WO2020257611A1 - Lipides cationiques comprenant une fraction hydroxy - Google Patents

Lipides cationiques comprenant une fraction hydroxy Download PDF

Info

Publication number
WO2020257611A1
WO2020257611A1 PCT/US2020/038678 US2020038678W WO2020257611A1 WO 2020257611 A1 WO2020257611 A1 WO 2020257611A1 US 2020038678 W US2020038678 W US 2020038678W WO 2020257611 A1 WO2020257611 A1 WO 2020257611A1
Authority
WO
WIPO (PCT)
Prior art keywords
chch
lipid
protein
mrna
composition
Prior art date
Application number
PCT/US2020/038678
Other languages
English (en)
Inventor
Shrirang KARVE
Frank Derosa
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.
Publication of WO2020257611A1 publication Critical patent/WO2020257611A1/fr

Links

Classifications

    • 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
    • A61K31/713Double-stranded nucleic acids or oligonucleotides
    • 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
    • A61K47/16Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing nitrogen, e.g. nitro-, nitroso-, azo-compounds, nitriles, cyanates
    • A61K47/18Amines; Amides; Ureas; Quaternary ammonium compounds; Amino acids; Oligopeptides having up to five amino acids
    • 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
    • A61K47/16Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing nitrogen, e.g. nitro-, nitroso-, azo-compounds, nitriles, cyanates
    • A61K47/18Amines; Amides; Ureas; Quaternary ammonium compounds; Amino acids; Oligopeptides having up to five amino acids
    • A61K47/186Quaternary ammonium compounds, e.g. benzalkonium chloride or cetrimide
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/127Liposomes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • C12N15/88Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation using microencapsulation, e.g. using amphiphile liposome vesicle
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/53DNA (RNA) vaccination
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55555Liposomes; Vesicles, e.g. nanoparticles; Spheres, e.g. nanospheres; Polymers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies

