Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
SUBCUTANEOUS DELIVERY OF MESSENGER RNA
Document Type and Number:
WIPO Patent Application WO/2019/222277
Kind Code:
A1
Abstract:
The present invention provides, among other things, methods of formulating nucleic acid-containing nanoparticles with an mRNA encoding an enzyme to afford efficient delivery of payload to a cell or tissue of interest via subcutaneous administration. The resulting payload can be efficiently delivered to the liver and other organs or tissues of a treated subject.

Inventors:
KARVE SHRIRANG (US)
DEROSA FRANK (US)
BHAVSAR ZARNA (US)
HEARTLEIN MICHAEL (US)
Application Number:
PCT/US2019/032300
Publication Date:
November 21, 2019
Filing Date:
May 14, 2019
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
TRANSLATE BIO INC (US)
International Classes:
A61P43/00; A61K38/47; A61K48/00
Domestic Patent References:
WO2015138348A12015-09-17
WO2017218524A12017-12-21
WO2014144196A12014-09-18
WO2010053572A22010-05-14
WO2012170930A12012-12-13
WO2015184256A22015-12-03
WO2013063468A12013-05-02
WO2010042877A12010-04-15
WO2005121348A12005-12-22
WO2010014474A22010-02-04
WO2013149140A12013-10-03
WO2016118725A12016-07-28
WO2016118724A12016-07-28
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
WO2005121348A12005-12-22
Foreign References:
US20170061176W2017-11-10
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
US8093367B22012-01-10
US8304529B22012-11-06
US201261617468P2012-03-29
US4897355A1990-01-30
US5171678A1992-12-15
US5334761A1994-08-02
US5744335A1998-04-28
US201514790562A2015-07-02
US201462020163P2014-07-02
US201662420413P2016-11-10
US201762580155P2017-11-01
US4737323A1988-04-12
US20120041663W2012-06-08
Other References:
SEAN C SEMPLE ET AL: "Rational design of cationic lipids for siRNA delivery", NATURE BIOTECHNOLOGY, GALE GROUP INC, NEW YORK, vol. 28, no. 2, 1 February 2010 (2010-02-01), pages 172 - 176, XP002633693, ISSN: 1087-0156, [retrieved on 20100117], DOI: 10.1038/NBT.1602
STERN ET AL., CHEM. REV., vol. 106, no. 3, 2006, pages 818 - 839
HUMPHREY JH ET AL.: "International Standard for Hyaluronidase", BULL WORLD HEALTH ORGAN., vol. 16, no. 2, 1957, pages 291 - 294
"GenBank", Database accession no. BC110183.1
JEMIELITY, J. ET AL.: "Novel 'anti-reverse' cap analogs with superior translational properties", RNA, vol. 9, 2003, pages 1108 - 1122, XP002466761, DOI: doi:10.1261/rna.5430403
JEMIELITY, J. ET AL., RNA, vol. 9, 2003, pages 1108 - 1122
GRUDZIEN, E ET AL., RNA, vol. 10, 2004, pages 1479 - 1487
GRUDZIEN-NOGALSKA, E. ET AL., RNA, vol. 13, 2007, pages 1745 - 1755
YOKOE ET AL., NATURE BIOTECHNOLOGY, vol. 14, 1996, pages 1252 - 1256
GAO ET AL., BIOCHEM. BIOPHYS. RES. COMM., vol. 179, 1991, pages 280
FEIGNER ET AL., PROC. NAT'L ACAD. SCI., vol. 84, 1987, pages 7413
BEHR ET AL., PROC. NAT.'L ACAD. SCI., vol. 86, 1989, pages 6982
SEMPLE ET AL., 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
J. MCCLELLANM. C. KING, CELL, vol. 141, 2010, pages 210 - 217
WHITEHEAD ET AL., NATURE COMMUNICATIONS, vol. 5, 2014, pages 4277
WOLF ET AL., BIOTECHNIQUES, vol. 23, 1997, pages 139
BLOOMFIELD, ANN. REV. BIOPHYS. BIOENG., vol. 10, 1981, pages 421 - 150
HEYES, J.PALMER, L.BREMNER, K.MACLACHLAN, I.: "Cationic lipid saturation influences intracellular delivery of encapsulated nucleic acids", J. CONTR. REL., vol. 107, 2005, pages 276 - 287, XP008157522, DOI: doi:10.1016/j.jconrel.2005.06.014
SEMPLE, S.C. ET AL.: "Rational Design of Cationic Lipids for siRNA Delivery", NATURE BIOTECH., vol. 28, 2010, pages 172 - 176
LOVE, K.T. ET AL.: "Lipid-like materials for low-dose in vivo gene silencing", PNAS, vol. 107, 2010, pages 1864 - 1869, XP055077922, DOI: doi:10.1073/pnas.0910603106
FECHTER, P.BROWNLEE, G.G.: "Recognition of mRNA cap structures by viral and cellular proteins", J. GEN. VIROLOGY, vol. 86, 2005, pages 1239 - 1249, XP002516933, DOI: doi:10.1099/VIR.0.80755-0
Attorney, Agent or Firm:
MENDEZ, Julio, J. et al. (US)
Download PDF:
Claims:
We claim:

1. A method for subcutaneous delivery of a messenger RNA (mRNA) to a subject in need thereof, the method comprising: administering subcutaneously to the subject a composition comprising:

an mRNA encoding a protein or polypeptide, and

an mRNA encoding a hyaluronidase.

2. The method of claim 1 wherein the mRNA encoding a protein or polypeptide is a

therapeutic mRNA.

3. The method of claim 1, wherein the hyaluronidase is a mammalian hyaluronidase.

4. The method of claim 3, wherein the mammalian hyaluronidase is a bovine hyaluronidase.

5. The method of claim 3, wherein the mammalian hyaluronidase is a human hyaluronidase.

6. The method of claim 1, wherein the hyaluronidase mRNA has a polynucleotide sequence having at least 80% sequence identity to a sequence selected from any one of SEQ ID NOs: 9, 10, and 12.

7. The method of any one of the preceding claims, wherein the therapeutic mRNA or the hyaluronidase mRNA comprises one or more modified nucleotides.

8. The method of any one of the preceding claims, wherein the therapeutic mRNA or the hyaluronidase mRNA are individually capped and tailed.

9. The method of claim 1 wherein the therapeutic mRNA and the hyaluronidase mRNA are encapsulated in one or more lipid nanoparticles.

10. The method of claim 9, wherein the one or more lipid nanoparticles comprise a cationic lipid.

11. The method of claim 10, wherein the cationic lipid is selected from a group consisting of cKK-E 12 (3 ,6-bis(4-(bis(2-hydroxydodecyl)amino)butyl)piperazine-2,5-dione) , OF-02, Target 23, Target 24, ICE, HGT5000, HGT5001, HGT4003, DOTAP (l,2-dioleyl-3- trimethylammonium propane), DODAP (l,2-dioleyl-3-dimethylammonium propane), DOTMA (l,2-di-0-octadecenyl-3-trimethylammonium propane), DLinDMA, DLin- KC2-DMA, dialkylamino-based, imidazole-based, and guanidinium-based cationic lipids.

12. The method of claim 9, wherein the one or more lipid nanoparticles comprise a PEG- modified lipid.

13. The composition of claim 9, wherein the PEG-modified lipid constitutes at least 1%, at least 2%, at least 3%, at least 4%, at least 5%, at least 6%, at least 7%, at least 8%, at least 9%, or at least 10% of the total lipids in the lipid nanoparticle.

14. The method of any of the above claims where the lipid nanoparticle comprises a

liposome.

15. The method of claim 1 wherein the therapeutic mRNA and the hyaluronidase mRNA are encapsulated in a single liposome.

16. The method of claim 1 wherein the therapeutic mRNA is administered in a composition separate from that of the hyaluronidase mRNA composition.

17. The method of claim 1 wherein the therapeutic mRNA and the hyaluronidase mRNA are administered in a single formulation.

18. The method of claim 1 wherein the therapeutic mRNA and the hyaluronidase mRNA are administered in separate formulations.

19. The method of claim 1 wherein the therapeutic mRNA and the hyaluronidase mRNA are administered separately.

20. The method of claim 1 wherein the hyaluronidase mRNA mRNA is administered prior to administering the therapeutic mRNA composition.

21. The method of claim 1 wherein the hyaluronidase-encoding mRNA is administered 0.1 hours, 0.2 hours, 0.3 hours, 0.4 hours, 0.5 hours, 1 hour, 2 hours, 3 hours, 4 hours, 5 hours or 6 hours prior to administering the therapeutic mRNA composition.

22. The method of claim 1 wherein the hyaluronidase-encoding mRNA is administered 1 hour, 2 hours, 3 hours, 4 hours, 5 hours or 6 hours prior to administering the first mRNA composition.

23. The method of claim 1, wherein the therapeutic mRNA is expressed in the liver.

24. The method of claim 1, wherein the therapeutic mRNA is expressed in the lung.

25. The method of claim 1, wherein the therapeutic mRNA is expressed in the kidney.

26. The method of claim 1, wherein the therapeutic mRNA is expressed in the subcutaneous tissue.

27. The method of claim 1, wherein the therapeutic mRNA is expressed in the serum.

28. The method of claim 1, wherein the administering the mRNA subcutaneously comprises subcutaneous injection.

29. The method of claim 1, wherein the administering the hyaluronidase mRNA

subcutaneously comprises topical administration.

30. A method of messenger RNA (mRNA) delivery for in vivo protein expression,

comprising,

administering via subcutaneous injection to a subject

a) an mRNA encoding a protein, and

b) an mRNA encoding a hyaluronidase enzyme.

31. A method for treating a disease, disorder or condition in a subject, comprising delivering subcutaneously to the subject a therapeutic mRNA encoding a protein or a polypeptide, and a helper mRNA encoding a hyaluronidase, wherein the therapeutic mRNA-encoded protein or polypeptide is deficient in the subject.

32. The method of claim 31, wherein therapeutic mRNA is expressed in the liver.

33. The method of claim 31, wherein therapeutic mRNA is expressed in the lung.

34. The method of claim 31, wherein the disease, disorder or condition is selected from

ornithine transcarbamylase (OTC) deficiency, Phenylalanine hydroxylase (PAH) deficiency (phenylketonuria, PKU), arginino succinate synthase 1 (ASS1) deficiency, erythropoietin (EPO) deficiency,, Fabry disease; hemophilic diseases (such as, e.g., hemophilia B (FIX), hemophilia A (FVIII); SMNl-related spinal muscular atrophy (SMA); amyotrophic lateral sclerosis (AFS); GAFT-related galactosemia; COF4A5- related disorders including Alport syndrome; galactocerebrosidase deficiencies; X-linked adrenoleukodystrophy; Friedreich’s ataxia; Pelizaeus-Merzbacher disease; TSC1 and TSC2-related tuberous sclerosis; Sanfilippo B syndrome (MPS IIIB); the FMRl-related disorders which include Fragile X syndrome, Fragile X-Associated Tremor/ Ataxia Syndrome and Fragile X Premature Ovarian Failure Syndrome; Prader-Willi syndrome; hereditary hemorrhagic telangiectasia (AT); Niemann-Pick disease Type Cl; the neuronal ceroid lipofuscinoses-related diseases including Juvenile Neuronal Ceroid Fipofuscinosis (JNCF), Juvenile Batten disease, Santavuori-Haltia disease, Jansky-Bielschowsky disease, and PTT-l and TPP1 deficiencies; EIF2B1, EIF2B2, EIF2B3, EIF2B4 and EIF2B 5 -related childhood ataxia with central nervous system hypomyelination/vanishing white matter; CACNA1A and CACNB4-related Episodic Ataxia Type 2; the MECP2- related disorders including Classic Rett Syndrome, MECP2-related Severe Neonatal Encephalopathy and PPM-X Syndrome; CDKF5-related Atypical Rett Syndrome;

Kennedy’s disease (SBMA); Notch-3 related cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIF); SCN1A and SCN1B- related seizure disorders; the Polymerase G-related disorders which include Alpers- Huttenlocher syndrome, POFG-related sensory ataxic neuropathy, dysarthria, and ophthalmoparesis, and autosomal dominant and recessive progressive external ophthalmoplegia with mitochondrial DNA deletions; X-Linked adrenal hypoplasia; X- linked agammaglobulinemia; and Wilson’s disease.

35. The method of claim 31, wherein the disease is ornithine transcarbamylase deficiency.

36. The method of claim 1 or 31, wherein the therapeutic mRNA encodes human ornithine transcarbamylase (OTC).

Description:
SUBCUTANEOUS DELIVERY OF MESSENGER RNA

CROSS REFERENCE TO RELATED APPLICATION

[0001] This application claims priority to U.S. Provisional Application Serial No.

62/671,820, filed May 15, 2018, which is incorporated by reference herein in its entirety.

SEQUENCE LISTING

[0002] The present specification makes reference to a Sequence Listing (submitted electronically as a .txt file named“MRT-l252WO_ST25” on May 13, 2019). The .txt file was generated May 13, 2019 and is 26,169 bytes in size. The entire contents of the Sequence Listing are herein incorporated by reference.

BACKGROUND

[0003] Messenger RNA therapy (MRT) is becoming an increasingly important approach for the treatment of a variety of diseases. MRT involves administration of messenger RNA (mRNA) to a patient in need of the therapy for production of the protein encoded by the mRNA within the patient’s body. Lipid nanoparticles are commonly being used to deliver mRNA for efficient in vivo delivery of mRNA and it is now possible to deliver specific mRNA-loaded lipid nanoparticles systemically via intravenous delivery. However, for increase in patient comfort and compliance, improvements in subcutaneous methods of delivery of therapeutic mRNA are greatly needed. SUMMARY OF INVENTION

[0004] The present invention provides, among other things, improved methods and compositions for the effective in vivo delivery of mRNA via subcutaneous administration. In particular, an mRNA encoding a protein of therapeutic interest is injected subcutaneously with an mRNA encoding an enzyme that is capable of degrading extracellular matrices such as a hyaluronidase, for efficient exposure of the theraepeutic mRNA to the circulation. As described herein, a first mRNA encoding a protein of therapeutic interest when administered with a second mRNA encoding hyaluronidase, results in unexpectedly efficient delivery of the first therapeutic mRNA, accompanied with its efficient protein expression in vivo , particularly in the liver. The mRNAs are encapsulated in lipid nanoparticles (LNPs). In some embodiments the therapeutic mRNA is encapsulated in lipid nanoparticles (LNPs). In some embodiments both the therapeutic mRNA and the hyaluronidase mRNA are encapsulated in lipid nanoparticles (LNPs). Although hyaluronidase had been used to enhance subcutaneous delivery of small molecule and protein drugs, it was uncertain prior to the inventors’ recent investigations if hyaluronidase could also be effective in facilitating subcutaneous delivery of mRNA, in particular, mRNA encapsulated in lipid nanoparticles (LNPs), in view of the significant size differences and the complexity of the LNP-mRNA formulations. Many mRNA-loaded LNPs have sizes close to or around about 100 nM, which is at least five times as large as a typical protein (typical proteins including antibodies have an average size below 20 nm). It was further uncertain whether delivery of mRNA-LNPs in presence of an mRNA encoding hyaluronidase could be effective in augmenting subcutaneous uptake and delivery of mRNA-LNPs. In view of efficient mRNA delivery and high protein expression in the liver following subcutaneous delivery using hyaluronidase enzyme, which was recently reported for the first time in the Applicant’s application PCT/US 17/61176, filed on 11- 10-2017, hereby fully incorporated by reference), the present invention is particularly useful in treating metabolic diseases such as ornithine transcarbamylase (OTC) deficiency. Using an mRNA encoding a hyaluronidase in the same or a separate formulation to deliver a therapeutic mRNA, a robust and sustained delivery and distribution of the therapeutic mRNA can be achieved with surprising ease and cost-effectiveness. Without wishing to be bound by a theory, it is likely that the mRNA encoding hyaluronidase is readily distributed and translated at the site of administration and in turn helps in uptake and efficient distribution of the therapeutic mRNA as a result of the function of the translated hyaluronidase in situ. The hyaluronidase based administration as provided in the present application increases the efficiency of subcutaneous delivery of mRNA, which is more patient friendly compared to other administration routes such as intravenous (IV) or intramuscular (IM), can reduce healthcare costs and increase patient compliance and throughput at the hospital.

[0005] In one aspect, the present invention provides a method for subcutaneous delivery of a messenger RNA (mRNA) to a subject in need thereof, the method comprising: administering subcutaneously to the subject a composition comprising: an mRNA encoding a protein or polypeptide, and an mRNA encoding a hyaluronidase.

[0006] In some embodiments, the mRNA encoding a protein or polypeptide is a therapeutic mRNA. In some embodiments, the protein or polypeptide encoded by the mRNA, i.e. the therapeutic mRNA as described herein, encodes a protein or polypeptide selected from a group consisting of: erythropoietin (EPO), Phenylalanine hydroxylase (PAH), argininosuccinate synthase 1 (ASS1), ocl-anti-trypsin (A1AT), Factor IX (FIX ), Factor VIII (FVIII),

carboxypeptidase N, alpha galactosidase (GFA), ornithine carbamoyltransferase (OTC), human growth hormone (hOtt), SLC3A1 encoded protein, SLC3A9 encoded protein, COL4A5 encoded protein, FXN encoded protein, GNS encoded protein, HGSNAT encoded protein, NAGLU encoded protein, SGSH encoded protein, MUT encoded protein methyl malonyl CoA mutase and ATP7B encoded protein ATPase 2.

[0007] In some embodiments, the mRNA encoding a protein or a polypeptide, which is a therapeutic mRNA, has a length of or greater than about 0.5 kb, 1 kb, 1.5 kb, 2 kb, 2.5 kb, 3 kb, 3.5 kb, 4 kb, 4.5 kb, 5 kb, 6kb, 7 kb, 8 kb, 9 kb, 10 kb, 11 kb, 12 kb, 13 kb, 14 kb, or 15 kb.

[0008] In some embodiments, the mRNA encoding hyaluronidase is a helper mRNA, which encodes a mammalian hyaluronidase selected from a bovine hyaluronidase, a porcine hyaluronidase, an equine hyaluronidase, an ovine hyaluronidase and a human hyaluronidase.

[0009] In some embodiments, the mRNA encoding the hyaluronidase comprises a polynucleotide sequence having at least 80% identity to SEQ ID NO: 9, 10 or 12.

[0010] In some embodiments the mRNA encoding the protein or polypeptide and the mRNA encoding a hyaluronidase enzyme are individually capped and tailed.

[0011] In some embodiments the mRNA encoding the protein or polypeptide and the mRNA encoding a hyaluronidase enzyme are encapsulated in a lipid nanoparticles (FNPs).

[0012] In some embodiments, the lipid nanoparticles comprise a cationic lipid, which is selected from a group consisting of CKK-E12 (3,6-bis(4-(bis(2- hydroxydodecyl)amino)butyl)piperazine-2,5-dione), OF-02, Target 23, Target 24, ICE,

HGT5000, HGT5001, HGT4003, DOTAP (l,2-dioleyl-3-trimethylammonium propane),

DODAP (l,2-dioleyl-3-dimethylammonium propane), DOTMA (l,2-di-0-octadecenyl-3- trimethylammonium propane), DLinDMA, DODAC, DDAB, DMRIE, DOSPA, DOGS, DODMA, DMDMA, DODAC, DLenDMA, DMRIE, CLinDMA, CpLinDMA, DMOBA, DOcarbDAP, DLinDAP, DLincarbDAP, DLinCDAP, KLin-K-DMA, DLin-K-XTC2-DMA, DLin-KC2-DMA, dialkylamino-based, imidazole-based, and guanidinium-based cationic lipids.

