WO2025052180A2 - Lipids and lipid nanoparticles - Google Patents

Lipids and lipid nanoparticles Download PDF

Info

Publication number
WO2025052180A2
WO2025052180A2 PCT/IB2024/000526 IB2024000526W WO2025052180A2 WO 2025052180 A2 WO2025052180 A2 WO 2025052180A2 IB 2024000526 W IB2024000526 W IB 2024000526W WO 2025052180 A2 WO2025052180 A2 WO 2025052180A2
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
occurrence
independently selected
alkenyl
lipid
Prior art date
Application number
PCT/IB2024/000526
Other languages
French (fr)
Other versions
WO2025052180A3 (en
Inventor
Örn ALMARSSON
John Lucas
Original Assignee
Axelyf ehf.
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 Axelyf ehf. filed Critical Axelyf ehf.
Publication of WO2025052180A2 publication Critical patent/WO2025052180A2/en
Publication of WO2025052180A3 publication Critical patent/WO2025052180A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C237/00Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by amino groups
    • C07C237/02Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by amino groups having the carbon atoms of the carboxamide groups bound to acyclic carbon atoms of the carbon skeleton
    • C07C237/04Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by amino groups having the carbon atoms of the carboxamide groups bound to acyclic carbon atoms of the carbon skeleton the carbon skeleton being acyclic and saturated
    • C07C237/12Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by amino groups having the carbon atoms of the carboxamide groups bound to acyclic carbon atoms of the carbon skeleton the carbon skeleton being acyclic and saturated having the nitrogen atom of at least one of the carboxamide groups bound to an acyclic carbon atom of a hydrocarbon radical substituted by carboxyl groups
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C219/00Compounds containing amino and esterified hydroxy groups bound to the same carbon skeleton
    • C07C219/02Compounds containing amino and esterified hydroxy groups bound to the same carbon skeleton having esterified hydroxy groups and amino groups bound to acyclic carbon atoms of the same carbon skeleton
    • C07C219/04Compounds containing amino and esterified hydroxy groups bound to the same carbon skeleton having esterified hydroxy groups and amino groups bound to acyclic carbon atoms of the same carbon skeleton the carbon skeleton being acyclic and saturated
    • C07C219/06Compounds containing amino and esterified hydroxy groups bound to the same carbon skeleton having esterified hydroxy groups and amino groups bound to acyclic carbon atoms of the same carbon skeleton the carbon skeleton being acyclic and saturated having the hydroxy groups esterified by carboxylic acids having the esterifying carboxyl groups bound to hydrogen atoms or to acyclic carbon atoms of an acyclic saturated carbon skeleton
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C219/00Compounds containing amino and esterified hydroxy groups bound to the same carbon skeleton
    • C07C219/02Compounds containing amino and esterified hydroxy groups bound to the same carbon skeleton having esterified hydroxy groups and amino groups bound to acyclic carbon atoms of the same carbon skeleton
    • C07C219/04Compounds containing amino and esterified hydroxy groups bound to the same carbon skeleton having esterified hydroxy groups and amino groups bound to acyclic carbon atoms of the same carbon skeleton the carbon skeleton being acyclic and saturated
    • C07C219/16Compounds containing amino and esterified hydroxy groups bound to the same carbon skeleton having esterified hydroxy groups and amino groups bound to acyclic carbon atoms of the same carbon skeleton the carbon skeleton being acyclic and saturated having at least one of the hydroxy groups esterified by an inorganic acid or a derivative thereof
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C237/00Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by amino groups
    • C07C237/02Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by amino groups having the carbon atoms of the carboxamide groups bound to acyclic carbon atoms of the carbon skeleton
    • C07C237/04Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by amino groups having the carbon atoms of the carboxamide groups bound to acyclic carbon atoms of the carbon skeleton the carbon skeleton being acyclic and saturated
    • C07C237/06Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by amino groups having the carbon atoms of the carboxamide groups bound to acyclic carbon atoms of the carbon skeleton the carbon skeleton being acyclic and saturated having the nitrogen atoms of the carboxamide groups bound to hydrogen atoms or to acyclic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C255/00Carboxylic acid nitriles
    • C07C255/01Carboxylic acid nitriles having cyano groups bound to acyclic carbon atoms
    • C07C255/24Carboxylic acid nitriles having cyano groups bound to acyclic carbon atoms containing cyano groups and singly-bound nitrogen atoms, not being further bound to other hetero atoms, bound to the same saturated acyclic carbon skeleton
    • C07C255/25Aminoacetonitriles
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D207/00Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D207/02Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D207/18Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having one double bond between ring members or between a ring member and a non-ring member
    • C07D207/22Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having one double bond between ring members or between a ring member and a non-ring member with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D207/24Oxygen or sulfur atoms
    • C07D207/262-Pyrrolidones
    • C07D207/2632-Pyrrolidones with only hydrogen atoms or radicals containing only hydrogen and carbon atoms directly attached to other ring carbon atoms
    • C07D207/272-Pyrrolidones with only hydrogen atoms or radicals containing only hydrogen and carbon atoms directly attached to other ring carbon atoms with substituted hydrocarbon radicals directly attached to the ring nitrogen atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D213/78Carbon atoms having three bonds to hetero atoms, with at the most one bond to halogen, e.g. ester or nitrile radicals
    • C07D213/79Acids; Esters
    • C07D213/80Acids; Esters in position 3
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D213/78Carbon atoms having three bonds to hetero atoms, with at the most one bond to halogen, e.g. ester or nitrile radicals
    • C07D213/81Amides; Imides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D213/78Carbon atoms having three bonds to hetero atoms, with at the most one bond to halogen, e.g. ester or nitrile radicals
    • C07D213/81Amides; Imides
    • C07D213/82Amides; Imides in position 3
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D319/00Heterocyclic compounds containing six-membered rings having two oxygen atoms as the only ring hetero atoms
    • C07D319/041,3-Dioxanes; Hydrogenated 1,3-dioxanes
    • C07D319/061,3-Dioxanes; Hydrogenated 1,3-dioxanes not condensed with other rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D493/00Heterocyclic compounds containing oxygen atoms as the only ring hetero atoms in the condensed system
    • C07D493/12Heterocyclic compounds containing oxygen atoms as the only ring hetero atoms in the condensed system in which the condensed system contains three hetero rings
    • C07D493/18Bridged systems
    • 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
    • A61K9/127Synthetic bilayered vehicles, e.g. liposomes or liposomes with cholesterol as the only non-phosphatidyl surfactant
    • A61K9/1271Non-conventional liposomes, e.g. PEGylated liposomes or liposomes coated or grafted with polymers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/51Nanocapsules; Nanoparticles
    • A61K9/5107Excipients; Inactive ingredients
    • A61K9/5123Organic compounds, e.g. fats, sugars
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/07Optical isomers
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/12Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains three hetero rings
    • C07D491/18Bridged systems

