WO2018119163A1 - Ionizable cationic lipid for rna delivery - Google Patents

Ionizable cationic lipid for rna delivery Download PDF

Info

Publication number
WO2018119163A1
WO2018119163A1 PCT/US2017/067756 US2017067756W WO2018119163A1 WO 2018119163 A1 WO2018119163 A1 WO 2018119163A1 US 2017067756 W US2017067756 W US 2017067756W WO 2018119163 A1 WO2018119163 A1 WO 2018119163A1
Authority
WO
WIPO (PCT)
Prior art keywords
atx
etoac
solution
added
compound
Prior art date
Application number
PCT/US2017/067756
Other languages
French (fr)
Inventor
Joseph E. Payne
Padmanabh Chivukula
Steven P. Tanis
Priya Karmali
Original Assignee
Payne Joseph E
Padmanabh Chivukula
Tanis Steven P
Priya Karmali
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
Priority claimed from US15/387,067 external-priority patent/US10383952B2/en
Priority to IL290592A priority Critical patent/IL290592B2/en
Priority to CA3046885A priority patent/CA3046885C/en
Priority to RS20240048A priority patent/RS65078B1/en
Priority to DK17826404.0T priority patent/DK3558943T3/en
Priority to KR1020197021199A priority patent/KR102299053B1/en
Priority to AU2017379059A priority patent/AU2017379059B2/en
Priority to FIEP17826404.0T priority patent/FI3558943T3/en
Priority to CN201780086949.XA priority patent/CN110325511B/en
Priority to EP17826404.0A priority patent/EP3558943B1/en
Application filed by Payne Joseph E, Padmanabh Chivukula, Tanis Steven P, Priya Karmali filed Critical Payne Joseph E
Priority to JP2019533410A priority patent/JP7437935B2/en
Priority to EP23196560.9A priority patent/EP4310075A3/en
Priority to CN202210319695.4A priority patent/CN114917203A/en
Priority to SI201731472T priority patent/SI3558943T1/en
Priority to HRP20231742TT priority patent/HRP20231742T1/en
Priority to PL17826404.0T priority patent/PL3558943T3/en
Priority to IL306099A priority patent/IL306099A/en
Priority to KR1020217027797A priority patent/KR102385562B1/en
Publication of WO2018119163A1 publication Critical patent/WO2018119163A1/en
Priority to IL267511A priority patent/IL267511B/en
Priority to AU2021200663A priority patent/AU2021200663B2/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C333/00Derivatives of thiocarbamic acids, i.e. compounds containing any of the groups, the nitrogen atom not being part of nitro or nitroso groups
    • C07C333/02Monothiocarbamic acids; Derivatives thereof
    • C07C333/04Monothiocarbamic acids; Derivatives thereof having nitrogen atoms of thiocarbamic groups bound to hydrogen atoms or to acyclic carbon atoms
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics

Definitions

  • nucleic acids are currently being developed as therapeutics for the treatment of a number of diseases.
  • these molecules are being developed, there has been developed a need to produce them in a form that is stable and has a long shelf-life and that can be easily incorporated into an anhydrous organic or anhydrous polar aprotic solvent to enable encapsulations of the nucleic acids without the side-reactions that can occur in a polar aqueous solution or nonpolar solvents.
  • the description herein relates to novel lipid compositions that facilitate the intracellular delivery of biologically active and therapeutic molecules.
  • the description relates also to pharmaceutical compositions that comprise such lipid compositions, and that are useful to deliver therapeutically effective amounts of biologically active molecules into the cells of patients.
  • the delivery of a therapeutic compound to a subject is important for its therapeutic effects and usually it can be impeded by limited ability of the compound to reach targeted cells and tissues. Improvement of such compounds to enter the targeted cells of tissues by a variety of means of delivery is crucial.
  • the description herein relates the novel lipids, in compositions and methods for preparation that facilitate the targeted intracellular delivery of biological active molecules.
  • biologically active molecules for which effective targeting to a patient's tissues is often not achieved include: numerous proteins including immunoglobulin proteins, polynucleotides such as genomic DNA, cDNA, or mRNA antisense
  • polynucleotides and many low molecular weight compounds, whether synthetic or naturally occurring, such as the peptide hormones and antibiotics.
  • nucleic acids are currently being developed as therapeutics for the treatment of a number of diseases. These nucleic acids include mRNA for gene expression, DNA in gene therapy, plasmids, small interfering nucleic acids (siNA), siRNA, and microRNA (miRNA) for use in RNA interference (RNAi), antisense molecules, ribozymes, antagomirs, and aptamers.
  • mRNA small interfering nucleic acids
  • siRNA small interfering nucleic acids
  • miRNA microRNA
  • antisense molecules ribozymes
  • antagomirs antagomirs
  • aptamers RNA interference
  • Ri is a branched chain alkyl consisting of 10 to 31 carbons
  • R2 is a linear alkyl, alkenyl, or alkynyl consisting of 2 to 20 carbons, or a branched chain alkyl consisting of 10 to 31 carbons;
  • Li and L2 are the same or different, each a linear alkane of 1 to 20 carbons or a linear alkene of 2 to 20 carbons;
  • Xi is S or O
  • R3 is a linear or branched alkylene consisting of 1 to 6 carbons
  • R4 and R5 are the same or different, each a hydrogen or a linear or branched alkyl consisting of 1 to 6 carbons; or
  • Ri is a branched, noncyclic alkyl or alkenyl of 8, 9, 10, 11, 12, 13, 14, 16, 17, 18, 19,
  • Li is linear alkane of 1 to 15 carbons
  • R21S a linear alkyl or alkenyl of 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15 carbons or a branched, noncyclic alkyl or alkenyl of 8, 9, 10, 11, 12, 13, 14, 16, 17, 18, 19, 20,
  • L2 is a linear alkane of 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15 carbons
  • X is O or S
  • R3 is a linear alkane of 1, 2, 3, 4, 5, or 6 carbons
  • R4 and R5 are the same or different, each a linear or branched, noncyclic alkyl of 1, 2, 3, 4, 5, or 6 carbons;
  • Ri has 8, 9, 10, 11, 12, 13, 14, 16, or 17 carbons.
  • Ri comprises two identical alkyl or alkenyl groups.
  • Ri comprises an alkenyl group.
  • R2 is an alkenyl
  • R2 is a branched, noncyclic alkyl.
  • L2 has 4, 5, 6, or 7 carbons.
  • the compound is selected from the group consisting of compounds of formulas ATX-0082, ATX-0085, ATX-0083, ATX-0121, ATX-0091, ATX- 0102, ATX-0098, ATX-0092, ATX-0084, ATX-0095, ATX-0125, ATX-0094, ATX-0109, ATX-0110, ATX-0118, ATX-0108, ATX-0107, ATX-0093, ATX-0097, and ATX-0096
  • Ri is a branched, noncyclic alkyl or alkenyl of 12, 13, 14, 16, 17, 18, 19, 20, 21, or 22 carbons;
  • Li is a linear alkane of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 carbons;
  • R21S linear alkyl or alkenyl of 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15 carbons or a
  • L 2 is a linear alkane of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15 carbons
  • X is O or S
  • R3 is linear alkane of 1, 2, 3, 4, 5, or 6 carbons
  • R4 and R5 are the same or different, each a linear or branched, noncyclic alkyl of 1, 2, 3, 4, 5, or 6 carbons;
  • a 1 mM solution of the compound has a pKa of 5.6 to 7.0 as measured by
  • the compound has a c-LogD value is between 10 and 14;
  • Ri has 12, 13, 14, 16, or 17 carbons.
  • Ri comprises two identical alkyl or alkenyl groups.
  • Ri comprises an alkyl group.
  • R2 is an alkenyl
  • R2 is a branched, noncyclic alkyl.
  • Li and L2 independently have 1, 2, or 3 carbons.
  • Li and L2 both have 3 carbons.
  • R3 is propylene or butylene.
  • the compound's c-LogD value is at least 11 and its measured pKa more basic than 6.
  • the compound is selected from the group consisting of compounds of formulas ATX-0111, ATX-0132, ATX-0134, ATX-0100, ATX-0117, ATX-0114, ATX-01 15. ATX-0101, ATX-0106.
  • Ri is a branched, noncyclic alkyl of 12, 13, 14, 16, 17, 18, 19, 20, 21 , or 22 carbons;
  • Li is a linear alkane of 1 , 2, or 3carbons;
  • R2 is linear alkenyl of 8, 9, 10, 11 , or 12 carbons or a branched, noncyclic alkyl of 12,
  • L2 is a linear alkane of 1 , 2, or 3 carbons
  • X is O or S
  • R3 is linear alkane of 2 or 3 carbons
  • R4 and R5 are the same or different, each a linear or branched, noncyclic alkyl of 1 or 2 carbons; or a pharmaceutically acceptable salt or solvate thereof.
  • cationic lipids described herein are in a
  • the pharmaceutical composition preferably comprises a lipid nanoparticle comprising a nucleic acid, preferably a RNA polynucleotide.
  • the lipid nanoparticle preferably increases the lifetime of RNA in the circulation.
  • the lipid nanoparticle therein delivers the nucleic acid to cells in the body.
  • the nucleic acid has an activity of suppressing the expression of a target gene.
  • the nucleic acid has an activity of increasing production of a protein it encodes upon expression in cells of the body.
  • compositions for introducing a nucleic acid into a cell of a mammal by using any of the compositions, above.
  • the cell may be in a liver, lung, kidney, brain, blood, spleen, or bone.
  • the composition preferably is administered intravenously, subcutaneously, intraperitoneally, or intrathecally.
  • the composition preferably is administered intravenously, subcutaneously, intraperitoneally, or intrathecally.
  • compositions described herein are used in a method for treating cancer or inflammatory disease.
  • the disease may be one selected from the group consisting of immune disorder, cancer, renal disease, fibrotic disease, genetic abnormality, inflammation, and cardiovascular disorder.
  • FIG. 1 shows the synthetic pathway of ATX-0043 from hexanoate (SM 1), 4-aminobutanoic acid (SM 2), and 4-bromobutyric acid (SM 3). Intermediates (Ints) 1-8 and reactions are described in Example 2.
  • FIG. 2 shows the synthetic pathway of ATX-0057 from octanoate (SM 1), 4- aminobutanoic acid (SM 2), and 4-bromobutyric acid (SM 3). Ints 1-8 and reactions are described in Example 3.
  • FIG. 3 shows the synthetic pathway of ATX-0058 from SM 1, SM 2 and SM3, which are the same as in FIG. 2. Ints 1-7 and reactions are described in Example 4.
  • FIG. 4 shows the synthetic pathway of ATX-0081 from SM 1, SM 2 and SM3, which are the same as in FIG. 2. Ints 1-8 and reactions are described in Example 5.
  • FIG. 5 shows the synthetic pathway of ATX-0082 from SM 1, SM 2 and SM3, which are the same as in FIG. 2. Ints 1-7 and reactions are described in Example 6.
  • FIG. 6 shows the synthetic pathway of ATX-0086 from SM 1, SM 2 and SM3, which are the same as in FIG. 2. Ints 1-8 and reactions are described in Example 7.
  • FIG. 7 shows the synthetic pathway of ATX-0087 from SM 1, SM 2 and SM3, which are the same as in FIG. 2. Ints 1-8 and reactions are described in Example 8.
  • FIG. 8 shows the synthetic pathway of ATX-0088 from SM 1, SM 2 and SM3, which are the same as in FIG. 2. Ints 1-8 and reactions are described in Example 9.
  • FIG. 9 shows the synthetic pathway of ATX-0083 from SM 1, SM 2 and SM3, which are the same as in FIG. 2. Ints 1-8 and reactions are described in Example 10.
  • FIG. 10 shows the synthetic pathway of ATX-0084 from SM 1, SM 2 and SM3, which are the same as in FIG. 2. Ints 1-8 and reactions are described in Example 11.
  • FIG. 11 shows the synthetic pathway of ATX-0061 from SM 1 and SM 2, which are the same as in FIG. 1. Ints 1-5 and reactions are described in Example 12.
  • FIG. 12 shows the synthetic pathway of ATX-0063 from SM 1 and SM 2, which are the same as in FIG. 1. Ints 1-5 and reactions are described in Example 13.
  • FIG. 13 shows the synthetic pathway of ATX-0064 from SM 1 and SM 2, which are the same as in FIG. 1. Ints 1-5 and reactions are described in Example 14.
  • FIG. 14 shows the synthetic pathway of ATX-0081. Ints 1-6 and reactions are described in Example 15.
  • FIG. 15 shows the synthetic pathway of ATX-0085 from SM 1, SM 2 and SM3, which are the same as in FIG. 2. Ints 1-11 and reactions are described in Example 16.
  • FIG. 16 shows the synthetic pathway of ATX-0134. Ints 1-6 and reactions are described in Example 17.
  • FIG. 17 shows the EPO mRNA levels (ng/ml) following injection of 0.03 mg/kg and 0.1 mg/kg mRNA in nanoparticles comprising ATX-002, ATX-0057, ATX-0081. ATX-0082, ATX-0083, ATX-0084, ATX-0085, ATX-0086. or ATX-0087 cationic lipid into mice.
  • FIG. 18 shows the anti-Factor VII knockdown activity of liposomes with ATX-0057 and ATX-0058 vs. the activity of ATX-002 and control (PBS alone).
  • FIG. 19 shows the anti-EPO knockdown activity of liposomes with ATX- 0057 vs. the activity of ATX-002.
  • composition means a product comprising the specified ingredients in the specified amounts, as well as any product that results, directly or indirectly, from
  • “In combination with” means the administration of a compound of formula I with other medicaments in the methods of treatment of this invention, meaning that the compounds of formula I and the other medicaments are administered sequentially or concurrently in separate dosage forms, or are administered concurrently in the same dosage form.
  • “Mammal” means a human or other mammal, or means a human being.
  • Patient means both human and other mammals, preferably human.
  • Alkyl means a saturated or unsaturated, straight or branched, hydrocarbon chain.
  • the alkyl group has 1-18 carbons, i.e. is a Ci-Cls group, or is a C1-C12 group, a C1-C6 group, or a C1-C4 group.
  • the alkyl group has zero branches (i.e. , is a straight chain), one branch, two branches, or more than two branches.
  • Alkenyl is an unsaturated alkyl that may have one double bond, two double bonds, or more than two double bonds.
  • Alkynyl is an unsaturated alkyl that may have one triple bond, two triple bonds, or more than two triple bonds.
  • Alkyl chains may be optionally substituted with 1 substituent (i.e. , the alkyl group is mono-substituted), or 1-2 substituents, or 1-3 substituents, or 1-4 substituents, etc.
  • the substituents may be selected from the group consisting of hydroxy, amino, alkylamino, boronyl, carboxy, nitro, cyano, and the like.
  • the alkyl group incorporates one or more heteroatoms, the alkyl group is referred to herein as a heteroalkyl group.
  • the substituents on an alkyl group are hydrocarbons, then the resulting group is simply referred to as a substituted alkyl.
  • the alkyl group including substituents has less than 25, 24, 23, 22, 21, 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, or 7 carbons.
  • “Lower alkyl” means a group having one to six carbons in the chain which chain may be straight or branched.
  • suitable alkyl groups include methyl, ethyl, n-propyl, isopropyl, n-butyl, t-butyl, n-pentyl, and hexyl.
  • Alkoxy means an alkyl-O-group wherein alkyl is as defined above.
  • Non- limiting examples of alkoxy groups include: methoxy, ethoxy, n-propoxy, isopropoxy, n- butoxy and heptoxy.
  • the bond to the parent moiety is through the ether oxygen.
  • Alkoxyalkyl means an alkoxy-alkyl-group in which the alkoxy and alkyl are as previously described. Preferred alkoxyalkyl comprise a lower alkyl group. The bond to the parent moiety is through the alkyl.
  • Alkylaryl means an alkyl-aryl-group in which the alkyl and aryl are as previously described. Preferred alkylaryls comprise a lower alkyl group. The bond to the parent moiety is through the aryl.
  • Aminoalkyl means an NH2-alkyl-group, wherein alkyl is as defined above, bound to the parent moiety through the alkyl group.
  • Carboxyalkyl means an HOOC-alkyl-group, wherein alkyl is as defined above, bound to the parent moiety through the alkyl group.
  • “Commercially available chemicals” and the chemicals used in the Examples set forth herein may be obtained from standard commercial sources, where such sources include, for example, Acros Organics (Pittsburgh, Pa.), Sigma-Adrich Chemical (Milwaukee, Wis.), Avocado Research (Lancashire, U.K.), Bionet (Cornwall, U.K.), Boron Molecular (Research Triangle Park, N.C.), Combi-Blocks (San Diego, Calif), Eastman Organic Chemicals, Eastman Kodak Company (Rochester, N.Y.), Fisher Scientific Co. (Pittsburgh, Pa.), Frontier Scientific (Logan, Utah), ICN Biomedicals, Inc.
  • Halo means fluoro, chloro, bromo, or iodo groups. Preferred are fluoro, chloro or bromo, and more preferred are fluoro and chloro.
  • Halogen means fluorine, chlorine, bromine, or iodine. Preferred are fluorine, chlorine and bromine.
  • Heteroalkyl means a saturated or unsaturated, straight or branched, chain containing carbon and at least one heteroatom.
  • the heteroalkyl group may, in various embodiments, have one heteroatom, or 1-2 heteroatoms, or 1-3 heteroatoms, or 1-4 heteroatoms.
  • the heteroalkyl chain contains from 1 to 18 (i.e., 1-18) member atoms (carbon and heteroatoms), and in various embodiments contain 1-12, or 1-6, or 1-4 member atoms.
  • the heteroalkyl group has zero branches (i.e., is a straight chain), one branch, two branches, or more than two branches.
  • the hetereoalkyl group is saturated.
  • the heteroalkyl group is unsaturated.
  • the unsaturated heterolkyl may have one double bond, two double bonds, more than two double bonds, and/or one triple bond, two triple bonds, or more than two triple bonds.
  • Heteroalkyl chains may be substituted or unsubstituted.
  • the heteroalkyl chain is unsubstituted.
  • the heteroalkyl chain is substituted.
  • a substituted heteroalkyl chain may have 1 substituent (i.e. , by monosubstituted), or may have, e.g. , 1-2 substituents, or 1-3 substituents, or 1-4 substituents.
  • Exemplary heteroalkyl substituents include esters (— C(O)— O— R) and carbonyls (— C(O)— ).
  • Hydroxyalkyl means an HO-alkyl-group, in which alkyl is previously defined. Preferred hydroxyalkyls contain lower alkyl. Non-limiting examples of suitable hydroxyalkyl groups include hydroxymethyl and 2 -hydroxy ethyl.
  • Hydrate means a solvate wherein the solvent molecule is H2O.
  • Lipid means an organic compound that comprises an ester of fatty acid and is characterized by being insoluble in water, but soluble in many organic solvents. Lipids are usually divided into at least three classes: (1) “simple lipids,” which include fats and oils as well as waxes; (2) “compound lipids,” which include phospholipids, glycolipids, cationic lipids, non-cationic lipids, neutral lipids, and anionic lipids, all described in more detail herein; and (3) "derived lipids” such as steroids.
  • Lipid particle means a lipid formulation that can be used to deliver a therapeutic nucleic acid (e.g. , mRNA) to a target site of interest (e.g., cell, tissue, organ, and the like).
  • the lipid particle is a nucleic acid-lipid particle, which is typically formed from a cationic lipid, a non-cationic lipid (e.g. , a phospholipid), a conjugated lipid that prevents aggregation of the particle (e.g. , a PEG-lipid), and optionally cholesterol.
  • the therapeutic nucleic acid e.g., mRNA
  • Lipid particles typically have a mean diameter of from 30 nm to 150 nm, from 40 nm to 150 nm, from 50 nm to 150 nm, from 60 nm to 130 nm, from 70 nm to 110 nm, from 70 nm to 100 nm, from 80 nm to 100 nm, from 90 nm to 100 nm, from 70 to 90 nm, from 80 nm to 90 nm, from 70 nm to 80 nm, or 30 nm, 35 nm, 40 nm, 45 nm, 50 nm, 55 nm, 60 nm, 65 nm, 70 nm, 75 nm, 80 nm, 85 nm, 90 nm, 95 nm, 100 nm, 105 nm, 110 nm, 1 15 nm, 120 nm, 125 nm, 130 nm, 135 nm, 140 nm, 145 nm,
  • Lipid encapsulated means a lipid particle that provides a therapeutic nucleic acid such as an mRNA with full encapsulation, partial encapsulation, or both.
  • the nucleic acid e.g. , mRNA
  • the nucleic acid is fully encapsulated in the lipid particle.
  • Lipid conjugate means a conjugated lipid that inhibits aggregation of lipid particles.
  • 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, cationic PEG lipids, polyoxazoline (POZ)-lipid conjugates, polyamide oligomers, and mixtures thereof.
  • 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 phosphat
  • PEG 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. , non-ester-containing linker moieties and ester-containing linker moieties.
  • non-ester-containing linker moieties such as amides or carbamates, are used.
  • Amphipathic lipid means the material in which the hydrophobic portion of the lipid material orients into a hydrophobic phase, while the hydrophilic portion orients toward the aqueous phase.
  • Hydrophilic characteristics derive from the presence of polar or charged groups such as carbohydrates, phosphate, carboxylic, sulfate, amino, sulfhydryl, nitro, hydroxyl, and other like groups. Hydrophobicity can be conferred by the inclusion of apolar groups that include, but are not limited to, long-chain saturated and unsaturated aliphatic hydrocarbon groups and such groups substituted by one or more aromatic, cycloaliphatic, or heterocyclic group(s). Examples of amphipathic compounds include, but are not limited to, phospholipids, aminolipids, and sphingolipids.
  • phospholipids include, but are not limited to, phosphatidylcholine, phosphatidylethanolamine, phosphatidylserine, phosphatidylinositol, phosphatidic acid, palmitoyloleoyl phosphatidylcholine, lysophosphatidylcholine, lysophosphatidylethanolamine, dipalmitoylphosphatidylcholine, dioleoylphosphatidylcholine, distearoylphosphatidylcholine, and dilinoleoylphosphatidylcholine.
  • amphipathic lipids Other compounds lacking in phosphorus, such as sphingolipid, glycosphingolipid families, diacylglycerols, and ⁇ -acyloxyacids, are also within the group designated as amphipathic lipids. Additionally, the amphipathic lipids described above can be mixed with other lipids including triglycerides and sterols.
  • Neutral lipid means a lipid species that exist either in an uncharged or neutral zwitterionic form at a selected pH. At physiological pH, such lipids include, for example, diacylphosphatidylcholine, diacylphosphatidylethanolamine, ceramide,
  • Non-cationic lipid means an amphipathic lipid or a neutral lipid or anionic lipid, and is described in more detail below.
  • Anionic lipid means a lipid that is negatively charged at physiological pH. These lipids include, but are not limited to, phosphatidylglycerols, cardiolipins,
  • diacylphosphatidylserines diacylphosphatidic acids, N-dodecanoyl
  • phosphatidylethanolamines N-succinyl phosphatidylethanolamines, N- glutarylphosphatidylethanolamines, lysylphosphatidylglycerols,
  • POPG palmitoyloleyolphosphatidylglycerol
  • Hydrophilic lipids means compounds having apolar groups 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.
  • “Cationic lipid” and “amino lipid” are used interchangeably mean those lipids and salts thereof having one, two, three, or more fatty acid or fatty alkyl chains and a pH-titratable amino head group (e.g. , an alkylamino or dialkylamino head group).
  • the cationic lipid is typically protonated (i.e. , positively charged) at a pH below the pKa of the cationic lipid and is substantially neutral at a pH above the pKa.
  • the cationic lipids of the invention may also be termed titratable cationic lipids.
  • the cationic lipids comprise: a protonatable tertiary amine (e.g. , pH-titratable) head group; Ci8 alkyl chains, wherein each alkyl chain independently has 0 to 3 (e.g., 0, 1, 2, or 3) double bonds; and ether, ester, or ketal linkages between the head group and alkyl chains.
  • a protonatable tertiary amine e.g. , pH-titratable
  • Ci8 alkyl chains wherein each alkyl chain independently has 0 to 3 (e.g., 0, 1, 2, or 3) double bonds
  • ether, ester, or ketal linkages between the head group and alkyl chains e.g., 1, 2, or 3
  • Such cationic lipids include, but are not limited to, DSDMA, DODMA, DLinDMA, DLenDMA, ⁇ - DLenDMA, DLin-K-DMA, DLin-K-C2-DMA (also known as DLin-C2K-DMA, XTC2, and C2K), DLin-K-C3 -DM A, DLin-K-C4-DMA, DLen-C2K-DMA, y-DLen-C2K-DMA, DLin- M-C2-DMA (also known as MC2), DLin-M-C3 -DMA (also known as MC3) and (DLin-MP- DMA)(also known as 1-Bl 1).
  • Substituted means substitution with specified groups other than hydrogen, or with one or more groups, moieties, or radicals which can be the same or different, with each, for example, being independently selected.
  • Antisense nucleic acid means a non-enzymatic nucleic acid molecule that binds to target RNA by means of RNA-RNA or RNA-DNA or RNA-PNA (protein nucleic acid; Egholm et al., 1993 Nature 365, 566) interactions and alters the activity of the target RNA (for a review, see Stein and Cheng, 1993 Science 261, 1004 and Woolf et al, U.S. Pat. No. 5,849,902).
  • antisense molecules are complementary to a target sequence along a single contiguous sequence of the antisense molecule.
  • an antisense molecule can bind to substrate such that the substrate molecule forms a loop, and/or an antisense molecule can bind such that the antisense molecule forms a loop.
  • the antisense molecule can be complementary to two (or even more) noncontiguous substrate sequences or two (or even more) non-contiguous sequence portions of an antisense molecule can be complementary to a target sequence or both.
  • antisense DNA can be used to target RNA by means of DNA-RNA interactions, thereby activating RNase H, which digests the target RNA in the duplex.
  • the antisense oligonucleotides can comprise one or more RNAse H activating region, which is capable of activating RNAse H cleavage of a target RNA.
  • Antisense DNA can be synthesized chemically or expressed via the use of a single stranded DNA expression vector or equivalent thereof.
  • Antisense RNA is an RNA strand having a sequence complementary to a target gene mRNA, that can induce RNAi by binding to the target gene mRNA.
  • Antisense RNA is an RNA strand having a sequence complementary to a target gene mRNA, and thought to induce RNAi by binding to the target gene mRNA.
  • Sense RNA has a sequence complementary to the antisense RNA, and annealed to its complementary antisense RNA to form iNA. These antisense and sense RNAs have been conventionally synthesized with an RNA synthesizer.
  • Nucleic acid means deoxyribonucleotides or ribonucleotides and polymers thereof in single- or double-stranded form.
  • the term encompasses nucleic acids containing known nucleotide analogs or modified backbone residues or linkages, which are synthetic, naturally occurring, and non-naturally occurring, which have similar binding properties as the reference nucleic acid, and which are metabolized in a manner similar to the reference nucleotides. Examples of such analogs include, without limitation, phosphorothioates, phosphoramidates, methyl phosphonates, chiral-methyl phosphonates, 2'-0-methyl ribonucleotides, peptide-nucleic acids (PNAs).
  • PNAs peptide-nucleic acids
  • RNA means a molecule comprising at least one ribonucleotide residue.
  • ribonucleotide is meant a nucleotide with a hydroxyl group at the 2' position of a ⁇ -D-ribo- furanose moiety.
  • the terms include double-stranded RNA, single-stranded RNA, isolated RNA such as partially purified RNA, essentially pure RNA, synthetic RNA, recombinantly produced RNA, as well as altered RNA that differs from naturally occurring RNA by the addition, deletion, substitution, and/or alteration of one or more nucleotides.
  • Such alterations can include addition of non-nucleotide material, such as to the end(s) of an interfering RNA or internally, for example at one or more nucleotides of the RNA.
  • Nucleotides in the RNA molecules of the instant invention can also comprise non-standard nucleotides, such as non- naturally occurring nucleotides or chemically synthesized nucleotides or deoxynucleotides. These altered RNAs can be referred to as analogs or analogs of naturally-occurring RNA.
  • ribonucleic acid and "RNA” refer to a molecule containing at least one ribonucleotide residue, including siRNA, antisense RNA, single stranded RNA, microRNA, mRNA, noncoding RNA, and multivalent RNA.
  • a ribonucleotide is a nucleotide with a hydroxyl group at the 2' position of a B-D-ribo-furanose moiety.
  • RNA double-stranded RNA
  • single-stranded RNA isolated RNA such as partially purified RNA, essentially pure RNA, synthetic RNA, recombinantly produced RNA, as well as modified and altered RNA that differs from naturally occurring RNA by the addition, deletion, substitution, modification, and/or alteration of one or more nucleotides.
  • Alterations of an RNA can include addition of non-nucleotide material, such as to the end(s) of an interfering RNA or internally, for example at one or more nucleotides of an RNA nucleotides in an RNA molecule include non-standard nucleotides, such as non-naturally occurring nucleotides or chemically synthesized nucleotides or deoxynucleotides. These altered RNAs can be referred to as analogs.
  • Nucleotides means natural bases (standard) and modified bases well known in the art. Such bases are generally located at the ⁇ position of a nucleotide sugar moiety. Nucleotides generally comprise a base, sugar, and a phosphate group. The nucleotides can be unmodified or modified at the sugar, phosphate, and/or base moiety, (also referred to interchangeably as nucleotide analogs, modified nucleotides, non-natural nucleotides, non-standard nucleotides and other; see, for example, Usman and McSwiggen, supra; Eckstein, et al, International PCT Publication No.
  • base modifications that can be introduced into nucleic acid molecules include: inosine, purine, pyridin-4-one, pyridin-2-one, phenyl, pseudouracil, 2,4,6- trimethoxy benzene, 3-methyl uracil, dihydrouridine, naphthyl, aminophenyl, 5- alkylcytidines (e.g. , 5-methylcytidine), 5-alkyluridines (e.g.
  • modified bases in this aspect is meant nucleotide bases other than adenine, guanine, cytosine, and uracil at ⁇ position or their equivalents.
  • Complementary nucleotide bases means a pair of nucleotide bases that form hydrogen bonds with each other.
  • nucleic acid can form hydrogen bond(s) with another nucleic acid sequence either by traditional Watson-Crick or by other non-traditional modes of binding.
  • miRNA means single-stranded RNA molecules of 21-23 nucleotides in length, which regulate gene expression miRNAs are encoded by genes that are transcribed from DNA but not translated into protein (non-coding RNA); instead they are processed from primary transcripts known as pri-miRNA to short stem-loop structures called pre-miRNA and finally to functional miRNA. Mature miRNA molecules are partially complementary to one or more messenger RNA (mRNA) molecules, and their main function is to downregulate gene expression
  • mRNA messenger RNA
  • siRNA mean a class of double-stranded RNA molecules, 16-40 nucleotides in length, that play a variety of roles in biology. Most notably, siRNA is involved in the RNA interference (RNAi) pathway, where it interferes with the expression of a specific gene. In addition to their role in the RNAi pathway, siRNAs also act in RNAi-related pathways, e.g. , as an antiviral mechanism or in shaping the chromatin structure of a genome; the complexity of these pathways is only now being elucidated.
  • RNAi RNA interference
  • RNAi means an RNA-dependent gene silencing process that is controlled by the RNA-induced silencing complex (RISC) and is initiated by short double-stranded RNA molecules in a cell, where they interact with the catalytic RISC component argonaute.
  • RISC RNA-induced silencing complex
  • the double-stranded RNA or RNA-like iNA or siRNA is exogenous (coming from infection by a virus with an RNA genome or from transfected iNA or siRNA), the RNA or iNA is imported directly into the cytoplasm and cleaved to short fragments by the enzyme dicer.
  • the initiating dsRNA can also be endogenous (originating in the cell), as in pre- microRNAs expressed from RNA-coding genes in the genome.
  • RNA-induced silencing complex RISC
  • argonaute proteins endonucleases
  • salts denotes acidic salts formed with inorganic and/or organic acids, as well as basic salts formed with inorganic and/or organic bases.
  • compound of formula I contain both a basic moiety, such as, but not limited to, a pyridine or imidazole, and an acidic moiety, such as, but not limited to, a carboxylic acid, zwitterions ("inner salts") may be formed and are included within the term “salt(s)" as used herein.
  • the salts can be pharmaceutically acceptable (i. e.
  • salts of a compound of formula I may be formed, for example, by reacting a compound of formula I with an amount of acid or base, such as an equivalent amount, in a medium such as one in which the salt precipitates or in an aqueous medium followed by lyophilization.
  • Exemplary acid addition salts include acetates, adipates, alginates, ascorbates, aspartates, benzoates, benzenesulfonates, bisulfates, borates, butyrates, citrates, camphorates, camphorsulfonates, cyclopentanepropionates, digluconates, dodecylsulfates, ethanesulfonates, fumarates, glucoheptanoates, glycerophosphates, hemisulfates, heptanoates, hexanoates, hydrochlorides, hydrobromides, hydroiodides, 2 -hydroxy ethanesulfonates, lactates, maleates, methanesulfonates, 2-napthalenesulfonates, nicotinates, nitrates, oxalates, pectinates, persulfates, 3-phenylpropionates
  • Exemplary basic salts include ammonium salts, alkali metal salts such as sodium, lithium, and potassium salts, alkaline earth metal salts such as calcium and magnesium salts, salts with organic bases (for example, organic amines) such as benzathines, dicyclohexylamines, hydrabamines (formed with N,N-bis(dehydroabietyl)ethylenediamine), N-methyl-D-glucamines, N-methyl-D-glucamides, t-butyl amines, and salts with amino acids such as arginine or lysine.
  • Basic nitrogen-containing groups may be quartemized with agents such as lower alkyl halides (e.g.
  • dialkyl sulfates e.g., dimethyl, diethyl, dibutyl, and diamyl sulfates
  • long chain halides e.g., decyl, lauryl, myristyl, and stearyl chlorides, bromides, and iodides
  • arylalkyl halides e.g., benzyl and
  • Compound of formula I can exist in unsolvated and solvated forms, including hydrated forms.
  • the solvated forms with pharmaceutically acceptable solvents such as water, ethanol, and the like, are equivalent to the unsolvated forms for the purposes of this disclosure.
  • polymorphs of the compound of this disclosure i.e. , polymorphs of the compound of formula I are within the scope of this disclosure.
  • Individual stereoisomers of the compound of this disclosure may, for example, be substantially free of other isomers, or may be admixed, for example, as racemates or with all other, or other selected, stereoisomers.
  • the chiral centers of the compound herein can have the S or R configuration as defined by the IUPAC 1974 Recommendations.
  • the use of the terms “salt”, “solvate”, and the like, is intended to equally apply to the salt and solvate of enantiomers, stereoisomers, rotamers, tautomers, racemates, or prodrugs of the disclosed compound.
  • Classes of compounds that can be used as the chemotherapeutic agent include: alkylating agents, antimetabolites, natural products and their derivatives, hormones and steroids (including synthetic analogs), and synthetics. Examples of compounds within these classes are given below.
  • a compound of formula I includes a pharmaceutically acceptable salt thereof, in a lipid composition, comprising a nanoparticle or a bilayer of lipid molecules.
  • the lipid bilayer preferably further comprises a neutral lipid or a polymer.
  • the lipid composition preferably comprises a liquid medium.
  • the composition preferably further encapsulates a nucleic acid.
  • the nucleic acid preferably has an activity of suppressing the expression of the target gene by utilizing RNA interference (RNAi).
  • RNAi RNA interference
  • the lipid composition preferably further comprises a nucleic acid and a neutral lipid or a polymer.
  • the lipid composition preferably encapsulates the nucleic acid.
  • the description provides lipid particles comprising one or more therapeutic mRNA molecules encapsulated within the lipid particles.
  • the mRNA is fully encapsulated within the lipid portion of the lipid particle such that the mRNA in the lipid particle is resistant in aqueous solution to nuclease degradation.
  • the lipid particles described herein are substantially non-toxic to mammals such as humans.
  • the lipid particles typically have a mean diameter of from 30 nm to 150 nm, from 40 nm to 150 nm, from 50 nm to 150 nm, from 60 nm to 130 nm, from 70 nm to 110 nm, or from 70 to 90 nm.
  • the lipid particles of the invention also typically have a lipid:RNA ratio (mass/mass ratio) of from 1 : 1 to 100: 1, from 1 : 1 to 50: 1, from 2: 1 to 25: 1, from 3: 1 to 20: 1, from 5: 1 to 15: 1, or from 5: 1 to 10: 1, or from 10: 1 to 14: 1, or from 9: 1 to 20: 1.
  • the lipid particles have a lipid: RNA ratio (mass/mass ratio) of 12: 1.
  • the lipid particles have a lipid: mRNA ratio (mass/mass ratio) of 13: 1.
  • the lipid particles comprise an mRNA, a cationic lipid (e.g., one or more cationic lipids or salts thereof described herein), a phospholipid, and a conjugated lipid that inhibits aggregation of the particles (e.g. , one or more PEG-lipid conjugates).
  • the lipid particles can also include cholesterol.
  • the lipid particles may comprise at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more mRNA that express one or more polypeptides.
  • the mRNA may be fully encapsulated within the lipid portion of the particle, thereby protecting the nucleic acid from nuclease degradation.
  • a lipid particle comprising an mRNA is fully encapsulated within the lipid portion of the particle, thereby protecting the nucleic acid from nuclease degradation.
  • the mRNA in the lipid particle is not substantially degraded after exposure of the particle to a nuclease at 37°C for at least 20, 30, 45, or 60 minutes.
  • the mRNA in the lipid particle is not substantially degraded after incubation of the particle in serum at 37°C for at least 30, 45, or 60 minutes or at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, or 36 hours.
  • the mRNA is complexed with the lipid portion of the particle.
  • the nucleic acid-lipid particle compositions are substantially non-toxic to mammals such as humans.
  • “Fully encapsulated” means that the nucleic acid (e.g. , mRNA) in the nucleic acid-lipid particle is not significantly degraded after exposure to serum or a nuclease assay that would significantly degrade free RNA. When fully encapsulated, preferably less than 25% of the nucleic acid in the particle is degraded in a treatment that would normally degrade 100% of free nucleic acid, more preferably less than 10%, and most preferably less than 5% of the nucleic acid in the particle is degraded. “Fully encapsulated” also means that the nucleic acid-lipid particles do not rapidly decompose into their component parts upon in vivo administration.
  • the present invention provides a nucleic acid-lipid particle composition comprising a plurality of nucleic acid-lipid particles.
  • the lipid particle comprises mRNA that is fully encapsulated within the lipid portion of the particles, such that from 30% to 100%, from 40% to 100%, from 50% to 100%, from 60% to 100%, from 70% to 100%, from 80% to 100%, from 90% to 100%, from 30% to 95%, from 40% to 95%, from 50% to 95%, from 60% to 95%, from 70% to 95%, from 80% to 95%, from 85% to 95%, from 90% to 95%, from 30% to 90%, from 40% to 90%, from 50% to 90%, from 60% to 90%, from 70% to 90%, from 80% to 90%, or at least 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% (or any fraction thereof or range therein) of the particles have the mRNA encapsulated therein.
  • the proportions of the components can be varied and the delivery efficiency of a particular formulation can be measured using assays know in the art.
  • the description includes synthesis of certain cationic lipid compounds.
  • the compounds are particularly suitable for delivering polynucleotides to cells and tissues as demonstrated in subsequent sections.
  • the lipomacrocycle compound described herein may be used for other purposes as well as, for example, recipients and additives.
  • the cationic lipid compounds may be combined with an agent to form microparticles, nanoparticles, liposomes, or micelles.
  • the agent to be delivered by the particles, liposomes, or micelles may be in the form of a gas, liquid, or solid, and the agent may be a polynucleotide, protein, peptide, or small molecule.
