Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
DI-THIOESTER CATIONIC LIPIDS
Document Type and Number:
WIPO Patent Application WO/2020/227085
Kind Code:
A1
Abstract:
The present invention provides, in part, di-thioester cationic lipid compounds or a pharmaceutically acceptable salt 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:
KARVE SHRIRANG (US)
DEROSA FRANK (US)
Application Number:
PCT/US2020/031000
Publication Date:
November 12, 2020
Filing Date:
May 01, 2020
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
TRANSLATE BIO INC (US)
International Classes:
C07D295/08; A61K31/495; A61P35/00; C07C327/00
Domestic Patent References:
WO2015184256A22015-12-03
WO2011141705A12011-11-17
WO2019226925A12019-11-28
WO2020097384A12020-05-14
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
WO2018089801A12018-05-17
Foreign References:
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
US20160032356A12016-02-04
US201762464327P2017-02-27
US4897355A1990-01-30
US5171678A1992-12-15
US5334761A1994-08-02
US5744335A1998-04-28
US5885613A1999-03-23
US5780014A1998-07-14
Other References:
S. M. BERGE ET AL.: "pharmaceutically acceptable salts", 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., RNA, vol. 9, 2003, pages 1108 - 1122
GRUDZIEN, E., RNA, vol. 10, 2004, pages 1479 - 1487
GRUDZIEN-NOGALSKA, E., 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
J. MCCLELLANM. C. KING, CELL, vol. 141, 2010, pages 210 - 217
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.'1 ACAD. SCI., vol. 86, 1989, pages 6982
SEMPLE, NATURE BIOTECH., vol. 28, 2010, pages 172 - 176
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, BIOTECHNIQUES, vol. 23, 1997, pages 139
KLIBANOV, 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
Attorney, Agent or Firm:
ESPINO, Christine, G. et al. (US)
Download PDF:
Claims:
CLAIMS

What is claimed is:

1. A cationic lipid having the following structure:

or a pharmaceutically acceptable salt thereof, wherein

Substructure A is 3-9-membered heterocyclyl, C3-C6 cycloalkyl, 5-10-membered heteroaryl, or C6-C10 aryl;

each R1 is independently C6-C30 aliphatic;

each m is independently an integer of 0-6; and

each n is independently an integer of 1-12.

2. The cationic lipid of claim 1, wherein Substructure A is 5-6-membered heterocyclyl,

C5-Cs cycloalkyl, or C6-C10 aryl.

3. The cationic lipid of claim 1 or 2, wherein m is 0, 1, or 2.

4. The cationic lipid of any one of claims 1-3, having the following structure:

or a pharmaceutically acceptable salt thereof.

5. The cationic lipid of any one of claims 1-3, having the following structure:

or a pharmaceutically acceptable salt thereof.

6. The cationic lipid of any one of claims 1-3, having the following structure:

or a pharmaceutically acceptable salt thereof.

7. The cationic lipid of any one of claims 1-3, having the following structure:

or a pharmaceutically acceptable salt thereof.

8. A cationic lipid having the following structure:

or a pharmaceutically acceptable salt thereof, wherein

L1 is C2-C14 alkenyl;

each R1 is independently C6-C30 aliphatic; and

each n Is independently an integer of 1-12.

9. The cationic lipid of claim 8, wherein said C2-C12 alkenyl comprises 1, 2, or 3 carbon-carbon double bonds.

10. The cationic lipid of claim 8 or 9, having the following structure:

or a pharmaceutically acceptable salt thereof, wherein

each R2 and R3 is Independently hydrogen or C1-C6 alkyl.

11. The cationic lipid of claim 10, wherein R2 and R3 are both hydrogen.

12. The cationic lipid of claim 10, wherein one of R2 and R3 is hydrogen, and the other is unsubstituted C1-C6 alkyl.

13. The cationic lipid of any one of claims 10-12, wherein R2 and R3 are cis to each other.

14. The cationic lipid of any one of claims 10-12, wherein R2 and R3 are trans to each other.

15. The cationic lipid of claim 8 or 9, having the following structure:

or a pharmaceutically acceptable salt thereof, wherein

a is an integer of 0-6;

b is an integer of 0-2; and

c is an integer of 0-3;

and wherein

at least one of a and b is not zero, and

when a or b is 1, then c is not zero.

16. The cationic lipid of claim 15, wherein one and only one of a and b is not zero.

17. The cationic lipid of claim 15 or 16, wherein a is 2, and each of b and c is zero.

18. The cationic lipid of claim 15 or 16, wherein a is 1, b is 0, and c is 1.

19. A cationic lipid having the following structure:

or a pharmaceutically acceptable salt thereof, wherein

L1 is C1-C12 aliphatic or C1-C12 heteroaliphatic comprising an oxo (=O), -OH, -COR4, or -COzR4; each R1 is Independently C6-C30 aliphatic;

each R4 is independently hydrogen or C1-C6 alkyl; and

each n is independently an integer of 1-12.

20. The cationic lipid of claim 19, having the following structure:

or a pharmaceutically acceptable salt thereof, wherein

R5 Is hydrogen;

R6 is -OH or -COCH3; or

R5 and R6 combine to form an oxo substituent.

21. The cationic lipid of claim 19, having the following structure:

or a pharmaceutically acceptable salt thereof, wherein

a is an integer of 2-6;

b is 0 or 1.

22. The cationic lipid of claim 19, having the following structure:

or a pharmaceutically acceptable salt thereof.

23. The cationic lipid of claim 19, having the following structure:

or a pharmaceutically acceptable salt thereof.

24. A cationic lipid having the following structure

or a pharmaceutically acceptable salt thereof, wherein each R1 is independently C6-C30 aliphatic; and

each n is independently an integer of 1-12.

25. A cationic lipid having the following structure

or a pharmaceutically acceptable salt thereof, wherein each R1 is independently C6-C30 aliphatic; and

each n is independently an integer of 1-12.

26. The cationic lipid of any one of claims 1-25, wherein n is 1, 2, or 3.

27. The cationic lipid of any one of claims 1-26, wherein each R1 is C6-C24 alkyl.

28. The cationic lipid of claim 27, wherein each R1 is C8H17, C10H21, C12H25, CnH29, or C16H33.

29. The cationic lipid of any one of claims 1-26, wherein each R1 is C6-C24 alkene.

30. The cationic lipid of claim 29, wherein each R1 is C6H12CH=CHCH2CH=CHC5H11.

31. A cationic lipid that is any one of Compounds 1-378, or a pharmaceutically acceptable salt thereof.

32. 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 1-31.

33. The composition of claim 32, comprising an mRNA encoding for cystic fibrosis

transmembrane conductance regulator (CFTR) protein.

34. The composition of claim 32, comprising an mRNA encoding for ornithine transcarbamylase (OTC) protein.

35. A composition comprising a nucleic acid encapsulated within a liposome, wherein the liposome comprises a cationic lipid according to any one of claims 1-31.

36. The composition of claim 35, wherein the nucleic acid is an mRNA encoding a peptide or protein.

37. The composition of claim 36, 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.

38. The composition of claim 37, wherein the mRNA encodes cystic fibrosis transmembrane conductance regulator (CFTR) protein.

39. The composition of claim 36, 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.

40. The composition of claim 39, wherein the mRNA encodes ornithine transcarbamylase (OTC) protein.

41. The composition of claim 32 or 35, wherein the mRNA encodes a peptide or protein for use in vaccine.

42. The composition of claim 41, wherein the mRNA encodes an antigen.

Description:
DI-THIOESTER CATIONIC LIPIDS

RELATED APPLICATIONS

[001] This application claims benefit of U.S. Provisional Application Number 62/842,848, filed on May 3, 2019, the entire disclosure of which is hereby incorporated by reference.

BACKGROUND

[002] 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

[003] The present invention provides, among other things, a novel class of di-thloester cationic lipid compounds for improved in vivo delivery of therapeutic agents, such as nucleic acids. It is contemplated that the compounds provided herein are capable of highly effective in vivo delivery while maintaining a favorable toxicity profile.

[004] In an aspect, provided herein are cationic lipids having a structure according to Formula (A),

or a pharmaceutically acceptable salt thereof, wherein

Substructure A is 3-9-membered heterocyclyl, C 3 -C 6 cycloalkyl, 5-10-membered heteroaryl, or C 6 -C 10 aryl;

each R 1 is independently C 6 -C 30 aliphatic;

each m is independently an integer of 0-6; and

each n is independently an integer of 1-12.

[005] In embodiments, Substructure A is 5-6-membered heterocyclyl, C 5 -C 6 cycloalkyl, or C 6 -C 10 aryl.

[006] In embodiments, m is 0, 1, or 2. [007] In embodiments, a cationic lipid of Formula (A) has the following structure:

or a pharmaceutically acceptable salt thereof.

[008] In embodiments, a cationic lipid of Formula (A) has the following structure:

or a pharmaceutically acceptable salt thereof.

[009] In embodiments, a cationic lipid of Formula (A) has the following structure:

(iii-i), or a pharmaceutically acceptable salt thereof.

[010] In embodiments, a cationic lipid of Formula (A) has the following structure: or a pharmaceutically acceptable salt thereof.

[011] In another aspect, a cationic lipid has the following structure:

or a pharmaceutically acceptable salt thereof, wherein

L 1 is C 2 -C 14 alkenyl;

each R 1 is independently C 6 -C 30 aliphatic; and

each n is independently an integer of 1-12.

[012] In embodiments, the C 2 -C 12 alkenyl comprises 1, 2, or 3 carbon-carbon double bonds.

[013] In embodiments, a cationic lipid of Formula (B) has the following structure:

or a pharmaceutically acceptable salt thereof, wherein

each R 2 and R 3 is Independently hydrogen or C 1 -C 6 alkyl.

[014] In embodiments, R 2 and R 3 are both hydrogen.

[015] In embodiments, wherein one of R 2 and R 3 is hydrogen, and the other is unsubstituted C 1 -C 6 alkyl.

[016] In embodiments, R 2 and R 3 are cis to each other.

[017] In embodiments, R 2 and R 3 are trans to each other.

[018] In embodiments, a cationic lipid of Formula (B) has the following structure:

or a pharmaceutically acceptable salt thereof, wherein a is an integer of 0-6;

b is an integer of 0-2; and

c is an integer of 0-3;

and wherein

at least one of a and b is not zero, and

when a or b is 1, then c is not zero.

[019] In embodiments, one and only one of a and b is not zero.

[020] In embodiments, a is 2, and each of b and c is zero.

[021] In embodiments, a is 1, b is 0, and c is 1.

[022] In another aspect, a cationic lipid has the following structure:

or a pharmaceutically acceptable salt thereof, wherein

L 1 is C 1 -C 12 aliphatic or C 1 -C 12 heteroaliphatic comprising an oxo (=O), -OH, -COR 4 , or -CO 2 R 4 ; each R 1 is Independently C 6 -C 30 aliphatic;

each R 4 is independently hydrogen or C 1 -C 6 alkyl; and

each n is independently an integer of 1-12.

[023] In embodiments, a cationic lipid of Formula (C) has the following structure:

or a pharmaceutically acceptable salt thereof, wherein

R 5 is hydrogen;

R 6 is -OH or -COCH 3 ; or

R 5 and R 6 combine to form an oxo substituent. [024] In embodiments, a cationic lipid of Formula (C) has the following structure:

or a pharmaceutically acceptable salt thereof, wherein

a is an integer of 2-6;

b is 0 or 1.

[025] In embodiments, a cationic lipid of Formula (C) has the following structure:

or a pharmaceutically acceptable salt thereof.

[026] In embodiments, a cationic lipid of Formula (C) has the following structure:

or a pharmaceutically acceptable salt thereof.

[027] In another aspect, a cationic lipid has the following structure:

or a pharmaceutically acceptable salt thereof, wherein

each R 1 is independently C 6 -C 30 aliphatic; and

each n is independently an integer of 1-12. [028] In another aspect, a cationic lipid has the following structure:

or a pharmaceutically acceptable salt thereof, wherein

each R 1 is independently C 6 -C 30 aliphatic; and

each n is independently an integer of 1-12.

[029] In embodiments, n is 1, 2, or 3.

[030] In embodiments, each R 1 is C 6 -C 24 alkyl.

[031] In embodiments, each R 1 is C 8 H 17 , C 10 H 21 , C 12 H 25 , C 14 H 29 , or C 16 H 33 .

[032] In embodiments, each R 1 is C 6 -C 24 alkene.

[033] In embodiments, each R 1 is C 6 H 12 CH=CHCH 2 CH=CHC 5 H 11 .

[034] In embodiments, a cationic lipid is any one of Compounds 1-378.

[035] In another aspect, the invention features a composition comprising any liposome (e.g., a liposome encapsulating an mRNA encoding a protein) described herein. In embodiments, the liposome comprises a cationic lipid described herein.

[036] In embodiments, an mRNA encodes for cystic fibrosis transmembrane conductance

regulator (CFTR) protein.

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

[038] In another aspect, the invention features a composition comprising a nucleic acid

encapsulated within a liposome as described herein. In embodiments, the liposome comprises a cationic lipid described herein.

[039] 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.

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

[041] 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. [042] In embodiments, an mRNA encodes for cystic fibrosis transmembrane conductance regulator (CFTR) protein.

[043] 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.

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

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

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

[047] 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 DRAWINGS

[048] FIG. 1 depicts in vivo protein production resulting from the delivery of mRNA using lipid nanoparticles comprising Compound 79 or Compound 98 as described herein. As shown in this Figure, use of these compounds results in high levels of in vivo protein production 6 hours after administration. Further, Compound 98 also results In high levels of in vivo protein production even 24 hours after administration.

DETAILED DESCRIPTION OF CERTAIN EMBODIMENTS

Definitions

[049] 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.