Definitions

  • Lipid nanoparticles are a type of nanoscale delivery system composed of lipids that have gained significant attention and validation in the field of medicine. These particles have shown great potential for the delivery of various therapeutic agents, including nucleic acids such as DNA, RNA, and siRNA. LNPs offer several advantages over other delivery systems, including intracellular delivery of sensitive nucleic acids like mRNA, biocompatibility, biodegradability, and the ability to encapsulate hydrophobic and hydrophilic molecules.
  • LNPs as a drug delivery system
  • researchers recognized the potential of lipids to form stable nanoparticles and protect encapsulated molecules.
  • the initial focus of LNP research was on gene therapy, where the delivery of nucleic acids posed significant challenges due to their inherent instability and the need for efficient intracellular delivery.
  • advancements in lipid chemistry, formulation techniques, and manufacturing processes have contributed to the refinement of LNP-based delivery systems.
  • LNPs have shown tremendous promise in the field of medicine, particularly in the delivery of nucleic acid-based therapeutics. This includes the delivery of small interfering RNA (siRNA) for gene silencing, messenger RNA (mRNA) for protein synthesis, and gene editing tools such as CRISPR-Cas9. LNPs protect the encapsulated nucleic acids from degradation, enhance their cellular uptake, and facilitate their release at the target site, thereby improving therapeutic efficacy.
  • siRNA small interfering RNA
  • mRNA messenger RNA
  • CRISPR-Cas9 gene editing tools
  • LNP-based delivery systems face barriers such as uptake by the reticuloendothelial system (RES) and clearance by the liver and kidneys, which can limit their circulation time and reduce their therapeutic efficacy.
  • RES reticuloendothelial system
  • LNPs may induce an immune response due to their foreign nature, leading to adverse reactions or decreased therapeutic outcomes.
  • LNPs pose significant challenges. Maintaining batch-to-batch consistency, controlling particle size, and optimizing production processes are crucial for the successful translation of LNPs from the laboratory to large-scale manufacturing.
  • lipid compounds e.g., cationic lipids, ionizable lipids (generally amines, tertiary substituted), polymer-conjugated lipids, and structural lipids, that afford efficient delivery of the therapeutic agents, sufficient activity of the therapeutic agents (e.g., expression of mRNA after delivery), optimal pharmacokinetics, and/or other suitable physiological, biological, and/or therapeutic properties.
  • the present invention provides for lipids that may be formulated in a delivery vehicle to facilitate the encapsulation of a wide range of payloads including therapeutic and diagnostic agents, such as, without limitation, nucleic acids (e.g., RNA or DNA), proteins, peptides, and small molecule active pharmaceutical ingredients (APIs).
  • therapeutic and diagnostic agents such as, without limitation, nucleic acids (e.g., RNA or DNA), proteins, peptides, and small molecule active pharmaceutical ingredients (APIs).
  • the lipid compounds of the present invention can be used in combination with other lipid components, such as neutral lipids, sterols and polymer conjugated lipids, to form lipid nanoparticles for delivery of payloads both in vitro and in vivo, for therapeutic or prophylactic purposes, including vaccination.
  • the present invention further provides for lipid nanoparticles (LNPs) comprising said lipids as well as methods of administering LNPs to a subject, e.g., delivering an mRNA and achieving prolonged expression of a desired polypeptide in the animal or human subject.
  • LNPs lipid nanoparticles
  • the invention provides for a method of delivering and/or producing a polypeptide of interest in a cell.
  • the invention provides for a method of treating a disease, disorder, or condition in a subject, comprising the step of administering the foregoing lipid nanoparticle and/or lipid nanoparticle composition, to a subject in need of such treatment.
  • the lipid particle and/or lipid particle composition may be also delivered to a subject as a component of a vaccine or diagnostic composition.
  • FIGs. 1A-1B Size assessed by Dynamic Light Scattering (DLS) of LNPs with and without fifth component.
  • FIG. 1A MC3 LNPs with 45mol% MC3 without and with AAP modifiers (50% denotes a control without AAP and having 50mol % MC3) with compositions according to Table 1.
  • FIG. IB Size assessed by DLS of LNPs with compositions according to Table 1.
  • MC3 is the chemical named (6Z,9Z,28Z,31Z)-heptatriaconta-6,9,28,31-tetraen-19-yl 4- (dimethylamino)butanoate.
  • FIGs. 2A-2B Polydispersity of LNPs with and without fifth component.
  • FIG. 2A MC3 LNPs with 45mol% MC3 without and with AAP modifiers (50% denotes a control without AAP and containing 50mol % MC3).
  • FIG. 2B Polydispersity of LNPs with compositions and structures of ionizable lipids per Table 1.
  • FIGs. 3A-3B Size and polydispersity of LNPs with and without fifth component, on day 14 after formulation and storage in refrigerator.
  • FIG. 3A MC3 LNP sizes with 45mol% MC3 without and with AAP modifiers (50% denotes a control without AAP and containing 50mol % MC3) according to Table 1.
  • FIG. 3B Polydispersity of LNPs with compositions according to Table 1.
  • FIGs. 4A-4B In vitro expression of Firefly Luciferase in HEK293 cells, measured by luminescence, expressed as relative light units (RLU). MC3 LNPs with 45mol% MC3 with AAP and without modifiers (FIG. 4A). Novel ionizable lipid-based LNPs with same mRNA (FIG. 4B) show proportional increase in luminescence with increased dose. Compositions are according to Table 1.
  • FIGs. 5A-5B In vitro expression of Firefly Luciferase in Huh7 cells, measured by luminescence, expressed as relative light units (RLU). MC3 LNPs with 45mol% MC3 with AAP and without modifiers (FIG. 5A). Novel ionizable lipid-based LNPs with same mRNA (FIG. 5B) show proportional increase in luminescence with increased dose. Compositions are according to Table 1.
  • FIGs. 6A-6B Viability of cells in the in vitro expression of Firefly Luciferase in HEK293T cells, expressed as relative fluorescence units (RFU) using TiterGlo kit.
  • MC3 LNPs with 45mol% MC3 with AAP and without modifiers (FIG. 6A).
  • Novel ionizable lipid-based LNPs with same mRNA (FIG. 6B) show proportional increase in luminescence with increased dose.
  • Compositions are according to Table 1.
  • FIGs. 7A-7B Viability of cells in the in vitro expression of Firefly Luciferase in Huh7 cells, expressed as relative fluorescence units (RFU) using TiterGlo kit.
  • IVIC3 LNPs with 45mol% IVIC3 with AAP and without modifiers (FIG. 7A).
  • Novel ionizable lipid-based LNPs with same mRNA (FIG. 7B) show proportional increase in luminescence with increased dose.
  • Compositions are according to Table 1.
  • FIG. 8 Fluorescence emission of LNP formulations in Table 2. Irradiation of the pure Compound (89) in ethanol at 480 nm is associated with emission band at around 560 nm. Irradiation of the LNP formulations at 480 nm is associated with negligible emission at around 560 nm, showing fluorescence quenching.
  • DLS Dynamic Light Scattering
  • FIG. 14 Expression of Luciferase from modified mRNA with LNPs (see Table 3) containing modified mRNA encoding Luciferase and ionizable lipid Compound 30 or Compound 52b y in vivo intravenous dosing in Balb/C mice.
  • administering refers to introducing a composition or agent of the present invention (e.g., an LNP comprising a nucleic acid payload) into a subject, organ, tissue or cells for therapeutic, pharmacokinetic, diagnostic purposes, or research purposes.
  • administering includes in vivo, in vitro, ex vivo and in utero administration.
  • the introduction of a composition or agent into a subject is by any route of administration that is suitable for the specific composition or agent.
  • ionizable cationic lipids have a pKa' in the range of about 4 to about 7 as measured in the particle context (TNS assay).
  • ionizable lipid may include "cleavable lipid" or "SS-cleavable lipid”.
  • an LNP comprising an ionizable lipid has a an pKa' (TNS) between 4-5, 5-6 between 4-5, 5-6, or 6-7.
  • a "non-cationic lipid” is an anionic or neutral lipid.
  • hydrophobic lipid refers to compounds having a polar group(s) that include, but are not limited to, long-chain saturated and unsaturated aliphatic hydrocarbon groups and such groups optionally substituted by one or more aromatic, cycloaliphatic, or heterocyclic group(s). Suitable examples include, but are not limited to, diacylglycerol, dialkylglycerol, N,N-dialkylamino, l,2-diacyloxy-3-aminopropane, and l,2-dialkyl-3-aminopropane.
  • hydroxyalkyl means a linear monovalent hydrocarbon radical or a branched monovalent hydrocarbon radical, substituted with one or two hydroxy groups, provided that if two hydroxy groups are present, they are not on the same carbon atom.
  • Representative examples include, but are not limited to, hydroxymethyl, 2-hydroxyethyl, 2-hydroxypropyl, 3-hydroxypropyl, l-(hydroxymethyl)-2- methylpropyl, 2-hydroxybutyl, 3-hydroxybutyl, 4- hydroxybutyl, 2,3-dihydroxypropyl, 1- (hydroxymethyl)-2-hydroxyethyl, 2,3-dihydroxybutyl, 3,4-dihydroxybutyl and 2- (hydroxymethyl)-3-hydroxypropyl, preferably 2- hydroxyethyl, 2,3-dihydroxypropyl, and 1- (hydroxymethyl)-2-hydroxyethyl.
  • a C1-C6 hydroxyalkyl means a linear monovalent hydrocarbon radical of one to six carbon atoms or a branched monovalent hydrocarbon radical of three to six carbons substituted with either one hydroxy group or two hydroxy groups on different carbon atoms. Where the alkyl is substituted with an alkene, the group is a "hydroxyalkenyl" group.
  • bonds encompasses chemical conjugation, adsorption (physisorption and/or chemisorption).
  • bonds encompassed by the term “linked” are covalent interactions and noncovalent interactions (e.g., hydrogen bonds, ionic bonds, van der Waal bonds, and hydrophobic bonds).
  • neutral lipid refers to a lipid that exist either in an uncharged or neutral zwitterionic form in a pH range comprising pH 4 - 7.4.
  • lipids include, for example, diacylphosphatidylcholine, diacylphosphatidylethanolamine, ceramide, sphingomyelin, cephalin, cholesterol, cerebrosides, and diacylglycerols.
  • non-fusogenic cationic lipid is meant a cationic lipid that can condense and/or encapsulate the nucleic acid cargo, such as mRNA, but does not have, or negligible, fusogenic activity with a cell plasma membrane.
  • cleavable lipid or "SS-cleavable lipid” refers to a lipid comprising a disulfide bond cleavable unit.
  • Cleavable lipids may include cleavable disulfide bond ("SS") containing lipid- like materials that comprise a pH-sensitive tertiary amine and self-degradable phenyl ester.
  • SS cleavable disulfide bond
  • a SS-cleavable lipid can be an SS-OP lipid (COATSOME® SS-OP), an SS-M lipid (COATSOME® SS-M), an SS-E lipid (COATSOME® SS-E), an SS-EC lipid (COATSOME® SS-EC), an SS-LC lipid (COATSOME® SS-LC), an SS-OC lipid (COATSOME® SS-OC), and an SS-PalmE lipid (see, for example, Formulae l-IV), or a lipid described in Togashi R, et al. J Control Release 2018 Jun 10;279:262-270, US Patent 9,708,628, or US Patent 10,385,030.
  • non-cationic lipid refers to a neutral lipid or anionic lipid.
  • nucleic acid refers to a polymer containing at least two nucleotides (i.e., deoxyribonucleotides or ribonucleotides) in either single- or double-stranded form and includes DNA, RNA, DNA-RNA hybrids, as well as analogs and modified forms thereof.
  • DNA may be in the form of, e.g., antisense molecules, plasmid DNA, DNA-DNA duplexes, precondensed DNA, PCR products, vectors (PI, PAC, BAC, YAC, artificial chromosomes), expression cassettes, chimeric sequences, chromosomal DNA, or derivatives and combinations of these groups.
  • DNA may be in the form of minicircle, plasmid, bacmid, minigene, ministring DNA (linear covalently closed DNA vector), closed-ended linear duplex DNA (CELiD or ceDNA), doggyboneTM DNA, dumbbell shaped DNA, minimalistic immunological-defined gene expression (MIDGE)-vector, viral vector or nonviral vectors.
  • RNA may be in the form of small interfering RNA (siRNA), dicer-substrate dsRNA, small hairpin RNA (shRNA), asymmetrical interfering RNA (aiRNA), microRNA (miRNA), mRNA, rRNA, tRNA, gRNA, viral RNA (vRNA), and combinations thereof.
  • Nucleic acids include nucleic acids containing known nucleotide analogs or modified backbone residues or linkages, which are synthetic, naturally occurring, and non-naturally occurring, and which have similar binding properties as the reference nucleic acid.
  • analogs and/or modified residues include, without limitation, phosphorothioates, phosphorodiamidate morpholino oligomer (morpholino), phosphoramidates, methyl phosphonates, chiral-methyl phosphonates, 2'-O-methyl ribonucleotides, locked nucleic acid (LNATM), and peptide nucleic acids (PNAs).
  • nucleic acids containing known analogues of natural nucleotides that have similar binding properties as the reference nucleic acid.
  • a particular nucleic acid sequence also implicitly encompasses conservatively modified variants thereof (e.g., degenerate codon substitutions), alleles, orthologs, SNPs, and complementary sequences as well as the sequence explicitly indicated.
  • nucleic acid therapeutic refers to any modality of therapeutic using a nucleic acid as an active pharmaceutical ingredient of therapeutic agent to treat a disease or disorder.
  • pharmaceutically acceptable carrier or “pharmaceutically acceptable excipient” includes any of the standard pharmaceutical carriers/excipients, such as a phosphate buffered saline solution, TRIS/sucrose buffer, water, emulsions such as an oil/water or water/oil, and various types of wetting agents.
  • pharmaceutically acceptable carrier or “pharmaceutically acceptable excipient” includes any of the standard pharmaceutical carriers/excipients, such as a phosphate buffered saline solution, TRIS/sucrose buffer, water, emulsions such as an oil/water or water/oil, and various types of wetting agents.
  • the term also encompasses any of the agents approved by a regulatory agency of the US Federal government or listed in the US Pharmacopeia for use in animals, including humans, as well as any carrier or diluent that does not cause significant irritation to a subject and does not abrogate the biological activity and properties of the administered compound.
  • subject refers to a human or animal, to whom treatment, including prophylactic treatment, with the therapeutic nucleic acid according to the present disclosure, is provided.
  • Animals include mammals, birds and fish.
  • the animal is a mammal, e.g., primate, rodent, lagomorph, companion animal or livestock.
  • Primates include humans, chimpanzees, cynomolgus monkeys, spider monkeys, and macaques, e.g., rhesus macaque.
  • Rodents include mice, rats, and hamsters.
  • Livestock include cows, horses, pigs, sheep and goats.
  • the subject is a human.
  • a human subject can be of any age, gender, race or ethnic group.
  • terapéuticaally effective amount of an active agent (e.g., a TNA described herein) are used interchangeably to refer to an amount that is sufficient to produce a desired effect, e.g., expression or inhibition of a target gene/sequence or disease modification.
  • an “effective amount” may be an amount sufficient to produce an increase in expression of a target polypeptide in comparison to the normal expression level, if any, detected in the absence of the messenger RNA.
  • Suitable assays for measuring expression of a target gene or target sequence include, examination of protein or RNA levels using techniques known to those of skill in the art such as dot blots, northern blots, in situ hybridization, ELISA, immunoprecipitation, enzyme function, fluorescence or luminescence of suitable reporter proteins, as well as phenotypic assays.
  • the terms include prophylactic or preventative amounts of an active agent is an amount sufficient to eliminate or reduce the risk, lessen the severity, or delay the onset of a disease, disorder or condition.
  • dose and “dosage” is the amount of an active agent administered at any given time. Dosage levels are based on a variety of factors, including the specific disease or disorder, the age, weight, sex, medical condition of the patient, the severity of the condition, the route of administration, and the particular active agent or agents employed. The dosage regimen can be determined routinely by a physician using standard methods.
  • therapeutic effect refers to a consequence of treatment, the results of which are judged to be desirable, safely achieved, and beneficial.
  • a therapeutic effect can include, directly or indirectly, the arrest, reduction, or elimination of a disease manifestation or progression.
  • treat may be therapeutic, prophylactic or palliative, and include abrogating, inhibiting, delaying, slowing or reversing the progression of a disease, disorder or condition; ameliorating clinical symptoms of a disease, disorder or condition; or preventing or reducing the appearance of clinical symptoms of a disease, disorder or condition.
  • alkyl refers to saturated monovalent hydrocarbon radical. Alkyls may be linear or branched and may be optionally substituted. Examples include C1-20 alkyl, Cus alkyl, Cue alkyl, Ci-14 alkyl, C1-12 alkyl, C1-10 alkyl, C1-9 alkyl, C1-8 alkyl, C1-7 alkyl, C1-6 alkyl, C1-5 alkyl, C1-4 alkyl, and C1-C 3 alkyl.
  • Examples further include methyl, ethyl, 1-propyl, 2-propyl, 1-butyl, 2- methyl-l-propyl, 2-butyl, 2-methyl-2-propyl, 1-pentyl, 2-pentyl, 3-pentyl, 2-methyl-2-butyl, 3-methyl-2-butyl, 3-methyl-l-butyl, 2-methyl-l-butyl, 1-hexyl, 2-hexyl, 3-hexyl, 2-methyl-2- pentyl, 3-methyl-2-pentyl, 4-methyl-2-pentyl, 3-methyl-3-pentyl, 2-methyl-3-pentyl, 2,3- dimethyl-2-butyl, 3,3-dimethyl-2-butyl, 1-heptyl, 1-octyl, and the like.
  • Heteroaryl refers to a monovalent group with one attachment point derived from heteroarene by removal of a hydrogen atom from a ring atom.
  • Heteroaryls of the present invention include 5-18- membered aromatic radicals (e.g., C5-C13 heteroaryl), preferably 5-10-membered aromatic groups, that includes one or more ring heteroatoms selected from nitrogen, oxygen and sulfur, and which may be a monocyclic or polycyclic (e.g., a bicyclic, tricyclic or tetracyclic ring system).
  • a polycyclic heteroaryl group may be fused or non-fused.
  • the heteroatom(s) in the heteroaryl radical are optionally oxidized.
  • One or more nitrogen atoms, if present, are optionally quaternized.
  • “Pharmaceutically acceptable salt” as used herein refers to pharmaceutically acceptable organic or inorganic salts of an ionizable lipid of the disclosure.
  • Spirocycle and “spiroheterocycle” refer is a 5- to 20-membered bicyclic ring system functional group, including spi ro [cycloa Ikyl] and spiro[cycloalkenyl] with both rings connected through a carbon single atom.
  • a spirocycle/spiroheterocycle can be fully saturated or can be partially unsaturated.
  • the rings can be different in size and nature, or identical in size and nature. Examples include spiropentanyl, spriohexanyl, spiroheptanyl, spirooctanyl, spirononanyl, or spirodecanyl.
  • One or both of the rings in a spiro(hetero)cyclcle can be fused to another ring carbocyclic, heterocyclic, aromatic, or heteroaromatic ring.
  • a (C5-C14) spirocycloalkyl e.g., is a spirocycle containing between 5 and 14 carbon atoms.
  • “Spiroheterocycloalkyl” or “spiroheterocyclyl” is understood to mean a spirocyclyl as defined above wherein at least one of the rings is a heterocycle, i.e., a ring containing a heteroatom.
  • the heteroatom is oxygen, sulfur, or nitrogen.
  • pharmaceutically acceptable salt refers to pharmaceutically acceptable organic or inorganic salts of an ionizable lipid of the disclosure.
  • Hydrogen atoms connected to carbons may be substituted with deuterium ( 2 H) atoms.
  • the headgroup Connecting the headgroup to the is an optional linker moiety which is preferably biodegradable for safe clearance after nucleic acid delivery.
  • the headgroup is essential for self-assembly and phospholipid membrane fusion within the lipid nanoparticle.
  • the headgroup-linker- is connected to a hydrophobic tail group.
  • Ionizable cationic lipids typically have hydrophobic hydrocarbon chains as or as part(s) of the tail group. These chains promote self-assembly of the lipid nanoparticles. They also facilitate membrane fusion during cellular uptake.
  • the hydrophobic tails further contribute to the overall stability of the lipid nanoparticle.
  • the invention provides for a lipid nanoparticle (LNP) comprising a lipid of the present invention.
  • LNP lipid nanoparticle
  • the present disclosure provides for a LNP composition comprising a plurality of LNPs and at least one pharmaceutically acceptable carrier, diluent or excipient.
  • the LNPs of the invention are lipid vesicles with a diameter that is typically in the range of 25-1000 nm.
  • LNPs of the invention comprise multiple lipids, at least one of which is positively charged (cationic) at low pH (enabling RNA complexation and endosomal escape).
  • the cationic lipid is preferably an ionizable cationic lipid that is substantially in the neutral form in an LNP at physiological pH.
  • the LNP may further comprises a non-cationic, structural/helper lipid, a sterol (to provide membrane fluidity) and a polymer conjugated lipid (to prevent aggregation).
  • LNPs of the invention may comprise a targeting moiety, such as a protein or peptide or a cluster of peptides and/or small molecule targeting ligands.
  • a targeting moiety such as a protein or peptide or a cluster of peptides and/or small molecule targeting ligands.
  • LNPs of the invention may comprise a labelling moiety, such as a fluorophore small molecule (BODIPY and the like) for tracking purposes with confocal microscopy and fluorescence imaging methods.
  • a labelling moiety such as a fluorophore small molecule (BODIPY and the like) for tracking purposes with confocal microscopy and fluorescence imaging methods.
  • LNPs of the invention may further comprise a diagnostic or therapeutic agent and be used to deliver the agent, such as TNA, to a cell, tissue or organ.
  • the LNP comprises a therapeutic agent such as a TNA (e.g., mRNA), protein, peptide or other sensitive cargo encapsulated or contained in the lipid portion of the particle, thereby protecting it from enzymatic degradation, excretion or immunogenic or other reaction.
  • the lipid particles of the disclosure have a mean diameter of: from about 25 nm to about 125 nm, about 50 nm to about 100 nm, about 100 nm to about 200 nm, about 200 nm to about 300 nm, from about 300 nm to about 400 nm, from about 400 nm to about 500 nm, from about 500 nm to about 750 nm, from about 750 nm to about 1000 nm, from about 50 nm to about 500 nm, from about 25 nm to about 1000 nm, less than about 1000 nm, less than about 750 nm, less than about 500 nm, less than about 250 nm, less than about 150 nm, less than about 100 nm, less than about 75 nm, less than about 50 nm, less than about 40 nm, less than about 30 nm, less than about 25 nm, or less than about 20 nm.
  • Lipid particle size can be determined, e.g., by quasi-elastic light scattering using a Malvern Zetasizer Nano ZS (Malvern, UK) or Wyatt Dynapro DLS (Wyatt Technologies, Santa Barbara CA).
  • the LNPs may be relatively homogenous.
  • a polydispersity index may be used to indicate the homogeneity of the LNPs.
  • a small, for example less than 0.3 or less than 0.2, polydispersity index generally indicates a narrow particle size distribution.
  • a composition of the LNPs described herein may have a polydispersity index from about 0 to about 0.25 or to about 0.30, such as 0.01, 0.02, 0.03, 0.04, 0.05, 0.06, 0.07, 0.08, 0.09, 0.10, 0.11, 0.12, 0.13, 0.14, 0.15, 0.16, 0.17, 0.18, 0.19, 0.20, 0.21, 0.22, 0.23, 0.24, 0.25, 0.26, 0.27, 0.28, 0.29 or 0.30.
  • the polydispersity index of the LNP composition may be from about 0 to about 0.30 or 0.05 to 0.20.
  • the LNP comprises: a (ionizable) cationic lipid, a sterol or a derivative thereof, a non-cationic lipid, and a polymer-conjugated lipid.
  • the LNP comprises more than one (ionizable) cationic lipid, more than one sterol or a derivative thereof, more than one non-cationic lipid, and/or more than one polymer-conjugated lipid.
  • the lipid particle (e.g., lipid nanoparticle) comprises a cationic lipid, a noncationic phospholipid, cholesterol and a PEGylated lipid (polymer conjugated lipid).
  • the cationic lipid, non-cationic phospholipid, cholesterol and a PEGylated lipid are present in a molar ratio of about 50:7:40:3. 50: 10:38.5: 1.5, respectively.
  • the LNP comprises about: 40-50 mol%, 45-50 mol%, 50-55 mol%, 45-46 mol%, 46-47 mol%, 47-48 mol%, 48-49 mol%, or 49- 50 mol%, 45 mol%, 45.5 mol%, 46 mol%, 46.5 mol%, 47 mol%, 47.5 mol%, 48 mol%, 48.5 mol%, 49 mol%, or 49.5 mol% (ionizable) cationic lipid.
  • the LNP comprises about: 5-25 mol%, 5-15 mol%, 10-12 mol%, 5-6 mol%, 6-7 mol%, 7-8 mol%, 8-9 mol%, 9-10 mol%, 10-11 mol%, 11-12 mol%, 12-13 mol%, 13-14 mol%, or 14- 15 mol% non-cationic lipid.
  • the LNP comprises about: 25-55 mol%, 30-45 mol%, 35-40 mol%, 30-31 mol%, 31-32 mol%, 32-33 mol%, 33-34 mol%, 35-35 mol%, 35-36 mol%, 36-37 mol%, 38-38 mol%, 38-39 mol%, or 39-40 mol% sterol.
  • the LNP comprises about: 0.5-15 mol%, 1-5 mol%, 1-3 mol%, 1.5-2.5 mol%, 1-2 mol%, 2-3 mol%, 3-4 mol%, or 4-5 mol% polymer conjugated lipid, e.g., PEGylated lipid.
  • the lipid nanoparticle comprises a total lipid content that is 20-60 mol% (ionizable) cationic lipid, 5-25 mol% non-cationic lipid, 25-55 mol% sterol, and 0.5-15 mol% polymer conjugated lipid, e.g., PEGylated lipid.
  • the lipid nanoparticle comprises a total lipid content that is 40-50 mol% (ionizable) cationic lipid, 5-15 mol% non-cationic lipid, 30-45 mol% sterol, and 1-5 mol% polymer conjugated lipid, e.g., PEGylated lipid. In one embodiment, the lipid nanoparticle comprises a total lipid content that is 45-50 mol% (ionizable) cationic lipid, 10-12 mol% noncationic lipid, 35-40 mol% sterol, and 1-3 mol% polymer conjugated lipid, e.g., PEGylated lipid.
  • the lipid nanoparticle comprises a total lipid content that is 45-50 mol% (ionizable) cationic lipid, 10-12 mol% non-cationic lipid, 35-40 mol% sterol, and 1.5-2.5 mol% polymer conjugated lipid, e.g., PEGylated lipid conjugate.
  • the lipid nanoparticle of the present invention comprises a cationic lipid.
  • the cationic lipid is an ionizable cationic lipid.
  • the ionizable cationic lipid is positively charged at low pH, which facilitates association with the negatively charged nucleic acid.
  • the ionizable cationic lipid is neutral at physiological pH (pH 7.4). The ability of these lipids to ionize at low pH aids in endosomal escape of the nucleic acid into the cytoplasm.
  • the invention provides for a pharmaceutical composition
  • a pharmaceutical composition comprising a lipid nanoparticle, wherein the lipid nanoparticle comprises a cationic lipid.
  • the cationic lipid is an ionizable cationic lipid.
  • LNPs can be used to deliver a diagnostic or therapeutic agent to a target cell, tissue or organ in a subject.
  • Exemplary ionizable cationic lipids are described in International PCT patent publications WO2022/246571, W02018/011633, WO2017/117528, WO2017/099823, WO2017/075531, WO2017/049245, W02017/004143, W02016/081029, WO2015/199952, WO2015/095346, W02015/095340, W02015/074085, WO2015/061467, WO2013/148541, WO2013/126803, WO2013/116126, W02013/089151, WO2013/086373, WO2013/086354, WO2013/086322, WO2013/049328, WO2013/033563, W02013/016058, W02013/006825, W02012/162210, WO2012/099755, WO2012/054365, WO2012/044638, W02012/040184, W02012/031043, W02012/
  • Further examples include 3-(didodecylamino)-Nl,Nl,4-tridodecyl-l-piperazineethanamine (KL1O), Nl-[2-(didodecylamino)ethyl]-Nl,N4,N4-tridodecyl-l,4- piperazinediethanamine (KL22), 14,25-ditridecyl-15, 18,21 ,24-tetraaza-octatriacontane (KL25), 1.2-dilinoleyloxy-N,N- dimethylaminopropane (DLin-DMA), 2,2-dilinoleyl-4-dimethylaminomethyl-[l,3]-dioxolane (DLin-K-DMA), (6Z,9Z,28Z,31Z)-heptatriacont-6,9,28,31-tetraene-19-yl 4- (dimethylamino)butanoate
  • the ionizable cationic lipid has a structural formula selected from the group consisting of: Compound (1):
  • R 3 is selected from: -H, optionally substituted C1-C6 alkyl, -[CH2]i-eOH, optionally substituted C 2 -C 6 alkenyl, -P(O)(OR 17 )(OR 18 ), -P(O)(OR 17 )(NR 19 ), -P(O)(NR 19 )(NR 20 ) or R 4 ;
  • R 2 is selected from: is absent (i.e., a lone pair on N), -H or optionally substituted C1-C6 alkyl; provided that when R 2 is -H or optionally substituted C1-C6 alkyl, the nitrogen atom, to which R 1 , R 2 , and R 3 are all bonded to, is protonated;
  • R 4 for each occurrence is independently selected from:
  • R 55 for each occurrence is independently selected from:
  • a 1 , A 3 , A 4 , A 5 , A 6 , A 7 , A 8 , A 12 and A 13 are each independently selected
  • R 6 for each occurrence is independently selected from: a bond, -R 58 -N(R 42 )-R 58 -, -R 58 - CH(R 42 )-R 58 -, C 1 -C 12 alkylene or C 2 -C 12 alkenylene;
  • R 7 for each occurrence is independently selected from: a bond (i.e., when present A 2 is bonded directly t optionally substituted C1-C18 alkylene, or optionally substituted C 2 -C 18 alkenylene;
  • R 8 and R 9 are each independently selected from:
  • R 12 for each occurrence is independently selected from: -H, optionally substituted C1-C16 alkyl or optionally substituted C 2 -C 16 alkenyl;
  • R 13 for each occurrence is independently selected from: optionally substituted C1-C16 alkyl or optionally substituted C2-C16 alkenyl;
  • R 14 for each occurrence, is independently selected from: -H, optionally substituted C1-C10 alkyl, optionally substituted C1-C10 alkenyl or optionally substituted C1-C10 alkynyl;
  • R 29 through R 34 are each independently selected from: -CH 2 -, -NH-, -S-, or -O-;
  • R 38 is selected from: -H, C1-C24 alkylene or C1-C24 alkenylene;
  • R 41 for each occurrence is independently selected from: a bond or C1-C5 alkylene
  • R 42 for each occurrence is independently selected from: C6-C24 alkyl, C6-C24 alkyl carbonate, C6-C24 alkyl ether, C6-C24 alkyl ester, -R 58 -A 1 -R 59 or -R 58 -A 1 -R 59 -A 2 -R 60 , wherein R 42 has between 6-24 total carbon atoms;
  • R 61 , R 73 and R 107 are each independently selected from:
  • R 118 and R 119 are independently selected from R 1 or R 55 ; n is an integer selected from: 1, 2, 3, 4, 5 or 6; and k is an integer selected from: 1, 2, 3, 4, 5 or 6.
  • said carbocyclyl or heterocyclyl is independently an optionally substituted group selected from: cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclopropenyl, cyclobutenyl, cyclopentenyl, cyclohexenyl, cycloheptenyl, cyclobutadienyl, cyclopentadienyl, cyclohexadienyl, cycloheptadienyl or cycloheptatrienyl.
  • said carbocyclyl or heterocyclyl is independently an optionally substituted group selected from: lH-pyrrolizidinyl, 1,2-dihydroquinolinyl, 1,5- naphthyridinyl, 1,8-naphthyridinyl, lH-indazolyl, lH-isochromenyl, lH-pyrrolizidinyl, 1- naphthyl, 2H-benzo[b][l,4]oxazinyl, 2H-benzo[e][l,2]oxazinyl, 2h-chromenyl, 2-naphthyl, 4H-quinolizinyl, adeninyl, azaindazolyl, azaindolyl, benzimidazolyl, benzo[b]thiophenyl, benzo[c][l,2,5]thiadiazolyl, benzo[c]isothiazolyl, benzo[c
  • said carbocyclyl or heterocyclyl is independently an optionally substituted group selected from: furyl, imidazolyl, isothiazolyl, isoxazolyl, oxadiazolyl, oxazolyl, pyrrolyl, pyrazolyl, pyridazinyl, pyridyl (pyridinyl), pyrimidinyl, thiadiazolyl, thienyl, tetrazolyl, thiazolyl, triazolyl, , azepinyl, azetidinyl, dioxothiomorpholinyl, imidazolidinyl, morpholinyl, oxanyl, oxazinyl, oxazolidinyl, oxepinyl, oxetanyl, piperazinyl, piperidinyl, pyranyl, pyrrolidinyl, tetrahydrofuranyl,
  • said carbocyclyl or heterocyclyl is independently an optionally substituted group selected from: adamantanyl, azabicyclo[3.1.0]hexanyl, 3- azabicyclo[3.1.1]heptanyl, bicyclo[2.1.1]hexyl, bicyclo[2.2.1]heptyl, bicyclo[2.2.1]heptenyl, 3-oxa-8-azabicyclo[3.2.1]octanyl, 6-oxa-3-azabicyclo[3.1.1]heptanyl, 8-Methyl-8- azabicyclo[3.2.1]octanyl, 8-oxa-3-azabicyclo[3.2.1]octanyl, 3-oxa-6- azabicyclo[3.1.1]heptanyl, tricyclo[2.2.1.0(2,6)]heptanyl, 6,6-dimethylbicyclo[3.1.1]heptyl, or 2,6,6-trimethylbicyclo[3.1.1]heptyl
  • said carbocyclyl or heterocyclyl is independently an optionally substituted group selected from:
  • said carbocyclylene or heterocyclylene is independently an optionally substituted group selected from: furandiyl, imidazolediyl, isothiazolediyl, isoxazolediyl, oxadiazolediyl, oxazolediyl, pyrazolediyl, pyrrolediyl, pyridazinediyl, pyridinediyl, pyrimidinediyl, thiadiazolediyl, thiendiyl, tetrazolediyl, thiazolediyl, triazolediyl, azepinediyl, azetidinediyl, dioxothiomorpholinediyl, imidazolidinediyl, morpholinediyl, oxanediyl, oxazinediyl, oxazolidinediyl, o
  • said carbocyclylene or heterocyclylene is independently an optionally substituted group selected from: cyclopropylene (cyclopropanediyl), cyclobutylene (cyclobutanediyl), cyclopentylene (cylcopentanediyl), cyclohexylene (cyclohexenediyl), cycloheptylene (cycloheptanediyl), cyclopropenediyl, cyclobutenylenediyl, cyclopentenylenediyl, cyclohexenediyl, cycloheptenediyl, cyclobutadienediyl, cyclopentadienediyl, cyclohexadienediyl or cycloheptadienediyl, cycloheptatrienediyl.
  • said carbocyclylene or heterocyclylene is independently an optionally substituted group selected from: adamantanediyl, azabicyclo[3.1.0]hexanediyl, 3- azabicyclo[3.1.1]heptanediyl, bicyclo[2.1.1] hexyl, bicyclo[2.2.1]heptyl, bicyclo[2.2.1]heptenyl, 3-oxa-8-azabicyclo[3.2.1]octanediyl, 6-oxa-3- azabicyclo[3.1.1]heptanediyl, 8-oxa-3-azabicyclo[3.2.1]octanediyl, 3-oxa-6- azabicyclo[3.1.1]heptanediyl, tricyclo[2.2.1.0(2,6)]heptanyl, 6,6- dimethylbicyclo[3.1.1]heptyl, or 2,6,6-trimethylbicyclo[3.
  • said cycloalkylene in each instance, is independently selected from:
  • said carbocyclylene or heterocyclylene is independently an optionally substituted group selected from: In some embodiments, said carbocyclylene or heterocvclvlene is independently an optionally substituted group selected from:
  • R117 is selected from
  • the longest linear chain may contain non-carbon atoms including oxygen, nitrogen, sulfur and silicon, however, once the longest chain is identified, only the carbon atoms are counted; or c) .
  • a longest linear chain i.e., excluding side groups
  • a total number of atoms (including non-carbon atoms) that is selected from: 10-24, 12-22, 14-22, 16-20, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23 or 24 atoms (including non-carbon atoms); wherein each variable, for each occurrence, is independently as described above for any one of Compounds (1) - (4) or for any embodiment of any compound herein.
  • said compound comprises two R 55 groups independently selected from:
  • said R 55 said group has: a) a total number of carbon atoms selected from: 10-24, 12-22, 14-22, 16-20, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23 or 24 carbon atoms; or b) a longest linear chain (i.e., excluding side groups) with a total number of carbon atoms or total number of atoms (i.e., carbon and heteroatoms) that is selected from: 10-24, 12-22, 14-22, 16-20, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23 or 24 carbon atoms.
  • R 55 for each occurrence, is independently: wherein,
  • R 79, f or eac h occurrence are each independently selected from: C1-C12 alkyl or C1-C12 alkenyl;
  • R 61 , R 73 and R 107 are each independently selected from: R 62 , R 63 , R 67 , R 68 , R 74 , R 76 , R 108 , and R 110 , for each occurrence, are each independently selected from: C1-C12 alkylene or C1-C12 alkenylene;
  • R 55 for each occurrence, is independently: wherein:
  • the ionizable cationic lipid has a structural formula selected from:
  • R 16 for each occurrence, is independently selected from: a bond or — [CH2]k-;
  • R 62 , R 63 , R 64 , R 67 , R 68 , R 69 , R 74 , R 75 , R 76 and R 77 are each independently selected from: a bond, C1-C24 alkylene or C1-C24 alkenylene; rence, are each independently selected from:
  • R 66 , R 71 and R 79 are each independently selected from: C1-C24 alkyl or C1-C24 alkenyl;
  • a 3 , A 4 , A 5 , A 6 , A 7 and A 8 and R 1 , for each occurrence, and each remaining variable, for each occurrence, is independently as described above for any one of Compounds ( l)-(4) or any embodiment of any compound herein.
  • R 67 -A 5 -R 68 -A 6 -R 69 -R 70 -R 71 each independently has a total of 10-24 carbon atoms.
  • the ionizable cationic lipid is one of:
  • R 85 and R 90 are each independently selected from: a bond or -[CH 2 ]I-8-;
  • R 87 for each occurrence, is independently selected from: a bond or -[CH2]I-6-;
  • R 88 and R 89 are each independently -[CH2]I-IOCH 3 ;
  • R 92 for each occurrence, is -[CH 2 ]2-i6CH 3 ;
  • R 93 and R 94 are each independently selected from: -H, -D, -CH 3 , -CD 3 , - CH 2 CH 3 , -CD2CH 3 , where D denotes deuterium ( 2 H);
  • R 16 for each occurrence, is independently selected from: a bond or -[CH 2 ]k-;
  • R 104 for each occurrence, is independently selected from:
  • R 85 -R 92 and R 97 are each independently as described above for any one of Compounds (8), (10) or (11); and wherein: each remaining variable, for each occurrence, is independently as described above for any one of Compounds (l)-(8) or any embodiment of any compound herein.
  • R 85 and R 90 are the same. In some other embodiments, for any one of Compounds (1)-(12), R 85 and R 90 are each independently -[CH2]4-5--
  • R 92 is -[CH 2 ] 8 -I 2 -.
  • R 85 , R 86 , R 87 , R 88 and R 89 together, have between 14-24 or between 16-22 total carbon atoms.
  • R 90 , R 91 and R 92 together, have between 10-20 or between 12-18 total carbon atoms. In some embodiments, for any one of Compounds (1)-(12), R 88 and R 89 are identical.
  • R 85 , R 86 , R 87 and R 88 together have a total number of carbon atoms that differs by +/- 3, +/- 2 or +/-1, or is equal to the total number of carbon atoms of R 90 , R 91 and R 92 , combined.
  • _R 93 and/or R 94 are/is -H. In some embodiments, _R 95 and/or R 96 are/is -OH.
  • R 61 When R 61 is J- for any one of compounds 1-12, the carbon of R 61 may be a chiral center. In some embodiments, the chiral center is in an R configuration and in other embodiments, an S configuration.
  • the ionizable cationic lipid has a structural formula selected from any one of:
  • R 80 for each occurrence, is independently selected from: a bond, R 16 or - [d-hJi-e-O- or -O-;
  • R 16 for each occurrence, is independently selected from: a bond or -[CFhJk-;
  • R 81 , R 82 , R 83 , and R 84 are each independently selected from: a bond or - [CH 2 ]I-IO-;
  • R 61 , R 93 , R 94 , R 95 , R 96 , R 97 R 98 and each remaining variable, for each occurrence, is independently as described above for any one of Compounds ( l)-( 12) or any embodiment of any compound herein.
  • R 81 and R 83 are each independently -[CH 2 ]5-9-- In other embodiments, for any one of Compounds (1)-(19), R 82 and R 84 are each independently -[Cl-hjo-e-- In other embodiments, for any one of Compounds (1)- (19), R 81 and R 83 are each independently -[C]-hjs-io- and R 82 and R 84 are each independently - [CH2]O-6".
  • R 81 and R 82 have between 7-13 total carbon atoms. In other embodiments, for any one of Compounds (1)- (19), R 83 and R 84 , combined, have between 7-13 total carbon atoms. In other embodiments for any one of Compounds ( l)-( 19), R 81 and R 82 , combined, have between 8-12 total carbon atoms. In other embodiments, for any one of Compounds (1)-(19), R 83 and R 84 , combined, have between 8-12 total carbon atoms.
  • R 81 and R 82 , combined, and R 83 and R 84 each independently have between 7-13 or between 8-12 total carbon atoms. In other embodiments, for any one of Compounds (1)-(19), R 81 and R 82 , combined, and R 83 and R 84 , combined, each have identical number of total carbon atoms which is between 7-13 or between 8-12. In other embodiments, for any one of Compounds (1)-(19), R 81 and R 82 , combined, and R 83 and R 84 , combined, have a different number of total carbon atoms.
  • the carbon atom between R 81 and R 83 is a chiral center.
  • the chiral center is in an R configuration and in other embodiments, an S configuration.
  • the ionizable cationic lipid has a structural formula selected from any one of:
  • the ionizable cationic lipid has a structural formula selected from:
  • R 1 , R 3 , R 16 , R 36 , A 1 , and each remaining variable, for each occurrence, is independently as described above for any one of Compounds (l)-(28) or any embodiment of any compound herein.
  • an ionizable cationic lipid of the invention comprising a chiral center, e.g., as indicated by an *
  • the chiral center is in an R-configuration, in other embodiments an S-configuration, and in still further embodiments, either in an R- or an S- configuration.
  • R 1 and R 3 are each independently, -H, optionally substituted C1-C6 alkyl, optionally substituted C2-C6 alkenyl, optionally substituted C1-C6 hydroxyalkyl or optionally substituted C2-C6 hydroxyalkenyl.
  • R 1 and R 3 are each independently selected from: -H, methyl, ethyl, propyl, isopropyl, butyl, isobutyl or tertbutyl.
  • R 2 is absent or -H.
  • R 1 and R 3 are each independently selected from: -H, -D, -CH 3 , -CD 3 , -CH 2 CH 3 , - CD2CH 3 , where D denotes deuterium ( 2 H); and R 2 is absent.
  • R 1 and R 3 are each independently selected from: -H, C1-C6 alkyl, C2-C6 alkenyl, C1-C5 alkyl, C2-C5 alkenyl, C1-C4 alkyl, C2-C4 alkenyl, Ce alkyl, C5 alkyl, C4 alkyl, C 3 alkyl, C2 alkyl, Ci alkyl, C 6 alkenyl, or C 5 alkenyl, or C 4 alkenyl, or C 3 alkenyl, or C 2 alkenyl.
  • R 1 and R 3 are each independently selected from: -H or C1-C3 alkyl.
  • R T and R 3 are identical.
  • R 2 is absent.
  • R 4 for each occurrence, is independently selected from: or ; wherein R 41 ,
  • R 35 , R 55 , R 96 and each remaining variable, for each occurrence, is independently as described above for any one of Compounds (l)-(54) or any embodiment of any compound herein.
  • R 6 for each occurrence, is independently selected from C3-C24 branched alkylene or C3-C24 branched alkenylene.
  • R 6 for each occurrence, is independently selected from: C1 -C3 alkylene, C1-C9 alkylene, C2-C9 alkenylene, C1-C7 alkylene, C2-C7 alkenylene, C1 -C5 alkylene, C2-C5 alkenylene, C2-C8 alkylene, C2-C8 alkenylene, C3-C7 alkylene, C3-C7 alkenylene, C5-C7 alkylene, C5-C7 alkenylene, C12 alkylene, CH alkylene, C10 alkylene, C9 alkylene, Cs alkylene, C7 alkylene, Ce alkylene, C5 alkylene, C4 alkylene, C3 alkylene, C2 alkylene, Ci alkylene, C12 alkenylene, C11 alkenylene, Cwalkenylene, C 9 alkenylene, C8 alken
  • R 8 and R 9 are each independently C6-C12 alkyl or C6-C12 alkenyl.
  • R 8 is Ce-Cio alkyl or Ce-Cio alkenyl.
  • R 8 and R 9 together, have a total of 12-20 carbon atoms.
  • R 14 is C1-C3 alkyl, e.g., methyl or ethyl.
  • R 8 and R 9 are each independently selected from: -C10-C18 alkenyl comprising 1, 2, 3 or 4 cis double bonds, - C10-C18 alkenyl ester comprising 1, 2, 3 or 4 cis double bonds, optionally substituted -C6-C18 alkyl, -C6-718 alkyl ester, -C6-C18 alkyl ether, -C6-C18 alkyl carbonate, -C6-C18 alkenyl, -C6-C18 alkenyl ester, -C6-C18 alkenyl ether, or -C6-C18 alkenyl carbonate.
  • R 8 and R 9 are each independently -C10-C18 alkenyl comprising 2 cis (Z configuration) double bonds.
  • R 8 and R 9 are each independently selected from: optionally substituted C6-C18 alkyl, C6-718 alkylester, C6-C18 alkyl ether, C6-C18 alkyl carbonate C6-C18 alkenyl, C6-C18 alkenylester, C6-C18 alkenylether, or C6-C18 alkenylcarbonate.
  • R 8 and R 9 are each independently selected from: C6-C14 alkyl, C6-C14 alkenyl, Cs-C12 alkyl, C8-C12 alkenyl, C16 alkyl, C15 alkyl, C14 alkyl, C13 alkyl, C12 alkyl, C11 alkyl, C10 alkyl, C9 alkyl, Cs alkyl, C7 alkyl, C16 alkenyl, C15 alkenyl, C14 alkenyl, C13 alkenyl, C12 alkenyl, C11 alkenyl, C10 alkenyl, C9, alkenyl, C8 alkenyl, or C7 alkenyl; wherein R 8 and R 9 , combined, have more than 15 total carbon atoms.
  • R 8 and R 9 have an equal number of carbon atoms or are identical: wherein R 8 and R 9 , combined, have more than 15 total carbon atoms. In some embodiments, R 8 and R 9 differ in total carbon atoms from each other; R 8 and R 9 differ by one or two total carbon atoms from each other; or R 8 and R 9 differ by one total carbon atoms.
  • R 8 is C 7 alkyl and R 9 is C 8 alkyl; R 8 is C 8 alkyl and R 9 is C 7 al kyl;R 8 is C 8 alkyl and R 9 is C 9 alkyl; R 8 is C 9 alkyl and R 9 is C8 alkyl; R 8 is C9 alkyl and R 9 is C10 a I kyl;R 8 is C10 alkyl and R 9 is C9 alkyl; R 8 is C10 alkyl and R 9 is C11 alkyl; R 8 is C11 alkyl and R 9 is C10 alkyl; R 8 is C11 alkyl and R 9 is C12 alkyl; R 8 is C12 alkyl and R 9 is C11 alkyl; R 8 is C7 alkyl and R 9 is C9 alkyl; R 8 is C9 alkyl and R 9 is C7 alkyl; R 8 is C8 alkyl and R 9 is C10 a I kyl;R 8 is C10 alkyl and R 9 is C
  • R 10 and R 11 are each independently selected from: C1-C16 unbranched alkyl or C2-C16 unbranched alkenyl.
  • R 16 is C1-C3 alkylene.
  • R 29 , R 30 and R 31 are each -CH2-.
  • R 32 , R 33 and R 34 are each -O-.
  • R 35 is -OH.
  • R 29 , R 30 and R 31 are each -CH2-, and R 32 , R 33 and R 34 are each -O-. In some embodiments, for an ionizable cationic lipid of the present invention, R 29 , R 30 and R 31 are each -CH2-, R 32 and R 33 are each -O- and R 35 is -OH.
  • R 55 for each occurrence is independently:
  • R 55 for each occurrence is independently selected from: or wherein:
  • R 5 for each occurrence is independently selected from: -H, optionally substituted C1-C16 alkyl, optionally substituted C1-C16 alkyl ester, optionally substituted C 2 -Ci6 alkenyl, optionally substituted C 2 -Ci6 alkenyl ester, ; wherein, R 10 and R 11 are each independently selected from: C1-C12 unbranched alkyl, C2-C12, unbranched alkenyl, C 2 -Ci 2 unbranched alkyl, C 2 -C 12 unbranched alkenyl, C5-C7, unbranched alkyl, or C5-C7 unbranched alkenyl.
  • R 10 and R 11 are each independently selected from; C16 unbranched alkyl, C15 unbranched alkyl, C14 unbranched alkyl, C13 unbranched alkyl, C12 unbranched alkyl, C11 unbranched alkyl, C10 unbranched alkyl, C9 unbranched alkyl, Cs unbranched alkyl, C7 unbranched alkyl, Ce unbranched alkyl, C5 unbranched alkyl, C4 unbranched alkyl, C3 unbranched alkyl, C2 unbranched alkyl, or Ci unbranched alkyl.
  • R 10 and R 11 are each independently selected from; C 16 unbranched alkenyl, C15 unbranched alkenyl, C14 unbranched alkenyl, C13 unbranched alkenyl, C12 unbranched alkenyl, C11 unbranched alkenyl, C10 unbranched alkenyl, C9 unbranched alkenyl, C8 unbranched alkenyl, C7 unbranched alkenyl, Ce unbranched alkenyl, C5 unbranched alkenyl, C4 unbranched alkenyl, C3 unbranched alkenyl, or C2 alkenyl; and R 15 , R 6 , A 2 , R 7 , R 8 , R 9 and any remaining variables are as described for any one of Compounds (l)-(54) or any embodiment of any compound herein.
  • R 10 and R 11 are each independently selected from; C2-C10 unbranched alkyl, or C2-C10 unbranched alkenyl
  • R 55 for each occurrence is independently selected from: wherein:
  • R 55 for each occurrence is independently selected from: or wherein:
  • R 55 for each occurrence is independently selected from: or ; wherein: R 15 , R 6 , A 2 , R 7 , R 8 and R 9 is as described for any one of Compounds (l)-(54) or any embodiment of any compound herein.
  • R 55 for each occurrence is independently selected from: A 2 , R 7 , R 8 and R 9 is as described for any one of Compounds (l)-(54) or any embodiment of any compound herein.
  • R 5 for each occurrence is independently selected from: C1-C14 unbranched alkyl, C2-C14 unbranched alkenyl, or , wherein: R 10 and R 11 for each occurrence, are each independently selected from: C1-C12 unbranched alkyl, C2-C12, unbranched alkenyl, C2-C12 unbranched alkyl, C2-C12 unbranched alkenyl, C5-C7, unbranched alkyl, or C5-C7 unbranched alkenyl.
  • R 10 and R 11 are each independently selected from; Ci6 unbranched alkyl, C15 unbranched alkyl, C14 unbranched alkyl, C13 unbranched alkyl, C12 unbranched alkyl, Cn unbranched alkyl, C10 unbranched alkyl, C9 unbranched alkyl, Cs unbranched alkyl, C7 unbranched alkyl, Ce unbranched alkyl, C5 unbranched alkyl, C4 unbranched alkyl, C3 unbranched alkyl, C2 unbranched alkyl, or Ci unbranched alkyl.
  • R 10 and R 11 are each independently selected from; Ci6 unbranched alkenyl, C15 unbranched alkenyl, C14 unbranched alkenyl, C13 unbranched alkenyl, C12 unbranched alkenyl, Cn unbranched alkenyl, C10 unbranched alkenyl, C9 unbranched alkenyl, Cs unbranched alkenyl, C7 unbranched alkenyl, Ce unbranched alkenyl, C5 unbranched alkenyl, C4 unbranched alkenyl, C3 unbranched alkenyl, or C2 alkenyl.
  • R 10 and R 11 are each independently selected from; C2-C10 unbranched alkyl, or C2-C10 unbranched alkenyl. In further embodiments, R 10 and R 11 are identical. In some embodiments, R 5 , for each occurrence, is independently selected from: C1-C16 unbranched alkyl, C1-C16 unbranched alkyl ester, unbranched C2-C16 alkenyl or C2-C16 unbranched alkenyl ester.
  • R 5 for each occurrence, is independently selected from:C3-Ci6 branched alkyl, C3-C16 branched alkyl ester, C3-C16 branched alkenyl, or C3-C16 branched alkenyl ester.
  • R 41 -R 45 and each remaining variable, for each occurrence, is independently as described above for any one of Compounds (l)-(54) or any embodiment of any compound herein.
  • each R 55 has 12-50, 18-30, 20-30, or 12-24 total carbon atoms.
  • each R 55 comprises 0-6, 0-3, 1-3, 1, 2, 3, 4, 5 or 6 esters; 0-6, 0-3, 1-3, 1, 2, 3, 4, 5 or 6 ethers; and/or 0-6, 0-3, 1-3, 1, 2, 3, 4, 5 or 6 carbonates.
  • R 16 for each occurrence, is independently selected from: a bond or -[CH 2 ]k-;
  • R 55 for each occurrence is independently selected from:
  • R 16 , R 57 , A 1 , A 2 , R 61 , R 85 , R 86 , R 87 , R 88 , R 89 , R 90 , R 91 , R 92 , R 97 and each remaining variable, for each occurrence, is independently as described above for any one of Compounds (l)-(54) or any embodiment of any compound herein.
  • R 16 , R 57 , A 1 , R 61 , R 81 , R 82 , R 83 , R 84 , R 97 and each remaining variable, for each occurrence, is independently as described above for any one of Compounds (l)-(54) or any embodiment of any compound herein; and wherein each cyclopropyl (cPr) fused to adjacent chain
  • R 55 for each occurrence is independently selected from: or
  • R 55 for each occurrence is independently selected from:
  • R 55 is:
  • the ionizable cationic lipid has a structural formula selected from the group consisting of: Compound (121):
  • Me is methyl
  • R 55 is as defined for any one of Compounds (l)-(54) or for any embodiment herein.
  • the present invention provides for a lipid comprising lipid tail, wherein the tail is one more -R 55 .
  • the lipid further comprises a head group and an optional linker.
  • the head group is an amine containing group.
  • the head group is an amine containing ionizable cationic head group.
  • the ionizable cationic lipid is a non-fusogenic lipid.
  • a non- fusogenic lipid is meant a cationic lipid that can condense and/or encapsulate a payload, e.g., a therapeutic nucleic acid, but has insufficient fusogenic activity to effectively delivery the payload across cellular membranes.
  • the lipid nanoparticles have mean diameter of 20-75 nm or 30-100 nm.
  • the pKa' i.e., the apparent pKa, of formulated cationic lipids in particles, can be correlated with the effectiveness of the LNPs for delivery of nucleic acids.
  • the pKa' of a cationic lipid can be determined in lipid nanoparticles, e.g., using an assay based on fluorescence of 2-(p- toluidino)-6-napthalene sulfonic acid (TNS).
  • Lipid nanoparticles in PBS at a concentration of 0.4 mM total lipid are prepared using standard methods.
  • TNS can be prepared as a 100 mM stock solution in distilled water and mixed into buffers of different pH.
  • Vesicles can be diluted to 24 mM lipid in 2 mL of buffered solutions containing, 10 mM HEPES, 10 mM MES, 10 mM ammonium acetate, 130 mM NaCI, where the pH ranges from 2.5 to 11.
  • An aliquot of the TNS solution can be added to give a final concentration of 1 mM and following vortex mixing fluorescence intensity is measured at room temperature in a SLM Aminco Series 2 Luminescence Spectrophotometer using excitation and emission wavelengths of 321 nm and 445 nm.
  • a sigmoidal best fit analysis can be applied to the fluorescence data and the pKa' is measured as the pH giving rise to half-maximal fluorescence intensity.
  • the LNP comprises about 30% to about 80%, about 30% to about 70%, about 30% to about 60%, about 30% to about 50%, or about 30% to about 40%, about 40% to about 80%, about 30% to about 70%, about 40% to about 60%, about 40% to about 50%, about 50% to about 80%, about 50% to about 70%, 50% to about 60%, about 60% to about 80%, or about 70% to about 80% (ionizable) cationic lipid.
  • the lipid particles can further comprise a component, such as a sterol, to provide membrane integrity and stability of the lipid particle.
  • a component such as a sterol
  • an exemplary sterol that can be used in the lipid particle is cholesterol, or a derivative thereof.
  • Non-limiting examples of cholesterol derivatives include 5-a-cholestanol (5a-Cholestan-3
  • the cholesterol derivative is a polar analogue such as cholesteryl-(4'-hydroxy)-butyl ether.
  • cholesterol derivative is cholesteryl hemisuccinate.
  • the sterol is a sea cucumber sulphated sterol, e.g., cholest-5-en-3
  • the lipid is fecosterol, sitosterol, campesterol, stigmasterol, brassicasterol, ergosterol, tomatidine, tomatine, ursolic acid or alpha-tocopherol.
  • Exemplary cholesterol derivatives are described in International Patent Application Publication No. W02009/127060 and U.S. Patent Application Publication No. US2010/0130588.
  • the component providing membrane integrity can comprise 0-50% (mol) of total lipids present in the lipid particle (e.g., lipid nanoparticle). In some embodiments, such a component is 20-50% (mol) of total lipids present in the lipid particle (e.g., lipid nanoparticle). In some embodiments, such a component is 30-40% (mol) of total lipids present in the lipid particle (e.g., lipid nanoparticle). In some embodiments, such a component is 35-45% (mol) of total lipids present in the lipid particle (e.g., lipid nanoparticle). In some embodiments, such a component is 38-42% (mol) of total lipid present in the lipid particle (e.g., lipid nanoparticle).
  • the LNP comprises a sterol, wherein the sterol is present at a molar percentage of about 20% to about 50%. According to some embodiments, the LNP comprises a sterol, wherein the sterol is present at a molar percentage of about 30% to about 50%. According to some embodiments, the LNP comprises a sterol, wherein the sterol is present at a molar percentage of about 40% to about 50%. According to some embodiments, the LNP comprises a sterol, wherein the sterol is present at a molar percentage of about 20% to about 40%. According to some embodiments, the LNP comprises a sterol, wherein the sterol is present at a molar percentage of about 30% to about 40%. In some embodiments, the LNP comprises more than one structural lipid, e.g., two or more sterols.
  • the non-cationic lipid is typically a phospholipid and serves to increase fusogenicity and/or increase stability of the LNP, including during formation.
  • Non-cationic lipids include amphipathic lipids, neutral lipids and anionic lipids. Accordingly, the non-cationic lipid can be a neutral uncharged, zwitterionic, or anionic lipid.
  • non-cationic lipids include, but are not limited to, distearoyl-sn-glycerophosphoethanolamine, distearoylphosphatidylcholine (DSPC), dioleylphosphatidylcholine (DOPC), dipalmitoylphosphatidylcholine (DPPC), dioleylphosphatidylglycerol (DOPG), dipalmitoylphosphatidylglycerol (DPPG), dioleyl-phosphatidylethanolamine (DOPE), palmitoyloleylphosphatidylcholine (POPC), palmitoyloleylphosphatidylethanolamine (POPE), dioleyl-phosphatidylethanolamine 4-(N-maleimidomethyl)-cyclohexane-l-carboxylate (DOPE-mal), dipalmitoyl-phosphatidylethanolamine (DPPE), dimyristoylphosphoethanolamine (DMPE), distearoyl-phosphatidyl,
  • acyl groups in these lipids are preferably acyl groups derived from fatty acids having C10-C24 carbon chains, e.g., lauroyl, myristoyl, palmitoyl, stearoyl, or oleyl.
  • Preferred helper lipid DSPC, DPPC, DMPC, DOPC, POPC, DOPE, and SM.
  • the nucleobase is an alternative cytosine.
  • Exemplary nucleobases and nucleosides having an alternative cytosine include 5-aza-cytosine, 6-aza-cytosine, pseudoisocytidine, 3-methyl-cytosine (m3C), N4-acetyl-cytosine (ac4C), 5-formyl-cytosine (f5C), N4-methyl-cytosine (m4C), 5-methyl-cytosine (m5C), 5-halo-cytosine (e.g., 5-iodo- cytosine), 5-hydroxymethyl-cytosine (hm5C), 1-methyl-pseudoisocytidine, pyrrolo-cytosine, pyrrolo-pseudoisocytidine, 2-thio-cytosine (s2C), 2-thio-5-methyl-cytosine, 4-thio- pseudoisocytidine, 4-thio-l-methyl-pseudoisoc
  • the nucleobase is an alternative guanine.
  • Exemplary nucleobases and nucleosides having an alternative guanine include inosine (I), 1-methyl-inosine (mil), wyosine (imG), methylwyosine (mimG), 4-demethyl-wyosine (imG-14), iso wyo sine (imG2), wybutosine (yW), peroxywybutosine (o2yW), hydroxywybutosine (OHyW), undermodified hydroxywybutosine (OHyW*), 7-deaza-guanine, queuosine (Q), epoxyqueuosine (oQ), galacto syl-queuo sine (galQ), manno syl-queuo sine (manQ), 7-cyano-7-deaza-guanine (preQO), 7-aminomethyl-7-deaza-guanine (preQi), arch
  • Messenger RNA may be prepared according to any available technique known in the art.
  • Messenger RNA may be prepared by, for example, enzymatic synthesis which provides a process of template-directed synthesis of RNA molecules from an engineered DNA template comprised of an upstream bacteriophage promoter sequence linked to a downstream sequence encoding the gene of interest.
  • Template DNA can be prepared for in vitro transcription from several sources with appropriate techniques which are well known in the art including, but not limited to, plasmid DNA and polymerase chain reaction amplification.
  • RNA polymerase adenosine, guanosine, uridine and cytidine ribonucleoside triphosphates (rNTPs) under conditions that support polymerase activity while minimizing potential degradation of the resultant mRNA transcripts.
  • rNTPs ribonucleoside triphosphates
  • In vitro transcription can be performed using a variety of commercially available kits including, but not limited to RiboMax Large Scale RNA Production System (Promega), MegaScript Transcription kits (Life Technologies) as well as with commercially available reagents including RNA polymerases and rNTPs.
  • the methodology for in vitro transcription of mRNA is well-known in the art.
  • the lipid particle formulation is an aqueous solution.
  • the lipid particle (e.g., lipid nanoparticle) formulation is a lyophilized powder.
  • the disclosure provides for a lipid particle formulation further comprising one or more pharmaceutical excipients.
  • the lipid particle (e.g., lipid nanoparticle) formulation further comprises sucrose, Tris buffer, trehalose and/or glycine.
  • Pharmaceutical compositions for therapeutic purposes can be formulated as a solution, microemulsion, dispersion, liposomes, or other ordered structure suitable for high TNA (e.g., mRNA) concentration.
  • Sterile injectable solutions can be prepared by incorporating the TNA (e.g., mRNA) in the required amount in an appropriate buffer (e.g., pharmaceutically acceptable excipient) with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization.
  • TNA e.g., mRNA
  • an appropriate buffer e.g., pharmaceutically acceptable excipient
  • the disclosure provides for a lipid particle (e.g., lipid nanoparticle) that is either unilamellar or multilamellar in structure.
  • a lipid particle (e.g., lipid nanoparticle) formulation that comprises multi-vesicular particles and/or foam-based particles.
  • lipid particle e.g., lipid nanoparticle
  • lipid particle size By controlling the composition and concentration of the lipid conjugate, one can control the lipid particle size.
  • the pKa' of formulated cationic lipids can be correlated with the effectiveness of the LNPs for deli very of nucleic acids (see Jayaraman et al., Angewandte Chemie, International Edition (2012), 51(34), 8529-8533; Semple et al., Nature Biotechnology 28, 172-176 (2010), both of which are incorporated by reference in their entireties).
  • the preferred range of pKa' for the ionizable lipid particle is about 6-7.
  • the pKa' of the cationic lipid can be determined in lipid particles (e.g., lipid nanoparticles) using an assay based on fluorescence of 2-(p-toluidino)-6-napthalene sulfonic acid (TNS).
  • Pharmaceutical compositions or formulations can optionally comprise one or more additional active substances, e.g., therapeutically and/or prophylactically active substances.
  • Pharmaceutical compositions or formulations of the present invention can be sterile and/or pyrogen-free.
  • compositions are administered to humans, human patients or subjects.
  • active ingredient generally refers to a TNA, e.g., mRNA, to be delivered as described herein.
  • Formulations and pharmaceutical compositions described herein can be prepared by any method known or hereafter developed in the art of pharmacology.
  • compositions and formulations described herein may contain at least one TNA, such as a mRNA.
  • the composition or formulation can contain 1, 2, 3, 4 or 5 TNAs.
  • the composition or formulation can comprise a TNA in linear and/or circular form, and in single-stranded and/or doublestranded form.
  • compositions and formulations are principally directed to pharmaceutical compositions and formulations that are suitable for administration to humans, it will be understood by the skilled artisan that such compositions are generally suitable for administration to any other animal, e.g., to non-human animals, e.g. non-human mammals.
  • Pharmaceutically acceptable excipient includes, but are not limited to, any and all solvents, dispersion media, or other liquid vehicles, dispersion or suspension aids, diluents, granulating and/or dispersing agents, surface active agents, isotonic agents, thickening or emulsifying agents, preservatives, binders, lubricants or oil, coloring, sweetening or flavoring agents, stabilizers, antioxidants, antimicrobial or antifungal agents, osmolality adjusting agents, pH adjusting agents, buffers, chelators, cyoprotectants, and/or bulking crosslinked polyvinyl pyrrolidone (crospovidone), cellulose, methylcellulose, carboxymethyl cellulose, cross-linked sodium carboxymethyl cellulose (croscarmellose), magnesium aluminum silicate (V EEG UM®), sodium lauryl sulfate, etc., and/or combinations thereof.
  • crospovidone crosslinked polyvinyl pyrrolidone
  • Exemplary surface active agents and/or emulsifiers include, but are not limited to, natural emulsifiers (e.g., acacia, agar, alginic acid, sodium alginate, tragacanth, chondrux, cholesterol, xanthan, pectin, gelatin, egg yolk, casein, wool fat, cholesterol, wax, and lecithin), sorbitan fatty acid esters (e.g., polyoxyethylene sorbitan monooleate [TWEEN®80], sorbitan monopalmitate [SPAN®40], glyceryl monooleate, polyoxyethylene esters, polyethylene glycol fatty acid esters ( e.g., CREMOPHOR®), polyoxyethylene ethers (e.g., polyoxyethylene lauryl ether [BRIJ®30]), PLURONIC® block copolymers, e.g.
  • natural emulsifiers e.g., acacia, agar,
  • Oxidation is a potential degradation pathway for TNAs, especially for liquid or freeze-dried DNA formulations.
  • antioxidants can be added to the formulations.
  • Exemplary antioxidants include, but are not limited to, alpha tocopherol, ascorbic acid, ascorbyl palmitate, benzyl alcohol, butylated hydroxyanisole, m-cresol, methionine, butylated hydroxytoluene, monothioglycerol, sodium or potassium metabisulfite, propionic acid, propyl gallate, sodium ascorbate, etc., and combinations thereof.
  • Exemplary chelating agents include, but are not limited to, ethylenediaminetetraacetic acid (EDTA), disodium edetate, diethylenetriaminepentaacetic acid (DTPA, in ionized forms), citric acid monohydrate, fumaric acid, malic acid, phosphoric acid, sodium edetate, tartaric acid, Trisodium edetate, etc., and combinations thereof.
  • EDTA ethylenediaminetetraacetic acid
  • DTPA diethylenetriaminepentaacetic acid
  • citric acid monohydrate fumaric acid, malic acid, phosphoric acid, sodium edetate, tartaric acid, Trisodium edetate, etc., and combinations thereof.
  • antimicrobial or antifungal agents include, but are not limited to, benzalkonium chloride, benzethonium chloride, methyl paraben, ethyl paraben, propyl paraben, butyl paraben, benzoic acid, hydroxybenzoic acid, potassium or sodium benzoate, potassium or sodium sorbate, sodium propionate, sorbic acid, etc., and combinations thereof.
  • Exemplary preservatives include, but are not limited to, vitamin A, vitamin C, vitamin E, beta-carotene, citric acid, ascorbic acid, butylated hydroxyanisole, ethylenediamine, sodium lauryl sulfate (SLS), sodium lauryl ether sulfate (SLES), p-amino benzoic acid, methyl and/or propyl parabens, etc., and combinations thereof.
  • the pH of polynucleotide solutions is maintained between pH 5 and pH 8 to improve stability.
  • cryoprotectants include, but are not limited to mannitol, sucrose, trehalose, lactose, glycerol, dextrose, etc., and combinations thereof.
  • the pharmaceutical composition or formulation described here can contain a bulking agent in lyophilized polynucleotide formulations to yield a "pharmaceutically elegant" cake, stabilize the lyophilized polynucleotides during long term (e.g., 36-month) storage.
  • Exemplary bulking agents of the present invention can include, but are not limited to sucrose, trehalose, mannitol, glycine, lactose, raffinose, and combinations thereof.
  • the pharmaceutical composition or formulation further comprises a delivery agent.
  • the pharmaceutical compositions can be presented in unit dosage form.
  • a unit dosage form will typically be adapted to one or more specific routes of administration of the pharmaceutical composition.
  • the unit dosage form is adapted for intravenous, intramuscular, or subcutaneous administration.
  • the unit dosage form is adapted for intrathecal or intracerebroventricular administration.
  • the unit dosage form is adapted for administration by inhalation.
  • the unit dosage form is adapted for administration by a vaporizer.
  • the unit dosage form is adapted for administration by a nebulizer.
  • the unit dosage form is adapted for administration by an aerosolizer.
  • the LNP/TNA is for administration at a dose of about 0.02 pg to about 50 mg, about 0.02 pg to about 0.2 pg, or about 0.2 pg to about 2.0 pg, about 1 pg to about 25 pg, about 25 pg to about 50 pg, about 50 pg to about 100 pg, about 100 pg to about 200 pg, about 200 pg to about 300 pg, about 300 pg to about 400 pg, about 400 pg to about 500 pg, about 500 pg to about 750 pg, about 750 pg to about 1.0 mg, about 1 mg to about 10 mg, about 10 mg to about 25 mg, about 25 mg to about 50 mg.
  • the pharmaceutical composition comprising an LNP and a TNA can be used to deliver any TNA in accordance with the description above to treat, prevent, or ameliorate the symptoms associated with any disorder related to gene expression.
  • disease states include, but are not-limited to: cystic fibrosis (and other diseases of the lung), hemophilia A, hemophilia B, thalassemia, anemia and other blood disorders, AIDS, Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, epilepsy, and other neurological disorders, cancer, diabetes mellitus, muscular dystrophies (e.g., Duchenne, Becker), Hurler's disease, adenosine deaminase deficiency, metabolic defects, retinal degenerative diseases (and other diseases of the eye), mitochondriopathies (e.g., Leber's hereditary optic neuropathy (LHON), Leigh syndrome, and subacute sclerosing encephalopathy), my
  • the ceDNA vectors as disclosed herein can be advantageously used in the treatment of individuals with metabolic disorders (e.g., ornithine transcarbamoylase deficiency).
  • the pharmaceutical composition comprising a LNP and a TNA can be used to treat, ameliorate, and/or prevent a disease or disorder caused by mutation in a gene or gene product (i.e., a genetic disorder) include, but are not limited to, metabolic diseases or disorders (e.g., Fabry disease, Gaucher disease, phenylketonuria (PKU), glycogen storage disease); urea cycle diseases or disorders (e.g., ornithine transcarbamoylase (OTC) deficiency); lysosomal storage diseases or disorders (e.g., metachromatic leukodystrophy (MLD), mucopolysaccharidosis Type II (MPSII; Hunter syndrome)); liver diseases or disorders (e.g., progressive familial intrahepatic cholest
  • MLD metachromat
  • the genetic disorder is hemophilia A, hemophilia B, phenylketonuria (PKU, Gaucher disease Types I, II and III, Stargardt macular dystrophy, Leber congenital amaurosis (LCA), Usher syndrome, wet AMD.
  • the pharmaceutical composition comprising an LNP and a TNA may be employed to deliver a heterologous nucleotide sequence, e.g., to correct an abnormal level and/or function of a gene product, such as an absence of, or a defect in, a protein, that results in the disease or disorder.
  • the TNA in lipid nanoparticles as described herein can produce a functional protein and/or modify levels of the protein to alleviate or reduce symptoms resulting from, or confer benefit to, a particular disease or disorder caused by the absence or a defect in the protein.
  • the TNA may be used for producing a functional protein or increased expression of a protein, such as OTC enzyme, Factor VIII, Factor IX, and Factor X, phenylalanine hydroxylase enzyme, alpha galactosidase or beta glucocerebrosidase, arylsulfatase A, iduronate-2-sulfatase, cystic fibrosis transmembrane conductance regulator, G6Pase enzyme, ATP8B 1, ABCB 11, ABCB4, or TJP2.
  • a protein such as OTC enzyme, Factor VIII, Factor IX, and Factor X, phenylalanine hydroxylase enzyme, alpha galactosidase or beta glucocere
  • exemplary TNA encode a protein selected from: lysosomal enzymes (e.g., hexosaminidase A, iduronate sulfatase, associated, erythropoietin, angiostatin, endostatin, superoxide dismutase, globin, leptin, catalase, tyrosine hydroxylase, as well as cytokines (e.g., a interferon, b-interferon, interferon-gamma, interleukin-2, interleukin-4, interleukin 12, granulocyte- macrophage colony stimulating factor, lymphotoxin, and the like), peptide growth factors and hormones (e.g., somatotropin, insulin, insulin-like growth factors 1 and 2, platelet derived growth factor (PDGF), epidermal growth factor (EGF), fibroblast growth factor (FGF), nerve growth factor (NGF), neurotrophic factor-3 and 4, brain-derived lys
  • the pharmaceutical compositions comprising a lipid nanoparticle (LNP) and a therapeutic nucleic acid (TNA), as described herein, can be administered to an organism for transduction of cells in vivo.
  • the TNA can be administered to an organism for transduction of cells ex vivo.
  • administration is by any of the routes normally used for introducing a molecule into ultimate contact with blood or tissue cells. Suitable methods of administering such nucleic acids are available and well known to those of skill in the art, and, although more than one route can be used to administer a particular composition, a particular route can often provide a more immediate and more effective reaction than another route.
  • Exemplary modes of administration of the pharmaceutical composition of the invention include oral, rectal, buccal (e.g., sublingual), transmucosal (regardless of anatomical location), parenteral , including, but not limited to intravenous, intraarterial, subcutaneous, intradermal, intracranial, intramuscular (including administration to skeletal, diaphragm and/or cardiac muscle], intrapleural, intracerebral, intraosseous (including bone marrow), and intraarticular, intranasal, inhalation (e.g., via an aerosol), vaginal, intrathecal, intraocular, transdermal, intraendothelial, in utero (or in ovo), topical (e.g., to both skin and mucosal surfaces, including airway surfaces, and transdermal administration), intralymphatic, intranodal and the like, as well as direct tissue or organ injection (e.g., to liver, eye, skeletal muscle, cardiac muscle, diaphragm muscle or brain).
  • Administration of the pharmaceutical composition can be to any site in a subject, including, without limitation, a site selected from the group consisting of the brain, a skeletal muscle, a smooth muscle, the heart, the diaphragm, the airway epithelium, the liver, the kidney, the spleen, the pancreas, the skin, and the eye.
  • the pharmaceutical composition can be administered to skeletal muscle includes but is not limited to administration to skeletal muscle in the limbs (e.g., upper arm, lower arm, upper leg, and/or lower leg), back, neck, head (e.g., tongue), thorax, abdomen, pelvis/perineum, and/or digits, by intravenous administration, intra-arterial administration, intraperitoneal administration, limb perfusion, (optionally, isolated limb perfusion of a leg and/or arm; see, e.g., Arruda et al. 2005, Blood 105: 3458-3464), and/or direct intramuscular injection.
  • limbs e.g., upper arm, lower arm, upper leg, and/or lower leg
  • head e.g., tongue
  • thorax e.g., abdomen, pelvis/perineum, and/or digits
  • intravenous administration e.g., intra-arterial administration, intraperitoneal administration, limb perfusion, (option
  • pharmaceutical composition is administered to cardiac muscle, including left atrium, right atrium, left ventricle, right ventricle and/or septum, e.g., by intravenous administration, intra-arterial administration such as intra-aortic administration, direct cardiac injection (e.g., into left atrium, right atrium, left ventricle, right ventricle), and/or coronary artery perfusion.
  • intravenous administration intra-arterial administration such as intra-aortic administration
  • direct cardiac injection e.g., into left atrium, right atrium, left ventricle, right ventricle
  • coronary artery perfusion e.g., coronary artery perfusion.
  • Administration to diaphragm muscle can be by any suitable method including intravenous administration, intra-arterial administration, and/or intra-peritoneal administration.
  • Administration to smooth muscle can be by any suitable method including intravenous administration, intra-arterial administration, and/or intra-peritoneal administration.
  • administration can be to endothelial cells present in, near, and/or on smooth muscle.
  • pharmaceutical composition comprising is administered to the CNS (e.g., to the brain or to the eye).
  • the pharmaceutical composition may be introduced into the spinal cord, brainstem (medulla oblongata, pons), midbrain (hypothalamus, thalamus, epithalamus, pituitary gland, substantia nigra, pineal gland), cerebellum, telencephalon (corpus striatum, cerebrum including the occipital, temporal, parietal and frontal lobes, cortex, basal ganglia, hippocampus and porta amygdala), limbic system, neocortex, corpus striatum, cerebrum, and inferior colliculus.
  • the pharmaceutical compositions may also be administered to different regions of the eye such as the retina, cornea and/or optic nerve., e.g., via subretinal injection, suprachoroidal injection, or intravitreal injection
  • the pharmaceutical composition may be delivered into the cerebrospinal fluid (e.g., by lumbar puncture).
  • the pharmaceutical composition may be administered to the desired region(s) of the CNS by any route known in the art, including but not limited to, intrathecal, intra-ocular, intracerebral, intraventricular, intravenous (e.g., in the presence of a sugar such as mannitol), intranasal, intra-aural, intra-ocular (e.g., intra- vitreous, sub-retinal, anterior chamber) and peri-ocular (e.g., sub-Tenon's region) delivery as well as intramuscular delivery with retrograde delivery to motor neurons.
  • repeat administrations of the therapeutic product can be made until the appropriate level of expression has been achieved.
  • a therapeutic nucleic acid can be administered and re-dosed multiple times. Examples
  • Another batch with 2.0 g of heptadecan-9- yl 4-(benzylamino)butanoate (52-6 was carried out by following the above procedure and both the crude mixtures were combined and purified by silica gel flash chromatography using 0-30% ethyl acetate-dichloromethane to obtain decyl 4-(benzyl(4-(heptadecan-9- yloxy)-4-oxobutyl)amino)butanoate (52-7, 2.35 g, 61%).
  • the 1 H NMR showed some impurities, and this product was used in the next step without further purification.
  • decyl 4-((4-(heptadecan-9-yloxy)-4-oxobutyl)amino)butanoate (52-8) A solution of decyl 4-(benzyl(4-(heptadecan-9-yloxy)-4-oxobutyl)amino)butanoate (36-7, 2.35 g, 3.6 mmol) in ethyl acetate (100 mL) was stirred overnight under hydrogen atmosphere in the presence of 10% Pd/C (wet, 500 mg). The reaction mixture was filtered through celite rinsing with ethyl acetate and dichloromethane.
  • the crude was purified by silica gel flash chromatography using 0-30% ethyl acetate-dichloromethane to obtain decyl 4- ((cyanomethyl)(4-(heptadecan-9-yloxy)-4-oxobutyl)amino)butanoate (52-9, 148 mg, 10%) and heptadecan-9-yl 4-(2-oxopyrrolidin-l-yl)butanoate (750 mg, 72%).
  • the acetonitrile-substituted tertiary amine is treated with HCI gas in methanol-containing solvent.
  • the orthoester is isolated from unreacted material and any methyl esters side product occurring by transesterification.
  • the decyl 4-(((trimethoxy)methyl)(4-(heptadecan-9-yloxy)-4-oxobutyl)amino)butanoate (52- 10) is treated with Tris(hydroxymethyl)nitromethane to form the bicyclic system with a nitro group opposite to the lipid tails.
  • the resulting nitro-bicyclo lipid compound is reduced with hydrogen gas over PtO2 to produce the desired amino-bicyclo lipid (Compound 36).
  • the amino lipid is purified by column chromatography over alumina, taking care to purify out the unreacted nitro analog.
  • Acidic hydrolysis of bicyclic ring system in Compound 36 is accomplished without hydrolysis of the carboxyl esters of the tails by mild acid treatment to obtain Compound 52.
  • heptadecan-9-ol (51-14, 18.50 g, 72.0 mmol) was added followed by diisopropylethylamine (9.30 g, 12.5 mL, 72.0 mmol) and the reaction mixture stirred at room temperature overnight. The reaction was quenched with sodium carbonate (IM), washed with water and the solvent removed to get the crude product (51- 15) which was used without further purification.
  • IM sodium carbonate
  • an ionizable cationic lipid of the invention molecular weight range approx. 500-1000 to formulate an LNP dispersion with a smallmedium size mRNA (for example 500-4000 nucleotides)
  • An ionizable lipid of the invention, 1,2- distearoyl-sn-glycero-3-phosphocholine (DSPC), cholesterol, and 1,2-dimyristoyl-rac- glycero-3-methylpolyoxyethylene (DMG-PEG) are combined in a 40:15:43:2 molar ratio in anhydrous ethanol at a concentration 12.5 mM (1.85 mg/mL).
  • mRNA was diluted in RNase- free 50 mM citrate buffer pH 4.0 to obtain a lipid:mRNA mass ratio of 10:1 (ionizable lipidmucleotide 3:1 molar ratio, often referred to as N:P ratio).
  • the lipid solution in ethanol is then rapidly mixed in a 3:1 (aqueous to ethanol) volume ratio through a micromixer chip using a Nanoassemblr or Egnyte benchtop instrument (Precision Nanosystems) at a total flow rate (TFR) of 12 mL/min.
  • TFR total flow rate
  • This combination of concentrations and mixing ratio results in a 3:1 molar ratio of ionizable cationic lipid to mRNA phosphate groups and a total lipid to mRNA mass ratio of approximately 10:1.
  • An alternative approach to achieve 3:1 molar ratio of ionizable cationic lipid to mRNA phosphate groups and a total lipid to mRNA mass ratio of approximately 20:1 uses 25 mM lipid stock (double the concentration in ethanol) with the same mRNA concentration at the start and the same mixing configuration. This composition may be preferably in some cases for more complete encapsulation.
  • the mixed LNP dispersion in ethanol/water resulting from the encapsulation step is diluted 10-fold into 50 mM citrate buffer at pH 6 and subjected to tangential flow filtration (TFF) using a 300k molecular weight cut-off membrane (mPES) until concentrated to the original volume. Subsequently, the citrate buffer is replaced with a buffer containing 10 mM Tris buffer at pH
  • the LNP dispersion is concentrated to a volume that leads to no more than about 0.5 mg/mL mRNA concentration, as measured by Ribogreen (QUANT-IT kit, ThermoFisher) with TritonX to access the RNA for quantification. Then the LNP is filtered using a 0.2 micron PES syringe filter, aliquoted into vials, and frozen at l°C/min using a Corning® CoolCell® LX Cell Freezing Container until the samples reach -80°C.
  • RNA concentration a percentage of input RNA recovered (% recovery), and the encapsulation efficiency (%encaps.) are determined using a Ribogreen assay, which is described elsewhere.
  • the Z-avg diameter (nm) and polydispersity index (PDI) are measured using dynamic light scattering (Malvern Zetasizer) following a 1:100 dilution in phosphate buffered saline (PBS).
  • PBS phosphate buffered saline
  • the messenger RNA-containing LNP composition is characterized using analytical methods to determine the loading of messenger RNA, the percentage of messenger RNA that is encapsulated, and the size of the particles.
  • the total amount of messenger RNA contained in the sample and the percentage of that messenger RNA that is encapsulated is determined using a fluorescence assay employing Ribogreen, a dye that becomes more emissive upon binding messenger RNA and the fluorescence relates quantitatively to the amount of RNA to which the dye binds.
  • the total amount of messenger RNA is determined by disrupting the LNP with 1 wt% Triton-X 100 to expose the encapsulated messenger RNA, adding the dye, and comparing the emission intensity against a standard curve prepared using ribosomal RNA.
  • the amount of unencapsulated messenger RNA is measured in a similar manner with the detergent disruption of the LNP is omitted. With the total amount of messenger RNA known and the amount of unencapsulated messenger RNA known, the percent encapsulated messenger RNA is calculated by the following formula:
  • RNATOTAL RNAUNENCAPSULATED/ RNATOTAL
  • RNATOTAL and RNAUNENCAPSULATED are, respectively, the concentrations of total messenger RNA and unencapsulated messenger RNA.
  • the total messenger RNA content varies based on formulation, but generally fall in the range of 0.030-0.200 mg/mL.
  • the size of LNP is measured using dynamic light scattering of a sample diluted 1:100 in PBS buffer.
  • pKa' is determined as the EC50 of this curve where half of the ionizable amines are expected to be protonated.
  • an LNP to transfect cultured cells with a TNA e.g., mRNA
  • a TNA e.g., mRNA
  • in vitro assay based on the percentage of cells expressing the protein of interest. Specifically, 1 million BHK-21 cells are co-incubated with mRNA-LNP of varying concentration (highest 6.25 ng per well, lowest 0.39 ng per well) in 0.3 mL of media for 17- 19 hours at 37°C with 5% CO2.
  • the in vivo potency of messenger RNA-LNPs may be evaluated using two related, but distinct, approaches. Firstly, to quantify the location, relative amount, and the duration of protein expression, LNPs formulated with a messenger RNA expressing luciferase is injected into mice, for example intramuscularly in the hind leg muscles. At defined time points, such as daily, the mice are administered luciferin, and the bioluminescence is imaged and quantified. Secondly, the ability of messenger RNA-LNPs to act as a vaccine is evaluated by measuring the antibody- and cell based immune response following a prime-boost vaccination schedule.
  • a priming vaccination given on Day 0 via intramuscular injection is followed 21 days later with a boosting vaccination.
  • the mice are sacrificed and relevant tissues, such as serum, the spleen, or lymph nodes are collected for further analysis. Serum is analyzed using an ELISA to determine the antigen- specific antibody response.
  • Splenocytes are analyzed for antigen- specific cytokine production, for example by using flow cytometry. Taken together, these in vivo assays can demonstrate that the messenger RNA-LNP is able to induce protein expression of the antigen of interest that initiates a productive antigen- specific immune response required for effective vaccination.
  • An ionizable lipid (IL) of the invention and/or known lipid e.g., DLin-MC3-DMA ("MC3")
  • MC3 1,2- distearoyl-sn-glycero-3-phosphocholine
  • chol cholesterol
  • AAP astaxanthin amino prodrug
  • compound 90 or 89 which are ionizable
  • DMG-PEG 1,2- dimyristoyl-rac-glycero-3-methylpolyoxyethylene
  • MC3 DLin-MC3-DMA
  • DSPC 1,2- distearoyl-sn-glycero-3-phosphocholine
  • DOPE dioleyl-phosphatidylethanolamine
  • a particular example is IL:DSPC:chol:AAP:DMG-PEG, having molar ratio of 45:12:39:2:2, in anhydrous ethanol at a concentration 12.5 mM (1.85 mg/mL).
  • Modified mRNA encoding for firefly Luciferase using 5-methoxyuridine substitutions for all uridines was diluted in RNase- free 50 mM acetate buffer pH 4.0 to obtain a lipid:mRNA mass ratio of 10:1 (ionizable lipidmucleotide 4.5:1 molar ratio, often referred to as N:P ratio).
  • the lipid solution in ethanol is then rapidly mixed in a 3:1 (aqueous to ethanol) volume ratio through a micromixer chip using a Nanoassemblr, Egnyte benchtop instrument (Precision Nanosystems) or similar mixer setup at a total flow rate (TFR) of 12 mL/min.
  • This combination of concentrations and mixing ratio results in a 3:1 to 4.5:1 molar ratio of ionizable lipid to mRNA phosphate groups and a total lipid to mRNA mass ratio of approximately 10:1 - 15:1.
  • the mixed LNP dispersion in ethanol/water resulting from the encapsulation step is diluted 10-fold into 50 mM citrate buffer at pH 6 and subjected to dialysis against phosphate buffered saline (PBS).
  • PBS phosphate buffered saline
  • tangential flow filtration (TFF) using a 300kDa molecular weight cut-off membrane (mPES) is applied until concentrated to the original volume.
  • TFF tangential flow filtration
  • mPES molecular weight cut-off membrane
  • the citrate buffer is replaced with a buffer containing 10 mM Tris buffer at pH 7.5 (measured at around 20°C), 80 mM sodium chloride, and 3% sucrose using diafiltration with 10 diavolumes.
  • the LNP dispersion is concentrated to a volume that leads to no more than about 0.5 mg/mL mRNA concentration, as measured by Ribogreen (QUANT-IT kit, ThermoFisher) with TritonX to rupture LNPs and access the encapsulated RNA for quantification. Then the LNP is filtered using a 0.2 micron PES syringe filter, aliquoted into vials, and frozen at l°C/min using a Corning® CoolCell® LX Cell Freezing Container until the samples reach -80°C. Samples are stored at -80°C and thawed on wet ice before analysis or use.
  • RNA concentration The total RNA concentration, the percentage of input RNA recovered (% recovery), and the encapsulation efficiency (%encapsulation) are determined using a Ribogreen assay.
  • the Z-avg diameter (nm) and polydispersity index (PDI) are measured using dynamic light scattering (Malvern Zetasizer) following a 1:100 dilution in phosphate buffered saline (PBS). LNPs are then stored refrigerated and in the dark.
  • PBS phosphate buffered saline
  • the messenger RNA-containing LNP composition is characterized using analytical methods to determine the loading of messenger RNA, the percentage of messenger RNA that is encapsulated, and the size of the particles.
  • the total amount of messenger RNA contained in the sample and the percentage of that messenger RNA that is encapsulated is determined using a fluorescence assay employing Ribogreen, a dye that becomes more emissive upon binding messenger RNA.
  • the total amount of messenger RNA is determined by disrupting the LNP with 1 wt% Triton-X 100 to expose the encapsulated messenger RNA, adding the dye, and comparing the emission intensity against a standard curve prepared using ribosomal RNA.
  • the amount of unencapsulated messenger RNA is measured in a similar manner with the detergent disruption of the LNP is omitted. With the total amount of messenger RNA known and the amount of unencapsulated messenger RNA known, the percent encapsulated messenger RNA is calculated thus:
  • RNATOTAL RNATOTAL - RNAUNENCAPSULATED/ RNATOTAL
  • RNATOTAL and RNAUNENCAPSULATED are, respectively, the concentrations of total messenger RNA and unencapsulated messenger RNA.
  • the total messenger RNA content varies based on formulation, but generally fall in the range of 0.030-0.200 mg/mL.
  • the size distribution parameters of LNP are measured using dynamic light scattering of a sample diluted 1:100 in PBS buffer.
  • Fluorescence imaging and spectroscopy can be used to identify and quantify xanthophyll components in the LNP.
  • the fluorescence is read on a plate reader at 25°C with an excitation setting of 450 nm and an emission setting of 560 nm.
  • Luciferase from modified mRNA by in vitro cell transfection with LNPs.
  • LNPs from the preparations entrapping the Firefly Luciferase mRNA with uridine modifications were dosed into wells containing a single cell line, either HEK293 or Huh7, at doses of 125, 250 and 500 ng of mRNA, in addition to a PBS blank dose denoted as 0 ng mRNA.
  • the luminescence from the expressed Luciferase is tested after dosing of Luciferin dye, a substrate for the enzyme which is luminescent, and the signal is proportional to the enzyme activity. Viability of cells is measured by Promega OneGlo + Tox luciferase detection kit.
  • helper lipid specifically the phospholipids
  • DSPC used in most formulations with RNA, including ONPATTRO and the mRNA vaccines for Covidl9
  • DOPC and DOPE DOPC
  • MC3 D-Lin-MC3-DIVIA
  • MC3 is used for the effective delivery of RNAs in vivo, although it is known to be reactogenic induce immuno-inflammatory responses (either TH1 or TH2).
  • luciferase Functional expression of luciferase was measured by luminescence, a direct enzymatic activity readout, in human embryonic kidney 293T cells (HEK293T) and in hepatic cell line Huh-7 (both epithelial-like), with increasing mRNA payload (125-250-500ng).
  • LNP formulations containing both standard and reduced molar ratio of MC3, with or without Axelyf AAP proprietary ionizable lipids induced dose-dependent luciferase expression luciferase expression, with or without AAP modifier components showed satisfactory in HEK293T cells ( Figure 4A and 4B, respectively) with notably lower expression for Compound (52) in formulations 7 and 9.
  • formulation Compound (30) using DSPC yielded a strikingly high expression of luciferase in HEK293T cells as compared with all other formulations, suggesting good mRNA stability, and formulation Compound (52) DOPE drove expression in Huh7, whereas other formulations had weaker activity. After one month at +4°C, different formulations retained different tropism.
  • Fluorescence spectral scan revealed emission of Compound (89) and Astaxanthin 2% in EtOH around 575 nm with 480 nm excitation, but little fluorescence of the lipid nanoparticles containing these compounds ( Figure 9).
  • LNPs compositions with ionisable lipids Cpd 51 or Cpd 53 were made and compared to LNP with MC3 and LP-01 which are widely used in the literature.
  • LNP containing Cpd 51 or Cpd 53 had Z-average values in line with control LNPs ( Figure 13A). PDI values were lower than MC3 LNPs, but in line with
  • LNPs containing Cpd 51 or Cpd 53 were equipotent to LP-01 in terms of in vitro expression of Luciferase in HEK293T and Huh7 cells as a result of transfection of different amount of modified mRNA.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Inorganic Chemistry (AREA)
  • Saccharide Compounds (AREA)
  • Organic Low-Molecular-Weight Compounds And Preparation Thereof (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

The present invention provides for lipids that may be formulated in a delivery vehicle to facilitate the encapsulation of a wide range of payloads including therapeutic and diagnostic agents, such as, without limitation, nucleic acids (e.g., RNA or DNA), proteins, peptides, and small molecule active pharmaceutical ingredients (APIs).