[0013] In some embodiments, the lipid nanoparticle comprises one or more non-cationic lipids. In some embodiments, the one or more non-cationic lipids are selected from the group consisting of DSPC (l,2-distearoyl-sn-glycero-3-phosphocholine), DPPC (l,2-dipalmitoyl-sn- glycero-3-phosphocholine), DOPE (l,2-dioleyl-sn-glycero-3-phosphoethanolamine), DOPC (l,2-dioleyl-sn-glycero-3-phosphotidylcholine) DPPE (l,2-dipalmitoyl-sn-glycero-3- phosphoethanolamine), DMPE (l,2-dimyristoyl-sn-glycero-3-phosphoethanolamine), DOPG (,2- dioleoyl-sn-glycero-3-phospho-(l'-rac-glycerol)) and combinations thereof.

[0014] In some embodiments, the liposome comprises a PEGylated lipid. In some embodiments, the PEGylated lipid constitutes at least 1%, at least 2%, at least 3%, at least 4%, at least 5%, at least 6%, at least 7%, at least 8%, at least 9%, or at least 10% of the total lipids in the liposome. In some embodiments, the PEGylated lipid constitutes at least 5% of the total lipids in the liposome. In some embodiments, the PEGylated lipid constitutes about 5% of the total lipids in the liposome. In some embodiments, the PEGylated lipid constitutes 10% or less, 9% or less, 8% or less, 7% or less, 6% or less, 5% or less, 4% or less, or 3% or less of the total lipids in the liposome. In some embodiments, the PEGylated lipid constitutes 5% or less of the total lipids in the liposome.

[0015] In some embodiments, the mRNA comprises unmodified nucleotides. In some embodiments, the mRNA comprises one or more modified nucleotides. In some embodiments, the one or more modified nucleotides comprise pseudouridine, N-l -methyl-pseudouridine, 2- aminoadenosine, 2-thiothymidine, inosine, pyrrolo-pyrimidine, 3-methyl adenosine, 5- methylcytidine, C-5 propynyl-cytidine, C-5 propynyl-uridine, 2-aminoadenosine, C5- bromouridine, C5-fluorouridine, C5-iodouridine, C5-propynyl-uridine, C5-propynyl-cytidine, C5-methylcytidine, 2-aminoadenosine, 7-deazaadenosine, 7-deazaguanosine, 8-oxoadenosine, 8- oxoguanosine, 0(6)-methylguanine, 4’thiouridine, 4’-thiocytidine, and/or 2-thiocytidine.

[0016] In some embodiments the mRNA encoding the protein or polypeptide and the mRNA encoding a hyaluronidase enzyme are encapsulated in the lipid nanoparticle (LNP). In some embodiments, the mRNA encoding the protein or polypeptide and the mRNA encoding a hyaluronidase enzyme are encapsulated in the separate LNPs. In some embodiments, the mRNA encoding the protein or polypeptide and the mRNA encoding a hyaluronidase enzyme are encapsulated in separate LNPs having non-identical compositions. [0017] In some embodiments, the therapeutic mRNA and the hyaluronidase-encoding mRNA are administered simultaneously. In some embodiments, the therapeutic mRNA and the hyaluronidase-encoding mRNA are administered sequentially. In some embodiments the hyaluronidase-encoding mRNA is administered 0.1 hours, 0.2 hours, 0.3 hours, 0.4 hours, 0.5 hours, 1 hour, 2 hours, 3 hours, 4 hours, 5 hours or 6 hours prior to administering the therapeutic mRNA composition. In some embodiments, the hyaluronidase-encoding mRNA is administered 1 hour, 2 hours, 3 hours, 4 hours, 5 hours or 6 hours prior to administering the therapeutic mRNA composition.

[0018] In some embodiments, the protein encoded by the therapeutic mRNA is expressed in the liver. In some embodiments, the protein encoded by the therapeutic mRNA is expressed in the kidney. In some embodiments, the protein encoded by the therapeutic mRNA is expressed in the lung. In some embodiments, the protein encoded by the therapeutic mRNA is detectable in the serum. In some embodiments, the expression of the protein encoded by the therapeutic mRNA is detectable at least 24 hours, 2 days, 3 days, 4 days, 5 days, 6 days, or 1 week after single administration.

[0019] In some embodiments, the therapeutic mRNA is administered at a dose of at least

0.5 mg/Kg of body weight. In some embodiments, the therapeutic mRNA is administered at a dose of about 1 mg/Kg, about 2 mg/Kg, about 3 mg/Kg, about 4 mg/Kg, about 5 mg/Kg, about 6 mg/Kg, about 7 mg/Kg, about 8 mg/Kg, about 9 mg/Kg, about 10 mg/Kg, about 11 mg/Kg, about 12 mg/Kg, about 13 mg/Kg, about 14 mg/Kg, about 15 mg/Kg, about 16 mg/Kg, about 17 mg/Kg, about 18 mg/Kg, about 19 mg/Kg, about 20 mg/Kg, about 25 mg/Kg, about 30 mg/Kg or about 50 mg/Kg of body weight.

[0020] In some embodiments, about 0.1 - 100 mg of mRNA encoding the hyaluronidase is administered. In some embodiments, about 0.5 - 90 mg of mRNA encoding the hyaluronidase is administered. In some embodiments, about 1- 80 mg of mRNA encoding the hyaluronidase is administered. In some embodiments, about 2- 70 mg of mRNA encoding the hyaluronidase is administered. In some embodiments, about 3- 60 mg of mRNA encoding the hyaluronidase is administered. In some embodiments, about 4- 50 mg of mRNA encoding the hyaluronidase is administered. In some embodiments, about 5- 50 mg of mRNA encoding the hyaluronidase is administered.

[0021] In some embodiments, the mRNA encoding the hyaluronidase is administered at a dose amount equivalent for translating to produce an expected amount of at least about 1U hyaluronidase enzyme per mg of the therapeutic RNA to be delivered. In some embodiments, hyaluronidase mRNA is administered at a dose equivalent of at least 2U per mg of the therapeutic RNA, at least 5U per mg of the therapeutic RNA, at least 10U per mg of the therapeutic RNA, at least 20U per mg of the therapeutic mRNA, at least 30U per mg of the therapeutic mRNA, at least 40U per mg of the therapeutic mRNA, at least 50U per mg of the therapeutic mRNA, at least 100U per mg of the therapeutic mRNA, at least 200U per mg of the therapeutic mRNA, at least 300U per mg of the therapeutic mRNA, at least 400U per mg of the therapeutic mRNA, at least 500U per mg of the therapeutic mRNA, at least 1000U per mg of the therapeutic RNA, at least 2000U per mg of the therapeutic RNA, at least 3000U per mg of the therapeutic RNA, at least 4000U per mg of the therapeutic RNA, or at least 5000U per mg of the therapeutic RNA delivered. In one aspect, the present invention provides a method for treating a disease, disorder or condition in a subject, comprising delivering subcutaneously to the subject a therapeutic mRNA encoding a protein or a polypeptide, and a helper mRNA encoding a hyaluronidase, wherein the therapeutic mRNA-encoded protein or polypeptide is deficient in the subject. The disease, disorder or condition herein is selected from ornithine transcarbamylase (OTC) deficiency, Phenylalanine hydroxylase (PAH) deficiency (phenylketonuria, PKU), arginino succinate synthase 1 (ASS1) deficiency, erythropoietin (EPO) deficiency, Fabry disease; hemophilic diseases (such as, e.g., hemophilia B (FIX), hemophilia A (FVIII); SMN1- related spinal muscular atrophy (SMA); amyotrophic lateral sclerosis (AFS); GAFT -related galactosemia; COF4A5 -related disorders including Alport syndrome; galactocerebrosidase deficiencies; X-linked adrenoleukodystrophy; Friedreich’s ataxia; Pelizaeus-Merzbacher disease; TSC1 and TSC2-related tuberous sclerosis; Sanfilippo B syndrome (MPS MB); the FMR1- related disorders which include Fragile X syndrome, Fragile X-Associated Tremor/ Ataxia Syndrome and Fragile X Premature Ovarian Failure Syndrome; Prader-Willi syndrome;

hereditary hemorrhagic telangiectasia (AT); Niemann-Pick disease Type Cl; the neuronal ceroid lipofuscinoses-related diseases including Juvenile Neuronal Ceroid Fipofuscinosis (JNCF), Juvenile Batten disease, Santavuori-Haltia disease, Jansky-Bielschowsky disease, and PTT-l and TPP1 deficiencies; EIF2B1, EIF2B2, EIF2B3, EIF2B4 and EIF2B5-related childhood ataxia with central nervous system hypomyelination/vanishing white matter; CACNA1A and

CACNB4-related Episodic Ataxia Type 2; the MECP2-related disorders including Classic Rett Syndrome, MECP2-related Severe Neonatal Encephalopathy and PPM-X Syndrome; CDKF5- related Atypical Rett Syndrome; Kennedy’s disease (SBMA); Notch-3 related cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy

(CADASIF); SCN1A and SCNlB-related seizure disorders; the Polymerase G-related disorders which include Alpers-Huttenlocher syndrome, POFG-related sensory ataxic neuropathy, dysarthria, and ophthalmoparesis, and autosomal dominant and recessive progressive external ophthalmoplegia with mitochondrial DNA deletions; X-Linked adrenal hypoplasia; X-linked agammaglobulinemia; and Wilson’s disease.

[0022] In one embodiment, the present disclosure provides a method of treating ornithine transcarbamylase (OTC deficiency) by mRNA therapy. The method comprises administering to a subject in need of treatment a composition for subcutaneous delivery comprising messenger RNA encoding OTC protein and an mRNA encoding a hyaluronidase enzyme.

[0023] In some embodiments, the OTC mRNA is encapsulated within a nanoparticle. In some embodiments, the nanoparticle is a lipid-based or polymer-based nanoparticle. In some embodiments, the lipid-based nanoparticle is a liposome.

[0024] In some embodiments, the subcutaneous injection results in expression of the

OTC protein in the liver of the subject.

[0025] In some embodiments, the subcutaneous injection delivers mRNA to hepatocytes.

In some embodiments, the subcutaneous injection results in OTC expression in hepatocytes.

[0026] In some embodiments, the subcutaneous injection results in expression of the

OTC protein in the serum of the subject.

[0027] In some embodiments, the expression of the protein encoded by the mRNA is detectable at least 24 hours, 2 days, 3 days, 4 days, 5 days, 6 days, 1 week, 2 weeks, 3 weeks, 4 weeks, or 1 month post-administration.

[0028] In some embodiments, OTC expression after mRNA administration can be detected by a functional assay.

[0029] In some embodiments, the administering of the composition results in an increased OTC protein expression or activity level in serum of the subject as compared to a control level. In some embodiments, the control level is a baseline serum OTC protein expression or activity level in the subject prior to the treatment. In some embodiments, the control level is a reference level indicative of the average serum OTC protein expression or activity level in OTC patients without treatment.

[0030] In some embodiments, the administering of the composition results in a reduced urinary orotic acid level in the subject as compared to a control orotic acid level. In some embodiments, the control orotic acid level is a baseline urinary orotic acid level in the subject prior to the treatment. In some embodiments, the control orotic acid level is a reference level indicative of the average urinary orotic acid level in OTC patients without treatment.

[0031] In some embodiments, wherein the administering of the composition results in an increased citrulline level in serum of the subject as compared to a control citrulline level. In some embodiments, the control citrulline level is a baseline serum citrulline level in the subject prior to the treatment. In some embodiments, the control citrulline level is a reference level indicative of the average serum citrulline level in OTC patients without treatment.

[0032] In some embodiments, the mRNA encoding the OTC protein and the mRNA encoding the hyaluronidase enzyme are injected simultaneously.

[0033] In some embodiments, the mRNA encoding the OTC protein and the mRNA encoding the hyaluronidase enzyme are injected in one composition.

[0034] In some embodiments, the mRNA encoding the OTC protein and the mRNA encoding the hyaluronidase enzyme are injected in separate compositions.

[0035] In some embodiments, the mRNA encoding the OTC protein and the mRNA encoding the hyaluronidase enzyme are injected sequentially.

[0036] In some embodiments, the mRNA encoding the OTC protein and the mRNA encoding the hyaluronidase enzyme are injected in a volume of less than 20 ml, less than 15 ml, less than 10 ml, less than 5ml, less than 4 ml, less than 3 ml, less than 2 ml, or less than 1 ml.

[0037] In some embodiments, the subcutaneous injection is performed once a week or less frequently. In some embodiments, the subcutaneous injection is performed twice a month or less frequently. In some embodiments, the subcutaneous injection is performed once a month or less frequently.

[0038] In another aspect, the present invention provides for a composition for treating ornithine transcarbamylase (OTC deficiency), comprising an mRNA encoding an ornithine transcarbamylase (OTC) protein, and an mRNA encoding a hyaluronidase enzyme.

[0039] In some embodiments, the mRNA encoding hyaluronidase enzyme is

administered at a dose 20 mg/mL or less. In some embodiments, the mRNA encoding hyaluronidase enzyme is administered at a dose 18 mg/mL or less. In some embodiments, the mRNA encoding hyaluronidase enzyme is administered at a dose 16 mg/mL or less. In some embodiments, the mRNA encoding hyaluronidase enzyme is administered at a dose 14 mg/mL or less. In some embodiments, the mRNA encoding hyaluronidase enzyme is administered at a dose 12 mg/mL or less. In some embodiments, the mRNA encoding hyaluronidase enzyme is administered at a dose 10 mg/mL or less. In some embodiments, the mRNA encoding hyaluronidase enzyme is administered at a dose 9 mg/mL or less. In some embodiments, the mRNA encoding hyaluronidase enzyme is administered at a dose 8 mg/mL or less. In some embodiments, the mRNA encoding hyaluronidase enzyme is administered at a dose 7 mg/mL or less. In some embodiments, the mRNA encoding hyaluronidase enzyme is administered at a dose 6 mg/mL or less. In some embodiments, the mRNA encoding hyaluronidase enzyme is administered at a dose 5 mg/mL or less. In some embodiments, the mRNA encoding hyaluronidase enzyme is administered at a dose 4 mg/mL or less. In some embodiments, the mRNA encoding hyaluronidase enzyme is administered at a dose 3 mg/mL or less. In some embodiments, the mRNA encoding hyaluronidase enzyme is administered at a dose 2 mg/mL or less. In some embodiments, the mRNA encoding hyaluronidase enzyme is administered at a dose 1 mg/mL or less. In some embodiments, the mRNA encoding hyaluronidase enzyme is administered at a dose ranging between 1- 20 mg/mL.

[0040] In some embodiments, the mRNA is encapsulated within a nanoparticle.

[0041] In some embodiments, the nanoparticle is a lipid-based or polymer-based nanoparticle.

[0042] In some embodiments, the composition is a liquid form.

[0043] In another embodiment the composition is a lyophilized powder.

[0044] In one aspect, the invention provides a container containing a composition described above. The container is a vial or a syringe. The syringe may be prefilled for single subcutaneous administration. The vial may contain lyophilized powder or liquid form of the composition.

[0045] In this application, the use of“or” means“and/or” unless stated otherwise. As used in this disclosure, the term“comprise” and variations of the term, such as“comprising” and “comprises,” are not intended to exclude other additives, components, integers or steps. As used in this application, the terms“about” and“approximately” are used as equivalents. Both terms are meant to cover any normal fluctuations appreciated by one of ordinary skill in the relevant art.

[0046] Other features, objects, and advantages of the present invention are apparent in the detailed description, drawings and claims that follow. It should be understood, however, that the detailed description, the drawings, and the claims, while indicating embodiments of the present invention, are given by way of illustration only, not limitation. Various changes and modifications within the scope of the invention will become apparent to those skilled in the art.

DEFINITIONS

[0047] 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.

[0048] Animal·. As used herein, the term“animal” refers to any member of the animal kingdom. In some embodiments,“animal” refers to humans, at any stage of development. In some embodiments,“animal” refers to non-human animals, at any stage of development. In certain embodiments, the non-human animal is a mammal ( e.g ., a rodent, a mouse, a rat, a rabbit, a monkey, a dog, a cat, a sheep, cattle, a primate, and/or a pig). In some embodiments, animals include, but are not limited to, mammals, birds, reptiles, amphibians, fish, insects, and/or worms. In some embodiments, an animal may be a transgenic animal, genetic ally- engineered animal, and/or a clone.

[0049] 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).

[0050] 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).

[0051] Encapsulation: As used herein, the term“encapsulation,” or grammatical equivalent, refers to the process of confining an individual mRNA molecule within a

nanoparticle.

[0052] Expression: As used herein,“expression” of a nucleic acid sequence refers to translation of an mRNA into a polypeptide, assemble multiple polypeptides into an intact protein (e.g., enzyme) and/or post-translational modification of a polypeptide or fully assembled protein (e.g., enzyme). In this application, the terms“expression” and“production,” and grammatical equivalent, are used inter-changeably.

[0053] 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.

[0054] Hyaluronidase: As used herein, the term "hyaluronidase" refers to the family of enzymes that are capable of degrading hyaluronic acid (hyaluronan). [0055] 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.

[0056] In Vitro As used herein, the term“m 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.

[0057] 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).

[0058] Local distribution or delivery: As used herein, the terms“local distribution,”

“local delivery,” or grammatical equivalent, refer to tissue specific delivery or distribution.

Typically, local distribution or delivery requires a protein (e.g., enzyme) encoded by mRNAs be translated and expressed intracellularly or with limited secretion that avoids entering the patient’ s circulation system.

[0059] Messenger RNA (mRNA): As used herein, the term“messenger RNA (mRNA)” refers to a polynucleotide that encodes at least one polypeptide. mRNA as used herein encompasses both modified and unmodified RNA. mRNA may contain one or more coding and non-coding regions. mRNA can be purified from natural sources, produced using recombinant expression systems and optionally purified, chemically synthesized, etc. Where appropriate, e.g., in the case of chemically synthesized molecules, mRNA can comprise nucleoside analogs such as analogs having chemically modified bases or sugars, backbone modifications, etc. An mRNA sequence is presented in the 5’ to 3’ direction unless otherwise indicated. In some embodiments, an mRNA is or comprises natural nucleosides (e.g., adenosine, guanosine, cytidine, uridine); nucleoside analogs (e.g., 2-aminoadenosine, 2-thiothymidine, inosine, pyrrolo- pyrimidine, 3-methyl adenosine, 5-methylcytidine, C-5 propynyl-cytidine, C-5 propynyl-uridine, 2-aminoadenosine, C5-bromouridine, C5-fluorouridine, C5-iodouridine, C5-propynyl-uridine, C5-propynyl-cytidine, C5-methylcytidine, 2-aminoadenosine, 7-deazaadenosine, 7- deazaguanosine, 8-oxoadenosine, 8-oxoguanosine, 0(6)-methylguanine, and 2-thiocytidine); chemically modified bases; biologically modified bases (e.g., methylated bases); intercalated bases; modified sugars ( e.g ., 2’-fluororibose, ribose, 2’-deoxyribose, arabinose, and hexose); and/or modified phosphate groups (e.g., phosphorothioates and 5’-/V-phosphoramidite linkages).