  • the lipomacrocycle compounds may be combined with other cationic lipid compounds, polymers (synthetic or natural), surfactants, cholesterol, carbohydrates, proteins, or lipids, to form the particles. These particles may then optionally be combined with a pharmaceutical excipient to form a pharmaceutical composition.
  • the present description provides novel cationic lipid compounds and drug delivery systems based on the use of such cationic lipid compounds.
  • the system may be used in the pharmaceutical/drug delivery arts to deliver polynucleotides, proteins, small molecules, peptides, antigen, or drugs, to a patient, tissue, organ, or cell.
  • These novel compounds may also be used as materials for coating, additives, excipients, materials, or bioengineering.
  • the cationic lipid compounds of the present description provide for several different uses in the drug delivery art.
  • the amine-containing portion of the cationic lipid compounds may be used to complex polynucleotides, thereby enhancing the delivery of polynucleotide and preventing their degradation.
  • the cationic lipid compounds may also be used in the formation of picoparticles, nanoparticles, microparticles, liposomes, and micelles containing the agent to be delivered.
  • the cationic lipid compounds are biocompatible and biodegradable, and the formed particles are also biodegradable and biocompatible and may be used to provide controlled, sustained release of the agent to be delivered.
  • These and their corresponding particles may also be responsive to pH changes given that these are protonated at lower pH. They may also act as proton sponges in the delivery of an agent to a cell to cause endosome lysis.
  • the cationic lipid compounds are relatively non- cytotoxic.
  • the cationic lipid compounds may be biocompatible and biodegradable.
  • the cationic lipid may have a measured pKa (in the formulation milieu) in the range of approximately 5.5 to approximately 7.5, more preferably between approximately 6.0 and approximately 7.0. It may be designed to have a desired pKa between approximately 3.0 and approximately 9.0, or between approximately 5.0 and approximately 8.0.
  • the cationic lipid compounds described herein are particularly attractive for drug delivery for several reasons: they contain amino groups for interacting with DNA, RNA, other polynucleotides, and other negatively charged agents, for buffering the pH, for causing endo-osmolysis, for protecting the agent to be delivered, they can be synthesized from commercially available starting materials; and/or they are pH responsive and can be engineered with a desired pKa.
  • Non-limiting examples of non-cationic lipids include phospholipids such as lecithin, phosphatidylethanolamine, lysolecithin, lysophosphatidylethanolamine,
  • phosphatidylserine phosphatidylinositol
  • sphingomyelin egg sphingomyelin (ESM)
  • cephalin phosphatidylserine
  • phosphatidylinositol phosphatidylinositol
  • sphingomyelin egg sphingomyelin (ESM)
  • cephalin phosphatidic acid
  • cerebrosides phosphatidic acid
  • dicetylphosphate dicetylphosphate
  • DSPC distearoylphosphatidylcholine
  • DOPC dioleoylphosphatidylcholine
  • DPPC dipalmitoylphosphatidylcholine
  • DOPG dioleoylphosphatidylglycerol
  • dipalmitoylphosphatidylglycerol DPPG
  • dioleoylphosphatidylethanolamine DOPE
  • palmitoyloleoyl-phosphatidylcholine POPC
  • palmitoyloleoyl-phosphatidylethanolamine POPE
  • palmitoyloleyol-phosphatidylglycerol POPG
  • dipalmitoyl- phosphatidylethanolamine DPPE
  • dimyristoyl- phosphatidylethanolamine DMPE
  • distearoyl-phosphatidylethanolamine DSPE
  • monomethyl-phosphatidylethanolamine dimethyl-phosphatidylethanolamine
  • dielaidoyl- phosphatidylethanolamine DEPE
  • 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 oleoyl.
  • non-cationic lipids include sterols such as cholesterol and derivatives thereof.
  • cholesterol derivatives include polar analogues such as 5a-cholestanol, 5a-coprostanol, cholesteryl-(2'-hydroxy)-ethyl ether, cholesteryl-(4'- hydroxy)-butyl ether, and 6-ketocholestanol; non-polar analogues such as 5a- cholestane, cholestenone, 5a-cholestanone, 5a-cholestanone, and cholesteryl decanoate; and mixtures thereof.
  • the cholesterol derivative is a polar analogue such as cholesteryl-(4'-hydroxy)-butyl ether.
  • the non-cationic lipid present in lipid particles comprises or consists of a mixture of one or more phospholipids and cholesterol or a derivative thereof. In other embodiments, the non-cationic lipid present in the lipid particles comprises or consists of one or more phospholipids, e.g., a cholesterol-free lipid particle formulation. In yet other embodiments, the non-cationic lipid present in the lipid particles comprises or consists of cholesterol or a derivative thereof, e.g. , a phospholipid-free lipid particle formulation.
  • non-cationic lipids include nonphosphorous containing lipids such as, e.g. , stearylamine, dodecylamine, hexadecylamine, acetyl palmitate, glycerol ricinoleate, hexadecyl stearate, isopropyl myristate, amphoteric acrylic polymers, triethanolamine-lauryl sulfate, alkyl-aryl sulfate polyethyloxylated fatty acid amides, dioctadecyldimethyl ammonium bromide, ceramide, and sphingomyelin.
  • nonphosphorous containing lipids such as, e.g. , stearylamine, dodecylamine, hexadecylamine, acetyl palmitate, glycerol ricinoleate, hexadecyl stearate, isopropyl my
  • the non-cationic lipid comprises from 10 mol % to 60 mol %, from 20 mol % to 55 mol %, from 20 mol % to 45 mol %, 20 mol % to 40 mol %, from 25 mol % to 50 mol %, from 25 mol % to 45 mol %, from 30 mol % to 50 mol %, from 30 mol % to 45 mol %, from 30 mol % to 40 mol %, from 35 mol % to 45 mol %, from 37 mol % to 42 mol %, or 35 mol %, 36 mol %, 37 mol %, 38 mol %, 39 mol %, 40 mol %, 41 mol %, 42 mol %, 43 mol %, 44 mol %, or 45 mol % (or any fraction thereof or range therein) of the total lipid present in the particle.
  • the mixture may comprise up to 40 mol %, 45 mol %, 50 mol %, 55 mol %, or 60 mol % of the total lipid present in the particle.
  • the phospholipid component in the mixture may comprise from 2 mol % to 20 mol %, from 2 mol % to 15 mol %, from 2 mol % to 12 mol %, from 4 mol % to 15 mol %, or from 4 mol % to 10 mol % (or any fraction thereof or range therein) of the total lipid present in the particle.
  • the phospholipid component in the mixture comprises from 5 mol % to 10 mol %, from 5 mol % to 9 mol %, from 5 mol % to 8 mol %, from 6 mol % to 9 mol %, from 6 mol % to 8 mol %, or 5 mol %, 6 mol %, 7 mol %, 8 mol %, 9 mol %, or 10 mol % (or any fraction thereof or range therein) of the total lipid present in the particle.
  • the cholesterol component in the mixture may comprise from 25 mol % to 45 mol %, from 25 mol % to 40 mol %, from 30 mol % to 45 mol %, from 30 mol % to 40 mol %, from 27 mol % to 37 mol %, from 25 mol % to 30 mol %, or from 35 mol % to 40 mol % (or any fraction thereof or range therein) of the total lipid present in the particle.
  • the cholesterol component in the mixture comprises from 25 mol % to 35 mol %, from 27 mol % to 35 mol %, from 29 mol % to 35 mol %, from 30 mol % to 35 mol %, from 30 mol % to 34 mol %, from 31 mol % to 33 mol %, or 30 mol %, 31 mol %, 32 mol %, 33 mol %, 34 mol %, or 35 mol % (or any fraction thereof or range therein) of the total lipid present in the particle.
  • the cholesterol or derivative thereof may comprise up to 25 mol %, 30 mol %, 35 mol %, 40 mol %, 45 mol %, 50 mol %, 55 mol %, or 60 mol % of the total lipid present in the particle.
  • the cholesterol or derivative thereof in the phospholipid-free lipid particle formulation may comprise from 25 mol % to 45 mol %, from 25 mol % to 40 mol %, from 30 mol % to 45 mol %, from 30 mol % to 40 mol %, from 31 mol % to 39 mol %, from 32 mol % to 38 mol %, from 33 mol % to 37 mol %, from 35 mol % to 45 mol %, from 30 mol % to 35 mol %, from 35 mol % to 40 mol %, or 30 mol %, 31 mol %, 32 mol %, 33 mol %, 34 mol %, 35 mol %, 36 mol %, 37 mol %, 38 mol %, 39 mol %, or 40 mol % (or any fraction thereof or range therein) of the total lipid present in the particle.
  • the non-cationic lipid comprises from 5 mol % to 90 mol %, from 10 mol % to 85 mol %, from 20 mol % to 80 mol %, 10 mol % (e.g. , phospholipid only), or 60 mol % (e.g. , phospholipid and cholesterol or derivative thereof) (or any fraction thereof or range therein) of the total lipid present in the particle.
  • the percentage of non-cationic lipid present in the lipid particles is a target amount, and that the actual amount of non-cationic lipid present in the formulation may vary, for example, by ⁇ 5 mol %.
  • a composition containing a cationic lipid compound may be 30-70% cationic lipid compound, 0-60 % cholesterol, 0-30% phospholipid and 1-10% polyethylene glycol (PEG).
  • the composition is 30-40% cationic lipid compound, 40- 50% cholesterol, and 10-20% PEG.
  • the composition is 50-75% cationic lipid compound, 20-40% cholesterol, and 5-10% phospholipid, and 1-10% PEG.
  • the composition may contain 60-70% cationic lipid compound, 25-35% cholesterol, and 5- 10% PEG.
  • the composition may contain up to 90% cationic lipid compound and 2-15% helper lipid.
  • the formulation may be a lipid particle formulation, for example containing 8-30% compound, 5-30% helper lipid , and 0-20% cholesterol; 4-25% cationic lipid, 4-25% helper lipid, 2- 25% cholesterol, 10- 35% cholesterol-PEG, and 5% cholesterol-amine; or 2- 30% cationic lipid, 2-30% helper lipid, 1- 15% cholesterol, 2- 35% cholesterol-PEG, and 1- 20% cholesterol-amine; or up to 90% cationic lipid and 2-10% helper lipids, or even 100% cationic lipid.
  • the lipid particles described herein may further comprise a lipid conjugate.
  • the conjugated lipid is useful in that it prevents the aggregation of particles.
  • Suitable conjugated lipids include, but are not limited to, PEG-lipid conjugates, cationic-polymer- lipid conjugates, and mixtures thereof.
  • the lipid conjugate is a PEG-lipid.
  • PEG-lipids include, but are not limited to, PEG coupled to dialkyloxypropyls (PEG-DAA), PEG coupled to diacylglycerol (PEG-DAG), PEG coupled to phospholipids such as phosphatidylethanolamine (PEG-PE), PEG conjugated to ceramides, PEG conjugated to cholesterol or a derivative thereof, and mixtures thereof.
  • PEG is a linear, water-soluble polymer of ethylene PEG repeating units with two terminal hydroxyl groups.
  • PEGs are classified by their molecular weights; and include the following: monomethoxypoly ethylene glycol (MePEG-OH), monomethoxypoly ethylene glycol- succinate (MePEG-S), monomethoxypoly ethylene glycol-succinimidyl succinate (MePEG-S- NHS), monomethoxypoly ethylene glycol-amine (MePEG-NEh),
  • MePEG-TRES monomethoxypoly ethylene glycol-tresylate
  • MePEG-IM monomethoxypoly ethylene glycol-imidazolyl-carbonyl
  • HO-PEG-S HO-PEG-S-NHS
  • the PEG moiety of the PEG-lipid conjugates described herein may comprise an average molecular weight ranging from 550 daltons to 10,000 daltons. In certain instances, the PEG moiety has an average molecular weight of from 750 daltons to 5,000 daltons (e.g. , from 1,000 daltons to 5,000 daltons, from 1,500 daltons to 3,000 daltons, from 750 daltons to 3,000 daltons, from 750 daltons to 2,000 daltons). In preferred embodiments, the PEG moiety has an average molecular weight of 2,000 daltons or 750 daltons.
  • the PEG can be optionally substituted by an alkyl, alkoxy, acyl, or aryl group.
  • the PEG 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 to a lipid can be used including, e.g., non-ester-containing linker moieties and ester-containing linker moieties.
  • the linker moiety is a non-ester-containing linker moiety.
  • Suitable non-ester-containing linker moieties include, but are not limited to, amido (- C(O)NH-), amino (-NR-), carbonyl (-C(O)-), carbamate (-NHC(O)O-), urea (-NHC(O)NH-), disulphide (-S-S-), ether (-0-), succinyl (- (0)CCH 2 CH 2 C(0)-), succinamidyl (- NHC(0)CH2CH2C(0)NH-), ether, disulphide, as well as combinations thereof (such as a linker containing both a carbamate linker moiety and an amido linker moiety).
  • a carbamate linker is used to couple the PEG to the lipid.
  • an ester-containing linker moiety is used to couple the PEG to the lipid.
  • Suitable ester-containing linker moieties include, e.g., carbonate (- OC(O)O-), succinoyl, phosphate esters (-O-(O)POH-O-), sulfonate esters, and combinations thereof.
  • Phosphatidylethanolamines having a variety of acyl chain groups of varying chain lengths and degrees of saturation can be conjugated to PEG to form the lipid conjugate.
  • Such phosphatidylethanolamines are commercially available, or can be isolated or synthesized using conventional techniques known to those of skill in the art.
  • Phosphatidylethanolamines containing saturated or unsaturated fatty acids with carbon chain lengths in the range of C10 to C20 are preferred. Phosphatidylethanolamines with mono- or di- unsaturated fatty acids and mixtures of saturated and unsaturated fatty acids can also be used. Suitable phosphatidylethanolamines include, but are not limited to, dimyristoyl- phosphatidylethanolamine (DMPE), dipalmitoyl-phosphatidylethanolamine (DPPE), dioleoyl-phosphatidylethanolamine (DOPE), and distearoyl-phosphatidylethanolamine (DSPE).
  • DMPE dimyristoyl- phosphatidylethanolamine
  • DPPE dipalmitoyl-phosphatidylethanolamine
  • DOPE dioleoyl-phosphatidylethanolamine
  • DSPE distearoyl-phosphatidylethanolamine
  • diacylglycerol or “DAG” includes a compound having 2 fatty acyl chains, R 1 and R 2 , both of which have independently between 2 and 30 carbons bonded to the 1- and 2-position of glycerol by ester linkages.
  • the acyl groups can be saturated or have varying degrees of unsaturation. Suitable acyl groups include, but are not limited to, lauroyl (C12), myristoyl (C14), palmitoyl (Ci6), stearoyl (Cis), and icosoyl (C20).
  • R 1 and R 2 are the same, i.e. , R 1 and R 2 are both myristoyl (i.e. , dimyristoyl), R 1 and R 2 are both stearoyl (i.e. , distearoyl).
  • dialkyloxy propyl or "DAA” includes a compound having 2 alkyl chains, Ri and R2 , both of which have independently between 2 and 30 carbons.
  • the alkyl groups can be saturated or have varying degrees of unsaturation.
  • the PEG-DAA conjugate is a PEG-didecyloxypropyl (C10) conjugate, a PEG-dilauryloxy propyl (C12) conjugate, a PEG-dimyristyloxypropyl (C14) conjugate, a PEG-dipalmityloxy propyl (Ci6) conjugate, or a PEG-distearyloxy propyl (Cis) conjugate.
  • the PEG preferably has an average molecular weight of 750 or 2,000 daltons.
  • the terminal hydroxyl group of the PEG is substituted with a methyl group.
  • hydrophilic polymers can be used in place of PEG.
  • suitable polymers that can be used in place of PEG include, but are not limited to, polyvinylpyrrolidone, polymethyloxazoline, polyethyloxazoline,
  • the lipid conjugate (e.g. , PEG-lipid) comprises from 0.1 mol % to 2 mol %, from 0.5 mol % to 2 mol %, from 1 mol % to 2 mol %, from 0.6 mol % to 1.9 mol %, from 0.7 mol % to 1.8 mol %, from 0.8 mol % to 1.7 mol %, from 0.9 mol % to 1.6 mol %, from 0.9 mol % to 1.8 mol %, from 1 mol % to 1.8 mol %, from 1 mol % to 1.7 mol %, from 1.2 mol % to 1.8 mol %, from 1.2 mol % to 1.7 mol %, from 1.2 mol % to 1.8 mol %, from 1.2 mol % to 1.7 mol %, from 1.3 mol % to 1.6 mol %, or from 1.4 mol % to 1.5 mol % (or any
  • the lipid conjugate (e.g., PEG-lipid) comprises from 0 mol % to 20 mol %, from 0.5 mol % to 20 mol %, from 2 mol % to 20 mol %, from 1.5 mol % to 18 mol %, from 2 mol % to 15 mol %, from 4 mol % to 15 mol %, from 2 mol % to 12 mol %, from 5 mol % to 12 mol %, or 2 mol % (or any fraction thereof or range therein) of the total lipid present in the particle.
  • PEG-lipid comprises from 0 mol % to 20 mol %, from 0.5 mol % to 20 mol %, from 2 mol % to 20 mol %, from 1.5 mol % to 18 mol %, from 2 mol % to 15 mol %, from 4 mol % to 15 mol %, from 2 mol % to 12 mol %, from 5 mol
  • the lipid conjugate (e.g. , PEG-lipid) comprises from 4 mol % to 10 mol %, from 5 mol % to 10 mol %, from 5 mol % to 9 mol %, from 5 mol % to 8 mol %, from 6 mol % to 9 mol %, from 6 mol % to 8 mol %, or 5 mol %, 6 mol %, 7 mol %, 8 mol %, 9 mol %, or 10 mol % (or any fraction thereof or range therein) of the total lipid present in the particle.
  • PEG-lipid comprises from 4 mol % to 10 mol %, from 5 mol % to 10 mol %, from 5 mol % to 9 mol %, from 5 mol % to 8 mol %, from 6 mol % to 9 mol %, from 6 mol % to 8 mol %, or 5 mol %, 6 mol %, 7 mol
  • the percentage of lipid conjugate (e.g., PEG-lipid) present in the lipid particles of the invention is a target amount, and the actual amount of lipid conjugate present in the formulation may vary, for example, by ⁇ 2 mol %.
  • concentration of the lipid conjugate can be varied depending on the lipid conjugate employed and the rate at which the lipid particle is to become fusogenic.
  • composition and concentration of the lipid conjugate By controlling the composition and concentration of the lipid conjugate, 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 becomes fusogenic.
  • other variables including, e.g. , pH, temperature, or ionic strength, can be used to vary and/or control the rate at which the lipid particle becomes fusogenic.
  • Other methods which can be used to control the rate at which the lipid particle becomes fusogenic will become apparent to those of skill in the art upon reading this disclosure. Also, by controlling the composition and
  • concentration of the lipid conjugate one can control the lipid particle size.
  • the nucleic acid-lipid compositions of this disclosure may be administered by various routes, for example, to effect systemic delivery via intravenous, parenteral, intraperitoneal, or topical routes.
  • a siRNA may be delivered intracellularly, for example, in cells of a target tissue such as lung or liver, or in inflamed tissues.
  • this disclosure provides a method for delivery of siRNA in vivo.
  • a nucleic acid-lipid composition may be administered intravenously, subcutaneously, or intraperitoneally to a subject.
  • the disclosure provides methods for in vivo delivery of interfering RNA to the lung of a mammalian subject.
  • this disclosure provides a method of treating a disease or disorder in a mammalian subject.
  • a therapeutically effective amount of a composition of this disclosure containing a nucleic, a cationic lipid, an amphiphile, a phospholipid, cholesterol, and a PEG-linked cholesterol may be administered to a subject having a disease or disorder associated with expression or overexpression of a gene that can be reduced, decreased, downregulated, or silenced by the composition.
  • compositions and methods of the disclosure may be administered to subjects by a variety of mucosal administration modes, including by oral, rectal, vaginal, intranasal, intrapulmonary, or transdermal or dermal delivery, or by topical delivery to the eyes, ears, skin, or other mucosal surfaces.
  • the mucosal tissue layer includes an epithelial cell layer.
  • the epithelial cell can be pulmonary, tracheal, bronchial, alveolar, nasal, buccal, epidermal, or gastrointestinal.
  • Compositions of this disclosure can be administered using conventional actuators such as mechanical spray devices, as well as pressurized, electrically activated, or other types of actuators.
  • compositions of this disclosure may be administered in an aqueous solution as a nasal or pulmonary spray and may be dispensed in spray form by a variety of methods known to those skilled in the art.
  • Pulmonary delivery of a composition of this disclosure is achieved by administering the composition in the form of drops, particles, or spray, which can be, for example, aerosolized, atomized, or nebulized.
  • Particles of the composition, spray, or aerosol can be in either a liquid or a solid form.
  • Preferred systems for dispensing liquids as a nasal spray are disclosed in U.S. Pat. No. 4,511,069.
  • Such formulations may be conveniently prepared by dissolving compositions according to the present disclosure in water to produce an aqueous solution, and rendering said solution sterile.
  • the formulations may be presented in multi-dose containers, for example in the sealed dispensing system disclosed in U.S. Pat. No. 4,511,069.
  • Other suitable nasal spray delivery systems have been described in TRANSDERMAL SYSTEMIC MEDICATION, Y. W. Chien ed., Elsevier Publishers, New York, 1985; and in U.S. Pat. No. 4,778,810.
  • Additional aerosol delivery forms may include, e.g. , compressed air-Jet-, ultrasonic-, and piezoelectric nebulizers, which deliver the biologically active agent dissolved or suspended in a pharmaceutical solvent, e.g., water, ethanol, or mixtures thereof.
  • Nasal and pulmonary spray solutions of the present disclosure typically comprise the drug or drug to be delivered, optionally formulated with a surface active agent, such as a nonionic surfactant (e.g., polysorbate-80), and one or more buffers.
  • a surface active agent such as a nonionic surfactant (e.g., polysorbate-80)
  • the nasal spray solution further comprises a propellant.
  • the pH of the nasal spray solution may be from pH 6.8 to 7.2.
  • pharmaceutical solvents employed can also be a slightly acidic aqueous buffer of pH 4-6.
  • Other components may be added to enhance or maintain chemical stability, including preservatives, surfactants, dispersants, or gases.
  • this disclosure is a pharmaceutical product which includes a solution containing a composition of this disclosure and an actuator for a pulmonary, mucosal, or intranasal spray or aerosol.
  • a dosage form of the composition of this disclosure can be liquid, in the form of droplets or an emulsion, or in the form of an aerosol.
  • a dosage form of the composition of this disclosure can be solid, which can be reconstituted in a liquid prior to administration.
  • the solid can be administered as a powder.
  • the solid can be in the form of a capsule, tablet, or gel.
  • the biologically active agent can be combined with various pharmaceutically acceptable additives, as well as a base or carrier for dispersion of the active agent(s).
  • additives include pH control agents such as arginine, sodium hydroxide, glycine, hydrochloric acid, citric acid, and mixtures thereof.
  • Other additives include local anesthetics (e.g., benzyl alcohol), isotonizing agents (e.g. , sodium chloride, mannitol, sorbitol), adsorption inhibitors (e.g., Tween 80), solubility enhancing agents (e.g., cyclodextrins and derivatives thereof), stabilizers (e.g., serum albumin), and reducing agents (e.g., glutathione).
  • local anesthetics e.g., benzyl alcohol
  • isotonizing agents e.g. , sodium chloride, mannitol, sorbitol
  • adsorption inhibitors e.g., Tween 80
  • solubility enhancing agents e.g., cyclodextrins and derivatives thereof
  • the tonicity of the formulation is typically adjusted to a value at which no substantial, irreversible tissue damage will be induced in the mucosa at the site of administration.
  • the tonicity of the solution is adjusted to a value of 1/3 to 3, more typically 1/2 to 2, and most often 3/4 to 1.7.
  • the biologically active agent may be dispersed in a base or vehicle, which may comprise a hydrophilic compound having a capacity to disperse the active agent and any desired additives.
  • the base may be selected from a wide range of suitable carriers, including but not limited to, copolymers of polycarboxylic acids or salts thereof, carboxylic anhydrides (e.g., maleic anhydride) with other monomers (e.g. , methyl(meth)acrylate, acrylic acid, etc.), hydrophilic vinyl polymers such as polyvinyl acetate, polyvinyl alcohol,
  • polyvinylpyrrolidone cellulose derivatives such as hydroxymethylcellulose
  • hydroxypropylcellulose, etc. and natural polymers such as chitosan, collagen, sodium alginate, gelatin, hyaluronic acid, and nontoxic metal salts thereof.
  • a biodegradable polymer is selected as a base or carrier, for example, polylactic acid, poly(lactic acid-gly colic acid) copolymer, polyhydroxybutyric acid, poly(hydroxybutyric acid-gly colic acid) copolymer, and mixtures thereof.
  • synthetic fatty acid esters such as poly glycerin fatty acid esters, sucrose fatty acid esters, etc., can be employed as carriers.
  • Hydrophilic polymers and other carriers can be used alone or in combination, and enhanced structural integrity can be imparted to the carrier by partial crystallization, ionic bonding, crosslinking, and the like.
  • the carrier can be provided in a variety of forms, including fluid or viscous solutions, gels, pastes, powders, microspheres, and films for direct application to the nasal mucosa. The use of a selected carrier in this context may result in promotion of absorption of the biologically active agent.
  • Formulations for mucosal, nasal, or pulmonary delivery may contain a hydrophilic low molecular weight compound as a base or excipient.
  • a hydrophilic low molecular weight compound provides a passage medium through which a water-soluble active agent, such as a physiologically active peptide or protein, may diffuse through the base to the body surface where the active agent is absorbed.
  • the hydrophilic low molecular weight compound optionally absorbs moisture from the mucosa or the administration atmosphere and dissolves the water-soluble active peptide.
  • the molecular weight of the hydrophilic low molecular weight compound is generally not more than 10,000 and preferably not more than 3,000.
  • hydrophilic low molecular weight compounds include polyol compounds, such as oligo-, di- and monosaccarides including sucrose, mannitol, lactose, L-arabinose, D-erythrose, D-ribose, D-xylose, D-mannose, D-galactose, lactulose, cellobiose, gentibiose, glycerin, polyethylene glycol, and mixtures thereof.
  • hydrophilic low molecular weight compounds include N-methylpyrrolidone, alcohols (e.g., oligovinyl alcohol, ethanol, ethylene glycol, propylene glycol, etc.), and mixtures thereof.
  • compositions of this disclosure may alternatively contain as pharmaceutically acceptable carriers substances as required to approximate physiological conditions, such as pH adjusting and buffering agents, tonicity adjusting agents, and wetting agents, for example, sodium acetate, sodium lactate, sodium chloride, potassium chloride, calcium chloride, sorbitan monolaurate, triethanolamine oleate, and mixtures thereof.
  • pharmaceutically acceptable carriers include, for example, pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharin, talcum, cellulose, glucose, sucrose, magnesium carbonate, and the like.
  • the biologically active agent may be administered in a time-release formulation, for example in a composition which includes a slow release polymer.
  • the active agent can be prepared with carriers that will protect against rapid release, for example a controlled release vehicle such as a polymer, microencapsulated delivery system, or bioadhesive gel.
  • Prolonged delivery of the active agent, in various compositions of the disclosure can be brought about by including in the composition agents that delay absorption, for example, aluminum monosterate hydrogels and gelatin.
  • Example 1 Exemplary lipids
  • the first pair of numbers denotes the number of carbons in the ester, including the carbonyl, for Li and L2; the pair of numbers in the parenthesis denotes the number of carbons in each branch of the branched alkyl, Ri or if R2 is also branched, R1/R2 (an asterisk denotes a double bond); the number of carbons in R3 is given; and the last line denotes the substitution for R4 and R5.
  • the ATX number is given for reference herein. Calculated LogD values (c-LogD) and calculated pKa (c-pKa) values are given, as well as measured pKa in parenthesis
  • c-LogD and c-pKa values are generated by ACD Labs Structure Designer vl2.0. Bioactivity is percentage in vivo Factor VII knockdown at a dose of 0.03 mg/kg, unless otherwise designated.
  • FIG. 1 shows the synthetic pathway of ATX-0043 that is described further as follows.
  • reaction mass was quenched with saturated NH4CI solution (20 ml) and then EtOAc (20 mL) was added. Organic layer was separated and the aqueous layer was washed with EtOAc (2 x20 mL). Combined organic layer was concentrated and the resulting crude was subjected to column chromatography. Progress of the reaction was monitored by TLC (60% EtOAc/Hex; Rf: 0.5; PMA charring).
  • FIG. 2 shows the synthetic pathway of ATX-0057 that is described further as follows.
  • Quantity produced 80 g (crude; required compound and alcohol).
  • the first purification was done using silica gel (60-120 mesh) 22 g of crude compound was adsorbed on 60 g of silica gel and poured onto 500 g of silica gel taken in the column. Compound was eluted at 35% EtOAc/hexane.
  • the second purification was done using neutral alumina with HPLC grade solvents. Crude compound, 7.5 g, was adsorbed on 18 g of neutral alumina and the resulting was poured onto 130 g of neutral alumina taken in the column. Compound was eluted at 10% EtOAc/hexane. Yield, 29%; confirmed by 3 ⁇ 4 NMR, HPLC, and Mass.
  • FIG. 3 shows the synthetic pathway of ATX-0058 that is described further as follows.
  • ATX-0058 Step 2 [0234] To a solution of 28 g hexyl magnesium bromide (1 eq.) in THF (100 ml), stirred at 0°C under nitrogen atmosphere, was added 36.8 g N-methoxy-N-methyloctanamide (1.3 eq.) in 200 ml THF and the resulting reaction mixture was stirred at room temperature for 5 hours.
  • ATX-0058 Step 7 [0251] To a solution of 20 g 4-bromo butyric acid (1 eq.) dissolved in DCM (150 ml), cooled to 0°C was added 1.5 eq. EDC.HC1, 3 eq. Et 3 N, and 0.1 eq. DMAP sequentially with 10 minutes interval. To this resulting solution 0.7 eq. (Z)-non-2-en-l-ol was added, by dissolving in 100 ml DCM, using a funnel, and stirred at room temperature for 24 hours under nitrogen atmosphere.
  • a first purification was done using silica gel (60-120 mesh). 5.0 g of crude compound was adsorbed on 9 g of silica gel and poured onto 90 g of silica gel taken in the column. Compound was eluted at 35% EtOAc/hexane. A second purification was done using neutral alumina with HPLC grade solvents. 1.5 g of crude compound was adsorbed on 4 g of neutral alumina and the resulting was poured onto 40 g of neutral alumina taken in the column. Compound was eluted at 10% EtOAc/hexane. Quantity produced, 1.2 g; yield, 21%; confirmed by 3 ⁇ 4 NMR; HPLC; Mass.
  • FIG. 4 shows the synthetic pathway of ATX-0081 that is described further as follows.
  • Quantity produced 8.5 g (crude; required compound and alcohol).
  • the first purification was done using silica gel (60-120 mesh) of crude compound was adsorbed on 60 g of silica gel and poured onto 500 g of silica gel taken in the column. Compound was eluted at 35% EtOAc/hexane.
  • the second purification was done using neutral alumina with HPLC grade solvents. Crude compound was adsorbed on 18 g of neutral alumina and the resulting was poured onto 130 g of neutral alumina taken in the column. Compound was eluted at 10% EtOAc/hexane. Quantity produced, 1.5 g; yield, 45%; confirmed by 3 ⁇ 4 NMR, HPLC, and Mass.
  • FIG. 5 shows the synthetic pathway of ATX-0082 that is described further as follows.
  • Quantity produced 8 g (crude; required compound and alcohol).
  • the second purification was done using neutral alumina with HPLC grade solvents. Crude compound was adsorbed on 18 g of neutral alumina and the resulting was poured onto 130 g of neutral alumina taken in the column. Compound was eluted at 10% EtOAc/hexane. Quantity produced, 1.2 g; yield, 43%; confirmed by 3 ⁇ 4 NMR, HPLC, and Mass.
  • FIG. 6 shows the synthetic pathway of ATX-0086 that is described further as follows.
  • Quantity produced 8 g (crude; required compound and alcohol).
  • the first purification was done using silica gel (60-120 mesh) of crude compound was adsorbed on 60 g of silica gel and poured onto 500 g of silica gel taken in the column. Compound was eluted at 35% EtOAc/hexane.
  • the second purification was done using neutral alumina with HPLC grade solvents. Crude compound was adsorbed on 18 g of neutral alumina and the resulting was poured onto 130 g of neutral alumina taken in the column. Compound was eluted at 10% EtOAc/hexane. Quantity produced, 1.2 g; yield, 43%; confirmed by 3 ⁇ 4 NMR, HPLC, and Mass.
  • FIG. 7 shows the synthetic pathway of ATX-0087 that involves nine steps.
  • Quantity produced 15 g (crude; required compound and alcohol).
  • the second purification was done using neutral alumina with HPLC grade solvents. Crude compound was adsorbed on 18 g of neutral alumina and the resulting was poured onto 130 g of neutral alumina taken in the column. Compound was eluted at 10% EtOAc/hexane. Quantity produced, 1.2 g; yield, 43%; confirmed by 3 ⁇ 4 NMR, HPLC, and Mass.
  • FIG. 8 shows the synthetic pathway of ATX-0088 that is described further as follows.
  • FIG. 9 shows the synthetic pathway of ATX-0083 that is described further as follows.
  • ATX-0083 Step 4 [0439] To a solution of 50 g 4-aminobutanoic acid (1 eq.) dissolved in THF, 490 ml 1 N aqueous NaOH solution (1 eq.) was added at 0°C, followed by 140 ml Boc anhydride (1.3 eq.), sequentially using additional funnel, over a period of 15 minutes. The resulting solution was stirred at room temperature for 4 hours.
  • a first purification was done using neutral alumina. Crude compound, dissolved in hexane, was loaded at the top of neutral alumina (700 g loaded in the column). Compound was eluted at 8-10% EtOAc/hexane. A second purification was done using silica gel (100-200 mesh). Compound, dissolved in hexane, was loaded at the top of silica gel (500g loaded in the column). Compound was eluted at 20-25% EtOAc/hexane.
  • FIG. 10 shows the synthetic pathway of ATX-0084 that is described further as follows.
  • a -0084 Step 1 [0465] In a 500 ml single neck round bottom flask, 30 g heptanoic acid (1 eq.) dissolved in of DCM (200 ml) was taken and then added 26.7 g oxalyl chloride (1.5 eq.) slowly at 0°C, stirring under nitrogen atmosphere and then added 1 ml DMF (catalytic). The resulting reaction mixture was stirred at room temperature for 2 hours.
  • a -0084 Step 3 [0474] To a solution of 30 g tridecan-7-one (1 eq.) dissolved in 200 ml MeOH/THF, 8.5 g sodium borohydride (0.5 eq.) was added at 0°C and the resulting solution was stirred at room temperature for 2 hours.
  • a first purification was done using silica gel (100-200 mesh). 4.6 g of crude compound was adsorbed on 10.0 g of silica gel and poured onto 90.0 g of silica gel taken in the column. Compound was eluted at 50% EtOAc/hexane. A second purification was done using neutral alumina with HPLC grade solvents. 2.0 g of crude compound was adsorbed on 6.0 g of neutral alumina and the resulting was poured onto 40.0 g of neutral alumina taken in the column. Compound was eluted at 20% EtOAc/hexane. Quantity produced, 1.2 g; yield, 38 % (300 mg mixture).
  • Example 12 Synthesis of ATX-0061
  • FIG. 11 shows the synthetic pathway of ATX-0061 that is described further as follows
  • FIG. 12 shows the synthetic pathway of ATX-0063 that is described further as follows.
  • ATX-0063 Ste 1
  • reaction was monitored by TLC (60% EtOAc/hexane; Rf: 0.3), starting was absent from reaction product.
  • FIG. 13 shows the synthetic pathway of ATX-0064 that is described further as follows.
  • ATX-0064 Ste 1
  • reaction mass was diluted with saturated NaHCCb solution (80 ml), organic layer was separated, aqueous layer was washed with DCM (40 ml), dried over sodium sulphate and concentrated under reduced pressure.
  • FIG. 14 shows the synthetic pathway of ATX-0081 that is described further as follows. [0602] ATX-0081: Step 1 Ethyl formate
  • reaction mass was quenched with sat. NH4CI solution (500 mL). The organic layer was separated and the aqueous layer was washed with EtOAc (3 x 100 mL). Combined organic layer was dried over anh.Na2S04 and concentrated under reduced pressure.
  • reaction mass was quenched with water (250 mL) and the organic layer was separated.
  • the aqueous layer was washed with DCM (2 x 150 mL).
  • the combined organic layers were concentrated under reduced pressure.
  • the resulting crude was washed with sat.NaHCC solution (150 mL) and then extracted with EtOAc (2 xl50 mL). The organic layer was separated and concentrated under reduced pressure.
  • FIG. 15 shows the synthetic pathway of ATX-0085 that is described further as follows.
  • reaction mass was quenched with sat. NH4CI solution (200 mL). The organic layer was separated and the aqueous layer was washed with ether (2 x 100 mL). The combined organic layers were dried over anh.Na2S04 and concentrated under reduced pressure.
  • FIG. 16 shows the synthetic pathway of ATX-0134 that is described further as follows.
  • Quantity produced 30.0 g (crude).
  • Example 15 Synthesis of ATX-0044 and ATX-0091 to ATX-0133.
  • ATX-0044, ATX-0085, ATX-0111, ATX-0132, ATX-0100, ATX-0117, ATX-0114, ATX-0115, ATX-0101, ATX-0106, ATX-0116, ATX-0122, ATX-0123, ATX- 0124, ATX-0126, ATX-0129, and ATX-0133 were synthesized using the methods of the previous examples.
  • pK a of cationic lipids in LNP or micellar formulations were measured by the procedure of Jayaraman, 2012, Angew. Chem. Int. Ed. , 51 :8529-33, hereby incorporated by reference.
  • Lipid micelles or LNPs are diluted to 1 mM total lipids in universal buffer with a pH range between 3 and 12 in presence of 0.06 mg/mL 6-(/ toluidino)-2-naphthalenesulfonic acid sodium salt (TNS) reagent (Sigma Aldrich), a pH sensitive fluorescence probe.
  • TNS 6-(/ toluidino)-2-naphthalenesulfonic acid sodium salt
  • the anionic TNS molecule fluoresces when associated with the surface of positively charged membranes but is not fluorescent when free in solution, allowing measurement of pKa.
  • the TNS signal is measured on a spectral plate reader.
  • the TNS signal is plotted as function of the pH and analyzed using a non
  • Reagents used in the assay include
  • Reagents are sterile filtered through a 0.2 ⁇ filter.
  • Preparation of UB solution includes adding 5 mL of 1 M HC1 to a 350 mL stock solution.
  • Example 17 In vivo EPO mRNA stability
  • Terminal blood collection was performed via cardiac puncture under 2% isoflurane at 6 hours after formulation injections. Blood was collected into 0.109 M citrate buffer tube and processed by centrifugation at 5000 rpm for 10 minutes. Serum was collected and epo mRNA levels were analyzed. Results are shown at FIG. 17. Results show a substantial improvement over ATX-0002 for ATX-0057, ATX-0081, ATX-0082, ATX-0083, ATX- 0084, ATX-0085, ATX-0086, and ATX-0087.
  • Example 18 In vivo mouse Factor VII silencing and EPO expression
  • Factor VII a blood-clotting factor
  • hepatocytes the cells comprising the liver parenchyma.
  • gene silencing indicates successful delivery to parenchyma, as opposed to delivery to the cells of the reticulo-endothelial system (e.g., Kupffer cells).
  • Factor VII is a secreted protein that can be readily measured in serum, obviating the need to euthanize animals.
  • Silencing at the mRNA level can be readily determined by measuring levels of protein. This is because the protein's short half-life (2-5 hour).
  • Compositions with siRNA directed to Factor VIII were formulated with the lipid, and comparator sample phosphate-buffered saline (PBS).
  • PBS comparator sample phosphate-buffered saline
  • Female C57BL/6 mice (6-8 week old) were used for FVII siRNA knockdown (KD) experiments.
  • Table 1 shows knockdown resulting from lipid nanoparticles comprising the lipids disclosed herein.
  • mice Female Balb/c mice (6-8 week old) were used for evaluation of epo protein expression in vivo following delivery of lipid encapsulated mouse epo mRNA. All formulations were administered intravenously via tail vein injection at a dose of 0.03 and 0.1 mg/kg at a dosing volume of 5mL/kg. Terminal blood collection was performed via cardiac puncture under 2% isoflurane at 6 hours after formulation injections. Blood was collected into 0.109 M citrate buffer tube and processed by centrifugation at 5000 rpm for 10 minutes. Serum was collected and epo protein levels were analyzed by epo ELISA assay (R&D systems).
  • a standard curve was constructed using samples from PBS-injected mice and relative Factor VII expression was determined by comparing treated groups to untreated PBS control. The results showed that epo mRNA is expressed at substantially higher amounts in ATX-0057 nanoparticles than ATX-002 at 0.1 mg/ml (FIG. 19).

