Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
CATIONIC LIPID COMPOUNDS AND COMPOSITIONS THEREOF FOR USE IN THE DELIVERY OF MESSENGER RNA
Document Type and Number:
WIPO Patent Application WO/2020/106903
Kind Code:
A1
Abstract:
The compounds disclosed herein compound of Formula (I), substructures thereof, and pharmaceutically acceptable salts thereof. The compounds provided herein can be useful for delivery and expression of mRNA and encoded protein, e.g., as a component of liposomal delivery vehicle, and accordingly can be useful for treating various diseases, disorders and conditions, such as those associated with deficiency of one or more proteins.

Inventors:
DEROSA FRANK (US)
KARVE SHRIRANG (US)
ZHANG YI (US)
Application Number:
PCT/US2019/062504
Publication Date:
May 28, 2020
Filing Date:
November 20, 2019
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
TRANSLATE BIO INC (US)
International Classes:
C07C215/24; A61K9/127; A61K31/7105; A61K35/15; A61K47/18; A61K47/50; A61K48/00; C12N15/88
Domestic Patent References:
WO2011141705A12011-11-17
WO2013149140A12013-10-03
WO2012068176A12012-05-24
WO2010144740A12010-12-16
WO2013149140A12013-10-03
WO2010053572A22010-05-14
WO2016118725A12016-07-28
WO2016118724A12016-07-28
WO2013063468A12013-05-02
WO2016205691A12016-12-22
WO2015184256A22015-12-03
WO2016004202A12016-01-07
WO2015199952A12015-12-30
WO2017004143A12017-01-05
WO2017075531A12017-05-04
WO2017117528A12017-07-06
WO2017049245A22017-03-23
WO2017173054A12017-10-05
WO2015095340A12015-06-25
WO2012170889A12012-12-13
WO2010042877A12010-04-15
WO2005121348A12005-12-22
Foreign References:
EP3147277A12017-03-29
US9186325B22015-11-17
US4373071A1983-02-08
US4401796A1983-08-30
US4415732A1983-11-15
US4458066A1984-07-03
US4500707A1985-02-19
US4668777A1987-05-26
US4973679A1990-11-27
US5047524A1991-09-10
US5132418A1992-07-21
US5153319A1992-10-06
US5262530A1993-11-16
US5700642A1997-12-23
US20160031928A12016-02-04
US8093367B22012-01-10
US8304529B22012-11-06
US4897355A1990-01-30
US5171678A1992-12-15
US5334761A1994-08-02
US5744335A1998-04-28
US5885613A1999-03-23
Other References:
S. M. BERGE ET AL., PHARMACEUTICALLY ACCEPTABLE SALTS IN DETAIL IN J. PHARMACEUTICAL SCIENCES, vol. 66, 1977, pages 1 - 19
JEMIELITY, J. ET AL.: "Novel 'anti-reverse' cap analogs with superior translational properties", RNA, vol. 9, 2003, pages 1108 - 1122, XP002466761, DOI: 10.1261/rna.5430403
JEMIELITY, J. ET AL., RNA, vol. 9, 2003, pages 1108 - 1122
GRUDZIEN, E. ET AL., RNA, vol. 10, 2004, pages 1479 - 1487
GRUDZIEN-NOGALSKA, E. ET AL., RNA, vol. 13, 2007, pages 1745 - 1755
YOKOE ET AL., NATURE BIOTECHNOLOGY, vol. 14, 1996, pages 1252 - 1256
GAO ET AL., BIOCHEM. BIOPHYS. RES. COMM., vol. 179, 1991, pages 280
LASIC, TRENDS BIOTECHNOL., vol. 16, 1998, pages 307 - 321
SEMPLE, S.C. ET AL.: "Rational Design of Cationic Lipids for siRNA Delivery", NATURE BIOTECH., vol. 141, 2010, pages 172 - 176
WHITEHEAD ET AL., NATURE COMMUNICATIONS, vol. 5, 2014, pages 4277
FEIGNER ET AL., PROC. NAT'L ACAD. SCI., vol. 84, 1987, pages 7413
BEHR ET AL., PROC. NAT.'I ACAD. SCI., vol. 86, 1989, pages 6982
HEYES, J. ET AL., J CONTROLLED RELEASE, vol. 107, 2005, pages 276 - 287
MORRISSEY, DV. ET AL., NAT. BIOTECHNOL., vol. 23, no. 8, 2005, pages 1003 - 1007
WOLF ET AL., BIOTECHNIQUES, vol. 23, 1997, pages 139
KLIBANOV ET AL., FEBS LETTERS, vol. 268, no. 1, 1990, pages 235 - 237
LUBKE ET AL.: "Proteomics of the Lysosome", BIOCHIM BIOPHYS ACTA, vol. 1793, 2009, pages 625 - 635, XP026073291, DOI: 10.1016/j.bbamcr.2008.09.018
FECHTER, P.BROWNLEE, G.G.: "Recognition of mRNA cap structures by viral and cellular proteins", J. GEN. VIROLOGY, vol. 86, 2005, pages 1239 - 1249, XP002516933, DOI: 10.1099/VIR.0.80755-0
HEYES, J.PALMER, L.BREMNER, K.MACLACHLAN, I.: "Cationic lipid saturation influences intracellular delivery of encapsulated nucleic acids", J. CONTR. REL., vol. 107, 2005, pages 276 - 287, XP008157522, DOI: 10.1016/j.jconrel.2005.06.014
LOVE, K.T. ET AL.: "Lipid-like materials for low-dose in vivo gene silencing", PNAS, vol. 107, 2010, pages 1864 - 1869, XP055077922, DOI: 10.1073/pnas.0910603106
Attorney, Agent or Firm:
ESPINO, Christine, G. et al. (US)
Download PDF:
Claims:
CLAIMS

WHAT IS CLAI MED IS:

1. A cationic lipid having a structure according to Formula (I):

wherein: each L1 is independently C -Ci -alkenylene;

each A1 is independently a linker group that is a covalent bond, O, S, N H, S-S, an amide, an ester, a thioester, or an anhydride group;

each R1 is an ionizable nitrogen-containing group;

each m is independently an integer of 6 to 20; and

each n is independently an integer of 1 to 6.

2. The cationic lipid of claim 1, wherein at least one m is 8.

3. The cationic lipid of claim 1, wherein each m is 8.

4. The cationic lipid of any one of claims 1-3, wherein each L1 is independently

-CH=CH-CH2- or -CH=CH-CH2-CH=CH-CH2-.

5. The cationic lipid of any one of claims 1-4, wherein each L1 is -CH=CH-CH -.

6. The cationic lipid of any one of claims 1-4, wherein each L1 is -CH=CH-CH -CH=CH-CH -.

7. The cationic lipid of any one of claims 1-6, wherein at least one A1 is a covalent bond.

8. The cationic lipid of any one of claims 1-6, wherein each A1 is a covalent bond.

9. The cationic lipid of any one of claims 1-6, wherein at least one A1 is O.

10. The cationic lipid of any one of claims 1-6 and 9, wherein each A1 is O.

11. The cationic lipid of any one of claims 1-6, wherein at least one A1 is OC(O).

12. The cationic lipid of any one of claims 1-6 and 11, wherein each A1 is OC(O).

13. The cationic lipid of any one of claims 1-12, wherein n is 2.

14. The cationic lipid of any one of claims 1-12, wherein n is 3.

15. The cationic lipid of any one of claims 1-14, wherein each R1 is independently selected from

16. The cationic lipid of any one of claims 1-4, having the structure selected from:

or a pharmaceutically acceptable salt thereof.

17. A composition comprising an mRNA encoding a protein, encapsulated within a liposome, wherein the liposome comprises a cationic lipid according to any one of claims 1-16.

18. The composition of claim 17, comprising an mRNA encoding for cystic fibrosis

transmembrane conductance regulator (CFTR) protein.

19. The composition of claim 17, comprising an mRNA encoding for ornithine transcarbamylase (OTC) protein.

20. A composition comprising a nucleic acid encapsulated within a liposome, wherein the liposome comprises a cationic lipid according to any one of claims 1-16.

21. The composition of claim 20, wherein the nucleic acid is an mRNA encoding a peptide or protein.

22. The composition of claim 21, wherein the mRNA encodes a peptide or protein for use in the delivery to or treatment of the lung of a subject or a lung cell.

23. The composition of claim 22, wherein the mRNA encodes cystic fibrosis transmembrane conductance regulator (CFTR) protein.

24. The composition of claim 21, wherein the mRNA encodes a peptide or protein for use in the delivery to or treatment of the liver of a subject or a liver cell.

25. The composition of claim 24, wherein the mRNA encodes ornithine transcarbamylase (OTC) protein.

26. The composition of claim 17 or 20, wherein the mRNA encodes a peptide or protein for use in a vaccine.

27. The composition of claim 26, wherein the mRNA encodes an antigen.

Description:
CATIONIC LIPID COMPOUNDS

RELATED APPLICATIONS

[0001] This application claims priority to U.S. Provisional Application Serial Number

62/770,309, filed on November 21, 2018, the entire disclosure of which is hereby incorporated by reference.

INCORPORATION BY REFERENCE OF SEQUENCE LISTING

[0002] The instant application contains a Sequence Listing that is submitted herewith electronically in ASCII format and is hereby incorcorporated by reference in its entirety. Said ASCII copy, created November 20, 2019, is 4,131 bytes in size.

BACKGROUND

[0003] Delivery of nucleic acids has been explored extensively as a potential therapeutic option for certain disease states. In particular, messenger RNA (mRNA) therapy has become an increasingly important option for treatment of various diseases, including for those associated with deficiency of one or more proteins.

SUMMARY

[0004] The present invention provides, among other things, compounds useful in for delivery of mRNA. Delivery of mRNA provided by compounds described herein can result in targeted delivery, reduce administration frequency, improve patient tolerability, and provide more potent and less toxic mRNA therapy for the treatment of a variety of diseases, including but not limited to cancer, cardiovascular, cystic fibrosis, infectious, and neurological diseases.

[0005] In an aspect, provided herein are cationic lipids having a structure according to

Formula (I):

wherein: each L 1 is independently C -Ci -alkenylene;

each A 1 is independently a linker group that is a covalent bond, O, S, NH, S-S, an amide, an ester, a thioester, or an anhydride group; each R 1 is an ionizable nitrogen-containing group;

each m is independently an integer of 6 to 20; and

each n is independently an integer of 1 to 6.

[0006] In embodiments, at least one m is 8.

[0007] In embodiments, each m is 8.

[0008] In embodiments, each L 1 is independently -CH=CH-CH - or -CH=CH-CH -CH=CH-CH -.

[0009] In embodiments, each L 1 is -CH=CH-CH -.

[0010] In embodiments, each L 1 is -CH=CH-CH -CH=CH-CH -.

[0011] In embodiments, at least one A 1 is a covalent bond.

[0012] In embodiments, each A 1 is a covalent bond.

[0013] In embodiments, at least one A 1 is 0.

[0014] In embodiments, each A 1 is 0.

[0015] In embodiments, at least one A 1 is OC(O).

[0016] In embodiments, each A 1 is OC(O).

[0017] In embodiments, n is 2.

[0018] In embodiments, n is 3.

[0019] In embodiments, each R 1 is independently selected from

[0020] In embodiments, the cationic lipid has a structure according to Compound (1),

or a pharmaceutically acceptable salt thereof.

[0021] In embodiments, the cationic lipid has a structure according to Compound (2),

or a pharmaceutically acceptable salt thereof.

[0022] In embodiments, the cationic lipid has a structure according to Compound (3),

or a pharmaceutically acceptable salt thereof.

[0023] In embodiments, the cationic lipid has a structure according to Compound (4),

or a pharmaceutically acceptable salt thereof.

[0024] In another aspect, the invention features a composition comprising any liposome

( e.g ., a liposome encapsulating an mRNA encoding a protein) described herein. [0025] In embodiments, an mRNA encodes for cystic fibrosis transmembrane conductance regulator (CFTR) protein.

[0026] In embodiments, an mRNA encodes for ornithine transcarbamylase (OTC) protein.

[0027] In another aspect, the invention features a composition comprising a nucleic acid encapsulated within a liposome as described herein.

[0028] In embodiments, a composition further comprises one more lipids selected from the group consisting of one or more cationic lipids, one or more non-cationic lipids, and one or more PEG-modified lipids.

[0029] In embodiments, a nucleic acid is an mRNA encoding a peptide or protein.

[0030] In embodiments, an mRNA encodes a peptide or protein for use in the delivery to or treatment of the lung of a subject or a lung cell.

[0031] In embodiments, an mRNA encodes for cystic fibrosis transmembrane conductance regulator (CFTR) protein.

[0032] In embodiments, an mRNA encodes a peptide or protein for use in the delivery to or treatment of the liver of a subject or a liver cell.

[0033] In embodiments, an mRNA encodes for ornithine transcarbamylase (OTC) protein.

[0034] In embodiments, an mRNA encodes a peptide or protein for use in vaccine.

[0035] In embodiments, an mRNA encodes an antigen.

[0036] In some aspects, the present invention provides methods of treating a disease in a subject comprising administering to the subject a composition as described herein.

BRIEF DESCRIPTION OF THE DRAWINGS

[0037] The drawings are for illustration purposes only, not for limitation.

[0038] FIG. 1 is a bar graph showing human EPO protein expression in mice dosed subcutaneously with hEPO mRNA encapsulated in LNP formulations comprising compound (3) or compound (4), respectively, compared to saline.

[0039] FIG. 2 shows FFL luciferase detection from FFL luciferase protein production in individual organs post-administration of mice dosed subcutaneously with FFL mRNA encapsulated in an LNP formulation comprising compound (4). DETAILED DESCRIPTION OF CERTAIN EMBODIMENTS

Definitions

[0040] In order for the present invention to be more readily understood, certain terms are first defined below. Additional definitions for the following terms and other terms are set forth throughout the specification. The publications and other reference materials referenced herein to describe the background of the invention and to provide additional detail regarding its practice are hereby incorporated by reference.

[0041] Amino acid: As used herein, the term "amino acid," in its broadest sense, refers to any compound and/or substance that can be incorporated into a polypeptide chain. In some embodiments, an amino acid has the general structure H2N-C(H)(R)-COOH. In some embodiments, an amino acid is a naturally occurring amino acid. In some embodiments, an amino acid is a synthetic amino acid; in some embodiments, an amino acid is a d-amino acid; in some embodiments, an amino acid is an l-amino acid. "Standard amino acid" refers to any of the twenty standard I- amino acids commonly found in naturally occurring peptides. "Nonstandard amino acid" refers to any amino acid, other than the standard amino acids, regardless of whether it is prepared synthetically or obtained from a natural source. As used herein, "synthetic amino acid"

encompasses chemically modified amino acids, including but not limited to salts, amino acid derivatives (such as amides), and/or substitutions. Amino acids, including carboxy- and/or amino- terminal amino acids in peptides, can be modified by methylation, amidation, acetylation, protecting groups, and/or substitution with other chemical groups that can change the peptide's circulating half-life without adversely affecting their activity. Amino acids may participate in a disulfide bond. Amino acids may comprise one or posttranslational modifications, such as association with one or more chemical entities ( e.g ., methyl groups, acetate groups, acetyl groups, phosphate groups, formyl moieties, isoprenoid groups, sulfate groups, polyethylene glycol moieties, lipid moieties, carbohydrate moieties, biotin moieties, etc.). The term "amino acid" is used interchangeably with "amino acid residue," and may refer to a free amino acid and/or to an amino acid residue of a peptide. It will be apparent from the context in which the term is used whether it refers to a free amino acid or a residue of a peptide.

[0042] Animal: As used herein, the term "animal" refers to any member of the animal kingdom. In some embodiments, "animal" refers to humans, at any stage of development. In some embodiments, "animal" refers to non-human animals, at any stage of development. In certain embodiments, the non-human animal is a mammal {e.g., a rodent, a mouse, a rat, a rabbit, a monkey, a dog, a cat, a sheep, cattle, a primate, and/or a pig). In some embodiments, animals include, but are not limited to, mammals, birds, reptiles, amphibians, fish, insects, and/or worms. In some embodiments, an animal may be a transgenic animal, genetically-engineered animal, and/or a clone.

[0043] Approximately or about: As used herein, the term "approximately" or "about," as applied to one or more values of interest, refers to a value that is similar to a stated reference value. In certain embodiments, the term "approximately" or "about" refers to a range of values that fall within 25%, 20%, 19%, 18%, 17%, 16%, 15%, 14%, 13%, 12%, 11%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, or less in either direction (greater than or less than) of the stated reference value unless otherwise stated or otherwise evident from the context (except where such number would exceed 100% of a possible value).

[0044] Biologically active: As used herein, the term "biologically active" refers to a characteristic of any agent that has activity in a biological system, and particularly in an organism.

For instance, an agent that, when administered to an organism, has a biological effect on that organism, is considered to be biologically active.

[0045] Delivery: As used herein, the term "delivery" encompasses both local and systemic delivery. For example, delivery of mRNA encompasses situations in which an mRNA is delivered to a target tissue and the encoded protein is expressed and retained within the target tissue (also referred to as "local distribution" or "local delivery"), and situations in which an mRNA is delivered to a target tissue and the encoded protein is expressed and secreted into patient's circulation system ( e.g ., serum) and systematically distributed and taken up by other tissues (also referred to as "systemic distribution" or "systemic delivery").

[0046] Expression: As used herein, "expression" of a nucleic acid sequence refers to translation of an mRNA into a polypeptide, assemble multiple polypeptides into an intact protein {e.g., enzyme) and/or post-translational modification of a polypeptide or fully assembled protein {e.g., enzyme). In this application, the terms "expression" and "production," and grammatical equivalent, are used inter-changeably.

[0047] Functional: As used herein, a "functional" biological molecule is a biological molecule in a form in which it exhibits a property and/or activity by which it is characterized.

[0048] Half-life: As used herein, the term "half-life" is the time required for a quantity such as nucleic acid or protein concentration or activity to fall to half of its value as measured at the beginning of a time period. [0049] Helper lipid: The term ' ' helper lipid "' as used herein refers to any neutral or zwitterionic lipid material including cholesterol. Without wishing to be held to a particular theory, helper lipids may add stability, rigidity, and/or fluidity within lipid bilayers/nanopartides.

[0050] Improve, increase, or reduce: As used herein, the terms "improve," "increase" or

"reduce," or grammatical equivalents, indicate values that are relative to a baseline measurement, such as a measurement in the same individual prior to initiation of the treatment described herein, or a measurement in a control subject (or multiple control subject) in the absence of the treatment described herein. A "control subject" is a subject afflicted with the same form of disease as the subject being treated, who is about the same age as the subject being treated.

[0051] In Vitro: As used herein, the term“in vitro" refers to events that occur in an artificial environment, e.g., in a test tube or reaction vessel, in cell culture, etc., rather than within a multi cellular organism.

[0052] In Vivo: As used herein, the term“in vivo" refers to events that occur within a multi cellular organism, such as a human and a non-human animal. In the context of cell-based systems, the term may be used to refer to events that occur within a living cell (as opposed to, for example, in vitro systems).

[0053] Isolated: As used herein, the term "isolated" refers to a substance and/or entity that has been (1) separated from at least some of the components with which it was associated when initially produced (whether in nature and/or in an experimental setting), and/or (2) produced, prepared, and/or manufactured by the hand of man. Isolated substances and/or entities may be separated from about 10%, about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, about 91%, about 92%, about 93%, about 94%, about 95%, about 96%, about 97%, about 98%, about 99%, or more than about 99% of the other components with which they were initially associated. In some embodiments, isolated agents are about 80%, about 85%, about 90%, about 91%, about 92%, about 93%, about 94%, about 95%, about 96%, about 97%, about 98%, about 99%, or more than about 99% pure. As used herein, a substance is "pure" if it is substantially free of other components. As used herein, calculation of percent purity of isolated substances and/or entities should not include excipients {e.g., buffer, solvent, water, etc. ).

