, lymph nodes, skin, and/or cerebrospinal fluid.
  • the tissue to be targeted in the liver include, but are not limited to the liver, kidney, heart, spleen, serum, brain, skeletal muscle, lymph nodes, skin, and/or cerebrospinal fluid.
  • compositions of the present invention can be inhaled (for nasal, tracheal, or bronchial delivery); compositions of the present invention can be injected into the site of injury, disease manifestation, or pain, for example; compositions can be provided in lozenges for oral, tracheal, or esophageal application; can be supplied in liquid, tablet or capsule form for administration to the stomach or intestines, can be supplied in suppository form for rectal or vaginal application; or can even be delivered to the eye by use of creams, drops, or even injection. [0355] In embodiments, administration is via pulmonary delivery.
  • pulmonary delivery refers to delivery to lung via, e.g., nasal cavity, trachea, bronchi, bronchioles, and/or other pulmonary system.
  • a composition described herein is formulated for nebulization.
  • the delivery vehicle may be in an aerosolized composition which can be inhaled.
  • pulmonary delivery involves inhalation (e.g., for nasal, tracheal, or bronchial delivery).
  • a composition is nebulized prior to inhalation.
  • the present invention provides methods for delivering a composition having full-length mRNA molecules encoding a peptide or polypeptide of interest for use in the treatment of a subject, e.g., a human subject or a cell of a human subject or a cell that is treated and delivered to a human subject.
  • a subject e.g., a human subject or a cell of a human subject or a cell that is treated and delivered to a human subject.
  • the present invention provides a method for producing a therapeutic composition comprising full-length mRNA that encodes a peptide or polypeptide for use in the delivery to or treatment of the lung of a subject or a lung cell.
  • the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for cystic fibrosis transmembrane conductance regulator (CFTR) protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for ATP-binding cassette sub-family A member 3 protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for dynein axonemal intermediate chain 1 protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for dynein axonemal heavy chain 5 (DNAH5) protein.
  • CFTR cystic fibrosis transmembrane conductance regulator
  • the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for alpha-1-antitrypsin protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for forkhead box P3 (FOXP3) protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes one or more surfactant protein, e.g., one or more of surfactant A protein, surfactant B protein, surfactant C protein, and surfactant D protein.
  • surfactant protein e.g., one or more of surfactant A protein, surfactant B protein, surfactant C protein, and surfactant D protein.
  • the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes a peptide or polypeptide for use in the delivery to or treatment of the liver of a subject or a liver cell.
  • peptides and polypeptides can include those associated with a urea cycle disorder, associated with a lysosomal storage disorder, with a glycogen storage disorder, associated with an amino acid metabolism disorder, associated with a lipid metabolism or fibrotic disorder, associated with methylmalonic acidemia, or associated with any other metabolic disorder for which delivery to or treatment of the liver or a liver cell with enriched full-length mRNA provides therapeutic benefit.
  • the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for a protein associated with a urea cycle disorder. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for ornithine transcarbamylase (OTC) protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for arginosuccinate synthetase 1 protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for carbamoyl phosphate synthetase I protein.
  • OTC ornithine transcarbamylase
  • the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for arginosuccinate lyase protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for arginase protein. [0360] In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for a protein associated with a lysosomal storage disorder. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for alpha galactosidase protein.
  • the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for glucocerebrosidase protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for iduronate-2-sulfatase protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for iduronidase protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for N- acetyl-alpha-D-glucosaminidase protein.
  • the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for heparan N-sulfatase protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for galactosamine-6 sulfatase protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for beta-galactosidase protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for lysosomal lipase protein.
  • the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for arylsulfatase B (N-acetylgalactosamine-4-sulfatase) protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for transcription factor EB (TFEB). [0361] In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for a protein associated with a glycogen storage disorder. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for acid alpha-glucosidase protein.
  • TFEB transcription factor EB
  • the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for glucose-6-phosphatase (G6PC) protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for liver glycogen phosphorylase protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for muscle phosphoglycerate mutase protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for glycogen debranching enzyme.
  • G6PC glucose-6-phosphatase
  • the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for a protein associated with amino acid metabolism. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for phenylalanine hydroxylase enzyme. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for glutaryl-CoA dehydrogenase enzyme. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for propionyl-CoA caboxylase enzyme.
  • the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for oxalase alanine-glyoxylate aminotransferase enzyme. [0363] In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for a protein associated with a lipid metabolism or fibrotic disorder. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for a mTOR inhibitor. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for ATPase phospholipid transporting 8B1 (ATP8B1) protein.
  • ATP8B1 ATP8B1
  • the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for one or more NF-kappa B inhibitors, such as one or more of I-kappa B alpha, interferon-related development regulator 1 (IFRD1), and Sirtuin 1 (SIRT1).
  • the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for PPAR-gamma protein or an active variant.
  • the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for a protein associated with methylmalonic acidemia.
  • the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for methylmalonyl CoA mutase protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for methylmalonyl CoA epimerase protein. [0365] In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA for which delivery to or treatment of the liver can provide therapeutic benefit. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for ATP7B protein, also known as Wilson disease protein.
  • the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for porphobilinogen deaminase enzyme. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for one or clotting enzymes, such as Factor VIII, Factor IX, Factor VII, and Factor X. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for human hemochromatosis (HFE) protein.
  • HFE human hemochromatosis
  • the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes a peptide or polypeptide for use in the delivery to or treatment of the cardiovasculature of a subject or a cardiovascular cell.
  • the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for vascular endothelial growth factor A protein.
  • the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for relaxin protein.
  • the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for bone morphogenetic protein-9 protein.
  • the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for bone morphogenetic protein-2 receptor protein.
  • the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes a peptide or polypeptide for use in the delivery to or treatment of the muscle of a subject or a muscle cell.
  • the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for dystrophin protein.
  • the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for frataxin protein.
  • the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes a peptide or polypeptide for use in the delivery to or treatment of the cardiac muscle of a subject or a cardiac muscle cell.
  • the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for a protein that modulates one or both of a potassium channel and a sodium channel in muscle tissue or in a muscle cell.
  • the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for a protein that modulates a Kv7.1 channel in muscle tissue or in a muscle cell.
  • the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for a protein that modulates a Nav1.5 channel in muscle tissue or in a muscle cell.
  • the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes a peptide or polypeptide for use in the delivery to or treatment of the nervous system of a subject or a nervous system cell.
  • the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for survival motor neuron 1 protein.
  • the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for survival motor neuron 2 protein.
  • the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for frataxin protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for ATP binding cassette subfamily D member 1 (ABCD1) protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for CLN3 protein. [0369] In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes a peptide or polypeptide for use in the delivery to or treatment of the blood or bone marrow of a subject or a blood or bone marrow cell.
  • ABCD1 ATP binding cassette subfamily D member 1
  • the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for beta globin protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for Bruton’s tyrosine kinase protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for one or clotting enzymes, such as Factor VIII, Factor IX, Factor VII, and Factor X.
  • the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes a peptide or polypeptide for use in the delivery to or treatment of the kidney of a subject or a kidney cell. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for collagen type IV alpha 5 chain (COL4A5) protein. [0371] In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes a peptide or polypeptide for use in the delivery to or treatment of the eye of a subject or an eye cell.
  • the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for ATP-binding cassette sub-family A member 4 (ABCA4) protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for retinoschisin protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for retinal pigment epithelium-specific 65 kDa (RPE65) protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for centrosomal protein of 290 kDa (CEP290).
  • ABCA4 ATP-binding cassette sub-family A member 4
  • an mRNA encodes for an antigen from an infectious agent.
  • the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes a peptide or polypeptide for use in the delivery of or treatment with a vaccine for a subject or a cell of a subject.
  • the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antigen from an infectious agent, such as a virus.
  • the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antigen from influenza virus.
  • the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antigen from respiratory syncytial virus. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antigen from rabies virus. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antigen from cytomegalovirus. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antigen from rotavirus.
  • the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antigen from a hepatitis virus, such as hepatitis A virus, hepatitis B virus, or hepatis C virus.
  • a hepatitis virus such as hepatitis A virus, hepatitis B virus, or hepatis C virus.
  • the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antigen from human papillomavirus.
  • the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antigen from a herpes simplex virus, such as herpes simplex virus 1 or herpes simplex virus 2.
  • the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antigen from a human immunodeficiency virus, such as human immunodeficiency virus type 1 or human immunodeficiency virus type 2.
  • a human immunodeficiency virus such as human immunodeficiency virus type 1 or human immunodeficiency virus type 2.
  • the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antigen from a human metapneumovirus.
  • the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antigen from a human parainfluenza virus, such as human parainfluenza virus type 1, human parainfluenza virus type 2, or human parainfluenza virus type 3.
  • the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antigen from malaria virus. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antigen from zika virus. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antigen from chikungunya virus. [0374] In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antigen associated with a cancer of a subject or identified from a cancer cell of a subject.
  • the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antigen determined from a subject’s own cancer cell, i.e., to provide a personalized cancer vaccine. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antigen expressed from a mutant KRAS gene. [0375] In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antibody. In certain embodiments, the antibody can be a bi-specific antibody. In certain embodiments, the antibody can be part of a fusion protein.
  • the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antibody to OX40. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antibody to VEGF. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antibody to tissue necrosis factor alpha. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antibody to CD3. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antibody to CD19.
  • the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an immunomodulator. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for Interleukin 12. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for Interleukin 23. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for Interleukin 36 gamma.
  • the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for a constitutively active variant of one or more stimulator of interferon genes (STING) proteins.
  • STING interferon genes
  • the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an endonuclease.
  • the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an RNA-guided DNA endonuclease protein, such as Cas 9 protein.
  • the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for a meganuclease protein.
  • the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for a transcription activator-like effector nuclease protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for a zinc finger nuclease protein.
  • exemplary therapeutic uses result from the delivery of mRNA encoding a secreted protein. Accordingly, in embodiments, the compositions and methods of the invention provide for delivery of mRNA encoding a secreted protein.
  • compositions and methods of the invention provide for delivery of mRNA encoding one or more secreted proteins listed in Table 1; thus, compositions of the invention may comprise an mRNA encoding a protein listed in Table 1 (or a homolog thereof) along with other components set out herein, and methods of the invention may comprise preparing and/or administering a composition comprising an mRNA encoding a protein listed in Table 1 (or a homolog thereof) along with other components set out herein Table 1.
  • Secreted Proteins secreted Proteins
  • compositions and methods of the invention provide for the delivery of one or more mRNAs encoding one or more additional exemplary proteins listed in Table 2; thus, compositions of the invention may comprise an mRNA encoding a protein listed in Table 2 (or a homolog thereof) along with other components set out herein, and methods of the invention may comprise preparing and/or administering a composition comprising an mRNA encoding a protein chosen from the proteins listed in Table 2 (or a homolog thereof) along with other components set out herein. Table 2. Additional Exemplary Proteins
  • the Uniprot IDs set forth in Table 1 and Table 2 refer to the human versions the listed proteins and the sequences of each are available from the Uniprot database. Sequences of the listed proteins are also generally available for various animals, including various mammals and animals of veterinary or industrial interest.
  • compositions and methods of the invention provide for the delivery of one or more mRNAs encoding one or more proteins chosen from mammalian homologs or homologs from an animal of veterinary or industrial interest of the secreted proteins listed in Table 1 and Table 2; thus, compositions of the invention may comprise an mRNA encoding a protein chosen from mammalian homologs or homologs from an animal of veterinary or industrial interest of a protein listed in Table 1 and Table 2 along with other components set out herein, and methods of the invention may comprise preparing and/or administering a composition comprising an mRNA encoding a protein chosen from mammalian homologs or homologs from an animal of veterinary or industrial interest of a protein listed in Table 1 and Table 2 along with other components set out herein.
  • mammalian homologs are chosen from mouse, rat, hamster, gerbil, horse, pig, cow, llama, alpaca, mink, dog, cat, ferret, sheep, goat, or camel homologs.
  • animal of veterinary or industrial interest is chosen from the mammals listed above and/or chicken, duck, turkey, salmon, catfish, or tilapia.
  • the compositions and methods of the invention provide for the delivery of mRNA encoding a lysosomal protein chosen from Table 3.
  • compositions and methods of the invention provide for the delivery of one or more mRNAs encoding one or more lysosomal and/or related proteins listed in Table 3; thus, compositions of the invention may comprise an mRNA encoding a protein listed in Table 3 (or a homolog thereof) along with other components set out herein, and methods of the invention may comprise preparing and/or administering a composition comprising an mRNA encoding a protein chosen from the proteins listed in Table 3 (or a homolog thereof) along with other components set out herein. Table 3. Lysosomal and Related Proteins
  • compositions and methods of the invention provide for the delivery of mRNA encoding a therapeutic protein (e.g., cytosolic, transmembrane or secreted) such as those listed in Table 4.
  • a therapeutic protein e.g., cytosolic, transmembrane or secreted
  • compositions and methods of the invention provide for the delivery of an mRNA encoding a therapeutic protein useful in treating a disease or disorder (i.e., indication) listed in Table 4; thus, compositions of the invention may comprise an mRNA encoding a therapeutic protein listed or not listed in Table 4 (or a homolog thereof, as discussed below) along with other components set out herein for treating a disease or disorder (i.e., indication) listed in Table 4, and methods of the invention may comprise preparing and/or administering a composition comprising an mRNA encoding a such a protein (or a homolog thereof, as discussed below) along with other components set out herein for treatment of a disease or disorder listed in Table 4.
  • Table 4 Exemplary Indications and Related Proteins
  • the present invention is used to prevent, treat and/or cure a subject affected with a disease or disorder listed or associated with the proteins listed in Tables 1, 2, 3, or 4.
  • an mRNA encodes one or more of Cystic Fibrosis Transmembrane
  • CFTR Conductance Regulator
  • ASS1 argininosuccinate synthetase
  • SNS1 survival motor neuron 1
  • PAH phenylalanine hydroxylase
  • Cationic lipids described herein can be prepared according to the exemplary synthesis of
  • a cationic lipid described herein can be prepared by conjugating a thiol with a di-carboxylic acid under suitable conditions.
  • exemplary di-carboxylic acids are described in Table A
  • exemplary thiols are described in Table B.
  • suitable cationic lipids include those resulting from any combination of the precursors described in Table A and Table B. Table A. Dicarboxylic Acids
  • Example 3 Lipid Nanoparticle Formulation Using Thioester Cationic Lipids
  • Cationic lipids described herein can be used in the preparation of lipid nanoparticles according to methods known in the art.
  • suitable methods include methods described in
  • Process A relates to a conventional method of encapsulating mRNA by mixing mRNA with a mixture of lipids, without first pre-forming the lipids into lipid nanoparticles.
  • Process A (“A”) relates to a conventional method of encapsulating mRNA by mixing mRNA with a mixture of lipids, without first pre-forming the lipids into lipid nanoparticles.
  • an ethanol lipid solution and an aqueous buffered solution of mRNA were prepared separately.
  • a solution of mixture of lipids (cationic lipid, helper lipids, zwitterionic lipids, PEG lipids etc.) was prepared by dissolving lipids in ethanol.
  • the mRNA solution was prepared by dissolving the mRNA in citrate buffer, resulting in mRNA at a
  • a second exemplary process for lipid nanoparticle formulation is Process B of WO 2018/089801 (see, e.g., Example 2 and Figure 2 of WO 2018/089801).
  • Process B refers to a process of encapsulating messenger RNA (mRNA) by mixing pre-formed lipid nanoparticles with mRNA.
  • mRNA messenger RNA
  • a range of different conditions, such as varying temperatures (i.e., heating or not heating the mixture), buffers, and concentrations, may be employed in Process B.
  • lipids dissolved in ethanol and citrate buffer were mixed using a pump system. The instantaneous mixing of the two streams resulted in the formation of empty lipid nanoparticles, which was a self- assembly process.
  • the resultant formulation mixture was empty lipid nanoparticles in citrate buffer containing alcohol.
  • the formulation was then subjected to a TFF purification process wherein buffer exchange occurred.
  • IV Intravenous
  • lipid nanoparticle formulations comprising a thioester cationic lipid and mRNA encoding hEPO (Table 5) was undertaken in order to study mRNA delivery and resultant hEPO expression.
  • Male CD1 mice at 6-8 weeks old were given a single intravenous injection of the LNP formulations at a dosage level of 1 mg/kg.
  • Blood samples were collected by tail snip at 6 and 24 hours post-dose.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Molecular Biology (AREA)
  • Engineering & Computer Science (AREA)
  • Dispersion Chemistry (AREA)
  • Biophysics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Immunology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Oil, Petroleum & Natural Gas (AREA)
  • Biochemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Medicinal Preparation (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Organic Low-Molecular-Weight Compounds And Preparation Thereof (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)

Abstract

Disclosed are cationic lipids which are compounds of Formula (I), (II), (III), (IV), (V), or (VI). Cationic lipids provided herein can be useful for delivery and expression of mRNA and encoded protein, e.g., as a component of liposomal delivery vehicle, and accordingly can be useful for treating various diseases, disorders and conditions, such as those associated with deficiency of one or more proteins.