Definitions

  • mRNA messenger RNA
  • the present invention provides, among other things, lipids useful in for delivery of mRNA.
  • 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.
  • the present invention provides lipids (e.g., a lipid of Formula (I) or (II)) described herein.
  • the present invention provides a liposome encapsulating an mRNA encoding a protein wherein the liposome comprises a lipid described herein (e.g., a lipid of Formula (I) or (I I)).
  • a lipid described herein e.g., a lipid of Formula (I) or (I I)
  • the present invention provides a nucleic acid encapsulated within a
  • the liposome comprises a lipid described herein (e.g., a lipid of Formula (I) or
  • a lipid has a structure according to Formula (I):
  • L 1 is independently C 2 -Ci 0 alkylene, C -Ci 0 alkenylene, or C -Ci 0 alkynylene;
  • X is independently OR 1 , SR 1 , or NfR 1 ) ⁇
  • R is H or unsubstituted Ci-C alkyl
  • each R and R is independently C-C o-alkyl, C-C o-alkenyl, or C-C -alkynyl.
  • X is OH
  • L is C-Ci alkylene
  • L is unsubstituted C-Ci alkylene.
  • a lipid has a structure according to Formula (II),
  • n is an integer of - .
  • n is 2, 3, 4, or 5.
  • each R and R is unsubstituted C -C -alkyl, unsubstituted C -C -alkenyl, or unsubstituted C -C -alkynyl.
  • each R and R is unsubstituted C -C -alkyl, or each R and R is
  • each of R and R is -C HI 3 , -C HI 5 , -C HI 7 , -C H , -CI H I, -CuH 23 , -CI H 25 , - CI H 27 , -Ci H , -Ci H i, -Ci H 33 , -CI H , -CieH 37 , -CigH g, -C H , -C IH 43 , -C H , -C H 47 , -C H 49 , or
  • each of R 2 and R 3 is C 6 -Ci 2 alkyl substituted by-0(CO)R 6 or -C(0)OR 6 , wherein R 6 is unsubstituted C 6 -Ci alkyl. In embodiments, R 6 is unsubstituted linear C 6 -Ci 4 alkyl. In embodiments, R 6 is unsubstituted branched C 6 -Ci alkyl.
  • each of R 2 and R 3 is (CH 2 ) 7 C(0)0(CH 2 ) 2 CH(CsHn) 2 or
  • each R 2 and R 3 is independently a same R 2 and R 3 .
  • a cationic lipid is any one of Compounds (l)-(33), or a pharmaceutically acceptable salt thereof.
  • the invention features a composition comprising any liposome (e.g., a liposome encapsulating an mRNA encoding a protein) described herein.
  • any liposome e.g., a liposome encapsulating an mRNA encoding a protein
  • an mRNA encodes for cystic fibrosis transmembrane conductance
  • CTR CTR regulator
  • an mRNA encodes for ornithine transcarbamylase (OTC) protein.
  • OTC ornithine transcarbamylase
  • the invention features a composition comprising a nucleic acid encapsulated within a liposome as described herein.
  • a composition further comprises 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.
  • a nucleic acid is an mRNA encoding a peptide or polypeptide.
  • a mRNA encodes a peptide or polypeptide for use in the delivery to or treatment of the lung of a subject or a lung cell.
  • a mRNA encodes a peptide or polypeptide for use in the delivery to or treatment of the lung of a subject or a lung cell.
  • an mRNA encodes for cystic fibrosis transmembrane conductance
  • CTR CTR regulator
  • a mRNA encodes a peptide or polypeptide for use in the delivery to or treatment of the liver of a subject or a liver cell.
  • a mRNA encodes for ornithine transcarbamylase (OTC) protein.
  • a mRNA encodes a peptide or polypeptide for use in vaccine.
  • a mRNA encodes an antigen.
  • the present invention provides methods of treating a disease in a subject comprising administering to the subject a composition as described herein.
  • FIG. 1 depicts in vivo protein production resulting from the delivery of mRNA using lipid nanoparticles, each comprising a lipid selected from Compounds 5-7, Compounds 10-13, Compound 19, Compound 21, and Compound 24, as described herein. As shown in this Figure, use of these compounds allow high levels of in vivo protein production even 6 hours after administration.
  • amino acid in its broadest sense, refers to any compound and/or substance that can be incorporated into a polypeptide chain.
  • 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 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.
  • 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.).
  • 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.
  • 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
  • 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.
  • mammals 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.
  • 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.
  • 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”).
  • patient's 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).
  • intact protein e.g., enzyme
  • post-translational modification e.g., enzyme
  • 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 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 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.).
  • Liposome refers to any lamellar, multilamellar, or solid nanoparticle vesicle.
  • a liposome as used herein can be formed by mixing one or more lipids or by mixing one or more lipids and polymer(s).
  • a liposome suitable for the present invention contains a cationic lipids(s) and optionally non-cationic lipid(s), optionally cholesterol-based lipid(s), and/or optionally PEG-modified lipid(s).
  • messenger RNA 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.
  • 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, 0(6)-
  • nucleic acid refers to any compound and/or substance that is or can be incorporated into a polynucleotide chain.
  • a 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 (IncRNA), 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
  • mRNA messenger RNA
  • mmRNA modified messenger RNA
  • IncRNA micro-RNA
  • miRNA multimeric coding nucleic acid
  • PCNA
  • 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).
  • DNA deoxyribonucleic acid
  • ssDNA single-stranded DNA
  • dsDNA double-stranded DNA
  • cDNA complementary DNA
  • nucleic acid encompasses both RNA and DNA.
  • 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., PI, PAC, BAC, YAC, artificial chromosomes), expression cassettes, chimeric sequences, chromosomal DNA, or derivatives of these groups.
  • PCR polymerase chain reaction
  • vectors e.g., PI, PAC, BAC, YAC, artificial chromosomes
  • expression cassettes e.g., 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 (IncRNA), micro-RNA (miRNA), piwi-interacting RNA (piRNA), small interfering RNA (siRNA), transacting siRNA (tasiRNA), repeat associated siRNA (rasiRNA), 73
  • patient 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).
  • 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.
  • compositions 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 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.
  • inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid
  • 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.
  • 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 + (C I- 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.
  • 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.”
  • 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
  • a therapeutically effective amount 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 refers to C -C hydrocarbons and includes both saturated and unsaturated hydrocarbons.
  • An aliphatic may be linear, branched, or cyclic.
  • Ci-C aliphatics can include Ci-C alkyls (e.g., linear or branched Ci-C saturated alkyls), C -C 2 o alkenyls (e.g., linear or branched C -C 2 o dienyls, linear or branched Ce-C 2 o trienyls, and the like), and C -C alkynyls (e.g., linear or branched C -C alkynyls).
  • Ci-C aliphatics can include C -C cyclic aliphatics (e.g., C -C cycloalkyls, C -C cycloalkenyls, or Cg-C
  • 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', -C0 2 FI, - C0 2 R', -CN, -OH, -OR', -OCOR', -OC0 2 R', -NH 2 ,
  • R' independently is Ci-C 20 aliphatic (e.g., Ci-C 20 alkyl, Ci-Ci 5 alkyl, Ci-Cio alkyl, or Ci-C 3 alkyl).
  • R' independently is an unsubstituted alkyl (e.g., unsubstituted Ci-C 20 alkyl, Ci-Ci 5 alkyl, Ci-Cio alkyl, or Ci-C 3 alkyl).
  • R' independently is unsubstituted C 1 -C 3 alkyl.
  • the aliphatic is unsubstituted.
  • the aliphatic does not include any heteroatoms.
  • alkyl means acyclic linear and branched hydrocarbon groups, e.g. "C -C 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.
  • 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 H, -CO R', -CN, -OH, -OR',
  • 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 C 1 -C 20 alkyl, C 1 -C 15 alkyl, C 1 -C 10 alkyl, or C 1 -C 3 alkyl).
  • R' independently is unsubstituted C 1 -C 3 alkyl.
  • the alkyl is substituted (e.g., with 1, 2, 3, 4, 5, or 6 substituent groups as described herein).
  • 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.
  • Alkylene represents a saturated divalent straight or branched chain hydrocarbon group and is exemplified by methylene, ethylene, isopropylene and the like.
  • alkenylene 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
  • 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', -C0 2 H, -C0 2 R',
  • 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 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 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 2 o 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.
  • 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', -C0 2 FI, -C0 2 R', -CN, -OH, -OR', -OCOR', -OCO 2 R', -NH 2 , -NHR', -N(R') 2 , -SR' or-S0 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 unsubstituted C 1 -C 3 alkyl.
  • the alkenyl is unsubstituted.
  • the alkenyl is substituted (e.g., with 1, 2, 3, 4, 5, or 6 substituent groups as described herein).
  • 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.
  • 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. "C 2 -C 2 o 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.