[0054] Liposome: As used herein, the term "liposome" refers to any lamellar, multilamellar, or solid nanoparticle vesicle. Typically, a liposome as used herein can be formed by mixing one or more lipids or by mixing one or more lipids and polymer(s). I n some embodiments, a liposome suitable for the present invention contains a cationic lipids(s) and optionally non-cationic lipid(s), optionally cholesterol-based lipid(s), and/or optionally PEG-modified lipid(s). [0055] messenger RNA (mRNA): As used herein, the term "messenger RNA (mRNA)" or

"mRNA" refers to a polynucleotide that encodes at least one polypeptide. mRNA as used herein encompasses both modified and unmodified RNA. The term "modified mRNA" relates to mRNA comprising at least one chemically modified nucleotide. mRNA may contain one or more coding and non-coding regions. mRNA can be purified from natural sources, produced using recombinant expression systems and optionally purified, chemically synthesized, etc. Where appropriate, e.g., in the case of chemically synthesized molecules, mRNA can comprise nucleoside analogs such as analogs having chemically modified bases or sugars, backbone modifications, etc. An mRNA sequence is presented in the 5' to 3' direction unless otherwise indicated. In some embodiments, an mRNA is or comprises natural nucleosides {e.g., adenosine, guanosine, cytidine, uridine); nucleoside analogs {e.g., 2-aminoadenosine, 2-thiothymidine, inosine, pyrrolo-pyrimidine, 3-methyl adenosine, 5-methylcytidine, C-5 propynyl-cytidine, C-5 propynyl-uridine, 2-aminoadenosine, C5-bromouridine, C5-fluorouridine, C5-iodouridine, C5-propynyl-uridine, C5-propynyl-cytidine, C5-methylcytidine, 2- aminoadenosine, 7-deazaadenosine, 7-deazaguanosine, 8-oxoadenosine, 8-oxoguanosine, 0(6)- methylguanine, and 2-thiocytidine); chemically modified bases; biologically modified bases {e.g., methylated bases); intercalated bases; modified sugars {e.g., 2'-fluororibose, ribose, 2'-deoxyribose, arabinose, and hexose); and/or modified phosphate groups {e.g., phosphorothioates and S'-N- phosphoramidite linkages).

[0056] Nucleic acid: As used herein, the term "nucleic acid," in its broadest sense, refers to any compound and/or substance that is or can be incorporated into a polynucleotide chain. In some embodiments, a nucleic acid is a compound and/or substance that is or can be incorporated into a polynucleotide chain via a phosphodiester linkage. In some embodiments, "nucleic acid" refers to individual nucleic acid residues {e.g., nucleotides and/or nucleosides). In some embodiments, "nucleic acid" refers to a polynucleotide chain comprising individual nucleic acid residues. In some embodiments, "nucleic acid" encompasses RNA as well as single and/or double-stranded DNA and/or cDNA. In some embodiments, "nucleic acid" encompasses ribonucleic acids (RNA), including but not limited to any one or more of interference RNAs (RNAi), small interfering RNA (si RNA), short hairpin RNA (shRNA), antisense RNA (aRNA), messenger RNA (mRNA), modified messenger RNA (mmRNA), long non-coding RNA (IncRNA), micro-RNA (miRNA) multimeric coding nucleic acid (MCNA), polymeric coding nucleic acid (PCNA), guide RNA (gRNA) and CRISPR RNA (crRNA). In some embodiments, "nucleic acid" encompasses deoxyribonucleic acid (DNA), including but not limited to any one or more of single-stranded DNA (ssDNA), double-stranded DNA (dsDNA) and

complementary DNA (cDNA). In some embodiments, "nucleic acid" encompasses both RNA and DNA. In embodiments, DNA may be in the form of antisense DNA, plasmid DNA, parts of a plasmid DNA, pre-condensed DNA, a product of a polymerase chain reaction (PCR), vectors ( e.g ., PI, PAC, BAC, YAC, artificial chromosomes), expression cassettes, chimeric sequences, chromosomal DNA, or derivatives of these groups. In embodiments, RNA may be in the form of messenger RNA (mRNA), ribosomal RNA (rRNA), signal recognition particle RNA (7 SL RNA or SRP RNA), transfer RNA (tRNA), transfer-messenger RNA (tmRNA), small nuclear RNA (snRNA), small nucleolar RNA (snoRNA), SmY RNA, small Cajal body-specific RNA (scaRNA), guide RNA (gRNA), ribonuclease P (RNase P), Y RNA, telomerase RNA component (TERC), spliced leader RNA (SL RNA), antisense RNA (aRNA or asRNA), cis-natural antisense transcript (cis-NAT), CRISPR RNA (crRNA), long noncoding RNA (IncRNA), micro- RNA (miRNA), piwi-interacting RNA (piRNA), small interfering RNA (si RNA), transacting siRNA (tasiRNA), repeat associated siRNA (rasiRNA), 73K RNA, retrotransposons, a viral genome, a viroid, satellite RNA, or derivatives of these groups. In some embodiments, a nucleic acid is a mRNA encoding a protein such as an enzyme.

[0057] Patient: As used herein, the term "patient" or "subject" refers to any organism to which a provided composition may be administered, e.g., for experimental, diagnostic, prophylactic, cosmetic, and/or therapeutic purposes. Typical patients include animals {e.g., mammals such as mice, rats, rabbits, non-human primates, and/or humans). In some embodiments, a patient is a human. A human includes pre- and post-natal forms.

[0058] Pharmaceutically acceptable: The term "pharmaceutically acceptable", as used herein, refers to substances that, within the scope of sound medical judgment, are suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.

[0059] Pharmaceutically acceptable salt: Pharmaceutically acceptable salts are well known in the art. For example, S. M. Berge et al., describes pharmaceutically acceptable salts in detail in J. Pharmaceutical Sciences (1977) 66:1-19. Pharmaceutically acceptable salts of the compounds of this invention include those derived from suitable inorganic and organic acids and bases. Examples of pharmaceutically acceptable, nontoxic acid addition salts are salts of an amino group formed with inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid or with organic acids such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid or rnalonic acid or by using other methods used in the art such as ion exchange. Other pharmaceutically acceptable salts include adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, formate, fumarate, glucoheptonate, glycerophosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2- hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectinate, persulfate, 3-phenylpropionate, phosphate, picrate, pivalate, propionate, stearate, succinate, sulfate, tartrate, thiocyanate, p-toluenesulfonate, undecanoate, valerate salts, and the like. Salts derived from appropriate bases include alkali metal, alkaline earth metal, ammonium and N + (C I -4 alkyl)4 salts. Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like. Further pharmaceutically acceptable salts include, when appropriate, nontoxic ammonium, quaternary ammonium, and amine cations formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, sulfonate and aryl sulfonate. Further pharmaceutically acceptable salts include salts formed from the quarternization of an amine using an appropriate electrophile, e.g., an alkyl halide, to form a quarternized alkylated amino salt.

[0060] Systemic distribution or delivery: As used herein, the terms "systemic distribution,"

"systemic delivery," or grammatical equivalent, refer to a delivery or distribution mechanism or approach that affect the entire body or an entire organism. Typically, systemic distribution or delivery is accomplished via body's circulation system, e.g., blood stream. Compared to the definition of "local distribution or delivery."

[0061] Subject: As used herein, the term "subject" refers to a human or any non-human animal {e.g., mouse, rat, rabbit, dog, cat, cattle, swine, sheep, horse or primate). A human includes pre- and post-natal forms. In many embodiments, a subject is a human being. A subject can be a patient, which refers to a human presenting to a medical provider for diagnosis or treatment of a disease. The term "subject" is used herein interchangeably with "individual" or "patient." A subject can be afflicted with or is susceptible to a disease or disorder but may or may not display symptoms of the disease or disorder.

[0062] Substantially: As used herein, the term "substantially" refers to the qualitative condition of exhibiting total or near-total extent or degree of a characteristic or property of interest. One of ordinary skill in the biological arts will understand that biological and chemical phenomena rarely, if ever, go to completion and/or proceed to completeness or achieve or avoid an absolute result. The term "substantially" is therefore used herein to capture the potential lack of completeness inherent in many biological and chemical phenomena.

[0063] Target tissues: As used herein, the term "target tissues" refers to any tissue that is affected by a disease to be treated. In some embodiments, target tissues include those tissues that display disease-associated pathology, symptom, or feature. [0064] Therapeutically effective amount As used herein, the term "therapeutically effective amount" of a therapeutic agent means an amount that is sufficient, when administered to a subject suffering from or susceptible to a disease, disorder, and/or condition, to treat, diagnose, prevent, and/or delay the onset of the symptom(s) of the disease, disorder, and/or condition. It will be appreciated by those of ordinary skill in the art that a therapeutically effective amount is typically administered via a dosing regimen comprising at least one unit dose.

[0065] Treating: As used herein, the term "treat," "treatment," or "treating" refers to any method used to partially or completely alleviate, ameliorate, relieve, inhibit, prevent, delay onset of, reduce severity of and/or reduce incidence of one or more symptoms or features of a particular disease, disorder, and/or condition. Treatment may be administered to a subject who does not exhibit signs of a disease and/or exhibits only early signs of the disease for the purpose of decreasing the risk of developing pathology associated with the disease.

[0066] Aliphatic: As used herein, the term aliphatic refers to C 1 -C 40 hydrocarbons and includes both saturated and unsaturated hydrocarbons. An aliphatic may be linear, branched, or cyclic. For example, C 1 -C 20 aliphatics can include C 1 -C 20 alkyls ( ., linear or branched C 1 -C 20 saturated alkyls), C 2 -C 20 alkenyls {e.g., linear or branched C 4 -C 20 dienyls, linear or branched C 6 -C 20 trienyls, and the like), and C 2 -C 20 alkynyls {e.g., linear or branched C 2 -C 20 alkynyls). C 1 -C 20 aliphatics can include C 3 -C 20 cyclic aliphatics {e.g., C 3 -C 20 cycloalkyls, C 4 -C 20 cycloalkenyls, or C 8 -C 20

cycloalkynyls). In certain embodiments, the aliphatic may comprise one or more cyclic aliphatic and/or one or more heteroatoms such as oxygen, nitrogen, or sulfur and may optionally be substituted with one or more substituents such as alkyl, halo, alkoxyl, hydroxy, amino, aryl, ether, ester or amide. An aliphatic group is unsubstituted or substituted with one or more substituent groups as described herein. For example, an aliphatic may be substituted with one or more {e.g., 1, 2, 3, 4, 5, or 6 independently selected substituents) of halogen, -COR', -CO 2 FI, -CO 2 R', -CN, -OFI, -OR', -OCOR', -OCO 2 R', -NH 2 ,

-NFIR', -N(R') 2 , -SR' or-S0 2 R', wherein each instance of R' independently is C 1 -C 20 aliphatic {e.g., C 1 -C 20 alkyl, C 1 -C 15 alkyl, C 1 -C 10 alkyl, or C 1 -C 3 alkyl). In embodiments, R' independently is an unsubstituted alkyl {e.g., unsubstituted C 1 -C 20 alkyl, C 1 -C 15 alkyl, C 1 -C 10 alkyl, or C 1 -C 3 alkyl). In embodiments, R' independently is unsubstituted C 1 -C 3 alkyl. In embodiments, the aliphatic is unsubstituted. In embodiments, the aliphatic does not include any heteroatoms.

[0067] Alkyl: As used herein, the term "alkyl" means acyclic linear and branched hydrocarbon groups, e.g. "C 1 -C 20 alkyl" refers to alkyl groups having 1-20 carbons. An alkyl group may be linear or branched. Examples of alkyl groups include, but are not limited to, methyl, ethyl, n- propyl, isopropyl, butyl, isobutyl, sec-butyl, tert-butyl, pentyl, isopentyl tert-pentylhexyl, Isohexyletc. Other alkyl groups will be readily apparent to those of skill in the art given the benefit of the present disclosure. An alkyl group may be unsubstituted or substituted with one or more substituent groups as described herein. For example, an alkyl group may be substituted with one or more ( e.g ., 1, 2, 3, 4, 5, or 6 independently selected substituents) of halogen, -COR', -CO 2 H, -CO 2 R', -CN, -OH, -OR', -OCOR', -OCO 2 R', -IMH 2 , -NHR', -N(R') 2 , -SR' or-S0 2 R', wherein each instance of R' independently is Ci- C 20 aliphatic {e.g., C 1 -C 20 alkyl, C 1 -C 15 alkyl, C 1 -C 10 alkyl, or C 1 -C 3 alkyl). In embodiments, R' independently is an unsubstituted alkyl {e.g., unsubstituted C 1 -C 20 alkyl, C 1 -C 15 alkyl, C 1 -C 10 alkyl, or C 1 -C 3 alkyl). In embodiments, R' independently is unsubstituted C 1 -C 3 alkyl. In embodiments, the alkyl is substituted {e.g., with 1, 2, 3, 4, 5, or 6 substituent groups as described herein). In embodiments, an alkyl group is substituted with a-OH group and may also be referred to herein as a "hydroxyalkyl" group, where the prefix denotes the -OH group and "alkyl" is as described herein.

[0068] Alkylene: Affixing the suffix "-ene" to a group indicates the group is a divalent moiety. The term "alkylene," as used herein, represents a saturated divalent straight or branched chain hydrocarbon group and is exemplified by methylene, ethylene, isopropylene and the like. Likewise, the term "alkenylene" as used herein represents an unsaturated divalent straight or branched chain hydrocarbon group having one or more unsaturated carbon-carbon double bonds that may occur in any stable point along the chain, and the term "alkynylene" herein represents an unsaturated divalent straight or branched chain hydrocarbon group having one or more unsaturated carbon-carbon triple bonds that may occur in any stable point along the chain. In certain embodiments, an alkylene, alkenylene, or alkynylene group may comprise one or more cyclic aliphatic and/or one or more heteroatoms such as oxygen, nitrogen, or sulfur and may optionally be substituted with one or more substituents such as alkyl, halo, alkoxyl, hydroxy, amino, aryl, ether, ester or amide. For example, an alkylene, alkenylene, or alkynylene may be substituted with one or more {e.g., 1, 2, 3, 4, 5, or 6 independently selected substituents) of halogen, -COR', -CO 2 H, -CO 2 R', - CN, -OH, -OR', -OCOR', -OC0 2 R', -NH 2 , -NHR', -N(R') 2 , -SR' or-S0 2 R', wherein each instance of R' independently is C 1 -C 20 aliphatic {e.g., C 1 -C 20 alkyl, C 1 -C 15 alkyl, C 1 -C 10 alkyl, or C 1 -C 3 alkyl). In embodiments, R' independently is an unsubstituted alkyl {e.g., unsubstituted C 1 -C 20 alkyl, C 1 -C 15 alkyl, C 1 -C 10 alkyl, or C 1 -C 3 alkyl). In embodiments, R' independently is unsubstituted C 1 -C 3 alkyl. In certain embodiments, an alkylene, alkenylene, or alkynylene is unsubstituted. In certain embodiments, an alkylene, alkenylene, or alkynylene does not include any heteroatoms.

[0069] Alkenyl: As used herein, "alkenyl" means any linear or branched hydrocarbon chains having one or more unsaturated carbon-carbon double bonds that may occur in any stable point along the chain, e.g. "C2-C20 alkenyl" refers to an alkenyl group having 2-20 carbons. For example, an alkenyl group includes prop-2-enyl, but-2-enyl, but-3-enyl, 2-methylprop-2-enyl, hex-2-enyl, hex-5- enyl, 2,3-dimethylbut-2-enyl, and the like. In embodiments, the alkenyl comprises 1, 2, or 3 carbon- carbon double bond. In embodiments, the alkenyl comprises a single carbon-carbon double bond.

In embodiments, multiple double bonds ( e.g ., 2 or 3) are conjugated. An alkenyl group may be unsubstituted or substituted with one or more substituent groups as described herein. For example, an alkenyl group may be substituted with one or more {e.g., 1, 2, 3, 4, 5, or 6 independently selected substituents) of halogen, -COR', -C0 2 H, -C0 2 R', -CN, -OH, -OR', -OCOR', -0C0 2 R', -NH 2 , -NHR', -N(R') 2 , -SR' or-S0 2 R', wherein each instance of R' independently is Ci-C 2 o aliphatic {e.g., Ci-C 2 o alkyl, C 1 -C 15 alkyl, Ci-Cio alkyl, or C 1 -C 3 alkyl). In embodiments, R' independently is an unsubstituted alkyl {e.g., unsubstituted Ci-C 2 o alkyl, C 1 -C 15 alkyl, C 1 -C 10 alkyl, or C 1 -C 3 alkyl). In embodiments, R' independently is unsubstituted C 1 -C 3 alkyl. In embodiments, the alkenyl is unsubstituted. In embodiments, the alkenyl is substituted {e.g., with 1, 2, 3, 4, 5, or 6 substituent groups as described herein). In embodiments, an alkenyl group is substituted with a-OH group and may also be referred to herein as a "hydroxyalkenyl" group, where the prefix denotes the -OH group and "alkenyl" is as described herein.

[0070] Alkynyl: As used herein, "alkynyl" means any hydrocarbon chain of either linear or branched configuration, having one or more carbon-carbon triple bonds occurring in any stable point along the chain, e.g. "C 2 -C 2 o alkynyl" refers to an alkynyl group having 2-20 carbons. Examples of an alkynyl group include prop-2-ynyl, but-2-ynyl, but-3-ynyl, pent-2-ynyl, 3-methylpent-4-ynyl, hex-2- ynyl, hex-5-ynyl, etc. In embodiments, an alkynyl comprises one carbon-carbon triple bond. An alkynyl group may be unsubstituted or substituted with one or more substituent groups as described herein. For example, an alkynyl group may be substituted with one or more {e.g., 1, 2, 3, 4, 5, or 6 independently selected substituents) of halogen, -COR', -C0 2 H, -C0 2 R', -CN, -OH, -OR', -OCOR', -OC0 2 R', -NH 2 , -NHR', -N(R') 2 , -SR' or-S0 2 R', wherein each instance of R' independently is Ci-C 2 o aliphatic {e.g., Ci-C 2 o alkyl, C 1 -C 15 alkyl, C 1 -C 10 alkyl, or C 1 -C 3 alkyl). In embodiments, R' independently is an unsubstituted alkyl {e.g., unsubstituted Ci-C 2 o alkyl, C 1 -C 15 alkyl, C 1 -C 10 alkyl, or C 1 -C 3 alkyl). In embodiments, R' independently is unsubstituted C 1 -C 3 alkyl. In embodiments, the alkynyl is unsubstituted. In embodiments, the alkynyl is substituted {e.g., with 1, 2, 3, 4, 5, or 6 substituent groups as described herein).

[0071] Amino and alkylamino: As used herein, "amino" refers to groups of the form—

N(R') 2 wherein each R' is independently selected from hydrogen, alkyl, alkenyl, alkynyl, and aryl as described herein. "Alkylamino" includes both mono-alkylamino and dialkylamino, unless specified. "Mono-alkylamino" means an -NH(alkyl) group, in which alkyl is as defined herein. "Dialkylamino" means an -N(alkyl) 2 group, in which each alkyl may be the same or different and are each as defined herein for alkyl. In embodiments, an alkyl group is a Ci-C 6 alkyl group. The group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the nitrogen atom.

[0072] Amide: As used herein, the term "amide" refers to a group having

the amide functional group:

An amide group may have 1, 2, or 3 points of attachment to the molecule. Exemplary amide groups include -C(0)N(R') 2 , -C(0)NH R', -C(0)NH 2 , -C(0)NH- ,-C(0)NR'-, -NHC(0)-,and -NR'C(O)-, wherein each instance of R' independently is Ci-C 2 o aliphatic ( e.g ., Ci-C 2 o alkyl, C 1 -C 15 alkyl, Ci-Cio alkyl, or C 1 -C 3 alkyl), or two R' can combine to form a 3- to 10-membered nitrogen-containing heterocyclyl. In embodiments, R' independently is an unsubstituted alkyl {e.g., unsubstituted Ci- C 2 o alkyl, C 1 -C 15 alkyl, C 1 -C 10 alkyl, or C 1 -C 3 alkyl). In embodiments, R' independently is unsubstituted C 1 -C 3 alkyl. In embodiments, the alkenyl is unsubstituted. In embodiments, the alkenyl is substituted {e.g., with 1, 2, 3, 4, 5, or 6 substituent groups as described herein). In embodiments, an alkenyl group is substituted with a-OH group and may also be referred to herein as a "hydroxyalkenyl" group, where the prefix denotes the -OH group and "alkenyl" is as described herein.