Description

THIOESTER CATIONIC LIPIDS CROSS-REFERENCE TO RELATED APPLICATIONS
[0001] The present application claims benefit of U.S. Application No.62/676,147, filed May 24, 2018;
.U.S. Application No.62/748,097, filed October 19, 2018; and U.S. Application No.62/750,013, filed October 24, 2018, each of which is incorporated by reference in its entirety. BACKGROUND
[0002] Delivery of nucleic acids has been explored extensively as a potential therapeutic option for certain disease states. In particular, messenger RNA (mRNA) therapy has become an increasingly important option for treatment of various diseases, including for those associated with deficiency of one or more proteins. SUMMARY
[0003] The present invention provides, among other things, cationic lipids useful in for delivery of mRNA. Delivery of mRNA provided by cationic lipids described herein can result in targeted delivery, reduce administration frequency, improve patient tolerability, and provide more potent and less toxic mRNA therapy for the treatment of a variety of diseases, including but not limited to cancer, cardiovascular, cystic fibrosis, infectious, and neurological diseases. [0004] In one aspect, the present invention provides a cationic lipid of Formula (I):
Figure imgf000003_0001
wherein
R1 is hydrogen, C6-C30 alkyl, C6-C30 alkenyl, or C6-C30 alkynyl, or Substructure Y; each a and b is an integer of 0-6;
each XA1 and XB1 is independently O or S;
each LA and LB is independently C1-C10 alkylene; C2-C10 alkenylene; or C2-C10 alkynylene; XA2 is independently NH, NRA, CH2, or CHRA;
XB2 is independently NH, NRB, CH2, or CHRB; each RA and RB is independently C6-C30 alkyl, C6-C30 alkenyl, or C6-C30 alkynyl; and
Substructure
Figure imgf000004_0001
.
[0005] In another aspect, the present invention provides a cationic lipid of Formula (II):
Figure imgf000004_0004
wherein
R1 is hydrogen, C6-C30 alkyl, C6-C30 alkenyl, or C6-C30 alkynyl, or Substructure Z;
each a and b is an integer of 0-6;
each XA1 and XB1 is independently O or S;
each LA and LB is independently C1-C10 alkylene; C2-C10 alkenylene; or C2-C10 alkynylene; XA2 is independently NH, NRA, CH2, or CHRA;
XB2 is independently NH, NRB, CH2, or CHRB;
each RA and RB is independently C6-C30 alkyl, C6-C30 alkenyl, or C6-C30 alkynyl; and
Substructure
Figure imgf000004_0002
. [0006] In yet another aspect, the present invention provides a cationic lipid of Formula (III):
Figure imgf000004_0003
wherein
each a is an integer of 0-6;
each XA1 is independently O or S;
each LA is independently C1-C10 alkylene; C2-C10 alkenylene; or C2-C10 alkynylene; XA2 is independently NH, NRA, CH2, or CHRA; and
each RA is independently C6-C30 alkyl, C6-C30 alkenyl, or C6-C30 alkynyl. [0007] In still another aspect, the present invention provides cationic lipid having a structure according to Formula (IV),
Figure imgf000005_0001
wherein
R1 is hydrogen, C1-C30 alkyl, C2-C30 alkenyl, or C2-C30 alkynyl;
each a and b is an integer of 0-6;
each XA1 and XB1 is independently O or S;
each LA and LB is independently C1-C10 alkylene; C2-C10 alkenylene; or C2-C10 alkynylene; LC is independently–C(O)– or–(CH2)b–;
XA2 is independently NH, NRA, CH2, or CHRA;
XB2 is independently NH, NRB, CH2, or CHRB; and
each RA and RB is independently C6-C30 alkyl, C6-C30 alkenyl, or C6-C30 alkynyl. [0008] In embodiments, LC is–C(O)–. In embodiments, LC is–(CH2)b–. [0009] In embodiments, a cationic lipid has a structure according to Formula (I-A),
Figure imgf000005_0002
[0010] In embodiments, a cationic lipid has a structure according to Formula (I-A’),
Figure imgf000006_0001
[0011] In embodiments, a cationic lipid has a structure according to Formula (IV-A):
Figure imgf000006_0002
[0012] In embodiments, each XA1 and XB1 is O, or each XA1 and XB1 is S. In embodiments, each XA1 and XB1 is O. In embodiments, each XA1 and XB1 is S.
[0013] In embodiments, each a and b is independently 0, 1, or 2.
[0014] In embodiments, each XA2 is NRA or CHRA.
[0015] In embodiments, each XB2 is NRB or CHRB.
[0016] In embodiments, a cationic lipid has a structure according to Formula (I-B),
Figure imgf000006_0003
[0017] In embodiments, a cationic lipid has a structure according to Formula (I-B’),
Figure imgf000007_0001
[0018] In embodiments, a cationic lipid has a structure according to Formula (I-B”),
Figure imgf000007_0002
wherein d is independently an integer of 0-5. [0019] In embodiments, a cationic lipid has a structure according to Formula (II-A),
Figure imgf000007_0003
[0020] In embodiments, a cationic lipid has a structure according to Formula (III-A),
Figure imgf000007_0004
[0021] In embodiments, a cationic lipid has a structure according to Formula (IV-B),
Figure imgf000007_0005
[0022] In embodiments, a cationic lipid has a structure according to Formula (IV-B’),
Figure imgf000008_0001
[0023] In embodiments, a cationic lipid has a structure according to Formula (IV-B”),
Figure imgf000008_0002
[0024] In another aspect, the present invention provides a cationic lipid has a structure according to Formula (V),
Figure imgf000008_0003
wherein
each LA and LB is independently C1-C10 alkylene; C2-C10 alkenylene; or C2-C10 alkynylene; and
each RA and RB is independently C6-C30 alkyl, C6-C30 alkenyl, C6-C30 alkynyl, or C1-C15 alkylene-C(O)2- C1-C15 alkyl. [0025] In embodiments, each LA is C1-C10 alkylene. [0026] In embodiments, each LB is C1-C10 alkylene. [0027] In embodiments, each LA and LB is unsubstituted C1-C10 alkylene.
[0028] In embodiments, a cationic lipid has a structure according to Formula (I-C),
Figure imgf000009_0001
wherein each c is independently an integer of 2-10.
[0029] In embodiments, a cationic lipid has a structure according to Formula (I-C’),
Figure imgf000009_0002
wherein each c is independently an integer of 2-10.
[0030] In embodiments, a cationic lipid has a structure according to Formula (I-C”),
Figure imgf000009_0003
, wherein each c is independently an integer of 2-10, and d is independently an integer of 0-5. In embodiments, d is 0, 1, 2, 3, or 4.
[0031] In embodiments, a cationic lipid has a structure according to Formula (II-B),
Figure imgf000009_0004
wherein each c is independently an integer of 2-10. [0032] In embodiments, a cationic lipid has a structure according to Formula (III-B),
Figure imgf000010_0001
wherein each c is independently an integer of 2-10.
[0033] In embodiments, a cationic lipid has a structure according to Formula (III-C),
Figure imgf000010_0002
wherein each c is independently an integer of 2-10.
[0034] In embodiments, a cationic lipid has a structure according to Formula (III-C’),
Figure imgf000010_0003
wherein each c is independently an integer of 2-10.
[0035] In embodiments, a cationic lipid has a structure according to Formula (IV-C”),
Figure imgf000011_0001
, wherein each c is independently an integer of 2- 10. [0036] In another aspect, the present invention provides a cationic lipid has a structure according to Formula (VI),
Figure imgf000011_0002
,
wherein
each RA and RB is independently C1-C30 alkyl, C2-C30 alkenyl, C2-C30 alkynyl, or C1- C15 alkylene-C(O)2- C1-C15 alkyl; and
each c is independently an integer of 2-10.
[0037] In embodiments, each c is 2, 3, or 4. In embodiments, each c is 4, 5, 6, 7, 8, 9, or 10. In
embodiments, each c is 4. [0038] In embodiments, each RA is C6-C20 alkyl or C6-C20 alkenyl. In embodiments, each RA is
unsubstituted C6-C20 alkyl, C6-C20 hydroxyalkyl, unsubstituted C6-C20 alkenyl, or C6-C20 hydroxyalkenyl. [0039] In embodiments, each RB is C6-C20 alkyl or C6-C20 alkenyl. In embodiments, each RB is
unsubstituted C6-C20 alkyl, C6-C20 hydroxyalkyl, unsubstituted C6-C20 alkenyl, or C6-C20 hydroxyalkenyl. [0040] In embodiments, each RA and RB is C6-C20 hydroxyalkyl, or each RA and RB is C6-C20
hydroxyalkenyl. In embodiments, each RA and RB is -CH2CH(OH)C10H21, or each RA and RB is -CH2CH(OH)(CH2)6(CH=CH)CH2(CH=CH)C5H11. In embodiments, each RA and RB
is -CH2CH(OH)C10H21. [0041] In embodiments, each RA and RB is -CH2CH(OH)RC, and wherein RC is selected from the group consisting of:
Figure imgf000012_0001
. [0042] The present invention also provides a cationic lipid that is any one of compounds 1-156.
[0043] In embodiments, a cationic lipid is
Figure imgf000012_0002
Figure imgf000013_0001
[0044] The present invention also provides a cationic lipid that is:
Figure imgf000014_0001
Figure imgf000014_0002
[0045] In another aspect, the invention features a composition comprising an mRNA encoding a
peptide or a polypeptide, encapsulated within a liposome, wherein the liposome comprises one or more cationic lipids, one or more non-cationic lipids, one or more cholesterol-based lipids and one or more PEG-modified lipids, wherein at least one cationic lipid is as described herein (e.g., a cationic lipid of any of Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III-B), (IV-A), (IV-B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)-(156)). [0046] In embodiments, a composition comprises an mRNA encoding for cystic fibrosis transmembrane conductance regulator (CFTR) protein. [0047] In embodiments, a composition comprises an mRNA encoding for ornithine transcarbamylase (OTC) protein. [0048] In embodiments, a composition comprises an mRNA encoding for an antigen (e.g., an antigen from an infectious agent). [0049] In another aspect, the invention features a composition comprising a nucleic acid encapsulated within a liposome, wherein the liposome comprises a cationic lipid as described herein (e.g., a cationic lipid of any of Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III-B), (IV-A), (IV-B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)-(156)). [0050] In embodiments, a composition further comprises one or more lipids selected from the group consisting of one or more cationic lipids, one or more non-cationic lipids, and one or more PEG- modified lipids. [0051] In embodiments, a nucleic acid is an mRNA encoding a peptide or polypeptide. [0052] In embodiments, an mRNA encodes a peptide or polypeptide for use in the delivery to or
treatment of the lung of a subject or a lung cell. In embodiments, an mRNA encodes cystic fibrosis transmembrane conductance regulator (CFTR) protein. [0053] In embodiments, an mRNA encodes a peptide or polypeptide for use in the delivery to or
treatment of the liver of a subject or a liver cell. In embodiments, an mRNA encodes ornithine transcarbamylase (OTC) protein. [0054] In embodiments, an mRNA encodes a peptide or polypeptide for use in a vaccine. In
embodiments, an mRNA encodes an antigen (e.g., an antigen from an infectious agent). [0055] In embodiments, a composition is formulated for intravenous (IV) administration. In
embodiments, a composition is formulated for intramuscular (IM) administration. In embodiments, a composition is formulated for administration by inhalation (e.g., a composition is formulated for nebulization). [0056] In some aspects, the present invention provides methods of treating a disease in a subject comprising administering to the subject a composition (e.g., a pharmaceutical composition) as described herein (e.g., a composition comprising a cationic lipid of any of Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III-B), (IV-A), (IV-B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)-(156)). BRIEF DESCRIPTION OF DRAWINGS
[0057] FIG.1 shows hEPO protein expression following intravenous (IV) administration of lipid
nanoparticle formulations comprising a cationic lipid described herein and mRNA encoding hEPO. Protein expression was determined using ELISA.
DETAILED DESCRIPTION OF CERTAIN EMBODIMENTS
Definitions
[0058] In order for the present invention to be more readily understood, certain terms are first defined below. Additional definitions for the following terms and other terms are set forth throughout the specification. The publications and other reference materials referenced herein to describe the background of the invention and to provide additional detail regarding its practice are hereby incorporated by reference. [0059] Amino acid: As used herein, the term“amino acid,” in its broadest sense, refers to any
compound and/or substance that can be incorporated into a polypeptide chain. In some embodiments, an amino acid has the general structure H2N–C(H)(R)–COOH. In some embodiments, an amino acid is a naturally occurring amino acid. In some embodiments, an amino acid is a synthetic amino acid; in some embodiments, an amino acid is a d-amino acid; in some
embodiments, an amino acid is an l-amino acid.“Standard amino acid” refers to any of the twenty standard l-amino acids commonly found in naturally occurring peptides.“Nonstandard amino acid” refers to any amino acid, other than the standard amino acids, regardless of whether it is prepared synthetically or obtained from a natural source. As used herein,“synthetic amino acid”
encompasses chemically modified amino acids, including but not limited to salts, amino acid derivatives (such as amides), and/or substitutions. Amino acids, including carboxy- and/or amino- terminal amino acids in peptides, can be modified by methylation, amidation, acetylation, protecting groups, and/or substitution with other chemical groups that can change the peptide’s circulating half-life without adversely affecting their activity. Amino acids may participate in a disulfide bond. Amino acids may comprise one or posttranslational modifications, such as association with one or more chemical entities (e.g., methyl groups, acetate groups, acetyl groups, phosphate groups, formyl moieties, isoprenoid groups, sulfate groups, polyethylene glycol moieties, lipid moieties, carbohydrate moieties, biotin moieties, etc.). The term“amino acid” is used interchangeably with“amino acid residue,” and may refer to a free amino acid and/or to an amino acid residue of a peptide. It will be apparent from the context in which the term is used whether it refers to a free amino acid or a residue of a peptide. [0060] Animal: As used herein, the term“animal” refers to any member of the animal kingdom. In some embodiments,“animal” refers to humans, at any stage of development. In some
embodiments,“animal” refers to non-human animals, at any stage of development. In certain embodiments, the non-human animal is a mammal (e.g., a rodent, a mouse, a rat, a rabbit, a monkey, a dog, a cat, a sheep, cattle, a primate, and/or a pig). In some embodiments, animals include, but are not limited to, mammals, birds, reptiles, amphibians, fish, insects, and/or worms. In some embodiments, an animal may be a transgenic animal, genetically-engineered animal, and/or a clone. [0061] Approximately or about: As used herein, the term“approximately” or“about,” as applied to one or more values of interest, refers to a value that is similar to a stated reference value. In certain embodiments, the term“approximately” or“about” refers to a range of values that fall within 25%, 20%, 19%, 18%, 17%, 16%, 15%, 14%, 13%, 12%, 11%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, or less in either direction (greater than or less than) of the stated reference value unless otherwise stated or otherwise evident from the context (except where such number would exceed 100% of a possible value). [0062] Biologically active: As used herein, the term“biologically active” refers to a characteristic of any agent that has activity in a biological system, and particularly in an organism. For instance, an agent that, when administered to an organism, has a biological effect on that organism, is considered to be biologically active. [0063] Delivery: As used herein, the term“delivery” encompasses both local and systemic delivery. For example, delivery of mRNA encompasses situations in which an mRNA is delivered to a target tissue and the encoded protein is expressed and retained within the target tissue (also referred to as“local distribution” or“local delivery”), and situations in which an mRNA is delivered to a target tissue and the encoded protein is expressed and secreted into patient’s circulation system (e.g., serum) and systematically distributed and taken up by other tissues (also referred to as“systemic distribution” or“systemic delivery”). [0064] Expression: As used herein,“expression” of a nucleic acid sequence refers to translation of an mRNA into a polypeptide, assemble multiple polypeptides into an intact protein (e.g., enzyme) and/or post-translational modification of a polypeptide or fully assembled protein (e.g., enzyme). In this application, the terms“expression” and“production,” and grammatical equivalent, are used inter-changeably. [0065] Functional: As used herein, a“functional” biological molecule is a biological molecule in a form in which it exhibits a property and/or activity by which it is characterized. [0066] Half-life: As used herein, the term“half-life” is the time required for a quantity such as nucleic acid or protein concentration or activity to fall to half of its value as measured at the beginning of a time period. [0067] Improve, increase, or reduce: As used herein, the terms“improve,”“increase” or“reduce,” or grammatical equivalents, indicate values that are relative to a baseline measurement, such as a measurement in the same individual prior to initiation of the treatment described herein, or a measurement in a control subject (or multiple control subject) in the absence of the treatment described herein. A“control subject” is a subject afflicted with the same form of disease as the subject being treated, who is about the same age as the subject being treated. [0068] In Vitro: As used herein, the term“in vitro” refers to events that occur in an artificial
environment, e.g., in a test tube or reaction vessel, in cell culture, etc., rather than within a multi- cellular organism. [0069] In Vivo: As used herein, the term“in vivo” refers to events that occur within a multi-cellular organism, such as a human and a non-human animal. In the context of cell-based systems, the term may be used to refer to events that occur within a living cell (as opposed to, for example, in vitro systems). [0070] Isolated: As used herein, the term“isolated” refers to a substance and/or entity that has been (1) separated from at least some of the components with which it was associated when initially produced (whether in nature and/or in an experimental setting), and/or (2) produced, prepared, and/or manufactured by the hand of man. Isolated substances and/or entities may be separated from about 10%, about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, about 91%, about 92%, about 93%, about 94%, about 95%, about 96%, about 97%, about 98%, about 99%, or more than about 99% of the other components with which they were initially associated. In some embodiments, isolated agents are about 80%, about 85%, about 90%, about 91%, about 92%, about 93%, about 94%, about 95%, about 96%, about 97%, about 98%, about 99%, or more than about 99% pure. As used herein, a substance is“pure” if it is substantially free of other components. As used herein, calculation of percent purity of isolated substances and/or entities should not include excipients (e.g., buffer, solvent, water, etc.). [0071] messenger RNA (mRNA): As used herein, the term“messenger RNA (mRNA)” or“mRNA” refers to a polynucleotide that encodes at least one polypeptide. mRNA as used herein encompasses both modified and unmodified RNA. The term“modified mRNA” related to mRNA comprising at least one chemically modified nucleotide. mRNA may contain one or more coding and non-coding regions. mRNA can be purified from natural sources, produced using recombinant expression systems and optionally purified, chemically synthesized, etc. Where appropriate, e.g., in the case of chemically synthesized molecules, mRNA can comprise nucleoside analogs such as analogs having chemically modified bases or sugars, backbone modifications, etc. An mRNA sequence is presented in the 5’ to 3’ direction unless otherwise indicated. In some embodiments, an mRNA is or comprises natural nucleosides (e.g., adenosine, guanosine, cytidine, uridine); nucleoside analogs (e.g., 2- aminoadenosine, 2-thiothymidine, inosine, pyrrolo-pyrimidine, 3-methyl adenosine, 5- methylcytidine, C-5 propynyl-cytidine, C-5 propynyl-uridine, 2-aminoadenosine, C5-bromouridine, C5-fluorouridine, C5-iodouridine, C5-propynyl-uridine, C5-propynyl-cytidine, C5-methylcytidine, 2- aminoadenosine, 7-deazaadenosine, 7-deazaguanosine, 8-oxoadenosine, 8-oxoguanosine, O(6)- methylguanine, and 2-thiocytidine); chemically modified bases; biologically modified bases (e.g., methylated bases); intercalated bases; modified sugars (e.g., 2’-fluororibose, ribose, 2’-deoxyribose, arabinose, and hexose); and/or modified phosphate groups (e.g., phosphorothioates and 5’-N- phosphoramidite linkages). [0072] Nucleic acid: As used herein, the term“nucleic acid,” in its broadest sense, refers to any
compound and/or substance that is or can be incorporated into a polynucleotide chain. In some embodiments, a nucleic acid is a compound and/or substance that is or can be incorporated into a polynucleotide chain via a phosphodiester linkage. In some embodiments,“nucleic acid” refers to individual nucleic acid residues (e.g., nucleotides and/or nucleosides). In some embodiments, “nucleic acid” refers to a polynucleotide chain comprising individual nucleic acid residues. In some embodiments,“nucleic acid” encompasses RNA as well as single and/or double-stranded DNA and/or cDNA. In some embodiments,“nucleic acid” encompasses ribonucleic acids (RNA), including but not limited to any one or more of interference RNAs (RNAi), small interfering RNA (siRNA), short hairpin RNA (shRNA), antisense RNA (aRNA), messenger RNA (mRNA), modified messenger RNA (mmRNA), long non-coding RNA (lncRNA), micro-RNA (miRNA) multimeric coding nucleic acid (MCNA), polymeric coding nucleic acid (PCNA), guide RNA (gRNA) and CRISPR RNA (crRNA). In some embodiments,“nucleic acid” encompasses deoxyribonucleic acid (DNA), including but not limited to any one or more of single-stranded DNA (ssDNA), double-stranded DNA (dsDNA) and
complementary DNA (cDNA). In some embodiments,“nucleic acid” encompasses both RNA and DNA. In embodiments, DNA may be in the form of antisense DNA, plasmid DNA, parts of a plasmid DNA, pre-condensed DNA, a product of a polymerase chain reaction (PCR), vectors (e.g., P1, PAC, BAC, YAC, artificial chromosomes), expression cassettes, chimeric sequences, chromosomal DNA, or derivatives of these groups. In embodiments, RNA may be in the form of messenger RNA (mRNA), ribosomal RNA (rRNA), signal recognition particle RNA (7 SL RNA or SRP RNA), transfer RNA (tRNA), transfer-messenger RNA (tmRNA), small nuclear RNA (snRNA), small nucleolar RNA (snoRNA), SmY RNA, small Cajal body-specific RNA (scaRNA), guide RNA (gRNA), ribonuclease P (RNase P), Y RNA, telomerase RNA component (TERC), spliced leader RNA (SL RNA), antisense RNA (aRNA or asRNA), cis-natural antisense transcript (cis-NAT), CRISPR RNA (crRNA), long noncoding RNA (lncRNA), micro- RNA (miRNA), piwi-interacting RNA (piRNA), small interfering RNA (siRNA), transacting siRNA (tasiRNA), repeat associated siRNA (rasiRNA), 73K RNA, retrotransposons, a viral genome, a viroid, satellite RNA, or derivatives of these groups. In some embodiments, a nucleic acid is a mRNA encoding a protein such as an enzyme. [0073] Patient: As used herein, the term“patient” or“subject” refers to any organism to which a
provided composition may be administered, e.g., for experimental, diagnostic, prophylactic, cosmetic, and/or therapeutic purposes. Typical patients include animals (e.g., mammals such as mice, rats, rabbits, non-human primates, and/or humans). In some embodiments, a patient is a human. A human includes pre- and post-natal forms. [0074] Pharmaceutically acceptable: The term“pharmaceutically acceptable”, as used herein, refers to substances that, within the scope of sound medical judgment, are suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio. [0075] Pharmaceutically acceptable salt: Pharmaceutically acceptable salts are well known in the art.
For example, S. M. Berge et al., describes pharmaceutically acceptable salts in detail in J.
Pharmaceutical Sciences (1977) 66:1-19. Pharmaceutically acceptable salts of the compounds of this invention include those derived from suitable inorganic and organic acids and bases. Examples of pharmaceutically acceptable, nontoxic acid addition salts are salts of an amino group formed with inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid or with organic acids such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid or rnalonic acid or by using other methods used in the art such as ion exchange. Other pharmaceutically acceptable salts include adipate, alginate, ascorbate, aspartate,
benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, formate, fumarate, glucoheptonate, glycerophosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2- hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectinate, persulfate, 3-phenylpropionate, phosphate, picrate, pivalate, propionate, stearate, succinate, sulfate, tartrate, thiocyanate, p-toluenesulfonate, undecanoate, valerate salts, and the like. Salts derived from appropriate bases include alkali metal, alkaline earth metal, ammonium and N+(C1-4 alkyl)4 salts. Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like. Further pharmaceutically acceptable salts include, when appropriate, nontoxic ammonium. quaternary ammonium, and amine cations formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, sulfonate and aryl sulfonate. Further pharmaceutically acceptable salts include salts formed from the quarternization of an amine using an appropriate electrophile, e.g., an alkyl halide, to form a quarternized alkylated amino salt. [0076] Systemic distribution or delivery: As used herein, the terms“systemic distribution,”“systemic delivery,” or grammatical equivalent, refer to a delivery or distribution mechanism or approach that affect the entire body or an entire organism. Typically, systemic distribution or delivery is accomplished via body’s circulation system, e.g., blood stream. Compared to the definition of“local distribution or delivery.” [0077] Subject: As used herein, the term“subject” refers to a human or any non-human animal (e.g., mouse, rat, rabbit, dog, cat, cattle, swine, sheep, horse or primate). A human includes pre- and post-natal forms. In many embodiments, a subject is a human being. A subject can be a patient, which refers to a human presenting to a medical provider for diagnosis or treatment of a disease. The term“subject” is used herein interchangeably with“individual” or“patient.” A subject can be afflicted with or is susceptible to a disease or disorder but may or may not display symptoms of the disease or disorder. [0078] Substantially: As used herein, the term“substantially” refers to the qualitative condition of exhibiting total or near-total extent or degree of a characteristic or property of interest. One of ordinary skill in the biological arts will understand that biological and chemical phenomena rarely, if ever, go to completion and/or proceed to completeness or achieve or avoid an absolute result. The term“substantially” is therefore used herein to capture the potential lack of completeness inherent in many biological and chemical phenomena. [0079] Target tissues: As used herein, the term“target tissues” refers to any tissue that is affected by a disease to be treated. In some embodiments, target tissues include those tissues that display disease-associated pathology, symptom, or feature. [0080] Therapeutically effective amount: As used herein, the term“therapeutically effective amount” of a therapeutic agent means an amount that is sufficient, when administered to a subject suffering from or susceptible to a disease, disorder, and/or condition, to treat, diagnose, prevent, and/or delay the onset of the symptom(s) of the disease, disorder, and/or condition. It will be appreciated by those of ordinary skill in the art that a therapeutically effective amount is typically administered via a dosing regimen comprising at least one unit dose. [0081] Treating: As used herein, the term“treat,”“treatment,” or“treating” refers to any method used to partially or completely alleviate, ameliorate, relieve, inhibit, prevent, delay onset of, reduce severity of and/or reduce incidence of one or more symptoms or features of a particular disease, disorder, and/or condition. Treatment may be administered to a subject who does not exhibit signs of a disease and/or exhibits only early signs of the disease for the purpose of decreasing the risk of developing pathology associated with the disease. [0082] Aliphatic: As used herein, the term aliphatic refers to C1-C40 hydrocarbons and includes both saturated and unsaturated hydrocarbons. An aliphatic may be linear, branched, or cyclic. For example, C1-C20 aliphatics can include C1-C20 alkyls (e.g., linear or branched C1-C20 saturated alkyls), C2-C20 alkenyls (e.g., linear or branched C4-C20 dienyls, linear or branched C6-C20 trienyls, and the like), and C2-C20 alkynyls (e.g., linear or branched C2-C20 alkynyls). C1-C20 aliphatics can include C3-C20 cyclic aliphatics (e.g., C3-C20 cycloalkyls, C4-C20 cycloalkenyls, or C8-C20 cycloalkynyls). In certain embodiments, the aliphatic may comprise one or more cyclic aliphatic and/or one or more heteroatoms such as oxygen, nitrogen, or sulfur and may optionally be substituted with one or more substituents such as alkyl, halo, alkoxyl, hydroxy, amino, aryl, ether, ester or amide. An aliphatic group is unsubstituted or substituted with one or more substituent groups as described herein. For example, an aliphatic may be substituted with one or more (e.g., 1, 2, 3, 4, 5, or 6 independently selected substituents) of halogen, -COR’, -CO2H, -CO2R’, -CN, -OH, -OR’, -OCOR’, -OCO2R’, -NH2, -NHR’, -N(R’)2, -SR’ or-SO2R’, wherein each instance of R’ independently is C1-C20 aliphatic (e.g., C1- C20 alkyl, C1-C15 alkyl, C1-C10 alkyl, or C1-C3 alkyl). In embodiments, R’ independently is an unsubstituted alkyl (e.g., unsubstituted C1-C20 alkyl, C1-C15 alkyl, C1-C10 alkyl, or C1-C3 alkyl). In embodiments, R’ independently is unsubstituted C1-C3 alkyl. In embodiments, the aliphatic is unsubstituted. In embodiments, the aliphatic does not include any heteroatoms. [0083] Alkyl: As used herein, the term“alkyl” means acyclic linear and branched hydrocarbon groups, e.g.“C1-C20 alkyl” refers to alkyl groups having 1-20 carbons. An alkyl group may be linear or branched. Examples of alkyl groups include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, butyl, isobutyl, sec-butyl, tert-butyl, pentyl, isopentyl tert-pentylhexyl, Isohexyletc. Other alkyl groups will be readily apparent to those of skill in the art given the benefit of the present disclosure. An alkyl group may be unsubstituted or substituted with one or more substituent groups as described herein. For example, an alkyl group may be substituted with one or more (e.g., 1, 2, 3, 4, 5, or 6 independently selected substituents) of halogen, -COR’, -CO2H, -CO2R’, -CN, -OH, -OR’, -OCOR’, -OCO2R’, -NH2, -NHR’, -N(R’)2, -SR’ or-SO2R’, wherein each instance of R’ independently is C1- C20 aliphatic (e.g., C1-C20 alkyl, C1-C15 alkyl, C1-C10 alkyl, or C1-C3 alkyl). In embodiments, R’ independently is an unsubstituted alkyl (e.g., unsubstituted C1-C20 alkyl, C1-C15 alkyl, C1-C10 alkyl, or C1-C3 alkyl). In embodiments, R’ independently is unsubstituted C1-C3 alkyl. In embodiments, the alkyl is substituted (e.g., with 1, 2, 3, 4, 5, or 6 substituent groups as described herein). In embodiments, an alkyl group is substituted with a–OH group and may also be referred to herein as a “hydroxyalkyl” group, where the prefix denotes the–OH group and“alkyl” is as described herein. [0084] Alkylene: The term“alkylene,” as used herein, represents a saturated divalent straight or
branched chain hydrocarbon group and is exemplified by methylene, ethylene, isopropylene and the like. Likewise, the term“alkenylene” as used herein represents an unsaturated divalent straight or branched chain hydrocarbon group having one or more unsaturated carbon-carbon double bonds that may occur in any stable point along the chain, and the term“alkynylene” herein represents an unsaturated divalent straight or branched chain hydrocarbon group having one or more unsaturated carbon-carbon triple bonds that may occur in any stable point along the chain. In certain embodiments, an alkylene, alkenylene, or alkynylene group may comprise one or more cyclic aliphatic and/or one or more heteroatoms such as oxygen, nitrogen, or sulfur and may optionally be substituted with one or more substituents such as alkyl, halo, alkoxyl, hydroxy, amino, aryl, ether, ester or amide. For example, an alkylene, alkenylene, or alkynylene may be substituted with one or more (e.g., 1, 2, 3, 4, 5, or 6 independently selected substituents) of halogen, -COR’, -CO2H, -CO2R’, -CN, -OH, -OR’, -OCOR’, -OCO2R’, -NH2, -NHR’, -N(R’)2, -SR’ or-SO2R’, wherein each instance of R’ independently is C1-C20 aliphatic (e.g., C1-C20 alkyl, C1-C15 alkyl, C1-C10 alkyl, or C1-C3 alkyl). In embodiments, R’ independently is an unsubstituted alkyl (e.g., unsubstituted C1-C20 alkyl, C1-C15 alkyl, C1-C10 alkyl, or C1-C3 alkyl). In embodiments, R’ independently is unsubstituted C1-C3 alkyl. In certain embodiments, an alkylene, alkenylene, or alkynylene is unsubstituted. In certain embodiments, an alkylene, alkenylene, or alkynylene does not include any heteroatoms. [0085] Alkenyl: As used herein,“alkenyl” means any linear or branched hydrocarbon chains having one or more unsaturated carbon-carbon double bonds that may occur in any stable point along the chain, e.g.“C2-C20 alkenyl” refers to an alkenyl group having 2-20 carbons. For example, an alkenyl group includes prop-2-enyl, but-2-enyl, but-3-enyl, 2-methylprop-2-enyl, hex-2-enyl, hex-5-enyl, 2,3- dimethylbut-2-enyl, and the like. In embodiments, the alkenyl comprises 1, 2, or 3 carbon-carbon double bond. In embodiments, the alkenyl comprises a single carbon-carbon double bond. In embodiments, multiple double bonds (e.g., 2 or 3) are conjugated. An alkenyl group may be unsubstituted or substituted with one or more substituent groups as described herein. For example, an alkenyl group may be substituted with one or more (e.g., 1, 2, 3, 4, 5, or 6
independently selected substituents) of halogen, -COR’, -CO2H, -CO2R’, -CN, -OH, -OR’, -OCOR’, - OCO2R’, -NH2, -NHR’, -N(R’)2, -SR’ or-SO2R’, wherein each instance of R’ independently is C1-C20 aliphatic (e.g., C1-C20 alkyl, C1-C15 alkyl, C1-C10 alkyl, or C1-C3 alkyl). In embodiments, R’ independently is an unsubstituted alkyl (e.g., unsubstituted C1-C20 alkyl, C1-C15 alkyl, C1-C10 alkyl, or C1-C3 alkyl). In embodiments, R’ independently is unsubstituted C1-C3 alkyl. In embodiments, the alkenyl is unsubstituted. In embodiments, the alkenyl is substituted (e.g., with 1, 2, 3, 4, 5, or 6 substituent groups as described herein). In embodiments, an alkenyl group is substituted with a–OH group and may also be referred to herein as a“hydroxyalkenyl” group, where the prefix denotes the–OH group and“alkenyl” is as described herein. [0086] Alkynyl: As used herein,“alkynyl” means any hydrocarbon chain of either linear or branched configuration, having one or more carbon-carbon triple bonds occurring in any stable point along the chain, e.g.“C2-C20 alkynyl” refers to an alkynyl group having 2-20 carbons. Examples of an alkynyl group include prop-2-ynyl, but-2-ynyl, but-3-ynyl, pent-2-ynyl, 3-methylpent-4-ynyl, hex-2-ynyl, hex- 5-ynyl, etc. In embodiments, an alkynyl comprises one carbon-carbon triple bond. An alkynyl group may be unsubstituted or substituted with one or more substituent groups as described herein. For example, an alkynyl group may be substituted with one or more (e.g., 1, 2, 3, 4, 5, or 6
independently selected substituents) of halogen, -COR’, -CO2H, -CO2R’, -CN, -OH, -OR’, -OCOR’, -OCO2R’, -NH2, -NHR’, -N(R’)2, -SR’ or-SO2R’, wherein each instance of R’ independently is C1-C20 aliphatic (e.g., C1-C20 alkyl, C1-C15 alkyl, C1-C10 alkyl, or C1-C3 alkyl). In embodiments, R’ independently is an unsubstituted alkyl (e.g., unsubstituted C1-C20 alkyl, C1-C15 alkyl, C1-C10 alkyl, or C1-C3 alkyl). In embodiments, R’ independently is unsubstituted C1-C3 alkyl. In embodiments, the alkynyl is unsubstituted. In embodiments, the alkynyl is substituted (e.g., with 1, 2, 3, 4, 5, or 6 substituent groups as described herein). [0087] Cycloalkyl: As used herein, the term“cycloalkyl” means a nonaromatic, saturated, cyclic group, e.g.“C3-C10 cycloalkyl.” In embodiments, a cycloalkyl is monocyclic. In embodiments, a cycloalkyl is polycyclic (e.g., bicyclic or tricyclic). In polycyclic cycloalkyl groups, individual rings can be fused, bridged, or spirocyclic. Examples of a cycloalkyl group include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, norbornanyl, bicyclo[3.2.1]octanyl, octahydro-pentalenyl, and spiro[4.5]decanyl, and the like. The term“cycloalkyl” may be used interchangeably with the term“carbocycle”. A cycloalkyl group may be unsubstituted or substituted with one or more substituent groups as described herein. For example, a cycloalkyl group may be substituted with one or more (e.g., 1, 2, 3, 4, 5, or 6 independently selected substituents) of halogen, -COR’, -CO2H, -CO2R’, -CN, -OH, -OR’, -OCOR’, -OCO2R’, -NH2, -NHR’, -N(R’)2, -SR’ or-SO2R’, wherein each instance of R’ independently is C1-C20 aliphatic (e.g., C1-C20 alkyl, C1-C15 alkyl, C1-C10 alkyl, or C1-C3 alkyl). In embodiments, R’ independently is an unsubstituted alkyl (e.g., unsubstituted C1-C20 alkyl, C1-C15 alkyl, C1-C10 alkyl, or C1-C3 alkyl). In embodiments, R’ independently is unsubstituted C1-C3 alkyl. In embodiments, the cycloalkyl is unsubstituted. In embodiments, the cycloalkyl is substituted (e.g., with 1, 2, 3, 4, 5, or 6 substituent groups as described herein). [0088] Halogen: As used herein, the term“halogen” means fluorine, chlorine, bromine, or iodine. Cationic Lipids