  • 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 independently selected substituents) of halogen, -COR', -C0 2 H, -C0 2 R', - CN, -OH, -OR', -OCOR',
  • R' independently is C -C 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).
  • R' independently is unsubstituted C 1 -C 3 alkyl.
  • the alkynyl is unsubstituted.
  • the alkynyl is substituted (e.g., with 1, 2, 3, 4, 5, or 6 substituent groups as described herein).
  • Aryl refers to an optionally substituted Ce-waromatic hydrocarbon moiety comprising one to three aromatic rings.
  • the aryl group is a Ce-ioaryl group (i.e., phenyl and naphthyl).
  • Aryl groups include, without limitation, optionally substituted phenyl, naphthyl, or anthracenyl.
  • aryl and “ar-”, as used herein, also include groups in which an aryl ring is fused to one or more cycloaliphatic rings to form an optionally substituted cyclic structure such as a tetrahydronaphthyl, indenyl, or indanyl ring.
  • aryl may be used interchangeably with the terms “aryl group”, “aryl ring”, and “aromatic ring”.
  • Cycloalkyl means a nonaromatic, saturated, cyclic group, e.g. "C 3 -C 10 cycloalkyl.”
  • a cycloalkyl is monocyclic.
  • a cycloalkyl is polycyclic (e.g., bicyclic or tricyclic). In polycyclic cycloalkyl groups, individual rings can be fused, bridged, or spirocyclic.
  • cycloalkyl groups 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', - CO 2 H, -CO 2 R', -CN, -OH, -OR', -OCOR', -OC0 2 R', -NH 2 , -NHR', -N(R') 2 , -SR' or-S0 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).
  • substituents e.g., 1, 2, 3, 4, 5, or 6 independently selected substituents
  • 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).
  • Halogen means fluorine, chlorine, bromine, or iodine.
  • heteroalkenyl is meant a branched or unbranched alkenyl group having from 2 to 14 carbon atoms in addition to 1, 2, 3 or 4 heteroatoms independently selected from the group consisting of N, O, S, and P.
  • a heteroalkenyl may optionally include monocyclic, bicyclic, or tricyclic rings, in which each ring desirably has three to six members.
  • the heteroalkenyl group may be substituted or unsubstituted.
  • heteroalkynyl is meant a branched or unbranched alkynyl group having from 2 to 14 carbon atoms in addition to 1, 2, 3 or 4 heteroatoms independently selected from the group consisting of N, O, S, and P.
  • a heteroalkynyl may optionally include monocyclic, bicyclic, or tricyclic rings, in which each ring desirably has three to six members.
  • the heteroalkynyl group may be substituted or unsubstituted.
  • Heteroalkyl is meant a branched or unbranched alkyl group having from 1 to 14 carbon atoms in addition to 1, 2, 3 or 4 heteroatoms independently selected from the group consisting of N, O, S, and P.
  • Heteroalkyls include, without limitation, tertiary amines, secondary amines, ethers, thioethers, amides, thioamides, carbamates, thiocarbamates, hydrazones, imines, phosphodiesters, phosphoramidates, sulfonamides, and disulfides.
  • a heteroalkyl may optionally include monocyclic, bicyclic, or tricyclic rings, in which each ring desirably has three to six members.
  • the heteroalkyl group may be substituted or unsubstituted.
  • heteroalkyls include, without limitation, polyethers, such as methoxymethyl and ethoxyethyl.
  • Heteroaryl The terms “heteroaryl” and “heteroar-”, used alone or as part of a larger
  • heteroaryl group refers to groups having 5 to 14 ring atoms, preferably 5, 6, 9, or 10 ring atoms; having 6, 10, or 14 p electrons shared in a cyclic array; and having, in addition to carbon atoms, from one to five heteroatoms.
  • a heteroaryl group may be mono-, bi-, tri-, or polycyclic, for example, mono-, bi-, or tricyclic (e.g., mono- or bicyclic).
  • heteroatom refers to nitrogen, oxygen, or sulfur, and includes any oxidized form of nitrogen or sulfur, and any quaternized form of a basic nitrogen.
  • a nitrogen atom of a heteroaryl may be a basic nitrogen atom and may also be optionally oxidized to the corresponding N-oxide.
  • a heteroaryl When a heteroaryl is substituted by a hydroxy group, it also includes its corresponding tautomer.
  • the terms "heteroaryl” and “heteroar-”, as used herein, also include groups in which a heteroaromatic ring is fused to one or more aryl, cycloaliphatic, or heterocycloaliphatic rings.
  • heteroaryl groups include thienyl, furanyl, pyrrolyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, oxazolyl, isoxazolyl, oxadiazolyl, thiazolyl, isothiazolyl, thiadiazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, indolizinyl, purinyl, naphthyridinyl, pteridinyl, indolyl, isoindolyl, benzothienyl, benzofuranyl, dibenzofuranyl, indazolyl, benzimidazolyl, benzthiazolyl, quinolyl, isoquinolyl, cinnolinyl, phtha lazinyl, quinazolinyl, quinoxalinyl, 4H-quinoliz
  • heteroaryl may be used interchangeably with the terms “heteroaryl ring”, “heteroaryl group”, or “heteroaromatic”, any of which terms include rings that are optionally substituted.
  • heteroarylkyl refers to an alkyl group substituted by a heteroaryl, wherein the alkyl and heteroaryl portions independently are optionally substituted.
  • heterocyclyl As used herein, the terms “heterocycle”, “heterocyclyl”, “heterocyclic radical”, and “heterocyclic ring” are used interchangeably and refer to a stable 3- to 8-membered monocyclic or 7-10-membered bicyclic heterocyclic moiety that is either saturated or partially unsaturated, and having, in addition to carbon atoms, one or more, such as one to four, heteroatoms, as defined above.
  • nitrogen includes a substituted nitrogen.
  • the nitrogen may be N (as in 3,4-dihydro-2H-pyrrolyl), NH (as in pyrrolidinyl), or NR + (as in N-substituted pyrrolidinyl).
  • a heterocyclic ring can be attached to its pendant group at any heteroatom or carbon atom that results in a stable structure and any of the ring atoms can be optionally substituted.
  • saturated or partially unsaturated heterocyclic radicals include, without limitation, tetrahydrofuranyl, tetrahydrothienyl, piperidinyl, decahydroquinolinyl, oxazolidinyl, piperazinyl, dioxanyl, dioxolanyl, diazepinyl, oxazepinyl, thiazepinyl, morpholinyl, and thiamorpholinyl.
  • a heterocyclyl group may be mono-, bi-, tri-, or polycyclic, preferably mono-, bi-, or tricyclic, more preferably mono- or bicyclic.
  • heterocyclylalkyl refers to an alkyl group substituted by a heterocyclyl, wherein the alkyl and heterocyclyl portions independently are optionally substituted. Additionally, a heterocyclic ring also includes groups in which the heterocyclic ring is fused to one or more aryl rings.
  • 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.
  • lipids lipids, compositions comprising such lipids, and related methods of their use.
  • 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.
  • lipids described herein can provide one or more desired characteristics or properties. That is, in certain embodiments, lipids described herein can be characterized as having one or more properties that afford such compounds advantages relative to other similarly classified lipids.
  • 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.
  • 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.
  • a lipid has a structure according to Formula (I):
  • L 1 is independently C 2 -Ci 0 alkylene, C -Ci 0 alkenylene, or C -Ci 0 alkynylene;
  • X 1 is independently OR 1 , SR 1 , or Nf R 1 ) ⁇
  • R 1 is H or unsubstituted Ci-C 6 alkyl
  • each R 2 and R 3 is independently C 6 -C 30 -alkyl, C 6 -C 30 -alkenyl, or C 6 -C 30 -alkynyl.
  • X 1 is independently OR 1 . In embodiments, X 1 is OH.
  • X 1 is independently SR 1 . In embodiments, X 1 is SH.
  • X 1 is independently N(R 1 ) 2 . In embodiments, X 1 is NH 2 .
  • L 1 is C 2 -Ci 0 alkylene. In embodiments, L 1 is unsubstituted C 2 -Ci 0 alkylene.
  • L 1 is C -C alkenylene. In embodiments, L 1 is unsubstituted C -C
  • L 1 is C -C alkynylene. In embodiments, L 1 is unsubstituted C -C
  • a lipid has a structure according to Formula (I I),
  • n is an integer of 2-10.
  • n is 2, 3, 4, or 5. In embodiments, n is 2. In embodiments, n is 3. In
  • n is 4. In embodiments, n is 5.
  • n is 6. In embodiments, n is 7. In embodiments, n is 8. In embodiments, n is 9. In embodiments, n is 10.
  • each R 2 and R 3 is unsubstituted C 6 -C 3 o-alkyl, unsubstituted C 6 -C 3 o-all ⁇ enyl, or unsubstituted C 6 -C 3 o-all ⁇ ynyl. In embodiments, each R 2 and R 3 is unsubstituted C 6 -C 3 o-alkyl. In embodiments, each R 2 and R 3 is unsubstituted C 6 -C 3 o-alkenyl. In embodiments, each R 2 and R 3 is unsubstituted C 6 -C 30 -alkynyl.
  • each R 2 and R 3 is unsubstituted C 6 -C 22 -alkyl, or each R 2 and R 3 is
  • each R 2 and R 3 is unsubstituted C 6 -C 22 -alkyl. In embodiments, each R 2 and R 3 is unsubstituted C 6 -C 22 -alkenyl.
  • each of R and R is -C HI 3 , -C HI 5 , -C Hi , -C H , -CI H I, -CuH 23 , -CI H 25 , -
  • each of R 2 and R 3 is C 6 -Ci 2 alkyl substituted by-0(CO)R 6 or -C(0)OR 6 , wherein R 6 is unsubstituted C 6 -Ci alkyl.
  • R 6 is unsubstituted linear C 6 -Ci alkyl.
  • R 6 is unsubstituted branched Ce-Ci 4 alkyl.
  • each of R 2 and R 3 is (CH 2 ) 7 C(0)0(CH 2 ) 2 CH(C 5 Hn) 2 or
  • each R 2 and R 3 is independently a same R 2 and R 3 .
  • a lipid is any one of Compounds (l)-(33) of Table A, or a pharmaceutically acceptable salt thereof.
  • a lipid is Compound 1. In embodiments, a lipid is Compound 2. In
  • a lipid is Compound 3. In embodiments, a lipid is Compound 4. In embodiments, a lipid is Compound 5. In embodiments, a lipid is Compound 6. In embodiments, a lipid is Compound 7. In embodiments, a lipid is Compound 8. In embodiments, a lipid is Compound 9. In embodiments, a lipid is Compound 10. In embodiments, a lipid is Compound 11. In embodiments, a lipid is Compound 12.