[0060] 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.

[0061] 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.

[0062] Subcutaneous administration : As used herein, the term“subcutaneous administration” or“subcutaneous injection” refers to a bolus injection into the subcutis which is the tissue layer between the skin and the muscle.

[0063] 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.

[0064] 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.

[0065] Systemic distribution or delivery: As used herein, the terms“systemic

distribution,”“systemic delivery,” or grammatical equivalent, refer to a delivery or distribution mechanism or approach that affect the entire body or an entire organism. Typically, systemic distribution or delivery is accomplished via body’s circulation system, e.g., blood stream.

Compared to the definition of“local distribution or delivery.” [0066] 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.

[0067] Therapeutic mRNA: As used herein, the term therapeutic mRNA is used to designate the mRNA that is intended for mRNA therapy. A therapeutic mRNA may designate an mRNA which encodes a protein or polypeptide which is deficient in a subject in need for therapy. It is interchangeably used with the term‘first mRNA’ throughout the specification, without any presumption as to the temporal sequence of delivery with respect to, for example, a second mRNA.

[0068] 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.

[0069] 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.

DETAILED DESCRIPTION

[0070] The present invention provides, among other things, improved methods and compositions of mRNA delivery for messenger RNA therapy via subcutaneous route by administering the mRNA of interest (the first mRNA) with a second mRNA encoding a hyaluronidase enzyme. The second mRNA helps or augments the cellular uptake and distribution of the mRNA. The mRNA payload was efficiently delivered to the livers (and other organs or tissues) of treated animals. Such a hyaluronidase based method has major benefits to creating new delivery profiles of otherwise intolerable drugs.

[0071] Among other things, the present invention provides methods and compositions for the treatment of ornithine transcarbamylase (OTC) deficiency by administering via subcutaneous injection to a subject in need of treatment an mRNA encoding an ornithine transcarbamylase (OTC) protein and a second mRNA encoding a hyaluronidase enzyme. The invention may also be used to treat various other diseases, disorders and conditions in particular metabolic diseases, disorders and conditions.

[0072] Various aspects of the invention are described in detail in the following sections.

The use of sections is not meant to limit the invention. Each section can apply to any aspect of the invention. In this application, the use of“or” means“and/or” unless stated otherwise.

Hyaluronidase enzymes

[0073] Various hyluronidase enzymes may be used to practice the present invention. For example, there are three groups of hyluronidases based on their mechanisms of action. Two of the groups are cndo-b-Zn- acetyl -hexosaminidases. One group includes the vertebrate enzymes that utilize substrate hydrolysis. The vertebrate hyaluronidases (EC 3.2.1.35) are endo-b-/n- acetyl-hexosaminidases employing substrate hydrolysis for catalysis. The vertebrate

hyaluronans also have transglycosidase activities, with the ability to cross -link chains of HA and the potential ability to cross-link chains of HA with ChS or Ch. The vertebrate hyaluronidases degrade HA through a non-processive endolytic process, generating mostly tetrasaccharides. Mammalian hyaluronidases are members of the group of carbohydrate-active enzymes (CAZy), termed glycosidase family 56, defined as cndo^-acctyl -hexosaminidases that utilize hydrolysis in catalysis of HA at the b 1 ,4 glycosidic linkages.

[0074] The second group, which is predominantly bacterial, includes the eliminases that function by b-elimination of the glycosidic linkage with introduction of an unsaturated bond. Bacterial hyaluronidases are also endo^-acetyl-hexosaminidases, but utilize the lyase mechanism. They belong to a different CAZy family, to polysaccharide lyase family 8. In general, these polysaccharide lyases (EC 4.2.2.*) cleave by b-elimination, resulting in a double bond at the new non-reducing end. The hyaluronate lyases (EC 4.2.2.1; bacterial Hyal) consists of only one subgroup within family 8 that also include: chondroitin ABC lyases (EC 4.2.2.4), chondroitin AC lyases (EC 4.2.2.5), and xanthan lyases (EC 4.2.2.12). All of these bacterial enzymes, hyaluronidases, chondroitinases, and xanthanases, share significant sequence, structural, and mechanistic homology.

[0075] The third group is the endo^-glucuronidases. These are found in leeches, which are annelids, and in certain crustaceans.

[0076] In addition, there are six known genes coding for hyaluronidase-like sequences in human genome, Hyal-l, Hyal-2, Hyal-3, Hyal-4, and PH-20/Spaml and a pseudogene Phyall (not translated), all of which have high degree of homology. Mice also have six genes coding for hyaluronidases which have high degree of homology with human genes (Stern et ah, Chem. Rev. 2006, 106(3): 818-839). In some embodiments, hyaluronidase may also be obtained from cows or pigs as a sterile preparation which is free of any other animal substance.

[0077] Bovine PH-20 is a commonly used hyaluronidase, and is available commercially in a reasonably pure form (Sigma catalog no. H3631, Type VI-S, from bovine testes, with an activity of 3,000 to 15,000 national formulary units (NFU) units/mg).

[0078] Hyaluronidase for injection can be obtained commercially in powder form or in solution. For example, an FDA approved bovine testicular hyaluronidase enzyme is available as a colorless oderless solution.

[0079] In some embodiments, an International Unit for hyaluronidase may be defined as the activity of 0.1 mg of the International Standard Preparation, and is equal to one turbidity reducing unit (TRU) (Humphrey JH et al.,“International Standard for Hyaluronidase,” Bull World Health Organ. 1957; 16(2): 291-294) based on the following reaction:

Hyaluronidase

Hyaluronic acid - > Di and monosaccharides + smaller hyaluronic acid fragments

Accordingly, one unit of Hyaluronidase activity will cause a change in Aeon of 0.330 per minute at pH 5.3 at 37 °C in a 2.0 ml reaction mixture (45 minute assay). % Transmittance is determined at 600 nm, Light path = 1 cm.

[0080] In some embodiments, an artificially synthesized bovine hyaluronidase PH-20 mRNA may be used for the present purpose.

[0081] In some embodiments, the bovine hyaluronidase mRNA used herein has a greater than 80% sequence identity to SEQ ID NO: 9 (GenBank ID No.: BC110183.1). In some embodiments, the bovine hyaluronidase mRNA used herein has greater than 90% sequence identity to SEQ ID NO: 9. In some embodiments, the mRNA has a sequence identity of greater than 91%, greater than 92%, greater than 93%, greater than 94%, greater than 95%, or greater than 98% sequence identity to SEQ ID NO: 9. In some embodiments, the bovine hyaluronidase mRNA used herein has 100% identity to SEQ ID NO: 9. In some embodiments the bovine hyaluronidase mRNA encodes for a PH-20 hyaluronidase which is about 90% identical to SEQ

ID NO: 10 (GenBank ID No.: BC110183.1, cds sequence). In some embodiments, the mRNA encoded PH-20 hyaluronidase has a sequence identity of greater than 91%, greater than 92%, greater than 93%, greater than 94%, greater than 95%, or greater than 98% sequence identity the sequence of SEQ ID NO: 10. In some embodiments, the bovine hyaluronidase has 100% identity to SEQ ID NO: 10. In some embodiments, the bovine hyaluronidase mRNA encodes a protein which has an amino acid sequence having at least about 90% sequence identity with that of SEQ ID NO: 11. (GenBank ID No.: AAI10184.1). In some embodiments, the mRNA encodes a protein having amino acid sequence identity of greater than 91%, greater than 92%, greater than 93%, greater than 94%, greater than 95%, or greater than 98% sequence identity to SEQ ID NO: 11.

An exemplary bovine hyaluronidase mRNA sequence is given below:

GGTTTATCTCTGTTCTTGGTGAGGAGACAGACAGAATTGACTGCTGTGCTCATCCGC

GAGGGTAAATGTG CTCAGCTCTT TATGGAGTAGTGGAGACGGGCAGAGATGACAA

GAT G A AGC A ACTTGC A A A AC ATT CCT A A AT AC G A AGG A AG A AG A AT ATTT A A AT G A

AATCATCATTATTCATTTTTATCCATCAAAG TGGCTTCATTCTGTGTTCATATCTTGC

ATCAAATATTAGGTACACCAAAGCGTGTAGGAG AAAAAAGTGCCTTTCACAGTCAT

CGCTCTTTGTGATGAGAATGCTGAGGC GCCACCATAT CTCCTTTCGGAGCTTTGCTG

GGTCTAGCGGAACACCCCAGGCAGTGTTCACCTTCCTTCTGCTTCCGTGTTGTTTGG C

TCTGGACTTCAGAGCACCCCCTCTTATTTCAAACACTTCTTTCCTCTGGGCCTGGAA T

GCCCCAGTTGAACGTTGTGTTAACAGAAGATTTCAACTACCTCCAGATCTGAGACTC

TTCTCTGTAAAAGGAAGCCCCCAGAAAAGTGCTACCGGACAATTTATTACATTATTT

T ATGCTGAT AGACTTGGCT ACT ATCCTC AT ATAGATGAAA AAACAGGCAAAACCGT

ATTCGGAGGAATTCCCCAGTTGGGAAACTTAAAAAGTCATATGGAGAAAGCAAAAA

ATGACATTGCCTATTACATACCAAATGACAGCGTGGGCTTGGCGGTCATTGACTGGG

AAAACTGGAGGCCTACCTGGGCAAGAAACTGGAAACCTAAAGATGTTTACAGGGAT

GAGTCAGTTGAGTTGGTTCTGCAAAAAAATCCGCAACTCAGTTTCCCAGAGGCTTCC

AAGATTGCAAAAGTGGATTTTGAGACAGCAGGAAAGAGTTTCATGCAAGAGACTTT

AAAACTGGGAAAATTACTTCGGCCAAATCACTTATGGGGTTATTATCTTTTTCCTGA

TTGTTACAATCATAATCATAACCAACCTACTTACAATGGAAATTGCCCTGATGTGAA

AAAAGGAGAAATGATGATCTCGAGTGGTTGTGGAAGGAAAGCACTGCCCTTTTCCC

TTCTGTTTATTTGAATATCAGGTTAAAATCTACTCAAAATGCTGCCTTGTATGTTCG T

AATCGTGTCCAGGAAGCCATTCGGTTGTCTAAAATAGCGAGTGTCGAAAGTCCACTT

CCGGTTTTTGTATATGCCCGTCCAGTTTTTACTGATGGGTCTTCAACATATCTTTCT C

AGGGTGACCTTGTGAATTCGGTTGGTGAGATCGTTTCTCTAGGTGCCTCTGGGATTA

TAATGTGGGGCAGTCTCAATCTAAGCTTATCTATGCAATCTTGCATGAACCTAGGCA

CT

TACTTGAACACTACACTGAATCCTTACATAATCAACGTCACCCTAGCCGCCAAAATG TGCAG CCAAGTGCTTTGCCACAATGAAGGAGTGTGTACAAGGAAACACTGGAATTC AAGCGACTATCTTCACCTGAACCCAATGAATTTTGCTATTCAAACTGGGGAAGGTGG

AAAATACACAGTACCTGGGACAGTCACACTTGAAGACTTGCAAAAGTTTTCTGATAC

ATTTTATTGCAGTTGTTATGCCAACATCCACTGTAAGAAGAGAGTTGATATAAAAAA

TGTTCATAGTGTTAACGTGTGTATGGCAGAAGACATTTGTATAGACAGCCCTGTGAA

GTTACAACCCAGTGATCATTCCTCCAGCCAGGAGGCATCTACTACCACCTTCAGCAG

TATCTCACCCTCCACTACAACTGCCACAGTATCTCCATGTACTCCTGAGAAACACTC

CCCTGAGTGCCTCAAAGTCAGGTGTTCGGAAGTCATCCCCAACGTCACCCAAAAGG

CGTGTCAAAGTGTTAAATTGAAGAACATTTCCTATCAGTCACCTATTCAAAATATTA

A A AAT C A A AC A ACCT ATT A A A ATT A A ATT C AGT A A A A A A A A A A A A A A A A A A A A A A

AAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAA

AAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAGAAAAAAA

AAAAAAAA (SEQ ID NO: 9)

Another exemplary bovine hyaluronidase mRNA sequence is given below:

ATGAGAATGCTGAGGCGCCACCATATCTCCTTTCGGAGCTTTGCTGGGTCTAGCGG

AACACCCCAGGCAGTGTTCACCTTCCTTCTGCTTCCGTGTTGTTTGGCTCTGGACTT C

AGAGCACCCCCTCTTATTTCAAACACTTCTTTCCTCTGGGCCTGGAATGCCCCAGTT G

AACGTTGTGTTAACAGAAGATTTCAACTACCTCCAGATCTGAGACTCTTCTCTGTAA

AAGGAAGCCCCCAGAAAAGTGCTACCGGACAATTTATTACATTATTTTATGCTGATA

GACTTGGCTACTATCCTCATATAGATGAAAAAACAGGCAAAACCGTATTCGGAGGA

ATTCCCCAGTTGGGAAACTTAAAAAGTCATATGGAGAAAGCAAAAAATGACATTGC

CTATTACATACCAAATGACAGCGTGGGCTTGGCGGTCATTGACTGGGAAAACTGGA

GGCCTACCTGGGCAAGAAACTGGAAACCTAAAGATGTTTACAGGGATGAGTCAGTT

GAGTTGGTTCTGCAAAAAAATCCGCAACTCAGTTTCCCAGAGGCTTCCAAGATTGCA

AAAGTGGATTTTGAGACAGCAGGAAAGAGTTTCATGCAAGAGACTTTAAAACTGGG

AAAATTACTTCGGCCAAATCACTTATGGGGTTATTATCTTTTTCCTGATTGTT

ACAATCATAATCATAACCAACCTACTTACAATGGAAATTGCCCTGATGTAGAAAAA

AGGAGAAATGATGATCTCGAGTGGTTGTGGAAGGAAAGCACTGCCCTTTTCCCTTCT

GTTTATTTGAATATCAGGTTAAAATCTACTCAAAATGCTGCCTTGTATGTTCGTAAT C

GTGTCCAGGAAGCCATTCGGTTGTCTAAAATAGCGAGTGTCGAAAGTCCACTTCCGG

TTTTTGTATATGCCCGTCCAGTTTTTACTGATGGGTCTTCAACATATCTTTCTCAGG G

TGACCTTGTGAATTCGGTTGGTGAGATCGTTTCTCTAGGTGCCTCTGGGATTATAAT G

TGGGGCAGTCTCAATCTAAGCTTATCTATGCAATCTTGCATGAACCTAGGCACTTAC

TTGAACACTACACTGAATCCTTACATAATCAACGTCACCCTAGCCGCCAAAATGTGC AGCCAAGTGCTTTGCCACAATGAAGGAGTGTGTACAAGGAAACACTGGAATTCAAG

CGACTATCTTCACCTGAACCCAATGAATTTTGCTATTCAAACTGGGGAAGGTGGAAA

ATACACAGTACCTGGGACAGTCACACTTGAAGACTTGCAAAAGTTTTCTGATACATT

TTATTGCAGTTGTTATGCCAACATCCACTGTAAGAAGAGAGTTGATATAAAAAATGT

TCATAGTGTTAACGTGTGTATGGCAGAAGACATTTGTATAGACAGCCCTGTGAAGTT

ACAACCCAGTGATCATTCCTCCAGCCAGGAGGCATCTACTACCACCTTCAGCAGTAT

CTCACCCTCCACTACAACTGCCACAGTATCTCCATGTACTCCTGAGAAACACTCCCC

TGAGTGCCTCAAAGTCAGGTGTTCGGAAGTCATCCCCAACGTCACCCAAAAGGCGT

GTCAAAGTGTTAAATTGAAGAACATTTCCTATCAGTCACCTATTCAAAATATTAAAA

ATCAAACAACCTATTA (SEQ ID NO: 10)

An exemplary translated protein sequence is:

MRMLRRHHIS FRS F AGS S GTPQ A VFTFLLLPCCLALDFRAPPLIS NT S FLW AWN AP VERC VNRRF QLPPDLRLF S VKGS PQKS ATGQFITLF Y ADRLG Y YPHIDEKTGKT VF GGIPQLGN LKSHMEKAKNDIAYYIPN DS V GLA VID WEN WRPTW ARNWKPKD V YRDES VELVLQK NPQLSFPEAS KIAKVDFETAGKS FMQETLKLGKLLRPNHLW GY YLFPDC YNHNHN QPT YN GNCPD VEKRRNDDLEWLWKES T ALFPS V YLNIRLKS TQN A AL Y VRNR V QE AIRLS KI AS VES PLP VFV Y ARP VFTDGS S T YLS QGDLVN S V GEIV S LG AS GIIMW GS LNLS LS MQS C MNLGTYLNTTLNPYIINVTLAAKMCSQVLCHNEGVCTRKHWNSSDYLHLNPMNFAIQT GEGGKYTVPGTVTLEDLQKFSDTFYCSCYANIHCKKRVDIKNVHSVNVCMAEDICIDSP VKLQPS DHS S S QE AS TTTFS S IS PS TTT AT V S PCTPEKHS PECLK VRC S E VIPN VTQKAC QS VKLKNIS YQS PIQNIKN QTT Y (SEQ ID NO: 11).

[0082] In some embodiments, an artificially synthesized human hyaluronidase mRNA is administered for subcutaneous delivery of a therapeutic mRNA. The human hyaluronidase mRNA administered for subcutaneous delivery of a therapeutic mRNA has greater than 80% sequence identity to SEQ ID NO: 12 (GenBank ID No: AF040710). In some embodiments, the human hyaluronidase mRNA used herein has greater than 90% sequence identity to SEQ ID NO: 12. In some embodiments, the mRNA has a sequence identity of greater than 91%, greater than 92%, greater than 93%, greater than 94%, greater than 95%, or greater than 98% sequence identity to SEQ ID NO: 12. In some embodiments, the human hyaluronidase mRNA used herein has 100% identity to SEQ ID NO: 12. In some embodiments, the human hyaluronidase mRNA encodes a protein which has an amino acid sequence having at least about 90% sequence identity with that of SEQ ID NO: 13. (GenBank ID No: AAC70915.1). In some embodiments, the mRNA encodes a protein having amino acid sequence identity of greater than 91%, greater than 92%, greater than 93%, greater than 94%, greater than 95%, or greater than 98% sequence identity to SEQ ID NO: 13.