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

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

[051] 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, a bovine, 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.

[052] 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).

[053] 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.

[054] 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").

[055] 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 equivalents thereof, are used interchangeably.

[056] 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.

[057] 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.

[058] Helper lipid: The term "helper lipid" as used herein refers to any neutral or zwitterionlc 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/nanoparticles.

[059] 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.

[060] 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.

[061] 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).

[062] 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.).

[063] 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). In 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).

[064] messenger RNA (mRNA): As used herein, the term "messenger RNA (mRNA)" or "mRNA" refers to a polynucleotide that encodes at least one polypeptide. mRNA as used herein encompasses both modified and unmodified RNA. The term "modified mRNA" related to mRNA comprising at least one chemically modified nucleotide. mRNA may contain one or more coding and non-coding regions. mRNA can be purified from natural sources, produced using recombinant expression systems and optionally purified, chemically synthesized, etc. Where appropriate, e.g., in the case of chemically synthesized molecules, mRNA can comprise nucleoside analogs such as analogs having chemically modified bases or sugars, backbone modifications, etc. An mRNA sequence is presented In the 5' to 3' direction unless otherwise indicated. In some embodiments, an mRNA is or comprises natural nucleosides (e.g., adenosine, guanosine, cytidine, uridine); nucleoside analogs (e.g., 2-aminoadenosine, 2-thiothymidine, Inosine, pyrrolo-pyrimidine, 3-methyl adenosine, 5-methylcytidine, C-5 propynyl-cytidine, C-5 propynyl-uridine, 2-aminoadenosine, C5-bromouridine, C5-fluorouridine, C5-iodouridine, C5- propynyl-uridine, C5-propynyl-cytidine, C5-methylcytidine, 2-aminoadenosine, 7- deazaadenosine, 7-deazagua nosine, 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, arablnose, and hexose); and/or modified phosphate groups (e.g., phosphorothloates and 5'-/V-phosphoramidite linkages). [065] 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 (siRNA), transacting siRNA (tasiRNA), repeat associated siRNA (rasiRNA), 73K RNA, retrotransposons, a viral genome, a viroid, satellite RNA, or derivatives of these groups. In some embodiments, a nucleic acid is a mRNA encoding a protein such as an enzyme.

[066] 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. [067] 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.

[068] 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 malonlc 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 1-4 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 quartemlzation of an amine using an appropriate electrophile, e.g., an alkyl halide, to form a quarternized alkylated amino salt.

[069] Systemic distribution or delivery. As used herein, the terms "systemic distribution" or

"systemic delivery," or grammatical equivalents thereof, 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." [070] 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.

[071] 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.

[072] 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.

[073] 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.

[074] 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.

[075] Aliphatic: As used herein, the term aliphatic refers to C1.C40 hydrocarbons and includes both saturated and unsaturated hydrocarbons. An aliphatic may be linear, branched, or cyclic. For example, C 1 -C 20 allphatics can include C 1 -C 20 alkyls (e.g., linear or branched C 1 -C 20 saturated alkyls), C2-C20 alkenyls (e.g., linear or branched C2-C20 monoalkenyls, linear or branched C4-C20 dienyls, linear or branched C6-C20 trienyls, and the like), and C2-C20 alkynyls (e.g., linear or branched C2-C20 alkynyls). C 1 -C 20 aliphatics can include C3-C20 cyclic aliphatics (e.g., C3-C20 cycloalkyls, C 4 -C 20 cycloalkenyls, or C 8 -C 20 cycloalkynyls). For example, C 3-6 cycloaliphatic groups Include, without limitation, cyclopropyl (C 3 ), cyclobutyl ( C 4 ), cyclopentyl (C 5 ), cyclopentenyl (C 5 ), cyclohexyl (C 6 ), cyclohexenyl (C 6 ), cyclohexadienyl (C 6 ) and the like. In embodiments, an aliphatic group is acyclic (e.g., linear or branched C 1 -C 20 saturated alkyls, linear or branched C 2 -C 20 monoalkenyls, linear or branched C 4 -C 20 dienyls, linear or branched C 6 -C 20 trienyls, or linear or branched C 2 -C 20 alkynyls). In embodiments, an aliphatic group is cyclic (a cyclic aliphatic or a cycloaliphatic). Fully saturated cycloaliphatics can be termed "cycloalkyl". Partially unsaturated cycloalkyl groups can be termed "cycloalkenyl" if the carbocycle contains at least one double bond, or "cycloalkynyl" if the carbocycle contains at least one triple bond. In certain embodiments, the aliphatic may comprise one or more cyclic aliphatic and/or one or more heteroatoms such as oxygen, nitrogen, or sulfur (a "heteroaliphatic" group) and/or 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 H, -CO 2 R', -CN, -OH, -OR', -OCOR', -OCO 2 R', -NH 2 ,

-NHR', -N(R')2, -SR' or-SO 2 R', wherein each instance of R* independently is C 1 -C 20 aliphatic (e.g., C 1 -C 20 alkyl, C 1 -C 15 alkyl, C 1 -C 10 alkyl, or C 1 -C 3 alkyl). 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.

[076] Alkyl: As used herein, the term "alkyl" means acyclic linear and branched hydrocarbon groups, e.g. "C 1 -C 30 alkyl" refers to alkyl groups having 1-30 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, isohexyl, etc. The term "lower alkyl" means an alkyl group straight chain or branched alkyl having 1 to 6 carbon atoms. 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', -NH 2 , -NHR', -N(R') 2 , -SR' or- SO 2 R', wherein each instance of R' independently is C 1 -C 20 aliphatic (e.g., C 1 -C 20 alkyl, C 1 -C 1 s 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 1 s 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 "hydroxyalkyt" group, where the prefix denotes the -OH group and "alkyl" is as described herein.

[077] Affixing the suffix "-ene" to a group indicates the group is a divalent moiety, e.g., arylene is the divalent moiety of aryl, and heteroarylene is the divalent moiety of heteroaryl.

[078] Alkylene: The term "alkylene," as used herein, represents a saturated divalent straight or branched chain hydrocarbon group and is exemplified by methylene, ethylene, isopropylene and the like. Likewise, the term "alkenylene" as used herein represents an unsaturated divalent straight or branched chain hydrocarbon group having one or more unsaturated carbon-carbon double bonds that may occur In any stable point along the chain, and the term "alkynylene" herein represents an unsaturated divalent straight or branched chain hydrocarbon group having one or more unsaturated carbon-carbon triple bonds that may occur in any stable point along the chain. In certain embodiments, an alkylene, alkenylene, or alkynylene group may comprise one or more cyclic aliphatic and/or one or more heteroatoms such as oxygen, nitrogen, or sulfur and may optionally be substituted with one or more substituents such as alkyl, halo, alkoxyl, hydroxy, amino, aryl, ether, ester or amide. For example, an alkylene, alkenylene, or alkynylene may be substituted with one or more (e.g., 1, 2, 3, 4, 5, or 6 independently selected substituents) of halogen, -COR', -CO 2 H, -CO 2 R',

-CN, -OH, -OR', -OCOR', -OCO 2 R', -NH 2 , -NHR', -N(R') 2 , -SR' or -SO 2 R', wherein each instance of R* independently is C 1 -C 20 aliphatic (e.g., C 1 -C 20 alkyl, C 1 -C 15 alkyl, C 1 -C 10 alkyl, or C 1 -C 3 alkyl). 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.

[079] 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. "C z -C3o alkenyl" refers to an alkenyl group having 2-30 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', -CO 2 H, -CO 2 R', -CN, -OH, -OR ' , -OCOR', -OCO 2 R', -NH 2 , -NHR', -N(R') 2 , -SR' or-SO 2 R', wherein each instance of R' independently is C 1 -C 20 aliphatic ( e.g ., C 1 -C 20 alkyl, C 1 -C 15 alkyl, C 1 -C 10 alkyl, or C 1 -C 3 alkyl). 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 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.

[080] 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 30 alkynyl" refers to an alkynyl group having 2-30 carbons. Examples of an alkynyl group include prop-2-ynyl, but-2-ynyl, but-3-ynyl, pent-2-ynyl, 3-methylpent-4-ynyl, hex-2-ynyl, hex-5-ynyl, etc. In embodiments, an alkynyl comprises one carbon-carbon triple bond. An alkynyl group may be unsubstituted or substituted with one or more substituent groups as described herein. For example, an alkynyl group may be substituted with one or more (e.g., 1, 2, 3, 4, 5, or 6 independently selected substituents) of halogen, -COR', -CO2H, -CO 2 R', - CN, -OH, -OR', -OCOR',

-OCO2R', -NH2, -NHR', -N(R') 2 , -SR' or-SO 2 R', wherein each instance of R' independently is C 1 -C 20 aliphatic (e.g., C 1 -C 20 alkyl, C 1 -C 15 alkyl, C 1 -C 10 alkyl, or C1-C3 alkyl). In embodiments, R' independently is an unsubstituted alkyl (e.g., unsubstituted C 1 -C 20 alkyl, C1-C15 alkyl, C 1 -C 10 alkyl, or C1-C3 alkyl). In embodiments, R' Independently Is unsubstituted C1-C3 alkyl. In embodiments, the alkynyl is unsubstituted. In embodiments, the alkynyl is substituted (e.g., with 1, 2, 3, 4, 5, or 6 substituent groups as described herein).

[081] Aryl: The term "aryl" used alone or as part of a larger moiety as in "aralkyl," refers to a monocyclic, bicyclic, or tricyclic carbocyclic ring system having a total of six to fourteen ring members, wherein said ring system has a single point of attachment to the rest of the molecule, at least one ring In the system is aromatic and wherein each ring In the system contains 4 to 7 ring members. In embodiments, an aryl group has 6 ring carbon atoms ("C 6 aryl," e.g., phenyl).

In some embodiments, an aryl group has 10 ring carbon atoms (“C 10 aryl," e.g., naphthyl such as 1-naphthyl and 2-naphthyl). In some embodiments, an aryl group has 14 ring carbon atoms ("C14 aryl," e.g., anthracyl). "Aryl" also includes ring systems wherein the aryl ring, as defined above, is fused with one or more carbocyclyl or heterocyclyl groups wherein the radical or point of attachment Is on the aryl ring, and in such instances, the number of carbon atoms continue to designate the number of carbon atoms in the aryl ring system. Exemplary aryls include phenyl, naphthyl, and anthracene.

[082] Arylene: The term "arylene" as used herein refers to an aryl group that is divalent (that is, having two points of attachment to the molecule). Exemplary arylenes include phenylene (e.g., unsubstituted phenylene or substituted phenylene).

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

[084] Heteroallphatlc: The term "heteroaliphatlc," as used herein, means aliphatic groups (e.g., as described herein) wherein one or two carbon atoms are independently replaced by one or more heteroatoms. Heteroaliphatlc groups may be substituted or unsubstituted, branched or unbranched, cyclic or acyclic, and include "heteroalkyl," "heteroalkylene," and

"heterocycloalkyl" groups (e.g., as described herein).

[085] 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, phosphoramldates, 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.

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

[087] Heteroaryl: The term "heteroaryl," as used herein, is fully unsaturated heteroatom- containing ring wherein at least one ring atom is a heteroatom such as, but not limited to, nitrogen and oxygen.

[088] Heterocycloalkyl/Heterocyclyl: The terms "heterocycloalkyl" and "heterocyclyl" as used herein, is a non-aromatic ring wherein at least one atom is a heteroatom such as, but not limited to, nitrogen, oxygen, sulfur, or phosphorus, and the remaining atoms are carbon. Examples of heterocycloalkyl groups include, but are not limited to, pyrrolidinyl, tetrahydrofuranyl, dihydrofuranyl, tetrahydrothienyl, tetrahydropyranyl, dihydropyranyl, tetrahydrothiopyranyl, piperidino, morpholino, thiomorpholino, thioxanyl, piperazinyl, azetidinyl, oxetanyl, thietanyl, homopiperidinyl, oxepanyl, thlepanyl, oxazepinyl, dlazepinyl, thiazepinyl, 1, 2,3,6- tetrahydropyridinyl, 2-pyrrolinyl, 3-pyrrolinyl, Indolinyl, 2H-pyranyl, 4H-pyranyl, dloxanyl, 1,3- dioxolanyl, pyrazolinyl, dithianyl, dithiolanyl, dihydropyranyl, dihydrothienyl, dihydrofuranyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, 3-azabicyclo[3.1.0]hexanyl, 3- azablcyclo[4.1.0]heptanyl, 3H-lndolyl and qulnollzinyl. The heterocycloalkyl group can be substituted or unsubstituted.

Compounds of the Invention

[089] Provided herein are compounds which are cationic lipids. Liposomal-based vehicles are considered an attractive carrier for therapeutic agents and remain subject to continued development efforts. While liposomal-based vehicles that comprise certain lipid components have shown promising results with regard 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.

[090] 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.

[091] Described herein a novel class of di-thioester cationic lipid compounds for improved in vivo delivery of therapeutic agents, such as nucleic acids. In particular, a di-thioester cationic lipid described herein may be used as a cationic lipid, optionally with other 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.

[092] 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 (A)

[093] Cationic lipids of the present invention include compounds having a structure according to Formula (A),

or a pharmaceutically acceptable salt thereof, wherein

Substructure A is 3-9-membered heterocyclyl, C 3 -C 6 cycloalkyl, 5-10-membered heteroaryl, or C 6 -C 10 aryl;

each R 1 is independently C 6 -C 30 aliphatic;

each m is independently an integer of 0-6; and

each n is independently an integer of 1-12.

[094] In embodiments, Substructure A is 5-6-membered heterocyclyl, C 5 -C 6 cycloalkyl, or C 6 -C 10 aryl. In embodiments, Substructure A is 3-9-membered heterocyclyl. In embodiments.