  • a lipid is Compound 13. In embodiments, a lipid is Compound 14. In embodiments, a lipid is Compound 15. In embodiments, a lipid is Compound 16. In embodiments, a lipid is Compound 17. In embodiments, a lipid is Compound 18. In embodiments, a lipid is Compound 19. In embodiments, a lipid is Compound 20. In embodiments, a lipid is Compound 21. In embodiments, a lipid is Compound 22. In embodiments, a lipid is Compound 23. In embodiments, a lipid is Compound 24.
  • a lipid is Compound 25. In embodiments, a lipid is Compound 26. In embodiments, a lipid is Compound 27. In embodiments, a lipid is Compound 28. In embodiments, a lipid is Compound 29. In embodiments, a lipid is Compound 30. In embodiments, a lipid is Compound 31. In embodiments, a lipid is Compound 32. In embodiments, a lipid is Compound 33.
  • Lipids described herein can be prepared according to methods known in the art, including the exemplary method of Scheme A.
  • An a,w-amino acid (Al) can be combined with excess terminal epoxide (A2) under basic conditions to afford the ring-opened product A3.
  • the hydroxyl groups can be protected (e.g., silyl protecting groups using TBSCI) to afford the protected compound A4.
  • the carboxylic acid can be reduced (e.g., with LiAIFU) to provide the protected compound A5.
  • A5 can then be deprotected to provide a lipid A6 as described herein.
  • a compound A3 can be directly reduced to provide a lipid A6 as described herein.
  • Cationic lipids described herein e.g., a lipid of Formulas (I) or (II) such as any of Compounds (l)-(33)
  • a lipid of Formulas (I) or (II) such as any of Compounds (l)-(33)
  • 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. 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 desired amino acid sequence e.g., an enzyme sequence
  • 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
  • a secondary structure can also be analyzed to calculate stabilizing and destabilizing properties or, respectively, regions of the RNA.
  • 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., PI, 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 (IncRNA), 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. 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.
  • 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.
  • 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.
  • mRNAs may contain RNA backbone modifications.
  • a RNA backbone modifications typically, a RNA backbone modifications.
  • 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'-0-(l-thiophosphate)), boranophosphates, positively charged guanidinium groups etc., which means by replacing the phosphodiester linkage by other anionic, cationic or neutral groups.
  • mRNAs may contain sugar modifications.
  • 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'-0- alkyloligoribonucleotide, 2'-deoxy-2'-C-alkyloligoribonucleotide (2'-0-methylcytidine 5'- triphosphate,
  • 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 typically 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.
  • GTP guanosine triphosphate
  • mRNAs include a 5' and/or 3' untranslated region.
  • 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. In some embodiments, a 3' untranslated region may be between 50 and 500 nucleotides in length or longer.
  • mRNAs include a 5' cap structure. In some embodiments, the
  • cap structures include, but are not limited to, m7G(5')ppp(5')(2'OMeG), m7G(5')ppp(5')(2'OMeA), m7(3'OMeG)(5')ppp(5')(2'OMeG), m7(3'OMeG)(5')ppp(5')(2'OMeA), m7G(5')ppp
  • cap structure is m7G(5')ppp(5')(2'OMeG). Additional cap structures are described in published US Application No. US 2016/0032356 and U.S. Provisional Application 62/464,327, filed February 27, 2017, which are incorporated herein by reference.
  • 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.
  • ARCA Anti-Reverse Cap Analog
  • modified ARCA which is generally a modified cap analog in which the 2' or 3' OH group is replaced with -OCH 3 .
  • 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), or anti reverse cap analogs (e.g., ARCA; m 7 , 2 0me GpppG, m 72 d GpppG, m 7 ’ 30me GpppG, m 7,3 d GpppG and their tetraphosphate derivatives) (see, e.g., Jemielity, J. et al., " Novel ' anti-reverse ' cap analogs with superior translational properties", RNA, 9: 1108-1122
  • a suitable cap is a 7-methyl guanylate (“m 7 G") linked via a
  • 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 CapO structure.
  • CapO structures lack a 2'-0-methyl residue of the ribose attached to bases 1 and 2.
  • the cap is a Capl structure.
  • Capl structures have a 2'-0-methyl residue at base 2.
  • the cap is a Cap2 structure.
  • Cap2 structures have a 2'-0-methyl residue attached to both bases 2 and 3.
  • 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.
  • 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)).
  • 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
  • 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.
  • 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.
  • 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.
  • 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 e.g., a lipid of Formulas (I) or (II) such as any of Compounds (l)-(33)
  • pharmaceutical and liposomal compositions comprising such lipids
  • 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 lipid as described herein (e.g., a lipid of Formulas (I) or (II) such as any of Compounds (l)-(33)).
  • a delivery vehicle comprising a lipid as described herein (e.g., a lipid of Formulas (I) or (II) such as any of Compounds (l)-(33)).
  • delivery vehicle As used herein, the terms "delivery vehicle,” “transfer vehicle,” “nanoparticle” or
  • the present invention provides a composition (e.g., a pharmaceutical
  • compositions comprising a lipid described herein (e.g., a lipid of Formulas (I) or (II) such as any of Compounds (l)-(33)) and one or more polynucleotides.
  • a composition e.g., a pharmaceutical composition
  • compositions 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 lipid described herein (e.g., a lipid of Formulas (I) or (II) such as any of Compounds (l)-(33)) 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).
  • a liposomal formulation comprising a lipid described herein (e.g., a lipid of Formulas (I) or (II) such as any of Compounds (l)-(33)
  • 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 encapsulated materials 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 e.g., liposomal delivery vehicles
  • 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 polynucleotides e.g., mRNA
  • 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.
  • Such 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 composition is a 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).
  • the compounds described herein 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).
  • encapsulated materials e.g., one or more therapeutic agents
  • liposomal delivery vehicles e.g., lipid nanoparticles
  • lipid nanoparticles are usually
  • 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
  • encapsulate materials such as for example, one or more biologically-active polynucleotides (e.g., mRNA).
  • a composition (e.g., a pharmaceutical composition) comprises an mRNA encoding a protein, encapsulated within a liposome.
  • 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 lipid as described herein (e.g., a lipid of Formulas (I) or (II) such as any of Compounds (l)-(33)) .
  • 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. In embodiments, a composition comprises an mRNA encoding for ornithine transcarbamylase (OTC) protein.
  • CFTR cystic fibrosis transmembrane conductance regulator
  • OTC ornithine transcarbamylase
  • 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 lipid of Formulas (I) or (II) such as any of Compounds (l)-(33)) as described herein.
  • any cationic lipid e.g., a lipid of Formulas (I) or (II) such as any of Compounds (l)-(33)
  • 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 net positive charge e.g., a lipid nanoparticle
  • a liposomal delivery vehicle e.g., a lipid nanoparticle
  • a net negative charge e.g., 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 lipid of Formulas (I) or (II) such as any of Compounds (l)-(33)) .
  • the amount of a lipid as described herein e.g., a lipid of Formulas (I) or (II) such as any of Compounds (l)-(33)
  • a lipid as described herein e.g., a lipid of Formulas (I) or (II) such as any of Compounds (l)-(33)
  • wt% percentage of the combined dry weight of all lipids of a composition
  • a lipid as described herein e.g., a lipid of Formulas (I) or (II) such as any of Compounds (l)-(33)
  • a lipid as described herein is present in an amount that is about 0.5 wt% to about 30 wt% (e.g., about 0.5 wt% to about 50 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 lipid as described herein e.g., a lipid of Formulas (I) or (II) such as any of Compounds (l)-(33)
  • a lipid as described herein is present in an amount that is about 1 wt% to about 50 wt%, about 1 wt% to about 40 wt%, 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%, about 5 wt% to about 25 wt%, about 10 wt% to about 30 wt %, or about 20 wt% to about 40 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 lipid as described herein e.g., a lipid of Formulas (I) or (II) such as any of Compounds (l)-(33)
  • a lipid as described herein 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 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%, 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
  • the amount of a 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%, 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 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 lipid described herein (e.g., a lipid of Formulas (I) or (II) such as any of Compounds (l)-(33)) .