[0073] Anhydride: The term "anhydride" linkage is characterized by two acyl groups joined by an oxygen atom, having the general structure:

[0074] Ester and Thioester: The term ester linkage refers to— OC(=0)— or— C(=0)0— ; the term thioester linkage refers to— SC(=0)— or— C(=0)S— .

[0075] Halogen: As used herein, the term "halogen" means fluorine, chlorine, bromine, or iodine.

[0076] Heteroalkyl: The term "heteroalkyl" is meant a branched or unbranched alkyl, alkenyl, or alkynyl group having from 1 to 14 carbon atoms in addition to 1, 2, 3 or 4 heteroatoms independently selected from the group consisting of N, O, S, and P. Heteroalkyls include tertiary amines, secondary amines, ethers, thioethers, amides, thioamides, carbamates, thiocarbamates, hydrazones, imines, phosphodiesters, phosphoramidates, sulfonamides, and disulfides. A heteroalkyl group may optionally include monocyclic, bicyclic, or tricyclic rings, in which each ring desirably has three to six members. Examples of heteroalkyls include polyethers, such as methoxymethyl and ethoxyethyl.

[0077] Heteroalkylene: The term "heteroalkylene," as used herein, represents a divalent form of a heteroalkyl group as described herein.

[0078] Heterocyclyl: As used herein, the terms "heterocycle", "heterocyclyl", "heterocyclic radical", and "heterocyclic ring" are used interchangeably and refer to a stable 3- to 8-membered monocyclic or 7-10-membered bicyclic heterocyclic moiety that is either saturated or partially unsaturated, and having, in addition to carbon atoms, one or more, such as one to four, heteroatoms, as defined above. When used in reference to a ring atom of a heterocycle, the term "nitrogen" includes a substituted nitrogen. As an example, in a saturated or partially unsaturated ring having 0-3 heteroatoms selected from oxygen, sulfur or nitrogen, the nitrogen may be N (as in 3,4-dihydro-2H-pyrrolyl), NH (as in pyrrolidinyl), or N R + (as in N-substituted pyrrolidinyl).

Compounds of the Invention

[0079] Liposomal-based vehicles are considered an attractive carrier for therapeutic agents and remain subject to continued development efforts. While liposomal-based vehicles that comprise certain lipid components have shown promising results with regards to encapsulation, stability and site localization, there remains a great need for improvement of liposomal-based delivery systems. For example, a significant drawback of liposomal delivery systems relates to the construction of liposomes that have sufficient cell culture or in vivo stability to reach desired target cells and/or intracellular compartments, and the ability of such liposomal delivery systems to efficiently release their encapsulated materials to such target cells.

[0080] In particular, there remains a need for improved lipids compounds that demonstrate improved pharmacokinetic properties and which are capable of delivering macromolecules, such as nucleic acids to a wide variety cell types and tissues with enhanced efficiency. Importantly, there also remains a particular need for novel lipid compounds that are characterized as having reduced toxicity and are capable of efficiently delivering encapsulated nucleic acids and polynucleotides to targeted cells, tissues and organs.

[0081] Described herein a novel class of cationic lipid compounds for improved in vivo delivery of therapeutic agents, such as nucleic acids. In particular, a biodegradable compound described herein may be used to as a cationic lipid, together with other non-cationic lipids, to formulate a lipid based nanoparticle (e.g., liposome) for encapsulating therapeutic agents, such as nucleic acids (e.g., DNA, siRNA, mRNA, microRNA) for therapeutic use. [0082] In embodiments, compounds described herein can provide one or more desired characteristics or properties. That is, in certain embodiments, compounds described herein can be characterized as having one or more properties that afford such compounds advantages relative to other similarly classified lipids. For example, compounds disclosed herein can allow for the control and tailoring of the properties of liposomal compositions ( e.g ., lipid nanoparticles) of which they are a component. In particular, compounds disclosed herein can be characterized by enhanced transfection efficiencies and their ability to provoke specific biological outcomes. Such outcomes can include, for example enhanced cellular uptake, endosomal/lysosomal disruption capabilities and/or promoting the release of encapsulated materials {e.g., polynucleotides) intracellularly. Additionally, the compounds disclosed herein have advantageous pharmacokinetic properties, biodistribution, and efficiency {e.g., due to the different disassociate rates of the polymer group used).

Compounds of Formula (I)

[0083] Provided herein are cationic lipids having a structure according to Formula (I):

wherein: each L 1 is independently C 2 -Ci 2 -alkenylene;

each A 1 is independently a linker group that is a covalent bond, 0, S, NH, S-S, an amide, an ester, a thioester, or an anhydride group;

each R 1 is an ionizable nitrogen-containing group;

each m is independently an integer of 6 to 20; and

each n is independently an integer of 1 to 6.

[0084] In embodiments, L 1 is independently C 2 -Ci 2 -alkenylene comprising one carbon- carbon double bond. In embodiments, L 1 is independently C 6 -Ci 2 -alkenylene comprising one carbon-carbon double bond or two carbon-carbon double bonds. In embodiments, L 1 comprises one carbon-carbon double bond. In embodiments, L 1 comprises two carbon-carbon double bonds.

[0085] In embodiments, each L 1 is independently -CH=CH-CH 2 - or -CH=CH-CH 2 -CH=CH-CH 2 -.

[0086] In embodiments, each L 1 is -CH=CH-CH 2 -. [0087] In embodiments, each L 1 is -CH=CH-CH -CH=CH-CH -.

[0088] In embodiments, at least one A 1 is a covalent bond. In embodiments, each A 1 is a covalent bond.

[0089] In embodiments, at least one A 1 is NH. In embodiments, each A 1 is NH.

[0090] In embodiments, at least one A 1 is S-S. In embodiments, each A 1 is S-S.

[0091] In embodiments, at least one A 1 is 0. In embodiments, each A 1 is 0.

[0092] In embodiments, at least one A 1 is an amide. In embodiments, each A 1 is an amide.

In embodiments, at least one A 1 is NHC(O) or (O)CNH. In embodiments, at least one A 1 is NHC(O). In embodiments, each A 1 is NHC(O) or (O)CNH. In embodiments, each A 1 is NHC(O).

[0093] In embodiments, at least one A 1 is an ester. In embodiments, each A 1 is an ester. In embodiments, at least one A 1 is OC(O) or (O)CO. In embodiments, at least one A 1 is OC(O). In embodiments, each A 1 is OC(O) or (O)CO. In embodiments, each A 1 is OC(O).

[0094] In embodiments, at least one A 1 is a thioester. In embodiments, each A 1 is a thioester. In embodiments, at least one A 1 is OC(S) or (S)CO. In embodiments, at least one A 1 is OC(S). In embodiments, each A 1 is OC(S) or (S)CO. In embodiments, each A 1 is OC(S).

[0095] In embodiments, at least one A 1 is an anhydride. In embodiments, each A 1 is an anhydride.

[0096] In embodiments, at least one m is 6 or each m is 6. In embodiments, at least one m is 7 or each m is 7. In embodiments, at least one m is 8 or each m is 8. In embodiments, at least one m is 9 or each m is 9. In embodiments, at least one m is 10 or each m is 10. In embodiments, at least one m is 11 or each m is 11. In embodiments, at least one m is 12 or each m is 12. In embodiments, at least one m is 13 or each m is 13. In embodiments, at least one m is 14 or each m is 14. In embodiments, at least one m is 15 or each m is 15. In embodiments, at least one m is 16 or each m is 16. In embodiments, at least one m is 17 or each m is 17. In embodiments, at least one m is 18 or each m is 18. In embodiments, at least one m is 19 or each m is 19. In embodiments, at least one m is 20 or each m is 20.

[0097] In embodiments, at least one m is 8. In embodiments, each m is 8.

[0098] In embodiments, at least one n is 1 or each n is 1. In embodiments, at least one n is

2 or each n is 2. In embodiments, at least one n is 3 or each n is 3. In embodiments, at least one n is

4 or each n is 4. In embodiments, at least one n is 5 or each n is 5. In embodiments, at least one n is

6 or each n is 6. [0099] In embodiments, each n is 2.

[0100] In embodiments, each n is 3.

[0101] In embodiments, each R 1 is independently an ionizable nitrogen-containing group that is imidazole, guanidinium, imine, enamine, amino, an optionally-substituted alkyl amino (e.g., an alkyl amino such as dimethylamino) and an optionally-substituted pyridine (e.g., pyridine or nitropyridine).

H. ¾ Me x

, N— I /H

[0102] In embodiments, each R 1 is independently selected from H Me

[ ,

Exemplary Compounds

[0104] Exemplary compounds of the invention include Compounds (1), (2) (3), and (4),

or a pharmaceutically acceptable salt thereof.

Synthesis of Compounds of the Invention

[0105] The compounds described herein ( e.g a compound of Formula (I) such as

Compounds (1)— (4)) can be prepared according to methods known in the art.

[0106] For example, compound (3) may be prepared according to the method shown in

Scheme 1.

Scheme 1.

[0107] For example, compound (4) may be prepared according to the method shown in

Scheme 2.

Nucleic Acids

[0108] The compounds described herein ( e.g ., a compound of Formula (I) such as

Compounds (1)— (4)) can be used to prepare compositions useful for the delivery of nucleic acids.

Synthesis of Nucleic Acids

[0109] Nucleic acids according to the present invention may be synthesized according to any known methods. For example, mRNAs according to the present invention may be synthesized via in vitro transcription (IVT). Briefly, IVT is typically performed with a linear or circular DNA template containing a promoter, a pool of ribonucleotide triphosphates, a buffer system that may include DTT and magnesium ions, and an appropriate RNA polymerase ( e.g ., T3, T7, mutated T7 or SP6 RNA polymerase), DNAse I, pyrophosphatase, and/or RNAse inhibitor. The exact conditions will vary according to the specific application.

[0110] In some embodiments, for the preparation of mRNA according to the invention, a

DNA template is transcribed in vitro. A suitable DNA template typically has a promoter, for example a T3, T7, mutated T7 or SP6 promoter, for in vitro transcription, followed by desired nucleotide sequence for desired mRNA and a termination signal.

[0111] Desired mRNA sequence(s) according to the invention may be determined and incorporated into a DNA template using standard methods. For example, starting from a desired amino acid sequence {e.g., an enzyme sequence), a virtual reverse translation is carried out based on the degenerated genetic code. Optimization algorithms may then be used for selection of suitable codons. Typically, the G/C content can be optimized to achieve the highest possible G/C content on one hand, taking into the best possible account the frequency of the tRNAs according to codon usage on the other hand. The optimized RNA sequence can be established and displayed, for example, with the aid of an appropriate display device and compared with the original (wild-type) sequence. A secondary structure can also be analyzed to calculate stabilizing and destabilizing properties or, respectively, regions of the RNA.

[0112] As described above, the term "nucleic acid," in its broadest sense, refers to any compound and/or substance that is or can be incorporated into a polynucleotide chain. DNA may be in the form of antisense DNA, plasmid DNA, parts of a plasmid DNA, pre-condensed DNA, a product of a polymerase chain reaction (PCR), vectors {e.g., PI, PAC, BAC, YAC, artificial chromosomes), expression cassettes, chimeric sequences, chromosomal DNA, or derivatives of these groups. RNA may be in the form of messenger RNA (mRNA), ribosomal RNA (rRNA), signal recognition particle RNA (7 SL RNA or SRP RNA), transfer RNA (tRNA), transfer-messenger RNA (tmRNA), small nuclear RNA (snRNA), small nucleolar RNA (snoRNA), SmY RNA, small Cajal body-specific RNA (scaRNA), guide RNA (gRNA), ribonuclease P (RNase P), Y RNA, telomerase RNA component (TERC), spliced leader RNA (SL RNA), antisense RNA (aRNA or asRNA), cis-natural antisense transcript (cis-NAT), CRISPR RNA (crRNA), long noncoding RNA (IncRNA), microRNA (miRNA), piwi-interacting RNA (pi RNA), small interfering RNA (si RNA), transacting siRNA (tasiRNA), repeat associated siRNA (rasiRNA), 73K RNA, retrotransposons, a viral genome, a viroid, satellite RNA, or derivatives of these groups. In some embodiments, a nucleic acid is a mRNA encoding a protein. Synthesis of mRNA

[0113] mRNAs according to the present invention may be synthesized according to any of a variety of known methods. For example, mRNAs according to the present invention may be synthesized via in vitro transcription (IVT). Briefly, IVT is typically performed with a linear or circular DNA template containing a promoter, a pool of ribonucleotide triphosphates, a buffer system that may include DTT and magnesium ions, and an appropriate RNA polymerase ( e.g ., T3, T7 or SP6 RNA polymerase), DNAse I, pyrophosphatase, and/or RNAse inhibitor. The exact conditions will vary according to the specific application. The exact conditions will vary according to the specific application. The presence of these reagents is undesirable in the final product according to several embodiments and may thus be referred to as impurities and a preparation containing one or more of these impurities may be referred to as an impure preparation. In some embodiments, the in vitro transcribing occurs in a single batch.

[0114] In some embodiments, for the preparation of mRNA according to the invention, a

DNA template is transcribed in vitro. A suitable DNA template typically has a promoter, for example a T3, T7 or SP6 promoter, for in vitro transcription, followed by desired nucleotide sequence for desired mRNA and a termination signal.

[0115] Desired mRNA sequence(s) according to the invention may be determined and incorporated into a DNA template using standard methods. For example, starting from a desired amino acid sequence {e.g., an enzyme sequence), a virtual reverse translation is carried out based on the degenerated genetic code. Optimization algorithms may then be used for selection of suitable codons. Typically, the G/C content can be optimized to achieve the highest possible G/C content on one hand, taking into the best possible account the frequency of the tRNAs according to codon usage on the other hand. The optimized RNA sequence can be established and displayed, for example, with the aid of an appropriate display device and compared with the original (wild-type) sequence. A secondary structure can also be analyzed to calculate stabilizing and destabilizing properties or, respectively, regions of the RNA.

Modified mRNA

[0116] In some embodiments, mRNA according to the present invention may be synthesized as unmodified or modified mRNA. Modified mRNA comprise nucleotide modifications in the RNA. A modified mRNA according to the invention can thus include nucleotide modification that are, for example, backbone modifications, sugar modifications or base modifications. In some embodiments, mRNAs may be synthesized from naturally occurring nucleotides and/or nucleotide analogues (modified nucleotides) including, but not limited to, purines (adenine (A), guanine (G)) or pyrimidines (thymine (T), cytosine (C), uracil (U)), and as modified nucleotides analogues or derivatives of purines and pyrimidines, such as e.g. 1-methyl-adenine, 2-methyl-adenine, 2- methylthio-N-6-isopentenyl-adenine, N6-methyl-adenine, N6-isopentenyl-adenine, 2-thio-cytosine, 3-methyl-cytosine, 4-acetyl-cytosine, 5-methyl-cytosine, 2,6-diaminopurine, 1-methyl-guanine, 2- methyl-guanine, 2,2-dimethyl-guanine, 7-methyl-guanine, inosine, 1-methyl-inosine, pseudouracil (5-uracil), dihydro-uracil, 2-thio-uracil, 4-thio-uracil, 5-carboxymethylaminomethyl-2-thio-uracil, 5- (carboxyhydroxymethyl)-uracil, 5-fluoro-uracil, 5-bromo-uracil, 5-carboxymethylaminomethyl-uracil, 5-methyl-2-thio-uracil, 5-methyl-uracil, N-uracil-5-oxyacetic acid methyl ester, 5- methylaminomethyl-uracil, 5-methoxyaminomethyl-2-thio-uracil, 5'-methoxycarbonylmethyl-uracil, 5-methoxy-uracil, uracil-5-oxyacetic acid methyl ester, uracil-5-oxyacetic acid (v), 1-methyl- pseudouracil, queosine, beta.-D-mannosyl-queosine, wybutoxosine, and phosphoramidates, phosphorothioates, peptide nucleotides, methylphosphonates, 7-deazaguanosine, 5-methylcytosine and inosine. The preparation of such analogues is known to a person skilled in the art e.g., from the U.S. Pat. No. 4,373,071, U.S. Pat. No. 4,401,796, U.S. Pat. No. 4,415,732, U.S. Pat. No. 4,458,066, U.S. Pat. No. 4,500,707, U.S. Pat. No. 4,668,777, U.S. Pat. No. 4,973,679, U.S. Pat. No. 5,047,524, U.S. Pat. No. 5,132,418, U.S. Pat. No. 5,153,319, U.S. Pat. Nos. 5,262,530 and 5,700,642, the disclosures of which are incorporated by reference in their entirety.

[0117] In some embodiments, mRNAs may contain RNA backbone modifications. Typically, a backbone modification is a modification in which the phosphates of the backbone of the nucleotides contained in the RNA are modified chemically. Exemplary backbone modifications typically include, but are not limited to, modifications from the group consisting of

methylphosphonates, methylphosphoramidates, phosphoramidates, phosphorothioates {e.g.

cytidine 5'-0-(l-thiophosphate)), boranophosphates, positively charged guanidinium groups etc., which means by replacing the phosphodiester linkage by other anionic, cationic or neutral groups.

[0118] In some embodiments, mRNAs may contain sugar modifications. A typical sugar modification is a chemical modification of the sugar of the nucleotides it contains including, but not limited to, sugar modifications chosen from the group consisting of 4'-thio-ribonucleotide (see, e.g., US Patent Application Publication No. US 2016/0031928, incorporated by reference herein), 2'- deoxy-2'-fluoro-oligoribonucleotide (2'-fluoro-2'-deoxycytidine 5'-triphosphate, 2'-fluoro-2'- deoxyuridine 5'-triphosphate), 2'-deoxy-2'-deamine-oligoribonucleotide (2'-amino-2'-deoxycytidine 5'-triphosphate, 2'-amino-2'-deoxyuridine 5'-triphosphate), 2'-0-alkyloligoribonucleotide, 2'-deoxy- 2'-C-alkyloligoribonucleotide (2'-0-methylcytidine 5'-triphosphate, 2'-methyluridine 5'- triphosphate), 2'-C-alkyloligoribonucleotide, and isomers thereof (2'-aracytidine 5'-triphosphate, 2'- arauridine 5'-triphosphate), or azidotriphosphates (2'-azido-2'-deoxycytidine 5'-triphosphate, 2'- azido-2'-deoxyuridine 5'-triphosphate).

[0119] In some embodiments, mRNAs may contain modifications of the bases of the nucleotides (base modifications). A modified nucleotide which contains a base modification is also called a base-modified nucleotide. Examples of such base-modified nucleotides include, but are not limited to, 2-amino-6-chloropurine riboside 5'-triphosphate, 2-aminoadenosine 5'-triphosphate, 2- thiocytidine 5'-triphosphate, 2-thiouridine 5'-triphosphate, 4-thiouridine 5'-triphosphate, 5- aminoallylcytidine 5'-triphosphate, 5-aminoallyluridine 5'-triphosphate, 5-bromocytidine 5'- triphosphate, 5-bromouridine 5'-triphosphate, 5-iodocytidine 5'-triphosphate, 5-iodouridine 5'- triphosphate, 5-methylcytidine 5'-triphosphate, 5-methyluridine 5'-triphosphate, 6-azacytidine 5'- triphosphate, 6-azauridine 5'-triphosphate, 6-chloropurine riboside 5'-triphosphate, 7- deazaadenosine 5'-triphosphate, 7-deazaguanosine 5'-triphosphate, 8-azaadenosine 5'-triphosphate, 8-azidoadenosine 5'-triphosphate, benzimidazole riboside 5'-triphosphate, Nl-methyladenosine 5'- triphosphate, Nl-methylguanosine 5'-triphosphate, N6-methyladenosine 5'-triphosphate, 06- methylguanosine 5'-triphosphate, pseudouridine 5'-triphosphate, puromycin 5'-triphosphate or xanthosine 5'-triphosphate.

[0120] Typically, mRNA synthesis includes the addition of a "cap" on the N-terminal (5') end, and a "tail" on the C-terminal (3') end. The presence of the cap is important in providing resistance to nucleases found in most eukaryotic cells. The presence of a "tail" serves to protect the mRNA from exonuclease degradation.