Description

Lipids and Lipid Nanoparticles
Cross-Reference to Related Applications
This application claims the benefit of U.S. Provisional Application Serial No. 63/536,997, filed September 7, 2023, and U.S. Provisional Application Serial No. 63/664,820, filed June 27, 2024, the disclosure of which is hereby incorporated by reference in its entirety, including all figures, tables and amino acid or nucleic acid sequences.
Background of the invention
Lipid nanoparticles (LNPs) are a type of nanoscale delivery system composed of lipids that have gained significant attention and validation in the field of medicine. These particles have shown great potential for the delivery of various therapeutic agents, including nucleic acids such as DNA, RNA, and siRNA. LNPs offer several advantages over other delivery systems, including intracellular delivery of sensitive nucleic acids like mRNA, biocompatibility, biodegradability, and the ability to encapsulate hydrophobic and hydrophilic molecules.
The development of LNPs as a drug delivery system can be traced back to the early 1990s. Researchers recognized the potential of lipids to form stable nanoparticles and protect encapsulated molecules. The initial focus of LNP research was on gene therapy, where the delivery of nucleic acids posed significant challenges due to their inherent instability and the need for efficient intracellular delivery. Over the years, advancements in lipid chemistry, formulation techniques, and manufacturing processes have contributed to the refinement of LNP-based delivery systems.
LNPs have shown tremendous promise in the field of medicine, particularly in the delivery of nucleic acid-based therapeutics. This includes the delivery of small interfering RNA (siRNA) for gene silencing, messenger RNA (mRNA) for protein synthesis, and gene editing tools such as CRISPR-Cas9. LNPs protect the encapsulated nucleic acids from degradation, enhance their cellular uptake, and facilitate their release at the target site, thereby improving therapeutic efficacy.
One of the significant breakthroughs in LNP-based medicine came with the development of COVID-19 mRNA vaccines. The mRNA vaccines from Pfizer-BioNTech and Moderna, which use LNPs as delivery vehicles, demonstrated remarkable effectiveness in combating COVID- 19. These vaccines not only showcased the potential of LNPs but also paved the way for the rapid development and deployment of mRNA-based vaccines for other diseases.
Despite the progress made, there are several hurdles and problems that remain with LNP- based delivery systems. One of the main challenges is achieving efficient and targeted delivery to specific tissues or cells. LNPs face barriers such as uptake by the reticuloendothelial system (RES) and clearance by the liver and kidneys, which can limit their circulation time and reduce their therapeutic efficacy.
Another challenge is the potential for toxicity and immune responses. LNPs may induce an immune response due to their foreign nature, leading to adverse reactions or decreased therapeutic outcomes.
Furthermore, the scalability and manufacturing of LNPs pose significant challenges. Maintaining batch-to-batch consistency, controlling particle size, and optimizing production processes are crucial for the successful translation of LNPs from the laboratory to large-scale manufacturing.
There exists a need to develop new lipid compounds, e.g., cationic lipids, ionizable lipids (generally amines, tertiary substituted), polymer-conjugated lipids, and structural lipids, that afford efficient delivery of the therapeutic agents, sufficient activity of the therapeutic agents (e.g., expression of mRNA after delivery), optimal pharmacokinetics, and/or other suitable physiological, biological, and/or therapeutic properties.
Brief Summary of the Invention
The present invention provides for lipids that may be formulated in a delivery vehicle to facilitate the encapsulation of a wide range of payloads including therapeutic and diagnostic agents, such as, without limitation, nucleic acids (e.g., RNA or DNA), proteins, peptides, and small molecule active pharmaceutical ingredients (APIs).
The lipid compounds of the present invention can be used in combination with other lipid components, such as neutral lipids, sterols and polymer conjugated lipids, to form lipid nanoparticles for delivery of payloads both in vitro and in vivo, for therapeutic or prophylactic purposes, including vaccination. Thus, the present invention further provides for lipid nanoparticles (LNPs) comprising said lipids as well as methods of administering LNPs to a subject, e.g., delivering an mRNA and achieving prolonged expression of a desired polypeptide in the animal or human subject.
In further embodiments, the invention provides for a method of delivering and/or producing a polypeptide of interest in a cell.
In further embodiments, the invention provides for a method of treating a disease, disorder, or condition in a subject, comprising the step of administering the foregoing lipid nanoparticle and/or lipid nanoparticle composition, to a subject in need of such treatment. The lipid particle and/or lipid particle composition may be also delivered to a subject as a component of a vaccine or diagnostic composition.
Brief Description of the Drawings
FIGs. 1A-1B. Size assessed by Dynamic Light Scattering (DLS) of LNPs with and without fifth component. FIG. 1A) MC3 LNPs with 45mol% MC3 without and with AAP modifiers (50% denotes a control without AAP and having 50mol % MC3) with compositions according to Table 1. FIG. IB) Size assessed by DLS of LNPs with compositions according to Table 1. MC3 is the chemical named (6Z,9Z,28Z,31Z)-heptatriaconta-6,9,28,31-tetraen-19-yl 4- (dimethylamino)butanoate.
FIGs. 2A-2B. Polydispersity of LNPs with and without fifth component. FIG. 2A) MC3 LNPs with 45mol% MC3 without and with AAP modifiers (50% denotes a control without AAP and containing 50mol % MC3). FIG. 2B) Polydispersity of LNPs with compositions and structures of ionizable lipids per Table 1.
FIGs. 3A-3B. Size and polydispersity of LNPs with and without fifth component, on day 14 after formulation and storage in refrigerator. FIG. 3A) MC3 LNP sizes with 45mol% MC3 without and with AAP modifiers (50% denotes a control without AAP and containing 50mol % MC3) according to Table 1. FIG. 3B) Polydispersity of LNPs with compositions according to Table 1.
FIGs. 4A-4B. In vitro expression of Firefly Luciferase in HEK293 cells, measured by luminescence, expressed as relative light units (RLU). MC3 LNPs with 45mol% MC3 with AAP and without modifiers (FIG. 4A). Novel ionizable lipid-based LNPs with same mRNA (FIG. 4B) show proportional increase in luminescence with increased dose. Compositions are according to Table 1.
FIGs. 5A-5B. In vitro expression of Firefly Luciferase in Huh7 cells, measured by luminescence, expressed as relative light units (RLU). MC3 LNPs with 45mol% MC3 with AAP and without modifiers (FIG. 5A). Novel ionizable lipid-based LNPs with same mRNA (FIG. 5B) show proportional increase in luminescence with increased dose. Compositions are according to Table 1.
FIGs. 6A-6B. Viability of cells in the in vitro expression of Firefly Luciferase in HEK293T cells, expressed as relative fluorescence units (RFU) using TiterGlo kit. MC3 LNPs with 45mol% MC3 with AAP and without modifiers (FIG. 6A). Novel ionizable lipid-based LNPs with same mRNA (FIG. 6B) show proportional increase in luminescence with increased dose. Compositions are according to Table 1.
FIGs. 7A-7B. Viability of cells in the in vitro expression of Firefly Luciferase in Huh7 cells, expressed as relative fluorescence units (RFU) using TiterGlo kit. IVIC3 LNPs with 45mol% IVIC3 with AAP and without modifiers (FIG. 7A). Novel ionizable lipid-based LNPs with same mRNA (FIG. 7B) show proportional increase in luminescence with increased dose. Compositions are according to Table 1.
FIG. 8. Fluorescence emission of LNP formulations in Table 2. Irradiation of the pure Compound (89) in ethanol at 480 nm is associated with emission band at around 560 nm. Irradiation of the LNP formulations at 480 nm is associated with negligible emission at around 560 nm, showing fluorescence quenching. FIG. 9. Size and polydispersity by Dynamic Light Scattering (DLS) of LNPs with and without fifth component Compound (89) and astaxanthin. Left side: Z-average size of LNPs. Right side: Polydispersity of LNPs with compositions and structures of ionizable lipids per Table 2. Composition labels are according to Table 1.
FIG. 10. Expression of Luciferase from modified mRNA by in vitro cell transfection with LNPs with varying amounts of Compound (89) additive in HEK293T cells (left) and Huh7 cells (right). Compositions are according to Table 1.
FIG. 11. Viability of cells tested with mRNA LNPs in vitro cell transfection, with formulations in Table 2 having varying amounts of Compound (89) additive in HEK293T cells (left) and Huh7 cells (right). Compositions are according to Table 1.
FIGs. 12A-12B. Characterization of LNPs containing MC3 and ionizable lipids Compound 51 or Compound 53, with additional components as per Table 2. FIG. 12A: Average Z diameter; FIG. 12B: Polydispersity (PDI).
FIGs. 13A-13B. Expression of Luciferase from modified mRNA with LNPs containing Compound 51 or Compound 53 by in vitro cell transfection of HEK293T cells (FIG. 13A) and Huh7 cells (FIG. 13B). LP-01 is the chemical named 3-((4,4-bis(octyloxy)butanoyl)oxy)-2-((((3- (diethylamino)propoxy)carbonyl)oxy)methyl)propyl (9Z,12Z)-octadeca-9,12-dienoate.
FIG. 14. Expression of Luciferase from modified mRNA with LNPs (see Table 3) containing modified mRNA encoding Luciferase and ionizable lipid Compound 30 or Compound 52b y in vivo intravenous dosing in Balb/C mice.
Detailed Description of the Invention
Definitions
"Administration", "administering" and variants thereof refers to introducing a composition or agent of the present invention (e.g., an LNP comprising a nucleic acid payload) into a subject, organ, tissue or cells for therapeutic, pharmacokinetic, diagnostic purposes, or research purposes. "Administration" includes in vivo, in vitro, ex vivo and in utero administration. The introduction of a composition or agent into a subject is by any route of administration that is suitable for the specific composition or agent. Routes of administration include oral, pulmonary, intranasal, parenteral (intravenous, intramuscular, intraperitoneal, or subcutaneous), rectally, intravesical, intranodal, intralymphatical, intratumoral and topical administration.
The term "cationic lipid" refers to any lipid that is or can be positively charged. In some embodiments, the cationic lipid is an ionizable lipid, i.e., an ionizable cationic lipid, which is predominantly protonated and positively charged at a pH that exceeds about 2 units of the pKa value, known as the negative log(10) value of the acid dissociation constant (Ka) of the protonated, positively charged form of the lipid. Cationic and cationic ionizable lipids are highly water-insoluble, even when charged, due to the lipophilic nature of the overall molecule. Therefore, the thermodynamic pKa of an ionizable lipid is generally not measured but is estimated or calculated, and the measurement of pKa is instead done at the particle level by a TNS method (Heyes J et al., J Control Release 2005, pp. 276-287). A pKa measured in this way, on LNPs containing among other lipids the ionizable lipid, is termed "apparent pKa", "pKa(app)" or "LNP pKa" and is generally denoted as pKa'. The pKa' is about 2 to 4 units lower than the pKa, based on estimation methodology proposed for correlating the pKa and pKa' (e.g. Carrasco et al., Comm Biol, 2021, vol 4:956). For the purposes of relating functional aspects of ionizable lipids and their LNPs we use pKa' in the following sections.
One function of ionizable lipids is to facilitate endosomal release, by changing LNP structure via interaction with the endosomal membrane of LNPs within endosomes. This occurs about pH 5.5 and below. In some embodiments, ionizable cationic lipid in an LNP is predominantly neutral at physiological pH (pH 7.4) and is ionized and positively charged in the same LNP at a pH that is below physiological pH (e.g., between pH 4-5, preferably about pH 4.5). In some embodiments, the ionizable cationic lipid is about neutral at physiological pH (7.4 pH) and becomes protonated when introduced to an environment where the pH is about 4.5 or the pH inside of endosomes. It will be understood by one of ordinary skill in the art that the addition or removal of protons as a function of pH is a fast equilibrium process, and that the reference to a charged or a neutral lipid refers to the nature of the predominant species and does not require that all of the lipid be present in the charged or neutral form, whether in isolation or in an LNP. Generally, ionizable cationic lipids have a pKa' in the range of about 4 to about 7 as measured in the particle context (TNS assay). In some embodiments, ionizable lipid may include "cleavable lipid" or "SS-cleavable lipid". Corresponding quaternary lipids of all ionizable cationic lipids described herein (i.e., where a nitrogen atom is protonated and has four substituents) are contemplated within the scope of this disclosure. In some embodiments, an LNP comprising an ionizable lipid has a an pKa' (TNS) between 4-5, 5-6 between 4-5, 5-6, or 6-7. A "non-cationic lipid" is an anionic or neutral lipid.
The term "anionic lipid" refers to any lipid that is negatively charged at pH 7.4 (physiological pH). These lipids include, but are not limited to, phosphatidylglycerols, cardiolipins, diacylphosphatidylserines, diacylphosphatidic acids, N-dodecanoyl phosphatidylethanolamines, N-succinyl phosphatidylethanolamines, N- glutarylphosphatidylethanolamines, lysylphosphatidylglycerols, palmitoyloleyolphosphatidylglycerol (POPG), and other anionic modifying groups joined to neutral lipids.
The term "hydrophobic lipid" refers to compounds having a polar group(s) that include, but are not limited to, long-chain saturated and unsaturated aliphatic hydrocarbon groups and such groups optionally substituted by one or more aromatic, cycloaliphatic, or heterocyclic group(s). Suitable examples include, but are not limited to, diacylglycerol, dialkylglycerol, N,N-dialkylamino, l,2-diacyloxy-3-aminopropane, and l,2-dialkyl-3-aminopropane. The term "hydroxyalkyl" means a linear monovalent hydrocarbon radical or a branched monovalent hydrocarbon radical, substituted with one or two hydroxy groups, provided that if two hydroxy groups are present, they are not on the same carbon atom. Representative examples include, but are not limited to, hydroxymethyl, 2-hydroxyethyl, 2-hydroxypropyl, 3-hydroxypropyl, l-(hydroxymethyl)-2- methylpropyl, 2-hydroxybutyl, 3-hydroxybutyl, 4- hydroxybutyl, 2,3-dihydroxypropyl, 1- (hydroxymethyl)-2-hydroxyethyl, 2,3-dihydroxybutyl, 3,4-dihydroxybutyl and 2- (hydroxymethyl)-3-hydroxypropyl, preferably 2- hydroxyethyl, 2,3-dihydroxypropyl, and 1- (hydroxymethyl)-2-hydroxyethyl. A C1-C6 hydroxyalkyl means a linear monovalent hydrocarbon radical of one to six carbon atoms or a branched monovalent hydrocarbon radical of three to six carbons substituted with either one hydroxy group or two hydroxy groups on different carbon atoms. Where the alkyl is substituted with an alkene, the group is a "hydroxyalkenyl" group.
The term "linked" encompasses chemical conjugation, adsorption (physisorption and/or chemisorption). The types of bonds encompassed by the term "linked" are covalent interactions and noncovalent interactions (e.g., hydrogen bonds, ionic bonds, van der Waal bonds, and hydrophobic bonds).
The term "neutral lipid" refers to a lipid that exist either in an uncharged or neutral zwitterionic form in a pH range comprising pH 4 - 7.4. At physiological pH, such lipids include, for example, diacylphosphatidylcholine, diacylphosphatidylethanolamine, ceramide, sphingomyelin, cephalin, cholesterol, cerebrosides, and diacylglycerols.
The term "non-fusogenic cationic lipid" is meant a cationic lipid that can condense and/or encapsulate the nucleic acid cargo, such as mRNA, but does not have, or negligible, fusogenic activity with a cell plasma membrane.
The term "cleavable lipid" or "SS-cleavable lipid" refers to a lipid comprising a disulfide bond cleavable unit. Cleavable lipids may include cleavable disulfide bond ("SS") containing lipid- like materials that comprise a pH-sensitive tertiary amine and self-degradable phenyl ester. For example, a SS-cleavable lipid can be an SS-OP lipid (COATSOME® SS-OP), an SS-M lipid (COATSOME® SS-M), an SS-E lipid (COATSOME® SS-E), an SS-EC lipid (COATSOME® SS-EC), an SS-LC lipid (COATSOME® SS-LC), an SS-OC lipid (COATSOME® SS-OC), and an SS-PalmE lipid (see, for example, Formulae l-IV), or a lipid described in Togashi R, et al. J Control Release 2018 Jun 10;279:262-270, US Patent 9,708,628, or US Patent 10,385,030.
The term "non-cationic lipid" refers to a neutral lipid or anionic lipid.
The term "nucleic acid," refers to a polymer containing at least two nucleotides (i.e., deoxyribonucleotides or ribonucleotides) in either single- or double-stranded form and includes DNA, RNA, DNA-RNA hybrids, as well as analogs and modified forms thereof. DNA may be in the form of, e.g., antisense molecules, plasmid DNA, DNA-DNA duplexes, precondensed DNA, PCR products, vectors (PI, PAC, BAC, YAC, artificial chromosomes), expression cassettes, chimeric sequences, chromosomal DNA, or derivatives and combinations of these groups. DNA may be in the form of minicircle, plasmid, bacmid, minigene, ministring DNA (linear covalently closed DNA vector), closed-ended linear duplex DNA (CELiD or ceDNA), doggybone™ DNA, dumbbell shaped DNA, minimalistic immunological-defined gene expression (MIDGE)-vector, viral vector or nonviral vectors. RNA may be in the form of small interfering RNA (siRNA), dicer-substrate dsRNA, small hairpin RNA (shRNA), asymmetrical interfering RNA (aiRNA), microRNA (miRNA), mRNA, rRNA, tRNA, gRNA, viral RNA (vRNA), and combinations thereof. Nucleic acids include nucleic acids containing known nucleotide analogs or modified backbone residues or linkages, which are synthetic, naturally occurring, and non-naturally occurring, and which have similar binding properties as the reference nucleic acid. Examples of such analogs and/or modified residues include, without limitation, phosphorothioates, phosphorodiamidate morpholino oligomer (morpholino), phosphoramidates, methyl phosphonates, chiral-methyl phosphonates, 2'-O-methyl ribonucleotides, locked nucleic acid (LNA™), and peptide nucleic acids (PNAs). Unless specifically limited, the term encompasses nucleic acids containing known analogues of natural nucleotides that have similar binding properties as the reference nucleic acid. Unless otherwise indicated, a particular nucleic acid sequence also implicitly encompasses conservatively modified variants thereof (e.g., degenerate codon substitutions), alleles, orthologs, SNPs, and complementary sequences as well as the sequence explicitly indicated.
The phrases "nucleic acid therapeutic", "therapeutic nucleic acid" and "TNA" are used interchangeably and refer to any modality of therapeutic using a nucleic acid as an active pharmaceutical ingredient of therapeutic agent to treat a disease or disorder.
The term "pharmaceutically acceptable carrier" or "pharmaceutically acceptable excipient "includes any of the standard pharmaceutical carriers/excipients, such as a phosphate buffered saline solution, TRIS/sucrose buffer, water, emulsions such as an oil/water or water/oil, and various types of wetting agents. The term also encompasses any of the agents approved by a regulatory agency of the US Federal government or listed in the US Pharmacopeia for use in animals, including humans, as well as any carrier or diluent that does not cause significant irritation to a subject and does not abrogate the biological activity and properties of the administered compound.
The term "subject" or "patient" refers to a human or animal, to whom treatment, including prophylactic treatment, with the therapeutic nucleic acid according to the present disclosure, is provided. Animals include mammals, birds and fish. Preferably, the animal is a mammal, e.g., primate, rodent, lagomorph, companion animal or livestock. Primates include humans, chimpanzees, cynomolgus monkeys, spider monkeys, and macaques, e.g., rhesus macaque. Rodents include mice, rats, and hamsters. Livestock include cows, horses, pigs, sheep and goats. Preferably, the subject is a human. A human subject can be of any age, gender, race or ethnic group. The terms "therapeutic amount", "therapeutically effective amount", "effective amount" "amount effective", or "pharmaceutically effective amount" of an active agent (e.g., a TNA described herein) are used interchangeably to refer to an amount that is sufficient to produce a desired effect, e.g., expression or inhibition of a target gene/sequence or disease modification. When the TNA is an mRNA, then an "effective amount" may be an amount sufficient to produce an increase in expression of a target polypeptide in comparison to the normal expression level, if any, detected in the absence of the messenger RNA. Suitable assays for measuring expression of a target gene or target sequence include, examination of protein or RNA levels using techniques known to those of skill in the art such as dot blots, northern blots, in situ hybridization, ELISA, immunoprecipitation, enzyme function, fluorescence or luminescence of suitable reporter proteins, as well as phenotypic assays. The terms include prophylactic or preventative amounts of an active agent is an amount sufficient to eliminate or reduce the risk, lessen the severity, or delay the onset of a disease, disorder or condition.
The terms "dose" and "dosage" is the amount of an active agent administered at any given time. Dosage levels are based on a variety of factors, including the specific disease or disorder, the age, weight, sex, medical condition of the patient, the severity of the condition, the route of administration, and the particular active agent or agents employed. The dosage regimen can be determined routinely by a physician using standard methods.
As used herein the term "therapeutic effect" refers to a consequence of treatment, the results of which are judged to be desirable, safely achieved, and beneficial. A therapeutic effect can include, directly or indirectly, the arrest, reduction, or elimination of a disease manifestation or progression.
The terms "treat," "treating," and/or "treatment" may be therapeutic, prophylactic or palliative, and include abrogating, inhibiting, delaying, slowing or reversing the progression of a disease, disorder or condition; ameliorating clinical symptoms of a disease, disorder or condition; or preventing or reducing the appearance of clinical symptoms of a disease, disorder or condition.
The term "alkyl" refers to saturated monovalent hydrocarbon radical. Alkyls may be linear or branched and may be optionally substituted. Examples include C1-20 alkyl, Cus alkyl, Cue alkyl, Ci-14 alkyl, C1-12 alkyl, C1-10 alkyl, C1-9 alkyl, C1-8 alkyl, C1-7 alkyl, C1-6 alkyl, C1-5 alkyl, C1-4 alkyl, and C1-C 3 alkyl. Examples further include methyl, ethyl, 1-propyl, 2-propyl, 1-butyl, 2- methyl-l-propyl, 2-butyl, 2-methyl-2-propyl, 1-pentyl, 2-pentyl, 3-pentyl, 2-methyl-2-butyl, 3-methyl-2-butyl, 3-methyl-l-butyl, 2-methyl-l-butyl, 1-hexyl, 2-hexyl, 3-hexyl, 2-methyl-2- pentyl, 3-methyl-2-pentyl, 4-methyl-2-pentyl, 3-methyl-3-pentyl, 2-methyl-3-pentyl, 2,3- dimethyl-2-butyl, 3,3-dimethyl-2-butyl, 1-heptyl, 1-octyl, and the like.
The term "alkyl ester" refers to -R50-C(=O)O-R51 or -R50-OC(=O)-R51, wherein R50 is alkylene and R51 is alkyl. The term "alkenyl ester" to -R52-C(=O)O-R53 or -R52-OC(=O)-R53, wherein R52 is alkylene or alkenylene and R53 is alkyl or alkenyl, with the proviso that R52 and/or R53 contain at least one carbon-carbon double bond.
The term "alkyl carbonate" refers to -R50-O-C(=O)O-R51 or -R50-OC(=O)O-R51, wherein R50 is alkylene and R51 is alkyl.
The term "alkenyl carbonate" to -R52-O-C(=O)O-R53 or -R52-OC(=O)O-R53, wherein R52 is alkylene or alkenylene and R53 is alkyl or alkenyl, with the proviso that R52 and/or R53 contain at least one carbon-carbon double bond.
The term "alkylene" refers to a saturated bivalent hydrocarbon radical. Alkylenes may be linear or branched and may be optionally substituted. Examples include C1-20alkylene groups, such as C1-20 alkylene, C1-18 alkylene, Cue alkylene, C1-14 alkylene, C1-12 alkylene, C1-10 alkylene, C1-9 alkylene, C1-8 alkylene, C1-7 alkylene, C1-6 alkylene, C1-5 alkylene, C1-4 alkylene, and C1-C 3 alkylene.
The term "alkene" refers to a group consisting of at least two carbon atoms and at least one carbon-carbon double bond, and an "alkyne" moiety refers to a group consisting of at least two carbon atoms and at least one carbon-carbon triple bond. The alkyl moiety, whether saturated or unsaturated, may be branched, straight chain, or cyclic.
The term "alkenyl" refers to a hydrocarbon radical with one or more carbon-carbon double bonds, including radicals having "cis" and "trans" orientations, or by an alternative nomenclature, "E" and "Z" orientations. Alkenyls may be linear or branched and may be optionally substituted. Examples include C2-20 alkenyl groups, such as, C2-20 alkenyl, C2-18 alkenyl, C2-16 alkenyl, C2-14 alkenyl, C2-12 alkenyl, C2-10 alkenyl, C2-9 alkenyl, C2-8 alkenyl, C2-7 alkenyl, C2-6 alkenyl, C2-5 alkenyl, C2-4 alkenyl, and C2-3 alkenyl.
"Alkenylene" refers to an aliphatic bivalent hydrocarbon radical with one or more carboncarbon double bonds, including radicals having "cis" and "trans" orientations, or by an alternative nomenclature, "Z" and "E" orientations, respectively. Alkenylenes may be linear or branched and may be optionally substituted. Examples include C2-20 alkenylene groups, such as, C2-20 alkenylene, C2-18 alkenylene, C2-16 alkenylene, C2-14 alkenylene, C2-12 alkenylene, C2-10 alkenylene, C2-9 alkenylene, C2-8 alkenylene, C2-7 alkenylene, C2-6 alkenylene, C2-5 alkenylene, C2-4 alkenylene, and C2-3 alkenylene.
"Alkynyl" refers to a hydrocarbon monovalent radical with one or more carbon-carbon triple bonds. "Alkynylene" refers to a hydrocarbon bivalent radical with one or more carboncarbon triple bonds (i.e., a group derived from an alkyne with two attachment points).
"Aryl" refers to a monovalent aromatic group derived from an arene by removal of a hydrogen atom from a ring carbon atom. As used herein, aryl includes substituted or unsubstituted single-ring aromatic groups and polycyclic ring systems having two or more cyclic rings in which two or more carbons are common to two adjoining rings, wherein at least one of the rings is aromatic.
"Arylyene" or "arenediyl" refers to a bivalent aromatic group derived from an arene by removal of a hydrogen atom from two different ring carbon atoms, forming a group with two attachment points. In multi-ring systems, the two hydrogens may be removed from the same ring or different rings.
Cyclic groups of the present invention include monocyclic and bicyclic groups (bridged or fused).
"Carbocycle" and "carbocyclic" refers to a C3-C20 monocyclic or polycyclic (e.g., bicyclic or tricyclic), saturated, partially saturated or unsaturated ring(s), in which all the atoms composing the ring are carbon atoms. Ring moieties include fused, spirocyclic and bridged bicyclic rings. Saturated carbocyclic rings include, for example, "cycloalkyl" rings, e.g., cyclopropyl, cyclobutyl, etc. Carbocyclyl is a monovalent radical of a carbocycle, i.e., a carbocycle functional group with one attachment point. Carbocyclediyl or carbocyclene is a bivalent radical of a carbocycle, i.e., a carbocycle functional group with two attachment points.
"Cycloalkyl" refers to a monovalent saturated carbocyclic ring radical. Cycloalkyls may be optionally substituted. Cycloalkyl groups include groups having from 3 to 18 ring atoms. Cycloalkyl groups include, but are not limited to: cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cycloheptyl, cyclooctyl, cyclononyl, cyclodecyl and norbornyl.
"Cycloalkylene" as used herein refers to a bivalent, saturated, 3-18-membered carbocyclic ring radical with two attachment points. Specific monocyclic cycloalkylene groups include, but are not limited to, cyclopropylene, cyclobutylene, cyclopentylene, cyclohexylene, cycloheptylene, cyclooctylene, cyclononylene, cyclodecylene, cycloundecylene, cyclododecylene, and the like. In one embodiment, the cycloalkylene is cyclopropylene.
The terms "include", "includes" and "including" are open-ended and not limited to examples provided.
"Halogen" refers to F, Cl, Br or I.
"Heterocycle" "heterocyclic" and "heterocyclic ring" are used interchangeably and refer to 5-20 aromatic or 3-20 aliphatic cyclic group having at least one ring heteroatom and at least one ring carbon atom. In one embodiment, the heteroatom is oxygen, sulfur, or nitrogen. A heterocycle containing more than one heteroatom may contain different heteroatoms. "Heterocyclyl" is a monovalent radical of a heterocycle and have one attachment point. An aromatic heterocyclyl is also referred to a "heteroaryl". Heterocyclene or heterocyclediyl refers to a bivalent heterocycle group with two attachment points. Heterocyclyl and heterocyclene moieties include both monocyclic and multicyclic (e.g., bicyclic or tricyclic) ring moieties. Ring moieties include fused, spirocyclic and bridged bicyclic rings and may comprise one or more heteroatoms in one or more of the rings. Either ring of a bicyclic heterocycle may be saturated, partially unsaturated or aromatic. The heterocycle may be attached to the rest of the molecule via a ring carbon atom or a ring nitrogen atom. Examples of heterocyclic functional groups include aziridinyl, diaziridinyl, thiaziridinyl, azetidinyl, diazetidinyl, triazetidinyl, thiadiazetidinyl, thiazetidinyl, pyrrolidinyl, pyrazolidinyl, imidazolinyl, isothiazolidinyl, thiazolidinyl, piperidinyl, piperazinyl, hexahydropyrimidinyl, azepanyl, and azocanyl.
"Heteroarene" is an aromatic compound derived from an arene by replacement of one or more methine (-C=) and/or vinylene (-CH=CH-) groups by trivalent or divalent heteroatoms, respectively.
"Heteroaryl" refers to a monovalent group with one attachment point derived from heteroarene by removal of a hydrogen atom from a ring atom.
"Heteroarylene" or "heteroarenediyl" refers to a bivalent group derived from a heteroarene by removal of a hydrogen atom from two different ring atoms, forming a group with two attachment points. The hydrogens may independently be removed from either a carbon or nitrogen atom (as available). In multi-ring systems, the two hydrogens may be removed from the same ring or different rings. Heteroaryls of the present invention include 5-18- membered aromatic radicals (e.g., C5-C13 heteroaryl), preferably 5-10-membered aromatic groups, that includes one or more ring heteroatoms selected from nitrogen, oxygen and sulfur, and which may be a monocyclic or polycyclic (e.g., a bicyclic, tricyclic or tetracyclic ring system). A polycyclic heteroaryl group may be fused or non-fused. The heteroatom(s) in the heteroaryl radical are optionally oxidized. One or more nitrogen atoms, if present, are optionally quaternized.
"Pharmaceutically acceptable salt" as used herein refers to pharmaceutically acceptable organic or inorganic salts of an ionizable lipid of the disclosure.
"Spirocycle" and "spiroheterocycle" refer is a 5- to 20-membered bicyclic ring system functional group, including spi ro [cycloa Ikyl] and spiro[cycloalkenyl] with both rings connected through a carbon single atom. A spirocycle/spiroheterocycle can be fully saturated or can be partially unsaturated. The rings can be different in size and nature, or identical in size and nature. Examples include spiropentanyl, spriohexanyl, spiroheptanyl, spirooctanyl, spirononanyl, or spirodecanyl. One or both of the rings in a spiro(hetero)cyclcle can be fused to another ring carbocyclic, heterocyclic, aromatic, or heteroaromatic ring. A (C5-C14) spirocycloalkyl, e.g., is a spirocycle containing between 5 and 14 carbon atoms. "Spiroheterocycloalkyl" or "spiroheterocyclyl" is understood to mean a spirocyclyl as defined above wherein at least one of the rings is a heterocycle, i.e., a ring containing a heteroatom. In one embodiment, the heteroatom is oxygen, sulfur, or nitrogen. "Amine" or "amino" as used herein interchangeably refers to a functional group that contains a basic nitrogen atom with a lone pair. The term "pharmaceutically acceptable salt" as used herein refers to pharmaceutically acceptable organic or inorganic salts of an ionizable lipid of the disclosure.
Ranges set forth herein are inclusive of the end values.
Hydrogen atoms connected to carbons may be substituted with deuterium (2H) atoms.
Where two or more variables within a chain of variables are absent and designated as "a bond", the absent variables are, together, considered as one bond connecting the present adjacent variables. As an example, for the group -R62-R123-R63-R73-R74-R66, when R73 and R74 are each "a bond", then the groups is read as -R62-R123-R63-R66.
Double bonds may be indicated as "=" or implicit based on valency rules.
All patents and other publications; including literature references, issued patents, published patent applications, and co-pending patent applications; cited throughout this application are expressly incorporated herein by reference in their entireties.
Lipid
Lipids used in delivery of nucleic acids, e.g., mRNA include a head group and optional linker moiety and a hydrocarbon containing lipid tail. The head group is typically an amine- containing group when used as a typical ionizable lipid. An ionizable cationic lipid comprise a hydrophilic ionizable amine headgroup. Some cationic lipids have a permanently charged quaternary ammonium group as their head. Other ionizable cationic lipids have a pKa- dependent head group. This headgroup is responsible for condensing mRNA or other nucleic acids. The headgroup provides positive charge to interact with the negatively charged nucleic acids. Connecting the headgroup to the is an optional linker moiety which is preferably biodegradable for safe clearance after nucleic acid delivery. The headgroup is essential for self-assembly and phospholipid membrane fusion within the lipid nanoparticle. The headgroup-linker- is connected to a hydrophobic tail group. Ionizable cationic lipids typically have hydrophobic hydrocarbon chains as or as part(s) of the tail group. These chains promote self-assembly of the lipid nanoparticles. They also facilitate membrane fusion during cellular uptake. The hydrophobic tails further contribute to the overall stability of the lipid nanoparticle.
In one aspect, the invention provides for a lipid as disclosed herein. The lipid may be used, e.g., in a lipid vesicle including liposomes, in a lipid film, and lipid nanoparticles (LNPs). The lipid may be a cationic lipid. The lipid may further be a ionizable cationic lipid.
Lipid nanoparticles
In another aspect, the invention provides for a lipid nanoparticle (LNP) comprising a lipid of the present invention. In some embodiments, the present disclosure provides for a LNP composition comprising a plurality of LNPs and at least one pharmaceutically acceptable carrier, diluent or excipient. The LNPs of the invention are lipid vesicles with a diameter that is typically in the range of 25-1000 nm. LNPs of the invention comprise multiple lipids, at least one of which is positively charged (cationic) at low pH (enabling RNA complexation and endosomal escape).
The cationic lipid is preferably an ionizable cationic lipid that is substantially in the neutral form in an LNP at physiological pH. In addition to a (ionizable) cationic lipid, the LNP may further comprises a non-cationic, structural/helper lipid, a sterol (to provide membrane fluidity) and a polymer conjugated lipid (to prevent aggregation).
LNPs of the invention may comprise a targeting moiety, such as a protein or peptide or a cluster of peptides and/or small molecule targeting ligands.
LNPs of the invention may comprise a labelling moiety, such as a fluorophore small molecule (BODIPY and the like) for tracking purposes with confocal microscopy and fluorescence imaging methods.
LNPs of the invention may further comprise a diagnostic or therapeutic agent and be used to deliver the agent, such as TNA, to a cell, tissue or organ. In some embodiments, the LNP comprises a therapeutic agent such as a TNA (e.g., mRNA), protein, peptide or other sensitive cargo encapsulated or contained in the lipid portion of the particle, thereby protecting it from enzymatic degradation, excretion or immunogenic or other reaction.
LNP Components
According to some embodiments, the lipid particles of the disclosure have a mean diameter of: from about 25 nm to about 125 nm, about 50 nm to about 100 nm, about 100 nm to about 200 nm, about 200 nm to about 300 nm, from about 300 nm to about 400 nm, from about 400 nm to about 500 nm, from about 500 nm to about 750 nm, from about 750 nm to about 1000 nm, from about 50 nm to about 500 nm, from about 25 nm to about 1000 nm, less than about 1000 nm, less than about 750 nm, less than about 500 nm, less than about 250 nm, less than about 150 nm, less than about 100 nm, less than about 75 nm, less than about 50 nm, less than about 40 nm, less than about 30 nm, less than about 25 nm, or less than about 20 nm. Lipid particle (e.g., lipid nanoparticle) size can be determined, e.g., by quasi-elastic light scattering using a Malvern Zetasizer Nano ZS (Malvern, UK) or Wyatt Dynapro DLS (Wyatt Technologies, Santa Barbara CA).
In some embodiments, the LNPs may be relatively homogenous. A polydispersity index may be used to indicate the homogeneity of the LNPs. A small, for example less than 0.3 or less than 0.2, polydispersity index generally indicates a narrow particle size distribution. A composition of the LNPs described herein may have a polydispersity index from about 0 to about 0.25 or to about 0.30, such as 0.01, 0.02, 0.03, 0.04, 0.05, 0.06, 0.07, 0.08, 0.09, 0.10, 0.11, 0.12, 0.13, 0.14, 0.15, 0.16, 0.17, 0.18, 0.19, 0.20, 0.21, 0.22, 0.23, 0.24, 0.25, 0.26, 0.27, 0.28, 0.29 or 0.30. In some embodiments, the polydispersity index of the LNP composition may be from about 0 to about 0.30 or 0.05 to 0.20. In one aspect, the LNP comprises: a (ionizable) cationic lipid, a sterol or a derivative thereof, a non-cationic lipid, and a polymer-conjugated lipid. In one embodiment, the LNP comprises more than one (ionizable) cationic lipid, more than one sterol or a derivative thereof, more than one non-cationic lipid, and/or more than one polymer-conjugated lipid. In another embodiment, the lipid particle (e.g., lipid nanoparticle) comprises a cationic lipid, a noncationic phospholipid, cholesterol and a PEGylated lipid (polymer conjugated lipid). In a further embodiment, the cationic lipid, non-cationic phospholipid, cholesterol and a PEGylated lipid are present in a molar ratio of about 50:7:40:3. 50: 10:38.5: 1.5, respectively. In one embodiment, of the total lipid content, the LNP comprises about: 40-50 mol%, 45-50 mol%, 50-55 mol%, 45-46 mol%, 46-47 mol%, 47-48 mol%, 48-49 mol%, or 49- 50 mol%, 45 mol%, 45.5 mol%, 46 mol%, 46.5 mol%, 47 mol%, 47.5 mol%, 48 mol%, 48.5 mol%, 49 mol%, or 49.5 mol% (ionizable) cationic lipid. In one embodiment of the total lipid content, the LNP comprises about: 5-25 mol%, 5-15 mol%, 10-12 mol%, 5-6 mol%, 6-7 mol%, 7-8 mol%, 8-9 mol%, 9-10 mol%, 10-11 mol%, 11-12 mol%, 12-13 mol%, 13-14 mol%, or 14- 15 mol% non-cationic lipid. In one embodiment of the total lipid content, the LNP comprises about: 25-55 mol%, 30-45 mol%, 35-40 mol%, 30-31 mol%, 31-32 mol%, 32-33 mol%, 33-34 mol%, 35-35 mol%, 35-36 mol%, 36-37 mol%, 38-38 mol%, 38-39 mol%, or 39-40 mol% sterol. In one embodiment of the total lipid content, the LNP comprises about: 0.5-15 mol%, 1-5 mol%, 1-3 mol%, 1.5-2.5 mol%, 1-2 mol%, 2-3 mol%, 3-4 mol%, or 4-5 mol% polymer conjugated lipid, e.g., PEGylated lipid. In one embodiment the lipid nanoparticle comprises a total lipid content that is 20-60 mol% (ionizable) cationic lipid, 5-25 mol% non-cationic lipid, 25-55 mol% sterol, and 0.5-15 mol% polymer conjugated lipid, e.g., PEGylated lipid. In one embodiment, the lipid nanoparticle comprises a total lipid content that is 40-50 mol% (ionizable) cationic lipid, 5-15 mol% non-cationic lipid, 30-45 mol% sterol, and 1-5 mol% polymer conjugated lipid, e.g., PEGylated lipid. In one embodiment, the lipid nanoparticle comprises a total lipid content that is 45-50 mol% (ionizable) cationic lipid, 10-12 mol% noncationic lipid, 35-40 mol% sterol, and 1-3 mol% polymer conjugated lipid, e.g., PEGylated lipid. In one embodiment, the lipid nanoparticle comprises a total lipid content that is 45-50 mol% (ionizable) cationic lipid, 10-12 mol% non-cationic lipid, 35-40 mol% sterol, and 1.5-2.5 mol% polymer conjugated lipid, e.g., PEGylated lipid conjugate.
Cationic and ionizable lipids
In some aspects, the lipid nanoparticle of the present invention comprises a cationic lipid. In some aspects, the cationic lipid is an ionizable cationic lipid. The ionizable cationic lipid is positively charged at low pH, which facilitates association with the negatively charged nucleic acid. The ionizable cationic lipid is neutral at physiological pH (pH 7.4). The ability of these lipids to ionize at low pH aids in endosomal escape of the nucleic acid into the cytoplasm.
In one aspect, the invention provides for a pharmaceutical composition comprising a lipid nanoparticle, wherein the lipid nanoparticle comprises a cationic lipid. In some embodiments, the cationic lipid is an ionizable cationic lipid. Such LNPs can be used to deliver a diagnostic or therapeutic agent to a target cell, tissue or organ in a subject.
Exemplary ionizable cationic lipids are described in International PCT patent publications WO2022/246571, W02018/011633, WO2017/117528, WO2017/099823, WO2017/075531, WO2017/049245, W02017/004143, W02016/081029, WO2015/199952, WO2015/095346, W02015/095340, W02015/074085, WO2015/061467, WO2013/148541, WO2013/126803, WO2013/116126, W02013/089151, WO2013/086373, WO2013/086354, WO2013/086322, WO2013/049328, WO2013/033563, W02013/016058, W02013/006825, W02012/162210, WO2012/099755, WO2012/054365, WO2012/044638, W02012/040184, W02012/031043, W02012/016184, W02012/000104, W02011/153120, W02011/141705, W02011/141704, W02011/090965, W02011/071860, W02011/066651, W02011/038160, W02011/022460, W02011/000107, W02011/000106, W02010/144740 , W02010/129709, W02010/088537, W02010/054406 , W02010/054405, W02010/054401, W02010/054384, W02010/048536, W02010/042877, W02009/132131, W02009/127060, W02009/086558, W02008/042973, W02006/069782, W02006/007712, WO2005/121348, and W02005/120152; and US patent publications US2018/0028664, US2018/0005363, US2017/0119904, US2017/0210697, US2016/0376224, US2016/0317458, US2016/0311759, US2016/0151284, US2015/0376115, US2015/0239926, US2015/0203446, US2015/0141678, US2015/0140070, US2015/0064242, US2015/0057373, US2014/0308304, US2014/0255472, US2014/0200257, US2014/0141070, US2014/0039032, US2013/0338210, US2013/0323269, US2013/0303587, US2013/0274523, US2013/0274504, US2013/0274504, US2013/0202684, US2013/0195920, US2013/0178541, US2013/0123338, US2013/0116307, US2013/0090372, US2013/0065939, US2012/0202871, US2012/0149894, US2012/0128760, US2012/0101148, US2012/0058144, US2012/0027796, US2010/0324120, US2010/0130588, US2010/0062967, US201/0256175, US201/0117125, US201/0076335, US2009/0023673, US2006/0083780, US2006/0051405, US2006/0008910, and US2003/0022649, each of which are incorporated by reference in their entireties.
Further examples include 3-(didodecylamino)-Nl,Nl,4-tridodecyl-l-piperazineethanamine (KL1O), Nl-[2-(didodecylamino)ethyl]-Nl,N4,N4-tridodecyl-l,4- piperazinediethanamine (KL22), 14,25-ditridecyl-15, 18,21 ,24-tetraaza-octatriacontane (KL25), 1.2-dilinoleyloxy-N,N- dimethylaminopropane (DLin-DMA), 2,2-dilinoleyl-4-dimethylaminomethyl-[l,3]-dioxolane (DLin-K-DMA), (6Z,9Z,28Z,31Z)-heptatriacont-6,9,28,31-tetraene-19-yl 4- (dimethylamino)butanoate (DLin-MC3-DMA), 2.2-dilinoleyl-4-(2-dimethylaminoethyl)-[l,3]- dioxolane (DLin-KC2-DMA), l,2-dioleyloxy-N,N-dimethylaminopropane (DODMA), 2-({8- [(3P)-cholest-5-en-3-yloxy]octyl}oxy)-N,N-dimethyl-3-[(9Z,12Z)- octadeca-9,12-dien-l- yloxy]propan-l -amine (Octyl-CLinDMA), (2R)-2-({8-[(3P)-cholest-5-en-3-yloxy]octyl}oxy)-N,N- dimethyl-3-[(9Z,12Z)-octadeca-9,12-dien-l-yloxy]propan-l-amine (Octyl-CLinDMA (2R)), (2S)-2-({8-[(3P)-cholest-5-en-3-yloxy]octyl}oxy)-N,N-dimethyl-3-[(9Z,12Z)-octadeca-9,12- dien-l-yloxy]propan-l-amine (Octyl-CLinDMA (2S)), ((4-hydroxybutyl)azanediyl)bis(hexane- 6,l-diyl)bis(2-hexyldecanoate)), 3-((4,4-bis(octyloxy)butanoyl)oxy)-2-((((3- (diethylamino)propoxy)carbonyl)oxy)methyl)propyl (9Z,12Z)-octadeca-9,12-dienoate, and 8- [(2-hydroxyethyl)[6-oxo-6-(undecyloxy)hexyl]amino]-octanoic acid, 1-octylnonyl ester.
In one aspect, the ionizable cationic lipid has a structural formula selected from the group consisting of: Compound (1):
Figure imgf000017_0001
(2),
Compound (3):
Figure imgf000017_0002
(4); or a pharmaceutically acceptable salt or co-crystal thereof, wherein: R1 for each occurrence is independently selected from: -H, optionally substituted C1-C6 alkyl, -[CFhJi-eOH, optionally substituted C2-C6 alkenyl, -C(=O)CH2N(R72)2, -P(O)(OR17)(OR18), - P(O)(OR17)(NR19), or -P(O)(NR19)(NR20);
R3 is selected from: -H, optionally substituted C1-C6 alkyl, -[CH2]i-eOH, optionally substituted C2-C6 alkenyl, -P(O)(OR17)(OR18), -P(O)(OR17)(NR19), -P(O)(NR19)(NR20) or R4;
R2 is selected from: is absent (i.e., a lone pair on N), -H or optionally substituted C1-C6 alkyl; provided that when R2 is -H or optionally substituted C1-C6 alkyl, the nitrogen atom, to which R1, R2, and R3 are all bonded to, is protonated;
R4 for each occurrence is independently selected from:
Figure imgf000018_0001
R55 for each occurrence, is independently selected from:
Figure imgf000018_0002
Figure imgf000019_0001
A1, A3, A4, A5, A6, A7, A8, A12 and A13, for each occurrence, are each independently selected
5 from: a bond, -O-, -C(=O)-, -C(=S)-, -OC(=O)-, -C(=O)O-, -OC(=O)O-, -OC(=S)-, -C(=S)O-, - OC(=S)O-,-S-C(=O)-, -C(=O)-S-, -S-S-, -S(OH)-, -S(=O)-, -S(=O)2-, -S(OH)2-, -C(=O)N(R14)-, - N(R14)C(=O)-, -N(R14)C(=O)N(R14)-, -O-C(=O)C(R14)2C(=O)O-, -C(=O)O-C(R14)2C(=O)O-, -O- C(=O)C(R14)2-O-C(=O)-, -O-C(R14)2C(=O)O-, -O-C(=O)C(R14)2C-O-, -O-C(R14)2-O-C(=O)-, -C(=O)- O-C(R14)2C-O-, -O-C(=O)-O-R41-O-C(=O)O-, -0-C(=0)-[CH2]O-4-C(=0)-0-, -P(OH)-, -P(OH)( R14)-, - P(=O)(OH)-, -P(O R14)2-, -P(O R14)- or -P(=O)(O R14)-, -CH(R14)-, -C(R14)2-, -CH(OH)-, -CH(O R14)-, -CH(NH2)-, -CH(NH[R14])-, -C(NH[OH])-, -C(NH[O R14])-, -N(R14)-, -S-, -C(=O)C(R14)2C(=O)-, - C(R14)2C(=O)-, -C(=O)C(R14)2-O-, -C(R14)2C(R14)2-, -C(R25)2-O-, -O-C(R14)2C(R14)2-, -O- C(R14)2C(=O)-, -C(=O)-O- R41-O-C(=O)-;
R41 for each occurrence, are each independently an optionally substituted group selected from: a bond, linear or branched Ci-Cs alkylene, Ci-Cs alkenylene or Ci-Cs alkynylene; C3-C10 heterocyclylene or C3-C10 carbocyclylene; or -R114-C3-C6-carbocyclylene-R115-, -R114-C3-Ce- heterocyclylene-R115-, -R114-C3-C6-cycloakylene-R115- -R114-CH=CH-CH2-CH=CH-R115- , -R114- CH=CH-CH2-cPr-R115-, -R114-cPr-CH2-CH=CH-R115-, -R114-cPr-CH2-cPr-R115-, -R114-CH=CH-CD2- CH=CH-R115-, -R114-CH=CH-CD2-cPr-R115-, -R114-cPr-CD2-CH=CH-R115- or -R114-cPr-CD2-cPr-R115- ; wherein, R114 and R115 are each independently selected from: a bond or linear or branched C1-C6 alkylene, C1-C6 alkenylene or C1-C6 alkynylene; wherein -cPr- is cyclopropylene (cyclopropane-1,2 diyl); each double bond or cPr has a cis configuration; and D denotes deuterium (2H);
A2 for each occurrence, is independently selected from: a bond, -O-, -OC(=O)-, -C(=O)O-, - OC(=O)O-, -S-C(=O)-, -C(=O)-S-, -S-S-, -C(=O)N(R14)-, -N(R14)C(=O)-, -N(R14)C(=O)N(R14)-, -O- C(=O)C(R14)2C(=O)O-, -C(=O)O-C(R14)2C(=O)O-, -O-C(=O)C(R14)2-O-C(=O)-, -O-C(R14)2C(=O)O-, - O-C(=O)C(R14)2C-O-, -O-C(R14)2-O-C(=O)-, -C(=O)-O-C(R14)2C-O- or -O-C(=O)-O-R41-O-C(=O)O-;
R6 for each occurrence, is independently selected from: a bond, -R58-N(R42)-R58-, -R58- CH(R42)-R58-, C1-C12 alkylene or C2-C12 alkenylene;
R7 for each occurrence, is independently selected from: a bond (i.e., when present A2 is bonded directly t
Figure imgf000020_0001
optionally substituted C1-C18 alkylene, or optionally substituted C2-C18 alkenylene;
R8 and R9, for each occurrence, are each independently selected from:
Figure imgf000020_0002
R42;
R12 for each occurrence, is independently selected from: -H, optionally substituted C1-C16 alkyl or optionally substituted C2-C16 alkenyl; R13 for each occurrence, is independently selected from: optionally substituted C1-C16 alkyl or optionally substituted C2-C16 alkenyl;
R14, for each occurrence, is independently selected from: -H, optionally substituted C1-C10 alkyl, optionally substituted C1-C10 alkenyl or optionally substituted C1-C10 alkynyl;
R15 for each occurrence, is independently selected from: a bond, -R41-N(R42)-R58-, -R41- CH(R42)-R58-, or C1-C6 alkylene;
R16 for each occurrence, is independently selected from: a bond or — [CH2]k-;
R17 and R18 are each independently selected from: -H, C1-C5 alkyl, an alkali metal cation, an alkaline earth metal cation, ammonium cation, methyl ammonium cation, or a pharmaceutically acceptable base;
R19 and R20, for each occurrence, are each independently selected from: -OH, formyl, acetyl, pivaloyl, -NH2, -NH(CH3), -NH(CH2CH3), -N(CH3)2, -NHC(=O)H, -NHC(=O)CH3, or C3-C5-alkyl;
R29 through R34, for each occurrence, are each independently selected from: -CH2-, -NH-, -S-, or -O-;
R35for each occurrence, is independently selected from: -OH or C1-C6 acyl; optionally substituted C1-C6 alkyl, C1-C6 alkyl ester, C1-C6 alkyl ether or C1-C6 alkyl carbonate;
R36 and R37, are each independently selected from: -H, -C(=O)C1-C6 alkyl, -C(=O)C2-C6 alkenyl, -C(=O)-O-C1-C6 alkyl, or -C(=O)-O-C2-C6 alkenyl, C1-C6 alkyl, or C2-C6 alkenyl;
R38 is selected from: -H, C1-C24 alkylene or C1-C24 alkenylene;
R41 for each occurrence, is independently selected from: a bond or C1-C5 alkylene;
R42 for each occurrence, is independently selected from: C6-C24 alkyl, C6-C24 alkyl carbonate, C6-C24 alkyl ether, C6-C24 alkyl ester, -R58-A1-R59 or -R58-A1-R59-A2-R60, wherein R42 has between 6-24 total carbon atoms;
R58 R59, and R60, for each occurrence, are each independently selected from: a bond, -O-, -O- C(=O)-, -OC(=O)O-, C1-C24 alkylene or C2-C24 alkenylene;
R62, R63, R64, R67, R68, R69, R74, R75, R76, R77, R108, R109, R110 and R111 for each occurrence, are each independently selected from: a bond, C1-C10 alkylene, C1-C10 alkenylene, C1-C10 alkynylene, C3-C7 cycloalkylene, C3-C7 cycloalkenylene, C5-C10-spirocycloalkylene, C3-C10- carbocyclylene, C3-Ci0-heterocyclylene, ,-R114-C3-C6-cycloalkylene-R115-, -R114-CH=CH-CH2- CH=CH-R115- , -R114-CH=CH-CH2-cPr-R115-, -R114-cPr-CH2-CH=CH-R115-, -R114-cPr-CH2-cPr-R115-, - R114-CH=CH-CD2-CH=CH-R115-, -R114-CH=CH-CD2-cPr-R115-, -R114-cPr-CD2-CH=CH-R115- or -R114- cPr-CD2-cPr-R115-; wherein, R114 and R115 are each independently selected from: a bond, C1- C10 alkylene, C1-C10 alkenylene or C1-C10 alkynylene; -cPr- is cyclopropylene (cyclopropane- 1,2 diyl); each double bond or cPr has a cis configuration; and D denotes deuterium (2H); R114 and R115 for each occurrence, are each independently, selected from: a bond, C1-C10 alkylene, C1-C10 alkenylene or C1-C10 alkynylene; -cPr- is cyclopropylene (cyclopropane-1,2 diyl); each double bond or cPr has a cis configuration; and D denotes deuterium (2H);
R61, R73 and R107, for each occurrence, are each independently selected from:
Figure imgf000022_0001
R116, for each occurrence, is independently selected from: -H, -F, -CF3, -Cl, -Br, -I, -CH3, - CH2R14, -CHR14 2, -CR143, -OH, -OR14, -NH2, -NHR14, -N(R14)2, -SH, -SR14, -S(=O)H, -S(=O)R14, - S(=O)2H, -S(=O)2R14, -PH2, -PHR14, -PR142, -P(OH)2, -POHR14, -PR142, -P(OR14)2, -P(O)H2, - P(O)HR14, -P(O)R142, -P(O)(OH)2, -P(O)(OR14)OH, -P(O)(OR14)2, -SiH3, -Si H2R14, -SiH R14 2 or - SiR14 3;
R65, R70, R78, R105 and R112, for each occurrence, are each independently selected from: a bond, C1-C10 alkylene, C1-C10 alkenylene, C1-C10 alkynylene, C3-C8 cycloalkylene, C3-C8 cycloalkenylene, C5-C10-spirocycloalkylene, C3-C10-carbocyclylene, C3-C10-heterocyclylene, - CH=CH-CH2-CH=CH- , C3-C6-cycloakylene, -CH=CH-CH2-cPr-, -cPr-CH2-CH=CH-, -cPr-CH2-cPr-, - CH=CH-CD2-CH=CH- , -CH=CH-CD2-cPr-, -cPr-CD2-CH=CH- -or -cPr-CD2-cPr-, where -cPr- is cyclopropylene, each double bond or cPr has a cis configuration and D denotes deuterium (2H);
R66, R71, R79, R106 and R113, for each occurrence, are each independently selected from: -H, linear or branched C1-C10 alkyl, C1-C10 alkenyl, or C1-C10 alkynyl, C5-C10-spirocycloalkyl, C3-C10- carbocyclyl, C3-C10-heterocyclyl, optionally substituted aryl, optionally substituted heteroaryl, C3-C18-heterocycloalkyl, optionally substituted aryl, optionally substituted heteroaryl, -R26-CH=CH-CH2-CH=CH-R28, -R26-CH=CH-CH2-cPr-R28, -R26-cPr-CH2-CH=CH-R28, - R26-cPr-CH2-cPr-R28, -R26-CH=CH-CD2-CH=CH-R28, -R26-CH=CH-CD2-cPr-R28, -R26-cPr-cPr-R28, - R26-cPr-cBu-R28, -R26-cBu-cBu-R28, -R26-cBu-cPr-R28, -R26-cHx-cBu-R28, -R26-cHx-cBu-R28, -R26- cHx-cBu-R28, -R26-cPr-cHx-R28, -R26-cBu-cHxR28, -R26-cPr-R26-cPr-R28, -R26-cPr-R26-cBu-R28, -R26- cBu-R26-cBu-R28, -R26-cBu-R26-cPr-R28, -R26-cHx-R26-cBu-R28, -R26-cHx-R26-cBu-R28, -R26-cHx- R26-cBu-R28, -R26-cPr-R26-cHx-R28, -R26-cBu-R26-cHxR28, -R26-c-CD2-cPr-R28, -R26-cPr-CD2-cPr- R28, -R26-cPr-CD2-cPr-R28, -R26-cPr-CD2-CH=CH-R28, -R26-cPr-CD2-cPr-R28 or -R26-C3-C6- cycloakylene-R26-C3-C6 cycloakylene-R28; wherein, R26 for each occurrence, is independently selected from: a bond, C1-C10 alkylene, C1-C10 alkenylene or C1-C10 alkynylene; and R28 for each occurrence, is independently selected from: -H, C1-C10 alkyl, C1-C10 alkenyl or C1-C10 alkynyl; -cPr- is cyclopropylene (cyclopropane-1,2 diyl); -cBu- is cyclobutylene; -cHx- is cyclohexylene; each double bond or -cPr- has a cis configuration; and D denotes deuterium (2H); R72, for each occurrence, is independently selected from -H, optionally substituted C1-C6 alkyl, -[CH2]1-6OH or optionally substituted C2-C6 alkenyl;