Abstract

What is described is a compound of formula (I) consisting of a compound in which R1 is a branched chain alkyl consisting of 10 to 31 carbons; R2 is a linear alkyl, alkenyl, or alkynyl consisting of 2 to 20 carbons, or a branched chain alkyl consisting of 10 to 31 carbons; L1 and L2 are the same or different, each a linear alkane of 1 to 20 carbons or a linear alkene of 2 to 20 carbons; X1 is S or O; R3 is a linear or branched alkylene consisting of 1 to 6 carbons; and R4 and R5 are the same or different, each a hydrogen or a linear or branched alkyl consisting of 1 to 6 carbons; or a pharmaceutically acceptable salt thereof.

Description

IONIZABLE CATIONIC LIPID FOR RNA DELIVERY
CROSS REFERENCE TO RELATED APPLICATIONS
[0001] This application claims priority to continuation-in-part application U.S. Patent Application No. 15/387,067 filed December 21, 2016.
BACKGROUND
[0002] A number of different types of nucleic acids are currently being developed as therapeutics for the treatment of a number of diseases. As these molecules are being developed, there has been developed a need to produce them in a form that is stable and has a long shelf-life and that can be easily incorporated into an anhydrous organic or anhydrous polar aprotic solvent to enable encapsulations of the nucleic acids without the side-reactions that can occur in a polar aqueous solution or nonpolar solvents.
[0003] The description herein relates to novel lipid compositions that facilitate the intracellular delivery of biologically active and therapeutic molecules. The description relates also to pharmaceutical compositions that comprise such lipid compositions, and that are useful to deliver therapeutically effective amounts of biologically active molecules into the cells of patients.
[0004] The delivery of a therapeutic compound to a subject is important for its therapeutic effects and usually it can be impeded by limited ability of the compound to reach targeted cells and tissues. Improvement of such compounds to enter the targeted cells of tissues by a variety of means of delivery is crucial. The description herein relates the novel lipids, in compositions and methods for preparation that facilitate the targeted intracellular delivery of biological active molecules.
[0005] Examples of biologically active molecules for which effective targeting to a patient's tissues is often not achieved include: numerous proteins including immunoglobulin proteins, polynucleotides such as genomic DNA, cDNA, or mRNA antisense
polynucleotides; and many low molecular weight compounds, whether synthetic or naturally occurring, such as the peptide hormones and antibiotics.
[0006] One of the fundamental challenges now facing medical practitioners is that a number of different types of nucleic acids are currently being developed as therapeutics for the treatment of a number of diseases. These nucleic acids include mRNA for gene expression, DNA in gene therapy, plasmids, small interfering nucleic acids (siNA), siRNA, and microRNA (miRNA) for use in RNA interference (RNAi), antisense molecules, ribozymes, antagomirs, and aptamers. As these nucleic acids are being developed, there is a need to produce lipid formulations that are easy to make and can be readily delivered to a target tissue.
SUMMARY
[0007] One aspect of what is described is a compound of formula I
Figure imgf000004_0001
consisting of a compound in which
Ri is a branched chain alkyl consisting of 10 to 31 carbons;
R2 is a linear alkyl, alkenyl, or alkynyl consisting of 2 to 20 carbons, or a branched chain alkyl consisting of 10 to 31 carbons;
Li and L2 are the same or different, each a linear alkane of 1 to 20 carbons or a linear alkene of 2 to 20 carbons;
Xi is S or O;
R3 is a linear or branched alkylene consisting of 1 to 6 carbons; and
R4 and R5 are the same or different, each a hydrogen or a linear or branched alkyl consisting of 1 to 6 carbons; or
a pharmaceutically acceptable salt thereof.
[0008] Another aspect of what is described is a compound of formula I
Figure imgf000005_0001
wherein
Ri is a branched, noncyclic alkyl or alkenyl of 8, 9, 10, 11, 12, 13, 14, 16, 17, 18, 19,
20, 21, or 22 carbons;
Li is linear alkane of 1 to 15 carbons;
R21S a linear alkyl or alkenyl of 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15 carbons or a branched, noncyclic alkyl or alkenyl of 8, 9, 10, 11, 12, 13, 14, 16, 17, 18, 19, 20,
21, or 22 carbons;
L2 is a linear alkane of 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15 carbons;
X is O or S;
R3 is a linear alkane of 1, 2, 3, 4, 5, or 6 carbons; and
R4 and R5 are the same or different, each a linear or branched, noncyclic alkyl of 1, 2, 3, 4, 5, or 6 carbons;
or a pharmaceutically acceptable salt or solvate thereof.
[0009] In one embodiment, Ri has 8, 9, 10, 11, 12, 13, 14, 16, or 17 carbons.
[0010] In another embodiment, Ri comprises two identical alkyl or alkenyl groups.
[0011] In another embodiment, Ri comprises an alkenyl group.
[0012] In another embodiment, R2 is an alkenyl.
[0013] In another embodiment, R2 is a branched, noncyclic alkyl.
[0014] In another embodiment, L2 has 4, 5, 6, or 7 carbons.
[0015] Preferably, the compound is selected from the group consisting of compounds of formulas ATX-0082, ATX-0085, ATX-0083, ATX-0121, ATX-0091, ATX- 0102, ATX-0098, ATX-0092, ATX-0084, ATX-0095, ATX-0125, ATX-0094, ATX-0109, ATX-0110, ATX-0118, ATX-0108, ATX-0107, ATX-0093, ATX-0097, and ATX-0096
Figure imgf000006_0001
Figure imgf000007_0001
Figure imgf000008_0001
-6-
Figure imgf000009_0001
Figure imgf000010_0001
wherein
Ri is a branched, noncyclic alkyl or alkenyl of 12, 13, 14, 16, 17, 18, 19, 20, 21, or 22 carbons;
Li is a linear alkane of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 carbons;
R21S linear alkyl or alkenyl of 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15 carbons or a
branched, noncyclic alkyl of 10, 11, 12, 13, 14, 16, 17, 18, 19, 20, 21, or 22 carbons;
L2 is a linear alkane of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15 carbons
X is O or S;
R3 is linear alkane of 1, 2, 3, 4, 5, or 6 carbons;
R4 and R5 are the same or different, each a linear or branched, noncyclic alkyl of 1, 2, 3, 4, 5, or 6 carbons;
a 1 mM solution of the compound has a pKa of 5.6 to 7.0 as measured by
fluorescence of 6-(/ toluidino)-2-naphthalenesulfonic; and
the compound has a c-LogD value is between 10 and 14;
or a pharmaceutically acceptable salt or solvate thereof.
[0017] In one embodiment, Ri has 12, 13, 14, 16, or 17 carbons.
[0018] In another embodiment, Ri comprises two identical alkyl or alkenyl groups.
[0019] In another embodiment, Ri comprises an alkyl group.
[0020] In another embodiment, R2 is an alkenyl.
[0021] In another embodiment, R2 is a branched, noncyclic alkyl.
[0022] In another embodiment, Li and L2 independently have 1, 2, or 3 carbons.
[0023] In another embodiment, Li and L2 both have 3 carbons.
[0024] In another embodiment, R3 is propylene or butylene.
[0025] In another embodiment, the compound's c-LogD value is at least 11 and its measured pKa more basic than 6. [0026] In another embodiment, the compound is selected from the group consisting of compounds of formulas ATX-0111, ATX-0132, ATX-0134, ATX-0100, ATX-0117, ATX-0114, ATX-01 15. ATX-0101, ATX-0106. ATX-0116, ATX-0086, ATX-0058, ATX- 0081, ATX-0123, ATX-0122, ATX-0057, ATX-0088, ATX-0087, ATX-0124, ATX-0126, ATX-0129, and ATX-0123.
Figure imgf000011_0001
Figure imgf000012_0001
Figure imgf000013_0001
- 11 -
Figure imgf000014_0001
- 12-
Figure imgf000015_0001
- 13 -
Figure imgf000016_0001
[0027] Another aspect of what is describe is a compound of formula I
Figure imgf000016_0002
wherein
Ri is a branched, noncyclic alkyl of 12, 13, 14, 16, 17, 18, 19, 20, 21 , or 22 carbons; Li is a linear alkane of 1 , 2, or 3carbons;
R2 is linear alkenyl of 8, 9, 10, 11 , or 12 carbons or a branched, noncyclic alkyl of 12,
13, 14, 16, or 17 carbons;
L2 is a linear alkane of 1 , 2, or 3 carbons
X is O or S;
R3 is linear alkane of 2 or 3 carbons;
R4 and R5 are the same or different, each a linear or branched, noncyclic alkyl of 1 or 2 carbons; or a pharmaceutically acceptable salt or solvate thereof.
[0028] In another embodiment, cationic lipids described herein are in a
pharmaceutical composition. The pharmaceutical composition preferably comprises a lipid nanoparticle comprising a nucleic acid, preferably a RNA polynucleotide. The lipid nanoparticle preferably increases the lifetime of RNA in the circulation. In another embodiment, upon administration of the pharmaceutical composition, the lipid nanoparticle therein delivers the nucleic acid to cells in the body. Preferably, the nucleic acid has an activity of suppressing the expression of a target gene. Alternatively, the nucleic acid has an activity of increasing production of a protein it encodes upon expression in cells of the body.
[0029] What is also described herein is a method for introducing a nucleic acid into a cell of a mammal by using any of the compositions, above. The cell may be in a liver, lung, kidney, brain, blood, spleen, or bone. The composition preferably is administered intravenously, subcutaneously, intraperitoneally, or intrathecally. Preferably, the
compositions described herein are used in a method for treating cancer or inflammatory disease. The disease may be one selected from the group consisting of immune disorder, cancer, renal disease, fibrotic disease, genetic abnormality, inflammation, and cardiovascular disorder.
BRIEF DESCRIPTION OF THE DRAWINGS
[0030] FIG. 1 shows the synthetic pathway of ATX-0043 from hexanoate (SM 1), 4-aminobutanoic acid (SM 2), and 4-bromobutyric acid (SM 3). Intermediates (Ints) 1-8 and reactions are described in Example 2.
[0031] FIG. 2 shows the synthetic pathway of ATX-0057 from octanoate (SM 1), 4- aminobutanoic acid (SM 2), and 4-bromobutyric acid (SM 3). Ints 1-8 and reactions are described in Example 3.
[0032] FIG. 3 shows the synthetic pathway of ATX-0058 from SM 1, SM 2 and SM3, which are the same as in FIG. 2. Ints 1-7 and reactions are described in Example 4.
[0033] FIG. 4 shows the synthetic pathway of ATX-0081 from SM 1, SM 2 and SM3, which are the same as in FIG. 2. Ints 1-8 and reactions are described in Example 5.
[0034] FIG. 5 shows the synthetic pathway of ATX-0082 from SM 1, SM 2 and SM3, which are the same as in FIG. 2. Ints 1-7 and reactions are described in Example 6.
[0035] FIG. 6 shows the synthetic pathway of ATX-0086 from SM 1, SM 2 and SM3, which are the same as in FIG. 2. Ints 1-8 and reactions are described in Example 7. [0036] FIG. 7 shows the synthetic pathway of ATX-0087 from SM 1, SM 2 and SM3, which are the same as in FIG. 2. Ints 1-8 and reactions are described in Example 8.
[0037] FIG. 8 shows the synthetic pathway of ATX-0088 from SM 1, SM 2 and SM3, which are the same as in FIG. 2. Ints 1-8 and reactions are described in Example 9.
[0038] FIG. 9 shows the synthetic pathway of ATX-0083 from SM 1, SM 2 and SM3, which are the same as in FIG. 2. Ints 1-8 and reactions are described in Example 10.
[0039] FIG. 10 shows the synthetic pathway of ATX-0084 from SM 1, SM 2 and SM3, which are the same as in FIG. 2. Ints 1-8 and reactions are described in Example 11.
[0040] FIG. 11 shows the synthetic pathway of ATX-0061 from SM 1 and SM 2, which are the same as in FIG. 1. Ints 1-5 and reactions are described in Example 12.
[0041] FIG. 12 shows the synthetic pathway of ATX-0063 from SM 1 and SM 2, which are the same as in FIG. 1. Ints 1-5 and reactions are described in Example 13.
[0042] FIG. 13 shows the synthetic pathway of ATX-0064 from SM 1 and SM 2, which are the same as in FIG. 1. Ints 1-5 and reactions are described in Example 14.
[0043] FIG. 14 shows the synthetic pathway of ATX-0081. Ints 1-6 and reactions are described in Example 15.
[0044] FIG. 15 shows the synthetic pathway of ATX-0085 from SM 1, SM 2 and SM3, which are the same as in FIG. 2. Ints 1-11 and reactions are described in Example 16.
[0045] FIG. 16 shows the synthetic pathway of ATX-0134. Ints 1-6 and reactions are described in Example 17.
[0046] FIG. 17 shows the EPO mRNA levels (ng/ml) following injection of 0.03 mg/kg and 0.1 mg/kg mRNA in nanoparticles comprising ATX-002, ATX-0057, ATX-0081. ATX-0082, ATX-0083, ATX-0084, ATX-0085, ATX-0086. or ATX-0087 cationic lipid into mice.
[0047] FIG. 18 shows the anti-Factor VII knockdown activity of liposomes with ATX-0057 and ATX-0058 vs. the activity of ATX-002 and control (PBS alone).
[0048] FIG. 19 shows the anti-EPO knockdown activity of liposomes with ATX- 0057 vs. the activity of ATX-002.
DETAILED DESCRIPTION OF ILLUSTRATIVE EMBODIMENTS
Definitions
[0049] "At least one" means one or more (e.g. , 1-3, 1-2, or 1). [0050] "Composition" means a product comprising the specified ingredients in the specified amounts, as well as any product that results, directly or indirectly, from
combination of the specified ingredients in the specified amounts.
[0051] "In combination with" means the administration of a compound of formula I with other medicaments in the methods of treatment of this invention, meaning that the compounds of formula I and the other medicaments are administered sequentially or concurrently in separate dosage forms, or are administered concurrently in the same dosage form.
[0052] "Mammal" means a human or other mammal, or means a human being.
[0053] "Patient" means both human and other mammals, preferably human.
[0054] "Alkyl" means a saturated or unsaturated, straight or branched, hydrocarbon chain. In various embodiments, the alkyl group has 1-18 carbons, i.e. is a Ci-Cls group, or is a C1-C12 group, a C1-C6 group, or a C1-C4 group. Independently, in various embodiments, the alkyl group has zero branches (i.e. , is a straight chain), one branch, two branches, or more than two branches. "Alkenyl" is an unsaturated alkyl that may have one double bond, two double bonds, or more than two double bonds. "Alkynyl" is an unsaturated alkyl that may have one triple bond, two triple bonds, or more than two triple bonds. Alkyl chains may be optionally substituted with 1 substituent (i.e. , the alkyl group is mono-substituted), or 1-2 substituents, or 1-3 substituents, or 1-4 substituents, etc. The substituents may be selected from the group consisting of hydroxy, amino, alkylamino, boronyl, carboxy, nitro, cyano, and the like. When the alkyl group incorporates one or more heteroatoms, the alkyl group is referred to herein as a heteroalkyl group. When the substituents on an alkyl group are hydrocarbons, then the resulting group is simply referred to as a substituted alkyl. In various aspects, the alkyl group including substituents has less than 25, 24, 23, 22, 21, 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, or 7 carbons.
[0055] "Lower alkyl" means a group having one to six carbons in the chain which chain may be straight or branched. Non-limiting examples of suitable alkyl groups include methyl, ethyl, n-propyl, isopropyl, n-butyl, t-butyl, n-pentyl, and hexyl.
[0056] "Alkoxy" means an alkyl-O-group wherein alkyl is as defined above. Non- limiting examples of alkoxy groups include: methoxy, ethoxy, n-propoxy, isopropoxy, n- butoxy and heptoxy. The bond to the parent moiety is through the ether oxygen. [0057] "Alkoxyalkyl" means an alkoxy-alkyl-group in which the alkoxy and alkyl are as previously described. Preferred alkoxyalkyl comprise a lower alkyl group. The bond to the parent moiety is through the alkyl.
[0058] "Alkylaryl" means an alkyl-aryl-group in which the alkyl and aryl are as previously described. Preferred alkylaryls comprise a lower alkyl group. The bond to the parent moiety is through the aryl.
[0059] "Aminoalkyl" means an NH2-alkyl-group, wherein alkyl is as defined above, bound to the parent moiety through the alkyl group.
[0060] "Carboxyalkyl" means an HOOC-alkyl-group, wherein alkyl is as defined above, bound to the parent moiety through the alkyl group.
[0061] "Commercially available chemicals" and the chemicals used in the Examples set forth herein may be obtained from standard commercial sources, where such sources include, for example, Acros Organics (Pittsburgh, Pa.), Sigma-Adrich Chemical (Milwaukee, Wis.), Avocado Research (Lancashire, U.K.), Bionet (Cornwall, U.K.), Boron Molecular (Research Triangle Park, N.C.), Combi-Blocks (San Diego, Calif), Eastman Organic Chemicals, Eastman Kodak Company (Rochester, N.Y.), Fisher Scientific Co. (Pittsburgh, Pa.), Frontier Scientific (Logan, Utah), ICN Biomedicals, Inc. (Costa Mesa, Calif), Lancaster Synthesis (Windham, N.H.), Maybridge Chemical Co. (Cornwall, U.K.), Pierce Chemical Co. (Rockford, 111.), Riedel de Haen (Hannover, Germany), Spectrum Quality Product, Inc. (New Brunswick, N.J.), TCI America (Portland, Oreg.), and Wako Chemicals USA, Inc.
(Richmond, Va.).
[0062] "Compounds described in the chemical literature" may be identified through reference books and databases directed to chemical compounds and chemical reactions, as known to one of ordinary skill in the art. Suitable reference books and treatise that detail the synthesis of reactants useful in the preparation of compounds disclosed herein, or provide references to articles that describe the preparation of compounds disclosed herein, include for example, "Synthetic Organic Chemistry", John Wiley and Sons, Inc. New York; S. R.
Sandler et al, "Organic Functional Group Preparations," 2nd Ed., Academic Press, New York, 1983; H. O. House, "Modem Synthetic Reactions," 2nd Ed., W. A. Benjamin, Inc. Menlo Park, Calif, 1972; T. L. Glichrist, "Heterocyclic Chemistry," 2nd Ed. John Wiley and Sons, New York, 1992; J. March, "Advanced Organic Chemistry: reactions, Mechanisms and Structure," 5th Ed., Wiley Interscience, New York, 2001; Specific and analogous reactants may also be identified through the indices of known chemicals prepared by the Chemical Abstract Service of the American Chemical Society, which are available in most public and university libraries, as well as through online databases (the American Chemical Society, Washington, D.C. may be contacted for more details). Chemicals that are known but not commercially available in catalogs may be prepared by custom chemical synthesis houses, where many of the standard chemical supply houses (such as those listed above) provide custom synthesis services.
[0063] "Halo" means fluoro, chloro, bromo, or iodo groups. Preferred are fluoro, chloro or bromo, and more preferred are fluoro and chloro.
[0064] "Halogen" means fluorine, chlorine, bromine, or iodine. Preferred are fluorine, chlorine and bromine.
[0065] "Heteroalkyl" means a saturated or unsaturated, straight or branched, chain containing carbon and at least one heteroatom. The heteroalkyl group may, in various embodiments, have one heteroatom, or 1-2 heteroatoms, or 1-3 heteroatoms, or 1-4 heteroatoms. In one aspect the heteroalkyl chain contains from 1 to 18 (i.e., 1-18) member atoms (carbon and heteroatoms), and in various embodiments contain 1-12, or 1-6, or 1-4 member atoms. Independently, in various embodiments, the heteroalkyl group has zero branches (i.e., is a straight chain), one branch, two branches, or more than two branches. Independently, in one embodiment, the hetereoalkyl group is saturated. In another embodiment, the heteroalkyl group is unsaturated. In various embodiments, the unsaturated heterolkyl may have one double bond, two double bonds, more than two double bonds, and/or one triple bond, two triple bonds, or more than two triple bonds. Heteroalkyl chains may be substituted or unsubstituted. In one embodiment, the heteroalkyl chain is unsubstituted. In another embodiment, the heteroalkyl chain is substituted. A substituted heteroalkyl chain may have 1 substituent (i.e. , by monosubstituted), or may have, e.g. , 1-2 substituents, or 1-3 substituents, or 1-4 substituents. Exemplary heteroalkyl substituents include esters (— C(O)— O— R) and carbonyls (— C(O)— ).
[0066] "Hydroxyalkyl" means an HO-alkyl-group, in which alkyl is previously defined. Preferred hydroxyalkyls contain lower alkyl. Non-limiting examples of suitable hydroxyalkyl groups include hydroxymethyl and 2 -hydroxy ethyl.
[0067] "Hydrate" means a solvate wherein the solvent molecule is H2O.
[0068] "Lipid" means an organic compound that comprises an ester of fatty acid and is characterized by being insoluble in water, but soluble in many organic solvents. Lipids are usually divided into at least three classes: (1) "simple lipids," which include fats and oils as well as waxes; (2) "compound lipids," which include phospholipids, glycolipids, cationic lipids, non-cationic lipids, neutral lipids, and anionic lipids, all described in more detail herein; and (3) "derived lipids" such as steroids.
[0069] "Lipid particle" means a lipid formulation that can be used to deliver a therapeutic nucleic acid (e.g. , mRNA) to a target site of interest (e.g., cell, tissue, organ, and the like). In preferred embodiments, the lipid particle is a nucleic acid-lipid particle, which is typically formed from a cationic lipid, a non-cationic lipid (e.g. , a phospholipid), a conjugated lipid that prevents aggregation of the particle (e.g. , a PEG-lipid), and optionally cholesterol. Typically, the therapeutic nucleic acid (e.g., mRNA) may be encapsulated in the lipid portion of the particle, thereby protecting it from enzymatic degradation.
[0070] Lipid particles typically have a mean diameter of from 30 nm to 150 nm, from 40 nm to 150 nm, from 50 nm to 150 nm, from 60 nm to 130 nm, from 70 nm to 110 nm, from 70 nm to 100 nm, from 80 nm to 100 nm, from 90 nm to 100 nm, from 70 to 90 nm, from 80 nm to 90 nm, from 70 nm to 80 nm, or 30 nm, 35 nm, 40 nm, 45 nm, 50 nm, 55 nm, 60 nm, 65 nm, 70 nm, 75 nm, 80 nm, 85 nm, 90 nm, 95 nm, 100 nm, 105 nm, 110 nm, 1 15 nm, 120 nm, 125 nm, 130 nm, 135 nm, 140 nm, 145 nm, or 150 nm, and are substantially non-toxic. In addition, nucleic acids, when present in the lipid particles of the present invention, are resistant in aqueous solution to degradation with a nuclease.
[0071] "Lipid encapsulated" means a lipid particle that provides a therapeutic nucleic acid such as an mRNA with full encapsulation, partial encapsulation, or both. In a preferred embodiment, the nucleic acid (e.g. , mRNA) is fully encapsulated in the lipid particle.
[0072] "Lipid conjugate" means a conjugated lipid that inhibits aggregation of lipid particles. 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, cationic PEG lipids, polyoxazoline (POZ)-lipid conjugates, polyamide oligomers, and mixtures thereof. PEG 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. , non-ester-containing linker moieties and ester-containing linker moieties. In certain preferred embodiments, non-ester-containing linker moieties, such as amides or carbamates, are used. [0073] "Amphipathic lipid" means the material in which the hydrophobic portion of the lipid material orients into a hydrophobic phase, while the hydrophilic portion orients toward the aqueous phase. Hydrophilic characteristics derive from the presence of polar or charged groups such as carbohydrates, phosphate, carboxylic, sulfate, amino, sulfhydryl, nitro, hydroxyl, and other like groups. Hydrophobicity can be conferred by the inclusion of apolar groups that include, but are not limited to, long-chain saturated and unsaturated aliphatic hydrocarbon groups and such groups substituted by one or more aromatic, cycloaliphatic, or heterocyclic group(s). Examples of amphipathic compounds include, but are not limited to, phospholipids, aminolipids, and sphingolipids.
[0074] Representative examples of phospholipids include, but are not limited to, phosphatidylcholine, phosphatidylethanolamine, phosphatidylserine, phosphatidylinositol, phosphatidic acid, palmitoyloleoyl phosphatidylcholine, lysophosphatidylcholine, lysophosphatidylethanolamine, dipalmitoylphosphatidylcholine, dioleoylphosphatidylcholine, distearoylphosphatidylcholine, and dilinoleoylphosphatidylcholine. Other compounds lacking in phosphorus, such as sphingolipid, glycosphingolipid families, diacylglycerols, and β-acyloxyacids, are also within the group designated as amphipathic lipids. Additionally, the amphipathic lipids described above can be mixed with other lipids including triglycerides and sterols.
[0075] "Neutral lipid" means a lipid species that exist either in an uncharged or neutral zwitterionic form at a selected pH. At physiological pH, such lipids include, for example, diacylphosphatidylcholine, diacylphosphatidylethanolamine, ceramide,
sphingomyelin, cephalin, cholesterol, cerebrosides, and diacylglycerols.
[0076] "Non-cationic lipid" means an amphipathic lipid or a neutral lipid or anionic lipid, and is described in more detail below.
[0077] "Anionic lipid" means a lipid that is negatively charged at 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.
[0078] "Hydrophobic lipids" means compounds having apolar groups 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.
[0079] "Cationic lipid" and "amino lipid" are used interchangeably mean those lipids and salts thereof having one, two, three, or more fatty acid or fatty alkyl chains and a pH-titratable amino head group (e.g. , an alkylamino or dialkylamino head group). The cationic lipid is typically protonated (i.e. , positively charged) at a pH below the pKa of the cationic lipid and is substantially neutral at a pH above the pKa. The cationic lipids of the invention may also be termed titratable cationic lipids. In some embodiments, the cationic lipids comprise: a protonatable tertiary amine (e.g. , pH-titratable) head group; Ci8 alkyl chains, wherein each alkyl chain independently has 0 to 3 (e.g., 0, 1, 2, or 3) double bonds; and ether, ester, or ketal linkages between the head group and alkyl chains. Such cationic lipids include, but are not limited to, DSDMA, DODMA, DLinDMA, DLenDMA, γ- DLenDMA, DLin-K-DMA, DLin-K-C2-DMA (also known as DLin-C2K-DMA, XTC2, and C2K), DLin-K-C3 -DM A, DLin-K-C4-DMA, DLen-C2K-DMA, y-DLen-C2K-DMA, DLin- M-C2-DMA (also known as MC2), DLin-M-C3 -DMA (also known as MC3) and (DLin-MP- DMA)(also known as 1-Bl 1).
[0080] "Substituted" means substitution with specified groups other than hydrogen, or with one or more groups, moieties, or radicals which can be the same or different, with each, for example, being independently selected.
[0081] "Antisense nucleic acid", means a non-enzymatic nucleic acid molecule that binds to target RNA by means of RNA-RNA or RNA-DNA or RNA-PNA (protein nucleic acid; Egholm et al., 1993 Nature 365, 566) interactions and alters the activity of the target RNA (for a review, see Stein and Cheng, 1993 Science 261, 1004 and Woolf et al, U.S. Pat. No. 5,849,902). Typically, antisense molecules are complementary to a target sequence along a single contiguous sequence of the antisense molecule. However, in certain embodiments, an antisense molecule can bind to substrate such that the substrate molecule forms a loop, and/or an antisense molecule can bind such that the antisense molecule forms a loop. Thus, the antisense molecule can be complementary to two (or even more) noncontiguous substrate sequences or two (or even more) non-contiguous sequence portions of an antisense molecule can be complementary to a target sequence or both. In addition, antisense DNA can be used to target RNA by means of DNA-RNA interactions, thereby activating RNase H, which digests the target RNA in the duplex. The antisense oligonucleotides can comprise one or more RNAse H activating region, which is capable of activating RNAse H cleavage of a target RNA. Antisense DNA can be synthesized chemically or expressed via the use of a single stranded DNA expression vector or equivalent thereof. "Antisense RNA" is an RNA strand having a sequence complementary to a target gene mRNA, that can induce RNAi by binding to the target gene mRNA. "Antisense RNA" is an RNA strand having a sequence complementary to a target gene mRNA, and thought to induce RNAi by binding to the target gene mRNA. "Sense RNA" has a sequence complementary to the antisense RNA, and annealed to its complementary antisense RNA to form iNA. These antisense and sense RNAs have been conventionally synthesized with an RNA synthesizer.
[0082] "Nucleic acid" means deoxyribonucleotides or ribonucleotides and polymers thereof in single- or double-stranded form. The term encompasses nucleic acids containing known nucleotide analogs or modified backbone residues or linkages, which are synthetic, naturally occurring, and non-naturally occurring, which have similar binding properties as the reference nucleic acid, and which are metabolized in a manner similar to the reference nucleotides. Examples of such analogs include, without limitation, phosphorothioates, phosphoramidates, methyl phosphonates, chiral-methyl phosphonates, 2'-0-methyl ribonucleotides, peptide-nucleic acids (PNAs).
[0083] "RNA" means a molecule comprising at least one ribonucleotide residue. By "ribonucleotide" is meant a nucleotide with a hydroxyl group at the 2' position of a β-D-ribo- furanose moiety. The terms include double-stranded RNA, single-stranded RNA, isolated RNA such as partially purified RNA, essentially pure RNA, synthetic RNA, recombinantly produced RNA, as well as altered RNA that differs from naturally occurring RNA by the addition, deletion, substitution, and/or alteration of one or more nucleotides. Such alterations can include addition of non-nucleotide material, such as to the end(s) of an interfering RNA or internally, for example at one or more nucleotides of the RNA. Nucleotides in the RNA molecules of the instant invention can also comprise non-standard nucleotides, such as non- naturally occurring nucleotides or chemically synthesized nucleotides or deoxynucleotides. These altered RNAs can be referred to as analogs or analogs of naturally-occurring RNA. As used herein, the terms "ribonucleic acid" and "RNA" refer to a molecule containing at least one ribonucleotide residue, including siRNA, antisense RNA, single stranded RNA, microRNA, mRNA, noncoding RNA, and multivalent RNA. A ribonucleotide is a nucleotide with a hydroxyl group at the 2' position of a B-D-ribo-furanose moiety. These terms include double-stranded RNA, single-stranded RNA, isolated RNA such as partially purified RNA, essentially pure RNA, synthetic RNA, recombinantly produced RNA, as well as modified and altered RNA that differs from naturally occurring RNA by the addition, deletion, substitution, modification, and/or alteration of one or more nucleotides. Alterations of an RNA can include addition of non-nucleotide material, such as to the end(s) of an interfering RNA or internally, for example at one or more nucleotides of an RNA nucleotides in an RNA molecule include non-standard nucleotides, such as non-naturally occurring nucleotides or chemically synthesized nucleotides or deoxynucleotides. These altered RNAs can be referred to as analogs.
[0084] "Nucleotides" means natural bases (standard) and modified bases well known in the art. Such bases are generally located at the Γ position of a nucleotide sugar moiety. Nucleotides generally comprise a base, sugar, and a phosphate group. The nucleotides can be unmodified or modified at the sugar, phosphate, and/or base moiety, (also referred to interchangeably as nucleotide analogs, modified nucleotides, non-natural nucleotides, non-standard nucleotides and other; see, for example, Usman and McSwiggen, supra; Eckstein, et al, International PCT Publication No. WO 92/07065; Usman, et al, International PCT Publication No. WO 93/15187; Uhlman & Peyman, supra, all are hereby incorporated by reference herein). There are several examples of modified nucleic acid bases known in the art as summarized by Limbach, et al, Nucleic Acids Res. 22:2183, 1994. Some of the non-limiting examples of base modifications that can be introduced into nucleic acid molecules include: inosine, purine, pyridin-4-one, pyridin-2-one, phenyl, pseudouracil, 2,4,6- trimethoxy benzene, 3-methyl uracil, dihydrouridine, naphthyl, aminophenyl, 5- alkylcytidines (e.g. , 5-methylcytidine), 5-alkyluridines (e.g. , ribothymidine), 5-halouridine (e.g., 5-bromouridine) or 6-azapyrimi dines or 6-alkylpyrimi dines (e.g., 6-methyluridine), propyne, and others (Burgin, et al, Biochemistry 35: 14090, 1996; Uhlman & Peyman, supra). By "modified bases" in this aspect is meant nucleotide bases other than adenine, guanine, cytosine, and uracil at Γ position or their equivalents.
[0085] "Complementary nucleotide bases" means a pair of nucleotide bases that form hydrogen bonds with each other. Adenine (A) pairs with thymine (T) or with uracil (U) in RNA, and guanine (G) pairs with cytosine (C). Complementary segments or strands of nucleic acid that hybridize (i.e. join by hydrogen bonding) with each other. By
"complementary" it is meant that a nucleic acid can form hydrogen bond(s) with another nucleic acid sequence either by traditional Watson-Crick or by other non-traditional modes of binding.
[0086] "MicroRNAs" (miRNA) means single-stranded RNA molecules of 21-23 nucleotides in length, which regulate gene expression miRNAs are encoded by genes that are transcribed from DNA but not translated into protein (non-coding RNA); instead they are processed from primary transcripts known as pri-miRNA to short stem-loop structures called pre-miRNA and finally to functional miRNA. Mature miRNA molecules are partially complementary to one or more messenger RNA (mRNA) molecules, and their main function is to downregulate gene expression
[0087] "Small interfering RNA (siRNA)" and "short interfering RNA" and
"silencing RNA" mean a class of double-stranded RNA molecules, 16-40 nucleotides in length, that play a variety of roles in biology. Most notably, siRNA is involved in the RNA interference (RNAi) pathway, where it interferes with the expression of a specific gene. In addition to their role in the RNAi pathway, siRNAs also act in RNAi-related pathways, e.g. , as an antiviral mechanism or in shaping the chromatin structure of a genome; the complexity of these pathways is only now being elucidated.
[0088] "RNAi" means an RNA-dependent gene silencing process that is controlled by the RNA-induced silencing complex (RISC) and is initiated by short double-stranded RNA molecules in a cell, where they interact with the catalytic RISC component argonaute. When the double-stranded RNA or RNA-like iNA or siRNA is exogenous (coming from infection by a virus with an RNA genome or from transfected iNA or siRNA), the RNA or iNA is imported directly into the cytoplasm and cleaved to short fragments by the enzyme dicer. The initiating dsRNA can also be endogenous (originating in the cell), as in pre- microRNAs expressed from RNA-coding genes in the genome. The primary transcripts from such genes are first processed to form the characteristic stem-loop structure of pre-miRNA in the nucleus, then exported to the cytoplasm to be cleaved by dicer. Thus, the two dsRNA pathways, exogenous and endogenous, converge at the RISC complex. The active components of an RNA-induced silencing complex (RISC) are endonucleases called argonaute proteins, which cleave the target mRNA strand complementary to their bound siRNA or iNA. As the fragments produced by dicer are double-stranded, they could each in theory produce a functional siRNA or iNA. However, only one of the two strands, which is known as the guide strand, binds the argonaute protein and directs gene silencing. The other anti-guide strand or passenger strand is degraded during RISC activation. [0089] Compound of Formula I
[0090] Reference to a compound of formula I herein is understood to include reference to salts thereof, unless otherwise indicated. The term "salt(s)", as employed herein, denotes acidic salts formed with inorganic and/or organic acids, as well as basic salts formed with inorganic and/or organic bases. In addition, when compound of formula I contain both a basic moiety, such as, but not limited to, a pyridine or imidazole, and an acidic moiety, such as, but not limited to, a carboxylic acid, zwitterions ("inner salts") may be formed and are included within the term "salt(s)" as used herein. The salts can be pharmaceutically acceptable (i. e. , non-toxic, physiologically acceptable) salts, although other salts are also useful. Salts of a compound of formula I may be formed, for example, by reacting a compound of formula I with an amount of acid or base, such as an equivalent amount, in a medium such as one in which the salt precipitates or in an aqueous medium followed by lyophilization.
[0091] Exemplary acid addition salts include acetates, adipates, alginates, ascorbates, aspartates, benzoates, benzenesulfonates, bisulfates, borates, butyrates, citrates, camphorates, camphorsulfonates, cyclopentanepropionates, digluconates, dodecylsulfates, ethanesulfonates, fumarates, glucoheptanoates, glycerophosphates, hemisulfates, heptanoates, hexanoates, hydrochlorides, hydrobromides, hydroiodides, 2 -hydroxy ethanesulfonates, lactates, maleates, methanesulfonates, 2-napthalenesulfonates, nicotinates, nitrates, oxalates, pectinates, persulfates, 3-phenylpropionates, phosphates, picrates, pivalates, propionates, salicylates, succinates, sulfates, sulfonates (such as those mentioned herein), tartarates, thiocyanates, toluenesulfonates (also known as tosylates) undecanoates, and the like.
Additionally, acids which are generally considered suitable for the formation of
pharmaceutically useful salts from basic pharmaceutical compound are discussed, for example, by S. Berge et al, J. Pharmaceutical Sciences (1977) 66(1)1-19; P. Gould,
International J. Pharmaceutics (1986) 33 201-217; Anderson et al, The Practice of Medicinal Chemistry (1996), Academic Press, New York; and in The Orange Book (Food & Drug Administration, Washington, D.C. on their website). These disclosures are incorporated by reference herein.
[0092] Exemplary basic salts include ammonium salts, alkali metal salts such as sodium, lithium, and potassium salts, alkaline earth metal salts such as calcium and magnesium salts, salts with organic bases (for example, organic amines) such as benzathines, dicyclohexylamines, hydrabamines (formed with N,N-bis(dehydroabietyl)ethylenediamine), N-methyl-D-glucamines, N-methyl-D-glucamides, t-butyl amines, and salts with amino acids such as arginine or lysine. Basic nitrogen-containing groups may be quartemized with agents such as lower alkyl halides (e.g. , methyl, ethyl, propyl, and butyl chlorides, bromides, and iodides), dialkyl sulfates (e.g., dimethyl, diethyl, dibutyl, and diamyl sulfates), long chain halides (e.g., decyl, lauryl, myristyl, and stearyl chlorides, bromides, and iodides), arylalkyl halides (e.g., benzyl and phenethyl bromides), and others.
[0093] All such acid and base salts are intended to be pharmaceutically acceptable salts within the scope of the disclosure and all acid and base salts are considered equivalent to the free forms of the corresponding compound of formula I for purposes of the disclosure.
[0094] Compound of formula I can exist in unsolvated and solvated forms, including hydrated forms. In general, the solvated forms, with pharmaceutically acceptable solvents such as water, ethanol, and the like, are equivalent to the unsolvated forms for the purposes of this disclosure.
[0095] Compound of formula I and salts, solvates thereof, may exist in their tautomeric form (for example, as an amide or imino ether). All such tautomeric forms are contemplated herein as part of the present disclosure.
[0096] Also within the scope of the present disclosure are polymorphs of the compound of this disclosure (i.e. , polymorphs of the compound of formula I are within the scope of this disclosure).
[0097] All stereoisomers (for example, geometric isomers, optical isomers, and the like) of the present compound (including those of the salts, solvates, and prodrugs of the compound as well as the salts and solvates of the prodrugs), such as those which may exist due to asymmetric carbons on various substituents, including enantiomeric forms (which may exist even in the absence of asymmetric carbons), rotameric forms, atropisomers, and diastereomeric forms, are contemplated within the scope of this disclosure. Individual stereoisomers of the compound of this disclosure may, for example, be substantially free of other isomers, or may be admixed, for example, as racemates or with all other, or other selected, stereoisomers. The chiral centers of the compound herein can have the S or R configuration as defined by the IUPAC 1974 Recommendations. The use of the terms "salt", "solvate", and the like, is intended to equally apply to the salt and solvate of enantiomers, stereoisomers, rotamers, tautomers, racemates, or prodrugs of the disclosed compound.
[0098] Classes of compounds that can be used as the chemotherapeutic agent (antineoplastic agent) include: alkylating agents, antimetabolites, natural products and their derivatives, hormones and steroids (including synthetic analogs), and synthetics. Examples of compounds within these classes are given below.
Lipid Particles
[0099] A compound of formula I includes a pharmaceutically acceptable salt thereof, in a lipid composition, comprising a nanoparticle or a bilayer of lipid molecules. The lipid bilayer preferably further comprises a neutral lipid or a polymer. The lipid composition preferably comprises a liquid medium. The composition preferably further encapsulates a nucleic acid. The nucleic acid preferably has an activity of suppressing the expression of the target gene by utilizing RNA interference (RNAi). The lipid composition preferably further comprises a nucleic acid and a neutral lipid or a polymer. The lipid composition preferably encapsulates the nucleic acid.
[0100] The description provides lipid particles comprising one or more therapeutic mRNA molecules encapsulated within the lipid particles.
[0101] In some embodiments, the mRNA is fully encapsulated within the lipid portion of the lipid particle such that the mRNA in the lipid particle is resistant in aqueous solution to nuclease degradation. In other embodiments, the lipid particles described herein are substantially non-toxic to mammals such as humans. The lipid particles typically have a mean diameter of from 30 nm to 150 nm, from 40 nm to 150 nm, from 50 nm to 150 nm, from 60 nm to 130 nm, from 70 nm to 110 nm, or from 70 to 90 nm. The lipid particles of the invention also typically have a lipid:RNA ratio (mass/mass ratio) of from 1 : 1 to 100: 1, from 1 : 1 to 50: 1, from 2: 1 to 25: 1, from 3: 1 to 20: 1, from 5: 1 to 15: 1, or from 5: 1 to 10: 1, or from 10: 1 to 14: 1, or from 9: 1 to 20: 1. In one embodiment, the lipid particles have a lipid: RNA ratio (mass/mass ratio) of 12: 1. In another embodiment, the lipid particles have a lipid: mRNA ratio (mass/mass ratio) of 13: 1.
[0102] In preferred embodiments, the lipid particles comprise an mRNA, a cationic lipid (e.g., one or more cationic lipids or salts thereof described herein), a phospholipid, and a conjugated lipid that inhibits aggregation of the particles (e.g. , one or more PEG-lipid conjugates). The lipid particles can also include cholesterol. The lipid particles may comprise at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more mRNA that express one or more polypeptides.
[0103] In the nucleic acid-lipid particles, the mRNA may be fully encapsulated within the lipid portion of the particle, thereby protecting the nucleic acid from nuclease degradation. In preferred embodiments, a lipid particle comprising an mRNA is fully encapsulated within the lipid portion of the particle, thereby protecting the nucleic acid from nuclease degradation. In certain instances, the mRNA in the lipid particle is not substantially degraded after exposure of the particle to a nuclease at 37°C for at least 20, 30, 45, or 60 minutes. In certain other instances, the mRNA in the lipid particle is not substantially degraded after incubation of the particle in serum at 37°C for at least 30, 45, or 60 minutes or at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, or 36 hours. In other embodiments, the mRNA is complexed with the lipid portion of the particle. One of the benefits of the formulations of the present invention is that the nucleic acid-lipid particle compositions are substantially non-toxic to mammals such as humans.
[0104] "Fully encapsulated" means that the nucleic acid (e.g. , mRNA) in the nucleic acid-lipid particle is not significantly degraded after exposure to serum or a nuclease assay that would significantly degrade free RNA. When fully encapsulated, preferably less than 25% of the nucleic acid in the particle is degraded in a treatment that would normally degrade 100% of free nucleic acid, more preferably less than 10%, and most preferably less than 5% of the nucleic acid in the particle is degraded. "Fully encapsulated" also means that the nucleic acid-lipid particles do not rapidly decompose into their component parts upon in vivo administration.
[0105] In the context of nucleic acids, full encapsulation may be determined by performing a membrane-impermeable fluorescent dye exclusion assay, which uses a dye that has enhanced fluorescence when associated with nucleic acid. Encapsulation is determined by adding the dye to a liposomal formulation, measuring the resulting fluorescence, and comparing it to the fluorescence observed upon addition of a small amount of nonionic detergent. Detergent-mediated disruption of the liposomal bilayer releases the encapsulated nucleic acid, allowing it to interact with the membrane-impermeable dye. Nucleic acid encapsulation may be calculated as E = (Io - I)/Io, where/and Io refers to the fluorescence intensities before and after the addition of detergent.