[0089] Liposomal-based vehicles are considered an attractive carrier for therapeutic agents and remain subject to continued development efforts. While liposomal-based vehicles that comprise a cationic lipid component have shown promising results with regards to encapsulation, stability and site localization, there remains a great need for improvement of liposomal-based delivery systems. For example, a significant drawback of liposomal delivery systems relates to the construction of liposomes that have sufficient cell culture or in vivo stability to reach desired target cells and/or intracellular compartments, and the ability of such liposomal delivery systems to efficiently release their encapsulated materials to such target cells. [0090] In particular, there remains a need for improved cationic lipids that demonstrate improved pharmacokinetic properties and which are capable of delivering macromolecules, such as nucleic acids to a wide variety cell types and tissues with enhanced efficiency. Importantly, there also remains a particular need for novel cationic lipids that are characterized as having reduced toxicity and are capable of efficiently delivering encapsulated nucleic acids and polynucleotides to targeted cells, tissues and organs. [0091] Described herein are novel cationic lipids, compositions comprising such lipids, and related methods of their use. In embodiments, the compounds described herein are useful as liposomal compositions or as components of liposomal compositions to facilitate the delivery to, and subsequent transfection of one or more target cells. [0092] Cationic lipids disclosed herein comprise a basic, ionizable functional group (e.g., an amine or a nitrogen-containing heteroaryl as described herein), which is present in neutral or charged form. [0093] In embodiments, cationic lipids described herein can provide one or more desired
characteristics or properties. That is, in certain embodiments, cationic lipids described herein can be characterized as having one or more properties that afford such compounds advantages relative to other similarly classified lipids. For example, cationic lipids disclosed herein can allow for the control and tailoring of the properties of liposomal compositions (e.g., lipid nanoparticles) of which they are a component. In particular, cationic lipids disclosed herein can be characterized by enhanced transfection efficiencies and their ability to provoke specific biological outcomes. Such outcomes can include, for example enhanced cellular uptake, endosomal/lysosomal disruption capabilities and/or promoting the release of encapsulated materials (e.g., polynucleotides) intracellularly. Cationic Lipids of Formula (I), (II), (III), (IV), (V), and (VI)
[0094] In one aspect, the present invention provides a cationic lipid of Formula (I):
Figure imgf000026_0001
,
wherein
R1 is hydrogen, C6-C30 alkyl, C6-C30 alkenyl, or C6-C30 alkynyl, Substructure Y, or
Substructure Z;
each a and b is an integer of 0-6;
each XA1 and XB1 is independently O or S;
each LA and LB is independently C1-C10 alkylene; C2-C10 alkenylene; or C2-C10 alkynylene; XA2 is independently NH, NRA, CH2, or CHRA;
XB2 is independently NH, NRB, CH2, or CHRB;
each RA and RB is independently C6-C30 alkyl, C6-C30 alkenyl, or C6-C30 alkynyl; and
Substructure
Figure imgf000026_0002
.
[0095] In embodiments, R1 is hydrogen. In embodiments, R1 is C6-C30 alkyl. In embodiments, R1 is C6-C30 alkenyl. In embodiments, R1 is C6-C30 alkynyl. In embodiments, R1 is Substructure Y. [0096] In one aspect, the present invention provides a cationic lipid of Formula (II):
Figure imgf000026_0003
wherein
R1 is hydrogen, C6-C30 alkyl, C6-C30 alkenyl, or C6-C30 alkynyl, or Substructure Z;
each a and b is an integer of 0-6;
each XA1 and XB1 is independently O or S;
each LA and LB is independently C1-C10 alkylene; C2-C10 alkenylene; or C2-C10 alkynylene; XA2 is independently NH, NRA, CH2, or CHRA; XB2 is independently NH, NRB, CH2, or CHRB;
each RA and RB is independently C6-C30 alkyl, C6-C30 alkenyl, or C6-C30 alkynyl; and
Substructure Z is
Figure imgf000027_0002
[0097] In embodiments, R1 is hydrogen. In embodiments, R1 is C6-C30 alkyl. In embodiments, R1 is C6-C30 alkenyl. In embodiments, R1 is C6-C30 alkynyl. In embodiments, R1 is Substructure Z. [0098] In another aspect, the present invention provides a cationic lipid of Formula (III):
Figure imgf000027_0003
wherein
each a is an integer of 0-6;
each XA1 is independently O or S;
each LA is independently C1-C10 alkylene; C2-C10 alkenylene; or C2-C10 alkynylene;
XA2 is independently NH, NRA, CH2, or CHRA; and
each RA is independently C6-C30 alkyl, C6-C30 alkenyl, or C6-C30 alkynyl. [0099] In embodiments, a cationic lipid has a structure according to Formula (IV),
Figure imgf000027_0001
, wherein
R1 is hydrogen, C1-C30 alkyl, C2-C30 alkenyl, or C2-C30 alkynyl;
each a and b is an integer of 0-6;
each XA1 and XB1 is independently O or S;
each LA and LB is independently C1-C10 alkylene; C2-C10 alkenylene; or C2-C10 alkynylene; LC is independently–C(O)– or–(CH2)b–; XA2 is independently NH, NRA, CH2, or CHRA;
XB2 is independently NH, NRB, CH2, or CHRB; and
each RA and RB is independently C6-C30 alkyl, C6-C30 alkenyl, or C6-C30 alkynyl. [0100] In another aspect, the present invention provides a cationic lipid has a structure according to Formula (V),
Figure imgf000028_0001
wherein
each LA and LB is independently C1-C10 alkylene; C2-C10 alkenylene; C2-C10 alkynylene; and each RA and RB is independently C6-C30 alkyl, C6-C30 alkenyl, C6-C30 alkynyl, or , or C1-C15 alkylene-C(O)2- C1-C15 alkylene.
[0101] In embodiments, a cationic lipid has a structure according to Formula (I-A),
Figure imgf000028_0002
[0102] In embodiments, a cationic lipid has a structure according to Formula (I-A’),
Figure imgf000028_0003
[0103] In embodiments, a cationic lipid has a structure according to Formula (II-A),
Figure imgf000029_0001
[0104] In embodiments, a cationic lipid has a structure according to Formula (IV-A):
Figure imgf000029_0002
[0105] In embodiments, each XA1 and XB1 is O, or each XA1 and XB1 is S. In embodiments, each XA1 and XB1 is O. In embodiments, each XA1 and XB1 is S. [0106] In embodiments, each a and b is independently 0, 1, or 2. In embodiments, each a is 0. In
embodiments, each a is 1. In embodiments, each a is 2. In embodiments, each b is independently 0. In embodiments, each b is 1. In embodiments, each b is 2. In embodiments, a is the same as b. In embodiments, a differs from b. In embodiments, each a and b is 0. In embodiments, each a and b is 1. In embodiments, each a and b is 2. In embodiments, each a is 0 and each b is 1. In
embodiments, each a is 0 and each b is 2. In embodiments, each a is 1 and each b is 0. In embodiments, each a is 1 and each b is 2. In embodiments, each a is 2 and each b is 0. In embodiments, each a is 2 and each b is 1. [0107] In embodiments, each XA2 is NRA or CHRA. In embodiments, each XA2 is NRA. In embodiments, each XA2 is CHRA. [0108] In embodiments, each XB2 is NRB or CHRB. In embodiments, each XB2 is NRB. In embodiments, each XB2 is CHRB. [0109] In embodiments, each XA2 is NRA, and each XB2 is NRB. In embodiments, each XA2 is CHRA, and each XB2 is CHRB. [0110] In embodiments, a cationic lipid has a structure according to Formula (I-B),
Figure imgf000030_0001
[0111] In embodiments, a cationic lipid has a structure according to Formula (I-B’),
Figure imgf000030_0002
[0112] In embodiments, a cationic lipid has a structure according to Formula (I-B”),
Figure imgf000030_0003
wherein d is independently an integer of 0-5. In embodiments, d is 0. In embodiments, d is 1. In embodiments, d is 2. In embodiments, d is 3. In embodiments, d is 4. In embodiments, d is 5. [0113] In embodiments, a cationic lipid has a structure according to Formula (II-A),
Figure imgf000030_0004
[0114] In embodiments, a cationic lipid has a structure according to Formula (III-A),
Figure imgf000031_0001
[0115] In embodiments, a cationic lipid has a structure according to Formula (IV-B),
Figure imgf000031_0002
[0116] In embodiments, a cationic lipid has a structure according to Formula (IV-B’),
Figure imgf000031_0003
[0117] In embodiments, a cationic lipid has a structure according to Formula (IV-B”),
Figure imgf000031_0004
[0118] In embodiments, each LA is C1-C10 alkylene (e.g., C2-C10 alkylene, C4-C10 alkylene, or C4-C8
alkylene). In embodiments, each LA is unsubstituted C1-C10 alkylene (e.g., unsubstituted C2-C10 alkylene, unsubstituted C4-C10 alkylene, or unsubstituted C4-C8 alkylene). In embodiments, each LA is substituted C1-C10 alkylene (e.g., substituted C2-C10 alkylene, substituted C4-C10 alkylene, or substituted C4-C8 alkylene). In embodiments, each LA is -(CH2)4-. In embodiments, each LA is -(CH2)2-. In embodiments, each LA is -(CH2)3-. In embodiments, each LA is -(CH2)5-. [0119] In embodiments, each RA and RB is unsubstituted C6-C30 alkenyl. In embodiments, each RA and RB is (unsubstituted C3-C15 alkylene)-C(O)2-(unsubstituted C3-C15 alkyl). [0120] In embodiments, each LB is C1-C10 alkylene (e.g., C2-C10 alkylene, C4-C10 alkylene, or C4-C8
alkylene). In embodiments, each LB is unsubstituted C1-C10 alkylene (e.g., unsubstituted C2-C10 alkylene, unsubstituted C4-C10 alkylene, or unsubstituted C4-C8 alkylene). In embodiments, each LB is substituted C1-C10 alkylene (e.g., substituted C2-C10 alkylene, substituted C4-C10 alkylene, or substituted C4-C8 alkylene). In embodiments, each LB is -(CH2)4-. In embodiments, each LB is -(CH2)2-. In embodiments, each LB is -(CH2)3-. In embodiments, each LB is -(CH2)5-. [0121] In embodiments, each LA and LB is C1-C10 alkylene. In embodiments, each LA and LB is substituted C1-C10 alkylene. In embodiments, each LA and LB is unsubstituted C1-C10 alkylene. In embodiments, each LA and LB is -CH2-. In embodiments, each LA and LB is -(CH2)2-. In embodiments, each LA and LB is -(CH2)3-. In embodiments, each LA and LB is -(CH2)4-. In embodiments, each LA and LB is -(CH2)5-. In embodiments, each LA and LB is -(CH2)6-. In embodiments, each LA and LB is -(CH2)7-. In
embodiments, each LA and LB is -(CH2)8-. In embodiments, each LA and LB is -(CH2)9-. In
embodiments, each LA and LB is -(CH2)10-.
[0122] In embodiments, a cationic lipid has a structure according to Formula (I-C),
Figure imgf000032_0001
wherein each c is independently an integer of 2-10. [0123] In embodiments, a cationic lipid has a structure according to Formula (I-C’),
Figure imgf000032_0002
wherein each c is independently an integer of 2-10. [0124] In embodiments, a cationic lipid has a structure according to Formula (I-C”),
Figure imgf000033_0001
, wherein each c is independently an integer of 2-10, and d is independently an integer of 0-5. In embodiments, d is 0, 1, 2, 3, or 4.
[0125] In embodiments, a cationic lipid has a structure according to Formula (II-B),
Figure imgf000033_0002
wherein each c is independently an integer of 2-10.
[0126] In embodiments, a cationic lipid has a structure according to Formula (III-B),
Figure imgf000033_0003
wherein each c is independently an integer of 2-10.
[0127] In embodiments, a cationic lipid has a structure according to Formula (III-C),
Figure imgf000033_0004
wherein each c is independently an integer of 2-10. [0128] In embodiments, a cationic lipid has a structure according to Formula (IV-C’),
Figure imgf000034_0001
wherein each c is independently an integer of 2-10. [0129] In embodiments, a cationic lipid has a structure according to Formula (IV-C”),
Figure imgf000034_0002
, wherein each c is independently an integer of 2- 10. [0130] In another aspect, the present invention provides a cationic lipid has a structure according to Formula (VI),
Figure imgf000034_0003
,
wherein
each RA and RB is independently C6-C30 alkyl, C6-C30 alkenyl, C6-C30 alkynyl or C1-C15 alkylene-C(O)2- C1-C15 alkylene; and
each c is independently an integer of 2-10.
[0131] In embodiments, each c is 4, 5, 6, 7, 8, 9, or 10. In embodiments, each c is 2, 3, or 4. In
embodiments, each c is 2. In embodiments, each c is 3. In embodiments, each c is 4. In
embodiments, each c is 5. In embodiments, each c is 6. In embodiments, each c is 7. In
embodiments, each c is 8. In embodiments, each c is 9. In embodiments, each c is 10. [0132] In embodiments, each RA and RB is unsubstituted C6-C30 alkenyl. In embodiments, each RA and RB is (unsubstituted C3-C15 alkylene)-C(O)2-(unsubstituted C3-C15 alkyl). [0133] In embodiments, each RA is C6-C30 alkyl (e.g., C6-C20 alkyl) or a C6-C30 alkenyl (e.g., C6-C20 alkenyl).
In embodiments, each RA is unsubstituted C6-C30 alkyl, C6-C30 hydroxyalkyl, unsubstituted C6-C30 alkenyl, or C6-C30 hydroxyalkenyl. In embodiments, each RA is unsubstituted C6-C20 alkyl, C6-C20 hydroxyalkyl, unsubstituted C6-C20 alkenyl, or C6-C20 hydroxyalkenyl. [0134] In embodiments, each RA is C6-C30 alkyl. In embodiments, each RA is unsubstituted C6-C30 alkyl.
In embodiments, each RA is substituted C6-C30 alkyl. In embodiments, each RA is C6-C30 hydroxyalkyl. In embodiments, each RA is a linear C6-C30 alkyl. In embodiments, each RA is a branched C6-C30 alkyl. [0135] In embodiments, each RA is C6-C20 alkyl. In embodiments, each RA is unsubstituted C6-C20 alkyl.
In embodiments, each RA is substituted C6-C20 alkyl. In embodiments, each RA is C6-C20 hydroxyalkyl. In embodiments, each RA is a linear C6-C20 alkyl. In embodiments, each RA is a branched C6-C20 alkyl. [0136] In embodiments, each RA is C6-C30 alkenyl. In embodiments, each RA is unsubstituted C6-C30 alkenyl. In embodiments, each RA is substituted C6-C30 alkenyl. In embodiments, each RA is C6-C30 hydroxyalkenyl. In embodiments, each RA is a linear C6-C30 alkenyl. In embodiments, each RA is a branched C6-C30 alkenyl. In embodiments, a C6-C30 alkenyl is a monoalkenyl, a dienyl, or a trienyl. [0137] In embodiments, each RA is C6-C20 alkenyl. In embodiments, each RA is unsubstituted C6-C20 alkenyl. In embodiments, each RA is substituted C6-C20 alkenyl. In embodiments, each RA is C6-C20 hydroxyalkenyl. In embodiments, each RA is a linear C6-C20 alkenyl. In embodiments, each RA is a branched C6-C20 alkenyl. In embodiments, a C6-C20 alkenyl is a monoalkenyl, a dienyl, or a trienyl. [0138] In embodiments, each RA is C4-C10 alkylene-C(O)2- C4-C10 alkyl. In embodiments, each RA is unsubstituted C4-C10 alkylene-C(O)2- C4-C10 alkyl. In embodiments, each RA is substituted C4-C10 alkylene-C(O)2- C4-C10 alkyl. In embodiments, each RA is C4-C10 alkylene-C(O)2- C4-C10 hydroxyalkyl. [0139] In embodiments, each RB is C6-C30 alkyl (e.g., C6-C20 alkyl) or a C6-C30 alkenyl (e.g., C6-C20 alkenyl).
In embodiments, each RB is unsubstituted C6-C30 alkyl, C6-C30 hydroxyalkyl, unsubstituted C6-C30 alkenyl, or C6-C30 hydroxyalkenyl. In embodiments, each RB is unsubstituted C6-C20 alkyl, C6-C20 hydroxyalkyl, unsubstituted C6-C20 alkenyl, or C6-C20 hydroxyalkenyl. [0140] In embodiments, each RB is C6-C20 alkyl. In embodiments, each RB is unsubstituted C6-C20 alkyl.
In embodiments, each RB is substituted C6-C20 alkyl. In embodiments, each RB is C6-C20 hydroxyalkyl. In embodiments, each RB is a linear C6-C20 alkyl. In embodiments, each RB is a branched C6-C20 alkyl. [0141] In embodiments, each RB is C6-C30 alkenyl. In embodiments, each RB is unsubstituted C6-C30 alkenyl. In embodiments, each RB is substituted C6-C30 alkenyl. In embodiments, each RB is C6-C30 hydroxyalkenyl. In embodiments, each RB is a linear C6-C30 alkenyl. In embodiments, each RB is a branched C6-C30 alkenyl. In embodiments, a C6-C30 alkenyl is a monoalkenyl, a dienyl, or a trienyl. [0142] In embodiments, each RB is C6-C20 alkenyl. In embodiments, each RB is unsubstituted C6-C20 alkenyl. In embodiments, each RB is substituted C6-C20 alkenyl. In embodiments, each RB is C6-C20 hydroxyalkenyl. In embodiments, each RB is a linear C6-C20 alkenyl. In embodiments, each RB is a branched C6-C20 alkenyl. In embodiments, a C6-C20 alkenyl is a monoalkenyl, a dienyl, or a trienyl. [0143] In embodiments, each RB is C4-C10 alkylene-C(O)2- C4-C10 alkyl. In embodiments, each RB is
unsubstituted C4-C10 alkylene-C(O)2- C4-C10 alkyl. In embodiments, each RB is substituted C4-C10 alkylene-C(O)2- C4-C10 alkyl. In embodiments, each RB is C4-C10 alkylene-C(O)2- C4-C10 hydroxyalkyl. [0144] In embodiments, RA is the same as RB. In embodiments RA differs from RB. [0145] In embodiments, each RA and RB is C6-C30 alkyl. In embodiments, each RA and RB is unsubstituted C6-C30 alkyl. In embodiments, each RA and RB is substituted C6-C30 alkyl. In embodiments, each RA and RB is C6-C30 hydroxyalkyl. In embodiments, each RA and RB is a linear C6-C30 alkyl. In
embodiments, each RA and RB is a branched C6-C30 alkyl. [0146] In embodiments, each RA and RB is C6-C20 alkyl. In embodiments, each RA and RB is unsubstituted C6-C20 alkyl. In embodiments, each RA and RB is substituted C6-C20 alkyl. In embodiments, each RA and RB is C6-C20 hydroxyalkyl. In embodiments, each RA and RB is a linear C6-C20 alkyl. In
embodiments, each RA and RB is a branched C6-C20 alkyl. [0147] In embodiments, each RA and RB is C6-C30 alkenyl. In embodiments, each RA and RB is
unsubstituted C6-C30 alkenyl. In embodiments, each RA and RB is substituted C6-C30 alkenyl. In embodiments, each RA and RB is C6-C30 hydroxyalkenyl. In embodiments, each RA and RB is a linear C6-C30 alkenyl. In embodiments, each RA and RB is a branched C6-C30 alkenyl. In embodiments, a C6- C30 alkenyl is a monoalkenyl, a dienyl, or a trienyl. [0148] In embodiments, each RA and RB is C6-C20 alkenyl. In embodiments, each RA and RB is
unsubstituted C6-C20 alkenyl. In embodiments, each RA and RB is substituted C6-C20 alkenyl. In embodiments, each RA and RB is C6-C20 hydroxyalkenyl. In embodiments, each RA and RB is a linear C6-C20 alkenyl. In embodiments, each RA and RB is a branched C6-C20 alkenyl. In embodiments, a C6- C20 alkenyl is a monoalkenyl, a dienyl, or a trienyl. [0149] In embodiments, each RA and RB is C6-C30 hydroxyalkyl, or each RA and RB is C6-C30
hydroxyalkenyl. In embodiments, each RA and RB is C6-C20 hydroxyalkyl, or each RA and RB is C6-C20 hydroxyalkenyl. [0150] In embodiments, each RA and RB is C4-C10 alkylene-C(O)2- C4-C10 alkyl. In embodiments, each RA and RB is unsubstituted C4-C10 alkylene-C(O)2- C4-C10 alkyl. In embodiments, each RA and RB is substituted C4-C10 alkylene-C(O)2- C4-C10 alkyl. In embodiments, each RA and RB is C4-C10 alkylene- C(O)2- C4-C10 hydroxyalkyl. [0151] In embodiments, each RA and RB is C6-C30 hydroxyalkyl. In embodiments, each RA and RB is C6-C20 hydroxyalkyl. In embodiments, each RA and RB is -CH2CH(OH)C10H21. In embodiments, each RA and RB is -CH2CH(OH)C8H17. In embodiments, each RA and RB is -CH2CH(OH)C12H25. In embodiments, each RA and RB is -CH2CH(OH)C14H29. In embodiments, each RA and RB is -CH2CH(OH)C16H33. In embodiments, each RA and RB is -CH2CH(OH)C18H37. [0152] In embodiments, each RA and RB is C6-C30 hydroxyalkenyl. In embodiments, each RA and RB is C6- C20 hydroxyalkenyl. In embodiments, each RA and RB is -CH2CH(OH)(CH2)6(CH=CH)CH2(CH=CH)C5H11. In embodiments, each RA and RB is -CH2CH(OH)(CH2)6(CH=CH)C8H17. In embodiments, each RA and RB is -CH2CH(OH)(CH2)6(CH=CH)CH2(CH=CH)CH2(CH=CH)C2H5. [0153] In embodiments, each RA and RB is C4-C10 alkylene-C(O)2- C4-C10 hydroxyalkyl. In embodiments, each RA and RB is -CH2CH(OH)(CH2)7C(O)2C7H15. [0154] In embodiments, each RA and RB is -CH2CH(OH)RC, and wherein RC is selected from the group consisting of:
Figure imgf000037_0001
.
[0155] In embodiments, a C6-C30 alkyl (e.g., each RA and/or each RB) is a C8-26 alkyl. In embodiments, a C6-C30 alkyl (e.g., each RA and/or each RB) is a straight-chain C8-26 alkyl. [0156] In embodiments, a C6-C30 alkyl (e.g., each RA and/or each RB) is CH3(CH2)6CH2-, CH3(CH2)7CH2-, CH3(CH2)8CH2-, CH3(CH2)9CH2-, CH3(CH2)10CH2-, CH3(CH2)11CH2-, CH3(CH2)12CH2-, CH3(CH2)13CH2-, CH3(CH2)14CH2-, CH3(CH2)15CH2-, CH3(CH2)16CH2-, CH3(CH2)17CH2-, CH3(CH2)18CH2-, CH3(CH2)19CH2-, CH3(CH2)20CH2-, CH3(CH2)21CH2-, CH3(CH2)22CH2-, CH3(CH2)23CH2- or CH3(CH2)24CH2-. [0157] In embodiments, a C6-C30 alkyl (e.g., each RA and/or each RB) is CH3(CH2)13CH2-, CH3(CH2)14CH2-, CH3(CH2)15CH2-, CH3(CH2)16CH2-, CH3(CH2)17CH2- or CH3(CH2)18CH2-. [0158] In embodiments, a C6-C30 alkyl (e.g., each RA and/or each RB) is CH3(CH2)14CH2-, CH3(CH2)15CH2- or CH3(CH2)16CH2-. [0159] In embodiments, a C6-C30 alkenyl (e.g., each RA and/or each RB) is a C8-26 alkenyl having one or two carbon-carbon double bonds. [0160] In embodiments, a C6-C30 alkenyl (e.g., each RA and/or each RB) is cis-CH3(CH2)3CH=CH(CH2)7CH2-, cis-CH3(CH2)5CH=CH(CH2)7CH2-, cis-CH3(CH2)8CH=CH(CH2)4CH2-, cis-CH3(CH2)7CH=CH(CH2)7CH2-, cis- CH3(CH2)9CH=CH(CH2)7CH2-, cis-CH3(CH2)7CH=CH(CH2)9CH2-, trans-CH3(CH2)7CH=CH(CH2)7CH2-, trans-CH3(CH2)5CH=CH(CH2)9CH2-, cis-CH3(CH2)9CH=CH(CH2)7CH2-, cis-CH3(CH2)7CH=CH(CH2)11CH2-, cis- CH3(CH2)7CH=CH(CH2)13CH2-, cis,cis-CH3(CH2)4CH=CHCH2CH=CH(CH2)7CH2-,
cis,cis-CH3(CH2)4CH=CHCH2CH=CH(CH2)9CH2- or cis,cis-CH3(CH2)4CH=CHCH2CH=CH(CH2)11CH2-. [0161] In embodiments, a C6-C30 alkenyl (e.g., each RA and/or each RB) is cis-CH3(CH2)3CH=CH(CH2)7CH2-, cis-CH3(CH2)5CH=CH(CH2)7CH2-, cis-CH3(CH2)8CH=CH(CH2)4CH2-, cis-CH3(CH2)7CH=CH(CH2)7CH2-, cis- CH3(CH2)9CH=CH(CH2)7CH2-, trans-CH3(CH2)7CH=CH(CH2)7CH2-,
cis,cis-CH3(CH2)4CH=CHCH2CH=CH(CH2)7CH2- or cis,cis-CH3(CH2)4CH=CHCH2CH=CH(CH2)9CH2-. [0162] In embodiments, a C6-C30 alkenyl (e.g., each RA and/or each RB) is cis-CH3(CH2)7CH=CH(CH2)7CH2-, cis-CH3(CH2)9CH=CH(CH2)7CH2-, cis,cis-CH3(CH2)4CH=CHCH2CH=CH(CH2)7CH2- or
cis,cis-CH3(CH2)4CH=CHCH2CH=CH(CH2)9CH2-. [0163] In embodiments, a C6-C30 alkenyl (e.g., each RA and/or each RB) is cis-CH3(CH2)7CH=CH(CH2)7CH2- or cis,cis-CH3(CH2)4CH=CH-CH2CH=CH(CH2)7CH2-. [0164] In embodiments, a C6-C30 alkenyl (e.g., each RA and/or each RB) is C8-26 aliphatic having three, four, five or six carbon-carbon double bonds. [0165] In embodiments, a C6-C30 alkenyl (e.g., each RA and/or each RB) is
cis,cis,cis-CH3CH2CH=CHCH2CH=CHCH2CH=CH(CH2)7CH2-,
cis,cis,cis-CH3(CH2)4CH=CHCH2CH=CHCH2CH=CH(CH2)4CH2-, cis,cis,cis-CH3(CH2)4CH=CHCH2CH=CHCH2CH2CH=CH(CH2)3CH2-,
trans,trans,trans-CH3(CH2)7CH=CHCH2CH=CHCH2CH=CH(CH2)3CH2-,
cis,cis,cis-CH3(CH2)4CH=CHCH2CH=CHCH2CH=CH(CH2)6CH2-,
cis,cis,cis-CH3CH2CH=CHCH2CH=CHCH2CH=CH(CH2)9CH2-,
cis,cis,cis,cis-CH3CH2CH=CHCH2CH=CHCH2CH=CHCH2CH=CH(CH2)4CH2-,
cis,cis,cis,cis-CH3(CH2)4CH=CHCH2CH=CHCH2CH=CHCH2CH=CH(CH2)3CH2-,
cis,cis,cis,cis-CH3CH2CH=CHCH2CH=CHCH2CH=CHCH2CH=CH(CH2)6CH2-,
cis,cis,trans,trans,cis-CH3(CH2)4CH=CHCH=CHCH=CHCH=CHCH2CH=CH(CH2)3CH2-,
cis,cis,cis,cis,cis-CH3CH2CH=CHCH2CH=CHCH2CH=CHCH2CH=CHCH2CH=CH(CH2)3CH2-,
cis,cis,cis,cis,cis-CH3(CH2)4CH=CHCH2CH=CHCH2CH=CHCH2CH=CHCH2CH=CH(CH2)2CH2-,
cis,cis,cis,cis,cis-CH3CH2CH=CHCH2CH=CHCH2CH=CHCH2CH=CHCH2CH=CH(CH2)5CH2-,
cis,cis,cis,cis,cis-CH3CH2CH=CHCH2CH=CHCH2CH=CHCH2CH=CHCH2CH=CH(CH2)7CH2-,
cis,cis,cis,cis,cis,cis-CH3CH2CH=CHCH2CH=CHCH2CH=CHCH2CH=CHCH2CH=CHCH2CH=CH(CH2)2CH2-, or cis,cis,cis,cis,cis,cis- CH3CH2CH=CHCH2CH=CHCH2CH=CHCH2CH=CHCH2CH=CHCH2CH=CH(CH2)4CH2-. [0166] In embodiments, a C6-C30 alkenyl (e.g., each RA and/or each RB) is
cis,cis,cis-CH3CH2CH=CHCH2CH=CHCH2CH=CH(CH2)7CH2-,
cis,cis,cis,cis-CH3CH2CH=CHCH2CH=CHCH2CH=CHCH2CH=CH(CH2)4CH2-,
cis,cis,trans,trans,cis-CH3(CH2)4CH=CHCH=CHCH=CHCH=CHCH2CH=CH(CH2)3CH2-, cis,cis,cis,cis,cis,cis- CH3CH2CH=CHCH2CH=CHCH2CH=CHCH2CH=CHCH2CH=CHCH2CH=CH(CH2)2CH2-. [0167] In embodiments, a C6-C30 alkenyl (e.g., each RA and/or each RB) is
cis,cis,cis-CH3CH2CH=CHCH2CH=CHCH2CH=CH(CH2)7CH2- or
cis,cis,cis,cis-CH3CH2CH=CHCH2CH=CHCH2CH=CHCH2CH=CH(CH2)4CH2-. [0168] In some embodiments, each RA and/or each RB independently is an aliphatic chain of a saturated or unsaturated fatty acid, i.e., R'-(CH2)- for a fatty acid R'-C(O)-. In some embodiments, each RA and/or each RB independently is the aliphatic chain of caprylic, pelargonic, capric, undecylic, lauric, tridecyclic, myristic, pentadecylic, margaric, stearic, nonadecylic, arachidic, heneicosylic, behenic, triosylic, lignoceric, oleic, linoleic, pentacosylic or cerotic acid. In some embodiments, each RA and/or each RB is the aliphatic chain of caprylic, pelargonic, capric, undecylic, lauric, tridecyclic, myristic, pentadecylic, or margaric acid. In some embodiments, each RA and/or each RB is the aliphatic chain of lauric, tridecyclic, myristic, or pentadecylic acid. In some embodiments, each RA and/or each RB is the aliphatic chain of lauric or myristic acid. In some embodiments, each RA and/or each RB is the aliphatic chain of stearic, nonadecylic, arachidic, heneicosylic, behenic, triosylic, lignoceric, oleic, linoleic, pentacosylic or cerotic acid. In some embodiments, each RA and/or each RB is the aliphatic chain of lignoceric, oleic, linoleic, pentacosylic or cerotic acid. In some embodiments, each RA and/or each RB is the aliphatic chain of oleic, linoleic or pentacosylic acid. In some embodiments, each RA and/or each RB is the aliphatic chain of oleic or linoleic acid. In some embodiments, each RA and/or each RB is the aliphatic chain of oleic acid. In some embodiments, each RA and/or each RB is the aliphatic chain of linoleic acid. Exemplary Cationic Lipids
[0169] Exemplary cationic lipids include cationic lipids such as Cationic Lipid (1), (2) (3), (4), (5), and (6),
Figure imgf000040_0001
Figure imgf000041_0001
[0170] In embodiments, a cationic lipid is Compound (1). In embodiments, a cationic lipid is
Compound (2). In embodiments, a cationic lipid is Compound (3). In embodiments, a cationic lipid is Compound (4). In embodiments, a cationic lipid is Compound (5). In embodiments, a cationic lipid is Compound (6). [0171] The present invention also provides a cationic lipid that is:
Figure imgf000042_0001
Figure imgf000042_0002
[0172] In embodiments, a cationic lipid is Compound (7). In embodiments, a cationic lipid is
Compound (8). In embodiments, a cationic lipid is Compound (9). In embodiments, a cationic lipid is Compound (10). In embodiments, a cationic lipid is Compound (11). In embodiments, a cationic lipid is Compound (12). In embodiments, a cationic lipid is Compound (13). In embodiments, a cationic lipid is Compound (14). In embodiments, a cationic lipid is Compound (15). In embodiments, a cationic lipid is Compound (16). [0173] In embodiments, a cationic lipid described herein can be obtained from combination of any of the diacids in Table A and the thiols of Table B, including of the following compounds as described herein.
Figure imgf000043_0001
Figure imgf000044_0001
Figure imgf000045_0001
Figure imgf000046_0001
Figure imgf000047_0001
Figure imgf000048_0001
Figure imgf000049_0001
Figure imgf000050_0001
Figure imgf000051_0001
Figure imgf000052_0001
Figure imgf000053_0001
Figure imgf000054_0001
Figure imgf000055_0001
Figure imgf000056_0001
Figure imgf000057_0001
Figure imgf000058_0001
Figure imgf000059_0001
Figure imgf000060_0001
Figure imgf000061_0001
Figure imgf000062_0001
Figure imgf000063_0001
Figure imgf000064_0001
[0174] In embodiments, a cationic lipid is Compound (17). In embodiments, a cationic lipid is
Compound (18). In embodiments, a cationic lipid is Compound (19). In embodiments, a cationic lipid is Compound (20). In embodiments, a cationic lipid is Compound (21). In embodiments, a cationic lipid is Compound (22). In embodiments, a cationic lipid is Compound (23). In
embodiments, a cationic lipid is Compound (24). In embodiments, a cationic lipid is Compound (25). In embodiments, a cationic lipid is Compound (26). In embodiments, a cationic lipid is Compound (27). In embodiments, a cationic lipid is Compound (28). In embodiments, a cationic lipid is Compound (29). In embodiments, a cationic lipid is Compound (30). In embodiments, a cationic lipid is Compound (31). In embodiments, a cationic lipid is Compound (32). In embodiments, a cationic lipid is Compound (33). In embodiments, a cationic lipid is Compound (34). In
embodiments, a cationic lipid is Compound (35). In embodiments, a cationic lipid is Compound (36). In embodiments, a cationic lipid is Compound (37). In embodiments, a cationic lipid is Compound (38). In embodiments, a cationic lipid is Compound (39). In embodiments, a cationic lipid is Compound (40). In embodiments, a cationic lipid is Compound (41). In embodiments, a cationic lipid is Compound (42). In embodiments, a cationic lipid is Compound (43). In embodiments, a cationic lipid is Compound (44). In embodiments, a cationic lipid is Compound (45). In
embodiments, a cationic lipid is Compound (46). [0175] In embodiments, a cationic lipid is Compound (7). In embodiments, a cationic lipid is
Compound (8). In embodiments, a cationic lipid is Compound (9). In embodiments, a cationic lipid is Compound (10). In embodiments, a cationic lipid is Compound (11). In embodiments, a cationic lipid is Compound (12). In embodiments, a cationic lipid is Compound (13). In embodiments, a cationic lipid is Compound (14). In embodiments, a cationic lipid is Compound (15). In
embodiments, a cationic lipid is Compound (16). In embodiments, a cationic lipid is Compound (47). In embodiments, a cationic lipid is Compound (48). In embodiments, a cationic lipid is Compound (49). In embodiments, a cationic lipid is Compound (50). In embodiments, a cationic lipid is Compound (51). In embodiments, a cationic lipid is Compound (52). In embodiments, a cationic lipid is Compound (53). In embodiments, a cationic lipid is Compound (54). In embodiments, a cationic lipid is Compound (55). In embodiments, a cationic lipid is Compound (56). In
embodiments, a cationic lipid is Compound (57). In embodiments, a cationic lipid is Compound (58). In embodiments, a cationic lipid is Compound (59). In embodiments, a cationic lipid is Compound (60). In embodiments, a cationic lipid is Compound (61). In embodiments, a cationic lipid is Compound (62). In embodiments, a cationic lipid is Compound (63). In embodiments, a cationic lipid is Compound (64). In embodiments, a cationic lipid is Compound (65). In embodiments, a cationic lipid is Compound (66). [0176] In embodiments, a cationic lipid is Compound (67). In embodiments, a cationic lipid is
Compound (68). In embodiments, a cationic lipid is Compound (69). In embodiments, a cationic lipid is Compound (70). In embodiments, a cationic lipid is Compound (71). In embodiments, a cationic lipid is Compound (72). In embodiments, a cationic lipid is Compound (73). In
embodiments, a cationic lipid is Compound (74). In embodiments, a cationic lipid is Compound (75). In embodiments, a cationic lipid is Compound (76). In embodiments, a cationic lipid is Compound (77). In embodiments, a cationic lipid is Compound (78). In embodiments, a cationic lipid is Compound (79). In embodiments, a cationic lipid is Compound (80). In embodiments, a cationic lipid is Compound (81). In embodiments, a cationic lipid is Compound (82). In embodiments, a cationic lipid is Compound (83). In embodiments, a cationic lipid is Compound (84). In
embodiments, a cationic lipid is Compound (85). In embodiments, a cationic lipid is Compound (86). In embodiments, a cationic lipid is Compound (87). In embodiments, a cationic lipid is Compound (88). In embodiments, a cationic lipid is Compound (89). In embodiments, a cationic lipid is Compound (90). In embodiments, a cationic lipid is Compound (91). In embodiments, a cationic lipid is Compound (92). In embodiments, a cationic lipid is Compound (93). In embodiments, a cationic lipid is Compound (94). In embodiments, a cationic lipid is Compound (95). In
embodiments, a cationic lipid is Compound (96). [0177] In embodiments, a cationic lipid is Compound (97). In embodiments, a cationic lipid is
Compound (98). In embodiments, a cationic lipid is Compound (99). In embodiments, a cationic lipid is Compound (100). In embodiments, a cationic lipid is Compound (101). In embodiments, a cationic lipid is Compound (102). In embodiments, a cationic lipid is Compound (103). In embodiments, a cationic lipid is Compound (104). In embodiments, a cationic lipid is Compound (105). In embodiments, a cationic lipid is Compound (106). In embodiments, a cationic lipid is Compound (107). In embodiments, a cationic lipid is Compound (108). In embodiments, a cationic lipid is Compound (109). In embodiments, a cationic lipid is Compound (110). In embodiments, a cationic lipid is Compound (111). In embodiments, a cationic lipid is Compound (112). In embodiments, a cationic lipid is Compound (113). In embodiments, a cationic lipid is Compound (114). In embodiments, a cationic lipid is Compound (115). In embodiments, a cationic lipid is Compound (116). In embodiments, a cationic lipid is Compound (117). In embodiments, a cationic lipid is Compound (118). In embodiments, a cationic lipid is Compound (119). In embodiments, a cationic lipid is Compound (120). In embodiments, a cationic lipid is Compound (121). In embodiments, a cationic lipid is Compound (122). In embodiments, a cationic lipid is Compound (123). In embodiments, a cationic lipid is Compound (124). In embodiments, a cationic lipid is Compound (125). In embodiments, a cationic lipid is Compound (126). [0178] In embodiments, a cationic lipid is Compound (127). In embodiments, a cationic lipid is Compound (128). In embodiments, a cationic lipid is Compound (129). In embodiments, a cationic lipid is Compound (130). In embodiments, a cationic lipid is Compound (131). In embodiments, a cationic lipid is Compound (132). In embodiments, a cationic lipid is Compound (133). In embodiments, a cationic lipid is Compound (134). In embodiments, a cationic lipid is Compound (135). In embodiments, a cationic lipid is Compound (136). In embodiments, a cationic lipid is Compound (137). In embodiments, a cationic lipid is Compound (138). In embodiments, a cationic lipid is Compound (139). In embodiments, a cationic lipid is Compound (140). In embodiments, a cationic lipid is Compound (141). In embodiments, a cationic lipid is Compound (142). In embodiments, a cationic lipid is Compound (143). In embodiments, a cationic lipid is Compound (144). In embodiments, a cationic lipid is Compound (145). In embodiments, a cationic lipid is Compound (146). In embodiments, a cationic lipid is Compound (147). In embodiments, a cationic lipid is Compound (148). In embodiments, a cationic lipid is Compound (149). In embodiments, a cationic lipid is Compound (150). In embodiments, a cationic lipid is Compound (151). In embodiments, a cationic lipid is Compound (152). In embodiments, a cationic lipid is Compound (153). In embodiments, a cationic lipid is Compound (154). In embodiments, a cationic lipid is Compound (155). In embodiments, a cationic lipid is Compound (156).
Synthesis of Cationic Lipids
[0179] Cationic lipids described herein (e.g., a cationic lipid of any of Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III-B), (IV-A), (IV-B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)-(156)).can be prepared according to methods known in the art. For example, cationic lipids of the present invention may be prepared via thioesterification of a starting carboxylic acid followed by deprotection.
[0180] In some embodiments, the cationic lipids described (e.g., Cationic Lipid (1) herein may be prepared according to Scheme 1.
Scheme 1
Figure imgf000068_0001
[0181] For example, an alkylating agent such as Compound A can be treated with a thiol reagent such as benzyl mercaptan to afford a bis-protected difunctional intermediate such as Compound B. Deprotection of the phthalimide moiety of Compound B affords the nucleophilic Compound C, which in turn can be treated with another electrophile such as an epoxide (Compound D) to provide a tertiary amine (Compound E). The hydroxyl groups of Compound E can be protected using protecting groups and conditions known in the art (e.g., TBSCl/imidazole) to afford the
corresponding hydroxyl-protected Compound F. The–SH functionality can then be deprotected (e.g., using reducing conditions such as Na/NH3) to afford nucleophilic thiol Compound G. [0182] Thiols such as Compound G can be coupled to poly-carboxylic acids (e.g., Compound H) to form thioester lipids (e.g., Compound J). When such lipids comprise protecting groups, deprotection (e.g., HF/pyridine when PG = TBS), can provide the cationic lipids described herein (e.g., Cationic Lipid 1). Nucleic Acids
[0183] Cationic lipids described herein (e.g., a cationic lipid of any of Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III-B), (IV-A), (IV-B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)-(156)) can be used to prepare compositions useful for the delivery of nucleic acids. Synthesis of Nucleic Acids
[0184] Nucleic acids according to the present invention may be synthesized according to any known methods. For example, mRNAs according to the present invention may be synthesized via in vitro transcription (IVT). Briefly, IVT is typically performed with a linear or circular DNA template containing a promoter, a pool of ribonucleotide triphosphates, a buffer system that may include DTT and magnesium ions, and an appropriate RNA polymerase (e.g., T3, T7, mutated T7 or SP6 RNA polymerase), DNAse I, pyrophosphatase, and/or RNAse inhibitor. The exact conditions will vary according to the specific application. [0185] In some embodiments, for the preparation of mRNA according to the invention, a DNA template is transcribed in vitro. A suitable DNA template typically has a promoter, for example a T3, T7, mutated T7 or SP6 promoter, for in vitro transcription, followed by desired nucleotide sequence for desired mRNA and a termination signal. [0186] Desired mRNA sequence(s) according to the invention may be determined and incorporated into a DNA template using standard methods. For example, starting from a desired amino acid sequence (e.g., an enzyme sequence), a virtual reverse translation is carried out based on the degenerated genetic code. Optimization algorithms may then be used for selection of suitable codons. Typically, the G/C content can be optimized to achieve the highest possible G/C content on one hand, taking into the best possible account the frequency of the tRNAs according to codon usage on the other hand. The optimized RNA sequence can be established and displayed, for example, with the aid of an appropriate display device and compared with the original (wild-type) sequence. A secondary structure can also be analyzed to calculate stabilizing and destabilizing properties or, respectively, regions of the RNA. [0187] As described above, the term“nucleic acid,” in its broadest sense, refers to any compound