An exemplary human hyaluronidase mRNA sequence is given below:

ATGACCACGCAACTGGGCCCAGCCCTGGTGCTGGGGGTGGCCCTGTGCCTGGGTTGT

GGCCAGCCCCTACCACAGGTCCCTGAACGCCCCTTCTCTGTGCTGTGGAATGTACCC

TCAGCACACTGTGAGGCCCGCTTTGGTGTGCACCTGCCACTCAATGCTCTGGGCATC

ATAGCCAACCGTGGCCAGCATTTTCACGGTCAGAACATGACCATTTTCTACAAGAAC

CAACTCGGCCTCTATCCCTACTTTGGACCCAGGGGCACAGCTCACAATGGGGGCATC

CCCCAGGCTTTGCCCCTTGACCGCCACCTGGCACTGGCTGCCTACCAGATCCACCAC

AGCCTGAGACCTGGCTTTGCTGGCCCAGCAGTGCTGGATTGGGAGGAGTGGTGTCC

ACTCTGGGCTGGGAACTGGGGCCGCCGCCGAGCTTATCAGGCAGCCTCTTGGGCTTG

GGCACAGCAGGTATTCCCTGACCTGGACCCTCAGGAGCAGCTCTACAAGGCCTATA

CTGGCTTTGAGCAGGCGGCCCGTGCACTGATGGAGGATACGCTGCGGGTGGCCCAG

GCACTACGGCCCCATGGACTCTGGGGCTTCTATCACTACCCAGCCTGTGGCAATGGC

TGGCATAGTATGGCTTCCAACTATACCGGCCGCTGCCATGCAGCCACCCTTGCCCGC

AACACTCAACTGCATTGGCTCTGGGCCGCCTCCAGTGCCCTCTTCCCCAGCATCTAC

CTCCCACCCAGGCTGCCACCTGCCCACCACCAGGCCTTTGTCCGACATCGCCTGGAG

GAGGCCTTCCGTGTGGCCCTTGTTGGGCACCGACATCCCCTGCCTGTCCTGGCCTAT

GTCCGCCTCACACACCGGAGATCTGGGAGGTTCCTGTCCCAGGATGACCTTGTGCAG

TCCATTGGTGTGAGTGCAGCACTAGGGGCAGCCGGCGTGGTGCTCTGGGGGGACCT

GAGCCTCTCCAGCTCTGAGGAGGAGTGCTGGCATCTCCATGACTACCTGGTGGACAC

CTTGGGCCCCTATGTGATCAATGTGACCAGGGCAGCGATGGCCTGCAGTCACCAGC

GGTGCCATGGCCACGGGCGCTGTGCCCGGCGAGATCCAGGACAGATGGAAGCCTTT

CTACACCTGTGGCCAGACGGCAGCCTTGGAGATTGGAAGTCCTTCAGCTGCCACTGT

TACTGGGGCTGGGCTGGCCCCACCTGCCAGGAGCCCAGCCTGGGCCTAAAGAAGCA

GTATAAAGCCAGGGCCCCTGCCACTGCCTCTTCTTTTCCCTGCTGCCACTTTTCCAG T

CCTGGAACTACTCTGTCCCACTCTTGCTCTATTCAGTTTACAGTCAACCCTCCCAAG C

ACACACCCCGCTTCCCTTGGAATCCCTGA (SEQ ID NO: 12)

An exemplary human hyaluronidase protein sequence is given below:

MTTQLGP ALVLG V ALCLGC GQPLPQ VPERPF S VLWN VPS AHCE ARF G VHLPLN ALGIIA NRGQHFHGQNMTIFYKNQLGLYPYFGPRGTAHNGGIPQALPLDRHLALAAYQIHHSLRP GFAGPAVLDWEEWCPLWAGNWGRRRAYQAASWAWAQQVFPDLDPQEQLYKAYTGF EQAARALMEDTLRVAQALRPHGLWGFYHYPACGNGWHSMASNYTGRCHAATLARNT QLHWLW A AS S ALFPS IYLPPRLPP AHHQ AFVRHRLEE AFRV ALV GHRHPLP VLA Y VRLT HRRS GRFLS QDDLVQS IG V S A ALG A AG V VLW GDLS LS S S EEEC WHLHD YLVDTLGP Y VI NVTRAAMACSHQRCHGHGRCARRDPGQMEAFLHLWPDGSLGDWKSFSCHCYWGWA GPTCQEPSLGLKKQYKARAPATASSFPCCHFSSPGTTLSHSCSIQFTVNPPKHTPRFPWN P

(SEQ ID NO: 13)

[0083] In some embodiments, an mRNA encoding the full length or a fragment of the hyaluronidase is used.

[0084] An exemplary recombinant hyaluronidase dose of hyaluronidase is about 1 Unit to 50,000 Units. Accordingly, the hyaluronidase mRNA is administered at a dose equivalent so as to translate to a protein of the amount of less than 40,000U, less than 30,000U, less than 20,000U, less than l0,000U, less than 9000U, less than 8000U, less than 7000U, less than 6000U, less than 5000U less than 4000U, less than 3000U, less than 2000U, less than 1000U, less than 900U, less than 800U, less than 700U, less than 600U, or less than 500U. In some embodiments, the hyaluronidase mRNA is administered at a dose equivalent so as to translate to a protein of the amount of at least 1U, at least 5U, at least 10U, at least 20U, at least 30U, at least 40U, at least 50U, at least 60U, at least 70U, at least 80U, at least 100U, or at least 150U. In some other embodiments, the hyaluronidase mRNA is administered at a dose equivalent so as to translate to a protein of the amount of at least 160U, at least 180U, at least 200U, at least 220U, at least 240U, at least 260U, at least 280U, at least 300U, at least 320U, at least 340U, at least 360U, at least 380U, or at least 400U. In one or more embodiments, a porcine (pig)

hyaluronidase is used at a dose ranging between 1-50,000 Units. The hyaluronidase mRNA is administered at a dose equivalent so as to translate to a protein of the amount of less than 40,000U, less than 30,000U, less than 20,000U, less than l0,000U, less than 9000U, less than 8000U, less than 7000U, less than 6000U, less than 5000U less than 4000U, less than 3000U, less than 2000U, less than 1000U, less than 900U, less than 800U, less than 700U, less than 600U, or less than 500U. The method of any one of the preceding claims, wherein the hyaluronidase mRNA is administered at a dose equivalent so as to translate to a protein of the of at least 1U, at least 5U, at least 10U, at least 20U, at least 30U, at least 40U, at least 50U, at least 60U, at least 70U, at least 80U, at least 100U, or at least 150U. In some other embodiments, the hyaluronidase mRNA is administered at a dose equivalent so as to translate to a protein of the amount of at least 160U, at least 180U, at least 200U, at least 220U, at least 240U, at least 260U, at least 280U, at least 300U, at least 320U, at least 340U, at least 360U, at least 380U, or at least 400U.

[0085] In one or more embodiments, hyaluronidase mRNA is administered

simultaneously with the therapeutic mRNA. In some embodiments, hyaluronidase may be administered prior to the administration of the mRNA. In some embodiments, the mRNA and the hyaluronidase enzyme are part of the same formulation. In some embodiments, the RNA and the hyaluronidase enzyme are injected as separate formulations.

[0086] In some embodiments, the mRNA encoding hyaluronidase may be administered in an aqueous solution. In some embodiments, the mRNA encoding hyaluronidase in saline solution. In some embodiments the hyaluronidase enzyme is part of the mRNA formulation and is present in the same solution, the solution comprising mRNA-encapsulated lipid nanoparticles. In some embodiments a lyophilized preparation comprising the mRNA-encapsulated lipid and the hyaluronidase enzyme is formulated for therapeutic use.

Messenger RN A (mRNA)

[0087] The present invention may be used to deliver any mRNA. As used herein, mRNA is the type of RNA that carries information from DNA to the ribosome for translation of the encoded protein. mRNAs 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), DNAsel, pyrophosphatase, and/or RNAse inhibitor. The exact conditions will vary according to the specific application.

[0088] In some embodiments, in vitro synthesized mRNA may be purified before formulation and encapsulation to remove undesirable impurities including various enzymes and other reagents used during mRNA synthesis.

[0089] The present invention may be used to deliver mRNAs of a variety of lengths. In some embodiments, the present invention may be used to deliver in vitro synthesized mRNA of or greater than about 1 kb, 1.5 kb, 2 kb, 2.5 kb, 3 kb, 3.5 kb, 4 kb, 4.5 kb, 5 kb 6 kb, 7 kb, 8 kb, 9 kb, 10 kb, 11 kb, 12 kb, 13 kb, 14 kb, 15 kb, or 20 kb in length. In some embodiments, the present invention may be used to deliver in vitro synthesized mRNA ranging from about 1-20 kb, about 1-15 kb, about 1-10 kb, about 5-20 kb, about 5-15 kb, about 5-12 kb, about 5-10 kb, about 8-20 kb, or about 8-15 kb in length. [0090] The present invention may be used to deliver mRNA that is unmodified or mRNA containing one or more modifications that typically enhance stability. In some embodiments, modifications are selected from modified nucleotides, modified sugar phosphate backbones, and 5’ and/or 3’ untranslated region (UTR).

[0091] In some embodiments, modifications of mRNA may include modifications of the nucleotides of the RNA. A modified mRNA according to the invention can include, 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, l-methyl- inosine, pseudouracil (5-uracil), dihydrouracil, 2-thio-uracil, 4-thio-uracil, 5- carboxymethylaminomethyl-2-thio-uracil, 5-(carboxyhydroxymethyl)-uracil, 5-fluoro-uracil, 5- bromo-uracil, 5-carboxymethylaminomethyl-uracil, 5-methyl-2-thio-uracil, 5-methyl-uracil, N- uracil-5-oxyacetic acid methyl ester, 5-methylaminomethyl-uracil, 5-methoxyaminomethyl-2- thio-uracil, 5'-methoxycarbonylmethyl-uracil, 5-methoxy-uracil, uracil-5-oxyacetic acid methyl ester, uracil- 5 -oxy acetic acid (v), 1 -methyl-pseudouracil, queosine, .beta.-D-mannosyl-queosine, wybutoxosine, and phosphoramidates, phosphorothioates, peptide nucleotides,

methylphosphonates, 7-deazaguanosine, 5-methylcytosine and inosine. The preparation of such analogues is known to a person skilled in the art e.g. from the U.S. Pat. No. 4,373,071, U.S. Pat. No. 4,401,796, U.S. Pat. No. 4,415,732, U.S. Pat. No. 4,458,066, U.S. Pat. No. 4,500,707, U.S. Pat. No. 4,668,777, U.S. Pat. No. 4,973,679, U.S. Pat. No. 5,047,524, U.S. Pat. No. 5,132,418, U.S. Pat. No. 5,153,319, U.S. Pat. Nos. 5,262,530 and 5,700,642, the disclosure of which is included here in its full scope by reference.

[0092] In some embodiments, mRNAs may contain RNA backbone modifications.

Typically, a backbone modification is a modification in which the phosphates of the backbone of the nucleotides contained in the RNA are modified chemically. Exemplary backbone

modifications typically include, but are not limited to, modifications from the group consisting of methylphosphonates, methylphosphoramidates, phosphoramidates, phosphorothioates (e.g.

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

[0093] 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 2'-deoxy-2'-fluoro- oligoribonucleotide (2'-fluoro-2'-deoxycytidine 5'-triphosphate, 2'-fluoro-2'-deoxyuridine 5'- triphosphate), 2'-deoxy-2'-deamine-oligoribonucleotide (2'-amino-2'-deoxycytidine 5'- triphosphate, 2'-amino-2'-deoxyuridine 5'-triphosphate), 2'-0-alkyloligoribonucleotide, 2'-deoxy- 2'-C-alkyloligoribonucleotide (2'-0-methylcytidine 5'-triphosphate, 2'-methyluridine 5'- triphosphate), 2'-C-alkyloligoribonucleotide, and isomers thereof (2'-aracytidine 5'-triphosphate, 2'-arauridine 5'-triphosphate), or azidotriphosphates (2'-azido-2'-deoxycytidine 5'-triphosphate, 2'-azido-2'-deoxyuridine 5'-triphosphate).

[0094] In some embodiments, mRNAs may contain modifications of the bases of the nucleotides (base modifications). A modified nucleotide which contains a base modification is also called a base-modified nucleotide. Examples of such base-modified nucleotides include, but are not limited to, 2-amino-6-chloropurine riboside 5'-triphosphate, 2-aminoadenosine 5'- triphosphate, 2-thiocytidine 5'-triphosphate, 2-thiouridine 5'-triphosphate, 4-thiouridine 5'- triphosphate, 5-aminoallylcytidine 5'-triphosphate, 5-aminoallyluridine 5'-triphosphate, 5- bromocytidine 5'-triphosphate, 5-bromouridine 5'-triphosphate, 5-iodocytidine 5'-triphosphate, 5- iodouridine 5'-triphosphate, 5-methylcytidine 5'-triphosphate, 5-methyluridine 5'-triphosphate, 6- azacytidine 5'-triphosphate, 6-azauridine 5 '-triphosphate, 6-chloropurine riboside 5'-triphosphate, 7-deazaadenosine 5'-triphosphate, 7-deazaguanosine 5'-triphosphate, 8-azaadenosine 5'- triphosphate, 8-azidoadenosine 5'-triphosphate, benzimidazole riboside 5'-triphosphate, Nl- methyladenosine 5'-triphosphate, Nl-methylguanosine 5'-triphosphate, N6-methyladenosine 5'- triphosphate, 06-methylguanosine 5'-triphosphate, pseudouridine 5'-triphosphate, puromycin 5'- triphosphate or xanthosine 5'-triphosphate.

[0095] Typically, mRNA synthesis includes the addition of a“cap” on the 5’ end, and a

“tail” on the 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.

[0096] 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’ inverted triphosphate linkage; and the 7-nitrogen of guanine is then methylated by a methyltransferase. 2’-0-methylation may also occur at the first base and/or second base following the 7-methyl guanosine triphosphate residues. Examples of cap structures include, but are not limited to, m7GpppNp-RNA, m7GpppNmp-RNA and m7GpppNmpNmp-RNA (where m indicates 2’-Omethyl residues).

[0097] 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.

[0098] 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.

[0099] In some embodiments, a 3’ untranslated region includes one or more of a polyadenylation signal, a binding site for proteins that affect an mRNA’s stability of location in a cell, or one or more binding sites for miRNAs. In some embodiments, a 3’ untranslated region may be between 50 and 500 nucleotides in length or longer.

Cap structure

[0100] 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’inverted 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.

[0101] 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.

[0102] A common cap for mRNA produced by in vitro transcription is m 7 G(5')ppp(5')G, which has been used as the dinucleotide cap in transcription with T7 or SP6 RNA polymerase in vitro to obtain RNAs having a cap structure in their 5'-termini. The prevailing method for the in vitro synthesis of capped mRNA employs a pre-formed dinucleotide of the form

m 7 G(5')ppp(5')G (“m 7 GpppG”) as an initiator of transcription.

[0103] To date, a usual form of a synthetic dinucleotide cap used in in vitro translation experiments is the Anti-Reverse Cap Analog (“ARCA”) or modified ARCA, which is generally a modified cap analog in which the 2' or 3' OH group is replaced with -OCH 3 .

[0104] Additional cap analogs include, but are not limited to, a chemical structures selected from the group consisting of m 7 GpppG, m 7 GpppA, m 7 GpppC; unmethylated cap analogs (e.g., GpppG); dimethylated cap analog (e.g., m 2,7 GpppG), trimethylated cap analog (e.g., m 2 2 7 GpppG), dimethylated symmetrical cap analogs (e.g., m 7 Gpppm 7 G), or anti reverse cap analogs (e.g., ARCA; m 7 , 20me GpppG, m 72 d GpppG, m 7,3 ° me GpppG, m 7,3 d GpppG and their tetraphosphate derivatives) (see, e.g., Jemielity, J. et al.,“ Novel‘anti-reverse’ cap analogs with superior translational properties” , RNA, 9: 1108-1122 (2003)).

[0105] 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.

[0106] In some embodiments, the cap is a CapO structure. CapO structures lack a 2'-0- methyl residue of the ribose attached to bases 1 and 2. In some embodiments, the cap is a Capl structure. Capl structures have a 2'-0-methyl residue at base 2. In some embodiments, the cap is a Cap2 structure. Cap2 structures have a 2'-0-methyl residue attached to both bases 2 and 3.

[0107] 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 ah, 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 ah, RNA, 10: 1479-1487 (2004)), phosphorothioate cap analogs (described in Grudzien-Nogalska, E., et ah, RNA, 13: 1745-1755 (2007)), and cap analogs (including biotinylated cap analogs) described in U.S. Patent Nos. 8,093,367 and 8,304,529, incorporated by reference herein.

Tail structure

[0108] 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)).

[0109] In some embodiments, mRNAs include a 3’ tail structure. Typically, a tail structure includes a poly(A) and/or poly(C) tail. A poly-A or poly-C tail on the 3' terminus of mRNA typically includes at least 50 adenosine or cytosine nucleotides, at least 150 adenosine or cytosine nucleotides, at least 200 adenosine or cytosine nucleotides, at least 250 adenosine or cytosine nucleotides, at least 300 adenosine or cytosine nucleotides, at least 350 adenosine or cytosine nucleotides, at least 400 adenosine or cytosine nucleotides, at least 450 adenosine or cytosine nucleotides, at least 500 adenosine or cytosine nucleotides, at least 550 adenosine or cytosine nucleotides, at least 600 adenosine or cytosine nucleotides, at least 650 adenosine or cytosine nucleotides, at least 700 adenosine or cytosine nucleotides, at least 750 adenosine or cytosine nucleotides, at least 800 adenosine or cytosine nucleotides, at least 850 adenosine or cytosine nucleotides, at least 900 adenosine or cytosine nucleotides, at least 950 adenosine or cytosine nucleotides, or at least 1 kb adenosine or cytosine nucleotides, respectively. In some embodiments, a poly-A or poly-C tail may be about 10 to 800 adenosine or cytosine nucleotides (e.g., about 10 to 200 adenosine or cytosine nucleotides, about 10 to 300 adenosine or cytosine nucleotides, about 10 to 400 adenosine or cytosine nucleotides, about 10 to 500 adenosine or cytosine nucleotides, about 10 to 550 adenosine or cytosine nucleotides, about 10 to 600 adenosine or cytosine nucleotides, about 50 to 600 adenosine or cytosine nucleotides, about 100 to 600 adenosine or cytosine nucleotides, about 150 to 600 adenosine or cytosine nucleotides, about 200 to 600 adenosine or cytosine nucleotides, about 250 to 600 adenosine or cytosine nucleotides, about 300 to 600 adenosine or cytosine nucleotides, about 350 to 600 adenosine or cytosine nucleotides, about 400 to 600 adenosine or cytosine nucleotides, about 450 to 600 adenosine or cytosine nucleotides, about 500 to 600 adenosine or cytosine nucleotides, about 10 to 150 adenosine or cytosine nucleotides, about 10 to 100 adenosine or cytosine nucleotides, about 20 to 70 adenosine or cytosine nucleotides, or about 20 to 60 adenosine or cytosine nucleotides) respectively. In some embodiments, a tail structure includes is a combination of poly(A) and poly(C) tails with various lengths described herein. In some embodiments, a tail structure includes at least 50%, 55%, 65%, 70%, 75%, 80%, 85%, 90%, 92%, 94%, 95%, 96%, 97%, 98%, or 99% adenosine nucleotides. In some embodiments, a tail structure includes at least 50%, 55%, 65%, 70%, 75%, 80%, 85%, 90%, 92%, 94%, 95%, 96%, 97%, 98%, or 99% cytosine nucleotides.

[0110] In some embodiments, the length of the poly A or poly C tail is adjusted to control the stability of a modified sense mRNA molecule of the invention and, thus, the transcription of protein. For example, since the length of the poly A tail can influence the half- life of a sense mRNA molecule, the length of the poly A tail can be adjusted to modify the level of resistance of the mRNA to nucleases and thereby control the time course of polynucleotide expression and/or polypeptide production in a target cell.

5’ and 3’ Untranslated Region

[0111] 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.