R96, for each occurrence, is independently selected from: -OH, -O-C1-C4 alkyl, -O-C(=O)-O-C1- C4 alkyl or -O-C(=O)-C1-C5 alkyl;
R118 and R119 are independently selected from R1 or R55; n is an integer selected from: 1, 2, 3, 4, 5 or 6; and k is an integer selected from: 1, 2, 3, 4, 5 or 6.
In some embodiments, said carbocyclyl or heterocyclyl is independently an optionally substituted group selected from: cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclopropenyl, cyclobutenyl, cyclopentenyl, cyclohexenyl, cycloheptenyl, cyclobutadienyl, cyclopentadienyl, cyclohexadienyl, cycloheptadienyl or cycloheptatrienyl.
In some embodiments, said carbocyclyl or heterocyclyl is independently an optionally substituted group selected from: lH-pyrrolizidinyl, 1,2-dihydroquinolinyl, 1,5- naphthyridinyl, 1,8-naphthyridinyl, lH-indazolyl, lH-isochromenyl, lH-pyrrolizidinyl, 1- naphthyl, 2H-benzo[b][l,4]oxazinyl, 2H-benzo[e][l,2]oxazinyl, 2h-chromenyl, 2-naphthyl, 4H-quinolizinyl, adeninyl, azaindazolyl, azaindolyl, benzimidazolyl, benzo[b]thiophenyl, benzo[c][l,2,5]thiadiazolyl, benzo[c]isothiazolyl, benzo[c]thiophenyl, benzo[d]isothiazolyl, benzo[d]isoxazolyl, benzo[d]oxazolyl, benzo[d]thiazolyl, benzofuryl, benzyl, cinnolinyl, cumenyl, decahydroisoquinolinyl, decahydroquinolinyl, guaninyl, indazolyl, indenyl, indolyl, indolinyl, indolizinyl, isobenzofuran, isoindolyl, isoquinolinyl, phenyl, phthalazinyl, pteridinyl, purinyl, pyrido[2,3-b]pyrazinyl, pyrido[4,3-d]pyrimidinyl, pyrimidinyl, quinazolinyl, quinolinyl, quinoxalinyl, tetrahydroquinolinyl, tolyl or xylyl.
In some embodiments, said carbocyclyl or heterocyclyl is independently an optionally substituted group selected from: furyl, imidazolyl, isothiazolyl, isoxazolyl, oxadiazolyl, oxazolyl, pyrrolyl, pyrazolyl, pyridazinyl, pyridyl (pyridinyl), pyrimidinyl, thiadiazolyl, thienyl, tetrazolyl, thiazolyl, triazolyl, , azepinyl, azetidinyl, dioxothiomorpholinyl, imidazolidinyl, morpholinyl, oxanyl, oxazinyl, oxazolidinyl, oxepinyl, oxetanyl, piperazinyl, piperidinyl, pyranyl, pyrrolidinyl, tetrahydrofuranyl, tetrahydropyranyl, thianyl, thiomorpholinyl or thiopyranyl.
In some embodiments, said carbocyclyl or heterocyclyl is independently an optionally substituted group selected from: adamantanyl, azabicyclo[3.1.0]hexanyl, 3- azabicyclo[3.1.1]heptanyl, bicyclo[2.1.1]hexyl, bicyclo[2.2.1]heptyl, bicyclo[2.2.1]heptenyl, 3-oxa-8-azabicyclo[3.2.1]octanyl, 6-oxa-3-azabicyclo[3.1.1]heptanyl, 8-Methyl-8- azabicyclo[3.2.1]octanyl, 8-oxa-3-azabicyclo[3.2.1]octanyl, 3-oxa-6- azabicyclo[3.1.1]heptanyl, tricyclo[2.2.1.0(2,6)]heptanyl, 6,6-dimethylbicyclo[3.1.1]heptyl, or 2,6,6-trimethylbicyclo[3.1.1]heptyl, 5-azaspiro[2.3]hexanyl, 2-azaspiro[3.3]heptanyl, 2- oxa-6-azaspiro[3.3]heptanyl, spiro[2.2]pentanyl, spiro[3.3]heptanyl, spiro[2.3]hexanyl or spiro[2.5]octanyl, spiro[4.5]decanyl.
In some embodiments, said carbocyclyl or heterocyclyl is independently an optionally substituted group selected from:
Figure imgf000024_0001
In some embodiments, said carbocyclylene or heterocyclylene is independently an optionally substituted group selected from: furandiyl, imidazolediyl, isothiazolediyl, isoxazolediyl, oxadiazolediyl, oxazolediyl, pyrazolediyl, pyrrolediyl, pyridazinediyl, pyridinediyl, pyrimidinediyl, thiadiazolediyl, thiendiyl, tetrazolediyl, thiazolediyl, triazolediyl, azepinediyl, azetidinediyl, dioxothiomorpholinediyl, imidazolidinediyl, morpholinediyl, oxanediyl, oxazinediyl, oxazolidinediyl, oxepinediyl, oxetanediyl, piperazinediyl, piperidinediyl, pyranyl, pyrrolidindiyl, tetrahydrofurandiyl, tetrahydropyrandiyl, thianediyl, thiomorpholinediyl or thiopyrandiyl.
In some embodiments, said carbocyclylene or heterocyclylene is independently an optionally substituted group selected from: cyclopropylene (cyclopropanediyl), cyclobutylene (cyclobutanediyl), cyclopentylene (cylcopentanediyl), cyclohexylene (cyclohexenediyl), cycloheptylene (cycloheptanediyl), cyclopropenediyl, cyclobutenylenediyl, cyclopentenylenediyl, cyclohexenediyl, cycloheptenediyl, cyclobutadienediyl, cyclopentadienediyl, cyclohexadienediyl or cycloheptadienediyl, cycloheptatrienediyl.
In some embodiments, said carbocyclylene or heterocyclylene is independently an optionally substituted group selected from: adamantanediyl, azabicyclo[3.1.0]hexanediyl, 3- azabicyclo[3.1.1]heptanediyl, bicyclo[2.1.1] hexyl, bicyclo[2.2.1]heptyl, bicyclo[2.2.1]heptenyl, 3-oxa-8-azabicyclo[3.2.1]octanediyl, 6-oxa-3- azabicyclo[3.1.1]heptanediyl, 8-oxa-3-azabicyclo[3.2.1]octanediyl, 3-oxa-6- azabicyclo[3.1.1]heptanediyl, tricyclo[2.2.1.0(2,6)]heptanyl, 6,6- dimethylbicyclo[3.1.1]heptyl, or 2,6,6-trimethylbicyclo[3.1.1]heptyl, , 5- azaspiro[2.3]hexanediyl, 2-azaspiro[3.3]heptanediyl, 2-oxa-6-azaspiro[3.3]heptanediyl, spiro[2.2]pentanyl, spiro[3.3]heptanyl, spiro[2.3]hexanediyl, spiro[2.5]octanediyl or spiro[4.5]decanediyl.
In some embodiments, said cycloalkylene, in each instance, is independently selected from:
Figure imgf000025_0001
In some embodiments for any one or more variable selected from R3, R4, Rll, R12, R15, R16, R17, R18, R62, R63, R64, R65, R67, R68, R69, R70, R74, R75, R76, R77, R78, R105, R108, R109, R110, Rill, R112, R115 or R191, said carbocyclylene or heterocyclylene is independently an optionally substituted group selected from:
Figure imgf000025_0002
Figure imgf000025_0003
In some embodiments, said carbocyclylene or heterocvclvlene is independently an optionally
Figure imgf000026_0001
substituted group selected from:
Figure imgf000026_0002
; wherein, R117 is selected from
Figure imgf000026_0003
In some embodiments, for any one of Compounds (l)-(4), -R62-A3-R63-A4-R64-R65-R66, -R62-A3-
R63-A4-R 64-R73-R74_A7_R75_R65_R6^ _R62_A3_R63_A4_R64_R73_R76_A8_R77_R78_R79; _R67_A5_R68_A6_R69_
R7O-R71, -R67_A5.R68.A6.R69.R1O7.R1O8.A12.R1O9.R1O5.R1O6 _R67_A5_R68_A6_R69_R107_R110_A13_R111_
R112-R113, -R62-A3-R63-A4-R64-R65-R66, -R62-A3-R63-A4-R64-R74-A7-R75-R65-R66, -R62-A3-R63-A4-R64-
R76-A8-R77-R78_R79; -R67-A5-R68_A6_R69_R7O_R71; _R67_A5_R68_A6_R69_R108_A12_R109_R105_R106 A N D _
R67-A5-R68-A6-R69-R110-A13-R111-R112-R113, for each occurrence, independently has: a) a total number of carbon atoms selected from: 10-24, 12-22, 14-22, 16-20, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23 or 24 carbon atoms; b) a longest linear chain (i.e., excluding side groups) with a total number of carbon atoms that is selected from: 10-24, 12-22, 14-22, 16-20, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23 or 24 carbon atoms. Note that the longest linear chain may contain non-carbon atoms including oxygen, nitrogen, sulfur and silicon, however, once the longest chain is identified, only the carbon atoms are counted; or c) .a longest linear chain (i.e., excluding side groups) with a total number of atoms (including non-carbon atoms) that is selected from: 10-24, 12-22, 14-22, 16-20, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23 or 24 atoms (including non-carbon atoms); wherein each variable, for each occurrence, is independently as described above for any one of Compounds (1) - (4) or for any embodiment of any compound herein.
For clarity, the "longest liner chain" refers only to those atoms that directly form the chain, such that no bond between any two chain atoms can be broken without breaking the chain. In some embodiments, for any one of Compounds (l)-(4), R55, for each occurrence, is independently selected from:
R62-A3-R66, -R62-A3-R63-R66, -R62-A3-R63-A4-R66, -A5-R68-A6-R69-R70-R71, -R62-A3-R63-A4-R64-R66
A4_R64_R74_A7_R75_R66; Qr -R62-A3-R63-A4-R64-R74-A7-R75-R65-R66;
; wherein each variable, for each occurrence, is independently as described above for any one of Compounds (1) - (4) or for any embodiment of any compound herein. In some further embodiments, said R55 said group has: a) a total number of carbon atoms selected from: 10-24, 12-22, 14-22, 16-20, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23 or 24 carbon atoms; or b) a longest linear chain (i.e., excluding side groups) with a total number of carbon atoms or total number of atoms (i.e., carbon and heteroatoms) that is selected from: 10-24, 12-22, 14-22, 16-20, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23 or 24 carbon atoms.
In some embodiments, for any one of Compounds (l)-(4), said compound comprises two R55 groups independently selected from:
-R62-A: -R66, -R62-A3-R63-R66, -R62-A3-R63-A4-R66, -A5-R68-A6-R69-R70-R71, -R62-A3-R63-A4-R64-R66, -
R62-A3-R63-A4-R64-R74-R66, -R62-A3-R63-A4-R64-R74-R66, -R62-A3-R63-A4-R64-R74-A7-R66, -R62-A3-R63
A4-R64-R74-A7-R75-R66, or -R62-A3-R63-A4-R64-R74-A7-R75-R65-R66; wherein, each variable, for each occurrence, is independently as described above for any one of Compounds (1) - (4) or for any embodiment of any compound herein. In some further embodiments, said R55 said group has: a) a total number of carbon atoms selected from: 10-24, 12-22, 14-22, 16-20, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23 or 24 carbon atoms; or b) a longest linear chain (i.e., excluding side groups) with a total number of carbon atoms or total number of atoms (i.e., carbon and heteroatoms) that is selected from: 10-24, 12-22, 14-22, 16-20, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23 or 24 carbon atoms.
In some embodiments, for any one of Compounds (l)-(4), R55, for each occurrence, is independently selected from:
-OC(=O)C(R42)[CH2]nC(=O)O-R41-CH(C4-Ci2 alkyl)2, -R41-N(R42)([CH2]nC(=O)O-R41-CH(C4-Ci2 alkyl)2, -OC(=O)C(R43)[CH2]nC(=O)O-R41-CH(C4-Ci2 alkyl)2, -R41-N(R43)([CH2]nC(=O)O-R41- CH(C4-Ci2 alkyl)2, -R41-C(R42)[CH2]nC(=O)O-R41-CH(C4-Ci2 alkyl)2 or -R41-CH(R43)(R44), -R41- N(R43)(R44); wherein,
R43 for each occurrence, is independently selected from: -C(=O)O-C6-C24 alkyl, -OC(=O)-C6-C24 alkyl, -OC(=O)O-C6-C24 alkyl, -C(=O)O-C6-C24 alkenyl, -O-C(=O)-C6-C24 alkenyl, -OC(=O)O-C6-C24 alkenyl-, -C2-C22 alkylene-O-C2-C22 alkyl, -C2-C22 alkenylene-O-C2-C22 alkyl, -C2-C22 alkylene-O- C2-C22 alkenyl, -C1-C22 alkylene-C(=O)O-C1-C22 alkyl, -C1-C22 alkylene-O-C(=O)O-C1-C22 alkyl, - C1-C22 alkylene-O-C(=O)-C1-C22 alkyl, -C2-C22 alkenylene-C(=O)O-C1-C22 alkyl, -C2-C22 alkenylene-O-C(=O)-C1-C22 alkyl, -C2-C22 alkenylene-O-C(=O)-Ci-C22 alkyl, -C1-C22 alkylene- C(=O)O-C2-C22 alkenyl, -C1-C22 alkylene-O-C(=O)O-C2-C22 alkenyl, -C1-C22 alkylene-O-C(=0)-C2- C22 alkenyl, -C2-C22 alkenylene-C(=O)O-C2-C22 alkenyl, -C2-C22 alkenylene-O-C(=O)O-C2-C22 alkenyl, -C2-C22 alkenylene-O-C(=O)-C2-C22 alkenyl; wherein R43 has 6-24 total carbon atoms;
R44 for each occurrence, is independently selected from: -R45-C(=O)O-R45-CH(C6-C12 a I ky 1)2, - R45- OC(=O) - R45-CH(C6-C12 alkyl)2, -R45-OC(=O)O-R45-CH(C6-C12 alkyl)2, R45-C(=O)O-R45-CH(C6- C12 alkenyl)2, -R45- OC(=O) - R45-CH(C6-C12 alkenyl)2, -R45-OC(=O)O- R45-CH(C6-C12 alkenyl)2; wherein R44 has 6-24 total carbon atoms and wherein R45 is, for each occurrence, independently selected from: a bond, C1-C6 alkylene or C1-C6 alkenylene; and each remaining variable, for each occurrence, is independently as described above for any one of Compounds (1) - (4) or for any embodiment of any compound herein.
In some embodiments, for any one of Compounds (l)-(4), R55, for each occurrence, is independently selected from:
Figure imgf000028_0001
Figure imgf000029_0001
wherein:
R16 for each occurrence, is independently selected from: a bond or — [CH2]k-; k is an integer selected from: 1, 2, 3, 4, 5 or 6;
Figure imgf000029_0002
R62, R63, R67, R68, R74, R76, R108, and R110 are each independently selected from: C1-C12 alkylene or C1-C12 alkenylene; R66, R71, R79, R106 and R113 are each independently selected from: C1-C12 alkyl or C1-C12 alkenyl;
A3 and A5 are each independently selected from: -O-, -OC(=O)-, -C(=O)O- or -OC(=O)O-; and
A4, A6, A7, A8, A12, A13, R64, R65, R69, R70, R75, R77, R78, R105, R109, R110, Rmand R112 are a bond; and each remaining variable, for each occurrence, is independently as described above for any one of Compounds (1) - (4) or for any embodiment of any compound herein. In some further embodiments, R62, R63, R66 and R74 together, contain a total of 10-22 carbon atoms. In some other embodiments, R62, R63 and R66, together, contain a total of 10-22 carbon atoms. In some other embodiments, R62, R63, R76 and R79, together, contain a total of 10-22 carbon atoms. In some other embodiments, R67, R68 and R71, together, contain a total of ID- 22 carbon atoms. In some other embodiments, R67, R68, R108 and R106, together, contain a total of 10-22 carbon atoms. In some other embodiments, R67, R68, R110 and R113, together, contain a total of 10-22 carbon atoms. In some other embodiments, R63 a bond. In other embodiments, R63 and R68 are absent.
In some embodiments, for any one of Compounds ( l)-(4), R55, for each occurrence, is independently:
Figure imgf000030_0001
wherein,
A3 and A5, for each occurrence, are each independently selected from: -O-, -OC(=O)-, - C(=O)O- or -OC(=O)O-;
R16, for each occurrence, is independently selected from: a bond or -[CH2]k-; k is an integer selected from: 1, 2, 3, 4, 5 or 6; ccurrence, are each independently selected from:
Figure imgf000030_0002
Figure imgf000030_0003
R62, R63, R67, R68, R74, R76, for each occurrence, are each independently selected from: C1-C12 alkylene or C1-C12 alkenylene;
R66, R7i a nc| R79, for each occurrence, are each independently selected from: C1-C12 alkyl or C1-C12 alkenyl;
R116, for each occurrence, and any remaining variable, for each occurrence, is independently as described above for any one of Compounds (1) - (4) or for any embodiment of any compound herein; and wherein, R62, R63, R66 and R74, together, contain 10-22 total carbon atoms; R62, R63, R76 and R79, together, contain 10-22 total carbon atoms; and R67, R68 and R71, together, contain 10- 22 total carbon atoms. In some other embodiments, R63 a bond. In some other embodiments, R63 and R68 are absent.
In some embodiments, -R62-A3-R63-R73-R74-R66, -R62-A3-R63-R73-R76-R79, and -R67-A5-R68-R71, independently has: c) a total number of carbon atoms selected from: 10-24, 12-22, 14-22, 16-20, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23 or 24 carbon atoms; d) a longest linear chain with a total number of carbon atoms that is selected from: 10- 24, 12-22, 14-22, 16-20, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23 or 24 carbon atoms; or e) a longest linear chain with a total number of atoms that is selected from: 10-24, 12-22, 14-22, 16-20, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23 or 24 atoms.
In some embodiments, for any one of Compounds ( l)-(4), R55, for each occurrence, is independently:
Figure imgf000031_0003
wherein:
A3 and A5, for each occurrence, are each independently selected from: -O-, -OC(=O)-, - C(=O)O- or -OC(=O)O-;
R16 for each occurrence, is independently selected from: a bond or -[CH2]k-; k is an integer selected from: 1, 2, 3, 4, 5 or 6;
R61, R73 and R107, for each occurrence, are each independently selected from:
Figure imgf000031_0001
Figure imgf000031_0002
R62, R63, R67, R68, R74, R76, R108, and R110, for each occurrence, are each independently selected from: C1-C12 alkylene or C1-C12 alkenylene;
R66, R79, R106 and R113, for each occurrence, are each independently selected from: C1-C12 alkyl or C1-C12 alkenyl;
R116 and any remaining variables, for each occurrence, is independently as described above for any one of Compounds (1) - (4) or for any embodiment of any compound herein. In some further embodiments, R62, R63, R66 and R74, together, contain 10-22 total carbon atoms; R62, R63, R76 and R79, together, contain 10-22 total carbon atoms; and R67, R68, R108 and R106, together, contain 10-22 total carbon atoms; and, R67, R68, R110 and R113, together, contain 10-22 total carbon atoms. In some other embodiments, R63 a bond. In other embodiments, R63 and R68 are absent. In some other embodiments, R73 and R107 are
Figure imgf000032_0001
In some embodiments, for any one of Compounds ( l)-(4), R55, for each occurrence, is independently:
Figure imgf000032_0002
wherein:
A3 and A5, for each occurrence, are each independently selected from: -O-, -OC(=O)-, - C(=O)O- or -OC(=O)O-;
R16, for each occurrence, is independently selected from: a bond or -[CH2]k-; k is an integer selected from: 1, 2, 3, 4, 5 or 6;
R61, R73 and R107, for each occurrence, are each independently selected from:
Figure imgf000032_0003
Figure imgf000032_0004
R62, R63, R67, R68, for each occurrence, are each independently selected from: C1-C12 alkylene or C1-C12 alkenylene;
R66 and R71, for each occurrence, are each independently selected from: C1-C12 alkyl or C1-C12 alkenyl; R116 and any remaining variables, for each occurrence, is independently as described above for any one of Compounds (1) - (4) or for any embodiment of any compound herein. In some further embodiments, R62, R63 and R66 together, contain 10-22 total carbon atoms. In some other embodiments, R67, R68 and R71, together, contain 10-22 total carbon atoms. In some other embodiments, R63 a bond. In other embodiments, R63 and R68 are a bond. In some embodiments, for any one of Compounds ( l)-(4), R55, for each occurrence, is independently selected from:
Figure imgf000033_0001
; or
Figure imgf000034_0001
; wherein,
A3-A8, A12, A13, R62-R69, R73-R79, and R105-R113 and any remaining variables, for each occurrence, are as defined above for any one of Compounds ( l)-(4) or for any embodiment of any compound herein. In some further embodiments, a compound selected from any one of Compounds (l)-(4) comprises a first and a second R55 with longest linear chains that differ in length by 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 atoms. In some embodiments, said first and second R55 have longest linear chains with a different number of carbon atoms. In some further embodiments, a first R55 has a longest linear chain with 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 less carbon atoms than a longest linear chain of a second R55.
In some embodiments, the ionizable cationic lipid has a structural formula selected from:
Compound (5):
Figure imgf000034_0002
Compound (7):
Figure imgf000035_0001
(7), or
Compound (8):
Figure imgf000035_0002
(8); wherein:
R16, for each occurrence, is independently selected from: a bond or — [CH2]k-;
R62, R63, R64, R67, R68, R69, R74, R75, R76 and R77, for each occurrence, are each independently selected from: a bond, C1-C24 alkylene or C1-C24 alkenylene; rence, are each independently selected from:
Figure imgf000035_0003
Figure imgf000035_0004
R65, R70 and R78, for each occurrence, are each independently selected from: a bond, - CH=CH-CH2-CH=CH- , -CH=CH-CH2-cPr-, -cPr-CH2-CH=CH-, -cPr-CH2-cPr-, -CH=CH-CD2-CH=CH- , -CH=CH-CD2-cPr-, -cPr-CD2-CH=CH- -or -cPr-CD2-cPr-, where -cPr- is cyclopropylene, each double bond or cPr has a cis configuration and D denotes deuterium (2H);
R66, R71 and R79, for each occurrence, are each independently selected from: C1-C24 alkyl or C1-C24 alkenyl;
A3, A4, A5, A6, A7 and A8 and R1, for each occurrence, and each remaining variable, for each occurrence, is independently as described above for any one of Compounds ( l)-(4) or any embodiment of any compound herein. In some further embodiments, -R62-A3-R63-A4-R64-R65-
Figure imgf000035_0005
R67-A5-R68-A6-R69-R70-R71, each independently has a total of 10-24 carbon atoms. In some embodiments, the ionizable cationic lipid is one of:
Compound (9)
Figure imgf000036_0003
(10),
Compound (11)
Figure imgf000036_0001
Compound (12)
Figure imgf000036_0002
or a pharmaceutically acceptable salt or co-crystal thereof; wherein: R61. for each occurrence, is independently selected from:
Figure imgf000037_0001
Figure imgf000037_0002
R85 and R90, for each occurrence, are each independently selected from: a bond or -[CH2]I-8-;
R86 and R91, for each occurrence, are each independently selected from: a bond -O-, -O- C(O=)-, -O-C(=O)-O- or -C(=O)-O-;
R87, for each occurrence, is independently selected from: a bond or -[CH2]I-6-;
R88 and R89, for each occurrence, are each independently -[CH2]I-IOCH3;
R92, for each occurrence, is -[CH2]2-i6CH3;
R93 and R94, for each occurrence, are each independently selected from: -H, -D, -CH3, -CD3, - CH2CH3, -CD2CH3, where D denotes deuterium (2H);
R95, R96, R100, R101 and R102, for each occurrence, are each independently selected from: -OH, -O-C1-C4 alkyl, -O-C(=O)-O-Ci-C4 alkyl or -O-C(=O)-Ci-C4 alkyl;
R97, for each occurrence, is independently selected from: -[CH2]I-6-, -[CH2]I-6-C(=O)-O-, -
[CH2]I-6-C(=O)-O-, -[CH2]I-6-O- or -[CH2]I-6-C(=O)-, wherein:
H V-
Figure imgf000037_0003
when R61 is Z or X , then R97 is selected from: -[CH2]I-6- or -[CH2]I-6-C(=O)-O- and when R , then R97 is selected from: -[CH2]I-6-, -[CH2]I-6-C(=O)-O-, -[CH2]I-6-O- or - [CH2]I-6
Figure imgf000037_0004
R16, for each occurrence, is independently selected from: a bond or -[CH2]k-;
R98, for each occurrence, is independently selected from: a bond, -C(=O)-, or -C(=O)-O-; and
R104, for each occurrence, is independently selected from:
Figure imgf000038_0001
wherein: R85-R92 and R97, for each occurrence, are each independently as described above for any one of Compounds (8), (10) or (11); and wherein: each remaining variable, for each occurrence, is independently as described above for any one of Compounds (l)-(8) or any embodiment of any compound herein.
In some embodiments, for any one of Compounds (1)-(12), R85 and R90 are the same. In some other embodiments, for any one of Compounds (1)-(12), R85 and R90 are each independently -[CH2]4-5--
In some embodiments, for any one of Compounds (1)-(12), R86 and R91 are -C(O=)-O-. In some other embodiments, for any one of Compounds (1)-(12), R86 and/or R91 are/a bond. In some other embodiments, for any one of Compounds (1)-(12), R86 and/or R91 are/is -C(=O)- O-. In some embodiments, for any one of Compounds (1)-(12), R88 and R89are each independently -[CH2]6-IO-.
In some embodiments, for any one of Compounds (1)-(12), R92 is -[CH2]8-I2-.
In some embodiments, for any one of Compounds (1)-(12), R85, R86, R87, R88 and R89, together, have between 14-24 or between 16-22 total carbon atoms.
In some embodiments, for any one of Compounds (1)-(12), R90, R91 and R92, together, have between 10-20 or between 12-18 total carbon atoms. In some embodiments, for any one of Compounds (1)-(12), R88 and R89 are identical.
In some embodiments, for any one of Compounds (1)-(12), R85, R86, R87 and R88, together have a total number of carbon atoms that differs by +/- 3, +/- 2 or +/-1, or is equal to the total number of carbon atoms of R90, R91 and R92, combined.
In some embodiments, for any one of Compounds (1)-(12),_R93 and/or R94 are/is -H. In some embodiments, _R95 and/or R96 are/is -OH.
In some embodiments, for any one of Compounds (1)-(12), R85 and R90 are each independently -[CH2]4-5-; R86 and R91 are -C(O=)-O-; R88 and R89are each independently - [CH2]6-IO-; R92 is -[CH2]8-I2-; R93 and/or R94 are/is -H; and R95 and/or R96 are/is -OH.
Figure imgf000039_0001
When R61 is J- for any one of compounds 1-12, the carbon of R61 may be a chiral center. In some embodiments, the chiral center is in an R configuration and in other embodiments, an S configuration.
In some embodiments, the ionizable cationic lipid has a structural formula selected from any one of:
Compound (13)
Figure imgf000039_0002
(13),
Compound (14)
Figure imgf000040_0001
Figure imgf000041_0001
( ) or a pharmaceutically acceptable salt or co-crystal thereof; wherein:
R80, for each occurrence, is independently selected from: a bond, R16 or - [d-hJi-e-O- or -O-;
R16, for each occurrence, is independently selected from: a bond or -[CFhJk-;
R81, R82, R83, and R84, for each occurrence, are each independently selected from: a bond or - [CH2]I-IO-;
R61, R93, R94, R95, R96, R97 R98 and each remaining variable, for each occurrence, is independently as described above for any one of Compounds ( l)-( 12) or any embodiment of any compound herein.
In some embodiments, for any one of Compounds (1)-(19), R81 and R83 are each independently -[CH2]5-9-- In other embodiments, for any one of Compounds (1)-(19), R82 and R84 are each independently -[Cl-hjo-e-- In other embodiments, for any one of Compounds (1)- (19), R81 and R83 are each independently -[C]-hjs-io- and R82 and R84 are each independently - [CH2]O-6".
In other embodiments, for any one of Compounds (1)-(19), R81 and R82, combined, have between 7-13 total carbon atoms. In other embodiments, for any one of Compounds (1)- (19), R83 and R84, combined, have between 7-13 total carbon atoms. In other embodiments for any one of Compounds ( l)-( 19), R81 and R82, combined, have between 8-12 total carbon atoms. In other embodiments, for any one of Compounds (1)-(19), R83 and R84, combined, have between 8-12 total carbon atoms. In other embodiments, for any one of Compounds (1)-(19), R81 and R82, combined, and R83 and R84, combined, each independently have between 7-13 or between 8-12 total carbon atoms. In other embodiments, for any one of Compounds (1)-(19), R81 and R82, combined, and R83 and R84, combined, each have identical number of total carbon atoms which is between 7-13 or between 8-12. In other embodiments, for any one of Compounds (1)-(19), R81 and R82, combined, and R83 and R84, combined, have a different number of total carbon atoms. In other embodiments, for any one of Compounds ( l)-( 19), when R81 and R82, combined, and R83 and R84, combined, have a different number of total carbon atoms, the carbon atom between R81 and R83 is a chiral center. In some further embodiments, the chiral center is in an R configuration and in other embodiments, an S configuration. In some embodiments, the ionizable cationic lipid has a structural formula selected from any one of:
Compound (20)
Figure imgf000042_0001
(22),
Compound (23)
Figure imgf000043_0001
(24),
Compound (25)
Figure imgf000043_0002
Compound (27)
Figure imgf000044_0002
(28), or a pharmaceutically acceptable salt or co-crystal thereof; wherein: R61, R85, R86, R87, R88, R89, R90, R91, R92, R93, R94, R96, R97 and each remaining variable, for each occurrence, is independently as described above for any one of Compounds (l)-( 19) or other embodiment of any compound herein.
In some embodiments, the ionizable cationic lipid has a structural formula selected from:
Compound (29)
Figure imgf000044_0001
(29),
Compound (30)
Figure imgf000045_0001
(33),
Compound (34)
Figure imgf000046_0001
(35),
Compound (36)
Figure imgf000046_0002
(37),
Compound (38)
Figure imgf000047_0001
Compound (40)
Figure imgf000047_0002
(41),
Compound (42)
Figure imgf000048_0001
Compound (46)
Figure imgf000049_0003
Compound (48)
Figure imgf000049_0001
(48) or
Compound (49)
Figure imgf000049_0002
(49),
Compound (50R)
Figure imgf000050_0001
Compound (53)
Figure imgf000051_0001
or a pharmaceutically acceptable salt or co-crystal thereof; wherein:
R1, R3, R16, R36, A1, and each remaining variable, for each occurrence, is independently as described above for any one of Compounds (l)-(28) or any embodiment of any compound herein.
In some embodiments for an ionizable cationic lipid of the invention comprising a chiral center, e.g., as indicated by an *, the chiral center is in an R-configuration, in other embodiments an S-configuration, and in still further embodiments, either in an R- or an S- configuration.
In some embodiments for an ionizable cationic lipid of the invention, R1 and R3, for each occurrence, are each independently, -H, optionally substituted C1-C6 alkyl, optionally substituted C2-C6 alkenyl, optionally substituted C1-C6 hydroxyalkyl or optionally substituted C2-C6 hydroxyalkenyl. In other embodiments, R1 and R3, for each occurrence, are each independently selected from: -H, methyl, ethyl, propyl, isopropyl, butyl, isobutyl or tertbutyl. In further embodiments, R2 is absent or -H. In further embodiments, R1 and R3, for each occurrence, are each independently selected from: -H, -D, -CH3, -CD3, -CH2CH3, - CD2CH3, where D denotes deuterium (2H); and R2 is absent.
In some embodiments for an ionizable cationic lipid of the present invention, R1 and R3 , for each occurrence, are each independently selected from: -H, C1-C6 alkyl, C2-C6 alkenyl, C1-C5 alkyl, C2-C5 alkenyl, C1-C4 alkyl, C2-C4 alkenyl, Ce alkyl, C5 alkyl, C4 alkyl, C3 alkyl, C2 alkyl, Ci alkyl, C6 alkenyl, or C5 alkenyl, or C4 alkenyl, or C3 alkenyl, or C2 alkenyl. In some further embodiments, R1 and R3 are each independently selected from: -H or C1-C3 alkyl. In some embodiments, RT and R3 are identical.
In some embodiments for an ionizable cationic lipid of the present invention, R2 is absent.
In other embodiments, for an ionizable cationic lipid of the present invention, R4, for each occurrence, is independently selected from:
Figure imgf000052_0001
Figure imgf000052_0002
or ; wherein R41,
R35, R55, R96 and each remaining variable, for each occurrence, is independently as described above for any one of Compounds (l)-(54) or any embodiment of any compound herein.
In some embodiments for an ionizable cationic lipid of the present invention, R6, for each occurrence, is independently selected from C3-C24 branched alkylene or C3-C24 branched alkenylene.
In some embodiments for an ionizable cationic lipid of the present invention, R6, for each occurrence, is independently selected from: C1 -C3 alkylene, C1-C9 alkylene, C2-C9 alkenylene, C1-C7 alkylene, C2-C7 alkenylene, C1 -C5 alkylene, C2-C5 alkenylene, C2-C8 alkylene, C2-C8 alkenylene, C3-C7 alkylene, C3-C7 alkenylene, C5-C7 alkylene, C5-C7 alkenylene, C12 alkylene, CH alkylene, C10 alkylene, C9 alkylene, Cs alkylene, C7 alkylene, Ce alkylene, C5 alkylene, C4 alkylene, C3 alkylene, C2 alkylene, Ci alkylene, C12 alkenylene, C11 alkenylene, Cwalkenylene, C9 alkenylene, C8 alkenylene, C7 alkenylene, C6 alkenylene, C5 alkenylene, C4 alkenylene, C3 alkenylene, C2 alkenylene.
In some embodiments for an ionizable cationic lipid of the present invention, R8 and R9, for each occurrence, are each independently C6-C12 alkyl or C6-C12 alkenyl. In other embodiments, R8 is Ce-Cio alkyl or Ce-Cio alkenyl. In other embodiments, R8 and R9, together, have a total of 12-20 carbon atoms.
In some embodiments for an ionizable cationic lipid of the present invention, R8, R9, R12, and R13, each independently have 1, 2 or 3, C=C double bonds. In further embodiments, at least one of the C=C double bonds is of Z configuration. In further embodiments, each C=C double bond is in a Z configuration. In further embodiments, for an ionizable cationic lipid of the invention, R14 is C1-C3 alkyl, e.g., methyl or ethyl. In further embodiments, R8 and R9, are each independently selected from: -C10-C18 alkenyl comprising 1, 2, 3 or 4 cis double bonds, - C10-C18 alkenyl ester comprising 1, 2, 3 or 4 cis double bonds, optionally substituted -C6-C18 alkyl, -C6-718 alkyl ester, -C6-C18 alkyl ether, -C6-C18 alkyl carbonate, -C6-C18 alkenyl, -C6-C18 alkenyl ester, -C6-C18 alkenyl ether, or -C6-C18 alkenyl carbonate. In further embodiments, R8 and R9, are each independently -C10-C18 alkenyl comprising 2 cis (Z configuration) double bonds. In some embodiments for an ionizable cationic lipid of the present invention, R8 and R9 are each independently selected from: optionally substituted C6-C18 alkyl, C6-718 alkylester, C6-C18 alkyl ether, C6-C18 alkyl carbonate C6-C18 alkenyl, C6-C18 alkenylester, C6-C18 alkenylether, or C6-C18 alkenylcarbonate.
In some embodiments for an ionizable cationic lipid of the present invention, R8 and R9, for each occurrence, are each independently selected from: C6-C14 alkyl, C6-C14 alkenyl, Cs-C12 alkyl, C8-C12 alkenyl, C16 alkyl, C15 alkyl, C14 alkyl, C13 alkyl, C12 alkyl, C11 alkyl, C10 alkyl, C9 alkyl, Cs alkyl, C7 alkyl, C16 alkenyl, C15 alkenyl, C14 alkenyl, C13 alkenyl, C12 alkenyl, C11 alkenyl, C10 alkenyl, C9, alkenyl, C8 alkenyl, or C7 alkenyl; wherein R8 and R9, combined, have more than 15 total carbon atoms. In some embodiments, R8 and R9 have an equal number of carbon atoms or are identical: wherein R8 and R9, combined, have more than 15 total carbon atoms. In some embodiments, R8 and R9 differ in total carbon atoms from each other; R8 and R9 differ by one or two total carbon atoms from each other; or R8 and R9 differ by one total carbon atoms.
In some embodiments, for an ionizable cationic lipid of the present invention, R8 is C7 alkyl and R9 is C8 alkyl; R8 is C8 alkyl and R9 is C7 al kyl;R8 is C8 alkyl and R9 is C9 alkyl; R8 is C9 alkyl and R9 is C8 alkyl; R8 is C9 alkyl and R9 is C10 a I kyl;R8 is C10 alkyl and R9 is C9 alkyl; R8 is C10 alkyl and R9 is C11 alkyl; R8 is C11 alkyl and R9 is C10 alkyl; R8 is C11 alkyl and R9 is C12 alkyl; R8 is C12 alkyl and R9 is C11 alkyl; R8 is C7 alkyl and R9 is C9 alkyl; R8 is C9 alkyl and R9 is C7 alkyl; R8 is C8 alkyl and R9 is C10 alkyl; R8 is C10 alkyl and R9 is C8 alkyl; R8 is C9 alkyl and R9 is C11 alkyl; R8 is C11 alkyl and R9 is C9 alkyl; R8 is C10 alkyl and R9 is C12 alkyl; R8 is C12 alkyl and R9 is C10 alkyl; R8 is C11 alkyl and R9 is Cs alkyl; or R8 is C8 alkyl and R9 is C11 alkyl.
In some embodiments for an ionizable cationic lipid of the present invention, R10 and R11, for each occurrence, are each independently selected from: C1-C16 unbranched alkyl or C2-C16 unbranched alkenyl.
In some embodiments, for an ionizable cationic lipid of the present invention, R16 is C1-C3 alkylene.
In some embodiments, for an ionizable cationic lipid of the present invention, R29, R30 and R31 are each -CH2-.
In some embodiments, for an ionizable cationic lipid of the present invention, R32, R33 and R34 are each -O-.
In some embodiments, for an ionizable cationic lipid of the present invention, R35 is -OH.
In some embodiments, for an ionizable cationic lipid of the present invention, R29, R30 and R31 are each -CH2-, and R32, R33 and R34are each -O-. In some embodiments, for an ionizable cationic lipid of the present invention, R29, R30 and R31 are each -CH2-, R32 and R33 are each -O- and R35 is -OH.
In some embodiments for an ionizable cationic lipid of the present invention, R42 is - [CH2]nC(=O)O[CH2]r; wherein n is as defined above for Compounds (l)-(4) or any embodiment of any compound herein; and r is an integer selected from 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16 or 17. In some embodiments for an ionizable cationic lipid comprising an R42 variable, R42, for each occurrence, is independently selected from: -R46-C(=O)O-R47-, - R46-OC(=O)-R47-, -R46-OC(=O)O-R47-,or -R46-O-R47-, wherein; R46, for each occurrence, is independently selected from C1-C12 alkylene, C1-C12 alkylene ester, C1-C12 alkylene ether, Ci- Ci2 alkylene carbonate; and R47, for each occurrence, is independently selected from -C1-C12 alkyl, -Ci-Ci2 alkyl ester, -C1-C12 alkyl ether, or -C1-C12 alkyl carbonate.
In some embodiments, for an ionizable cationic lipid of the present invention, R55 for each occurrence, is independently:
Figure imgf000054_0001
; wherein A1, R6, A2, R7, R8 and R9, are as described for any one of Compounds (l)-(28) or any embodiment of any compound herein.
In some embodiments, for an ionizable cationic lipid of the present invention, R55 for each occurrence, is independently selected from:
Figure imgf000054_0002
or wherein:
R5 for each occurrence is independently selected from: -H, optionally substituted C1-C16 alkyl, optionally substituted C1-C16 alkyl ester, optionally substituted C2-Ci6 alkenyl, optionally substituted C2-Ci6 alkenyl ester,
Figure imgf000054_0003
; wherein, R10 and R11 are each independently selected from: C1-C12 unbranched alkyl, C2-C12, unbranched alkenyl, C2-Ci2 unbranched alkyl, C2-C12 unbranched alkenyl, C5-C7, unbranched alkyl, or C5-C7 unbranched alkenyl. In further embodiments, R10 and R11 are each independently selected from; C16 unbranched alkyl, C15 unbranched alkyl, C14 unbranched alkyl, C13 unbranched alkyl, C12 unbranched alkyl, C11 unbranched alkyl, C10 unbranched alkyl, C9 unbranched alkyl, Cs unbranched alkyl, C7 unbranched alkyl, Ce unbranched alkyl, C5 unbranched alkyl, C4 unbranched alkyl, C3 unbranched alkyl, C2 unbranched alkyl, or Ci unbranched alkyl. In further embodiments, R10 and R11 are each independently selected from; C16 unbranched alkenyl, C15 unbranched alkenyl, C14 unbranched alkenyl, C13 unbranched alkenyl, C12 unbranched alkenyl, C11 unbranched alkenyl, C10 unbranched alkenyl, C9 unbranched alkenyl, C8 unbranched alkenyl, C7 unbranched alkenyl, Ce unbranched alkenyl, C5 unbranched alkenyl, C4 unbranched alkenyl, C3 unbranched alkenyl, or C2 alkenyl; and R15, R6, A2, R7, R8, R9 and any remaining variables are as described for any one of Compounds (l)-(54) or any embodiment of any compound herein. In further embodiments, R10 and R11 are each independently selected from; C2-C10 unbranched alkyl, or C2-C10 unbranched alkenyl. In further embodiments, R10 and R11 are identical.
In some embodiments, for an ionizable cationic lipid of the present invention, R55 for each occurrence, is independently selected from:
Figure imgf000055_0001
wherein:
R15, R5, R6, A2, R7, R8 and R9 is as described for any one of Compounds (l)-(54) or any embodiment of any compound herein.
In some embodiments, for an ionizable cationic lipid of the present invention, R55 for each occurrence, is independently selected from:
Figure imgf000055_0002
Figure imgf000056_0001
or wherein:
R15, R5, R6, R7, R8 and R9 is as described for any one of Compounds (l)-(54) or any embodiment of any compound herein.
In some embodiments, for an ionizable cationic lipid of the present invention, R55 for each occurrence, is independently selected from:
Figure imgf000056_0002
or ; wherein: R15, R6, A2, R7, R8 and R9 is as described for any one of Compounds (l)-(54) or any embodiment of any compound herein.
In some embodiments, for an ionizable cationic lipid of the present invention, R55 for each occurrence, is independently selected from:
Figure imgf000056_0003
A2, R7, R8 and R9 is as described for any one of Compounds (l)-(54) or any embodiment of any compound herein.
In some embodiments for an ionizable cationic lipid of the present invention, R5 for each occurrence is independently selected from: C1-C14 unbranched alkyl, C2-C14 unbranched alkenyl, or
Figure imgf000057_0001
, wherein: R10 and R11 for each occurrence, are each independently selected from: C1-C12 unbranched alkyl, C2-C12, unbranched alkenyl, C2-C12 unbranched alkyl, C2-C12 unbranched alkenyl, C5-C7, unbranched alkyl, or C5-C7 unbranched alkenyl. In further embodiments, R10 and R11 are each independently selected from; Ci6 unbranched alkyl, C15 unbranched alkyl, C14 unbranched alkyl, C13 unbranched alkyl, C12 unbranched alkyl, Cn unbranched alkyl, C10 unbranched alkyl, C9 unbranched alkyl, Cs unbranched alkyl, C7 unbranched alkyl, Ce unbranched alkyl, C5 unbranched alkyl, C4 unbranched alkyl, C3 unbranched alkyl, C2 unbranched alkyl, or Ci unbranched alkyl. In further embodiments, R10 and R11 are each independently selected from; Ci6 unbranched alkenyl, C15 unbranched alkenyl, C14 unbranched alkenyl, C13 unbranched alkenyl, C12 unbranched alkenyl, Cn unbranched alkenyl, C10 unbranched alkenyl, C9 unbranched alkenyl, Cs unbranched alkenyl, C7 unbranched alkenyl, Ce unbranched alkenyl, C5 unbranched alkenyl, C4 unbranched alkenyl, C3 unbranched alkenyl, or C2 alkenyl. In further embodiments, R10 and R11 are each independently selected from; C2-C10 unbranched alkyl, or C2-C10 unbranched alkenyl. In further embodiments, R10 and R11 are identical. In some embodiments, R5, for each occurrence, is independently selected from: C1-C16 unbranched alkyl, C1-C16 unbranched alkyl ester, unbranched C2-C16 alkenyl or C2-C16 unbranched alkenyl ester. In some embodiments, R5, for each occurrence, is independently selected from:C3-Ci6 branched alkyl, C3-C16 branched alkyl ester, C3-C16 branched alkenyl, or C3-C16 branched alkenyl ester. In some embodiments, for an ionizable cationic lipid of the present invention, R55 for each occurrence, is independently selected from: -OC(=O)C(R42)[CH2]nC(=O)O-R41-CH(C4-Ci2 alkyl)2, -R41-N(R42)([CH2]nC(=O)O-R41-CH(C4-Ci2 alkyl)2, -OC(=O)C(R43)[CH2]nC(=O)O-R41-CH(C4- C12 alkyl)2, -R41-N(R43)([CH2]nC(=O)O-R41-CH(C4-Ci2 alkyl)2, -R41-C(R42)[CH2]nC(=O)O-R41- CH(C4-Ci2 alkyl)2, -R41-CH(R43)(R44), or -R41-N(R43)(R44); wherein
R41-R45 and each remaining variable, for each occurrence, is independently as described above for any one of Compounds (l)-(54) or any embodiment of any compound herein.
In some embodiments for an ionizable cationic lipid of the present invention, A2 is -OC(=O)-, -OC(=O)R16C(=O)O- or -C(=O)O-.
In some embodiments for an ionizable cationic lipid of the present invention, each R55 has 12-50, 18-30, 20-30, or 12-24 total carbon atoms. In further embodiments, each R55 comprises 0-6, 0-3, 1-3, 1, 2, 3, 4, 5 or 6 esters; 0-6, 0-3, 1-3, 1, 2, 3, 4, 5 or 6 ethers; and/or 0-6, 0-3, 1-3, 1, 2, 3, 4, 5 or 6 carbonates.
In some embodiments for an ionizable cationic lipid of the present invention, R55 for each occurrence, is independently selected from: -O-C(=O)C(R42)[CH2]nC(=O)O-R41-CH(C4-Ci2 alkyl)2, -R41-N(-R42)(-[CH2]nC(=O)O-R41-CH(C4-Ci2 alkyl)2, -O-C(=O)C(R43)-[CH2]nC(=O)O-R41-CH(C4-Ci2 alkyl)2, -R41-N(-R43)(-[CH2]nC(=O)O-R41- CH(C4-CI2 alkyl)2, -R41-C(R42)[CH2]nC(=O)O-R41-CH(C4-Ci2 alkyl)2, -R41-CH(R43)(R44), or -R41- N(R43)(R44).
In some embodiments for an ionizable cationic lipid of the present invention, R55 for each occurrence, is independently selected from: -R16-N(-R48)-R49, -R16-CH(-R48)-R49, -R16-OC(=O)- R16-N(-R48)-R49, -R16-OC(=O)CH(-R48)-R49, or -R16-C(=O)O-R16-CH(-R48)-R49; wherein: a) R48 is -R16-O-CH(-[CH2]5-9 CH3)2 and R49 is -R16-C(=O)O-[CH2]6-I2CH3, b) R48 is -CH2O-C(=O)O-CH(-[CH2]7 CH3)2 and R49 is -R16-O-C(=O)O-[CH2]7-I3CH3, c) R48 is -R16C(=O)O-CH([CH2]7 CH3)2 and R49 is -R16C(=O)O-[CH2]7CH3, d) R48 is -[CH2]3C(=O)O-CH(-[CH2]7 CH3)2 and R49 is -[CH2]3C(=O)O-[CH2]7CH3, e) R48 is -CH2C(=O)O-CH(-[CH2]9 CH3)2 and R49 is -C(=O)O-[CH2]I0CH3, f) R48 is -[CH2]3C(=O)O-CH(-[CH2]7CH3)2 and R49 is -[CH2]3C(=O)O-[CH2]9CH3 g) R48 is -[CH2]2C(=O)O-CH(-[CH2]7 CH3)2 and R49 is -[CH2]2C(=O)O-[CH2]9CH3, or h) R48 is -R16-C(=O)O-R16-CH(-R57-CH3)2 and R49 is -[CH2]8-I6CH3;
R16, for each occurrence, is independently selected from: a bond or -[CH2]k-;
R57, for each occurrence, is independently selected from: a bond or -[CH2]m-; wherein m is an integer selected from: 1, 2, 3, 4, 5, 6, 7 or 8; and each remaining variable, for each occurrence, is independently as described above for any Compound or for any other embodiment of any compound herein.
In some embodiments, for an ionizable cationic lipid of the present invention, R55 for each occurrence, is independently selected from:
Figure imgf000058_0001
Figure imgf000059_0001
Figure imgf000060_0001
; wherein: R16, R57, A1, A2, R61, R85, R86, R87, R88, R89, R90, R91, R92, R97 and each remaining variable, for each occurrence, is independently as described above for any one of Compounds (l)-(54) or any embodiment of any compound herein.
In some embodiments, R55for each occurrence, is independently selected from:
Figure imgf000060_0002
Figure imgf000061_0002
; wherein:
R16, R57, A1, R61, R81, R82, R83, R84, R97 and each remaining variable, for each occurrence, is independently as described above for any one of Compounds (l)-(54) or any embodiment of any compound herein; and wherein each cyclopropyl (cPr) fused to adjacent chain
4 carbons, i.e., , and each -C=C-, as shown in any one of the structures, has a cis (or
E) configuration.
In some embodiments, R55for each occurrence, is independently selected from:
Figure imgf000061_0001
or
Figure imgf000062_0001
; wherein:
R16, R57, A1, and each remaining variable, for each occurrence, is independently as described above for any one of Compounds (l)-(54) or any embodiment of any compound herein; and wherein each -C=C-, as shown in any one of the structures has a cis configuration. In some embodiments, R55for each occurrence, is independently selected from:
Figure imgf000062_0002
Figure imgf000063_0001
; wherein, R16, R61, R62 and R67, for each occurrence, are each independently as described above for any one of Compounds (1)- (54) or any embodiment of any compound herein; and wherein each -C=C-, as shown in any one of the structures has a cis configuration. In some embodiments, R55 is:
Figure imgf000063_0002
In one aspect, the ionizable cationic lipid has a structural formula selected from the group consisting of: Compound (121):
Figure imgf000063_0003
(121),
Compound (122):
Figure imgf000063_0004
(122),
Compound (123):
Figure imgf000064_0001
(123),
Compound (124):
Figure imgf000064_0002
(124),
Compound (125):
Figure imgf000064_0004
(126),
Compound (127):
Figure imgf000064_0003
(127), Compound (128):
Figure imgf000065_0001
Me is methyl; and,
R55 is as defined for any one of Compounds (l)-(54) or for any embodiment herein.
In another aspect, the present invention provides for a lipid comprising lipid tail, wherein the tail is one more -R55. In some embodiments, the lipid further comprises a head group and an optional linker. In some embodiments, the head group is an amine containing group. In some further embodiments, the head group is an amine containing ionizable cationic head group.
In some embodiments the ionizable cationic lipid is a non-fusogenic lipid. By a "non- fusogenic lipid" is meant a cationic lipid that can condense and/or encapsulate a payload, e.g., a therapeutic nucleic acid, but has insufficient fusogenic activity to effectively delivery the payload across cellular membranes. In some embodiments, the lipid nanoparticles have mean diameter of 20-75 nm or 30-100 nm.
The pKa', i.e., the apparent pKa, of formulated cationic lipids in particles, can be correlated with the effectiveness of the LNPs for delivery of nucleic acids. The pKa' of a cationic lipid can be determined in lipid nanoparticles, e.g., using an assay based on fluorescence of 2-(p- toluidino)-6-napthalene sulfonic acid (TNS). Lipid nanoparticles in PBS at a concentration of 0.4 mM total lipid are prepared using standard methods. TNS can be prepared as a 100 mM stock solution in distilled water and mixed into buffers of different pH. Vesicles can be diluted to 24 mM lipid in 2 mL of buffered solutions containing, 10 mM HEPES, 10 mM MES, 10 mM ammonium acetate, 130 mM NaCI, where the pH ranges from 2.5 to 11. An aliquot of the TNS solution can be added to give a final concentration of 1 mM and following vortex mixing fluorescence intensity is measured at room temperature in a SLM Aminco Series 2 Luminescence Spectrophotometer using excitation and emission wavelengths of 321 nm and 445 nm. A sigmoidal best fit analysis can be applied to the fluorescence data and the pKa' is measured as the pH giving rise to half-maximal fluorescence intensity.
In one embodiment, as a molar percent of total lipids, the LNP comprises about 30% to about 80%, about 30% to about 70%, about 30% to about 60%, about 30% to about 50%, or about 30% to about 40%, about 40% to about 80%, about 30% to about 70%, about 40% to about 60%, about 40% to about 50%, about 50% to about 80%, about 50% to about 70%, 50% to about 60%, about 60% to about 80%, or about 70% to about 80% (ionizable) cationic lipid.
Sterol
In one embodiment, the lipid particles (e.g., lipid nanoparticles) can further comprise a component, such as a sterol, to provide membrane integrity and stability of the lipid particle. In one embodiment, an exemplary sterol that can be used in the lipid particle is cholesterol, or a derivative thereof. Non-limiting examples of cholesterol derivatives include 5-a-cholestanol (5a-Cholestan-3|3-ol), 5-|3-coprostanol, cholesteryl-(2'-hydroxy)-ethyl ether, cholesteryl-( 4' -hydroxy)-butyl ether, and 6-ketocholestanol, 5a-cholestane, cholestenone, 5-a-cholestanone, 5 p-cholestanone, allocholesterol, epi-allocholesterol and cholesteryl decanoate; and mixtures thereof. In some embodiments, the cholesterol derivative is a polar analogue such as cholesteryl-(4'-hydroxy)-butyl ether. In some embodiments, cholesterol derivative is cholesteryl hemisuccinate. In some embodiments, the sterol is a sea cucumber sulphated sterol, e.g., cholest-5-en-3|3-yl hydrogen sulfate or 24-methylene- cholesterol sulfate (J. Oleo Sci. 71, (3) 401-410 (2022)). In some embodiments, the lipid is fecosterol, sitosterol, campesterol, stigmasterol, brassicasterol, ergosterol, tomatidine, tomatine, ursolic acid or alpha-tocopherol.
Exemplary cholesterol derivatives are described in International Patent Application Publication No. W02009/127060 and U.S. Patent Application Publication No. US2010/0130588.
In one embodiment, the component providing membrane integrity, such as a sterol, can comprise 0-50% (mol) of total lipids present in the lipid particle (e.g., lipid nanoparticle). In some embodiments, such a component is 20-50% (mol) of total lipids present in the lipid particle (e.g., lipid nanoparticle). In some embodiments, such a component is 30-40% (mol) of total lipids present in the lipid particle (e.g., lipid nanoparticle). In some embodiments, such a component is 35-45% (mol) of total lipids present in the lipid particle (e.g., lipid nanoparticle). In some embodiments, such a component is 38-42% (mol) of total lipid present in the lipid particle (e.g., lipid nanoparticle).
According to some embodiments, the LNP comprises a sterol, wherein the sterol is present at a molar percentage of about 20% to about 50%. According to some embodiments, the LNP comprises a sterol, wherein the sterol is present at a molar percentage of about 30% to about 50%. According to some embodiments, the LNP comprises a sterol, wherein the sterol is present at a molar percentage of about 40% to about 50%. According to some embodiments, the LNP comprises a sterol, wherein the sterol is present at a molar percentage of about 20% to about 40%. According to some embodiments, the LNP comprises a sterol, wherein the sterol is present at a molar percentage of about 30% to about 40%. In some embodiments, the LNP comprises more than one structural lipid, e.g., two or more sterols.
Non-cationic lipid
The non-cationic lipid is typically a phospholipid and serves to increase fusogenicity and/or increase stability of the LNP, including during formation. Non-cationic lipids include amphipathic lipids, neutral lipids and anionic lipids. Accordingly, the non-cationic lipid can be a neutral uncharged, zwitterionic, or anionic lipid. Exemplary non-cationic lipids include, but are not limited to, distearoyl-sn-glycerophosphoethanolamine, distearoylphosphatidylcholine (DSPC), dioleylphosphatidylcholine (DOPC), dipalmitoylphosphatidylcholine (DPPC), dioleylphosphatidylglycerol (DOPG), dipalmitoylphosphatidylglycerol (DPPG), dioleyl-phosphatidylethanolamine (DOPE), palmitoyloleylphosphatidylcholine (POPC), palmitoyloleylphosphatidylethanolamine (POPE), dioleyl-phosphatidylethanolamine 4-(N-maleimidomethyl)-cyclohexane-l-carboxylate (DOPE-mal), dipalmitoyl-phosphatidylethanolamine (DPPE), dimyristoylphosphoethanolamine (DMPE), distearoyl-phosphatidylethanolamine (DSPE), monomethylphosphatidylethanolamine (such as 16-O-monomethyl PE), dimethylphosphatidylethanolamine (such as 16-O-dimethyl PE), 18-1-trans PE, l-stearoyl-2-oleyl- phosphatidyethanolamine (SOPE), hydrogenated soy phosphatidylcholine (HSPC), egg phosphatidylcholine (EPC), dioleyl-phosphatidylserine (DOPS), sphingomyelin (SM), 1,2- diundecanoyl-sn-glycero-phosphocholine (DUPC), dimyristoyl-phosphatidylcholine (DMPC), dimyristoyl-phosphatidylglycerol (DMPG), distearoyl-phosphatidylglycerol (DSPG), dierucoyl- phosphatidylcholine (DEPC), palmitoyl-oleyl-phosphatidylglycerol (POPG), dielaidoylphosphatidylethanolamine (DEPE), l,2-dilauroyl-sn-glycero-3-phosphoethanolamine (D LPE); l,2-diphytanoyl-sn-glycero-3-phosphoethanolamine (DPHyPE); 1,2-di-O-octadecenyl- sn-glycero-3-phosphocholine (18:0 Diether PC), l-oleyl-2-cholesterylhemisuccinoyl-sn- glycero-3-phosphocholine (OChemsPC), l-hexadecyl-sn-glycero-3-phosphocholine (C16 Lyso PC), l,2-dilinolenoyl-sn-glycero-3- phosphocholine, 1 ,2-diarachidonoyl-sn-glycero-3- phosphocholine, 1 ,2-didocosahexaenoyl-sn-glycero-3-phosphocholine, 1,2-diphytanoyl-sn- glycero-3-phosphoethanolamine (ME 16.0 PE), l,2-distearoyl-sn-glycero-3- phosphoethanolamine, 1.2-dilinoleyl-sn-glycero-3-phosphoethanolamine, 1,2-dilinolenoyl- sn-glycero-3-phosphoethanolamine, ,2-diarachidonoyl-sn-glycero-3-phosphoethanolamine, l,2-didocosahexaenoyl-sn-glycero-3-phosphoethanolamine, phosphatidylethanolamine, lysolecithin, lysophosphatidylethanolamine, phosphatidylserine, phosphatidylinositol, sphingomyelin (SM), egg sphingomyelin (ESM), dihydrosphingomyelin, cephalin, cardiolipin, phosphatidicacid, cerebrosides, dicetylphosphate, lysophosphatidylcholine, dilinoleyl- phosphatidylcholine, or mixtures thereof. It is to be understood that other diacylphosphatidylcholine and diacylphosphatidylethanolamine phospholipids can also be used. The acyl groups in these lipids are preferably acyl groups derived from fatty acids having C10-C24 carbon chains, e.g., lauroyl, myristoyl, palmitoyl, stearoyl, or oleyl. Preferred helper lipid: DSPC, DPPC, DMPC, DOPC, POPC, DOPE, and SM.