and/or substance that is or can be incorporated into a polynucleotide chain. DNA may be in the form of antisense DNA, plasmid DNA, parts of a plasmid DNA, pre-condensed DNA, a product of a polymerase chain reaction (PCR), vectors (e.g., P1, PAC, BAC, YAC, artificial chromosomes), expression cassettes, chimeric sequences, chromosomal DNA, or derivatives of these groups. RNA may be in the form of messenger RNA (mRNA), ribosomal RNA (rRNA), signal recognition particle RNA (7 SL RNA or SRP RNA), transfer RNA (tRNA), transfer-messenger RNA (tmRNA), small nuclear RNA (snRNA), small nucleolar RNA (snoRNA), SmY RNA, small Cajal body-specific RNA (scaRNA), guide RNA (gRNA), ribonuclease P (RNase P), Y RNA, telomerase RNA component (TERC), spliced leader RNA (SL RNA), antisense RNA (aRNA or asRNA), cis-natural antisense transcript (cis-NAT), CRISPR RNA (crRNA), long noncoding RNA (lncRNA), microRNA (miRNA), piwi-interacting RNA (piRNA), small interfering RNA (siRNA), transacting siRNA (tasiRNA), repeat associated siRNA (rasiRNA), 73K RNA, retrotransposons, a viral genome, a viroid, satellite RNA, or derivatives of these groups. In some embodiments, a nucleic acid is a mRNA encoding a protein. Synthesis of mRNA
[0188] mRNAs according to the present invention may be synthesized according to any of a variety of known methods. For example, mRNAs according to the present invention may be synthesized via in vitro transcription (IVT). Briefly, IVT is typically performed with a linear or circular DNA template containing a promoter, a pool of ribonucleotide triphosphates, a buffer system that may include DTT and magnesium ions, and an appropriate RNA polymerase (e.g., T3, T7 or SP6 RNA polymerase), DNAse I, pyrophosphatase, and/or RNAse inhibitor. The exact conditions will vary according to the specific application. The exact conditions will vary according to the specific application. The presence of these reagents is undesirable in the final product according to several embodiments and may thus be referred to as impurities and a preparation containing one or more of these impurities may be referred to as an impure preparation. In some embodiments, the in vitro transcribing occurs in a single batch. [0189] In some embodiments, for the preparation of mRNA according to the invention, a DNA template is transcribed in vitro. A suitable DNA template typically has a promoter, for example a T3, T7 or SP6 promoter, for in vitro transcription, followed by desired nucleotide sequence for desired mRNA and a termination signal. [0190] Desired mRNA sequence(s) according to the invention may be determined and incorporated into a DNA template using standard methods. For example, starting from a desired amino acid sequence (e.g., an enzyme sequence), a virtual reverse translation is carried out based on the degenerated genetic code. Optimization algorithms may then be used for selection of suitable codons. Typically, the G/C content can be optimized to achieve the highest possible G/C content on one hand, taking into the best possible account the frequency of the tRNAs according to codon usage on the other hand. The optimized RNA sequence can be established and displayed, for example, with the aid of an appropriate display device and compared with the original (wild-type) sequence. A secondary structure can also be analyzed to calculate stabilizing and destabilizing properties or, respectively, regions of the RNA. Modified mRNA
[0191] In some embodiments, mRNA according to the present invention may be synthesized as
unmodified or modified mRNA. Modified mRNA comprise nucleotide modifications in the RNA. A modified mRNA according to the invention can thus include nucleotide modification that are, for example, backbone modifications, sugar modifications or base modifications. In some
embodiments, mRNAs may be synthesized from naturally occurring nucleotides and/or nucleotide analogues (modified nucleotides) including, but not limited to, purines (adenine (A), guanine (G)) or pyrimidines (thymine (T), cytosine (C), uracil (U)), and as modified nucleotides analogues or derivatives of purines and pyrimidines, such as e.g.1-methyl-adenine, 2-methyl-adenine, 2- methylthio-N-6-isopentenyl-adenine, N6-methyl-adenine, N6-isopentenyl-adenine, 2-thio-cytosine, 3-methyl-cytosine, 4-acetyl-cytosine, 5-methyl-cytosine, 2,6-diaminopurine, 1-methyl-guanine, 2- methyl-guanine, 2,2-dimethyl-guanine, 7-methyl-guanine, inosine, 1-methyl-inosine, pseudouracil (5-uracil), dihydro-uracil, 2-thio-uracil, 4-thio-uracil, 5-carboxymethylaminomethyl-2-thio-uracil, 5- (carboxyhydroxymethyl)-uracil, 5-fluoro-uracil, 5-bromo-uracil, 5-carboxymethylaminomethyl-uracil, 5-methyl-2-thio-uracil, 5-methyl-uracil, N-uracil-5-oxyacetic acid methyl ester, 5- methylaminomethyl-uracil, 5-methoxyaminomethyl-2-thio-uracil, 5'-methoxycarbonylmethyl-uracil, 5-methoxy-uracil, uracil-5-oxyacetic acid methyl ester, uracil-5-oxyacetic acid (v), 1-methyl- pseudouracil, queosine, .beta.-D-mannosyl-queosine, wybutoxosine, and phosphoramidates, phosphorothioates, peptide nucleotides, methylphosphonates, 7-deazaguanosine, 5-methylcytosine and inosine. The preparation of such analogues is known to a person skilled in the art e.g., from the U.S. Pat. No.4,373,071, U.S. Pat. No.4,401,796, U.S. Pat. No.4,415,732, U.S. Pat. No.4,458,066, U.S. Pat. No.4,500,707, U.S. Pat. No.4,668,777, U.S. Pat. No.4,973,679, U.S. Pat. No.5,047,524, U.S. Pat. No.5,132,418, U.S. Pat. No.5,153,319, U.S. Pat. Nos.5,262,530 and 5,700,642, the disclosures of which are incorporated by reference in their entirety. [0192] In some embodiments, mRNAs may contain RNA backbone modifications. Typically, a backbone modification is a modification in which the phosphates of the backbone of the nucleotides contained in the RNA are modified chemically. Exemplary backbone modifications typically include, but are not limited to, modifications from the group consisting of methylphosphonates,
methylphosphoramidates, phosphoramidates, phosphorothioates (e.g. cytidine 5'-O-(1- thiophosphate)), boranophosphates, positively charged guanidinium groups etc., which means by replacing the phosphodiester linkage by other anionic, cationic or neutral groups. [0193] In some embodiments, mRNAs may contain sugar modifications. A typical sugar modification is a chemical modification of the sugar of the nucleotides it contains including, but not limited to, sugar modifications chosen from the group consisting of 4'-thio-ribonucleotide (see, e.g., US Patent Application Publication No. US 2016/0031928, incorporated by reference herein), 2'-deoxy-2'- fluoro-oligoribonucleotide (2'-fluoro-2'-deoxycytidine 5'-triphosphate, 2'-fluoro-2'-deoxyuridine 5'- triphosphate), 2'-deoxy-2'-deamine-oligoribonucleotide (2'-amino-2'-deoxycytidine 5'-triphosphate, 2'-amino-2'-deoxyuridine 5'-triphosphate), 2'-O-alkyloligoribonucleotide, 2'-deoxy-2'-C- alkyloligoribonucleotide (2'-O-methylcytidine 5'-triphosphate, 2'-methyluridine 5'-triphosphate), 2'- C-alkyloligoribonucleotide, and isomers thereof (2'-aracytidine 5'-triphosphate, 2'-arauridine 5'- triphosphate), or azidotriphosphates (2'-azido-2'-deoxycytidine 5'-triphosphate, 2'-azido-2'- deoxyuridine 5'-triphosphate). [0194] In some embodiments, mRNAs may contain modifications of the bases of the nucleotides (base modifications). A modified nucleotide which contains a base modification is also called a base- modified nucleotide. Examples of such base-modified nucleotides include, but are not limited to, 2- amino-6-chloropurine riboside 5'-triphosphate, 2-aminoadenosine 5'-triphosphate, 2-thiocytidine 5'- triphosphate, 2-thiouridine 5'-triphosphate, 4-thiouridine 5'-triphosphate, 5-aminoallylcytidine 5'- triphosphate, 5-aminoallyluridine 5'-triphosphate, 5-bromocytidine 5'-triphosphate, 5-bromouridine 5'-triphosphate, 5-iodocytidine 5'-triphosphate, 5-iodouridine 5'-triphosphate, 5-methylcytidine 5'- triphosphate, 5-methyluridine 5'-triphosphate, 6-azacytidine 5'-triphosphate, 6-azauridine 5'- triphosphate, 6-chloropurine riboside 5'-triphosphate, 7-deazaadenosine 5'-triphosphate, 7- deazaguanosine 5'-triphosphate, 8-azaadenosine 5'-triphosphate, 8-azidoadenosine 5'-triphosphate, benzimidazole riboside 5'-triphosphate, N1-methyladenosine 5'-triphosphate, N1-methylguanosine 5'-triphosphate, N6-methyladenosine 5'-triphosphate, O6-methylguanosine 5'-triphosphate, pseudouridine 5'-triphosphate, puromycin 5'-triphosphate or xanthosine 5'-triphosphate. [0195] Typically, mRNA synthesis includes the addition of a“cap” on the N-terminal (5’) end, and a “tail” on the C-terminal (3’) end. The presence of the cap is important in providing resistance to nucleases found in most eukaryotic cells. The presence of a“tail” serves to protect the mRNA from exonuclease degradation. [0196] Thus, in some embodiments, mRNAs include a 5’ cap structure. A 5’ cap is typically added as follows: first, an RNA terminal phosphatase removes one of the terminal phosphate groups from the 5’ nucleotide, leaving two terminal phosphates; guanosine triphosphate (GTP) is then added to the terminal phosphates via a guanylyl transferase, producing a 5’5’5 triphosphate linkage; and the 7- nitrogen of guanine is then methylated by a methyltransferase. Examples of cap structures include, but are not limited to, m7G(5')ppp (5'(A,G(5')ppp(5')A and G(5')ppp(5')G. [0197] In some embodiments, mRNAs include a 3’ poly(A) tail structure. A poly-A tail on the 3'
terminus of mRNA typically includes about 10 to 300 adenosine nucleotides (e.g., about 10 to 200 adenosine nucleotides, about 10 to 150 adenosine nucleotides, about 10 to 100 adenosine nucleotides, about 20 to 70 adenosine nucleotides, or about 20 to 60 adenosine nucleotides). In some embodiments, mRNAs include a 3’ poly(C) tail structure. A suitable poly-C tail on the 3' terminus of mRNA typically include about 10 to 200 cytosine nucleotides (e.g., about 10 to 150 cytosine nucleotides, about 10 to 100 cytosine nucleotides, about 20 to 70 cytosine nucleotides, about 20 to 60 cytosine nucleotides, or about 10 to 40 cytosine nucleotides). The poly-C tail may be added to the poly-A tail or may substitute the poly-A tail. [0198] In some embodiments, mRNAs include a 5’ and/or 3’ untranslated region. In some
embodiments, a 5’ untranslated region includes one or more elements that affect an mRNA’s stability or translation, for example, an iron responsive element. In some embodiments, a 5’ untranslated region may be between about 50 and 500 nucleotides in length. [0199] In some embodiments, a 3’ untranslated region includes one or more of a polyadenylation signal, a binding site for proteins that affect an mRNA’s stability of location in a cell, or one or more binding sites for miRNAs. In some embodiments, a 3’ untranslated region may be between 50 and 500 nucleotides in length or longer. Cap structure
[0200] In some embodiments, mRNAs include a 5’ cap structure. A 5’ cap is typically added as follows: first, an RNA terminal phosphatase removes one of the terminal phosphate groups from the 5’ nucleotide, leaving two terminal phosphates; guanosine triphosphate (GTP) is then added to the terminal phosphates via a guanylyl transferase, producing a 5’5’5 triphosphate linkage; and the 7- nitrogen of guanine is then methylated by a methyltransferase. Examples of cap structures include, but are not limited to, m7G(5')ppp (5'(A,G(5')ppp(5')A and G(5')ppp(5')G. [0201] Naturally occurring cap structures comprise a 7-methyl guanosine that is linked via a
triphosphate bridge to the 5'-end of the first transcribed nucleotide, resulting in a dinucleotide cap of m7G(5')ppp(5')N, where N is any nucleoside. In vivo, the cap is added enzymatically. The cap is added in the nucleus and is catalyzed by the enzyme guanylyl transferase. The addition of the cap to the 5' terminal end of RNA occurs immediately after initiation of transcription. The terminal nucleoside is typically a guanosine, and is in the reverse orientation to all the other nucleotides, i.e., G(5')ppp(5')GpNpNp. [0202] A common cap for mRNA produced by in vitro transcription is m7G(5')ppp(5')G, which has been used as the dinucleotide cap in transcription with T7 or SP6 RNA polymerase in vitro to obtain RNAs having a cap structure in their 5'-termini. The prevailing method for the in vitro synthesis of caPPEd mRNA employs a pre-formed dinucleotide of the form m7G(5')ppp(5')G (“m7GpppG”) as an initiator of transcription. [0203] To date, a usual form of a synthetic dinucleotide cap used in in vitro translation experiments is the Anti-Reverse Cap Analog (“ARCA”) or modified ARCA, which is generally a modified cap analog in which the 2' or 3' OH group is replaced with -OCH3. [0204] Additional cap analogs include, but are not limited to, a chemical structures selected from the group consisting of m7GpppG, m7GpppA, m7GpppC; unmethylated cap analogs (e.g., GpppG);
dimethylated cap analog (e.g., m2,7GpppG), trimethylated cap analog (e.g., m2,2,7GpppG), dimethylated symmetrical cap analogs (e.g., m7Gpppm7G), or anti reverse cap analogs (e.g., ARCA; m7,2'OmeGpppG, m72'dGpppG, m7,3'OmeGpppG, m7,3'dGpppG and their tetraphosphate derivatives) (see, e.g., Jemielity, J. et al.,“Novel‘anti-reverse’ cap analogs with superior translational properties”, RNA, 9: 1108-1122 (2003)). [0205] In some embodiments, a suitable cap is a 7-methyl guanylate (“m7G”) linked via a triphosphate bridge to the 5'-end of the first transcribed nucleotide, resulting in m7G(5')ppp(5')N, where N is any nucleoside. A preferred embodiment of a m7G cap utilized in embodiments of the invention is m7G(5')ppp(5')G. [0206] In some embodiments, the cap is a Cap0 structure. Cap0 structures lack a 2'-O-methyl residue of the ribose attached to bases 1 and 2. In some embodiments, the cap is a Cap1 structure. Cap1 structures have a 2'-O-methyl residue at base 2. In some embodiments, the cap is a Cap2 structure. Cap2 structures have a 2'-O-methyl residue attached to both bases 2 and 3. [0207] A variety of m7G cap analogs are known in the art, many of which are commercially available.
These include the m7GpppG described above, as well as the ARCA 3'-OCH3 and 2'-OCH3 cap analogs (Jemielity, J. et al., RNA, 9: 1108-1122 (2003)). Additional cap analogs for use in embodiments of the invention include N7-benzylated dinucleoside tetraphosphate analogs (described in Grudzien, E. et al., RNA, 10: 1479-1487 (2004)), phosphorothioate cap analogs (described in Grudzien-Nogalska, E., et al., RNA, 13: 1745-1755 (2007)), and cap analogs (including biotinylated cap analogs) described in U.S. Patent Nos.8,093,367 and 8,304,529, incorporated by reference herein. Tail structure
[0208] Typically, the presence of a“tail” serves to protect the mRNA from exonuclease degradation.
The poly A tail is thought to stabilize natural messengers and synthetic sense RNA. Therefore, in certain embodiments a long poly A tail can be added to an mRNA molecule thus rendering the RNA more stable. Poly A tails can be added using a variety of art-recognized techniques. For example, long poly A tails can be added to synthetic or in vitro transcribed RNA using poly A polymerase (Yokoe, et al. Nature Biotechnology.1996; 14: 1252-1256). A transcription vector can also encode long poly A tails. In addition, poly A tails can be added by transcription directly from PCR products. Poly A may also be ligated to the 3' end of a sense RNA with RNA ligase (see, e.g., Molecular Cloning A Laboratory Manual, 2nd Ed., ed. by Sambrook, Fritsch and Maniatis (Cold Spring Harbor
Laboratory Press: 1991 edition)). [0209] In some embodiments, mRNAs include a 3’ poly(A) tail structure. Typically, the length of the poly A tail can be at least about 10, 50, 100, 200, 300, 400 at least 500 nucleotides. In some embodiments, a poly-A tail on the 3' terminus of mRNA typically includes about 10 to 300 adenosine nucleotides (e.g., about 10 to 200 adenosine nucleotides, about 10 to 150 adenosine nucleotides, about 10 to 100 adenosine nucleotides, about 20 to 70 adenosine nucleotides, or about 20 to 60 adenosine nucleotides). In some embodiments, mRNAs include a 3’ poly(C) tail structure. A suitable poly-C tail on the 3' terminus of mRNA typically include about 10 to 200 cytosine nucleotides (e.g., about 10 to 150 cytosine nucleotides, about 10 to 100 cytosine nucleotides, about 20 to 70 cytosine nucleotides, about 20 to 60 cytosine nucleotides, or about 10 to 40 cytosine nucleotides). The poly- C tail may be added to the poly-A tail or may substitute the poly-A tail. [0210] In some embodiments, the length of the poly A or poly C tail is adjusted to control the stability of a modified sense mRNA molecule of the invention and, thus, the transcription of protein. For example, since the length of the poly A tail can influence the half-life of a sense mRNA molecule, the length of the poly A tail can be adjusted to modify the level of resistance of the mRNA to nucleases and thereby control the time course of polynucleotide expression and/or polypeptide production in a target cell. 5’ and 3’ Untranslated Region
[0211] In some embodiments, mRNAs include a 5’ and/or 3’ untranslated region. In some
embodiments, a 5’ untranslated region includes one or more elements that affect an mRNA’s stability or translation, for example, an iron responsive element. In some embodiments, a 5’ untranslated region may be between about 50 and 500 nucleotides in length. [0212] In some embodiments, a 3’ untranslated region includes one or more of a polyadenylation signal, a binding site for proteins that affect an mRNA’s stability of location in a cell, or one or more binding sites for miRNAs. In some embodiments, a 3’ untranslated region may be between 50 and 500 nucleotides in length or longer. [0213] Exemplary 3' and/or 5' UTR sequences can be derived from mRNA molecules which are stable (e.g., globin, actin, GAPDH, tubulin, histone, or citric acid cycle enzymes) to increase the stability of the sense mRNA molecule. For example, a 5’ UTR sequence may include a partial sequence of a CMV immediate-early 1 (IE1) gene, or a fragment thereof to improve the nuclease resistance and/or improve the half-life of the polynucleotide. Also contemplated is the inclusion of a sequence encoding human growth hormone (hGH), or a fragment thereof to the 3’ end or untranslated region of the polynucleotide (e.g., mRNA) to further stabilize the polynucleotide. Generally, these modifications improve the stability and/or pharmacokinetic properties (e.g., half-life) of the polynucleotide relative to their unmodified counterparts, and include, for example modifications made to improve such polynucleotides’ resistance to in vivo nuclease digestion.
Pharmaceutical Formulations of Cationic Lipids and Nucleic Acids
[0214] In certain embodiments cationic lipids described herein described (e.g., a cationic lipid of any of Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III- B), (IV-A), (IV-B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)-(156)), as well as pharmaceutical and liposomal compositions comprising such lipids, can be used in formulations to facilitate the delivery of encapsulated materials (e.g., one or more polynucleotides such as mRNA) to, and subsequent transfection of one or more target cells. For example, in certain embodiments cationic lipids described herein (and compositions such as liposomal compositions comprising such lipids) are characterized as resulting in one or more of receptor-mediated endocytosis, clathrin- mediated and caveolae-mediated endocytosis, phagocytosis and macropinocytosis, fusogenicity, endosomal or lysosomal disruption and/or releasable properties that afford such compounds advantages relative other similarly classified lipids. [0215] According to the present invention, a nucleic acid, e.g., mRNA encoding a protein (e.g., a full length, fragment or portion of a protein) as described herein may be delivered via a delivery vehicle comprising a cationic lipid as described (e.g., a cationic lipid of any of Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III-B), (IV-A), (IV-B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)-(156)). [0216] As used herein, the terms“delivery vehicle,”“transfer vehicle,”“nanoparticle” or grammatical equivalent, are used interchangeably. [0217] For example, the present invention provides a composition (e.g., a pharmaceutical composition) comprising a cationic lipid described herein (e.g., a cationic lipid of any of Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III-B), (IV-A), (IV-B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)-(156)) and one or more polynucleotides. A composition (e.g., a pharmaceutical composition) may further comprise one or more cationic lipids, one or more non-cationic lipids, one or more cholesterol-based lipids and/or one or more PEG-modified lipids. [0218] In certain embodiments a composition exhibits an enhanced (e.g., increased) ability to transfect one or more target cells. Accordingly, also provided herein are methods of transfecting one or more target cells. Such methods generally comprise the step of contacting the one or more target cells with the cationic lipids and/or pharmaceutical compositions disclosed herein (e.g., a liposomal formulation comprising a cationic lipid described herein (e.g., a cationic lipid of any of Formulas (I)- (VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III-B), (IV-A), (IV- B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)-(156)) encapsulating one or more polynucleotides) such that the one or more target cells are transfected with the materials encapsulated therein (e.g., one or more polynucleotides). As used herein, the terms“transfect” or “transfection” refer to the intracellular introduction of one or more encapsulated materials (e.g., nucleic acids and/or polynucleotides) into a cell, or preferably into a target cell. The introduced polynucleotide may be stably or transiently maintained in the target cell. The term“transfection efficiency” refers to the relative amount of such encapsulated material (e.g., polynucleotides) up- taken by, introduced into and/or expressed by the target cell which is subject to transfection. In practice, transfection efficiency may be estimated by the amount of a reporter polynucleotide product produced by the target cells following transfection. In certain embodiments, the compounds and pharmaceutical compositions described herein demonstrate high transfection efficiencies thereby improving the likelihood that appropriate dosages of the encapsulated materials (e.g., one or more polynucleotides) will be delivered to the site of pathology and subsequently expressed, while at the same time minimizing potential systemic adverse effects or toxicity associated with the compound or their encapsulated contents. [0219] Following transfection of one or more target cells by, for example, the polynucleotides
encapsulated in the one or more lipid nanoparticles comprising the pharmaceutical or liposomal compositions disclosed herein, the production of the product (e.g., a polypeptide or protein) encoded by such polynucleotide may be preferably stimulated and the capability of such target cells to express the polynucleotide and produce, for example, a polypeptide or protein of interest is enhanced. For example, transfection of a target cell by one or more compounds or pharmaceutical compositions encapsulating mRNA will enhance (i.e., increase) the production of the protein or enzyme encoded by such mRNA. [0220] Further, delivery vehicles described herein (e.g., liposomal delivery vehicles) may be prepared to preferentially distribute to other target tissues, cells or organs, such as the heart, lungs, kidneys, spleen. In embodiments, the lipid nanoparticles of the present invention may be prepared to achieve enhanced delivery to the target cells and tissues. For example, polynucleotides (e.g., mRNA) encapsulated in one or more of the compounds or pharmaceutical and liposomal compositions described herein can be delivered to and/or transfect targeted cells or tissues. In some
embodiments, the encapsulated polynucleotides (e.g., mRNA) are capable of being expressed and functional polypeptide products produced (and in some instances excreted) by the target cell, thereby conferring a beneficial property to, for example the target cells or tissues. Such
encapsulated polynucleotides (e.g., mRNA) may encode, for example, a hormone, enzyme, receptor, polypeptide, peptide or other protein of interest.
Liposomal Delivery Vehicles
[0221] In some embodiments, a composition is a suitable delivery vehicle. In embodiments, a
composition is a liposomal delivery vehicle, e.g., a lipid nanoparticle. [0222] The terms“liposomal delivery vehicle” and“liposomal composition” are used interchangeably. [0223] Enriching liposomal compositions with one or more of the cationic lipids disclosed herein may be used as a means of improving (e.g., reducing) the toxicity or otherwise conferring one or more desired properties to such enriched liposomal composition (e.g., improved delivery of the encapsulated polynucleotides to one or more target cells and/or reduced in vivo toxicity of a liposomal composition). Accordingly, also contemplated are pharmaceutical compositions, and in particular liposomal compositions, that comprise one or more of the cationic lipids disclosed herein. [0224] Thus, in certain embodiments, the compounds described herein (e.g., a cationic lipid of any of Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III- B), (IV-A), (IV-B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)-(156)) are cationic lipids that may be used as a component of a liposomal composition to facilitate or enhance the delivery and release of encapsulated materials (e.g., one or more therapeutic agents) to one or more target cells (e.g., by permeating or fusing with the lipid membranes of such target cells). [0225] As used herein, liposomal delivery vehicles, e.g., lipid nanoparticles, are usually characterized as microscopic vesicles having an interior aqua space sequestered from an outer medium by a membrane of one or more bilayers. Bilayer membranes of liposomes are typically formed by amphiphilic molecules, such as lipids of synthetic or natural origin that comprise spatially separated hydrophilic and hydrophobic domains (Lasic, Trends Biotechnol., 16: 307-321, 1998). Bilayer membranes of the liposomes can also be formed by amphophilic polymers and surfactants (e.g., polymerosomes, niosomes, etc.). In the context of the present invention, a liposomal delivery vehicle typically serves to transport a desired mRNA to a target cell or tissue. [0226] In certain embodiments, such compositions (e.g., liposomal compositions) are loaded with or otherwise encapsulate materials, such as for example, one or more biologically-active
polynucleotides (e.g., mRNA). [0227] In embodiments, a composition (e.g., a pharmaceutical composition) comprises an mRNA
encoding a protein, encapsulated within a liposome. In embodiments, a liposome comprises one or more cationic lipids, one or more non-cationic lipids, one or more cholesterol-based lipids and one or more PEG-modified lipids, and at least one cationic lipid is a cationic lipid as described herein (e.g., a cationic lipid of any of Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III-B), (IV-A), (IV-B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)-(156)). In embodiments, a composition comprises an mRNA encoding for a protein (e.g., any protein described herein). In embodiments, a composition comprises an mRNA encoding for cystic fibrosis transmembrane conductance regulator (CFTR) protein. In embodiments, a composition comprises an mRNA encoding for ornithine transcarbamylase (OTC) protein. In embodiments, an mRNA encodes for an antigen (e.g., an antigen from an infectious agent. [0228] In embodiments, a composition (e.g., a pharmaceutical composition) comprises a nucleic acid encapsulated within a liposome, wherein the liposome comprises any cationic lipid (e.g., a cationic lipid of any of Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II- B), (III-A), (III-B), (IV-A), (IV-B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)- (156)) as described herein. [0229] In embodiments, a nucleic acid is an mRNA encoding a peptide or polypeptide. In
embodiments, an mRNA encodes a peptide or polypeptide for use in the delivery to or treatment of the lung of a subject or a lung cell (e.g., an mRNA encodes cystic fibrosis transmembrane conductance regulator (CFTR) protein). In embodiments, an mRNA encodes a peptide or polypeptide for use in the delivery to or treatment of the liver of a subject or a liver cell (e.g., an mRNA encodes ornithine transcarbamylase (OTC) protein). Still other exemplary mRNAs are described herein. [0230] In embodiments, a liposomal delivery vehicle (e.g., a lipid nanoparticle) can have a net positive charge. [0231] In embodiments, a liposomal delivery vehicle (e.g., a lipid nanoparticle) can have a net negative charge. [0232] In embodiments, a liposomal delivery vehicle (e.g., a lipid nanoparticle) can have a net neutral charge. [0233] In embodiments, a lipid nanoparticle that encapsulates a nucleic acid (e.g., mRNA encoding a peptide or polypeptide) comprises one or more cationic lipids described herein (e.g., a cationic lipid of any of Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III-B), (IV-A), (IV-B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)-(156)). [0234] For example, the amount of a cationic lipid as described herein (e.g., a cationic lipid of any of Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III- B), (IV-A), (IV-B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)-(156)) in a composition can be described as a percentage (“wt%”) of the combined dry weight of all lipids of a composition (e.g., the combined dry weight of all lipids present in a liposomal composition). [0235] In embodiments of the pharmaceutical compositions described herein, a cationic lipid as
described herein (e.g., a cationic lipid of any of Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III-B), (IV-A), (IV-B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV- C”), or any of Compounds (1)-(156)) is present in an amount that is about 0.5 wt% to about 30 wt% (e.g., about 0.5 wt% to about 20 wt%) of the combined dry weight of all lipids present in a composition (e.g., a liposomal composition). [0236] In embodiments, a cationic lipid as described herein (e.g., a cationic lipid of any of Formulas (I)- (VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III-B), (IV-A), (IV- B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)-(156)) is present in an amount that is about 1 wt% to about 30 wt%, about 1 wt% to about 20 wt%, about 1 wt% to about 15 wt%, about 1 wt% to about 10 wt%, or about 5 wt% to about 25 wt% of the combined dry weight of all lipids present in a composition (e.g., a liposomal composition). In embodiments, a cationic lipid as described herein (e.g., a cationic lipid of any of Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III-B), (IV-A), (IV-B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV- C”), or any of Compounds (1)-(156)) is present in an amount that is about 0.5 wt% to about 5 wt%, about 1 wt% to about 10 wt%, about 5 wt% to about 20 wt%, or about 10 wt% to about 20 wt% of the combined molar amounts of all lipids present in a composition such as a liposomal delivery vehicle. [0237] In embodiments, the amount of a cationic lipid as described herein (e.g., a cationic lipid of any of Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III-B), (IV-A), (IV-B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)-(156)) is present in an amount that is at least about 5 wt%, about 10 wt%, about 15 wt%, about 20 wt%, about 25 wt%, about 30 wt%, about 35 wt%, about 40 wt%, about 45 wt%, about 50 wt%, about 55 wt%, about 60 wt%, about 65 wt%, about 70 wt%, about 75 wt%, about 80 wt%, about 85 wt%, about 90 wt%, about 95 wt%, about 96 wt%, about 97 wt%, about 98 wt%, or about 99 wt% of the combined dry weight of total lipids in a composition (e.g., a liposomal composition). [0238] In embodiments, the amount of a cationic lipid as described herein (e.g., a cationic lipid of any of Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III-B), (IV-A), (IV-B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)-(156)) is present in an amount that is no more than about 5 wt%, about 10 wt%, about 15 wt%, about 20 wt%, about 25 wt%, about 30 wt%, about 35 wt%, about 40 wt%, about 45 wt%, about 50 wt%, about 55 wt%, about 60 wt%, about 65 wt%, about 70 wt%, about 75 wt%, about 80 wt%, about 85 wt%, about 90 wt%, about 95 wt%, about 96 wt%, about 97 wt%, about 98 wt%, or about 99 wt% of the combined dry weight of total lipids in a composition (e.g., a liposomal composition). [0239] In embodiments, a composition (e.g., a liposomal delivery vehicle such as a lipid nanoparticle) comprises about 0.1 wt% to about 20 wt% (e.g., about 0.1 wt% to about 15 wt%) of a cationic lipid described herein (e.g., a cationic lipid of any of Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III-B), (IV-A), (IV-B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV- C”), or any of Compounds (1)-(156)). In embodiments, a delivery vehicle (e.g., a liposomal delivery vehicle such as a lipid nanoparticle) comprises about 0.5 wt%, about 1 wt%, about 3 wt%, about 5 wt%, or about 10 wt% a cationic lipid described herein (e.g., a cationic lipid of any of Formulas (I)- (VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III-B), (IV-A), (IV- B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)-(156)). In embodiments, a delivery vehicle (e.g., a liposomal delivery vehicle such as a lipid nanoparticle) comprises up to about 0.5 wt%, about 1 wt%, about 3 wt%, about 5 wt%, about 10 wt%, about 15 wt%, or about 20 wt% of a cationic lipid described herein (e.g., a cationic lipid of any of Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III-B), (IV-A), (IV-B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)-(156)). In embodiments, the percentage results in an improved beneficial effect (e.g., improved delivery to targeted tissues such as the liver or the lung). [0240] The amount of a cationic lipid as described herein (e.g., a cationic lipid of any of Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III-B), (IV-A), (IV-B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)-(156)) in a composition also can be described as a percentage (“mol%”) of the combined molar amounts of total lipids of a composition (e.g., the combined molar amounts of all lipids present in a liposomal delivery vehicle). [0241] In embodiments of pharmaceutical compositions described herein, a cationic lipid as described herein (e.g., a cationic lipid of any of Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III-B), (IV-A), (IV-B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)-(156)) is present in an amount that is about 0.5 mol% to about30 mol% (e.g., about 0.5 mol% to about20 mol%) of the combined molar amounts of all lipids present in a composition such as a liposomal delivery vehicle. [0242] In embodiments, a cationic lipid as described herein (e.g., a cationic lipid of any of Formulas (I)- (VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III-B), (IV-A), (IV- B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)-(156)) is present in an amount that is about 0.5 mol% to about 5 mol%, about 1 mol% to about 10 mol%, about 5 mol% to about 20 mol%, or about 10 mol% to about 20 mol% of the combined molar amounts of all lipids present in a composition such as a liposomal delivery vehicle. In embodiments, a cationic lipid as described herein (e.g., a cationic lipid of any of Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III-B), (IV-A), (IV-B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)-(156)) is present in an amount that is about 1 mol% to about 30 mol%, about 1 mol% to about 20 mol%, about 1 mol% to about 15 mol%, about 1 mol% to about 10 mol%, or about 5 mol% to about 25 mol% of the combined dry weight of all lipids present in a composition such as a liposomal delivery vehicle [0243] In certain embodiments, a cationic lipid as described herein (e.g., a cationic lipid of any of
Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III- B), (IV-A), (IV-B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)-(156)) can comprise from about 0.1 mol% to about 50 mol%, or from 0.5 mol% to about 50 mol%, or from about 1 mol% to about 25 mol%, or from about 1 mol% to about 10 mol% of the total amount of lipids in a composition (e.g., a liposomal delivery vehicle). [0244] In certain embodiments, a cationic lipid as described herein (e.g., a cationic lipid of any of
Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III- B), (IV-A), (IV-B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)-(156)) can comprise greater than about 0.1 mol%, or greater than about 0.5 mol%, or greater than about 1 mol%, or greater than about 5 mol% of the total amount of lipids in the lipid nanoparticle. [0245] In certain embodiments, a cationic lipid as described herein (e.g., a cationic lipid of any of Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III- B), (IV-A), (IV-B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)-(156)) can comprise less than about 25 mol%, or less than about 10 mol%, or less than about 5 mol%, or less than about 1 mol% of the total amount of lipids in a composition (e.g., a liposomal delivery vehicle). [0246] In embodiments, the amount of a cationic lipid as described herein (e.g., a cationic lipid of any of Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III-B), (IV-A), (IV-B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)-(156)) is present in an amount that is at least about 5 mol%, about 10 mol%, about 15 mol%, about 20 mol%, about 25 mol%, about 30 mol%, about 35 mol%, about 40 mol%, about 45 mol%, about 50 mol%, about 55 mol%, about 60 mol%, about 65 mol%, about 70 mol%, about 75 mol%, about 80 mol%, about 85 mol%, about 90 mol%, about 95 mol%, about 96 mol%, about 97 mol%, about 98 mol%, or about 99 mol% of the combined dry weight of total lipids in a composition (e.g., a liposomal composition). [0247] In embodiments, the amount of a cationic lipid as described herein (e.g., a cationic lipid of any of Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III-B), (IV-A), (IV-B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)-(156)) is present in an amount that is no more than about 5 mol%, about 10 mol%, about 15 mol%, about 20 mol%, about 25 mol%, about 30 mol%, about 35 mol%, about 40 mol%, about 45 mol%, about 50 mol%, about 55 mol%, about 60 mol%, about 65 mol%, about 70 mol%, about 75 mol%, about 80 mol%, about 85 mol%, about 90 mol%, about 95 mol%, about 96 mol%, about 97 mol%, about 98 mol%, or about 99 mol% of the combined dry weight of total lipids in a composition (e.g., a liposomal composition). [0248] In embodiments, the percentage results in an improved beneficial effect (e.g., improved delivery to targeted tissues such as the liver or the lung). [0249] In embodiments, a composition further comprises one more lipids (e.g., one more lipids