Substructure A is C 3 -C 6 cycloalkyl. In embodiments, Substructure A is 5-10-membered heteroaryl. In embodiments, Substructure A is C 6 -C 10 aryl.

[095] In embodiments, m is 0, 1, or 2. In embodiments, m is 0. In embodiments, m is 1. In

embodiments, m is 2.

[096] In embodiments, n is 1, 2, or 3. In embodiments, n is 1. In embodiments, n is 2. In

embodiments, n is 3.

[097] In embodiments, each R 1 is C 6 -C 30 alkyl. In embodiments, each R 1 is unsubstituted C 6 -C 30 alkyl. In embodiments, each R 1 is substituted C 6 -C 30 alkyl. In embodiments, each R 1 is C 6 -C 24 alkyl. In embodiments, each R 1 is unsubstituted C 6 -C 24 alkyl. In embodiments, each R 1 is substituted C 6 -C 24 alkyl. In embodiments, an alkyl is a branched alkyl. In embodiments, an alkyl is a linear alkyl.

[098] In embodiments, each R 1 is C 6 -C 30 alkenyl. In embodiments, each R 1 is unsubstituted C 6 -C 30 alkenyl. In embodiments, each R 1 is substituted C 6 -C 30 alkenyl. In embodiments, each R 1 is C 6 -C 24 alkenyl. In embodiments, each R 1 is unsubstituted C 6 -C 24 alkenyl. In embodiments, each R 1 is substituted C 6 -C 24 alkenyl. In embodiments, an alkenyl is a branched alkenyl. In embodiments, an alkenyl is a linear alkenyl. In embodiments, an alkenyl is a mono-alkenyl. In embodiments, an alkenyl is a dienyl. In embodiments, an alkenyl Is a trienyl.

[099] In embodiments, each R 1 is C 6 -C 30 alkynyl. In embodiments, each R 1 is unsubstituted C 6 -C 30 alkynyl. In embodiments, each R 1 is substituted C 6 -C 30 alkynyl. In embodiments, each R 1 is C 6 -C 24 alkynyl. In embodiments, each R 1 is unsubstituted C 6 -C 24 alkynyl. In embodiments, each R 1 is substituted C 6 -C 24 alkynyl. In embodiments, an alkenyl is a branched alkynyl. In embodiments, an alkenyl is a linear alkynyl.

[0100] In embodiments, each R 1 is C 6 -C 24 alkyl.

[0101] In embodiments, each R 1 is unsubstituted C 6 -C 24 alkyl. In embodiments, each R 1 is C 8 H 17 , C 10 H 21 , C 12 H 25 , CnH 29 , or C 16 H 33 .

[0102] In embodiments, each R 1 is C 6 -C 24 alkenyl. In embodiments, a C 6 -C 24 alkenyl has one, two, or three carbon-carbon double bonds.

[0103] In embodiments, each R 1 is unsubstituted C 6 -C 24 alkenyl. In embodiments, each R 1 is - C 6 H 12 CH=CHCH 2 CH=CHC 5 H 11 .

[0104] In embodiments, a cationic lipid of Formula (A) has the following structure:

or a pharmaceutically acceptable salt thereof.

[0105] In embodiments, a cationic lipid of Formula (A) has the following structure:

or a pharmaceutically acceptable salt thereof. [0106] In embodiments, a cationic lipid of Formula (A) has the following structure:

or a pharmaceutically acceptable salt thereof.

[0107] In embodiments, a cationic lipid of Formula (A) has the following structure:

or a pharmaceutically acceptable salt thereof.

Compounds of Formula (B)

[0108] In another aspect, a cationic lipid has the following structure:

or a pharmaceutically acceptable salt thereof, wherein

L 1 is C 2 -C 14 alkenyl;

each R 1 is independently C 6 -C 30 aliphatic; and

each n is independently an integer of 1-12.

[0109] In embodiments, the C 2 -C 12 alkenyl comprises 1, 2, or 3 carbon-carbon double bonds.

[0110] In embodiments, n is 1, 2, or 3. In embodiments, n is 1. In embodiments, n is 2. In

embodiments, n is 3.

[0111] In embodiments, each R 1 is C 6 -C 30 alkyl. In embodiments, each R 1 is unsubstituted C 6 -C 30 alkyl. In embodiments, each R 1 is substituted C 6 -C 30 alkyl. In embodiments, each R 1 is C 6 -C 24 alkyl. In embodiments, each R 1 is unsubstituted C 6 -C 24 alkyl. In embodiments, each R 1 is substituted C 6 -C 24 alkyl. In embodiments, an alkyl is a branched alkyl. In embodiments, an alkyl Is a linear alkyl. [0112] In embodiments, each R 1 is C 6 -C 30 alkenyl. In embodiments, each R 1 is unsubstituted C 6 -C 30 alkenyl. In embodiments, each R 1 is substituted C 6 -C 30 alkenyl. In embodiments, each R 1 is C 6 -C 24 alkenyl. In embodiments, each R 1 is unsubstituted C 6 -C 24 alkenyl. In embodiments, each R 1 is substituted C 6 -C 24 alkenyl. In embodiments, an alkenyl is a branched alkenyl. In embodiments, an alkenyl is a linear alkenyl. In embodiments, an alkenyl is a mono-alkenyl. In embodiments, an alkenyl is a dienyl. In embodiments, an alkenyl is a trienyl.

[0113] In embodiments, each R 1 Is C 6 -C 30 alkynyl. In embodiments, each R 1 is unsubstituted C 6 -C 30 alkynyl. In embodiments, each R 1 is substituted C 6 -C 30 alkynyl. In embodiments, each R 1 is C 6 -C 24 alkynyl. In embodiments, each R 1 is unsubstituted C 6 -C 24 alkynyl. In embodiments, each R 1 is substituted C 6 -C 24 alkynyl. In embodiments, an alkenyl Is a branched alkynyl. In embodiments, an alkenyl is a linear alkynyl.

[0114] In embodiments, each R 1 Is C 6 -C 24 alkyl.

[0115] In embodiments, each R 1 is unsubstituted C 6 -C 24 alkyl. In embodiments, each R 1 Is C 8 H 17 , C 10 H 21 , C 12 H 25 , C 14 H 29 , or. C 16 H 33

[0116] In embodiments, each R 1 is C 6 -C 24 alkenyl. In embodiments, a C 6 -C 24 alkenyl has one, two, or three carbon-carbon double bonds.

[0117] In embodiments, each R 1 Is unsubstituted C 6 -C 24 alkenyl. In embodiments, each R 1 Is - C 6 H 12 CH=CHCH 2 CH=CHC 5 H 11 .

[0118] In embodiments, a cationic lipid of Formula (B) has the following structure:

or a pharmaceutically acceptable salt thereof, wherein

each R 2 and R 3 Is independently hydrogen or C 1 -C 6 alkyl.

[0119] In embodiments, R 2 and R 3 are both hydrogen.

[0120] In embodiments, wherein one of R 2 and R 3 is hydrogen, and the other is unsubstituted C 1 -C 6 alkyl. In embodiments, an unsubstituted C 1 -C 6 alkyl is methyl or ethyl.

[0121] In embodiments, R 2 and R 3 are cls to each other.

[0122] In embodiments, R 2 and R 3 are trans to each other. [0123] In embodiments, a cationic lipid of Formula (B) has the following structure:

or a pharmaceutically acceptable salt thereof, wherein

a is an integer of 0-6;

b is an Integer of 0-2; and

c is an integer of 0-3;

and wherein

at least one of a and b is not zero, and

when a or b is 1, then c is not zero.

[0124] In embodiments, one and only one of a and b is not zero.

[0125] In embodiments a is 0. In embodiments, a is 1, 2, 3, 4, 5, or 6.

[0126] In embodiments, b is 0. In embodiments, b is 1 or 2.

[0127] In embodiments, c is 0. In embodiments, c is 1, 2, or 3.

[0128] In embodiments, a is 2, and each of b and c is zero.

[0129] In embodiments, a is 1, b is 0, and c is 1.

Compounds of Formula (C)

[0130] In another aspect, a cationic lipid has the following structure:

or a pharmaceutically acceptable salt thereof, wherein

L 1 is C 1 -C 12 aliphatic or C 1 -C 12 heteroaliphatic comprising an oxo (=O), -OH, -COR 4 , or -COzR 4 ; each R 1 is independently C 6 -C 30 aliphatic;

each R 4 is Independently hydrogen or C 1 -C 6 alkyl; and

each n is independently an integer of 1-12.

[0131] In embodiments, L 1 is C 1 -C 12 aliphatic comprising an oxo (=O), -OH, or -COCH 3 group.

[0132] In embodiments, L 1 is C 1 -C 12 heteroaliphatic comprising an oxo (=O), -OH, or -COCH 3 group.

In embodiments, L 1 is C 1 -C 12 heteroaliphatic comprising one or two ethylene glycol moieties. [0133] In embodiments, n is 1, 2, or 3. In embodiments, n is 1. In embodiments, n is 2. In embodiments, n is 3.

[0134] In embodiments, each R 1 is C 6 -C 30 alkyl. In embodiments, each R 1 is unsubstituted C 6 -C 30 alkyl. In embodiments, each R 1 is substituted C 6 -C 30 alkyl. In embodiments, each R 1 is C 6 -C 24 alkyl. In embodiments, each R 1 is unsubstituted C 6 -C 24 alkyl. In embodiments, each R 1 is substituted C 6 -C 24 alkyl. In embodiments, an alkyl is a branched alkyl. In embodiments, an alkyl is a linear alkyl.

[0135] In embodiments, each R 1 is C 6 -C 30 alkenyl. In embodiments, each R 1 is unsubstituted C 6 -C 30 alkenyl. In embodiments, each R 1 is substituted C 6 -C 30 alkenyl. In embodiments, each R 1 is C 6 -C 24 alkenyl. In embodiments, each R 1 is unsubstituted C 6 -C 24 alkenyl. In embodiments, each R 1 is substituted C 6 -C 24 alkenyl. In embodiments, an alkenyl is a branched alkenyl. In embodiments, an alkenyl is a linear alkenyl. In embodiments, an alkenyl is a mono-alkenyl. In embodiments, an alkenyl is a dienyl. In embodiments, an alkenyl is a trienyl.

[0136] In embodiments, each R 1 is C 6 -C 30 alkynyl. In embodiments, each R 1 is unsubstituted C 6 -C 30 alkynyl. In embodiments, each R 1 is substituted C 6 -C 30 alkynyl. In embodiments, each R 1 is C 6 -C 24 alkynyl. In embodiments, each R 1 is unsubstituted C 6 -C 24 alkynyl. In embodiments, each R 1 is substituted C 6 -C 24 alkynyl. In embodiments, an alkenyl is a branched alkynyl. In embodiments, an alkenyl is a linear alkynyl.

[0137] In embodiments, each R 1 is C 6 -C 24 alkyl.

[0138] In embodiments, each R 1 is unsubstituted C 6 -C 24 alkyl. In embodiments, each R 1 is C 8 H 17 , C 10 H 21 , C 12 H 25 , C 14 H 29 , or. C 16 H 33

[0139] In embodiments, each R 1 is C 6 -C 24 alkenyl. In embodiments, a C 6 -C 24 alkenyl has one, two, or three carbon-carbon double bonds.

[0140] In embodiments, each R 1 is unsubstituted C 6 -C 24 alkenyl. In embodiments, each R 1 is - C 6 H 12 CH=CHCH 2 CH=CHC 5 H 11 .

[0141] In embodiments, a cationic lipid of Formula (C) has the following structure:

or a pharmaceutically acceptable salt thereof, wherein

R 5 Is hydrogen;

R 6 is -OH or -COCH 3 ; or R 5 and R 6 combine to form an oxo substituent.

[0142] In embodiments, R 5 is hydrogen and R 6 is -OH.

[0143] In embodiments, R 5 is hydrogen and R 6 is -COCH 3 .

[0144] In embodiments, R 5 and R 6 combine to form an oxo substituent.

[0145] In embodiments, a cationic lipid of Formula (C) has the following structure:

or a pharmaceutically acceptable salt thereof, wherein

a is an integer of 2-6;

b is 0 or 1.

[0146] In embodiments, a cationic lipid of Formula (C) has the following structure:

or a pharmaceutically acceptable salt thereof.

[0147] In embodiments, a cationic lipid of Formula (C) has the following structure:

or a pharmaceutically acceptable salt thereof. Comoounds of Formula (D)

[0148] In another aspect, a cationic lipid has the following structure:

or a pharmaceutically acceptable salt thereof, wherein

each R 1 is independently C 6 -C 30 aliphatic; and

each n is independently an integer of 1-12.

[0149] In embodiments, n is 1, 2, or 3. In embodiments, n is 1. In embodiments, n is 2. In

embodiments, n is 3.

[0150] In embodiments, each R 1 is C 6 -C 30 alkyl. In embodiments, each R 1 is unsubstituted C 6 -C 30 alkyl. In embodiments, each R 1 is substituted C 6 -C 30 alkyl. In embodiments, each R 1 is C 6 -C 24 alkyl. In embodiments, each R 1 is unsubstituted C 6 -C 24 alkyl. In embodiments, each R 1 is substituted C 6 -C 24 alkyl. In embodiments, an alkyl is a branched alkyl. In embodiments, an alkyl is a linear alkyl.

[0151] In embodiments, each R 1 Is C 6 -C 30 alkenyl. In embodiments, each R 1 Is unsubstituted C 6 -C 30 alkenyl. In embodiments, each R 1 is substituted C 6 -C 30 alkenyl. In embodiments, each R 1 is C 6 -C 24 alkenyl. In embodiments, each R 1 is unsubstituted C 6 -C 24 alkenyl. In embodiments, each R 1 is substituted C 6 -C 24 alkenyl. In embodiments, an alkenyl is a branched alkenyl. In embodiments, an alkenyl Is a linear alkenyl. In embodiments, an alkenyl is a mono-alkenyl. In embodiments, an alkenyl is a dienyl. In embodiments, an alkenyl Is a trienyl.