In some embodiments, the non-cationic lipid can comprise 0-20% (mol) of the total lipid present in the lipid nanoparticle. In some embodiments, the non-cationic lipid comprises 0.5-15% (mol) of the total lipid present in the lipid particle (e.g., lipid nanoparticle). In some embodiments, the non-cationic lipid comprises 5-12% (mol) or 5-10% (mol) of the total lipid present in the lipid particle (e.g., lipid nanoparticle). In some embodiments, the non-cationic lipid comprises about 6% (mol), about 7.0% (mol), about 7.5% (mol), about 8.0% (mol), about 9.0% (mol), about 10% (mol), or about 11 % (mol) of the total lipid present in the lipid particle (e.g., lipid nanoparticle).
Exemplary non-cationic lipids are described in International Patent Application Publication No. WO2017/099823 and US Patent Application Publication No. US2018/0028664, the contents of both of which are incorporated herein by reference in their entirety.
According to some embodiments, the LNP comprises a non-cationic lipid, wherein the noncationic lipid is present at a molar percentage of about 2 % to about 20%. According to some embodiments, the LNP comprises a non-cationic lipid, wherein the non-cationic lipid is present at a molar percentage of about 5% to about 20%, about 10% to about 20%, about 15% to about 20%, about 10% to about 20%, or about 10% to about 15%.
Polymer conjugated lipids
In one embodiment, the lipid particle (e.g., lipid nanoparticle) can further comprise a polymer conjugated lipid molecule, e.g., polyethylene glycol (PEG)-conjugated ("PEGylated") lipid. Generally, these are used to inhibit aggregation of lipid particle (e.g., lipid nanoparticle) and/or provide steric stabilization. Exemplary conjugated lipids include, but are not limited to, PEG-lipid conjugates, polyoxazoline (POZ)-lipid conjugates, polyamide - lipid conjugates (such as ATTA-lipid conjugates), cationic-polymer lipid (CPL) conjugates, and mixtures thereof. In some embodiments, the conjugated lipid molecule is a PEGylated lipid, for example, a (methoxy polyethylene glycol)-conjugated lipid. In some other embodiments, the PEGylated lipid is PEG2000-DMG (dimyristoylglycerol). Exemplary PEGylated lipids include, but are not limited to, PEG-diacylglycerol (DAG) (such as l-(monomethoxy- polyethyleneglycol)-2,3-dimyristoylglycerol (PEG-DMG)), PEG-dialkyloxypropyl (DAA), PEG- phospholipid, PEG-ceramide (Cer), a pegylated phosphatidylethanolamine (PEG-PE), PEG succinate diacylglycerol (PEGS-DAG) l,2-dimyristoyl-rac-glycero-3-methoxypolyethylene glycol-2000 (DMG-PEG 2000), R-3-[((jo-methoxy polyethylene glycol)2000)carbamoyl)]-l,2- dimyristyloxy-propyl-3-amine (PEG-c-DOMG), l,2-Dimyristoyl-sn-Glycero-3-
Phosphoethanolamine (DMPE) conjugated Polyethylene Glycol, l,2-Dilauroyl-sn-Glycero-3- Phosphoethanolamine (DLPE) conjugated Polyethylene Glycol, dipalmitoyl phosphatidylcholine (DPPC) conjugated Polyethylene Glycol, 1, 2-Distearoyl-sn-glycero-3- phosphoethanolamine-Poly(ethylene glycol) (DSPE-PEG), PEG dialkoxypropylcarbamate, N- (carbonyl-methoxypolyethylene glycol 2000)-l,2-distearoyl-sn-glycero-3- phosphoethanolamine sodium salt,
Figure imgf000069_0001
or a mixture thereof. PEG mono-fatty acid esters may have hydroxy on the PEG terminus opposite to the hydrophobic lipid end, as in
Figure imgf000069_0002
A polymer conjugated lipid may be used in a LNP formulation, alone or in mixtures with other surface lipids.
Additional exemplary PEG-lipid conjugates are described, for example, in US patents US5885613, US6287591, US8936942B2 and US patent applications US2003/0077829, US2003/0077829, US2005/0175682, US2008/0020058, US2011/0117125, US2010/0130588, US2016/0376224, and US2017/0119904, the contents of all of which are incorporated herein by reference in their entireties.
In some embodiments, the PEG-DAA PEGylated lipid is, for example, PEG- dilauroyloxypropyl, PEG- dimyristyoloxypropyl, PEG-dipalmitoyloxypropyl, or PEG- distearoyloxypropyl. The PEG-lipid can be, e.g., one or more of PEG-DMG, PEG- dilauroylglycerol, PEG-dipalmitoylglycerol, PEG-disteroylglycerol, PEG-dilauroylglycamide, PEG-dimyristoylglycamide, PEG-dipalmitoylglycamide, PEG- disteroylglycamide, PEG- cholesterol (l-[8'-( Cholest-5-en-3 [beta]-oxy)carboxamido-3',6'-dioxaoctanyl] carbamoyl- [omega]-methyl-poly(ethylene glycol), PEG-DMB (3,4-Ditetradecoxylbenzyl-[omega]-methyl- poly(ethylene glycol) ether), or 1,2-dimyristoyl-sn-glycero- 3-phosphoethanolamine-N- [methoxy(polyethylene glycol)-2000] . In some embodiments, the PEG-lipid is selected from PEG-DMG and/or l,2-dimyristoyl-sn-glycero-3- phosphoethanolamine-N- [methoxy(polyethylene glycol)-2000]. In some embodiments, the PEGylated lipid is selected from one or more of: N-(Carbonyl- methoxypolyethyleneglycoln)-l,2-dimyristoyl-sn-glycero-3 -phosphoethanolamine (DMPE- PEGn, where n (representing PEG average molecular weight) is 350, 500, 750, 1000 or 2000); N-(Carbonyl-methoxypolyethyleneglycoln)-l,2-distearoyl-sn-glycero-3- phosphoethanolamine (DSPE-PEGn, where n is 350, 500, 750, 1000, 2000, or 5000); DSPE- polyglycerin-cyclohexyl-carboxylic acid, DSPE-polyglycelin-2-methylglutaric acid; 1,2- Distearoyl-sn-Glycero-3-Phosphoethanolamine (DSPE) conjugated Polyethylene Glycol (DSPE-PEG-OH); polyethylene glycol-dimyristolglycerol (PEG-DMG); polyethylene glycoldistearoyl glycerol (PEG-DSG); or N-octanoyl-sphingosine-l-{succinyl[methoxy(polyethylene glycol)2000 (Cs PEG2000 Ceramide). In some examples of DMPE-PEGn, where n is 350, 500, 750, 1000 or 2000, the PEG-lipid is N-(Carbonyl-methoxypolyethyleneglycol 2000)-l,2- dimyristoyl-sn-glycero-3-phosphoethanolamine (DMPE-PEG 2,000). In some examples of DSPE-PEGn, where n (representing PEG average molecular weight) is 350, 500, 750, 1000 2000, or 5000 the PEG-lipid is N-(Carbonylmethoxypolyethyleneglycol 2000)-l,2-distearoyl- sn-glycero-3-phosphoethanolamine (DSPE-PEG 2000). In some embodiments, the PEGylated lipid is DSPE-PEG-OH. In some embodiments, the PEGylated lipid is DSPE-PEG-azide. In some embodiments, the PEGylated lipid is PEG-DMG. In some embodiments, the PEGylated lipid is PEG-DSG. In some embodiments, the conjugated lipid, e.g., PEGylated lipid, includes a tissue-specific ligand, e.g., first or second ligand. For example, DSPE-PEG conjugated with a GalNAc ligand, DSGPEG conjugated with a GalNAc ligand. In one embodiment, lipids conjugated with a molecule other than a PEG can also be used in place of PEG-lipid. For example, polyoxazoline (POZ)-lipid conjugates, polyamide-lipid conjugates, such as ATTA- lipid conjugates and polysarcosine (PSAR) lipid conjugates, and cationic polymer lipid (CPL) conjugates, such as with chitosan free amine oligomers, can be used in place of or in addition to the PEG-lipid. Exemplary conjugated lipids, i.e., PEG-lipids, POZ-lipid conjugates, ATTA-lipid conjugates and cationic polymer-lipids are described in the International Patent Application Publication Nos. WO 1996/010392, WO1998/051278, W02002/087541, W02005/026372, WO2008/147438, W02009/086558, W02012/000104, WO2017/117528, WO2017/099823, WO2015/199952, W02017/004143, WO2015/095346, W02012/000104, W02012/000104, and W02010/006282, U.S. Patent Application Publication Nos.
US2003/0077829, US2005/0175682, US2008/0020058, US2011/0117125, US2013/0303587, US2018/0028664, US2015/0376115, US2016/0376224, US2016/0317458, US2013/0303587, US2013/0303587, and US20110123453, and U.S. Patent Nos. US5,885,613, US6,287,591, US6, 320,017, and US6,586,559.
According to some embodiments, the LNP comprises at least one PEGylated lipid, wherein the PEGylated lipid is present at a molar percentage of about 0.5% to about 20% of the total lipid present in the lipid nanoparticle. In some embodiments, the LNP comprises at least one PEGylated lipid, wherein the PEGylated lipid is present at a molar percentage of about 2.1% to about 10% of the total lipid present in the lipid nanoparticle. In some embodiments, the LNP comprises about 1-5% (mol), about 2-4% (mol), about 2-3% (mol), about 1-3% (mol), about 0.75-2.5% (mol), about 0.75-2.0% (mol), about 0.75-1.8% (mol), about 1-2% (mol), about 0.75-1.5% (mol), about 1- 1.8% (mol), about 1-1.5% (mol), about 1-1.3% (mol), about 1-1.2% (mol), about 0.75-1.5% (mol), about 0.75-1.25% (mol), about 1.5-1.8% (mol), about 1.2-1.5% (mol), about 2% (mol), about 2.5% (mol), about 3% (mol), about 3.5% (mol) or about 4% (mol) of PEGylated lipid, based on the total lipid present in the lipid nanoparticle. In some embodiments, the LNP comprises at least one PEGylated lipid, wherein the PEGylated lipid is present at a molar percentage of about 5% to about 10%, about 7% to about 10%, about 2.1 % to about 8%, about 2.1 % to about 5%, about 5% to about 8%, about 1 % to about 2%. about 1.2% to about 2%. about 1.5% to about 2%. about 1.75% to about 2%, about 1 % to about 1.5%, about 1.25% to about 1.5%, or about 1.5% to about 1.75%.
In some embodiments, the lipid particles (e.g., lipid nanoparticles) is conjugated with other moieties to prevent aggregation. Such lipid conjugates include, but are not limited to, PEG- lipid conjugates such as, e.g., PEG coupled to dialkyloxypropyls (e.g., PEG-DAA conjugates), PEG coupled to diacylglycerols (e.g., PEG-DAG conjugates), PEG coupled to cholesterol, PEG coupled to phosphatidylethanolamines, and PEG conjugated to ceramides (see, e.g., U.S. Patent No. 5,885,613), cationic PEG lipids, polyoxazoline (POZ)-lipid conjugates (e.g., POZ- DAA conjugates; see, e.g., U.S. Provisional Application No. 61/294,828, filed Jan. 13, 2010, and U.S. Provisional Application No. 61/295,140, filed Jan. 14, 2010), polyamide oligomers (e.g., ATTA-lipid conjugates), and/or mixtures thereof. Additional examples of POZ-lipid conjugates are described in PCT Publication No. WO 2010/006282. PEG, PSAR or POZ can be conjugated directly to the lipid or may be linked to the lipid via a linker moiety. Any linker moiety suitable for coupling the PEG or the POZ to a lipid can be used including, e.g., nonester containing linker moieties and ester-containing linker moieties. In certain preferred embodiments, non-ester containing linker moieties, such as amides or carbamates, are used. The disclosures of each of the above patent documents are herein incorporated by reference in their entirety for all purposes.
Additional Lipids
Lipid nanoparticles, including those of the present invention, may comprise one or more additional lipid(s), including triglycerides, fatty acids, carotenoids (including carotenes and xanthophylls), other classes of lipids, and derivatives (including conjugates) thereof. Note that some of these additional lipids may also fall into one of the categories above.
Additional lipids may be present to improve stability, fusogenicity, endosomal escape, tolerability/safety, efficacy, tissue tropism/targeting, etc.
One aspect of the invention provides for a lipid particle, e.g. LNP, comprising an additional lipid. These lipid particles include but are not limited to lipid particles comprising an ionizable cationic lipid of the present invention.
In some embodiments, the lipid particle comprises a lipid selected from: Compound (55A)
Figure imgf000072_0001
or
Compound (55B)
Figure imgf000072_0002
(55B).
In another embodiment, the lipid particle comprises a carotene or xanthophyll. In a further embodiment, the lipid particle comprises a xanthophyll selected from astaxanthin, zeaxanthin, lutein, cryptoxanthin, antheraxanthin, fucoxanthin, neoxanthin or violaxanthin.
The chemical structure:
Figure imgf000072_0003
is free astaxanthin with the S,S'-stereochemistry of the secondary alcohol carbons.
The xanthophyll may be esterified or unesterified, present as a single isomer, a racemate or mixture of isomers. Double bonds may be in an E or Z configuration. Astaxanthin, e.g., can exist as a (3S, 3'S), (3S, 3'R) or (3R, 3'R) isomer. In another embodiment, said compound is selected from any one of Compounds (56) - (70):
Figure imgf000073_0001
Figure imgf000074_0001
: wherein A9, A10 and All are each independently selected from: R103 or -H; wherein at least one of A9, A10 or All is R103; and wherein R103, for each occurrence, is independently selected from:
Figure imgf000075_0001
Figure imgf000076_0001
In another embodiment, said compound is selected from any one of Compounds (71)-(120):
Figure imgf000077_0001
Figure imgf000078_0001
10 (81),
Figure imgf000078_0002
(82),
Figure imgf000079_0001
(83),
Figure imgf000079_0002
(84),
Figure imgf000079_0004
(86),
Figure imgf000079_0003
(87),
Figure imgf000080_0001
(88),
Figure imgf000080_0002
(89),
Figure imgf000080_0004
(91),
Figure imgf000080_0003
(92),
Figure imgf000081_0001
Figure imgf000082_0001
10 (102),
Figure imgf000083_0001
10 (107),
Figure imgf000084_0001
Figure imgf000085_0001
(119), or
Figure imgf000086_0001
(120).
Therapeutic Nucleic Acids (TNA)
The lipid particles of the present invention comprise a payload/cargo. In one aspect the payload is therapeutic nucleic acid (TNA). The length of the TNA can vary and include nucleic acid of 5-50,000 nucleotides in length. The nucleic acid can be in any form, including singlestranded DNA, single-stranded RNA, double-stranded DNA or double-stranded RNA, or hybrids thereof. The nucleotides may be modified, unmodified or a combination thereof. The TNA may be chemically synthesized. Synthesis of mRNA includes in vitro transcription. In some embodiments, the TNA is selected from the group consisting of minigenes, plasmids, minicircles, antisense oligonucleotides (ASO), closed-ended (ceDNA), ministring, doggybone, protelomere closed ended DNA, dumbbell linear DNA, asymmetrical interfering RNA (aiRNA), microRNA (miRNA), messenger RNA (mRNA), small interfering RNA (siRNA), small activating RNA (saRNA), self-amplifying RNA (SAM), ribozymes, dicer substrate dsRNA, small hairpin RNA (shRNA), tRNA, tRNA derived RNA fragments (tRFs), rRNA, piwi-interacting RNA (piRNA), guide RNA (gRNA), DNA viral vectors, viral RNA vector, non-viral vector and any combination thereof.
The payload of an LNP may include more than one TNA. For example, in the case of Cas9- CRISPR, a guide RNA (gRNA), together with a plasmid or mRNA encoding the Cas9 protein may be incorporated into a single LNP. Alternatively, LNPs containing different TNAs may be combined into a single formulation.
According to some embodiments, the LNP has a total lipid to TNA ratio of about 10: 1 to about 40:1, for example 10:1 to 30:1 or 10:1 to 20:1 or 10:1 to 15:1.
A preferred payload of an LNP of the present invention is an mRNA. An mRNA of the invention may include a nucleic acid sequence encoding a polypeptide of interest (e.g., a coding region), a first flanking region located at the 5'-terminus of the first region (e.g., a 5'- UTR), a second flanking region located at the 3'-terminus of the first region (e.g., a 3'-UTR), at least one 5'-cap region, and a 3 '-stabilizing region. In some embodiments, a messenger RNA further includes a poly-A region or a Kozak sequence (e.g., in the 5'-UTR). In some cases, messenger RNAs may contain one or more intronic sequences capable of being excised from the messenger RNA. In some embodiments, a messenger RNA may include a 5' cap structure, a chain terminating nucleotide, a stem loop, a poly A sequence, and/or a polyadenylation signal. Any one of the regions of a messenger RNA may include one or more alternative components (e.g., an alternative nucleoside). For example, the 3'- stabilizing region may contain an alternative nucleoside such as an L-nucleoside, an inverted thymidine, or a 2'-O-methyl nucleoside and/or the coding region, 5'-UTR, 3'- UTR, or cap region may include an alternative nucleoside such as a 5 -substituted uridine (e.g., 5- methoxy uridine), a 1-substituted pseudouridine (e.g., 1-methyl-pseudouridine or 1-ethyl- pseudouridine), and/or a 5-substituted cytidine (e.g., 5-methyl-cytidine).
An mRNA of the invention may include an internal ribosome entry site (IRES). An IRES may act as a sole ribosome binding site, or as one of multiple ribosome binding sites. A messenger RNA containing more than one functional ribosome binding site may encode several peptides or polypeptides that are translated independently by the ribosomes. Suitable IRES sequences that may be useful include those from picomaviruses (e.g. FMDV), pest viruses (CFFV), polio viruses (PV), encephalomyocarditis viruses (ECMV), foot-and- mouth disease viruses (FMDV), hepatitis C viruses (HCV), classical swine fever viruses (CSFV), murine leukemia virus (MEV), simian immune deficiency viruses (S1V) and cricket paralysis viruses (CrPV).
An mRNA of the invention may comprise a first region of linked nucleosides encoding an antigenic polypeptide, a first flanking region located at the 5'-terminus of the first region (e.g., a 5'-UTR), a second flanking region located at the 3'-terminus of the first region (e.g., a 3'-UTR), at least one 5'-cap region, and a 3'- stabilizing region.
Messenger RNA nucleotides may be naturally or non-naturally occurring. The 5'-UTR, (b) the open reading frame (ORF), (c) the 3'-UTR, (d) the poly A tail, and any combination of (a, b, c, or d above) comprise naturally occurring canonical nucleotides A (adenosine), G (guanosine), C (cytosine), U (uridine), or T (thymidine). In some embodiments, the nucleobase is an alternative uracil. Exemplary nucleobases and nucleosides having an alternative uracil include pseudouridine (psi), pyridin-4-one ribonucleoside, 5-aza-uracil, 6- aza-uracil, 2-thio-5-aza-uracil, 2-thio-u raci I (s2U), 4-thio-uracil (s4U), 4-thio-pseudouridine, 2-thio-pseudouridine, 5- hydroxy-uracil, 5-aminoallyl-uracil, 5-halo-uracil (e.g., 5-iodo-uracil or 5-bromo-uracil), 3-methyl-uracil (m3U), 5-methoxy-uracil (mo5U), uracil 5-oxyacetic acid (cmo5U), uracil 5-oxyacetic acid methyl ester (mcmo5U), 5-carboxymethyl-uracil (cm5U), 1 - carboxymethyl-pseudouridine, 5-carboxyhydroxymethyl-uracil (chm5U), 5- carboxyhydroxymethyl-uracil methyl ester (mchm5U), 5-methoxycarbonylmethyl-uracil (mcm5U), 5-methoxycarbonylmethyl-2-thio-uracil (mcm5s2U), 5-aminomethyl- 2-thio-uracil (nm5s2U), 5-methylaminomethyl-uracil (mnm5U), 5-methylaminomethyl- 2-thio-uracil (mnm5s2U), 5-methylaminomethyl-2-seleno-uracil (mnm5se2U), 5-carbamoylmethyl-uracil (ncm5U), 5-carboxymethylaminomethyl-uracil (cmnm5U), 5- carboxymethylaminomethyl-2- thio-uracil (cmnm5s2U), 5-propynyl-uracil, 1-propynyl-pseudouracil, 5-taurinomethyl-uracil (rm5U), 1-taurinomethyl-pseudouridine, 5-taurinomethyl-2-thio-uracil (rm5s2U), 1- taurinomethyl-4-thio-pseudouridine, 5-methyl-uracil (m5U, i.e., having the nucleobase deoxythymine), 1-methyl-pseudouridine, 1-ethyl-pseudouridine (Etly), 5-methyl-2-thio- uracil (m5s2U), 1 -methyl-4-thio-pseudouridine, 4-thio- 1-methyl-pseudouridine, 3-methyl- pseudouridine, 2-thio- 1-methyl-pseudouridine, 1 methyl- 1-deaza-pseudouridine, 2-thio- 1- methyl- 1-deaza-pseudouridine, dihydrouracil (D), dihydropseudouridine, 5,6-dihydrouracil, 5- methyl-dihydrouracil (m5D), 2-thio-dihydrouracil, 2-thio-dihydropseudouridine, 2- methoxy-uracil, 2-methoxy-4-thio-uracil, 4-methoxy-pseudouridine, 4-methoxy-2-thio- pseudouridine, Nl-methyl-pseudouridine, 3-(3-amino-3-carboxypropyl)uracil (acp3U), 1- methyl-3-(3-amino-3-carboxypropyl)pseudouridine, 5-(isopentenylaminomethyl)uracil (inm5U), 5-(isopentenylaminomethyl)-2-thio-uracil (inm5s2U), 5,2'-O-dimethyl-uridine (m5Um), 2-thio-2'-O_methyl-uridine (s2Um), 5- methoxycarbonylmethyl-2'-O-methyl- uridine (mcm5Um), 5-carbamoylmethyl-2'-O- methyl-uridine (ncm5Um), 5- carboxymethylaminomethyl-2'-O-methyl-uridine (cmnm5Um), 3, 2'-O-dimethyl- uridine (m3Um), and 5-(isopentenylaminomethyl)-2'-O-methyl-uridine (inm5Um), 1-thio-uracil, deoxythymidine, 5-(2-carbomethoxyvinyl)-uracil, 5-(carbamoylhydroxymethyl)-uracil, 5- carbamoylmethyl-2-thio-uracil, 5- carboxymethyl-2-thio-uracil, 5-cyanomethyl-uracil, 5- methoxy-2-thio-uracil, and 5-[3-(l-E-propenylamino)]uracil. In one embodiment, the modified uracil is pseudouridine. In one embodiment, the modified uracil is Nl-methyl- pseudouridine.
In some embodiments, the mRNA one or more alternative components which impart useful properties including increased stability and/or the lack of a substantial induction of the innate immune response of a cell into which the poly messenger RNA is introduced. For example, an alternative messenger RNA exhibits reduced degradation in a cell into which the messenger RNA is introduced, relative to a corresponding unaltered messenger RNA. These alternative species may enhance the efficiency of protein production, intracellular retention of the messenger RNA, and/or viability of contacted cells, as well as possess reduced immunogenicity.
In some embodiments, the nucleobase is an alternative cytosine. Exemplary nucleobases and nucleosides having an alternative cytosine include 5-aza-cytosine, 6-aza-cytosine, pseudoisocytidine, 3-methyl-cytosine (m3C), N4-acetyl-cytosine (ac4C), 5-formyl-cytosine (f5C), N4-methyl-cytosine (m4C), 5-methyl-cytosine (m5C), 5-halo-cytosine (e.g., 5-iodo- cytosine), 5-hydroxymethyl-cytosine (hm5C), 1-methyl-pseudoisocytidine, pyrrolo-cytosine, pyrrolo-pseudoisocytidine, 2-thio-cytosine (s2C), 2-thio-5-methyl-cytosine, 4-thio- pseudoisocytidine, 4-thio-l-methyl-pseudoisocytidine, 4-thio-l-methyl-l-deaza-pseudo isocytidine, 1-methyl- 1-deaza-pseudoisocytidine, zebularine, 5-aza-zebularine, 5-methyl- zebularine, 5-aza-2-thio-zebularine, 2-thio-zebularine, 2-methoxy-cytosine, 2-methoxy-5- methyl-cytosine, 4-methoxy-pseudoisocytidine, 4-methoxy-l-methyl-pseudoisocytidine, lysidine (k2C), 5,2'-O- dimethyl-cytidine (m5Cm), N4-acetyl-2'-O-methyl-cytidine (ac4Cm), N4,2'-O- dimethyl-cytidine (m4Cm), 5-formyl-2'-O-methyl-cytidine (f5Cm), N4,N4,2'-O- trimethyl-cytidine (m42Cm), 1 -thio-cytosine, 5-hydroxy-cytosine, 5-(3-azidopropyl)- cytosine, and 5-(2-azidoethyl)-cytosine. In one example, the modified cytosine is 5-methyl- cytosine. [0075] In some embodiments, the nucleobase is an alternative adenine. Exemplary nucleobases and nucleosides having an alternative adenine include 2-amino-purine, 2,6- diaminopurine, 2-amino-6-halo-purine (e.g., 2-amino-6-chloro-purine), 6-halo-purine (e.g., 6-chloro-purine), 2-amino-6-methyl-purine, 8-azido-adenine, 7-deaza-adenine, 7-deaza-8- aza-adenine, 7-deaza-2-amino-purine, 7-deaza-8-aza-2-amino-purine, 7-deaza-2,6- diaminopurine, 7-deaza-8-aza-2,6-diaminopurine, 1-methyl-adenine (mlA), 2-methyl- adenine (m2A), N6-methyl-adenine (m6A), 2-methylthio-N6-methyl-adenine (ms2m6A), N6- isopentenyl-adenine (i6A), 2-methylthio-N6-isopentenyl-adenine (ms2i6A), N6-(cis- hydroxyisopentenyl)adenine (io6A), 2-methylthio-N6-(cis-hydroxyisopentenyl)adenine (ms2io6A), N6-glycinylcarbamoyl-adenine (g6A), N6-threonylcarbamoyl-adenine (t6A), N6- methyl-N6-threonylcarbamoyl-adenine (m6t6A), 2-methylthio-N6-threonylcarbamoyl- adenine (ms2g6A), N6,N6-dimethyl-adenine (m62A), N6-hydroxynorvalylcarbamoyl-adenine (hn6A), 2-methylthio-N6-hydroxynorvalylcarbamoyl-adenine (ms2hn6A), N6-acetyl- adenine (ac6A), 7-methyl- adenine, 2-methylthio-adenine, 2-methoxy-adenine, N6,2'-O-dimethyl- adenosine (m6Am), N6,N6,2'-O-trimethyl-adenosine (m62Am), l,2'-O-dimethyl-adenosine (mlAm), 2-amino-N6-methyl-purine, 1-thio-adenine, 8-azido-adenine, N6-(19-amino- pentaoxanonadecyl)-adenine, 2,8-dimethyl-adenine, N6-formyl-adenine, and N6-hydroxy methyl-adenine.
In some embodiments, the nucleobase is an alternative guanine. Exemplary nucleobases and nucleosides having an alternative guanine include inosine (I), 1-methyl-inosine (mil), wyosine (imG), methylwyosine (mimG), 4-demethyl-wyosine (imG-14), iso wyo sine (imG2), wybutosine (yW), peroxywybutosine (o2yW), hydroxywybutosine (OHyW), undermodified hydroxywybutosine (OHyW*), 7-deaza-guanine, queuosine (Q), epoxyqueuosine (oQ), galacto syl-queuo sine (galQ), manno syl-queuo sine (manQ), 7-cyano-7-deaza-guanine (preQO), 7-aminomethyl-7-deaza-guanine (preQi), archaeosine (G+), 7-deaza-8-aza-guanine, 6-thio-guanine, 6-thio-7-deaza-guanine, 6-thio-7-deaza-8-aza-guanine, 7-methyl-guanine (m7G), 6- thio-7-methyl-guanine, 7-methyl-inosine, 6-methoxy-guanine, 1-methyl-guanine (mIG), N2-methyl-guanine (m2G), N2,N2-dimethyl-guanine (m22G), N2,7-dimethyl-guanine (m2,7G), N2, N2,7- dimethyl-guanine (m2,2,7G), 8-oxo-guanine, 7-methyl-8-oxo-guanine, 1- methyl-6-thio-guanine, N2-methyl-6-thio-guanine, N2,N2-dimethyl-6-thio-guanine, N2- methyl-2'-O-methyl-guanosine (m2Gm), N2,N2-dimethyl-2'-O-methyl-guanosine (m22Gm), 1- methyl-2'-O-methyl-guanosine (mIGm), N2,N7-dimethyl-2'-O-methyl-guanosine (m2,7Gm), 2'-O-methyl-inosine (Im), l,2'-O-dimethyl-inosine (mllm), 1-thio-guanine, and O- 6-methyl-guanine.
The alternative nucleobase of a nucleotide can be independently a purine, a pyrimidine, a purine or pyrimidine analog. For example, the nucleobase can be an alternative to adenine, cytosine, guanine, uracil, or hypoxanthine. In another embodiment, the nucleobase can also include, for example, naturally-occurring and synthetic derivatives of a base, including pyrazolo[3,4-d]pyrimidines, 5-methylcytosine (5-me-C), 5-hydroxymethyl cytosine, xanthine, hypoxanthine, 2-aminoadenine, 6-methyl and other alkyl derivatives of adenine and guanine, 2-propyl and other alkyl derivatives of adenine and guanine, 2-thiou raci I, 2- thiothymine and 2-thiocytosine, 5-propynyl uracil and cytosine, 6-azo uracil, cytosine and thymine, 5-uracil (pseudo uracil), 4-thiouracil, 8-halo (e.g., 8-bromo), 8-amino, 8-thiol, 8- thioalkyl, 8-hydroxy and other 8-substituted adenines and guanines, 5-halo particularly 5- bromo, 5-trifiuoromethyl and other 5-substituted uracils and cytosines, 7-methylguanine and 7-methyladenine, 8- azaguanine and 8-azaadenine, deazaguanine, 7-deazaguanine, 3- deazaguanine, deazaadenine, 7-deazaadenine, 3-deazaadenine, pyrazolo[3,4-d]pyrimidine, imidazo[l,5-a] 1,3,5 triazinones, 9-deazapurines, imidazo[4,5-d]pyrazines, thiazolo[4,5- d]pyrimidines, pyrazin-2-ones, 1,2,4-triazine, pyridazine; or 1,3,5 triazine.
An mRNA of the invention may be prepared according to any available technique known in the art. Messenger RNA may be prepared by, for example, enzymatic synthesis which provides a process of template-directed synthesis of RNA molecules from an engineered DNA template comprised of an upstream bacteriophage promoter sequence linked to a downstream sequence encoding the gene of interest. Template DNA can be prepared for in vitro transcription from several sources with appropriate techniques which are well known in the art including, but not limited to, plasmid DNA and polymerase chain reaction amplification.
Transcription of the RNA occurs in vitro using the appropriate linearized DNA template in the presence of the corresponding RNA polymerase and adenosine, guanosine, uridine and cytidine ribonucleoside triphosphates (rNTPs) under conditions that support polymerase activity while minimizing potential degradation of the resultant mRNA transcripts. In vitro transcription can be performed using a variety of commercially available kits including, but not limited to RiboMax Large Scale RNA Production System (Promega), MegaScript Transcription kits (Life Technologies) as well as with commercially available reagents including RNA polymerases and rNTPs. The methodology for in vitro transcription of mRNA is well-known in the art.
The desired in vitro transcribed messenger RNA is then purified from the undesired components of the transcription or associated reactions. Techniques for the isolation of the messenger RNA transcripts are well known in the art and include phenol/chloroform extraction or precipitation with either alcohol in the presence of monovalent cations or lithium chloride.
In some embodiments, the messenger RNA associated with the LNP is a self-amplifying messenger RNA (SAM) molecule. In certain embodiments, the SAM is derived from or based on an alphavirus. Such SAM molecules are known in the art and can be produced using replication elements derived from, for example, alphaviruses substituting the structural viral proteins with a nucleotide sequence encoding a protein of interest. The target cell to which the SAM is delivered generates an exponential increase of encoded gene products, such as proteins or antigens, which can accumulate in the cells or be secreted therefrom. The SAM may contain one or more genes selected from the group consisting of viral replicases, viral proteases, viral helicases and other nonstructural viral proteins, and may also comprise 5'- and 3'-end cis-active replication sequences and, optionally, a heterologous sequence that encodes a desired amino acid sequence. A subgenomic promoter that directs expression of the heterologous sequence may be present. In embodiments, the heterologous sequence may be fused in frame to other coding regions in the SAM and/or be under the control of an internal ribosome entry site (IRES).
In some embodiments, the RNA associated with the LNP is a small activating RNA (saRNA), which is a TNA that causes enhancement of endogenous messenger RNA transcription. The saRNA molecules are small double-stranded nucleic acids (See e.g., Voutila et al. Mol Ther. 2017 Dec 6;25(12):2705-2714.
The RNA of the invention may encode one or more polypeptide antigens that contain a range of epitopes such as epitopes capable of eliciting either a helper T-cell response or a cytotoxic T-cell response, or both. In some embodiments, the RNA may be engineered to express multiple nucleotide sequences, from two or more open reading frames, thereby allowing co- expression of proteins, such as two or more antigens together with cytokines or other immunomodulators, which can enhance the generation of an immune response. Such a SAM molecule might be useful in the simultaneous production of various proteins as a bivalent or multivalent vaccine.
The RNA, including mRNA and self-amplifying RNA may be prepared using any suitable method known in the art. An RNA molecule that contains modified nucleotides can be prepared by transcribing a DNA that encodes the RNA molecule using a suitable DNA- dependent RNA polymerase, such as T7 phage RNA polymerase, SP6 phage RNA polymerase, T3 phage RNA polymerase, and the like, or mutants of the polymerases which allow efficient incorporation of modified nucleotides into RNA. The incorporation of nucleotide analogs into an RNA may be employed to alter the stability of such RNA molecule, to increase resistance against RNases, to establish replication after introduction into appropriate host cells ("infectivity" of the RNA), and/or to induce or reduce innate and adaptive immune responses.
Process of making lipid nanoparticles
LNPs of the invention can be made using approaches which are well-known in the art of formulation. For example, suitable LNPs can be formed using mixing processes such as microfluidics, including herringbone micromixing, and T-junction mixing of two fluid streams, one of which contains an RNA, e.g., mRNA, typically in an aqueous solution, and the other of which has the various required lipid components, typically in ethanol.
Lipid particles (e.g., lipid nanoparticles) can form spontaneously upon mixing of the nucleic acid, e.g., mRNA, and the lipid(s). Depending on the desired particle size distribution, the resultant nanoparticle mixture can be extruded through a membrane (e.g., 100 nm cut-off) using, for example, a thermobarrel extruder, such as Lipex Extruder (Northern Lipids, Inc). In some cases, the extrusion step can be omitted. Ethanol removal and simultaneous buffer exchange can be accomplished by, for example, dialysis or tangential flow filtration. In one embodiment, the lipid nanoparticles are formed as described in Example 3 described in U.S. Provisional Application No. 63/194,620.
Methods for preparing LNPs are disclosed, e.g., in W02022/261101 WO2019051289, US2013/0037977, US2010/0015218, US2013/0156845, US2013/0164400, US2012/0225129, US2010/0130588, US2007/0042031, US2004/0142025, Kulkarni et al., 2018, ACS Nano, 12:4787 and Kulkarni et al., 2017, Nanoscale, 36: 133347, the content of each of which is incorporated herein by reference in its entirety. In some embodiments, lipid particles (e.g., lipid nanoparticles) can be prepared using a continuous mixing method, a direct dilution process, or an in-line dilution process. The processes and apparatuses for apparatuses for preparing lipid nanoparticles using direct dilution and in-line dilution processes are described, e.g., in US2007/0042031. The processes and apparatuses for preparing lipid nanoparticles using stepwise dilution processes are described in US2004/0142025.
According to some embodiments, the disclosure provides for an LNP comprising a TNA and an ionizable lipid. For example, a lipid nanoparticle formulation that is made and loaded with a TNA is disclosed in WO2019051289. In one embodiment, the lipid particles (e.g., lipid nanoparticles) can be prepared by an impinging jet process (see e.g., W02022/261101). According to some embodiments, the TNA is encapsulated in the lipid(s) thereby protecting it from degradation by a nuclease, e.g., in an aqueous solution. In one embodiment, the TNA in the lipid nanoparticle is not substantially degraded after exposure of the lipid particle to a nuclease at 37°C. for at least about 20, 30, 45, or 60 minutes.
The efficiency of encapsulation of the TNA, e.g., mRNA, within the LNPs may be at least 50%, for example about 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99%. In some embodiments, the encapsulation efficiency may be at least 80%. In certain embodiments, the encapsulation efficiency may be at least 90%.
Encapsulation of TNA in the lipid nanoparticles can be determined by performing a membrane-impermeable fluorescent dye exclusion assay, which uses a dye that has enhanced fluorescence when associated with nucleic acid, for example, an OLIGREEN® assay or PICOGREEN® assay. Generally, encapsulation is determined by adding the dye to the lipid particle formulation, measuring the resulting fluorescence, and comparing it to the fluorescence observed upon addition of a small amount of non-ionic detergent. Detergent mediated disruption of the lipid bilayer releases the encapsulated TNA, allowing it to interact with the membrane-impermeable dye. Encapsulation of ceDNA can be calculated as E= (Io - 1 )/lo, where I and Io refer to the fluorescence intensities before and after the addition of detergent, respectively.
Compositions
In one embodiment, the lipid particle formulation is an aqueous solution. In one embodiment, the lipid particle (e.g., lipid nanoparticle) formulation is a lyophilized powder. According to some aspects, the disclosure provides for a lipid particle formulation further comprising one or more pharmaceutical excipients. In one embodiment, the lipid particle (e.g., lipid nanoparticle) formulation further comprises sucrose, Tris buffer, trehalose and/or glycine. Pharmaceutical compositions for therapeutic purposes can be formulated as a solution, microemulsion, dispersion, liposomes, or other ordered structure suitable for high TNA (e.g., mRNA) concentration. Sterile injectable solutions can be prepared by incorporating the TNA (e.g., mRNA) in the required amount in an appropriate buffer (e.g., pharmaceutically acceptable excipient) with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization.
In one embodiment, lipid particles (e.g., lipid nanoparticles) are solid core particles that possess at least one lipid bilayer. In one embodiment, the lipid particles (e.g., lipid nanoparticles) have a non-bilayer structure, i.e., a non-lamellar (i.e., non-bilayer) morphology. Without limitations, the non-bilayer morphology can include, for example, three dimensional tubes, rods, cubic symmetries, etc. The non-lamellar morphology (i.e., non-bilayer structure) of the lipid particles (e.g., lipid nanoparticles) can be determined using analytical techniques known to and used by those of skill in the art. Such techniques include, but are not limited to, Cryo-Transmission Electron Microscopy ("Cryo-TEM"), Differential Scanning calorimetry ("DSC"), X-Ray Diffraction, and the like. For example, the morphology of the lipid particles (lamellar vs. non-lamellar) can readily be assessed and characterized using, e.g., Cryo-TEM analysis as described in US2010/0130588. In one embodiment, the lipid particles (e.g., lipid nanoparticles) having a non-lamellar morphology are electron dense. In one embodiment, the disclosure provides for a lipid particle (e.g., lipid nanoparticle) that is either unilamellar or multilamellar in structure. In some aspects, the disclosure provides for a lipid particle (e.g., lipid nanoparticle) formulation that comprises multi-vesicular particles and/or foam-based particles. By controlling the composition and concentration of the lipid components, one can control the rate at which the lipid conjugate exchanges out of the lipid particle and, in turn, the rate at which the lipid particle (e.g., lipid nanoparticle) becomes fusogenic. In addition, other variables including, for example, pH, temperature, or ionic strength, can be used to vary and/or control the rate at which the lipid particle (e.g., lipid nanoparticle) becomes fusogenic. By controlling the composition and concentration of the lipid conjugate, one can control the lipid particle size. In one embodiment, the pKa' of formulated cationic lipids can be correlated with the effectiveness of the LNPs for deli very of nucleic acids (see Jayaraman et al., Angewandte Chemie, International Edition (2012), 51(34), 8529-8533; Semple et al., Nature Biotechnology 28, 172-176 (2010), both of which are incorporated by reference in their entireties). In one embodiment, the preferred range of pKa' for the ionizable lipid particle is about 6-7. In one embodiment, the pKa' of the cationic lipid can be determined in lipid particles (e.g., lipid nanoparticles) using an assay based on fluorescence of 2-(p-toluidino)-6-napthalene sulfonic acid (TNS). Pharmaceutical compositions or formulations can optionally comprise one or more additional active substances, e.g., therapeutically and/or prophylactically active substances. Pharmaceutical compositions or formulations of the present invention can be sterile and/or pyrogen-free. General considerations in the formulation and/or manufacture of pharmaceutical agents can be found, for example, in Remington: The Science and Practice of Pharmacy 21st ed., Lippincott Williams & Wilkins, 2005 (incorporated herein by reference in its entirety). In some embodiments, compositions are administered to humans, human patients or subjects. For the purposes of the present disclosure, the phrase "active ingredient" generally refers to a TNA, e.g., mRNA, to be delivered as described herein. Formulations and pharmaceutical compositions described herein can be prepared by any method known or hereafter developed in the art of pharmacology.
A pharmaceutical composition or formulation in accordance with the present disclosure can be prepared, packaged, and/or sold in bulk, as a single unit dose, and/or as a plurality of single unit doses. As used herein, a "unit dose" refers to a discrete amount of the pharmaceutical composition comprising a predetermined amount of the active ingredient. The amount of the active ingredient is generally equal to the dosage of the active ingredient that would be administered to a subject and/or a convenient fraction of such a dosage such as, for example, one-half or one-third of such a dosage.
Relative amounts of a TNA, the pharmaceutically acceptable excipient, and/or any additional ingredients in a pharmaceutical composition in accordance with the present disclosure can vary, depending upon the identity, size, and/or condition of the subject being treated and further depending upon the route by which the composition is to be administered. The compositions and formulations described herein may contain at least one TNA, such as a mRNA. As a non-limiting example, the composition or formulation can contain 1, 2, 3, 4 or 5 TNAs. In some embodiments, the composition or formulation can comprise a TNA in linear and/or circular form, and in single-stranded and/or doublestranded form. Although the descriptions of pharmaceutical compositions and formulations provided herein are principally directed to pharmaceutical compositions and formulations that are suitable for administration to humans, it will be understood by the skilled artisan that such compositions are generally suitable for administration to any other animal, e.g., to non-human animals, e.g. non-human mammals.
Pharmaceutically acceptable excipient, includes, but are not limited to, any and all solvents, dispersion media, or other liquid vehicles, dispersion or suspension aids, diluents, granulating and/or dispersing agents, surface active agents, isotonic agents, thickening or emulsifying agents, preservatives, binders, lubricants or oil, coloring, sweetening or flavoring agents, stabilizers, antioxidants, antimicrobial or antifungal agents, osmolality adjusting agents, pH adjusting agents, buffers, chelators, cyoprotectants, and/or bulking crosslinked polyvinyl pyrrolidone (crospovidone), cellulose, methylcellulose, carboxymethyl cellulose, cross-linked sodium carboxymethyl cellulose (croscarmellose), magnesium aluminum silicate (V EEG UM®), sodium lauryl sulfate, etc., and/or combinations thereof. Exemplary surface active agents and/or emulsifiers include, but are not limited to, natural emulsifiers ( e.g., acacia, agar, alginic acid, sodium alginate, tragacanth, chondrux, cholesterol, xanthan, pectin, gelatin, egg yolk, casein, wool fat, cholesterol, wax, and lecithin), sorbitan fatty acid esters (e.g., polyoxyethylene sorbitan monooleate [TWEEN®80], sorbitan monopalmitate [SPAN®40], glyceryl monooleate, polyoxyethylene esters, polyethylene glycol fatty acid esters ( e.g., CREMOPHOR®), polyoxyethylene ethers (e.g., polyoxyethylene lauryl ether [BRIJ®30]), PLURONIC® block copolymers, e.g. POLOXAMER® 188, etc. and/or combinations thereof. Exemplary binding agents include, but are not limited to, starch, gelatin, sugars (e.g., sucrose, glucose, dextrose, dextrin, molasses, lactose, lactitol, mannitol), amino acids (e.g., glycine), natural and synthetic gums (e.g., acacia, sodium alginate), ethylcellulose, hydroxyethylcellulose, hydroxypropyl methylcellulose, etc., and combinations thereof.
Oxidation is a potential degradation pathway for TNAs, especially for liquid or freeze-dried DNA formulations. To prevent oxidation, antioxidants can be added to the formulations. Exemplary antioxidants include, but are not limited to, alpha tocopherol, ascorbic acid, ascorbyl palmitate, benzyl alcohol, butylated hydroxyanisole, m-cresol, methionine, butylated hydroxytoluene, monothioglycerol, sodium or potassium metabisulfite, propionic acid, propyl gallate, sodium ascorbate, etc., and combinations thereof. Exemplary chelating agents include, but are not limited to, ethylenediaminetetraacetic acid (EDTA), disodium edetate, diethylenetriaminepentaacetic acid (DTPA, in ionized forms), citric acid monohydrate, fumaric acid, malic acid, phosphoric acid, sodium edetate, tartaric acid, Trisodium edetate, etc., and combinations thereof. Exemplary antimicrobial or antifungal agents include, but are not limited to, benzalkonium chloride, benzethonium chloride, methyl paraben, ethyl paraben, propyl paraben, butyl paraben, benzoic acid, hydroxybenzoic acid, potassium or sodium benzoate, potassium or sodium sorbate, sodium propionate, sorbic acid, etc., and combinations thereof. Exemplary preservatives include, but are not limited to, vitamin A, vitamin C, vitamin E, beta-carotene, citric acid, ascorbic acid, butylated hydroxyanisole, ethylenediamine, sodium lauryl sulfate (SLS), sodium lauryl ether sulfate (SLES), p-amino benzoic acid, methyl and/or propyl parabens, etc., and combinations thereof. In some embodiments, the pH of polynucleotide solutions is maintained between pH 5 and pH 8 to improve stability. Exemplary buffers to control pH can include, but are not limited to sodium phosphate, sodium citrate, sodium succinate, histidine (or histidine HCI), sodium malate, sodium carbonate, etc., and/or combinations thereof. Exemplary lubricating agents include, but are not limited to, magnesium stearate, calcium stearate, stearic acid, silica, talc, malt, hydrogenated vegetable oils, polyethylene glycol, sodium benzoate, sodium or magnesium lauryl sulfate, etc., and combinations thereof. The pharmaceutical composition or formulation described here can contain a cryoprotectant to stabilize a polynucleotide described herein during freezing. Exemplary cryoprotectants include, but are not limited to mannitol, sucrose, trehalose, lactose, glycerol, dextrose, etc., and combinations thereof. The pharmaceutical composition or formulation described here can contain a bulking agent in lyophilized polynucleotide formulations to yield a "pharmaceutically elegant" cake, stabilize the lyophilized polynucleotides during long term (e.g., 36-month) storage. Exemplary bulking agents of the present invention can include, but are not limited to sucrose, trehalose, mannitol, glycine, lactose, raffinose, and combinations thereof. In some embodiments, the pharmaceutical composition or formulation further comprises a delivery agent. The delivery agent of the present disclosure can include, without limitation, liposomes, lipid nanoparticles, lipidoids, polymers, lipoplexes, microvesicles, exosomes, extracellular vesicles, peptides, proteins, cells transfected with polynucleotides, hyaluronidase, nanoparticle mimics, nanotubes, conjugates, and combinations thereof.
Unit Dosage
In one embodiment, the pharmaceutical compositions can be presented in unit dosage form. A unit dosage form will typically be adapted to one or more specific routes of administration of the pharmaceutical composition. In some embodiments, the unit dosage form is adapted for intravenous, intramuscular, or subcutaneous administration. In some embodiments, the unit dosage form is adapted for intrathecal or intracerebroventricular administration. In some embodiments, the unit dosage form is adapted for administration by inhalation. In some embodiments, the unit dosage form is adapted for administration by a vaporizer. In some embodiments, the unit dosage form is adapted for administration by a nebulizer. In some embodiments, the unit dosage form is adapted for administration by an aerosolizer. In some embodiments, the unit dosage form is adapted for oral administration, for buccal administration, or for sublingual administration. In some embodiments, the pharmaceutical composition is formulated for topical administration. The amount of active ingredient which can be combined with a carrier material to produce a single dosage form will generally be that amount of the compound which produces a therapeutic effect. According to some embodiments, the LNP/TNA is for administration at a dose of about 0.02 pg to about 50 mg, about 0.02 pg to about 0.2 pg, or about 0.2 pg to about 2.0 pg, about 1 pg to about 25 pg, about 25 pg to about 50 pg, about 50 pg to about 100 pg, about 100 pg to about 200 pg, about 200 pg to about 300 pg, about 300 pg to about 400 pg, about 400 pg to about 500 pg, about 500 pg to about 750 pg, about 750 pg to about 1.0 mg, about 1 mg to about 10 mg, about 10 mg to about 25 mg, about 25 mg to about 50 mg.
Methods of Using
The pharmaceutical compositions comprising a lipid nanoparticle (LN P) and a therapeutic nucleic acid (TNA) can be used to introduce a nucleic acid sequence (e.g., a TNA) into a host cell. In one embodiment, the host cell is in vitro. In one embodiment, the host cell is in vivo. According to some embodiments, the subject is a human. In one embodiment, introduction of a nucleic acid sequence in a host cell using the pharmaceutical compositions comprising a lipid nanoparticle (LN P) and a therapeutic nucleic acid (TNA), as described herein, can be monitored with appropriate biomarkers from treated patients to assess gene expression.
Provided herein are methods of treating a disease or disorder in a subject comprising introducing into a cell in need thereof (for example, a muscle cell or tissue, or other affected cell type) of the subject a therapeutically effective amount of pharmaceutical composition comprising a lipid nanoparticle (LNP) and a therapeutic nucleic acid (TNA), wherein the LNP comprises an ApoE polypeptide, or a fragment thereof and/or an ApoB polypeptide, or a fragment thereof, linked to the LNP. While the TNA lipid nanoparticles can be introduced in the presence of a carrier, such a carrier is not required. Provided herein are methods for providing a subject in need thereof with a diagnostically- or therapeutically- effective amount of the pharmaceutical composition comprising a LNP and a TNA.
In general, the pharmaceutical composition comprising an LNP and a TNA can be used to deliver any TNA in accordance with the description above to treat, prevent, or ameliorate the symptoms associated with any disorder related to gene expression. Illustrative disease states include, but are not-limited to: cystic fibrosis (and other diseases of the lung), hemophilia A, hemophilia B, thalassemia, anemia and other blood disorders, AIDS, Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, epilepsy, and other neurological disorders, cancer, diabetes mellitus, muscular dystrophies (e.g., Duchenne, Becker), Hurler's disease, adenosine deaminase deficiency, metabolic defects, retinal degenerative diseases (and other diseases of the eye), mitochondriopathies (e.g., Leber's hereditary optic neuropathy (LHON), Leigh syndrome, and subacute sclerosing encephalopathy), myopathies (e.g., facioscapulohumeral myopathy (FSHD) and cardiomyopathies), diseases of solid organs (e.g., brain, liver, kidney, heart), and the like. In some embodiments, the ceDNA vectors as disclosed herein can be advantageously used in the treatment of individuals with metabolic disorders (e.g., ornithine transcarbamoylase deficiency). In one embodiment, the pharmaceutical composition comprising a LNP and a TNA can be used to treat, ameliorate, and/or prevent a disease or disorder caused by mutation in a gene or gene product (i.e., a genetic disorder) include, but are not limited to, metabolic diseases or disorders (e.g., Fabry disease, Gaucher disease, phenylketonuria (PKU), glycogen storage disease); urea cycle diseases or disorders (e.g., ornithine transcarbamoylase (OTC) deficiency); lysosomal storage diseases or disorders (e.g., metachromatic leukodystrophy (MLD), mucopolysaccharidosis Type II (MPSII; Hunter syndrome)); liver diseases or disorders (e.g., progressive familial intrahepatic cholestasis (PFIC); blood diseases or disorders (e.g., hemophilia (A and B), thalassemia, and anemia); cancers and tumors, and genetic diseases or disorders (e.g., cystic fibrosis). According to some embodiments, the genetic disorder is hemophilia A, hemophilia B, phenylketonuria (PKU, Gaucher disease Types I, II and III, Stargardt macular dystrophy, Leber congenital amaurosis (LCA), Usher syndrome, wet AMD. In one embodiment, the pharmaceutical composition comprising an LNP and a TNA may be employed to deliver a heterologous nucleotide sequence, e.g., to correct an abnormal level and/or function of a gene product, such as an absence of, or a defect in, a protein, that results in the disease or disorder. The TNA in lipid nanoparticles as described herein can produce a functional protein and/or modify levels of the protein to alleviate or reduce symptoms resulting from, or confer benefit to, a particular disease or disorder caused by the absence or a defect in the protein. For example, the TNA may be used for producing a functional protein or increased expression of a protein, such as OTC enzyme, Factor VIII, Factor IX, and Factor X, phenylalanine hydroxylase enzyme, alpha galactosidase or beta glucocerebrosidase, arylsulfatase A, iduronate-2-sulfatase, cystic fibrosis transmembrane conductance regulator, G6Pase enzyme, ATP8B 1, ABCB 11, ABCB4, or TJP2. In one embodiment, exemplary TNA encode a protein selected from: lysosomal enzymes (e.g., hexosaminidase A, iduronate sulfatase, associated, erythropoietin, angiostatin, endostatin, superoxide dismutase, globin, leptin, catalase, tyrosine hydroxylase, as well as cytokines (e.g., a interferon, b-interferon, interferon-gamma, interleukin-2, interleukin-4, interleukin 12, granulocyte- macrophage colony stimulating factor, lymphotoxin, and the like), peptide growth factors and hormones (e.g., somatotropin, insulin, insulin-like growth factors 1 and 2, platelet derived growth factor (PDGF), epidermal growth factor (EGF), fibroblast growth factor (FGF), nerve growth factor (NGF), neurotrophic factor-3 and 4, brain-derived neurotrophic factor (BDNF), glial derived growth factor (GDNF), transforming growth factor-a and -b, and the like), receptors (e.g., tumor necrosis factor receptor). In some exemplary embodiments, the transgene encodes a monoclonal antibody specific for one or more desired targets. In some exemplary embodiments, the antibody may be a full-length antibody or antibody fragment, e.g., antigen binding fragment, thereof.
Administration