[0121] Thus, in some embodiments, mRNAs include a 5' cap structure. A 5' cap is typically added as follows: first, an RNA terminal phosphatase removes one of the terminal phosphate groups from the 5' nucleotide, leaving two terminal phosphates; guanosine triphosphate (GTP) is then added to the terminal phosphates via a guanylyl transferase, producing a 5'5'5 triphosphate linkage; and the 7-nitrogen of guanine is then methylated by a methyltransferase. Examples of cap structures include, but are not limited to, m7G(5')ppp (5'(A,G(5')ppp(5')A and G(5')ppp(5')G.

[0122] In some embodiments, mRNAs include a 3' poly(A) tail structure. A poly-A tail on the 3' terminus of mRNA typically includes about 10 to 300 adenosine nucleotides ( e.g ., about 10 to 200 adenosine nucleotides, about 10 to 150 adenosine nucleotides, about 10 to 100 adenosine nucleotides, about 20 to 70 adenosine nucleotides, or about 20 to 60 adenosine nucleotides). In some embodiments, mRNAs include a 3' poly(C) tail structure. A suitable poly-C tail on the 3' terminus of mRNA typically include about 10 to 200 cytosine nucleotides {e.g., about 10 to 150 cytosine nucleotides, about 10 to 100 cytosine nucleotides, about 20 to 70 cytosine nucleotides, about 20 to 60 cytosine nucleotides, or about 10 to 40 cytosine nucleotides). The poly-C tail may be added to the poly-A tail or may substitute the poly-A tail.

[0123] In some embodiments, mRNAs include a 5' and/or 3' untranslated region. In some embodiments, a 5' untranslated region includes one or more elements that affect an mRNA's stability or translation, for example, an iron responsive element. In some embodiments, a 5' untranslated region may be between about 50 and 500 nucleotides in length.

[0124] In some embodiments, a 3' untranslated region includes one or more of a polyadenylation signal, a binding site for proteins that affect an mRNA's stability of location in a cell, or one or more binding sites for miRNAs. In some embodiments, a 3' untranslated region may be between 50 and 500 nucleotides in length or longer.

Cap structure

[0125] In some embodiments, mRNAs include a 5' cap structure. A 5' cap is typically added as follows: first, an RNA terminal phosphatase removes one of the terminal phosphate groups from the 5' nucleotide, leaving two terminal phosphates; guanosine triphosphate (GTP) is then added to the terminal phosphates via a guanylyl transferase, producing a 5'5'5 triphosphate linkage; and the 7-nitrogen of guanine is then methylated by a methyltransferase. Examples of cap structures include, but are not limited to, m7G(5')ppp (5'(A,G(5')ppp(5')A and G(5')ppp(5')G.

[0126] Naturally occurring cap structures comprise a 7-methyl guanosine that is linked via a triphosphate bridge to the 5'-end of the first transcribed nucleotide, resulting in a dinucleotide cap of m 7 G(5')ppp(5')N, where N is any nucleoside. In vivo, the cap is added enzymatically. The cap is added in the nucleus and is catalyzed by the enzyme guanylyl transferase. The addition of the cap to the 5' terminal end of RNA occurs immediately after initiation of transcription. The terminal nucleoside is typically a guanosine, and is in the reverse orientation to all the other nucleotides, i.e., G(5')ppp(5')GpNpNp.

[0127] A common cap for mRNA produced by in vitro transcription is m 7 G(5')ppp(5')G, which has been used as the dinucleotide cap in transcription with T7 or SP6 RNA polymerase in vitro to obtain RNAs having a cap structure in their 5'-termini. The prevailing method for the in vitro synthesis of caPPEd mRNA employs a pre-formed dinucleotide of the form m 7 G(5')ppp(5')G ("m 7 GpppG") as an initiator of transcription.

[0128] To date, a usual form of a synthetic dinucleotide cap used in in vitro translation experiments is the Anti-Reverse Cap Analog ("ARCA") or modified ARCA, which is generally a modified cap analog in which the 2' or 3' OH group is replaced with -OCH 3 . [0129] Additional cap analogs include, but are not limited to, a chemical structures selected from the group consisting of m 7 GpppG, m 7 GpppA, m 7 GpppC; unmethylated cap analogs ( e.g ., GpppG); dimethylated cap analog {e.g., m 2,7 GpppG), trimethylated cap analog {e.g., m 2,2,7 GpppG), dimethylated symmetrical cap analogs {e.g., m 7 Gpppm 7 G), or anti reverse cap analogs {e.g., ARCA; m 7 , 2Ome GpppG, m 72 d GpppG, m 7,3 0me GpppG, m 7,3 d GpppG and their tetraphosphate derivatives) (see, e.g., Jemielity, J. et al.,“Novel 'anti-reverse' cap analogs with superior translational properties" , RNA, 9: 1108-1122 (2003)).

[0130] In some embodiments, a suitable cap is a 7-methyl guanylate ("m 7 G") linked via a triphosphate bridge to the 5'-end of the first transcribed nucleotide, resulting in m 7 G(5')ppp(5')N, where N is any nucleoside. A preferred embodiment of a m 7 G cap utilized in embodiments of the invention is m 7 G(5')ppp(5')G.

[0131] In some embodiments, the cap is a CapO structure. CapO structures lack a 2'-0- methyl residue of the ribose attached to bases 1 and 2. In some embodiments, the cap is a Capl structure. Capl structures have a 2'-0-methyl residue at base 2. I n some embodiments, the cap is a Cap2 structure. Cap2 structures have a 2'-0-methyl residue attached to both bases 2 and 3.

[0132] A variety of m 7 G cap analogs are known in the art, many of which are commercially available. These include the m 7 GpppG described above, as well as the ARCA 3'-OCH 3 and 2'-OCH 3 cap analogs (Jemielity, J. et al., RNA, 9: 1108-1122 (2003)). Additional cap analogs for use in embodiments of the invention include N7-benzylated dinucleoside tetraphosphate analogs (described in Grudzien, E. et al., RNA, 10: 1479-1487 (2004)), phosphorothioate cap analogs (described in Grudzien-Nogalska, E., et al., RNA, 13: 1745-1755 (2007)), and cap analogs (including biotinylated cap analogs) described in U.S. Patent Nos. 8,093,367 and 8,304,529, incorporated by reference herein.

Tail structure

[0133] Typically, the presence of a "tail" serves to protect the m RNA from exonuclease degradation. The poly A tail is thought to stabilize natural messengers and synthetic sense RNA. Therefore, in certain embodiments a long poly A tail can be added to an m RNA molecule thus rendering the RNA more stable. Poly A tails can be added using a variety of art-recognized techniques. For example, long poly A tails can be added to synthetic or in vitro transcribed RNA using poly A polymerase (Yokoe, et al. Nature Biotechnology. 1996; 14: 1252-1256). A transcription vector can also encode long poly A tails. In addition, poly A tails can be added by transcription directly from PCR products. Poly A may also be ligated to the 3' end of a sense RNA with RNA ligase (see, e.g., Molecular Cloning A Laboratory Manual, 2nd Ed., ed. by Sambrook, Fritsch and Maniatis (Cold Spring Harbor Laboratory Press: 1991 edition)).

[0134] In some embodiments, mRNAs include a 3' poly(A) tail structure. Typically, the length of the poly A tail can be at least about 10, 50, 100, 200, 300, 400 at least 500 nucleotides. In some embodiments, a poly-A tail on the 3' terminus of mRNA typically includes about 10 to 300 adenosine nucleotides {e.g., about 10 to 200 adenosine nucleotides, about 10 to 150 adenosine nucleotides, about 10 to 100 adenosine nucleotides, about 20 to 70 adenosine nucleotides, or about 20 to 60 adenosine nucleotides). In some embodiments, mRNAs include a 3' poly(C) tail structure. A suitable poly-C tail on the 3' terminus of mRNA typically include about 10 to 200 cytosine nucleotides {e.g., about 10 to 150 cytosine nucleotides, about 10 to 100 cytosine nucleotides, about 20 to 70 cytosine nucleotides, about 20 to 60 cytosine nucleotides, or about 10 to 40 cytosine nucleotides). The poly-C tail may be added to the poly-A tail or may substitute the poly-A tail.

[0135] In some embodiments, the length of the poly A or poly C tail is adjusted to control the stability of a modified sense mRNA molecule of the invention and, thus, the transcription of protein. For example, since the length of the poly A tail can influence the half-life of a sense mRNA molecule, the length of the poly A tail can be adjusted to modify the level of resistance of the mRNA to nucleases and thereby control the time course of polynucleotide expression and/or polypeptide production in a target cell.

5' and 3' Untranslated Region

[0136] In some embodiments, mRNAs include a 5' and/or 3' untranslated region. In some embodiments, a 5' untranslated region includes one or more elements that affect an mRNA's stability or translation, for example, an iron responsive element. In some embodiments, a 5' untranslated region may be between about 50 and 500 nucleotides in length.

[0137] In some embodiments, a 3' untranslated region includes one or more of a polyadenylation signal, a binding site for proteins that affect an mRNA's stability of location in a cell, or one or more binding sites for miRNAs. In some embodiments, a 3' untranslated region may be between 50 and 500 nucleotides in length or longer.

[0138] Exemplary 3' and/or 5' UTR sequences can be derived from mRNA molecules which are stable {e.g., globin, actin, GAPDH, tubulin, histone, or citric acid cycle enzymes) to increase the stability of the sense mRNA molecule. For example, a 5' UTR sequence may include a partial sequence of a CMV immediate-early 1 (I El) gene, or a fragment thereof to improve the nuclease resistance and/or improve the half-life of the polynucleotide. Also contemplated is the inclusion of a sequence encoding human growth hormone (hGH), or a fragment thereof to the 3' end or untranslated region of the polynucleotide ( e.g ., mRNA) to further stabilize the polynucleotide. Generally, these modifications improve the stability and/or pharmacokinetic properties {e.g., half- life) of the polynucleotide relative to their unmodified counterparts, and include, for example modifications made to improve such polynucleotides' resistance to in vivo nuclease digestion.

Pharmaceutical Formulations of Cationic Lipids and Nucleic Acids

[0139] In certain embodiments, the compounds described herein {e.g., a compound of

Formula (I) such as Compounds ( 1)— (4)), as well as pharmaceutical and liposomal compositions comprising such lipids, can be used in formulations to facilitate the delivery of encapsulated materials {e.g., one or more polynucleotides such as mRNA) to, and subsequent transfection of one or more target cells. For example, in certain embodiments cationic lipids described herein (and compositions such as liposomal compositions comprising such lipids) are characterized as resulting in one or more of receptor-mediated endocytosis, clathrin-mediated and caveolae-mediated endocytosis, phagocytosis and macropinocytosis, fusogenicity, endosomal or lysosomal disruption and/or releasable properties that afford such compounds advantages relative other similarly classified lipids.

[0140] According to the present invention, a nucleic acid, e.g., mRNA encoding a protein

{e.g., a full length, fragment or portion of a protein) as described herein may be delivered via a delivery vehicle comprising a compound as described herein {e.g., a compound of Formula (I) such as Compounds (l)-(4)).

[0141] As used herein, the terms "delivery vehicle," "transfer vehicle," "nanoparticle" or grammatical equivalent, are used interchangeably.

[0142] For example, the present invention provides a composition {e.g., a pharmaceutical composition) comprising a compound described herein {e.g., a compound of Formula (I) such as Compounds (1)— (4)) and one or more polynucleotides. A composition {e.g., a pharmaceutical composition) may further comprise one or more cationic lipids, one or more non-cationic lipids, one or more cholesterol-based lipids and/or one or more PEG-modified lipids.

[0143] In certain embodiments a composition exhibits an enhanced {e.g., increased) ability to transfect one or more target cells. Accordingly, also provided herein are methods of transfecting one or more target cells. Such methods generally comprise the step of contacting the one or more target cells with the cationic lipids and/or pharmaceutical compositions disclosed herein {e.g., a liposomal formulation comprising a compound described herein {e.g., a compound of Formula (I) such as Compounds (1)— (4)) encapsulating one or more polynucleotides) such that the one or more target cells are transfected with the materials encapsulated therein ( e.g ., one or more

polynucleotides). As used herein, the terms "transfect" or "transfection" refer to the intracellular introduction of one or more encapsulated materials (e.g., nucleic acids and/or polynucleotides) into a cell, or preferably into a target cell. The introduced polynucleotide may be stably or transiently maintained in the target cell. The term "transfection efficiency" refers to the relative amount of such encapsulated material (e.g., polynucleotides) up-taken by, introduced into and/or expressed by the target cell which is subject to transfection. In practice, transfection efficiency may be estimated by the amount of a reporter polynucleotide product produced by the target cells following transfection. In certain embodiments, the compounds and pharmaceutical compositions described herein demonstrate high transfection efficiencies thereby improving the likelihood that appropriate dosages of the encapsulated materials (e.g., one or more polynucleotides) will be delivered to the site of pathology and subsequently expressed, while at the same time minimizing potential systemic adverse effects or toxicity associated with the compound or their encapsulated contents.

[0144] Following transfection of one or more target cells by, for example, the

polynucleotides encapsulated in the one or more lipid nanoparticles comprising the pharmaceutical or liposomal compositions disclosed herein, the production of the product (e.g., a polypeptide or protein) encoded by such polynucleotide may be preferably stimulated and the capability of such target cells to express the polynucleotide and produce, for example, a polypeptide or protein of interest is enhanced. For example, transfection of a target cell by one or more compounds or pharmaceutical compositions encapsulating mRNA will enhance (i.e., increase) the production of the protein or enzyme encoded by such mRNA.

[0145] Further, delivery vehicles described herein (e.g., liposomal delivery vehicles) may be prepared to preferentially distribute to other target tissues, cells or organs, such as the heart, lungs, kidneys, spleen. In embodiments, the lipid nanoparticles of the present invention may be prepared to achieve enhanced delivery to the target cells and tissues. For example, polynucleotides (e.g., mRNA) encapsulated in one or more of the compounds or pharmaceutical and liposomal compositions described herein can be delivered to and/or transfect targeted cells or tissues. In some embodiments, the encapsulated polynucleotides (e.g., mRNA) are capable of being expressed and functional polypeptide products produced (and in some instances excreted) by the target cell, thereby conferring a beneficial property to, for example the target cells or tissues. Such encapsulated polynucleotides (e.g., mRNA) may encode, for example, a hormone, enzyme, receptor, polypeptide, peptide or other protein of interest. Liposomal Delivery Vehicles

[0146] In some embodiments, a composition is a suitable delivery vehicle. In embodiments, a composition is a liposomal delivery vehicle, e.g., a lipid nanoparticle.

[0147] The terms "liposomal delivery vehicle" and "liposomal composition" are used interchangeably.

[0148] Enriching liposomal compositions with one or more of the cationic lipids disclosed herein may be used as a means of improving {e.g., reducing) the toxicity or otherwise conferring one or more desired properties to such enriched liposomal composition {e.g., improved delivery of the encapsulated polynucleotides to one or more target cells and/or reduced in vivo toxicity of a liposomal composition). Accordingly, also contemplated are pharmaceutical compositions, and in particular liposomal compositions, that comprise one or more of the cationic lipids disclosed herein.

[0149] Thus, in certain embodiments, the compounds described herein {e.g., a compound of Formula (I) such as Compounds ( 1)— (4)) may be used as a component of a liposomal composition to facilitate or enhance the delivery and release of encapsulated materials {e.g., one or more therapeutic agents) to one or more target cells {e.g., by permeating or fusing with the lipid membranes of such target cells).

[0150] As used herein, liposomal delivery vehicles, e.g., lipid nanoparticles, are usually characterized as microscopic vesicles having an interior aqua space sequestered from an outer medium by a membrane of one or more bilayers. Bilayer membranes of liposomes are typically formed by amphiphilic molecules, such as lipids of synthetic or natural origin that comprise spatially separated hydrophilic and hydrophobic domains (Lasic, Trends Biotechnol., 16: 307-321, 1998). Bilayer membranes of the liposomes can also be formed by amphophilic polymers and surfactants {e.g., polymerosomes, niosomes, etc.). In the context of the present invention, a liposomal delivery vehicle typically serves to transport a desired mRNA to a target cell or tissue.

[0151] In certain embodiments, such compositions {e.g., liposomal compositions) are loaded with or otherwise encapsulate materials, such as for example, one or more biologically-active polynucleotides {e.g., mRNA).

[0152] In embodiments, a composition {e.g., a pharmaceutical composition) comprises an mRNA encoding a protein, encapsulated within a liposome. In embodiments, a liposome comprises one or more cationic lipids, one or more non-cationic lipids, one or more cholesterol-based lipids and one or more PEG-modified lipids, and wherein at least one PEG-modified lipid is a compound as described herein {e.g., a compound of Formula (I) such as Compounds (l)-(4)). In embodiments, a composition comprises an mRNA encoding for a protein ( e.g ., any protein described herein).

In embodiments, a composition comprises an mRNA encoding for cystic fibrosis transmembrane conductance regulator (CFTR) protein. In embodiments, a composition comprises an mRNA encoding for ornithine transcarbamylase (OTC) protein.

[0153] In embodiments, a composition (e.g., a pharmaceutical composition) comprises a nucleic acid encapsulated within a liposome, wherein the liposome comprises any compound described herein (e.g., a compound of Formula (I) such as Compounds (1)— (4)) as described herein.

[0154] In embodiments, a nucleic acid is an mRNA encoding a peptide or protein. In embodiments, an mRNA encodes a peptide or protein for use in the delivery to or treatment of the lung of a subject or a lung cell (e.g., an mRNA encodes cystic fibrosis transmembrane conductance regulator (CFTR) protein). In embodiments, an mRNA encodes a peptide or protein for use in the delivery to or treatment of the liver of a subject or a liver cell (e.g., an mRNA encodes ornithine transcarbamylase (OTC) protein). Still other exemplary mRNAs are described herein.

[0155] In embodiments, a liposomal delivery vehicle (e.g., a lipid nanoparticle) can have a net positive charge.

[0156] In embodiments, a liposomal delivery vehicle (e.g., a lipid nanoparticle) can have a net negative charge.

[0157] In embodiments, a liposomal delivery vehicle (e.g., a lipid nanoparticle) can have a net neutral charge.

[0158] In embodiments, a lipid nanoparticle that encapsulates a nucleic acid (e.g., mRNA encoding a peptide or protein) comprises one or more compounds described herein ((e.g., a compound of Formula (I) such as Compounds (l)-(4)).

[0159] For example, the amount of a compound as described herein (e.g., a compound of

Formula (I) such as Compounds ( 1)— (4)) in a composition can be described as a percentage ("wt%") of the combined dry weight of all lipids of a composition (e.g., the combined dry weight of all lipids present in a liposomal composition).

[0160] In embodiments of the pharmaceutical compositions described herein, a compound as described herein (e.g., a compound of Formula (I) such as Compounds (1)— (4)) is present in an amount that is about 0.5 wt% to about 30 wt% (e.g., about 0.5 wt% to about 20 wt%) of the combined dry weight of all lipids present in a composition (e.g., a liposomal composition).

[0161] In embodiments, a compound as described herein (e.g., a compound of Formula (I) such as Compounds (1)— (4)) is present in an amount that is about 1 wt% to about 30 wt%, about 1 wt% to about 20 wt%, about 1 wt% to about 15 wt%, about 1 wt% to about 10 wt%, or about 5 wt% to about 25 wt% of the combined dry weight of all lipids present in a composition ( e.g ., a liposomal composition). In embodiments, a compound as described herein {e.g., a compound of Formula (I) such as Compounds (1)— (4)) is present in an amount that is about 0.5 wt% to about 5 wt%, about 1 wt% to about 10 wt%, about 5 wt% to about 20 wt%, or about 10 wt% to about 20 wt% of the combined molar amounts of all lipids present in a composition such as a liposomal delivery vehicle.

[0162] In embodiments, the amount of a compound as described herein {e.g., a compound of Formula (I) such as Compounds ( 1)— (4)) is present in an amount that is at least about 5 wt%, about 10 wt%, about 15 wt%, about 20 wt%, about 25 wt%, about 30 wt%, about 35 wt%, about 40 wt%, about 45 wt%, about 50 wt%, about 55 wt%, about 60 wt%, about 65 wt%, about 70 wt%, about 75 wt%, about 80 wt%, about 85 wt%, about 90 wt%, about 95 wt%, about 96 wt%, about 97 wt%, about 98 wt%, or about 99 wt% of the combined dry weight of total lipids in a composition {e.g., a liposomal composition).