[0152] In embodiments, each R 1 is C 6 -C 30 alkynyl. In embodiments, each R 1 is unsubstituted C 6 -C 30 alkynyl. In embodiments, each R 1 is substituted C 6 -C 30 alkynyl. In embodiments, each R 1 is C 6 -C 24 alkynyl. In embodiments, each R 1 is unsubstituted C 6 -C 24 alkynyl. In embodiments, each R 1 is substituted C 6 -C 24 alkynyl. In embodiments, an alkenyl Is a branched alkynyl. In embodiments, an alkenyl is a linear alkynyl.

[0153] In embodiments, each R 1 is C 6 -C 24 alkyl.

[0154] In embodiments, each R 1 is unsubstituted C 6 -C 24 alkyl. In embodiments, each R 1 is C 8 H 17 , C 10 H 21 , C 12 H 25 , CnH 29 , or C 16 H 33 .

[0155] In embodiments, each R 1 is C 6 -C 24 alkenyl. In embodiments, a C 6 -Cz* alkenyl has one, two, or three carbon-carbon double bonds.

[0156] In embodiments, each R 1 is unsubstituted C 6 -C 24 alkenyl. In embodiments, each R 1 is - C 6 H 12 CH=CHCH 2 CH=CHC 5 H 11 . Compounds of Formula (E)

[0157] In another aspect, a cationic lipid has the following structure:

or a pharmaceutically acceptable salt thereof, wherein

each R 1 is independently C 6 -C 30 aliphatic; and

each n is independently an integer of 1-12.

[0158] In embodiments, n is 1, 2, or 3. In embodiments, n is 1. In embodiments, n is 2. In

embodiments, n is 3.

[0159] In embodiments, each R 1 is C 6 -C 30 alkyl. In embodiments, each R 1 is unsubstituted C 6 -C 30 alkyl. In embodiments, each R 1 is substituted C 6 -C 30 alkyl. In embodiments, each R 1 is C 6 -C 24 alkyl. In embodiments, each R 1 is unsubstituted C 6 -C 24 alkyl. In embodiments, each R 1 is substituted C 6 -C 24 alkyl. In embodiments, an alkyl is a branched alkyl. In embodiments, an alkyl is a linear alkyl.

[0160] In embodiments, each R 1 is C 6 -C 30 alkenyl. In embodiments, each R 1 is unsubstituted C 6 -C 30 alkenyl. In embodiments, each R 1 is substituted C 6 -C 30 alkenyl. In embodiments, each R 1 is C 6 -C 24 alkenyl. In embodiments, each R 1 is unsubstituted C 6 -C 24 alkenyl. In embodiments, each R 1 is substituted C 6 -C 24 alkenyl. In embodiments, an alkenyl is a branched alkenyl. In embodiments, an alkenyl is a linear alkenyl. In embodiments, an alkenyl is a mono-alkenyl. In embodiments, an alkenyl is a dienyl. In embodiments, an alkenyl is a trienyl.

[0161] In embodiments, each R 1 is C 6 -C 30 alkynyl. In embodiments, each R 1 is unsubstituted C 6 -C 30 alkynyl. In embodiments, each R 1 is substituted C 6 -C 30 alkynyl. In embodiments, each R 1 is C 6 -C 24 alkynyl. In embodiments, each R 1 is unsubstituted C 6 -C 24 alkynyl. In embodiments, each R 1 is substituted C 6 -C 24 alkynyl. In embodiments, an alkenyl is a branched alkynyl. In embodiments, an alkenyl is a linear alkynyl.

[0162] In embodiments, each R 1 is C 6 -C 24 alkyl.

[0163] In embodiments, each R 1 is unsubstituted C 6 -C 24 alkyl. In embodiments, each R 1 is C 8 H 17 , C 10 H 21 , C 12 H 25 , CnH 29 , or C 16 H 33 . [0164] In embodiments, each R 1 is C 6 -C 24 alkenyl. In embodiments, a C 6 -C 24 alkenyl has one, two, or three carbon-carbon double bonds.

[0165] In embodiments, each R 1 is unsubstituted C 6 -C 24 alkenyl. In embodiments, each R 1 is - C 6 H 12 CH=CHCH 2 CH=CHC 5 H 11 .

Exemplary Compounds

[0166] In embodiments, a cationic lipid is any one of Compounds 1-378.

[0167] In embodiments, a cationic lipid is Compound 1. In embodiments, a cationic lipid is

Compound 2. In embodiments, a cationic lipid is Compound 3. In embodiments, a cationic lipid is Compound 4. In embodiments, a cationic lipid is Compound 5. In embodiments, a cationic lipid is Compound 6. In embodiments, a cationic lipid is Compound 7. In embodiments, a cationic lipid is Compound 8. In embodiments, a cationic lipid is Compound 9. In embodiments, a cationic lipid is Compound 10. In embodiments, a cationic lipid is Compound 11. In embodiments, a cationic lipid is Compound 12. In embodiments, a cationic lipid is Compound 13. In embodiments, a cationic lipid is Compound 14. In embodiments, a cationic lipid is

Compound 15. In embodiments, a cationic lipid is Compound 16. In embodiments, a cationic lipid is Compound 17. In embodiments, a cationic lipid is Compound 18. In embodiments, a cationic lipid is Compound 19. In embodiments, a cationic lipid is Compound 20. In

embodiments, a cationic lipid is Compound 21. In embodiments, a cationic lipid is Compound 22. In embodiments, a cationic lipid is Compound 23. In embodiments, a cationic lipid is

Compound 24. In embodiments, a cationic lipid is Compound 25. In embodiments, a cationic lipid is Compound 26. In embodiments, a cationic lipid is Compound 27. In embodiments, a cationic lipid is Compound 28. In embodiments, a cationic lipid is Compound 29. In

embodiments, a cationic lipid is Compound 30.

[0168] In embodiments, a cationic lipid is Compound 31. In embodiments, a cationic lipid is

Compound 32. In embodiments, a cationic lipid is Compound 33. In embodiments, a cationic lipid is Compound 34. In embodiments, a cationic lipid is Compound 35. In embodiments, a cationic lipid is Compound 36. In embodiments, a cationic lipid is Compound 37. In

embodiments, a cationic lipid is Compound 38. In embodiments, a cationic lipid is Compound 39. In embodiments, a cationic lipid is Compound 40. In embodiments, a cationic lipid is

Compound 41. In embodiments, a cationic lipid is Compound 42. In embodiments, a cationic lipid is Compound 43. In embodiments, a cationic lipid is Compound 44. In embodiments, a cationic lipid is Compound 45. In embodiments, a cationic lipid is Compound 46. In

embodiments, a cationic lipid is Compound 47. In embodiments, a cationic lipid is Compound 48. In embodiments, a cationic lipid is Compound 49. In embodiments, a cationic lipid is

Compound 50. In embodiments, a cationic lipid is Compound 51. In embodiments, a cationic lipid is Compound 52. In embodiments, a cationic lipid is Compound 53. In embodiments, a cationic lipid is Compound 54. In embodiments, a cationic lipid is Compound 55. In

embodiments, a cationic lipid is Compound 56. In embodiments, a cationic lipid is Compound 57. In embodiments, a cationic lipid is Compound 58. In embodiments, a cationic lipid is

Compound 59. In embodiments, a cationic lipid is Compound 60.

[0169] In embodiments, a cationic lipid is Compound 61. In embodiments, a cationic lipid is

Compound 62. In embodiments, a cationic lipid is Compound 63. In embodiments, a cationic lipid is Compound 64. In embodiments, a cationic lipid is Compound 65. In embodiments, a cationic lipid is Compound 66. In embodiments, a cationic lipid is Compound 67. In

embodiments, a cationic lipid is Compound 68. In embodiments, a cationic lipid is Compound 69. In embodiments, a cationic lipid is Compound 70. In embodiments, a cationic lipid is

Compound 71. In embodiments, a cationic lipid is Compound 72. In embodiments, a cationic lipid is Compound 73. In embodiments, a cationic lipid is Compound 74. In embodiments, a cationic lipid is Compound 75. In embodiments, a cationic lipid is Compound 76. In

embodiments, a cationic lipid is Compound 77. In embodiments, a cationic lipid is Compound 78. In embodiments, a cationic lipid is Compound 79. In embodiments, a cationic lipid is

Compound 80. In embodiments, a cationic lipid is Compound 81. In embodiments, a cationic lipid is Compound 82. In embodiments, a cationic lipid is Compound 83. In embodiments, a cationic lipid is Compound 84. In embodiments, a cationic lipid is Compound 85. In

embodiments, a cationic lipid is Compound 86. In embodiments, a cationic lipid is Compound 87. In embodiments, a cationic lipid is Compound 88. In embodiments, a cationic lipid is

Compound 89. In embodiments, a cationic lipid is Compound 90.

[0170] In embodiments, a cationic lipid is Compound 91. In embodiments, a cationic lipid is

Compound 92. In embodiments, a cationic lipid is Compound 93. In embodiments, a cationic lipid is Compound 94. In embodiments, a cationic lipid is Compound 95. In embodiments, a cationic lipid is Compound 96. In embodiments, a cationic lipid is Compound 97. In embodiments, a cationic lipid is Compound 98. In embodiments, a cationic lipid is Compound 99. In embodiments, a cationic lipid is Compound 100. In embodiments, a cationic lipid is

Compound 101. In embodiments, a cationic lipid is Compound 102. In embodiments, a cationic lipid is Compound 103. In embodiments, a cationic lipid is Compound 104. In embodiments, a cationic lipid is Compound 105. In embodiments, a cationic lipid is Compound 106. In embodiments, a cationic lipid is Compound 107. In embodiments, a cationic lipid is

Compound 108. In embodiments, a cationic lipid is Compound 109. In embodiments, a cationic lipid is Compound 110. In embodiments, a cationic lipid is Compound 111. In embodiments, a cationic lipid is Compound 112. In embodiments, a cationic lipid is Compound 113. In embodiments, a cationic lipid is Compound 114. In embodiments, a cationic lipid is

Compound 115. In embodiments, a cationic lipid is Compound 116. In embodiments, a cationic lipid is Compound 117. In embodiments, a cationic lipid is Compound 118. In embodiments, a cationic lipid is Compound 119. In embodiments, a cationic lipid is Compound 120.

[0171] In embodiments, a cationic lipid is Compound 121. In embodiments, a cationic lipid is

Compound 122. In embodiments, a cationic lipid is Compound 123. In embodiments, a cationic lipid is Compound 124. In embodiments, a cationic lipid is Compound 125. In embodiments, a cationic lipid is Compound 126. In embodiments, a cationic lipid is Compound 127. In embodiments, a cationic lipid is Compound 128. In embodiments, a cationic lipid is

Compound 129. In embodiments, a cationic lipid is Compound 130. In embodiments, a cationic lipid is Compound 131. In embodiments, a cationic lipid is Compound 132. In embodiments, a cationic lipid is Compound 133. In embodiments, a cationic lipid is Compound 134. In embodiments, a cationic lipid is Compound 135. In embodiments, a cationic lipid is

Compound 136. In embodiments, a cationic lipid is Compound 137. In embodiments, a cationic lipid is Compound 138. In embodiments, a cationic lipid is Compound 139. In embodiments, a cationic lipid is Compound 140. In embodiments, a cationic lipid is Compound 141. In embodiments, a cationic lipid is Compound 142. In embodiments, a cationic lipid is

Compound 143. In embodiments, a cationic lipid is Compound 144. In embodiments, a cationic lipid is Compound 145. In embodiments, a cationic lipid is Compound 146. In embodiments, a cationic lipid is Compound 147. In embodiments, a cationic lipid is Compound 148. In embodiments, a cationic lipid is Compound 149. In embodiments, a cationic lipid is

Compound 150.

[0172] In embodiments, a cationic lipid is Compound 151. In embodiments, a cationic lipid is

Compound 152. In embodiments, a cationic lipid is Compound 153. In embodiments, a cationic lipid is Compound 154. In embodiments, a cationic lipid is Compound 155. In embodiments, a cationic lipid is Compound 156. In embodiments, a cationic lipid is Compound 157. In embodiments, a cationic lipid is Compound 158. In embodiments, a cationic lipid is

Compound 159. In embodiments, a cationic lipid is Compound 160. In embodiments, a cationic lipid is Compound 161. In embodiments, a cationic lipid is Compound 162. In embodiments, a cationic lipid is Compound 163. In embodiments, a cationic lipid is Compound 164. In embodiments, a cationic lipid is Compound 165. In embodiments, a cationic lipid is

Compound 166. In embodiments, a cationic lipid is Compound 167. In embodiments, a cationic lipid is Compound 168. In embodiments, a cationic lipid is Compound 169. In embodiments, a cationic lipid is Compound 170. In embodiments, a cationic lipid is Compound 171. In embodiments, a cationic lipid is Compound 172. In embodiments, a cationic lipid is

Compound 173. In embodiments, a cationic lipid is Compound 174. In embodiments, a cationic lipid is Compound 175. In embodiments, a cationic lipid is Compound 176. In embodiments, a cationic lipid is Compound 177. In embodiments, a cationic lipid is Compound 178. In embodiments, a cationic lipid is Compound 179. In embodiments, a cationic lipid is

Compound 180.