  • a lipid described herein e.g., a lipid of Formulas (I) or (II) such as any of Compounds (l)-(33)
  • a delivery vehicle e.g., a liposomal delivery vehicle such as a lipid nanoparticle
  • a delivery vehicle comprises about 0.5 wt%, about 1 wt%, about 3 wt%, about 5 wt%, or about 10 wt% a lipid described herein (e.g., a lipid of Formulas (I) or (II) such as any of Compounds (l)-(33)) .
  • a delivery vehicle e.g., a liposomal delivery vehicle such as a lipid nanoparticle
  • a delivery vehicle 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 lipid described herein (e.g., a lipid of Formulas (I) or (II) such as any of Compounds (l)-(33)) .
  • the percentage results in an improved beneficial effect (e.g., improved delivery to targeted tissues such as the liver or the lung).
  • the amount of a lipid as described herein e.g., a lipid of Formulas (I) or (II) such as any of Compounds (l)-(33)
  • a lipid as described herein e.g., a lipid of Formulas (I) or (II) such as any of Compounds (l)-(33)
  • mol% a percentage 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).
  • a lipid as described herein e.g., a lipid of Formulas (I) or (II) such as any of Compounds (l)-(33)
  • a composition such as a liposomal delivery vehicle.
  • a lipid as described herein e.g., a lipid of Formulas (I) or (II) such as any of Compounds (l)-(33)
  • a lipid as described herein 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%, about 10 mol% to about 20 mol%, about 20 mol% to about 30 mol%, about 30 mol% to about 40 mol%, about 40 mol% to about 50 mol%, or about 50 mol% to about 60 mol% of the combined molar amounts of all lipids present in a composition such as a liposomal delivery vehicle.
  • a lipid as described herein e.g., a lipid of Formulas (I) or (II) such as any of Compounds (l)-(33)
  • a lipid as described herein is present in an amount that is about about 1 mol% to about 50 mol%, about 1 mol% to about 40 mol%, 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
  • a lipid as described herein 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 lipid as described herein 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%, or greater than about 10 mol%, or greater than about 15 mol%, or greater than about 20 mol%, or greater than 25 mol%, or greater than 30 mol%, or greater than 35 mol%, or greater than 40 mol%, or greater than 45 mol%, or greater than 50 mol% of the total amount of lipids in the lipid nanoparticle.
  • a lipid as described herein can comprise less than about 50 mol%, or less than about 45mol%, or less than about 40 mol% or less than about 30%, less than about 25 mol%, or less than about 20 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).
  • the amount of a 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%, 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).
  • a composition e.g., a liposomal composition
  • the amount of a 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%, 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).
  • a composition e.g., a liposomal composition
  • the percentage results in an improved beneficial effect (e.g., improved delivery to targeted tissues such as the liver or the lung).
  • 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).
  • 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.
  • 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 e.g., lipid nanoparticle
  • a nucleic acid e.g., mRNA encoding a peptide or polypeptide
  • a composition comprises one or more cationic lipids as described herein (e.g., a lipid of Formulas (I) or (I I) such as any of Compounds (l)-(33)) 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 lipid of Formulas (I) or (II) such as any of Compounds (l)-(33)) ; 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.
  • a lipid nanoparticle that encapsulates a nucleic acid comprises one or more cationic lipids as described herein (e.g., a lipid of Formulas (I) or (II) such as any of Compounds (l)-(33)) , 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.
  • 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.
  • a composition may comprise one or more further cationic lipids.
  • liposomes may comprise one or more further cationic lipids.
  • 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:
  • Ri and R 2 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 Li 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 C 1 -C 30 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-l-amine ("HGT5000”), having a compound structure of:
  • compositions include the cationic lipid (15Z, 18Z)-N,N-dimethyl-6-((9Z,12Z)-octadeca-9,12-dien-l-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-l-yl) tetracosa-5,15,18-trien- 1 -amine ("HGT5002”), having a compound structure of:
  • compositions include cationic lipids described as aminoalcohol lipidoids in International Patent Publication WO 2010/053572, 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 2016/118725, 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:
  • 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.
  • 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 a
  • 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.
  • the cationic lipids of the compositions include a cationic lipid having a compound structure of:
  • 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 structure:
  • 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.
  • 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 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 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:
  • R is independently selected from -(CH 2 ) n Q and -(CH ) nCHQR;
  • Q is selected from the group consisting of -OR, -OH, -0(CH 2 )nN(R) 2 , -0C(0)R, -CX 3 , -CN, -N(R)C(0)R, -N(H)C(0)R, -N(R)S(0) 2 R, -N(H)S(0) 2 R, -N(R)C(0)N(R) 2 , -N(H)C(0)N(R) 2 , -N(H)C(0)N(R) 2 , -N(H)C(0)N(H)(R), -N(R)C(S)N(R) 2 , -N(H)C(S)N(R) 2 , - N(H)C(S)N(R) 2 , - N(H)C(S)N(R), and a heterocycle;
  • compositions include a cationic lipid having a compound
  • compositions include a cationic lipid having a compound structure of:
  • compositions include a cationic lipid having a compound
  • compositions include a cationic lipid having a compound
  • 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.
  • compositions include a cationic lipid having a compound
  • 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 lipids
  • compositions include a cationic lipid of the following formula:
  • Ri 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 R 2 is selected from the group consisting of one of the following two formulas:
  • R 3 and R 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 compound structure of:
  • compositions include a cationic lipid, "HGT4002", having a compound structure of:
  • compositions include a cationic lipid, "HGT4003", having a compound structure of:
  • compositions include a cationic lipid, "HGT4004", having a compound structure of:
  • compositions include a cationic lipid "HGT4005", having a compound structure of:
  • 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. No. 5,334,761); l,2-Dioleoyl-3- Dimethylammonium-Propane (“DODAP”); l,2-Dioleoyl-3-Trimethylammonium-Propane (“DOTAP”).
  • DOGS 5-carboxyspermylglycinedioctadecylamide
  • DOSPA 2,3-dioleyloxy-N- [2(spermine-carboxa
  • Additional exemplary cationic lipids suitable for the compositions also include: 1,2- distearyloxy-N,N-dimethyl-3-aminopropane ( "DSDMA”); l,2-dioleyloxy-N,N-dimethyl-3- aminopropane (“DODMA”); 1 ,2-dilinoleyloxy-N,N-dimethyl-3-aminopropane (“DLinDMA”); 1,2- 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- dimethyl
  • 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- Dilinoleyl-4-dimethylaminoethyl-[l,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)-Nl,N16-diundecyl-4,7,10,13- tetraazahexadecane- 1,16-diamide (“NC98-5").
  • XTC 2,2- Dilinoleyl-4-dimethylaminoethyl-[l,3]-dioxolane
  • 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%).
  • 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%).
  • 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.
  • compositions may also comprise one or more non-cationic ("helper") lipids.
  • 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),
  • DOPG dioleoylphosphatidylglycerol
  • DPPG dipalmitoylphosphatidylglycerol
  • 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 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%. 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%.
  • 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%.
  • 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%. 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%.
  • 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), l,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 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 wt%, greater than about 10 wt%, greater than about 20 wt%, greater than about 30 wt%, or greater than about 40 wt%.
  • 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-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.
  • particularly useful exchangeable lipids are PEG-ceramides having shorter acyl chains (e.g., C M or Cie).
  • a PEG-modified lipid is 1,2-dimyristoyl-sn-glycerol, methoxypolyethylene glycol (DMG-PEG2000).
  • 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 C -C length.
  • 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).
  • 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 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).
  • compositions e.g., to construct liposomal compositions
  • encapsulated materials e.g., one or more therapeutic polynucleotides
  • target cells e.g., by permeating or fusing with the lipid membranes of such target cells.