[0163] In embodiments, the amount of a compound as described herein {{e.g., a compound of Formula (I) such as Compounds (1)— (4)) is present in an amount that is no more than about 5 wt%, about 10 wt%, about 15 wt%, about 20 wt%, about 25 wt%, about 30 wt%, about 35 wt%, about 40 wt%, about 45 wt%, about 50 wt%, about 55 wt%, about 60 wt%, about 65 wt%, about 70 wt%, about 75 wt%, about 80 wt%, about 85 wt%, about 90 wt%, about 95 wt%, about 96 wt%, about 97 wt%, about 98 wt%, or about 99 wt% of the combined dry weight of total lipids in a composition {e.g., a liposomal composition).

[0164] In embodiments, a composition {e.g., a liposomal delivery vehicle such as a lipid nanoparticle) comprises about 0.1 wt% to about 20 wt% {e.g., about 0.1 wt% to about 15 wt%) of a compound described herein {e.g., a compound of Formula (I) such as Compounds (l)-(4)). In embodiments, a delivery vehicle {e.g., a liposomal delivery vehicle such as a lipid nanoparticle) comprises about 0.5 wt%, about 1 wt%, about 3 wt%, about 5 wt%, or about 10 wt% a compound described herein {e.g., a compound of Formula (I) such as Compounds (l)-(4)). In embodiments, a delivery vehicle {e.g., a liposomal delivery vehicle such as a lipid nanoparticle) comprises up to about 0.5 wt%, about 1 wt%, about 3 wt%, about 5 wt%, about 10 wt%, about 15 wt%, or about 20 wt% of a compound described herein {e.g., a compound of Formula (I) such as Compounds (l)-(4)). In embodiments, the percentage results in an improved beneficial effect {e.g., improved delivery to targeted tissues such as the liver or the lung).

[0165] The amount of a compound as described herein {e.g., a compound of Formula (I) such as Compounds (1)— (4)) in a composition also can be described as a percentage ("mol%") of the combined molar amounts of total lipids of a composition ( e.g ., the combined molar amounts of all lipids present in a liposomal delivery vehicle).

[0166] In embodiments of pharmaceutical compositions described herein, a compound as described herein (e.g., a compound of Formula (I) such as Compounds (1)— (4)) is present in an amount that is about 0.5 mol% to about30 mol% (e.g., about 0.5 mol% to about20 mol%) of the combined molar amounts of all lipids present in a composition such as a liposomal delivery vehicle.

[0167] In embodiments, a compound as described herein (e.g., a compound of Formula (I) such as Compounds (1)— (4)) is present in an amount that is about 0.5 mol% to about 5 mol%, about 1 mol% to about 10 mol%, about 5 mol% to about 20 mol%, or about 10 mol% to about 20 mol% of the combined molar amounts of all lipids present in a composition such as a liposomal delivery vehicle. In embodiments, a compound as described herein (e.g., a compound of Formula (I) such as Compounds (1)— (4)) is present in an amount that is about 1 mol% to about 30 mol%, about 1 mol% to about 20 mol%, about 1 mol% to about 15 mol%, about 1 mol% to about 10 mol%, or about 5 mol% to about 25 mol% of the combined dry weight of all lipids present in a composition such as a liposomal delivery vehicle

[0168] In certain embodiments, a compound as described herein (e.g., a compound of

Formula (I) such as Compounds (1)— (4)) can comprise from about 0.1 mol% to about 50 mol%, or from 0.5 mol% to about 50 mol%, or from about 1 mol% to about 25 mol%, or from about 1 mol% to about 10 mol% of the total amount of lipids in a composition (e.g., a liposomal delivery vehicle).

[0169] In certain embodiments, a compound as described herein (e.g., a compound of

Formula (I) such as Compounds ( 1)— (4)) can comprise greater than about 0.1 mol%, or greater than about 0.5 mol%, or greater than about 1 mol%, or greater than about 5 mol% of the total amount of lipids in the lipid nanoparticle.

[0170] In certain embodiments, a compound as described (e.g., a compound of Formula (I) such as Compounds (1)— (4)) can comprise less than about 25 mol%, or less than about 10 mol%, or less than about 5 mol%, or less than about 1 mol% of the total amount of lipids in a composition (e.g., a liposomal delivery vehicle).

[0171] In embodiments, the amount of a compound as described herein (e.g., a compound of Formula (I) such as Compounds ( 1)— (4)) is present in an amount that is at least about 5 mol%, about 10 mol%, about 15 mol%, about 20 mol%, about 25 mol%, about 30 mol%, about 35 mol%, about 40 mol%, about 45 mol%, about 50 mol%, about 55 mol%, about 60 mol%, about 65 mol%, about 70 mol%, about 75 mol%, about 80 mol%, about 85 mol%, about 90 mol%, about 95 mol%, about 96 mol%, about 97 mol%, about 98 mol%, or about 99 mol% of the combined dry weight of total lipids in a composition ( e.g ., a liposomal composition).

[0172] In embodiments, the amount of a compound as described herein {e.g., a compound of Formula (I) such as Compounds ( 1)— (4)) is present in an amount that is no more than about 5 mol%, about 10 mol%, about 15 mol%, about 20 mol%, about 25 mol%, about 30 mol%, about 35 mol%, about 40 mol%, about 45 mol%, about 50 mol%, about 55 mol%, about 60 mol%, about 65 mol%, about 70 mol%, about 75 mol%, about 80 mol%, about 85 mol%, about 90 mol%, about 95 mol%, about 96 mol%, about 97 mol%, about 98 mol%, or about 99 mol% of the combined dry weight of total lipids in a composition {e.g., a liposomal composition).

[0173] In embodiments, the percentage results in an improved beneficial effect {e.g., improved delivery to targeted tissues such as the liver or the lung).

[0174] In embodiments, a composition further comprises one more lipids {e.g., one more lipids selected from the group consisting of one or more cationic lipids, one or more non-cationic lipids, one or more cholesterol-based lipids, and one or more PEG-modified lipids).

[0175] In certain embodiments, such pharmaceutical {e.g., liposomal) compositions comprise one or more of a PEG-modified lipid, a non-cationic lipid and a cholesterol lipid. In embodiments, such pharmaceutical {e.g., liposomal) compositions comprise: one or more PEG- modified lipids; one or more non-cationic lipids; and one or more cholesterol lipids. In

embodiments, such pharmaceutical {e.g., liposomal) compositions comprise: one or more PEG- modified lipids and one or more cholesterol lipids.

[0176] In embodiments, a composition {e.g., lipid nanoparticle) that encapsulates a nucleic acid {e.g., mRNA encoding a peptide or protein) comprises one or more compounds as described herein {e.g., a compound of Formula (I) such as Compounds (1)— (4)) and one or more lipids selected from the group consisting of a cationic lipid, a non-cationic lipid, and a PEGylated lipid.

[0177] In embodiments, a composition {e.g., lipid nanoparticle) that encapsulates a nucleic acid {e.g., mRNA encoding a peptide or protein) comprises one or more compound as described herein {e.g., a compound of Formula (I) such as Compounds (1)— (4)); one or more lipids selected from the group consisting of a cationic lipid, a non-cationic lipid, and a PEGylated lipid; and further comprises a cholesterol-based lipid.

[0178] In embodiments, a lipid nanoparticle that encapsulates a nucleic acid {e.g., mRNA encoding a peptide or protein) comprises one or more compound as described herein {{e.g., a compound of Formula (I) such as Compounds (1)— (4)), as well as one or more lipids selected from the group consisting of a cationic lipid, a non-cationic lipid, a PEGylated lipid, and a cholesterol- based lipid.

[0179] According to various embodiments, the selection of cationic lipids, non-cationic lipids and/or PEG-modified lipids which comprise the lipid nanoparticle, as well as the relative molar ratio of such lipids to each other, is based upon the characteristics of the selected lipid(s), the nature of the intended target cells, the characteristics of the mRNA to be delivered. Additional considerations include, for example, the saturation of the alkyl chain, as well as the size, charge, pH, pKa, fusogenicity and toxicity of the selected lipid(s). Thus, the molar ratios may be adjusted accordingly.

Cationic Lipids

[0180] In addition to any of the compounds as described herein {{e.g., a compound of

Formula (I) such as Compounds (1)— (4)), a composition may comprise one or more cationic lipids. In some embodiments, liposomes may comprise one or more cationic lipids. As used herein, the phrase "cationic lipid" refers to any of a number of lipid species that have a net positive charge at a selected pH, such as physiological pH. Several cationic lipids have been described in the literature, many of which are commercially available.

[0181] In some embodiments, liposomes may comprise one or more cationic lipids. As used herein, the phrase "cationic lipid" refers to any of a number of lipid species that have a net positive charge at a selected pH, such as physiological pH. Several cationic lipids have been described in the literature, many of which are commercially available.

[0182] Suitable cationic lipids for use in the compositions include the cationic lipids as described in International Patent Publication WO 2010/144740, which is incorporated herein by reference. In certain embodiments, the compositions include a cationic lipid, (6Z,9Z,28Z,31Z)- heptatriaconta-6,9,28,31-tetraen-19-yl 4-(dimethylamino) butanoate, having a compound structure of: and pharmaceutically acceptable salts thereof. [0183] Other suitable cationic lipids for use in the compositions include ionizable cationic lipids as described in International Patent Publication WO 2013/149140, which is incorporated herein by reference. In some embodiments, the compositions include a cationic lipid of one of the following formulas:

or a pharmaceutically acceptable salt thereof, wherein Ri and R are each independently selected from the group consisting of hydrogen, an optionally substituted, variably saturated or unsaturated C 1 -C 20 alkyl and an optionally substituted, variably saturated or unsaturated C 6 -C 20 acyl; wherein U and L are each independently selected from the group consisting of hydrogen, an optionally substituted C 1 -C 30 alkyl, an optionally substituted variably unsaturated C 1 -C 30 alkenyl, and an optionally substituted C 1 -C 30 alkynyl; wherein m and o are each independently selected from the group consisting of zero and any positive integer (e.g., where m is three); and wherein n is zero or any positive integer (e.g., where n is one).

[0184] In certain embodiments, the compositions include the cationic lipid (15Z, 18Z)-N,N- dimethyl-6-(9Z,12Z)-octadeca-9,12-dien-l -yl) tetracosa- 15,18-dien-l-amine ("HGT5000"), having a compound structure of: and pharmaceutically acceptable salts thereof.

[0185] In certain embodiments, the compositions include the cationic lipid (15Z, 18Z)-N,N- dimethyl-6-((9Z,12Z)-octadeca-9,12-dien-l-yl) tetracosa-4,15,18-trien-l -amine ("HGT5001"), having a compound structure of:

and pharmaceutically acceptable salts thereof. [0186] In certain embodiments, the include the cationic lipid and (15Z,18Z)-N,N-dimethyl-6-

((9Z,12Z)-octadeca-9,12-dien-l-yl) tetracosa-5,15,18-trien- 1 -amine ("HGT5002"), having a compound structure of:

and pharmaceutically acceptable salts thereof.

[0187] Other suitable cationic lipids for use in the compositions include cationic lipids described as aminoalcohol lipidoids in International Patent Publication WO 2010/053572, which is incorporated herein by reference. In certain embodiments, the compositions include a cationic lipid having a compound structure of:

and pharmaceutically acceptable salts thereof.

[0188] Other suitable cationic lipids for use in the compositions include the cationic lipids as described in International Patent Publication WO 2016/118725, which is incorporated herein by reference. In certain embodiments, the compositions include a cationic lipid having a compound structure of:

and pharmaceutically acceptable salts thereof.

[0189] Other suitable cationic lipids for use in the compositions include the cationic lipids as described in International Patent Publication WO 2016/118724, which is incorporated herein by reference. In certain embodiments, the compositions include a cationic lipid having a compound structure of:

and pharmaceutically acceptable salts thereof.

[0190] Other suitable cationic lipids for use in the compositions include a cationic lipid having the formula of 14,25-ditridecyl 15,18,21,24-tetraaza-octatriacontane, and pharmaceutically acceptable salts thereof.

[0191] Other suitable cationic lipids for use in the compositions include the cationic lipids as described in International Patent Publications WO 2013/063468 and WO 2016/205691, each of which are incorporated herein by reference. In some embodiments, the compositions include a cationic lipid of the following formula:

or pharmaceutically acceptable salts thereof, wherein each instance of R L is independently optionally substituted C -C alkenyl.

[0192] In certain embodiments, the compositions include a cationic lipid having a compound structure of:

and pharmaceutically acceptable salts thereof.

[0193] In certain embodiments, the compositions include a cationic lipid having a compound structure of:

(OF-02) and pharmaceutically acceptable salts thereof.

[0194] In certain embodiments, the compositions include a cationic lipid having a compound structure of:

and pharmaceutically acceptable salts thereof.

[0195] In certain embodiments, the compositions include a cationic lipid having a compound structure of:

and pharmaceutically acceptable salts thereof. [0196] Other suitable cationic lipids for use in the compositions include the cationic lipids as described in International Patent Publication WO 2015/184256, which is incorporated herein by reference. In some embodiments, the compositions include a cationic lipid of the following formula :

or a pharmaceutically acceptable salt thereof, wherein each X independently is O or S; each Y independently is O or S; each m independently is 0 to 20; each n independently is 1 to 6; each RA is independently hydrogen, optionally substituted Cl-50 alkyl, optionally substituted C2-50 alkenyl, optionally substituted C2-50 alkynyl, optionally substituted C3-10 carbocyclyl, optionally substituted 3-14 membered heterocyclyl, optionally substituted C6-14 aryl, optionally substituted 5-14 membered heteroaryl or halogen; and each R B is independently hydrogen, optionally substituted Cl-50 alkyl, optionally substituted C2-50 alkenyl, optionally substituted C2-50 alkynyl, optionally substituted C3-10 carbocyclyl, optionally substituted 3-14 membered heterocyclyl, optionally substituted C6-14 aryl, optionally substituted 5-14 membered heteroaryl or halogen. In certain embodiments, the compositions include a cationic lipid, "Target 23", having a compound structure of:

(Target 23) and pharmaceutically acceptable salts thereof. [0197] Other suitable cationic lipids for use in the compositions include the cationic lipids as described in International Patent Publication WO 2016/004202, which is incorporated herein by reference. In some embodiments, the compositions include a cationic lipid having the compound structure:

or a pharmaceutically acceptable salt thereof.

[0198] In some embodiments, the compositions include a cationic lipid having the compound structure:

or a pharmaceutically acceptable salt thereof.

[0199] In some embodiments, the compositions include a cationic lipid having the compound structure:

or a pharmaceutically acceptable salt thereof. [0200] Other suitable cationic lipids for use in the compositions include the cationic lipids as described in J. McClellan, M. C. King, Cell 2010, 141, 210-217 and in Whitehead et al., Nature Communications (2014) 5:4277, which is incorporated herein by reference. In certain embodiments, the cationic lipids of the compositions include a cationic lipid having a compound structure of:

and pharmaceutically acceptable salts thereof.

[0201] Other suitable cationic lipids for use in the compositions include the cationic lipids as described in International Patent Publication WO 2015/199952, which is incorporated herein by reference.

[0202] In some embodiments, the compositions include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof.

[0203] In some embodiments, the compositions include a cationic lipid having the compound structure: and pharmaceutically acceptable salts thereof.

[0204] In some embodiments, the compositions include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof.

[0205] In some embodiments, the compositions include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof.

[0206] In some embodiments, the compositions include a cationic lipid having the compound structure: and pharmaceutically acceptable salts thereof.

[0207] In some embodiments, the compositions include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof.

[0208] In some embodiments, the compositions include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof.

[0209] In some embodiments, the compositions include a cationic lipid having the compound structure: and pharmaceutically acceptable salts thereof.

[0210] In some embodiments, the compositions include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof.

[0211] In some embodiments, the compositions include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof.

[0212] In some embodiments, the compositions include a cationic lipid having the compound structure: and pharmaceutically acceptable salts thereof.

[0213] In some embodiments, the compositions include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof.

[0214] In some embodiments, the compositions include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof.

[0215] Other suitable cationic lipids for use in the compositions include the cationic lipids as described in International Patent Publication WO 2017/004143, which is incorporated herein by reference. In some embodiments, the compositions include a cationic lipid having the compound structure: and pharmaceutically acceptable salts thereof.

[0216] In some embodiments, the compositions include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof.

[0217] In some embodiments, the compositions include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof.

[0218] In some embodiments, the compositions include a cationic lipid having the compound structure:

[0219] In some embodiments, the compositions include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof.

[0220] In some embodiments, the compositions include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof.

[0221] In some embodiments, the compositions include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof.

[0222] In some embodiments, the compositions include a cationic lipid having the compound structure: and pharmaceutically acceptable salts thereof.

[0223] In some embodiments, the compositions include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof.

[0224] In some embodiments, the compositions include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof.

[0225] In some embodiments, the compositions include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof.

[0226] In some embodiments, the compositions include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof.

[0227] In some embodiments, the compositions include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof.

[0228] In some embodiments, the compositions include a cationic lipid having the compound structure: and pharmaceutically acceptable salts thereof.

[0229] In some embodiments, the compositions include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof.

[0230] In some embodiments, the compositions include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof.

[0231] In some embodiments, the compositions include a cationic lipid having the compound structure: and pharmaceutically acceptable salts thereof.

[0232] Other suitable cationic lipids for use in the compositions include the cationic lipids as described in International Patent Publication WO 2017/075531, which is incorporated herein by reference. In some embodiments, the compositions include a cationic lipid of the following formula:

or a pharmaceutically acceptable salt thereof, wherein one of L 1 or L 2 is -0(C=0)-, -(C=0)0-, - C(=0)-, -0-, -S(0) x , -S-S-, -C(=0)S-, -SC(=0)-, -NR a C(=0)-, -C(=0)NR a -, NR a C(=0)NR a -, -OC(=0)NR a -, or -NR a C(=0)0-; and the other of L 1 or L 2 is -0(C=0)-, -(0=0)0-, -C(=0)-, -0-, -S(O) x , -S-S-, -C(=0)S- , SC(=0)-, -NR a C(=0)-, -C(=0)NR a -, ,NR a C(=0)NR a -, -OC(=0)NR a - or -NR a C(=0)0- or a direct bond; G 1 and G 2 are each independently unsubstituted C -C alkylene or C -C alkenylene; G 3 is C -C alkylene, C -C alkenylene, C -C cycloalkylene, C -C cycloalkenylene; R a is H or C -C alkyl; R 1 and R 2 are each independently C -C alkyl or C -C alkenyl; R 3 is H, OR 5 , CN, -C(=0)OR 4 , - OC(=0)R 4 or -NR 5 C(=0)R 4 ; R 4 is C -C alkyl; R 5 is H or Oi-Ob alkyl; and x is 0, 1 or 2.

[0233] Other suitable cationic lipids for use in the compositions include the cationic lipids as described in International Patent Publication WO 2017/117528, which is incorporated herein by reference. In some embodiments, the compositions include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof. [0234] In some embodiments, the compositions include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof.

[0235] In some embodiments, the compositions include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof.

[0236] Other suitable cationic lipids for use in the compositions include the cationic lipids as described in International Patent Publication WO 2017/049245, which is incorporated herein by reference. In some embodiments, the cationic lipids of the compositions and methods of the present invention include a compound of one of the following formulas:

and pharmaceutically acceptable salts thereof. For any one of these four formulas, R is independently selected from -(CH 2 ) n Q and -(CH2) n CHQR; Q is selected from the group consisting of -OR, -OH, -0(CH 2 )nN(R) 2 , -OC(0)R, -CX 3 , -CN, -N(R)C(0)R, -N(H)C(0)R, -N(R)S(0) 2 R, -N(H)S(0) 2 R, -N(R)C(0)N(R) 2 , -N(H)C(0)N(R) 2 , -N(H)C(0)N(H)(R), -N(R)C(S) N(R) 2 , -N(H)C(S)N(R) 2 , - N(H)C(S)N(H)(R), and a heterocycle; R is independently selected from the group consisting of C1-3 alkyl, C 2 -3 alkenyl, and H; and n is 1, 2, or 3.

[0237] In certain embodiments, the compositions include a cationic lipid having a compound structure of:

and pharmaceutically acceptable salts thereof.