[0173] In embodiments, a cationic lipid is Compound 181. In embodiments, a cationic lipid is Compound 182. In embodiments, a cationic lipid is Compound 183. In embodiments, a cationic lipid is Compound 184. In embodiments, a cationic lipid is Compound 185. In embodiments, a cationic lipid is Compound 186. In embodiments, a cationic lipid is Compound 187. In embodiments, a cationic lipid is Compound 188. In embodiments, a cationic lipid is

Compound 189. In embodiments, a cationic lipid is Compound 190. In embodiments, a cationic lipid is Compound 191. In embodiments, a cationic lipid is Compound 192. In embodiments, a cationic lipid is Compound 193. In embodiments, a cationic lipid is Compound 194. In embodiments, a cationic lipid is Compound 195. In embodiments, a cationic lipid is

Compound 196. In embodiments, a cationic lipid is Compound 197. In embodiments, a cationic lipid is Compound 198. In embodiments, a cationic lipid is Compound 199. In embodiments, a cationic lipid is Compound 200. In embodiments, a cationic lipid is Compound 201. In embodiments, a cationic lipid is Compound 202. In embodiments, a cationic lipid is

Compound 203. In embodiments, a cationic lipid is Compound 204. In embodiments, a cationic lipid is Compound 205. In embodiments, a cationic lipid is Compound 206. In embodiments, a cationic lipid is Compound 207. In embodiments, a cationic lipid is Compound 208. In embodiments, a cationic lipid is Compound 209. In embodiments, a cationic lipid is

Compound 210. [0174] In embodiments, a cationic lipid is Compound 211. In embodiments, a cationic lipid is Compound 212. In embodiments, a cationic lipid is Compound 213. In embodiments, a cationic lipid is Compound 214. In embodiments, a cationic lipid is Compound 215. In embodiments, a cationic lipid is Compound 216. In embodiments, a cationic lipid is Compound 217. In embodiments, a cationic lipid is Compound 218. In embodiments, a cationic lipid is

Compound 219. In embodiments, a cationic lipid is Compound 220. In embodiments, a cationic lipid is Compound 221. In embodiments, a cationic lipid is Compound 222. In embodiments, a cationic lipid is Compound 223. In embodiments, a cationic lipid is Compound 224. In embodiments, a cationic lipid is Compound 225. In embodiments, a cationic lipid is

Compound 226. In embodiments, a cationic lipid is Compound 227. In embodiments, a cationic lipid is Compound 228. In embodiments, a cationic lipid is Compound 229. In embodiments, a cationic lipid is Compound 230. In embodiments, a cationic lipid is Compound 231. In embodiments, a cationic lipid is Compound 232. In embodiments, a cationic lipid is

Compound 233. In embodiments, a cationic lipid is Compound 234. In embodiments, a cationic lipid is Compound 235. In embodiments, a cationic lipid is Compound 236. In embodiments, a cationic lipid is Compound 237. In embodiments, a cationic lipid is Compound 238. In embodiments, a cationic lipid is Compound 239. In embodiments, a cationic lipid is

Compound 240.

[0175] In embodiments, a cationic lipid is Compound 241. In embodiments, a cationic lipid is Compound 242. In embodiments, a cationic lipid is Compound 243. In embodiments, a cationic lipid is Compound 244. In embodiments, a cationic lipid is Compound 245. In embodiments, a cationic lipid is Compound 246. In embodiments, a cationic lipid is Compound 247. In embodiments, a cationic lipid is Compound 248. In embodiments, a cationic lipid is

Compound 249. In embodiments, a cationic lipid is Compound 250. In embodiments, a cationic lipid is Compound 251. In embodiments, a cationic lipid is Compound 252. In embodiments, a cationic lipid is Compound 253. In embodiments, a cationic lipid is Compound 254. In embodiments, a cationic lipid is Compound 255. In embodiments, a cationic lipid is

Compound 256. In embodiments, a cationic lipid is Compound 257. In embodiments, a cationic lipid is Compound 258. In embodiments, a cationic lipid is Compound 259. In embodiments, a cationic lipid is Compound 260. In embodiments, a cationic lipid is Compound 261. In embodiments, a cationic lipid is Compound 262. In embodiments, a cationic lipid is

Compound 263. In embodiments, a cationic lipid is Compound 264. In embodiments, a cationic lipid is Compound 265. In embodiments, a cationic lipid is Compound 266. In embodiments, a cationic lipid is Compound 267. In embodiments, a cationic lipid is Compound 268. In embodiments, a cationic lipid is Compound 269. In embodiments, a cationic lipid is

Compound 270.

[0176] In embodiments, a cationic lipid is Compound 271. In embodiments, a cationic lipid Is Compound 272. In embodiments, a cationic lipid is Compound 273. In embodiments, a cationic lipid is Compound 274. In embodiments, a cationic lipid is Compound 275. In embodiments, a cationic lipid is Compound 276. In embodiments, a cationic lipid is Compound 277. In embodiments, a cationic lipid is Compound 278. In embodiments, a cationic lipid is

Compound 279. In embodiments, a cationic lipid is Compound 280. In embodiments, a cationic lipid is Compound 281. In embodiments, a cationic lipid is Compound 282. In embodiments, a cationic lipid is Compound 283. In embodiments, a cationic lipid is Compound 284. In embodiments, a cationic lipid is Compound 285. In embodiments, a cationic lipid is

Compound 286. In embodiments, a cationic lipid is Compound 287. In embodiments, a cationic lipid is Compound 288. In embodiments, a cationic lipid is Compound 289. In embodiments, a cationic lipid is Compound 290. In embodiments, a cationic lipid is Compound 291. In embodiments, a cationic lipid is Compound 292. In embodiments, a cationic lipid is

Compound 293. In embodiments, a cationic lipid is Compound 294. In embodiments, a cationic lipid is Compound 295. In embodiments, a cationic lipid is Compound 296. In embodiments, a cationic lipid is Compound 297. In embodiments, a cationic lipid is Compound 298. In embodiments, a cationic lipid is Compound 299. In embodiments, a cationic lipid is

Compound 300.

[0177] In embodiments, a cationic lipid is Compound 301. In embodiments, a cationic lipid is Compound 302. In embodiments, a cationic lipid is Compound 303. In embodiments, a cationic lipid is Compound 304. In embodiments, a cationic lipid is Compound 305. In embodiments, a cationic lipid is Compound 306. In embodiments, a cationic lipid is Compound 307. In embodiments, a cationic lipid is Compound 308. In embodiments, a cationic lipid is

Compound 309. In embodiments, a cationic lipid is Compound 310. In embodiments, a cationic lipid is Compound 311. In embodiments, a cationic lipid is Compound 312. In embodiments, a cationic lipid is Compound 313. In embodiments, a cationic lipid is Compound 314. In embodiments, a cationic lipid is Compound 315. In embodiments, a cationic lipid is

Compound 316. In embodiments, a cationic lipid is Compound 317. In embodiments, a cationic lipid is Compound 318. In embodiments, a cationic lipid is Compound 319. In embodiments, a cationic lipid is Compound 320. In embodiments, a cationic lipid is Compound 321. In embodiments, a cationic lipid is Compound 322. In embodiments, a cationic lipid is

Compound 323. In embodiments, a cationic lipid is Compound 324. In embodiments, a cationic lipid is Compound 325. In embodiments, a cationic lipid is Compound 326. In embodiments, a cationic lipid is Compound 327. In embodiments, a cationic lipid is Compound 328. In embodiments, a cationic lipid is Compound 329. In embodiments, a cationic lipid is

Compound 330.

[0178] In embodiments, a cationic lipid is Compound 331. In embodiments, a cationic lipid is Compound 332. In embodiments, a cationic lipid is Compound 333. In embodiments, a cationic lipid is Compound 334. In embodiments, a cationic lipid is Compound 335. In embodiments, a cationic lipid is Compound 336. In embodiments, a cationic lipid is Compound 337. In embodiments, a cationic lipid is Compound 338. In embodiments, a cationic lipid is

Compound 339. In embodiments, a cationic lipid is Compound 340. In embodiments, a cationic lipid is Compound 341. In embodiments, a cationic lipid is Compound 342. In embodiments, a cationic lipid is Compound 343. In embodiments, a cationic lipid is Compound 344. In embodiments, a cationic lipid is Compound 345. In embodiments, a cationic lipid is

Compound 346. In embodiments, a cationic lipid is Compound 347. In embodiments, a cationic lipid is Compound 348. In embodiments, a cationic lipid is Compound 349. In embodiments, a cationic lipid is Compound 350. In embodiments, a cationic lipid is Compound 351. In embodiments, a cationic lipid is Compound 352. In embodiments, a cationic lipid is

Compound 353. In embodiments, a cationic lipid is Compound 354. In embodiments, a cationic lipid is Compound 355. In embodiments, a cationic lipid is Compound 356. In embodiments, a cationic lipid is Compound 357. In embodiments, a cationic lipid is Compound 358. In embodiments, a cationic lipid is Compound 359. In embodiments, a cationic lipid is

Compound 360.

[0179] In embodiments, a cationic lipid is Compound 361. In embodiments, a cationic lipid is Compound 362. In embodiments, a cationic lipid is Compound 363. In embodiments, a cationic lipid is Compound 364. In embodiments, a cationic lipid is Compound 365. In embodiments, a cationic lipid is Compound 366. In embodiments, a cationic lipid is Compound 367. In embodiments, a cationic lipid is Compound 368. In embodiments, a cationic lipid is

Compound 369. In embodiments, a cationic lipid is Compound 370. In embodiments, a cationic lipid is Compound 371. In embodiments, a cationic lipid is Compound 372. In embodiments, a cationic lipid is Compound 373. In embodiments, a cationic lipid is Compound 374. In embodiments, a cationic lipid is Compound 375. In embodiments, a cationic lipid is

Compound 376. In embodiments, a cationic lipid is Compound 377. In embodiments, a cationic lipid is Compound 378. Svnthesis of Comoounds of the Invention

[0180] The compounds described herein (e.g., a compound of any of Formulae (A)-(E), such as any of Formulae (l)-(IX) or any one of Compounds 1-378) can be prepared according to methods known in the art, including the exemplary syntheses of the Examples provided herein.

Nucleic Acids

[0181] The compounds described herein (e.g., a compound of any of Formulae (A)-(E), such as any of Formulae (l)-(IX) or any one of Compounds 1-378) can be used to prepare compositions useful for the delivery of nucleic acids.

Synthesis of Nucleic Acids

[0182] 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.

[0183] 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.

[0184] 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.

[0185] 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 (sea RNA), 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 (siRNA), transacting siRNA (tasiRNA), repeat associated siRNA (rasiRNA), 73K RNA, retrotransposons, a viral genome, a vlroid, satellite RNA, or derivatives of these groups. In some embodiments, a nucleic acid is a mRNA encoding a protein.

Synthesis of mRNA

[0186] 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 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.

[0187] 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.

[0188] 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

[0189] 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, S'-methoxycarbonylmethyl-uracil, 5-methoxy-uracil, uracil-5-oxyacetic acid methyl ester, uracil-5-oxyacetic acid (v), 1-methyl-pseudouracil, queuosine, beta-D-mannosyl- queuosine, 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.

[0190] In some embodiments, mRNAs may contain RNA backbone modifications. Typically, a

backbone modification is a modification in which the phosphates of the backbone of the nucleotides contained in the RNA are modified chemically. Exemplary backbone modifications typically include, but are not limited to, modifications from the group consisting of

methylphosphonates, methylphosphoramidates, phosphoramidates, phosphorothioates (e.g., cytidine 5'-O-(l-thiophosphate)), boranophosphates, positively charged guanidlnlum groups etc., which means by replacing the phosphodiester linkage by other anionic, cationic or neutral groups.

[0191] 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-oligo ribonucleotide (2'-fluoro-2'-deoxycytidine S'-triphosphate, 2'- fluoro-2'-deoxyuridine 5'-triphosphate), 2'-deoxy-2'-deamine-oligoribonucleotide (2'-amino-2'- deoxycytidine S'-triphosphate, 2'-amino-2'-deoxyuridine S'-triphosphate), 2'-O- alkyloligoribonucleotide, 2'-deoxy-2'-C-alkyloligoribonucleotide (2'-O-methylcytidine S'- triphosphate, 2'-methyluridine S'-triphosphate), 2'-C-alkyloligoribonucleotide, and isomers thereof (2'-aracytldine S'-triphosphate, 2'-arauridine S'-triphosphate), or azidotriphosphates (2‘- azido-2'-deoxycytidine S'-triphosphate, 2'-azido-2'-deoxyuridlne S'-triphosphate).

[0192] 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 S'-triphosphate, 2-amlnoadenosine S'-triphosphate, 2-thiocytidine S'-triphosphate, 2-thiourldine S'-triphosphate, 4-thiouridine S'-triphosphate, 5- aminoallylcytidine S'-triphosphate, 5-aminoallyluridine S'-triphosphate, 5-bromocytidine S'- triphosphate, 5-bromouridine S'-triphosphate, 5-iodocytidine S'-triphosphate, 5-iodouridine S'- triphosphate, 5-methylcytidine S'-triphosphate, 5-methyluridine S'-triphosphate, 6-azacytidine S'-triphosphate, 6-azauridine S'-triphosphate, 6-chloropurine riboside S'-triphosphate, 7- deazaadenosine S'-triphosphate, 7-deazaguanosine S'-triphosphate, 8-azaadenosine S'- triphosphate, 8-azidoadenosine S'-triphosphate, benzimidazole riboside S'-triphosphate, Nl- methyladenosine S'-triphosphate, Nl-methylguanosine S'-triphosphate, N6-methyladenosine S'- triphosphate, 06-methylguanosine S'-triphosphate, pseudouridine S'-triphosphate, puromycin S'-triphosphate or xanthosine S'-triphosphate.