[0106] In other embodiments, the present invention provides a nucleic acid-lipid particle composition comprising a plurality of nucleic acid-lipid particles.
[0107] The lipid particle comprises mRNA that is fully encapsulated within the lipid portion of the particles, such that from 30% to 100%, from 40% to 100%, from 50% to 100%, from 60% to 100%, from 70% to 100%, from 80% to 100%, from 90% to 100%, from 30% to 95%, from 40% to 95%, from 50% to 95%, from 60% to 95%, from 70% to 95%, from 80% to 95%, from 85% to 95%, from 90% to 95%, from 30% to 90%, from 40% to 90%, from 50% to 90%, from 60% to 90%, from 70% to 90%, from 80% to 90%, or at least 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% (or any fraction thereof or range therein) of the particles have the mRNA encapsulated therein.
[0108] Depending on the intended use of the lipid particles, the proportions of the components can be varied and the delivery efficiency of a particular formulation can be measured using assays know in the art.
Cationic lipids
[0109] The description includes synthesis of certain cationic lipid compounds. The compounds are particularly suitable for delivering polynucleotides to cells and tissues as demonstrated in subsequent sections. The lipomacrocycle compound described herein may be used for other purposes as well as, for example, recipients and additives.
[0110] The synthetic methods for the cationic lipid compounds can be synthesized with the skills in the art. Those skilled in the art will recognize other methods to produce these compounds, and also to produce the other compounds of the description.
[0111] The cationic lipid compounds may be combined with an agent to form microparticles, nanoparticles, liposomes, or micelles. The agent to be delivered by the particles, liposomes, or micelles may be in the form of a gas, liquid, or solid, and the agent may be a polynucleotide, protein, peptide, or small molecule. The lipomacrocycle compounds may be combined with other cationic lipid compounds, polymers (synthetic or natural), surfactants, cholesterol, carbohydrates, proteins, or lipids, to form the particles. These particles may then optionally be combined with a pharmaceutical excipient to form a pharmaceutical composition.
[0112] The present description provides novel cationic lipid compounds and drug delivery systems based on the use of such cationic lipid compounds. The system may be used in the pharmaceutical/drug delivery arts to deliver polynucleotides, proteins, small molecules, peptides, antigen, or drugs, to a patient, tissue, organ, or cell. These novel compounds may also be used as materials for coating, additives, excipients, materials, or bioengineering.
[0113] The cationic lipid compounds of the present description provide for several different uses in the drug delivery art. The amine-containing portion of the cationic lipid compounds may be used to complex polynucleotides, thereby enhancing the delivery of polynucleotide and preventing their degradation. The cationic lipid compounds may also be used in the formation of picoparticles, nanoparticles, microparticles, liposomes, and micelles containing the agent to be delivered. Preferably, the cationic lipid compounds are biocompatible and biodegradable, and the formed particles are also biodegradable and biocompatible and may be used to provide controlled, sustained release of the agent to be delivered. These and their corresponding particles may also be responsive to pH changes given that these are protonated at lower pH. They may also act as proton sponges in the delivery of an agent to a cell to cause endosome lysis.
[0114] In certain embodiments, the cationic lipid compounds are relatively non- cytotoxic. The cationic lipid compounds may be biocompatible and biodegradable. The cationic lipid may have a measured pKa (in the formulation milieu) in the range of approximately 5.5 to approximately 7.5, more preferably between approximately 6.0 and approximately 7.0. It may be designed to have a desired pKa between approximately 3.0 and approximately 9.0, or between approximately 5.0 and approximately 8.0. The cationic lipid compounds described herein are particularly attractive for drug delivery for several reasons: they contain amino groups for interacting with DNA, RNA, other polynucleotides, and other negatively charged agents, for buffering the pH, for causing endo-osmolysis, for protecting the agent to be delivered, they can be synthesized from commercially available starting materials; and/or they are pH responsive and can be engineered with a desired pKa.
Neutral Helper Lipids
[0115] Non-limiting examples of non-cationic lipids include phospholipids such as lecithin, phosphatidylethanolamine, lysolecithin, lysophosphatidylethanolamine,
phosphatidylserine, phosphatidylinositol, sphingomyelin, egg sphingomyelin (ESM), cephalin, cardiolipin, phosphatidic acid, cerebrosides, dicetylphosphate,
distearoylphosphatidylcholine (DSPC), dioleoylphosphatidylcholine (DOPC),
dipalmitoylphosphatidylcholine (DPPC), dioleoylphosphatidylglycerol (DOPG),
dipalmitoylphosphatidylglycerol (DPPG), dioleoylphosphatidylethanolamine (DOPE), palmitoyloleoyl-phosphatidylcholine (POPC), palmitoyloleoyl-phosphatidylethanolamine (POPE), palmitoyloleyol-phosphatidylglycerol (POPG), dioleoylphosphatidylethanolamine 4- (N-maleimidomethyl)-cyclohexane- 1 - carboxylate (DOPE-mal), dipalmitoyl- phosphatidylethanolamine (DPPE), dimyristoyl- phosphatidylethanolamine (DMPE), distearoyl-phosphatidylethanolamine (DSPE), monomethyl-phosphatidylethanolamine, dimethyl-phosphatidylethanolamine, dielaidoyl- phosphatidylethanolamine (DEPE), stearoyloleoyl-phosphatidylethanolamine (SOPE), lysophosphatidylcholine, dilinoleoylphosphatidylcholine, and mixtures thereof. 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 oleoyl.
[0116] Additional examples of non-cationic lipids include sterols such as cholesterol and derivatives thereof. Non-limiting examples of cholesterol derivatives include polar analogues such as 5a-cholestanol, 5a-coprostanol, cholesteryl-(2'-hydroxy)-ethyl ether, cholesteryl-(4'- hydroxy)-butyl ether, and 6-ketocholestanol; non-polar analogues such as 5a- cholestane, cholestenone, 5a-cholestanone, 5a-cholestanone, and cholesteryl decanoate; and mixtures thereof. In preferred embodiments, the cholesterol derivative is a polar analogue such as cholesteryl-(4'-hydroxy)-butyl ether.
[0117] In some embodiments, the non-cationic lipid present in lipid particles comprises or consists of a mixture of one or more phospholipids and cholesterol or a derivative thereof. In other embodiments, the non-cationic lipid present in the lipid particles comprises or consists of one or more phospholipids, e.g., a cholesterol-free lipid particle formulation. In yet other embodiments, the non-cationic lipid present in the lipid particles comprises or consists of cholesterol or a derivative thereof, e.g. , a phospholipid-free lipid particle formulation.
[0118] Other examples of non-cationic lipids include nonphosphorous containing lipids such as, e.g. , stearylamine, dodecylamine, hexadecylamine, acetyl palmitate, glycerol ricinoleate, hexadecyl stearate, isopropyl myristate, amphoteric acrylic polymers, triethanolamine-lauryl sulfate, alkyl-aryl sulfate polyethyloxylated fatty acid amides, dioctadecyldimethyl ammonium bromide, ceramide, and sphingomyelin.
[0119] In some embodiments, the non-cationic lipid comprises from 10 mol % to 60 mol %, from 20 mol % to 55 mol %, from 20 mol % to 45 mol %, 20 mol % to 40 mol %, from 25 mol % to 50 mol %, from 25 mol % to 45 mol %, from 30 mol % to 50 mol %, from 30 mol % to 45 mol %, from 30 mol % to 40 mol %, from 35 mol % to 45 mol %, from 37 mol % to 42 mol %, or 35 mol %, 36 mol %, 37 mol %, 38 mol %, 39 mol %, 40 mol %, 41 mol %, 42 mol %, 43 mol %, 44 mol %, or 45 mol % (or any fraction thereof or range therein) of the total lipid present in the particle.
[0120] In embodiments where the lipid particles contain a mixture of phospholipid and cholesterol or a cholesterol derivative, the mixture may comprise up to 40 mol %, 45 mol %, 50 mol %, 55 mol %, or 60 mol % of the total lipid present in the particle. [0121] In some embodiments, the phospholipid component in the mixture may comprise from 2 mol % to 20 mol %, from 2 mol % to 15 mol %, from 2 mol % to 12 mol %, from 4 mol % to 15 mol %, or from 4 mol % to 10 mol % (or any fraction thereof or range therein) of the total lipid present in the particle. In certain preferred embodiments, the phospholipid component in the mixture comprises from 5 mol % to 10 mol %, from 5 mol % to 9 mol %, from 5 mol % to 8 mol %, from 6 mol % to 9 mol %, from 6 mol % to 8 mol %, or 5 mol %, 6 mol %, 7 mol %, 8 mol %, 9 mol %, or 10 mol % (or any fraction thereof or range therein) of the total lipid present in the particle.
[0122] In other embodiments, the cholesterol component in the mixture may comprise from 25 mol % to 45 mol %, from 25 mol % to 40 mol %, from 30 mol % to 45 mol %, from 30 mol % to 40 mol %, from 27 mol % to 37 mol %, from 25 mol % to 30 mol %, or from 35 mol % to 40 mol % (or any fraction thereof or range therein) of the total lipid present in the particle. In certain preferred embodiments, the cholesterol component in the mixture comprises from 25 mol % to 35 mol %, from 27 mol % to 35 mol %, from 29 mol % to 35 mol %, from 30 mol % to 35 mol %, from 30 mol % to 34 mol %, from 31 mol % to 33 mol %, or 30 mol %, 31 mol %, 32 mol %, 33 mol %, 34 mol %, or 35 mol % (or any fraction thereof or range therein) of the total lipid present in the particle.
[0123] In embodiments where the lipid particles are phospholipid-free, the cholesterol or derivative thereof may comprise up to 25 mol %, 30 mol %, 35 mol %, 40 mol %, 45 mol %, 50 mol %, 55 mol %, or 60 mol % of the total lipid present in the particle.
[0124] In some embodiments, the cholesterol or derivative thereof in the phospholipid-free lipid particle formulation may comprise from 25 mol % to 45 mol %, from 25 mol % to 40 mol %, from 30 mol % to 45 mol %, from 30 mol % to 40 mol %, from 31 mol % to 39 mol %, from 32 mol % to 38 mol %, from 33 mol % to 37 mol %, from 35 mol % to 45 mol %, from 30 mol % to 35 mol %, from 35 mol % to 40 mol %, or 30 mol %, 31 mol %, 32 mol %, 33 mol %, 34 mol %, 35 mol %, 36 mol %, 37 mol %, 38 mol %, 39 mol %, or 40 mol % (or any fraction thereof or range therein) of the total lipid present in the particle.
[0125] In other embodiments, the non-cationic lipid comprises from 5 mol % to 90 mol %, from 10 mol % to 85 mol %, from 20 mol % to 80 mol %, 10 mol % (e.g. , phospholipid only), or 60 mol % (e.g. , phospholipid and cholesterol or derivative thereof) (or any fraction thereof or range therein) of the total lipid present in the particle. [0126] The percentage of non-cationic lipid present in the lipid particles is a target amount, and that the actual amount of non-cationic lipid present in the formulation may vary, for example, by ± 5 mol %.
[0127] A composition containing a cationic lipid compound may be 30-70% cationic lipid compound, 0-60 % cholesterol, 0-30% phospholipid and 1-10% polyethylene glycol (PEG). Preferably, the composition is 30-40% cationic lipid compound, 40- 50% cholesterol, and 10-20% PEG. In other preferred embodiments, the composition is 50-75% cationic lipid compound, 20-40% cholesterol, and 5-10% phospholipid, and 1-10% PEG. The composition may contain 60-70% cationic lipid compound, 25-35% cholesterol, and 5- 10% PEG. The composition may contain up to 90% cationic lipid compound and 2-15% helper lipid.
[0128] The formulation may be a lipid particle formulation, for example containing 8-30% compound, 5-30% helper lipid , and 0-20% cholesterol; 4-25% cationic lipid, 4-25% helper lipid, 2- 25% cholesterol, 10- 35% cholesterol-PEG, and 5% cholesterol-amine; or 2- 30% cationic lipid, 2-30% helper lipid, 1- 15% cholesterol, 2- 35% cholesterol-PEG, and 1- 20% cholesterol-amine; or up to 90% cationic lipid and 2-10% helper lipids, or even 100% cationic lipid.
Lipid Conjugates
[0129] In addition to cationic, the lipid particles described herein may further comprise a lipid conjugate. The conjugated lipid is useful in that it prevents the aggregation of particles. Suitable conjugated lipids include, but are not limited to, PEG-lipid conjugates, cationic-polymer- lipid conjugates, and mixtures thereof.
[0130] In a preferred embodiment, the lipid conjugate is a PEG-lipid. Examples of PEG- lipids include, but are not limited to, PEG coupled to dialkyloxypropyls (PEG-DAA), PEG coupled to diacylglycerol (PEG-DAG), PEG coupled to phospholipids such as phosphatidylethanolamine (PEG-PE), PEG conjugated to ceramides, PEG conjugated to cholesterol or a derivative thereof, and mixtures thereof.
[0131] PEG is a linear, water-soluble polymer of ethylene PEG repeating units with two terminal hydroxyl groups. PEGs are classified by their molecular weights; and include the following: monomethoxypoly ethylene glycol (MePEG-OH), monomethoxypoly ethylene glycol- succinate (MePEG-S), monomethoxypoly ethylene glycol-succinimidyl succinate (MePEG-S- NHS), monomethoxypoly ethylene glycol-amine (MePEG-NEh),
monomethoxypoly ethylene glycol-tresylate (MePEG-TRES), monomethoxypoly ethylene glycol-imidazolyl-carbonyl (MePEG-IM), as well as such compounds containing a terminal hydroxyl group instead of a terminal methoxy group (e.g., HO-PEG-S, HO-PEG-S-NHS,
Figure imgf000037_0001
[0132] The PEG moiety of the PEG-lipid conjugates described herein may comprise an average molecular weight ranging from 550 daltons to 10,000 daltons. In certain instances, the PEG moiety has an average molecular weight of from 750 daltons to 5,000 daltons (e.g. , from 1,000 daltons to 5,000 daltons, from 1,500 daltons to 3,000 daltons, from 750 daltons to 3,000 daltons, from 750 daltons to 2,000 daltons). In preferred embodiments, the PEG moiety has an average molecular weight of 2,000 daltons or 750 daltons.
[0133] In certain instances, the PEG can be optionally substituted by an alkyl, alkoxy, acyl, or aryl group. The PEG 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 to a lipid can be used including, e.g., non-ester-containing linker moieties and ester-containing linker moieties. In a preferred embodiment, the linker moiety is a non-ester-containing linker moiety. Suitable non-ester-containing linker moieties include, but are not limited to, amido (- C(O)NH-), amino (-NR-), carbonyl (-C(O)-), carbamate (-NHC(O)O-), urea (-NHC(O)NH-), disulphide (-S-S-), ether (-0-), succinyl (- (0)CCH2CH2C(0)-), succinamidyl (- NHC(0)CH2CH2C(0)NH-), ether, disulphide, as well as combinations thereof (such as a linker containing both a carbamate linker moiety and an amido linker moiety). In a preferred embodiment, a carbamate linker is used to couple the PEG to the lipid.
[0134] In other embodiments, an ester-containing linker moiety is used to couple the PEG to the lipid. Suitable ester-containing linker moieties include, e.g., carbonate (- OC(O)O-), succinoyl, phosphate esters (-O-(O)POH-O-), sulfonate esters, and combinations thereof.
[0135] Phosphatidylethanolamines having a variety of acyl chain groups of varying chain lengths and degrees of saturation can be conjugated to PEG to form the lipid conjugate. Such phosphatidylethanolamines are commercially available, or can be isolated or synthesized using conventional techniques known to those of skill in the art.
Phosphatidylethanolamines containing saturated or unsaturated fatty acids with carbon chain lengths in the range of C10 to C20 are preferred. Phosphatidylethanolamines with mono- or di- unsaturated fatty acids and mixtures of saturated and unsaturated fatty acids can also be used. Suitable phosphatidylethanolamines include, but are not limited to, dimyristoyl- phosphatidylethanolamine (DMPE), dipalmitoyl-phosphatidylethanolamine (DPPE), dioleoyl-phosphatidylethanolamine (DOPE), and distearoyl-phosphatidylethanolamine (DSPE).
[0136] The term "diacylglycerol" or "DAG" includes a compound having 2 fatty acyl chains, R1 and R2, both of which have independently between 2 and 30 carbons bonded to the 1- and 2-position of glycerol by ester linkages. The acyl groups can be saturated or have varying degrees of unsaturation. Suitable acyl groups include, but are not limited to, lauroyl (C12), myristoyl (C14), palmitoyl (Ci6), stearoyl (Cis), and icosoyl (C20). In preferred embodiments, R1 and R2 are the same, i.e. , R1 and R2 are both myristoyl (i.e. , dimyristoyl), R1 and R2 are both stearoyl (i.e. , distearoyl).
[0137] The term "dialkyloxy propyl" or "DAA" includes a compound having 2 alkyl chains, Ri and R2 , both of which have independently between 2 and 30 carbons. The alkyl groups can be saturated or have varying degrees of unsaturation.
[0138] Preferably, the PEG-DAA conjugate is a PEG-didecyloxypropyl (C10) conjugate, a PEG-dilauryloxy propyl (C12) conjugate, a PEG-dimyristyloxypropyl (C14) conjugate, a PEG-dipalmityloxy propyl (Ci6) conjugate, or a PEG-distearyloxy propyl (Cis) conjugate. In these embodiments, the PEG preferably has an average molecular weight of 750 or 2,000 daltons. In particular embodiments, the terminal hydroxyl group of the PEG is substituted with a methyl group.
[0139] In addition to the foregoing, other hydrophilic polymers can be used in place of PEG. Examples of suitable polymers that can be used in place of PEG include, but are not limited to, polyvinylpyrrolidone, polymethyloxazoline, polyethyloxazoline,
polyhydroxypropyl methacrylamide, polymethacrylamide and polydimethylacrylamide, polylactic acid, poly gly colic acid, and derivatized celluloses such as hydroxymethylcellulose or hydroxy ethylcellulose.
[0140] In some embodiments, the lipid conjugate (e.g. , PEG-lipid) comprises from 0.1 mol % to 2 mol %, from 0.5 mol % to 2 mol %, from 1 mol % to 2 mol %, from 0.6 mol % to 1.9 mol %, from 0.7 mol % to 1.8 mol %, from 0.8 mol % to 1.7 mol %, from 0.9 mol % to 1.6 mol %, from 0.9 mol % to 1.8 mol %, from 1 mol % to 1.8 mol %, from 1 mol % to 1.7 mol %, from 1.2 mol % to 1.8 mol %, from 1.2 mol % to 1.7 mol %, from 1.3 mol % to 1.6 mol %, or from 1.4 mol % to 1.5 mol % (or any fraction thereof or range therein) of the total lipid present in the particle. In other embodiments, the lipid conjugate (e.g., PEG-lipid) comprises from 0 mol % to 20 mol %, from 0.5 mol % to 20 mol %, from 2 mol % to 20 mol %, from 1.5 mol % to 18 mol %, from 2 mol % to 15 mol %, from 4 mol % to 15 mol %, from 2 mol % to 12 mol %, from 5 mol % to 12 mol %, or 2 mol % (or any fraction thereof or range therein) of the total lipid present in the particle.
[0141] In further embodiments, the lipid conjugate (e.g. , PEG-lipid) comprises from 4 mol % to 10 mol %, from 5 mol % to 10 mol %, from 5 mol % to 9 mol %, from 5 mol % to 8 mol %, from 6 mol % to 9 mol %, from 6 mol % to 8 mol %, or 5 mol %, 6 mol %, 7 mol %, 8 mol %, 9 mol %, or 10 mol % (or any fraction thereof or range therein) of the total lipid present in the particle.
[0142] The percentage of lipid conjugate (e.g., PEG-lipid) present in the lipid particles of the invention is a target amount, and the actual amount of lipid conjugate present in the formulation may vary, for example, by ± 2 mol %. One of ordinary skill in the art will appreciate that the concentration of the lipid conjugate can be varied depending on the lipid conjugate employed and the rate at which the lipid particle is to become fusogenic.
[0143] By controlling the composition and concentration of the lipid conjugate, 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 becomes fusogenic. In addition, other variables including, e.g. , pH, temperature, or ionic strength, can be used to vary and/or control the rate at which the lipid particle becomes fusogenic. Other methods which can be used to control the rate at which the lipid particle becomes fusogenic will become apparent to those of skill in the art upon reading this disclosure. Also, by controlling the composition and
concentration of the lipid conjugate, one can control the lipid particle size.
[0144] Compositions and Formulations for Administration
[0145] The nucleic acid-lipid compositions of this disclosure may be administered by various routes, for example, to effect systemic delivery via intravenous, parenteral, intraperitoneal, or topical routes. In some embodiments, a siRNA may be delivered intracellularly, for example, in cells of a target tissue such as lung or liver, or in inflamed tissues. In some embodiments, this disclosure provides a method for delivery of siRNA in vivo. A nucleic acid-lipid composition may be administered intravenously, subcutaneously, or intraperitoneally to a subject. In some embodiments, the disclosure provides methods for in vivo delivery of interfering RNA to the lung of a mammalian subject.
[0146] In some embodiments, this disclosure provides a method of treating a disease or disorder in a mammalian subject. A therapeutically effective amount of a composition of this disclosure containing a nucleic, a cationic lipid, an amphiphile, a phospholipid, cholesterol, and a PEG-linked cholesterol may be administered to a subject having a disease or disorder associated with expression or overexpression of a gene that can be reduced, decreased, downregulated, or silenced by the composition.
[0147] The compositions and methods of the disclosure may be administered to subjects by a variety of mucosal administration modes, including by oral, rectal, vaginal, intranasal, intrapulmonary, or transdermal or dermal delivery, or by topical delivery to the eyes, ears, skin, or other mucosal surfaces. In some aspects of this disclosure, the mucosal tissue layer includes an epithelial cell layer. The epithelial cell can be pulmonary, tracheal, bronchial, alveolar, nasal, buccal, epidermal, or gastrointestinal. Compositions of this disclosure can be administered using conventional actuators such as mechanical spray devices, as well as pressurized, electrically activated, or other types of actuators.
[0148] Compositions of this disclosure may be administered in an aqueous solution as a nasal or pulmonary spray and may be dispensed in spray form by a variety of methods known to those skilled in the art. Pulmonary delivery of a composition of this disclosure is achieved by administering the composition in the form of drops, particles, or spray, which can be, for example, aerosolized, atomized, or nebulized. Particles of the composition, spray, or aerosol can be in either a liquid or a solid form. Preferred systems for dispensing liquids as a nasal spray are disclosed in U.S. Pat. No. 4,511,069. Such formulations may be conveniently prepared by dissolving compositions according to the present disclosure in water to produce an aqueous solution, and rendering said solution sterile. The formulations may be presented in multi-dose containers, for example in the sealed dispensing system disclosed in U.S. Pat. No. 4,511,069. Other suitable nasal spray delivery systems have been described in TRANSDERMAL SYSTEMIC MEDICATION, Y. W. Chien ed., Elsevier Publishers, New York, 1985; and in U.S. Pat. No. 4,778,810. Additional aerosol delivery forms may include, e.g. , compressed air-Jet-, ultrasonic-, and piezoelectric nebulizers, which deliver the biologically active agent dissolved or suspended in a pharmaceutical solvent, e.g., water, ethanol, or mixtures thereof.
[0149] Nasal and pulmonary spray solutions of the present disclosure typically comprise the drug or drug to be delivered, optionally formulated with a surface active agent, such as a nonionic surfactant (e.g., polysorbate-80), and one or more buffers. In some embodiments of the present disclosure, the nasal spray solution further comprises a propellant. The pH of the nasal spray solution may be from pH 6.8 to 7.2. The
pharmaceutical solvents employed can also be a slightly acidic aqueous buffer of pH 4-6. Other components may be added to enhance or maintain chemical stability, including preservatives, surfactants, dispersants, or gases.
[0150] In some embodiments, this disclosure is a pharmaceutical product which includes a solution containing a composition of this disclosure and an actuator for a pulmonary, mucosal, or intranasal spray or aerosol.
[0151] A dosage form of the composition of this disclosure can be liquid, in the form of droplets or an emulsion, or in the form of an aerosol.
[0152] A dosage form of the composition of this disclosure can be solid, which can be reconstituted in a liquid prior to administration. The solid can be administered as a powder. The solid can be in the form of a capsule, tablet, or gel.
[0153] To formulate compositions for pulmonary delivery within the present disclosure, the biologically active agent can be combined with various pharmaceutically acceptable additives, as well as a base or carrier for dispersion of the active agent(s).
Examples of additives include pH control agents such as arginine, sodium hydroxide, glycine, hydrochloric acid, citric acid, and mixtures thereof. Other additives include local anesthetics (e.g., benzyl alcohol), isotonizing agents (e.g. , sodium chloride, mannitol, sorbitol), adsorption inhibitors (e.g., Tween 80), solubility enhancing agents (e.g., cyclodextrins and derivatives thereof), stabilizers (e.g., serum albumin), and reducing agents (e.g., glutathione). When the composition for mucosal delivery is a liquid, the tonicity of the formulation, as measured with reference to the tonicity of 0.9% (w/v) physiological saline solution taken as unity, is typically adjusted to a value at which no substantial, irreversible tissue damage will be induced in the mucosa at the site of administration. Generally, the tonicity of the solution is adjusted to a value of 1/3 to 3, more typically 1/2 to 2, and most often 3/4 to 1.7.
[0154] The biologically active agent may be dispersed in a base or vehicle, which may comprise a hydrophilic compound having a capacity to disperse the active agent and any desired additives. The base may be selected from a wide range of suitable carriers, including but not limited to, copolymers of polycarboxylic acids or salts thereof, carboxylic anhydrides (e.g., maleic anhydride) with other monomers (e.g. , methyl(meth)acrylate, acrylic acid, etc.), hydrophilic vinyl polymers such as polyvinyl acetate, polyvinyl alcohol,
polyvinylpyrrolidone, cellulose derivatives such as hydroxymethylcellulose,
hydroxypropylcellulose, etc., and natural polymers such as chitosan, collagen, sodium alginate, gelatin, hyaluronic acid, and nontoxic metal salts thereof. Often, a biodegradable polymer is selected as a base or carrier, for example, polylactic acid, poly(lactic acid-gly colic acid) copolymer, polyhydroxybutyric acid, poly(hydroxybutyric acid-gly colic acid) copolymer, and mixtures thereof. Alternatively or additionally, synthetic fatty acid esters such as poly glycerin fatty acid esters, sucrose fatty acid esters, etc., can be employed as carriers. Hydrophilic polymers and other carriers can be used alone or in combination, and enhanced structural integrity can be imparted to the carrier by partial crystallization, ionic bonding, crosslinking, and the like. The carrier can be provided in a variety of forms, including fluid or viscous solutions, gels, pastes, powders, microspheres, and films for direct application to the nasal mucosa. The use of a selected carrier in this context may result in promotion of absorption of the biologically active agent.
[0155] Formulations for mucosal, nasal, or pulmonary delivery may contain a hydrophilic low molecular weight compound as a base or excipient. Such hydrophilic low molecular weight compounds provide a passage medium through which a water-soluble active agent, such as a physiologically active peptide or protein, may diffuse through the base to the body surface where the active agent is absorbed. The hydrophilic low molecular weight compound optionally absorbs moisture from the mucosa or the administration atmosphere and dissolves the water-soluble active peptide. The molecular weight of the hydrophilic low molecular weight compound is generally not more than 10,000 and preferably not more than 3,000. Examples of hydrophilic low molecular weight compounds include polyol compounds, such as oligo-, di- and monosaccarides including sucrose, mannitol, lactose, L-arabinose, D-erythrose, D-ribose, D-xylose, D-mannose, D-galactose, lactulose, cellobiose, gentibiose, glycerin, polyethylene glycol, and mixtures thereof. Further examples of hydrophilic low molecular weight compounds include N-methylpyrrolidone, alcohols (e.g., oligovinyl alcohol, ethanol, ethylene glycol, propylene glycol, etc.), and mixtures thereof.
[0156] The compositions of this disclosure may alternatively contain as pharmaceutically acceptable carriers substances as required to approximate physiological conditions, such as pH adjusting and buffering agents, tonicity adjusting agents, and wetting agents, for example, sodium acetate, sodium lactate, sodium chloride, potassium chloride, calcium chloride, sorbitan monolaurate, triethanolamine oleate, and mixtures thereof. For solid compositions, conventional nontoxic pharmaceutically acceptable carriers can be used which include, for example, pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharin, talcum, cellulose, glucose, sucrose, magnesium carbonate, and the like. [0157] In certain embodiments of the disclosure, the biologically active agent may be administered in a time-release formulation, for example in a composition which includes a slow release polymer. The active agent can be prepared with carriers that will protect against rapid release, for example a controlled release vehicle such as a polymer, microencapsulated delivery system, or bioadhesive gel. Prolonged delivery of the active agent, in various compositions of the disclosure can be brought about by including in the composition agents that delay absorption, for example, aluminum monosterate hydrogels and gelatin.
[0158] While this disclosure has been described in relation to certain embodiments, and many details have been set forth for purposes of illustration, it will be apparent to those skilled in the art that this disclosure includes additional embodiments, and that some of the details described herein may be varied considerably without departing from this disclosure. This disclosure includes such additional embodiments, modifications, and equivalents. In particular, this disclosure includes any combination of the features, terms, or elements of the various illustrative components and examples.
Examples
Example 1: Exemplary lipids
[0159] Exemplary compounds of formula I are provided in Table 1. The structure of the Compound is shown in the first column. The designation of the compound is give according to formula I
Figure imgf000043_0001
The first pair of numbers denotes the number of carbons in the ester, including the carbonyl, for Li and L2; the pair of numbers in the parenthesis denotes the number of carbons in each branch of the branched alkyl, Ri or if R2 is also branched, R1/R2 (an asterisk denotes a double bond); the number of carbons in R3 is given; and the last line denotes the substitution for R4 and R5. The ATX number is given for reference herein. Calculated LogD values (c-LogD) and calculated pKa (c-pKa) values are given, as well as measured pKa in parenthesis
(measured in the formulation milieu). The c-LogD and c-pKa values are generated by ACD Labs Structure Designer vl2.0. Bioactivity is percentage in vivo Factor VII knockdown at a dose of 0.03 mg/kg, unless otherwise designated.
Table1
Figure imgf000044_0001
Figure imgf000045_0001
Figure imgf000046_0001
Figure imgf000047_0001
Figure imgf000048_0001
Figure imgf000049_0001
Figure imgf000050_0001
Figure imgf000051_0001
Figure imgf000052_0001
Figure imgf000053_0001
Figure imgf000054_0001
Figure imgf000055_0001
Figure imgf000056_0001
[0160] Example 2: Synthesis of ATX-0043
[0161] FIG. 1 shows the synthetic pathway of ATX-0043 that is described further as follows.
[0162] ATX-0043: Step 1
Figure imgf000056_0002
[0163] In a 500 mL single neck round bottom flask, 25 g hexanoic acid (SM 1; 1 eq.) dissolved in dichloromethane (DCM; 200 mL) was taken and then added 27.6 ml oxalyl chloride (1.5 eq.) slowly at 0°C, stirring under nitrogen atmosphere and then added 0.5 ml dimethylformamide (DMF; catalytic). The resulting reaction mixture was stirred at room temperature for 2 hours.
[0164] In a separate 1 liter two neck found bottom flask, to 31.4 g Ν,Ο- dimethylhydroxylamine hydrochloride (1.5 eq.) in DCM (200 ml), was added 89.8 ml triethylamine (Et3N, 3 eq.) using additional funnel, stirred at 0°C. To this resulting solution, the above acid chloride, after concentration under reduced pressure, was added under nitrogen atmosphere by dissolving in DCM (100 ml), dropwise using addition funnel for 20 minutes. The resulting reaction solution was stirred at room temperature for 3 hours under nitrogen atmosphere. [0165] Progress of the reaction was monitored by thin layer chromatography (TLC) (20% ethylacetate (EtOAc)/hexane; Rf: 0.5). Reaction mass was diluted with water (300 ml). Organic layer was separated and the aqueous layer was washed with DCM (3 x 100ml). Combined organic layer was concentrated under reduced pressure.
[0166] Crude compound was subjected to column chromatography using (60-120 silica gel; 10% EtOAc/hexane). Quantity produced, 20.0 g; yield, 58%.
[0167] ATX-0043: Step 2
Figure imgf000057_0001
[0168] To a solution of 33 g pentyl magnesium bromide (1.5 eq.) in tetrahydrofuran (THF; 100 ml), taken in a 500 ml two neck round bottom flask, stirred at 0°C under nitrogen atmosphere, was added 20 g N-methoxy-N-methyl hexanamide (1 eq.) solution (dissolved in 200 ml of THF) and the resulting reaction mixture was stirred at room temperature for 4 hours.
[0169] Progress of the reaction was monitored by TLC (10% EtOAc in hexane; Rf: 0.7). Reaction mass was quenched with saturated NH4CI solution (150 ml) and then EtOAc (300 ml) was added. Organic layer was separated and the aqueous layer was washed with EtOAc (2 x 100 ml). Combined organic layers were concentrated under reduced pressure.
[0170] Crude compound was subjected to column chromatography using (60-120 mesh silica gel; 2% EtOAc/hexane). Quantity produced, 15.0 g; yield, 66%.
[0171] AT -0043: Step 3
Figure imgf000057_0002
[0172] To a solution of 15 g undecan-6-one (1 eq.) dissolved in 25 ml methanol (MeOH) in 150 ml THF, 4.9 g sodium borohydride (1.5 eq.) was added at 0°C and the resulting solution was stirred at room temperature for 2 hour.
[0173] Progress of the reaction was monitored by TLC (10% EtOAc in hexane; Rf: 0.5). Reaction mass was quenched with saturated NH4CI solution (100 ml). Solvent was removed under reduced pressure and the resulting crude was portioned between EtOAc (150 ml) and water (150 ml). Organic layer was separated and the aqueous layer was washed with EtOAc (3 x 100 ml). Combined organic layers were concentrated under reduced pressure to get white solid. Quantity produced, 14.0 g; yield, 93%. [0174] ATX-0043: Step 4
Figure imgf000058_0001
[0175] To a solution of 15 g 4-aminobutanoic acid (1 eq.) dissolved in 150 ml THF, 145 ml aqueous 1 N NaOH solution (1 eq.) was added at 0°C, followed by 43.4 ml Boc anhydride (1.3 eq.), sequentially using additional funnel, over a period of 15 minutes each one. The resulting solution was stirred at room temperature for 4 hours.
[0176] Progress of the reaction was monitored by TLC (10% MeOH in chloroform (CHCh); Rf: 0.5). Reaction mass was quenched with 5% HCl (150 ml) and then EtOAc (100 ml) was added. Organic layer was separated and the aqueous layer was washed with EtOAc (2 x 100 ml). Combined organic layer was concentrated under reduced pressure to get gummy liquid. Quantity produced, 20.0 g; yield, 68%.
[ -0043: Step 5
Figure imgf000058_0002
[0178] To a solution of 12 g 4-((tert-butoxycarbonyl) amino) butanoic acid (1 eq.) dissolved in DCM (200 ml), cooled to below 0°C was added 14.7 g l-ethyl-3-(3- dimethylaminopropyl)carbodiimide (EDC).HCl (1.3 eq.), 10.6 ml Et3N (1.3 eq.), and 0.72 g 4-Dimethylaminopyridine (DMAP; 0.1 eq.) sequentially under nitrogen atmosphere with 10- minute intervals. To this resulting solution alcohol was added at the same temperature, by dissolving in DCM (50 ml), using additional funnel, and stirred at room temperature for 24 hours under nitrogen atmosphere.
[0179] Progress of the reaction was monitored by TLC (10% EtOAc in hexane; Rf: 0.5). Reaction mass was quenched with water (100 ml) and then organic layer was separated. Aqueous layer was washed with DCM (2 x 50 ml). Combined organic layer was
concentrated under reduced pressure. The resulting crude was washed with saturated
NaHC03 solution (100 ml) and then extracted with EtOAc (2 x 50 ml). Organic layer was concentrated under reduced pressure and proceeded to next step with crude. Quantity produced, 8.5 g; yield, 48%.
-0043: Step 6
Figure imgf000058_0003
[0181] To a solution 8.5 g undecan-6-yl 4-((tert-butoxycarbonyl) amino) butanoate (1 eq.) dissolved in 70 ml DCM, was added trifluoroacetic acid (TFA; 10 eq.) at 0°C and stirred at room temperature for 4 hours under nitrogen atmosphere.
[0182] Progress of the reaction was monitored by TLC (70% EtOAc/hexane; Rf: 0.2). Reaction mass was concentrated under reduced pressure. The resulting crude was washed with saturated NaHCC solution (150 ml) and then extracted with EtOAc (2 x 100 ml). Organic layer was separated and concentrated under reduced pressure.
[0183] Crude compound was subjected to column chromatography using (60-120 mesh silica gel; 4% MeOH/CHCh and 1ml of triethylamine), and alcohol was recovered. Quantity produced, 5.0 g; yield, 33% (with respect to alcohol).
[0184] ATX-0043: Step 7
Figure imgf000059_0001
[0185] To a solution of 14 g 4-bromo butyric acid (1 eq.) dissolved in DCM (100 ml), cooled to below 0°C was added 21 g EDC.HCl (1.3 eq.), 15.2 ml Et3N (1.3 eq.), and 1 g DMAP (0.1 eq.) sequentially under nitrogen atmosphere with 10-minute intervals. To this resulting solution 8.3 g (Z)-non-2-en-l-ol (0.7 eq.) was added, by dissolving in 50 ml of DCM, using additional funnel, and stirred at room temperature for 16 hours under nitrogen atmosphere.
[0186] Progress of the reaction was monitored by TLC (10% EtOAc in hexane; Rf: 0.7). Reaction mass was quenched with water (50 ml) and then organic layer was separated. Aqueous layer was washed with DCM (2 x 50 ml). Combined organic layer was concentrated under reduced pressure. The resulting crude was washed with saturated NaHCC solution (100 ml) and then extracted with EtOAc (2 x 50 ml). Organic layer was separated and concentrated under reduced pressure.
[0187] Crude compound was subjected to column chromatography (60-120 mesh silica gel) using 5% EtOAc/hexane. Quantity produced, 11.0 g; yield, 64%.
[0188] ATX-0043: Step 8
Figure imgf000059_0002
[0189] To a 250 ml round bottom flask, 2 g undecan-6-yl 4-aminobutanoate (1 eq.) and 2.2 g (Z)-non-2-en-l-yl 4-bromobutanoate (1 eq.) in DMF, 1.2 g potassium carbonate (1.2 eq.) was added and the resulting mixture was refluxed at 90°C for 4 hours under nitrogen atmosphere.
[0190] Progress of the reaction was monitored by TLC (10% MeOH in CHCh; Rf: 0.5). Ice water was added to the reaction mass and then extracted with EtOAc and dried over sodium sulphate and concentrated under reduced pressure.
[0191] Crude compound was subjected to column chromatography (100-200 mesh silica gel) using 15% EtOAc/hexane. Starting amine and bromo compounds were recovered. Quantity produced, 1.45 g; yield, 40%.
[0192] ATX-0043: Step 9
Figure imgf000060_0001
[0193] To a solution of 1.45 g (Z)-non-2-en-l-yl 4-((4-oxo-4-(undecan-6- yloxy)butyl)amino)butanoate (1 eq.) dissolved in dry DCM, was added 1.29 ml triethylamine 3 eq.) and 360 mg triphosgene (0.4 eq.) with 5 minute intervals at 0°C under nitrogen atmosphere. The resulting solution was stirred at room temperature, under nitrogen atmosphere for 1 hour. The resulting reaction mass was concentrated under reduced pressure and kept under nitrogen atmosphere.
[0194] To 360 mg sodium hydride (5.5 eq.) dissolved in dry THF (20 ml), in a 2 neck 250 ml round bottom flask stirred at 0°C under nitrogen atmosphere, was added 2.1 g 2- (dimethylamino)ethane-l -thiol hydrochloride (5.5 eq.) in THF (30 ml) and kept stirring for 5 minutes under nitrogen atmosphere. To this resulting solution the above carbonyl chloride dissolved in THF (50 mL) was added using additional funnel slowly for about 15 minutes, added to this resulting solution and stirred at room temperature for 1 hour. [0195] Reaction mass was quenched with saturated NH4CI solution (20 ml) and then EtOAc (20 mL) was added. Organic layer was separated and the aqueous layer was washed with EtOAc (2 x20 mL). Combined organic layer was concentrated and the resulting crude was subjected to column chromatography. Progress of the reaction was monitored by TLC (60% EtOAc/Hex; Rf: 0.5; PMA charring).
[0196] Purification was done using silica gel (100-200 mesh; 18% EtOAc/hexane) chromatography. Quantity produced, 500 mg; yield, 26%; confirmed by ¾ NMR; HPLC; and mass spectroscopy (Mass).
[0197] ATX-0043 / RL-43A: !H-NMR (PPM, 400 MHz, CDCh): δ = 5.63 (m, 1), 5.54 (m, 1), 4.87 (m, 1), 4.63 (d, J = 7.0, 2), 3.37 (brs, 4), 3.03 (t, J = 7.0, 2), 2.27 (s, 6), 2.22- 2.32 (4), 2.09 (m, 2), 1.80-1.90 (4), 1.45-1.55 (4), 1.20-1.40 (22), 0.83-0.92 (9).
Example 3: Synthesis of ATX-0057
[0198] FIG. 2 shows the synthetic pathway of ATX-0057 that is described further as follows.
[0199] ATX-0057: Step 1: N-methoxy-N-methyloctanamide
Figure imgf000061_0001
[0200] In a 2 liter, two neck round bottom flask, octanoic acid (1 eq.) dissolved in DCM (300 ml) was taken and then added 1.5 eq. oxalyl chloride slowly at 0°C, stirring under nitrogen atmosphere. The resulting reaction mixture was stirred at room temperature for 2 hours. In a separate 2 liter, two neck round bottom flask, to 2 eq. Ν,Ο- dimethylhydroxylamine hydrochloride in DCM (200 ml), was added 3 eq. trimethylamine using additional funnel, stirred at 0°C. To this resulting solution, the above acid chloride, after concentration under reduced pressure, was added under nitrogen atmosphere by dissolving in DCM (150 ml), dropwise using addition funnel for 20 minutes. The resulting reaction solution was stirred at room temperature for 3 hours under nitrogen atmosphere.
[0201] Progress of the reaction was monitored by TLC (20% EtOAc/hexane; Rf: 0.5). Reaction mass was diluted with water (250 ml). Organic layer was separated and the aqueous layer was washed with DCM (3 x 100 ml). Combined organic layer was concentrated under reduced pressure. Crude compound was subjected to column
chromatography using (60-120 mesh silica gel; 10% EtOAc/hexane). Quantity produced, 85 g; yield, 65%. [0202] ATX-0057: Step 2: hexadecane-8-one
Figure imgf000062_0001
[0203] To a solution of octyl magnesium bromide in THF (100 ml), taken in a 1 liter, two neck round bottom flask, stirred at 0°C under nitrogen atmosphere, was added N- methoxy-N-methyloctanamide solution (dissolved in 200 ml THF) and the resulting reaction mixture was stirred at room temperature for 4 hours.
[0204] Progress of the reaction was monitored by TLC (10% EtOAc in hexane; Rf: 0.7). Reaction mass was quenched with saturated NH4CI solution (250 ml) and then EtOAc (350 ml) was added. The organic layer was separated and the aqueous layer was washed with EtOAc (2 x 100 ml). Combined organic layers were concentrated under reduced pressure. Crude compound was subjected to column chromatography using (60-120 mesh silica gel; 2% EtOAc/hexane). Quantity produced, 65 g; yield, 63%.
[0205] -0057: Step 3: hexadecane-8-ol
Figure imgf000062_0002
[0206] To a solution of hexadecan-8-one (1 eq.) dissolved in MeOH/THF, 1 eq. sodium borohydride was added at 0°C and the resulting solution was stirred at room temperature for 1.5 hours.
[0207] Progress of the reaction was monitored by TLC (10% EtOAc in hexane; Rf: 0.5). Reaction mass was quenched with saturated NH4CI solution (75 ml). Solvent was removed under reduced pressure and the resulting crude was portioned between EtOAc (150 ml) and water (100 ml). Organic layer was separated and the aqueous layer was washed with EtOAc (3 x 100 ml). Combined organic layers were concentrated under reduced pressure to get white solid. Quantity produced, 60 g; yield, 91%.
[0208] A -0057: Step 4: 4-((tert-butoxycarbonyl) amino)butanoic acid
Figure imgf000062_0003
[0209] To a solution of 4-aminobutanoic acid, dissolved in THF, aqueous 1 N NaOH solution was added at 0°C, followed by Boc anhydride, sequentially using additional funnel, over a period of 15 minutes. The resulting solution was stirred at room temperature for 4 hours. [0210] Progress of the reaction was monitored by TLC (10% MeOH in CHCh; Rf: 0.5). Reaction mass was quenched with 5% HC1 (250 ml) and then EtOAc (300 ml) was added. Organic layer was separated and the aqueous layer was washed with EtOAc (3 x 150 ml). Combined organic layer was concentrated under reduced pressure to obtain a gummy liquid. Quantity produced, 80 g; yield, 81%.
[0211] ATX-0057: Step 5: hexadecan-8-yl 4-((tert- butoxycarbo
Figure imgf000063_0001