[0238] In certain embodiments, the compositions include a cationic lipid having a compound structure of:

and pharmaceutically acceptable salts thereof. [0239] In certain embodiments, the compositions include a cationic lipid having a compound structure of:

and pharmaceutically acceptable salts thereof.

[0240] In certain embodiments, the compositions include a cationic lipid having a compound structure of:

and pharmaceutically acceptable salts thereof.

[0241] Other suitable cationic lipids for use in the compositions include the cationic lipids as described in International Patent Publication WO 2017/173054 and WO 2015/095340, each of which is incorporated herein by reference. In certain embodiments, the compositions include a cationic lipid having a compound structure of:

and pharmaceutically acceptable salts thereof.

[0242] In certain embodiments, the compositions include a cationic lipid having a compound structure of: and pharmaceutically acceptable salts thereof.

[0243] In certain embodiments, the compositions include a cationic lipid having a compound structure of:

and pharmaceutically acceptable salts thereof.

[0244] In certain embodiments, the compositions include a cationic lipid having a compound structure of:

and pharmaceutically acceptable salts thereof.

[0245] Other suitable cationic lipids for use in the compositions include cholesterol-based cationic lipids. In certain embodiments, the compositions include imidazole cholesterol ester or "ICE", having a compound structure of: and pharmaceutically acceptable salts thereof.

[0246] Other suitable cationic lipids for use in the compositions include cleavable cationic lipids as described in International Patent Publication WO 2012/170889, which is incorporated herein by reference. In some embodiments, the compositions include a cationic lipid of the following formula: wherein Ri is selected from the group consisting of imidazole, guanidinium, amino, imine, enamine, an optionally-substituted alkyl amino (e.g., an alkyl amino such as dimethylamino) and pyridyl; wherein R is selected from the group consisting of one of the following two formulas:

and wherein R and R are each independently selected from the group consisting of an optionally substituted, variably saturated or unsaturated C -C alkyl and an optionally substituted, variably saturated or unsaturated C -C acyl; and wherein n is zero or any positive integer (e.g., one, two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, fourteen, fifteen, sixteen, seventeen, eighteen, nineteen, twenty or more). In certain embodiments, the compositions include a cationic lipid, "HGT4001", having a compound structure of:

and pharmaceutically acceptable salts thereof.

[0247] In certain embodiments, the compositions include a cationic lipid, "HGT4002", having a compound structure of:

and pharmaceutically acceptable salts thereof.

[0248] In certain embodiments, the compositions include a cationic lipid, "HGT4003", having a compound structure of:

(HGT4003)

and pharmaceutically acceptable salts thereof.

[0249] In certain embodiments, the compositions include a cationic lipid, "HGT4004", having a compound structure of:

and pharmaceutically acceptable salts thereof. [0250] In certain embodiments, the compositions include a cationic lipid "HGT4005", having a compound structure of: and pharmaceutically acceptable salts thereof.

[0251] In some embodiments, the compositions include the cationic lipid, N-[l-(2,3- dioleyloxy)propyl]-N,N,N-trimethylammonium chloride ("DOTMA"). (Feigner et al. (Proc. Nat'l Acad. Sci. 84, 7413 (1987); U.S. Pat. No. 4,897,355, which is incorporated herein by reference). DOTMA can be formulated alone or can be combined with a neutral lipid ( e.g ., dioleoylphosphatidyl- ethanolamine or "DOPE") or still other cationic or non-cationic lipids into a liposomal transfer vehicle or a lipid nanoparticle, and such liposomes can be used to enhance the delivery of nucleic acids into target cells. Other cationic lipids suitable for the compositions include, for example, 5- carboxyspermylglycinedioctadecylamide ("DOGS"); 2,3-dioleyloxy-N-[2(spermine- carboxamido)ethyl]-N,N-dimethyl-l-propanaminium ("DOSPA") (Behr et al. Proc. Nat.'l Acad. Sci. 86, 6982 (1989), U.S. Pat. No. 5,171,678; U.S. Pat. No. 5,334,761); l,2-Dioleoyl-3-Dimethylammonium- Propane ("DODAP"); l,2-Dioleoyl-3-Trimethylammonium-Propane ("DOTAP").

[0252] Additional exemplary cationic lipids suitable for the compositions also include: 1,2- distearyloxy-N,N-dimethyl-3-aminopropane ( "DSDMA"); l,2-dioleyloxy-N,N-dimethyl-3- aminopropane ("DODMA"); 1 ,2-dilinoleyloxy-N,N-dimethyl-3-aminopropane ("DLinDMA"); 1,2- dilinolenyloxy-N,N-dimethyl-3-aminopropane ("DLenDMA"); N-dioleyl-N,N-dimethylammonium chloride ("DODAC"); N,N-distearyl-N,N-dimethylarnrnonium bromide ("DDAB"); N-(l,2- dimyristyloxyprop-3-yl)-N,N-dimethyl-N-hydroxyethyl ammonium bromide ("DMRIE"); 3- dimethylamino-2-(cholest-5-en-3-beta-oxybutan-4-oxy)-l-(cis, cis-9,12-octadecadienoxy)propane ("CLinDMA"); 2-[5'-(cholest-5-en-3-beta-oxy)-3'-oxapentoxy)-3-dimethy l-l-(cis,cis-9', 1-2'- octadecadienoxy)propane ("CpLinDMA"); N,N-dimethyl-3,4-dioleyloxybenzylamine ("DMOBA"); 1 ,2- N,N'-dioleylcarbamyl-3-dimethylaminopropane ("DOcarbDAP"); 2,3-Dilinoleoyloxy-N,N- dimethylpropylamine ("DLinDAP"); l,2-N,N'-Dilinoleylcarbamyl-3-dimethylaminopropane

("DLincarbDAP"); 1 ,2-Dilinoleoylcarbamyl-3-dimethylaminopropane ("DLinCDAP"); 2,2-dilinoleyl-4- dimethylaminomethyl-[l,3]-dioxolane ("DLin-K-DMA"); 2-((8-[(3P)-cholest-5-en-3-yloxy]octyl)oxy)-N, N-dimethyl-3-[(9Z, 12Z)-octadeca-9, 12-dien-l -yloxy]propane- 1-amine ("Octyl-CLinDMA"); (2R)-2- ((8-[(3beta)-cholest-5-en-3-yloxy]octyl)oxy)-N, N-dimethyl-3-[(9Z, 12Z)-octadeca-9, 12-dien-l- yloxy]propan-l -amine ("Octyl-CLinDMA (2R)"); (2S)-2-((8-[(3P)-cholest-5-en-3-yloxy]octyl)oxy)-N, fsl- dimethyh3-[(9Z, 12Z)-octadeca-9, 12-dien-l -yloxy]propan-l -amine ("Octyl-CLinDMA (2S)"); 2,2- dilinoleyl-4-dimethylaminoethyl-[l,3]-dioxolane ("DLin-K-XTC2-DMA"); and 2-(2,2-di((9Z,12Z)- octadeca-9,1 2-dien- l-yl)-l ,3-dioxolan-4-yl)-N,N-dimethylethanamine ("DLin-KC2-DMA") (see, WO 2010/042877, which is incorporated herein by reference; Semple et al., Nature Biotech. 28: 172-176 (2010)). (Heyes, J., et al., J Controlled Release 107: 276-287 (2005); Morrissey, DV., et al., Nat.

Biotechnol. 23(8): 1003-1007 (2005); International Patent Publication WO 2005/121348). In some embodiments, one or more of the cationic lipids comprise at least one of an imidazole, dialkylamino, or guanidinium moiety.

[0253] In some embodiments, one or more cationic lipids suitable for the compositions include 2,2-Dilinoleyl-4-dimethylaminoethyl-[l,3]-dioxolane ("XTC"); (3aR,5s,6aS)-N,N-dimethyl- 2,2-di((9Z,12Z)-octadeca-9,12-dienyl)tetrahydro-3aH-cyclopen ta[d] [1 ,3]dioxol-5-amine ("ALNY- 100") and/or 4,7,13-tris(3-oxo-3-(undecylamino)propyl)-Nl,N16-diundecyl-4 ,7,10,13- tetraazahexadecane-1, 16-diamide ("NC98-5").

[0254] In some embodiments, the compositions include one or more cationic lipids that constitute at least about 5%, 10%, 20%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, or 70%, measured by weight, of the total lipid content in the composition, e.g., a lipid nanoparticle. In some embodiments, the compositions include one or more cationic lipids that constitute at least about 5%, 10%, 20%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, or 70%, measured as a mol %, of the total lipid content in the composition, e.g., a lipid nanoparticle. In some embodiments, the compositions include one or more cationic lipids that constitute about 30-70 % (e.g., about 30-65%, about 30-60%, about 30-55%, about 30-50%, about 30-45%, about 30-40%, about 35-50%, about 35-45%, or about 35-40%), measured by weight, of the total lipid content in the composition, e.g., a lipid nanoparticle. In some embodiments, the compositions include one or more cationic lipids that constitute about 30-70 % (e.g., about 30-65%, about 30-60%, about 30-55%, about 30-50%, about 30-45%, about 30- 40%, about 35-50%, about 35-45%, or about 35-40%), measured as mol %, of the total lipid content in the composition, e.g., a lipid nanoparticle.

Helper Lipids

[0255] Compositions ( e.g ., liposomal compositions) may also comprise one or more helper lipids. Such helper lipids include non-cationic lipids. As used herein, the phrase "non-cationic lipid" refers to any neutral, zwitterionic or anionic lipid. As used herein, the phrase "anionic lipid" refers to any of a number of lipid species that carry a net negative charge at a selected pH, such as physiological pH. Non-cationic lipids include, but are not limited to, distearoylphosphatidylcholine (DSPC), dioleoylphosphatidylcholine (DOPC), dipalmitoylphosphatidylcholine (DPPC),

dioleoylphosphatidylglycerol (DOPG), dipalmitoylphosphatidylglycerol (DPPG),

dioleoylphosphatidylethanolamine (DOPE), palmitoyloleoylphosphatidylcholine (POPC), palmitoyloleoyl-phosphatidylethanolamine (POPE), dioleoyl-phosphatidylethanolamine 4-(N- maleimidomethyl)-cyclohexane-l-carboxylate (DOPE-mal), dipalmitoyl phosphatidyl ethanolamine (DPPE), dimyristoylphosphoethanolamine (DMPE), distearoyl-phosphatidyl-ethanolamine (DSPE), 16- O-monomethyl PE, 16-O-dimethyl PE, 18-1-trans PE, l-stearoyl-2-oleoyl-phosphatidyethanolamine (SOPE), or a mixture thereof. In embodiments, a non-cationic or helper lipid is

dioleoylphosphatidylethanolamine (DOPE).

[0256] In some embodiments, a non-cationic lipid is a neutral lipid, i.e., a lipid that does not carry a net charge in the conditions under which the composition is formulated and/or administered.

[0257] In some embodiments, a non-cationic lipid may be present in a molar ratio (mol%) of about 5% to about 90%, about 5% to about 70%, about 5% to about 50%, about 5% to about 40%, about 5% to about 30%, about 10 % to about 70%, about 10% to about 50%, or about 10% to about 40% of the total lipids present in a composition. In some embodiments, total non-cationic lipids may be present in a molar ratio (mol%) of about 5% to about 90%, about 5% to about 70%, about 5% to about 50%, about 5% to about 40%, about 5% to about 30%, about 10 % to about 70%, about 10% to about 50%, or about 10% to about 40% of the total lipids present in a composition. In some embodiments, the percentage of non-cationic lipid in a liposome may be greater than about 5 mol%, greater than about 10 mol%, greater than about 20 mol%, greater than about 30 mol%, or greater than about 40 mol%. In some embodiments, the percentage total non-cationic lipids in a liposome may be greater than about 5 mol%, greater than about 10 mol%, greater than about 20 mol%, greater than about 30 mol%, or greater than about 40 mol%. In some embodiments, the percentage of non-cationic lipid in a liposome is no more than about 5 mol%, no more than about 10 mol%, no more than about 20 mol%, no more than about 30 mol%, or no more than about 40 mol%. In some embodiments, the percentage total non-cationic lipids in a liposome may be no more than about 5 mol%, no more than about 10 mol%, no more than about 20 mol%, no more than about 30 mol%, or no more than about 40 mol%.

[0258] In some embodiments, a non-cationic lipid may be present in a weight ratio (wt%) of about 5% to about 90%, about 5% to about 70%, about 5% to about 50%, about 5% to about 40%, about 5% to about 30%, about 10 % to about 70%, about 10% to about 50%, or about 10% to about 40% of the total lipids present in a composition. In some embodiments, total non-cationic lipids may be present in a weight ratio (wt%) of about 5% to about 90%, about 5% to about 70%, about 5% to about 50%, about 5% to about 40%, about 5% to about 30%, about 10 % to about 70%, about 10% to about 50%, or about 10% to about 40% of the total lipids present in a composition. In some embodiments, the percentage of non-cationic lipid in a liposome may be greater than about 5 wt%, greater than about 10 wt%, greater than about 20 wt%, greater than about 30 wt%, or greater than about 40 wt%. In some embodiments, the percentage total non-cationic lipids in a liposome may be greater than about 5 wt%, greater than about 10 wt%, greater than about 20 wt%, greater than about 30 wt%, or greater than about 40 wt%. In some embodiments, the percentage of non-cationic lipid in a liposome is no more than about 5 wt%, no more than about 10 wt%, no more than about 20 wt%, no more than about 30 wt%, or no more than about 40 wt%. In some embodiments, the percentage total non-cationic lipids in a liposome may be no more than about 5 wt%, no more than about 10 wt%, no more than about 20 wt%, no more than about 30 wt%, or no more than about 40 wt%.

Cholesterol-based Lipids

[0259] In some embodiments, a composition ( e.g ., a liposomal composition) comprises one or more cholesterol-based lipids. For example, suitable cholesterol-based lipids include cholesterol and, for example, DC-Chol (N,N-dimethyl-N-ethylcarboxamidocholesterol), l,4-bis(3-N-oleylamino- propyljpiperazine (Gao, et al. Biochem. Biophys. Res. Comm. 179, 280 (1991); Wolf et al.

BioTechniques 23, 139 (1997); U.S. Pat. No. 5,744,335), or imidazole cholesterol ester (ICE), which has the following structure,

[0260] In some embodiments, a cholesterol-based lipid may be present in a molar ratio

(mol%) of about 1% to about 30%, or about 5% to about 20% of the total lipids present in a liposome. In some embodiments, the percentage of cholesterol-based lipid in the lipid nanoparticle may be greater than about 5 mol%, greater than about 10 mol%, greater than about 20 mol%, greater than about 30 mol%, or greater than about 40 mol%. In some embodiments, the percentage of cholesterol-based lipid in the lipid nanoparticle may be no more than about 5 mol%, no more than about 10 mol%, no more than about 20 mol%, no more than about 30 mol%, or no more than about 40 mol%.

[0261] In some embodiments, a cholesterol-based lipid may be present in a weight ratio

(wt%) of about 1% to about 30%, or about 5% to about 20% of the total lipids present in a liposome. In some embodiments, the percentage of cholesterol-based lipid in the lipid nanoparticle may be greater than about 5 wt%, greater than about 10 wt%, greater than about 20 wt%, greater than about 30 wt%, or greater than about 40 wt%. In some embodiments, the percentage of cholesterol- based lipid in the lipid nanoparticle may be no more than about 5 wt%, no more than about 10 wt%, no more than about 20 wt%, no more than about 30 wt%, or no more than about 40 wt%.

PEGylated Lipids

[0262] In some embodiments, a composition ( e.g ., a liposomal composition) comprises one or more further PEGylated lipids.

[0263] For example, the use of polyethylene glycol (PEG)-modified phospholipids and derivatized lipids such as derivatized ceramides (PEG-CER), including N-octanoyl-sphingosine-1- [succinyl(methoxy polyethylene glycol)-2000] (C8 PEG-2000 ceramide) is also contemplated by the present invention in combination with one or more of compounds described herein {e.g., a compound of Formula (I) such as Compounds (1)— (4)) and, in some embodiments, other lipids together which comprise the liposome. In some embodiments, particularly useful exchangeable lipids are PEG-ceramides having shorter acyl chains {e.g., CM or Cis).

[0264] Contemplated further PEG-modified lipids (also referred to herein as a PEGylated lipid, which term is interchangeable with PEG-modified lipid) include, but are not limited to, a polyethylene glycol chain of up to 5 kDa in length covalently attached to a lipid with alkyl chain(s) of C -C length. In some embodiments, a PEG-modified or PEGylated lipid is PEGylated cholesterol or PEG-2K. The addition of such components may prevent complex aggregation and may also provide a means for increasing circulation lifetime and increasing the delivery of the lipid-nucleic acid composition to the target cell, (Klibanov et al. (1990) FEBS Letters, 268 (1): 235-237), or they may be selected to rapidly exchange out of the formulation in vivo (see U.S. Pat. No. 5,885,613).

[0265] Further PEG-modified phospholipid and derivatized lipids of the present invention may be present in a molar ratio (mol%) from about 0% to about 15%, about 0.5% to about 15%, about 1% to about 15%, about 4% to about 10%, or about 2% of the total lipid present in the composition {e.g., a liposomal composition). [0266] Further PEG-modified phospholipid and derivatized lipids of the present invention may be present in a weight ratio (wt%) from about 0% to about 15%, about 0.5% to about 15%, about 1% to about 15%, about 4% to about 10%, or about 2% of the total lipid present in the composition ( e.g ., a liposomal composition).

Pharmaceutical Formulations and Therapeutic Uses

[0267] Compounds described herein {e.g., a compound of Formula (I) such as Compounds

( 1)— (4)) may be used in the preparation of compositions {e.g., to construct liposomal compositions) that facilitate or enhance the delivery and release of encapsulated materials {e.g., one or more therapeutic polynucleotides) to one or more target cells {e.g., by permeating or fusing with the lipid membranes of such target cells).

[0268] For example, when a liposomal composition {e.g., a lipid nanoparticle) comprises or is otherwise enriched with one or more of the compounds disclosed herein, the phase transition in the lipid bilayer of the one or more target cells may facilitate the delivery of the encapsulated materials {e.g., one or more therapeutic polynucleotides encapsulated in a lipid nanoparticle) into the one or more target cells.

[0269] Similarly, in certain embodiments compounds described herein {e.g., a compound of

Formula (I) such as Compounds (1)— (4)) may be used to prepare liposomal vehicles that are characterized by their reduced toxicity in vivo. In certain embodiments, the reduced toxicity is a function of the high transfection efficiencies associated with the compositions disclosed herein, such that a reduced quantity of such composition may administered to the subject to achieve a desired therapeutic response or outcome.

[0270] Thus, pharmaceutical formulations comprising a compound described {e.g., a compound of Formula (I) such as Compounds (1)— (4)) and nucleic acids provided by the present invention may be used for various therapeutic purposes. To facilitate delivery of nucleic acids in vivo, a compound described herein {e.g., a compound of Formula (I) such as Compounds (1)— (4)) and nucleic acids can be formulated in combination with one or more additional pharmaceutical carriers, targeting ligands or stabilizing reagents. In some embodiments, a compound described herein {e.g., a compound of Formula (I) such as Compounds (1)— (4)) can be formulated via pre-mixed lipid solution. In other embodiments, a composition comprising a compound described herein {e.g., a compound of Formula (I) such as Compounds (1)— (4)) can be formulated using post-insertion techniques into the lipid membrane of the nanoparticles. Techniques for formulation and administration of drugs may be found in "Remington's Pharmaceutical Sciences," Mack Publishing Co., Easton, Pa., latest edition. [0271] Suitable routes of administration include, for example, oral, rectal, vaginal, transmucosal, pulmonary including intratracheal or inhaled, or intestinal administration; parenteral delivery, including intradermal, transdermal (topical), intramuscular, subcutaneous, intramedullary injections, as well as intrathecal, direct intraventricular, intravenous, intraperitoneal, or intranasal.

In particular embodiments, the intramuscular administration is to a muscle selected from the group consisting of skeletal muscle, smooth muscle and cardiac muscle. In some embodiments the administration results in delivery of the nucleic acids to a muscle cell. In some embodiments the administration results in delivery of the nucleic acids to a hepatocyte (i.e., liver cell).