[0193] Typically, mRNA synthesis includes the addition of a "cap" on the N-termlnal (S') end, and a "tail" on the C-termlnal (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.

[0194] 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.

[0195] 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.

[0196] 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.

[0197] In some embodiments, a 3' untranslated region includes one or more of a polyadenylatlon 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

[0198] In some embodiments, in some embodiments, mRNAs (e.g., mRNAs encoding CFTR) 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' triphosphate linkage; and the 7-nitrogen of guanine is then methylated by a methyltransferase. In some embodiments, the nucleotide forming the cap is further methylated at the 3'position. In some embodiments, the nucleotide directly adjacent to the cap is further methylated at the 2' position. Examples of cap structures include, but are not limited to, m7G(5')ppp(5 , )(2'OMeG), m7G(5')ppp(5')(2'OMeA),

m7(3OMeG)(5')ppp(5')(2'OMeG), m7(3'OMeG)(5')ppp(5')(2'OMeA), m7G(5‘)ppp

(5'( ,G(5')prr(5')A and G(5')ppp(5')G. In a specific embodiment, the cap structure is m7G(5')ppp(5')(2OMeG). [0199] 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.

[0200] In some embodiments, the 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 S'-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.

[0201] In some embodiments, a 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 .

[0202] 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; un methylated cap analogs (e.g., GpppG); dimethylated cap analog (e.g., m^GpppG), trimethylated cap analog (e.g., m 2,2,7 GpppG), dimethylated symmetrical cap analogs (e.g., m 7 Gpppm 7 G), or anti reverse cap analogs (e.g., ARCA; m 7 , 2 ' Ome GpppG, m 72d GpppG, m 7,3 ' Ome GpppG, m 7,3,d GpppG and their tetraphosphate derivatives) (see, e.g., Jemielity, J. et al. , * Novel 1 anti-reverse’ cap analogs with superior translational properties?, RNA, 9: 1108-1122 (2003)).

[0203] 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.

[0204] In some embodiments, the cap is a CapO structure. CapO structures lack a 2 -O-methyl residue of the rlbose attached to bases 1 and 2. In some embodiments, the cap Is a Capl structure. Capl structures have a 2 -O-methyl residue at base 2. In some embodiments, the cap is a Cap2 structure. Cap2 structures have a 2 -O-methyl residue attached to both bases 2 and 3.

[0205] 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.

[0206] Additional cap structures are also described in published US Application No. US

2016/0032356 and U.S. Provisional Application 62/464,327, filed February 27, 2017, which are Incorporated herein by reference.

Tail structure

[0207] Typically, the presence of a "tail" serves to protect the mRNA from exonuclease degradation.

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

[0208] 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.

[0209] 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 tall 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.

S' and 3' Untranslated Region

[0210] 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.

[0211] 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 mlRNAs. In some embodiments, a 3' untranslated region may be between 50 and 500 nucleotides in length or longer.

[0212] Exemplary 3‘ and/or 5‘ UTR sequences can be derived from mRNA molecules which are stable (e.g., globin, actin, GAPDH, tubulin, histone, or citric acid cycle enzymes) to increase the stability of the sense mRNA molecule. For example, a 5' UTR sequence may include a partial sequence of a CMV immediate-early 1 (IE1) gene, or a fragment thereof to Improve the nuclease resistance and/or improve the half-life of the polynucleotide. Also contemplated Is the Inclusion of a sequence encoding human growth hormone (hGH), or a fragment thereof to the 3' end or untranslated region of the polynucleotide (e.g., mRNA) to further stabilize the polynucleotide. Generally, these modifications Improve the stability and/or pharmacokinetic properties (e.g., half-life) of the polynucleotide relative to their unmodified counterparts, and include, for example modifications made to Improve such polynucleotides' resistance to in vivo nuclease digestion.

Pharmaceutical Formulations of Cationic Upids and Nucleic Adds

[0213] In certain embodiments, the compounds described herein (e.g., a compound of any of Formulae (A)-(E), such as any of Formulae (l)-(IX) or any one of Compounds 1-378), 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. [0214] 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 any of Formulae (A)- (E), such as any of Formulae (l)-(IX) or any one of Compounds 1-378).

[0215] As used herein, the terms "delivery vehicle," "transfer vehicle," "nanoparticle," or

grammatical equivalents thereof, are used interchangeably.

[0216] For example, the present invention provides a composition (e.g., a pharmaceutical

composition) comprising a compound described herein (e.g., a compound of any of Formulae (A)-(E), such as any of Formulae (l)-(IX) or any one of Compounds 1-378) and one or more polynucleotides. A composition (e.g., a pharmaceutical composition) may further comprise one or more cationic lipids, one or more non-cationlc lipids, one or more cholesterol-based lipids and/or one or more PEG-modified lipids.

[0217] 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 any of Formulae (A)-(E), such as any of Formulae (l)-(IX) or any one of Compounds 1-378) 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. [0218] 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.

[0219] 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

[0220] In some embodiments, a composition is a suitable delivery vehicle. In embodiments, a composition is a liposomal delivery vehicle, e.g., a lipid nanoparticle.

[0221] The terms "liposomal delivery vehicle" and "liposomal composition" are used

interchangeably.

[0222] 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.

[0223] Thus, in certain embodiments, the compounds described herein (e.g., a compound of any of Formulae (A)-(E), such as any of Formulae (l)-(IX) or any one of Compounds 1-378) 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).

[0224] 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.

[0225] In certain embodiments, such compositions (e.g., liposomal compositions) are loaded with or otherwise encapsulate materials, such as for example, one or more biologically-acthze polynucleotides (e.g., mRNA).

[0226] 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-cationlc 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 any of Formulae (A)-(E), such as any of Formulae (l)-(IX) or any one of Compounds 1-378). 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.

[0227] 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 any of Formulae (A)-(E), such as any of Formulae (l)-(IX) or any one of Compounds 1-378) as described herein.

[0228] 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. [0229] In embodiments, a liposomal delivery vehicle (e.g., a lipid nanoparticle) can have a net positive charge.

[0230] In embodiments, a liposomal delivery vehicle (e.g., a lipid nanoparticle) can have a net negative charge.

[0231] In embodiments, a liposomal delivery vehicle (e.g., a lipid nanoparticle) can have a net neutral charge.

[0232] 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 any of Formulae (A)-(E), such as any of Formulae (l)-(IX) or any one of Compounds 1-378).

[0233] For example, the amount of a compound as described herein (e.g., a compound of any of Formulae (A)-(E), such as any of Formulae (l)-(IX) or any one of Compounds 1-378) 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).

[0234] In embodiments of the pharmaceutical compositions described herein, a compound as described herein (e.g., a compound of any of Formulae (A)-(E), such as any of Formulae (l)-(IX) or any one of Compounds 1-378) 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).

[0235] In embodiments, a compound as described herein (e.g., a compound of any of Formulae (A)- (E), such as any of Formulae (l)-(IX) or any one of Compounds 1-378) 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 any of Formulae (A)-(E), such as any of Formulae (l)-(IX) or any one of Compounds 1-378) 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.

[0236] In embodiments, the amount of a compound as described herein (e.g., a compound of any of Formulae (A)-(E), such as any of Formulae (l)-(IX) or any one of Compounds 1-378) 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).

[0237] In embodiments, the amount of a compound as described herein ((e.g., a compound of any of Formulae (A)-(E), such as any of Formulae (l)-(IX) or any one of Compounds 1-378) 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).

[0238] 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 any of Formulae (A)-(E), such as any of Formulae (l)-(IX) or any one of Compounds 1-378). 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% of a compound described herein (e.g., a compound of any of Formulae (A)-(E), such as any of Formulae (l)-(IX) or any one of Compounds 1-378). 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 any of Formulae (A)-(E), such as any of Formulae (l)-(IX) or any one of Compounds 1-378). In embodiments, the percentage results in an improved beneficial effect (e.g., improved delivery to targeted tissues such as the liver or the lung).

[0239] The amount of a compound as described herein (e.g., a compound of any of Formulae (A)- (E), such as any of Formulae (l)-(IX) or any one of Compounds 1-378) 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).

[0240] In embodiments of pharmaceutical compositions described herein, a compound as

described herein (e.g., a compound of any of Formulae (A)-(E), such as any of Formulae (l)-(IX) or any one of Compounds 1-378) is present in an amount that is about 0.5 mol% to about 30 mol% (e.g., about 0.5 mol% to about 20 mol%) of the combined molar amounts of all lipids present in a composition such as a liposomal delivery vehicle.

[0241] In embodiments, a compound as described herein (e.g., a compound of any of Formulae (A)- (E), such as any of Formulae (l)-(IX) or any one of Compounds 1-378) 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 any of Formulae (A)-(E), such as any of Formulae (l)-(IX) or any one of Compounds 1-378) is present in an amount that is about 1 mot% 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

[0242] In certain embodiments, a compound as described herein (e.g., a compound of any of Formulae (A)-(E), such as any of Formulae (l)-(IX) or any one of Compounds 1-378) 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).

[0243] In certain embodiments, a compound as described herein (e.g., a compound of any of Formulae (A)-(E), such as any of Formulae (l)-(IX) or any one of Compounds 1-378) 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.

[0244] In certain embodiments, a compound as described (e.g., a compound of any of Formulae (A)-(E), such as any of Formulae (l)-(IX) or any one of Compounds 1-378) 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).

[0245] In embodiments, the amount of a compound as described herein (e.g., a compound of any of Formulae (A)-(E), such as any of Formulae (l)-(IX) or any one of Compounds 1-378) 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).

[0246] In embodiments, the amount of a compound as described herein (e.g., a compound of any of Formulae (A)-(E), such as any of Formulae (l)-(IX) or any one of Compounds 1-378) 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).

[0247] In embodiments, the percentage results in an improved beneficial effect (e.g., improved delivery to targeted tissues such as the liver or the lung).

[0248] 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).

[0249] 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.

[0250] 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 any of Formulae (A)-(E), such as any of Formulae (l)-(IX) or any one of Compounds 1-378) and one or more lipids selected from the group consisting of a cationic lipid, a non-cationic lipid, and a PEGylated lipid.

[0251] 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 any of Formulae (A)-(E), such as any of Formulae (l)-(IX) or any one of Compounds 1-378); 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.

[0252] 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 any of Formulae (A)-(E), such as any of Formulae (l)-(IX) or any one of Compounds 1-378), 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.

[0253] 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, fusogeniclty and toxicity of the selected llpld(s). Thus, the molar ratios may be adjusted accordingly.

Cationic Lipids

[0254] In addition to any of the compounds as described herein (e.g., a compound of any of

Formulae (A)-(E), such as any of Formulae (l)-(IX) or any one of Compounds 1-378), a composition may comprise one or more additional cationic lipids.

[0255] In some embodiments, liposomes may comprise one or more additional cationic lipids. As used herein, the phrase "cationic lipid" refers to any of a number of lipid species that have a net positive charge at a selected pH, such as physiological pH. Several cationic lipids have been described in the literature, many of which are commercially available.

[0256] Suitable additional cationic lipids for use in the compositions include the cationic lipids as described in International Patent Publication WO 2010/144740, which is incorporated herein by reference. In certain embodiments, the compositions include a cationic lipid, (6Z,9Z,28Z,31Z)- heptatriaconta-6,9,28,31-tetraen-19-yl 4-(dimethylamino) butanoate, having a compound structure of:

and pharmaceutically acceptable salts thereof.

[0257] Other suitable additional cationic lipids for use in the compositions include ionizable cationic lipids as described in International Patent Publication WO 2013/149140, which is incorporated herein by reference. In some embodiments, the compositions include a cationic lipid of one of the following formulas:

or a pharmaceutically acceptable salt thereof, wherein R 1 and Rz are each independently selected from the group consisting of hydrogen, an optionally substituted, variably saturated or unsaturated C 1 -C 20 alkyl and an optionally substituted, variably saturated or unsaturated C 6 -C 20 acyl; wherein L 1 and Lz are each independently selected from the group consisting of hydrogen, an optionally substituted C1-C30 alkyl, an optionally substituted variably unsaturated C1-C30 alkenyl, and an optionally substituted C1-C30 alkynyl; wherein m and o are each independently selected from the group consisting of zero and any positive integer (e.g., where m is three); and wherein n is zero or any positive integer (e.g., where n is one). In certain embodiments, the compositions include the cationic lipid (15Z, 18Z)-N,N-dimethyl-6-(9Z,12Z)-octadeca-9,12-dien-l - yl) tetracosa- 15,18-dien- 1-amine ("HGT5000"), having a compound structure of:

and pharmaceutically acceptable salts thereof. In certain embodiments, the compositions include the cationic lipid (15Z, 18Z)-N,N-dimethyl-6-((9Z,12Z)-octadeca-9,12-dien-l-yl) tetracosa- 4,15,18-trien-l -amine ("HGTSOOl"), having a compound structure of:

and pharmaceutically acceptable salts thereof. In certain embodiments, the include the cationic lipid and (15Z,18Z)-N,N-dimethyl-6-((9Z,12Z)-octadeca-9,12-dien-l-yl) tetracosa-5,15,18-trien- 1 -amine ("HGT5002"), having a compound structure of:

and pharmaceutically acceptable salts thereof.

[0258] Other suitable additional cationic lipids for use in the compositions include cationic lipids described as aminoalcohol lipidoids in International Patent Publication WO 2010/053572, which is incorporated herein by reference. In certain embodiments, the compositions include a cationic lipid having a compound structure of:

and pharmaceutically acceptable salts thereof.

[0259] Other suitable additional cationic lipids for use in the compositions include the cationic lipids as described in International Patent Publication WO 2016/118725, which is incorporated herein by reference. In certain embodiments, the compositions include a cationic lipid having a compound structure of:

and pharmaceutically acceptable salts thereof.