In one embodiment, the pharmaceutical compositions comprising a lipid nanoparticle (LNP) and a therapeutic nucleic acid (TNA), as described herein, can be administered to an organism for transduction of cells in vivo. In one embodiment, the TNA can be administered to an organism for transduction of cells ex vivo. Generally, administration is by any of the routes normally used for introducing a molecule into ultimate contact with blood or tissue cells. Suitable methods of administering such nucleic acids are available and well known to those of skill in the art, and, although more than one route can be used to administer a particular composition, a particular route can often provide a more immediate and more effective reaction than another route. Exemplary modes of administration of the pharmaceutical composition of the invention include oral, rectal, buccal (e.g., sublingual), transmucosal (regardless of anatomical location), parenteral , including, but not limited to intravenous, intraarterial, subcutaneous, intradermal, intracranial, intramuscular (including administration to skeletal, diaphragm and/or cardiac muscle], intrapleural, intracerebral, intraosseous (including bone marrow), and intraarticular, intranasal, inhalation (e.g., via an aerosol), vaginal, intrathecal, intraocular, transdermal, intraendothelial, in utero (or in ovo), topical (e.g., to both skin and mucosal surfaces, including airway surfaces, and transdermal administration), intralymphatic, intranodal and the like, as well as direct tissue or organ injection (e.g., to liver, eye, skeletal muscle, cardiac muscle, diaphragm muscle or brain). Administration of the pharmaceutical composition can be to any site in a subject, including, without limitation, a site selected from the group consisting of the brain, a skeletal muscle, a smooth muscle, the heart, the diaphragm, the airway epithelium, the liver, the kidney, the spleen, the pancreas, the skin, and the eye. The pharmaceutical composition can be administered to skeletal muscle includes but is not limited to administration to skeletal muscle in the limbs (e.g., upper arm, lower arm, upper leg, and/or lower leg), back, neck, head (e.g., tongue), thorax, abdomen, pelvis/perineum, and/or digits, by intravenous administration, intra-arterial administration, intraperitoneal administration, limb perfusion, (optionally, isolated limb perfusion of a leg and/or arm; see, e.g., Arruda et al. 2005, Blood 105: 3458-3464), and/or direct intramuscular injection. In one embodiment, pharmaceutical composition is administered to cardiac muscle, including left atrium, right atrium, left ventricle, right ventricle and/or septum, e.g., by intravenous administration, intra-arterial administration such as intra-aortic administration, direct cardiac injection (e.g., into left atrium, right atrium, left ventricle, right ventricle), and/or coronary artery perfusion. Administration to diaphragm muscle can be by any suitable method including intravenous administration, intra-arterial administration, and/or intra-peritoneal administration. Administration to smooth muscle can be by any suitable method including intravenous administration, intra-arterial administration, and/or intra-peritoneal administration. In one embodiment, administration can be to endothelial cells present in, near, and/or on smooth muscle. In one embodiment, pharmaceutical composition comprising is administered to the CNS (e.g., to the brain or to the eye). The pharmaceutical composition may be introduced into the spinal cord, brainstem (medulla oblongata, pons), midbrain (hypothalamus, thalamus, epithalamus, pituitary gland, substantia nigra, pineal gland), cerebellum, telencephalon (corpus striatum, cerebrum including the occipital, temporal, parietal and frontal lobes, cortex, basal ganglia, hippocampus and porta amygdala), limbic system, neocortex, corpus striatum, cerebrum, and inferior colliculus. The pharmaceutical compositions may also be administered to different regions of the eye such as the retina, cornea and/or optic nerve., e.g., via subretinal injection, suprachoroidal injection, or intravitreal injection The pharmaceutical composition may be delivered into the cerebrospinal fluid (e.g., by lumbar puncture). The pharmaceutical composition may be administered to the desired region(s) of the CNS by any route known in the art, including but not limited to, intrathecal, intra-ocular, intracerebral, intraventricular, intravenous (e.g., in the presence of a sugar such as mannitol), intranasal, intra-aural, intra-ocular (e.g., intra- vitreous, sub-retinal, anterior chamber) and peri-ocular (e.g., sub-Tenon's region) delivery as well as intramuscular delivery with retrograde delivery to motor neurons. In one embodiment, repeat administrations of the therapeutic product can be made until the appropriate level of expression has been achieved. Thus, in one embodiment, a therapeutic nucleic acid can be administered and re-dosed multiple times. Examples
Synthesis of amino lipids is carried out with standard methods of organic synthetic methodology. All cited sources, for example, references, publications, databases, database entries, and synthesis art cited herein, are incorporated into this application by reference, even if not expressly stated in the citation. All solvents and reagents are/were obtained commercially and used as such unless noted otherwise.
Synthesis of decyl 4-(((4-amino-2,6,7-trioxabicyclo[2.2.2]octan-l-yl)methyl)(4- (heptadecan-9-yloxy)-4-oxobutyl)amino)butanoate (52).
To a cold solution of 4-bromobutanoic acid (52-1, 5.0 g, 30 mmol) in dichloromethane (20 mL) was added oxalyl chloride (3.85 mL, 45 mmol) followed by dimethylformamide (1 drop). The resultant mixture was stirred at 0 °C for 2 h and the solvent was then evaporated in a rotary evaporator. The residue was dissolved in dichloromethane (20 mL), cooled to 0 °C, and decan-9-ol (52-2, 7.1 g, 45 mmol) was added followed by pyridine (4.84 mL, 60 mmol). The resultant mixture was stirred at ambient temperature overnight. The mixture was quenched with ice, extracted with dichloromethane (2 x 25 mL). The combined organic layer was washed with water, brine, dried over anhydrous sodium sulfate and evaporated under vacuo. The crude product was purified by silica gel flash chromatography with 0-10% ethyl acetate-dichloromethane to obtain decyl 4-bromobutanoate (52-3, 8.09 g, 88%). 1H NMR (400 MHz, CDCI3, ppm): δ 4.04-4.07 (m, 2H), 3.43-3.48 (m, 2H), 2.45-2.49 (m, 2H), 2.09- 2.18 (m, 2H), 1.45-1.6 (m, 2H), 1.24-1.31 (m, 14H), 0.84-0.89 (m, 3H).
Figure imgf000100_0001
To a cold solution of 4-bromobutanoic acid (52-1, 5.0 g, 30 mmol) in dichloromethane (20 mL) was added oxalyl chloride (3.85 mL, 45 mmol) followed by dimethylformamide (1 drop). The resultant mixture was stirred at 0°C for 2 h and the solvent was then evaporated in a rotary evaporator. The residue was dissolved in dichloromethane (20 mL), cooled to 0 °C, and heptadecan-9-ol (52-4, 11.5 g, 45 mmol) was added followed by pyridine (4.84 mL, 60 mmol). The resultant mixture was stirred at ambient temperature overnight. The mixture was quenched with ice, extracted with dichloromethane (2 x 25 mL). The combined organic layer was washed with water, brine, dried over anhydrous sodium sulfate and evaporated under vacuo. The crude product was purified by silica gel flash chromatography with 0-10% ethyl acetate-dichloromethane to obtain heptadecan-9-yl 4-bromobutanoate (52-5,11.35 g, 94%).1H NMR (400 MHz, CDCI3, ppm): δ 4.82-4.92 (m, 1H), 3.45-3.62 (m, 2H), 2.47-2.52 (m, 2H), 2.05-2.18 (m, 2H), 1.45-1.6 (m, 4H), 1.21-1.31 (m, 24H), 0.86-0.89 (m, 6H).
Figure imgf000100_0002
Synthesis of heptadecan-9-yl 4-(benzylamino)butanoate (Compound 52-6)
Figure imgf000101_0001
A mixture of heptadecan-9-yl 4-bromobutanoate (52-3, 3.75 g, 9.27 mmol) and benzylamine (4.97 g, 46.38 mmol) in ethanol (100 mL) was refluxed at 80 °C for 48 h. The solvent was removed, and the product was purified by silica gel flash chromatography with 0-10% ethyl acetate-dichloromethane to obtain the desired compound, with 0-10% ethyl acetatedichloromethane to obtain heptadecan-9-yl 4-(benzylamino)butanoate (52-6, 2.45 g, 88%). The 1H NMR showed some impurities, and this product was used in the next step without further purification. 1H NMR (400 MHz, CDCI3, ppm): δ 7.22-7.34 (m, 5H), 4.82-4.88 (m, 1H), 4.44 (br s, 1H), 3.77 (s, 2H), 3.45-3.62 (m, 2H), 2.63-2.67 (m, 2H), 2.33-2.37 (m, 2H), 1.80- 1.86 (m, 2H), 1.48-1.52 (m, 2H), 1.21-1.31 (m, 24H), 0.84-0.88 (m, 6H).
Synthesis of decyl 4-(benzyl(4-(heptadecan-9-yloxy)-4-oxobutyl)amino)butanoate (52-7)
Figure imgf000101_0002
To a solution of heptadecan-9-yl 4-(benzylamino)butanoate (52-6, 500 mg, 1.16 mmol) and decyl 4-bromobutanoate ,(52-3,532 mg, 1.74 mmol) in acetonitrile (100 mL) was added potassium carbonate (480 mg, 3.5 mmol) and potassium iodide (96 mg, 0.6 mmol) and the reaction was heated at 180 °C for 48 h. The reaction mixture was filtered, washed with methylene chloride and the solvents evaporated. Another batch with 2.0 g of heptadecan-9- yl 4-(benzylamino)butanoate (52-6 was carried out by following the above procedure and both the crude mixtures were combined and purified by silica gel flash chromatography using 0-30% ethyl acetate-dichloromethane to obtain decyl 4-(benzyl(4-(heptadecan-9- yloxy)-4-oxobutyl)amino)butanoate (52-7, 2.35 g, 61%). The 1H NMR showed some impurities, and this product was used in the next step without further purification. 1H NMR (400 MHz, CDCI3, ppm): δ 7.21-7.29 (m, 5H), 4.81-4.87 (m, 1H), 3.98-4.03 (m, 2H), 3.54 (s, 2H), 2.28-2.43 (m, 8H), 1.75-1.81 (m, 4H), 1.45-1.53 (m, 4H), 1.18-1.45 (m, 40H), 0.85-0.88 (m, 9H). APCI: m/z 658.6
Synthesis of decyl 4-((4-(heptadecan-9-yloxy)-4-oxobutyl)amino)butanoate (52-8)
Figure imgf000101_0003
A solution of decyl 4-(benzyl(4-(heptadecan-9-yloxy)-4-oxobutyl)amino)butanoate (36-7, 2.35 g, 3.6 mmol) in ethyl acetate (100 mL) was stirred overnight under hydrogen atmosphere in the presence of 10% Pd/C (wet, 500 mg). The reaction mixture was filtered through celite rinsing with ethyl acetate and dichloromethane. The filtrate was evaporated to obtain the desired compound, decyl 4-((4-(heptadecan-9-yloxy)-4- oxobutyl)amino)butanoate (52-8, 1.5 g, 75%). (400H M NHMz,R CDCI3, ppm): δ 4.84-4.87 (m, 1H), 4.01-4.05 (m, 2H), 3.62-3.64 (m, 1H), 2.61-2.65 (m, 2H), 2.28-2.40 (m, 4H), 1.75-1.88 (m, 4H), 1.50-1.65 (m, 6H), 1.19-1.43 (m, 40H), 0.83-0.89 (m, 9H). APCI: m/z 568.5
Synthesis of decyl 4-((cyanomethyl)(4-(heptadecan-9-yloxy)-4-oxobutyl)amino)butanoate (52-9) and heptadecan-9-yl 4-(2-oxopyrrolidin-l-yl)butanoate
Figure imgf000102_0001
To a solution of decyl 4-((4-(heptadecan-9-yloxy)-4-oxobutyl)amino)butanoate (52-8, 1.45 g, 2.55 mmol) and chloroacetonitrile (289 mg, 3.83 mmol) in acetonitrile (50 mL) was added potassium carbonate (1.06 g, 7.66 mmol) and potassium iodide (212 mg, 1.3 mmol) and the reaction was heated at 180 °C for 48 h. The reaction mixture was filtered, washed with methylene chloride and the solvents evaporated. The crude was purified by silica gel flash chromatography using 0-30% ethyl acetate-dichloromethane to obtain decyl 4- ((cyanomethyl)(4-(heptadecan-9-yloxy)-4-oxobutyl)amino)butanoate (52-9, 148 mg, 10%) and heptadecan-9-yl 4-(2-oxopyrrolidin-l-yl)butanoate (750 mg, 72%). decyl 4-((cyanomethyl)(4-(heptadecan-9-yloxy)-4-oxobutyl)amino)butanoate (52-9):1H NMR (400 MHz, CDCI3, ppm): δ 4.83-4.89 (m, 1H), 4.04-4.07 (m, 2H), 3.56 (s, 2H), 2.53-2.56 (m, 4H), 2.30-2.35 (m, 4H), 1.50-1.78 (m, 8H), 1.19-1.36 (m, 40H), 0.84-0.89 (m, 9H). heptadecan-9-yl 4-(2-oxopyrrolidin-l-yl)butanoate: (410H0 N MMHRz, CDCI3, ppm): 64.84- 4.89 (m, 1H), 3.29-3.42 (m, 4H), 2.29-2.38 (m, 4H), 1.99-2.08 (m, 2H), 1.80-1.90 (m, 2H), 1.65-1.47(m, 2H), 1.45-1.55 (m, 2H), 1.20-1.32 (m, 24H), 0.86-0.89 (m, 6H).
Synthesis of decyl 4-(((trimethoxy)methyl)(4-(heptadecan-9-yloxy)-4- oxobutyl)amino)butanoate (Compound 52-10)
The acetonitrile-substituted tertiary amine is treated with HCI gas in methanol-containing solvent. The orthoester is isolated from unreacted material and any methyl esters side product occurring by transesterification.
Figure imgf000103_0001
Synthesis of decyl 4-((2-(2-amino-3-hydroxy-2-(hydroxymethyl)propoxy)-2-oxoethyl)(4 - (heptadecan-9-yloxy)-4-oxobutyl)amino)butanoate (Compound 52)
The decyl 4-(((trimethoxy)methyl)(4-(heptadecan-9-yloxy)-4-oxobutyl)amino)butanoate (52- 10) is treated with Tris(hydroxymethyl)nitromethane to form the bicyclic system with a nitro group opposite to the lipid tails. The resulting nitro-bicyclo lipid compound is reduced with hydrogen gas over PtO2 to produce the desired amino-bicyclo lipid (Compound 36). The amino lipid is purified by column chromatography over alumina, taking care to purify out the unreacted nitro analog.
Figure imgf000103_0002
Acidic hydrolysis of bicyclic ring system in Compound 36 is accomplished without hydrolysis of the carboxyl esters of the tails by mild acid treatment to obtain Compound 52.
Figure imgf000103_0003
Figure imgf000104_0001
Synthetic Scheme for Compound 51, octyl 10-(2-hydroxyethyl)-16-octyl-14-oxo-7, 13,15- trioxa-10-azatetracosanoate, is shown below.
Figure imgf000105_0001
Compound 51
Synthesis of 6-(2-(benzyloxy)ethoxy)hexan-l-ol (51-3 in scheme for Compound 51)
Figure imgf000105_0002
Chemical Formula: C15H24O3
Exact Mass: 252.17
Molecular Weight: 252.35
Figure imgf000105_0003
To a suspension of sodium hydride (3.94 g, 60% dispersion in oil, 98 mmol) in THF (200 mL) was added dropwise a solution of 2-(benzyloxy)ethan-l-ol (51-2, 10.0 g, 65 mmol) in THF (50 mL) at 0 °C. It was stirred at 0 °C for 1 h. Then a solution of 6-bromohexan-l-ol (51-1, 14.3 g, 78 mmol) was added and the reaction mixture stirred at room temperature overnight. It was quenched with ice, extracted with hexane (3 X 100 mL). The organic layer was washed with brine, dried with anhydrous sodium sulfate and the solvent removed to get the crude compound. The crude compound was purified by flash chromatography on silica gel using 40% ethyl acetate in hexane to get pure 6-(2-(benzyloxy)ethoxy)hexan-l-ol (51-3, 13.9 g, 84%. 1H NMR (400 MHz, CDCI3, ppm): δ 7.26-7.35 (m, 5H), 4.57 (s, 2H), 3.39-3.65 (m, 8H), 3.80 (m, 1H), 1.36-1.58 (m, 8H).
Synthesis of 6-(2-(benzyloxy)ethoxy)hexanoic acid (51-4 in scheme for Compound 51)
Figure imgf000106_0001
Chemical Formula: C15H22O4 Exact Mass: 266.15 Molecular Weight: 266.34
Figure imgf000106_0002
To a solution of 6-(2-(benzyloxy)ethoxy)hexan-l-ol (51-3, 4.0 g, 16 mmol) in acetone (100 mL) at 0 °C, was added Jones reagent (10.0 mL, 2M, 32 mmol). The reaction mixture was stirred at 0 °C for 3h. The reaction was quenched with isopropanol. The solvent was then removed, and the crude product taken in dichloromethane (100 mL). the organic layer was washed with sodium hydroxide (IN, 3X50 mL). The aqueous part was then acidified with hydrochloric acid (IN) and extracted with ethyl acetate. The ethyl acetate layer was washed with brine, dried over anhydrous sodium sulfate and solvent removed to give the product, 6-(2-(benzyloxy)ethoxy) hexanoic acid (51-4, 2.4 g), which was directly used in the next step.
Synthesis of octyl 6-(2-(benzyloxy)ethoxy)hexanoate (51-6 in scheme for Compound 51)
Figure imgf000106_0003
Chemical Formula: C23H38O4 Exact Mass: 378.28 Molecular Weight: 378.55
Figure imgf000106_0004
To a solution of 6-(2-(benzyloxy)ethoxy) hexanoic acid (51-4, 2.1 g, 8 mmol) in dichloromethane (50 mL) at 0 °C, was added oxalyl chloride (1.54 g, 1.06 mL, 12.1 mmol) followed by /V,/V-Dimethylformamide (0.05 mL) under nitrogen atmosphere. The reaction mixture was stirred at 0 °C for 2 h. The solvent was removed, and the crude reaction mixture dried in rotavapor for 2 h. Then the crude acid chloride was dissolved in dichloromethane (50 mL) and octan-l-ol (51-5, 1.58 g, 1.9 mL, 12.1 mmol) was added at 0 °C followed by pyridine (1.28 g, 1.31 mL, 16.1 mmol). The reaction mixture was stirred at room temperature for 18 h under nitrogen. The reaction mixture was quenched with ice. The organic layer was separated washed with brine, dried with anhydrous sodium sulfate and the solvent removed to get the crude product which was purified by flash chromatography to provide octyl 6-(2-(benzyloxy)ethoxy)hexanoate (51-6, 650 mg, 21%) as a colorless oiI. 1H NMR (400 MHz, CDCI3, ppm): δ 7.26-7.35 (m, 5H), 4.57 (s, 2H), 4.05 (t, 2H, J 6.8 Hz), 3.60 (s, 4H), 3.46 (t, 2H, J 6.8 Hz), 2.30 (t, 2H, J 6.6 Hz), 1.59-1.72 (m, 8H), 1.26-1.42 (m, 10H), 0.88 (t, 3H, J 6.8 Hz).
Synthesis of octyl 6-(2-hydroxyethoxy)hexanoate (51-7 in scheme for Compound 51)
Figure imgf000107_0001
Chemical Formula: C16H32O4 Exact Mass: 288.23 Molecular Weight: 288.43
To a solution of octyl 6-(2-(benzyloxy)ethoxy)hexanoate (51-6, 1.75 g, 4.62 mmol) in ethyl acetate (50 mL) was added palladium hydroxide on carbon (20%) (325 mg, 0.46 mmol)and the reaction mixture stirred at room temperature under hydrogen atmosphere for 18 h. The reaction mixture was filtered through a bed of celite, washed with ethyl acetate and dichloromethane and the solvent removed to get the crude product octyl 6-(2- hydroxyethoxy)hexanoate (51-7, 1.33 g, 99 %) in quantitative yield. The crude product was carried forward to the next reaction. 1H NMR (400 MHz, CDCI3, ppm): δ 4.05 (t, 2H, J 6.8 Hz), 3.73 (t, 2H, J 4.5 Hz), 3.53 (t, 2H, J 4.4 Hz), 3.48 (t, 2H, J 6.4 Hz), 2.31 (t, 2H, J 7.2 Hz), 1.58- 1.70 (m, 6H), 1.20-1.42 (m, 13H), 0.88 (t, 3H, J 6.8 Hz).
Synthesis of octyl 6-(2-bromoethoxy)hexanoate (51-8 in scheme for Compound 51)
Figure imgf000107_0002
Chemical Formula: C-ieHsiBrOa Exact Mass: 350.15 Molecular Weight: 351.33
Figure imgf000107_0003
To a solution of octyl 6-(2-hydroxyethoxy)hexanoate (51-7, 1.33 g, 4.61 mmol) in dichloromethane (100 mL) at room temperature was added carbon tetrabromide (2.14 g, 6.46 mmol) followed by triphenylphosphine (1.69 g, 6.46 mmol). The reaction mixture was stirred at room temperature for 2 h. Solid silica was added to the reaction mixture and the solvent removed. The silica was directly loaded and purified by flash chromatography to get octyl 6-(2-bromoethoxy)hexanoate (51-8, 1.299 g, 80 %) as colorless oil. (400 MHz, 1H NMR CDCI3, ppm): δ 4.05 (t, 2H, J 6.6 Hz), 3.73 (t, 2H, J 6.2 Hz), 3.44-3.52 (m, 4H), 2.31 (t, 2H, J 7.6 Hz), 1.57-1.72 (m, 6H), 1.21-144 (m, 12H), 0.88 (t, 3H, J 6.6 Hz).
Synthesis of octyl 6-(2-((2-hydroxyethyl)amino)ethoxy)hexanoate (51-10 in scheme for Compound 51)
Figure imgf000108_0001
To a solution of octyl 6-(2-bromoethoxy)hexanoate (51-8, 1.29 g, 3.67 mmol) in ethanol (50 mL) was added ethanolamine (51-9, 4.49 g, 4.44 mL, 73.4 mmol) and the reaction mixture refluxed for 18 h at 80 °C. The solvent was removed, and the crude reaction mixture purified by flash chromatography to obtain octyl 6-(2-((2-hydroxyethyl)amino)ethoxy)hexanoate (51- 10, 1.01 g, 83 %) as an yellow oil.
The crude compound NMR is below, the material is carried forward to be purified in the next step. 1H NM (4R00 MHz, CDCI3, ppm): δ 4.05 (t, 2H, J 6.8 Hz), 3.36-3.58 (m, 6H), 2.79 (t, 4H, J 5.2 Hz), 2.31 (t, 2H, J 7.4 Hz), 2.07 (bs, 2H), 1.58-1.66 (m, 6H), 1.20-1.39 (m, 12H), 0.88 (t, 3H, J 6.8 Hz).
Synthesis of 2-bromoethyl heptadecan-9-yl carbonate (15 in scheme for Compound 51)
Figure imgf000108_0002
To a solution of 2-bromoethan-l-ol (51-11, 3.00 g, 24.0 mmol) in dichloromethane (50 mL) was added 4-nitrophenyl carbonochloridate (51-12, 5.80 g, 29.0 mmol) followed by DMAP (0.58 g, 5.0 mmol) and pyridine (3.80 g, 2.15 mL, 48.0 mmol) and the reaction mixture stirred at room temperature for 1 h. Then, heptadecan-9-ol (51-14, 18.50 g, 72.0 mmol) was added followed by diisopropylethylamine (9.30 g, 12.5 mL, 72.0 mmol) and the reaction mixture stirred at room temperature overnight. The reaction was quenched with sodium carbonate (IM), washed with water and the solvent removed to get the crude product (51- 15) which was used without further purification.
Synthesis of octyl 10-(2-hydroxyethyl)-16-octyl-14-oxo-7,13,15-trioxa-10- azatetracosanoate (Compound 51)
Figure imgf000109_0001
To a solution of octyl 6-(2-((2-hydroxyethyl)amino)ethoxy)hexanoate (51-10, 400 mg, 1.21 mmol) in acetonitrile (50 mL) was added 2-bromoethyl heptadecan-9-yl carbonate (51-15, 737 mg, 1.81 mmol) followed by potassium carbonate (500 mg, 3.62 mmol) and potassium iodide (100 mg, 0.60 mmol). The reaction mixture was then heated to reflux at 98 °C for 48 h. The reaction mixture initially turned canary yellow and then slowly the color changes and becomes colorless. The solvent was removed, and the crude compound purified by flash chromatography, followed by reverse phase chromatography using acetonitrile and water in 0.1% trifluoroacetic acid. The pure product was dissolved in dichloromethane (5.0 mL) and solid sodium bicarbonate (100 mg, 1.19 mmol) was added and stirred for 1 hour. It was then filtered, and the solvent removed to get the pure product octyl 10-(2-hydroxyethyl)-16- octyl-14-oxo-7,13,15-trioxa-10-azatetracosanoate (Compound 51 150 mg, 18%) as colorless oil. 1H NMR (400 MHz, CD3OD, ppm): δ 4.70-4.73 (m, 1H), 4.51 (t, 2H, J 4.8 Hz), 4.05 (t, 2H, J 6.6 Hz), 3.88 (t, 2H, J 5.2 Hz), 3.79 (t, 2H, J 4.8 Hz), 3.69 (t, 2H, J 4.8 Hz), 3.51-3.54 (m 4H),
3.44 (m, 2H), 2.33 (t, 2H, J 7.4 Hz), 1.58-1.65 (m 9H), 1.29-1.41 (m, 38H), 0.88-0.91 (m, 9H). APCI: m/z 658.5, HPLC-ELSD purity = >99%. HPLC-CAD purity = >97%.
Synthetic Scheme for Compound 53, 2-((2-((3-(heptadecan-9-yloxy)-3- oxopropanoyl)oxy)ethyl)(2-hydroxyethyl)amino)ethyl octyl adipate:
Figure imgf000110_0001
Compound 53
Synthesis of benzyl octyl adipate (53-2 in scheme for Compound 53)
Figure imgf000110_0002
Chemical Formula: C21H32O4 Exact Mass: 348.23
Figure imgf000110_0003
0 Molecular Weight: 348.48
To a solution of benzyl adipate (53-1, 2.1 g, 8 mmol) in dichloromethane (50 mL) at 0 °C was added oxalyl chloride (1.54 g, 1.06 mL, 12.1 mmol) followed by /V,/V-Dimethylformamide (0.05 mL) under nitrogen atmosphere. The reaction mixture was stirred at 0 °C for 2 h. The solvent was removed, and the crude reaction mixture dried in rotavapor for 2 h. Then the crude acid chloride was dissolved in dichloromethane (50 mL) and octan-l-ol (51-5, 1.58 g, 1.9 mL, 12.1 mmol) was added at 0 °C followed by pyridine (1.28 g, 1.31 mL, 16.1 mmol). The reaction mixture was stirred at room temperature for 18 h under nitrogen. The reaction mixture was quenched with ice. The organic layer was separated washed with brine, dried with anhydrous sodium sulfate and the solvent removed to get the crude product which was purified by flash chromatography to get benzyl octyl adipate (53-2, 650 mg, 21.3 %) as a colorless oil^H NMR (400 MHz, CDCh, ppm): δ 7.32-7.36 (m, 5H), 5.11 (s, 2H), 4.05 (t, 2H, J 6.8 Hz), 2.29-2.40 (m, 4H), 1.56-1.72 (m, 6H), 1.21-1.36 (m, 10H), 0.88 (t, 3H, J 6.8 Hz).
Synthesis of octyl adipate(53-3 in scheme for Compound 53)
Figure imgf000111_0001
To a solution of benzyl octyl adipate (53-2 10.0 g, 29 mmol) in ethyl acetate (100 mL) was added palladium hydroxide on carbon (20%) (1.5 g, 14 mmol) and the reaction mixture stirred at room temperature under hydrogen atmosphere for 18 h. The reaction mixture was filtered through a bed of celite, washed with ethyl acetate and dichloromethane and the solvent removed to get the crude product to get the product octyl adipate(53-3, 4.6 g, 49%), which was directly used in the next step. (4001H M NHMz,R CDCI3, ppm): δ 4.06 (t, 2H, J 6.8 Hz), 2.31-2.40 (m, 4H), 1.59-1.69 (m, 6H), 1.12-1.38 (m, 10H), 0.88 (t, 3H, J 6.8 Hz)
Synthesis of 3-(heptadecan-9-yloxy)-3-oxopropanoic acid (53-5in scheme for Compound 53)
Figure imgf000111_0002
A solution of 2, 2-dimethyl-l,3-dioxane-4, 6-dione (53-4, 4.0 g, 28 mmol) and heptadecan-9- 01 (51-14, 7.12 g, 28 mmol) in toluene (100 mL) was refluxed at 120 °C for 4 h. It was cooled and the solvent removed under vacuum to get the crude product, which was purified by flash chromatography using ethyl acetate and hexane to get pure 3-(heptadecan-9-yloxy)-3- oxopropanoic acid (53-5, 5.94 g, 62%) .[pH NMR (400 MHz, CDCI3, ppm): δ 4.93-4.99 (m, 1H), 3.42 (s, 2H), 1.54-1.56 (bm, 5H), 1.12-1.42 (m, 24H), 0.87 (t, 6H, J 6.6 Hz).
Synthesis of 2-((2-((tert-butyldimethylsilyl)oxy)ethyl)amino)ethan-l-ol (53-6 in scheme for Compound 53)
Figure imgf000111_0003
A solution of (2-bromoethyl)(tert-butyl)dimethylsilane (10.0 g, 42 mmol) and ethanolamine (51-9, 51.0 g, 50.5 mL, 840 mmol) in acetonitrile (130 mL) was heated to reflux at 90 °C for 3 h. It was cooled and poured into ice-water and extracted with ethyl acetate (2 X 100 mL). The organic layer was washed with brine, dried with anhydrous sodium sulfate, the solvent removed to get the crude product which was purified by flash chromatography to get 2-((2- ((tert-butyldimethylsilyl)oxy)ethyl)amino)ethan-l-ol (53-6, 3.78 g, 41%). 1H NMR (400 MHz, CDCI3, ppm): δ 3.72 (t, 2H, J 5.4 Hz), 3.64 (t, 2H, J 5.0 Hz), 2.80 (t, 2H, J 5.2 Hz), 2.74 (t, 2H, J 5.2 Hz), 2.31 (bs, 2H), 0.90 (s, 9H), 0.07 (s, 6H).
Synthesis of 2-((2-(benzyloxy)ethyl)(2-((tert-butyldimethylsilyl)oxy)ethyl)amino)ethan-l-ol (53-7 in scheme for Compound 53)
Figure imgf000112_0001
To a solution of 2-((2-((tert-butyldimethylsilyl)oxy)ethyl)amino)ethan-l-ol (53-6, 3.5 g, 16 mmol) in acetonitrile (20 mL) was added ((2-bromoethoxy)methyl)benzene (5.15 g, 24 mmol) followed by potassium carbonate (6.6 g, 48 mmol) and potassium iodide (1.3 g, 8 mmol). The reaction mixture was then heated to reflux at 98 °C for 48 h. The solvent was removed and the crude compound purified by flash chromatography to get the pure product 2-((2-(benzyloxy)ethyl)(2-((tert-butyldimethylsilyl)oxy)ethyl)amino)ethan-l-ol (53-7, 3.87 g, 68 %). 1H N (M40R0 MHz, CDCI3, ppm): δ 7.26-7.34 (m, 5H), 4.51 (s, 2H), 3.66 (t, 2H, J 6.0 Hz), 3.52-3.56 (m, 4H), 2.83 (t, 2H, J 5.8 Hz), 2.71-2.75 (m, 4H), 0.88 (s, 9H), 0.04 (s, 6H).
Synthesis of 2-((2-(benzyloxy)ethyl)(2-((tert-butyldimethylsilyl)oxy)ethyl)amino)ethyl octyl adipate (53-8 in scheme for Compound 53)
Figure imgf000112_0002
To a solution of octyl adipate (53-3, 1.5 g, 5.8 mmol) in dichloromethane (10 mL) at 0 °C was added oxalyl chloride (1.10 g, 0.75 mL, 8.7 mmol) followed by /V,/V-Dimethylformamide (0.05 mL) under nitrogen atmosphere. The reaction mixture was stirred at 0 °C for 2 h. The solvent was removed, and the crude reaction mixture dried in rotavapor for 2 h. Then the crude acid chloride was dissolved in dichloromethane (10 mL) and 2-((2-(benzyloxy) ethyl)(2- ((tert-butyldimethylsilyl)oxy)ethyl)amino)ethan-l-ol (53-7, 3.08 g, 8.7 mmol) was added at 0 °C followed by pyridine (0.92 g, 0.94 mL, 11.6 mmol). The reaction mixture was stirred at room temperature for 18 h under nitrogen. The reaction mixture was quenched with ice. The organic layer was separated washed with brine, dried with anhydrous sodium sulfate and the solvent removed to get the crude product which was purified by flash chromatography to get 2-((2-( benzy loxy)ethy l)(2-((tert- butyldimethylsilyl)oxy)ethyl)amino)ethyl octyl adipate (53-8 2.61 g, 76 %) as a colorless oil. XH NMR (400 MHz, CDCI3, ppm): δ 7.26-7.34 (m, 5H), 4.51 (s, 2H), 4.12 (t, 2H, J 5.6 Hz), 4.05 (t, 2H, J 6.8 Hz), 3.66 (t, 2H, J 6.4 Hz), 3.53 (t, 2H, J 6.8 Hz), 2.83-2.85 (m, 4H), 2.72 (t, 2H, J 5.2 Hz), 2.25-2.34 (m, 4H), 1.58-1.72 (m, 6H), 1.20-1.39 (m, 10H), 0.86-0.89 (m, 12H), 0.04 (s, 6H).
Synthesis of 2-((2-(benzyloxy)ethyl)(2-hydroxyethyl)amino)ethyl octyl adipate (53-9 in scheme for Compound 53)
Figure imgf000113_0001
To a solution 2-((2-(benzyloxy)ethyl)(2-((tert-butyldimethylsilyl)oxy)ethyl)amino)ethyl octyl adipate (53-8, 1.4 g, 2.5 mmol) in tetra hydrofuran (10 mL) at 0 °C, was added hydrofluoric acid - pyridine (2.5 mL, 70% HF - 30% Pyridine) and the reaction mixture stirred at room temperature for 18 h. The reaction was quenched with saturated sodium bi-carbonate solution, extracted with dichloromethane. The organic layer was washed with brine, dried with anhydrous sodium sulfate and the solvent removed to get the crude product which was used purified using silica gel column to get pure 2-((2-(benzyloxy)ethyl)(2- hydroxyethyl)amino)ethyl octyl adipate (53-9, 830 mg, 73%). (400 M 1HHz, N CMDCRI3, ppm): δ 7.26-7.35 (m, 5H), 4.51 (s, 2H), 4.15 (t, 2H, J 5.6 Hz), 4.05 (t, 2H, J 6.8 Hz), 3.52-3.55 (m, 4H), 2.80-2.86 (m, 4H), 2.73 (t, 2H, J 5.2 Hz), 2.28-2.32 (m, 4H), 1.59-1.68 (m, 7H), 1.20- 1.39 (m, 10H), 0.88 (t, 3H, J 7.2 Hz). Synthesis of 2-((2-(benzyloxy)ethyl)(2-((3-(heptadecan-9-yloxy)-3-oxopropanoyl)oxy)ethyl) amino)ethyl octyl adipate 53-10 in scheme for Compound 53)
Figure imgf000114_0001
Chemical Formula: C47H81NO9 Exact Mass: 803.59 Molecular Weight: 804.16
Figure imgf000114_0002
To a solution of 3-(heptadecan-9-yloxy)-3-oxopropanoic acid (53-5, 290 mg, 0.85 mmol) in dichloromethane (10 mL) at 0 °C was added oxalyl chloride (161 mg, 0.1 mL, 1.27 mmol) followed by /V,/V-Dimethylformamide (0.05 mL) under nitrogen atmosphere. The reaction mixture was stirred at 0 °C for 2 h. The solvent was removed, and the crude reaction mixture dried in rotavapor for 2 h. Then the crude acid chloride was dissolved in dichloromethane (10 mL) and 2-((2-(benzyloxy)ethyl)(2-hydroxyethyl)amino)ethyl octyl adipate (53-9, 609 mg, 1.27 mmol) was added at 0 °C followed by pyridine (134 mg, 0.14 mL, 1.69 mmol). The reaction mixture was stirred at room temperature for 18 h under nitrogen. The reaction mixture was quenched with ice. The organic layer was separated washed with brine, dried with anhydrous sodium sulfate and the solvent removed to get the crude product which was purified by flash chromatography to get the pure product 2-((2- (benzyloxy)ethyl)(2-((3-(heptadecan-9-yloxy)-3-oxopropanoyl)oxy)ethyl)amino)ethyl octyl adipate (53-10, 200 mg, 29 %) as colorless oil. (4001H M NHMz,R CDCI3, ppm): δ 7.26-7.36 (m, 5H), 4.88-4.91 (m, 1H), 4.51 (s, 2H), 4.19 (t, 2H, J 6.4 Hz), 4.11 (t, 2H, J 6.2 Hz), 4.05 (t, 2H, J 6.8 Hz), 3.53 (t, 2H, J 5.8 Hz), 3.34 (s, 2H), 2.81-2.86 (m, 6H), 2.29-2.32 (m, 4H), 1.48- 1.65 (m, 9H), 1.20-1.39 (m, 35H), 0.85-0.89 (m, 9H).
Synthesis 2-((2-((3-(heptadecan-9-yloxy)-3-oxopropanoyl)oxy)ethyl)(2- hydroxyethyl)amino) ethyl octyl adipate (Compound 53)
Figure imgf000115_0001
To a solution of 2-((2-(benzyloxy)ethyl)(2-((3-(heptadecan-9-yloxy)-3- oxopropanoyl)oxy)ethyl) amino)ethyl octyl adipate (53-10, 200 mg, 0.25 mmol) in ethyl acetate (20 mL) at room temperature was added palladium hydroxide on carbon (20 %) (100 mg, 0.14 mmol) and the reaction mixture stirred under hydrogen atmosphere for 24 h. MS showed complete conversion. The reaction mixture was filtered through a bed of celite, washed with ethyl acetate, dichloromethane. The solvent was removed and the crude product purified by flash chromatography to get pure 2-((2-((3-(heptadecan-9-yloxy)-3- oxopropanoyl)oxy)ethyl)(2-hydroxyethyl)amino)ethyl octyl adipate (Compound 53, 100 mg, 56%) as colorless oil. 1H ( N40M0R MHz, CD3OD, ppm): δ 4.88-4.91 (m, 1H), 4.20 (t, 2H, J 5.8 Hz), 4.13 (t, 2H, J 5.6 Hz), 4.04 (t, 2H, J 6.8 Hz), 3.51-3.53 (m, 2H), 3.37 (s, 2H), 2.72-2.84 (m, 8H), 2.29-2.33 (m, 4H), 1.51-1.65 (m 9H), 1.24-1.28 (m, 34H), 0.85-0.88 (m, 9H). APCI: m/z 714.6, HPLC-ELSD purity = >99%. HPLC-CAD purity = >97%. HPLC conditions:
Column: Agela C18 column, 4.6 X 50 mm, 3 pm.
Eluents: A: Acetonitrile with 0.1 % TFA, B: Water with 0.1 % TFA.
Flow rate: 1 mL/min; Column Temperature: Room temperature (25 °C, not controlled)
Gradient elution on HPLC:
Figure imgf000115_0002
ELSD detector parameters: Evaporator temperature = 60C, Nebulizer Temperature = 60C, Gas
Flow Rate = 1.60 SLM
UPLC conditions: Aquity C18 column, 3.0 X 150 mm, 1.7 pm.
Eluents: A: Water with 0.1 % TFA, B: Acetonitrile with 0.1 % TFA.
Flow rate: 0.5 mL/min; Column Temperature: 55 °C; Preheater Temperature: 55 °C
Gradient elution on UPLC:
Figure imgf000116_0001
CAD detector: Evaporator temperature setting 35°C
Formulation of LNPs with a messenger RNA
This is a general description of the use of an ionizable cationic lipid of the invention (molecular weight range approx. 500-1000) to formulate an LNP dispersion with a smallmedium size mRNA (for example 500-4000 nucleotides) An ionizable lipid of the invention, 1,2- distearoyl-sn-glycero-3-phosphocholine (DSPC), cholesterol, and 1,2-dimyristoyl-rac- glycero-3-methylpolyoxyethylene (DMG-PEG) are combined in a 40:15:43:2 molar ratio in anhydrous ethanol at a concentration 12.5 mM (1.85 mg/mL). mRNA was diluted in RNase- free 50 mM citrate buffer pH 4.0 to obtain a lipid:mRNA mass ratio of 10:1 (ionizable lipidmucleotide 3:1 molar ratio, often referred to as N:P ratio). The lipid solution in ethanol is then rapidly mixed in a 3:1 (aqueous to ethanol) volume ratio through a micromixer chip using a Nanoassemblr or Egnyte benchtop instrument (Precision Nanosystems) at a total flow rate (TFR) of 12 mL/min. This combination of concentrations and mixing ratio results in a 3:1 molar ratio of ionizable cationic lipid to mRNA phosphate groups and a total lipid to mRNA mass ratio of approximately 10:1. An alternative approach to achieve 3:1 molar ratio of ionizable cationic lipid to mRNA phosphate groups and a total lipid to mRNA mass ratio of approximately 20:1 uses 25 mM lipid stock (double the concentration in ethanol) with the same mRNA concentration at the start and the same mixing configuration. This composition may be preferably in some cases for more complete encapsulation. The mixed LNP dispersion in ethanol/water resulting from the encapsulation step is diluted 10-fold into 50 mM citrate buffer at pH 6 and subjected to tangential flow filtration (TFF) using a 300k molecular weight cut-off membrane (mPES) until concentrated to the original volume. Subsequently, the citrate buffer is replaced with a buffer containing 10 mM Tris buffer at pH
7.5 (measured at 20-25C), 80 mM sodium chloride, and 3% sucrose using diafiltration with 10 diavolumes. The LNP dispersion is concentrated to a volume that leads to no more than about 0.5 mg/mL mRNA concentration, as measured by Ribogreen (QUANT-IT kit, ThermoFisher) with TritonX to access the RNA for quantification. Then the LNP is filtered using a 0.2 micron PES syringe filter, aliquoted into vials, and frozen at l°C/min using a Corning® CoolCell® LX Cell Freezing Container until the samples reach -80°C. Samples are stored at -80°C and thawed on wet ice before analysis or use. The total RNA concentration, the percentage of input RNA recovered (% recovery), and the encapsulation efficiency (%encaps.) are determined using a Ribogreen assay, which is described elsewhere. The Z-avg diameter (nm) and polydispersity index (PDI) are measured using dynamic light scattering (Malvern Zetasizer) following a 1:100 dilution in phosphate buffered saline (PBS).
Biophysical characterization of lipid nanoparticles
The messenger RNA-containing LNP composition is characterized using analytical methods to determine the loading of messenger RNA, the percentage of messenger RNA that is encapsulated, and the size of the particles. The total amount of messenger RNA contained in the sample and the percentage of that messenger RNA that is encapsulated is determined using a fluorescence assay employing Ribogreen, a dye that becomes more emissive upon binding messenger RNA and the fluorescence relates quantitatively to the amount of RNA to which the dye binds. The total amount of messenger RNA is determined by disrupting the LNP with 1 wt% Triton-X 100 to expose the encapsulated messenger RNA, adding the dye, and comparing the emission intensity against a standard curve prepared using ribosomal RNA. The amount of unencapsulated messenger RNA is measured in a similar manner with the detergent disruption of the LNP is omitted. With the total amount of messenger RNA known and the amount of unencapsulated messenger RNA known, the percent encapsulated messenger RNA is calculated by the following formula:
Percent Encapsulation (%) = ((RNATOTAL - RNAUNENCAPSULATED)/ RNATOTAL) X 100 where RNATOTAL and RNAUNENCAPSULATED are, respectively, the concentrations of total messenger RNA and unencapsulated messenger RNA. The total messenger RNA content varies based on formulation, but generally fall in the range of 0.030-0.200 mg/mL. The size of LNP is measured using dynamic light scattering of a sample diluted 1:100 in PBS buffer. pKa' Protocol
In a black 96-well plate, solutions of mRNA-LNP (final assay concentration 2 pg/mL total RNA) in a series of buffers ranging from pH 4 to 9.5 are prepared. Buffers between pH 4 to
7.6 are prepared from disodium phosphate and citric acid. Buffers from 7.8 to 9.5 are prepared by titrating Tris buffer with 10 N sodium hydroxide. To each well is added 6-(p- Toluidino)-2-naphthalenesulfonic acid sodium salt (TNS) in water to a final assay concentration of 6 pM. The fluorescence is read on a plate reader at 25°C with an excitation setting of 321 nm and an emission setting of 445 nm. The intensity values are plotted as a function of pH using GraphPad Prism and are fit with a sigmoidal dose response curve. The apparent pKa (pKa') is determined as the EC50 of this curve where half of the ionizable amines are expected to be protonated.
Characterization of the in vitro cellular potency of LNPs
The ability of an LNP to transfect cultured cells with a TNA, e.g., mRNA, is characterized using an in vitro assay based on the percentage of cells expressing the protein of interest. Specifically, 1 million BHK-21 cells are co-incubated with mRNA-LNP of varying concentration (highest 6.25 ng per well, lowest 0.39 ng per well) in 0.3 mL of media for 17- 19 hours at 37°C with 5% CO2.
Characterization of the in vivo potency of lipid nanoparticles
The in vivo potency of messenger RNA-LNPs may be evaluated using two related, but distinct, approaches. Firstly, to quantify the location, relative amount, and the duration of protein expression, LNPs formulated with a messenger RNA expressing luciferase is injected into mice, for example intramuscularly in the hind leg muscles. At defined time points, such as daily, the mice are administered luciferin, and the bioluminescence is imaged and quantified. Secondly, the ability of messenger RNA-LNPs to act as a vaccine is evaluated by measuring the antibody- and cell based immune response following a prime-boost vaccination schedule. For example, a priming vaccination given on Day 0 via intramuscular injection (i.m.) is followed 21 days later with a boosting vaccination. After an additional 21 days (Day 42 of the experiment), the mice are sacrificed and relevant tissues, such as serum, the spleen, or lymph nodes are collected for further analysis. Serum is analyzed using an ELISA to determine the antigen- specific antibody response. Splenocytes are analyzed for antigen- specific cytokine production, for example by using flow cytometry. Taken together, these in vivo assays can demonstrate that the messenger RNA-LNP is able to induce protein expression of the antigen of interest that initiates a productive antigen- specific immune response required for effective vaccination.
All patents and other publications; including literature references, issued patents, published patent applications, and co-pending patent applications; cited throughout this application are expressly incorporated herein by reference for the purpose of describing and disclosing, for example, the methodologies described in such publications that might be used in connection with the technology described herein. These publications are provided solely for their disclosure prior to the filing date of the present application. Nothing in this regard should be construed as an admission that the inventors are not entitled to antedate such disclosure by virtue of prior disclosure or for any other reason. All statements as to the date or representation as to the contents of these documents is based on the information available to the applicants and does not constitute any admission as to the correctness of the dates or contents of these documents. The description of embodiments of the disclosure is not intended to be exhaustive or to limit the disclosure to the precise form disclosed. While specific embodiments of, and examples for, the disclosure are described herein for illustrative purposes, various equivalent modifications are possible within the scope of the disclosure, as those skilled in the relevant art will recognize. For example, while method steps or functions are presented in a given order, alternative embodiments may perform functions in a different order, or functions may be performed substantially concurrently. The teachings of the disclosure provided herein can be applied to other procedures or methods as appropriate. The various embodiments described herein can be combined to provide further embodiments. Aspects of the disclosure can be modified, if necessary, to employ the compositions, functions and concepts of the above references and application to provide yet further embodiments of the disclosure. Moreover, due to biological functional equivalency considerations, some changes can be made in protein structure without affecting the biological or chemical action in kind or amount. These and other changes can be made to the disclosure in light of the detailed description. All such modifications are intended to be included within the scope of the appended claims. Specific elements of any of the foregoing embodiments can be combined or substituted for elements in other embodiments. Furthermore, while advantages associated with certain embodiments of the disclosure have been described in the context of these embodiments, other embodiments may also exhibit such advantages, and not all embodiments need necessarily exhibit such advantages to fall within the scope of the disclosure. The technology described herein is further illustrated by the following examples which in no way should be construed as being further limiting. It should be understood that this disclosure is not limited in any manner to the particular methodology, protocols, and reagents, etc., described herein and as such can vary. The terminology used herein is for the purpose of describing particular embodiments only and is not intended to limit the scope of the present disclosure, which is defined solely by the claims.
LNP formulation with astaxanthin prodrug in LNPs.
The following description is an example of formulating an LNP dispersion with a smallmedium size mRNA (for example 500-4000 nucleotides) and a fifth lipid component additive. An ionizable lipid (IL) of the invention and/or known lipid (e.g., DLin-MC3-DMA ("MC3")) or LP01, 1,2- distearoyl-sn-glycero-3-phosphocholine (DSPC), cholesterol (chol), astaxanthin amino prodrug (AAP, e.g. compound 90 or 89, which are ionizable) and 1,2- dimyristoyl-rac-glycero-3-methylpolyoxyethylene (DMG-PEG) are combined in ratios that produces a nanoparticle dispersion having suitable size and polydispersity, exhibiting RNA encapsulation and activity in a biological assay.
Figure imgf000119_0001
DLin-MC3-DMA ("MC3")
Figure imgf000120_0001
1,2- distearoyl-sn-glycero-3-phosphocholine ("DSPC")
Figure imgf000120_0002
dioleyl-phosphatidylethanolamine ("DOPE")
A particular example is IL:DSPC:chol:AAP:DMG-PEG, having molar ratio of 45:12:39:2:2, in anhydrous ethanol at a concentration 12.5 mM (1.85 mg/mL). Modified mRNA encoding for firefly Luciferase using 5-methoxyuridine substitutions for all uridines, was diluted in RNase- free 50 mM acetate buffer pH 4.0 to obtain a lipid:mRNA mass ratio of 10:1 (ionizable lipidmucleotide 4.5:1 molar ratio, often referred to as N:P ratio). The lipid solution in ethanol is then rapidly mixed in a 3:1 (aqueous to ethanol) volume ratio through a micromixer chip using a Nanoassemblr, Egnyte benchtop instrument (Precision Nanosystems) or similar mixer setup at a total flow rate (TFR) of 12 mL/min. This combination of concentrations and mixing ratio results in a 3:1 to 4.5:1 molar ratio of ionizable lipid to mRNA phosphate groups and a total lipid to mRNA mass ratio of approximately 10:1 - 15:1. The mixed LNP dispersion in ethanol/water resulting from the encapsulation step is diluted 10-fold into 50 mM citrate buffer at pH 6 and subjected to dialysis against phosphate buffered saline (PBS). Alternatively, tangential flow filtration (TFF) using a 300kDa molecular weight cut-off membrane (mPES) is applied until concentrated to the original volume. Subsequent to TFF, the citrate buffer is replaced with a buffer containing 10 mM Tris buffer at pH 7.5 (measured at around 20°C), 80 mM sodium chloride, and 3% sucrose using diafiltration with 10 diavolumes. The LNP dispersion is concentrated to a volume that leads to no more than about 0.5 mg/mL mRNA concentration, as measured by Ribogreen (QUANT-IT kit, ThermoFisher) with TritonX to rupture LNPs and access the encapsulated RNA for quantification. Then the LNP is filtered using a 0.2 micron PES syringe filter, aliquoted into vials, and frozen at l°C/min using a Corning® CoolCell® LX Cell Freezing Container until the samples reach -80°C. Samples are stored at -80°C and thawed on wet ice before analysis or use. The total RNA concentration, the percentage of input RNA recovered (% recovery), and the encapsulation efficiency (%encapsulation) are determined using a Ribogreen assay. The Z-avg diameter (nm) and polydispersity index (PDI) are measured using dynamic light scattering (Malvern Zetasizer) following a 1:100 dilution in phosphate buffered saline (PBS). LNPs are then stored refrigerated and in the dark.
Biophysical characterization of lipid nanoparticles
The messenger RNA-containing LNP composition is characterized using analytical methods to determine the loading of messenger RNA, the percentage of messenger RNA that is encapsulated, and the size of the particles. The total amount of messenger RNA contained in the sample and the percentage of that messenger RNA that is encapsulated is determined using a fluorescence assay employing Ribogreen, a dye that becomes more emissive upon binding messenger RNA. The total amount of messenger RNA is determined by disrupting the LNP with 1 wt% Triton-X 100 to expose the encapsulated messenger RNA, adding the dye, and comparing the emission intensity against a standard curve prepared using ribosomal RNA. The amount of unencapsulated messenger RNA is measured in a similar manner with the detergent disruption of the LNP is omitted. With the total amount of messenger RNA known and the amount of unencapsulated messenger RNA known, the percent encapsulated messenger RNA is calculated thus:
Percent Encapsulation (%) = ((RNATOTAL - RNAUNENCAPSULATED)/ RNATOTAL) X 100 where RNATOTAL and RNAUNENCAPSULATED are, respectively, the concentrations of total messenger RNA and unencapsulated messenger RNA. The total messenger RNA content varies based on formulation, but generally fall in the range of 0.030-0.200 mg/mL. The size distribution parameters of LNP are measured using dynamic light scattering of a sample diluted 1:100 in PBS buffer.
Fluorescence imaging and spectroscopy can be used to identify and quantify xanthophyll components in the LNP. The fluorescence is read on a plate reader at 25°C with an excitation setting of 450 nm and an emission setting of 560 nm.
Table 1. LNP compositions used to generate data presented in Figures 1-7 and 9-11. RNA content and encapsulation was generated with RiboGreen assay.
Figure imgf000121_0001
Figure imgf000122_0001
Table 2. LNP compositions used to generate data presented in Figures 12 and 13. RNA content and encapsulation was generated with RiboGreen assay
Figure imgf000123_0001
Table 3. LNP compositions used to generate data presented in Figure 14.
Figure imgf000123_0002
Figure imgf000124_0001
Expression of Luciferase from modified mRNA by in vitro cell transfection with LNPs.
LNPs from the preparations entrapping the Firefly Luciferase mRNA with uridine modifications were dosed into wells containing a single cell line, either HEK293 or Huh7, at doses of 125, 250 and 500 ng of mRNA, in addition to a PBS blank dose denoted as 0 ng mRNA. After 6 hours, the luminescence from the expressed Luciferase is tested after dosing of Luciferin dye, a substrate for the enzyme which is luminescent, and the signal is proportional to the enzyme activity. Viability of cells is measured by Promega OneGlo + Tox luciferase detection kit.
A set of experiments was done to test the potential of LNPs compositions with proprietary lipids designed by and synthesized.
The choice of helper lipid, specifically the phospholipids, was aimed to compare DSPC (used in most formulations with RNA, including ONPATTRO and the mRNA vaccines for Covidl9) as well as DOPC and DOPE.
Additionally, evaluation of MC3 (D-Lin-MC3-DIVIA) as a potent and ionizable cationic lipid was done. MC3 is used for the effective delivery of RNAs in vivo, although it is known to be reactogenic induce immuno-inflammatory responses (either TH1 or TH2).
In the studies, all the ionizable lipid formulations tested containing MC3 with or without AAP modifiers, Compound (89) and Compound (90) (Figure 1A) or with compositions and structures of Axelyf ionizable lipids as per Table 1 (Figure IB) yielded particles withing expected optimal range. Additionally, the polydispersity (PDI) measured using Stunner (rotating angle dynamic light scattering, Unchained Labs) Protocol was also within ranges expected from this type of lipid nanoparticles (Figure 2A and Figure 2B). Importantly, most were able to yield an RNA encapsulation efficiency >80%, which is a lower threshold for what is considered good encapsulation of mRNA (Table 1). The presence of astaxanthin compounds did not dramatically affect the measured properties of the particles, although Compound (90) addition resulted in overall larger particles. Notably, assessment of size (Figure 3A) and polydispersity (Figure 3B) of LNPs with and without AAP modifier component on day 14 after formulation showed that particles retained their overall characteristic in storage.
All MC3-based LNPs showed similar pKa' values in line with literature reports (value range of 6.3-6.6).
Functional expression of luciferase was measured by luminescence, a direct enzymatic activity readout, in human embryonic kidney 293T cells (HEK293T) and in hepatic cell line Huh-7 (both epithelial-like), with increasing mRNA payload (125-250-500ng). LNP formulations containing both standard and reduced molar ratio of MC3, with or without Axelyf AAP proprietary ionizable lipids induced dose-dependent luciferase expression luciferase expression, with or without AAP modifier components showed satisfactory in HEK293T cells (Figure 4A and 4B, respectively) with notably lower expression for Compound (52) in formulations 7 and 9. Dose dependent luciferase expression with (Figure 5A) or without (Figure 5B) AAP modifier was also observed in Huh7. In this cell type, formulation with Compound (52), i.e., formulations 7 and 9 showed the highest luciferase expression, suggesting possible in vivo differential tropism.
All the LNPs formulation tested showed acceptable minimal toxicity at the tested RNA concentrations based on cell viability measurements in HEK293T cells (Figure 6A and Figure 6B.) and in Huh7 cells (Figure 7A and 7B), regardless of the presence of AAP modifiers or lack thereof.
After a one-month storage at +4°C, the size and polydispersity of particles showed maintained consistency, with the exception of MC3 Compound (90 containing LNPs, which, already larger than other compositions, showed an increase in size over the observation period/ Encapsulation efficiency of particles did not appear to change significantly on storage.
After one month, dose-dependent expression of luciferase was consistent with earlier experiments in all MC3-based LNPs, with only minor differences among the various formulations (in both cell types). Notably, expression of luciferase in Huh-7 induced by MC3 LNPs, including the ones with the astaxanthin derivatives, was higher than in 293T cells.
Conversely, formulation Compound (30) using DSPC yielded a strikingly high expression of luciferase in HEK293T cells as compared with all other formulations, suggesting good mRNA stability, and formulation Compound (52) DOPE drove expression in Huh7, whereas other formulations had weaker activity. After one month at +4°C, different formulations retained different tropism.
In order to further evaluate the contribution of astaxanthin derivative Compound (89), a further set of experiments was performed comparing MC3 LNPs containing increasing molar percentages (2%, 4-%, 8%) of Compound (89), with unmodified 2% astaxanthin as control. Visible light assessment of the color of formulations with increasing concentrations of Compound (89)-and astaxanthin showed a progressively increasing red color of Compound (89) formulations.
Fluorescence spectral scan revealed emission of Compound (89) and Astaxanthin 2% in EtOH around 575 nm with 480 nm excitation, but little fluorescence of the lipid nanoparticles containing these compounds (Figure 9).
All of the ionizable lipid formulations tested produced measurable particles, with the size (Figure 11, left) and PDI (Figure 11, right) within expected from this type of lipid nanoparticles. All LNPs were able to yield an RNA encapsulation efficiency >90% (Table 1). Astaxanthin compounds did not dramatically affect the measured size or encapsulation properties of the particles.
Transfection potential and cytotoxicity of all LNPs were measured once again measured in HEK293T and Huh7. All Compound (89) formulations showed slightly higher expression of luciferase in both cell lines in compared to MC3 control, with 4% Compound (89) showing also better viability. Astaxanthin inclusion in LNPs did not provide the same increase in luciferase expression over control that was seen with Compound (89) in either cell line (Figure 12). In HEK293T and Huh7 cells there appeared to be minimal toxicity of LNPs with Compound (89) at tested RNA concentrations based on cell viability measurements when compared to MC3 LNPs alone. However, formulation with 8% Compound (89) showed some toxicity in both cell lines at the highest concentration (500ng; Figure 13).
LNPs compositions with ionisable lipids Cpd 51 or Cpd 53 were made and compared to LNP with MC3 and LP-01 which are widely used in the literature.
LNP containing Cpd 51 or Cpd 53 (with either DOPE, DOPC, or DSPC) had Z-average values in line with control LNPs (Figure 13A). PDI values were lower than MC3 LNPs, but in line with
LP-01 LNPs (Figure 13B).
LNPs containing Cpd 51 or Cpd 53 were equipotent to LP-01 in terms of in vitro expression of Luciferase in HEK293T and Huh7 cells as a result of transfection of different amount of modified mRNA.