[0272] Alternatively or additionally, pharmaceutical formulations of the invention may be administered in a local rather than systemic manner, for example, via injection of the

pharmaceutical formulation directly into a targeted tissue, preferably in a sustained release formulation. Local delivery can be affected in various ways, depending on the tissue to be targeted. Exemplary tissues in which delivered mRNA may be delivered and/or expressed include, but are not limited to the liver, kidney, heart, spleen, serum, brain, skeletal muscle, lymph nodes, skin, and/or cerebrospinal fluid. In embodiments, the tissue to be targeted in the liver. For example, aerosols containing compositions of the present invention can be inhaled (for nasal, tracheal, or bronchial delivery); compositions of the present invention can be injected into the site of injury, disease manifestation, or pain, for example; compositions can be provided in lozenges for oral, tracheal, or esophageal application; can be supplied in liquid, tablet or capsule form for administration to the stomach or intestines, can be supplied in suppository form for rectal or vaginal application; or can even be delivered to the eye by use of creams, drops, or even injection.

[0273] Compositions described herein can comprise mRNA encoding peptides including those described herein (e.g., a polypeptide such as a protein).

[0274] In embodiments, a mRNA encodes a polypeptide.

[0275] In embodiments, a mRNA encodes a protein.

[0276] Exemplary peptides encoded by mRNA (e.g., exemplary proteins encoded by mRNA) are described herein.

[0277] The present invention provides methods for delivering a composition having full- length mRNA molecules encoding a peptide or protein of interest for use in the treatment of a subject, e.g., a human subject or a cell of a human subject or a cell that is treated and delivered to a human subject. [0278] Accordingly, in certain embodiments the present invention provides a method for producing a therapeutic composition comprising full-length mRNA that encodes a peptide or protein for use in the delivery to or treatment of the lung of a subject or a lung cell. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for cystic fibrosis transmembrane conductance regulator (CFTR) protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for ATP-binding cassette sub-family A member 3 protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for dynein axonemal intermediate chain 1 protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for dynein axonemal heavy chain 5 (DNAH5) protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for alpha-l-antitrypsin protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for forkhead box P3 (FOXP3) protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes one or more surfactant protein, e.g., one or more of surfactant A protein, surfactant B protein, surfactant C protein, and surfactant D protein.

[0279] In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes a peptide or protein for use in the delivery to or treatment of the liver of a subject or a liver cell. Such peptides and polypeptides can include those associated with a urea cycle disorder, associated with a lysosomal storage disorder, with a glycogen storage disorder, associated with an amino acid metabolism disorder, associated with a lipid metabolism or fibrotic disorder, associated with methylmalonic acidemia, or associated with any other metabolic disorder for which delivery to or treatment of the liver or a liver cell with enriched full-length mRNA provides therapeutic benefit.

[0280] In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for a protein associated with a urea cycle disorder. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for ornithine transcarbamylase (OTC) protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for arginosuccinate synthetase 1 protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for carbamoyl phosphate synthetase I protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for arginosuccinate lyase protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for arginase protein.

[0281] In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for a protein associated with a lysosomal storage disorder. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for alpha galactosidase protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for glucocerebrosidase protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for iduronate-2-sulfatase protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for iduronidase protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for N-acetyl-alpha-D-glucosaminidase protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for heparan N-sulfatase protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for galactosamine-6 sulfatase protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for beta- galactosidase protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for lysosomal lipase protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for arylsulfatase B (N- acetylgalactosamine-4-sulfatase) protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for transcription factor EB (TFEB).

[0282] In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for a protein associated with a glycogen storage disorder. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for acid alpha- glucosidase protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for glucose-6-phosphatase (G6PC) protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for liver glycogen phosphorylase protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for muscle phosphoglycerate mutase protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for glycogen debranching enzyme.

[0283] In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for a protein associated with amino acid metabolism. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for phenylalanine hydroxylase enzyme. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for glutaryl-CoA dehydrogenase enzyme. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for propionyl-CoA caboxylase enzyme. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for oxalase alanine-glyoxylate aminotransferase enzyme.

[0284] In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for a protein associated with a lipid metabolism or fibrotic disorder. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for a mTOR inhibitor. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for ATPase phospholipid transporting 8B1 (ATP8B1) protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for one or more NF-kappa B inhibitors, such as one or more of l-kappa B alpha, interferon-related development regulator 1 (IFRD1), and Sirtuin 1 (SIRT1). In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for PPAR-gamma protein or an active variant.

[0285] In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for a protein associated with methylmalonic acidemia. For example, in certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for methylmalonyl CoA mutase protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for methylmalonyl CoA epimerase protein.

[0286] In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA for which delivery to or treatment of the liver can provide therapeutic benefit. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for ATP7B protein, also known as Wilson disease protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for porphobilinogen deaminase enzyme. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for one or clotting enzymes, such as Factor VIII, Factor IX, Factor VII, and Factor X. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for human hemochromatosis (FIFE) protein.

[0287] In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes a peptide or protein for use in the delivery to or treatment of the cardiovasculature of a subject or a cardiovascular cell. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for vascular endothelial growth factor A protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for relaxin protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for bone morphogenetic protein-9 protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for bone morphogenetic protein-2 receptor protein.

[0288] In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes a peptide or protein for use in the delivery to or treatment of the muscle of a subject or a muscle cell. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for dystrophin protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for frataxin protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes a peptide or protein for use in the delivery to or treatment of the cardiac muscle of a subject or a cardiac muscle cell. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for a protein that modulates one or both of a potassium channel and a sodium channel in muscle tissue or in a muscle cell. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for a protein that modulates a Kv7.1 channel in muscle tissue or in a muscle cell. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for a protein that modulates a Navi.5 channel in muscle tissue or in a muscle cell.

[0289] In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes a peptide or protein for use in the delivery to or treatment of the nervous system of a subject or a nervous system cell. For example, in certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for survival motor neuron 1 protein. For example, in certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for survival motor neuron 2 protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for frataxin protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for ATP binding cassette subfamily D member 1 (ABCD1) protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for CLN3 protein.

[0290] In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes a peptide or protein for use in the delivery to or treatment of the blood or bone marrow of a subject or a blood or bone marrow cell.

In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for beta globin protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for Bruton's tyrosine kinase protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for one or clotting enzymes, such as Factor VIII, Factor IX, Factor VII, and Factor X.

[0291] In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes a peptide or protein for use in the delivery to or treatment of the kidney of a subject or a kidney cell. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for collagen type IV alpha 5 chain (COL4A5) protein. [0292] In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes a peptide or protein for use in the delivery to or treatment of the eye of a subject or an eye cell. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for ATP-binding cassette sub-family A member 4 (ABCA4) protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for retinoschisin protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for retinal pigment epithelium-specific 65 kDa (RPE65) protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for centrosomal protein of 290 kDa (CEP290).

[0293] In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes a peptide or protein for use in the delivery of or treatment with a vaccine for a subject or a cell of a subject. For example, in certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antigen from an infectious agent, such as a virus. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antigen from influenza virus. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antigen from respiratory syncytial virus. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antigen from rabies virus. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antigen from cytomegalovirus. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antigen from rotavirus. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antigen from a hepatitis virus, such as hepatitis A virus, hepatitis B virus, or hepatis C virus. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antigen from human papillomavirus. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antigen from a herpes simplex virus, such as herpes simplex virus 1 or herpes simplex virus 2. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antigen from a human immunodeficiency virus, such as human immunodeficiency virus type 1 or human immunodeficiency virus type 2. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antigen from a human metapneumovirus. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antigen from a human parainfluenza virus, such as human parainfluenza virus type 1, human parainfluenza virus type 2, or human parainfluenza virus type 3. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antigen from malaria virus. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antigen from zika virus. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antigen from chikungunya virus.

[0294] In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antigen associated with a cancer of a subject or identified from a cancer cell of a subject. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antigen determined from a subject's own cancer cell, i.e., to provide a personalized cancer vaccine. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antigen expressed from a mutant KRAS gene.

[0295] In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antibody. In certain embodiments, the antibody can be a bi-specific antibody. In certain embodiments, the antibody can be part of a fusion protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antibody to 0X40. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antibody to VEGF. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antibody to tissue necrosis factor alpha. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antibody to CD3. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an antibody to CD19. [0296] In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an immunomodulator. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for Interleukin 12. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for Interleukin 23. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for Interleukin 36 gamma. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for a constitutively active variant of one or more stimulator of interferon genes (STING) proteins.

[0297] In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an endonuclease. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for an RNA-guided DNA endonuclease protein, such as Cas 9 protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for a meganuclease protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for a transcription activator-like effector nuclease protein. In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for a zinc finger nuclease protein.

[0298] In embodiments, exemplary therapeutic uses result from the delivery of mRNA encoding a secreted protein. Accordingly, in embodiments, the compositions and methods of the invention provide for delivery of mRNA encoding a secreted protein. In some embodiments, the compositions and methods of the invention provide for delivery of mRNA encoding one or more secreted proteins listed in Table 1; thus, compositions of the invention may comprise an mRNA encoding a protein listed in Table 1 (or a homolog thereof) along with other components set out herein, and methods of the invention may comprise preparing and/or administering a composition comprising an mRNA encoding a protein listed in Table 1 (or a homolog thereof) along with other components set out herein

Table 1. Secreted Proteins

[0299] In some embodiments, the compositions and methods of the invention provide for the delivery of one or more mRNAs encoding one or more additional exemplary proteins listed in Table 2; thus, compositions of the invention may comprise an mRNA encoding a protein listed in Table 2 (or a homolog thereof) along with other components set out herein, and methods of the invention may comprise preparing and/or administering a composition comprising an mRNA encoding a protein chosen from the proteins listed in Table 2 (or a homolog thereof) along with other components set out herein.

Table 2. Additional Exemplary Proteins

[0300] The Uniprot IDs set forth in Table 1 and Table 2 refer to the human versions the listed proteins and the sequences of each are available from the Uniprot database. Sequences of the listed proteins are also generally available for various animals, including various mammals and animals of veterinary or industrial interest. Accordingly, in some embodiments, compositions and methods of the invention provide for the delivery of one or more mRNAs encoding one or more proteins chosen from mammalian homologs or homologs from an animal of veterinary or industrial interest of the secreted proteins listed in Table 1 and Table 2; thus, compositions of the invention may comprise an mRNA encoding a protein chosen from mammalian homologs or homologs from an animal of veterinary or industrial interest of a protein listed in Table 1 and Table 2 along with other components set out herein, and methods of the invention may comprise preparing and/or administering a composition comprising an mRNA encoding a protein chosen from mammalian homologs or homologs from an animal of veterinary or industrial interest of a protein listed in

Table 1 and Table 2 along with other components set out herein. In some embodiments, mammalian homologs are chosen from mouse, rat, hamster, gerbil, horse, pig, cow, llama, alpaca, mink, dog, cat, ferret, sheep, goat, or camel homologs. In some embodiments, the animal of veterinary or industrial interest is chosen from the mammals listed above and/or chicken, duck, turkey, salmon, catfish, or tilapia.

[0301] In embodiments, the compositions and methods of the invention provide for the delivery of mRNA encoding a lysosomal protein chosen from Table 3. In some embodiments, the compositions and methods of the invention provide for the delivery of one or more mRNAs encoding one or more lysosomal and/or related proteins listed in Table 3; thus, compositions of the invention may comprise an mRNA encoding a protein listed in Table 3 (or a homolog thereof) along with other components set out herein, and methods of the invention may comprise preparing and/or administering a composition comprising an mRNA encoding a protein chosen from the proteins listed in Table 3 (or a homolog thereof) along with other components set out herein.

Table 3. Lysosomal and Related Proteins

[0302] Information regarding lysosomal proteins is available from Lubke et al., "Proteomics of the Lysosome," Biochim Biophys Acta. (2009) 1793: 625-635. In some embodiments, the protein listed in Table 3 and encoded by mRNA in the compositions and methods of the invention is a human protein. Sequences of the listed proteins are also available for various animals, including various mammals and animals of veterinary or industrial interest as described above.

[0303] In some embodiments, the compositions and methods of the invention provide for the delivery of mRNA encoding a therapeutic protein ( e.g ., cytosolic, transmembrane or secreted) such as those listed in Table 4. In some embodiments, the compositions and methods of the invention provide for the delivery of an mRNA encoding a therapeutic protein useful in treating a disease or disorder (i.e., indication) listed in Table 4; thus, compositions of the invention may comprise an mRNA encoding a therapeutic protein listed or not listed in Table 4 (or a homolog thereof, as discussed below) along with other components set out herein for treating a disease or disorder (i.e., indication) listed in Table 4, and methods of the invention may comprise preparing and/or administering a composition comprising an mRNA encoding a such a protein (or a homolog thereof, as discussed below) along with other components set out herein for treatment of a disease or disorder listed in Table 4.

Table 4. Exemplary Indications and Related Proteins

[0304] In some embodiments, the present invention is used to prevent, treat and/or cure a subject affected with a disease or disorder listed or associated with the proteins listed in Tables 1, 2, 3, or 4. In some embodiments, an mRNA encodes one or more of Cystic Fibrosis Transmembrane Conductance Regulator (CFTR), argininosuccinate synthetase (ASS1), Factor IX, survival motor neuron 1 (SMN1), or phenylalanine hydroxylase (PAH).

[0305] While certain compounds, compositions and methods of the present invention have been described with specificity in accordance with certain embodiments, the following examples serve only to illustrate the compounds of the invention and are not intended to limit the same. EXAMPLES

Example 1: Synthesis of Compounds of the Invention

Compound (3)

[0306] The compounds described ( e.g Compound (3)) herein can be prepared according to the exemplary synthesis of Scheme 1.

Scheme 1

Synthesis of Compound 3.2

[0307] To a solution of compound 3.1 (5 g, 22.4 mmol) in anhydrous acetonitrile (25 mL) was added potassium carbonate (3.5 g, 25 mmol) followed by triphenylphosphine (5.9 g, 22.4 mmol). After addition complete, the reaction mixture was heated to reflux and stirred overnight. The reaction mixture was cooled to room temperature and filtered through cotton. The solid was rinsed with acetonitrile (lOmL and 5 mL). To the filtrate was added, slowly, diethyl ether (200 mL). The mixture was stirred for 90 minutes and the clear phase was decanted. To the solid residue was added diethyl ether (50 mL) and it was stirred for 30 minutes. After decanting the ether phase, the solid was dried under high vacuum to yield 7.95 g (73%) of compound 3.2.

Synthesis of Compound 3.4

[0308] Compound 3.3 (20 g, 200 mmol) was dissolved in a 2M THF solution of

dimethylamine (300 mL, 600 mmol). This reaction mixture was stirred under nitrogen at room temperature for 2 days. The reaction mixture was concentrated under reduced pressure. The residue was dried under high vacuum to give crude product compound 3.4 (27 g; 93%) that was used directly for next step without further purification.

Synthesis of Compound 3.5

[0309] To a solution of compound 3.4 (27 g, crude) in dichloromethane (200 mL) and diethyl ether (200 mL) at RT was added Celite (30 g) and PCC (30 g). After addition, the reaction mixture was stirred at RT for 3 h and then filtered through Celite. The filtrate was concentrated to give crude product which was purified by flash chromatography on silica gel (0-10 % methanol in ethyl acetate) to yield 2.5 g (8.7%, 2 steps) of compound 3.5.

Synthesis of Compound 3.6

[0310] To a solution of compound 3.2 (7.85 g, 16 mmol) and 3.5 (3.7 g, 26 mmol) in anhydrous THF (75 mL) and anhydrous dichloromethane (75 mL) at -75 °C (dry ice-acetone bath) under nitrogen atmosphere was added a solution of NaHMDS (19 ml, 1 M in THF). After addition, the reaction mixture was stirred at -75 °C (dry ice-acetone bath) for 0.5 h and then warmed up slowly to room temperature and stirred overnight. Water (200 mL) was added in and the resulting mixture was extracted with dichloromethane (200 mL x 2). Combined extracts were washed with brine and dried over Na2S04. Filtration and concentration gave crude product which was purified by flash chromatography on silica gel (0-100% diethyl ether/hexane) to yield 2 g of compound 3.6 (57%). Synthesis of Compound 3.7

[0311] To a solution of compound 3.6 (460 mg, 1.7 mmol) in THF (8.5 mL) at RT was added a solution of p-toluene sulfonic acid monohydrate (50 mg, 1.7 mmol) in methanol (8.5 mL). After addition, the reaction mixture was stirred at RT overnight. The reaction mixture was diluted with ethyl acetate and washed with sat. aqueous sodium bicarbonate and brine and dried over Na2S04. Filtration and concentration gave crude product which was purified by flash chromatography on silica gel (dichloromethane/methanol gradient) to yield 270 mg of compound 3.7 (86%).

Synthesis of Compound 3.8

[0312] To a solution of compound 3.7 (630 mg, 3.4 mmol) in anhydrous dichloromethane

(50 mL) at 0-5 °C (ice-water bath) with vigorously stirring was added triphenylphosphine (981 mg,

3.8 mmol) followed by carbon tetrabromide (1.25 g, 3.8 mmol). After addition finished, the reaction was stirred at 0-5 °C (ice-water bath) for 1.5 h and then warmed up slowly to room temperature and stirred overnight. More triphenylphosphine (93 mg, 0.34 mmol) followed by carbon tetrabromide (120 mg, 0.34 mmol) were added to the reaction mixture and stirring was continued for 20 h. The reaction mixture was concentrated under reduced pressure. The residue was purified by flash chromatography on silica gel (0-10 % methanol in dichloromethane) to yield 1.3 g semi-solid which contained 0.6 g of compound 3.8 (71%).

Synthesis of Compound 3.9

[0313] A mixture of compound 3.8 (5 g, 20 mmol) and triphenylphosphine (5.8g, 22 mmol) in anhydrous acetonitrile (90 mL) was heated to reflux overnight. The solvent was removed under reduced pressure. The residue was dissolved in dichloromethane. Under stirring, diethyl ether was added to give a cloudy mixture which was kept at -20 °C overnight. After decanting the clear solvent phase, the residue was dried under high vacuum to give a white foamy solid 5.9 g. The clear solvent phase was concentrated and treated with dichloromethane/diethyl ether again to give another crop of product, 3.6 g. Combining the two crops of product yielded in total of 9.5 g (92%) of product compound 3.9 as a white solid.

Synthesis of Compound 3.11

[0314] To a solution of compound 3.10 (40 g, 0.18mol) in anhydrous dichloromethane

(550mL) at 0-5 °C (ice-water bath) with vigorously stirring was added imidazole (36 g, 0.54mol) followed by TBSCI (40 g, 0.27mol). After addition finished, the reaction was warmed up slowly to room temperature and stirred for 60 hours. The reaction mixture was filtered through Celite. The Celite was rinsed with dichloromethane (100 mL x 2). The combined filtrate was washed with brine and dried over Na 2 S0 4 . Filtration and concentration gave 68 g crude which was purified by flash chromatography on silica gel (0-25 % ethyl acetate in hexane) to yield 47.6 g (78%) of compound 3.11 as a colorless oil.

Synthesis of Compound 3.12

[0315] To a mixture of magnesium (2.1 g, 89 mmol) and iodine (10 mg) in anhydrous THF

(90 mL) at room temperature was added 1,2-dibromoethane (0.1 mL). After the reaction initiated, a solution of compound 3.11 (15 g, 44.5 mmol) was added slowly, note and the reaction system was kept at reflux during the addition with gently heating (oil bath). After addition finished, the reaction was heated (oil bath) at reflux for 1 h and then cooled to 0-5 °C (ice-water bath). A solution of ethyl formate (1.6 mL, 20 mmol) in anhydrous TFH F (10 mL) was added. After addition finished, the reaction was warmed up slowly to room temperature and stirred overnight. The reaction mixture was added slowly to sat. aq. ammonium chloride solution and adjusted pFH to 7 with 1 M HCI. The resulting mixture was extracted with dichloromethane (150 mL x 3). Combined extracts were washed with brine and dried over Na 2 S0 4 . Filtration and concentration gave 13 g crude product which was purified by flash chromatography on silica gel (0-30% ethyl acetate/hexane) to yield 7.6 g (70%) of compound 3.12 as a slightly yellow oil.