selected from the group consisting of one or more cationic lipids, one or more non-cationic lipids, and one or more PEG-modified lipids). [0250] In certain embodiments, such pharmaceutical (e.g., liposomal) compositions comprise one or more of a PEG-modified lipid, a non-cationic lipid and a cholesterol lipid. In embodiments, such pharmaceutical (e.g., liposomal) compositions comprise: one or more PEG-modified lipids; one or more non-cationic lipids; and one or more cholesterol lipids. In embodiments, such pharmaceutical (e.g., liposomal) compositions comprise: one or more PEG-modified lipids and one or more cholesterol lipids. [0251] In embodiments, a composition (e.g., lipid nanoparticle) that encapsulates a nucleic acid (e.g., mRNA encoding a peptide or polypeptide) comprises one or more cationic lipids as described herein (e.g., a cationic lipid of any of Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III-B), (IV-A), (IV-B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)-(156)) and one or more lipids selected from the group consisting of a cationic lipid, a non-cationic lipid, and a PEGylated lipid. [0252] In embodiments, a composition (e.g., lipid nanoparticle) that encapsulates a nucleic acid (e.g., mRNA encoding a peptide or polypeptide) comprises one or more cationic lipids as described herein (e.g., a cationic lipid of any of Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III-B), (IV-A), (IV-B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)-(156)); one or more lipids selected from the group consisting of a cationic lipid, a non-cationic lipid, and a PEGylated lipid; and further comprises a cholesterol-based lipid. [0253] In embodiments, a lipid nanoparticle that encapsulates a nucleic acid (e.g., mRNA encoding a peptide or polypeptide) comprises one or more cationic lipids as described herein (e.g., a cationic lipid of any of Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II- B), (III-A), (III-B), (IV-A), (IV-B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)- (156)), as well as one or more lipids selected from the group consisting of a cationic lipid, a non- cationic lipid, a PEGylated lipid, and a cholesterol-based lipid. [0254] In embodiments of lipid nanoparticles described herein, a lipid nanoparticle comprises one or more cationic lipids described herein (e.g., a cationic lipid of any of Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III-B), (IV-A), (IV-B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)-(156)), a non-cationic lipid (e.g., DOPE), a
PEGylated lipid (e.g., DMG-PEG2000), and a cholesterol-based lipid (e.g., cholesterol). [0255] According to various embodiments, the selection of cationic lipids, non-cationic lipids and/or PEG-modified lipids which comprise the lipid nanoparticle, as well as the relative molar ratio of such lipids to each other, is based upon the characteristics of the selected lipid(s), the nature of the intended target cells, the characteristics of the mRNA to be delivered. Additional considerations include, for example, the saturation of the alkyl chain, as well as the size, charge, pH, pKa, fusogenicity and toxicity of the selected lipid(s). Thus, the molar ratios may be adjusted accordingly. Further Cationic Lipids
[0256] In addition to any of the cationic lipids as described herein (e.g., a cationic lipid of any of
Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III- B), (IV-A), (IV-B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)-(156)), a composition may comprise one or more further cationic lipids. [0257] In some embodiments, liposomes may comprise one or more further cationic lipids. As used herein, the phrase“cationic lipid” refers to any of a number of lipid species that have a net positive charge at a selected pH, such as physiological pH. Several cationic lipids have been described in the literature, many of which are commercially available. [0258] In some embodiments, liposomes may comprise one or more additional cationic lipids. As used herein, the phrase“cationic lipid” refers to any of a number of lipid species that have a net positive charge at a selected pH, such as physiological pH. Several cationic lipids have been described in the literature, many of which are commercially available. [0259] Suitable additional cationic lipids for use in the compositions include the cationic lipids as described in International Patent Publication WO 2010/144740, which is incorporated herein by reference. In certain embodiments, the compositions include a cationic lipid, (6Z,9Z,28Z,31Z)- heptatriaconta-6,9,28,31-tetraen-19-yl 4-(dimethylamino) butanoate, having a compound structure of:
Figure imgf000085_0001
and pharmaceutically acceptable salts thereof.
[0260] Other suitable additional cationic lipids for use in the compositions include ionizable cationic lipids as described in International Patent Publication WO 2013/149140, which is incorporated herein by reference. In some embodiments, the compositions include a cationic lipid of one of the following formulas:
Figure imgf000085_0002
,
or a pharmaceutically acceptable salt thereof, wherein R1 and R2 are each independently selected from the group consisting of hydrogen, an optionally substituted, variably saturated or unsaturated C1-C20 alkyl and an optionally substituted, variably saturated or unsaturated C6-C20 acyl; wherein L1 and L2 are each independently selected from the group consisting of hydrogen, an optionally substituted C1-C30 alkyl, an optionally substituted variably unsaturated C1-C30 alkenyl, and an optionally substituted C1-C30 alkynyl; wherein m and o are each independently selected from the group consisting of zero and any positive integer (e.g., where m is three); and wherein n is zero or any positive integer (e.g., where n is one). In certain embodiments, the compositions include the cationic lipid (15Z, 18Z)-N,N-dimethyl-6-(9Z,12Z)-octadeca-9,12-dien-l -yl) tetracosa- 15,18-dien-1- amine (“HGT5000”), having a compound structure of:
Figure imgf000086_0001
and pharmaceutically acceptable salts thereof. In certain embodiments, the compositions include the cationic lipid (15Z, 18Z)-N,N-dimethyl-6-((9Z,12Z)-octadeca-9,12-dien-1-yl) tetracosa-4,15,18- trien-l -amine (“HGT5001”), having a compound structure of:
Figure imgf000086_0002
and pharmaceutically acceptable salts thereof. In certain embodiments, the include the cationic lipid and (15Z,18Z)-N,N-dimethyl-6-((9Z,12Z)-octadeca-9,12-dien-1-yl) tetracosa-5,15,18-trien- 1 - amine (“HGT5002”), having a compound structure of:
Figure imgf000086_0003
and pharmaceutically acceptable salts thereof.
[0261] Other suitable additional cationic lipids for use in the compositions include cationic lipids
described as aminoalcohol lipidoids in International Patent Publication WO 2010/053572, which is incorporated herein by reference. In certain embodiments, the compositions include a cationic lipid having a compound structure of:
Figure imgf000087_0001
and pharmaceutically acceptable salts thereof.
[0262] Other suitable additional cationic lipids for use in the compositions include the cationic lipids as described in International Patent Publication WO 2016/118725, which is incorporated herein by reference. In certain embodiments, the compositions include a cationic lipid having a compound structure of:
Figure imgf000087_0002
and pharmaceutically acceptable salts thereof.
[0263] Other suitable additional cationic lipids for use in the compositions include the cationic lipids as described in International Patent Publication WO 2016/118724, which is incorporated herein by reference. In certain embodiments, the compositions include a cationic lipid having a compound structure of:
Figure imgf000087_0003
and pharmaceutically acceptable salts thereof.
[0264] Other suitable cationic lipids for use in the compositions include a cationic lipid having the formula of 14,25-ditridecyl 15,18,21,24-tetraaza-octatriacontane, and pharmaceutically acceptable salts thereof. [0265] Other suitable additional cationic lipids for use in the compositions include the cationic lipids as described in International Patent Publications WO 2013/063468 and WO 2016/205691, each of which are incorporated herein by reference. In some embodiments, the compositions include a cationic lipid of the following formula:
Figure imgf000088_0001
or pharmaceutically acceptable salts thereof, wherein each instance of RL is independently optionally substituted C6-C40 alkenyl. In certain embodiments, the compositions include a cationic lipid having a compound structure of:
Figure imgf000088_0002
and pharmaceutically acceptable salts thereof.
[0266] In certain embodiments, the compositions include a cationic lipid having a compound structure of:
Figure imgf000089_0001
(OF-02)
and pharmaceutically acceptable salts thereof.
[0267] In certain embodiments, the compositions include a cationic lipid having a compound structure of:
Figure imgf000089_0002
and pharmaceutically acceptable salts thereof. [0268] In certain embodiments, the compositions include a cationic lipid having a compound structure of:
Figure imgf000090_0001
and pharmaceutically acceptable salts thereof.
[0269] Other suitable additional cationic lipids for use in the compositions include the cationic lipids as described in International Patent Publication WO 2015/184256, which is incorporated herein by reference. In some embodiments, the compositions include a cationic lipid of the following formula:
Figure imgf000090_0002
or a pharmaceutically acceptable salt thereof, wherein each X independently is O or S; each Y independently is O or S; each m independently is 0 to 20; each n independently is 1 to 6; each RA is independently hydrogen, optionally substituted C1-50 alkyl, optionally substituted C2-50 alkenyl, optionally substituted C2-50 alkynyl, optionally substituted C3-10 carbocyclyl, optionally substituted 3-14 membered heterocyclyl, optionally substituted C6-14 aryl, optionally substituted 5-14 membered heteroaryl or halogen; and each RB is independently hydrogen, optionally substituted C1- 50 alkyl, optionally substituted C2-50 alkenyl, optionally substituted C2-50 alkynyl, optionally substituted C3-10 carbocyclyl, optionally substituted 3-14 membered heterocyclyl, optionally substituted C6-14 aryl, optionally substituted 5-14 membered heteroaryl or halogen. In certain embodiments, the compositions include a cationic lipid,“Target 23”, having a compound structure of:
Figure imgf000091_0001
(Target 23)
and pharmaceutically acceptable salts thereof.
[0270] Other suitable additional cationic lipids for use in the compositions include the cationic lipids as described in International Patent Publication WO 2016/004202, which is incorporated herein by reference. In some embodiments, the compositions include a cationic lipid having the compound structure:
Figure imgf000091_0002
, w e e
or a pharmaceutically acceptable salt thereof.
[0271] In some embodiments, the compositions include a cationic lipid having the compound structure:
Figure imgf000091_0003
or a pharmaceutically acceptable salt thereof. [0272] In some embodiments, the compositions include a cationic lipid having the compound structure:
Figure imgf000092_0001
or a pharmaceutically acceptable salt thereof.
[0273] Other suitable additional cationic lipids for use in the compositions include the cationic lipids as described in J. McClellan, M. C. King, Cell 2010, 141, 210-217 and in Whitehead et al., Nature Communications (2014) 5:4277, which is incorporated herein by reference. In certain
embodiments, the cationic lipids of the compositions include a cationic lipid having a compound structure of:
Figure imgf000092_0002
and pharmaceutically acceptable salts thereof.
[0274] Other suitable additional cationic lipids for use in the compositions include the cationic lipids as described in International Patent Publication WO 2015/199952, which is incorporated herein by reference. In some embodiments, the compositions include a cationic lipid having the compound structure:
Figure imgf000092_0003
and pharmaceutically acceptable salts thereof. [0275] In some embodiments, the compositions include a cationic lipid having the compound structure:
Figure imgf000093_0001
and pharmaceutically acceptable salts thereof.
[0276] In some embodiments, the compositions include a cationic lipid having the compound structure:
Figure imgf000093_0002
and pharmaceutically acceptable salts thereof.
[0277] In some embodiments, the compositions include a cationic lipid having the compound structure:
Figure imgf000093_0003
and pharmaceutically acceptable salts thereof.
[0278] In some embodiments, the compositions include a cationic lipid having the compound structure:
Figure imgf000093_0004
and pharmaceutically acceptable salts thereof.
[0279] In some embodiments, the compositions include a cationic lipid having the compound structure:
Figure imgf000094_0001
and pharmaceutically acceptable salts thereof.
[0280] In some embodiments, the compositions include a cationic lipid having the compound structure:
Figure imgf000094_0002
and pharmaceutically acceptable salts thereof.
[0281] In some embodiments, the compositions include a cationic lipid having the compound structure:
Figure imgf000094_0003
and pharmaceutically acceptable salts thereof.
[0282] In some embodiments, the compositions include a cationic lipid having the compound structure:
Figure imgf000094_0004
and pharmaceutically acceptable salts thereof. [0283] In some embodiments, the compositions include a cationic lipid having the compound structure:
Figure imgf000095_0001
[0284] and pharmaceutically acceptable salts thereof. [0285] In some embodiments, the compositions include a cationic lipid having the compound structure:
Figure imgf000095_0002
and pharmaceutically acceptable salts thereof.
[0286] In some embodiments, the compositions include a cationic lipid having the compound structure:
Figure imgf000095_0003
and pharmaceutically acceptable salts thereof.
[0287] In some embodiments, the compositions include a cationic lipid having the compound
structure:
Figure imgf000095_0004
and pharmaceutically acceptable salts thereof.
[0288] Other suitable additional cationic lipids for use in the compositions include the cationic lipids as described in International Patent Publication WO 2017/004143, which is incorporated herein by reference. [0289] In some embodiments, the compositions include a cationic lipid having the compound structure:
Figure imgf000096_0001
and pharmaceutically acceptable salts thereof.
[0290] In some embodiments, the compositions include a cationic lipid having the compound structure:
Figure imgf000096_0002
and pharmaceutically acceptable salts thereof.
[0291] In some embodiments, the compositions include a cationic lipid having the compound
structure:
Figure imgf000096_0003
and pharmaceutically acceptable salts thereof.
[0292] In some embodiments, the compositions include a cationic lipid having the compound
structure:
Figure imgf000096_0004
and pharmaceutically acceptable salts thereof. [0293] In some embodiments, the compositions include a cationic lipid having the compound structure:
Figure imgf000097_0001
and pharmaceutically acceptable salts thereof.
[0294] In some embodiments, the compositions include a cationic lipid having the compound
structure:
Figure imgf000097_0002
and pharmaceutically acceptable salts thereof.
[0295] In some embodiments, the compositions include a cationic lipid having the compound structure:
Figure imgf000097_0003
and pharmaceutically acceptable salts thereof.
[0296] In some embodiments, the compositions include a cationic lipid having the compound structure:
Figure imgf000097_0004
and pharmaceutically acceptable salts thereof. [0297] In some embodiments, the compositions include a cationic lipid having the compound structure:
Figure imgf000098_0001
and pharmaceutically acceptable salts thereof.
[0298] In some embodiments, the compositions include a cationic lipid having the compound structure:
Figure imgf000098_0002
and pharmaceutically acceptable salts thereof.
[0299] In some embodiments, the compositions include a cationic lipid having the compound
structure:
Figure imgf000098_0003
and pharmaceutically acceptable salts thereof.
[0300] In some embodiments, the compositions include a cationic lipid having the compound structure:
Figure imgf000099_0001
and pharmaceutically acceptable salts thereof.
[0301] In some embodiments, the compositions include a cationic lipid having the compound structure:
Figure imgf000099_0002
and pharmaceutically acceptable salts thereof.
[0302] In some embodiments, the compositions include a cationic lipid having the compound
structure:
Figure imgf000099_0003
and pharmaceutically acceptable salts thereof. [0303] In some embodiments, the compositions include a cationic lipid having the compound structure:
Figure imgf000100_0001
and pharmaceutically acceptable salts thereof.
[0304] In some embodiments, the compositions include a cationic lipid having the compound
structure:
Figure imgf000100_0002
and pharmaceutically acceptable salts thereof.
[0305] In some embodiments, the compositions include a cationic lipid having the compound
structure:
Figure imgf000100_0003
and pharmaceutically acceptable salts thereof.
[0306] Other suitable additional cationic lipids for use in the compositions include the cationic lipids as described in International Patent Publication WO 2017/075531, which is incorporated herein by reference. In some embodiments, the compositions include a cationic lipid of the following formula:
Figure imgf000100_0004
or a pharmaceutically acceptable salt thereof, wherein one of L1 or L2 is -O(C=O)-, -(C=O)O-, -C(=O)-, -O-, -S(O)x, -S-S-, -C(=O)S-, -SC(=O)-, -NRaC(=O)-, -C(=O)NRa-, NRaC(=O)NRa-, -OC(=O)NRa-, or - NRaC(=O)O-; and the other of L1 or L2 is -O(C=O)-, -(C=O)O-, -C(=O)-, -O-, -S(O) x, -S-S-, -C(=O)S-, SC(=O)-, -NRaC(=O)-, -C(=O)NRa-, ,NRaC(=O)NRa-, -OC(=O)NRa- or -NRaC(=O)O- or a direct bond; G1 and G2 are each independently unsubstituted C1-C12 alkylene or C1-C12 alkenylene; G3 is C1-C24 alkylene, C1-C24 alkenylene, C3-C8 cycloalkylene, C3-C8 cycloalkenylene; Ra is H or C1-C12 alkyl; R1 and R2 are each independently C6-C24 alkyl or C6-C24 alkenyl; R3 is H, OR5, CN, -C(=O)OR4, -OC(=O)R4 or - NR5 C(=O)R4; R4 is C1-C12 alkyl; R5 is H or C1-C6 alkyl; and x is 0, 1 or 2.
[0307] Other suitable additional cationic lipids for use in the compositions include the cationic lipids as described in International Patent Publication WO 2017/117528, which is incorporated herein by reference. In some embodiments, the compositions include a cationic lipid having the compound structure:
Figure imgf000101_0001
and pharmaceutically acceptable salts thereof.
[0308] In some embodiments, the compositions include a cationic lipid having the compound structure:
Figure imgf000101_0002
and pharmaceutically acceptable salts thereof.
[0309] In some embodiments, the compositions include a cationic lipid having the compound structure:
Figure imgf000101_0003
and pharmaceutically acceptable salts thereof. [0310] Other suitable additional cationic lipids for use in the compositions include the cationic lipids as described in International Patent Publication WO 2017/049245, which is incorporated herein by reference. In some embodiments, the cationic lipids of the compositions and methods of the present invention include a compound of one of the following formulas:
Figure imgf000102_0001
,
and pharmaceutically acceptable salts thereof. For any one of these four formulas, R4 is independently selected from -(CH2)nQ and -(CH2) nCHQR; Q is selected from the group consisting of - OR, -OH, -O(CH2)nN(R)2, -OC(O)R, -CX3, -CN, -N(R)C(O)R, -N(H)C(O)R, -N(R)S(O)2R, -N(H)S(O)2R, - N(R)C(O)N(R)2, -N(H)C(O)N(R)2, -N(H)C(O)N(H)(R), -N(R)C(S)N(R)2, -N(H)C(S)N(R)2, -N(H)C(S)N(H)(R), and a heterocycle; R is independently selected from the group consisting of C1-3 alkyl, C2-3 alkenyl, and H; and n is 1, 2, or 3. [0311] In certain embodiments, the compositions include a cationic lipid having a compound structure of:
Figure imgf000103_0001
and pharmaceutically acceptable salts thereof.
[0312] In certain embodiments, the compositions include a cationic lipid having a compound structure of:
Figure imgf000103_0002
and pharmaceutically acceptable salts thereof.
[0313] In certain embodiments, the compositions include a cationic lipid having a compound structure of:
Figure imgf000103_0003
and pharmaceutically acceptable salts thereof.
[0314] In certain embodiments, the compositions include a cationic lipid having a compound structure of:
Figure imgf000103_0004
and pharmaceutically acceptable salts thereof.
[0315] Other suitable additional cationic lipids for use in the compositions include the cationic lipids as described in International Patent Publication WO 2017/173054 and WO 2015/095340, each of which is incorporated herein by reference. [0316] In certain embodiments, the compositions include a cationic lipid having a compound structure of:
Figure imgf000104_0001
and pharmaceutically acceptable salts thereof.
[0317] In certain embodiments, the compositions include a cationic lipid having a compound structure of:
Figure imgf000104_0002
and pharmaceutically acceptable salts thereof.
[0318] In certain embodiments, the compositions include a cationic lipid having a compound structure of:
Figure imgf000104_0003
and pharmaceutically acceptable salts thereof. [0319] In certain embodiments, the compositions include a cationic lipid having a compound structure of:
Figure imgf000105_0001
and pharmaceutically acceptable salts thereof.
[0320] Other suitable additional cationic lipids for use in the compositions include cholesterol-based cationic lipids. In certain embodiments, the compositions include imidazole cholesterol ester or “ICE”, having a compound structure of:
Figure imgf000105_0002
and pharmaceutically acceptable salts thereof.
[0321] Other suitable additional cationic lipids for use in the compositions include cleavable cationic lipids as described in International Patent Publication WO 2012/170889, which is incorporated herein by reference. In some embodiments, the compositions include a cationic lipid of the following formula:
Figure imgf000105_0003
,
wherein R1 is selected from the group consisting of imidazole, guanidinium, amino, imine, enamine, an optionally-substituted alkyl amino (e.g., an alkyl amino such as dimethylamino) and pyridyl; wherein R2 is selected from the group consisting of one of the following two formulas:
Figure imgf000105_0004
and wherein R3 and R4 are each independently selected from the group consisting of an optionally substituted, variably saturated or unsaturated C6-C20 alkyl and an optionally substituted, variably saturated or unsaturated C6-C20 acyl; and wherein n is zero or any positive integer (e.g., one, two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, fourteen, fifteen, sixteen, seventeen, eighteen, nineteen, twenty or more).
[0322] In certain embodiments, the compositions include a cationic lipid,“HGT4001”, having a
compound structure of:
Figure imgf000106_0001
(HGT4001)
and pharmaceutically acceptable salts thereof.
[0323] In certain embodiments, the compositions include a cationic lipid,“HGT4002”, having a
compound structure of:
Figure imgf000106_0002
and pharmaceutically acceptable salts thereof.
[0324] In certain embodiments, the compositions include a cationic lipid,“HGT4003”, having a
compound structure of:
Figure imgf000106_0003
and pharmaceutically acceptable salts thereof.
[0325] In certain embodiments, the compositions include a cationic lipid,“HGT4004”, having a compound structure of:
Figure imgf000106_0004
and pharmaceutically acceptable salts thereof.
[0326] In certain embodiments, the compositions include a cationic lipid“HGT4005”, having a
compound structure of:
Figure imgf000107_0001
and pharmaceutically acceptable salts thereof.
[0327] In some embodiments, the compositions include the cationic lipid, N-[l-(2,3-dioleyloxy)propyl]- N,N,N-trimethylammonium chloride (“DOTMA”). Feigner et al. (Proc. Nat'l Acad. Sci.84, 7413 (1987); U.S. Pat. No.4,897,355, each of which is incorporated herein by reference. DOTMA can be formulated alone or can be combined with a neutral lipid (e.g., dioleoylphosphatidyl-ethanolamine or“DOPE”) or still other cationic or non-cationic lipids into a liposomal transfer vehicle or a lipid nanoparticle, and such liposomes can be used to enhance the delivery of nucleic acids into target cells. Other cationic lipids suitable for the compositions include, for example, 5- carboxyspermylglycinedioctadecylamide (“DOGS”); 2,3-dioleyloxy-N-[2(spermine- carboxamido)ethyl]-N,N-dimethyl-l-propanaminium (“DOSPA”) (Behr et al. Proc. Nat.'l Acad. Sci.86, 6982 (1989), U.S. Pat. No.5,171,678; U.S. Pat. No.5,334,761); l,2-Dioleoyl-3-Dimethylammonium- Propane (“DODAP”); l,2-Dioleoyl-3-Trimethylammonium-Propane (“DOTAP”). [0328] Additional exemplary cationic lipids suitable for the compositions also include: l,2-distearyloxy- N,N-dimethyl-3-aminopropane (“DSDMA”); 1,2-dioleyloxy-N,N-dimethyl-3-aminopropane
(“DODMA”); 1 ,2-dilinoleyloxy-N,N-dimethyl-3-aminopropane (“DLinDMA”); l,2-dilinolenyloxy-N,N- dimethyl-3-aminopropane (“DLenDMA”); N-dioleyl-N,N-dimethylammonium chloride (“DODAC”); N,N-distearyl-N,N-dimethylarnrnonium bromide (“DDAB”); N-(l,2-dimyristyloxyprop-3-yl)-N,N- dimethyl-N-hydroxyethyl ammonium bromide (“DMRIE”); 3-dimethylamino-2-(cholest-5-en-3-beta- oxybutan-4-oxy)-l-(cis,cis-9,12-octadecadienoxy)propane (“CLinDMA”); 2-[5'-(cholest-5-en-3-beta- oxy)-3'-oxapentoxy)-3-dimethy l-l-(cis,cis-9', l-2'-octadecadienoxy)propane (“CpLinDMA”); N,N- dimethyl-3,4-dioleyloxybenzylamine (“DMOBA”); 1 ,2-N,N'-dioleylcarbamyl-3- dimethylaminopropane (“DOcarbDAP”); 2,3-Dilinoleoyloxy-N,N-dimethylpropylamine (“DLinDAP”); l,2-N,N'-Dilinoleylcarbamyl-3-dimethylaminopropane (“DLincarbDAP”); l ,2-Dilinoleoylcarbamyl-3- dimethylaminopropane (“DLinCDAP”); 2,2-dilinoleyl-4-dimethylaminomethyl-[l,3]-dioxolane (“DLin- K-DMA”); 2-((8-[(3P)-cholest-5-en-3-yloxy]octyl)oxy)-N, N-dimethyl-3-[(9Z, 12Z)-octadeca-9, 12-dien- 1 -yloxy]propane-1-amine (“Octyl-CLinDMA”); (2R)-2-((8-[(3beta)-cholest-5-en-3-yloxy]octyl)oxy)-N, N-dimethyl-3-[(9Z, 12Z)-octadeca-9, 12-dien-1-yloxy]propan-1 -amine (“Octyl-CLinDMA (2R)”); (2S)- 2-((8-[(3P)-cholest-5-en-3-yloxy]octyl)oxy)-N, fsl-dimethyh3-[(9Z, 12Z)-octadeca-9, 12-dien-1 - yloxy]propan-1 -amine (“Octyl-CLinDMA (2S)”); 2,2-dilinoleyl-4-dimethylaminoethyl-[l,3]-dioxolane (“DLin-K-XTC2-DMA”); and 2-(2,2-di((9Z,12Z)-octadeca-9,l 2-dien- 1-yl)-l ,3-dioxolan-4-yl)-N,N- dimethylethanamine (“DLin-KC2-DMA”) (see, WO 2010/042877, which is incorporated herein by reference; Semple et al., Nature Biotech.28: 172-176 (2010)). (Heyes, J., et al., J Controlled Release 107: 276-287 (2005); Morrissey, DV., et al., Nat. Biotechnol.23(8): 1003-1007 (2005); International Patent Publication WO 2005/121348). In some embodiments, one or more of the cationic lipids comprise at least one of an imidazole, dialkylamino, or guanidinium moiety. [0329] In some embodiments, one or more cationic lipids suitable for the compositions include 2,2- Dilinoley1-4-dimethylaminoethy1-[1,3]-dioxolane (“XTC”); (3aR,5s,6aS)-N,N-dimethyl-2,2- di((9Z,12Z)-octadeca-9,12-dienyl)tetrahydro-3aH-cyclopenta[d] [1 ,3]dioxol-5-amine (“ALNY-100”) and/or 4,7,13-tris(3-oxo-3-(undecylamino)propyl)-N1,N16-diundecyl-4,7,10,13-tetraazahexadecane- 1,16-diamide (“NC98-5”). [0330] In some embodiments, the percentage of total cationic lipids in a composition (e.g., a liposomal composition) may be no more than 10%, no more than 20%, no more than 30%, no more than 40%, no more than 50%, no more than 60%, no more than 70%, no more than 80%, no more than 90%, or no more than 95% of total lipids as measured by molar ratios (mol%) or by weight (wt%). [0331] In some embodiments, the percentage of total cationic lipids in a composition (e.g., a liposomal composition) may be greater than 10%, greater than 20%, greater than 30%, greater than 40%, greater than 50%, greater than 60%, greater than 70%, greater than 80%, greater than 90%, or greater than 95% of total lipids as measured by molar ratios (mol%) or by weight (wt%). [0332] In some embodiments, total cationic lipid(s) constitute(s) about 30-50 % (e.g., about 30-45%, about 30-40%, about 35-50%, about 35-45%, or about 35-40%) of the liposome by weight. In some embodiments, the cationic lipid constitutes about 30%, about 35%, about 40 %, about 45%, or about 50% of a composition (e.g., a liposomal composition) by molar ratio. In some embodiments, total cationic lipid(s) constitute(s) about 30-50 % (e.g., about 30-45%, about 30-40%, about 35-50%, about 35-45%, or about 35-40%) of the liposome by weight. In some embodiments, the cationic lipid constitutes about 30%, about 35%, about 40 %, about 45%, or about 50% of a composition (e.g., a liposomal composition) by weight. [0333] In some embodiments, the compositions include one or more cationic lipids that constitute at least about 5%, 10%, 20%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, or 70%, measured by weight, of the total lipid content in the composition, e.g., a lipid nanoparticle. In some embodiments, the compositions include one or more cationic lipids that constitute at least about 5%, 10%, 20%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, or 70%, measured as a mol%, of the total lipid content in the composition, e.g., a lipid nanoparticle. In some embodiments, the compositions include one or more cationic lipids that constitute about 30-70 % (e.g., about 30-65%, about 30-60%, about 30- 55%, about 30-50%, about 30-45%, about 30-40%, about 35-50%, about 35-45%, or about 35-40%), measured by weight, of the total lipid content in the composition, e.g., a lipid nanoparticle. In some embodiments, the compositions include one or more cationic lipids that constitute about 30-70 % (e.g., about 30-65%, about 30-60%, about 30-55%, about 30-50%, about 30-45%, about 30-40%, about 35-50%, about 35-45%, or about 35-40%), measured as mol %, of the total lipid content in the composition, e.g., a lipid nanoparticle.
Non-cationic/Helper Lipids
[0334] Compositions (e.g., liposomal compositions) may also comprise one or more non-cationic
(“helper”) lipids. As used herein, the phrase“non-cationic lipid” refers to any neutral, zwitterionic or anionic lipid. As used herein, the phrase“anionic lipid” refers to any of a number of lipid species that carry a net negative charge at a selected pH, such as physiological pH. Non-cationic lipids include, but are not limited to, distearoylphosphatidylcholine (DSPC), dioleoylphosphatidylcholine (DOPC), dipalmitoylphosphatidylcholine (DPPC), dioleoylphosphatidylglycerol (DOPG),
dipalmitoylphosphatidylglycerol (DPPG), dioleoylphosphatidylethanolamine (DOPE),
palmitoyloleoylphosphatidylcholine (POPC), palmitoyloleoyl-phosphatidylethanolamine (POPE), dioleoyl-phosphatidylethanolamine 4-(N-maleimidomethyl)-cyclohexane-l-carboxylate (DOPE-mal), dipalmitoyl phosphatidyl ethanolamine (DPPE), dimyristoylphosphoethanolamine (DMPE), distearoyl-phosphatidyl-ethanolamine (DSPE), 16-O-monomethyl PE, 16-O-dimethyl PE, 18-1-trans PE, l-stearoyl-2-oleoyl-phosphatidyethanolamine (SOPE), or a mixture thereof. [0335] In embodiments, a non-cationic or helper lipid is dioleoylphosphatidylethanolamine (DOPE). [0336] In some embodiments, a non-cationic lipid is a neutral lipid, i.e., a lipid that does not carry a net charge in the conditions under which the composition is formulated and/or administered. [0337] In some embodiments, a non-cationic lipid may be present in a molar ratio (mol%) of about 5% to about 90%, about 5% to about 70%, about 5% to about 50%, about 5% to about 40%, about 5% to about 30%, about 10 % to about 70%, about 10% to about 50%, or about 10% to about 40% of the total lipids present in a composition. In some embodiments, total non-cationic lipids may be present in a molar ratio (mol%) of about 5% to about 90%, about 5% to about 70%, about 5% to about 50%, about 5% to about 40%, about 5% to about 30%, about 10 % to about 70%, about 10% to about 50%, or about 10% to about 40% of the total lipids present in a composition. In some embodiments, the percentage of non-cationic lipid in a liposome may be greater than about 5 mol%, greater than about 10 mol%, greater than about 20 mol%, greater than about 30 mol%, or greater than about 40 mol%. In some embodiments, the percentage total non-cationic lipids in a liposome may be greater than about 5 mol%, greater than about 10 mol%, greater than about 20 mol%, greater than about 30 mol%, or greater than about 40 mol%. In some embodiments, the percentage of non-cationic lipid in a liposome is no more than about 5 mol%, no more than about 10 mol%, no more than about 20 mol%, no more than about 30 mol%, or no more than about 40 mol%. In some embodiments, the percentage total non-cationic lipids in a liposome may be no more than about 5 mol%, no more than about 10 mol%, no more than about 20 mol%, no more than about 30 mol%, or no more than about 40 mol%. [0338] In some embodiments, a non-cationic lipid may be present in a weight ratio (wt%) of about 5% to about 90%, about 5% to about 70%, about 5% to about 50%, about 5% to about 40%, about 5% to about 30%, about 10 % to about 70%, about 10% to about 50%, or about 10% to about 40% of the total lipids present in a composition. In some embodiments, total non-cationic lipids may be present in a weight ratio (wt%) of about 5% to about 90%, about 5% to about 70%, about 5% to about 50%, about 5% to about 40%, about 5% to about 30%, about 10 % to about 70%, about 10% to about 50%, or about 10% to about 40% of the total lipids present in a composition. In some embodiments, the percentage of non-cationic lipid in a liposome may be greater than about 5 wt%, greater than about 10 wt%, greater than about 20 wt%, greater than about 30 wt%, or greater than about 40 wt%. In some embodiments, the percentage total non-cationic lipids in a liposome may be greater than about 5 wt%, greater than about 10 wt%, greater than about 20 wt%, greater than about 30 wt%, or greater than about 40 wt%. In some embodiments, the percentage of non- cationic lipid in a liposome is no more than about 5 wt%, no more than about 10 wt%, no more than about 20 wt%, no more than about 30 wt%, or no more than about 40 wt%. In some embodiments, the percentage total non-cationic lipids in a liposome may be no more than about 5 wt%, no more than about 10 wt%, no more than about 20 wt%, no more than about 30 wt%, or no more than about 40 wt%.
Cholesterol-based Lipids
[0339] In some embodiments, a composition (e.g., a liposomal composition) comprises one or more cholesterol-based lipids. For example, suitable cholesterol-based lipids include cholesterol and , for example, DC-Chol (N,N-dimethyl-N-ethylcarboxamidocholesterol), 1,4-bis(3-N-oleylamino- propyl)piperazine (Gao, et al. Biochem. Biophys. Res. Comm.179, 280 (1991); Wolf et al.
BioTechniques 23, 139 (1997); U.S. Pat. No.5,744,335), or imidazole cholesterol ester (ICE), which has the following structure,
Figure imgf000111_0001
(“ICE”). [0340] In embodiments, a cholesterol-based lipid is cholesterol. [0341] In some embodiments, a cholesterol-based lipid may be present in a molar ratio (mol%) of about 1% to about 30%, or about 5% to about 20% of the total lipids present in a liposome. In some embodiments, the percentage of cholesterol-based lipid in the lipid nanoparticle may be greater than about 5 mol%, greater than about 10 mol%, greater than about 20 mol%, greater than about 30 mol%, or greater than about 40 mol%. In some embodiments, the percentage of cholesterol- based lipid in the lipid nanoparticle may be no more than about 5 mol%, no more than about 10 mol%, no more than about 20 mol%, no more than about 30 mol%, or no more than about 40 mol%. [0342] In some embodiments, a cholesterol-based lipid may be present in a weight ratio (wt%) of
about 1% to about 30%, or about 5% to about 20% of the total lipids present in a liposome. In some embodiments, the percentage of cholesterol-based lipid in the lipid nanoparticle may be greater than about 5 wt%, greater than about 10 wt%, greater than about 20 wt%, greater than about 30 wt%, or greater than about 40 wt%. In some embodiments, the percentage of cholesterol-based lipid in the lipid nanoparticle may be no more than about 5 wt%, no more than about 10 wt%, no more than about 20 wt%, no more than about 30 wt%, or no more than about 40 wt%.
PEGylated Lipids
[0343] In some embodiments, a composition (e.g., a liposomal composition) comprises one or more PEGylated lipids. [0344] For example, the use of polyethylene glycol (PEG)-modified phospholipids and derivatized lipids such as derivatized ceramides (PEG-CER), including N-octanoyl-sphingosine-1-[succinyl(methoxy polyethylene glycol)-2000] (C8 PEG-2000 ceramide) is also contemplated by the present invention in combination with one or more of the cationic and, in some embodiments, other lipids together which comprise the liposome. In some embodiments, particularly useful exchangeable lipids are PEG-ceramides having shorter acyl chains (e.g., C14 or C18). [0345] Contemplated PEG-modified lipids (also referred to herein as a PEGylated lipid, which term is interchangeable with PEG-modified lipid) include, but are not limited to, a polyethylene glycol chain of up to 5 kDa in length covalently attached to a lipid with alkyl chain(s) of C6-C20 length. In some embodiments, a PEG-modified or PEGylated lipid is PEGylated cholesterol or PEG-2K. The addition of such components may prevent complex aggregation and may also provide a means for increasing circulation lifetime and increasing the delivery of the lipid-nucleic acid composition to the target cell, (Klibanov et al. (1990) FEBS Letters, 268 (1): 235-237), or they may be selected to rapidly exchange out of the formulation in vivo (see U.S. Pat. No.5,885,613). [0346] In embodiments, a PEG-modified lipid is 1,2-dimyristoyl-sn-glycerol, methoxypolyethylene glycol (DMG-PEG2000). [0347] A PEG-modified phospholipid and derivatized lipids of the present invention may be present in a molar ratio (mol%) from about 0% to about 15%, about 0.5% to about 15%, about 1% to about 15%, about 4% to about 10%, or about 2% of the total lipid present in the composition (e.g., a liposomal composition). [0348] A PEG-modified phospholipid and derivatized lipids of the present invention may be present in a weight ratio (wt%) from about 0% to about 15%, about 0.5% to about 15%, about 1% to about 15%, about 4% to about 10%, or about 2% of the total lipid present in the composition (e.g., a liposomal composition).