[0112] In some embodiments, a 3’ untranslated region includes one or more of a polyadenylation signal, a binding site for proteins that affect an mRNA’s stability of location in a cell, or one or more binding sites for miRNAs. In some embodiments, a 3’ untranslated region may be between 50 and 500 nucleotides in length or longer.

[0113] 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.

[0114] While mRNA provided from in vitro transcription reactions may be desirable in some embodiments, other sources of mRNA are contemplated as within the scope of the invention including mRNA produced from bacteria, fungi, plants, and/or animals.

[0115] The present invention may be used to deliver mRNAs encoding a variety of proteins. Non-limiting examples of mRNAs suitable for the present invention include mRNAs encoding target proteins such as argininosuccinate synthetase (ASS 1), firefly luciferase (FFL), phenylalanine hydroxylase (PAH), and Ornithine transcarbamylase (OTC).

Exemplary mRNA sequences

[0116] In some embodiments, the present invention provides methods and compositions for delivering mRNA encoding a target protein to a subject for the treatment of the target protein deficiency. Exemplary mRNA sequences are shown below.

Construct design:

X - mRNA coding sequence - Y

5’ and 3’ UTR Sequences

X (5’ UTR Sequence) =

GGACAGAUCGCCUGGAGACGCCAUCCACGCUGUUUUGACCUCCAUAGAAGACACC GGGACCGAUCCAGCCUCCGCGGCCGGGAACGGUGCAUUGGAACGCGGAUUCCCCG U GCC AAGAGU G ACU C ACCGUCCUU GAC ACG (SEQ ID NO: 1)

Y (3’ UTR Sequence) =

CGGGU GGC AUCCCUGU GACCCCUCCCC AGU GCCUCUCCU GGCCCUGGAAGUU GCC ACUCCAGUGCCCACCAGCCUUGUCCUAAUAAAAUUAAGUUGCAUCAAGCU (SEQ

ID NO: 2)

OR

GGGUGGCAUCCCUGUGACCCCUCCCCAGUGCCUCUCCUGGCCCUGGAAGUUGCCA CUCCAGUGCCCACCAGCCUUGUCCUAAUAAAAUUAAGUUGCAUCAAAGCU (SEQ ID NO: 3) An exemplary full-length codon-optimized human ornithine transcarbamylase (QTC ) messenger RNA sequence is shown below:

GGACAGAUCGCCUGGAGACGCCAUCCACGCUGUUUUGACCUCCAUAGAAGACACC

GGGACCGAUCCAGCCUCCGCGGCCGGGAACGGUGCAUUGGAACGCGGAUUCCCCG

U GCC AAGAGU G ACU C ACCGUCCUU GAC ACGAU GCUGUUC AACCUUCGGAUCUU GC

UGAACAACGCUGCGUUCCGGAAUGGUCACAACUUCAUGGUCCGGAACUUCAGAUG

CGGCCAGCCGCUCCAGAACAAGGUGCAGCUCAAGGGGAGGGACCUCCUCACCCUG

AAAAACUUCACCGGAGAAGAGAUCAAGUACAUGCUGUGGCUGUCAGCCGACCUCA

AAUUCCGGAUCAAGCAGAAGGGCGAAUACCUUCCUUUGCUGCAGGGAAAGUCCCU

GGGGAUGAUCUUCGAGAAGCGCAGCACUCGCACUAGACUGUCAACUGAAACCGGC

UUCGCGCUGCUGGGAGGACACCCCUGCUUCCUGACCACCCAAGAUAUCCAUCUGG

GUGUGAACGAAUCCCUCACCGACACAGCGCGGGUGCUGUCGUCCAUGGCAGACGC

GGUCCUCGCCCGCGUGUACAAGCAGUCUGAUCUGGACACUCUGGCCAAGGAAGCC

UCCAUUCCUAUCAUUAAUGGAUUGUCCGACCUCUACCAUCCCAUCCAGAUUCUGG

CCGAUUAUCUGACUCUGCAAGAACAUUACAGCUCCCUGAAGGGGCUUACCCUUUC

GUGGAUCGGCGACGGCAACAACAUUCUGCACAGCAUUAUGAUGAGCGCUGCCAAG

UUU GGA AU GC ACCU CC AAGC AGCGACCCCGAAGGGAU ACGAGCC AGACGCCUCCG

UGACGAAGCUGGCUGAGCAGUACGCCAAGGAGAACGGCACUAAGCUGCUGCUCAC

CAACGACCCUCUCGAAGCCGCCCACGGUGGCAACGUGCUGAUCACCGAUACCUGG

AUCUCCAUGGGACAGGAGGAGGAAAAGAAGAAGCGCCUGCAAGCAUUUCAGGGG

UACCAGGUGACUAUGAAAACCGCCAAGGUCGCCGCCUCGGACUGGACCUUCUUGC

ACUGUCUGCCCAGAAAGCCCGAAGAGGUGGACGACGAGGUGUUCUACAGCCCGCG

GUCGCUGGUCUUUCCGGAGGCCGAAAACAGGAAGUGGACUAUCAUGGCCGUGAU

GGUGUCCCUGCUGACCGAUUACUCCCCGCAGCUGCAGAAACCAAAGUUCUGA

CGGGUGGCAUCCCUGUGACCCCUCCCCAGUGCCUCUCCUGGCCCUGGAAGUUGCC

ACUCCAGUGCCCACCAGCCUUGUCCUAAUAAAAUUAAGUUGCAUCAAGCU (SEQ

ID NO: 4).

An exemplary full length codon-optimized human ornithine transcarbamylase (PTC ) messenger

RNA sequence is shown below:

GGACAGAUCGCCUGGAGACGCCAUCCACGCUGUUUUGACCUCCAUAGAAGACACC GGGACCGAUCCAGCCUCCGCGGCCGGGAACGGUGCAUUGGAACGCGGAUUCCCCG U GCC AAGAGU G ACU C ACCGUCCUU GAC ACGAU GCUGUUC AACCUUCGGAUCUU GC UGAACAACGCUGCGUUCCGGAAUGGUCACAACUUCAUGGUCCGGAACUUCAGAUG

CGGCCAGCCGCUCCAGAACAAGGUGCAGCUCAAGGGGAGGGACCUCCUCACCCUG

AAAAACUUCACCGGAGAAGAGAUCAAGUACAUGCUGUGGCUGUCAGCCGACCUCA

AAUUCCGGAUCAAGCAGAAGGGCGAAUACCUUCCUUUGCUGCAGGGAAAGUCCCU

GGGGAUGAUCUUCGAGAAGCGCAGCACUCGCACUAGACUGUCAACUGAAACCGGC

UUCGCGCUGCUGGGAGGACACCCCUGCUUCCUGACCACCCAAGAUAUCCAUCUGG

GUGUGAACGAAUCCCUCACCGACACAGCGCGGGUGCUGUCGUCCAUGGCAGACGC

GGUCCUCGCCCGCGUGUACAAGCAGUCUGAUCUGGACACUCUGGCCAAGGAAGCC

UCCAUUCCUAUCAUUAAUGGAUUGUCCGACCUCUACCAUCCCAUCCAGAUUCUGG

CCGAUUAUCUGACUCUGCAAGAACAUUACAGCUCCCUGAAGGGGCUUACCCUUUC

GUGGAUCGGCGACGGCAACAACAUUCUGCACAGCAUUAUGAUGAGCGCUGCCAAG

UUUGGAAUGCACCUCCAAGCAGCGACCCCGAAGGGAUACGAGCCAGACGCCUCCG

UGACGAAGCUGGCUGAGCAGUACGCCAAGGAGAACGGCACUAAGCUGCUGCUCAC

CAACGACCCUCUCGAAGCCGCCCACGGUGGCAACGUGCUGAUCACCGAUACCUGG

AUCUCCAUGGGACAGGAGGAGGAAAAGAAGAAGCGCCUGCAAGCAUUUCAGGGG

UACCAGGUGACUAUGAAAACCGCCAAGGUCGCCGCCUCGGACUGGACCUUCUUGC

ACUGUCUGCCCAGAAAGCCCGAAGAGGUGGACGACGAGGUGUUCUACAGCCCGCG

GUCGCUGGUCUUUCCGGAGGCCGAAAACAGGAAGUGGACUAUCAUGGCCGUGAU

GGUGUCCCUGCUGACCGAUUACUCCCCGCAGCUGCAGAAACCAAAGUUCUGA

GGGUGGCAUCCCUGUGACCCCUCCCCAGUGCCUCUCCUGGCCCUGGAAGUUGCCA

CUCCAGUGCCCACCAGCCUUGUCCUAAUAAAAUUAAGUUGCAUCAAAGCU (SEQ

ID NO: 5).

Another exemplary full length codon-optimized human ornithine transcarbamylase (PTC) messenger RNA sequence is shown below:

GGACAGAUCGCCUGGAGACGCCAUCCACGCUGUUUUGACCUCCAUAGAAGACACC GGGACCGAUCCAGCCUCCGCGGCCGGGAACGGUGCAUUGGAACGCGGAUUCCCCG U GCC AAGAGU G ACU C ACCGUCCUU GAC ACGAU GCUGUUUAACCUGAGAAUUCU GC UGAACAACGCCGCGUUCAGGAACGGCCACAAUUUCAUGGUCCGCAACUUUAGAUG CGGACAGCCUCUCCAAAACAAGGUCCAGCUCAAGGGGCGGGACUUGCUGACCCUU AAGAACUUUACCGGCGAAGAGAUC AAGU AC AU GCUGU GGUU GUC AGCGGACCUG AAGUUCCGCAUCAAGCAGAAAGGGGAGUAUCUGCCGCUGCUCCAAGGAAAGUCGC UCGGCAUGAUCUUCGAGAAGCGCUCGACCAGAACCCGGCUGUCCACUGAAACUGG UUUCGCCCUUCUGGGUGGACACCCUUGUUUCCUGACAACCCAGGACAUCCAUCUG GGCGUGAACGAAAGCCUCACUGACACCGCCAGGGUGCUGAGCUCCAUGGCCGACG CUGUCCUUGCCCGGGUGUACAAGCAGUCCGAUCUGGACACUCUGGCCAAGGAAGC GUCCAUCCCGAUCAUUAACGGACUGUCCGACCUGUACCACCCGAUCCAGAUUCUG GCCGACUACCUGACCUU GC AAGAGC ACU AC AGCUC ACUGAAGGGCUU GACCCU GA GCUGGAUCGGCGACGGAAACAACAUUCUGCAUUCGAUCAUGAUGUCCGCGGCCAA GUUCGGAAUGCAUCUGCAGGCCGCAACUCCCAAGGGAUACGAACCUGAUGCGUCC GU GACUAAGCU GGCCGAGC AGUACGC AAAGGAAA ACGGC ACC AAGCU GCUGCU GA CCAACGACCCGCUCGAAGCUGCCCACGGAGGGAACGUGCUCAUUACCGACACUUG GAUCUCCAUGGGGCAGGAAGAAGAGAAGAAGAAGCGGCUCCAGGCAUUCCAGGG UUACCAGGUCACCAUGAAAACGGCCAAAGUGGCCGCUUCGGAUUGGACUUUCCUC C ACU GCCUUCCCCGC AAACCU GAGGAAGU GGAU GAU GAAGU GUUCUACUCCCC AC GCU CCCUC GU GUU CCCC G AGGCC G AG A AU C GG A AGU GG ACC AUU AU GGCC GU GAU GGUGUCACUGCUGACCGACUACAGCCCCCAACUGCAAAAGCCGAAGUUCUGA CGGGU GGC AUCCCUGU GACCCCUCCCC AGU GCCUCUCCU GGCCCUGGAAGUU GCC ACUCCAGUGCCCACCAGCCUUGUCCUAAUAAAAUUAAGUUGCAUCAAGCU (SEQ ID NO: 6)

Exemplary codon-optimized Human ASS 1 (CO-hASS 1 ) Coding Sequence

AU GAGC AGC AAGGGC AGCGU GGU GCUGGCCUAC AGCGGCGGCCU GGAC ACC AGCU

GCAUCCUGGUGUGGCUGAAGGAGCAGGGCUACGACGUGAUCGCCUACCUGGCCAA

CAUCGGCCAGAAGGAGGACUUCGAGGAGGCCCGCAAGAAGGCCCUGAAGCUGGGC

GCCAAGAAGGUGUUCAUCGAGGACGUGAGCCGCGAGUUCGUGGAGGAGUUCAUC

UGGCCCGCCAUCCAGAGCAGCGCCCUGUACGAGGACCGCUACCUGCUGGGCACCA

GCCUGGCCCGCCCCUGCAUCGCCCGCAAGCAGGUGGAGAUCGCCCAGCGCGAGGG

CGCCAAGUACGUGAGCCACGGCGCCACCGGC AAGGGC AACGACCAGGUGCGCUUC

GAGCUGAGCUGCUACAGCCUGGCCCCCCAGAUCAAGGUGAUCGCCCCCUGGCGCA

UGCCCGAGUUCUACAACCGCUUCAAGGGCCGCAACGACCUGAUGGAGUACGCCAA

GCAGCACGGCAUCCCCAUCCCCGUGACCCCCAAGAACCCCUGGAGCAUGGACGAG

AACCU GAU GC AC AU C AGCUACGAGGCCGGC AUCCU GGAGAACCCC AAGAACC AGG

CCCCCCCCGGCCUGUACACCAAGACCCAGGACCCCGCCAAGGCCCCCAACACCCCC

GAC AUCCUGGAGAUCGAGUUC AAGAAGGGCGU GCCCGU GAAGGU GACC AACGU G

AAGGACGGCACCACCCACCAGACCAGCCUGGAGCUGUUCAUGUACCUGAACGAGG UGGCCGGCAAGCACGGCGUGGGCCGCAUCGACAUCGUGGAGAACCGCUUCAUCGG CAUGAAGAGCCGCGGCAUCUACGAGACCCCCGCCGGCACCAUCCUGUACCACGCC CACCUGGACAUCGAGGCCUUCACCAUGGACCGCGAGGUGCGCAAGAUCAAGCAGG GCCUGGGCCUGAAGUUCGCCGAGCUGGUGUACACCGGCUUCUGGCACAGCCCCGA GUGCGAGUUCGUGCGCCACUGCAUCGCCAAGAGCCAGGAGCGCGUGGAGGGCAAG GU GC AGGU GAGCGU GCUGAAGGGCC AGGU GUAC AUCCU GGGCCGCGAGAGCCCCC U G AGCCU GU AC A AC G AGG AGCUGGU G AGC AU G A AC GU GC AGGGC G ACU AC G AGC CCACCGACGCCACCGGCUUCAUCAACAUCAACAGCCUGCGCCUGAAGGAGUACCA CCGCCU GC AGAGC AAGGU GACCGCC AAGU GA (SEQ ID NO: 7)

Exemplary codon-optimized Human PAH (CO-hPAH ) Coding Sequence

AUGAGCACCGCCGUGCUGGAGAACCCCGGCCUGGGCCGCAAGCUGAGCGACUUCG

GCCAGGAGACCAGCUACAUCGAGGACAACUGCAACCAGAACGGCGCCAUCAGCCU

GAUCUUCAGCCUGAAGGAGGAGGUGGGCGCCCUGGCCAAGGUGCUGCGCCUGUUC

GAGGAGAACGACGUGAACCUGACCCACAUCGAGAGCCGCCCCAGCCGCCUGAAGA

AGGACGAGUACGAGUUCUUCACCCACCUGGACAAGCGCAGCCUGCCCGCCCUGAC

CAACAUCAUCAAGAUCCUGCGCCACGACAUCGGCGCCACCGUGCACGAGCUGAGC

CGCGACAAGAAGAAGGACACCGUGCCCUGGUUCCCCCGCACCAUCCAGGAGCUGG

ACCGCUUCGCCAACCAGAUCCUGAGCUACGGCGCCGAGCUGGACGCCGACCACCC

CGGCUUCAAGGACCCCGUGUACCGCGCCCGCCGCAAGCAGUUCGCCGACAUCGCC

UACAACUACCGCCACGGCCAGCCCAUCCCCCGCGUGGAGUACAUGGAGGAGGAGA

AGAAGACCU GGGGC ACCGU GUUC AAGACCCU GAAGAGCCU GU AC AAGACCC ACGC

CUGCUACGAGUACAACCACAUCUUCCCCCUGCUGGAGAAGUACUGCGGCUUCCAC

GAGGAC AAC AUCCCCC AGCUGGAGGACGU GAGCC AGUUCCUGC AGACCUGC ACCG

GCUUCCGCCUGCGCCCCGUGGCCGGCCUGCUGAGCAGCCGCGACUUCCUGGGCGG

CCUGGCCUUCCGCGUGUUCCACUGCACCCAGUACAUCCGCCACGGCAGCAAGCCC

AUGUACACCCCCGAGCCCGACAUCUGCCACGAGCUGCUGGGCCACGUGCCCCUGU

UCAGCGACCGCAGCUUCGCCCAGUUCAGCCAGGAGAUCGGCCUGGCCAGCCUGGG

CGCCCCCGACGAGUACAUCGAGAAGCUGGCCACCAUCUACUGGUUCACCGUGGAG

UUCGGCCUGUGCAAGCAGGGCGACAGCAUCAAGGCCUACGGCGCCGGCCUGCUGA

GCAGCUUCGGCGAGCUGCAGUACUGCCUGAGCGAGAAGCCCAAGCUGCUGCCCCU

GGAGCUGGAGAAGACCGCCAUCCAGAACUACACCGUGACCGAGUUCCAGCCCCUG

UACUACGUGGCCGAGAGCUUCAACGACGCCAAGGAGAAGGUGCGCAACUUCGCCG CCACCAUCCCCCGCCCCUUCAGCGUGCGCUACGACCCCUACACCCAGCGCAUCGAG GUGCUGGACAACACCCAGCAGCUGAAGAUCCUGGCCGACAGCAUCAACAGCGAGA UCGGCAUCCUGUGCAGCGCCCUGCAGAAGAUCAAGUAA (SEQ ID NO: 8)

[0117] In some embodiments, a suitable mRNA sequence may encode a homolog or an analog of target protein. For example, a homolog or an analog of target protein may be a modified target protein containing one or more amino acid substitutions, deletions, and/or insertions as compared to a wild-type or naturally- occurring target protein while retaining substantial target protein activity. In some embodiments, an mRNA suitable for the present invention encodes an amino acid sequence at least 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more homologous to the above exemplary sequences. In some embodiments, an mRNA suitable for the present invention encodes a protein substantially identical to target protein. In some embodiments, an mRNA suitable for the present invention encodes an amino acid sequence at least 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more identical to the above exemplary sequences. In some embodiments, an mRNA suitable for the present invention encodes a fragment or a portion of target protein. In some embodiments, an mRNA suitable for the present invention encodes a fragment or a portion of target protein, wherein the fragment or portion of the protein still maintains target activity similar to that of the wild-type protein. In some embodiments, an mRNA suitable for the present invention has a nucleotide sequence at least 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%,

93%, 94%, 95%, 96%, 97%, 98%, 99% or more identical to the above exemplary sequences.

[0118] In some embodiments, a suitable mRNA encodes a fusion protein comprising a full length, fragment or portion of a target protein fused to another protein (e.g., an N or C terminal fusion). In some embodiments, the protein fused to the mRNA encoding a full length, fragment or portion of a target protein encodes a signal or a cellular targeting sequence.