Claims

We claim:
1. A lipid having a structural formula selected from the group consisting of:
Figure imgf000127_0001
and (4); or a pharmaceutically acceptable salt or cocrystal thereof, wherein, for each occurrence:
* denotes a chiral center;
R1 for each occurrence, is independently selected from: -H, optionally substituted C1-C6 alkyl, optionally substituted C2-C& alkenyl, -C(=O)CH2N(R72)2,
Figure imgf000127_0003
-P(O)(OR17)(OR18), -P(O)(OR17)(NR19), or -P(O)(NR19)(NR20);
R3 for each occurrence, is independently selected from: -H, optionally substituted C1-C6 alkyl, -[CI-hJi-eOH, optionally substituted C2-C6 alkenyl, -P(O)(OR17)(OR18), - P(O)(OR17)(NR19), -P(O)(NR19)(NR20) or R4;
R2 is selected from: is absent (i.e., a lone pair on N), -H or optionally substituted C1-C6 alkyl; provided that when R2 is -H or optionally substituted C1-C6 alkyl, the nitrogen atom, to which R1, R2, and R3 are all bonded to, is protonated;
R4 for each occurrence, is independently selected from:
Figure imgf000127_0002
or
R55 for each occurrence, is independently selected from:
Figure imgf000128_0001
Figure imgf000129_0001
A1, A3, A4, A5, A6, A7, A8, A12 and A13 for each occurrence, are each independently selected from: a bond, -O-, -C(=O)-, -C(=S)-, -OC(=O)-, -C(=O)O-, -OC(=O)O-, -OC(=S)-, -C(=S)O-, - OC(=S)O-,-S-C(=O)-, -C(=O)-S-, -S-S-, -S(OH)-, -S(=O)-, -S(=O)2-, -S(OH)2-, -C(=O)N(R14)-, - N(R14)C(=O)-, -N(R14)C(=O)N(R14)-, -O-C(=O)C(R14)2C(=O)O-, -C(=O)O-C(R14)2C(=O)O-, -O- C(=O)C(R14)2-O-C(=O)-, -O-C(R14)2C(=O)O-, -O-C(=O)C(R14)2C-O-, -O-C(R14)2-O-C(=O)-, - C(=O)-O-C(R14)2C-O-, -O-C(=O)-O-R41-O-C(=O)O-, -0-C(=0)-[CH2]O-4-C(=0)-0-, -P(OH)-, - P(OH)(R14)-, -P(=O)(OH)-, -P(O R14)2-, -P(O R14)- or -P(=O)(O R14)-;
A2 for each occurrence, is independently selected from: a bond, -O-, -OC(=O)-, -C(=O)O-, - OC(=O)O-, -S-C(=O)-, -C(=O)-S-, -S-S-, -C(=O)N(R14)-, -N(R14)C(=O)-, -N(R14)C(=O)N(R14)-, -O-C(=O)C(R14)2C(=O)O-, -C(=O)O-C(R14)2C(=O)O-, -O-C(=O)C(R14)2-O-C(=O)-, -O- C(R14)2C(=O)O-, -O-C(=O)C(R14)2C-O-, -O-C(R14)2-O-C(=O)-, -C(=O)-O-C(R14)2C-O- or -O- C(=O)-O-R41-O-C(=O)O-;
R6 for each occurrence, is independently selected from: a bond, -R58-N(R42)-R58-, -R58- CH(R42)-R58-, Ci-Ci2 alkylene or C2-Ci2 alkenylene;
R7 for each occurrence, is independently selected from: a bond, optionally substituted Ci- Ci8 alkylene, or optionally substituted C2-Cis alkenylene;
R8 and R9 for each occurrence, are each independently selected from:
Figure imgf000129_0002
R42;
R12 for each occurrence, is independently selected from: -H, optionally substituted Ci-Cie alkyl or optionally substituted C2-C16 alkenyl;
R13 for each occurrence, is independently selected from: optionally substituted C1-C16 alkyl or optionally substituted C2-C16 alkenyl;
R14, for each occurrence, is independently selected from: -H, optionally substituted C1-C10 alkyl, optionally substituted C1-C10 alkenyl or optionally substituted C1-C10 alkynyl; R15 for each occurrence, is independently selected from: a bond, -R41-N(R42)-R58-, -R41- CH(R42)-R58-, or C1-C6 alkylene;
R16 for each occurrence, is independently selected from: a bond or -[CH2]k-;
R17 and R18 for each occurrence, are each independently selected from: -H, C1-C5 alkyl, an alkali metal cation, an alkaline earth metal cation, ammonium cation, methyl ammonium cation, or a pharmaceutically acceptable base;
R19 and R20 for each occurrence, are each independently selected from: -OH, formyl, acetyl, pivaloyl, -NH2, -NH(CH3), -NH(CH2CH3), -N(CH3)2, -NHC(=O)H, -NHC(=O)CH3 or C3-C5- alkyl;
R29, R30, R31, R32, R33 and R34for each occurrence, are each independently selected from: - CH2-, -NH-, -S-, or -O-;
R35 for each occurrence, is independently selected from: -OH or C1-C6 acyl; optionally substituted C1-C6 alkyl, C1-C6 alkyl ester, C1-C6 alkyl ether or C1-C6 alkyl carbonate;
R36 and R37 are each independently selected from: -H, -C(=O)C1-C6 alkyl, -C(=O)C2-C6 alkenyl, -C(=O)-O-C1-C6 alkyl, or -C(=O)-O-C2-C6 alkenyl, C1-C6 alkyl, or C2-C6 alkenyl;
R38 for each occurrence, is independently selected from: -H, C1-C24 alkylene or C1-C24 alkenylene;
R41 for each occurrence, is independently selected from: a bond or C1-C5 alkylene;
R42 for each occurrence, is independently selected from: C6-C24 alkyl, C6-C24 alkyl carbonate, C6-C24 alkyl ether, C6-C24 alkyl ester, -R58-A1-R59 or -R58-A1-R59-A2-R60, wherein R42 has 6-24 total carbon atoms;
R58 R59, and R60 for each occurrence, are each independently selected from: a bond, -O-, - O-C(=O)-, -OC(=O)O-, Ci-C24 alkylene or C2-C24 alkenylene;
R62, R63, R64, R67, R68, R69, R74, R75, R76, R77, R108, R109, R110 and R111 for each occurrence, are each independently selected from: a bond, cycloalkylene , C1-C24 alkylene, C1-C24 alkenylene, -R114-C3-C6-cycloalkylene-R115-, -R114-CH=CH-CH2-CH=CH-R115- , -R114- CH=CH-CH2-cPr-R115-, -R114-cPr-CH2-CH=CH-R115-, -R114-cPr-CH2-cPr-R115-, -R114-CH=CH- CD2-CH=CH-R115-, -R114-CH=CH-CD2-cPr-R115-, -R114-cPr-CD2-CH=CH-R115- or -R114-cPr- CD2-cPr-R115-; where -cPr- is cyclopropylene (cyclopropane-1,2 diyl); each double bond or cPr has a cis configuration; and D denotes deuterium (2H);
R114 and R115 for each occurrence, are each independently selected from: a bond, C1-C10 alkylene, C1-C10 alkenylene C1-C10 alkynylene; R61, R73 and R107 for each occurrence, are each independently selected from:
Figure imgf000131_0001
Figure imgf000131_0002
R116 for each occurrence, is independently selected from: -H, -F, -CF3, -Cl, -Br, -I, -CH3, - CH2R14, -CHR14 2, -CR14 3, -OH, -OR14, -NH2, -NHR14, -N(R14)2, -SH, -SR14, -S(=O)H, - S(=O)R14, -S(=O)2H, -S(=O)2R14, -PH2, -PHR14, -PR14 2, -P(OH)2, -POHR14, -PR14 2, -P(OR14)2, -P(O)H2, -P(O)HR14, -P(O)R14 2, -P(O)(OH)2, -P(O)(OR14)OH, -P(O)(OR14)2, -SiH3, -SiH2R14, - SiHR14 2 or-SiR14 3;
R65, R70, R78, R105 and R112 for each occurrence, are each independently selected from: a bond, -CH=CH-CH2-CH=CH- , C3-C6-cycloakylene, -CH=CH-CH2-cPr-, -cPr-CH2-CH=CH-, - cPr-CH2-cPr-, -CH=CH-CD2-CH=CH- , -CH=CH-CD2-cPr-, -cPr-CD2-CH=CH- -or -cPr-CD2- cPr-, where -cPr- is cyclopropylene, each double bond or cPr has a cis configuration and D denotes deuterium (2H);
R66, R71, R79, R106 and R113 for each occurrence, are each independently selected from: -H, linear or branched C1-C24 alkyl, C1-C24 alkenyl, or C1-C24 alkynyl, C5-C2o-spirocycloalkyl, C3-Ci8-cycloalkyl, C3-Ci8-heterocycloalkyl, optionally substituted aryl, optionally substituted heteroaryl, -R26-CH=CH-CH2-CH=CH-R28, -R26-CH=CH-CH2-cPr-R28, -R26-cPr- CH2-CH=CH-R28, -R26-cPr-CH2-cPr-R28, -R26-CH=CH-CD2-CH=CH-R28, -R26-CH=CH-CD2-cPr- R28, -R26-cPr-cPr-R28, -R26-cPr-cBu-R28, -R26-cBu-cBu-R28, -R26-cBu-cPr-R28, -R26-cHx-cBu- R28, -R26-cHx-cBu-R28, -R26-cHx-cBu-R28, -R26-cPr-cHx-R28, -R26-cBu-cHxR28, -R26-cPr-R26- cPr-R28, -R26-cPr-R26-cBu-R28, -R26-cBu-R26-cBu-R28, -R26-cBu-R26-cPr-R28, -R26-cHx-R26- cBu-R28, -R26-cHx-R26-cBu-R28, -R26-cHx-R26-cBu-R28, -R26-cPr-R26-cHx-R28, -R26-cBu-R26- cHxR28, -R26-c-CD2-cPr-R28, -R26-cPr-CD2-cPr-R28, -R26-cPr-CD2-cPr-R28, -R26-cPr-CD2- CH=CH-R28, -R26-cPr-CD2-cPr-R28 or -R26-C3-Ce cycloakylene-R26-C3-Ce cycloakylene-R28; wherein, R26 for each occurrence, is independently selected from: a bond, C1-C10 alkylene, C1-C10 alkenylene or C1-C10 alkynylene; and R28 for each occurrence, is independently selected from: -H, C1-C10 alkyl, C1-C10 alkenyl or C1-C10 alkynyl; -cPr- is cyclopropylene (cyclopropane-1,2 diyl); -cBu- is cyclobutylene; -cHx- is cyclohexylene; each double bond or -cPr- has a cis configuration; and D denotes deuterium (2H);
R72 for each occurrence, is independently selected from; -H, optionally substituted C1-C6 alkyl, -[CH2]I-6OH or optionally substituted C2-C6 alkenyl;
R96 for each occurrence, is independently selected from: -OH, -O-C1-C4 alkyl, -O-C(=O)-O- C1-C4 alkyl or -O-C(=O)-C1-C5 alkyl;
R118 and R119 are independently selected from R1 or R55; n is an integer selected from: 1, 2, 3, 4, 5 or 6; and k is an integer selected from: 1, 2, 3, 4, 5 or 6.
2. The lipid of claim 1, comprising one or more cycloalkylene independently selected from:
Figure imgf000132_0001
3. The lipid of claim 1, wherein, -R62-A3-R63-A4-R64-R65-R66, -R62-A3-R63-A4-R64-R73-R74-A7- R75-R65-R66, -R62-A3-R63-A4-R64-R73-R76-A8-R77-R78-R79, -R67-A5-R68-A6-R69-R70-R71, -R67-A5- R68-A6-R69-R107-R108-A12-R109-R105-R106, and -R67-A5-R68-A6-R69-R107-R110-A13-R111-R112-R113, for each occurrence, independently has: f) a total number of carbon atoms selected from: 10-24, 12-22, 14-22, 16-20, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23 or 24 carbon atoms; g) a longest linear chain with a total number of carbon atoms that is selected from: 10- 24, 12-22, 14-22, 16-20, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23 or 24 carbon atoms; or h) a longest linear chain with a total number of atoms that is selected from: 10-24, 12-22, 14-22, 16-20, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23 or 24 atoms.
4. The lipid of claim 1, comprising two R55 variables selected from: a) -R62-A3-R63-A4-R64-R73-R74-A7-R75-R65-R66 and -R62-A3-R63-A4-R64-R73-R76-A8-R77-R78-R79, b) -R74-A7-R75-R65-R66 and -R76-A8-R77-R78-R79, c) -R62-A3-R63-A4-R64-R65-R66 and -R67-A5-R68-A6-R69-R70-R71, d) -R67_A5_R68_A6_R69_R1O7_R1O8_A12_R1O9_R1O5_R1O6 a n d _R67_A5_R68_A6_R69_R107_R110_A13_Rlll_
R112-R113, or e) -R1O8.A12.R1O9.R1O5.R1O6 a n d _R110_A13_Rlll_R112_Rll3
5. The lipid of claim 1, wherein R55, for each occurrence, is independently selected from:
Figure imgf000132_0002
d) -R67-A5-R68-A6-R69-R70-R71, e) -R67-A5-R68-A6-R69-R107-R108-A12-R109-R105-R106 f) _R67_A5_R68_A6_R69_RIO7_RIIO_AI3-RIII_RII2_RII3; g) -A3-R63-A4-R64-R73-R74-A7-R75-R65-R66, h) -A3-R63-A4-R64-R73-R76-A8-R77-R78-R79, i) -A3-R63-A4-R64-R65-R66, j) -A5-R68-A6-R69-R70-R71, k) -A5-R68-A6-R69-R107-R108-A12-R109-R105-R106 l) -A5-R68-A6-R69-R107-R110-A13-R111-R112-R113, m) -R74-A7-R75-R65-R66, n) -R76-A8-R77-R78-R79, o) _R108_A12_R109_R105_R106
_
P) Riio_Ai3-Riii_Rii2_Rii3 q) -R74-R66 or r) -R76-R79.
6. The lipid of claim 1, wherein R55, for each occurrence, is independently selected from:
-OC(=O)C(R42)[CH2]nC(=O)O-R41-CH(C4-Ci2 alkyl)2, -R41-N(R42)([CH2]nC(=O)O-R41-CH(C4-Ci2 alkyl)2, -OC(=O)C(R43)[CH2]nC(=O)O-R41-CH(C4-Ci2 alkyl)2, -R41-N(R43)([CH2]nC(=O)O-R41- CH(C4-CI2 alkyl)2, -R41-C(R42)[CH2]nC(=O)O-R41-CH(C4-Ci2 alkyl)2 or -R41-CH(R43)(R44), - R41-N(R43)(R44); wherein,
R43 for each occurrence, is independently selected from: -C(=O)O-C6-C24 alkyl, -OC(=O)-Ce- C24 alkyl, -OC(=O)O-C6-C24 alkyl, -C(=O)O-C6-C24 alkenyl, -O-C(=O)-C6-C24 alkenyl, - OC(=O)O-C6-C24 alkenyl-, -C2-C22 alkylene-O-C2-C22 alkyl, -C2-C22 alkenylene-O-C2-C22 alkyl, -C2-C22 alkylene-O-C2-C22 alkenyl, -Ci-C22 alkylene-C(=O)O-Ci-C22 alkyl, -Ci-C22 alkylene-O-C(=O)O-Ci-C22 alkyl, -Ci-C22 alkylene-O-C(=O)-Ci-C22 alkyl, -C2-C22 alkenylene-C(=O)O-Ci-C22 alkyl, -C2-C22 alkenylene-O-C(=O)-Ci-C22 alkyl, -C2-C22 alkenylene-O-C(=O)-Ci-C22 alkyl, -Ci-C22 alkylene-C(=O)O-C2-C22 alkenyl, -Ci-C22 alkylene-O-C(=O)O-C2-C22 alkenyl, -Ci-C22 alkylene-O-C(=O)-C2-C22 alkenyl, -C2-C22 alkenylene-C(=O)O-C2-C22 alkenyl, -C2-C22 alkenylene-O-C(=O)O-C2-C22 alkenyl, -C2-C22 alkenylene-O-C(=O)-C2-C22 alkenyl; wherein R43 has 6-24 total carbon atoms;
R44 for each occurrence, is independently selected from: -R45-C(=O)O-R45-CH(C6-CI2 al kyl)2, -R45- OC(=O) - R45-CH(C6-C12 alkyl)2, -R45-OC(=O)O-R45-CH(C6-C12 alkyl)2, R45-C(=O)O-R45- CH(C6-C12 alkenyl)2, -R45- OC(=O) - R45-CH(C6-C12 alkenyl)2, -R45-OC(=O)O- R45-CH(C6-CI2 alkenyl)2; wherein R44 has 6-24 total carbon atoms and wherein R45 is, for each occurrence, independently selected from: a bond, C1-C6 alkylene or C1-C6 alkenylene.
7. The lipid of claim 1, wherein R55, for each occurrence, is independently:
Figure imgf000134_0001
wherein,
A3 and A5 for each occurrence, are each independently selected from: -O-, -OC(=O)-, - C(=O)O- or -OC(=O)O-;
R16 for each occurrence, is independently selected from: a bond or — [CH2]k-; k is an integer selected from: 1, 2, 3, 4, 5 or 6; rence, are each independently selected from:
Figure imgf000134_0002
Figure imgf000134_0003
h occurrence, are each independently selected from: C1-C12 alkylene or C1-C12 alkenylene;
R66, R71 and R79 for each occurrence, are each independently selected from: C1-C12 alkyl or C1-C12 alkenyl.
8. The lipid of claim 7, wherein, R62, R63, R66 and R74, together, have 10-24 total carbon atoms; R62, R63, R76 and R79, together, have 10-24 total carbon atoms; and R67, R68 and R71, together, have 10-24 total carbon atoms.
9. The lipid of claim 7, wherein, -R62-A3-R63-R73-R74-R66, -R62-A3-R63-R73-R76-R79, and -R67- A5-R68-R71, independently has: i) a total number of carbon atoms selected from: 10-24, 12-22, 14-22, 16-20, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23 or 24 carbon atoms; j) a longest linear chain with a total number of carbon atoms that is selected from: 10- 24, 12-22, 14-22, 16-20, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23 or 24 carbon atoms; or k) a longest linear chain with a total number of atoms that is selected from: 10-24, 12-22, 14-22, 16-20, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23 or 24 atoms.
10. The lipid of claim 1, wherein R55, for each occurrence, is independently:
Figure imgf000135_0001
wherein:
A3 and A5 for each occurrence, are each independently selected from: -O-, -OC(=O)-, -
C(=O)O- or -OC(=O)O-;
R16 for each occurrence, is independently selected from: a bond or — [CH2]k-; k is an integer selected from: 1, 2, 3, 4, 5 or 6; each independently selected from:
Figure imgf000135_0002
Figure imgf000135_0003
each occurrence, are each independently selected from: C1-C12 alkylene or C1-C12 alkenylene;
R66, R79, R106 and R113 for each occurrence, are each independently selected from: C1-C12 alkyl or C1-C12 alkenyl; and wherein: R62, R63, R66 and R74, together, have 10-24 total carbon atoms; R62, R63, R76 and R79, together, have 10-24 total carbon atoms; and R67, R68, R108 and R106, together, have 10-24 total carbon atoms; and, R67, R68, R110 and R113, together, have 10-24 total carbon atoms.
11. The lipid of claim 7, wherein, -R62-A3-R63-R73-R74-R66, -R62-A3-R63-R73-R76-R79, -R67-A5-R68- R1O7_R1O8_R1O6 a n d _R67_A5_R68_RIO7_RIIO_RII3 independently have: a) a total number of carbon atoms selected from: 10-24, 12-22, 14-22, 16-20, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23 or 24 carbon atoms; b) a longest linear chain with a total number of carbon atoms that is selected from: 10- 24, 12-22, 14-22, 16-20, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23 or 24 carbon atoms; or c) a longest linear chain with a total number of atoms that is selected from: 10-24, 12-22, 14-22, 16-20, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23 or 24 atoms.
12. The lipid of claim 1, wherein R55, for each occurrence, is independently:
Figure imgf000136_0001
wherein:
A3 and A5 for each occurrence, are each independently selected from: -O-, -OC(=O)-, - C(=O)O- or -OC(=O)O-;
R16 for each occurrence, is independently selected from: a bond or — [CH2]k-; k is an integer selected from: 1, 2, 3, 4, 5 or 6;
R61, R73 and R107 for each occurrence, are each independently selected from:
Figure imgf000136_0002
Figure imgf000136_0003
R62, R63, R67, R68 for each occurrence, are each independently selected from: C1-C12 alkylene or C1-C12 alkenylene.
13. The lipid of claim 12, wherein, -R62-A3-R63-R66 and -R67-A5-R68-R71 independently have: a) a total number of carbon atoms selected from: 10-24, 12-22, 14-22, 16-20, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23 or 24 carbon atoms; b) a longest linear chain with a total number of carbon atoms that is selected from: 10- 24, 12-22, 14-22, 16-20, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23 or 24 carbon atoms; or c) a longest linear chain with a total number of atoms that is selected from: 10-24, 12-22, 14-22, 16-20, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23 or 24 atoms.
14. The lipid of claim 1, wherein R55, for each occurrence, is independently selected from:
Figure imgf000137_0001
15. The lipid of claim 1, having a structural formula selected from:
Figure imgf000138_0001
R16 for each occurrence, is independently selected from: a bond or — [CH2]k-;
R62, R63, R64, R67, R68, R69, R74, R75, R76 and R77 for each occurrence, are each independently selected from: a bond, C1-C24 alkylene or C1-C24 alkenylene;
R61 and R73 for each occurrence, are each independently selected from:
Figure imgf000138_0002
Figure imgf000138_0003
R65, R70 and R78 for each occurrence, are each independently selected from: a bond, - CH=CH-CH2-CH=CH- , -CH=CH-CH2-cPr-, -cPr-CH2-CH=CH-, -cPr-CH2-cPr-, -CH=CH-CD2- CH=CH- , -CH=CH-CD2-cPr-, -cPr-CD2-CH=CH- -or -cPr-CD2-cPr-, where -cPr- is cyclopropylene, each double bond or cPr has a cis configuration and D denotes deuterium (2H); and
R66, R71 and R79 for each occurrence, are each independently selected from: C1-C24 alkyl or Ci-C24 alkenyl.
16. The lipid of claim 15, wherein: -R62-A3-R63-A4-R64-R65-R66, -R62-A3-R63-A4-R64-R73-R74-A7- R75-R65-R66, -R62-A3-R63-A4-R64-R73-R76-A8-R77-R78-R79 and -R67-A5-R68-A6-R69-R70-R71, each independently has a total of 10-24 carbon atoms.
17. The lipid of claim 1, having a structural formula selected from:
Figure imgf000139_0001
Figure imgf000140_0001
(12); wherein:
R61 for each occurrence, is independently selected from:
Figure imgf000140_0002
R85 and R90 for each occurrence, are each independently selected from: a bond or -[CH2]I-S-
R86 and R91 for each occurrence, are each independently selected from: a bond -O-, -O-
C(O=)-, -O-C(=O)-O- or -C(=O)-O-;
R87 for each occurrence, is independently selected from: a bond or -[CH2]I-6-;
R88 and R89 for each occurrence, are each independently -[CH2]I-IOCH3;
R92 is -[CH2]2-I6CH3;
R93 and R94 for each occurrence, are each independently selected from: -H, -D, -CH3, -CD3, - CH2CH3, -CD2CH3, where D denotes deuterium (2H);
R95, R96, R100, R101 and R102 for each occurrence, are each independently selected from: - OH, -O-C1-C4 alkyl, -O-C(=O)-O-Ci-C4 alkyl or -O-C(=O)-CrC4 alkyl;
R97 for each occurrence, is independently selected from: -[CH2]I-6-, -[CH2]I-6-C(=O)-O-, - [CH2] -[CH2]I-6-O- or -[CH2]I-6-C(=O)-, wherein: when R61 then R97 for each occurrence, is independently selected from: - [CH2] I-6-C(=O)-O- and when R61 then R97 for each occurrence, is independently selected from: -
[CH2]
Figure imgf000140_0003
-C(=O)-O-, -[CH2]I-6-O- or -[CH2]I-6-C(=O)-; and
R16 for each occurrence, is independently selected from: a bond or -[CH2]k-;
R98 for each occurrence, is independently selected from: a bond, -C(=O)-, or -C(=O)-O-; and R104 for each occurrence, is independently selected from:
Figure imgf000141_0001
18. The lipid of claim 17, wherein: a) R85 and R90 are the same, b) R85 and R90 are each independently -[CH2]4-5-, c) one or both of R86 and R91 are/is -C(O=)-O-, d) one or both of R86 and R91 is/are a bond, e) R88 and R89 are each independently -[CH2]6-IO-, f) R92 is -[CH2]8-i2-, g) R85, R86, R87, R88 and R89, together, have between 14-24 or between 16-22 total carbon atoms, h) R90, R91 and R92, together, have 12-18 total carbon atoms, i) R88 and R89 are identical, j) R86, R87 and R88, together, and R90, R91 and R92, together, have total number of carbon atoms that differ by +/- 3, k) R93 and/or R94 are/is -H, or l) R95 and/or R96 are/is -OH.
19. The lipid of claim 1, having a structural formula selected from:
Figure imgf000142_0001
Figure imgf000143_0001
wherein:
R80 for each occurrence, is independently selected from: a bond, R16 or -[CH2]I-6-O- or -O-;
R16 for each occurrence, is independently selected from: a bond or -[CH2]k-;
R81, R82, R83 and R84 are each independently selected from: a bond or -[CH2]I-IO-;
R93 and R94 for each occurrence, are each independently selected from: -H, -D, -CH3, -CD3, - CH2CH3, -CD2CH3, where D denotes deuterium (2H);
R95 and R96 for each occurrence, are each independently selected from: -OH, -O-C1-C4 alkyl, -O-C(=O)-O-CrC4 alkyl or -O-C(=O)-Ci-C4 alkyl;
R97 for each occurrence, is independently selected from: -[CH2]I-6-, -[CH2]I-6-C(=O)-O-, -
[CH2] -[CH2]I-6-O- or -[CH2]I-6-C(=O)-, wherein: when R61 then R97 for each occurrence, is independently selected from: - [CH2] I-6-C(=O)-O- and when R61 then R97 for each occurrence, is independently selected from: - [CH2]
Figure imgf000143_0002
-C(=O)-O-, -[CH2]I-6-O- or -[CH2]I-6-C(=O)-; and R98 for each occurrence, is independently selected from: a bond, -C(=O)-, or -C(=O)-O-.
20. The lipid of claim 1, wherein: a) R81 and R83 are each independently -[CH2]s-9, and R82 and R84 are each independently - [CH2]O-6, b) R81 and R83 are each independently -[CH2]S-IO-, and R82 and R84 are each independently -[CH2]O-6-, c) R81 and R82, combined, have 7-13 total carbon atoms, d) R83 and R84, combined, have 7-13 total carbon atoms, e) R81 and R82, combined, have 8-12 total carbon atoms, f) R83 and R84, combined, have 8-12 total carbon atoms, g) R81 and R82, combined, and R83 and R84, combined, have an identical number of total carbon atoms which is between 7-13, or h) R81 and R82, combined, and R83 and R84, combined, have a different number of total carbon atoms.
21. The lipid of claim 1, having a structural formula selected from:
Figure imgf000144_0001
Figure imgf000145_0001
(27) or
Figure imgf000146_0001
(28); wherein:
R85 and R90 for each occurrence, are each independently selected from: a bond or
-[CH2]I-8-;
R86 and R91 for each occurrence, are each independently selected from: a bond -O-, -O- C(O=)-, -O-C(=O)-O- or -C(=O)-O-;
R87 for each occurrence, is independently selected from: a bond or -[CH2]I-6-;
R88 and R89 for each occurrence, are each independently -[CH2]I-I0CH3;
R92 is -[CH2]2.I6CH3;
R93 and R94 for each occurrence, are each independently selected from: -H, -D, -CH3, -CD3, -CH2CH3, -CD2CH3, where D denotes deuterium (2H);
R96 for each occurrence, is each independently selected from: -OH, -O-C1-C4 alkyl, -O- C(=O)-O-C1-C4 alkyl or -O-C(=O)-Ci-C4 alkyl;
R97 for each occurrence, is independently selected from: -[CH2]I_6-, -[CH2]I.6-C(=O)-O-,
-[ -, -[CH2]I-6-O- or -[CH2]I-6-C(=O)-, wherein: when
Figure imgf000146_0002
, then R97 for each occurrence, is independently selected from:
-[CH2]I-6- or -[CH2]I-6-C(=O)-O- and when R61 is ~L , then R97 for each occurrence, is independently selected from: -[CH2]I-6-, -[CH2]I-6-C(=O)-O-, -[CH2]I-6-O- or -[CH2]I-6-C(=O)-; and
R98 for each occurrence, is independently selected from: a bond, -C(=O)-, or -C(=O)-O-.
22. The lipid of claim 1, having a structural formula selected from:
Figure imgf000146_0003
(29),
Figure imgf000147_0001
Figure imgf000148_0001
Figure imgf000149_0001
Figure imgf000150_0001
Figure imgf000151_0001
Figure imgf000152_0001
23. The lipid of claim 1, wherein: a) R1 and R3, for each occurrence, are each independently, -H, optionally substituted Cr Ce alkyl, optionally substituted C2-C6 alkenyl, optionally substituted C1-C6 hydroxyalkyl or optionally substituted C2-C6 hydroxyalkenyl, b) R1 and R3, for each occurrence, are each independently selected from: -H, methyl, ethyl, propyl, isopropyl, butyl, isobutyl or tertbutyl, and R2 is absent or -H or c) R1 and R3 for each occurrence, are each independently selected from: -H, -D, -CH3, -CD3, -CH2CH3, -CD2CH3, where D denotes deuterium (2H), and R2 is absent, d) R1 and R3, for each occurrence, are each independently selected from: -H, C1-C6 alkyl, C2-C6 alkenyl, C1-C5 alkyl, C2-C5 alkenyl, C1-C4 alkyl, C2-C4 alkenyl, Ce alkyl, C5 alkyl, C4 alkyl, C3 alkyl, C2 alkyl, Ci alkyl, Ce alkenyl, or C5 alkenyl, or C4 alkenyl, or C3 alkenyl, or C2 alkenyl. In some further embodiments, e) R1 and R3, for each occurrence, are each independently selected from: -H or C1-C3 alkyl, or f) RT and R3 are identical and R2 is absent.
24. The lipid of claim 1, wherein:
R4 for each occurrence, is independently selected from
Figure imgf000152_0002
25. The lipid of claim 1, wherein: a) R6 for each occurrence, is independently selected from C3-C24 branched alkylene or C3- C24 branched alkenylene; b) R6 for each occurrence, is independently selected from: Ci -C3 alkylene, C1-C9 alkylene, C2-C9 alkenylene, C1-C7 alkylene, C2-C7 alkenylene, Ci -C5 alkylene, C2-C5 alkenylene, C2- C8 alkylene, C2-C8 alkenylene, C3-C7 alkylene, C3-C7 alkenylene, C5-C7 alkylene, C5-C7 alkenylene, C12 alkylene, C11 alkylene, C10 alkylene, C9 alkylene, C8 alkylene, C7 alkylene, C6 alkylene, C5 alkylene, C4 alkylene, C3 alkylene, C2 alkylene, Ci alkylene, C12 alkenylene, C11 alkenylene, Cwalkenylene, C9 alkenylene, C8 alkenylene, C7 alkenylene, Ce alkenylene, C5 alkenylene, C4 alkenylene, C3 alkenylene, C2 alkenylene; c) R8 and R9, are each independently C6-C12 alkyl or C6-C12 alkenyl; d) R8 is Ce-Cio alkyl or Ce-Cio alkenyl; e) R8 and R9, together, have a total of 12-20 carbon atoms; f) R8, R9, R12, and R13, each independently have 1, 2 or 3, C=C double bonds where at least one of the C=C double bonds is of Z configuration or where each C=C double bond is in a Z configuration; g) R14 is C1-C3 alkyl; h) R8 and R9, for each occurrence, are each independently selected from: -Cio-Cis alkenyl comprising 1, 2, 3 or 4 cis double bonds, -Cio-Cis alkenyl ester comprising 1, 2, 3 or 4 cis double bonds, optionally substituted -C6-C18 alkyl, -Ce-7i8 alkyl ester, -C6-C18 alkyl ether, -C6-C18 alkyl carbonate, -C6-C18 alkenyl, -C6-C18 alkenyl ester, -C6-C18 alkenyl ether, or -C6-C18 alkenyl carbonate; i) R8 and R9, are each independently -Cio-Cis alkenyl comprising 2 cis (Z configuration) double bonds. The lipid of claim 1, R8 and R9 for each occurrence, are each independently selected from: optionally substituted C6-C18 alkyl, Ce-7i8 alkylester, Ce- Ci8 alkyl ether, C6-C18 alkyl carbonate C6-C18 alkenyl, C6-C18 alkenylester, C6-C18 alkenylether, or C6-C18 alkenylcarbonate; j) R8 and R9 for each occurrence, are each independently selected from: C6-C14 alkyl, C6- C14 alkenyl, C8-C12 alkyl, C8-Ci2 alkenyl, Ci6 alkyl, C15 alkyl, C14 alkyl, C13 alkyl, C12 alkyl, C11 alkyl, C10 alkyl, C9 alkyl, C8 alkyl, C7 alkyl, Ci6 alkenyl, C15 alkenyl, C14 alkenyl, C13 alkenyl, C12 alkenyl, C11 alkenyl, C10 alkenyl, C9, alkenyl, C8 alkenyl, or C7 alkenyl; k) R8 and R9 have an equal number of carbon atoms; l) R8 and R9, combined, have more than 15 total carbon atoms; m) R8 and R9 differ in total carbon atoms from each other; n) R8 and R9 differ by one or two total carbon atoms from each other; o) R8 and R9 differ by one total carbon atoms; p) R8 is C7 alkyl and R9 is C8 alkyl, R8 is C8 alkyl and R9 is C7 alkyl, R8 is C8 alkyl and R9 is C9 alkyl, R8 is C9 alkyl and R9 is C8 alkyl, R8 is C9 alkyl and R9 is C10 alkyl ,R8 is C10 alkyl and R9 is C9 alkyl, R8 is C10 alkyl and R9 is C11 alkyl, R8 is C11 alkyl and R9 is C10 alkyl, R8 is C11 alkyl and R9 is C12 alkyl, R8 is C12 alkyl and R9 is C11 alkyl, R8 is C7 alkyl and R9 is C9 alkyl, R8 is C9 alkyl and R9 is C7 alkyl, R8 is C8 alkyl and R9 is C10 alkyl, R8 is C10 alkyl and R9 is C8 alkyl, R8 is C9 alkyl and R9 is C11 alkyl, R8 is C11 alkyl and R9 is C9 alkyl, R8 is C10 alkyl and R9 is C12 alkyl, R8 is C12 alkyl and R9 is C10 alkyl, R8 is C11 alkyl and R9 is C8 alkyl, or R8 is C8 alkyl and R9 is C11 alkyl; q) R10 and R11 for each occurrence, are each independently selected from: C1-C16 unbranched alkyl or C2-C16 unbranched alkenyl; r) R16 is C1-C3 alkylene; s) R29, R30 and R31 are each -CH2-; t) R32, R33 and R34 are each -O-; u) R35 is -OH; v) R29, R30 and R31 are each -CH2-, and R32, R33 and R34 are each -O-; w) R29, R30 and R31 are each -CH2-, R32 and R33 are each -O- and R35 is -OH; x) R42 is -[CH2]nC(=O)O[CH2]r; wherein r is an integer selected from 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16 or 17; y) R42 for each occurrence, is independently selected from: -R46-C(=O)O-R47-, -R46- OC(=O)-R47-, -R46-OC(=O)O-R47-,or -R46-O-R47-, wherein; R46 for each occurrence, is independently selected from C1-C12 alkylene, C1-C12 alkylene ester, C1-C12 alkylene ether, C1-C12 alkylene carbonate; and R47 for each occurrence, is independently selected from -C1-C12 alkyl, -C1-C12 alkyl ester, -C1-C12 alkyl ether, or -C1-C12 alkyl carbonate.
26. The lipid of claim 1, wherein: R55 for each occurrence, is independently selected from:
Figure imgf000154_0001
Figure imgf000155_0001
R5 for each occurrence, is independently selected from: -H, optionally substituted C1-C16 alkyl, optionally substituted C1-C16 alkyl ester, optionally substituted C2-C16 alkenyl, optionally substituted C2-C16 alkenyl ester,
Figure imgf000155_0002
; wherein, R10 and R11 for each occurrence, are each independently selected from: optionally substituted C1-C16 alkyl or optionally substituted C2-C16 alkenyl. T1. The lipid of claim 26, wherein: R5 for each occurrence, is independently selected from: C1-C14 unbranched alkyl, C2-C14 unbranched alkenyl, C1-C16 unbranched alkyl, C1-C16 unbranched alkyl ester, unbranched C2-Ci6 alkenyl, C2-Ci6 unbranched alkenyl ester, C3-C16 branched alkyl, C3-C16 branched alkyl ester, C3-C16 branched alkenyl, C3-C16 branched alkenyl ester or
Figure imgf000156_0001
, wherein: R10 and R11 for each occurrence, are each independently selected from: C1-C12 unbranched alkyl, C2-C12, unbranched alkenyl, C2-Ci2 unbranched alkyl, C2- Ci2 unbranched alkenyl, C5-C7, unbranched alkyl, or C5-C7 unbranched alkenyl, Ci6 unbranched alkyl, C15 unbranched alkyl, C14 unbranched alkyl, C13 unbranched alkyl, Ci2 unbranched alkyl, C11 unbranched alkyl, C10 unbranched alkyl, C9 unbranched alkyl, C8 unbranched alkyl, C7 unbranched alkyl, Ce unbranched alkyl, C5 unbranched alkyl, C4 unbranched alkyl, C3 unbranched alkyl, C2 unbranched alkyl, methyl, Ci6 unbranched alkenyl, C15 unbranched alkenyl, C14 unbranched alkenyl, C13 unbranched alkenyl, C12 unbranched alkenyl, C11 unbranched alkenyl, C10 unbranched alkenyl, C9 unbranched alkenyl, C8 unbranched alkenyl, C7 unbranched alkenyl, C6 unbranched alkenyl, C5 unbranched alkenyl, C4 unbranched alkenyl, C3 unbranched alkenyl, C2 alkenyl, C2-Ci0 unbranched alkyl or C2-C10 unbranched alkenyl.
28. The lipid of claim 1, wherein: a) R55 for each occurrence, is independently selected from: -OC(=O)C(R42)[CH2]nC(=O)O- R41-CH(C4-CI2 alkyl)2, -R41-N(R42)([CH2]nC(=O)O-R41-CH(C4-Ci2 alkyl)2, - OC(=O)C(R43)[CH2]nC(=O)O-R41-CH(C4-Ci2 alkyl)2, -R41-N(R43)([CH2]nC(=O)O-R41-CH(C4- Ci2 alkyl)2, -R41-C(R42)[CH2]nC(=O)O-R41-CH(C4-Ci2 alkyl)2, -R41-CH(R43)(R44), or -R41- N(R43)(R44); b) A2 for each occurrence, is independently selected from; -OC(=O)-, -OC(=O)R16C(=O)O- or -C(=O)O-; c) each R55 has 12-50, 18-30, 20-30, or 12-24 total carbon atoms; and d) each R55 comprises 0-6, 0-3, 1-3, 1, 2, 3, 4, 5 or 6 esters; 0-6, 0-3, 1-3, 1, 2, 3, 4, 5 or 6 ethers; and/or 0-6, 0-3, 1-3, 1, 2, 3, 4, 5 or 6 carbonates.
29. The lipid of claim 1, R55 for each occurrence, is independently selected from: -O- C(=O)C(R42)[CH2]nC(=O)O-R41-CH(C4-Ci2 alkyl)2, -R41-N(-R42)(-[CH2]nC(=O)O-R41-CH(C4- Ci2 alkyl)2, -O-C(=O)C(R43)-[CH2]nC(=O)O-R41-CH(C4-Ci2 alkyl)2, -R41-N(-R43)(- [CH2]nC(=O)O-R41— CH(C4-CI2 alkyl)2, -R41-C(R42)[CH2]nC(=O)O-R41-CH(C4-Ci2 alkyl)2, -R41- CH(R43)(R44), or -R41-N(R43)(R44).
30. The lipid of claim 1, wherein: R55, for each occurrence, is independently selected from: -R16-N(-R48)-R49, -R16-CH(-R48)-R49, -R16-OC(=O)-R16-N(-R48)-R49, -R16-OC(=O)CH(-R48)-R49, or -
R16-C(=O)O-R16-CH(-R48)-R49; wherein:
R16 for each occurrence, is independently selected from: a bond or -[CH2]k-; and
R48 and R49 are selected from : i) R48 is -R16-O-CH(-[CH2]5-9 CH3)2 and R49 is -R16-C(=O)O-[CH2]6-I2CH3, j) R48 is -CH2O-C(=O)O-CH(-[CH2]7 CH3)2 and R49 is -R16-O-C(=O)O-[CH2]7-I3CH3, k) R48 is -R16C(=O)O-CH([CH2]7 CH3)2 and R49 is -R16C(=O)O-[CH2]7CH3, l) R48 is -[CH2]3C(=O)O-CH(-[CH2]7 CH3)2 and R49 is -[CH2]3C(=O)O-[CH2]7CH3, m) R48 is -CH2C(=O)O-CH(-[CH2]9 CH3)2 and R49 is -C(=O)O-[CH2]I0CH3, n) R48 is -[CH2]3C(=O)O-CH(-[CH2]7CH3)2 and R49 is -[CH2]3C(=O)O-[CH2]9CH3 o) R48 is -[CH2]2C(=O)O-CH(-[CH2]7 CH3)2 and R49 is -[CH2]2C(=O)O-[CH2]9CH3, or p) R48 is -R16-C(=O)O-R16-CH(-R57-CH3)2 and R49 is -[CH2]8-I6CH3.
31. The lipid of claim 1, wherein: R55, for each occurrence, is independently selected from:
Figure imgf000157_0001
Figure imgf000158_0001
Figure imgf000159_0001
Figure imgf000160_0001
Figure imgf000161_0001
each cyclopropyl (cPr) and each -C=C- has a cis (or E) configuration;
R57 for each occurrence, is independently selected from: a bond or -[CFhJm-; wherein m is an integer selected from: 1, 2, 3, 4, 5, 6, 7 or 8;
R16 for each occurrence, is independently selected from: a bond or -[CFhJk-;
R61 for each occurrence, is independently selected from:
Figure imgf000161_0002
R116 for each occurrence, is independently selected from: -H, -F, -CF3, -Cl, -Br, -I, -CH3, - CH2R14, -CHR14 2, -CR14 3, -OH, -OR14, -NH2, -NHR14, -N(R14)2, -SH, -SR14, -S(=O)H, - S(=O)R14, -S(=O)2H, -S(=O)2R14, -PH2, -PHR14, -PR142, -P(OH)2, -POHR14, -PR142, -P(OR14)2, -P(O)H2, -P(O)HR14, -P(O)R142, -P(O)(OH)2, -P(O)(OR14)OH, -P(O)(OR14)2, -SiH3, -SiH2R14, - SiHR14 2 or-SiR14 3;
R81, R82, R83 and R84 for each occurrence, are each independently selected from: a bond or -
[CH2]I-IO-; R85 and R90 for each occurrence, are each independently selected from: a bond or -[CH2]I-S-
R86 and R91 for each occurrence, are each independently selected from: a bond -O-, -O- C(O=)-, -O-C(=O)-O- or -C(=O)-O-;
R87 for each occurrence, is independently selected from: a bond or -[CH2]I-6-;
R88 and R89 for each occurrence, are each independently -[CI-hJi-ioCHs;
R92 is -[CH2]2-I6CH3;
R97 for each occurrence, is independently selected from: -[CH2]I-6-, -[CH2]I-6-C(=O)-O-, - [CH2] -[CH2]I-6-C(=O)-, wherein: when R61
Figure imgf000162_0001
then R97 for each occurrence, is independently selected from: -[CH2]I-6- or -[CH2]I-6-C(=O)-O- and when R61 is ~L , then R97 for each occurrence, is independently selected from: - [CH2]I-6-, -[CH2]I-6-C(=O)-O-, -[CH2]I-6-O- or -[CH2]I-6-C(=O)-.
32. A lipid particle comprising the lipid if claim 1.
33. The lipid particle of claim 32, wherein said particle is a lipid nanoparticle or liposome.
34. The lipid nanoparticle of claim 33, wherein the lipid component further comprises one or more components selected from the group consisting of: a neutral lipid, a structural lipid and a polymer conjugated lipid.
35. The lipid nanoparticle of claim 34, wherein the neutral lipid is selected from one or more of the group consisting of: l,2-distearoyl-sn-glycero-3-phosphocholine (DSPC), 1,2- dioleyl- sn-glycero-3-phosphoethanolamine (DOPE), l,2-dilinoleyl-sn-glycero-3- phosphocholine (DLPC), 1,2-dimyristoyl-sn-glycero-phosphocholine (DMPC), 1,2- dioleyl- sn-glycero-3-phosphocholine (DOPC), l,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC), 1,2-diundecanoyl-sn-glycero-phosphocholine (DUPC), 1- palmitoyl-2-oleyl-sn- glycero-3-phosphocholine (POPC), 1,2-di-O-octadecenyl-sn- glycero-3-phosphocholine (18:0 Diether PC), l-oleyl-2-cholesterylhemisuccinoyl-sn- glycero-3-phosphocholine (OChemsPC), l-hexadecyl-sn-glycero-3-phosphocholine (C16 Lyso PC), 1,2-dilinolenoyl- sn-glycero-3-phosphocholine, 1,2-diarachidonoyl-sn- glycero-3-phosphocholine, 1,2- didocosahexaenoyl-sn-glycero-3-phosphocholine, l,2-diphytanoyl-sn-glycero-3- phosphoethanolamine (ME 16.0 PE), l,2-distearoyl-sn-glycero-3-phosphoethanolamine, l,2-dilinoleyl-sn-glycero-3-phosphoethanolamine, l,2-dilinolenoyl-sn-glycero-3- phosphoethanolamine, l,2-diarachidonoyl-sn-glycero-3-phosphoethanolamine, 1 ,2- didocosahexaenoyl-sn-glycero-3-phosphoethanolamine, 1 ,2- dioleyl-sn-glycero-3- phospho-rac-(l-glycerol) sodium salt (DOPG), and sphingomyelin.
36. The lipid nanoparticle of claim 34, wherein the structural lipid is selected from one or more of the group consisting of: cholesterol, fecosterol, sitosterol, campesterol, stigmasterol, brassicasterol, ergosterol, tomatidine, tomatine, ursolic acid and alphatocopherol, 5-a-cholestanol (5a-Cholestan-3|3-ol), 5-0-coprostanol, cholesteryl-(2' - hydroxy)-ethyl ether, cholesteryl-( 4' -hydroxy)-butyl ether, and 6-ketocholestanol, 5a- cholestane, cholestenone, 5-a-cholestanone, 5 p-cholestanone, allocholesterol, epiallocholesterol, cholesteryl decanoate, cholesteryl-(4'-hydroxy)-butyl ether, cholesteryl hemisuccinate, cholest-5-en-3|3-yl hydrogen sulfate and 24-methylene-cholesterol sulfate.
37. The lipid nanoparticle of claim 34, wherein the polymer conjugated lipid is selected from one or more of the group consisting of: PEGylated lipid selected from the group consisting of PEG-modified phosphatidylethanolamines, PEG-modified phosphatidic acids, PEG-modified ceramides, PEG-modified dialkylamines, PEG-modified diacylglycerols, and PEG-modified dialkylglycerols, optionally PEG-c-DOMG, PEG-DMG, PEG-DLPE, PEG-DMPE, PEG-DPPC, and PEG-DSPE.
38. The lipid nanoparticle of claim 34, comprising: about 25 mol % to about 60 mol % of a lipid selected from any one or more of Compound (1) - (54); about 2 mol % to about 25 mol % neutral lipid; about 18.5 mol % to about 60 mol % structural lipid; and about 0.2 mol % to about 10 mol % of PEGylated lipid.
39. The lipid nanoparticle of claim 34, wherein the lipid nanoparticle further comprises a diagnostic, prophylactic or therapeutic payload selected from a therapeutic nucleic acid, a protein a peptide or a small molecule.
40. The lipid nanoparticle of claim 39, wherein the therapeutic nucleic acid is an mRNA or a self-amplifying RNA.
41. The lipid nanoparticle of claim 34, wherein the lipid nanoparticle has a diameter of from about 30 nm to about 160 nm.
42. A pharmaceutical lipid nanoparticle composition comprising the lipid nanoparticle of claim 34 and a pharmaceutically acceptable carrier, excipient or diluent.
43. A method of delivering a lipid nanoparticle of claim 34 to a mammalian cell, comprising contacting the cell with the lipid nanoparticle.
44. The method of item 43, wherein the cell is a cell of a human subject.
45. A method of treating a disease, disorder or condition in a subject in need of such treatment, comprising administering a therapeutically effective amount of the pharmaceutical composition of claim 42 to the subject to thereby treat the disease, disorder or condition.
46. Use of a lipid or lipid nanoparticle of any one of claims 32-41 or the lipid nanoparticle composition of claim 11 in the manufacture of a medicament for the treatment of a disease, disorder or condition.
47. The method of claim 45, wherein the disease, disorder or condition is selected from the group consisting of a rare disease, an infectious disease, cancer, a proliferative disease, a genetic disease, an autoimmune disease, diabetes, a neurodegenerative disease, a cardiovascular disease, a reno-vascular disease and a metabolic disease.
PCT/IB2024/000526 2023-09-07 2024-09-09 Lipids and lipid nanoparticles WO2025052180A2 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US202363536997P 2023-09-07 2023-09-07
US63/536,997 2023-09-07
US202463664820P 2024-06-27 2024-06-27
US63/664,820 2024-06-27