Pharmaceutical Formulations and Therapeutic Uses
[0349] Cationic lipids described herein (e.g., a cationic lipid of any of Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III-B), (IV-A), (IV-B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)-(156)) may be used in the preparation of compositions (e.g., to construct liposomal compositions) that facilitate or enhance the delivery and release of encapsulated materials (e.g., one or more therapeutic polynucleotides) to one or more target cells (e.g., by permeating or fusing with the lipid membranes of such target cells). [0350] For example, when a liposomal composition (e.g., a lipid nanoparticle) comprises or is otherwise enriched with one or more of the compounds disclosed herein, the phase transition in the lipid bilayer of the one or more target cells may facilitate the delivery of the encapsulated materials (e.g., one or more therapeutic polynucleotides encapsulated in a lipid nanoparticle) into the one or more target cells. [0351] Similarly, in certain embodiments cationic lipids described herein (e.g., a cationic lipid of any of Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III- B), (IV-A), (IV-B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)-(156)) may be used to prepare liposomal vehicles that are characterized by their reduced toxicity in vivo. In certain embodiments, the reduced toxicity is a function of the high transfection efficiencies associated with the compositions disclosed herein, such that a reduced quantity of such composition may administered to the subject to achieve a desired therapeutic response or outcome. [0352] Thus, pharmaceutical formulations comprising a cationic lipid described herein (e.g., a cationic lipid of any of Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II- B), (III-A), (III-B), (IV-A), (IV-B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)- (156)) and nucleic acids provided by the present invention may be used for various therapeutic purposes. To facilitate delivery of nucleic acids in vivo, a cationic lipid described herein (e.g., a cationic lipid of any of Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III-B), (IV-A), (IV-B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)-(156)) and nucleic acids can be formulated in combination with one or more additional pharmaceutical carriers, targeting ligands or stabilizing reagents. In some embodiments, a cationic lipid described herein (e.g., a cationic lipid of any of Formulas (I)-(VI) such as any of Formulas (I-A)-(I- C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III-B), (IV-A), (IV-B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)-(156)) can be formulated via pre-mixed lipid solution. In other embodiments, a composition comprising a cationic lipid described herein (e.g., a cationic lipid of any of Formulas (I)-(VI) such as any of Formulas (I-A)-(I-C), (I-A’)-(I-C’), (I-B”), (I-C”), (II-A), (II-B), (III-A), (III-B), (IV-A), (IV-B), (IV-B’), (IV-B”), (IV-C), (IV-C’), or (IV-C”), or any of Compounds (1)-(156)) can be formulated using post-insertion techniques into the lipid membrane of the nanoparticles.
Techniques for formulation and administration of drugs may be found in“Remington’s
Pharmaceutical Sciences,” Mack Publishing Co., Easton, Pa., latest edition. [0353] Suitable routes of administration include, for example, oral, rectal, vaginal, transmucosal,
pulmonary including intratracheal or inhaled, or intestinal administration; parenteral delivery, including intradermal, transdermal (topical), intramuscular, subcutaneous, intramedullary injections, as well as intrathecal, direct intraventricular, intravenous, intraperitoneal, or intranasal. In particular embodiments, the intramuscular administration is to a muscle selected from the group consisting of skeletal muscle, smooth muscle and cardiac muscle. In some embodiments the administration results in delivery of the nucleic acids to a muscle cell. In some embodiments the administration results in delivery of the nucleic acids to a hepatocyte (i.e., liver cell). In embodiments, administration is intramuscular. In embodiments, administration is intravenous. [0354] Alternatively or additionally, pharmaceutical formulations of the invention may be administered in a local rather than systemic manner, for example, via injection of the pharmaceutical formulation directly into a targeted tissue, preferably in a sustained release formulation. Local delivery can be affected in various ways, depending on the tissue to be targeted. Exemplary tissues in which delivered mRNA may be delivered and/or expressed include, but are not limited to the liver, kidney, heart, spleen, serum, brain, skeletal muscle, lymph nodes, skin, and/or cerebrospinal fluid. In embodiments, the tissue to be targeted in the liver. For example, aerosols containing compositions of the present invention can be inhaled (for nasal, tracheal, or bronchial delivery); compositions of the present invention can be injected into the site of injury, disease manifestation, or pain, for example; compositions can be provided in lozenges for oral, tracheal, or esophageal application; can be supplied in liquid, tablet or capsule form for administration to the stomach or intestines, can be supplied in suppository form for rectal or vaginal application; or can even be delivered to the eye by use of creams, drops, or even injection. [0355] In embodiments, administration is via pulmonary delivery. As used herein, pulmonary delivery refers to delivery to lung via, e.g., nasal cavity, trachea, bronchi, bronchioles, and/or other pulmonary system. In embodiments, a composition described herein is formulated for nebulization. In embodiments, the delivery vehicle may be in an aerosolized composition which can be inhaled. In embodiments, pulmonary delivery involves inhalation (e.g., for nasal, tracheal, or bronchial delivery). In embodiments, a composition is nebulized prior to inhalation. [0356] The present invention provides methods for delivering a composition having full-length mRNA molecules encoding a peptide or polypeptide of interest for use in the treatment of a subject, e.g., a human subject or a cell of a human subject or a cell that is treated and delivered to a human subject. [0357] Accordingly, in certain embodiments the present invention provides a method for producing a therapeutic composition comprising full-length mRNA that encodes a peptide or polypeptide for use in the delivery to or treatment of the lung of a subject or a lung cell. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for cystic fibrosis transmembrane conductance regulator (CFTR) protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for ATP-binding cassette sub-family A member 3 protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for dynein axonemal intermediate chain 1 protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for dynein axonemal heavy chain 5 (DNAH5) protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for alpha-1-antitrypsin protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for forkhead box P3 (FOXP3) protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes one or more surfactant protein, e.g., one or more of surfactant A protein, surfactant B protein, surfactant C protein, and surfactant D protein. [0358] In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes a peptide or polypeptide for use in the delivery to or treatment of the liver of a subject or a liver cell. Such peptides and polypeptides can include those associated with a urea cycle disorder, associated with a lysosomal storage disorder, with a glycogen storage disorder, associated with an amino acid metabolism disorder, associated with a lipid metabolism or fibrotic disorder, associated with methylmalonic acidemia, or associated with any other metabolic disorder for which delivery to or treatment of the liver or a liver cell with enriched full-length mRNA provides therapeutic benefit. [0359] In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for a protein associated with a urea cycle disorder. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for ornithine transcarbamylase (OTC) protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for arginosuccinate synthetase 1 protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for carbamoyl phosphate synthetase I protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for arginosuccinate lyase protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for arginase protein. [0360] In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for a protein associated with a lysosomal storage disorder. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for alpha galactosidase protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for glucocerebrosidase protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for iduronate-2-sulfatase protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for iduronidase protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for N- acetyl-alpha-D-glucosaminidase protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for heparan N-sulfatase protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for galactosamine-6 sulfatase protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for beta-galactosidase protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for lysosomal lipase protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for arylsulfatase B (N-acetylgalactosamine-4-sulfatase) protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for transcription factor EB (TFEB). [0361] In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for a protein associated with a glycogen storage disorder. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for acid alpha-glucosidase protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for glucose-6-phosphatase (G6PC) protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for liver glycogen phosphorylase protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for muscle phosphoglycerate mutase protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for glycogen debranching enzyme. [0362] In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for a protein associated with amino acid metabolism. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for phenylalanine hydroxylase enzyme. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for glutaryl-CoA dehydrogenase enzyme. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for propionyl-CoA caboxylase enzyme. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for oxalase alanine-glyoxylate aminotransferase enzyme. [0363] In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for a protein associated with a lipid metabolism or fibrotic disorder. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for a mTOR inhibitor. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for ATPase phospholipid transporting 8B1 (ATP8B1) protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for one or more NF-kappa B inhibitors, such as one or more of I-kappa B alpha, interferon-related development regulator 1 (IFRD1), and Sirtuin 1 (SIRT1). In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for PPAR-gamma protein or an active variant. [0364] In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for a protein associated with methylmalonic acidemia. For example, in certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for methylmalonyl CoA mutase protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for methylmalonyl CoA epimerase protein. [0365] In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA for which delivery to or treatment of the liver can provide therapeutic benefit. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for ATP7B protein, also known as Wilson disease protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for porphobilinogen deaminase enzyme. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for one or clotting enzymes, such as Factor VIII, Factor IX, Factor VII, and Factor X. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for human hemochromatosis (HFE) protein. [0366] In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes a peptide or polypeptide for use in the delivery to or treatment of the cardiovasculature of a subject or a cardiovascular cell. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for vascular endothelial growth factor A protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for relaxin protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for bone morphogenetic protein-9 protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for bone morphogenetic protein-2 receptor protein. [0367] In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes a peptide or polypeptide for use in the delivery to or treatment of the muscle of a subject or a muscle cell. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for dystrophin protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for frataxin protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes a peptide or polypeptide for use in the delivery to or treatment of the cardiac muscle of a subject or a cardiac muscle cell. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for a protein that modulates one or both of a potassium channel and a sodium channel in muscle tissue or in a muscle cell. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for a protein that modulates a Kv7.1 channel in muscle tissue or in a muscle cell. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for a protein that modulates a Nav1.5 channel in muscle tissue or in a muscle cell. [0368] In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes a peptide or polypeptide for use in the delivery to or treatment of the nervous system of a subject or a nervous system cell. For example, in certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for survival motor neuron 1 protein. For example, in certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for survival motor neuron 2 protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for frataxin protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for ATP binding cassette subfamily D member 1 (ABCD1) protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for CLN3 protein. [0369] In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes a peptide or polypeptide for use in the delivery to or treatment of the blood or bone marrow of a subject or a blood or bone marrow cell. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for beta globin protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for Bruton’s tyrosine kinase protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for one or clotting enzymes, such as Factor VIII, Factor IX, Factor VII, and Factor X. [0370] In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes a peptide or polypeptide for use in the delivery to or treatment of the kidney of a subject or a kidney cell. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for collagen type IV alpha 5 chain (COL4A5) protein. [0371] In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes a peptide or polypeptide for use in the delivery to or treatment of the eye of a subject or an eye cell. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for ATP-binding cassette sub-family A member 4 (ABCA4) protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for retinoschisin protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for retinal pigment epithelium-specific 65 kDa (RPE65) protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for centrosomal protein of 290 kDa (CEP290). [0372] In embodiments, an mRNA encodes for an antigen from an infectious agent. [0373] In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes a peptide or polypeptide for use in the delivery of or treatment with a vaccine for a subject or a cell of a subject. For example, in certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antigen from an infectious agent, such as a virus. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antigen from influenza virus. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antigen from respiratory syncytial virus. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antigen from rabies virus. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antigen from cytomegalovirus. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antigen from rotavirus. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antigen from a hepatitis virus, such as hepatitis A virus, hepatitis B virus, or hepatis C virus. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antigen from human papillomavirus. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antigen from a herpes simplex virus, such as herpes simplex virus 1 or herpes simplex virus 2. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antigen from a human immunodeficiency virus, such as human immunodeficiency virus type 1 or human immunodeficiency virus type 2. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antigen from a human metapneumovirus. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antigen from a human parainfluenza virus, such as human parainfluenza virus type 1, human parainfluenza virus type 2, or human parainfluenza virus type 3. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antigen from malaria virus. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antigen from zika virus. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antigen from chikungunya virus. [0374] In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antigen associated with a cancer of a subject or identified from a cancer cell of a subject. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antigen determined from a subject’s own cancer cell, i.e., to provide a personalized cancer vaccine. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antigen expressed from a mutant KRAS gene. [0375] In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antibody. In certain embodiments, the antibody can be a bi-specific antibody. In certain embodiments, the antibody can be part of a fusion protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antibody to OX40. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antibody to VEGF. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antibody to tissue necrosis factor alpha. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antibody to CD3. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antibody to CD19. [0376] In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an immunomodulator. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for Interleukin 12. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for Interleukin 23. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for Interleukin 36 gamma. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for a constitutively active variant of one or more stimulator of interferon genes (STING) proteins. [0377] In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an endonuclease. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an RNA-guided DNA endonuclease protein, such as Cas 9 protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for a meganuclease protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for a transcription activator-like effector nuclease protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for a zinc finger nuclease protein. [0378] In embodiments, exemplary therapeutic uses result from the delivery of mRNA encoding a secreted protein. Accordingly, in embodiments, the compositions and methods of the invention provide for delivery of mRNA encoding a secreted protein. In some embodiments, the compositions and methods of the invention provide for delivery of mRNA encoding one or more secreted proteins listed in Table 1; thus, compositions of the invention may comprise an mRNA encoding a protein listed in Table 1 (or a homolog thereof) along with other components set out herein, and methods of the invention may comprise preparing and/or administering a composition comprising an mRNA encoding a protein listed in Table 1 (or a homolog thereof) along with other components set out herein Table 1. Secreted Proteins
Figure imgf000122_0001
Figure imgf000123_0001
Figure imgf000124_0001
Figure imgf000125_0001
Figure imgf000126_0001
Figure imgf000127_0001
Figure imgf000128_0001
Figure imgf000129_0001
Figure imgf000130_0001
Figure imgf000131_0001
Figure imgf000132_0001
Figure imgf000133_0001
Figure imgf000134_0001
Figure imgf000135_0001
Figure imgf000136_0001
Figure imgf000137_0001
Figure imgf000138_0001
Figure imgf000139_0001
Figure imgf000140_0001
Figure imgf000141_0001
Figure imgf000142_0001
Figure imgf000143_0001
Figure imgf000144_0001
Figure imgf000145_0001
Figure imgf000146_0001
Figure imgf000147_0001
Figure imgf000148_0001
Figure imgf000149_0001
Figure imgf000150_0001
Figure imgf000151_0001
Figure imgf000152_0001
Figure imgf000153_0001
Figure imgf000154_0001
Figure imgf000155_0001
Figure imgf000156_0001
Figure imgf000157_0001
Figure imgf000158_0001
Figure imgf000159_0001
Figure imgf000160_0001
Figure imgf000161_0001
Figure imgf000162_0001
Figure imgf000163_0001
Figure imgf000164_0001
Figure imgf000165_0001
Figure imgf000166_0001
Figure imgf000167_0001
Figure imgf000168_0001
Figure imgf000169_0001
[0379] In some embodiments, the compositions and methods of the invention provide for the delivery of one or more mRNAs encoding one or more additional exemplary proteins listed in Table 2; thus, compositions of the invention may comprise an mRNA encoding a protein listed in Table 2 (or a homolog thereof) along with other components set out herein, and methods of the invention may comprise preparing and/or administering a composition comprising an mRNA encoding a protein chosen from the proteins listed in Table 2 (or a homolog thereof) along with other components set out herein. Table 2. Additional Exemplary Proteins
Figure imgf000170_0001
[0380] The Uniprot IDs set forth in Table 1 and Table 2 refer to the human versions the listed proteins and the sequences of each are available from the Uniprot database. Sequences of the listed proteins are also generally available for various animals, including various mammals and animals of veterinary or industrial interest. Accordingly, in some embodiments, compositions and methods of the invention provide for the delivery of one or more mRNAs encoding one or more proteins chosen from mammalian homologs or homologs from an animal of veterinary or industrial interest of the secreted proteins listed in Table 1 and Table 2; thus, compositions of the invention may comprise an mRNA encoding a protein chosen from mammalian homologs or homologs from an animal of veterinary or industrial interest of a protein listed in Table 1 and Table 2 along with other components set out herein, and methods of the invention may comprise preparing and/or administering a composition comprising an mRNA encoding a protein chosen from mammalian homologs or homologs from an animal of veterinary or industrial interest of a protein listed in Table 1 and Table 2 along with other components set out herein. In some embodiments, mammalian homologs are chosen from mouse, rat, hamster, gerbil, horse, pig, cow, llama, alpaca, mink, dog, cat, ferret, sheep, goat, or camel homologs. In some embodiments, the animal of veterinary or industrial interest is chosen from the mammals listed above and/or chicken, duck, turkey, salmon, catfish, or tilapia. [0381] In embodiments, the compositions and methods of the invention provide for the delivery of mRNA encoding a lysosomal protein chosen from Table 3. In some embodiments, the compositions and methods of the invention provide for the delivery of one or more mRNAs encoding one or more lysosomal and/or related proteins listed in Table 3; thus, compositions of the invention may comprise an mRNA encoding a protein listed in Table 3 (or a homolog thereof) along with other components set out herein, and methods of the invention may comprise preparing and/or administering a composition comprising an mRNA encoding a protein chosen from the proteins listed in Table 3 (or a homolog thereof) along with other components set out herein. Table 3. Lysosomal and Related Proteins
Figure imgf000171_0001
Figure imgf000172_0001
Figure imgf000173_0001
[0382] Information regarding lysosomal proteins is available from Lubke et al.,“Proteomics of the Lysosome,” Biochim Biophys Acta. (2009) 1793: 625–635. In some embodiments, the protein listed in Table 3 and encoded by mRNA in the compositions and methods of the invention is a human protein. Sequences of the listed proteins are also available for various animals, including various mammals and animals of veterinary or industrial interest as described above. [0383] In some embodiments, the compositions and methods of the invention provide for the delivery of mRNA encoding a therapeutic protein (e.g., cytosolic, transmembrane or secreted) such as those listed in Table 4. In some embodiments, the compositions and methods of the invention provide for the delivery of an mRNA encoding a therapeutic protein useful in treating a disease or disorder (i.e., indication) listed in Table 4; thus, compositions of the invention may comprise an mRNA encoding a therapeutic protein listed or not listed in Table 4 (or a homolog thereof, as discussed below) along with other components set out herein for treating a disease or disorder (i.e., indication) listed in Table 4, and methods of the invention may comprise preparing and/or administering a composition comprising an mRNA encoding a such a protein (or a homolog thereof, as discussed below) along with other components set out herein for treatment of a disease or disorder listed in Table 4. Table 4. Exemplary Indications and Related Proteins
Figure imgf000173_0002
Figure imgf000174_0001
Figure imgf000175_0001
Figure imgf000176_0001
Figure imgf000177_0001
Figure imgf000178_0001
Figure imgf000179_0001
Figure imgf000180_0001
Figure imgf000181_0001
Figure imgf000182_0001
[0384] In some embodiments, the present invention is used to prevent, treat and/or cure a subject affected with a disease or disorder listed or associated with the proteins listed in Tables 1, 2, 3, or 4. In some embodiments, an mRNA encodes one or more of Cystic Fibrosis Transmembrane
Conductance Regulator (CFTR), argininosuccinate synthetase (ASS1), Factor IX, survival motor neuron 1 (SMN1), or phenylalanine hydroxylase (PAH). [0385] While certain compounds, compositions and methods of the present invention have been
described with specificity in accordance with certain embodiments, the following examples serve only to illustrate the compounds of the invention and are not intended to limit the same. EXAMPLES
Example 1: Synthesis of Cationic Lipids
[0386] Cationic lipids described herein can be prepared according to the exemplary synthesis of
Scheme 1. [0387] In embodiments, a cationic lipid described herein can be prepared by conjugating a thiol with a di-carboxylic acid under suitable conditions. Exemplary di-carboxylic acids are described in Table A, and exemplary thiols are described in Table B. Accordingly, suitable cationic lipids include those resulting from any combination of the precursors described in Table A and Table B. Table A. Dicarboxylic Acids
Figure imgf000183_0001
Table B. Thiols
Figure imgf000184_0001
Figure imgf000185_0001
Figure imgf000186_0001
Figure imgf000187_0001
Figure imgf000188_0001
Example 2: Exemplary Synthesis of Compound 16
[0388] Cationic lipids described herein can be prepared according to the exemplary synthesis of Scheme 2. An exemplary synthesis for Compound 16 is provided herein. Scheme 2
Figure imgf000189_0001
Synthesis of 2-(4-(Benzylthio)butyl)isoindoline-1,3-dione (D2)
Figure imgf000190_0001
[0389] To a mixture of sodium hydride (1.76 g, 44 mmol, 60% dispersion in mineral oil) in 60 mL N, N- dimethylformamide was added benzyl mercaptan (4.96 g, 40 mmol) at 0 oC. After stirring for 30 min, a solution of N-(4-bromobutyl)phthalimide D1 (12 g, 42.5 mmol) in 20 mL N, N-dimethylformamide was added, and the resulting mixture was allowed to warm slowly to room temperature and stirred overnight. The reaction mixture was quenched by methanol and saturated NH4Cl solution, and then extracted with ethyl acetate. The organic layer was washed by water (300 mL x 3), and then dried over Na2SO4. After filtration and concentration, the crude was purified by flash column
chromatography (330 g silica gel column, 0-60% ethyl acetate in hexane gradient) to give 10.5 g product D2 as white solid (Yield: 81%). Synthesis of 4-(Benzylthio)butan-1-amine (D3)
Figure imgf000190_0002
[0390] A mixture of 2-(4-(benzylthio)butyl)isoindoline-1,3-dione D2 (10.5 g, 32.5 mmol) and hydrazine hydrate (3.8 mL, 67 mmol) in methanol (300 mL) was heated under nitrogen atmosphere to gentle reflux for 4 h. After cooled to room temperature, the reaction mixture was filtered through Celite, and then washed with methanol (30 mL). The combined filtrate was concentrated under reduced pressure to afford 11.5 g (~ 50% purity) of crude product D3 as white solid, which was used for next step without purification. Synthesis of 1,1'-((4-(Benzylthio)butyl)azanediyl)bis(dodecan-2-ol) (D5)
Figure imgf000190_0003
[0391] A mixture of 4-(benzylthio)butan-1-amine D3 (11.5 g, ~ 50% pure, ~29.5 mmol),
diisopropylethylamine (15 mL) and 1,2-epoxydodecane D4 (26 g, 141 mmol) in 210 mL methanol was heated under nitrogen atmosphere to gentle reflux for 20 h. The reaction mixture was concentrated, and the crude was purified by flash column chromatography (330 g silica gel column, 0-40% ethyl acetate in hexane gradient) to give 12 g of product D5 as colorless oil (Yield: 66%, 2 steps). Synthesis of N-(4-(Benzylthio)butyl)-2-((tert-butyldimethylsilyl)oxy)-N-(2-((tert- butyldimethylsilyl)oxy)dodecyl)dodecan-1-amine (D6)
Figure imgf000191_0001
[0392] To a solution of 1,1'-((4-(benzylthio)butyl)azanediyl)bis(dodecan-2-ol) 5 (11.5 g, 29.5 mmol) in 60 mL anhydrous N, N-dimethylformamide was added imidazole (8.6 g, 127 mmol) and DMAP (0.5 g, 4 mmol), followed by tert-butyldimethylsilyl chloride (12.8 g, 85 mmol), and then the resulting mixture was stirred at room temperature under nitrogen atmosphere for 48 h. The reaction mixture was concentrated under reduced pressure, and the residue was partitioned between CH2Cl2 (300 mL) and water (300 mL). The organic layer was separated, and the aqueous layer was extracted with CH2Cl2 (300 mLx2). The combined organic phase was dried over Na2SO4. After filtration and concentration, the crude was purified by flash column chromatography (330 g silica gel column, 0- 25% ethyl acetate in hexane gradient) to give 13.6 g of product D6 as colorless oil (Yield: 81%). Synthesis of 4-(Bis(2-((tert-butyldimethylsilyl)oxy)dodecyl)amino)butane-1-thiol (D7)
Figure imgf000191_0002
[0393] In a 1 L three neck round bottom flask equipped with magnetic stirring bar, dry ice-acetone condenser and nitrogen inlet, N-(4-(benzylthio)butyl)-2-((tert-butyldimethylsilyl)oxy)-N-(2-((tert- butyldimethylsilyl)oxy)dodecyl)dodecan-1-amine D6 (13.6 g, 17 mmol) was dissolved in 250 mL anhydrous diethyl ether, which was then cooled to -78 °C in an dry ice-acetone bath Liquid ammonia (400 mL) was condensed into the reaction flask, and then sodium (1 g, 43 mmol) was then added in portions under nitrogen atmosphere to form a dark blue solution. After addition, the reaction was stirred at this temperature for one hour. The reaction was quenched by adding solid NH4Cl (20 g, 373 mmol). The dry ice-acetone bath was replaced with a water bath, and then the solvent was removed by blowing nitrogen gas. The white solid residue was triturated with diethyl ether (300 mL x 4). After concentration, the crude was purified by flash column chromatography (330 g silica gel column, 0-25% ethyl acetate in hexane gradient) to give 8.6 g product D7 as colorless oil (Yield: 71%). Synthesis of S,S-Bis(4-(bis(2-((tert-butyldimethylsilyl)oxy)dodecyl)amino)butyl) butanebis(thioate) (D9)
Figure imgf000192_0001
[0394] To a mixture of 4-(bis(2-((tert-butyldimethylsilyl)oxy)dodecyl)amino)butane-1-thiol 7 (600 mg, 0.86 mmol) and succinic acid D8 (50 mg, 0.43 mmol) in 15 mL CH2Cl2 was added DMAP (52 mg, 0.43 mmol) and EDCI (330 mg, 1.7 mmol). The resulting mixture was stirred at room temperature for 18 h. The reaction mixture was concentrated, and the crude was purified by flash column
chromatography (80 g silica gel column, 0-15% ethyl acetate in hexane gradient) to give 560 mg product D9 as colorless oil (Yield: 88%). Synthesis of S,S-Bis(4-(bis(2-hydroxydodecyl)amino)butyl) butanebis(thioate) (Cationic Lipid 16)
Figure imgf000192_0002
[0395] In a 100 ml Teflon flask, to a solution of S,S-bis(4-(bis(2-((tert- butyldimethylsilyl)oxy)dodecyl)amino)butyl) butanebis(thioate) D9 (560 mg, 0.38 mmol) in 16 mL anhydrous tetrahydrofuran was added HF-pyridine solution (8 mL, 308 mmol, 70%wt) dropwise at 0 o C, and the resulting mixture was allowed to warm slowly to room temperature and stirred overnight. The reaction mixture was diluted with CH2Cl2 (50 mL) and neutralized with aqueous Na2CO3 solution. The CH2Cl2 layer was separated, and the aqueous layer was extracted with CH2Cl2 (30 mLx3). The combined organic phase was dried over Na2SO4 and evaporated. The residue was purified by flash column chromatography (12 g silica gel column pre-deactivated with 1% triethylamine in hexane, eluent: 0-100% ethyl acetate in hexane gradient) to give 336 mg of product Cationic Lipid 16 as colorless oil (Yield: 85%). [0396] Other cationic lipids can be prepared according the same reaction procedures as described
above but with different tails derived from different epoxides D4.
Example 3: Lipid Nanoparticle Formulation Using Thioester Cationic Lipids
[0397] Cationic lipids described herein can be used in the preparation of lipid nanoparticles according to methods known in the art. For example, suitable methods include methods described in
International Publication No. WO 2018/089801, which is hereby incorporated by reference in its entirety. [0398] One exemplary process for lipid nanoparticle formulation is Process A of WO 2018/089801 (see, e.g., Example 1 and Figure 1 of WO 2018/089801). Process A (“A”) relates to a conventional method of encapsulating mRNA by mixing mRNA with a mixture of lipids, without first pre-forming the lipids into lipid nanoparticles. In an exemplary process, an ethanol lipid solution and an aqueous buffered solution of mRNA were prepared separately. A solution of mixture of lipids (cationic lipid, helper lipids, zwitterionic lipids, PEG lipids etc.) was prepared by dissolving lipids in ethanol. The mRNA solution was prepared by dissolving the mRNA in citrate buffer, resulting in mRNA at a
concentration of 0.0833mg/ml in citrate buffer with a pH of 4.5. The mixtures were then both heated to 65 °C prior to mixing. Then, these two solutions were mixed using a pump system. In some instances, the two solutions were mixed using a gear pump system. In certain embodiments, the two solutions were mixing using a‘T’ junction (or“Y” junction). The mixture was then purified by diafiltration with a TFF process. The resultant formulation concentrated and stored at 2-8 °C until further use. [0399] A second exemplary process for lipid nanoparticle formulation is Process B of WO 2018/089801 (see, e.g., Example 2 and Figure 2 of WO 2018/089801). Process B (“B”) refers to a process of encapsulating messenger RNA (mRNA) by mixing pre-formed lipid nanoparticles with mRNA. A range of different conditions, such as varying temperatures (i.e., heating or not heating the mixture), buffers, and concentrations, may be employed in Process B. In an exemplary process, lipids dissolved in ethanol and citrate buffer were mixed using a pump system. The instantaneous mixing of the two streams resulted in the formation of empty lipid nanoparticles, which was a self- assembly process. The resultant formulation mixture was empty lipid nanoparticles in citrate buffer containing alcohol. The formulation was then subjected to a TFF purification process wherein buffer exchange occurred. The resulting suspension of pre-formed empty lipid nanoparticles was then mixed with mRNA using a pump system. For certain cationic lipids, heating the solution post-mixing resulted in a higher percentage of lipid nanoparticles containing mRNA and a higher total yield of mRNA. [0400] Lipid nanoparticle formulations of Table 5 were prepared by using either Process A or Process B as described in WO 2018/089801. All of the lipid nanoparticle formulations comprised hEPO mRNA and the different lipids in following mol % ratios: Cationic Lipid: DMG-PEG2000; Cholesterol: DOPE = 40:5:25:30. Table 5. Exemplary lipid nanoparticle formulations
Figure imgf000194_0001
Example 4: In Vivo Expression of hEPO in CD1 Mice using Thioester Lipids
[0401] Intravenous (IV) administration of lipid nanoparticle formulations comprising a thioester cationic lipid and mRNA encoding hEPO (Table 5) was undertaken in order to study mRNA delivery and resultant hEPO expression. Male CD1 mice at 6-8 weeks old were given a single intravenous injection of the LNP formulations at a dosage level of 1 mg/kg. Blood samples were collected by tail snip at 6 and 24 hours post-dose. hEPO protein expression levels measured in the sera samples by ELISA (FIG.1). These studies show that thioester cationic lipids described herein are highly effective at delivery mRNA in vivo, resulting in high expression of the protein or polypeptide encoded by the delivered mRNA.