[0260] Other suitable additional cationic lipids for use in the compositions include the cationic lipids as described in International Patent Publication WO 2016/118724, which is incorporated herein by reference. In certain embodiments, the compositions include a cationic lipid having a compound structure of:

and pharmaceutically acceptable salts thereof.

[0261] Other suitable cationic lipids for use in the compositions include a cationic lipid having the formula of 14,25-dltridecyl 15,18, 21, 24-tetraaza-octatriacontane, and pharmaceutically acceptable salts thereof.

[0262] Other suitable additional cationic lipids for use in the compositions include the cationic lipids as described in International Patent 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 6 -C 40 alkenyl. In certain embodiments, the compositions include a cationic lipid having a compound structure of:

and pharmaceutically acceptable salts thereof.

[0263] In certain embodiments, the compositions include a cationic lipid having a compound structure of:

and pharmaceutically acceptable salts thereof.

[0264] In certain embodiments, the compositions include a cationic lipid having a compound structure of:

and pharmaceutically acceptable salts thereof.

[0265] In certain embodiments, the compositions include a cationic lipid having a compound

structure of:

and pharmaceutically acceptable salts thereof.

[0266] Other suitable additional cationic lipids for use in the compositions include the cationic lipids as described in International Patent Publication WO 2015/184256, which is incorporated herein by reference. In some embodiments, the compositions include a cationic lipid of the following formula:

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 R* is independently hydrogen, optionally substituted C1-50 alkyl, optionally substituted C2-50 alkenyl, optionally substituted C2-50 alkynyl, optionally substituted C3-10 carbocyclyl, optionally substituted 3-14 membered heterocyclyl, optionally substituted C6-14 aryl, optionally substituted 5-14 membered heteroaryl or halogen; and each 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:

and pharmaceutically acceptable salts thereof.

[0267] Other suitable additional cationic lipids for use In the compositions include the cationic lipids as described in International Patent Publication WO 2016/004202, which is incorporated herein by reference. In some embodiments, the compositions include a cationic lipid having the compound structure:

or a pharmaceutically acceptable salt thereof.

[0268] In some embodiments, the compositions include a cationic lipid having the compound structure:

or a pharmaceutically acceptable salt thereof.

[0269] In some embodiments, the compositions include a cationic lipid having the compound structure:

or a pharmaceutically acceptable salt thereof.

[0270] Other suitable additional cationic lipids for use In the compositions include the cationic lipids as described in J. McClellan, M. C. King, C 6 ll 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.

[0271] Other suitable additional cationic lipids for use In the compositions include the cationic lipids as described in International Patent Publication WO 2015/199952, which is incorporated herein by reference. In some embodiments, the compositions include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof.

[0272] In some embodiments, the compositions Include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof. [0273] In some embodiments, the compositions include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof.

[0274] In some embodiments, the compositions include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof.

[0275] In some embodiments, the compositions include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof.

[0276] In some embodiments, the compositions include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof.

[0277] In some embodiments, the compositions include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof.

[0278] In some embodiments, the compositions include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof.

[0279] In some embodiments, the compositions include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof. [0280] In some embodiments, the compositions include a cationic lipid having the compound structure:

[0281] and pharmaceutically acceptable salts thereof.

[0282] In some embodiments, the compositions include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof.

[0283] In some embodiments, the compositions include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof.

[0284] In some embodiments, the compositions Include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof.

[0285] Other suitable additional cationic lipids for use In the compositions include the cationic lipids as described in International Patent Publication WO 2017/004143, which is incorporated herein by reference.

[0286] In some embodiments, the compositions include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof.

[0287] In some embodiments, the compositions include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof.

[0288] In some embodiments, the compositions include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof.

[0289] In some embodiments, the compositions include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof.

[0290] In some embodiments, the compositions include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof.

[0291] In some embodiments, the compositions include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof.

[0292] In some embodiments, the compositions include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof. [0293] In some embodiments, the compositions include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof.

[0294] In some embodiments, the compositions include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof.

[0295] In some embodiments, the compositions include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof.

[0296] In some embodiments, the compositions include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof.

[0297] In some embodiments, the compositions include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof.

[0298] In some embodiments, the compositions include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof.

[0299] In some embodiments, the compositions Include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof. [0300] In some embodiments, the compositions include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof.

[0301] In some embodiments, the compositions include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof.

[0302] In some embodiments, the compositions include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof.

[0303] Other suitable additional cationic lipids for use in the compositions include the cationic lipids as described in International Patent Publication WO 2017/075531, which is incorporated herein by reference. In some embodiments, the compositions include a cationic lipid of the following formula:

or a pharmaceutically acceptable salt thereof, wherein one of L 1 or L 2 is -O(C=O)-, -(C=O)0-, - C(=O)-, -O-, -S(O)x, -S-S-, -C(=O)S-, -SC(=O)-, -NR a C(=O)-, -C(=O)NR a - NR a C(=O)NR a -, -OC(=O)NR a -, or -NR a C(=O)0-; and the other of L 1 or L 2 is -O(C=O)-, -(C=O)0-, -C(=O)-, -O-, -S(O)„, -S-S-, -C(=O)S- , SC(=O)-, -NR a C(=O)-, -C(=O)NR a -, ,NR a C(=O)NR a -, -OC(=O)NR a - or -NR a C(=O)0- or a direct bond; G 1 and G 2 are each Independently unsubstituted C 1 -C 12 alkylene or C 1 -C 12 alkenylene; G 3 is C 1 -C 24 alkylene, C 1 -C 24 alkenylene, C 3 -C 8 cycloalkylene, C 3 -C 8 cycloalkenylene; R a is H or C 1 -C 12 alkyl; R 1 and R 2 are each Independently C 6 -C 24 alkyl or C 6 -C 24 alkenyl; R 3 is H, OR 5 , CN, -C(=O)0R 4 , - 0C(=O)R* or -NR 5 C(=O)R*; R 4 is C 1 -C 12 alkyl; R 5 is H or C 1 -C 6 alkyl; and x is 0, 1 or 2.

[0304] Other suitable additional cationic lipids for use in the compositions include the cationic lipids as described in International Patent Publication WO 2017/117528, which is incorporated herein by reference. In some embodiments, the compositions include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof.

[0305] In some embodiments, the compositions include a cationic lipid having the compound

structure:

and pharmaceutically acceptable salts thereof. [0306] In some embodiments, the compositions include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof.

[0307] Other suitable additional cationic lipids for use In the compositions include the cationic lipids as described in International Patent Publication WO 2017/049245, which is incorporated herein by reference. In some embodiments, the cationic lipids of the compositions and methods of the present invention include a compound of one of the following formulas:

and pharmaceutically acceptable salts thereof. For any one of these four formulas, R 4 is independently selected from -(CH 2 )„Q and -(CH 2 ) n CHQR; Q is selected from the group consisting of -OR, -OH, -O(CH 2 )„N(R) 2 , -OC(O)R, -CX3, -CN, -N(R)C(O)R, -N(H)C(O)R, -N(R)S(O) 2 R, -N(H)S(O) 2 R, -N(R)C(O)N(R) 2 , -N(H)C(O)N(R) 2 , -N(H)C(O)N(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 C 1-3 alkyl, C 2-3 alkenyl, and H; and n is 1, 2, or 3.

[0308] In certain embodiments, the compositions include a cationic lipid having a compound

structure of:

and pharmaceutically acceptable salts thereof.

[0309] In certain embodiments, the compositions include a cationic lipid having a compound

structure of:

and pharmaceutically acceptable salts thereof.

[0310] In certain embodiments, the compositions include a cationic lipid having a compound

structure of:

and pharmaceutically acceptable salts thereof.

[0311] In certain embodiments, the compositions include a cationic lipid having a compound

structure of:

and pharmaceutically acceptable salts thereof.

[0312] Other suitable additional cationic lipids for use In the compositions Include the cationic lipids as described in International Patent Publication WO 2017/173054 and WO 2015/095340, each of which is incorporated herein by reference. [0313] In certain embodiments, the compositions include a cationic lipid having a compound structure of:

and pharmaceutically acceptable salts thereof.

[0314] In certain embodiments, the compositions include a cationic lipid having a compound structure of:

and pharmaceutically acceptable salts thereof.

[0315] In certain embodiments, the compositions Include a cationic lipid having a compound structure of:

and pharmaceutically acceptable salts thereof.

[0316] In certain embodiments, the compositions include a cationic lipid having a compound structure of:

and pharmaceutically acceptable salts thereof.

[0317] Other suitable additional cationic lipids for use in the compositions include cholesterol· based cationic lipids. In certain embodiments, the compositions Include imidazole cholesterol ester or "ICE", having a compound structure of:

and pharmaceutically acceptable salts thereof.

[0318] Other suitable additional cationic lipids for use in the compositions include cleavable

cationic lipids as described in International Patent Publication WO 2012/170889, which is incorporated herein by reference. In some embodiments, the compositions include a cationic lipid of the following formula:

wherein R 1 is selected from the group consisting of imidazole, guanidinium, amino, imine, enamine, an optionally-substituted alkyl amino (e.g., an alkyl amino such as dimethylamlno) and pyridyl; wherein R 2 is selected from the group consisting of one of the following two formulas:

and wherein R3 and R*are each Independently selected from the group consisting of an optionally substituted, variably saturated or unsaturated C 6 -C 20 alkyl and an optionally substituted, variably saturated or unsaturated C 6 -C 20 acyl; and wherein n Is zero or any positive integer (e.g., one, two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, fourteen, fifteen, sixteen, seventeen, eighteen, nineteen, twenty or more).

[0319] In certain embodiments, the compositions include a cationic lipid, "HGT4001", having a compound structure of:

and pharmaceutically acceptable salts thereof.

[0320] In certain embodiments, the compositions include a cationic lipid, "HGT4002", having a compound structure of:

and pharmaceutically acceptable salts thereof.

[0321] In certain embodiments, the compositions Include a cationic lipid, "HGT4003", having a compound structure of:

(HGT4003)

and pharmaceutically acceptable salts thereof.

[0322] In certain embodiments, the compositions include a cationic lipid, "HGT4004", having a compound structure of:

and pharmaceutically acceptable salts thereof.

[0323] In certain embodiments, the compositions Include a cationic lipid "HGT4005", having a compound structure of:

and pharmaceutically acceptable salts thereof.

[0324] In some embodiments, the compositions include the cationic lipid, N-[l-(2,3- dioleyloxy)propyl]-N,N,N-trimethylammonium chloride ("DOTMA"). Feigner et al. (Proc. Nat'l Acad. Sci. 84, 7413 (1987); U.S. Pat. No. 4,897,355, each of which is Incorporated herein by reference. DOTMA can be formulated alone or can be combined with a neutral lipid (e.g., dioleoylphosphatldyl-ethanolamine or "DOPE") or still other cationic or non-catlonic 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(spermlne-carboxamldo)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").

[0325] Additional exemplary cationic lipids suitable for the compositions also include: 1,2- distearyloxy-N,N-dimethyl-3-amlnopropane ( "DSDMA"); l,2-dioleyloxy-N,N-dimethyl-3- aminopropane ("DODMA"); 1 ,2-dllinoleyloxy-N,N-dimethyl-3-amlnopropane ("DLlnDMA"); 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-dlmethy 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 -yloxyjpropane- 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 -yloxyjpropan- 1 -amine (Octyl-CLinDMA (2S)"); 2,2-dilinoleyl-4-dimethylaminoethyl-[l,3]-dioxolane ("DLin-K- XTC2-DMA"); and 2-(2,2-di((9Z,12Z)-octadeca-9,l 2-dien- 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.

[0326] 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-cyclopenta[d ] [1 ,3]dioxol-5-amine ("ALNY- 100") and/or 4,7, 13-tris(3-oxo-3-(undecylamlno)propyl)-Nl,N16-diundecyl-4, 7, 10,13- tetraazahexadecane- 1,16-diamide ("NC98-5").

[0327] 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

[0328] 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), dioleoylphosphatidylethanolamlne (DOPE), palmitoyloleoylphosphatldylcholine (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-phosphatidyethanolamlne (SOPE), or a mixture thereof. In embodiments, a non-cationlc or helper lipid is dioleoylphosphatidylethanolamine (DOPE).

[0329] In some embodiments, a non-cationlc 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.

[0330] 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-cationlc lipid in a liposome is no more than about 5 mot%, 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%.

[0331] 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-cation ic 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-catlonic 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

[0332] 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- propyl)piperazine (Gao, et al. Biochem. Biophys. Res. Comm. 179, 280 (1991); Wolf et al.

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

[0333] 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%.

[0334] 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

[0335] In some embodiments, a composition (e.g., a liposomal composition) comprises one or more further PEGylated lipids.

[0336] 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 any of Formulae (A)-(E), such as any of Formulae (l)-(IX) or any one of Compounds 1-378) 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., Cu or CM).

[0337] Contemplated further PEG-modified lipids (also referred to herein as a PEGylated lipid, which term is Interchangeable with PEG-modifled 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 6 -C 20 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).

[0338] 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).

[0339] 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

[0340] Compounds described herein (e.g., a compound of any of Formulae (A)-(E), such as any of Formulae (l)-(IX) or any one of Compounds 1-378) 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).

[0341] 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.

[0342] Similarly, in certain embodiments compounds described herein (e.g., a compound of any of Formulae (A)-(E), such as any of Formulae (l)-(IX) or any one of Compounds 1-378) 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.

[0343] Thus, pharmaceutical formulations comprising a compound described (e.g., a compound of any of Formulae (A)-(E), such as any of Formulae (l)-(IX) or any one of Compounds 1-378) 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 any of Formulae (A)-(E), such as any of Formulae (l)-(IX) or any one of Compounds 1-378) 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 any of Formulae (A)-(E), such as any of Formulae (l)-(IX) or any one of Compounds 1-378) can be formulated via pre-mixed lipid solution. In other embodiments, a composition comprising a compound described herein (e.g., a compound of any of Formulae (A)- (E), such as any of Formulae (l)-(IX) or any one of Compounds 1-378) 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.