  • 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 lipid of Formulas (I) or (II) such as any of Compounds (l)-(33)
  • 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.
  • compositions comprising a lipid described herein (e.g., a lipid of
  • Formulas (I) or (II) such as any of Compounds (l)-(33)) and nucleic acids provided by the present invention may be used for various therapeutic purposes.
  • a lipid described herein e.g., a lipid of Formulas (I) or (II) such as any of Compounds (1)- (33)
  • nucleic acids can be formulated in combination with one or more additional pharmaceutical carriers, targeting ligands or stabilizing reagents.
  • a lipid described herein e.g., a lipid of Formulas (I) or (II) such as any of Compounds (l)-(33)
  • a composition comprising a lipid described herein (e.g., a lipid of Formulas (I) or (II) such as any of Compounds (l)-(33)) can be formulated using post-insertion techniques into the lipid membrane of the nanoparticles.
  • a lipid described herein e.g., a lipid of Formulas (I) or (II) such as any of Compounds (l)-(33)
  • post-insertion techniques into the lipid membrane of the nanoparticles.
  • 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.
  • the 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 any suitable pharmaceutical formulations of the invention.
  • tissue to be targeted 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.
  • 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.
  • 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.
  • 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.
  • 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-l-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.
  • FOXP3 forkhead box P3
  • 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.
  • one or more 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
  • 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.
  • the present invention provides a method for producing a
  • 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.
  • the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for ornithine transcarbamylase (OTC) protein.
  • OTC ornithine transcarbamylase
  • the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for arginosuccinate synthetase 1 protein.
  • the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for carbamoyl phosphate synthetase I protein.
  • 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. [0288] 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
  • 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.
  • 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).
  • TFEB transcription factor EB
  • the present invention provides a method for producing a
  • 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.
  • the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for acid alpha- glucosidase protein.
  • the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for glucose-e- phosphatase (G6PC) protein.
  • the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for liver glycogen phosphorylase protein.
  • the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for muscle phosphoglycerate mutase protein.
  • the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for glycogen debranching enzyme.
  • the present invention provides a method for producing a
  • 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.
  • the present invention provides a method for producing a
  • 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 l-kappa B alpha, interferon-related development regulator 1 (IFRD1), and Sirtuin 1 (SIRT1).
  • NF-kappa B inhibitors such as one or more of l-kappa B alpha, interferon-related development regulator 1 (IFRD1), and Sirtuin 1 (SIRT1).
  • IFRD1 interferon-related development regulator 1
  • SIRT1 Sirtuin 1
  • the present invention provides a method for producing a
  • 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.
  • the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for methylmalonyl CoA epimerase protein.
  • the present invention provides a method for producing a
  • 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.
  • 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.
  • 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 for human hemochromatosis (FIFE) protein.
  • FIFE human hemochromatosis
  • the present invention provides a method for producing a
  • 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
  • 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. 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 Navi.5 channel in muscle tissue or in a muscle cell.
  • the present invention provides a method for producing a
  • 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.
  • 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.
  • ABCD1 ATP binding cassette subfamily D member 1
  • the present invention provides a method for producing a
  • 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.
  • the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for beta globin protein.
  • the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for Bruton's tyrosine kinase protein.
  • 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.
  • 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.
  • the present invention provides a method for producing a
  • 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.
  • the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for retinoschisin protein.
  • 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.
  • the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for centrosomal protein of 290 kDa (CEP290).
  • the present invention provides a method for producing a
  • 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.
  • 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.
  • 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. 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
  • 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.
  • 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.
  • the present invention provides a method for producing a
  • 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.
  • 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.
  • the present invention provides a method for producing a
  • 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 0X40. 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
  • 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.
  • STING interferon genes
  • the present invention provides a method for producing a
  • 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.
  • the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for a zinc finger nuclease protein.
  • compositions and methods of the invention provide for delivery of mRNA encoding a secreted protein.
  • 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
  • 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.
  • 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.
  • the animal of veterinary or industrial interest is chosen from the mammals listed above and/or chicken, duck, turkey, salmon, catfish, or tilapia.
  • compositions and methods of the invention provide for the delivery of mRNA encoding a lysosomal protein chosen from Table 3.
  • 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.
  • 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.
  • 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
  • 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.
  • 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 Conductance Regulator (CFTR), argininosuccinate synthetase (ASS1), Factor IX, survival motor neuron 1 (SMN1), or phenylalanine hydroxylase (PAFI).
  • CFTR Cystic Fibrosis Transmembrane Conductance Regulator
  • ASS1 argininosuccinate synthetase
  • SNS1 survival motor neuron 1
  • PAFI phenylalanine hydroxylase
  • Cationic lipids described herein can be prepared according to methods known in the art such as those described in Scheme A, including Cationic Lipid (9).
  • Example 2 Lipid Nanoparticle Formulation Using Cationic Lipid (9) and In Vivo Expression of hEPO
  • Cationic lipids described herein can be used in the preparation of lipid nanoparticles
  • suitable methods include methods described in International Publication No. WO 2018/089801, which is hereby incorporated by reference in its entirety.
  • 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. 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.
  • a second exemplary process for lipid nanoparticle formulation is Process B of
  • Process B refers to a process of encapsulating messenger RNA (mRNA) by mixing pre-formed lipid nanoparticles with mRNA.
  • mRNA messenger RNA
  • 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.
  • heating the solution post-mixing resulted in a higher percentage of lipid nanoparticles containing mRNA and a higher total yield of mRNA.
  • IV administration to mice of lipid nanoparticle formulations comprising an exemplary cationic lipid and mRNA encoding hEPO can be undertaken in order to study mRNA delivery and resultant hEPO expression.
  • Lipid nanoparticle formulations comprising human erythropoietin (hEPO) mRNA, exemplary cationic lipid, DMG-PEG2000, Cholesterol and DOPE were administered intramuscularly to study mRNA delivery and resultant hEPO expression.
  • Male BALB/c mice at 6-8 weeks old are given a single injection of the LNP formulations into the gastrocnemius muscle at a dosage level of 0.1 ug. Blood samples were collected at 6 hours post-dose.
  • hEPO protein expression levels were measured in the sera samples by ELISA and presented in Figure 1. These studies show that the cationic lipids described herein are effective at delivery mRNA in vivo, resulting in expression of the protein or polypeptide encoded by the delivered mRNA.