Synthesis of Compound 3.13

[0316] To a mixture of compound 3.12 (7.6 g, 13.9 mmol), DMAP (1.7 g, 13.9 mmol) and anhydrous pyridine (3.5 mL, 41.8 mmol) in anhydrous dichloromethane (60 mL) and anhydrous toluene (6 mL) at 0-5 °C (ice-water bath) with vigorously stirring was added benzoyl chloride (2.1 mL, 18.1 mmol). After addition finished, the reaction was warmed up slowly to room temperature and stirred for 60 h. The reaction mixture was washed with brine and dried over Na 2 S0 4 . Filtration and concentration gave crude product which was purified by flash chromatography on silica gel (0-50% ethyl acetate/hexane) to yield 8.6 g (95%) of compound 3.13 as a slightly yellow oil.

Synthesis of Compound 3.14

[0317] To a solution of compound 3.13 (8.6 g, 13 mmol) in anhydrous TFH F (40 mL) at to room temperature under nitrogen atmosphere was dropped in a solution of TBAF (40 ml, 1 M in THF). After addition, the reaction mixture was stirred at to room temperature overnight. The reaction mixture was diluted with ethyl acetate and washed with brine and dried over Na 2 S0 4 . Filtration and concentration gave 12 g crude product. The crude was then purified by flash chromatography on silica gel (0-100% ethyl acetate/hexane) to yield 4.8 g (86%) of compound 3.14 as a light yellow oil.

Synthesis of Compound 3.15

[0318] To a mixture of PCC (2.6 g, 18 mmol) and Celite (2.6 g) in anhydrous

dichloromethane (40 mL) at 0-5 °C (ice-water bath) with vigorously stirring was dropped in a solution of compound 3.14 (2 g, 6 mmol) in anhydrous dichloromethane (10 mL). After addition finished, the reaction was stirred at 0-5 °C (ice-water bath) for 0.5 h and then warmed up slowly to room temperature and stirred overnight. The reaction mixture was filtered through a pad of silica gel.

The silica gel pad was washed with dichloromethane (50 mL x 3). The combined filtrate was concentrated to give 1.85 g crude product which was purified by flash chromatography on silica gel (0-100 % ethyl acetate in hexane) to yield 1.1 g (56%) of compound 3.15 as a colorless oil.

Synthesis of Compound 3.16

[0319] To a solution of compound 3.9 (5.55 g, 10.9 mmol) in anhydrous THF (40 mL) and anhydrous dichloromethane (20 mL) at -75 °C (dry ice-acetone bath) under nitrogen atmosphere was added a solution of NaHMDS (5.86 ml, 2 M in THF). After addition, the reaction mixture was stirred at -75 °C (dry ice-acetone bath) for 0.5 h and then at 0-5 °C (ice-water bath) for 2 h. The reaction system was re-cooled to -75 °C (dry ice-acetone bath). A solution of 3.15 (1.3 g, 3.1 mmol) in anhydrous THF (3.5 mL) and anhydrous dichloromethane (1.5 mL) was dropped in. After addition finished, the reaction was warmed up slowly to room temperature and stirred overnight. The reaction mixture was diluted with diethyl ether and washed with brine and dried over Na2S04. Filtration and concentration gave 6.8 g crude product. The crude was then purified by flash chromatography on silica gel (0-100% ethyl acetate/hexane) to yield 2.6 g light yellow semi-solid which contained product compound 3.16 and triphenylphosphine oxide in about 3:2 ratio. 2.3 g of the above semi-solid was triturated with a mixture of hexane and diethyl ether (v/v =3/1). The clear solution from trituration was decanted and concentrated. The resulting semi-solid residue was triturated with a mixture of hexane and diethyl ether. The clear solution from trituration was decanted and concentrated and the resulting semi-solid residue was purified by flash

chromatography on silica gel (0-100% ethyl acetate/hexane) to yield 1.06 g of compound 3.16 (53%) as a colorless oil.

Synthesis of Compound (3)

[0320] To a solution of compound 3.16 (1.06 g, 1.5 mmol) in anhydrous THF (40 mL) under nitrogen at -30- -40 °C (dry ice-acetonitrile bath) under nitrogen atmosphere was added a solution of UAIH4 (8.8 ml, 2 M in THF). After addition finished, the reaction mixture was stirred at -25 °C (dry ice-acetonitrile bath) for 40 minutes and then at 0-5 °C (ice-water bath) for 2.5 h. Sodium sulfate decahydrate was carefully added to the reaction mixture until gas evolution stopped. The resulting mixture was filtered through Celite. The Celite was rinsed with dichloromethane/methanol (v/v =9/1). The combined filtrate was concentrated to give 1.2 g crude which was purified by flash chromatography on silica gel (0-5 % methanol in dichloromethane that was previously saturated with N H 3 ) to yield 605 mg (70%) of Compound (3) as a colorless oil, which changed to a slightly yellow oil upon storage.

Compound (4)

[0321] The compounds described ( e.g ., Compound (4)) herein can be prepared according to the exemplary synthesis of Scheme 2.

Scheme 2

Synthesis of dimethyl (2Z,21Z)-12-(benzoyloxy)tricosa-2,21-dienedioate (compound 4.1)

[0322] To a solution of methyl 2-(bis(2,2,2-trifluoroethoxy)phosphoryl)acetate (2.5 g, 7.9 mmol) in anhydrous THF (90 mL), 18-crown ether (9.5 g, 36 mmol) and 3.16 was added. The resulting suspension was cooled with dry ice-acetone, and KHMDS (0.5M in toluene, 15.8 mL, 7.9 mmol) was added slowly with stirring. After the addition, the resulting reaction mixture was stirred at - 78 °C for 3 h. The reaction was quenched with saturated aqueous N H 4 CI solution and extracted with EtOAc (100 mL x 3). The combined organic phases were dried with Na2S04. After filtration, the solvent was evaporated. The resulting residue was purified by MPC (120 g column, eluted with EtOAc/Hexanes (0-30%), to provide 1.1 g (58%) of the desired product compound 4.1.

Synthesis of (2Z,21Z)-tricosa-2,21-diene-l,12,23-triol (compound 4.2)

[0323] To a solution of dimethyl (2Z,21Z)-12-(benzoyloxy)tricosa-2,21-dienedioate

(compound 4.1, 1.1 g, 2.08 mmol) in anhydrous THF (20 mL), DIBAL (1.0M in TH F, 20.8 mL, 20.8 mmol) was added slowly at - 78 °C. After the addition, the resulting reaction mixture was stirred at room temperature for 2 h, then cooled with ice bath. Saturated aqueous NFI CI solution (60 mL) was added and extracted with EtOAc (100 mL x 3). The combined organic phases were dried with Na S . After filtration, the solvent was evaporated. The resulting residue was purified by MPC (80 g column, eluted with MeOFI/DCM (0-15%), to provide 4.1 g (54%) of the desired product compound 4.2.

Synthesis of (2Z,21Z)-12-hydroxytricosa-2,21-diene-l,23-diyi bis(4- (dimethylamino)butanoate) (Compound (4))

[0324] To a solution of iodine (0.93 g, 3.67 mmol) in anhydrous DCM (40 mL), P(o-Tolyl

(1.1 g, 3.67 mmol), imidazole (0.65 g, 9.58 mmol) and 4-(dimethylamino)butanoic acid hydrogen chloride (0.41 g, 2.45 mmol) were added sequentially. The resulting mixture was stirred at room temperature for 0.5 h. Then a solution of (2Z,21Z)-tricosa-2,21-diene-l,12,23-triol (compound 4.2, 0.41 g, 1.1 mmol) in anhydrous DCM (50 mL) was added. The resulting reaction mixture was stirred at room temperature overnight. The reaction solution was washed with brine (50 mL x 3) and dried with Na SC> . After filtration, the solvent was evaporated. The residue was dissolved in DMSO and purified reverse phase MPC [80 g C-18 column, eluted with ACN/water (0.1% HCOOFH ) (0-100%)], to provide 0.19 g (28%) of pure desired product Compound (4).

Example 2: Synthesis and Expression of mRNA Formulation Comprising a Cationic Lipid of the Invention

[0325] An mRNA which encodes a protein is synthesized. Formulation Experimental

Details:

Messenger RNA Materials

[0326] Firefly Luciferase (FFL) and human erythropoietin (EPO) were synthesized by in vitro transcription from a plasmid DNA template encoding the gene, which was followed by the addition of a 5' cap structure (Capl) (Fechter and Brownlee, J. Gen. Virology 86:1239-1249(2005)) and a 3' poly(A) tail of approximately 200 nucleotides in length as determined by gel electrophoresis. 5' and 3' untranslated regions present in each mRNA product are represented as X and Y, respectively and defined as stated ( ide infra). [0327] Human erythropoietin (EPO) mRNA (SEQ ID NO:l):

XiAUGGGUGUGCACGAAUGUCCUGCUUGGCUGUGGCUCCUUCUCUCCCUGCUGUCCCUGC CUCUUGGAC

UCCCGGUGCUUGGAGCACCCCCGAGACUGAUCUGCGACAGCAGGGUGCUCGAGCGCU ACCUCCUGGAAG

CCAAGGAAGCCGAAAACAUCACUACUGGCUGCGCCGAACACUGCUCCCUGAACGAGA ACAUCACCGUGCC

GGACACCAAGGUCAACUUCUACGCGUGGAAGAGAAUGGAGGUCGGACAGCAAGCCGU GGAAGUGUGGC

AGGGACUUGCGCUCCUGUCGGAAGCCGUGCUGAGGGGACAAGCCCUGCUCGUGAACA GCUCACAGCCUU

GGGAGCCCCUGCAGCUGCAUGUCGACAAGGCCGUGUCCGGACUGCGCUCACUGACCA CUCUGCUGAGGG

CCUUGGGUGCCCAGAAAGAGGCUAUUUCCCCACCGGAUGCAGCCUCGGCAGCUCCUC UGCGGACCAUUA

CGGCGGACACCUUUCGGAAGCUGUUCCGCGUCUACAGCAAUUUCCUCCGGGGGAAGU UGAAACUGUAU

ACCGGCGAAGCCUGUCGGACUGGCGAUCGCUGAYi

[0328] Codon-Optimized Firefly Luciferase (FFL) mRNA (SEQ ID NO:2):

XiAUGGAAGAUGCCAAAAACAUUAAGAAGGGCCCAGCGCCAUUCUACCCACUCGAAGACG GGACCGCCGG

CGAGCAGCUGCACAAAGCCAUGAAGCGCUACGCCCUGGUGCCCGGCACCAUCGCCUU UACCGACGCACAU

AUCGAGGUGGACAUUACCUACGCCGAGUACUUCGAGAUGAGCGUUCGGCUGGCAGAA GCUAUGAAGCG

CUAUGGGCUGAAUACAAACCAUCGGAUCGUGGUGUGCAGCGAGAAUAGCUUGCAGUU CUUCAUGCCCG

UGUUGGGUGCCCUGUUCAUCGGUGUGGCUGUGGCCCCAGCUAACGACAUCUACAACG AGCGCGAGCUG

CUGAACAGCAUGGGCAUCAGCCAGCCCACCGUCGUAUUCGUGAGCAAGAAAGGGCUG CAAAAGAUCCUC

AACGUGCAAAAGAAGCUACCGAUCAUACAAAAGAUCAUCAUCAUGGAUAGCAAGACC GACUACCAGGGC

UUCCAAAGCAUGUACACCUUCGUGACUUCCCAUUUGCCACCCGGCUUCAACGAGUAC GACUUCGUGCCC

GAGAGCUUCGACCGGGACAAAACCAUCGCCCUGAUCAUGAACAGUAGUGGCAGUACC GGAUUGCCCAAG

GGCGUAGCCCUACCGCACCGCACCGCUUGUGUCCGAUUCAGUCAUGCCCGCGACCCC AUCUUCGGCAACC

AGAUCAUCCCCGACACCGCUAUCCUCAGCGUGGUGCCAUUUCACCACGGCUUCGGCA UGUUCACCACGCU

GGGCUACUUGAUCUGCGGCUUUCGGGUCGUGCUCAUGUACCGCUUCGAGGAGGAGCU AUUCUUGCGCA

GCUUGCAAGACUAUAAGAUUCAAUCUGCCCUGCUGGUGCCCACACUAUUUAGCUUCU UCGCUAAGAGCA

CUCUCAUCGACAAGUACGACCUAAGCAACUUGCACGAGAUCGCCAGCGGCGGGGCGC CGCUCAGCAAGG

AGGUAGGUGAGGCCGUGGCCAAACGCUUCCACCUACCAGGCAUCCGCCAGGGCUACG GCCUGACAGAAA

CAACCAGCGCCAUUCUGAUCACCCCCGAAGGGGACGACAAGCCUGGCGCAGUAGGCA AGGUGGUGCCCU

UCUUCGAGGCUAAGGUGGUGGACUUGGACACCGGUAAGACACUGGGUGUGAACCAGC GCGGCGAGCUG

UGCGUCCGUGGCCCCAUGAUCAUGAGCGGCUACGUUAACAACCCCGAGGCUACAAAC GCUCUCAUCGAC

AAGGACGGCUGGCUGCACAGCGGCGACAUCGCCUACUGGGACGAGGACGAGCACUUC UUCAUCGUGGAC

CGGCUGAAGAGCCUGAUCAAAUACAAGGGCUACCAGGUAGCCCCAGCCGAACUGGAG AGCAUCCUGCUG

CAACACCCCAACAUCUUCGACGCCGGGGUCGCCGGCCUGCCCGACGACGAUGCCGGC GAGCUGCCCGCCG

CAGUCGUCGUGCUGGAACACGGUAAAACCAUGACCGAGAAGGAGAUCGUGGACUAUG UGGCCAGCCAG

GUUACAACCGCCAAGAAGCUGCGCGGUGGUGUUGUGUUCGUGGACGAGGUGCCUAAA GGACUGACCGG CAAG UUGGACGCCCGCAAGAUCCGCGAGAUUCUCAU UAAGGCCAAGAAGGGCGGCAAGAUCGCCG UG UA

Yi

[0329] Xi (5' untranslated sequence) (SEQ I D N0:3):

GGACAGAUCGCCUGGAGACGCCAUCCACGCUGU UU UGACCUCCAUAGAAGACACCGGGACCGAUCCAGC

CUCCGCGGCCGGGAACGGUGCAU UGGAACGCGGAUUCCCCGUGCCAAGAGUGACUCACCGUCCU UGACA

CG

[0330] Yi (3' untranslated sequence) (SEQ I D N0:4):

CGGGUGGCAUCCCUGUGACCCCUCCCCAGUGCCUCUCCUGGCCCUGGAAGU UGCCACUCCAGUGCCCACC

AGCCU UGUCCUAAUAAAAUUAAGU UGCAUC

Formulation Protocol

[0331] Aliquots of 20 mg/mL ethanolic solutions of the compounds described herein, DOPE,

Cholesterol and DMG-PEG 2K were mixed and diluted with ethanol to the required concentration. Separately, an aqueous buffered solution ( 10 mM citrate/ 150 mM NaCI, pH 4.5) of h EPO mRNA was prepared form lmg/mL stock. The lipid solution and the aqueous m RNA solution were mixed using a pump system to yield a final suspension in 20% ethanol solution. The resulting nanoparticle suspension was diafiltrated with 10% trehalose solution.

[0332] Molar composition - DMG-PEG 2K: compound :CHOL: DOPE: :5:40:325:30.

[0333] Compound (3) formulation : 90.13 % encapsulation; Size - 119 nm.

[0334] Compound (4) formulation : 96 % encapsulation; Size - 76 nm.

Example 3: Exemplary Liposome Formulations for Delivery and Expression of mRNA

[0335] This example provides exemplary liposome formulations for effective delivery and expression of m RNA in vivo.

Lipid materials

[0336] The formulations described herein include a multi-component lipid mixture of varying ratios employing one or more cationic lipids described herein, one or more additional cationic lipids, helper lipids (e.g., non-cationic lipids and/or cholesterol-based lipids), and PEGylated lipids designed to encapsulate various nucleic acid-based materials. Additional cationic lipids can include (but not exclusively) DOTAP ( l,2-dioleyl-3-trimethylammonium propane), DODAP (1,2- dioleyl-3-dimethylammonium propane) , DOTMA (l,2-di-0-octadecenyl-3-trimethylammonium propane), DLin DMA (Heyes, J.; Palmer, L.; Bremner, K.; MacLachlan, I. "Cationic lipid saturation influences intracellular delivery of encapsulated nucleic acids" J. Contr. Rel. 2005, 107, 276-287), DLin-KC2-DMA (Semple, S.C. et al. "Rational Design of Cationic Lipids for siRNA Delivery" Nature Biotech. 2010, 28, 172-176), C12-200 (Love, K.T. et al. "Lipid-like materials for low-dose in vivo gene silencing" PNAS 2010, 107, 1864-1869), HGT4003, HGT5000, HGT5001, MC3, CKK-E12 (3,6-bis(4- (bis(2-hydroxydodecyl)amino)butyl)piperazine-2,5-dione), ICE, dialkylamino-based, imidazole-based, guanidinium-based, etc. Helper lipids can include (but not exclusively) DSPC (1,2-distearoyl-sn- glycero-3-phosphocholine), DPPC (l,2-dipalmitoyl-sn-glycero-3-phosphocholine), DOPE (1,2-dioleyl- sn-glycero-3-phosphoethanolamine), DPPE (l,2-dipalmitoyl-sn-glycero-3-phosphoethanolamine), DMPE (l,2-dimyristoyl-sn-glycero-3-phosphoethanolamine), DOPG (,2-dioleoyl-sn-glycero-3- phospho-(l'-rac-glycerol)), cholesterol, etc. The PEGylated lipids can include (but not exclusively) a poly(ethylene) glycol chain of up to 5 kDa in length covalently attached to a lipid with alkyl chain(s) of C -C length.

Polymeric Materials

[0337] Further formulations described herein may include various charged, polymeric materials which can include (but not exclusively) branched polyethyleneimine (PEI) (25 kDa) (Sigma #408727), protamine, PEGylated protamine, PLL, PEGylated PLL, etc.

mRNA Materials

[0338] Firefly Luciferase (FFL) and human erythropoietin (EPO) were synthesized by in vitro transcription from a plasmid DNA template encoding the gene, which was followed by the addition of a 5' cap structure (Capl) (Fechter, P.; Brownlee, G.G. "Recognition of mRNA cap structures by viral and cellular proteins" J. Gen. Virology 2005, 86, 1239-1249) and a 3' poly(A) tail of approximately 200 nucleotides in length as determined by gel electrophoresis. 5' and 3' untranslated regions present in each mRNA product are represented as X and Y, respectively, and defined as stated ( ide infra).

[0339] Exemplary mRNA sequences of human erythropoietin (EPO) and Codon-Optimized

Firefly Luciferase (FFL) are depicted in SEQ ID No. 1 and 2. Exemplary 5' and 3' UTR sequences are described in SEQ ID Nos. 3 and 4.

Example 4: In Vitro and In Vivo Studies

[0340] In Vitro Studies: In vitro transfections of unmodified mRNA were performed using

HEK293T cells. Transfections of one microgram of each mRNA construct are performed in separate wells using lipofectamine. Cells are harvested at select time points (e.g., 4 hours, 8 hours, 24 hours, 48 hours, 72 hours, etc.) and respective protein production are analyzed. For FFL mRNA, cell lysates are analyzed for luciferase production via bioluminescence assays. For EPO mRNA studies, cell supernatants are obtained and analyzed for EPO protein, using ELISA-based methods. A comparison of protein production over time is made.

[0341] In Vivo Studies: A comparison of protein production over time was made via injection of mRNA encapsulated nanoparticles (lipid or polymeric) into wild type mice (CD-I) versus unmodified mRNA delivered in identical fashion. Serum and organs were collected at select time points (e.g., 6 hr, 12 hr, 24 hr, 48 hr, 72 hr, etc.) and respective protein levels are monitored. FIG. 1 shows human EPO protein expression in mice dosed subcutaneously with hEPO mRNA encapsulated in LNP formulations comprising compound (3) or compound (4), respectively, compared to saline.

[0342] For FFL mRNA, liver homogenates were analyzed for luciferase production via bioluminescence assays. A comparison of protein production over time of is made. FIG. 2 shows FFL luciferase detection from FFL luciferase protein production in individual organs post-administration of mice dosed subcutaneously with FFL mRNA encapsulated in an LNP formulation comprising compound (4).