[0344] 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).

[0345] Alteratively 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.

[0346] Compositions described herein can comprise mRNA encoding peptides including those described herein (e.g., a polypeptide such as a protein).

[0347] In embodiments, a mRNA encodes a polypeptide.

[0348] In embodiments, a mRNA encodes a protein.

[0349] Exemplary peptides encoded by mRNA (e.g., exemplary proteins encoded by mRNA) are described herein.

[0350] 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.

[0351] 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 subfamily 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.

[0352] 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.

[0353] 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 arglnosuccinate 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.

[0354] 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).

[0355] 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.

[0356] 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.

[0357] 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.

[0358] 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.

[0359] In certain embodiments the present invention provides a method for producing a

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

In certain embodiments the present invention provides a method for producing a therapeutic composition having full-length mRNA that encodes for human hemochromatosis (HFE) protein.

[0360] 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.

[0361] 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 Navl.5 channel in muscle tissue or in a muscle cell.

[0362] 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.

[0363] 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.

[0364] 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.

[0365] 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).

[0366] 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.

[0367] 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.

[0368] 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 fulHength 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.

[0369] 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.

[0370] 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.

[0371] 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.

[0372] 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

[0373] 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. [0374] 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

[0375] Information regarding lysosomal proteins is available from Lubke et ai, "Proteomics of the Lysosome," Biochim BiophysActa. (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.

[0376] 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 (/.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 (/.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

[0377] 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). Delivery Methods

[0378] The route of delivery used in the methods of the invention allows for non-invasive, self- administration of the compounds of the invention (e.g., a compound of any of Formulae (A)-(E) y such as any of Formulae (l)-(IX) or any one of Compounds 1-378). In some embodiments, the methods involve intratracheal or pulmonary administration by aerosolization, nebulization, or instillation of a compositions comprising mRNA encoding a therapeutic protein in a suitable transfection or lipid carrier vehicles as described above. In some embodiments, the protein is encapsulated with a liposome. In some embodiments, the liposome comprises a lipid, which is a compound of the invention (e.g., a compound of any of Formulae (A)-(E), such as any of Formulae (l)-(IX) or any one of Compounds 1-378). As used herein below, administration of a compound of the invention includes administration of a composition comprising a compound of the invention.

[0379] Although the local cells and tissues of the lung represent a potential target capable of functioning as a biological depot or reservoir for production and secretion of the protein encoded by the mRNA, applicants have discovered that administration of the compounds of the invention (e.g., a compound of any of Formulae (A)-(E), such as any of Formulae (l)-(IX) or any one of Compounds 1-378) to the lung via aerosolization, nebulization, or instillation results In the distribution of even non-secreted proteins outside the lung cells. Without wishing to be bound by any particular theory, it is contemplated that nanoparticle compositions of the invention pass, through the lung airway-blood barrier, resulting in translation of the intact nanoparticle to non-lung cells and tissues, such as, e.g., the heart, the liver, the spleen, where it results in the production of the encoded protein in these non-lung tissues. Thus, the utility of the compounds of the invention (e.g., a compound of any of Formulae (A)-(E), such as any of Formulae (l)-(IX) or any one of Compounds 1-378) and methods of the invention extend beyond production of therapeutic protein in lung cells and tissues of the lung and can be used to delivery to non-lung target cells and/or tissues They are useful in the management and treatment of a large number of diseases, and in particular peripheral diseases which result from both secreted and non-secreted protein and/or enzyme deficiencies (e.g., one or more lysosomal storage disorders). In certain embodiments, the compounds of the invention (e.g., a compound of any of Formulae (A)-(E), such as any of Formulae (l)-(IX) or any one of Compounds 1-378), used in the methods of the invention result in the distribution of the mRNA encapsulated nanoparticles and production of the encoded protein in the liver, spleen, heart, and/or other non-lung cells. For example, administration of the compounds of the Invention (e.g., a compound of any of Formulae (A)-(E), such as any of Formulae (l)-(IX) or any one of Compounds 1-378), by aerosollzatlon, nebulization, or instillation to the lung will result In the composition Itself and Its protein product (e.g., functional beta galactosidase protein) will be detectable in both the local cells and tissues of the lung, as well as in peripheral target cells, tissues and organs as a result of translocation of the mRNA and delivery vehicle to non-lung cells.

[0380] In certain embodiments, the compounds of the Invention (e.g., a compound of any of

Formulae (A)-(E), such as any of Formulae (l)-(IX) or any one of Compounds 1-378) may be employed in the methods of the invention to specifically target peripheral cells or tissues.

Following the pulmonary delivery, it is contemplated the compounds of the invention (e.g., a compound of any of Formulae (A)-(E), such as any of Formulae (l)-(IX) or any one of Compounds 1-378) cross the lung airway-blood barrier and distribute into cells other than the local lung cells. Accordingly, the compounds disclosed herein (e.g., a compound of any of Formulae (A)-(E), such as any of Formulae (l)-(IX) or any one of Compounds 1-378) may be administered to a subject by way of the pulmonary route of administration, using a variety of approach known by those skilled in the art (e.g., by inhalation), and distribute to both the local target cells and tissues of the lung, as well as in peripheral non-lung cells and tissues (e.g., cells of the liver, spleen, kidneys, heart, skeletal muscle, lymph nodes, brain, cerebrospinal fluid, and plasma). As a result, both the local cells of the lung and the peripheral non-lung cells can serve as biological reservoirs or depots capable of producing and/or secreting a translation product encoded by one or more polynucleotides. Accordingly, the present invention is not limited to the treatment of lung diseases or conditions, but rather can be used as a non-invasive means of facilitating the delivery of polynucleotides, or the production of enzymes and proteins encoded thereby. In peripheral organs, tissues and cells (e.g., hepatocytes) which would otherwise be achieved only by systemic administration. Exemplary peripheral non-lung cells include, but are not limited to, hepatocytes, epithelial cells, hematopoietic cells, epithelial cells, endothelial cells, bone cells, stem cells, mesenchymal cells, neural cells, cardiac cells, adipocytes, vascular smooth muscle cells, cardiomyocytes, skeletal muscle cells, beta cells, pituitary cells, synovial lining cells, ovarian cells, testicular cells, fibroblasts, B cells, T cells, reticulocytes, leukocytes, granulocytes and tumor cells.

[0381] Following administration of the composition to the subject, the protein product encoded by the mRNA (e.g., a functional protein or enzyme) is detectable in the peripheral target tissues for at least about one to seven days or longer following administration of the compound to the subject. The amount of protein product necessary to achieve a therapeutic effect will vary depending on the condition being treated, the protein encoded, and the condition of the patient. For example, the protein product may be detectable in the peripheral target tissues at a concentration (e.g., a therapeutic concentration) of at least 0.025-1.5 pg/ml (e.g., at least 0.050 pg/ml, at least 0.075 pg/nnl, at least 0.1 pg/ml, at least 0.2 pg/ml, at least 0.3 pg/ml, at least 0.4 pg/ml, at least 0.5 pg/ml, at least 0.6 pg/nnl, at least 0.7 pg/ml, at least 0.8 pg/ml, at least 0.9 pg/ml, at least 1.0 pg/ml, at least 1.1 pg/ml, at least 1.2 pg/ml, at least 1.3 pg/ml, at least 1.4 pg/ml, or at least 1.5 pg/ml), for at least about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11,12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 35, 40, 45 days or longer following administration of the compound to the subject.

[0382] It has been demonstrated that nucleic acids can be delivered to the lungs by intratracheal administration of a liquid suspension of the compound and inhalation of an aerosol mist produced by a liquid nebulizer or the use of a dry powder apparatus such as that described in U.S. patent 5,780,014, incorporated herein by reference.

[0383] In certain embodiments, the compounds of the Invention (e.g., a compound of any of

Formulae (A)-(E), such as any of Formulae (l)-(IX) or any one of Compounds 1-378) may be formulated such that they may be aerosolized or otherwise delivered as a particulate liquid or solid prior to or upon administration to the subject. Such compounds may be administered with the assistance of one or more suitable devices for administering such solid or liquid particulate compositions (such as, e.g., an aerosolized aqueous solution or suspension) to generate particles that are easily respirable or Inhalable by the subject. In some embodiments, such devices (e.g., a metered dose inhaler, jet-nebulizer, ultrasonic nebulizer, dry-powder-inhalers, propellant- based inhaler or an insufflator) facilitate the administration of a predetermined mass, volume or dose of the compositions (e.g., about 0.5 mg/kg of mRNA per dose) to the subject. For example, in certain embodiments, the compounds of the invention (e.g., a compound of any of Formulae (A)-(E), such as any of Formulae (l)-(IX) or any one of Compounds 1-378) are administered to a subject using a metered dose inhaler containing a suspension or solution comprising the compound and a suitable propellant. In certain embodiments, the compounds of the invention (e.g., a compound of any of Formulae (A)-(E), such as any of Formulae (l)-(IX) or any one of Compounds 1-378) may be formulated as a particulate powder (e.g., respirable dry particles) intended for inhalation. In certain embodiments, compositions of the invention formulated as respirable particles are appropriately sized such that they may be respirable by the subject or delivered using a suitable device (e.g., a mean D50 or D90 particle size less than about 500mth, 400pm, 300mth, 250pm, 200 pm, 150pm, 100pm, 75pm, 50pm, 25pm, 20pm, 15pm, 12.5pm, 10pm, 5pm, 2.5pm or smaller). In yet other embodiments, the compounds of the invention (e.g., a compound of any of Formulae (A)-(E), such as any of Formulae (l)-(IX) or any one of Compounds 1-378) are formulated to include one or more pulmonary surfactants (e.g., lamellar bodies). In some embodiments, the compounds of the invention (e.g., a compound of any of Formulae (A)-(E), such as any of Formulae (l)-(IX) or any one of Compounds 1-378) are administered to a subject such that a concentration of at least 0.05 mg/kg, at least 0.1 mg/kg, at least 0.5 mg/kg, at least 1.0 mg/kg, at least 2.0 mg/kg, at least 3.0 mg/kg, at least 4.0 mg/kg, at least 5.0 mg/kg, at least 6.0 mg/kg, at least 7.0 mg/kg, at least 8.0 mg/kg, at least 9.0 mg/kg, at least 10 mg/kg, at least 15 mg/kg, at least 20 mg/kg, at least 25 mg/kg, at least 30 mg/kg, at least 35 mg/kg, at least 40 mg/kg, at least 45 mg/kg, at least 50 mg/kg, at least 55 mg/kg, at least 60 mg/kg, at least 65 mg/kg, at least 70 mg/kg, at least 75 mg/kg, at least 80 mg/kg, at least 85 mg/kg, at least 90 mg/kg, at least 95 mg/kg, or at least 100 mg/kg body weight is administered In a single dose. In some embodiments, the compounds of the invention (e.g., a compound of any of Formulae (A)-(E), such as any of Formulae (l)-(IX) or any one of Compounds 1-378) are administered to a subject such that a total amount of at least 0.1 mg, at least 0.5 mg, at least 1.0 mg, at least 2.0 mg, at least 3.0 mg, at least 4.0 mg, at least 5.0 mg, at least 6.0 mg, at least 7.0 mg, at least 8.0 mg, at least 9.0 mg, at least 10 mg, at least 15 mg, at least 20 mg, at least 25 mg, at least 30 mg, at least 35 mg, at least 40 mg, at least 45 mg, at least 50 mg, at least 55 mg, at least 60 mg, at least 65 mg, at least 70 mg, at least 75 mg, at least 80 mg, at least 85 mg, at least 90 mg, at least 95 mg or at least 100 mg mRNA Is administered in one or more doses.

EXAMPLES

[0384] 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.

[0385] In embodiments, a cationic lipid described herein can be prepared by conjugating a thiol with a di-carboxylic acid under suitable conditions. Exemplary dl-carboxylic acids are described In Table A, and exemplary thiols are described in Table B. Accordingly, suitable cationic lipids include those resulting from any combination of the precursors described in Table A and Table

B.

Table A. Dkarboxylic Adds

Example 1: Synthesis of Lipid Nanoparticle Formulations

[0386] In embodiments, cationic lipids described herein can be used in the preparation of lipid nanoparticles according to methods known in the art. For example, suitable methods include methods described in International Publication No. WO 2018/089801, which is hereby incorporated by reference in its entirety.

Example 2: In Vivo Expression of hEPO after IM Injection In BALB/c Mice:

[0387] Lipid nanoparticle formulations comprising human erythropoietin (hEPO) mRNA,a cationic Lipid, DMG-PEG2000, cholesterol and DOPE were administered intramuscularly to study mRNA delivery and resultant hEPO expression. Male BALB/c mice at 6-8 weeks old are given a single injection of the LNP formulations into the gastrocnemius muscle at a dosage level of 0.1 ug. Blood samples were collected at 6 and 24 hours post-dose. hEPO protein expression levels were measured in the sera samples by ELISA and presented in Figure 1. These studies show that the cationic lipids described herein are highly effective at delivery mRNA in vivo, resulting in high expression of the protein or polypeptide encoded by the delivered mRNA.

[0388] From the foregoing description, one skilled in the art can easily ascertain the essential characteristics of this invention, and without departing from the spirit and scope thereof, can make various changes and modifications of the invention to adapt it to various usages and conditions.

[0389] All references, patents or applications, U.S. or foreign, cited in the application are hereby incorporated by reference as if written herein in their entireties. Where any inconsistencies arise, material literally disclosed herein controls.