Abstract

L'invention concerne des lipides cationiques qui sont des composés de formule I. Les lipides cationiques de la présente invention peuvent être utiles pour l'administration et l'expression d'ARNm et de protéine codée, par exemple, en tant que composant d'un véhicule d'administration liposomal, et peuvent par conséquent être utiles pour traiter diverses maladies, troubles et états, tels que ceux associés à une déficience en une ou de plusieurs protéines.
PCT/US2020/038678 2019-06-21 2020-06-19 Lipides cationiques comprenant une fraction hydroxy WO2020257611A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201962864824P 2019-06-21 2019-06-21
US62/864,824 2019-06-21

Publications (1)

Publication Number Publication Date
WO2020257611A1 true WO2020257611A1 (fr) 2020-12-24

Family

ID=71528034

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2020/038678 WO2020257611A1 (fr) 2019-06-21 2020-06-19 Lipides cationiques comprenant une fraction hydroxy

Country Status (1)

Country Link
WO (1) WO2020257611A1 (fr)

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022221688A1 (fr) * 2021-04-15 2022-10-20 Translate Bio, Inc. "bons" lipides cationiques à base de substance tampon
WO2022247801A1 (fr) * 2021-05-28 2022-12-01 北京启辰生生物科技有限公司 Composé lipidique et son utilisation dans l'administration d'acide nucléique
WO2023031394A1 (fr) 2021-09-03 2023-03-09 CureVac SE Nouvelles nanoparticules lipidiques pour l'administration d'acides nucléiques
WO2023073228A1 (fr) 2021-10-29 2023-05-04 CureVac SE Arn circulaire amélioré pour exprimer des protéines thérapeutiques
WO2023081526A1 (fr) * 2021-11-08 2023-05-11 Orna Therapeutics, Inc. Compositions de nanoparticules lipidiques pour l'administration de polynucléotides circulaires
WO2023144330A1 (fr) 2022-01-28 2023-08-03 CureVac SE Inhibiteurs de facteurs de transcription codés par un acide nucleique
WO2023198857A1 (fr) * 2022-04-13 2023-10-19 Sanofi Lipides cationiques "bons" à base de tampon
WO2023227608A1 (fr) 2022-05-25 2023-11-30 Glaxosmithkline Biologicals Sa Vaccin à base d'acide nucléique codant pour un polypeptide antigénique fimh d'escherichia coli

Citations (43)

* 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 (fr) 2004-06-07 2005-12-22 Protiva Biotherapeutics, Inc. Arn interferant encapsule dans des lipides
WO2010042877A1 (fr) 2008-10-09 2010-04-15 Tekmira Pharmaceuticals Corporation Lipides aminés améliorés et procédés d'administration d'acides nucléiques
WO2010053572A2 (fr) 2008-11-07 2010-05-14 Massachusetts Institute Of Technology Lipidoïdes aminoalcool et leurs utilisations
WO2010144740A1 (fr) 2009-06-10 2010-12-16 Alnylam Pharmaceuticals, Inc. Formulation lipidique améliorée
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 (fr) 2011-06-08 2012-12-13 Shire Human Genetic Therapies, Inc. Lipides clivables
WO2013063468A1 (fr) 2011-10-27 2013-05-02 Massachusetts Institute Of Technology Dérivés d'aminoacides fonctionnalisés sur le n-terminal, capables de former des microsphères d'encapsulation de médicament
WO2013149140A1 (fr) 2012-03-29 2013-10-03 Shire Human Genetic Therapies, Inc. Lipides cationiques ionisables
WO2015095340A1 (fr) 2013-12-19 2015-06-25 Novartis Ag Lipides et compositions lipidiques pour le largage d'agents actifs
WO2015184256A2 (fr) 2014-05-30 2015-12-03 Shire Human Genetic Therapies, Inc. Lipides biodégradables pour l'administration d'acides nucléiques
WO2015199952A1 (fr) 2014-06-25 2015-12-30 Acuitas Therapeutics Inc. Nouveaux lipides et formulations nanoparticulaires lipidiques pour l'administration d'acides nucléiques
WO2016004202A1 (fr) 2014-07-02 2016-01-07 Massachusetts Institute Of Technology Lipidoïdes dérivés de polyamine-acide gras et leurs utilisations
US20160032356A1 (en) 2013-03-14 2016-02-04 Shire Human Genetic Therapies, Inc. Quantitative assessment for cap efficiency of messenger rna
US20160031928A1 (en) 2013-03-14 2016-02-04 Shire Human Genetic Therapies, Inc. RIBONUCLEIC ACIDs WITH 4'-THIO-MODIFIED NUCLEOTIDES AND RELATED METHODS
WO2016118725A1 (fr) 2015-01-23 2016-07-28 Moderna Therapeutics, Inc. Compositions de nanoparticules lipidiques
WO2016118724A1 (fr) 2015-01-21 2016-07-28 Moderna Therapeutics, Inc. Compositions de nanoparticules lipidiques
WO2016205691A1 (fr) 2015-06-19 2016-12-22 Massachusetts Institute Of Technology 2,5-pipérazinediones substituées par un alcényle, et leur utilisation dans des compositions destinées à l'administration d'un agent à un sujet ou une cellule
WO2017004143A1 (fr) 2015-06-29 2017-01-05 Acuitas Therapeutics Inc. Formulations de lipides et de nanoparticules de lipides pour l'administration d'acides nucléiques
WO2017049245A2 (fr) 2015-09-17 2017-03-23 Modernatx, Inc. Composés et compositions pour l'administration intracellulaire d'agents thérapeutiques
WO2017075531A1 (fr) 2015-10-28 2017-05-04 Acuitas Therapeutics, Inc. Nouveaux lipides et nouvelles formulations de nanoparticules de lipides pour l'administration d'acides nucléiques
WO2017117528A1 (fr) 2015-12-30 2017-07-06 Acuitas Therapeutics, Inc. Lipides et formulations de nanoparticules de lipides pour la libération d'acides nucléiques
WO2017173054A1 (fr) 2016-03-30 2017-10-05 Intellia Therapeutics, Inc. Formulations de nanoparticules lipidiques pour des composés crispr/cas
WO2018089801A1 (fr) 2016-11-10 2018-05-17 Translate Bio, Inc. Procédé amélioré de préparation de nanoparticules lipidiques chargées d'arnm
WO2018191750A2 (fr) * 2017-04-14 2018-10-18 The Broad Institute Inc. Nouvelle distribution de grandes charges utiles
WO2019226925A1 (fr) * 2018-05-24 2019-11-28 Translate Bio, Inc. Lipides cationiques de thioester

Patent Citations (43)