Claims

CLAIMS WHAT IS CLAIMED IS:
1. A cationic lipid having a structure according to Formula (I),
Figure imgf000195_0002
wherein
R1 is hydrogen, C6-C30 alkyl, C6-C30 alkenyl, or C6-C30 alkynyl, or Substructure Y;
each a and b is an integer of 0-6;
each XA1 and XB1 is independently O or S;
each LA and LB is independently C1-C10 alkylene; C2-C10 alkenylene; or C2-C10 alkynylene; XA2 is independently NH, NRA, CH2, or CHRA;
XB2 is independently NH, NRB, CH2, or CHRB;
each RA and RB is independently C6-C30 alkyl, C6-C30 alkenyl, or C6-C30 alkynyl; and
Substructure
Figure imgf000195_0001
.
2. The cationic lipid of claim 1, having a structure according to Formula (I-A),
Figure imgf000195_0003
3. The cationic lipid of claim 1, having a structure according to Formula (I-A’):
Figure imgf000196_0002
4. The cationic lipid of any one of claims 1-3, wherein each XA1 and XB1 is O, or each XA1 and XB1 is S.
5. The cationic lipid of claim 4, wherein each XA1 and XB1 is O.
6. The cationic lipid of any one of claims 1-5, wherein each a and b is independently 0, 1, or 2.
7. The cationic lipid of any one of claims 1-6, wherein each XA2 is NRA or CHRA.
8. The cationic lipid of any one of claims 1-7, wherein each XB2 is NRB or CHRB.
9. The cationic lipid of any one of claims 1-8, having a structure according to Formula (I-B),
Figure imgf000196_0001
10. The cationic lipid of any one of claims 1-8, having a structure according to Formula (I-B”):
Figure imgf000196_0003
wherein d is independently an integer of 0-5.
11. The cationic lipid of any one of claims 1-8, having a structure according to Formula (I-B’):
Figure imgf000197_0004
12. The cationic lipid of any one of claims 1-11, wherein each LA is C1-C10 alkylene.
13. The cationic lipid of any one of claims 1-12, wherein each LB is C1-C10 alkylene.
14. The cationic lipid of any one of claims 1-13, wherein each LA and LB is unsubstituted C1-C10 alkylene.
15. The cationic lipid of any one of claims 12-14, having a structure according to Formula (I-C),
Figure imgf000197_0001
wherein each c is independently an integer of 2-10.
16. The cationic lipid of any one of claims 12-14, having a structure according to Formula (I-C”),
Figure imgf000197_0002
wherein each c is independently an integer of 2-10, and d is independently an integer of 0-5.
17. The cationic lipid of claim 16, wherein d is 0, 1, 2, 3, or 4.
18. The cationic lipid of any one of claims 12-14, having a structure according to Formula (I-C’),
Figure imgf000197_0003
wherein each c is independently an integer of 2-10.
19. The cationic lipid of any one of claims 15-18, wherein each c is 2, 3, or 4.
20. The cationic lipid of any one of claims 15-18, wherein each c is 4, 5, 6, 7, 8, 9, or 10.
21. The cationic lipid of claim 20, wherein each c is 4.
22. The cationic lipid of any one of claims 1-21, wherein each RA is C6-C20 alkyl or C6-C20 alkenyl.
23. The cationic lipid of claim 22, wherein each RA is unsubstituted C6-C20 alkyl, C6-C20 hydroxyalkyl, unsubstituted C6-C20 alkenyl, or C6-C20 hydroxyalkenyl.
24. The cationic lipid of any one of claims 1-23, wherein each RB is C6-C20 alkyl or C6-C20 alkenyl.
25. The cationic lipid of claim 24, wherein each RB is unsubstituted C6-C20 alkyl, C6-C20 hydroxyalkyl, unsubstituted C6-C20 alkenyl, or C6-C20 hydroxyalkenyl.
26. The cationic lipid of any one of claims 1-25, wherein each RA and RB is C6-C20 hydroxyalkyl, or each RA and RB is C6-C20 hydroxyalkenyl.
27. The cationic lipid of claim 26, wherein each RA and RB is -CH2CH(OH)C10H21, or each RA and RB
is -CH2CH(OH)(CH2)6(CH=CH)CH2(CH=CH)C5H11.
28. The cationic lipid of claim 27, wherein each RA and RB is -CH2CH(OH)C10H21.
29. The cationic lipid of claim 26, wherein each RA and RB is -CH2CH(OH)RC, and wherein RC is selected from the group consisting of:
Figure imgf000198_0001
30. The cationic lipid of claim 1, having the following structure,
Figure imgf000199_0001
31. The cationic lipid of claim 1, having the following structure,
Figure imgf000199_0002
32. A cationic lipid having a structure according to Formula (II),
Figure imgf000199_0003
wherein
each a is independently an integer of 1-5;
b is independently an integer of 0-6;
each XA1 and XB1 is independently O or S;
each LA and LB is independently C1-C10 alkylene; C2-C10 alkenylene; or C2-C10 alkynylene; each XA2 is independently NH, NRA, CH2, or CHRA;
XB2 is independently NH, NRB, CH2, or CHRB; and
each RA and RB is independently C6-C30 alkyl, C6-C30 alkenyl, or C6-C30 alkynyl.
33. The cationic lipid of claim 32, wherein each XA1 and XB1 is O, or each XA1 and XB1 is S.
34. The cationic lipid of claim 33, wherein each XA1 and XB1 is O.
35. The cationic lipid of any one of claims 32-34, wherein each a is independently 1, 2, or 3; and b is independently 0, 1, or 2.
36. The cationic lipid of claim 35, wherein each a is 1, and b is 0.
37. The cationic lipid of any one of claims 32-36, wherein each XA2 is NRA or CHRA.
38. The cationic lipid of any one of claims 32-37, wherein each XB2 is NRB or CHRB.
39. The cationic lipid of any one of claims 32-38, having a structure according to Formula (II-A),
Figure imgf000200_0001
40. The cationic lipid of any one of claims 32-39, wherein each LA is C1-C10 alkylene.
41. The cationic lipid of any one of claims 32-40, wherein each LB is C1-C10 alkylene.
42. The cationic lipid of any one of claims 32-41, wherein each LA and LB is unsubstituted C1-C10 alkylene.
43. The cationic lipid of any one of claims 32-42, having a structure according to Formula (II-B),
Figure imgf000200_0002
wherein each c is independently an integer of 2-10.
44. The cationic lipid of claim 43, wherein each c is 2, 3, or 4.
45. The cationic lipid of claim 43, wherein each c is 4, 5, 6, 7, 8, 9, or 10.
46. The cationic lipid of claim 45, wherein each c is 4.
47. The cationic lipid of any one of claims 32-46, wherein each RA is C6-C20 alkyl or C6-C20 alkenyl.
48. The cationic lipid of claim 47, wherein each RA is unsubstituted C6-C20 alkyl, C6-C20 hydroxyalkyl, unsubstituted C6-C20 alkenyl, or C6-C20 hydroxyalkenyl.
49. The cationic lipid of any one of claims 32-48, wherein each RB is C6-C20 alkyl or C6-C20 alkenyl.
50. The cationic lipid of claim 49, wherein each RB is unsubstituted C6-C20 alkyl, C6-C20 hydroxyalkyl, unsubstituted C6-C20 alkenyl, or C6-C20 hydroxyalkenyl.
51. The cationic lipid of any one of claims 32-50, wherein each RA and RB is C6-C20 hydroxyalkyl, or each RA and RB is C6-C20 hydroxyalkenyl.
52. The cationic lipid of claim 51, wherein each RA and RB is -CH2CH(OH)C10H21, or each RA and RB is -CH2CH(OH)(CH2)6(CH=CH)CH2(CH=CH)C5H11.
53. The cationic lipid of claim 52, wherein each RA and RB is -CH2CH(OH)C10H21.
54. The cationic lipid of claim 51, wherein each RA and RB is-CH2CH(OH)RC, and wherein RC is selected from the group consisting of:
Figure imgf000201_0001
55. The cationic lipid of claim 32, having the following structure,
Figure imgf000202_0001
56. A cationic lipid having a structure according to Formula (III):
wherein
Figure imgf000202_0002
each a is independently an integer of 0-6;
each XA1 is independently O or S;
each LA is independently C1-C10 alkylene; C2-C10 alkenylene; or C2-C10 alkynylene;
XA2 is independently NH, NRA, CH2, or CHRA; and
each RA is independently C6-C30 alkyl, C6-C30 alkenyl, or C6-C30 alkynyl.
57. The cationic lipid of claim 56, wherein each XA1 is O, or each XA1 is S.
58. The cationic lipid of claim 57, wherein each XA1 is O.
59. The cationic lipid of any one of claims 56-58, wherein each a is independently 0, 1, or 2.
60. The cationic lipid of claim 59, wherein each a is 1.
61. The cationic lipid of any one of claims 56-60, wherein each XA2 is NRA or CHRA.
62. The cationic lipid of any one of claims 56-61, having a structure according to Formula (III-A),
Figure imgf000203_0001
(III-A).
63. The cationic lipid of any one of claims 56-62, wherein each LA is C1-C10 alkylene.
64. The cationic lipid of any one of claims 56-63, wherein each LA is unsubstituted C1-C10 alkylene.
65. The cationic lipid of any one of claims 56-64, having a structure according to Formula (III-B),
Figure imgf000203_0002
(III-B), wherein each c is independently an integer of 2-10.
66. The cationic lipid of claim 65, wherein each c is 2, 3, or 4.
67. The cationic lipid of claim 65, wherein each c is 4, 5, 6, 7, 8, 9, or 10.
68. The cationic lipid of claim 67, wherein each c is 4.
69. The cationic lipid of any one of claims 56-68, wherein each RA is C6-C20 alkyl or C6-C20 alkenyl.
70. The cationic lipid of claim 69, wherein each RA is unsubstituted C6-C20 alkyl, C6-C20 hydroxyalkyl, unsubstituted C6-C20 alkenyl, or C6-C20 hydroxyalkenyl.
71. The cationic lipid of any one of claims 56-70, wherein each RA is C6-C20 hydroxyalkyl, or each RA is C6- C20 hydroxyalkenyl.
72. The cationic lipid of claim 71, wherein each RA is -CH2CH(OH)C10H21, or each RA
is -CH2CH(OH)(CH2)6(CH=CH)CH2(CH=CH)C5H11.
73. The cationic lipid of claim 72, wherein each RA is -CH2CH(OH)C10H21.
74. The cationic lipid of claim 71, wherein each RA is-CH2CH(OH)RC, and wherein RC is selected from the group consisting of:
Figure imgf000204_0001
.
75. The cationic lipid of claim 56, having the structure,
Figure imgf000204_0002
76. A cationic lipid having a structure according to Formula (IV),
Figure imgf000205_0001
,
wherein
R1 is hydrogen, C1-C30 alkyl, C2-C30 alkenyl, or C2-C30 alkynyl;
each a and b is an integer of 0-6;
each XA1 and XB1 is independently O or S;
each LA and LB is independently C1-C10 alkylene; C2-C10 alkenylene; or C2-C10 alkynylene; LC is independently–C(O)– or–(CH2)b–;
XA2 is independently NH, NRA, CH2, or CHRA;
XB2 is independently NH, NRB, CH2, or CHRB; and
each RA and RB is independently C6-C30 alkyl, C6-C30 alkenyl, or C6-C30 alkynyl. 77. The cationic lipid of claim 76, wherein LC is–(CH2)b–. 78. The cationic lipid of claim 76, having a structure according to Formula (IV-A):
Figure imgf000205_0002
79. The cationic lipid of any one of claims 76-78, wherein each XA1 and XB1 is O, or each XA1 and XB1 is S.
80. The cationic lipid of claim 79, wherein each XA1 and XB1 is O.
81. The cationic lipid of any one of claims 76-80, wherein each a and b is independently 0, 1, or 2.
82. The cationic lipid of any one of claims 76-81, wherein each XA2 is NRA or CHRA.
83. The cationic lipid of any one of claims 76-82, wherein each XB2 is NRB or CHRB.
84. The cationic lipid of any one of claims 76-83, having a structure according to Formula (IV-B):
Figure imgf000206_0001
85. The cationic lipid of any one of claims 76-83, having a structure according to Formula (IV-B’):
Figure imgf000206_0002
86. The cationic lipid of any one of claims 76-85, wherein each LA is C1-C10 alkylene.
87. The cationic lipid of any one of claims 76-86, wherein each LB is C1-C10 alkylene.
88. The cationic lipid of any one of claims 76-87, wherein each LA and LB is unsubstituted C1-C10 alkylene.
89. The cationic lipid of any one of claims 86-88, having a structure according to Formula (IV-C):
Figure imgf000206_0003
wherein each c is independently an integer of 2-10.
90. The cationic lipid of any one of claims 86-88, having a structure according to Formula (IV-C’):
Figure imgf000206_0004
wherein each c is independently an integer of 2-10.
91. The cationic lipid of claim 76, wherein LC is–C(O)–.
92. The cationic lipid of claim 91, having a structure according to Formula (IV-B”),
Figure imgf000207_0001
93. The cationic lipid of claim 92, wherein each LA is C1-C10 alkylene.
94. The cationic lipid of claim 92 or 93, wherein each LB is C1-C10 alkylene.
95. The cationic lipid of any one of claims 92-94, wherein each LA and LB is unsubstituted C1-C10 alkylene.
96. The cationic lipid of claim 95, having a structure according to Formula (IV-C”):
Figure imgf000207_0002
wherein each c is independently an integer of 2-10.
97. The cationic lipid of any one of claims 89, 90, and 96 wherein each c is 2, 3, or 4.
98. The cationic lipid of any one of claims 89, 90, and 96 wherein each c is 4, 5, 6, 7, 8, 9, or 10.
99. The cationic lipid of claim 98, wherein each c is 4.
100. The cationic lipid of any one of claims 76-99, wherein each RA is C6-C20 alkyl or C6-C20 alkenyl.
101. The cationic lipid of claim 100, wherein each RA is unsubstituted C6-C20 alkyl, C6-C20 hydroxyalkyl, unsubstituted C6-C20 alkenyl, or C6-C20 hydroxyalkenyl.
102. The cationic lipid of any one of claims 76-101, wherein each RB is C6-C20 alkyl or C6-C20 alkenyl.
103. The cationic lipid of claim 102, wherein each RB is unsubstituted C6-C20 alkyl, C6-C20 hydroxyalkyl, unsubstituted C6-C20 alkenyl, or C6-C20 hydroxyalkenyl.
104. The cationic lipid of any one of claims 76-103, wherein each RA and RB is C6-C20 hydroxyalkyl, or each RA and RB is C6-C20 hydroxyalkenyl.
105. The cationic lipid of claim 104, wherein each RA and RB is -CH2CH(OH)C10H21, or each RA and RB is -CH2CH(OH)(CH2)6(CH=CH)CH2(CH=CH)C5H11.
106. The cationic lipid of claim 105, wherein each RA and RB is -CH2CH(OH)C10H21. 107. The cationic lipid of claim 104, wherein each RA and RB is-CH2CH(OH)RC, and wherein RC is selected from the group consisting of:
Figure imgf000208_0001
108. The cationic lipid of claim 76, having the following structure:
Figure imgf000208_0002
109. A cationic lipid having a structure according to Formula (V),
Figure imgf000209_0001
wherein
each LA and LB is independently C1-C10 alkylene; C2-C10 alkenylene; or C2-C10 alkynylene; and
each RA and RB is independently C6-C30 alkyl, C6-C30 alkenyl, C6-C30 alkynyl, or C1-C15 alkylene-C(O)2-C1-C15 alkyl.
110. The compound of claim 109, wherein each LA and LB is independently C1-C10 alkylene.
111. The compound of claim 109, wherein each LA and LB is independently C2-C10 alkenylene or C1-C10 alkylene-C(O)2- C1-C10 alkylene.
112. A cationic lipid according to Formula (VI),
Figure imgf000209_0002
wherein
each RA and RB is independently C1-C30 alkyl, C2-C30 alkenyl, C2-C30 alkynyl, or C1-C15 alkylene-C(O)2- C1-C15 alkyl; and
each c is independently an integer of 2-10.
113. The cationic lipid of claim 112, wherein each c is 2, 3, or 4.
114. The cationic lipid of claim 112, wherein each c is 4, 5, 6, 7, 8, 9, or 10.
115. The cationic lipid of claim 114, wherein each c is 4.
116. The cationic lipid of any one of claims 112-115, wherein each RA and RB is unsubstituted C6-C30 alkenyl.
117. The cationic lipid of any one of claims 112-115, wherein each RA and RB is (unsubstituted C3-C15 alkylene)-C(O)2-(unsubstituted C3-C15 alkyl).
118. The cationic lipid of any one of claims 112-115, wherein each RA and RB is-CH2CH(OH)RC, and wherein RC is selected from the group consisting of:
Figure imgf000210_0001
119. A cationic lipid that is any one of cationic lipids 1-156.
120. A cationic lipid that is:
Figure imgf000210_0002
121. A composition comprising an mRNA encoding a peptide or a polypeptide, encapsulated within a liposome, wherein the liposome comprises one or more cationic lipids, one or more non-cationic lipids, one or more cholesterol-based lipids and one or more PEG-modified lipids, wherein at least one cationic lipid is according to of any one of claims 1-120.
122. The composition of claim 121, comprising an mRNA encoding for cystic fibrosis transmembrane conductance regulator (CFTR) protein.
123. The composition of claim 121, comprising an mRNA encoding for ornithine transcarbamylase (OTC) protein.
124. The composition of claim 121, comprising an mRNA encoding for an antigen.
125. The composition of claim 124, wherein the antigen is from an infectious agent.
126. A composition comprising a nucleic acid encapsulated within a liposome, wherein the liposome comprises a cationic lipid according to of any one of claims 1-120.
127. The composition of claim 126, further comprising one or more lipids selected from the group consisting of one or more cationic lipids, one or more non-cationic lipids, and one or more PEG- modified lipids.
128. The composition of claim 126 or 127, wherein the nucleic acid is an mRNA encoding a peptide or polypeptide.
129. The composition of any one of claims 126-128, wherein the mRNA encodes a peptide or
polypeptide for use in the delivery to or treatment of the lung of a subject or a lung cell.
130. The composition of claim 129, wherein the mRNA encodes cystic fibrosis transmembrane
conductance regulator (CFTR) protein.
131. The composition of any one of claims 126-128, wherein the mRNA encodes a peptide or
polypeptide for use in the delivery to or treatment of the liver of a subject or a liver cell.
132. The composition of claim 131, wherein the mRNA encodes ornithine transcarbamylase (OTC) protein.
133. The composition of any one of claims 126-128, wherein the mRNA encodes a peptide or
polypeptide for use in a vaccine.
134. The composition of claim 133, wherein the mRNA encodes an antigen.
135. The composition of claim 134, wherein the antigen is from an infectious agent.
136. The composition of any one of claims 121-135, formulated for intravenous (IV) administration.
137. The composition of any one of claims 121-135, formulated for intramuscular (IM)
administration.
138. The composition of any one of claims 121-135, formulated for administration by inhalation.
139. The composition of claim 138, wherein the composition is formulated for nebulization.
PCT/US2019/033806 2018-05-24 2019-05-23 Thioester cationic lipids WO2019226925A1 (en)