[0212] To a solution of 4-((tert-butoxycarbonyl) amino)butanoic acid, dissolved in DCM (200 ml), cooled to below 0°C was added EDC.HC1, EtsN, and 4- dimethylaminopyridine (DMAP), sequentially under nitrogen atmosphere with 10 minutes interval. To this resulting solution, 1 eq. hexadecane-8-ol alcohol was added at the same temperature, by dissolving in DCM (150 ml), using additional funnel, and stirred at room temperature for 24 hours under nitrogen atmosphere.
[0213] Progress of the reaction was monitored by TLC (10% EtOAc in hexane; Rf: 0.5). Reaction mass was quenched with water (150 ml) and then organic layer was separated. Aqueous layer was washed with DCM (2 x 100 ml). Combined organic layer was concentrated under reduced pressure. The resulting crude was washed with saturated NaHC03 solution (150 ml) and then EtOAc (200 ml) was added. Organic layer was separated, concentrated under reduced pressure, and proceeded to next step with crude.
Quantity produced, 80 g (crude; required compound and alcohol).
-0057: Step 6: hexadecan-8-yl 4-aminobutanoate
Figure imgf000063_0002
[0215] To a solution of hexadecan-8-yl-4-((tert-butoxycarbonyl)amino)butanoate, dissolved in DCM, was added TFA at 0°C and stirred at room temperature for 3 hours under nitrogen atmosphere. Progress of the reaction was monitored by TLC (10% MeOH in CHCh; Rf: 0.3). Reaction mass was concentrated under reduced pressure. The resulting crude was washed with a saturated NaHC03 solution (300 ml) and then extracted with EtOAc (2 x 200 ml). The organic layer was separated and concentrated under reduced pressure. Crude compound was subjected to column chromatography using (60-120 mesh silica gel; 4% MeOH/CHCh and 1ml of triethylamine), and alcohol was recovered. Quantity produced, 40 g; yield, 59% for two steps; confirmed by Mass.
[0216] ATX-0057: Step 7: (Z)-non-2-en-l-yl 4-bromobutanoate
Figure imgf000064_0001
[0217] To a solution of 4-bromo butyric acid, dissolved in DCM (400 ml), cooled to below 0°C was added to EDC.HCl, Et3N, and DMAP sequentially under nitrogen atmosphere with 10-minute intervals. To this resulting solution (Z)-non-2-en-l-ol was added, by dissolving in 100 ml of DCM, using additional funnel, and stirred at room temperature for 24 hours under nitrogen atmosphere.
[0218] Progress of the reaction was monitored by TLC (10% EtOAc in hexane; Rf: 0.7). Reaction mass was quenched with water (300 ml) and then organic layer was separated. Aqueous layer was washed with DCM (2 x 150 ml). Combined organic layer was concentrated under reduced pressure. The resulting crude was washed with saturated NaHCCb solution (200 ml) and then extracted with EtOAc (150 ml). Organic layer was separated and concentrated under reduced pressure. Crude compound was subjected to column chromatography (60-120 mesh silica gel) using 5% EtOAc/hexane. Alcohol was recovered. Quantity produced, 27 g; yield, 51%.
[0219] ATX-0057: Step 8: hexadecan-8-yl (Z)-4-((4-(non-2-en-l-yloxy)-4-
Figure imgf000064_0002
[0220] To a solution of hexadecan-8-yl 4-aminobutanoate, (Z)-non-2-en-l-yl 4- bromobutanoate in acetonitrile (ACN), potassium carbonate was added and the resulting mixture was refluxed at 90°C for 4 hours under nitrogen atmosphere. Progress of the reaction was monitored by TLC (10% MeOH in CHCh; Rf: 0.5). Reaction mass was filtered, washed with ACN (20 ml), and the filtrate concentrated under reduced pressure. Crude compound was subjected to column chromatography (100-200 mesh silica gel) using 15%
EtOAc/hexane. Starting materials, amine and bromo compounds, were recovered. Quantity produced, 20 g; yield, 40%; confirmed by Mass. [02 -0057: Step 9
Figure imgf000065_0001
[0222] To a solution of hexadecan-8-yl (Z)-4-((4-(non-2-en-l-yloxy)-4-oxobutyl) amino) butanoate, dissolved in dry DCM, was added trimethylamine and triphosgene with 5 minutes interval at 0°C under nitrogen atmosphere. The resulting solution was stirred at room temperature, under nitrogen atmosphere for 1 hour. The resulting reaction mass was concentrated under reduced pressure and kept under nitrogen atmosphere.
[0223] To sodium hydride dissolved in dry THF (50 ml), in a two neck 100 ml round bottom flask stirred at 0°C under nitrogen atmosphere, was added 2- (dimethylamino)propane-l -thiol hydrochloride and kept stirring for 5 minutes under nitrogen atmosphere. To this resulting solution the above carbamoyl chloride, dissolved in THF (80 ml), was added via syringe slowly for about 10 minutes. The resulting solution was stirred at room temperature for 6 hours under nitrogen atmosphere.
[0224] Progress of the reaction was monitored by TLC (60% EtOAC/hexane; Rf: 0.5; PMA charring). Reaction mass was quenched with saturated NH4CI solution (75 ml) and then EtOAc (150 ml) was added. Organic layer was separated and the aqueous layer was washed with EtOAc (3 x 40 ml). Combined organic layer was concentrated and the resulting crude was subjected to column chromatography.
[0225] The first purification was done using silica gel (60-120 mesh) 22 g of crude compound was adsorbed on 60 g of silica gel and poured onto 500 g of silica gel taken in the column. Compound was eluted at 35% EtOAc/hexane. The second purification was done using neutral alumina with HPLC grade solvents. Crude compound, 7.5 g, was adsorbed on 18 g of neutral alumina and the resulting was poured onto 130 g of neutral alumina taken in the column. Compound was eluted at 10% EtOAc/hexane. Yield, 29%; confirmed by ¾ NMR, HPLC, and Mass.
[0226] ATX-0057 / RL-43C: Ή-ΝΜΙί (PPM, 400MHz, CDCh): δ = 5.63 (m, 1), 5.51 (m, 1), 4.68 (m, 1), 4.83 (d, J = 7.0, 2), 3.19 (brs, 4), 3.22 (m, 2), 2.52 (m, 2), 2.23-2.37 (4), 2.18 (s, 6), 2.08 (m, 2), 1.84-1.93 (4), 1.46-1.54 (4), 1.20-1.40 (30), 0.83-0.91 (9).
Example 4: Synthesis of ATX-0058
[0227] FIG. 3 shows the synthetic pathway of ATX-0058 that is described further as follows.
[0228] ATX-0058: Step 1
Figure imgf000066_0001
[0229] In 500 ml two neck round bottom flask under N2 atmosphere, 30 g 8- bromooctanoic acid (1 eq.) dissolved in 200 ml of DCM was taken and then added slowly to 26.7 ml oxalyl chloride (1.5 eq.) at 0°C, stirring under nitrogen atmosphere. The resulting reaction mixture was stirred at room temperature for 2 hours.
[0230] In a separate 1 liter two neck round bottom flask, 40.5 g Ν,Ο- dimethylhydroxylamine hydrochloride (2 eq.) in 300 ml DCM was added 87 ml
trimethylamine (3 eq.) stirred at 0°C. To this resulting solution, the above acid chloride was added after concentration under reduced pressure, by dissolving in 500 ml DCM, dropwise using addition funnel for 15 minutes. The resulting reaction solution was stirred at room temperature for 3 hours under nitrogen atmosphere.
[0231] Progress of the reaction was monitored by TLC (20% EtOAc/hexane; Rf: 0.5). Reaction mass was diluted with water (300 ml). Organic layer was separated and the aqueous layer was washed with DCM (2 x 100 ml). Combined organic layer was concentrated under reduced pressure.
[0232] Crude compound was subjected to column chromatography using (60-120 mesh silica gel; 10% EtOAc/hexane). Quantity produced, 28 g.
[0233] ATX-0058: Step 2
Figure imgf000066_0002
[0234] To a solution of 28 g hexyl magnesium bromide (1 eq.) in THF (100 ml), stirred at 0°C under nitrogen atmosphere, was added 36.8 g N-methoxy-N-methyloctanamide (1.3 eq.) in 200 ml THF and the resulting reaction mixture was stirred at room temperature for 5 hours.
[0235] Progress of the reaction was monitored by TLC (10% EtOAc/hexane; Rf: 0.7). Reaction mass was quenched with saturated NH4CI solution (100 ml). The organic layer was separated and the aqueous layer was washed with EtOAc (2 x 100 ml). Combined organic layer was concentrated under reduced pressure.
[0236] Crude compound was subjected to column chromatography using (60-120 mesh silica gel; 2% ethyl acetate/hexane). Quantity produced, 24 g; yield, 77%.
[0237] ATX-0058: Step 3
Figure imgf000067_0001
[0238] To a solution of 24 g tetradecan-7-one (1 eq.) dissolved in MeOH/THF, 4.27 g sodium borohydride (1 eq.) was added at 0°C and the resulting solution was stirred at room temperature for 1 hour.
[0239] Progress of the reaction was monitored by TLC (10% EtOAc/hexane; Rf: 0.5). Reaction mass was quenched with saturated NH4CI solution (50 ml). Methanol was reduced under reduced pressure. The resulting crude was portioned between EtOAc (200 ml) and water. Organic layer was separated and the aqueous layer was washed with EtOAc (2 x 80 ml). Combined organic layer was concentrated under reduced pressure to obtain a white solid. Quantity produced, 21.5 g; yield, 89%.
[0240] ATX-0058: Step 4
H0^^^NH2 ( oc)?Q , H0^^^N- Boc
O O H
[0241] To a solution of 20 g 4-aminobutanoic acid dissolved in 140 ml THF, 196 ml of aqueous IN NaOH solution was added at 0°C, followed by 36.8 g Boc anhydride, using a funnel. The resulting solution was stirred at room temperature for 4 hours.
[0242] Progress of the reaction was monitored by TLC (10% MeOH/CHCh; Rf: 0.5). Reaction mass was quenched with 5% HC1 (100 ml) and then EtOAc (200 ml) was added. The organic layer was separated and the aqueous layer was washed with EtOAc (2 x 100 ml). Combined organic layer was concentrated under reduced pressure to obtain a gummy liquid. Quantity produced, 30 g; yield, 76%. [0243] -0058: Step 5
Figure imgf000068_0001
[0244] To a solution of 10 g 4-((tert-butoxycarbonyl)amino)butanoic acid (1 eq.) dissolved in DCM (150 ml), cooled to below 0°C was added 12.2 g EDC.HCl (1.3 eq.), 20.4 ml Et3N (3 eq.), and 488 mg DMAP (0.1 eq.) sequentially with 10 minutes interval. To this resulting solution alcohol was added, by dissolving in DCM, using additional funnel, and stirred at room temperature for 24 hours under nitrogen atmosphere.
[0245] Progress of the reaction was monitored by TLC (10% EtOAc/hexane; Rf: 0.5). Reaction mass was quenched with water (100 ml) and the organic layer was separated. The aqueous layer was washed with DCM (2 x 50 ml). Combined organic layer was concentrated under reduced pressure. The resulting crude was washed with saturated NaHCC solution and EtOAc (100 ml) was added. The organic layer was separated and concentrated under reduced pressure, and proceeded to next step with crude. Quantity produced, 12.7 g (crude).
[0246] ATX-0058: Step 6
Figure imgf000068_0002
[0247] To a solution of 12.5 g tetradecan-7-yl 4-((tert- butoxycarbonyl)amino)butanoate (1 eq.) dissolved in 100 ml DCM, was added 23.9 ml TFA (10 eq.) at 0°C and stirred at room temperature for 3 hours under nitrogen atmosphere.
[0248] Progress of the reaction was monitored by TLC (10% MeOH/CHCb; Rf: 0.3). Reaction mass was concentrated under reduced pressure. The resulting crude was washed with saturated NaHCC solution (100 ml) and EtOAc (100 ml) was added. The organic layer was separated and concentrated under reduced pressure.
[0249] Crude compound was subjected to column chromatography using (60-120 mesh silica gel; 4% MeOH/CHCb) and alcohol was recovered. Quantity produced, 7 g for two steps; yield, 47%; confirmed by Mass.
[0250] ATX-0058: Step 7
Figure imgf000068_0003
[0251] To a solution of 20 g 4-bromo butyric acid (1 eq.) dissolved in DCM (150 ml), cooled to 0°C was added 1.5 eq. EDC.HC1, 3 eq. Et3N, and 0.1 eq. DMAP sequentially with 10 minutes interval. To this resulting solution 0.7 eq. (Z)-non-2-en-l-ol was added, by dissolving in 100 ml DCM, using a funnel, and stirred at room temperature for 24 hours under nitrogen atmosphere.
[0252] Progress of the reaction was monitored by TLC (10% EtOAc/hexane; Rf: 0.7). Reaction mass was quenched with water (100 ml) and then organic layer was separated. Aqueous layer was washed with DCM (2 x 100 ml). Combined organic layer was concentrated under reduced pressure. The resulting crude was washed with saturated NaHCC solution and EtOAc (150 ml) was added. The organic layer was separated and concentrated under reduced pressure.
[0253] Crude compound was subjected to column chromatography using (60-120 mesh silica gel; 5% EtOAc/hexane). Quantity produced, 17 g; yield, 69%; confirmed by ¾ NMR.
[0254] ATX-0058: Step 8
Figure imgf000069_0001
[0255] To a solution of 6 g tetradecan-7-yl 4-aminobutanoate (1 eq.), 5.8 g (Z)-non- 2-en-l-yl 4-bromobutanoate (1 eq.) in ACN (125 ml), 2.7 g potassium carbonate (1.2 eq.) was added and the resulting was refluxed at 90°C for 3 hours under nitrogen atmosphere.
[0256] Progress of the reaction was monitored by TLC (10% MeOH/CHCb; Rf: 0.5). Reaction mass was filtered and the filtrate concentrated under reduced pressure.
[0257] Crude compound was subjected to column chromatography using (100-200 mesh silica gel; 15% EtOAc/hexane). Quantity produced, 4.5 g; yield, 44%; confirmed by Mass.
[0258] ATX-0058: Step 9
Figure imgf000070_0001
[0259] To a solution of 4.4 g (Z)-non-2-en-l-yl 4-((4-oxo-4-(tetradecan-7- yloxy)butyl)amino)butanoate (1 eq.) dissolved in 30 ml dry DCM, was added 0.83 ml trimethylamine (3 eq.) and 418 mg triphosgene (0.5 eq.) with 5 minutes interval, at 0°C under nitrogen atmosphere. The resulting solution was stirred at room temperature, under nitrogen atmosphere for 1 hour. The resulting reaction mass was concentrated under reduced pressure and kept under nitrogen atmosphere.
[0260] To 192 mg sodium hydride (10 eq.) dissolved in dry THF (25 ml), in a two neck 100 ml round bottom flask, was added 564 mg 2-(dimethylamino)propane-l -thiol hydrochloride (5 eq.) at 0°C and kept stirring for 5 minutes under nitrogen atmosphere. To this resulting solution the above carbamoyl chloride, dissolved in THF (35 ml), was added via syringe slowly for about 10 minutes. The resulting solution was stirred at room temperature for 4 hours under nitrogen atmosphere.
[0261] Progress of the reaction was monitored by TLC (60% EtOAC/hexane; Rf: 0.5; PMA charring). Reaction mass was quenched with saturated NH4CI (30 ml) and then EtOAc (100 ml) was added. The organic layer was separated and the aqueous layer was washed with EtOAc (2 x 50 ml). Combined organic layer was concentrated and the resulting crude was subjected to column chromatography.
[0262] A first purification was done using silica gel (60-120 mesh). 5.0 g of crude compound was adsorbed on 9 g of silica gel and poured onto 90 g of silica gel taken in the column. Compound was eluted at 35% EtOAc/hexane. A second purification was done using neutral alumina with HPLC grade solvents. 1.5 g of crude compound was adsorbed on 4 g of neutral alumina and the resulting was poured onto 40 g of neutral alumina taken in the column. Compound was eluted at 10% EtOAc/hexane. Quantity produced, 1.2 g; yield, 21%; confirmed by ¾ NMR; HPLC; Mass.
[0263] ATX-0058 / RL-43B: ^-NMR (PPM, 400MHz, CDCh): δ = 5.65 (m, 1), 5.52 (m, 1), 4.86 (m, 1), 4.63 (d, J = 7.0, 2), 3.37 (brs, 4), 3.02 (t, J = 6.0, 2), 2.53 (t, J = 6.0, 2), 2.27-2.36 (4), 2.27 (s, 6), 2.09 (m, 2), 1.83-1.96 (4), 1.46-1.54 (4), 1.20-1.40 (26), 0.84- 0.91 (9).
Example 5: Synthesis of ATX-0081
[0264] FIG. 4 shows the synthetic pathway of ATX-0081 that is described further as follows.
[0265] -0081: Step 1
Figure imgf000071_0001
[0266] In a 2 liter, two neck round bottom flask, octanoic acid dissolved in DCM (200 ml) was taken and then added 1.5 eq. oxalyl chloride slowly at 0°C, stirring under nitrogen atmosphere. The resulting reaction mixture was stirred at room temperature for 2 hours. In a separate 2 liter, two neck round bottom flask, to 2 eq. Ν,Ο- dimethylhydroxylamine hydrochloride in DCM (200 ml), was added 3 eq. trimethylamine using additional funnel, stirred at 0°C. To this resulting solution, the above acid chloride, after concentration under reduced pressure, was added under nitrogen atmosphere by dissolving in DCM (150 ml), dropwise using addition funnel for 20 minutes. The resulting reaction solution was stirred at room temperature for 3 hours under nitrogen atmosphere.
[0267] Progress of the reaction was monitored by TLC (20% EtOAc/hexane; Rf 0.5). Reaction mass was diluted with water (250 ml). Organic layer was separated and the aqueous layer was washed with DCM (3 x 100 ml). Combined organic layer was concentrated under reduced pressure. Crude compound was subjected to column
chromatography using (60-120 mesh silica gel; 10% EtOAc/hexane). Quantity produced, 33 g; yield, 84%.
[0268] -0081: Step 2
Figure imgf000071_0002
[0269] To a solution of 22 g heptyl magnesium bromide (1.5 eq.) in THF (100 ml), taken in a 1 liter, two neck round bottom flask, stirred at 0°C under nitrogen atmosphere, was added N-methoxy-N-methyloctanamide (1 eq.) solution (dissolved in 200 ml THF) and the resulting reaction mixture was stirred at room temperature for 4 hours.
[0270] Progress of the reaction was monitored by TLC (10% EtOAc in hexane; Rf: 0.7). Reaction mass was quenched with saturated NH4CI solution (250 ml) and then EtOAc (350 ml) was added. The organic layer was separated and the aqueous layer was washed with EtOAc (2 x 100 ml). Combined organic layers were concentrated under reduced pressure. Crude compound was subjected to column chromatography using (60-120 mesh silica gel; 2% EtOAc/hexane). Quantity produced, 22 g; yield, 65%.
[0271] -0081: Step 3
Figure imgf000072_0001
[0272] To a solution of 22 g pentadecan-8-one (1 eq.) dissolved in MeOH/THF, 1.5 eq. sodium borohydride was added at 0°C and the resulting solution was stirred at room temperature for 1 hour.
[0273] Progress of the reaction was monitored by TLC (10% EtOAc in hexane; Rf: 0.5). Reaction mass was quenched with saturated NH4CI solution (75 ml). Solvent was removed under reduced pressure and the resulting crude was portioned between EtOAc (150 ml) and water (100 ml). Organic layer was separated and the aqueous layer was washed with EtOAc (3 x 100 ml). Combined organic layers were concentrated under reduced pressure to get white solid. Quantity produced, 20 g; yield, 90%.
[0274] A -0081: Step 4
Figure imgf000072_0002
[0275] To a solution of 50 g 4-aminobutanoic acid dissolved in 350 ml THF, 490 ml aqueous 1 N NaOH solution was added at 0°C, followed by 140 ml Boc anhydride, sequentially using additional funnel, over a period of 15 minutes. The resulting solution was stirred at room temperature for 4 hours.
[0276] Progress of the reaction was monitored by TLC (10% MeOH in CHCh; Rf: 0.5). Reaction mass was quenched with 5% HC1 (250 ml) and then EtOAc (300 ml) was added. Organic layer was separated and the aqueous layer was washed with EtOAc (3 x 150 ml). Combined organic layer was concentrated under reduced pressure to obtain a gummy liquid. Quantity produced, 80 g; yield, 81%.
[0277] -0081: Step 5
Figure imgf000073_0001
[0278] To a solution of 10 g 4-((tert-butoxycarbonyl) amino)butanoic acid, dissolved in DCM (250 ml), cooled to below 0°C was added 1.3 eq. EDC.HC1, Et3N, and 4- dimethylaminopyridine (DMAP), sequentially under nitrogen atmosphere with 10 minutes interval. To this resulting solution, 1 eq. pentadecane-7-ol alcohol was added at the same temperature, by dissolving in DCM (150 ml), using additional funnel, and stirred at room temperature for 24 hours under nitrogen atmosphere.
[0279] Progress of the reaction was monitored by TLC (10% EtOAc in hexane; Rf: 0.5). Reaction mass was quenched with water (150 ml) and then organic layer was separated. Aqueous layer was washed with DCM (2 x 100 ml). Combined organic layer was concentrated under reduced pressure. The resulting crude was washed with saturated NaHCC solution (150 ml) and then EtOAc (200 ml) was added. Organic layer was separated, concentrated under reduced pressure, and proceeded to next step with crude.
Quantity produced, 8.5 g (crude; required compound and alcohol).
[028 -0081: Step 6
Figure imgf000073_0002
[0281] To a solution of 8.5 g pentadecan-8-yl 4-((tert- butoxycarbonyl)amino)butanoate dissolved in 65 ml DCM, was added 10 eq. TFA at 0°C and stirred at room temperature for 3 hours under nitrogen atmosphere.
[0282] Progress of the reaction was monitored by TLC (10% MeOH in CHCh; Rf: 0.3). Reaction mass was concentrated under reduced pressure. The resulting crude was washed with a saturated NaHCC solution (300 ml) and then extracted with EtOAc (2 x 200 ml). The organic layer was separated and concentrated under reduced pressure. Crude compound was subjected to column chromatography using (60-120 mesh silica gel; 4% MeOH/CHCh and 1 ml of triethylamine), and alcohol was recovered. Quantity produced, 4 g for two steps; yield, 25%; confirmed by Mass. -0081: Step 7
Figure imgf000074_0001
[0284] To a solution of 4-bromo butyric acid, dissolved in DCM (300 ml), cooled to below 0°C was added to EDC.HCl, Et3N, and DMAP sequentially under nitrogen atmosphere with 10-minute intervals. To this resulting solution 20 g (Z)-non-2-en-l-ol was added, by dissolving in 100 ml of DCM, using additional funnel, and stirred at room temperature for 24 hours under nitrogen atmosphere.
[0285] Progress of the reaction was monitored by TLC (10% EtOAc in hexane; Rf: 0.7). Reaction mass was quenched with water (300 ml) and then organic layer was separated. Aqueous layer was washed with DCM (2 x 150 ml). Combined organic layer was concentrated under reduced pressure. The resulting crude was washed with saturated NaHCC solution (200 ml) and then extracted with EtOAc (150 ml). Organic layer was separated and concentrated under reduced pressure. Crude compound was subjected to column chromatography (60-120 mesh silica gel) using 5% EtOAc/hexane. Alcohol was recovered. Quantity produced, 19 g; yield, 55%.
-0081: Step 8
Figure imgf000074_0002
[0287] To a solution of 4.5 g pentadecan-8-yl 4-aminobutanoate, 1 eq. (Z)-non-2- en-l-yl 4-bromobutanoate in 70 ml acetonitrile (ACN), 1.4 eq. potassium carbonate was added and the resulting mixture was refluxed at 90°C for 4 hours under nitrogen atmosphere.
[0288] Progress of the reaction was monitored by TLC (10% MeOH in CHCh; Rf: 0.5). Reaction mass was filtered, washed with ACN (20 ml), and the filtrate concentrated under reduced pressure. Crude compound was subjected to column chromatography (100- 200 mesh silica gel) using 15% EtOAc/hexane. Starting materials, amine and bromo compounds, were recovered. Quantity produced, 2.1 g; yield, 27%; confirmed by Mass. [0289] ATX-0081: Step 9
Figure imgf000075_0001
[0290] To a solution of 2.1 g pentadecan-8-yl (Z)-4-((4-(non-2-en-l-yloxy)-4- oxobutyl) amino) butanoate, dissolved in 150 ml dry DCM, was added 3 eq. triethylamine and triphosgene with 5 minutes interval at 0°C under nitrogen atmosphere. The resulting solution was stirred at room temperature, under nitrogen atmosphere for 1 hour. The resulting reaction mass was concentrated under reduced pressure and kept under nitrogen atmosphere.
[0291] To 7 eq. sodium hydride dissolved in dry THF (80 ml), in a two neck 100 ml round bottom flask stirred at 0°C under nitrogen atmosphere, was added 3.5 eq. 2- (dimethylamino)propane-l -thiol hydrochloride and kept stirring for 5 minutes under nitrogen atmosphere. To this resulting solution the above carbamoyl chloride, dissolved in THF (80 ml), was added via syringe slowly for about 10 minutes. The resulting solution was stirred at 0°C to room temperature overnight under nitrogen atmosphere.
[0292] Progress of the reaction was monitored by TLC (60% EtOAC/hexane; Rf: 0.5; PMA charring). Reaction mass was quenched with saturated NH4CI solution (75 ml) and then EtOAc (150 ml) was added. Organic layer was separated and the aqueous layer was washed with EtOAc (3 x 40 ml). Combined organic layer was concentrated and the resulting crude was subjected to column chromatography.
[0293] The first purification was done using silica gel (60-120 mesh) of crude compound was adsorbed on 60 g of silica gel and poured onto 500 g of silica gel taken in the column. Compound was eluted at 35% EtOAc/hexane. The second purification was done using neutral alumina with HPLC grade solvents. Crude compound was adsorbed on 18 g of neutral alumina and the resulting was poured onto 130 g of neutral alumina taken in the column. Compound was eluted at 10% EtOAc/hexane. Quantity produced, 1.5 g; yield, 45%; confirmed by ¾ NMR, HPLC, and Mass.
[0294] ATX-0081 / RL-48B: Ή-ΝΜΙί (PPM, 500 MHz, CDCh): δ = 5.64 (m, 1), 5.52 (m, 1), 4.86 (m, 1), 4.63 (d, J = 7.0, 2), 3.31-3.44 (4), 3.02 (t, J = 7.0, 2), 2.52 (t, J = 7.0, 2), 2.26-2.36 (4), 2.27 (s, 6), 2.10 (m, 2), 1.84-1.95 (4), 1.46-1.54 (4), 1.20-1.40 (26), 0.85- 0.94 (9).
Example 6: Synthesis of ATX-0082
[0295] FIG. 5 shows the synthetic pathway of ATX-0082 that is described further as follows.
[0296] -0082: Step 1
Figure imgf000076_0001
[0297] In a 2 liter, two neck round bottom flask, 30 g octanoic acid dissolved in DCM (200 ml) was taken and then added 1.5 eq. oxalyl chloride slowly at 0°C, stirring under nitrogen atmosphere. The resulting reaction mixture was stirred at room temperature for 2 hours. In a separate 2 liter, two neck round bottom flask, to 2 eq. Ν,Ο- dimethylhydroxylamine hydrochloride in DCM (200 ml), was added 3 eq. trimethylamine using additional funnel, stirred at 0°C. To this resulting solution, the above acid chloride, after concentration under reduced pressure, was added under nitrogen atmosphere by dissolving in DCM (150 ml), dropwise using addition funnel for 20 minutes. The resulting reaction solution was stirred at room temperature for 3 hours under nitrogen atmosphere.
[0298] Progress of the reaction was monitored by TLC (20% EtOAc/hexane; Rf 0.5). Reaction mass was diluted with water (250 ml). Organic layer was separated and the aqueous layer was washed with DCM (3 x 100 ml). Combined organic layer was concentrated under reduced pressure. Crude compound was subjected to column
chromatography using (60-120 mesh silica gel; 10% EtOAc/hexane). Quantity produced, 33 g; yield, 84%. [0299] -0082: Step 2
Figure imgf000077_0001
[0300] To a solution of heptyl magnesium bromide (1.5 eq.) in THF (100 ml), taken in a 1 liter, two neck round bottom flask, stirred at 0°C under nitrogen atmosphere, was added 28 g N-methoxy-N-methyloctanamide (1 eq.) solution (dissolved in 200 ml THF) and the resulting reaction mixture was stirred at room temperature for 4 hours.
[0301] Progress of the reaction was monitored by TLC (10% EtOAc in hexane; Rf: 0.7). Reaction mass was quenched with saturated NH4CI solution (250 ml) and then EtOAc (350 ml) was added. The organic layer was separated and the aqueous layer was washed with EtOAc (2 x 100 ml). Combined organic layers were concentrated under reduced pressure. Crude compound was subjected to column chromatography using (60-120 mesh silica gel; 2% EtOAc/hexane). Quantity produced, 22 g; yield, 65%.
[0302] -0082: Step 3
Figure imgf000077_0002
[0303] To a solution of 22 g pentadecan-8-one (1 eq.) dissolved in MeOH/THF, 1.5 eq. sodium borohydride was added at 0°C and the resulting solution was stirred at room temperature for 1 hour.
[0304] Progress of the reaction was monitored by TLC (10% EtOAc in hexane; Rf: 0.5). Reaction mass was quenched with saturated NH4CI solution (75 ml). Solvent was removed under reduced pressure and the resulting crude was portioned between EtOAc (150 ml) and water (100 ml). Organic layer was separated and the aqueous layer was washed with EtOAc (3 x 100 ml). Combined organic layers were concentrated under reduced pressure to get white solid. Quantity produced, 20 g; yield, 90%.
[0305] ATX-0082: Step 4
H0^^^ NH2 (Boc)2Q , H0^^^ oc
O O
[0306] To a solution of 15 g 4-aminobutanoic acid dissolved in 120 ml THF, 185 ml aqueous 1 N NaOH solution was added at 0°C, followed by 50 ml Boc anhydride, sequentially using additional funnel, over a period of 15 minutes. The resulting solution was stirred at room temperature for 4 hours. [0307] Progress of the reaction was monitored by TLC (10% MeOH in CHCh; Rf: 0.5). Reaction mass was quenched with 5% HC1 (250 ml) and then EtOAc (300 ml) was added. Organic layer was separated and the aqueous layer was washed with EtOAc (3 x 150 ml). Combined organic layer was concentrated under reduced pressure to obtain a gummy liquid. Quantity produced, 27 g; yield, 85%.
[0308] -0082: Step 5
Figure imgf000078_0001
[0309] To a solution of 10 g 4-((tert-butoxycarbonyl) amino)butanoic acid, dissolved in DCM (250 ml), cooled to below 0°C was added 1.3 eq. EDC.HC1, Et3N, and 4- dimethylaminopyridine (DMAP), sequentially under nitrogen atmosphere with 10 minutes interval. To this resulting solution, 1 eq. pentadecane-7-ol alcohol was added at the same temperature, by dissolving in DCM (150 ml), using additional funnel, and stirred at room temperature for 24 hours under nitrogen atmosphere.
[0310] Progress of the reaction was monitored by TLC (10% EtOAc in hexane; Rf: 0.5). Reaction mass was quenched with water (150 ml) and then organic layer was separated. Aqueous layer was washed with DCM (2 x 100 ml). Combined organic layer was concentrated under reduced pressure. The resulting crude was washed with saturated NaHC03 solution (150 ml) and then EtOAc (200 ml) was added. Organic layer was separated, concentrated under reduced pressure, and proceeded to next step with crude.
Quantity produced, 8 g (crude; required compound and alcohol).
[0311] -0082: Step 6
Figure imgf000078_0002
[0312] To a solution of 8.0 g pentadecan-8-yl 4-((tert- butoxycarbonyl)amino)butanoate dissolved in 60 ml DCM, was added 10 eq. TFA at 0°C and stirred at room temperature for 3 hours under nitrogen atmosphere.
[0313] Progress of the reaction was monitored by TLC (10% MeOH in CHCh; Rf: 0.3). Reaction mass was concentrated under reduced pressure. The resulting crude was washed with a saturated NaHC03 solution (300 ml) and then extracted with EtOAc (2 x 200 ml). The organic layer was separated and concentrated under reduced pressure. Crude compound was subjected to column chromatography using (60-120 mesh silica gel; 4% MeOH/CHCh and 1 ml of triethylamine), and alcohol was recovered. Quantity produced, 4 g; yield, 25% for two steps; confirmed by Mass.
-0082: Step 7
Figure imgf000079_0001
[0315] To a solution of 4-bromo butyric acid, dissolved in DCM (400 ml), cooled to below 0°C was added to 1.5 eq. EDC.HC1, 3 eq. Et3N, and DMAP sequentially under nitrogen atmosphere with 10-minute intervals. To this resulting solution 20 g (Z)-non-2-en- l-ol was added, by dissolving in 100 ml of DCM, using additional funnel, and stirred at room temperature for 24 hours under nitrogen atmosphere.
[0316] Progress of the reaction was monitored by TLC (10% EtOAc in hexane; Rf: 0.7). Reaction mass was quenched with water (300 ml) and then organic layer was separated. Aqueous layer was washed with DCM (2 x 150 ml). Combined organic layer was concentrated under reduced pressure. The resulting crude was washed with saturated NaHCC solution (200 ml) and then extracted with EtOAc (150 ml). Organic layer was separated and concentrated under reduced pressure. Crude compound was subjected to column chromatography (60-120 mesh silica gel) using 5% EtOAc/hexane. Alcohol was recovered. Quantity produced, 18 g; yield, 55%.
[0 -0082: Step 8
Figure imgf000079_0002
[0318] To a solution of 4.0 g pentadecan-8-yl 4-aminobutanoate, 1 eq. (Z)-non-2- en-l-yl 4-bromobutanoate in 90 ml ACN, 1.4 eq. potassium carbonate was added and the resulting mixture was refluxed at 90°C for 4 hours under nitrogen atmosphere.
[0319] Progress of the reaction was monitored by TLC (10% MeOH in CHCh; Rf:
0.5). Reaction mass was filtered, washed with ACN (20 ml), and the filtrate concentrated under reduced pressure. Crude compound was subjected to column chromatography (100- 200 mesh silica gel) using 15% EtOAc/hexane. Starting materials (amine and bromo compounds) were recovered. Quantity produced, 2.2 g; yield, 30%; confirmed by Mass.
[0320] ATX-0082: Step 9
Figure imgf000080_0001
[0321] To a solution of 2.2 g pentadecan-8-yl (Z)-4-((4-(non-2-en-l-yloxy)-4- oxobutyl) amino) butanoate, dissolved in 25 ml dry DCM, was added 3 eq. triethylamine and triphosgene with 5 minutes interval at 0°C under nitrogen atmosphere. The resulting solution was stirred at room temperature, under nitrogen atmosphere for 1 hour. The resulting reaction mass was concentrated under reduced pressure and kept under nitrogen atmosphere.
[0322] To 7 eq. sodium hydride dissolved in dry THF (100 ml), in a two neck 100 ml round bottom flask stirred at 0°C under nitrogen atmosphere, was added 3.5 eq. 2- (dimethylamino)propane-l -thiol hydrochloride and kept stirring for 5 minutes under nitrogen atmosphere. To this resulting solution the above carbamoyl chloride, dissolved in THF (100 ml), was added via syringe slowly for about 10 minutes. The resulting solution was stirred at 0°C to room temperature overnight under nitrogen atmosphere.
[0323] Progress of the reaction was monitored by TLC (60% EtOAc/hexane; Rf 0.5; PMA charring). Reaction mass was quenched with saturated NH4CI solution (75 ml) and then EtOAc (150 ml) was added. Organic layer was separated and the aqueous layer was washed with EtOAc (3 x 40 ml). Combined organic layer was concentrated and the resulting crude was subjected to column chromatography. [0324] The first purification was done using silica gel (60-120 mesh) of crude compound was adsorbed on 60 g of silica gel and poured onto 500 g of silica gel taken in the column. Compound was eluted at 35% EtOAc/hexane. The second purification was done using neutral alumina with HPLC grade solvents. Crude compound was adsorbed on 18 g of neutral alumina and the resulting was poured onto 130 g of neutral alumina taken in the column. Compound was eluted at 10% EtOAc/hexane. Quantity produced, 1.2 g; yield, 43%; confirmed by ¾ NMR, HPLC, and Mass.
[0325] ATX-0082 / RL-47A: !H-NMR (PPM, 500 MHz, CDCh): δ = 5.64 (m, 1), 5.52 (m, 1), 4.87 (m, 1), 4.62 (d, J = 7.0, 2), 3.61 (t, J = 7.0, 2), 3.28-3.37 (2), 3.02 (t, J = 7.0, 2), 2.61 (m, 2), 2.52 (t, J = 7.0, 2), 2.31 (m, 2), 2.27 (s, 6), 2.10 (m, 2), 1.62-1.70 (6), 1.21- 1.40 (32), 0.85-0.91 (9).
Example 7: Synthesis of ATX-0086
[0326] FIG. 6 shows the synthetic pathway of ATX-0086 that is described further as follows.
[0327] ATX-0086: Step 1
Figure imgf000081_0001
[0328] In a 2 liter, two neck round bottom flask, 30 g octanoic acid dissolved in DCM (200 ml) was taken and then added 1.5 eq. oxalyl chloride slowly at 0°C, stirring under nitrogen atmosphere. The resulting reaction mixture was stirred at room temperature for 2 hours. In a separate 2 liter, two neck round bottom flask, to 2 eq. Ν,Ο- dimethylhydroxylamine hydrochloride in DCM (200 ml), was added 3 eq. trimethylamine using additional funnel, stirred at 0°C. To this resulting solution, the above acid chloride, after concentration under reduced pressure, was added under nitrogen atmosphere by dissolving in DCM (150 ml), dropwise using addition funnel for 20 minutes. The resulting reaction solution was stirred at room temperature for 3 hours under nitrogen atmosphere.
[0329] Progress of the reaction was monitored by TLC (20% EtOAc/hexane; Rf 0.5). Reaction mass was diluted with water (250 ml). Organic layer was separated and the aqueous layer was washed with DCM (3 x 100 ml). Combined organic layer was concentrated under reduced pressure. Crude compound was subjected to column
chromatography using (60-120 mesh silica gel; 10% EtOAc/hexane). Quantity produced, 38 g; yield, 84%; confirmed by Mass. [0330] -0086: Step 2
Figure imgf000082_0001
[0331] To a solution of hexyl magnesium bromide (1.5 eq.) in THF (100 ml), taken in a 1 liter, two neck round bottom flask, stirred at 0°C under nitrogen atmosphere, was added 38 g N-methoxy-N-methyloctanamide (1 eq.) solution (dissolved in 200 ml THF) and the resulting reaction mixture was stirred at room temperature for 4 hours.
[0332] Progress of the reaction was monitored by TLC (10% EtOAc in hexane; Rf: 0.7). Reaction mass was quenched with saturated NH4CI solution (250 ml) and then EtOAc (350 ml) was added. The organic layer was separated and the aqueous layer was washed with EtOAc (2 x 100 ml). Combined organic layers were concentrated under reduced pressure. Crude compound was subjected to column chromatography using (60-120 mesh silica gel; 2% EtOAc/hexane). Quantity produced, 44 g; yield, 65%; confirmed by Mass.
[0333] A -0086: Step 3
Figure imgf000082_0002
[0334] To a solution of 44 g tridecane-7-one (1 eq) dissolved in MeOH/THF, 1.5 eq. sodium borohydride was added at 0°C and the resulting solution was stirred at room temperature for 1 hour.
[0335] Progress of the reaction was monitored by TLC (10% EtOAc in hexane; Rf: 0.5). Reaction mass was quenched with saturated NH4CI solution (75 ml). Solvent was removed under reduced pressure and the resulting crude was portioned between EtOAc (150 ml) and water (100 ml). Organic layer was separated and the aqueous layer was washed with EtOAc (3 x 100 ml). Combined organic layers were concentrated under reduced pressure to get white solid. Quantity produced, 40 g; yield, 90%; confirmed by Mass.
[0336] ATX-0086: Step 4
H0^^^^NH2 (Boc)2Q , HO^^^^N. Boc
O O H
[0337] To a solution of 50 g 4-aminobutanoic acid dissolved in 350 ml THF, 490 ml aqueous 1 N NaOH solution was added at 0°C, followed by 140 ml Boc anhydride, sequentially using additional funnel, over a period of 15 minutes. The resulting solution was stirred at room temperature for 4 hours. [0338] Progress of the reaction was monitored by TLC (10% MeOH in CHCh; Rf: 0.5). Reaction mass was quenched with 5% HC1 (250 ml) and then EtOAc (300 ml) was added. Organic layer was separated and the aqueous layer was washed with EtOAc (3 x 150 ml). Combined organic layer was concentrated under reduced pressure to obtain a gummy liquid. Quantity produced, 80 g; yield, 81%; confirmed by Mass.
[0339] ATX-0086: Step 5
Figure imgf000083_0001
[0340] To a solution of 10 g 4-((tert-butoxycarbonyl) amino)butanoic acid, dissolved in DCM (250 ml), cooled to below 0°C was added 1.3 eq. EDC.HC1, Et3N, and 4- dimethylaminopyridine (DMAP), sequentially under nitrogen atmosphere with 10 minutes interval. To this resulting solution, 1 eq. pentadecane-7-ol alcohol was added at the same temperature, by dissolving in DCM (150 ml), using additional funnel, and stirred at room temperature for 24 hours under nitrogen atmosphere.
[0341] Progress of the reaction was monitored by TLC (10% EtOAc in hexane; Rf: 0.5). Reaction mass was quenched with water (150 ml) and then organic layer was separated. Aqueous layer was washed with DCM (2 x 100 ml). Combined organic layer was concentrated under reduced pressure. The resulting crude was washed with saturated NaHC03 solution (150 ml) and then EtOAc (200 ml) was added. Organic layer was separated, concentrated under reduced pressure, and proceeded to next step with crude.
Quantity produced, 8 g (crude; required compound and alcohol).
[ -0086: Step 6
Figure imgf000083_0002
[0343] To a solution of 8.0 g pentadecan-8-yl 4-((tert- butoxycarbonyl)amino)butanoate dissolved in 60 ml DCM, was added 10 eq. TFA at 0°C and stirred at room temperature for 3 hours under nitrogen atmosphere.
[0344] Progress of the reaction was monitored by TLC (10% MeOH in CHCh; Rf: 0.3). Reaction mass was concentrated under reduced pressure. The resulting crude was washed with a saturated NaHC03 solution (300 ml) and then extracted with EtOAc (2 x 200 ml). The organic layer was separated and concentrated under reduced pressure. Crude compound was subjected to column chromatography using (60-120 mesh silica gel; 4% MeOH/CHCh and lmL of triethylamine), and alcohol was recovered. Quantity produced, 3.5 g; yield, 52% for two steps; confirmed by Mass.
-0086: Step 7
Figure imgf000084_0001
[0346] To a solution of 4-bromo butyric acid, dissolved in DCM (400 ml), cooled to below 0°C was added to 1.5 eq. EDC.HC1, 2 eq. Et3N, and DMAP sequentially under nitrogen atmosphere with 10-minute intervals. To this resulting solution 20 g (Z)-non-2-en- l-ol was added, by dissolving in 100 ml of DCM, using additional funnel, and stirred at room temperature for 24 hours under nitrogen atmosphere.
[0347] Progress of the reaction was monitored by TLC (10% EtOAc in hexane; Rf: 0.7). Reaction mass was quenched with water (300 ml) and then organic layer was separated. Aqueous layer was washed with DCM (2 x 150 ml). Combined organic layer was concentrated under reduced pressure. The resulting crude was washed with saturated NaHCC solution (200 ml) and then extracted with EtOAc (150 ml). Organic layer was separated and concentrated under reduced pressure. Crude compound was subjected to column chromatography (60-120 mesh silica gel) using 5% EtOAc/hexane. Alcohol was recovered. Quantity produced, 18 g; yield, 55%.
-0086: Step 8
Figure imgf000084_0002
[0349] To a solution of 4.0 g tridecan-7-yl 4-aminobutanoate, 1 eq. (Z)-non-2-en-l- yl 4-bromobutanoate in 90 ml ACN, 1.4 eq. potassium carbonate was added and the resulting mixture was refluxed at 90°C for 4 hours under nitrogen atmosphere.
[0350] Progress of the reaction was monitored by TLC (10% MeOH in CHCh; Rf: 0.5). Reaction mass was filtered, washed with ACN (20 ml), and the filtrate concentrated under reduced pressure. Crude compound was subjected to column chromatography (100- 200 mesh silica gel) using 15% EtOAc/hexane. Starting materials, amine and bromo compounds, were recovered. Quantity produced, 2.2 g; yield, 30%; confirmed by Mass.
[0351] ATX-0086: Step 9
Figure imgf000085_0001
[0352] To a solution of 2.2 g tridecan-7-yl (Z)-4-((4-(non-2-en-l-yloxy)-4- oxobutyl) amino) butanoate, dissolved in 25 ml dry DCM, was added 3 eq. triethylamine and triphosgene with 5 minutes interval at 0°C under nitrogen atmosphere. The resulting solution was stirred at room temperature, under nitrogen atmosphere for 1 hour. The resulting reaction mass was concentrated under reduced pressure and kept under nitrogen atmosphere.
[0353] To 7 eq. sodium hydride dissolved in dry THF (100 ml), in a two neck 100 ml round bottom flask stirred at 0°C under nitrogen atmosphere, was added 3.5 eq. 2- (dimethylamino)propane-l -thiol hydrochloride and kept stirring for 5 minutes under nitrogen atmosphere. To this resulting solution the above carbamoyl chloride, dissolved in THF (100 ml), was added via syringe slowly for about 10 minutes. The resulting solution was stirred at 0°C to room temperature overnight under nitrogen atmosphere.
[0354] Progress of the reaction was monitored by TLC (60% EtOAC/hexane; Rf: 0.5; PMA charring). Reaction mass was quenched with saturated NH4CI solution (75 ml) and then EtOAc (150 ml) was added. Organic layer was separated and the aqueous layer was washed with EtOAc (3 x 40 ml). Combined organic layer was concentrated and the resulting crude was subjected to column chromatography.
[0355] The first purification was done using silica gel (60-120 mesh) of crude compound was adsorbed on 60 g of silica gel and poured onto 500 g of silica gel taken in the column. Compound was eluted at 35% EtOAc/hexane. The second purification was done using neutral alumina with HPLC grade solvents. Crude compound was adsorbed on 18 g of neutral alumina and the resulting was poured onto 130 g of neutral alumina taken in the column. Compound was eluted at 10% EtOAc/hexane. Quantity produced, 1.2 g; yield, 43%; confirmed by ¾ NMR, HPLC, and Mass.
[0356] ATX-0086 / RL-48A: Ή-ΝΜΙί (PPM, 500 MHz, CDCh): δ = 5.64 (m, 1), 5.51 (m, 10, 4.87 (m, 1), 4.63 (d, J = 7.0, 2), 3.30-3.44 (4), 3.02 (t, J = 7.0, 2), 2.52 (t, J = 7.0, 2), 2.26-2.36 (4), 2.27 (s, 6), 2.09 (m, 2), 1.82-1.96 (4), 1.46-1.54 (4), 1.21-1.40 (24), 0.84- 0.91 (9).
Example 8: Synthesis of ATX-0087
[0357] FIG. 7 shows the synthetic pathway of ATX-0087 that involves nine steps.
[0358] -0087: Step 1
Figure imgf000086_0001
[0359] In a 2 liter, two neck round bottom flask, 20 g octanoic acid dissolved in DCM (200 ml) was taken and then added 1.5 eq. oxalyl chloride slowly at 0°C, stirring under nitrogen atmosphere. The resulting reaction mixture was stirred at room temperature for 2 hours. In a separate 2 liter, two neck round bottom flask, to 2 eq. Ν,Ο- dimethylhydroxylamine hydrochloride in DCM (200 ml), was added 3 eq. trimethylamine using additional funnel, stirred at 0°C. To this resulting solution, the above acid chloride, after concentration under reduced pressure, was added under nitrogen atmosphere by dissolving in DCM (150 ml), dropwise using addition funnel for 20 minutes. The resulting reaction solution was stirred at room temperature for 3 hours under nitrogen atmosphere.
[0360] Progress of the reaction was monitored by TLC (20% EtOAc/hexane; Rf 0.5). Reaction mass was diluted with water (250 ml). Organic layer was separated and the aqueous layer was washed with DCM (3 x 100 ml). Combined organic layer was concentrated under reduced pressure. Crude compound was subjected to column
chromatography using (60-120 mesh silica gel; 10% EtOAc/hexane). Quantity produced, 20 g; yield, 84%. [0361] ATX-0087: Step 2
Figure imgf000087_0001
[0362] To a solution of hexyl magnesium bromide (1.5 eq.) in THF (100 ml), taken in a 1 liter, two neck round bottom flask, stirred at 0°C under nitrogen atmosphere, was added 20 g N-methoxy-N-methyloctanamide (1 eq.) solution (dissolved in 200 ml THF) and the resulting reaction mixture was stirred at room temperature for 4 hours.
[0363] Progress of the reaction was monitored by TLC (10% EtOAc in hexane; Rf: 0.7). Reaction mass was quenched with saturated NH4CI solution (250 ml) and then EtOAc (350 ml) was added. The organic layer was separated and the aqueous layer was washed with EtOAc (2 x 100 ml). Combined organic layers were concentrated under reduced pressure. Crude compound was subjected to column chromatography using (60-120 mesh silica gel; 2% EtOAc/hexane). Quantity produced, 25 g; yield, 65%.