Publications (2)

Publication Number Publication Date
WO2025052180A2 true WO2025052180A2 (en) 2025-03-13
WO2025052180A3 WO2025052180A3 (en) 2025-05-15

Family

ID=93563527

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2024/000526 WO2025052180A2 (en) 2023-09-07 2024-09-09 Lipids and lipid nanoparticles

Country Status (1)

Country Link
WO (1) WO2025052180A2 (en)

Citations (82)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1996010392A1 (en) 1994-09-30 1996-04-11 The University Of British Columbia Bilayer stabilizing components and their use in forming programmable fusogenic liposomes
WO1998051278A2 (en) 1997-05-14 1998-11-19 Inex Pharmaceuticals Corporation High efficiency encapsulation of charged therapeutic agents in lipid vesicles
US6320017B1 (en) 1997-12-23 2001-11-20 Inex Pharmaceuticals Corp. Polyamide oligomers
WO2002087541A1 (en) 2001-04-30 2002-11-07 Protiva Biotherapeutics Inc. Lipid-based formulations for gene transfer
US20030022649A1 (en) 2001-05-03 2003-01-30 Mitsubishi Denki Kabushiki Kaisha Signal reception method and device
US20040142025A1 (en) 2002-06-28 2004-07-22 Protiva Biotherapeutics Ltd. Liposomal apparatus and manufacturing methods
WO2005026372A1 (en) 2003-09-15 2005-03-24 Protiva Biotherapeutics, Inc. Polyethyleneglycol-modified lipid compounds and uses thereof
WO2005121348A1 (en) 2004-06-07 2005-12-22 Protiva Biotherapeutics, Inc. Lipid encapsulated interfering rna
WO2005120152A2 (en) 2004-06-07 2005-12-22 Protiva Biotherapeutics, Inc. Cationic lipids and methods of use
WO2006007712A1 (en) 2004-07-19 2006-01-26 Protiva Biotherapeutics, Inc. Methods comprising polyethylene glycol-lipid conjugates for delivery of therapeutic agents
WO2006069782A2 (en) 2004-12-27 2006-07-06 Silence Therapeutics Ag. Lipid complexes coated with peg and their use
US20070042031A1 (en) 2005-07-27 2007-02-22 Protiva Biotherapeutics, Inc. Systems and methods for manufacturing liposomes
US20080020058A1 (en) 2005-02-14 2008-01-24 Sirna Therapeutics, Inc. Lipid nanoparticle based compositions and methods for the delivery of biologically active molecules
WO2008042973A2 (en) 2006-10-03 2008-04-10 Alnylam Pharmaceuticals, Inc. Lipid containing formulations
WO2009086558A1 (en) 2008-01-02 2009-07-09 Tekmira Pharmaceuticals Corporation Improved compositions and methods for the delivery of nucleic acids
WO2009127060A1 (en) 2008-04-15 2009-10-22 Protiva Biotherapeutics, Inc. Novel lipid formulations for nucleic acid delivery
WO2009132131A1 (en) 2008-04-22 2009-10-29 Alnylam Pharmaceuticals, Inc. Amino lipid based improved lipid formulation
WO2010006282A2 (en) 2008-07-10 2010-01-14 Serina Therapeutics, Inc. Polyoxazolines with inert terminating groups, polyoxazolines prepared from protected initiating groups and related compounds
US20100015218A1 (en) 2007-02-16 2010-01-21 Vasant Jadhav Compositions and methods for potentiated activity of biologically active molecules
US20100076335A1 (en) 2001-09-25 2010-03-25 James Gharib System and Methods for Performing Surgical Procedures and Assessments
WO2010042877A1 (en) 2008-10-09 2010-04-15 Tekmira Pharmaceuticals Corporation Improved amino lipids and methods for the delivery of nucleic acids
WO2010048536A2 (en) 2008-10-23 2010-04-29 Alnylam Pharmaceuticals, Inc. Processes for preparing lipids
US20100117125A1 (en) 2006-11-06 2010-05-13 International Business Machines Corporation Semiconductor structures incorporating multiple crystallographic planes and methods for fabrication thereof
WO2010054405A1 (en) 2008-11-10 2010-05-14 Alnylam Pharmaceuticals, Inc. Novel lipids and compositions for the delivery of therapeutics
WO2010054384A1 (en) 2008-11-10 2010-05-14 Alnylam Pharmaceuticals, Inc. Lipids and compositions for the delivery of therapeutics
WO2010088537A2 (en) 2009-01-29 2010-08-05 Alnylam Pharmaceuticals, Inc. Improved lipid formulation
US20100256175A1 (en) 2009-04-03 2010-10-07 Bommannan D Bommi Treatment for drug addiction
WO2010129709A1 (en) 2009-05-05 2010-11-11 Alnylam Pharmaceuticals, Inc. Lipid compositions
WO2010144740A1 (en) 2009-06-10 2010-12-16 Alnylam Pharmaceuticals, Inc. Improved lipid formulation
WO2011000106A1 (en) 2009-07-01 2011-01-06 Protiva Biotherapeutics, Inc. Improved cationic lipids and methods for the delivery of therapeutic agents
WO2011000107A1 (en) 2009-07-01 2011-01-06 Protiva Biotherapeutics, Inc. Novel lipid formulations for delivery of therapeutic agents to solid tumors
WO2011022460A1 (en) 2009-08-20 2011-02-24 Merck Sharp & Dohme Corp. Novel cationic lipids with various head groups for oligonucleotide delivery
WO2011038160A2 (en) 2009-09-23 2011-03-31 Protiva Biotherapeutics, Inc. Compositions and methods for silencing genes expressed in cancer
WO2011066651A1 (en) 2009-12-01 2011-06-09 Protiva Biotherapeutics, Inc. Snalp formulations containing antioxidants
WO2011071860A2 (en) 2009-12-07 2011-06-16 Alnylam Pharmaceuticals, Inc. Compositions for nucleic acid delivery
WO2011090965A1 (en) 2010-01-22 2011-07-28 Merck Sharp & Dohme Corp. Novel cationic lipids for oligonucleotide delivery
WO2011141705A1 (en) 2010-05-12 2011-11-17 Protiva Biotherapeutics, Inc. Novel cationic lipids and methods of use thereof
WO2011141704A1 (en) 2010-05-12 2011-11-17 Protiva Biotherapeutics, Inc Novel cyclic cationic lipids and methods of use
WO2011153120A1 (en) 2010-06-04 2011-12-08 Merck Sharp & Dohme Corp. Novel low molecular weight cationic lipids for oligonucleotide delivery
WO2012000104A1 (en) 2010-06-30 2012-01-05 Protiva Biotherapeutics, Inc. Non-liposomal systems for nucleic acid delivery
WO2012016184A2 (en) 2010-07-30 2012-02-02 Alnylam Pharmaceuticals, Inc. Methods and compositions for delivery of active agents
WO2012031043A1 (en) 2010-08-31 2012-03-08 Novartis Ag Pegylated liposomes for delivery of immunogen-encoding rna
WO2012040184A2 (en) 2010-09-20 2012-03-29 Merck Sharp & Dohme Corp. Novel low molecular weight cationic lipids for oligonucleotide delivery
WO2012044638A1 (en) 2010-09-30 2012-04-05 Merck Sharp & Dohme Corp. Low molecular weight cationic lipids for oligonucleotide delivery
WO2012054365A2 (en) 2010-10-21 2012-04-26 Merck Sharp & Dohme Corp. Novel low molecular weight cationic lipids for oligonucleotide delivery
WO2012099755A1 (en) 2011-01-11 2012-07-26 Alnylam Pharmaceuticals, Inc. Pegylated lipids and their use for drug delivery
US20120225129A1 (en) 2010-08-20 2012-09-06 Cerulean Pharma Inc. Conjugates, particles, compositions, and related methods
WO2012162210A1 (en) 2011-05-26 2012-11-29 Merck Sharp & Dohme Corp. Ring constrained cationic lipids for oligonucleotide delivery
WO2013006825A1 (en) 2011-07-06 2013-01-10 Novartis Ag Liposomes having useful n:p ratio for delivery of rna molecules
WO2013016058A1 (en) 2011-07-22 2013-01-31 Merck Sharp & Dohme Corp. Novel bis-nitrogen containing cationic lipids for oligonucleotide delivery
US20130037977A1 (en) 2010-04-08 2013-02-14 Paul A. Burke Preparation of Lipid Nanoparticles
WO2013033563A1 (en) 2011-08-31 2013-03-07 Novartis Ag Pegylated liposomes for delivery of immunogen-encoding rna
WO2013049328A1 (en) 2011-09-27 2013-04-04 Alnylam Pharmaceuticals, Inc. Di-aliphatic substituted pegylated lipids
WO2013086354A1 (en) 2011-12-07 2013-06-13 Alnylam Pharmaceuticals, Inc. Biodegradable lipids for the delivery of active agents
WO2013086373A1 (en) 2011-12-07 2013-06-13 Alnylam Pharmaceuticals, Inc. Lipids for the delivery of active agents
WO2013086322A1 (en) 2011-12-07 2013-06-13 Alnylam Pharmaceuticals, Inc. Branched alkyl and cycloalkyl terminated biodegradable lipids for the delivery of active agents
US20130156845A1 (en) 2010-04-29 2013-06-20 Alnylam Pharmaceuticals, Inc. Lipid formulated single stranded rna
WO2013089151A1 (en) 2011-12-12 2013-06-20 協和発酵キリン株式会社 Lipid nanoparticles for drug delivery system containing cationic lipids
US20130164400A1 (en) 2011-11-04 2013-06-27 Nitto Denko Corporation Single use system for sterilely producing lipid-nucleic acid particles
WO2013116126A1 (en) 2012-02-01 2013-08-08 Merck Sharp & Dohme Corp. Novel low molecular weight, biodegradable cationic lipids for oligonucleotide delivery
WO2013126803A1 (en) 2012-02-24 2013-08-29 Protiva Biotherapeutics Inc. Trialkyl cationic lipids and methods of use thereof
WO2013148541A1 (en) 2012-03-27 2013-10-03 Merck Sharp & Dohme Corp. DIETHER BASED BIODEGRADABLE CATIONIC LIPIDS FOR siRNA DELIVERY
WO2015061467A1 (en) 2013-10-22 2015-04-30 Shire Human Genetic Therapies, Inc. Lipid formulations for delivery of messenger rna
WO2015074085A1 (en) 2013-11-18 2015-05-21 Arcturus Therapeutics, Inc. Ionizable cationic lipid for rna delivery
WO2015095340A1 (en) 2013-12-19 2015-06-25 Novartis Ag Lipids and lipid compositions for the delivery of active agents
WO2015095346A1 (en) 2013-12-19 2015-06-25 Novartis Ag Lipids and lipid compositions for the delivery of active agents
US20150239926A1 (en) 2013-11-18 2015-08-27 Arcturus Therapeutics, Inc. Asymmetric ionizable cationic lipid for rna delivery
WO2015199952A1 (en) 2014-06-25 2015-12-30 Acuitas Therapeutics Inc. Novel lipids and lipid nanoparticle formulations for delivery of nucleic acids
WO2016081029A1 (en) 2014-11-18 2016-05-26 Arcturus Therapeutics, Inc. Ionizable cationic lipid for rna delivery
US20160151284A1 (en) 2013-07-23 2016-06-02 Protiva Biotherapeutics, Inc. Compositions and methods for delivering messenger rna
US20160376224A1 (en) 2015-06-29 2016-12-29 Acuitas Therapeutics, Inc. Lipids and lipid nanoparticle formulations for delivery of nucleic acids
WO2017049245A2 (en) 2015-09-17 2017-03-23 Modernatx, Inc. Compounds and compositions for intracellular delivery of therapeutic agents
US20170119904A1 (en) 2015-10-28 2017-05-04 Acuitas Therapeutics, Inc. Novel lipids and lipid nanoparticle formulations for delivery of nucleic acids
WO2017099823A1 (en) 2015-12-10 2017-06-15 Modernatx, Inc. Compositions and methods for delivery of therapeutic agents
WO2017117528A1 (en) 2015-12-30 2017-07-06 Acuitas Therapeutics, Inc. Lipids and lipid nanoparticle formulations for delivery of nucleic acids
US9708628B2 (en) 2011-11-18 2017-07-18 Nof Corporation Cationic lipid having improved intracellular kinetics
US20180005363A1 (en) 2015-01-30 2018-01-04 Hitachi High-Technologies Corporation Pattern Matching Device and Computer Program for Pattern Matching
WO2018011633A1 (en) 2016-07-13 2018-01-18 Alcatel Lucent Underlying recessed component placement
WO2019051289A1 (en) 2017-09-08 2019-03-14 Generation Bio Co. Lipid nanoparticle formulations of non-viral, capsid-free dna vectors
US10385030B2 (en) 2015-01-30 2019-08-20 Nof Corporation Cationic lipid
WO2022246571A1 (en) 2021-05-28 2022-12-01 Nanovation Therapeutics Inc. Mc3-type lipids and use thereof in the preparation of lipid nanoparticles
WO2022261101A1 (en) 2021-06-07 2022-12-15 Generation Bio Co. Apoe and apob modified lipid nanoparticle compositions and uses thereof

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2448626A (en) * 1944-12-08 1948-09-07 Us Agriculture Alkylol amine salts of hydroxy fatty acids and process for their preparation
US2587546A (en) * 1948-10-30 1952-02-26 Standard Oil Dev Co Rust inhibiting composition
WO2016046673A1 (en) * 2014-09-28 2016-03-31 Mohan M Alapati Compositions and methods for the treatment of neurological diseases
CA3088546A1 (en) * 2018-01-29 2019-08-01 Merck Sharp & Dohme Corp. Stabilized rsv f proteins and uses thereof

Patent Citations (135)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1996010392A1 (en) 1994-09-30 1996-04-11 The University Of British Columbia Bilayer stabilizing components and their use in forming programmable fusogenic liposomes
US5885613A (en) 1994-09-30 1999-03-23 The University Of British Columbia Bilayer stabilizing components and their use in forming programmable fusogenic liposomes
WO1998051278A2 (en) 1997-05-14 1998-11-19 Inex Pharmaceuticals Corporation High efficiency encapsulation of charged therapeutic agents in lipid vesicles
US6287591B1 (en) 1997-05-14 2001-09-11 Inex Pharmaceuticals Corp. Charged therapeutic agents encapsulated in lipid particles containing four lipid components
US6320017B1 (en) 1997-12-23 2001-11-20 Inex Pharmaceuticals Corp. Polyamide oligomers
US6586559B2 (en) 1997-12-23 2003-07-01 Inex Pharmaceuticals Corporation Polyamide oligomers
WO2002087541A1 (en) 2001-04-30 2002-11-07 Protiva Biotherapeutics Inc. Lipid-based formulations for gene transfer
US20030077829A1 (en) 2001-04-30 2003-04-24 Protiva Biotherapeutics Inc.. Lipid-based formulations
US20030022649A1 (en) 2001-05-03 2003-01-30 Mitsubishi Denki Kabushiki Kaisha Signal reception method and device
US20100076335A1 (en) 2001-09-25 2010-03-25 James Gharib System and Methods for Performing Surgical Procedures and Assessments
US20040142025A1 (en) 2002-06-28 2004-07-22 Protiva Biotherapeutics Ltd. Liposomal apparatus and manufacturing methods
US8936942B2 (en) 2003-09-15 2015-01-20 Protiva Biotherapeutics, Inc. Polyethyleneglycol-modified lipid compounds and uses thereof
US20050175682A1 (en) 2003-09-15 2005-08-11 Protiva Biotherapeutics, Inc. Polyethyleneglycol-modified lipid compounds and uses thereof
WO2005026372A1 (en) 2003-09-15 2005-03-24 Protiva Biotherapeutics, Inc. Polyethyleneglycol-modified lipid compounds and uses thereof
WO2005121348A1 (en) 2004-06-07 2005-12-22 Protiva Biotherapeutics, Inc. Lipid encapsulated interfering rna
WO2005120152A2 (en) 2004-06-07 2005-12-22 Protiva Biotherapeutics, Inc. Cationic lipids and methods of use
US20060008910A1 (en) 2004-06-07 2006-01-12 Protiva Biotherapeuties, Inc. Lipid encapsulated interfering RNA
US20060083780A1 (en) 2004-06-07 2006-04-20 Protiva Biotherapeutics, Inc. Cationic lipids and methods of use
US20060051405A1 (en) 2004-07-19 2006-03-09 Protiva Biotherapeutics, Inc. Compositions for the delivery of therapeutic agents and uses thereof
WO2006007712A1 (en) 2004-07-19 2006-01-26 Protiva Biotherapeutics, Inc. Methods comprising polyethylene glycol-lipid conjugates for delivery of therapeutic agents
WO2006069782A2 (en) 2004-12-27 2006-07-06 Silence Therapeutics Ag. Lipid complexes coated with peg and their use
US20100062967A1 (en) 2004-12-27 2010-03-11 Silence Therapeutics Ag Coated lipid complexes and their use
US20080020058A1 (en) 2005-02-14 2008-01-24 Sirna Therapeutics, Inc. Lipid nanoparticle based compositions and methods for the delivery of biologically active molecules
US20070042031A1 (en) 2005-07-27 2007-02-22 Protiva Biotherapeutics, Inc. Systems and methods for manufacturing liposomes
US20090023673A1 (en) 2006-10-03 2009-01-22 Muthiah Manoharan Lipid containing formulations
WO2008042973A2 (en) 2006-10-03 2008-04-10 Alnylam Pharmaceuticals, Inc. Lipid containing formulations
WO2008147438A2 (en) 2006-10-24 2008-12-04 Sirna Therapeutics, Inc. Lipid nanoparticle based compositions and methods for the delivery of biologically active molecules
US20100117125A1 (en) 2006-11-06 2010-05-13 International Business Machines Corporation Semiconductor structures incorporating multiple crystallographic planes and methods for fabrication thereof
US20100015218A1 (en) 2007-02-16 2010-01-21 Vasant Jadhav Compositions and methods for potentiated activity of biologically active molecules
WO2009086558A1 (en) 2008-01-02 2009-07-09 Tekmira Pharmaceuticals Corporation Improved compositions and methods for the delivery of nucleic acids
US20110117125A1 (en) 2008-01-02 2011-05-19 Tekmira Pharmaceuticals Corporation Compositions and methods for the delivery of nucleic acids
WO2009127060A1 (en) 2008-04-15 2009-10-22 Protiva Biotherapeutics, Inc. Novel lipid formulations for nucleic acid delivery
US20100130588A1 (en) 2008-04-15 2010-05-27 Protiva Biotherapeutics, Inc. Novel lipid formulations for nucleic acid delivery
WO2009132131A1 (en) 2008-04-22 2009-10-29 Alnylam Pharmaceuticals, Inc. Amino lipid based improved lipid formulation
WO2010006282A2 (en) 2008-07-10 2010-01-14 Serina Therapeutics, Inc. Polyoxazolines with inert terminating groups, polyoxazolines prepared from protected initiating groups and related compounds
US20110123453A1 (en) 2008-07-10 2011-05-26 Serina Therapeutics, Inc. Polyoxazolines with Inert Terminating Groups, Polyoxazolines Prepared from Protected Initiating Groups and Related Compounds
WO2010042877A1 (en) 2008-10-09 2010-04-15 Tekmira Pharmaceuticals Corporation Improved amino lipids and methods for the delivery of nucleic acids
WO2010048536A2 (en) 2008-10-23 2010-04-29 Alnylam Pharmaceuticals, Inc. Processes for preparing lipids
WO2010054384A1 (en) 2008-11-10 2010-05-14 Alnylam Pharmaceuticals, Inc. Lipids and compositions for the delivery of therapeutics
WO2010054401A1 (en) 2008-11-10 2010-05-14 Alnylam Pharmaceuticals, Inc. Novel lipids and compositions for the delivery of therapeutics
WO2010054405A1 (en) 2008-11-10 2010-05-14 Alnylam Pharmaceuticals, Inc. Novel lipids and compositions for the delivery of therapeutics
US20120027796A1 (en) 2008-11-10 2012-02-02 Alnylam Pharmaceuticals, Inc. Novel lipids and compositions for the delivery of therapeutics
US20120058144A1 (en) 2008-11-10 2012-03-08 Alnylam Pharmaceuticals, Inc. Lipids and compositions for the delivery of therapeutics
WO2010054406A1 (en) 2008-11-10 2010-05-14 Alnylam Pharmaceuticals, Inc. Novel lipids and compositions for the delivery of therapeutics
US20120101148A1 (en) 2009-01-29 2012-04-26 Alnylam Pharmaceuticals, Inc. lipid formulation
WO2010088537A2 (en) 2009-01-29 2010-08-05 Alnylam Pharmaceuticals, Inc. Improved lipid formulation
US20100256175A1 (en) 2009-04-03 2010-10-07 Bommannan D Bommi Treatment for drug addiction
US20120128760A1 (en) 2009-05-05 2012-05-24 Alnylam Pharmaceuticals, Inc. Lipid compositions
WO2010129709A1 (en) 2009-05-05 2010-11-11 Alnylam Pharmaceuticals, Inc. Lipid compositions
US20100324120A1 (en) 2009-06-10 2010-12-23 Jianxin Chen Lipid formulation
WO2010144740A1 (en) 2009-06-10 2010-12-16 Alnylam Pharmaceuticals, Inc. Improved lipid formulation
US20120202871A1 (en) 2009-07-01 2012-08-09 Protiva Biotherapeutics, Inc. Cationic lipids and methods for the delivery of therapeutic agents
WO2011000107A1 (en) 2009-07-01 2011-01-06 Protiva Biotherapeutics, Inc. Novel lipid formulations for delivery of therapeutic agents to solid tumors
WO2011000106A1 (en) 2009-07-01 2011-01-06 Protiva Biotherapeutics, Inc. Improved cationic lipids and methods for the delivery of therapeutic agents
US20120149894A1 (en) 2009-08-20 2012-06-14 Mark Cameron Novel cationic lipids with various head groups for oligonucleotide delivery
WO2011022460A1 (en) 2009-08-20 2011-02-24 Merck Sharp & Dohme Corp. Novel cationic lipids with various head groups for oligonucleotide delivery
US20130065939A1 (en) 2009-09-23 2013-03-14 Protiva Biotherapeutics, Inc. Compositions and methods for silencing genes expressed in cancer
WO2011038160A2 (en) 2009-09-23 2011-03-31 Protiva Biotherapeutics, Inc. Compositions and methods for silencing genes expressed in cancer
WO2011066651A1 (en) 2009-12-01 2011-06-09 Protiva Biotherapeutics, Inc. Snalp formulations containing antioxidants
US20130338210A1 (en) 2009-12-07 2013-12-19 Alnylam Pharmaceuticals, Inc. Compositions for nucleic acid delivery
WO2011071860A2 (en) 2009-12-07 2011-06-16 Alnylam Pharmaceuticals, Inc. Compositions for nucleic acid delivery
WO2011090965A1 (en) 2010-01-22 2011-07-28 Merck Sharp & Dohme Corp. Novel cationic lipids for oligonucleotide delivery
US20130037977A1 (en) 2010-04-08 2013-02-14 Paul A. Burke Preparation of Lipid Nanoparticles
US20130156845A1 (en) 2010-04-29 2013-06-20 Alnylam Pharmaceuticals, Inc. Lipid formulated single stranded rna
WO2011141705A1 (en) 2010-05-12 2011-11-17 Protiva Biotherapeutics, Inc. Novel cationic lipids and methods of use thereof
US20130123338A1 (en) 2010-05-12 2013-05-16 Protiva Biotherapeutics, Inc. Novel cationic lipids and methods of use thereof
US20130116307A1 (en) 2010-05-12 2013-05-09 Protiva Biotherapeutics Inc. Novel cyclic cationic lipids and methods of use
WO2011141704A1 (en) 2010-05-12 2011-11-17 Protiva Biotherapeutics, Inc Novel cyclic cationic lipids and methods of use
US20130090372A1 (en) 2010-06-04 2013-04-11 Brian W. Budzik Novel Low Molecular Weight Cationic Lipids for Oligonucleotide Delivery
WO2011153120A1 (en) 2010-06-04 2011-12-08 Merck Sharp & Dohme Corp. Novel low molecular weight cationic lipids for oligonucleotide delivery
US20130303587A1 (en) 2010-06-30 2013-11-14 Protiva Biotherapeutics, Inc. Non-liposomal systems for nucleic acid delivery
WO2012000104A1 (en) 2010-06-30 2012-01-05 Protiva Biotherapeutics, Inc. Non-liposomal systems for nucleic acid delivery
US20130323269A1 (en) 2010-07-30 2013-12-05 Muthiah Manoharan Methods and compositions for delivery of active agents
WO2012016184A2 (en) 2010-07-30 2012-02-02 Alnylam Pharmaceuticals, Inc. Methods and compositions for delivery of active agents
US20120225129A1 (en) 2010-08-20 2012-09-06 Cerulean Pharma Inc. Conjugates, particles, compositions, and related methods
US20130202684A1 (en) 2010-08-31 2013-08-08 Lichtstrasse Pegylated liposomes for delivery of immunogen encoding rna
WO2012031043A1 (en) 2010-08-31 2012-03-08 Novartis Ag Pegylated liposomes for delivery of immunogen-encoding rna
WO2012040184A2 (en) 2010-09-20 2012-03-29 Merck Sharp & Dohme Corp. Novel low molecular weight cationic lipids for oligonucleotide delivery
US20130178541A1 (en) 2010-09-20 2013-07-11 Matthew G. Stanton Novel low molecular weight cationic lipids for oligonucleotide delivery
US20130274523A1 (en) 2010-09-30 2013-10-17 John A. Bawiec, III Low molecular weight cationic lipids for oligonucleotide delivery
WO2012044638A1 (en) 2010-09-30 2012-04-05 Merck Sharp & Dohme Corp. Low molecular weight cationic lipids for oligonucleotide delivery
WO2012054365A2 (en) 2010-10-21 2012-04-26 Merck Sharp & Dohme Corp. Novel low molecular weight cationic lipids for oligonucleotide delivery
US20130274504A1 (en) 2010-10-21 2013-10-17 Steven L. Colletti Novel Low Molecular Weight Cationic Lipids For Oligonucleotide Delivery
US20140200257A1 (en) 2011-01-11 2014-07-17 Alnylam Pharmaceuticals, Inc. Pegylated lipids and their use for drug delivery
WO2012099755A1 (en) 2011-01-11 2012-07-26 Alnylam Pharmaceuticals, Inc. Pegylated lipids and their use for drug delivery
WO2012162210A1 (en) 2011-05-26 2012-11-29 Merck Sharp & Dohme Corp. Ring constrained cationic lipids for oligonucleotide delivery
US20140141070A1 (en) 2011-07-06 2014-05-22 Andrew Geall Liposomes having useful n:p ratio for delivery of rna molecules
WO2013006825A1 (en) 2011-07-06 2013-01-10 Novartis Ag Liposomes having useful n:p ratio for delivery of rna molecules
WO2013016058A1 (en) 2011-07-22 2013-01-31 Merck Sharp & Dohme Corp. Novel bis-nitrogen containing cationic lipids for oligonucleotide delivery
WO2013033563A1 (en) 2011-08-31 2013-03-07 Novartis Ag Pegylated liposomes for delivery of immunogen-encoding rna
US20140255472A1 (en) 2011-08-31 2014-09-11 Andrew Geall Pegylated liposomes for delivery of immunogen-encoding rna
WO2013049328A1 (en) 2011-09-27 2013-04-04 Alnylam Pharmaceuticals, Inc. Di-aliphatic substituted pegylated lipids
US20150203446A1 (en) 2011-09-27 2015-07-23 Takeda Pharmaceutical Company Limited Di-aliphatic substituted pegylated lipids
US20130164400A1 (en) 2011-11-04 2013-06-27 Nitto Denko Corporation Single use system for sterilely producing lipid-nucleic acid particles
US9708628B2 (en) 2011-11-18 2017-07-18 Nof Corporation Cationic lipid having improved intracellular kinetics
US20130195920A1 (en) 2011-12-07 2013-08-01 Alnylam Pharmaceuticals, Inc. Biodegradable lipids for the delivery of active agents
WO2013086322A1 (en) 2011-12-07 2013-06-13 Alnylam Pharmaceuticals, Inc. Branched alkyl and cycloalkyl terminated biodegradable lipids for the delivery of active agents
US20140308304A1 (en) 2011-12-07 2014-10-16 Alnylam Pharmaceuticals, Inc. Lipids for the delivery of active agents
WO2013086373A1 (en) 2011-12-07 2013-06-13 Alnylam Pharmaceuticals, Inc. Lipids for the delivery of active agents
WO2013086354A1 (en) 2011-12-07 2013-06-13 Alnylam Pharmaceuticals, Inc. Biodegradable lipids for the delivery of active agents
WO2013089151A1 (en) 2011-12-12 2013-06-20 協和発酵キリン株式会社 Lipid nanoparticles for drug delivery system containing cationic lipids
US20140039032A1 (en) 2011-12-12 2014-02-06 Kyowa Hakko Kirin Co., Ltd. Lipid nano particles comprising cationic lipid for drug delivery system
WO2013116126A1 (en) 2012-02-01 2013-08-08 Merck Sharp & Dohme Corp. Novel low molecular weight, biodegradable cationic lipids for oligonucleotide delivery
US20150064242A1 (en) 2012-02-24 2015-03-05 Protiva Biotherapeutics, Inc. Trialkyl cationic lipids and methods of use thereof
WO2013126803A1 (en) 2012-02-24 2013-08-29 Protiva Biotherapeutics Inc. Trialkyl cationic lipids and methods of use thereof
US20150057373A1 (en) 2012-03-27 2015-02-26 Sirna Therapeutics, Inc DIETHER BASED BIODEGRADABLE CATIONIC LIPIDS FOR siRNA DELIVERY
WO2013148541A1 (en) 2012-03-27 2013-10-03 Merck Sharp & Dohme Corp. DIETHER BASED BIODEGRADABLE CATIONIC LIPIDS FOR siRNA DELIVERY
US20160151284A1 (en) 2013-07-23 2016-06-02 Protiva Biotherapeutics, Inc. Compositions and methods for delivering messenger rna
WO2015061467A1 (en) 2013-10-22 2015-04-30 Shire Human Genetic Therapies, Inc. Lipid formulations for delivery of messenger rna
US20150140070A1 (en) 2013-10-22 2015-05-21 Shire Human Genetic Therapies, Inc. Lipid formulations for delivery of messenger rna
US20150239926A1 (en) 2013-11-18 2015-08-27 Arcturus Therapeutics, Inc. Asymmetric ionizable cationic lipid for rna delivery
WO2015074085A1 (en) 2013-11-18 2015-05-21 Arcturus Therapeutics, Inc. Ionizable cationic lipid for rna delivery
US20150141678A1 (en) 2013-11-18 2015-05-21 Arcturus Therapeutics, Inc. Ionizable cationic lipid for rna delivery
US20160311759A1 (en) 2013-12-19 2016-10-27 Luis Brito Lipids and Lipid Compositions for the Delivery of Active Agents
WO2015095340A1 (en) 2013-12-19 2015-06-25 Novartis Ag Lipids and lipid compositions for the delivery of active agents
WO2015095346A1 (en) 2013-12-19 2015-06-25 Novartis Ag Lipids and lipid compositions for the delivery of active agents
US20160317458A1 (en) 2013-12-19 2016-11-03 Luis Brito Lipids and Lipid Compositions for the Delivery of Active Agents
US20150376115A1 (en) 2014-06-25 2015-12-31 Acuitas Therapeutics Inc. Novel lipids and lipid nanoparticle formulations for delivery of nucleic acids
WO2015199952A1 (en) 2014-06-25 2015-12-30 Acuitas Therapeutics Inc. Novel lipids and lipid nanoparticle formulations for delivery of nucleic acids
WO2016081029A1 (en) 2014-11-18 2016-05-26 Arcturus Therapeutics, Inc. Ionizable cationic lipid for rna delivery
US10385030B2 (en) 2015-01-30 2019-08-20 Nof Corporation Cationic lipid
US20180005363A1 (en) 2015-01-30 2018-01-04 Hitachi High-Technologies Corporation Pattern Matching Device and Computer Program for Pattern Matching
US20160376224A1 (en) 2015-06-29 2016-12-29 Acuitas Therapeutics, Inc. Lipids and lipid nanoparticle formulations for delivery of nucleic acids
WO2017004143A1 (en) 2015-06-29 2017-01-05 Acuitas Therapeutics Inc. Lipids and lipid nanoparticle formulations for delivery of nucleic acids
WO2017049245A2 (en) 2015-09-17 2017-03-23 Modernatx, Inc. Compounds and compositions for intracellular delivery of therapeutic agents
US20170210697A1 (en) 2015-09-17 2017-07-27 Modernatx, Inc. Compounds and compositions for intracellular delivery of therapeutic agents
WO2017075531A1 (en) 2015-10-28 2017-05-04 Acuitas Therapeutics, Inc. Novel lipids and lipid nanoparticle formulations for delivery of nucleic acids
US20170119904A1 (en) 2015-10-28 2017-05-04 Acuitas Therapeutics, Inc. Novel lipids and lipid nanoparticle formulations for delivery of nucleic acids
WO2017099823A1 (en) 2015-12-10 2017-06-15 Modernatx, Inc. Compositions and methods for delivery of therapeutic agents
US20180028664A1 (en) 2015-12-10 2018-02-01 Modernatx, Inc. Compositions and methods for delivery of agents
WO2017117528A1 (en) 2015-12-30 2017-07-06 Acuitas Therapeutics, Inc. Lipids and lipid nanoparticle formulations for delivery of nucleic acids
WO2018011633A1 (en) 2016-07-13 2018-01-18 Alcatel Lucent Underlying recessed component placement
WO2019051289A1 (en) 2017-09-08 2019-03-14 Generation Bio Co. Lipid nanoparticle formulations of non-viral, capsid-free dna vectors
WO2022246571A1 (en) 2021-05-28 2022-12-01 Nanovation Therapeutics Inc. Mc3-type lipids and use thereof in the preparation of lipid nanoparticles
WO2022261101A1 (en) 2021-06-07 2022-12-15 Generation Bio Co. Apoe and apob modified lipid nanoparticle compositions and uses thereof

Non-Patent Citations (10)

* Cited by examiner, † Cited by third party
Title
ARRUDA ET AL., BLOOD, vol. 105, 2005, pages 3458 - 3464
CARRASCO ET AL., COMM BIOL, vol. 4, 2021, pages 956
HEYES J ET AL., J CONTROL RELEASE, 2005, pages 276 - 287
J. OLEO SCI., vol. 71, no. 3, 2022, pages 401 - 410
JAYARAMAN ET AL., ANGEWANDTE CHEMIE, INTERNATIONAL EDITION, vol. 51, no. 34, 2012, pages 8529 - 8533
KULKARNI ET AL., ACS NANO, vol. 12, 2018, pages 4787
KULKARNI ET AL., NANOSCALE, vol. 36, 2017, pages 133347
SEMPLE ET AL., NATURE BIOTECHNOLOGY, vol. 28, 2010, pages 172 - 176
TOGASHI R ET AL., J CONTROL RELEASE, vol. 279, 10 June 2018 (2018-06-10), pages 262 - 270
VOUTILA ET AL., MOL THER., vol. 25, no. 12, 6 December 2017 (2017-12-06), pages 2705 - 2714

Also Published As

Publication number Publication date
WO2025052180A3 (en) 2025-05-15

Similar Documents

Publication Publication Date Title
EP4077272B1 (en) Lipid nanoparticle composition
JP7638973B2 (en) Carbonate-Containing Lipid Compounds and Compositions for Intracellular Delivery of Therapeutic Agents - Patent application
ES2992653T3 (en) Methods of preparing lipid nanoparticles
JP7463006B2 (en) Cationic lipids containing steroidal moieties
CA3113436A1 (en) Compounds and compositions for intracellular delivery of therapeutic agents
JP2025081495A (en) Branched tail lipid compounds and compositions for intracellular delivery of therapeutic agents - Patents.com
US20230053437A1 (en) Lipid compounds and lipid nanoparticle compositions
CA3056132A1 (en) Compounds and compositions for intracellular delivery of therapeutic agents
WO2022152109A2 (en) Lipid compounds and lipid nanoparticle compositions
TW202345863A (en) Mucosal administration methods and formulations
JP2020510072A (en) Lipid nanoparticle preparation
CN114401942A (en) Tris (hydroxymethyl) methylglycine and citrate esters
US11964052B2 (en) Lipid compounds and lipid nanoparticle compositions
US20240252650A1 (en) Lipid compounds and lipid nanoparticle compositions
TW202330457A (en) Lipid compounds and lipid nanoparticle compositions
EP4114470A1 (en) Compositions and methods for the treatment of ornithine transcarbamylase deficiency
AU2022207550C1 (en) Lipid compounds and lipid nanoparticle compositions
WO2024107906A2 (en) Ionizable lipids and lipid nanoparticle compositions for the delivery of nucleic acids
WO2025052180A2 (en) Lipids and lipid nanoparticles
TW201726614A (en) Compounds as cationic lipids
JP7709525B2 (en) Lipid Compounds and Lipid Nanoparticle Compositions
WO2024152512A1 (en) Lipid compound for delivery of therapeutic agent, preparation method therefor and use thereof
WO2024109798A1 (en) Lipid compound and lipid nanoparticle composition
RU2798342C2 (en) Compounds and compositions for intracellular delivery of therapeutic agents
WO2025102261A1 (en) Lipid compounds for delivering therapeutic agent, preparation method therefor and use thereof

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: 24808712

Country of ref document: EP

Kind code of ref document: A2