Lipid Nanoparticles

[0119] According to the present invention, mRNA may be encapsulated or complexed in nanoparticles. In some embodiments, nanoparticles are also referred to as“delivery vehicle,” “transfer vehicle”, or grammatical equivalents.

[0120] According to various embodiments, suitable nanoparticles include, but are not limited to polymer based carriers, such as polyethylenimine (PEI), lipid nanoparticles and liposomes, nanoliposomes, ceramide-containing nanoliposomes, proteoliposomes, both natural and synthetically-derived exosomes, natural, synthetic and semi-synthetic lamellar bodies, nanoparticulates, calcium phosphor-silicate nanoparticulates, calcium phosphate

nanoparticulates, silicon dioxide nanoparticulates, nanocrystalline particulates, semiconductor nanoparticulates, poly(D-arginine), sol-gels, nanodendrimers, starch-based delivery systems, micelles, emulsions, niosomes, multi-domain-block polymers (vinyl polymers, polypropyl acrylic acid polymers, dynamic polyconjugates), dry powder formulations, plasmids, viruses, calcium phosphate nucleotides, aptamers, peptides and other vectorial tags.

[0121] In some embodiments, the mRNA is encapsulated within one or more liposomes.

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). Thus, the term“liposome” as used herein

encompasses both lipid and polymer based nanoparticles. In some embodiments, a liposome suitable for the present invention contains cationic, non-cationic lipid(s), cholesterol-based lipid(s) and/or PEG-modified lipid(s).

PEGylated Lipids

[0122] In some embodiments, a suitable lipid solution includes one or more PEGylated lipids. For example, the use of polyethylene glycol (PEG) -modified phospholipids and derivatized lipids such as derivatized ceramides (PEG-CER), including N-Octanoyl- Sphingosine-l-[Succinyl(Methoxy Polyethylene Glycol)-2000] (C8 PEG-2000 ceramide) is also contemplated by the present invention. Contemplated PEG-modified lipids include, but are not limited to, a polyethylene glycol chain of up to 5 kDa in length covalently attached to a lipid with alkyl chain(s) of C6-C20 length. In some embodiments, a PEG-modified or PEGylated lipid is PEGylated cholesterol or PEG-2K. In some embodiments, particularly useful exchangeable lipids are PEG-ceramides having shorter acyl chains (e.g., C 14 or Cis).

[0123] PEG-modified phospholipid and derivatized lipids may constitute at least 1%, at least 2%, at least 3%, at least 4%, at least 5%, at least 6%, at least 7%, at least 8%, at least 9%, or at least l0%of the total lipids in the liposome.

Cationic Lipids

[0124] As used herein, the phrase“cationic lipids” 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. Particularly suitable cationic lipids for use in the compositions and methods of the invention include those described in international patent publications WO 2010/053572 (and particularly, C12-200 described at paragraph [00225]) and WO 2012/170930, both of which are incorporated herein by reference. In certain embodiments, cationic lipids suitable for the compositions and methods of the invention include an ionizable cationic lipid described in U.S. provisional patent application 61/617,468, filed March 29, 2012 (incorporated herein by reference), such as, e.g, (15Z, l8Z)-N,N-dimethyl-6-(9Z, l2Z)-octadeca-9, l2-dien-l -yl)tetracosa- l5,l8-dien- 1 -amine (HGT5000), ( 15Z, l8Z)-N,N-dimethyl-6-((9Z, l2Z)-octadeca-9, l2-dien- 1 -yl)tetracosa- 4,l5,l8-trien-l -amine (HGT5001), and (l5Z,l8Z)-N,N-dimethyl-6-((9Z, l2Z)-octadeca-9, 12- dien- 1 -yl)tetracosa-5, 15 , l8-trien- 1 -amine (HGT5002).

[0125] In some embodiments, cationic lipids suitable for the compositions and methods of the invention include cationic lipids such as such as 3,6-bis(4-(bis((9Z,l2Z)-2- hydroxyoctadeca-9,l2-dien-l-yl)amino)butyl)piperazine-2,5-di one (OF-02).

[0126] In some embodiments, cationic lipids suitable for the compositions and methods of the invention include a cationic lipid described in WO 2015/184256 A2 entitled

“Biodegradable lipids for delivery of nucleic acids” which is incorporated by reference herein such as 3-(4-(bis(2-hydroxydodecyl)amino)butyl)-6-(4-((2-hydroxydode cyl)(2- hydroxyundecyl)amino)butyl)-l,4-dioxane-2,5-dione (Target 23), 3-(5-(bis(2- hydroxydodecyl)amino)pentan-2-yl)-6-(5-((2-hydroxydodecyl)(2 - hydroxyundecyl)amino)pentan-2-yl)-l,4-dioxane-2,5-dione (Target 24).

[0127] In some embodiments, cationic lipids suitable for the compositions and methods of the invention include a cationic lipid described in WO 2013/063468 and in U.S. provisional application entitled“Lipid Formulations for Delivery of Messenger RNA”, both of which are incorporated by reference herein.

[0128] In some embodiments, one or more cationic lipids suitable for the present invention may be N-[l-(2,3-dioleyloxy)propyl]-N,N,N-trimethylammonium chloride or

"DOTMA". (Feigner et al. (Proc. Nat'l Acad. Sci. 84, 7413 (1987); U.S. Pat. No. 4,897,355). Other suitable cationic lipids include, for example, 5-carboxyspermylglycinedioctadecylamide or "DOGS , " 2,3 -dioleyloxy-N- [2(spermine-carboxamido)ethyl] -N,N-dimethyl-l-propanaminium or "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 or "DODAP", l,2-Dioleoyl-3- Trimethylammonium-Propane or "DOTAP".

[0129] Additional exemplary cationic lipids also include l,2-distearyloxy-N,N-dimethyl-

3-aminopropane or "DSDMA", l,2-dioleyloxy-N,N-dimethyl-3-aminopropane or "DODMA", 1 ,2-dilinoleyloxy-N,N-dimethyl-3-aminopropane or "DLinDMA", l,2-dilinolenyloxy-N,N- dimethyl-3-aminopropane or "DLenDMA", N-dioleyl-N,N-dimethylammonium chloride or "DODAC", N,N-distearyl-N,N-dimethylarnmonium bromide or "DDAB", N-(l,2- dimyristyloxyprop-3-yl)-N,N-dimethyl-N-hydroxyethyl ammonium bromide or "DMRIE", 3- dimethylamino-2-(cholest-5-en-3-beta-oxybutan-4-oxy)-l-(ci s,cis-9,l2- octadecadienoxy)propane or "CLinDMA", 2-[5'-(cholest-5-en-3-beta-oxy)-3'-oxapentoxy)-3- dimethy l-l-(cis,cis-9', l-2'-octadecadienoxy)propane or "CpLinDMA", N,N-dimethyl-3,4- dioleyloxybenzylamine or "DMOBA", 1 ,2-N,N'-dioleylcarbamyl-3-dimethylaminopropane or "DOcarbDAP", 2,3-Dilinoleoyloxy-N,N-dimethylpropylamine or "DLinDAP", l,2-N,N'- Dilinoleylcarbamyl-3-dimethylaminopropane or "DLincarbDAP", 1 ,2-Dilinoleoylcarbamyl-3- dimethylaminopropane or "DLinCDAP", 2,2-dilinoleyl-4-dimethylaminomethyl-[l,3]-dioxolane or "DLin-DMA", 2,2-dilinoleyl-4-dimethylaminoethyl-[l,3]-dioxolane or "DLin-K-XTC2- DMA", and 2-(2,2-di((9Z,l2Z)-octadeca-9,l 2-dien- l-yl)-l ,3-dioxolan-4-yl)-N,N- dimethylethanamine (DLin-KC2-DMA)) (see, WO 2010/042877; Semple et al., Nature Biotech. 28: 172-176 (2010)), or mixtures thereof. (Heyes, J., et al., J Controlled Release 107: 276-287 (2005); Morrissey, DV., et al., Nat. Biotechnol. 23(8): 1003-1007 (2005); PCT Publication WO2005/121348A1). In some embodiments, one or more of the cationic lipids comprise at least one of an imidazole, dialkylamino, or guanidinium moiety.

[0130] In some embodiments, one or more cationic lipids may be chosen from XTC (2,2-

Dilinoleyl-4-dimethylaminoethyl-[l,3]-dioxolane), MC3 (((6Z,9Z,28Z,3 lZ)-heptatriaconta- 6,9,28,3 l-tetraen-l9-yl 4-(dimethylamino)butanoate), ALNY-100 ((3aR,5s,6aS)-N,N-dimethyl- 2,2-di((9Z, 12Z)-octadeca-9, 12-dienyl)tetrahydro-3 aH-cyclopenta[d] [1 ,3] dioxol-5-amine)) , NC98-5 (4,7,l3-tris(3-oxo-3-(undecylamino)propyl)-Nl,Nl6-diundecyl- 4,7,l0,l3- tetraazahexadecane- 1 , 16-diamide),

[0131] The term“cationic lipids” refers to any of a number of lipid and lipidoid species that have a net positive charge at a selected pH, such as at physiological pH.

[0132] Suitable cationic lipids for use in the compositions and methods of the invention include the cationic lipids as described in International Patent Publication WO 2010/14474, which is incorporated herein by reference. In certain embodiments, the compositions and methods of the present invention include a cationic lipid, (6Z,9Z,28Z,3lZ)-heptatriaconta- 6,9,28,3 l-tetraen-l9-yl 4-(dimethylamino) butanoate, having a compound structure of: and pharmaceutically acceptable salts thereof.

[0133] Other suitable cationic lipids for use in the compositions and methods of the present invention include ionizable cationic lipids as described in International Patent Publication WO 2013/149140, which is incorporated herein by reference. In some embodiments, the

compositions and methods of the present invention include a cationic lipid of one of the following formulas:

or a pharmaceutically acceptable salt thereof, wherein Ri and R 2 are each independently selected from the group consisting of hydrogen, an optionally substituted, variably saturated or unsaturated C 1 -C 20 alkyl and an optionally substituted, variably saturated or unsaturated C 6 -C 20 acyl; wherein Li and L 2 are each independently selected from the group consisting of hydrogen, an optionally substituted C 1 -C 30 alkyl, an optionally substituted variably unsaturated C 1 -C 30 alkenyl, and an optionally substituted C 1 -C 30 alkynyl; wherein m and o are each independently selected from the group consisting of zero and any positive integer (e.g., where m is three); and wherein n is zero or any positive integer (e.g., where n is one). In certain embodiments, the compositions and methods of the present invention include the cationic lipid (15Z, 18Z)-N,N- dimethyl-6-(9Z,l2Z)-octadeca-9,l2-dien-l -yl) tetracosa- l5,l8-dien-l-amine (“HGT5000”), having a compound structure of:

(HGT-5000) and pharmaceutically acceptable salts thereof. In certain embodiments, the compositions and methods of the present invention include the cationic lipid (15Z, l8Z)-N,N-dimethyl-6- ((9Z,l2Z)-octadeca-9,l2-dien-l-yl) tetracosa-4,l5,l8-trien-l -amine (“HGT5001”), having a compound structure of:

(HGT-5001) and pharmaceutically acceptable salts thereof. In certain embodiments, the compositions and methods of the present invention include the cationic lipid and (l5Z,l8Z)-N,N-dimethyl-6- ((9Z,l2Z)-octadeca-9,l2-dien-l-yl) tetracosa-5,l5,l8-trien- 1 -amine (“HGT5002”), having a compound structure of:

(HGT-5002) and pharmaceutically acceptable salts thereof.

[0134] Other suitable cationic lipids for use in the compositions and methods of the invention 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 and methods of the present invention include a cationic lipid having a compound structure of:

and pharmaceutically acceptable salts thereof.

[0135] Other suitable cationic lipids for use in the compositions and methods of the invention include the cationic lipids as described in International Patent Publication WO 2016/118725, which is incorporated herein by reference. In certain embodiments, the compositions and methods of the present invention include a cationic lipid having a compound structure of: and pharmaceutically acceptable salts thereof.

[0136] Other suitable cationic lipids for use in the compositions and methods of the invention include the cationic lipids as described in International Patent Publication WO 2016/118724, which is incorporated herein by reference. In certain embodiments, the compositions and methods of the present invention include a cationic lipid having a compound structure of:

and pharmaceutically acceptable salts thereof.

[0137] Other suitable cationic lipids for use in the compositions and methods of the invention include a cationic lipid having the formula of 14, 25-ditridecyl l5,l8,2l,24-tetraaza- octatriacontane, and pharmaceutically acceptable salts thereof.

[0138] Other suitable cationic lipids for use in the compositions and methods of the invention 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 and methods of the present invention 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 4 o alkenyl. In certain embodiments, the compositions and methods of the present invention include a cationic lipid having a compound structure of: and pharmaceutically acceptable salts thereof. In certain embodiments, the compositions and methods of the present invention include a cationic lipid having a compound structure of:

and pharmaceutically acceptable salts thereof. In certain embodiments, the compositions and methods of the present invention include a cationic lipid having a compound structure of:

and pharmaceutically acceptable salts thereof. In certain embodiments, the compositions and methods of the present invention include a cationic lipid having a compound structure of:

and pharmaceutically acceptable salts thereof.

[0139] Other suitable cationic lipids for use in the compositions and methods of the invention include the cationic lipids as described in International Patent Publication WO 2015/184256, which is incorporated herein by reference. In some embodiments, the compositions and methods of the present invention include a cationic lipid of the following formula:

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

(Target 23)

[0140] and pharmaceutically acceptable salts thereof.

[0141] Other suitable cationic lipids for use in the compositions and methods of the invention include the cationic lipids as described in International Patent Publication WO 2016/004202, which is incorporated herein by reference. In some embodiments, the compositions and methods of the present invention include a cationic lipid having the compound structure:

or a pharmaceutically acceptable salt thereof. In some embodiments, the compositions and methods of the present invention include a cationic lipid having the compound structure:

or a pharmaceutically acceptable salt thereof. In some embodiments, the compositions and methods of the present invention include a cationic lipid having the compound structure:

or a pharmaceutically acceptable salt thereof.

[0142] Other suitable cationic lipids for use in the compositions and methods of the present invention include the cationic lipids as described in J. McClellan, M. C. King, Cell 2010, 141, 210-217 and in Whitehead et ah, Nature Communications (2014) 5:4277, which is incorporated herein by reference. In certain embodiments, the cationic lipids of the compositions and methods of the present invention include a cationic lipid having a compound structure of: and pharmaceutically acceptable salts thereof.

[0143] Other suitable cationic lipids for use in the compositions and methods of the invention include the cationic lipids as described in International Patent Publication WO 2015/199952, which is incorporated herein by reference. In some embodiments, the compositions and methods of the present invention include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof. In some embodiments, the compositions and methods of the present invention include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof. In some embodiments, the compositions and methods of the present invention include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof. In some embodiments, the compositions and methods of the present invention include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof. In some embodiments, the compositions and methods of the present invention include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof. In some embodiments, the compositions and methods of the present invention include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof. In some embodiments, the compositions and methods of the present invention include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof. In some embodiments, the compositions and methods of the present invention include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof. In some embodiments, the compositions and methods of the present invention include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof. In some embodiments, the compositions and methods of the present invention include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof. In some embodiments, the compositions and methods of the present invention include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof. In some embodiments, the compositions and methods of the present invention include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof. In some embodiments, the compositions and methods of the present invention include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof.

[0144] Other suitable cationic lipids for use in the compositions and methods of the invention include the cationic lipids as described in International Patent Publication WO 2017/004143, which is incorporated herein by reference. In some embodiments, the compositions and methods of the present invention include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof. In some embodiments, the compositions and methods of the present invention include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof. In some embodiments, the compositions and methods of the present invention include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof. In some embodiments, the compositions and methods of the present invention include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof. In some embodiments, the compositions and methods of the present invention include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof. In some embodiments, the compositions and methods of the present invention include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof. In some embodiments, the compositions and methods of the present invention include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof. In some embodiments, the compositions and methods of the present invention include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof. In some embodiments, the compositions and methods of the present invention include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof. In some embodiments, the compositions and methods of the present invention include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof. In some embodiments, the compositions and methods of the present invention include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof. In some embodiments, the compositions and methods of the present invention include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof. In some embodiments, the compositions and methods of the present invention include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof. In some embodiments, the compositions and methods of the present invention include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof. In some embodiments, the compositions and methods of the present invention include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof. In some embodiments, the compositions and methods of the present invention include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof. In some embodiments, the compositions and methods of the present invention include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof.

[0145] Other suitable cationic lipids for use in the compositions and methods of the invention include the cationic lipids as described in International Patent Publication WO 2017/075531, which is incorporated herein by reference. In some embodiments, the compositions and methods of the present invention include a cationic lipid of the following formula:

or a pharmaceutically acceptable salt thereof, wherein one of L 1 or L 2 is -0(C=0)-, -(C=0)0-, -

C(=0)-, -0-, -S(0) x , -S-S-, -C(=0)S-, -SC(=0)-, -NR a C(=0)-, -C(=0)NR a -, NR a C(=0)NR a -, -

OC(=0)NR a -, or -NR a C(=0)0-; and the other of L 1 or L 2 is -0(C=0)-, -(C=0)0-, -C(=0)-, -0-, -S(0) x, -S-S-, -C(=0)S-, SC(=0)-, -NR a C(=0)-, -C(=0)NR a -, ,NR a C(=0)NR a -, -0C(=0)NR a - or -NR a C(=0)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 -Cs 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(=0)0R 4 , -0C(=0)R 4 or -NR 5 C(=0)R 4 ; R 4 is C 1 -C 12 alkyl; R 5 is H or Ci-C 6 alkyl; and x is 0, 1 or 2.

[0146] Other suitable cationic lipids for use in the compositions and methods of the invention include the cationic lipids as described in International Patent Publication WO 2017/117528, which is incorporated herein by reference. In some embodiments, the compositions and methods of the present invention include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof. In some embodiments, the compositions and methods of the present invention include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof. In some embodiments, the compositions and methods of the present invention include a cationic lipid having the compound structure:

and pharmaceutically acceptable salts thereof.

[0147] Other suitable cationic lipids for use in the compositions and methods of the invention 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 ) n Q and -(CH 2 ) n CHQR; Q is selected from the group consisting of -OR, -OH, -0(CH 2 )„N(R) 2 , -0C(0)R, -CX 3 , -CN, -N(R)C(0)R, -N(H)C(0)R, - N(R)S(0) 2 R, -N(H)S(0) 2 R, -N(R)C(0)N(R) 2 , -N(H)C(0)N(R) 2 , -N(H)C(0)N(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; and n is 1, 2, or 3. In certain embodiments, the compositions and methods of the present invention include a cationic lipid having a compound structure of:

and pharmaceutically acceptable salts thereof. In certain embodiments, the compositions and methods of the present invention include a cationic lipid having a compound structure of:

and pharmaceutically acceptable salts thereof. In certain embodiments, the compositions and methods of the present invention include a cationic lipid having a compound structure of:

and pharmaceutically acceptable salts thereof. In certain embodiments, the compositions and methods of the present invention include a cationic lipid having a compound structure of:

and pharmaceutically acceptable salts thereof.