Priority Applications (7)

Application Number Priority Date Filing Date Title
JP2020565307A JP7488193B2 (en) 2018-05-24 2019-05-23 Thioester Cationic Lipids
CN201980048008.6A CN112437767B (en) 2018-05-24 2019-05-23 thioester cationic lipids
CA3100254A CA3100254A1 (en) 2018-05-24 2019-05-23 Thioester cationic lipids
AU2019275068A AU2019275068A1 (en) 2018-05-24 2019-05-23 Thioester cationic lipids
CN202311296793.1A CN117430538A (en) 2018-05-24 2019-05-23 Thioester cationic lipids
US17/057,929 US20220008338A1 (en) 2018-05-24 2019-05-23 Thioester Cationic Lipids
EP19731398.4A EP3802487A1 (en) 2018-05-24 2019-05-23 Thioester cationic lipids

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US201862676147P 2018-05-24 2018-05-24
US62/676,147 2018-05-24
US201862748097P 2018-10-19 2018-10-19
US62/748,097 2018-10-19
US201862750013P 2018-10-24 2018-10-24
US62/750,013 2018-10-24

Publications (1)

Publication Number Publication Date
WO2019226925A1 true WO2019226925A1 (en) 2019-11-28

Family

ID=66912933

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2019/033806 WO2019226925A1 (en) 2018-05-24 2019-05-23 Thioester cationic lipids

Country Status (7)

Country Link
US (1) US20220008338A1 (en)
EP (1) EP3802487A1 (en)
JP (1) JP7488193B2 (en)
CN (2) CN117430538A (en)
AU (1) AU2019275068A1 (en)
CA (1) CA3100254A1 (en)
WO (1) WO2019226925A1 (en)

Cited By (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020227085A1 (en) * 2019-05-03 2020-11-12 Translate Bio, Inc. Di-thioester cationic lipids
WO2020254535A1 (en) 2019-06-18 2020-12-24 Curevac Ag Rotavirus mrna vaccine
WO2020257611A1 (en) * 2019-06-21 2020-12-24 Translate Bio, Inc. Cationic lipids comprising an hydroxy moiety
WO2021123332A1 (en) 2019-12-20 2021-06-24 Curevac Ag Lipid nanoparticles for delivery of nucleic acids
WO2021156267A1 (en) 2020-02-04 2021-08-12 Curevac Ag Coronavirus vaccine
WO2021202694A1 (en) * 2020-04-01 2021-10-07 Translate Bio, Inc. Phenolic acid lipid based cationic lipids
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
WO2022043551A2 (en) 2020-08-31 2022-03-03 Curevac Ag Multivalent nucleic acid based coronavirus vaccines
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
WO2022207862A2 (en) 2021-03-31 2022-10-06 Curevac Ag Syringes containing pharmaceutical compositions comprising rna
US11471525B2 (en) 2020-02-04 2022-10-18 Curevac Ag Coronavirus vaccine
WO2022233880A1 (en) 2021-05-03 2022-11-10 Curevac Ag Improved nucleic acid sequence for cell type specific expression
WO2023009421A1 (en) * 2021-07-26 2023-02-02 Modernatx, Inc. Processes for preparing lipid nanoparticle 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
WO2023073228A1 (en) 2021-10-29 2023-05-04 CureVac SE Improved circular rna for expressing therapeutic proteins
WO2023144330A1 (en) 2022-01-28 2023-08-03 CureVac SE Nucleic acid encoded transcription factor inhibitors
WO2023227608A1 (en) 2022-05-25 2023-11-30 Glaxosmithkline Biologicals Sa Nucleic acid based vaccine encoding an escherichia coli fimh antigenic polypeptide
US11872280B2 (en) 2020-12-22 2024-01-16 CureVac SE RNA vaccine against SARS-CoV-2 variants
DE202023106198U1 (en) 2022-10-28 2024-03-21 CureVac SE Nucleic acid-based vaccine
WO2024089229A1 (en) 2022-10-28 2024-05-02 CureVac SE Improved formulations comprising lipid-based carriers encapsulating rna

Families Citing this family (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP7448488B2 (en) * 2018-05-15 2024-03-12 トランスレイト バイオ, インコーポレイテッド Subcutaneous delivery of messenger RNA
KR20210093232A (en) 2018-10-09 2021-07-27 더 유니버시티 오브 브리티시 콜롬비아 Compositions and systems and related methods comprising transfection competent vesicles free of organic solvent and detergent
CN115710196A (en) * 2021-08-23 2023-02-24 广州谷森制药有限公司 Novel cationic lipid compounds
CN115304756B (en) * 2022-01-30 2023-05-09 上海科技大学 Five-membered lipid nanoparticle as well as preparation method and application thereof
CN114380724B (en) * 2022-03-23 2022-05-24 深圳市瑞吉生物科技有限公司 Cationic lipid compounds and compositions for delivery of nucleic acids and uses
CN115286523B (en) * 2022-07-15 2024-03-19 中国科学院基础医学与肿瘤研究所(筹) Lipid molecules for delivery of active ingredients and preparation and use of compositions thereof
CN117088825A (en) * 2023-10-12 2023-11-21 成都威斯津生物医药科技有限公司 Ionizable lipid, pharmaceutical composition containing same and application thereof

Citations (41)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4373071A (en) 1981-04-30 1983-02-08 City Of Hope Research Institute Solid-phase synthesis of polynucleotides
US4401796A (en) 1981-04-30 1983-08-30 City Of Hope Research Institute Solid-phase synthesis of polynucleotides
US4415732A (en) 1981-03-27 1983-11-15 University Patents, Inc. Phosphoramidite compounds and processes
US4458066A (en) 1980-02-29 1984-07-03 University Patents, Inc. Process for preparing polynucleotides
US4500707A (en) 1980-02-29 1985-02-19 University Patents, Inc. Nucleosides useful in the preparation of polynucleotides
US4668777A (en) 1981-03-27 1987-05-26 University Patents, Inc. Phosphoramidite nucleoside compounds
US4897355A (en) 1985-01-07 1990-01-30 Syntex (U.S.A.) Inc. N[ω,(ω-1)-dialkyloxy]- and N-[ω,(ω-1)-dialkenyloxy]-alk-1-yl-N,N,N-tetrasubstituted ammonium lipids and uses therefor
US4973679A (en) 1981-03-27 1990-11-27 University Patents, Inc. Process for oligonucleo tide synthesis using phosphormidite intermediates
US5047524A (en) 1988-12-21 1991-09-10 Applied Biosystems, Inc. Automated system for polynucleotide synthesis and purification
US5132418A (en) 1980-02-29 1992-07-21 University Patents, Inc. Process for preparing polynucleotides
US5153319A (en) 1986-03-31 1992-10-06 University Patents, Inc. Process for preparing polynucleotides
US5171678A (en) 1989-04-17 1992-12-15 Centre National De La Recherche Scientifique Lipopolyamines, their preparation and their use
US5262530A (en) 1988-12-21 1993-11-16 Applied Biosystems, Inc. Automated system for polynucleotide synthesis and purification
US5334761A (en) 1992-08-28 1994-08-02 Life Technologies, Inc. Cationic lipids
US5700642A (en) 1995-05-22 1997-12-23 Sri International Oligonucleotide sizing using immobilized cleavable primers
US5744335A (en) 1995-09-19 1998-04-28 Mirus Corporation Process of transfecting a cell with a polynucleotide mixed with an amphipathic compound and a DNA-binding protein
US5885613A (en) 1994-09-30 1999-03-23 The University Of British Columbia Bilayer stabilizing components and their use in forming programmable fusogenic liposomes
WO2005121348A1 (en) 2004-06-07 2005-12-22 Protiva Biotherapeutics, Inc. Lipid encapsulated interfering rna
WO2010042877A1 (en) 2008-10-09 2010-04-15 Tekmira Pharmaceuticals Corporation Improved amino lipids and methods for the delivery of nucleic acids
WO2010053572A2 (en) 2008-11-07 2010-05-14 Massachusetts Institute Of Technology Aminoalcohol lipidoids and uses thereof
WO2010144740A1 (en) 2009-06-10 2010-12-16 Alnylam Pharmaceuticals, Inc. Improved lipid formulation
WO2011141705A1 (en) * 2010-05-12 2011-11-17 Protiva Biotherapeutics, Inc. Novel cationic lipids and methods of use thereof
US8093367B2 (en) 2007-10-31 2012-01-10 Applied Biosystems, Llc Preparation and isolation of 5′ capped mRNA
US8304529B2 (en) 2006-07-28 2012-11-06 Life Technologies Corporation Dinucleotide MRNA cap analogs
WO2012170889A1 (en) 2011-06-08 2012-12-13 Shire Human Genetic Therapies, Inc. Cleavable lipids
WO2013063468A1 (en) 2011-10-27 2013-05-02 Massachusetts Institute Of Technology Amino acid derivates functionalized on the n- terminal capable of forming drug incapsulating microspheres
WO2013149140A1 (en) 2012-03-29 2013-10-03 Shire Human Genetic Therapies, Inc. Ionizable cationic lipids
WO2015095340A1 (en) 2013-12-19 2015-06-25 Novartis Ag Lipids and lipid compositions for the delivery of active agents
WO2015184256A2 (en) 2014-05-30 2015-12-03 Shire Human Genetic Therapies, Inc. Biodegradable lipids for delivery of nucleic acids
WO2015199952A1 (en) 2014-06-25 2015-12-30 Acuitas Therapeutics Inc. Novel lipids and lipid nanoparticle formulations for delivery of nucleic acids
WO2016004202A1 (en) 2014-07-02 2016-01-07 Massachusetts Institute Of Technology Polyamine-fatty acid derived lipidoids and uses thereof
US20160031928A1 (en) 2013-03-14 2016-02-04 Shire Human Genetic Therapies, Inc. RIBONUCLEIC ACIDs WITH 4'-THIO-MODIFIED NUCLEOTIDES AND RELATED METHODS
WO2016118725A1 (en) 2015-01-23 2016-07-28 Moderna Therapeutics, Inc. Lipid nanoparticle compositions
WO2016118724A1 (en) 2015-01-21 2016-07-28 Moderna Therapeutics, Inc. Lipid nanoparticle compositions
WO2016205691A1 (en) 2015-06-19 2016-12-22 Massachusetts Institute Of Technology Alkenyl substituted 2,5-piperazinediones and their use in compositions for delivering an agent to a subject or cell
WO2017004143A1 (en) 2015-06-29 2017-01-05 Acuitas Therapeutics Inc. Lipids and lipid nanoparticle formulations for delivery of nucleic acids
WO2017049245A2 (en) 2015-09-17 2017-03-23 Modernatx, Inc. Compounds and compositions for intracellular delivery of therapeutic agents
WO2017075531A1 (en) 2015-10-28 2017-05-04 Acuitas Therapeutics, Inc. Novel lipids and lipid nanoparticle formulations for delivery of nucleic acids
WO2017117528A1 (en) 2015-12-30 2017-07-06 Acuitas Therapeutics, Inc. Lipids and lipid nanoparticle formulations for delivery of nucleic acids
WO2017173054A1 (en) 2016-03-30 2017-10-05 Intellia Therapeutics, Inc. Lipid nanoparticle formulations for crispr/cas components
WO2018089801A1 (en) 2016-11-10 2018-05-17 Translate Bio, Inc. Improved process of preparing mrna-loaded lipid nanoparticles

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2710300A (en) * 1953-08-07 1955-06-07 Gulf Research Development Co Dithiol adipates
FR2396545A1 (en) * 1977-07-08 1979-02-02 Elf Aquitaine PROTECTION OF THE SKIN AGAINST ULTRAVIOLET RAYS
CN1524899A (en) * 2003-02-28 2004-09-01 中国石油天然气股份有限公司 Special cross-linked polyethylene material for rotational molding and preparing process thereof
CN1274759C (en) * 2004-10-12 2006-09-13 四川大学 Weatherability type polyformaldehyde material and its preparation method
WO2013086373A1 (en) * 2011-12-07 2013-06-13 Alnylam Pharmaceuticals, Inc. Lipids for the delivery of active agents
DE21212055T1 (en) * 2011-12-07 2022-08-04 Alnylam Pharmaceuticals, Inc. BIODEGRADABLE LIPIDS TO RELEASE ACTIVE INGREDIENTS

Patent Citations (41)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5132418A (en) 1980-02-29 1992-07-21 University Patents, Inc. Process for preparing polynucleotides
US4458066A (en) 1980-02-29 1984-07-03 University Patents, Inc. Process for preparing polynucleotides
US4500707A (en) 1980-02-29 1985-02-19 University Patents, Inc. Nucleosides useful in the preparation of polynucleotides
US4415732A (en) 1981-03-27 1983-11-15 University Patents, Inc. Phosphoramidite compounds and processes
US4668777A (en) 1981-03-27 1987-05-26 University Patents, Inc. Phosphoramidite nucleoside compounds
US4973679A (en) 1981-03-27 1990-11-27 University Patents, Inc. Process for oligonucleo tide synthesis using phosphormidite intermediates
US4401796A (en) 1981-04-30 1983-08-30 City Of Hope Research Institute Solid-phase synthesis of polynucleotides
US4373071A (en) 1981-04-30 1983-02-08 City Of Hope Research Institute Solid-phase synthesis of polynucleotides
US4897355A (en) 1985-01-07 1990-01-30 Syntex (U.S.A.) Inc. N[ω,(ω-1)-dialkyloxy]- and N-[ω,(ω-1)-dialkenyloxy]-alk-1-yl-N,N,N-tetrasubstituted ammonium lipids and uses therefor
US5153319A (en) 1986-03-31 1992-10-06 University Patents, Inc. Process for preparing polynucleotides
US5262530A (en) 1988-12-21 1993-11-16 Applied Biosystems, Inc. Automated system for polynucleotide synthesis and purification
US5047524A (en) 1988-12-21 1991-09-10 Applied Biosystems, Inc. Automated system for polynucleotide synthesis and purification
US5171678A (en) 1989-04-17 1992-12-15 Centre National De La Recherche Scientifique Lipopolyamines, their preparation and their use
US5334761A (en) 1992-08-28 1994-08-02 Life Technologies, Inc. Cationic lipids
US5885613A (en) 1994-09-30 1999-03-23 The University Of British Columbia Bilayer stabilizing components and their use in forming programmable fusogenic liposomes
US5700642A (en) 1995-05-22 1997-12-23 Sri International Oligonucleotide sizing using immobilized cleavable primers
US5744335A (en) 1995-09-19 1998-04-28 Mirus Corporation Process of transfecting a cell with a polynucleotide mixed with an amphipathic compound and a DNA-binding protein
WO2005121348A1 (en) 2004-06-07 2005-12-22 Protiva Biotherapeutics, Inc. Lipid encapsulated interfering rna
US8304529B2 (en) 2006-07-28 2012-11-06 Life Technologies Corporation Dinucleotide MRNA cap analogs
US8093367B2 (en) 2007-10-31 2012-01-10 Applied Biosystems, Llc Preparation and isolation of 5′ capped mRNA
WO2010042877A1 (en) 2008-10-09 2010-04-15 Tekmira Pharmaceuticals Corporation Improved amino lipids and methods for the delivery of nucleic acids
WO2010053572A2 (en) 2008-11-07 2010-05-14 Massachusetts Institute Of Technology Aminoalcohol lipidoids and uses thereof
WO2010144740A1 (en) 2009-06-10 2010-12-16 Alnylam Pharmaceuticals, Inc. Improved lipid formulation
WO2011141705A1 (en) * 2010-05-12 2011-11-17 Protiva Biotherapeutics, Inc. Novel cationic lipids and methods of use thereof
WO2012170889A1 (en) 2011-06-08 2012-12-13 Shire Human Genetic Therapies, Inc. Cleavable lipids
WO2013063468A1 (en) 2011-10-27 2013-05-02 Massachusetts Institute Of Technology Amino acid derivates functionalized on the n- terminal capable of forming drug incapsulating microspheres
WO2013149140A1 (en) 2012-03-29 2013-10-03 Shire Human Genetic Therapies, Inc. Ionizable cationic lipids
US20160031928A1 (en) 2013-03-14 2016-02-04 Shire Human Genetic Therapies, Inc. RIBONUCLEIC ACIDs WITH 4'-THIO-MODIFIED NUCLEOTIDES AND RELATED METHODS
WO2015095340A1 (en) 2013-12-19 2015-06-25 Novartis Ag Lipids and lipid compositions for the delivery of active agents
WO2015184256A2 (en) 2014-05-30 2015-12-03 Shire Human Genetic Therapies, Inc. Biodegradable lipids for delivery of nucleic acids
WO2015199952A1 (en) 2014-06-25 2015-12-30 Acuitas Therapeutics Inc. Novel lipids and lipid nanoparticle formulations for delivery of nucleic acids
WO2016004202A1 (en) 2014-07-02 2016-01-07 Massachusetts Institute Of Technology Polyamine-fatty acid derived lipidoids and uses thereof
WO2016118724A1 (en) 2015-01-21 2016-07-28 Moderna Therapeutics, Inc. Lipid nanoparticle compositions
WO2016118725A1 (en) 2015-01-23 2016-07-28 Moderna Therapeutics, Inc. Lipid nanoparticle compositions
WO2016205691A1 (en) 2015-06-19 2016-12-22 Massachusetts Institute Of Technology Alkenyl substituted 2,5-piperazinediones and their use in compositions for delivering an agent to a subject or cell
WO2017004143A1 (en) 2015-06-29 2017-01-05 Acuitas Therapeutics Inc. Lipids and lipid nanoparticle formulations for delivery of nucleic acids
WO2017049245A2 (en) 2015-09-17 2017-03-23 Modernatx, Inc. Compounds and compositions for intracellular delivery of therapeutic agents
WO2017075531A1 (en) 2015-10-28 2017-05-04 Acuitas Therapeutics, Inc. Novel lipids and lipid nanoparticle formulations for delivery of nucleic acids
WO2017117528A1 (en) 2015-12-30 2017-07-06 Acuitas Therapeutics, Inc. Lipids and lipid nanoparticle formulations for delivery of nucleic acids
WO2017173054A1 (en) 2016-03-30 2017-10-05 Intellia Therapeutics, Inc. Lipid nanoparticle formulations for crispr/cas components
WO2018089801A1 (en) 2016-11-10 2018-05-17 Translate Bio, Inc. Improved process of preparing mrna-loaded lipid nanoparticles

Non-Patent Citations (18)

* Cited by examiner, † Cited by third party
Title
BEHR ET AL., PROC. NAT.'L ACAD. SCI., vol. 86, 1989, pages 6982
FEIGNER ET AL., PROC. NAT'L ACAD. SCI., vol. 84, 1987, pages 7413
GAO ET AL., BIOCHEM. BIOPHYS. RES. COMM., vol. 179, 1991, pages 280
GRUDZIEN, E. ET AL., RNA, vol. 10, 2004, pages 1479 - 1487
GRUDZIEN-NOGALSKA, E. ET AL., RNA, vol. 13, 2007, pages 1745 - 1755
HEYES, J. ET AL., J CONTROLLED RELEASE, vol. 107, 2005, pages 276 - 287
J. MCCLELLANM. C. KING, CELL, vol. 141, 2010, pages 210 - 217
JEMIELITY, J. ET AL., RNA, vol. 9, 2003, pages 1108 - 1122
JEMIELITY, J. ET AL.: "Novel 'anti-reverse' cap analogs with superior translational properties", RNA, vol. 9, 2003, pages 1108 - 1122, XP002466761, DOI: doi:10.1261/rna.5430403
KLIBANOV ET AL., FEBS LETTERS, vol. 268, no. 1, 1990, pages 235 - 237
LASIC, TRENDS BIOTECHNOL., vol. 16, 1998, pages 307 - 321
LUBKE ET AL.: "Proteomics of the Lysosome", BIOCHIM BIOPHYS ACTA, vol. 1793, 2009, pages 625 - 635, XP026073291, DOI: doi:10.1016/j.bbamcr.2008.09.018
MORRISSEY, DV. ET AL., NAT. BIOTECHNOL., vol. 23, no. 8, 2005, pages 1003 - 1007
S. M. BERGE ET AL.: "pharmaceutically acceptable salts", J. PHARMACEUTICAL SCIENCES, vol. 66, 1977, pages 1 - 19
SEMPLE ET AL., NATURE BIOTECH., vol. 28, 2010, pages 172 - 176
WHITEHEAD ET AL., NATURE COMMUNICATIONS, vol. 5, 2014, pages 4277
WOLF ET AL., BIOTECHNIQUES, vol. 23, 1997, pages 139
YOKOE ET AL., NATURE BIOTECHNOLOGY, vol. 14, 1996, pages 1252 - 1256

Cited By (35)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020227085A1 (en) * 2019-05-03 2020-11-12 Translate Bio, Inc. Di-thioester cationic lipids
WO2020254535A1 (en) 2019-06-18 2020-12-24 Curevac Ag Rotavirus mrna vaccine
WO2020257611A1 (en) * 2019-06-21 2020-12-24 Translate Bio, Inc. Cationic lipids comprising an hydroxy moiety
WO2021123332A1 (en) 2019-12-20 2021-06-24 Curevac Ag Lipid nanoparticles for delivery of nucleic acids
DE202021004123U1 (en) 2020-02-04 2022-10-26 Curevac Ag Coronavirus Vaccine
US11964012B2 (en) 2020-02-04 2024-04-23 CureVac SE Coronavirus vaccine
DE112021000012T5 (en) 2020-02-04 2021-11-18 Curevac Ag Coronavirus vaccine
US11596686B2 (en) 2020-02-04 2023-03-07 CureVac SE Coronavirus vaccine
DE202021003575U1 (en) 2020-02-04 2022-01-17 Curevac Ag Coronavirus Vaccine
DE202021004130U1 (en) 2020-02-04 2022-10-26 Curevac Ag Coronavirus Vaccine
WO2021156267A1 (en) 2020-02-04 2021-08-12 Curevac Ag Coronavirus vaccine
US11576966B2 (en) 2020-02-04 2023-02-14 CureVac SE Coronavirus vaccine
US11471525B2 (en) 2020-02-04 2022-10-18 Curevac Ag Coronavirus vaccine
US11964011B2 (en) 2020-02-04 2024-04-23 CureVac SE Coronavirus vaccine
EP4147717A1 (en) 2020-02-04 2023-03-15 CureVac SE Coronavirus vaccine
WO2021202694A1 (en) * 2020-04-01 2021-10-07 Translate Bio, Inc. Phenolic acid lipid based cationic lipids
CN115667207A (en) * 2020-04-01 2023-01-31 翻译生物公司 Cationic lipids based on phenolic acid lipids
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
WO2022043551A2 (en) 2020-08-31 2022-03-03 Curevac Ag Multivalent nucleic acid based coronavirus vaccines
WO2022135993A2 (en) 2020-12-22 2022-06-30 Curevac Ag Pharmaceutical composition comprising lipid-based carriers encapsulating rna for multidose administration
US11872280B2 (en) 2020-12-22 2024-01-16 CureVac SE RNA vaccine against SARS-CoV-2 variants
WO2022137133A1 (en) 2020-12-22 2022-06-30 Curevac Ag Rna vaccine against sars-cov-2 variants
US11918643B2 (en) 2020-12-22 2024-03-05 CureVac SE RNA vaccine against SARS-CoV-2 variants
WO2022162027A2 (en) 2021-01-27 2022-08-04 Curevac Ag Method of reducing the immunostimulatory properties of in vitro transcribed rna
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
WO2023009421A1 (en) * 2021-07-26 2023-02-02 Modernatx, Inc. Processes for preparing lipid nanoparticle compositions
WO2023031394A1 (en) 2021-09-03 2023-03-09 CureVac SE Novel lipid nanoparticles for delivery of nucleic acids
WO2023031392A2 (en) 2021-09-03 2023-03-09 CureVac SE Novel lipid nanoparticles for delivery of nucleic acids comprising phosphatidylserine
WO2023073228A1 (en) 2021-10-29 2023-05-04 CureVac SE Improved circular rna for expressing therapeutic proteins
WO2023144330A1 (en) 2022-01-28 2023-08-03 CureVac SE Nucleic acid encoded transcription factor inhibitors
WO2023227608A1 (en) 2022-05-25 2023-11-30 Glaxosmithkline Biologicals Sa Nucleic acid based vaccine encoding an escherichia coli fimh antigenic polypeptide
DE202023106198U1 (en) 2022-10-28 2024-03-21 CureVac SE Nucleic acid-based vaccine
WO2024089229A1 (en) 2022-10-28 2024-05-02 CureVac SE Improved formulations comprising lipid-based carriers encapsulating rna

Also Published As

Publication number Publication date
CN117430538A (en) 2024-01-23
JP7488193B2 (en) 2024-05-21
JP2021525240A (en) 2021-09-24
EP3802487A1 (en) 2021-04-14
CA3100254A1 (en) 2019-11-28
CN112437767B (en) 2023-10-27
AU2019275068A1 (en) 2020-12-03
US20220008338A1 (en) 2022-01-13
CN112437767A (en) 2021-03-02

Similar Documents

Publication Publication Date Title
JP7488193B2 (en) Thioester Cationic Lipids
AU2019278813B2 (en) Cationic lipids comprising a steroidal moiety
JP7384832B2 (en) Ribose cationic lipid
AU2019277355A1 (en) Phosphoester cationic lipids
EP3802507A1 (en) Vitamin cationic lipids
CN114401942B (en) Tris (hydroxymethyl) methylglycine and citrate lipids
WO2020257611A1 (en) Cationic lipids comprising an hydroxy moiety
EP3962902A1 (en) Di-thioester cationic lipids
EP3956303A1 (en) Cystine cationic lipids
WO2020106903A1 (en) Cationic lipid compounds and compositions thereof for use in the delivery of messenger rna
EP3959195A1 (en) Thioester cationic lipids
EP4149556A1 (en) Peg lipidoid compounds
WO2020243540A1 (en) Macrocyclic lipids
WO2020097376A1 (en) Multi-peg lipid compounds

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: 19731398

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 3100254

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2020565307

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2019275068

Country of ref document: AU

Date of ref document: 20190523

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2019731398

Country of ref document: EP

Effective date: 20210111