* 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 (fr) 2004-06-07 2005-12-22 Protiva Biotherapeutics, Inc. Arn interferant encapsule dans des lipides
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 (fr) 2008-10-09 2010-04-15 Tekmira Pharmaceuticals Corporation Lipides aminés améliorés et procédés d'administration d'acides nucléiques
WO2010053572A2 (fr) 2008-11-07 2010-05-14 Massachusetts Institute Of Technology Lipidoïdes aminoalcool et leurs utilisations
WO2010144740A1 (fr) 2009-06-10 2010-12-16 Alnylam Pharmaceuticals, Inc. Formulation lipidique améliorée
WO2012170889A1 (fr) 2011-06-08 2012-12-13 Shire Human Genetic Therapies, Inc. Lipides clivables
WO2013063468A1 (fr) 2011-10-27 2013-05-02 Massachusetts Institute Of Technology Dérivés d'aminoacides fonctionnalisés sur le n-terminal, capables de former des microsphères d'encapsulation de médicament
WO2013149140A1 (fr) 2012-03-29 2013-10-03 Shire Human Genetic Therapies, Inc. Lipides cationiques ionisables
US20160032356A1 (en) 2013-03-14 2016-02-04 Shire Human Genetic Therapies, Inc. Quantitative assessment for cap efficiency of messenger rna
US20160031928A1 (en) 2013-03-14 2016-02-04 Shire Human Genetic Therapies, Inc. RIBONUCLEIC ACIDs WITH 4'-THIO-MODIFIED NUCLEOTIDES AND RELATED METHODS
WO2015095340A1 (fr) 2013-12-19 2015-06-25 Novartis Ag Lipides et compositions lipidiques pour le largage d'agents actifs
WO2015184256A2 (fr) 2014-05-30 2015-12-03 Shire Human Genetic Therapies, Inc. Lipides biodégradables pour l'administration d'acides nucléiques
WO2015199952A1 (fr) 2014-06-25 2015-12-30 Acuitas Therapeutics Inc. Nouveaux lipides et formulations nanoparticulaires lipidiques pour l'administration d'acides nucléiques
WO2016004202A1 (fr) 2014-07-02 2016-01-07 Massachusetts Institute Of Technology Lipidoïdes dérivés de polyamine-acide gras et leurs utilisations
WO2016118724A1 (fr) 2015-01-21 2016-07-28 Moderna Therapeutics, Inc. Compositions de nanoparticules lipidiques
WO2016118725A1 (fr) 2015-01-23 2016-07-28 Moderna Therapeutics, Inc. Compositions de nanoparticules lipidiques
WO2016205691A1 (fr) 2015-06-19 2016-12-22 Massachusetts Institute Of Technology 2,5-pipérazinediones substituées par un alcényle, et leur utilisation dans des compositions destinées à l'administration d'un agent à un sujet ou une cellule
WO2017004143A1 (fr) 2015-06-29 2017-01-05 Acuitas Therapeutics Inc. Formulations de lipides et de nanoparticules de lipides pour l'administration d'acides nucléiques
WO2017049245A2 (fr) 2015-09-17 2017-03-23 Modernatx, Inc. Composés et compositions pour l'administration intracellulaire d'agents thérapeutiques
WO2017075531A1 (fr) 2015-10-28 2017-05-04 Acuitas Therapeutics, Inc. Nouveaux lipides et nouvelles formulations de nanoparticules de lipides pour l'administration d'acides nucléiques
WO2017117528A1 (fr) 2015-12-30 2017-07-06 Acuitas Therapeutics, Inc. Lipides et formulations de nanoparticules de lipides pour la libération d'acides nucléiques
WO2017173054A1 (fr) 2016-03-30 2017-10-05 Intellia Therapeutics, Inc. Formulations de nanoparticules lipidiques pour des composés crispr/cas
WO2018089801A1 (fr) 2016-11-10 2018-05-17 Translate Bio, Inc. Procédé amélioré de préparation de nanoparticules lipidiques chargées d'arnm
WO2018191750A2 (fr) * 2017-04-14 2018-10-18 The Broad Institute Inc. Nouvelle distribution de grandes charges utiles
WO2019226925A1 (fr) * 2018-05-24 2019-11-28 Translate Bio, Inc. Lipides cationiques de thioester

Non-Patent Citations (18)

* Cited by examiner, † Cited by third party
Title
BEHR ET AL., PROC. NAT.'I 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: 10.1261/rna.5430403
KEVIN J. KAUFFMAN ET AL: "Optimization of Lipid Nanoparticle Formulations for mRNA Delivery in Vivo with Fractional Factorial and Definitive Screening Designs", NANO LETTERS, vol. 15, no. 11, 20 October 2015 (2015-10-20), US, pages 7300 - 7306, XP055679418, ISSN: 1530-6984, DOI: 10.1021/acs.nanolett.5b02497 *
KLIBANOV ET AL., FEBS LETTERS, vol. 268, no. 1, 1990, pages 235 - 237
LUBKE ET AL.: "Proteomics of the Lysosome", BIOCHIM BIOPHYS ACTA, vol. 1793, 2009, pages 625 - 635, XP026073291, 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., 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 (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022221688A1 (fr) * 2021-04-15 2022-10-20 Translate Bio, Inc. "bons" lipides cationiques à base de substance tampon
WO2022247801A1 (fr) * 2021-05-28 2022-12-01 北京启辰生生物科技有限公司 Composé lipidique et son utilisation dans l'administration d'acide nucléique
WO2023031394A1 (fr) 2021-09-03 2023-03-09 CureVac SE Nouvelles nanoparticules lipidiques pour l'administration d'acides nucléiques
WO2023073228A1 (fr) 2021-10-29 2023-05-04 CureVac SE Arn circulaire amélioré pour exprimer des protéines thérapeutiques
WO2023081526A1 (fr) * 2021-11-08 2023-05-11 Orna Therapeutics, Inc. Compositions de nanoparticules lipidiques pour l'administration de polynucléotides circulaires
WO2023144330A1 (fr) 2022-01-28 2023-08-03 CureVac SE Inhibiteurs de facteurs de transcription codés par un acide nucleique
WO2023198857A1 (fr) * 2022-04-13 2023-10-19 Sanofi Lipides cationiques "bons" à base de tampon
WO2023227608A1 (fr) 2022-05-25 2023-11-30 Glaxosmithkline Biologicals Sa Vaccin à base d'acide nucléique codant pour un polypeptide antigénique fimh d'escherichia coli

Similar Documents

Publication Publication Date Title
AU2019278813B2 (en) Cationic lipids comprising a steroidal moiety
JP7441802B2 (ja) ビタミンカチオン性脂質
JP7384832B2 (ja) リボースカチオン性脂質
EP3801627A1 (fr) Lipides cationiques de phosphoesters
WO2019226925A1 (fr) Lipides cationiques de thioester
CN114401942B (zh) 三(羟甲基)甲基甘氨酸和柠檬酸脂质
WO2020257611A1 (fr) Lipides cationiques comprenant une fraction hydroxy
EP3962902A1 (fr) Lipides cationiques di-thioesters
WO2020214946A1 (fr) Lipides cationiques de cystine
EP3883917A1 (fr) Composés lipidiques cationiques et compositions associées destinés à être utilisés dans l'administration d'arn messager
EP3959195A1 (fr) Lipides cationiques de thioester
WO2020243540A1 (fr) Lipides macrocycliques
EP3877444A1 (fr) Composés lipidiques comportant plusieurs groupes peg

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: 20737765

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 20737765

Country of ref document: EP

Kind code of ref document: A1