[0148] Other suitable cationic lipids for use in the compositions and methods of the invention include the cationic lipids as described in International Patent Publication WO 2017/173054 and WO 2015/095340, each of which is incorporated herein by reference. In certain embodiments, the compositions and methods of the present invention include a cationic lipid having a compound structure of:

and pharmaceutically acceptable salts thereof. In certain embodiments, the compositions and methods of the present invention include a cationic lipid having a compound structure of:

and pharmaceutically acceptable salts thereof. In certain embodiments, the compositions and methods of the present invention include a cationic lipid having a compound structure of:

and pharmaceutically acceptable salts thereof. In certain embodiments, the compositions and methods of the present invention include a cationic lipid having a compound structure of:

and pharmaceutically acceptable salts thereof.

[0149] Other suitable cationic lipids for use in the compositions and methods of the invention include cholesterol-based cationic lipids. In certain embodiments, the compositions and methods of the present invention include imidazole cholesterol ester or“ICE”, having a compound structure of:

(ICE) and pharmaceutically acceptable salts thereof.

[0150] Other suitable cationic lipids for use in the compositions and methods of the present invention include cleavable cationic lipids as described in International Patent Publication WO 2012/170889, which is incorporated herein by reference. In some embodiments, the

compositions and methods of the present invention include a cationic lipid of the following formula: wherein Ri is selected from the group consisting of imidazole, guanidinium, amino, imine, enamine, an optionally-substituted alkyl amino (e.g., an alkyl amino such as dimethylamino) and pyridyl; wherein R2 is selected from the group consisting of one of the following two formulas:

and wherein R 3 and R 4 are each independently selected from the group consisting of an optionally substituted, variably saturated or unsaturated C6-C20 alkyl and an optionally substituted, variably saturated or unsaturated C6-C20 acyl; and wherein n is zero or any positive integer (e.g., one, two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, fourteen, fifteen, sixteen, seventeen, eighteen, nineteen, twenty or more). In certain

embodiments, the compositions and methods of the present invention include a cationic lipid, “HGT4001”, having a compound structure of:

(HGT4001) and pharmaceutically acceptable salts thereof. In certain embodiments, the compositions and methods of the present invention include a cationic lipid,“HGT4002”, having a compound structure of:

(HGT4002) and pharmaceutically acceptable salts thereof. In certain embodiments, the compositions and methods of the present invention include a cationic lipid,“HGT4003”, having a compound structure of:

(HGT4003) and pharmaceutically acceptable salts thereof. In certain embodiments, the compositions and methods of the present invention include a cationic lipid,“HGT4004”, having a compound structure of:

(HGT4004) and pharmaceutically acceptable salts thereof. In certain embodiments, the compositions and methods of the present invention include a cationic lipid“HGT4005”, having a compound structure of:

(HGT4005) and pharmaceutically acceptable salts thereof. [0151] In some embodiments, the compositions and methods of the present invention include the cationic lipid, N-[l-(2,3-dioleyloxy)propyl]-N,N,N-trimethylammonium chloride

(“DOTMA”). (Feigner et al. (Proc. Nat'l Acad. Sci. 84, 7413 (1987); U.S. Pat. No. 4,897,355, which is incorporated herein by reference). Other cationic lipids suitable for the compositions and methods of the present invention include, for example, 5- carboxyspermylglycinedioctadecylamide (“DOGS”); 2,3-dioleyloxy-N-[2(spermine- carboxamido)ethyl]-N,N-dimethyl-l-propanaminium (“DOSPA”) (Behr et al. Proc. Nat.'l Acad. Sci. 86, 6982 (1989), U.S. Pat. No. 5,171,678; U.S. Pat. No. 5,334,761); l,2-Dioleoyl-3- Dimethylammonium-Propane (“DODAP”); l,2-Dioleoyl-3-Trimethylammonium-Propane (“DOTAP”).

[0152] Additional exemplary cationic lipids suitable for the compositions and methods of the present invention also include: l,2-distearyloxy-N,N-dimethyl-3-aminopropane (“DSDMA”); l,2-dioleyloxy-N,N-dimethyl-3-aminopropane (“DODMA”); 1 ,2-dilinoleyloxy-N,N-dimethyl- 3-aminopropane (“DLinDMA”); l,2-dilinolenyloxy-N,N-dimethyl-3-aminopropane

(“DLenDMA”); N-dioleyl-N,N-dimethylammonium chloride (“DODAC”); N,N-distearyl-N,N- dimethylammonium 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,l2-octadecadienoxy)propane (“CLinDMA”); 2-[5'-(cholest-5-en-3- beta-oxy)-3'-oxapentoxy)-3-dimethy l-l-(cis,cis-9', l-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, l2Z)-octadeca-9, l2-dien-l -yloxy]propane-l -amine (“Octyl-CLinDM A”) ; (2R)-2-((8 - [(3beta)-cholest-5-en-3 -yloxy] octyl)oxy)-N, N-dimethyl-3 - [(9Z, l2Z)-octadeca-9, l2-dien-l-yloxy]propan-l -amine (“Octyl-CLinDMA (2R)”); (2S)-2-((8- [(3P)-cholest-5-en-3-yloxy]octyl)oxy)-N, fsl-dimethyh3-[(9Z, l2Z)-octadeca-9, l2-dien-l - yloxy]propan-l -amine (“Octyl-CLinDMA (2S)”); 2,2-dilinoleyl-4-dimethylaminoethyl-[l,3]- dioxolane (“DLin-K-XTC2-DMA”); and 2-(2,2-di((9Z,l2Z)-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.

[0153] In some embodiments, one or more cationic lipids suitable for the compositions and methods of the present invention include 2,2-Dilinoleyl-4-dimethylaminoethyl-[l,3]-dioxolane (“XTC”); (3aR,5s,6aS)-N,N-dimethyl-2,2-di((9Z,l2Z)-octadeca-9,l2-dien yl)tetrahydro-3aH- cyclopenta[d] [1 ,3]dioxol-5-amine (“ALNY-100”) and/or 4,7, l3-tris(3-oxo-3- (undecylamino)propyl)-Nl,Nl6-diundecyl-4,7,lO,l3-tetraazahex adecane-l,l6-diamide (“NC98-

5”)·

[0154] In some embodiments, the compositions of the present invention 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 of the present invention 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 of the present invention 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 of the present invention 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.

Non-cationic/Helper Lipids

[0155] As used herein, the phrase "non-cationic lipid" refers to any neutral, zwitterionic or anionic lipid. As used herein, the phrase "anionic lipid" refers to any of a number of lipid species that carry a net negative charge at a selected pH, such as physiological pH. Non-cationic lipids include, but are not limited to, distearoylphosphatidylcholine (DSPC),

dioleoylphosphatidylcholine (DOPC), dipalmitoylphosphatidylcholine (DPPC),

dioleoylphosphatidylglycerol (DOPG), dipalmitoylphosphatidylglycerol (DPPG),

dioleoylphosphatidylethanolamine (DOPE), palmitoyloleoylphosphatidylcholine (POPC), palmitoyloleoyl-phosphatidylethanolamine (POPE), dioleoyl-phosphatidylethanolamine 4-(N- maleimidomethyl)-cyclohexane-l-carboxylate (DOPE-mal), dipalmitoyl phosphatidyl ethanolamine (DPPE), dimyristoylphosphoethanolamine (DMPE), distearoyl-phosphatidyl- ethanolamine (DSPE), 16-O-monomethyl PE, l6-0-dimethyl PE, l8-l-trans PE, l-stearoyl-2- oleoyl-phosphatidyethanolamine (SOPE), or a mixture thereof.

[0156] In some embodiments, non-cationic lipids may constitute at least about 5%, 10%,

15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65% or 70% of the total lipids in a suitable lipid solution by weight or by molar. In some embodiments, non-cationic lipid(s) constitute(s) about 30-50 % (e.g., about 30-45%, about 30-40%, about 35-50%, about 35-45%, or about 35-40%) of the total lipids in a suitable lipid solution by weight or by molar.

Cholesterol-based Lipids

[0157] In some embodiments, a suitable lipid solution includes one or more cholesterol- based lipids. For example, suitable cholesterol-based cationic lipids include, for example, DC- Choi (N,N-dimethyl-N-ethylcarboxamidocholesterol), 1 ,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 ICE. In some embodiments, cholesterol-based lipid(s) constitute(s) at least about 5%, 10%, 20%, 30%, 40%, 50%, 60%, or 70% of the total lipids in a suitable lipid solution by weight or by molar. In some embodiments, cholesterol-based lipid(s) constitute(s) about 30-50 % (e.g., about 30-45%, about 30-40%, about 35-50%, about 35-45%, or about 35-40%) of the total lipids in a suitable lipid solution by weight or by molar.

[0158] Exemplary combinations of cationic lipids, non-cationic lipids, cholesterol-based lipids, and PEG-modified lipids are described in the Examples section. For example, a suitable lipid solution may contain CKK-E12, DOPE, cholesterol, and DMG-PEG2K; C 12-200, DOPE, cholesterol, and DMG-PEG2K; HGT5000, DOPE, cholesterol, and DMG-PEG2K; HGT5001, DOPE, cholesterol, and DMG-PEG2K; CKK-E12, DPPC, cholesterol, and DMG-PEG2K; 02- 200, DPPC, cholesterol, and DMG-PEG2K; HGT5000, DPPC, cholesterol, and DMG-PEG2K; or HGT5001, DPPC, cholesterol, and DMG-PEG2K. The selection of cationic lipids, non- cationic lipids and/or PEG-modified lipids which comprise the lipid mixture as well as the relative molar ratio of such lipids to each other, is based upon the characteristics of the selected lipid(s) and the nature of the and the characteristics of the mRNA to be encapsulated. Additional considerations include, for example, the saturation of the alkyl chain, as well as the size, charge, pH, pKa, fusogenicity and toxicity of the selected lipid(s). Thus the molar ratios may be adjusted accordingly.

mRNA-loaded Nanoparticles [0159] Any desired lipids may be mixed at any ratios suitable for encapsulating mRNAs.

In some embodiments, a suitable lipid solution contains a mixture of desired lipids including cationic lipids, non-cationic lipids, cholesterol and/or PEGylated lipids.

[0160] In some embodiments, a process for encapsulating mRNA in lipid nanoparticles comprises mixing an mRNA solution and a lipid solution, wherein the mRNA solution and/or the lipid solution are heated to a pre-determined temperature greater than ambient temperature prior to mixing to form lipid nanoparticles that encapsulate mRNA (see U.S. Patent Application Serial No. 14/790,562 entitled“Encapsulation of messenger RNA”, filed July 2, 2015 and its provisional U.S. patent application Serial No. 62/020,163, filed July 2, 2014, the disclosure of which are hereby incorporated in their entirety).

[0161] In some embodiments, a process for encapsulating mRNA in lipid nanoparticles comprises combining pre-formed lipid nanoparticles with mRNA (see U.S. Provisional

Application Serial No. 62/420,413, filed November 10, 2016 and U.S. Provisional Application Serial No. 62/580,155, filed November 1, 2017, the disclosures of which are hereby incorporated by reference). In some embodiments, combining pre-formed lipid nanoparticles with mRNA results in lipid nanoparticles that show improved efficacy of intracellular delivery of the mRNA. In some embodiments, combining pre-formed lipid nanoparticles with mRNA results in very high encapsulation efficiencies of mRNA encapsulated in lipid nanoparticles (i.e., in the range of 90-95%). In some embodiments, combining pre-formed lipid nanoparticles with mRNA is achieved with pump systems which maintain the lipid/mRNA (N/P) ratio constant throughout the process and which also afford facile scale-up.

[0162] Suitable liposomes in accordance with the present invention may be made in various sizes. In some embodiments, provided liposomes may be made smaller than previously known mRNA encapsulating liposomes. In some embodiments, decreased size of liposomes is associated with more efficient delivery of mRNA. Selection of an appropriate liposome size may take into consideration the site of the target cell or tissue and to some extent the application for which the liposome is being made.

[0163] In some embodiments, an appropriate size of liposome is selected to facilitate systemic distribution of antibody encoded by the mRNA. In some embodiments, it may be desirable to limit transfection of the mRNA to certain cells or tissues. For example, to target hepatocytes a liposome may be sized such that its dimensions are smaller than the fenestrations of the endothelial layer lining hepatic sinusoids in the liver; in such cases the liposome could readily penetrate such endothelial fenestrations to reach the target hepatocytes. [0164] Alternatively or additionally, a liposome may be sized such that the dimensions of the liposome are of a sufficient diameter to limit or expressly avoid distribution into certain cells or tissues. For example, a liposome may be sized such that its dimensions are larger than the fenestrations of the endothelial layer lining hepatic sinusoids to thereby limit distribution of the liposomes to hepatocytes.

[0165] In some embodiments, the size of a liposome is determined by the length of the largest diameter of the liposome particle. In some embodiments, a suitable liposome has a size no greater than about 250 nm (e.g., no greater than about 225 nm, 200 nm, 175 nm, 150 nm, 125 nm, 100 nm, 75 nm, or 50 nm). In some embodiments, a suitable liposome has a size ranging from about 10 - 250 nm (e.g., ranging from about 10 - 225 nm, 10 - 200 nm, 10 - 175 nm, 10 - 150 nm, 10 - 125 nm, 10 - 100 nm, 10 - 75 nm, or 10 - 50 nm). In some embodiments, a suitable liposome has a size ranging from about 100 - 250 nm (e.g., ranging from about 100 - 225 nm, 100 - 200 nm, 100 - 175 nm, 100 - 150 nm). In some embodiments, a suitable liposome has a size ranging from about 10 - 100 nm (e.g., ranging from about 10 - 90 nm, 10 - 80 nm, 10 - 70 nm, 10 - 60 nm, or 10 - 50 nm). In a particular embodiment, a suitable liposome has a size less than about 100 nm.

[0166] A variety of alternative methods known in the art are available for sizing of a population of liposomes. One such sizing method is described in U.S. Pat. No. 4,737,323, incorporated herein by reference. Sonicating a liposome suspension either by bath or probe sonication produces a progressive size reduction down to small ULV less than about 0.05 microns in diameter. Homogenization is another method that relies on shearing energy to fragment large liposomes into smaller ones. In a typical homogenization procedure, MLV are recirculated through a standard emulsion homogenizer until selected liposome sizes, typically between about 0.1 and 0.5 microns, are observed. The size of the liposomes may be determined by quasi-electric light scattering (QELS) as described in Bloomfield, Ann. Rev. Biophys.

Bioeng., 10:421-150 (1981), incorporated herein by reference. Average liposome diameter may be reduced by sonication of formed liposomes. Intermittent sonication cycles may be alternated with QELS assessment to guide efficient liposome synthesis.

Pharmaceutical Compositions

[0167] To facilitate expression of mRNA in vivo , delivery vehicles such as lipid nanoparticles, including liposomes, can be formulated in combination with one or more additional nucleic acids, carriers, targeting ligands or stabilizing reagents, or in pharmacological compositions where it is mixed with suitable excipients. In some embodiments, the lipid nanoparticles encapsulating mRNA are simultaneously administrated with hyaluronidase.

Techniques for formulation and administration of drugs may be found in "Remington's

Pharmaceutical Sciences," Mack Publishing Co., Easton, Pa., latest edition.

[0168] Provided liposomally-encapsulated or associated mRNAs, and compositions containing the same, may be administered and dosed in accordance with current medical practice, taking into account the clinical condition of the subject, the site and method of administration, the scheduling of administration, the subject's age, sex, body weight and other factors relevant to clinicians of ordinary skill in the art. The "effective amount" for the purposes herein may be determined by such relevant considerations as are known to those of ordinary skill in experimental clinical research, pharmacological, clinical and medical arts. In some embodiments, the amount administered is effective to achieve at least some stabilization, improvement or elimination of symptoms and other indicators as are selected as appropriate measures of disease progress, regression or improvement by those of skill in the art. For example, a suitable amount and dosing regimen is one that causes at least transient protein (e.g., enzyme) production.

[0169] Although the current invention focuses on subcutaneous delivery, which is a bolus injection into the subcutis (the tissue layer between the skin and the muscle), other 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, 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 mRNA to a muscle cell. In some embodiments the administration results in delivery of the mRNA to a hepatocyte (i.e., liver cell). In a particular embodiment, the intramuscular administration results in delivery of the mRNA to a muscle cell.

[0170] Alternatively or additionally, liposomally encapsulated mRNAs and compositions of the invention may be administered in a local rather than systemic manner.

[0171] Provided methods of the present invention contemplate single as well as multiple administrations of a therapeutically effective amount of the therapeutic agents (e.g., mRNA encoding a therapeutic protein) described herein. Therapeutic agents can be administered at regular intervals, depending on the nature, severity and extent of the subject’s condition (e.g., OTC deficiency). In some embodiments, a therapeutically effective amount of the therapeutic agent (e.g., mRNA encoding a therapeutic protein) of the present invention may be administered subcutaneously periodically at regular intervals (e.g., once every year, once every six months, once every five months, once every three months, bimonthly (once every two months), monthly (once every month), biweekly (once every two weeks), twice a month, once every 30 days, once every 28 days, once every 14 days, once every 10 days, once every 7 days, weekly, twice a week, daily or continuously.

[0172] In some embodiments, provided liposomes and/or compositions are formulated such that they are suitable for extended-release of the mRNA contained therein. Such extended- release compositions may be conveniently administered to a subject at extended dosing intervals. For example, in some embodiments, the compositions of the present invention are administered to a subject twice a day, daily or every other day. In a preferred embodiment, the compositions of the present invention are administered to a subject twice a week, once a week, once every 7 days, once every 10 days, once every 14 days, once every 28 days, once every 30 days, once every two weeks, once every three weeks, or more preferably once every four weeks, once a month, twice a month, once every six weeks, once every eight weeks, once every other month, once every three months, once every four months, once every six months, once every eight months, once every nine months or annually. Also contemplated are compositions and liposomes which are formulated for depot administration (e.g., intramuscularly, subcutaneously, intravitreally) to either deliver or release mRNA over extended periods of time. Preferably, the extended-release means employed are combined with modifications made to the mRNA to enhance stability.

[0173] As used herein, the term“therapeutically effective amount” is largely based on the total amount of the therapeutic agent contained in the pharmaceutical compositions of the present invention. Generally, a therapeutically effective amount is sufficient to achieve a meaningful benefit to the subject (e.g., treating, modulating, curing, preventing and/or ameliorating OTC deficiency). For example, a therapeutically effective amount may be an amount sufficient to achieve a desired therapeutic and/or prophylactic effect. Generally, the amount of a therapeutic agent (e.g., mRNA encoding a therapeutic protein) administered to a subject in need thereof will depend upon the characteristics of the subject. Such characteristics include the condition, disease severity, general health, age, sex and body weight of the subject. One of ordinary skill in the art will be readily able to determine appropriate dosages depending on these and other related factors. In addition, both objective and subjective assays may optionally be employed to identify optimal dosage ranges.

[0174] A therapeutically effective amount is commonly administered in a dosing regimen that may comprise multiple unit doses. For any particular therapeutic protein, a therapeutically effective amount (and/or an appropriate unit dose within an effective dosing regimen) may vary, for example, depending on route of administration, on combination with other pharmaceutical agents. Also, the specific therapeutically effective amount (and/or unit dose) for any particular patient may depend upon a variety of factors including the disorder being treated and the severity of the disorder; the activity of the specific pharmaceutical agent employed; the specific composition employed; the age, body weight, general health, sex and diet of the patient; the time of administration, route of administration, and/or rate of excretion or metabolism of the specific protein employed; the duration of the treatment; and like factors as is well known in the medical arts.