[0364] ATX-0087: Step 3
Figure imgf000087_0002
[0365] To a solution of 25 g tridecane-7-one (1 eq) dissolved in MeOH/THF, 1.5 eq. sodium borohydride was added at 0°C and the resulting solution was stirred at room temperature for 1 hour.
[0366] Progress of the reaction was monitored by TLC (10% EtOAc in hexane; Rf: 0.5). Reaction mass was quenched with saturated NH4CI solution (75 ml). Solvent was removed under reduced pressure and the resulting crude was portioned between EtOAc(150 ml) and water (100 ml). Organic layer was separated and the aqueous layer was washed with EtOAc (3 x 100 ml). Combined organic layers were concentrated under reduced pressure to get white solid. Quantity produced, 22 g; yield, 90%.
[0367] ATX-0087: Step 4
Figure imgf000087_0003
[0368] To a solution of 50 g 4-aminobutanoic acid dissolved in 350 ml THF, 490 ml aqueous 1 N NaOH solution was added at 0°C, followed by 140 ml Boc anhydride, sequentially using additional funnel, over a period of 15 minutes. The resulting solution was stirred at room temperature for 4 hours. [0369] Progress of the reaction was monitored by TLC (10% MeOH in CHCh; Rf: 0.5). Reaction mass was quenched with 5% HC1 (250 ml) and then EtOAc (300 ml) was added. Organic layer was separated and the aqueous layer was washed with EtOAc (3 x 150 ml). Combined organic layer was concentrated under reduced pressure to obtain a gummy liquid. Quantity produced, 80 g; yield, 81%.
[ -0087: Step 5
Figure imgf000088_0001
[0371] To a solution of 17 g 4-((tert-butoxycarbonyl) amino)butanoic acid, dissolved in DCM (250 ml), cooled to below 0°C was added 1.3 eq. EDC.HC1, Et3N, and 4- dimethylaminopyridine (DMAP), sequentially under nitrogen atmosphere with 10 minutes interval. To this resulting solution, 1 eq. tridecane-7-ol was added at the same temperature, by dissolving in DCM (150 ml), using additional funnel, and stirred at room temperature for 24 hours under nitrogen atmosphere.
[0372] Progress of the reaction was monitored by TLC (10% EtOAc in hexane; Rf: 0.5). Reaction mass was quenched with water (150 ml) and then organic layer was separated. Aqueous layer was washed with DCM (2 x 100 ml). Combined organic layer was concentrated under reduced pressure. The resulting crude was washed with saturated NaHC03 solution (150 ml) and then EtOAc (200 ml) was added. Organic layer was separated, concentrated under reduced pressure, and proceeded to next step with crude.
Quantity produced, 15 g (crude; required compound and alcohol).
-0087: Step 6
Figure imgf000088_0002
[0374] To a solution of 15.0 g pentadecan-8-yl 4-((tert- butoxycarbonyl)amino)butanoate dissolved in 80 ml DCM, was added 10 eq. TFA at 0°C and stirred at room temperature for 3 hours under nitrogen atmosphere.
[0375] Progress of the reaction was monitored by TLC (10% MeOH in CHCh; Rf: 0.3). Reaction mass was concentrated under reduced pressure. The resulting crude was washed with a saturated NaHC03 solution (300 ml) and then extracted with EtOAc (2 x 200 ml). The organic layer was separated and concentrated under reduced pressure. Crude compound was subjected to column chromatography using (60-120 mesh silica gel; 4% MeOH/CHCh and lmL of triethylamine), and alcohol was recovered. Quantity produced, 7 g; yield, 24% for two steps; confirmed by Mass.
[0376] ATX-0087: Step 7
HC
Figure imgf000089_0001
[0377] To a solution of 4-bromo butyric acid, dissolved in DCM (400 ml), cooled to below 0°C was added to 1.5 eq. EDC.HC1, 2 eq. Et3N, and DMAP sequentially under nitrogen atmosphere with 10-minute intervals. To this resulting solution 20 g (Z)-non-2-en- l-ol was added, by dissolving in 100 ml of DCM, using additional funnel, and stirred at room temperature for 24 hours under nitrogen atmosphere.
[0378] Progress of the reaction was monitored by TLC (10% EtOAc in hexane; Rf: 0.7). Reaction mass was quenched with water (300 ml) and then organic layer was separated. Aqueous layer was washed with DCM (2 x 150 ml). Combined organic layer was concentrated under reduced pressure. The resulting crude was washed with saturated NaHCC solution (200 ml) and then extracted with EtOAc (150 ml). Organic layer was separated and concentrated under reduced pressure. Crude compound was subjected to column chromatography (60-120 mesh silica gel) using 5% EtOAc/hexane. Alcohol was recovered. Quantity produced, 19 g; yield, 55%.
Figure imgf000089_0002
[0380] To a solution of 4.0 g tridecan-7-yl 4-aminobutanoate, 1 eq. (Z)-non-2-en-l- yl 4-bromobutanoate in 90 ml ACN, 1.4 eq. potassium carbonate was added and the resulting mixture was refluxed at 90°C for 4 hours under nitrogen atmosphere.
[0381] Progress of the reaction was monitored by TLC (10% MeOH in CHCh; Rf: 0.5). Reaction mass was filtered, washed with ACN (20 ml), and the filtrate concentrated under reduced pressure. Crude compound was subjected to column chromatography (100- 200 mesh silica gel) using 15% EtOAc/hexane. Starting materials, amine and bromo compounds, were recovered. Quantity produced, 2.2 g; yield, 30%; confirmed by Mass.
[0382] ATX-0087: Step 9
Figure imgf000090_0001
[0383] To a solution of 2.2 g tridecan-7-yl (Z)-4-((4-(non-2-en-l-yloxy)-4- oxobutyl) amino) butanoate, dissolved in 25 ml dry DCM, was added 3 eq. triethylamine and triphosgene with 5 minutes interval at 0°C under nitrogen atmosphere. The resulting solution was stirred at room temperature, under nitrogen atmosphere for 1 hour. The resulting reaction mass was concentrated under reduced pressure and kept under nitrogen atmosphere.
[0384] To 7 eq. sodium hydride dissolved in dry THF (100 ml), in a two neck 100 ml round bottom flask stirred at 0°C under nitrogen atmosphere, was added 3.5 eq. 2- (dimethylamino)propane-l -thiol hydrochloride and kept stirring for 5 minutes under nitrogen atmosphere. To this resulting solution the above carbamoyl chloride, dissolved in THF (100 ml), was added via syringe slowly for about 10 minutes. The resulting solution was stirred at 0°C to room temperature overnight under nitrogen atmosphere.
[0385] Progress of the reaction was monitored by TLC (60% EtOAc/hexane; Rf 0.5; PMA charring). Reaction mass was quenched with saturated NH4CI solution (75 ml) and then EtOAc (150 ml) was added. Organic layer was separated and the aqueous layer was washed with EtOAc (3 x 40 ml). Combined organic layer was concentrated and the resulting crude was subjected to column chromatography. [0386] The first purification was done using silica gel (60-120 mesh) of crude compound was adsorbed on 60 g of silica gel and poured onto 500 g of silica gel taken in the column. Compound was eluted at 35% EtOAc/hexane. The second purification was done using neutral alumina with HPLC grade solvents. Crude compound was adsorbed on 18 g of neutral alumina and the resulting was poured onto 130 g of neutral alumina taken in the column. Compound was eluted at 10% EtOAc/hexane. Quantity produced, 1.2 g; yield, 43%; confirmed by ¾ NMR, HPLC, and Mass.
[0387] ATX-0087 / RL-48C: Ή-ΝΜΙί (PPM, 500 MHz, CDCh): δ = 5.64 (m, 1), 5.52 (m, 1), 4.87 (m, 1), 4.63 (d, J=7.0, 2), 3.30-3.44 (4), 3.02 (t, J = 7.0, 2), 2.52 (t, J = 7.0, 2), 2.26-2.36 (4), 2.27 (s, 6), 2.09 (m, 2), 1.83-1.96 (4), 1.46-1.54 (4), 1.21-1.40 (32), 0.85- 0.90 (9).
Example 9: Synthesis of ATX-0088
[0388] FIG. 8 shows the synthetic pathway of ATX-0088 that is described further as follows.
[0389] -0088: Step 1
Figure imgf000091_0001
[0390] In 500 ml two neck round bottom flask under N2 atmosphere, 25 g 8- bromooctanoic acid (1 eq.) dissolved in 200 ml of DCM was taken and then added slowly to oxalyl chloride, 1.5 eq., at 0°C, stirring under nitrogen atmosphere. The resulting reaction mixture was stirred at room temperature for 2 hours.
[0391] In a separate 1 liter two neck round bottom flask, 2 eq. Ν,Ο- dimethylhydroxylamine hydrochloride in 300 ml DCM was added 3 eq. trimethylamine and stirred at 0°C. To this resulting solution, the above acid chloride was added after concentration under reduced pressure, by dissolving in 500 ml DCM, dropwise using addition funnel for 15 minutes. The resulting reaction solution was stirred at room temperature for 3 hours under nitrogen atmosphere.
[0392] Progress of the reaction was monitored by TLC (20% EtOAc/hexane; Rf 0.5). Reaction mass was diluted with water (300 ml). Organic layer was separated and the aqueous layer was washed with DCM (2 x 100 ml). Combined organic layer was concentrated under reduced pressure. [0393] Crude compound was subjected to column chromatography using (60-120 mesh silica gel; 10% EtOAC/hexane). Quantity produced, 21 g; yield, 66%.
[0394] ATX-0088: Step 2
Figure imgf000092_0001
[0395] To a solution of 1.3 eq. octyl magnesium bromide in THF (100 ml), stirred at 0°C under nitrogen atmosphere, was added 20 g N-methoxy-N-methyloctanamide in 100 ml THF and the resulting reaction mixture was stirred at room temperature for 4 hours.
[0396] Progress of the reaction was monitored by TLC (10% EtOAc/hexane; Rf: 0.7). Reaction mass was quenched with saturated NH4CI solution (100 ml). The organic layer was separated and the aqueous layer was washed with EtOAc (2 x 100 ml). Combined organic layer was concentrated under reduced pressure.
[0397] Crude compound was subjected to column chromatography using (60-120 mesh silica gel; 2% ethyl acetate/hexane). Quantity yield was 17.4 g; 68%.
[0398] -0088: Step 3
Figure imgf000092_0002
[0399] To a solution of 17 g hexadecan-7-one (1 eq.) dissolved in 135 ml
MeOH/THF, 1.5 eq. sodium borohydride was added at 0°C and the resulting solution was stirred at room temperature for 1 hour.
[0400] Progress of the reaction was monitored by TLC (10% EtOAc/hexane; Rf: 0.5). Reaction mass was quenched with saturated NH4CI solution (50 ml). Methanol was reduced under reduced pressure. The resulting crude was portioned between EtOAc (200 ml) and water. Organic layer was separated and the aqueous layer was washed with EtOAc (2 x 80 ml). Combined organic layer was concentrated under reduced pressure to obtain a white solid. Quantity produced, 14.5 g; yield, 85%.
[0401] ATX-0088: Step 4
HO^^\^ (Boc)?Q HC ^ ^. .. Boc
N
O o H
[0402] To a solution of 50 g 4-aminobutanoic acid dissolved in 350 ml THF, 490 ml of aqueous IN NaOH solution was added at 0°C, followed by 140 ml Boc anhydride, using a funnel. The resulting solution was stirred at room temperature for 4 hours. [0403] Progress of the reaction was monitored by TLC (10% MeOH/CHCb; Rf: 0.5). Reaction mass was quenched with 5% HC1 (100 ml) and then EtOAc (200 ml) was added. The organic layer was separated and the aqueous layer was washed with EtOAc (2 x 100 ml). Combined organic layer was concentrated under reduced pressure to obtain a gummy liquid. Quantity produced, 80 g; yield, 81%.
[0404 -0088: Step 5
Figure imgf000093_0001
[0405] To a solution of 1 eq. 4-((tert-butoxycarbonyl)amino)butanoic acid dissolved in DCM (200 ml), cooled to below 0°C was added 3 eq. EDC.HCl, Et3N (3 eq.), and DMAP (0.1 eq.) sequentially with 10 minutes interval. To this resulting solution alcohol was added, by dissolving in DCM, using additional funnel, and stirred at room temperature for 24 hours under nitrogen atmosphere.
[0406] Progress of the reaction was monitored by TLC (10% EtOAc/hexane; Rf: 0.5). Reaction mass was quenched with water (100 ml) and the organic layer was separated. The aqueous layer was washed with DCM (2 x 50 ml). Combined organic layer was concentrated under reduced pressure. The resulting crude was washed with saturated NaHCCb solution and EtOAc (100 ml) was added. The organic layer was separated and concentrated under reduced pressure, and proceeded to next step with crude. Quantity produced, 19 g (crude).
-0088: Step 6
Figure imgf000093_0002
[0408] To a solution of 19 g hexadecan-7-yl 4-((tert- butoxycarbonyl)amino)butanoate (1 eq.) dissolved in 140 ml DCM, was added 10 eq. TFA at 0°C and stirred at room temperature for 3 hours under nitrogen atmosphere.
[0409] Progress of the reaction was monitored by TLC (10% MeOH/CHCb; Rf: 0.3). Reaction mass was concentrated under reduced pressure. The resulting crude was washed with saturated NaHCCb solution (100 ml) and EtOAc (100 ml) was added. The organic layer was separated and concentrated under reduced pressure. [0410] Crude compound was subjected to column chromatography using (60-120 mesh silica gel; 4% MeOH/CHCh) and alcohol was recovered. Quantity produced, 9.4 g; yield, 50% for two steps; confirmed by Mass.
[0411] ATX-0088: Step 7
Figure imgf000094_0001
[0412] To a solution of 30 g 4-bromo butyric acid (1 eq.) dissolved in DCM (500 ml), cooled to 0°C was added 1.5 eq. EDC.HC1, 3 eq. Et3N, and 0.1 eq. DMAP sequentially with 10 minutes interval. To this resulting solution 0.7 eq. (Z)-non-2-en-l-ol was added, by dissolving in 100 ml DCM, using a funnel, and stirred at room temperature for 24 hours under nitrogen atmosphere.
[0413] Progress of the reaction was monitored by TLC (10% EtOAc/hexane; Rf: 0.7). Reaction mass was quenched with water (100 ml) and then organic layer was separated. Aqueous layer was washed with DCM (2 x 100 ml). Combined organic layer was concentrated under reduced pressure. The resulting crude was washed with saturated NaHCC solution and EtOAc (150 ml) was added. The organic layer was separated and concentrated under reduced pressure.
[0414] Crude compound was subjected to column chromatography using (60-120 mesh silica gel; 5% EtOAc/hexane). Quantity produced, 27 g; yield, 51%; confirmed by ¾ NMR.
-0088: Step 8
Figure imgf000094_0002
[0416] To a solution of 6 g hexadecan-8-yl 4-aminobutanoate (1 eq.), 1 eq.5 (Z)- non-2-en-l-yl 4-bromobutanoate in ACN (70 ml), 1.2 eq. potassium carbonate was added and the resulting was refluxed at 90°C for 3 hours under nitrogen atmosphere.
[0417] Progress of the reaction was monitored by TLC (10% MeOH/CHCb; Rf: 0.5). Reaction mass was filtered and the filtrate concentrated under reduced pressure. [0418] Crude compound was subjected to column chromatography using (100-200 mesh silica gel; 15% EtOAc/hexane). Quantity produced, 4.5g; yield, 44%; confirmed by Mass.
[0419] ATX-0088: Step 9
Figure imgf000095_0001
[0420] To a solution of 4.4 g (Z)-non-2-en-l-yl 4-((4-oxo-4-(tetradecan-7- yloxy)butyl)amino)butanoate (1 eq.) dissolved in 30 ml dry DCM, was added 0.83 ml trimethylamine (3 eq.) and 418 mg triphosgene (0.5 eq.) with 5 minutes interval, at 0°C under nitrogen atmosphere. The resulting solution was stirred at room temperature, under nitrogen atmosphere for 1 hour. The resulting reaction mass was concentrated under reduced pressure and kept under nitrogen atmosphere.
[0421] To 192 mg sodium hydride (10 eq.) dissolved in dry THF (25 ml), in a two neck 100 ml round bottom flask, was added 564 mg 2-(dimethylamino)propane-l -thiol hydrochloride (5 eq.) at 0°C and kept stirring for 5 minutes under nitrogen atmosphere. To this resulting solution the above carbamoyl chloride, dissolved in THF (35 ml), was added via syringe slowly for about 10 minutes. The resulting solution was stirred at room temperature for 4 hours under nitrogen atmosphere.
[0422] Progress of the reaction was monitored by TLC (60% EtOAC/hexane; Rf: 0.5; PMA charring). Reaction mass was quenched with saturated NH4CI solution (30 ml) and then EtOAc (100 ml) was added. The organic layer was separated and the aqueous layer was washed with EtOAc (2 x 50 ml). Combined organic layer was concentrated and the resulting crude was subjected to column chromatography. [0423] A first purification was done using silica gel (60-120 mesh). 5.0 g of crude compound was adsorbed on 9 g of silica gel and poured onto 90 g of silica gel taken in the column. Compound was eluted at 35% EtOAc/hexane. A second purification was done using neutral alumina with HPLC grade solvents. Crude compound, 1.5 g, was adsorbed on 4 g of neutral alumina and the resulting was poured onto 40 g of neutral alumina taken in the column. Compound was eluted at 10% EtOAc/hexane. Quantity produced, 1.2 g; yield, 21%; confirmed by ¾ NMR; HPLC; Mass.
[0424] ATX-0088 / RL-48D: ^-NMR (PPM, 500 MHz, CDCh): δ = 5.64 (m, 1), 5.51 (m, 1), 4.87 (m, 1), 4.63 (d, J = 7.0, 2), 3.30-3.44 (4), 2.90 (t, J = 7.0, 2), 2.46-2.55 (6), 2.26-2.37 (4), 2.09 (m, 2), 1.71-1.80 (4), 1.46-1.55 (4), 1.21-1.41 (32), 1.01 (t, J = 7.0, 6), 00.85-0.91 (9).
Example 10: Synthesis of ATX-0083
[0425] FIG. 9 shows the synthetic pathway of ATX-0083 that is described further as follows.
[0426] A -0083: Step 1
Figure imgf000096_0001
[0427] In a 500 ml single neck round bottom flask, 50 g octanoic acid (1 eq.) dissolved in of DCM (200 ml) was taken and then added 44.6 ml oxalyl chloride (1.5 eq.) slowly at 0°C, via additional funnel, stirring under nitrogen atmosphere and then added 1 ml DMF (catalytic). The resulting reaction mixture was stirred at room temperature for 2 hours.
[0428] In a separate 2 lit two neck round bottom flask to 67.4 g Ν,Ο- dimethylhydroxylamine hydrochloride (2 eq.) in DCM (300 ml), was added 144 ml triethylamine (3 eq.) using additional funnel, stirred at 0°C. To this resulting solution, the above acid chloride, after concentration under reduced pressure, was added under nitrogen atmosphere by dissolving in DCM (350 ml), dropwise using addition funnel for 20 minutes. The resulting reaction solution was stirred at room temperature for 3 hours under nitrogen atmosphere.
[0429] Progress of the reaction was monitored by TLC (20% EtOAc/hexane; Rf 0.5; PMA charring). Reaction mass was diluted with water (300 ml). Organic layer was separated and the aqueous layer was washed with DCM (3 x 100 ml). Combined organic layer was dried over anhydrous Na2S04 and concentrated under reduced pressure. [0430] Crude compound was subjected to column chromatography (60-120 mesh silica gel) using 10% EtOAc/hexane. Quantity produced, 55.0 g; yield, 84%
[0431] -0083: Step 2
Figure imgf000097_0001
[0432] To a solution of 55 g heptyl magnesium bromide (1 eq.) in ether, taken in a 2 1 two neck round bottom flask, stirred at 0°C under nitrogen atmosphere, was added 89.6 g N- methoxy-N-methyloctanamide solution (1.5 eq.) dissolved in 400 ml of dry ether and the resulting reaction solution was stirred at room temperature for 4 hours.
[0433] Progress of the reaction was monitored by TLC (10% EtOAc in hexane; Rf: 0.7; PMA charring). Reaction mass was quenched with saturated NH4CI solution (250 ml). Organic layer was separated and the aqueous layer was washed with ether (2 x 100 ml). Combined organic layer was dried over anh.Na2S04 and concentrated under reduced pressure.
[0434] Crude compound was subjected to column chromatography (60-120 mesh silica gel) using 2% EtOAc/hexane. Quantity produced, 50.0g; yield, 75%.
[0435] -0083: Step 3
Figure imgf000097_0002
[0436] To a solution of 50 g pentadecan-8-one (1 eq.) dissolved in 290 ml
MeOH/THF, 12.5 g sodium borohydride (1.5 eq.) was added at 0°C and the resulting solution was stirred at room temperature for 2 hours.
[0437] Progress of the reaction was monitored by TLC (10% EtOAc in hexane; Rf: 0.5; PMA charring). Reaction mass was quenched with saturated NH4CI solution (80 ml). Solvent was removed under reduced pressure and the resulting crude was partitioned between EtOAc (250 ml) and water (100 ml). Organic layer was separated and the aqueous layer was washed with EtOAc (3 x 80 ml). Combined organic layer was dried over anh.Na2S04 concentrated under reduced pressure and dried under vacuum to get white solid. Quantity produced, 46.0 g; yield, 90%.
[0438] ATX-0083: Step 4
Figure imgf000097_0003
[0439] To a solution of 50 g 4-aminobutanoic acid (1 eq.) dissolved in THF, 490 ml 1 N aqueous NaOH solution (1 eq.) was added at 0°C, followed by 140 ml Boc anhydride (1.3 eq.), sequentially using additional funnel, over a period of 15 minutes. The resulting solution was stirred at room temperature for 4 hours.
[0440] Progress of the reaction was monitored by TLC (10% MeOH in CHCh; Rf: 0.5). Reaction mass was quenched with 5% HC1 (350 ml) and then EtOAc (300 ml) was added. Organic layer was separated and the aqueous layer was washed with EtOAc (3 x 150 ml). Combined organic layer was dried over anhydrous Na2S04 and concentrated under reduced pressure to get gummy liquid. Quantity produced, 77.0 g; yield, 78%.
[0441] -0083: Step 5
Figure imgf000098_0001
[0442] Synthesis was done in 4 batches. In each, to a solution of 23 g 4-((tert- butoxycarbonyl) amino)butanoic acid (1 eq.) in DCM (400 ml), cooled to below 0°C, were added 32.3 g EDC.HC1 (1.5 eq.), 47 ml Et3N (3 eq.), and 1.3 g DMAP (0.1 eq.) sequentially under nitrogen atmosphere with 10 min interval. To this resulting solution 20 g pentadecan- 8-ol (0.77 eq.) was added, by dissolving in DCM (200 ml), using additional funnel, and stirred at room temperature for 24 hours under nitrogen atmosphere.
[0443] Progress of the reaction was monitored by TLC (10% EtOAc in hexane; Rf: 0.4). Reaction mass was quenched with water (250 ml) and then organic layer was separated. Aqueous layer was washed with DCM (2 x 100 ml). Combined organic layer was concentrated under reduced pressure. To this resulting crude was washed saturated NaHC03 solution (150 ml) and EtOAc (250 ml) was added. Organic layer was separated, dried over anh.Na2S04 and concentrated under reduced pressure and then proceeded to next step with crude. Quantity produced, 105 g (crude; required compound and alcohol)
[0444] ATX-0083: Step 6
Figure imgf000098_0002
[0445] To a solution of 105 g pentadecan-8-yl 4-((tert- butoxycarbonyl)amino)butanoate (1 eq.) dissolved in 450 ml DCM, was added 194 ml TFA (10 eq.) at 0°C and stirred at room temperature for 3 hours under nitrogen atmosphere. [0446] Progress of the reaction was monitored by TLC (10% MeOH in CHCh; Rf:
0.3).
[0447] Reaction mass was concentrated under reduced pressure. The resulting crude was stirred with saturated NaHCCbsolution (200 ml) for 10 minutes and then EtOAc (300 ml). Organic layer was separated and the aqueous layer was washed with EtOAc (2 x 100 ml). Combined organic layer was dried over anhydrous Na2S04 and concentrated under reduced pressure.
[0448] Crude compound was subjected to column chromatography (silica gel 60- 120 mesh) using 4% MeOH/CHCh and 1ml of triethylamine. Quantity produced, 60.0 g for two steps; yield, 54%.
[0 -0083: Step 7
Figure imgf000099_0001
[0450] Reaction was done in two batches, In each, to a solution of 20 g 6- bromohexanoic acid (1 eq.) dissolved in DCM (300 ml), cooled to below 0°C was added 29.3 g EDC.HC1 (1.5 eq.), 42.8 ml Et3N (3 eq.), and 1.2 g DMAP (0.1 eq.) sequentially under nitrogen atmosphere with 10-minute intervals. To this resulting solution 14.5 g (Z)-non-2-en- l-ol (1 eq.) was added (by dissolving in 100 ml of DCM) using additional funnel, and stirred at room temperature for 24 hours under nitrogen atmosphere.
[0451] Progress of the reaction was monitored by TLC (10% EtOAc in hexane; Rf: 0.7). Reaction mass was quenched with water (200 ml) and then organic layer was separated. Aqueous layer was washed with DCM (2 x 100 ml). Combined organic layer was concentrated under reduced pressure. The resulting crude was washed with saturated NaHC03 solution (150 ml) and then extracted with EtOAc (2 x 150 ml). Organic layer was separated, dried over anhydrous Na2S04 and concentrated under reduced pressure.
[0452] Crude compound was subjected to column chromatography (60-120 mesh silica gel) using 4% EtOAc/hexane. Alcohol reactant was recovered. Quantity produced, 36.0 g; yield, 55%. [0453] ATX-0083: Step 8
Figure imgf000100_0001
[0454] The reaction was done in six batches. In each, to a solution of 10 g pentadecan-8-yl 4-aminobutanoate (Int 6, 1 eq.), 10.1 g (Z)-non-2-en-l-yl 6-bromohexanoate (Int 7, 1 eq.) in 120 ml ACN, 6.1 g anhydrous potassium carbonate (1.4 eq.) was added and the resulting mixture was refluxed at 90°C for 4 hours under nitrogen atmosphere.
[0455] Progress of the reaction was monitored by TLC (10% MeOH in CHCh; Rf: 0.5). Reaction mass was filtered, washed with ACN (2 x 20 ml), and the filtrate concentrated under reduced pressure.
[0456] Crude compound was subjected to column chromatography (silica gel 100- 200 mesh) using 20-80% EtOAc/hexane. Starting materials were recovered. Quantity produced, 36.9 g; yield, 35%.
[0457] ATX-0083: Step 9
Figure imgf000100_0002
[0458] The reaction was done in three batches. In each, to a solution of 10 g (Z)- non-2-en-l-yl 6-((4-oxo-4-(pentadecan-8-yloxy)butyl)amino)hexanoate (1 eq.) dissolved in 100 ml dry DCM, was added 7.5 ml triethylamine (3 eq.) and 2.68 g triphosgene (0.5 eq.) with 5 minute intervals at 0°C under nitrogen atmosphere. The resulting solution was stirred at room temperature, under nitrogen atmosphere for 1 hour. The resulting reaction mass was concentrated under reduced pressure and kept under nitrogen atmosphere.
[0459] To a suspension of 3 g sodium hydride (7 eq.) in dry THF (100 ml), in a 2 neck 500 ml RB flask stirred at 0°C under nitrogen atmosphere, was added 8.9 g 2- (dimethylamino)ethane-l -thiol hydrochloride (3.5 eq.) and kept stirring for 5 minutes under nitrogen atmosphere. To this resulting solution the above carbamoyl chloride, dissolved in dry THF (200 ml), was added via syringe slowly for about 10 minutes. The resulting solution was stirred at room temperature overnight under nitrogen atmosphere.
[0460] Progress of the reaction was monitored by TLC (10% EtOAc/hexane; Rf: 0.5; PMA charring). Reaction mass was quenched with saturated NH4CI solution (100 ml) and then EtOAc (350 ml) was added. Organic layer was separated and the aqueous layer was washed with EtOAc (2 x 80 ml). Combined organic layer was dried over anhydrous Na2S04 and concentrated under reduced pressure.
[0461] A first purification was done using neutral alumina. Crude compound, dissolved in hexane, was loaded at the top of neutral alumina (700 g loaded in the column). Compound was eluted at 8-10% EtOAc/hexane. A second purification was done using silica gel (100-200 mesh). Compound, dissolved in hexane, was loaded at the top of silica gel (500g loaded in the column). Compound was eluted at 20-25% EtOAc/hexane. Final Compound (dissolved in hexane) was subjected to charcoal treatment (200 mg/g) and filtered through celite bed (after stirred for 20 minutes), and then passed through syringe end membrane filter (PTFE; 0.2 micron, 25 mm diameter). The resulting filtrate was concentrated under reduced pressure. Quantity produced, 15.5 g; yield, 41%.
[0462] ATX-0083 / RL-47B: !H-NMR (PPM, 500 MHz, CDCh): δ = 5.64 (m, 1), 5.52 (m, 1), 4.87 (m, 1), 4.62 (d, J = 7.0, 2), 3.24-3.42 (4), 3.02 (t, J = 7.0, 2), 2.53 (t, J = 7.0, 2), 2.26-2.34 (4), 2.26 (s, 6), 2.10 (m, 2), 1.45-1.70 (6), 1.20-1.41 (34), 0.84-0.92 (9).
Example 11: Synthesis of ATX-0084
[0463] FIG. 10 shows the synthetic pathway of ATX-0084 that is described further as follows.
[0464] A -0084: Step 1
Figure imgf000101_0001
[0465] In a 500 ml single neck round bottom flask, 30 g heptanoic acid (1 eq.) dissolved in of DCM (200 ml) was taken and then added 26.7 g oxalyl chloride (1.5 eq.) slowly at 0°C, stirring under nitrogen atmosphere and then added 1 ml DMF (catalytic). The resulting reaction mixture was stirred at room temperature for 2 hours.
[0466] In a separate 1 1 two neck round bottom flask, to 40.5 g Ν,Ο- dimethylhydroxylamine hydrochloride (2 eq.), in DCM (250 ml), was added 86.6 ml trimethylamine (3 eq.) using additional funnel, stirred at 0°C. To this resulting solution, the above acid chloride, after concentration under reduced pressure, was added under nitrogen atmosphere by dissolving in DCM (100 ml), dropwise using addition funnel for 20 minutes. The resulting reaction solution was stirred at room temperature for 3 hours under nitrogen atmosphere.
[0467] Progress of the reaction was monitored by TLC (20% EtOAc/hexane; Rf 0.5). Reaction mass was diluted with water (250 ml). Organic layer was separated and the aqueous layer was washed with DCM (3 x 100 ml). Combined organic layer was concentrated under reduced pressure.
[0468] Crude compound was subjected to column chromatography using (60-120 silica gel) using 10% EtOAc/hexane. Quantity produced, 38.0 g; yield, 84%.
[0469] -0084: Step 2
Figure imgf000102_0001
[0470] To a solution of 8 g hexyl magnesium bromide (1 eq.) in 250 ml dry ether, taken in a 1 liter two neck round bottom flask, stirred at 0°C under nitrogen atmosphere, was added 2.3 g N-methoxy-N-methyheptanamide (0.5 eq.) dissolved in 250 ml of ether and the resulting reaction mixture was stirred at room temperature for 4 hours.
[0471] Progress of the reaction was monitored by TLC (10% EtOAc in hexane; Rf: 0.7). Reaction mass was quenched with saturated NH4CI solution (200 ml). Organic layer was separated and the aqueous layer was washed with ether (2 x 100 ml). Combined organic layer was dried over anhydrous Na2S04 and concentrated under reduced pressure.
[0472] Crude compound was subjected to column chromatography using (60-120 mesh silica gel) using 2% EtOAc/hexane. Quantity produced, 30.8g; yield, 71%.
[0473] A -0084: Step 3
Figure imgf000102_0002
[0474] To a solution of 30 g tridecan-7-one (1 eq.) dissolved in 200 ml MeOH/THF, 8.5 g sodium borohydride (0.5 eq.) was added at 0°C and the resulting solution was stirred at room temperature for 2 hours.
[0475] Progress of the reaction was monitored by TLC (10% EtOAc/hexane; Rf: 0.5). Reaction mass was quenched with saturated NH4CI solution (80 ml). Solvent was removed under reduced pressure and the resulting crude was partitioned between EtOAc (200 ml) and water (100 ml). Organic layer was separated and the aqueous layer was washed with EtOAc (2 x 70 ml). Combined organic layers were concentrated under reduced pressure to get white solid. Quantity produced, 27.2 g; yield, 90%.
[0476] -0084: Step 4
Figure imgf000103_0001
[0477] To a solution of 5 g 6-aminohexanoic acid (1 eq.), dissolved in 120 ml THF, 125 ml of IN aqueous NaOH solution was added at 0°C, followed by 34 ml Boc anhydride (1.3 eq.), sequentially using additional funnel, over a period of 15 min. The resulting solution was stirred at room temperature for 4 hours.
[0478] Progress of the reaction was monitored by TLC (10% MeOH in CHCh; Rf: 0.5). Reaction mass was quenched with 5% HC1 (100 ml) and then EtOAc (150 ml) was added. Organic layer was separated and the aqueous layer was washed with EtOAc (2 x 100 ml). Combined organic layer was concentrated under reduced pressure to get gummy liquid. Quantity produced, 22.4 g; yield, 85%.
[04 -0084: Step 5
Figure imgf000103_0002
[0480] To a solution of 10 g 6-((tert-butoxycarbonyl)amino)hexanoic acid (1 eq.) dissolved in DCM (200 ml), cooled to below 0°C was added 10.7 g EDC.HCl (1.3 eq.), 18 ml Et3N (3 eq.), and 525 mg DMAP (0.1 eq.) sequentially under nitrogen atmosphere with 10- minute interval. To this resulting solution 6 g tridecan-7-ol (Int 3, 0.7 eq.) was added at the same temperature, by dissolving in DCM (50 ml), using additional funnel, and stirred at room temperature for 24 hours under nitrogen atmosphere.
[0481] Progress of the reaction was monitored by TLC (10% EtOAc in hexane; Rf: 0.4). Reaction mass was quenched with water (150 ml) and then organic layer was separated. Aqueous layer was washed with DCM (2 x 75 ml). Combined organic layer was concentrated under reduced pressure. The resulting crude was washed with saturated NaHCC solution (100 ml) and then extracted with EtOAc (2 x 100 ml) was added. Organic layer was separated and concentrated under reduced pressure, and proceeded to next step with crude. Quantity produced, 8.5 g (crude; required compound and alcohol).
-0084: Step 6
Figure imgf000104_0001
[0483] To a solution of 10 g tridecan-7-yl 6-((tert-butoxycarbonyl)amino)hexanoate (1 eq.) dissolved in 65 ml DCM, was added 18.5 ml TFA (10 eq.) at 0°C and stirred at room temperature for 3 hours under nitrogen atmosphere.
[0484] Progress of the reaction was monitored by TLC (10% MeOH in CHCh; Rf: 0.3). Reaction mass was concentrated under reduced pressure. The resulting crude was washed with saturated NaHCC solution (100 ml) and then extracted with EtOAc (3 x 100 ml). Organic layer was separated and concentrated under reduced pressure.
[0485] Crude compound was subjected to column chromatography using (60-120 mesh silica gel; 4% MeOH/CHCh and 1 ml triethylamine), and alcohol starting material was recovered. Quantity, 4.5 g in two steps; yield, 33%.
-0084: Step 7
Figure imgf000104_0002
[0487] To a solution of 20 g 6-bromohexanoic acid (1 eq.) dissolved in DCM (300 ml), cooled to below 0°C was added 29.3 g EDC.HCl (1.5 eq.), 42.8 ml Et3N (3 eq.), and 1.2 g DMAP (0.1 eq.) sequentially under nitrogen atmosphere with 10-minute interval. To this resulting solution 14.5 g (Z)-non-2-en-l-ol (1 eq.) was added, dissolved in 100 ml of DCM, using additional funnel, and stirred at room temperature for 24 hours under nitrogen atmosphere.
[0488] Progress of the reaction was monitored by TLC (10% EtOAc in hexane; Rf:
0.7). Reaction mass was quenched with water (200 ml) and then organic layer was separated.
Aqueous layer was washed with DCM (2 x 100 ml). Combined organic layer was concentrated under reduced pressure. The resulting crude was washed with saturated
NaHC03 solution (150 ml) and then extracted with EtOAc (2 x 150 ml). Organic layer was separated, dried over anh.Na2S04 and concentrated under reduced pressure. [0489] Crude compound was subjected to column chromatography (60-120 mesh silica gel) using 4% EtOAc/hexane. Alcohol starting material was recovered. Quantity produced, 18.0 g; yield, 55%.
[0490] ATX-0084: Step 8
Figure imgf000105_0001
[0491] To a solution of 4.5 g tridecan-7-yl 6-aminohexanoate (Int 6, 1 eq.) and 4.5 g (Z)-non-2-en-l-yl 6-bromohexanoate (Int 7, 1 eq.) in 90 ml ACN, 2.7 g potassium carbonate (1.4 eq.) was added and the resulting mixture was refluxed at 90°C for 4 hour under nitrogen atmosphere.
[0492] Progress of the reaction was monitored by TLC (10% MeOH in CHCh; Rf: 0.5). Reaction mass was filtered, washed with ACN (2 x 20 ml), and the filtrate concentrated under reduced pressure.
[0493] Crude compound was subjected to column chromatography (100-200 mesh silica gel) using 20% EtOAc/hexane. Starting materials were recovered. Quantity produced, 3.0 g; yield, 37%.
-0084: Step 9
Figure imgf000105_0002
[0495] To a solution of 2.5 g (Z)-non-2-en-l-yl 6-((6-oxo-6-(tridecan-7- yloxy)hexyl)amino)hexanoate (1 eq.) dissolved in 30 ml dry DCM, was added 1.8 ml triethylamine (3 eq.) and 672 mg triphosgene (0.5 eq.) with 5 minute interval at 0°C under nitrogen atmosphere. The resulting solution was stirred at room temperature under nitrogen atmosphere for 1 hour. The resulting reaction mass was concentrated under reduced pressure and kept under nitrogen atmosphere.
[0496] To a suspension of 761 mg sodium hydride in dry THF (50 ml), in a 2 neck 250 ml round bottom flask stirred at 0°C under nitrogen atmosphere, was added 2.2 g 2- (dimethylamino)ethane-l -thiol hydrochloride (3.5 eq.) and kept stirring for 5 minutes under nitrogen atmosphere. To the resulting solution the above carbamoyl chloride, dissolved in THF (60 ml), was added via syringe slowly for about 10 minutes. The resulting solution was stirred at room temperature overnight under nitrogen atmosphere.
[0497] Progress of the reaction was monitored by TLC (10% EtOAc/hexane; Rf: 0.5; PMA charring). Reaction mass was quenched with saturated NH4CI solution (60 ml) and then EtOAc (130 ml) was added. Organic layer was separated and the aqueous layer was washed with EtOAc (3 x 40 ml). Combined organic layer was concentrated and the resulting crude was subjected to column chromatography.
[0498] A first purification was done using silica gel (100-200 mesh). 4.6 g of crude compound was adsorbed on 10.0 g of silica gel and poured onto 90.0 g of silica gel taken in the column. Compound was eluted at 50% EtOAc/hexane. A second purification was done using neutral alumina with HPLC grade solvents. 2.0 g of crude compound was adsorbed on 6.0 g of neutral alumina and the resulting was poured onto 40.0 g of neutral alumina taken in the column. Compound was eluted at 20% EtOAc/hexane. Quantity produced, 1.2 g; yield, 38 % (300 mg mixture).
[0499] ATX-0084 / RL-47C: ¾-NMR (PPM, 500 MHz, CDCh): δ = 5.64 (m, 1), 5.52 (m, 1), 4.86 (m, 1), 4.62 (d, J = 7.0, 2), 3.22-3.35 (4), 3.01 (t, J = 7.0, 2), 2.53 (t, J = 7.0, 2), 2.25-2.34 (4), 2.27 (s, 6), 2.10 (m, 2), 1.45-1-73 (10), 1.20-1.40 (30), 00.84-0.91 (9). Example 12: Synthesis of ATX-0061
[0500] FIG. 11 shows the synthetic pathway of ATX-0061 that is described further as follows
[0501] ATX-0061 Step 1
Figure imgf000106_0001
[0502] 12 g glycine ester (1 eq.) was dissolved in THF (100 ml) and cooled to below 0°C. To this solution, 24.2 ml triethylamine (1.5 eq.) and 38.11 g Boc anhydride (1.5 eq.) through an additional funnel were added sequentially.
[0503] Progress of the reaction was monitored by TLC using 50% EtOAc/hexane;
Rf: 0.4.
[0504] Reaction mass was quenched with water and EtOAc (100 ml) was added, after 16 hours. Organic layer was separated, aqueous layer was washed with EtOAc (2 x 40 ml) and combined organic layers were dried over sodium sulphate and concentrated under reduced pressure.
[0505] Crude product was subjected to 60-120 silica gel (25% EtOAc/hexane). Quantity produced, 20.8; yield, 88%.
[0506] ATX-0061: Ste 2
Figure imgf000107_0001
[0507] To a solution of 18.9 g N-Boc glycine ester (1 eq.) dissolved in THF (130 ml) was added aqueous solution of 5.85 g LiOH (1.5eq.) and the resulting solution was stirred at room temperature for 4 hours.
[0508] The reaction was monitored by TLC (60% EtOAc/hexane; Rf: 0.3), SM is absent.
[0509] Reaction mass was concentrated and crude mass was quenched with 5% HC1 (pH 3) and then extracted with EtOAc (4 x 80 ml), dried over sodium sulphate and concentrated under reduced pressure to get the compound. Quantity produced, 15g; yield, 92%; confirmed by Mass.
-0061: Step 3
Figure imgf000107_0002
[0511] To a solution of 5 g N-Boc-glycine ester (Int 1, 1 eq.) dissolved in DCM (30 ml), cooled to below 0°C was added 4.5 ml Et3N (1.2 eq.) and 6.44 g EDC.HCl (1.2 eq.). To this reaction solution 5.12 g heptaden-9-ol (0.7 eq.) in 20 ml DCM was added and stirred at room temperature overnight.
[0512] Starting material observed to be absent by TLC (10% EtOAc/hexane; Rf: 0.6). Reaction mass was diluted with saturated NaHC03 solution, organic layer was separated, aqueous layer was washed with DCM (2 x 30 ml) and dried over sodium sulphate and concentrated under reduced pressure. Proceeded to next step with crude (6.8 g; mixture of product and alcohol).
[0513] ATX-0061: Step 4
Figure imgf000108_0001
[0514] 4 g heptadecan-9-yl (tert-butoxycarbonyl)glycinate (Int 2, 1 eq.) was dissolved in DCM (40 ml) and cooled 0°C, added 7.4 ml TFA (10 eq.) and stirred at room temperature for 1 hour.
[0515] Completion of reaction was checked in 2 hour by TLC (10% EtOAc/hexane; Rf: 0.5).
[0516] Reaction mass was concentrated under reduced pressure, residual mass was washed with saturated sodium bicarbonate solution (30 ml) and extracted with EtOAc (3 x 30 ml), organic layer dried over sodium sulphate and concentrated under reduced pressure to get Int 3
[0517] Crude product was subjected to column chromatography (silica, 60-120) using 1-3% MeOH/CHCh and 2 mL of Et3N. Quantity produced, 1 g; confirmed by ¾- NMR and Mass.
[0518] ATX-0061: Step 5
Figure imgf000108_0002
[0519] To a solution of 4 g bromo acetic acid (1 eq.) dissolved in DCM (35 ml), cooled to below 0°C was added 4.7 ml Et3N (1.2 eq.) and 354 mg DMAP (0.1 eq.), followed by 13.23 g HATU (1.2 eq.). To this reaction solution 2.88 g (Z)-non-2-en-l-ol (0.7 eq.) in 20 ml of DCM was added and stirred at room temperature overnight.
[0520] Reaction was monitored by TLC (10% EtOAc/hexane; Rf: 0.7).
[0521] Reaction mass was diluted with saturated NaHCCb solution (80 ml), organic layer was separated, aqueous layer was washed with DCM (40 ml), dried over sodium sulphate and concentrated under reduced pressure. The residual mass was purified by silica gel (60-120) column chromatography (1.5% EtOAc/hexane). Quantity produced, 4 g; yield, 52%. [0522] ATX-0061: Step 6
Figure imgf000109_0001
[0523] 1 g heptadecan-9-yl glycinate (Int 3, 1 eq.) was dissolved in THF (25 ml), added 0.5 ml TEA (1.3 eq.) and 1.08 g (Z)-non-2-en-l-yl 2-bromoacetate derivative (Int 4, 1.3 eq.), and stirred at room temperature for overnight.
[0524] Progress of the reaction was monitored by TLC (10%EtOAc/hexane; Rf: 0.4). Reaction mixture was diluted with water (30 ml) and extracted with EtOAc (20 ml x 2), combined organic layer was dried over sodium sulphate and concentrated under reduced pressure.
[0525] The residual mass was purified by column (silica gel; 100-200)
chromatography (2% EtOAc/hexane). Quantity produced, 700mg; yield, 47%; confirmed by Mass.
Step 7
Figure imgf000109_0002
[0527] To a solution of 700 mg heptadecan-9-yl (Z)-(2-(non-2-en-l -yloxy)-2- oxoethyl)glycinate) (1 eq.) dissolved in 15 ml DCM, cooled to below 5°C was added 0.4 ml Et3N (3 eq.), followed by 209 mg triphosgene (0.5 eq.) portion-wise for 10 minutes.
[0528] Progress of the reaction mixture monitored by TLC, reaction was completed for 0.5 hours, reaction mass was concentrated under reduced pressure.
[0529] To a solution of 423 mg N, N-dimethyl ethanethiol hydrochloride (3 eq.) in dry THF (10 ml) and DMF (3 ml), stirred at 0°C under nitrogen atmosphere was added 144 mg sodium hydride (6 eq.). After 10 minutes, to this reaction mass was added the above solution, by dissolving in THF (15 ml). The resulting solution was stirred at room temperature for 1 hour. [0530] Completion of the reaction was observed by TLC (10% MeOH/CHCb; Rf: 0.5), after 1 hour.
[0531] Reaction mass was quenched with saturated NH4CI solution (20 ml), water (20 ml) and EtOAc (30 ml) was added. Aqueous layer was washed with EtOAc (2 x 20 ml), and the combined organic layer was washed with brine solution (20 ml). Organic layer was dried over Na2S04 and concentrated under reduced pressure.
[0532] Crude was subjected to column chromatography using silica gel (100-200) with 15% EtOAC/hexane, and then with neutral alumina withl5% EtOAc/hexane, to get pure compound. Quantity produced, 520 mg; yield, 58%; confirmed by ¾-NMR, HPLC and
Mass.
[0533] ATX-0061 / RL-42D: ^-NMR (PPM, 400MHz, CDCh): δ = 5.67 (m, 1), 5.51 (m, 1), 4.92 (m, 1), 4.70 (m, 2), 4.16-4.27 (4), 3.07 (m, 2), 2.53 (m, 2), 2.27 (s, 6), 2.10 (m, 2), 1 -47-1.57 (4), 1.19-1.40 (32), 0.83-0.92 (9).
Example 13: Synthesis of ATX-0063
[0534] FIG. 12 shows the synthetic pathway of ATX-0063 that is described further as follows.
[0535] ATX-0063: Ste 1
Figure imgf000110_0001
ethyl glycinate
[0536] 12 g glycine ester (1 eq.) was dissolved in THF (100 mL) and cooled to below 0°C. To this solution, 24.2 ml triethylamine (1.5 eq.) and 38.1 1 g Boc anhydride (1.5 eq.) through an additional funnel were added sequentially.
[0537] Progress of the reaction was monitored by TLC using 50% EtOAc/hexane;
Rf: 0.4.
[0538] Reaction mass was quenched with water and EtOAc (100 ml) was added, after 16 hours. Organic layer was separated, aqueous layer was washed with EtOAc (2 x 40 ml) and combined organic layers were dried over sodium sulphate and concentrated under reduced pressure.
[0539] Crude product was subjected to 60-120 silica gel (25% EtOAc/hexane). Quantity produced, 20.8; yield, 88%. [0540] ATX-0063: Step 2
Figure imgf000111_0001
[0541] To a solution of 18.9 g N-Boc glycine ester (1 eq.) dissolved in THF (130 ml) was added aqueous solution of 5.85 g LiOH (1.5 eq.) and the resulting solution was stirred at room temperature for 4 hours.
[0542] The reaction was monitored by TLC (60% EtOAc/hexane; Rf: 0.3), starting was absent from reaction product.
[0543] The reaction mass was concentrated and crude mass was quenched with 5% HC1 (pH 3) and then extracted with EtO Ac (4 x 80 ml), dried over sodium sulphate and concentrated under reduced pressure to get the compound. Quantity produced, 15 g; yield, 92%; confirmed by Mass.
[05 -0063: Step 3
Figure imgf000111_0002
[0545] To a solution of 5 g N-Boc-glycine ester (Int 1, 1 eq.), dissolved in DCM (50 ml), cooled to below 0°C was added 4.5 ml Et3N (1.2 eq.) and 6.4 g EDC.HC1 (1.2 eq.). To this reaction solution 3.4 g undecan-6-ol (0.7 eq.) in 20 ml of DCM was added and stirred at room temperature overnight.
[0546] Starting material observed to be absent by TLC (15% EtOAc/hexane; Rf: 0.6). Reaction mass was diluted with saturated NaHC03 solution (20 ml), organic layer was separated, aqueous layer was washed with DCM (2 x 40 mL) and dried over sodium sulphate and concentrated under reduced pressure. Proceeded to next step with crude (5.5 g; mixture of product and alcohol), after column filtration.
-0063: Step 4
Figure imgf000111_0003
[0548] 3.3 g crude undecan-6-yl (tert-butoxycarbonyl)glycinate (Int 2, 1 eq.) was dissolved in DCM (20 ml) and cooled to 0°C, added 7.6 ml TFA (10 eq.) and stirred at room temperature for 1 hour. [0549] Completion of reaction was checked in 2 hours by TLC (10% MeOH/DCM; Rf: 0.5). Reaction mass was concentrated under reduced pressure, residual mass was washed with saturated sodium bicarbonate solution (50 ml) and extracted with EtOAc (3 x 25 ml), organic layer dried over sodium sulphate and concentrated under reduced pressure to get Int 3.
[0550] Crude product was subjected to column chromatography (silica, 60-120) using 1-3% MeOH/CHCh and 2 ml EtsN. Quantity produced, 1.2 g; yield, 40%; confirmed by ¾-NMR and Mass.
[0551] ATX-0063: Step 5
Figure imgf000112_0001
[0552] To a solution of 4 g bromo acetic acid (1 eq.) dissolved in DCM (35 mL), cooled to below 0°C was added 4.7 ml Et3N (1.2 eq.) followed by 13.23 g HATU (1.2 eq.) and 354 mg DMAP (0.1 eq.). To this reaction solution 2.88 g (Z)-non-2-en-l-ol (0.7 eq.) in 20mL of DCM was added and stirred at room temperature overnight.
[0553] Reaction was monitored by TLC (10% EtOAc/hexane; Rf: 0.7).
[0554] Reaction mass was diluted with saturated NaHCC solution (80 ml), organic layer was separated, aqueous layer was washed with DCM (40 ml), dried over sodium sulphate and concentrated under reduced pressure. The residual mass was purified by silica gel (60-120) column chromatography (1.5% EtOAc/hexane). Quantity produced, 4 g; yield, 52%.
[0555] ATX-0063: Step 6
Figure imgf000112_0002
[0556] 1.2 g undecan-6-yl glycinate (Int 3, 1 eq.) was dissolved in 25 ml THF, added 0.9 ml TEA (1.3 eq.) and 1.37 g (Z)-non-2-en-l-yl 2-bromoacetate (Int 4, 1 eq.), and stirred at room temperature overnight.
[0557] Progress of the reaction was monitored by TLC (10% EtOAc/hexane; Rf: 0.5). Reaction mixture was diluted with water (30 ml) and extracted with EtOAc (20 ml x 2), combined organic layer was dried over sodium sulphate and concentrated under reduced pressure.
[0558] The residual mass was purified by column (silica gel; 100-200)
chromatography (3% EtOAc/hexane). Quantity produce, 800 mg; yield, 37%; confirmed by Mass.
[0559] ATX-0063: Step 7
Figure imgf000113_0001
[0560] To a solution of 800 mg (Z)-non-2-en-l-yl (2-oxo-2-(undecan-6- yloxy)ethyl)glycinate (1 eq.) dissolved in DCM, cooled to below 5°C was added 0.4 ml Et3N (3 eq.), followed by 209 mg triphosgene (0.5 eq.) portion-wise for 10 minutes.
[0561] Progress of the reaction mixture monitored by TLC, reaction was completed for 1 hour, reaction mass was concentrated under reduced pressure.
[0562] To a solution of 423 mg N, N-dimethyl ethanethiol hydrochloride (3 eq.) in dry THF and DMF (10 ml and 5 ml, respectively), stirred at 0°C under nitrogen atmosphere was added 144 mg sodium hydride (6 eq.). After 10 minutes, to this reaction mass was added the above solution, by dissolving in THF. The resulting solution was stirred at room temperature for 1 hour.
[0563] Completion of the reaction was observed by TLC (70% EtOAc/hexane; Rf 0.4), after 1 hour. Reaction mass was quenched with saturated NH4CI solution (25 ml), water (20 ml) and EtOAc (20 ml) was added. Aqueous layer was washed with EtOAc (2 x 20 ml), and the combined organic layer was washed with brine solution (20 ml). Organic layer was dried over Na2S04 and concentrated under reduced pressure.
[0564] Crude was subjected to column chromatography using silica gel (100-200) with 20% EtOAc/hexane, and then with neutral alumina with 5% EtOAc/hexane, to get pure compound. Quantity produced, 510 mg; yield, 48%; confirmed by Ti-NMR, HPLC and Mass.
[0565] ATX-0063 / RL-42A: ^-NMR (PPM, 400MHz, CDCh): δ = 5.67 (m, 1), 5.52 (m, 1), 4.92 (m, 1), 4.70 (m, 2), 4.15-4.27 (4), 3.06 (m, 2), 2.53 (m, 2), 2.27 (s, 6), 2.09 (m, 2), 1.47-1.57 (4), 1.20-1.41 (20), 0.82-0.92 (9).
- I l l - Example 14: Synthesis of ATX-0064
[0566] FIG. 13 shows the synthetic pathway of ATX-0064 that is described further as follows.
[0567] ATX-0064: Ste 1
Figure imgf000114_0001
ethyl glycinate
[0568] 12 g ethyl glycinate (1 eq.) was dissolved in THF (100 ml) and cooled to below 0°C. To this resulting solution, 24.2 ml triethylamine (1.5 eq.) and 38.1 1 g Boc anhydride (1.5 eq.) through an additional funnel were added sequentially.
[0569] Progress of the reaction was monitored by TLC using 50% EtOAc/hexane;
Rf: 0.4.
[0570] Reaction mass was quenched with water and EtOAc (100 m) was added, after 16 hour. Organic layer was separated, aqueous layer was washed with EtOAc (2 x 40 ml) and combined organic layers were dried over sodium sulphate and concentrated under reduced pressure.
[0571] Crude product was subjected to 60-120 silica gel (25% EtOAc/hexane). Quantity produced, 20.8; yield, 88%.
[0572] ATX-0064: Ste 2
Figure imgf000114_0002
[0573] To a solution of 18.9 g N-Boc glycine ester (1 eq.) dissolved in THF (130 ml) was added aqueous solution of 5.85 g LiOH (1.5 eq.) and the resulting solution was stirred at room temperature for 4 hours.
[0574] The reaction was monitored by TLC (60% EtOAc/hexane; Rf: 0.3), starting material was absent from the reaction product.
[0575] Reaction mass was concentrated and crude mass was quenched with 5% HC1 (pH 3) and then extracted with EtOAc (4 x 80 ml), dried over sodium sulphate and concentrated under reduced pressure to get the compound. Quantity produced, 15 g; yield, 92%; confirmed by Mass. [0576] ATX-0064: Step 3
Figure imgf000115_0001
[0577] To a solution of 5 g N-Boc-glycine ester (Int 1, 1 eq.), dissolved in DCM (50 mL), cooled to below 0°C was added 4.5 ml Et3N (1.2 eq.) and 6.4 g EDC.HCl (1.2 eq.). To this reaction solution 4.84 g hexadecan-10-ol (0.7 eq.) in 15 ml of DCM was added and stirred at room temperature overnight.
[0578] Starting material observed to be absent by TLC (15% EtOAc/hexane; Rf: 0.6). Reaction mass was diluted with saturated NaHCC solution, organic layer was separated, aqueous layer was washed with DCM (2 x 30 ml) and dried over sodium sulphate and concentrated under reduced pressure.
[0579] Proceeded to next step with crude (5.5 g; mixture of product and alcohol) after column filtration.
-0064: Step 4
Figure imgf000115_0002
[0581] 3.85 g crude heptadecan-9-yl (tert-butoxycarbonyl)glycinate (Int 2, 1 eq.) was dissolved in 30 ml DCM and cooled 0°C, added 7.4 ml TFA (10 eq.), and stirred at room temperature for 1 hour.
[0582] Completion of reaction was checked in 2 hours by TLC (10% MeOH/DCM; Rf: 0.5).
[0583] Reaction mass was concentrated under reduced pressure, residual mass was washed with saturated sodium bicarbonate solution (30 ml) and extracted with EtOAc (3 x 30 ml), organic layer dried over sodium sulphate and concentrated under reduced pressure to afford Int 3.
[0584] Crude product was subjected to column chromatography (silica, 60-120) using 1-3% MeOH/CHCh and 2 ml of Et3N. Quantity produced, 2.2 g; confirmed by ¾- NMR and Mass. [0585] ATX-0064: Step 5
Figure imgf000116_0001
[0586] To a solution of 4 g bromo acetic acid (1 eq.) dissolved in DCM (35 mL), cooled to below 0°C was added 4.7 ml Et3N (1.2 eq.) followed by 13.23 g HATU (1.2 eq.) and 354 mg DMAP (0.1 eq.). To this reaction solution 2.88 g (Z)-non-2-en-l-ol (0.7 eq.) in 20 ml of DCM was added and stirred at room temperature overnight.
[0587] Reaction was monitored by TLC (10% EtOAc/hexane; Rf: 0.7).
[0588] Reaction mass was diluted with saturated NaHCCb solution (80 ml), organic layer was separated, aqueous layer was washed with DCM (40 ml), dried over sodium sulphate and concentrated under reduced pressure.
[0589] The residual mass was purified by silica gel (60-120) column
chromatography (1.5% EtOAc/hexane). Quantity produced, 4 g; yield, 52%.
[0590] ATX-0064: Step 6
Figure imgf000116_0002
[0591] 2.1 g hexadecan-8-yl glycinate (Int 3, 1 eq.) was dissolved in 50 ml THF, added 1.2 ml TEA (1.3 eq.) and 2.39 g (Z)-non-2-en-l-yl 2-bromoacetate (Int 4, 1.3 eq.), and stirred at room temperature overnight.
[0592] Progress of the reaction was monitored by TLC (10% EtOAc/hexane; Rf: 0.5). Reaction mixture was diluted with water (30 ml) and extracted with EtOAc (2 x 30 ml), combined organic layer was dried over sodium sulphate and concentrated under reduced pressure.
[0593] The residual mass was purified by column (silica gel; 100-200)
chromatography (3% EtOAc/hexane). Quantity produced, 2.2 g; yield, 65%; confirmed by Mass. Step 7
Figure imgf000117_0001
[0595] To a solution of 2.2 g heptadecan-9-yl (Z)-(2-(non-2-en-l-yloxy)-2- oxoethyl)glycinate) (1 eq.) dissolved in 15 ml DCM, cooled to below 5°C was added 1.6 ml Et3N (3 eq.), followed by 678 mg triphosgene (0.5 eq.) portion-wise for 10 minutes.
[0596] Progress of the reaction mixture monitored by TLC, reaction was completed for 1 hour, reaction mass was concentrated under reduced pressure.
[0597] To a solution of 3.94 g N, N-dimethyl ethanethiol hydrochloride (7 eq.) in dry THF and DMF (35 ml and 15 ml, respectively), stirred at 0°C under nitrogen atmosphere was added 672 mg sodium hydride (7 eq.). After 10 minutes, to this reaction mass was added the above solution, by dissolving in THF. The resulting solution was stirred at room temperature for 1 hour.
[0598] Completion of the reaction was observed by TLC (70% EtOAc/hexane; Rf 0.4), after 1 hour. Reaction mass was quenched with saturated NH4CI solution (25 ml), water (20 ml) and EtOAc (20 ml) was added. Aqueous layer was washed with EtOAc (20 ml x 2), and the combined organic layer was washed with brine solution (20 ml). Organic layer was dried over Na2S04 and concentrated under reduced pressure.
[0599] Crude was subjected to column chromatography using silica gel (100-200) with 25% EtOAc/hexane, and then with neutral alumina with 15-20% EtOAc/hexane, to get pure compound. Quantity produced, 1.0 mg; yield, 40%; confirmed by ¾-NMR, HPLC and Mass.
[0600] ATX-0064 / RL-42C: ^-NMR (PPM, 400MHz, CDCh): δ = 5.67 (m, 1), 5.50 (m, 1), 4.92 (m, 1), 4.70 (t, J = 7.0, 2), 3.06 (, m, 2), 2.53 (m, 2), 2.27 (s, 6), 1.47-1.57 (4), 1.17-1.40 (30), 0.82-0.93 (9).
Example 15: Synthesis of ATX-0081
[0601] FIG. 14 shows the synthetic pathway of ATX-0081 that is described further as follows. [0602] ATX-0081: Step 1
Figure imgf000118_0001
Ethyl formate
[0603] To a solution of hept l magnesium bromide in 370 mL dry THF (in situ generated) (1.5 eq.), cooled to -15°C under nitrogen atmosphere, was added ethylformate dissolved in 80 mL of THF (0.5 eq.) dropwise via addition funnel over 20 min, and the resulting reaction mixture was warmed to room temperature and stirred overnight.
[0604] Progress of the reaction was monitored by TLC (10% EtOAc in Hexane; Rf:
0.5).
[0605] Reaction mass was quenched with sat. NH4CI solution (500 mL). The organic layer was separated and the aqueous layer was washed with EtOAc (3 x 100 mL). Combined organic layer was dried over anh.Na2S04 and concentrated under reduced pressure.
[0606] Crude compound was subjected to column chromatography on silica gel (60- 120 mesh) using 20% EtO Ac/Hex. Quantity produced, 169.0 g; yield 60%.
[0607] ATX-0081: Step 2
Figure imgf000118_0002
[0608] To a solution of 100 g 4-aminobutanoic acid (1 eq.) in THF, was added IN. aq. NaOH solution (1.065 lit) (1.1 eq.) in an ice bath, followed by 1.3 eq. Boc anhydride, sequentially using addition funnel over a period of 15 min. The resulting solution was allowed to warm to RT and stirred for 4 hours.
[0609] Progress of the reaction was monitored by TLC (10% MeOH/CHCh;
[0610] Reaction mass was quenched with 5% HC1 (ILit) and then EtOAc (300 mL) was added. The organic layer was separated and the aqueous layer was washed with EtOAc (3 x 200 mL). The combined organic layers were concentrated under reduced pressure to get gummy liquid.
[0611] Crude compound was subjected to column chromatography on silica gel (60- 120 mesh) using 80-100% EtO Ac/Hex. Quantity produced, 161.0 g; yield, 82%.
[0612] -0081: Step 3
Figure imgf000118_0003
[0613] To a solution of 3 x 17.8 g 4-((tert-butoxycarbonyl) amino) butanoic acid in DCM (350 mL) (1 eq.), cooled in an ice bath was added 2 x 25.1 g EDC.HC1 (1.5 eq), Et3N (2.0 eq.), and 3 x 1.0 g DMAP (0.1 eq) sequentially under nitrogen atmosphere with 10 min interval. To this resulting solution 3 x 20.0 g alcohol (1.0 eq.) was added (in 150 mL of DCM) dropwise via additional funnel, at the same temperature, and the resulting reaction mass was allowed to warm to RT and stirred for 24 hours under nitrogen atmosphere.
[0614] Progress of the reaction was monitored by TLC (10% EtOAc in Hex; Rf:
0.5).
[0615] The reaction mass was quenched with water (450 mL) and the organic layer was separated. Aqueous layer was washed with DCM (3 x 100 mL). The combined organic layers were concentrated under reduced pressure. The resulting crude was stirred with sat. NaHCCbsolution (300 mL), for 5 min, and the aqueous phase was extracted with EtOAc (3 x 150 mL). The organic layer was separated, dried over anh. Na2S04 and concentrated under reduced pressure, and proceeded to next step with crude. Quantity produced, 69.0 g (crude; required compound and alcohol);
[0616] ATX-0081: Step 4
Figure imgf000119_0001
[0617] To a solution of 69.0 g pentadecan-8-yl 4-((tert-butoxycarbonyl) amino) butanoate (1 eq.) dissolved in DCM, was added TFA (10 eq.) in an ice bath and the resulting reaction solution was allowed to warm to RT and stirred for 3 hours under nitrogen atmosphere.
[0618] Progress of the reaction was monitored by TLC (10% MeOH in CHCh; Rf: 0.3). Reaction mass was concentrated under reduced pressure. The resulting crude was washed with sat.NaHCCbsolution (450 mL) and then extracted with EtOAc (3 x 150 mL). Combined organic layer was dried over anh.Na2S04 and concentrated under reduced pressure.
[0619] Crude compound was subjected to column chromatography using (60-120 mesh silica gel; 4%MeOH/CHCh and lmL of triethylamine), and alcohol was recovered. Quantity produced, 47.0 g; yield, 57 % for two steps
[0620] ATX-0081: Step 5 HO.
Figure imgf000120_0001
[0621] To a solution of 50.0 g 4-bromo butyric acid in DCM (450 mL) (1.0 eq.), taken in a 2L RB flask, cooled to below 0°C was added 86.0 g EDC.HC1 (1.5 eq.), 83.3 mL Et3N (2.0 eq.), and 3.6 g DMAP (0.1 eq.) sequentially under nitrogen atmosphere with lOmin interval. To this resulting solution 42.5 g alcohol (1.0 eq.) was added (by dissolving in 200 mL of DCM) using additional funnel and resulting solution was warmed to RT and stirred for 24h under nitrogen atmosphere.
[0622] Progress of the reaction was monitored by TLC (10% EtOAc in Hex; Rf:
0.7).
[0623] The reaction mass was quenched with water (250 mL) and the organic layer was separated. The aqueous layer was washed with DCM (2 x 150 mL). The combined organic layers were concentrated under reduced pressure. The resulting crude was washed with sat.NaHCC solution (150 mL) and then extracted with EtOAc (2 xl50 mL). The organic layer was separated and concentrated under reduced pressure.
[0624] Crude compound was subjected to column chromatography on silica gel (60- 120 mesh silica gel) using 5% EtO Ac/Hex. Quantity produced, 55.0 g; yield, 63 %
[0625] ATX-0081: Step 6
Figure imgf000120_0002
[0626] To a solution of 2 x 20.0 g pentadecan-8-yl 4-aminobutanoate (1 eq.), 2 x 18.5 g (Z)-non-2-en-l-yl 4-bromobutanoate (1 eq.) in 180 ml of ACN, 2 x 17.6 g potassium carbonate (2 eq.) was added and the resulting mixture was refluxed at 70°C for 5h under nitrogen atmosphere.
[0627] Progress of the reaction was monitored by TLC (10% MeOH/CHCb; Rf: 0.4). Reaction mass was filtered, washed with ACN (2 x 20 mL), and the filtrate concentrated under reduced pressure. [0628] Crude compound was subjected to column chromatography (100-200 mesh silica gel) using 30% EtO Ac/Hex. SMs (Amine) was recovered. Quantity produced, 24.0 g; yield, 36%.
[0629] ATX-0081: Step 7
Figure imgf000121_0001
[0630] To a solution of 24.0 g (Z)-non-2-en-l-yl 4-((4-oxo-4-(pentadecan-8- yloxy)butyl)amino)butanoate (1 eq.), dissolved in 250 mL dry DCM , 13.5 g triphosgene (1 eq.) was added and the reaction mixture was cooled to 0°C, and 18.4 mL pyridine(5 eq.) was added dropwise over a period of 10 min. The reaction mixture was stirred at 20°C for 4 h.
[0631] DCM was removed under reduced pressure and the mixture was taken up with pyridine (300 mL). After cooled to 0°C, 32.3 g dimethylaminoethanethiol hydrochloride salt (5 eq.) was added portion wise and the resulting solution was stirred for overnight at 20°C under nitrogen atmosphere.
[0632] The reaction mass, after TLC checking, was concentrated under reduced pressure to dryness. To this residue was added 250 mL EA and 200 mL (10%) citric acid. The organic phase was separated and then organic layer was washed again with 10% citric acid (100 mL) for one time and again 10% brine (200 mL) for one time. The resulting organic layer was dried overanh. Na2S04 and concentrated under reduced pressure to get crude product.
[0633] A first purification was done on silica gel (100-200 mesh; 500 g). In gradient elution, compound was eluted at 70%EtO Ac/Hex. After concentration compound (22.0 g) appeared in reddish color. A second purification was done using neutral alumina (400 g) with HPLC grade solvents. Compound was eluted at 15%EtO Ac/Hex and pure fractions were concentrated under reduced pressure to get 19.0 g of yellow liquid. [0634] The product (14.0 g) was diluted with 200 mL EtOH (HPLC grade), then added charcoal (50% WAV) 7.0 g to the solution and continued stirring at room temperature for overnight. The resulting solution was filtered through a pad of celite and the filtrate was concentrated. Finally, compound dissolved in 120 mL of 50% EtO Ac/Hex and filtered (to remove alumina and silica particles) through cotton plug and concentrated under reduced pressure. Final compound (11.5 g) appeared in light yellow color.
[0635] A second batch (5.0 g) was diluted with 80 mL EtOH (HPLC grade), then added charcoal (50% WAV) 2.5 g to the solution and continued stirring at room temperature for overnight. The resulting solution was filtered through a pad of celite and the filtrate was concentrated. Finally, compound dissolved in 60 mL of 50% EtO Ac/Hex and filtered (to remove alumina and silica particles) through cotton plug and concentrated under reduced pressure. Final compound (4.0 g) appeared in reddish yellow color. Quantity produced, 15.5 g; yield, 51%.
[0636] ATX-0081: !H-NMR (PPM, 400MHz, CDCh): δ =5.62 (m, 1), 5.51 (m, 1), 4.86 (m, 1), 4.62 (d, J = 6.0Hz, 2), 3.37 (brs, 4), 3.01 (t, J = 7.1Hz, 2), 2.51 (t, J = 7.1Hz, 2), 2.31 (m, 4), 2.26 (s, 6), 2.07 (m, 2), 1.89 (brs, 4), 1.42-1.57 (4), 1.16-1.40 (28), 0.82-0.91 (9) Example 15: Synthesis of ATX-0085
[0637] FIG. 15 shows the synthetic pathway of ATX-0085 that is described further as follows.
[0638] ATX-0085: Step 1
Figure imgf000122_0001
[0639] In a 500 mL lit single neck RB flask,, 30.0 g heptanoic acid (1 eq.) dissolved in of DCM (200 mL) was taken and then added 26.7 mL oxalyl chloride (1.5 eq.) slowly at 0°C, stirring under nitrogen atmosphere and then added I ml DMF (catalytic). The resulting reaction mixture was stirred at RT for 2h.
[0640] In a separate 1 lit two neck RB flask, to 40.5 g N,0-dimethylhydroxylamine hydrochloride (2 eq.) in DCM (250 mL), was added 86.6 mL trimethylamine (3 eq.) using additional funnel, stirred at 0°C. To this resulting solution, the above acid chloride, after concentration under reduced pressure, was added under nitrogen atmosphere by dissolving in DCM(100 mL), dropwise using addition funnel for 20min. The resulting reaction solution was stirred at RT for 3h under nitrogen atmosphere. [0641] Progress of the reaction was monitored by TLC (20% EtOAc/Hexane; Rf: 0.5). Reaction mass was diluted with water (250 mL).
[0642] The organic layer was separated and the aqueous layer was washed with DCM (3 x 100 mL). The combined organic layers were concentrated under reduced pressure.
[0643] Crude compound was subjected to column chromatography using (60-120 silica gel) using 10% EtO Ac/Hex. Quantity produced, 38.0 g; yield, 84%.
[0644] AT -0085: Step 2
Figure imgf000123_0001
[0645] To a solution of 62.3 g hexylmagnesium bromide (1.5 eq.) in ether, taken in a 1 L two neck RB flask, stirred at 0°C under nitrogen atmosphere, was added 38.0 g N- methoxy-N-methylheptanamide (1 eq.) dissolved in 250 mL of ether and the resulting reaction mixture was stirred at RT for 4 h.
[0646] Progress of the reaction was monitored by TLC (10% EtO Ac in Hexane; Rf:
0.7).
[0647] Reaction mass was quenched with sat. NH4CI solution (200 mL). The organic layer was separated and the aqueous layer was washed with ether (2 x 100 mL). The combined organic layers were dried over anh.Na2S04 and concentrated under reduced pressure.
[0648] Crude compound was subjected to column chromatography using (60-120 mesh silica gel) using 2% EtO Ac/Hex. Quantity produced, 30.8g; yield, 71%.
[0649] ATX-0085: Step 3
Figure imgf000123_0002
[0650] To a solution of 30.0 g tridecan-7-one (1 eq.), dissolved in 200 MeOH/THF (10: 1, v:v), 8.5 g sodium borohydride (1.5 eq.) was added at 0°C and the resulting solution was stirred at RT for 2 h.
[0651] Progress of the reaction was monitored by TLC (10% EtOAc/Hexane; Rf:
0.5).
[0652] Reaction mass was quenched with sat. NH4CI solution (80 mL). Solvent was removed under reduced pressure and the resulting crude was partitioned between EtO Ac (200 mL) and water (100 mL). Organic layer was separated and the aqueous layer was washed with EtOAc (2 x 70 mL). Combined organic layers were concentrated under reduced pressure to get white solid. Quantity produced, 27.2 g; yield, 90%.
[0653] ATX-0085: Step 4
Figure imgf000124_0001
[0654] To a solution of 50.0 g 4-aminobutanoic acid (1 eq.) dissolved in THF/aq. NaOH solution (490 mL) was added at 0°C, followed by 140 mL Boc anhydride (1.3 eq.), sequentially using additional funnel, over a period of 15 min. The resulting solution was stirred at RT for 4 hours.
[0655] Progress of the reaction was monitored by TLC (10% MeOH in CHCh; Rf:
[0656] Reaction mass was quenched with 5% HC1 (250 mL) and then EtOAc (300 mL) was added. Organic layer was separated and the aqueous layer was washed with EtOAc (3 x 150 mL). Combined organic layer was concentrated under reduced pressure to get gummy liquid. Quantity produced, 80.0 g; yield, 81%. Confirmed by 1H NMR. Note: Since Boc-Acid (Int 4) is having some impurity (20-30%) observed in the 1H NMR, it was reflected in step 5 & step 6 yields
[0657] A -0085: Step 5
Figure imgf000124_0002
[0658] To a solution of 10.0 g 4-((tert-butoxycarbonyl) amino)butanoic acid (1 eq.) dissolved in DCM (200 mL), cooled to below 0°C was added 12.2 g EDC.HCl (1.3 eq.), 20.4 mL Et3N (3 eq.), and 601 mg DMAP (0.1 eq.) sequentially under nitrogen atmosphere with lOmin interval. To this resulting solution alcohol was added at the same temperature, by dissolving in DCM (150mL), using additional funnel, and stirred at RT for 24 hours under nitrogen atmosphere.
[0659] Progress of the reaction was monitored by TLC (10% EtOAc in Hex; Rf:
0.5).
[0660] Reaction mass was quenched with water (150 mL) and then organic layer was separated. Aqueous layer was washed with DCM (2 x 75 mL). Combined organic layer was concentrated under reduced pressure. The resulting crude was washed with
sat.NaHC03Solution (lOOmL) and then EtOAc (200 mL) was added. Organic layer was separated and concentrated under reduced pressure, and proceeded to next step with crude. Quantity produced, 8.0 g (crude; required compound and alcohol).
-0085: Step 6
Figure imgf000125_0001
[0662] To a solution of 8.0 g tridecan-7-yl 4-((tert-butoxycarbonyl)amino)butanoate (1 eq.) dissolved in 65 mL DCM, was added 15.7 mL TFA (10 eq.) at 0°C and stirred at RT for 3 hours under nitrogen atmosphere.
[0663] Progress of the reaction was monitored by TLC (10% MeOH in CHCh; Rf:
[0664] Reaction mass was concentrated under reduced pressure. The resulting crude was washed with sat.NaHCC solution (100 mL) and then extracted with EtOAc (3 x 100 mL). Organic layer was separated and concentrated under reduced pressure. Crude compound was subjected to column chromatography using (60-120 mesh silicagel;
4%MeOH/CHCh and ImL of triethylamine), and alcohol was recovered. Quantity produced, 3.5 g; yield, 25% for two steps. Confirmed by Mass.
[0665] ATX-0085: Step 7
Figure imgf000125_0002
[0666] To 20.0 g hexanoic acid (1 eq.) dissolved in of DCM (150 mL) was added slowly 22.1 mL oxalyl chloride (1.5 eq.) at 0°C, stirring under nitrogen atmosphere and then added 1 mL DMF (catalytic). The resulting reaction mixture was stirred at RT for 2 hours.
[0667] In a separate flask, to 33.5 g N,0-dimethylhydroxylamine hydrochloride (2 eq.) in DCM (250 mL), was added 71.7 mL trimethylamine (3 eq.) using an addition funnel, stirred at 0°C. To this resulting solution, the above acid chloride, after concentration under reduced pressure, was added under nitrogen atmosphere by dissolving in DCM (100 mL), dropwise using addition funnel for 20min. The resulting reaction solution was stirred at RT for 3 hours under nitrogen atmosphere.
[0668] Progress of the reaction was monitored by TLC (20% EtOAc/Hexane; Rf: 0.5). Reaction mass was diluted with water (250 mL). Organic layer was separated and the aqueous layer was washed with DCM (2 x 100 mL). Combined organic layer was concentrated under reduced pressure.
[0669] Crude compound was subjected to column chromatography using (60-120 silica gel) using 10% EtO Ac/Hex. Quantity produced, 21.0 g; yield, 76%.
[0670] ATX-0085: Step 8
Figure imgf000126_0001
[0671] To a solution of 33.0 g pentylmagnesium bromide (1.5 eq.) in ether at 0°C under nitrogen atmosphere, was added 20.0 g N-methoxy-N-methylhexanamide (1 eq.) dissolved in 220 mL of ether and the resulting reaction mixture was stirred at RT for 4 hours.
[0672] Progress of the reaction was monitored by TLC (10% EtO Ac in Hexane; Rf:
0.6).
[0673] Reaction mass was quenched with sat. NH4CI solution (200 mL). Organic layer was separated and the aqueous layer was washed with ether (2 x 100 mL). Combined organic layers were dried over anh.Na2S04 and concentrated under reduced pressure. Crude compound was subjected to column chromatography using (60-120 mesh silica gel) using 2% EtO Ac/Hex. Quantity produced, 15.4g; yield, 72%.
[0674] ATX-0085: Step 9
Figure imgf000126_0002
[0675] To a solution 15.0 g of undecan-6-one (1 eq.) dissolved in 100 mL MeOH/ 15 mL THF, 4.9 g sodium borohydride (1.5 eq.) was added at 0°C and the resulting solution was stirred at RT for 2 h.
[0676] Progress of the reaction was monitored by TLC (10% EtOAc/Hexane; Rf:
0.5).
[0677] Reaction mass was quenched with sat. NH4CI solution (80 mL). Solvent was removed under reduced pressure and the resulting crude was partitioned between EtO Ac (200 mL) and water (100 mL). Organic layer was separated and the aqueous layer was washed with EtO Ac (2 x 70 mL). Combined organic layers were concentrated under reduced pressure to get white solid. Quantity produced, 13.9 g; white solid; yield, 92%
[0678] ATX-0085: Step 10
Figure imgf000127_0001
[0679] To a solution of 10.0 g 5-bromopentanoic acid (1 eq.), dissolved in DCM (300 mL), cooled to below 0°C was added 13.7 g EDC.HC1 (1.3 eq.), 23.0 mL Et3N (3 eq.), and 674 mg DMAP sequentially under nitrogen atmosphere with 10 min interval. To this resulting solution 9.5 g alcohol (1 eq.) was added at the same temperature, by dissolving in DCM (150 mL), using additional funnel, and stirred at RT for 24 hours under nitrogen atmosphere.
[0680] Progress of the reaction was monitored by TLC (10% EtO Ac in Hex; Rf:
0.7).
[0681] Reaction mass was quenched with water (200 mL) and then organic layer was separated. Aqueous layer was washed with DCM (2 x 100 mL). Combined organic layer was concentrated under reduced pressure. The resulting crude was washed with
sat.NaHC03Solution (100 mL) and then EtO Ac (3 x 100 mL) was added. The organic layers were separated and concentrated under reduced pressure, and proceeded to next step with crude. Crude compound was subjected to column chromatography using (60-120 mesh silica gel) using 5% EtO Ac/Hex, and SM (alcohol) was recovered. Quantity produce, 11.6 g; yield, 62%.
-0085: Step 11
Figure imgf000127_0002
[0683] To a solution of 4.5 g tridecan-7-yl 4-aminobutanoate (1 eq.), 5.2 g undecan- 6-yl 5-bromopentanoate (1 eq.) in ACN, 3.0 g potassium carbonate (1.4 eq.) was added and the resulting mixture was refluxed at 90°C for 4 hours under nitrogen atmosphere.
[0684] Progress of the reaction was monitored by TLC (10% MeOH/CHCh; Rf:
0.45).
[0685] Reaction mass was filtered, washed with ACN (2 x 20 mL), and the filtrate concentrated under reduced pressure. Crude compound was subjected to column
chromatography (100-200 mesh silica gel) using 20% EtO Ac/Hex. SMs (Amine and Bromo compound) were recovered. Quantity produced, 2.2 grams of the pure compound; yield, 25%; and 1.1 gram of the mixture. Confirmed by Mass.
[0686] ATX-0085:
Figure imgf000128_0001
[0687] To a solution 2.2 g of undecan-6-yl 5-((4-oxo-4-(tridecan-7- yloxy)butyl)amino)pentanoate (1 eq.) dissolved in dry DCM, was added 1.6 mL
trimethylamine (3 eq.) and 604 mg triphosgene (0.5 eq.) with 5 min interval at 0°C under nitrogen atmosphere. The resulting solution was stirred at RT, under nitrogen atmosphere for 1 hr. The resulting reaction mass was concentrated under reduced pressure and kept under nitrogen atmosphere.
[0688] To a suspension of 684 mg sodium hydride (7 eq.) in dry THF (30 mL), in a flask stirred at 0°C under nitrogen atmosphere, was added 2.0 g 2-(dimethylamino)ethane-l- thiol hydrochloride (3.5 eq.) and kept stirring for 5 min under nitrogen atmosphere. To this resulting solution the above carbamoyl chloride, dissolved in THF (50 mL), was added via syringe slowly for about 10 min. The resulting solution was stirred at RT overnight under nitrogen atmosphere.
[0689] Progress of the reaction was monitored by TLC (10% MeOH/CHCb; Rf: 0.5; PMA charring).
[0690] Reaction mass was quenched with sat NH4CI (40 mL) and then EtOAc (100 mL) was added. Organic layer was separated and the aqueous layer was washed with EtOAc (2 x 40 mL). The combined organic layers were concentrated and the resulting crude was subjected to column chromatography. A first purification was done using silica gel (100-200 mesh). 5.0 g of crude compound was adsorbed on 9.0 g of silica gel and poured onto 70 g of silica gel taken in the column. Compound was eluted at 50% EtO Ac/Hex. A second purification was done using neutral alumina with HPLC grade solvents. 2.0 g of crude compound was adsorbed on 7.0 g of neutral alumina and the resulting was poured onto 40 g of neutral alumina taken in the column. Compound was eluted at 25%EtO Ac/Hex. Quantity: 1.4 g; yield: 51%. Confirmed by ¾ NMR.
[0691] ATX-0085 ^-NMR (PPM, 400MHz, CDCh): δ = 4.87 (m, 2), 3.36 (brs, 4), 3.02 (t, J = 7.1Hz, 2), 2.52 (t, J = 7.1Hz, 2), 2.31 (m, 4), 2.27 (s, 6), 1.88 (brs, 2), 1.56-1.66 (4), 1.46-1.54 (8), 1.21-1.34 (30), 0.89 (m, 12).
Example 15: Synthesis of ATX-0134
[0692] FIG. 16 shows the synthetic pathway of ATX-0134 that is described further as follows.
[0693] ATX-0134: Step 1
9 BnNH2 9 Bn 9
Br^OMe " MeO -N- OMe
[0694] In a 1 L single neck RB flask, to a stirred solution of 50.0 g methyl 2- bromoacetate (1 eq.) in 400 mL of DMF, was added 90.3 g potassium carbonate (2 eq.) followed by 17.5 g benzyl amine (0.5 eq.), at ice bath temperature under nitrogen
atmosphere, and the resulting reaction mixture was allowed to warm to RT and continued stirring for 36 h.
[0695] Progress of the reaction was monitored by TLC (20% EtOAc/Hexane; Rf: 0.4; ninhydrin charring).
[0696] Reaction mass was diluted with ice water (1 L) and then EtOAc (250 mL) was added. Organic layer was separated and aqueous layer was washed with EtOAc (3 x 100 mL). Combined organic layer was again washed with ice water (500 mL) and the resultant organic layer was dried over anh.Na2S04 and concentrated under reduced pressure.
[0697] Crude compound was subjected to column chromatography on silica gel (60- 120 mesh) using 15-20% EtOAc/Hex. Quantity produced, 35.0 g; yield, 85%.
[0698] ATX-0134: Step 2
9 Bn 9 Pd/C 9 H 9
MeO'^/Nv-^OMe " MeO^^ ^OMe
[0699] 10.5 g 10% Pd/C was added to the reaction 500 mL hydrogenation flask, containing 35.0 g dimethyl 2,2'-(benzylazanediyl)diacetate in EtOAc, and the reaction mass was subjected to hydrogenation using Parr shaker (60 psi) until starting material disappeared
(2 h). [0700] Progress of the reaction was monitored by TLC (5% MeOH/CHC13; Rf: 0.5; ninhydrin charring).
[0701] Reaction mass was filtered through a pad of celite and was washed with EtOAc (2 x 60 mL). The combined filtrate was concentrated under reduced pressure.
Quantity produced, 30.0 g (crude).
[0702] ATX-0134: Step 3
Boc
MeO ^ OMe MeO ^ OMe
[0703] To a solution of 30.0 g dimethyl 2,2'-azanediyldiacetate (1.0 eq.) in THF, was added 38.7 mL triethylamine (1.5 eq.) followed by 55.5 mL Boc anhydride (1.3 eq.), under ice bath temperature, sequentially using an addition funnel. The resulting reaction solution was stirred at RT overnight under nitrogen atmosphere.
[0704] Progress of the reaction was monitored by TLC (40% EtOAc/Hexane; Rf:
0.5).
[0705] Reaction mass was diluted with water (250 mL) and then EtOAc (150 mL) was added. Organic layer was separated and aqueous layer was washed with EtOAc (2 x 100 mL). Combined organic layer was dried over anh.Na2S04 and concentrated under reduced pressure. Crude compound was subjected to column chromatography on silica gel (60-120 mesh) using 15% - 20% EtO Ac/Hex. Quantity produced, 35.0 g; yield, 96%.
[0706] ATX-0134: Step 4
9 Boc 9 LiOH 9 Boc 9
ϋ Ν ιι,— - Η<Λ-Ν-Λ>Η
[0707] To a solution of 35.0 g dimethyl 2,2'-((tert- butoxycarbonyl)azanediyl)diacetate (1.0 eq.) in THF stirred under ice bath temperature, was added aq. solution of 16.8 g lithium hydroxide (75 mL of 5.3 M) (3 eq.). The resulting reaction solution was stirred at RT for 5 hours under nitrogen atmosphere.
[0708] Progress of the reaction was monitored by TLC (10% MeOH/CHC13 ; Rf
0.2).
[0709] Reaction mass was quenched with 5%HC1 (400 mL) and then EtOAc (200 mL) was added. Organic layer was separated and aqueous layer was washed with EtOAc (2 x 100 mL). Combined organic layer was dried over anh.Na2S04 and concentrated under reduced pressure. Crude compound was subjected to column chromatography on silica gel (60-120 mesh) using 100%EtOAc. Quantity produced, 29.0 g; yield, 93%. [0710] ATX-0134: Step 5
Figure imgf000131_0001
[0711] 1 2,2'-((tert-butoxycarbonyl)azanediyl)diacetic acid 233.22 0.051 1.0
[0712] 2 Alcohol 228.42 0.102 2.0
[0713] 3 EDC.HC1 191.70 0.154 3.0
[0714] 4 Triethylamine 101 0.154 3.0
[0715] 5 DMAP 122.17 0.002 0.1
[0716] 6 DCM 400 mL
[0717] To a solution of 12.0 g 4,4'-((tert-butoxycarbonyl)azanediyl)dibutanoic acid (1 eq.) in 250 mL of DCM, cooled to between 0°C to 50°C (ice bath), was added 29.5 g EDC.HC1 (3 eq.), 21.4 mL Et3N (3 eq.), and 628 mg DMAP (0.1 eq.) sequentially under nitrogen atmosphere with 10 min interval. To this resulting solution 23.5 g alcohol (in 150 mL of DCM) (2 eq.) was added at the same temperature via addition funnel and the resulting reaction mass was allowed to warm to RT and stirred for 24 hours under nitrogen atmosphere.
[0718] Progress of the reaction was monitored by TLC (10% EtOAc in Hex; Rf: 0.6; PMA charring).
[0719] Reaction mass was quenched with water (100 mL) and then organic layer was separated. Aqueous layer was washed with DCM (2 x 80 mL). Combined organic layer was concentrated under reduced pressure. The resulting crude was stirred with
sat.NaHC03 solution (100 mL) for 5 min, to remove unreacted acid, and then EtOAc (2 x 80 mL) was added. Organic layer was dried over anh. Na2S04 and concentrated under reduced pressure. Crude compound was subjected to column chromatography on silica gel (60-120 mesh) using 2-3% EtO Ac/Hex. Quantity produced, 15.0 g; yield, 44%.
[0720] ATX-0134: Step 6
Figure imgf000131_0002
[0721] To a solution of 15.0 g di(pentadecan-8-yl) 2,2'-((tert- butoxycarbonyl)azanediyl)diacetate (1 eq.) in 120 mL of DCM, was added 17.7 mL TFA (10 eq.), between 0°C to 50°C (ice bath), and the reaction mass was allowed to warm to RT and stirred for 4 hours under nitrogen atmosphere.
[0722] Progress of the reaction was monitored by TLC (10% ETOAc/Hex; Rf: 0.5).
[0723] Reaction mass was concentrated under reduced pressure and the resulting crude was stirred with sat.NaHC03solution (100 mL) for 5 min (to remove traces of TFA) and the aqueous phase was extracted with EtOAc (3 x 100 mL). Organic layer was separated and concentrated under reduced pressure. Crude compound was subjected to column chromatography on silica gel (60-120 mesh) and compound was eluted with 10%
EtO Ac/Hex. Quantity produced, 10.4 g; yield, 82%.
[0724] ATX-0134: Step
Figure imgf000132_0001
[0725] To a solution of 1.5 g di(pentadecan-8-yl) 2,2'-azanediyldiacetate (1 eq.) in 20 mL of dry DCM, was added 1.1 mL trimethylamine (3 eq.) and 401 mg triphosgene (0.5 eq.) with 5 min interval under ice bath temperature and nitrogen atmosphere. The resulting solution was then allowed to warm to RT and stirred for lh under nitrogen atmosphere. After completion of starting material (checked by TLC), the resulting reaction mass was concentrated under reduced pressure and kept under nitrogen atmosphere. 1.1 g 3- (diethylamino)propane-l -thiol (3 eq.), dissolved in 10 mL of THF, was added to a suspension of 129 mg sodium hydride (2 eq.) in dry THF (10 mL), cooled to between 0°C to 50°C under nitrogen atmosphere, and stirring was continued for 5 min. To this resulting solution the above carbamoyl chloride, dissolved in THF (30 mL), was added at the same temperature via addition funnel for about 5 min. The resulting solution was warmed to RT and continued stirring overnight under nitrogen atmosphere.
[0726] Progress of the reaction was monitored by TLC (10% MeOH/CHC13 ; Rf 0.5; PMA charring). [0727] Reaction mass was quenched with sat NH4C1 (30 mL) and then EtOAc (50 mL) was added. Organic layer was separated and the aqueous layer was washed with EtOAc (2 x 40 mL). Combined organic layer was concentrated and the resulting crude was subjected to column chromatography. First purification was done using neutral alumina (100 g). In gradient elution, compound was eluted at 20%EtO Ac/Hex. Second purification was done using silica gel (100-200 mesh; 80 g) with HPLC grade solvents. Compound was eluted at 50%EtOAc/Hex. Quantity: 680 mg; yield: 34%.
[0728] ATX-0134 ^-NMR (PPM, 400MHz, CDCh): δ = 4.91 (m, 2), 4.21 (s, 2), 4.16 (s, 2), 2.95 (t, J = 7.1Hz, 2), 2.54 (m, 6), 1.79 (m, 2), 1.46-1.52 (8), 1.17-1.36 (40), 1.03 (t, J = 7.8Hz, 6), 0.87 (t, J = 7.1Hz, 12).
Example 15: Synthesis of ATX-0044 and ATX-0091 to ATX-0133.
[0729] ATX-0044, ATX-0085, ATX-0111, ATX-0132, ATX-0100, ATX-0117, ATX-0114, ATX-0115, ATX-0101, ATX-0106, ATX-0116, ATX-0122, ATX-0123, ATX- 0124, ATX-0126, ATX-0129, and ATX-0133 were synthesized using the methods of the previous examples.
Example 16: pKa values
[0730] pKa of cationic lipids in LNP or micellar formulations were measured by the procedure of Jayaraman, 2012, Angew. Chem. Int. Ed. , 51 :8529-33, hereby incorporated by reference. Lipid micelles or LNPs are diluted to 1 mM total lipids in universal buffer with a pH range between 3 and 12 in presence of 0.06 mg/mL 6-(/ toluidino)-2-naphthalenesulfonic acid sodium salt (TNS) reagent (Sigma Aldrich), a pH sensitive fluorescence probe. The anionic TNS molecule fluoresces when associated with the surface of positively charged membranes but is not fluorescent when free in solution, allowing measurement of pKa. The TNS signal is measured on a spectral plate reader. The TNS signal is plotted as function of the pH and analyzed using a non-linear (Boltzman) to determine the pKa.
[0731] Reagents used in the assay include
• Hepes free acid, CAS: 7365-45-9 (VWR, 0511-lKG or equivalent)
• MES, HPLC grade: (Sigma 105228- 100G or equivalent)
• Ammonium acetate, HPLC grade: (Sigma 90335-lOOmL or equivalent)
• Sodium chloride, HPLC grade (VWR EM-MX0475-1 or equivalent)
• TNS (Sigma-Aldrich T9792-250mg or equivalent)
• Hydrochloric acid
• DMSO DPBS without calcium and magnesium (GE, SH30028.O2 or equivalent) H2O, HPLC grade (OmniSolv, WX0004-1 or equivalent)
[0732] To prepare a stock solution of Universal Buffer (UB) in a polystyrene sterile storage bottle, prepare 1 L using the following table.
Figure imgf000134_0001
Reagents are sterile filtered through a 0.2 μιτι filter. Preparation of UB solution includes adding 5 mL of 1 M HC1 to a 350 mL stock solution.
[0733] To prepare a pH ladder, centrifuge tubes (50 mL) with 20 mL UB buffer are used. (pH ~3) and differing amounts of 2 N NaOH. Stock solutions of TNS at 1 mg/mL in DMSO. 60 of TNS at 1 mg/mL is added to 940 water to reach a working solution concentration of 0.06 mg/mL. Five test samples, 1 mL of LNP sample in PBS at 1 mM total lipid, and 1 reference sample are analyzed per plate. Samples at different pH values are prepared in in wells in which 25 μΐ. of 0.06 mg/mL TNS is added. 15 μΐ. of test samples is added per well. The microplate is incubated for 15 minutes at room temperature in the dark. TNS template can be used to format data from the plate reader. Perform non-linear
(Boltzman) regression analysis of the samples in Origin Pro 8 or equivalent software.
[0734] The results are shown in Table 1. In general increasing lipophilicity via the ester near the ionizable head group lowers the measured pKa while making the calculated pKa more basic. Adding lipophilicity in the alcohol while shortening the length of the ester chain can have a major impact on measured pKa. For example, shortening the ester to butanoate has no impact when the lipophilicity of the alcohol is increased to a branched alkyl group (see: ATX-0057 and ATX-0058 vs ATX-0002). A major impact is seen when shortening the ester to an acetate, e.g., ATX-0061 / ATX-0064 (Δ = -0.90) and ATX-0063 (Δ = -0.7) for 1 -branched / 1 -Z-2-nonenol ester; and ATX-0062/ATX-0065 (Δ = -3.20) for acetate / bis-branched ester with a Δ cLogP +4.0 vs ATX-0002.
[0735] Without conceding to be bound by theory, the results show the importance of increased lipophilicity (measured by cLogP) combined with chain shortening in lowering the measured pKa compared to predicted, and to increasing the bioactivity of the lipid nanoparticles.
Example 17: In vivo EPO mRNA stability
[0736] Levels of mRNA in plasma were measured and compared following injection of nanoparticles comprising different cationic lipids. Female Balb/c mice (6-8 week old) were used for evaluation of plasma erythropoietin (epo) levels in vivo following injection of lipid encapsulated mouse epo mRNA. All formulations were administered intravenously via tail vein injection at a dose of 0.03 and 0.1 mg/kg at a dosing volume of 5 ml/kg.
Terminal blood collection was performed via cardiac puncture under 2% isoflurane at 6 hours after formulation injections. Blood was collected into 0.109 M citrate buffer tube and processed by centrifugation at 5000 rpm for 10 minutes. Serum was collected and epo mRNA levels were analyzed. Results are shown at FIG. 17. Results show a substantial improvement over ATX-0002 for ATX-0057, ATX-0081, ATX-0082, ATX-0083, ATX- 0084, ATX-0085, ATX-0086, and ATX-0087.
Example 18: In vivo mouse Factor VII silencing and EPO expression
[0737] Using a liver-directed in vivo screen of the liposome libraries, a series of compounds were tested that facilitate high levels of siRNA mediated gene silencing in hepatocytes, the cells comprising the liver parenchyma. Factor VII, a blood-clotting factor, is a suitable target gene for assaying functional siRNA delivery to liver. Because this factor is produced specifically in hepatocytes, gene silencing indicates successful delivery to parenchyma, as opposed to delivery to the cells of the reticulo-endothelial system (e.g., Kupffer cells). Furthermore, Factor VII is a secreted protein that can be readily measured in serum, obviating the need to euthanize animals. Silencing at the mRNA level can be readily determined by measuring levels of protein. This is because the protein's short half-life (2-5 hour). Compositions with siRNA directed to Factor VIII were formulated with the lipid, and comparator sample phosphate-buffered saline (PBS). Female C57BL/6 mice (6-8 week old) were used for FVII siRNA knockdown (KD) experiments.
[0738] All formulations were administered intravenously via tail vein injection at a dose of 0.03 and 0.1 mg/kg at a dosing volume of 5 mg/kg. Terminal blood collection was performed via cardiac puncture under 2% isoflurane at 48 hours after formulation injections. Blood was collected into 0.109 M citrate buffer tube and processed by centrifugation at 1200 G for 10 min. Plasma was collected and Factor VII protein levels were analyzed by chromogenic assay (Biophen FVII, Aniara Corporation). A standard curve was constructed using samples from PBS-injected mice and relative Factor VII expression was determined by comparing treated groups to untreated PBS control. The results showed that ATX-0057 and ATX-0058 were substantially more effective than ATX-002 both at 0.03 and 0.1 mg/kg (FIG. 18).
[0739] Table 1 shows knockdown resulting from lipid nanoparticles comprising the lipids disclosed herein.
[0740] Epo expression following delivery of mRNA using lipid nanoparticles comprising the lipids described herein was measured.
[0741] Female Balb/c mice (6-8 week old) were used for evaluation of epo protein expression in vivo following delivery of lipid encapsulated mouse epo mRNA. All formulations were administered intravenously via tail vein injection at a dose of 0.03 and 0.1 mg/kg at a dosing volume of 5mL/kg. Terminal blood collection was performed via cardiac puncture under 2% isoflurane at 6 hours after formulation injections. Blood was collected into 0.109 M citrate buffer tube and processed by centrifugation at 5000 rpm for 10 minutes. Serum was collected and epo protein levels were analyzed by epo ELISA assay (R&D systems). A standard curve was constructed using samples from PBS-injected mice and relative Factor VII expression was determined by comparing treated groups to untreated PBS control. The results showed that epo mRNA is expressed at substantially higher amounts in ATX-0057 nanoparticles than ATX-002 at 0.1 mg/ml (FIG. 19).

Claims

What is Claimed:
1. A compound of formula I
Figure imgf000137_0001
wherein
Ri is a branched, noncyclic alkyl or alkenyl of 8, 9, 10, 11 , 12, 13, 14, 16, 17, 18,
19, 20, 21 , or 22 carbons;
Li is linear alkane of 1 to 15 carbons;
R2 is a linear alkyl or alkenyl of 5, 6, 7, 8, 9, 10, 1 1, 12, 13, 14 or 15 carbons or a branched, noncyclic alkyl or alkenyl of 8, 9, 10, 11 , 12, 13, 14, 16, 17, 18, 19,
20, 21, or 22 carbons;
L2 is a linear alkane of 4, 5, 6, 7, 8, 9, 10, 11 , 12, 13, 14 or 15 carbons; X is O or S;
R3 is a linear alkane of 1 , 2, 3, 4, 5, or 6 carbons; and
R4 and R5 are the same or different, each a linear or branched, noncyclic alkyl of 1 , 2, 3, 4, 5, or 6 carbons;
or a pharmaceutically acceptable salt or solvate thereof.
2. The compound of claim 1, wherein the compound is selected from the group consisting of compounds of formulas ATX-0082, ATX-0085, ATX-0083, ATX-0121, ATX- 0091, ATX-0102, ATX-0098, ATX-0092, ATX-0084, ATX-0095, ATX-0125, ATX-0094, ATX-0109, ATX-0110, ATX-0118, ATX-0108, ATX-0107, ATX-0093, ATX-0097, and ATX-0096
Figure imgf000138_0001
Figure imgf000139_0001
- 137 -
Figure imgf000140_0001
- 138 -
Figure imgf000141_0001
- 139-
Figure imgf000142_0001
The compound of claim 1, wherein Ri has 8, 9, 10, 11, 12, 13, 14, 16, or 17
The compound of claim 1, wherein Ri comprises two identical alkyl or alkenyl
The compound of claim 1, wherein Ri comprises an alkenyl group.
The compound of claim 1, wherein R2 is an alkenyl.
The compound of claim 1, wherein R2 is a branched, noncyclic alkyl.
The compound of claim 1, wherein L2 has 4, 5, 6, or 7 carbons. A compound of formula I
Figure imgf000142_0002
wherein
Ri is a branched, noncyclic alkyl or alkenyl of 12, 13, 14, 16, 17, 18, 19, 20, 21, or 22 carbons;
Li is a linear alkane of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 carbons; R.2 is linear alkyl or alkenyl of 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15 carbons or a branched, noncyclic alkyl of 10, 11, 12, 13, 14, 16, 17, 18, 19, 20, 21, or 22 carbons;
L2 is a linear alkane of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15 carbons X is O or S;
R.3 is linear alkane of 1, 2, 3, 4, 5, or 6 carbons;
R.4 and R.5 are the same or different, each a linear or branched, noncyclic alkyl of
1, 2, 3, 4, 5, or 6 carbons;
a 1 mM solution of the compound has a pKa of 5.6 to 7.0 as measured by fluorescence of 6-(y toluidino)-2-naphthalenesulfonic; and
the compound has a c-LogD value is between 10 and 14;
or a pharmaceutically acceptable salt or solvate thereof.
10. The compound of claim 9, wherein the compound is selected from the group consisting of compounds of formulas ATX-0111, ATX-0132, ATX-0134, ATX-0100, ATX- 0117, ATX-0114, ATX-0115, ATX-0101, ATX-0106, ATX-0116, ATX-0086, ATX-0058, ATX-0081, ATX-0123, ATX-0122, ATX-0057, ATX-0088, ATX-0087, ATX-0124, ATX- -0129, and ATX-0123.
Figure imgf000143_0001
Figure imgf000144_0001
Figure imgf000145_0001
Figure imgf000146_0001
- 144-
Figure imgf000147_0001
Figure imgf000148_0001
The compound of claim 9, wherein Ri has 12, 13, 14, 16, or 17 carbons.
The compound of claim 9, wherein Ri comprises two identical alkyl or alkenyl groups.
13. The compound of claim 9, wherein Ri comprises an alkyl group.
The compound of claim 9, wherein R2 is an alkenyl.
15. The compound of claim 9, wherein R2 is a branched, noncyclic alkyl.
16. The compound of claim 9, wherein Li and L2 independently have 1, 2, or 3 carbons.
17. The compound of claim 9, wherein Li and L2 both have 3 carbons.
18. The compound of claim 9, wherein R3 is propylene or butylene.
19. The compound of claim 9, wherein its c-LogD value is at least 11 and its measured pKa more basic than 6.
20. A compound of formula I
Figure imgf000149_0001
wherein
Ri is a branched, noncyclic alkyl of 12, 13, 14, 16, 17, 18, 19, 20, 21, or 22
carbons;
Li is a linear alkane of 1, 2, or 3 carbons;
R2 is linear alkenyl of 8, 9, 10, 1 1, or 12 carbons or a branched, noncyclic alkyl of
12, 13, 14, 16, or 17 carbons;
L2 is a linear alkane of 1, 2, or 3 carbons
X is O or S;
R3 is linear alkane of 2 or 3 carbons;
R4 and R5 are the same or different, each a linear or branched, noncyclic alkyl of 1 or 2 carbons;
or a pharmaceutically acceptable salt or solvate thereof.
PCT/US2017/067756 2016-12-21 2017-12-20 Ionizable cationic lipid for rna delivery WO2018119163A1 (en)

Priority Applications (19)

Application Number Priority Date Filing Date Title
KR1020217027797A KR102385562B1 (en) 2016-12-21 2017-12-20 ionizable cationic lipid for rna delivery
EP23196560.9A EP4310075A3 (en) 2016-12-21 2017-12-20 Ionizable cationic lipid for rna delivery
JP2019533410A JP7437935B2 (en) 2016-12-21 2017-12-20 Ionizable cationic lipids for RNA delivery
CN202210319695.4A CN114917203A (en) 2016-12-21 2017-12-20 Ionizable cationic lipids for RNA delivery
CA3046885A CA3046885C (en) 2016-12-21 2017-12-20 Ionizable cationic lipid for rna delivery
AU2017379059A AU2017379059B2 (en) 2016-12-21 2017-12-20 Ionizable cationic lipid for RNa delivery
FIEP17826404.0T FI3558943T3 (en) 2016-12-21 2017-12-20 Ionizable cationic lipid for rna delivery
CN201780086949.XA CN110325511B (en) 2016-12-21 2017-12-20 Ionizable cationic lipids for RNA delivery
EP17826404.0A EP3558943B1 (en) 2016-12-21 2017-12-20 Ionizable cationic lipid for rna delivery
IL290592A IL290592B2 (en) 2016-12-21 2017-12-20 Ionizable cationic lipid for rna delivery
RS20240048A RS65078B1 (en) 2016-12-21 2017-12-20 Ionizable cationic lipid for rna delivery
KR1020197021199A KR102299053B1 (en) 2016-12-21 2017-12-20 Ionizable Cationic Lipids for RNA Delivery
DK17826404.0T DK3558943T3 (en) 2016-12-21 2017-12-20 Ionizable cationic lipid for rna delivery
SI201731472T SI3558943T1 (en) 2016-12-21 2017-12-20 Ionizable cationic lipid for rna delivery
HRP20231742TT HRP20231742T1 (en) 2016-12-21 2017-12-20 Ionizable cationic lipid for rna delivery
PL17826404.0T PL3558943T3 (en) 2016-12-21 2017-12-20 Ionizable cationic lipid for rna delivery
IL306099A IL306099A (en) 2016-12-21 2017-12-20 Ionizable cationic lipid for rna delivery
IL267511A IL267511B (en) 2016-12-21 2019-06-19 Ionizable cationic lipid for rna delivery
AU2021200663A AU2021200663B2 (en) 2016-12-21 2021-02-02 Ionizable cationic lipid for rna delivery

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US15/387,067 2016-12-21
US15/387,067 US10383952B2 (en) 2016-12-21 2016-12-21 Ionizable cationic lipid for RNA delivery
USPCT/US2017/015886 2017-01-31
PCT/US2017/015886 WO2018118102A1 (en) 2016-12-21 2017-01-31 Ionizable cationic lipid for rna delivery

Publications (1)

Publication Number Publication Date
WO2018119163A1 true WO2018119163A1 (en) 2018-06-28

Family

ID=60943160

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2017/067756 WO2018119163A1 (en) 2016-12-21 2017-12-20 Ionizable cationic lipid for rna delivery

Country Status (1)

Country Link
WO (1) WO2018119163A1 (en)

Cited By (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019191780A1 (en) * 2018-03-30 2019-10-03 Arcturus Therapeutics, Inc. Lipid particles for nucleic acid delivery
WO2020118115A1 (en) 2018-12-06 2020-06-11 Arcturus Therapeutics, Inc. Compositions and methods for treating ornithine transcarbamylase deficiency
WO2020191103A1 (en) * 2019-03-19 2020-09-24 Arcturus Therapeutics, Inc. Method of making lipid-encapsulated rna nanoparticles
WO2020255062A1 (en) 2019-06-20 2020-12-24 Janssen Sciences Ireland Unlimited Company Lipid nanoparticle or liposome delivery of hepatitis b virus (hbv) vaccines
WO2021067598A1 (en) 2019-10-04 2021-04-08 Ultragenyx Pharmaceutical Inc. Methods for improved therapeutic use of recombinant aav
WO2021178510A1 (en) * 2020-03-03 2021-09-10 Arcturus Therapeutics, Inc. Compositions and methods for the treatment of ornithine transcarbamylase deficiency
WO2022008613A1 (en) 2020-07-08 2022-01-13 Janssen Sciences Ireland Unlimited Company Rna replicon vaccines against hbv
WO2022146654A1 (en) 2020-12-28 2022-07-07 Janssen Pharmaceuticals, Inc. Transcription activator-like effector nucleases (talens) targeting hbv
WO2023031394A1 (en) 2021-09-03 2023-03-09 CureVac SE Novel lipid nanoparticles for delivery of nucleic acids
WO2023073228A1 (en) 2021-10-29 2023-05-04 CureVac SE Improved circular rna for expressing therapeutic proteins
WO2023144330A1 (en) 2022-01-28 2023-08-03 CureVac SE Nucleic acid encoded transcription factor inhibitors
WO2023218420A1 (en) 2022-05-13 2023-11-16 Janssen Pharmaceuticals, Inc. Mrna compositions for inducing latent hiv-1 reversal
WO2023227608A1 (en) 2022-05-25 2023-11-30 Glaxosmithkline Biologicals Sa Nucleic acid based vaccine encoding an escherichia coli fimh antigenic polypeptide
US11938227B2 (en) 2020-09-13 2024-03-26 Arcturus Therapeutics, Inc. Lipid nanoparticles encapsulation of large RNA

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4511069A (en) 1981-06-04 1985-04-16 The Pharmasol Corporation Dispensing system
US4778810A (en) 1987-01-08 1988-10-18 Nastech Pharmaceutical Co., Inc. Nasal delivery of caffeine
WO1992007065A1 (en) 1990-10-12 1992-04-30 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. Modified ribozymes
WO1993015187A1 (en) 1992-01-31 1993-08-05 Massachusetts Institute Of Technology Nucleozymes
US5849902A (en) 1996-09-26 1998-12-15 Oligos Etc. Inc. Three component chimeric antisense oligonucleotides
WO2016081029A1 (en) * 2014-11-18 2016-05-26 Arcturus Therapeutics, Inc. Ionizable cationic lipid for rna delivery

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4511069A (en) 1981-06-04 1985-04-16 The Pharmasol Corporation Dispensing system
US4778810A (en) 1987-01-08 1988-10-18 Nastech Pharmaceutical Co., Inc. Nasal delivery of caffeine
WO1992007065A1 (en) 1990-10-12 1992-04-30 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. Modified ribozymes
WO1993015187A1 (en) 1992-01-31 1993-08-05 Massachusetts Institute Of Technology Nucleozymes
US5849902A (en) 1996-09-26 1998-12-15 Oligos Etc. Inc. Three component chimeric antisense oligonucleotides
WO2016081029A1 (en) * 2014-11-18 2016-05-26 Arcturus Therapeutics, Inc. Ionizable cationic lipid for rna delivery

Non-Patent Citations (7)

* Cited by examiner, † Cited by third party
Title
"TRANSDERMAL SYSTEMIC MEDICATION", 1985, ELSEVIER PUBLISHERS
ANDERSON ET AL.: "The Practice of Medicinal Chemistry", 1996, ACADEMIC PRESS
BURGIN ET AL., BIOCHEMISTRY, vol. 35, 1996, pages 14090
JAYARAMAN, ANGEW. CHEM. INT., vol. 51, 2012, pages 8529 - 33
LIMBACH ET AL., NUCLEIC ACIDS RES., vol. 22, 1994, pages 2183
P. GOULD, INTERNATIONAL J. PHARMACEUTICS, vol. 33, 1986, pages 201 - 217
S. BERGE ET AL., J. PHARMACEUTICAL SCIENCES, vol. 66, no. 1, 1977, pages 1 - 19

Cited By (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019191780A1 (en) * 2018-03-30 2019-10-03 Arcturus Therapeutics, Inc. Lipid particles for nucleic acid delivery
WO2020118115A1 (en) 2018-12-06 2020-06-11 Arcturus Therapeutics, Inc. Compositions and methods for treating ornithine transcarbamylase deficiency
EP4299750A2 (en) 2018-12-06 2024-01-03 Arcturus Therapeutics, Inc. Compositions and methods for treating ornithine transcarbamylase deficiency
US11685906B2 (en) 2018-12-06 2023-06-27 Arcturus Therapeutics, Inc. Compositions and methods for treating ornithine transcarbamylase deficiency
EP3942050A4 (en) * 2019-03-19 2023-02-22 Arcturus Therapeutics, Inc. Method of making lipid-encapsulated rna nanoparticles
WO2020191103A1 (en) * 2019-03-19 2020-09-24 Arcturus Therapeutics, Inc. Method of making lipid-encapsulated rna nanoparticles
CN113840926A (en) * 2019-03-19 2021-12-24 阿克丘勒斯治疗公司 Method for preparing lipid-encapsulated RNA nanoparticles
US11737979B2 (en) 2019-03-19 2023-08-29 Arcturus Therapeutics, Inc. Method of making lipid-encapsulated RNA nanoparticles
WO2020255062A1 (en) 2019-06-20 2020-12-24 Janssen Sciences Ireland Unlimited Company Lipid nanoparticle or liposome delivery of hepatitis b virus (hbv) vaccines
WO2021067598A1 (en) 2019-10-04 2021-04-08 Ultragenyx Pharmaceutical Inc. Methods for improved therapeutic use of recombinant aav
WO2021178510A1 (en) * 2020-03-03 2021-09-10 Arcturus Therapeutics, Inc. Compositions and methods for the treatment of ornithine transcarbamylase deficiency
WO2022008613A1 (en) 2020-07-08 2022-01-13 Janssen Sciences Ireland Unlimited Company Rna replicon vaccines against hbv
US11938227B2 (en) 2020-09-13 2024-03-26 Arcturus Therapeutics, Inc. Lipid nanoparticles encapsulation of large RNA
WO2022146654A1 (en) 2020-12-28 2022-07-07 Janssen Pharmaceuticals, Inc. Transcription activator-like effector nucleases (talens) targeting hbv
WO2023031394A1 (en) 2021-09-03 2023-03-09 CureVac SE Novel lipid nanoparticles for delivery of nucleic acids
WO2023073228A1 (en) 2021-10-29 2023-05-04 CureVac SE Improved circular rna for expressing therapeutic proteins
WO2023144330A1 (en) 2022-01-28 2023-08-03 CureVac SE Nucleic acid encoded transcription factor inhibitors
WO2023218420A1 (en) 2022-05-13 2023-11-16 Janssen Pharmaceuticals, Inc. Mrna compositions for inducing latent hiv-1 reversal
WO2023227608A1 (en) 2022-05-25 2023-11-30 Glaxosmithkline Biologicals Sa Nucleic acid based vaccine encoding an escherichia coli fimh antigenic polypeptide

Similar Documents

Publication Publication Date Title
AU2017379059B2 (en) Ionizable cationic lipid for RNa delivery
US10961188B2 (en) Ionizable cationic lipid for RNA delivery
EP3397614B1 (en) Ionizable cationic lipid
US9670152B2 (en) Ionizable cationic lipid for RNA delivery
US9580711B2 (en) Lipid particles with asymmetric cationic lipids for RNA delivery
EP3221293B1 (en) Ionizable cationic lipid for rna delivery
WO2018119163A1 (en) Ionizable cationic lipid for rna delivery

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 17826404

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 3046885

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2019533410

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 20197021199

Country of ref document: KR

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2017826404

Country of ref document: EP

Effective date: 20190722

ENP Entry into the national phase

Ref document number: 2017379059

Country of ref document: AU

Date of ref document: 20171220

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: P-2024/0048

Country of ref document: RS