[0175] In some embodiments, the therapeutically effective dose ranges from about 0.005 mg/kg to 500 mg/kg body weight, e.g., from about 0.005 mg/kg to 400 mg/kg body weight, from about 0.005 mg/kg to 300 mg/kg body weight, from about 0.005 mg/kg to 200 mg/kg body weight, from about 0.005 mg/kg to 100 mg/kg body weight, from about 0.005 mg/kg to 90 mg/kg body weight, from about 0.005 mg/kg to 80 mg/kg body weight, from about 0.005 mg/kg to 70 mg/kg body weight, from about 0.005 mg/kg to 60 mg/kg body weight, from about 0.005 mg/kg to 50 mg/kg body weight, from about 0.005 mg/kg to 40 mg/kg body weight, from about 0.005 mg/kg to 30 mg/kg body weight, from about 0.005 mg/kg to 25 mg/kg body weight, from about 0.005 mg/kg to 20 mg/kg body weight, from about 0.005 mg/kg to 15 mg/kg body weight, from about 0.005 mg/kg to 10 mg/kg body weight.

[0176] In some embodiments, the therapeutically effective dose is greater than about 0.1 mg/kg body weight, greater than about 0.5 mg/kg body weight, greater than about 1.0 mg/kg body weight, greater than about 3 mg/kg body weight, greater than about 5 mg/kg body weight, greater than about 10 mg/kg body weight, greater than about 15 mg/kg body weight, greater than about 20 mg/kg body weight, greater than about 30 mg/kg body weight, greater than about 40 mg/kg body weight, greater than about 50 mg/kg body weight, greater than about 60 mg/kg body weight, greater than about 70 mg/kg body weight, greater than about 80 mg/kg body weight, greater than about 90 mg/kg body weight, greater than about 100 mg/kg body weight, greater than about 150 mg/kg body weight, greater than about 200 mg/kg body weight, greater than about 250 mg/kg body weight, greater than about 300 mg/kg body weight, greater than about 350 mg/kg body weight, greater than about 400 mg/kg body weight, greater than about 450 mg/kg body weight, greater than about 500 mg/kg body weight. In a particular embodiment, the therapeutically effective dose is 1.0 mg/kg body weight. In some embodiments, the

therapeutically effective dose of 1.0 mg/kg body weight is administered intramuscularly or intravenously. [0177] Also contemplated herein are lyophilized pharmaceutical compositions comprising one or more of the liposomes disclosed herein and related methods for the use of such compositions as disclosed for example, in International Patent Application

PCT/US 12/41663, filed June 8, 2012, the teachings of which are incorporated herein by reference in their entirety. For example, lyophilized pharmaceutical compositions according to the invention may be reconstituted prior to administration or can be reconstituted in vivo. For example, a lyophilized pharmaceutical composition can be formulated in an appropriate dosage form (e.g., an intradermal dosage form such as a disk, rod or membrane) and administered such that the dosage form is rehydrated over time in vivo by the individual's bodily fluids.

[0178] Provided liposomes and compositions may be administered to any desired tissue.

In some embodiments, the provided liposomes and compositions comprising mRNA are delivered subcutaneously and the mRNA is expressed in a cell or tissue type other than the subcutis. In some embodiments, the mRNA encoding a target protein delivered by provided liposomes or compositions is expressed in the tissue in which the liposomes and/or compositions were administered. In some embodiments, the mRNA delivered is expressed in a tissue different from the tissue in which the liposomes and/or compositions were administered. 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.

[0179] In some embodiments, administering a provided composition results in increased expression of the mRNA administered, or increased activity level of the mRNA-encoded protein in a biological sample from a subject as compared to a baseline expression or activity level before treatment or administration. In some embodiments, administering a provided composition results in increased expression or activity level of the therapeutic protein encoded by the mRNA of a provided composition in a biological sample from a subject as compared to a baseline expression or activity level before treatment. Typically, the baseline level is measured immediately before treatment. Biological samples include, for example, whole blood, serum, plasma, urine and tissue samples (e.g., muscle, liver, skin fibroblasts). In some embodiments, administering a provided composition results in increased therapeutic protein (protein encoded by administered mRNA) expression or activity level by at least about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 95% as compared to the baseline level immediately before treatment. In some embodiments, administering a provided composition results in increased mRNA expression or activity level in a biological sample from a subject as compared to subjects who were not treated. In some embodiments, administering a provided composition results in increased expression or activity level of the therapeutic protein encoded by the mRNA of a provided composition in a biological sample from a subject as compared to subjects who were not treated.

[0180] According to various embodiments, the timing of expression of delivered mRNAs can be tuned to suit a particular medical need. In some embodiments, the expression of the protein encoded by delivered mRNA is detectable 1, 2, 3, 6, 12, 24, 48, 72, 96 hours, 1 week, 2 weeks, or 1 month after administration of provided liposomes and/or compositions.

[0181] In some embodiments, a therapeutically effective dose of the provided

composition, when administered regularly, results in increased citrulline production in a subject as compared to baseline citrulline production before treatment. Typically, the citrulline level before or after the treatment may be measured in a biological sample obtained from the subject such as blood, plasma or serum, urine, or solid tissue extracts. In some embodiments, treatment according to the present invention results in an increase of the citrulline level in a biological sample (e.g., plasma, serum, or urine) by at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, l-fold, 1.5-fold, 2-fold, 2.5-fold, or 3-fold as compared to the base line citrulline level.

[0182] According to the present invention, a therapeutically effective dose of the provided composition, when administered regularly, results in at least one symptom or feature of a protein deficiency being reduced in intensity, severity, or frequency or having delayed onset.

Therapeutic Application

[0183] The present invention may be used to treat various diseases, disorders and conditions. Of particular interest, monogenic disorders and disorders where administering an mRNA encoding a protein reduces one or more disease related symptoms, or ameliorates the disease symptoms, are candidates for therapeutic application using the present invention.

Exemplary therapeutic messenger RNAs for subcutaneous administration as delineated in the present application disclosure can be selected from any of the corresponding exemplary genes listed in Tables 1, 2, 3, 4, 5 or 6 having the related functions, or implicated in the disease or conditions as described.

TABLE 1

TABLE 2

TABLE 3

TABLE 4

TABLE 5

TABLE 6 - Secreted Proteins

[0184] In some embodiments, the present invention is useful in treating a disease or disorder listed in Table 1.

[0185] In some embodiments, the present invention is useful in delivering vaccines.

Vaccines delivered subcutaneously include vaccines against infectious diseases which include but are not limited to diphtheria, tetanus, pertussis, poliomyelitis, measles, mumps, rubella, haemophilus influenzae type b infections, hepatitis B, influenza, pneumococcal infections, cholera, hepatitis A, meningococcal disease, plague, rabies, bat lyssavirus, yellow fever, Japanese encephalitis, Q fever, tuberculosis, typhoid and varicella-zoster. Vaccines delivered subcutaneously may also include vaccines against cell proliferative disorders such as cancers. In some embodiments, subcutaneously delivered vaccines include cancer vaccines for

lymphoproliferative disorders. In some embodiments, the cancer vaccines include

subcutaneously delivered mRNA encoding immunogenic agents that direct cellular immune response against cancer cells, using the method of the invention. In some embodiments, a vaccine comprising mRNA encoding MHC-class specific peptides comprising one or more cancer antigenic epitopes is administered subcutaneously with an mRNA encoding hyaluronidase, which can result in superior systemic delivery of the vaccine and more robust antigenic response.

[0186] In some embodiments, the present invention is useful in treating a liver disease, for example OTC deficiency. Co-injection of mRNA encoding an OTC protein with a hyaluronidase enzyme results in an increased level of OTC enzyme (protein) in a liver cell (e.g., a hepatocyte) of a subject as compared to a baseline level before treatment. Typically, the baseline level is measured before treatment (e.g., up to 12 months prior to the treatment and in some instances, immediately before the treatment). In some embodiments, subcutaneous injection according to the present invention results in an increased OTC protein level in the liver cell by at least about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 95% as compared to a baseline level before treatment. In some embodiments, subcutaneous injection according to the present invention results in an increased OTC protein level in a liver cell as compared to the OTC protein level a liver cell of subjects who are not treated.

[0187] In some embodiments, subcutaneous injection according to the present invention results in an increased OTC protein level in plasma or serum of subject as compared to a baseline level before treatment. Typically, the baseline level is measured before treatment (e.g., up to 12 months prior to the treatment and in some instances, immediately before the treatment). In some embodiments, administering the provided composition results in an increased OTC protein level in plasma or serum by at least about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 95% as compared to a baseline level before treatment. In some embodiments, administering the provided composition results in an increased OTC protein level in plasma or serum as compared to an OTC protein level in plasma or serum of subjects who are not treated.

[0188] The compositions and methods of the invention provide for the delivery of mRNA to treat a number of disorders. In particular, the compositions and methods of the present invention are suitable for the treatment of diseases or disorders relating to the deficiency of proteins and/or enzymes that are excreted or secreted in the liver. These include but are not limited to: Phenylalanine hydroxylase (PAH) deficiency (classically known as phenylketonuria, PKU), argininosuccinate synthase 1 (ASS1) deficiency, which causes a liver urea cycle disorder citrullinaemia, erythropoietin (EPO) deficiency, which leads to anemia, erythropoietin being a protein produced both in the kidney and in the liver.

[0189] Disorders for which the present invention are useful include, but are not limited to, disorders such as Fabry disease; hemophilic diseases (such as, e.g., hemophilia B (FIX), hemophilia A (FVIII); SMNl-related spinal muscular atrophy (SMA); amyotrophic lateral sclerosis (ALS); GALT-related galactosemia; COL4A5 -related disorders including Alport syndrome; galactocerebrosidase deficiencies; X-linked adrenoleukodystrophy; Friedreich’s ataxia; Pelizaeus-Merzbacher disease; TSC1 and TSC2-related tuberous sclerosis; Sanfilippo B syndrome (MPS MB); the FMR1 -related disorders which include Fragile X syndrome, Fragile X-Associated Tremor/ Ataxia Syndrome and Fragile X Premature Ovarian Failure Syndrome; Prader-Willi syndrome; hereditary hemorrhagic telangiectasia (AT); Niemann-Pick disease Type Cl; the neuronal ceroid lipofuscinoses-related diseases including Juvenile Neuronal Ceroid Lipofuscinosis (JNCL), Juvenile Batten disease, Santavuori-Haltia disease, Jansky-Bielschowsky disease, and PTT-l and TPP1 deficiencies; EIF2B1, EIF2B2, EIF2B3, EIF2B4 and EIF2B5- related childhood ataxia with central nervous system hypomyelination/vanishing white matter; CACNA1A and CACNB4-related Episodic Ataxia Type 2; the MECP2-related disorders including Classic Rett Syndrome, MECP2-related Severe Neonatal Encephalopathy and PPM-X Syndrome; CDKL5-related Atypical Rett Syndrome; Kennedy’s disease (SBMA); Notch-3 related cerebral autosomal dominant arteriopathy with subcortical infarcts and

leukoencephalopathy (CADASIL); SCN1A and SCNlB-related seizure disorders; the

Polymerase G-related disorders which include Alpers-Huttenlocher syndrome, POLG-related sensory ataxic neuropathy, dysarthria, and ophthalmoparesis, and autosomal dominant and recessive progressive external ophthalmoplegia with mitochondrial DNA deletions; X-Linked adrenal hypoplasia; X-linked agammaglobulinemia; and Wilson’s disease.

[0190] In some embodiments, the nucleic acids, and in particular mRNA, of the invention may encode functional proteins or enzymes that are secreted into extracellular space. For example, the secreted proteins include clotting factors, components of the complement pathway, cytokines, chemokines, chemoattractants, protein hormones (e.g. EGF, PDF), protein components of serum, antibodies, secretable toll-like receptors, and others. In some

embodiments, the compositions of the present invention may include mRNA encoding erythropoietin, al -antitrypsin, carboxypeptidase N or human growth hormone.

EXAMPLES

[0191] 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. Lipid Materials

[0192] The formulations described in the following Examples, unless otherwise specified, contain a multi-component lipid mixture of varying ratios employing one or more cationic lipids, helper lipids (e.g., non-cationic lipids and/or cholesterol lipids) and PEGylated lipids designed to encapsulate various nucleic acid materials. Cationic lipids for the process can include, but are not limited to, CKK-E12 (3,6-bis(4-(bis(2- hydroxydodecyl)amino)butyl)piperazine-2,5-dione), OF-02, Target 23, Target 24, ICE,

HGT5000, HGT5001, HGT4003, DOTAP (l,2-dioleyl-3-trimethylammonium propane),

DODAP (l,2-dioleyl-3-dimethylammonium propane), DOTMA (l,2-di-0-octadecenyl-3- trimethylammonium propane), DLinDMA (Heyes, J.; Palmer, L.; Bremner, K.; MacLachlan, I. “Cationic lipid saturation influences intracellular delivery of encapsulated nucleic acids” J.

Contr. Rel. 2005, 107, 276-287), DLin-KC2-DMA (Semple, S.C. et al.“Rational Design of Cationic Lipids for siRNA Delivery” Nature Biotech. 2010, 28, 172-176), C12-200 (Love, K.T. et al.“Lipid-like materials for low-dose in vivo gene silencing” PNAS 2010, 107, 1864-1869), , dialkylamino-based, imidazole-based, guanidinium-based, etc. Helper lipids can include, but are not limited, to DSPC (l,2-distearoyl-sn-glycero-3-phosphocholine), DPPC (l,2-dipalmitoyl-sn- glycero-3-phosphocholine), DOPE (l,2-dioleyl-sn-glycero-3-phosphoethanolamine), DPPE (1,2- dipalmitoyl- sn-glycero-3 -phosphoethanolamine) , DMPE ( 1 ,2-dimyristoyl- sn-glycero-3 - phosphoethanolamine), DOPG (l,2-dioleoyl-sn-glycero-3-phospho-(l'-rac-glycerol)), DOPC (l,2-dioleyl-sn-glycero-3-phosphotidylcholine), cholesterol, etc. PEGylated lipids can include, but are not limited to, a poly(ethylene) glycol chain of up to 5 kDa in length covalently attached to a lipid with alkyl chain(s) of C6-C20 length. mRNA Materials

[0193] In some embodiments, codon-optimized messenger RNA encoding target protein was synthesized by in vitro transcription from a plasmid DNA template encoding the gene, which was followed by the addition of a 5’ cap structure (Cap 1) (Fechter, P.; Brownlee, G.G. “Recognition of mRNA cap structures by viral and cellular proteins” J. Gen. Virology 2005, 86, 1239-1249) and a 3’ poly(A). 5’ and 3’ untranslated regions present in each mRNA product are represented as X and Y, respectively and defined as stated previously.

Example 1. In vivo expression of firefly lucif erase protein in mice

[0194] This example illustrates an exemplary method of administering firefly luciferase

(FFL) mRNA-loaded LNPs and methods for analyzing firefly luciferase in target tissues in vivo. Wild type mice are treated with LNPs encapsulating mRNA encoding FFL at 20 mg/kg co formulated with hyaluronidase mRNA at 20 mg/kg by subcutaneous delivery. The luminescence produced by FFL protein is observed at 3, 24 and 48 hours post-subcutaneous administration. Significant luminescence is observed representing the successful production of active FFL protein in the livers of these mice. Further, sustained FFL activity is maintained for at least 24 hours with little to no decrease in intensity.

Example 2. In vivo activity of expressed hOTC in mice

[0195] This example shows a comparison of intravenous administration without hyaluronidase and subcutaneous administration with and without an mRNA encoding hyaluronidase in OTC KO spf sh mice and human OTC (hOTC) mRNA-loaded lipid

nanoparticles. In this example, hOTC and hyaluronidase mRNAs are present in the same formulation and therefore are administered simultaneously. The hOTC protein is shown to be enzymatically active, as determined by measuring levels of citrulline production using a custom ex vivo activity assay. Generally, the production of citrulline can be used to evaluate the activity of the expressed hOTC protein. Citrulline activity of hOTC protein is measured in the liver extracts of mice sacrificed 24 hours after the single dose of the lipid nanoparticles encapsulating hOTC mRNA at 20 mg/kg is delivered subcutaneously with and without hyaluronidase mRNA (20 mg/kg). Citrulline activity in the livers of saline-treated OTC KO mice is also measured. No significant hOTC protein activity is observed after subcutaneous administration of hOTC mRNA without hyaluronidase mRNA co-formulation. hOTC protein activity in those animals is similar to those seen in animals treated with saline. In contrast, hOTC protein activity (as evidenced by citrulline protein levels) is similar in the livers of mice which are administered the hOTC mRNA LNP composition intravenously and those administered the hOTC mRNA LNP composition formulated with hyaluronidase-encoding mRNA subcutaneously. A hyaluronidase mRNA dose dependence on the robustness of OTC mRNA expression can be tested using varying doses of hyaluronidase mRNA in the formulation.

Example 3. In vivo efficiency of CO-hOTC mRNA delivery in mice

[0196] This example shows a comparison of intravenous administration without hyaluronidase versus subcutaneous administration with and without the mRNA encoding hyaluronidase in OTC KO spf sh mice using CO-hOTC (codon-optimized human OTC) mRNA- loaded lipid nanoparticles. Subcutaneously delivered CO-hOTC mRNA lipid nanoparticles co- formulated with hyaluronidase mRNA are more effective than subcutaneously delivered mRNA lipid nanoparticles without the mRNA encoding hyaluronidase.

[0197] Efficiency of administration was determined by comparing CO-hOTC mRNA copy number in the livers of the various treatment groups. CO-hOTC mRNA copy number in the livers of mice is measured 24 hours after a single subcutaneous dose of 20 mg/kg CO-hOTC mRNA and 20 mg/kg hyaluronidase mRNA (SEQ ID NO: 12) LNP formulation. A control set comprise OTC mRNA, without hyaluronidase mRNA. For comparison, CO-hOTC mRNA copy number is also measured in livers of mice 24 hours after a CO-hOTC mRNA LNP solution is injected intravenously at 0.50 mg/kg. mOTC mRNA copy number is measured in the livers of saline-treated wild type (WT) mice, saline-treated OTC KO mice, and OTC KO mice treated intravenously with hOTC LNP solution, subcutaneously with hOTC LNP formulation free of hyaluronidase or subcutaneously with hOTC LNP co-formulated with hyaluronidase.

Example 4. In vivo expression of human erythropoietin (hEPO) in mice

[0198] This example illustrates an exemplary time course of human erythropoietin

(hEPO) protein expression following subcutaneous administration of hEPO encoding mRNA using the method disclosed, in comparison with intravenous administration of the same.

[0199] Male CD1 mice are administered either an intravenous dose of hEPO mRNA- loaded lipid nanoparticles at a dosage of 1 mg/kg or a subcutaneous dose of hEPO mRNA- loaded lipid nanoparticles at a dosage of 5 mg/kg co-formulated with 5 mg/kg hyaluronidase mRNA once on day 1. Human EPO protein expression is examined in serum samples by hEPO- specific ELISA for 4 days.

[0200] High level of EPO protein expression is observed in both intravenous- administered and subcutaneous-administered groups of mice at 6 hours after mRNA

administration (Day 1) and on Day 2. The expression level is compared to intravenous administration for the same mRNA LNP.

EQUIVALENTS

[0201] Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the invention described herein. The scope of the present invention is not intended to be limited to the above Description, but rather is as set forth in the following claims: