MRNA therapy for argininosuccinate synthetase deficiency

Information

  • Patent Grant
  • 11224642
  • Patent Number
    11,224,642
  • Date Filed
    Wednesday, October 22, 2014
    9 years ago
  • Date Issued
    Tuesday, January 18, 2022
    2 years ago
Abstract
The present invention provides, among other things, methods of treating Argininosuccinate Synthetase Deficiency (ASD), including administering to a subject in need of treatment a composition comprising an mRNA encoding argininosuccinate synthetase (ASS1) at an effective dose and an administration interval such that at least one symptom or feature of ASD is reduced in intensity, severity, or frequency or has delayed in onset. In some embodiments, the mRNA is encapsulated in a liposome comprising one or more cationic lipids, one or more non-cationic lipids, one or more cholesterol-based lipids and one or more PEG-modified lipids.
Description
BACKGROUND

Argininosuccinate Synthetase Deficiency (ASD) is an autosomal recessive metabolic genetic disorder characterized by a mutation in the gene for the enzyme argininosuccinate synthetase (ASS1), affecting its ability to bind to citrulline, aspartate and other molecules. Defects in the ASS protein disrupt the urea cycle and prevent the liver from properly processing excess nitrogen into urea. An accumulation of ammonia and other byproducts of the urea cycle (such as citrulline) is toxic and when it occurs during the first few days of life can lead to symptoms such as lack of energy (lethargy) poor feeding, vomiting, seizures and loss of consciousness. Currently, there is no cure for the disease and standard of care is through management of diet, minimizing foods that contain high amounts of protein, and dietary supplements of arginine and phenylacetate.


SUMMARY OF THE INVENTION

The present invention provides, among other things, improved methods and compositions for the treatment of Argininosuccinate Synthetase Deficiency (ASD) based on mRNA therapy. The invention encompasses the observation that administration of an mRNA encoding a human ASS1 protein, encapsulated within a liposome, resulted in highly efficient and sustained protein production in vivo and successful reduction of plasma ammonia levels, a clinically-relevant disease marker.


In one aspect, the present invention provides a method of treating ASD, including administering to a subject in need of treatment a composition comprising an mRNA encoding argininosuccinate synthetase (ASS1) at an effective dose and an administration interval such that at least one symptom or feature of ASD is reduced in intensity, severity, or frequency or has delayed in onset. In some embodiments, the mRNA is encapsulated within a liposome.


In another aspect, the present invention provides compositions for treating ASD comprising an mRNA encoding ASS1 at an effective dose amount encapsulated within a liposome.


In some embodiments, a suitable liposome comprises one or more cationic lipids, one or more non-cationic lipids, one or more cholesterol-based lipids and one or more PEG-modified lipids.


In some embodiments, the one or more cationic lipids are selected from the group consisting of C12-200, MC3, DLinDMA, DLinkC2DMA, cKK-E12, ICE (Imidazol-based), HGT5000, HGT5001, DODAC, DDAB, DMRIE, DOSPA, DOGS, DODAP, DODMA and DMDMA, DODAC, DLenDMA, DMRIE, CLinDMA, CpLinDMA, DMOBA, DOcarbDAP, DLinDAP, DLincarbDAP, DLinCDAP, KLin-K-DMA, DLin-K-XTC2-DMA, HGT4003, and combinations thereof.


In some embodiments, the one or more cationic lipids comprise a compound of formula I-c1-a:




embedded image



or a pharmaceutically acceptable salt thereof, wherein:


each R2 independently is hydrogen or C1-3 alkyl;


each q independently is 2 to 6;


each R′ independently is hydrogen or C1-3 alkyl; and each RL independently is C8-12 alkyl.


In some embodiments, the one or more cationic lipids comprise cKK-E12:




embedded image


In some embodiments, the one or more non-cationic lipids suitable for the invention are selected from DSPC (1,2-distearoyl-sn-glycero-3-phosphocholine), DPPC (1,2-dipalmitoyl-sn-glycero-3-phosphocholine), DOPE (1,2-dioleyl-sn-glycero-3-phosphoethanolamine), DOPC (1,2-dioleyl-sn-glycero-3-phosphotidylcholine) DPPE (1,2-dipalmitoyl-sn-glycero-3-phosphoethanolamine), DMPE (1,2-dimyristoyl-sn-glycero-3-phosphoethanolamine), DOPG (,2-dioleoyl-sn-glycero-3-phospho-(1′-rac-glycerol)), and combinations thereof.


In some embodiments, the one or more cholesterol-based lipids are selected from cholesterol, PEGylated cholesterol and DC-Chol (N,N-dimethyl-N-ethylcarboxamidocholesterol), 1,4-bis(3-N-oleylamino-propyl)piperazine.


In some embodiments, the liposome further comprises one or more PEG-modified lipids. In some embodiments, the one or more PEG-modified lipids comprise 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. In some embodiments, a PEG-modified lipid is a derivatized ceramide such as N-Octanoyl-Sphingosine-1-[Succinyl(Methoxy Polyethylene Glycol)-2000]. In some embodiments, a PEG-modified or PEGylated lipid is PEGylated cholesterol or Dimyristoylglycerol (DMG)-PEG-2K.


In some embodiments, a suitable liposome comprises a combination selected from cKK-E12, DOPE, cholesterol and DMG-PEG2K; C12-200, DOPE, cholesterol and DMG-PEG2K; HGT4003, DOPE, cholesterol and DMG-PEG2K; or ICE, DOPE, cholesterol and DMG-PEG2K.


In some embodiments, cationic lipids (e.g., cKK-E12, C12-200, ICE, and/or HGT4003) constitute about 30-60% (e.g., about 30-55%, about 30-50%, about 30-45%, about 30-40%, about 35-50%, about 35-45%, or about 35-40%) of the liposome by molar ratio. In some embodiments, cationic lipids (e.g., cKK-E12, C12-200, ICE, and/or HGT4003) constitute about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, or about 60% of the liposome by molar ratio.


In some embodiments, the ratio of cationic lipid(s) (e.g., cKK-E12, C12-200, ICE, and/or HGT4003) to non-cationic lipid(s) (e.g., DOPE) to cholesterol-based lipid(s) (e.g., cholesterol) to PEGylated lipid(s) (e.g., DMG-PEG2K) may be between about 30-60:25-35:20-30:1-15, respectively. In some embodiments, the ratio of cationic lipid(s) (e.g., cKK-E12, C12-200, ICE, and/or HGT4003) to non-cationic lipid(s) (e.g., DOPE) to cholesterol-based lipid(s) (e.g., cholesterol) to PEGylated lipid(s) (e.g., DMG-PEG2K) is approximately 40:30:20:10, respectively. In some embodiments, the ratio of cationic lipid(s) (e.g., cKK-E12, C12-200, ICE, and/or HGT4003) to non-cationic lipid(s) (e.g., DOPE) to cholesterol-based lipid(s) (e.g., cholesterol) to PEGylated lipid(s) (e.g., DMG-PEG2K) is approximately 40:30:25:5, respectively. In some embodiments, the ratio of cationic lipid(s) (e.g., cKK-E12, C12-200, ICE, and/or HGT4003) to non-cationic lipid(s) (e.g., DOPE) to cholesterol-based lipid(s) (e.g., cholesterol) to PEGylated lipid(s) (e.g., DMG-PEG2K) is approximately 40:32:25:3, respectively. In some embodiments, the ratio of cationic lipid(s) (e.g., cKK-E12, C12-200, ICE, and/or HGT4003) to non-cationic lipid(s) (e.g., DOPE) to cholesterol-based lipid(s) (e.g., cholesterol) to PEGylated lipid(s) (e.g., DMG-PEG2K) is approximately 50:25:20:5.


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 less than about 500 nm, 400 nm, 300 nm, 250 nm, 200 nm, 150 nm, 100 nm, 75 nm, or 50 nm. In some embodiments, a suitable liposome has a size less than about 100 nm, 90 nm, 80 nm, 70 nm, or 60 nm.


In some embodiments, the mRNA is administered at a dose ranging from about 0.1-5.0 mg/kg body weight, for example about 0.1-4.5, 0.1-4.0, 0.1-3.5, 0.1-3.0, 0.1-2.5, 0.1-2.0, 0.1-1.5, 0.1-1.0, 0.1-0.5, 0.1-0.3, 0.3-5.0, 0.3-4.5, 0.3-4.0, 0.3-3.5, 0.3-3.0, 0.3-2.5, 0.3-2.0, 0.3-1.5, 0.3-1.0, 0.3-0.5, 0.5-5.0, 0.5-4.5, 0.5-4.0, 0.5-3.5, 0.5-3.0, 0.5-2.5, 0.5-2.0, 0.5-1.5, or 0.5-1.0 mg/kg body weight. In some embodiments, the mRNA is administered at a dose of or less than about 5.0, 4.5, 4.0, 3.5, 3.0, 2.5, 2.0, 1.5, 1.0, 0.8, 0.6, 0.5, 0.4, 0.3, 0.2, or 0.1 mg/kg body weight.


In some embodiments, provided composition is administered intravenously. In some embodiments, provided composition is administered via pulmonary delivery. In certain embodiments, pulmonary delivery is performed by aerosolization, inhalation, nebulization or instillation. In some embodiments, provided compositions are formulated as respirable particles, nebulizable lipid, or inhalable dry powder.


In some embodiments, provided compositions are administered once daily, once a week, twice a month, once a month. In some embodiments, provided compositions are administered once every 7 days, once every 10 days, once every 14 days, once every 28 days or once every 30 days.


In some embodiments, the ASS1 protein is expressed in liver. In some embodiments, administering the provided composition results in the expression of an ASS1 protein level at or above about 100 ng/mg (e.g., at or above about 200 ng/mg, 400 ng/mg, 500 ng/mg, 1000 ng/mg, 2000 ng/mg or 3000 ng/mg) of total protein in the liver.


In some embodiments, administering of the composition results in increased serum ASS1 protein level. In some embodiments, administering of the composition results in increased serum ASS1 protein level by at least about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 1-fold, 2-fold, 3-fold, 4-fold or 5-fold as compared to the baseline serum ASS1 protein level before the treatment.


In some embodiments, administering of the composition results in reduced citrulline level in the subject as compared to the baseline citrulline level before the treatment. In some embodiments, administering of the composition results in reduced plasma citrulline level by at least about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 95% as compared to the baseline plasma citrulline level before the treatment. In some embodiments, administering of the composition results in reduced plasma citrulline level to less than about 2000 μM, 1500 μM, 1000 μM, 750 μM, 500 μM, 250 μM, 100 μM, 90 μM, 80 μM, 70 μM, 60 μM, 50 μM, 40 μM, or 30 μM.


In some embodiments, administering of the composition results in reduced ammonia level in the subject as compared to the baseline ammonia level before the treatment. In some embodiments, administering the provided composition results in reduction of ammonia levels to about 3000 μmol/L or less, about 2750 μmol/L or less, about 2500 μmol/L or less, about 2250 μmol/L or less, about 2000 μmol/L or less, about 1750 μmol/L or less, about 1500 μmol/L or less, about 1250 μmol/L or less, about 1000 μmol/L or less, about 750 μmol/L or less, about 500 μmol/L or less, about 250 μmol/L or less, about 100 μmol/L or less, or about 50 μmol/L or less in the plasma or serum. In a particular embodiment, administering the provided composition results in reduction of ammonia levels to about 50 μmol/L or less in plasma or serum.


In some embodiments, administering the provided composition results in reduced ammonia level in a biological sample by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50% at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, or at least about 95% as compared to baseline ammonia level before treatment. Suitable biological sample may be whole blood, serum, plasma, or urine.


In some embodiments, the mRNA is codon optimized. In some embodiments, the codon-optimized mRNA comprises SEQ ID NO:3, SEQ ID NO:13, SEQ ID NO: 14 or SEQ ID NO:15 (corresponding to codon-optimized human ASS1 mRNA sequences). In some embodiments, the mRNA comprises the 5′ UTR sequence of SEQ ID NO:4 (corresponding to 5′ UTR sequence X). In some embodiments, the mRNA comprises the 3′ UTR sequence of SEQ ID NO:5 (corresponding to a 3′ UTR sequence Y). In some embodiments, the mRNA comprises the 3′ UTR sequence of SEQ ID NO:6 (corresponding to a 3′ UTR sequence Y). In some embodiments, the codon-optimized mRNA comprises SEQ ID NO:7 or SEQ ID NO:8 (corresponding to codon-optimized human ASS1 mRNA sequence with 5′ UTR and 3′ UTR sequences).


In some embodiments, the mRNA comprises one or more modified nucleotides. In some embodiments, the one or more modified nucleotides comprise pseudouridine, N-1-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, O(6)-methylguanine, and/or 2-thiocytidine. In some embodiments, the mRNA is unmodified.


In particular embodiments, the present invention provides a composition for treating ASD comprising an mRNA encoding argininosuccinate synthetase (ASS1) at an effective dose amount encapsulated within a liposome, wherein the mRNA comprises SEQ ID NO:3, SEQ ID NO:13, SEQ ID NO:14 or SEQ ID NO:15, and further wherein the liposome comprises cationic or non-cationic lipid, cholesterol-based lipid and PEG-modified lipid.


In particular embodiments, the present invention provides a composition for treating ASD comprising an mRNA encoding argininosuccinate synthetase (ASS1) at an effective dose amount encapsulated within a liposome, wherein the mRNA comprises SEQ ID NO:7 or SEQ ID NO:8, and further wherein the liposome comprises cationic or non-cationic lipid, cholesterol-based lipid and PEG-modified lipid.


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.





BRIEF DESCRIPTION OF THE DRAWING

The drawings are for illustration purposes only not for limitation.



FIG. 1 depicts exemplary ASS1 protein levels detected via ELISA after treatment with human ASS1 mRNA-loaded cKK-E12-based lipid nanoparticles at various doses.



FIGS. 2A-2D depict exemplary Western blots comparing human ASS1 protein levels in liver as a function of dose after a single intravenous dose of human ASS1 mRNA-encapsulated lipid nanoparticles. CD 1 mice were sacrificed at 24 hours post-administration and livers were harvested and analyzed as described above. Human ASS1 protein was detected using 2H8 mouse monoclonal antibody. 50 micrograms total liver protein was loaded into each well. Recombinant human ASS1 protein was loaded on each gel as a positive control (R5 control).



FIG. 3 depicts an exemplary graph of accumulated human argininosuccinate synthetase (ASS1) protein levels as measured via ELISA. The protein detected was a result of its production from ASS1 mRNA delivered intravenously via a single dose of lipid nanoparticles (1.0 mg/kg encapsulated ASS1 mRNA) over time.



FIGS. 4A-4E depict exemplary Western blots of human ASS1 protein levels in liver over time after a single intravenous dose of human ASS1 mRNA-encapsulated lipid nanoparticles (1.0 mg/kg dose).



FIGS. 5A-5I depict detection of human ASS1 messenger RNA via in situ hybridization in the livers of treated mice. Exogenous mRNA is observable for at least 7 days post-administration after a single dose (1.0 mg/kg) of ASS1 mRNA-loaded cKK-E12-based lipid nanoparticles. Human ASS1 mRNA is detectable in sinusoidal cells as well as hepatocytes.



FIGS. 6A-6I depict exemplary immunohistochemical staining of ASS1 protein levels in mouse liver at various time points after administration of 1 mg/kg ASS1 mRNA containing cKK-E12 lipid nanoparticles. Human ASS1 protein is detectable in sinusoidal cells as well as hepatocytes. Human ASS1 protein is detectable for at least one week post-administration of a single dose of ASS1 mRNA-loaded lipid nanoparticles.



FIGS. 7A-7B depict low magnification (4×) immunohistochemical staining of ASS1 protein levels in mouse liver 24 hours after administration of 1 mg/kg ASS1 mRNA-containing cKK-E12 liposomes. A comparison to untreated mouse liver (left) demonstrates the widespread distribution of human ASS1 protein throughout the liver.



FIG. 8 depicts an exemplary graph of human argininosuccinate synthetase (ASS1) protein levels as measured via ELISA. The protein detected was a result of its production from ASS1 mRNA delivered intravenously via a single dose of various lipid nanoparticles.



FIG. 9 depicts 14C Arginine incorporation into proteins after transfection of ASS1 mRNA in an ASS1 KO cell line (SK (−)) as compared to a stably-expressing positive AS1 cell line (SK (+), Clone #5). Control represents lipofectamine-only treated SK (−) cells.



FIG. 10 depicts human ASS1 protein levels in rat liver 24 hours after administration of ASS1 mRNA-loaded lipid nanoparticles.



FIG. 11 depicts plasma ammonia levels in AAS1 knockout mice administered 1.0 mg/kg of ASS1 mRNA-loaded lipid nanoparticles every 14 days for 30 days.





DEFINITIONS

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


Alkyl: As used herein, “alkyl” refers to a radical of a straight-chain or branched saturated hydrocarbon group having from 1 to 15 carbon atoms (“C1-15 alkyl”). In some embodiments, an alkyl group has 1 to 3 carbon atoms (“C1-3 alkyl”). Examples of C1-3 alkyl groups include methyl (C1), ethyl (C2), n-propyl (C3), and isopropyl (C3). In some embodiments, an alkyl group has 8 to 12 carbon atoms (“C8-12 alkyl”). Examples of C8-12 alkyl groups include, without limitation, n-octyl (C8), n-nonyl (C9), n-decyl (C10), n-undecyl (C11), n-dodecyl (C12) and the like. The prefix “n-” (normal) refers to unbranched alkyl groups. For example, n-C8 alkyl refers to —(CH2)7CH3, n-C10 alkyl refers to —(CH2)9CH3, etc.


Amino acid: As used herein, term “amino acid,” in its broadest sense, refers to any compound and/or substance that can be incorporated into a polypeptide chain. In some embodiments, an amino acid has the general structure H2N—C(H)(R)—COOH. In some embodiments, an amino acid is a naturally occurring amino acid. In some embodiments, an amino acid is a synthetic amino acid; in some embodiments, an amino acid is a d-amino acid; in some embodiments, an amino acid is an 1-amino acid. “Standard amino acid” refers to any of the twenty standard 1-amino acids commonly found in naturally occurring peptides. “Nonstandard amino acid” refers to any amino acid, other than the standard amino acids, regardless of whether it is prepared synthetically or obtained from a natural source. As used herein, “synthetic amino acid” encompasses chemically modified amino acids, including but not limited to salts, amino acid derivatives (such as amides), and/or substitutions. Amino acids, including carboxy- and/or amino-terminal amino acids in peptides, can be modified by methylation, amidation, acetylation, protecting groups, and/or substitution with other chemical groups that can change the peptide's circulating half-life without adversely affecting their activity. Amino acids may participate in a disulfide bond. Amino acids may comprise one or posttranslational modifications, such as association with one or more chemical entities (e.g., methyl groups, acetate groups, acetyl groups, phosphate groups, formyl moieties, isoprenoid groups, sulfate groups, polyethylene glycol moieties, lipid moieties, carbohydrate moieties, biotin moieties, etc.). The term “amino acid” is used interchangeably with “amino acid residue,” and may refer to a free amino acid and/or to an amino acid residue of a peptide. It will be apparent from the context in which the term is used whether it refers to a free amino acid or a residue of a peptide.


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


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).


Biologically active: As used herein, the phrase “biologically active” refers to a characteristic of any agent that has activity in a biological system, and particularly in an organism. For instance, an agent that, when administered to an organism, has a biological effect on that organism, is considered to be biologically active.


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).


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.


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


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.


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.


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


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).


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


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.


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, O(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′-N-phosphoramidite linkages).


Nucleic acid: As used herein, the term “nucleic acid,” in its broadest sense, refers to any compound and/or substance that is or can be incorporated into a polynucleotide chain. In some embodiments, a nucleic acid is a compound and/or substance that is or can be incorporated into a polynucleotide chain via a phosphodiester linkage. In some embodiments, “nucleic acid” refers to individual nucleic acid residues (e.g., nucleotides and/or nucleosides). In some embodiments, “nucleic acid” refers to a polynucleotide chain comprising individual nucleic acid residues. In some embodiments, “nucleic acid” encompasses RNA as well as single and/or double-stranded DNA and/or cDNA.


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.


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.


Pharmaceutically acceptable salt: Pharmaceutically acceptable salts are well known in the art. For example, S. M. Berge et al., describes pharmaceutically acceptable salts in detail in J. Pharmaceutical Sciences (1977) 66:1-19. Pharmaceutically acceptable salts of the compounds of this invention include those derived from suitable inorganic and organic acids and bases. Examples of pharmaceutically acceptable, nontoxic acid addition salts are salts of an amino group formed with inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid or with organic acids such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid or malonic acid or by using other methods used in the art such as ion exchange. Other pharmaceutically acceptable salts include adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, formate, fumarate, glucoheptonate, glycerophosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectinate, persulfate, 3-phenylpropionate, phosphate, picrate, pivalate, propionate, stearate, succinate, sulfate, tartrate, thiocyanate, p-toluenesulfonate, undecanoate, valerate salts, and the like. Salts derived from appropriate bases include alkali metal, alkaline earth metal, ammonium and N+(C1-4 alkyl)4 salts. Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like. Further pharmaceutically acceptable salts include, when appropriate, nontoxic ammonium. quaternary ammonium, and amine cations formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, sulfonate and aryl sulfonate. Further pharmaceutically acceptable salts include salts formed from the quarternization of an amine using an appropriate electrophile, e.g., an alkyl halide, to form a quarternized alkylated amino salt.


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.”


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.


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.


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.


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.


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

The present invention provides, among other things, methods and compositions for treating Argininosuccinate Synthetase Deficiency (ASD) based on mRNA therapy. In particular, the present invention provides methods for treating ASD by administering to a subject in need of treatment a composition comprising an mRNA encoding argininosuccinate synthetase (ASS) at an effective dose and an administration interval such that at least one symptom or feature of ASD is reduced in intensity, severity, or frequency or has delayed in onset. 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 or non-cationic lipid(s), cholesterol-based lipid(s) and PEG-modified lipid(s).


Argininosuccinate Synthetase Deficiency (ASD)


The present invention may be used to treat a subject who is suffering from or susceptible to Argininosuccinate synthetase deficiency (ASD). ASD is an autosomal recessive metabolic genetic disorder characterized by a mutation in the gene for the enzyme argininosuccinate synthetase (ASS1). At least 50 mutations that cause type I ASD have been identified in the ASS1 gene. Most of these mutations involve single amino acid substitutions. Many of the mutations in the ASS1 gene likely affect the structure of the resulting protein and its ability to bind to citrulline, aspartate and other molecules. A few of the mutations in the ASS1 gene lead to the productions of an abnormally short version of the enzyme that cannot effectively play its role in the urea cycle.


Defects in the ASS1 protein disrupt the urea cycle and prevent the liver from properly processing excess nitrogen, which is generated when protein is used for energy, into urea. An accumulation of ammonia and other byproducts of the urea cycle (such as citrulline) is toxic and when it occurs during the first few days of life can lead to symptoms such as lack of energy (lethargy) poor feeding, vomiting, seizures and loss of consciousness. These medical problems can be life-threatening in many cases.


Compositions and methods described herein may be used to treat at least one symptom or feature of ASD.


Argininosuccinate Synthetase (ASS1)


In some embodiments, the present invention provides methods and compositions for delivering mRNA encoding ASS1 to a subject for the treatment of argininosuccinate synthetase deficiency (ASD). A suitable ASS1 mRNA encodes any full length, fragment or portion of an ASS1 protein which can be substituted for naturally-occurring ASS1 protein activity and/or reduce the intensity, severity, and/or frequency of one or more symptoms associated with ASD.


In some embodiments, a suitable mRNA sequence is an mRNA sequence encoding human ASS1 protein. The naturally-occurring human ASS1 mRNA sequence and the corresponding amino acid sequence are shown in Table 1:









TABLE 1





Human ASS1
















Human
GCCGGCGCGCCCCUGGGAGGGUGAGCCGGCGCCGGGCCCAGGCCCGGACCUG


ASS1
GUGGGAGGCGGGGGGAGGUGGGGACGAGGCCUGGGGAGGCGGGCCCCGCCC


(mRNA)
AUCUGCAGGUGGCUGUGAACGCUGAGCGGCUCCAGGCGGGGGCCGGGCCCGG



GGGCGGGGUCUGUGGCGCGCGUCCCCGCCACGUGUCCCCGGUCACCGGCCCU



GCCCCCGGGCCCUGUGCUUAUAACCUGGGAUGGGCACCCCUGCCAGUCCUGC



UCUGCCGCCUGCCACCGCUGCCCGAGCCCGAGUGGUUCACUGCACUGUGAAA



ACAGAUUCCAGACGCCGGGAACUCACGCCUCCAAUCCCAGACGCUAUGUCCA



GCAAAGGCUCCGUGGUUCUGGCCUACAGUGGCGGCCUGGACACCUCGUGCAU



CCUCGUGUGGCUGAAGGAACAAGGCUAUGACGUCAUUGCCUAUCUGGCCAA



CAUUGGCCAGAAGGAAGACUUCGAGGAAGCCAGGAAGAAGGCACUGAAGCU



UGGGGCCAAAAAGGUGUUCAUUGAGGAUGUCAGCAGGGAGUUUGUGGAGGA



GUUCAUCUGGCCGGCCAUCCAGUCCAGCGCACUGUAUGAGGACCGCUACCUC



CUGGGCACCUCUCUUGCCAGGCCCUGCAUCGCCCGCAAACAAGUGGAAAUCG



CCCAGCGGGAGGGGGCCAAGUAUGUGUCCCACGGCGCCACAGGAAAGGGGA



ACGAUCAGGUCCGGUUUGAGCUCAGCUGCUACUCACUGGCCCCCCAGAUAAA



GGUCAUUGCUCCCUGGAGGAUGCCUGAAUUCUACAACCGGUUCAAGGGCCGC



AAUGACCUGAUGGAGUACGCAAAGCAACACGGGAUUCCCAUCCCGGUCACUC



CCAAGAACCCGUGGAGCAUGGAUGAGAACCUCAUGCACAUCAGCUACGAGGC



UGGAAUCCUGGAGAACCCCAAGAACCAAGCGCCUCCAGGUCUCUACACGAAG



ACCCAGGACCCAGCCAAAGCCCCCAACACCCCUGACAUUCUCGAGAUCGAGU



UCAAAAAAGGGGUCCCUGUGAAGGUGACCAACGUCAAGGAUGGCACCACCC



ACCAGACCUCCUUGGAGCUCUUCAUGUACCUGAACGAAGUCGCGGGCAAGCA



UGGCGUGGGCCGUAUUGACAUCGUGGAGAACCGCUUCAUUGGAAUGAAGUC



CCGAGGUAUCUACGAGACCCCAGCAGGCACCAUCCUUUACCAUGCUCAUUUA



GACAUCGAGGCCUUCACCAUGGACCGGGAAGUGCGCAAAAUCAAACAAGGCC



UGGGCUUGAAAUUUGCUGAGCUGGUGUAUACCGGUUUCUGGCACAGCCCUG



AGUGUGAAUUUGUCCGCCACUGCAUCGCCAAGUCCCAGGAGCGAGUGGAAG



GGAAAGUGCAGGUGUCCGUCCUCAAGGGCCAGGUGUACAUCCUCGGCCGGG



AGUCCCCACUGUCUCUCUACAAUGAGGAGCUGGUGAGCAUGAACGUGCAGG



GUGAUUAUGAGCCAACUGAUGCCACCGGGUUCAUCAACAUCAAUUCCCUCAG



GCUGAAGGAAUAUCAUCGUCUCCAGAGCAAGGUCACUGCCAAAUAGACCCG



UGUACAAUGAGGAGCUGGGGCCUCCUCAAUUUGCAGAUCCCCCAAGUACAG



GCGCUAAUUGUUGUGAUAAUUUGUAAUUGUGACUUGUUCUCCCCGGCUGGC



AGCGUAGUGGGGCUGCCAGGCCCCAGCUUUGUUCCCUGGUCCCCCUGAAGCC



UGCAAACGUUGUCAUCGAAGGGAAGGGUGGGGGGCAGCUGCGGUGGGGAGC



UAUAAAAAUGACAAUUAAAAGAGACACUAGUCUUUUAUUUCUAAAAAAAAA



AAAAAAA(SEQ ID NO: 1)





Human
MSSKGSVVLAYSGGLDTSCILVWLKEQGYDVIAYLANIGQKEDFEEARKKALKLG


ASS1
AKKVFIEDVSREFVEEFIWPAIQSSALYEDRYLLGTSLARPCIARKQVEIAQREGAK


(Amino
YVSHGATGKGNDQVRFELSCYSLAPQIKVIAPWRMPEFYNRFKGRNDLMEYAKQ


Acid Seq.)
HGIPIPVTPKNPWSMDENLMHISYEAGILENPKNQAPPGLYTKTQDPAKAPNTPDIL



EIEFKKGVPVKVTNVKDGTTHQTSLELFMYLNEVAGKHGVGRIDIVENRFIGMKSR



GIYETPAGTILYHAHLDIEAFTMDREVRKIKQGLGLKFAELVYTGFWHSPECEFVR



HCIAKSQERVEGKVQVSVLKGQVYILGRESPLSLYNEELVSMNVQGDYEPTDATG



FININSLRLKEYHRLQSKVTAK (SEQ ID NO: 2)









In some embodiments, a suitable mRNA is a wild-type hASS1 mRNA of sequence (SEQ ID NO:1). In some embodiments, a suitable mRNA may be a codon optimized hASS1 sequence, such as the sequence shown below:









(SEQ ID NO: 3)


AUGAGCAGCAAGGGCAGCGUGGUGCUGGCCUACAGCGGCGGCCUGGACAC





CAGCUGCAUCCUGGUGUGGCUGAAGGAGCAGGGCUACGACGUGAUCGCCU





ACCUGGCCAACAUCGGCCAGAAGGAGGACUUCGAGGAGGCCCGCAAGAAG





GCCCUGAAGCUGGGCGCCAAGAAGGUGUUCAUCGAGGACGUGAGCCGCGA





GUUCGUGGAGGAGUUCAUCUGGCCCGCCAUCCAGAGCAGCGCCCUGUACG





AGGACCGCUACCUGCUGGGCACCAGCCUGGCCCGCCCCUGCAUCGCCCGC





AAGCAGGUGGAGAUCGCCCAGCGCGAGGGCGCCAAGUACGUGAGCCACGG





CGCCACCGGCAAGGGCAACGACCAGGUGCGCUUCGAGCUGAGCUGCUACA





GCCUGGCCCCCCAGAUCAAGGUGAUCGCCCCCUGGCGCAUGCCCGAGUUC





UACAACCGCUUCAAGGGCCGCAACGACCUGAUGGAGUACGCCAAGCAGCA





CGGCAUCCCCAUCCCCGUGACCCCCAAGAACCCCUGGAGCAUGGACGAGA





ACCUGAUGCACAUCAGCUACGAGGCCGGCAUCCUGGAGAACCCCAAGAAC





CAGGCCCCCCCCGGCCUGUACACCAAGACCCAGGACCCCGCCAAGGCCCC





CAACACCCCCGACAUCCUGGAGAUCGAGUUCAAGAAGGGCGUGCCCGUGA





AGGUGACCAACGUGAAGGACGGCACCACCCACCAGACCAGCCUGGAGCUG





UUCAUGUACCUGAACGAGGUGGCCGGCAAGCACGGCGUGGGCCGCAUCGA





CAUCGUGGAGAACCGCUUCAUCGGCAUGAAGAGCCGCGGCAUCUACGAGA





CCCCCGCCGGCACCAUCCUGUACCACGCCCACCUGGACAUCGAGGCCUUC





ACCAUGGACCGCGAGGUGCGCAAGAUCAAGCAGGGCCUGGGCCUGAAGUU





CGCCGAGCUGGUGUACACCGGCUUCUGGCACAGCCCCGAGUGCGAGUUCG





UGCGCCACUGCAUCGCCAAGAGCCAGGAGCGCGUGGAGGGCAAGGUGCAG





GUGAGCGUGCUGAAGGGCCAGGUGUACAUCCUGGGCCGCGAGAGCCCCCU





GAGCCUGUACAACGAGGAGCUGGUGAGCAUGAACGUGCAGGGCGACUACG





AGCCCACCGACGCCACCGGCUUCAUCAACAUCAACAGCCUGCGCCUGAAG





GAGUACCACCGCCUGCAGAGCAAGGUGACCGCCAAGUGA






Additional exemplary mRNA sequences are described in the Examples section below, for example, SEQ ID NO:7 and SEQ ID NO:8, both of which include 5′ and 3′ untranslated regions framing a coden-optimized ASS1-encoding mRNA.


In some embodiments, a suitable mRNA sequence may be an mRNA sequence a homolog or an analog of human ASS1 protein. For example, a homologue or an analogue of human ASS1 protein may be a modified human ASS1 protein containing one or more amino acid substitutions, deletions, and/or insertions as compared to a wild-type or naturally-occurring human ASS1 protein while retaining substantial ASS1 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 SEQ ID NO:2. In some embodiments, an mRNA suitable for the present invention encodes a protein substantially identical to human ASS1 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 SEQ ID NO:2. In some embodiments, an mRNA suitable for the present invention encodes a fragment or a portion of human ASS1 protein. In some embodiments, an mRNA suitable for the present invention encodes a fragment or a portion of human ASS1 protein, wherein the fragment or portion of the protein still maintains ASS1 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 SEQ ID NO:1, SEQ ID NO:3, SEQ ID NO:7, SEQ ID NO:8, SEQ ID NO:13, SEQ ID NO:14 or SEQ ID NO:15.


In some embodiments, a suitable mRNA encodes a fusion protein comprising a full length, fragment or portion of an ASS1 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 an ASS1 protein encodes a signal or a cellular targeting sequence.


Delivery Vehicles


According to the present invention, mRNA encoding an ASS1 protein (e.g., a full length, fragment or portion of an ASS1 protein) as described herein may be delivered as naked RNA (unpackaged) or via delivery vehicles. As used herein, the terms “delivery vehicle,” “transfer vehicle,” “nanoparticle” or grammatical equivalent, are used interchangeably.


In some embodiments, mRNAs encoding an ASS1 protein may be delivered via a single delivery vehicle. In some embodiments, mRNAs encoding an ASS1 protein may be delivered via one or more delivery vehicles each of a different composition. According to various embodiments, suitable delivery vehicles include, but are not limited to polymer based carriers, such as polyethyleneimine (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.


Liposomal Delivery Vehicles


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


Cationic Lipids


In some embodiments, liposomes may comprise one or more cationic lipids. As used herein, the phrase “cationic lipid” refers to any of a number of lipid species that have a net positive charge at a selected pH, such as physiological pH. Several cationic lipids have been described in the literature, many of which are commercially available. 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, CI 2-200 described at paragraph [00225]) and WO 2012/170930, both of which are incorporated herein by reference. In certain embodiments, the compositions and methods of the invention employ a lipid nanoparticles comprising an ionizable cationic lipid described in U.S. provisional patent application 61/617,468, filed Mar. 29, 2012 (incorporated herein by reference), such as, e.g, (15Z,18Z)—N,N-dimethyl-6-(9Z,12Z)-octadeca-9,12-dien-1-yl)tetracosa-15,18-dien-1-amine (HGT5000), (15Z,18Z)—N,N-dimethyl-6-((9Z,12Z)-octadeca-9,12-dien-1-yl)tetracosa-4,15,18-trien-1-amine (HGT5001), and (15Z,18Z)—N,N-dimethyl-6-((9Z,12Z)-octadeca-9,12-dien-1-yl)tetracosa-5,15,18-trien-1-amine (HGT5002).


In some embodiments, provided liposomes include a cationic lipid described in WO 2013063468 and in U.S. provisional application entitled “Lipid Formulations for Delivery of Messernger RNA” filed concurrently with the present application on even date, both of which are incorporated by reference herein.


In some embodiments, a cationic lipid comprises a compound of formula I-c1-a:




embedded image



or a pharmaceutically acceptable salt thereof, wherein:


each R2 independently is hydrogen or C1-3 alkyl;


each q independently is 2 to 6;


each R′ independently is hydrogen or C1-3 alkyl;


and each RL independently is C8-12 alkyl.


In some embodiments, each R2 independently is hydrogen, methyl or ethyl. In some embodiments, each R2 independently is hydrogen or methyl. In some embodiments, each R2 is hydrogen.


In some embodiments, each q independently is 3 to 6. In some embodiments, each q independently is 3 to 5. In some embodiments, each q is 4.


In some embodiments, each R′ independently is hydrogen, methyl or ethyl. In some embodiments, each R′ independently is hydrogen or methyl. In some embodiments, each R′ independently is hydrogen.


In some embodiments, each RL independently is C8-12 alkyl. In some embodiments, each RL independently is n-C8-12 alkyl. In some embodiments, each RL independently is C9-11 alkyl. In some embodiments, each RL independently is n-C9-11 alkyl. In some embodiments, each RL independently is C10 alkyl. In some embodiments, each RL independently is n-C10 alkyl.


In some embodiments, each R2 independently is hydrogen or methyl; each q independently is 3 to 5; each R′ independently is hydrogen or methyl; and each RL independently is C8-12 alkyl.


In some embodiments, each R2 is hydrogen; each q independently is 3 to 5; each R′ is hydrogen; and each RL independently is C8-12 alkyl.


In some embodiments, each R2 is hydrogen; each q is 4; each R′ is hydrogen; and each RL independently is C8-12 alkyl.


In some embodiments, a cationic lipid comprises a compound of formula I-g:




embedded image



or a pharmaceutically acceptable salt thereof, wherein each RL independently is C8-12 alkyl. In some embodiments, each RL independently is n-C8-12 alkyl. In some embodiments, each RL independently is C9-11 alkyl. In some embodiments, each RL independently is n-C9-11 alkyl. In some embodiments, each RL independently is C10 alkyl. In some embodiments, each RL is n-C10 alkyl.


In particular embodiments, provided liposomes include a cationic lipid cKK-E12, or (3,6-bis(4-(bis(2-hydroxydodecyl)amino)butyl)piperazine-2,5-dione). Structure of cKK-E12 is shown below:




embedded image


In some embodiments, the one or more cationic lipids may be N-[1-(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). DOTMA can be formulated alone or can be combined with the neutral lipid, dioleoylphosphatidyl-ethanolamine or “DOPE” or other cationic or non-cationic lipids into a liposomal transfer vehicle or a lipid nanoparticle, and such liposomes can be used to enhance the delivery of nucleic acids into target cells. Other suitable cationic lipids include, for example, 5-carboxyspermylglycinedioctadecylamide or “DOGS,” 2,3-dioleyloxy-N-[2(spermine-carboxamido)ethyl]-N,N-dimethyl-1-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), 1,2-Dioleoyl-3-Dimethylammonium-Propane or “DODAP”, 1,2-Dioleoyl-3-Trimethylammonium-Propane or “DOTAP”.


Additional exemplary cationic lipids also include 1,2-distearyloxy-N,N-dimethyl-3-aminopropane or “DSDMA”, 1,2-dioleyloxy-N,N-dimethyl-3-aminopropane or “DODMA”, 1,2-dilinoleyloxy-N,N-dimethyl-3-aminopropane or “DLinDMA”, 1,2-dilinolenyloxy-N,N-dimethyl-3-aminopropane or “DLenDMA”, N-dioleyl-N,N-dimethylammonium chloride or “DODAC”, N,N-distearyl-N,N-dimethylammonium bromide or “DDAB”, N-(1,2-dimyristyloxyprop-3-yl)-N,N-dimethyl-N-hydroxyethyl ammonium bromide or “DMRIE”, 3-dimethylamino-2-(cholest-5-en-3-beta-oxybutan-4-oxy)-1-(cis,cis-9,12-octadecadienoxy)propane or “CLinDMA”, 2-[5′-(cholest-5-en-3-beta-oxy)-3′-oxapentoxy)-3-dimethy 1-1-(cis,cis-9′,1-2′-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”, 1,2-N,N′-Dilinoleylcarbamyl-3-dimethylaminopropane or “DLincarbDAP”, 1,2-Dilinoleoylcarbamyl-3-dimethylaminopropane or “DLinCDAP”, 2,2-dilinoleyl-4-dimethylaminomethyl-[1,3]-dioxolane or “DLin-1-DMA”, 2,2-dilinoleyl-4-dimethylaminoethyl-[1,3]-dioxolane or “DLin-K-XTC2-DMA”, and 2-(2,2-di((9Z,12Z)-octadeca-9,12-dien-1-yl)-1,3-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, D V., 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.


In some embodiments, the one or more cationic lipids may be chosen from XTC (2,2-Dilinoley 1-4-dimethylaminoethyl-[1,3]-dioxolane), MC3 (((6Z,9Z,28Z,31Z)-heptatriaconta-6,9,28,31-tetraen-19-yl 4-(dimethylamino)butanoate), ALNY-100 ((3aR,5s,6aS)—N,N-dimethyl-2,2-di((9Z,12Z)-octadeca-9,12-dienyl)tetrahydro-3aH-cyclopenta[d][1,3]dioxol-5-amine)), NC98-5 (4,7,13-tris(3-oxo-3-(undecylamino)propyl)-N1,N16-diundecyl-4,7,10,13-tetraazahexadecane-1,16-diamide), DODAP (1,2-dioleyl-3-dimethylammonium propane), HGT4003 (WO 2012/170889, the teachings of which are incorporated herein by reference in their entirety), ICE (WO 2011/068810, the teachings of which are incorporated herein by reference in their entirety), HGT5000 (U.S. Provisional Patent Application No. 61/617,468, the teachings of which are incorporated herein by reference in their entirety) or HGT5001 (cis or trans) (Provisional Patent Application No. 61/617,468), aminoalcohol lipidoids such as those disclosed in WO2010/053572, DOTAP (1,2-dioleyl-3-trimethylammonium propane), DOTMA (1,2-di-O-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).


In some embodiments, the percentage of cationic lipid in a liposome may be greater than 10%, greater than 20%, greater than 30%, greater than 40%, greater than 50%, greater than 60%, or greater than 70%. In some embodiments, 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 liposome by weight. In some embodiments, the cationic lipid (e.g., cKK-E12) constitutes about 30%, about 35%, about 40%, about 45%, or about 50% of the liposome by molar ratio.


Non-Cationic/Helper Lipids


In some embodiments, provided liposomes contain one or more non-cationic (“helper”) lipids. As used herein, the phrase “non-cationic lipid” refers to any neutral, zwitterionic or anionic lipid. As used herein, the phrase “anionic lipid” refers to any of a number of lipid species that carry a net negative charge at a selected H, 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-1-carboxylate (DOPE-mal), dipalmitoyl phosphatidyl ethanolamine (DPPE), dimyristoylphosphoethanolamine (DMPE), distearoyl-phosphatidyl-ethanolamine (DSPE), 16-O-monomethyl PE, 16-O-dimethyl PE, 18-1-trans PE, 1-stearoyl-2-oleoyl-phosphatidyethanolamine (SOPE), or a mixture thereof.


In some embodiments, such non-cationic lipids may be used alone, but are preferably used in combination with other excipients, for example, cationic lipids. In some embodiments, the non-cationic lipid may comprise a molar ratio of about 5% to about 90%, or about 10% to about 70% of the total lipid present in a liposome. In some embodiments, a non-cationic lipid is a neutral lipid, i.e., a lipid that does not carry a net charge in the conditions under which the composition is formulated and/or administered. In some embodiments, the percentage of non-cationic lipid in a liposome may be greater than 5%, greater than 10%, greater than 20%, greater than 30%, or greater than 40%.


Cholesterol-Based Lipids


In some embodiments, provided liposomes comprise 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, the cholesterol-based lipid may comprise a molar ration of about 2% to about 30%, or about 5% to about 20% of the total lipid present in a liposome. In some embodiments, The percentage of cholesterol-based lipid in the lipid nanoparticle may be greater than 5, %, 10%, greater than 20%, greater than 30%, or greater than 40%.


PEGylated Lipids


In some embodiments, provided liposomes comprise 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-1-[Succinyl(Methoxy Polyethylene Glycol)-2000] (C8 PEG-2000 ceramide) is also contemplated by the present invention in combination with one or more of the cationic and, in some embodiments, other lipids together which comprise the liposome. 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. The addition of such components may prevent complex aggregation and may also provide a means for increasing circulation lifetime and increasing the delivery of the lipid-nucleic acid composition to the target cell, (Klibanov et al. (1990) FEBS Letters, 268 (1): 235-237), or they may be selected to rapidly exchange out of the formulation in vivo (see U.S. Pat. No. 5,885,613).


In some embodiments, particularly useful exchangeable lipids are PEG-ceramides having shorter acyl chains (e.g., C14 or C18). The PEG-modified phospholipid and derivitized lipids of the present invention may comprise a molar ratio from about 0% to about 15%, about 0.5% to about 15%, about 1% to about 15%, about 4% to about 10%, or about 2% of the total lipid present in the liposome.


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


Polymers


In some embodiments, a suitable delivery vehicle is formulated using a polymer as a carrier, alone or in combination with other carriers including various lipids described herein. Thus, in some embodiments, liposomal delivery vehicles, as used herein, also encompass polymer containing nanoparticles. Suitable polymers may include, for example, polyacrylates, polyalkycyanoacrylates, polylactide, polylactide-polyglycolide copolymers, polycaprolactones, dextran, albumin, gelatin, alginate, collagen, chitosan, cyclodextrins, protamine, PEGylated protamine, PLL, PEGylated PLL and polyethylenimine (PEI). When PEI is present, it may be branched PEI of a molecular weight ranging from 10 to 40 kDa, e.g., 25 kDa branched PEI (Sigma #408727).


A suitable liposome for the present invention may include one or more of any of the cationic lipids, non-cationic lipids, cholesterol lipids, PEGylated lipids and/or polymers described herein at various ratios. As non-limiting examples, a suitable liposome formulation may include a combination selected from cKK-E12, DOPE, cholesterol and DMG-PEG2K; C12-200, DOPE, cholesterol and DMG-PEG2K; HGT4003, DOPE, cholesterol and DMG-PEG2K; or ICE, DOPE, cholesterol and DMG-PEG2K.


In various embodiments, cationic lipids (e.g., cKK-E12, C12-200, ICE, and/or HGT4003) constitute about 30-60% (e.g., about 30-55%, about 30-50%, about 30-45%, about 30-40%, about 35-50%, about 35-45%, or about 35-40%) of the liposome by molar ratio. In some embodiments, the percentage of cationic lipids (e.g., cKK-E12, C12-200, ICE, and/or HGT4003) is or greater than about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, or about 60% of the liposome by molar ratio.


In some embodiments, the ratio of cationic lipid(s) to non-cationic lipid(s) to cholesterol-based lipid(s) to PEGylated lipid(s) may be between about 30-60:25-35:20-30:1-15, respectively. In some embodiments, the ratio of cationic lipid(s) to non-cationic lipid(s) to cholesterol-based lipid(s) to PEGylated lipid(s) is approximately 40:30:20:10, respectively. In some embodiments, the ratio of cationic lipid(s) to non-cationic lipid(s) to cholesterol-based lipid(s) to PEGylated lipid(s) is approximately 40:30:25:5, respectively. In some embodiments, the ratio of cationic lipid(s) to non-cationic lipid(s) to cholesterol-based lipid(s) to PEGylated lipid(s) is approximately 40:32:25:3, respectively. In some embodiments, the ratio of cationic lipid(s) to non-cationic lipid(s) to cholesterol-based lipid(s) to PEGylated lipid(s) is approximately 50:25:20:5.


Synthesis of mRNA


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


In some embodiments, for the preparation of mRNA according to the invention, a DNA template is transcribed in vitro. A suitable DNA template typically has a promoter, for example a T3, T7 or SP6 promoter, for in vitro transcription, followed by desired nucleotide sequence for desired mRNA and a termination signal.


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


Modified mRNA


In some embodiments, mRNA according to the present invention may be synthesized as unmodified or modified mRNA. Typically, mRNAs are modified to enhance stability. Modifications of mRNA can include, for example, modifications of the nucleotides of the RNA. An modified mRNA according to the invention can thus 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, 1-methyl-inosine, pseudouracil (5-uracil), dihydro-uracil, 2-thio-uracil, 4-thio-uracil, 5-carboxymethylaminomethyl-2-thio-uracil, 5-(carboxyhydroxymethyl)-uracil, 5-fluoro-uracil, 5-bromo-uracil, 5-carboxymethylaminomethyl-uracil, 5-methyl-2-thio-uracil, 5-methyl-uracil, N-uracil-5-oxyacetic acid methyl ester, 5-methylaminomethyl-uracil, 5-methoxyaminomethyl-2-thio-uracil, 5′-methoxycarbonylmethyl-uracil, 5-methoxy-uracil, uracil-5-oxyacetic acid methyl ester, uracil-5-oxyacetic acid (v), 1-methyl-pseudouracil, queosine, .beta.-D-mannosyl-queosine, wybutoxosine, and phosphoramidates, phosphorothioates, peptide nucleotides, methylphosphonates, 7-deazaguanosine, 5-methylcytosine and inosine. The preparation of such analogues is known to a person skilled in the art e.g. from the U.S. Pat. Nos. 4,373,071, 4,401,796, 4,415,732, 4,458,066, 4,500,707, 4,668,777, 4,973,679, 5,047,524, 5,132,418, 5,153,319, 5,262,530 and 5,700,642, the disclosures of which are incorporated by reference in their entirety.


In some embodiments, mRNAs (e.g., ASS1-encoding mRNAs) may contain RNA backbone modifications. Typically, a backbone modification is a modification in which the phosphates of the backbone of the nucleotides contained in the RNA are modified chemically. Exemplary backbone modifications typically include, but are not limited to, modifications from the group consisting of methylphosphonates, methylphosphoramidates, phosphoramidates, phosphorothioates (e.g. cytidine 5′-O-(1-thiophosphate)), boranophosphates, positively charged guanidinium groups etc., which means by replacing the phosphodiester linkage by other anionic, cationic or neutral groups.


In some embodiments, mRNAs (e.g., ASS1-encoding 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′-O-alkyloligoribonucleotide, 2′-deoxy-2′-C-alkyloligoribonucleotide (2′-O-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).


In some embodiments, mRNAs (e.g., ASS1-encoding 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, N1-methyladenosine 5′-triphosphate, N1-methylguanosine 5′-triphosphate, N6-methyladenosine 5′-triphosphate, O6-methylguanosine 5′-triphosphate, pseudouridine 5′-triphosphate, puromycin 5′-triphosphate or xanthosine 5′-triphosphate.


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


Thus, in some embodiments, mRNAs (e.g., ASS1-encoding mRNAs) include a 5′ cap structure. A 5′ cap is typically added as follows: first, an RNA terminal phosphatase removes one of the terminal phosphate groups from the 5′ nucleotide, leaving two terminal phosphates; guanosine triphosphate (GTP) is then added to the terminal phosphates via a guanylyl transferase, producing a 5′5′5 triphosphate linkage; and the 7-nitrogen of guanine is then methylated by a methyltransferase. Examples of cap structures include, but are not limited to, m7G(5′)ppp (5′(A,G(5′)ppp(5′)A and G(5′)ppp(5′)G.


In some embodiments, mRNAs (e.g., ASS1-encoding 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 (SEQ ID NO:9) (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 (SEQ ID NO:10) (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.


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.


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


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


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 m7G(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.


A common cap for mRNA produced by in vitro transcription is m7G(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 m7G(5′)ppp(5′)G (“m7GpppG”) as an initiator of transcription.


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 —OCH3.


Additional cap analogs include, but are not limited to, a chemical structures selected from the group consisting of m7GpppG, m7GpppA, m7GpppC; unmethylated cap analogs (e.g., GpppG); dimethylated cap analog (e.g., m2,7GpppG), trimethylated cap analog (e.g., m2,2,7GpppG), dimethylated symmetrical cap analogs (e.g., m7Gpppm7G), or anti reverse cap analogs (e.g., ARCA; m7,2′OmeGpppG, m72′dGpppG, m7,3′OmeGpppG, m73′dGpppG 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)).


In some embodiments, a suitable cap is a 7-methyl guanylate (“m7G”) linked via a triphosphate bridge to the 5′-end of the first transcribed nucleotide, resulting in m7G(5′)ppp(5′)N, where N is any nucleoside. A preferred embodiment of a m7G cap utilized in embodiments of the invention is m7G(5′)ppp(5′)G.


In some embodiments, the cap is a Cap0 structure. Cap0 structures lack a 2′-O-methyl residue of the ribose attached to bases 1 and 2. In some embodiments, the cap is a Cap1 structure. Cap1 structures have a 2′-O-methyl residue at base 2. In some embodiments, the cap is a Cap2 structure. Cap2 structures have a 2′-O-methyl residue attached to both bases 2 and 3.


A variety of m7G cap analogs are known in the art, many of which are commercially available. These include the m7GpppG described above, as well as the ARCA 3′-OCH3 and 2′-OCH3 cap analogs (Jemielity, J. et al., RNA, 9: 1108-1122 (2003)). Additional cap analogs for use in embodiments of the invention include N7-benzylated dinucleoside tetraphosphate analogs (described in Grudzien, E. et al., RNA, 10: 1479-1487 (2004)), phosphorothioate cap analogs (described in Grudzien-Nogalska, E., et al., RNA, 13: 1745-1755 (2007)), and cap analogs (including biotinylated cap analogs) described in U.S. Pat. Nos. 8,093,367 and 8,304,529, incorporated by reference herein.


Tail Structure


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)).


In some embodiments, mRNAs include a 3′ poly(A) tail structure. Typically, the length of the poly A tail can be at least about 10, 50, 100, 200, 300, 400 at least 500 nucleotides (SEQ ID NO: 11). In some embodiments, a poly-A tail on the 3′ terminus of mRNA typically includes about 10 to 300 adenosine nucleotides (SEQ ID NO:9) (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 (SEQ ID NO:10) (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.


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


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.


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. 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.


Formation of Liposomes


The liposomal transfer vehicles for use in the compositions of the invention can be prepared by various techniques which are presently known in the art. The liposomes for use in provided compositions can be prepared by various techniques which are presently known in the art. For example, multilamellar vesicles (MLV) may be prepared according to conventional techniques, such as by depositing a selected lipid on the inside wall of a suitable container or vessel by dissolving the lipid in an appropriate solvent, and then evaporating the solvent to leave a thin film on the inside of the vessel or by spray drying. An aqueous phase may then added to the vessel with a vortexing motion which results in the formation of MLVs. Uni-lamellar vesicles (ULV) can then be formed by homogenization, sonication or extrusion of the multilamellar vesicles. In addition, unilamellar vesicles can be formed by detergent removal techniques.


In certain embodiments, provided compositions comprise a liposome wherein the mRNA is associated on both the surface of the liposome and encapsulated within the same liposome. For example, during preparation of the compositions of the present invention, cationic liposomes may associate with the mRNA through electrostatic interactions. For example, during preparation of the compositions of the present invention, cationic liposomes may associate with the mRNA through electrostatic interactions.


In some embodiments, the compositions and methods of the invention comprise mRNA encapsulated in a liposome. In some embodiments, the one or more mRNA species may be encapsulated in the same liposome. In some embodiments, the one or more mRNA species may be encapsulated in different liposomes. In some embodiments, the mRNA is encapsulated in one or more liposomes, which differ in their lipid composition, molar ratio of lipid components, size, charge (Zeta potential), targeting ligands and/or combinations thereof. In some embodiments, the one or more liposome may have a different composition of cationic lipids, neutral lipid, PEG-modified lipid and/or combinations thereof. In some embodiments the one or more lipisomes may have a different molar ratio of cationic lipid, neutral lipid, cholesterol and PEG-modified lipid used to create the liposome.


The process of incorporation of a desired mRNA into a liposome is often referred to as “loading”. Exemplary methods are described in Lasic, et al., FEBS Lett., 312: 255-258, 1992, which is incorporated herein by reference. The liposome-incorporated nucleic acids may be completely or partially located in the interior space of the liposome, within the bilayer membrane of the liposome, or associated with the exterior surface of the liposome membrane. The incorporation of a nucleic acid into liposomes is also referred to herein as “encapsulation” wherein the nucleic acid is entirely contained within the interior space of the liposome. The purpose of incorporating a mRNA into a transfer vehicle, such as a liposome, is often to protect the nucleic acid from an environment which may contain enzymes or chemicals that degrade nucleic acids and/or systems or receptors that cause the rapid excretion of the nucleic acids. Accordingly, in some embodiments, a suitable delivery vehicle is capable of enhancing the stability of the mRNA contained therein and/or facilitate the delivery of mRNA to the target cell or tissue.


Liposome Size


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.


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.


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.


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).


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


To facilitate expression of mRNA in vivo, delivery vehicles such as 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. Techniques for formulation and administration of drugs may be found in “Remington's Pharmaceutical Sciences,” Mack Publishing Co., Easton, Pa., latest edition.


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.


Suitable routes of administration include, for example, oral, rectal, vaginal, transmucosal, pulmonary including intratracheal or inhaled, or intestinal administration; parenteral delivery, including intradermal, transdermal (topical), intramuscular, subcutaneous, intramedullary injections, as well as intrathecal, direct intraventricular, intravenous, intraperitoneal, or intranasal.


Alternatively or additionally, liposomally encapsulated mRNAs and compositions of the invention may be administered in a local rather than systemic manner, for example, via injection of the pharmaceutical composition directly into a targeted tissue, preferably in a sustained release formulation. Local delivery can be affected in various ways, depending on the tissue to be targeted. For example, aerosols containing compositions of the present invention can be inhaled (for nasal, tracheal, or bronchial delivery); compositions of the present invention can be injected into the site of injury, disease manifestation, or pain, for example; compositions can be provided in lozenges for oral, tracheal, or esophageal application; can be supplied in liquid, tablet or capsule form for administration to the stomach or intestines, can be supplied in suppository form for rectal or vaginal application; or can even be delivered to the eye by use of creams, drops, or even injection. Formulations containing provided compositions complexed with therapeutic molecules or ligands can even be surgically administered, for example in association with a polymer or other structure or substance that can allow the compositions to diffuse from the site of implantation to surrounding cells. Alternatively, they can be applied surgically without the use of polymers or supports.


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 ASS1 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., ASD). In some embodiments, a therapeutically effective amount of the therapeutic agents (e.g., mRNA encoding a ASS1 protein) of the present invention may be administered intrathecally 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), once every 30 days, once every 28 days, once every 14 days, once every 10 days, once every 7 days, weekly, daily or continuously).


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 one embodiment, 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, 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 a mRNA over extended periods of time. Preferably, the extended-release means employed are combined with modifications made to the mRNA to enhance stability.


As used herein, the term “therapeutically effective amount” is largely determined 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 ASD). 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 ASS1 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.


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.


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


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.


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 U.S. Provisional Application No. 61/494,882, filed Jun. 8, 2011, 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.


Provided liposomes and compositions may be administered to any desired tissue. In some embodiments, the mRNA 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.


According to the present invention, a therapeutically effective dose, when administered regularly, results in increased hepatic ASS1 levels. In some embodiments, a therapeutically effective dose, when administered regularly, results in reduced citrulline level in serum as compared to the baseline citrulline level before treatment. In some embodiments, a therapeutically effective dose, when administered regularly, results in reduced ammonia level in serum as compared to the baseline ammonia level before treatment.


In some embodiments, administering the provided composition results in increased expression of ASS1 protein in the liver as compared to baseline levels before the treatment. In some embodiment, administering the provided compositions results in an ASS1 protein level at or above about 3000 ng/mg, at or above about 2000 ng/mg, at or above about 1000 ng/mg, at or above about 500 ng/mg, at or above about 400 ng/mg, at or above about 200 ng/mg or at or above about 100 ng/mg of total protein in the liver. In a particular embodiment, administering the provided compositions results in an ASS1 protein level at or above 120 ng/mg of total protein in the liver.


In some embodiments, administering the provided composition results in increased ASS1 protein level in plasma or serum as compared to baseline level before the treatment. In some embodiments, administering the provided composition results in increased ASS1 protein level in plasma or serum by at least about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 95% as compared to baseline level before treatment.


In some embodiments, administering of the composition results in reduced citrulline and/or ammonia levels in the subject as compared to the baseline levels before treatment. Typically, the baseline levels are measured immediately before treatment. Typically, citrulline and/or ammonia levels are measured in a biological sample. Suitable biological samples include, for example, whole blood, plasma, serum, urine or cerebral spinal fluid.


In some embodiments, administering of the composition results in reduced citrulline level in a biological sample (e.g., a serum, plasma, or urine sample) by at least about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 95% as compared to the baseline citrulline level immediately before treatment. In some embodiments, administering of the composition results in reduced plasma citrulline level to less than about 2000 μM, 1500 μM, 1000 μM, 750 μM, 500 μM, 250 μM, 100 μM, 90 μM, 80 μM, 70 μM, 60 μM, 50 μM, 40 μM, or 30 μM.


In some embodiments, administering the composition results in reduced ammonia levels in a biological sample (e.g., a serum, plasma, or urine sample) by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, or at least about 95% as compared to baseline level immediately before treatment.


In some embodiments, administering the provided composition results in reduced ammonia levels in plasma or serum as compared to the baseline ammonia level immediately before treatment. In some embodiments, administering the provided composition results in reduced ammonia levels in plasma or serum as compared to the ammonia level in subjects who are not treated. In some embodiments, administering the composition results in reduction of ammonia levels to about 3000 μmol/L or less, about 2750 μmol/L or less, about 2500 μmol/L or less, about 2250 μmol/L or less, about 2000 μmol/L or less, about 1750 μmol/L or less, about 1500 μmol/L or less, about 1250 μmol/L or less, about 1000 μmol/L or less, about 750 μmol/L or less, about 500 μmol/L or less, about 250 μmol/L or less, about 100 μmol/L or less or about 50 μmol/L or less in the plasma or serum. In a particular embodiment, administering the composition results in reduction of ammonia levels to about 50 μmol/L or less in the plasma or serum.


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, and/or 96 hours after administration of provided liposomes and/or compositions. In some embodiments, the expression of the protein encoded by delivered mRNA is detectable 1 week, two weeks, and/or 1 month after administration.


EXAMPLES

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.


Example 1
Exemplary Liposome Formulations for ASS1 mRNA Delivery and Expression

This example provides exemplary liposome formulations for effective delivery and expression of ASS1 mRNA in vivo.


Lipid Materials


The formulations described herein include a multi-component lipid mixture of varying ratios employing one or more cationic lipids, helper lipids (e.g., non-cationic lipids and/or cholesterol-based lipids) and PEGylated lipids designed to encapsulate mRNA encoding ASS1 protein. Cationic lipids can include (but not exclusively) DOTAP (1,2-dioleyl-3-trimethylammonium propane), DODAP (1,2-dioleyl-3-dimethylammonium propane), DOTMA (1,2-di-O-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), cKK-E12 (3,6-bis(4-(bis(2-hydroxydodecyl)amino)butyl)piperazine-2,5-dione), HGT5000, HGT5001, HGT4003, ICE, dialkylamino-based, imidazole-based, guanidinium-based, etc. Helper lipids can include (but not exclusively) DSPC (1,2-distearoyl-sn-glycero-3-phosphocholine), DPPC (1,2-dipalmitoyl-sn-glycero-3-phosphocholine), DOPE (1,2-dioleyl-sn-glycero-3-phosphoethanolamine), DOPC (1,2-dioleyl-sn-glycero-3-phosphotidylcholine) DPPE (1,2-dipalmitoyl-sn-glycero-3-phosphoethanolamine), DMPE (1,2-dimyristoyl-sn-glycero-3-phosphoethanolamine), DOPG (2-dioleoyl-sn-glycero-3-phospho-(1′-rac-glycerol)), cholesterol, etc. The PEGylated lipids can include (but not exclusively) a poly(ethylene)glycol chain of up to 5 kDa in length covalently attached to a lipid with alkyl chain(s) of C6-C20 length.


Codon-optimized human argininosuccinate synthetase (ASS1) messenger RNA 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) tail of approximately 250 nucleotides in length (SEQ ID NO:12) as determined by gel electrophoresis. 5′ and 3′ untranslated regions present in each mRNA product are represented as X and Y, respectively and defined as stated (vide infra).


Exemplary Codon-Optimized Human Argininosuccinate Synthetase (ASS1) mRNAs


Construct Design:


X—SEQ ID NO:3—Y;


X—SEQ ID NO:13—Y;


X—SEQ ID NO:14—Y; and


X—SEQ ID NO:15—Y.


5′ and 3′ UTR Sequences









[SEQ ID NO.: 4]


X (5′ UTR Sequence) =


GGACAGAUCGCCUGGAGACGCCAUCCACGCUGUUUUGACCUCCAUAGAAG





ACACCGGGACCGAUCCAGCCUCCGCGGCCGGGAACGGUGCAUUGGAACGC





GGAUUCCCCGUGCCAAGAGUGACUCACCGUCCUUGACACG





[SEQ ID NO.: 5]


Y (3′ UTR Sequence) =


CGGGUGGCAUCCCUGUGACCCCUCCCCAGUGCCUCUCCUGGCCCUGGAAG





UUGCCACUCCAGUGCCCACCAGCCUUGUCCUAAUAAAAUUAAGUUGCAUC





AAGCU


OR





(SEQ ID NO.: 6)


GGGUGGCAUCCCUGUGACCCCUCCCCAGUGCCUCUCCUGGCCCUGGAAGU





UGCCACUCCAGUGCCCACCAGCCUUGUCCUAAUAAAAUUAAGUUGCAUCA





AAGCU






Exemplary codon-optimized human ASS1 mRNA sequences include SEQ ID NO:3 described in the detailed description section, and SEQ ID NO:13, SEQ ID NO:14 and SEQ ID NO:15 below:










SEQ ID NO: 13



AUGAGCUCAAAGGGAUCUGUGGUGCUGGCAUACUCGGGGGGAUUGGACACUUCA






UGCAUACUUGUCUGGUUGAAGGAACAGGGCUACGACGUGAUCGCCUACCUGGCU





AACAUCGGUCAAAAGGAGGACUUCGAGGAGGCCCGGAAGAAGGCCCUGAAGCUG





GGCGCGAAGAAAGUGUUCAUCGAGGACGUGUCCCGGGAAUUUGUGGAAGAGUUC





AUCUGGCCCGCCAUCCAAAGCAGCGCACUGUACGAGGAUAGAUACCUCCUCGGAA





CAUCCCUUGCCCGGCCAUGUAUUGCCAGGAAACAGGUGGAAAUCGCCCAGCGGGA





AGGAGCCAAAUACGUGUCCCACGGGGCGACCGGAAAGGGGAACGACCAAGUGCGC





UUCGAGCUGUCGUGCUACUCCCUGGCACCGCAGAUUAAGGUCAUCGCGCCGUGGA





GAAUGCCUGAAUUCUACAACCGCUUCAAGGGCCGCAACGAUCUGAUGGAAUACGC





CAAGCAGCACGGCAUCCCGAUCCCCGUGACCCCUAAGAACCCUUGGUCAAUGGAC





GAGAAUCUGAUGCACAUCAGCUACGAAGCGGGCAUCCUGGAGAACCCCAAGAAUC





AAGCUCCGCCCGGACUGUACACUAAGACUCAGGAUCCCGCUAAGGCGCCCAACAC





UCCUGAUAUUUUGGAAAUCGAAUUCAAGAAGGGUGUCCCAGUGAAGGUCACCAA





CGUGAAGGACGGCACUACCCACCAGACCUCGCUGGAACUGUUUAUGUAUCUGAAC





GAGGUGGCCGGCAAACAUGGAGUCGGCAGAAUCGAUAUUGUGGAGAACCGCUUU





AUUGGCAUGAAGUCCAGGGGGAUCUAUGAAACCCCGGCCGGAACCAUCCUCUACC





ACGCCCAUCUCGACAUUGAAGCGUUCACCAUGGACCGCGAGGUCCGCAAGAUUAA





GCAGGGCCUGGGACUCAAGUUCGCCGAGCUCGUGUACACCGGUUUCUGGCAUUCC





CCGGAAUGCGAAUUCGUGCGACACUGCAUUGCCAAGAGCCAGGAGCGGGUGGAA





GGAAAGGUCCAGGUGUCCGUGCUGAAGGGUCAAGUGUACAUCCUGGGGCGGGAG





UCCCCUCUUUCCCUGUACAACGAAGAACUGGUGUCGAUGAACGUGCAGGGAGACU





ACGAGCCGACCGACGCCACGGGUUUCAUUAACAUCAAUUCCCUGAGACUGAAGGA





GUACCACCGGCUCCAGUCCAAAGUCACCGCUAAGUGA (SEQ ID NO: 13),





SEQ ID NO: 14



AUGAGCUCAAAAGGAUCGGUGGUGCUGGCAUACUCGGGAGGAUUGGACACUUCA






UGUAUUCUUGUCUGGCUCAAGGAACAGGGCUACGACGUCAUUGCCUACCUGGCCA





ACAUCGGUCAGAAAGAGGACUUCGAGGAAGCCAGAAAGAAGGCCCUGAAGCUGG





GAGCCAAGAAGGUGUUCAUCGAGGACGUGUCCCGCGAAUUUGUGGAAGAAUUCA





UCUGGCCUGCCAUUCAAUCCUCCGCGCUCUACGAGGAUCGGUACCUUCUGGGAAC





UUCCUUGGCUCGCCCGUGCAUCGCCCGGAAACAAGUGGAGAUUGCACAGCGGGAA





GGAGCUAAGUACGUGUCCCACGGGGCCACUGGAAAGGGCAACGAUCAAGUGCGCU





UCGAGCUGUCCUGCUACUCCCUGGCGCCACAGAUCAAGGUCAUCGCGCCGUGGCG





GAUGCCCGAGUUCUAUAACCGCUUCAAGGGACGGAACGAUCUGAUGGAGUACGCC





AAGCAGCACGGCAUUCCGAUACCCGUGACCCCCAAGAACCCUUGGAGCAUGGACG





AGAACCUGAUGCAUAUCUCUUACGAAGCCGGGAUUCUCGAAAACCCUAAGAAUCA





GGCGCCGCCUGGCCUGUACACCAAAACCCAGGACCCCGCCAAGGCGCCGAACACG





CCCGACAUCCUCGAAAUCGAGUUCAAGAAGGGGGUGCCAGUGAAGGUCACCAACG





UGAAGGACGGAACCACCCAUCAGACCUCACUGGAACUCUUCAUGUACCUCAACGA





GGUCGCAGGGAAGCACGGCGUGGGGAGAAUCGACAUCGUGGAAAACAGGUUCAU





CGGCAUGAAGUCCCGGGGAAUCUACGAAACACCCGCCGGGACUAUCCUCUACCAC





GCCCACCUGGACAUUGAGGCCUUCACCAUGGAUAGAGAAGUGCGCAAGAUUAAGC





AGGGUCUGGGUCUGAAGUUCGCCGAGUUGGUCUACACCGGAUUCUGGCAUUCCCC





UGAAUGCGAAUUCGUGCGCCACUGCAUUGCCAAGAGCCAGGAAAGAGUGGAGGG





CAAAGUCCAAGUGUCGGUGCUGAAGGGCCAAGUGUACAUCCUGGGAAGGGAAAG





CCCGCUCUCCCUGUACAACGAGGAACUGGUGUCGAUGAACGUCCAGGGCGAUUAU





GAGCCGACUGACGCCACUGGUUUUAUCAAUAUCAACAGCCUGCGACUGAAGGAGU





ACCACCGGCUGCAGUCCAAGGUCACCGCUAAGUAG (SEQ ID NO: 14),





SEQ ID NO: 15



AUGAGCUCGAAAGGAUCCGUGGUUUUGGCAUACUCCGGUGGACUUGACACUUCA






UGCAUUUUGGUUUGGCUCAAAGAACAGGGCUACGAUGUGAUCGCCUACCUGGCG





AACAUCGGACAGAAAGAGGACUUUGAAGAGGCCCGCAAGAAGGCACUGAAGCUG





GGUGCCAAGAAAGUGUUUAUCGAGGAUGUGUCGAGAGAAUUCGUGGAAGAAUUC





AUUUGGCCAGCCAUUCAAAGCUCCGCGCUGUACGAGGACAGAUACCUCCUCGGCA





CCUCACUGGCCCGCCCUUGCAUCGCGCGCAAACAGGUCGAGAUCGCUCAAAGAGA





AGGAGCUAAAUACGUGUCACACGGCGCCACCGGAAAGGGAAAUGACCAAGUCCGC





UUCGAGCUGUCUUGCUACUCACUCGCUCCGCAAAUCAAGGUCAUCGCACCGUGGA





GGAUGCCCGAGUUCUACAACCGGUUCAAGGGGCGGAACGACCUGAUGGAGUACGC





GAAGCAGCACGGUAUCCCGAUCCCUGUCACCCCAAAGAACCCCUGGAGCAUGGAC





GAAAAUCUGAUGCACAUCAGCUACGAAGCAGGAAUCCUGGAGAACCCGAAAAAU





CAAGCACCUCCUGGACUGUACACUAAGACCCAGGACCCAGCCAAGGCCCCGAAUA





CCCCGGACAUCUUGGAAAUCGAGUUCAAGAAGGGGGUGCCAGUGAAGGUUACCA





AUGUCAAGGAUGGGACCACUCACCAAACUAGCCUGGAACUGUUCAUGUACCUGAA





CGAAGUGGCUGGAAAACAUGGCGUGGGAAGAAUCGAUAUCGUGGAGAACCGCUU





CAUCGGCAUGAAGUCAAGGGGAAUCUACGAAACUCCGGCCGGGACGAUACUGUA





UCAUGCGCAUCUCGACAUUGAAGCCUUUACUAUGGAUCGGGAAGUCCGAAAGAU





CAAACAGGGCUUGGGCCUCAAGUUUGCCGAGCUGGUGUACACGGGAUUCUGGCAC





UCGCCGGAAUGCGAAUUCGUGCGCCACUGUAUUGCGAAGUCCCAGGAGCGCGUGG





AAGGGAAGGUCCAAGUCUCCGUGCUCAAAGGACAGGUCUACAUCCUUGGACGGG





AGUCGCCCCUGUCGCUCUACAACGAAGAACUGGUGUCGAUGAACGUGCAGGGAGA





CUAUGAACCAACGGAUGCUACUGGUUUCAUCAACAUCAAUUCGCUGCGGCUUAAG





GAGUACCAUCGGCUGCAGUCCAAGGUCACCGCGAAGUAG(SEQ ID NO: 15).






An exemplary full-length codon-optimized human argininosuccinate synthetase (ASS1) messenger RNA sequence is shown below:










(SEQ ID NO: 7)



GGACAGAUCGCCUGGAGACGCCAUCCACGCUGUUUUGACCUCCAUAGAAGACACC






GGGACCGAUCCAGCCUCCGCGGCCGGGAACGGUGCAUUGGAACGCGGAUUCCCCG





UGCCAAGAGUGACUCACCGUCCUUGACACGAUGAGCAGCAAGGGCAGCGUGGUGC





UGGCCUACAGCGGCGGCCUGGACACCAGCUGCAUCCUGGUGUGGCUGAAGGAGCA





GGGCUACGACGUGAUCGCCUACCUGGCCAACAUCGGCCAGAAGGAGGACUUCGAG





GAGGCCCGCAAGAAGGCCCUGAAGCUGGGCGCCAAGAAGGUGUUCAUCGAGGACG





UGAGCCGCGAGUUCGUGGAGGAGUUCAUCUGGCCCGCCAUCCAGAGCAGCGCCCU





GUACGAGGACCGCUACCUGCUGGGCACCAGCCUGGCCCGCCCCUGCAUCGCCCGC





AAGCAGGUGGAGAUCGCCCAGCGCGAGGGCGCCAAGUACGUGAGCCACGGCGCCA





CCGGCAAGGGCAACGACCAGGUGCGCUUCGAGCUGAGCUGCUACAGCCUGGCCCC





CCAGAUCAAGGUGAUCGCCCCCUGGCGCAUGCCCGAGUUCUACAACCGCUUCAAG





GGCCGCAACGACCUGAUGGAGUACGCCAAGCAGCACGGCAUCCCCAUCCCCGUGA





CCCCCAAGAACCCCUGGAGCAUGGACGAGAACCUGAUGCACAUCAGCUACGAGGC





CGGCAUCCUGGAGAACCCCAAGAACCAGGCCCCCCCCGGCCUGUACACCAAGACC





CAGGACCCCGCCAAGGCCCCCAACACCCCCGACAUCCUGGAGAUCGAGUUCAAGA





AGGGCGUGCCCGUGAAGGUGACCAACGUGAAGGACGGCACCACCCACCAGACCAG





CCUGGAGCUGUUCAUGUACCUGAACGAGGUGGCCGGCAAGCACGGCGUGGGCCGC





AUCGACAUCGUGGAGAACCGCUUCAUCGGCAUGAAGAGCCGCGGCAUCUACGAGA





CCCCCGCCGGCACCAUCCUGUACCACGCCCACCUGGACAUCGAGGCCUUCACCAUG





GACCGCGAGGUGCGCAAGAUCAAGCAGGGCCUGGGCCUGAAGUUCGCCGAGCUGG





UGUACACCGGCUUCUGGCACAGCCCCGAGUGCGAGUUCGUGCGCCACUGCAUCGC





CAAGAGCCAGGAGCGCGUGGAGGGCAAGGUGCAGGUGAGCGUGCUGAAGGGCCA





GGUGUACAUCCUGGGCCGCGAGAGCCCCCUGAGCCUGUACAACGAGGAGCUGGUG





AGCAUGAACGUGCAGGGCGACUACGAGCCCACCGACGCCACCGGCUUCAUCAACA





UCAACAGCCUGCGCCUGAAGGAGUACCACCGCCUGCAGAGCAAGGUGACCGCCAA





GUGACGGGUGGCAUCCCUGUGACCCCUCCCCAGUGCCUCUCCUGGCCCUGGAAGU





UGCCACUCCAGUGCCCACCAGCCUUGUCCUAAUAAAAUUAAGUUGCAUCAAGCU.






In another example, a full length codon-optimized human argininosuccinate synthetase (ASS1) messenger RNA sequence is shown below:










(SEQ ID NO: 8)



GGACAGAUCGCCUGGAGACGCCAUCCACGCUGUUUUGACCUCCAUAGAAGACACC






GGGACCGAUCCAGCCUCCGCGGCCGGGAACGGUGCAUUGGAACGCGGAUUCCCCG





UGCCAAGAGUGACUCACCGUCCUUGACACGAUGAGCAGCAAGGGCAGCGUGGUGC





UGGCCUACAGCGGCGGCCUGGACACCAGCUGCAUCCUGGUGUGGCUGAAGGAGCA





GGGCUACGACGUGAUCGCCUACCUGGCCAACAUCGGCCAGAAGGAGGACUUCGAG





GAGGCCCGCAAGAAGGCCCUGAAGCUGGGCGCCAAGAAGGUGUUCAUCGAGGACG





UGAGCCGCGAGUUCGUGGAGGAGUUCAUCUGGCCCGCCAUCCAGAGCAGCGCCCU





GUACGAGGACCGCUACCUGCUGGGCACCAGCCUGGCCCGCCCCUGCAUCGCCCGC





AAGCAGGUGGAGAUCGCCCAGCGCGAGGGCGCCAAGUACGUGAGCCACGGCGCCA





CCGGCAAGGGCAACGACCAGGUGCGCUUCGAGCUGAGCUGCUACAGCCUGGCCCC





CCAGAUCAAGGUGAUCGCCCCCUGGCGCAUGCCCGAGUUCUACAACCGCUUCAAG





GGCCGCAACGACCUGAUGGAGUACGCCAAGCAGCACGGCAUCCCCAUCCCCGUGA





CCCCCAAGAACCCCUGGAGCAUGGACGAGAACCUGAUGCACAUCAGCUACGAGGC





CGGCAUCCUGGAGAACCCCAAGAACCAGGCCCCCCCCGGCCUGUACACCAAGACC





CAGGACCCCGCCAAGGCCCCCAACACCCCCGACAUCCUGGAGAUCGAGUUCAAGA





AGGGCGUGCCCGUGAAGGUGACCAACGUGAAGGACGGCACCACCCACCAGACCAG





CCUGGAGCUGUUCAUGUACCUGAACGAGGUGGCCGGCAAGCACGGCGUGGGCCGC





AUCGACAUCGUGGAGAACCGCUUCAUCGGCAUGAAGAGCCGCGGCAUCUACGAGA





CCCCCGCCGGCACCAUCCUGUACCACGCCCACCUGGACAUCGAGGCCUUCACCAUG





GACCGCGAGGUGCGCAAGAUCAAGCAGGGCCUGGGCCUGAAGUUCGCCGAGCUGG





UGUACACCGGCUUCUGGCACAGCCCCGAGUGCGAGUUCGUGCGCCACUGCAUCGC





CAAGAGCCAGGAGCGCGUGGAGGGCAAGGUGCAGGUGAGCGUGCUGAAGGGCCA





GGUGUACAUCCUGGGCCGCGAGAGCCCCCUGAGCCUGUACAACGAGGAGCUGGUG





AGCAUGAACGUGCAGGGCGACUACGAGCCCACCGACGCCACCGGCUUCAUCAACA





UCAACAGCCUGCGCCUGAAGGAGUACCACCGCCUGCAGAGCAAGGUGACCGCCAA





GUGAGGGUGGCAUCCCUGUGACCCCUCCCCAGUGCCUCUCCUGGCCCUGGAAGUU





GCCACUCCAGUGCCCACCAGCCUUGUCCUAAUAAAAUUAAGUUGCAUCAAAGCU.






Exemplary Formulation Protocols

A. cKK-E12


Aliquots of 50 mg/mL ethanolic solutions of cKK-E12, DOPE, cholesterol and DMG-PEG2K were mixed and diluted with ethanol to 3 mL final volume. Separately, an aqueous buffered solution (10 mM citrate/150 mM NaCl, pH 4.5) of ASS1 mRNA was prepared from a 1 mg/mL stock. The lipid solution was injected rapidly into the aqueous mRNA solution and shaken to yield a final suspension in 20% ethanol. The resulting nanoparticle suspension was filtered, diafiltrated with 1×PBS (pH 7.4), concentrated and stored at 2-8° C. Final concentration=0.64 mg/mL ASS1 mRNA (encapsulated). Zave=78 nm (Dv(50))=46 nm; Dv(90)=96 nm).


B. C12-200


Aliquots of 50 mg/mL ethanolic solutions of C12-200, DOPE, cholesterol and DMG-PEG2K were mixed and diluted with ethanol to 3 mL final volume. Separately, an aqueous buffered solution (10 mM citrate/150 mM NaCl, pH 4.5) of ASS1 mRNA was prepared from a 1 mg/mL stock. The lipid solution was injected rapidly into the aqueous mRNA solution and shaken to yield a final suspension in 20% ethanol. The resulting nanoparticle suspension was filtered, diafiltrated with 1×PBS (pH 7.4), concentrated and stored at 2-8° C. Final concentration=0.82 mg/mL ASS1 mRNA (encapsulated). Zave=86 nm (Dv(50)=50 nm; Dv(90)=101 nm).


C. HGT4003


Aliquots of 50 mg/mL ethanolic solutions of HGT4003, DOPE, cholesterol and DMG-PEG2K were mixed and diluted with ethanol to 3 mL final volume. Separately, an aqueous buffered solution (10 mM citrate/150 mM NaCl, pH 4.5) of ASS1 mRNA was prepared from a 1 mg/mL stock. The lipid solution was injected rapidly into the aqueous mRNA solution and shaken to yield a final suspension in 20% ethanol. The resulting nanoparticle suspension was filtered, diafiltrated with 1×PBS (pH 7.4), concentrated and stored at 2-8° C. Final concentration=0.82 mg/mL ASS1 mRNA (encapsulated). Zave=86 nm (Dv(50)=50 nm; Dv(90)=101 nm).


D. ICE


Aliquots of 50 mg/mL ethanolic solutions of ICE, DOPE, cholesterol and DMG-PEG2K were mixed and diluted with ethanol to 3 mL final volume. Separately, an aqueous buffered solution (10 mM citrate/150 mM NaCl, pH 4.5) of ASS1 mRNA was prepared from a 1 mg/mL stock. The lipid solution was injected rapidly into the aqueous mRNA solution and shaken to yield a final suspension in 20% ethanol. The resulting nanoparticle suspension was filtered, diafiltrated with 1×PBS (pH 7.4), concentrated and stored at 2-8° C. Final concentration=0.91 mg/mL ASS1 mRNA (encapsulated). Zave=81 nm (Dv(50)=48 nm; Dv(90)=96 nm).


E. HGT5001


Aliquots of 50 mg/mL ethanolic solutions of HGT5001, DOPE, cholesterol and DMG-PEG2K were mixed and diluted with ethanol to 3 mL final volume. Separately, an aqueous buffered solution (10 mM citrate/150 mM NaCl, pH 4.5) of ASS1 mRNA was prepared from a 1 mg/mL stock. The lipid solution was injected rapidly into the aqueous mRNA solution and shaken to yield a final suspension in 20% ethanol. The resulting nanoparticle suspension was filtered, diafiltrated with 1×PBS (pH 7.4), concentrated and stored at 2-8° C. Final concentration=0.20 mg/mL ASS1 mRNA (encapsulated). Zave=87.0 nm (Dv(50)=75 nm; Dv(90)=103 nm).


F. HGT5000


Aliquots of 50 mg/mL ethanolic solutions of HGT5000, DOPE, cholesterol and DMG-PEG2K were mixed and diluted with ethanol to 3 mL final volume. Separately, an aqueous buffered solution (10 mM citrate/150 mM NaCl, pH 4.5) of ASS1 mRNA was prepared from a 1 mg/mL stock. The lipid solution was injected rapidly into the aqueous mRNA solution and shaken to yield a final suspension in 20% ethanol. The resulting nanoparticle suspension was filtered, diafiltrated with 1×PBS (pH 7.4), concentrated and stored at 2-8° C. Final concentration=0.20 mg/mL ASS1 mRNA (encapsulated). Zave=81 nm (Dv(50)=67 nm; Dv(90)=97 nm).


G. DLinKC2DMA


Aliquots of 50 mg/mL ethanolic solutions of DLinKC2DMA, DOPE, cholesterol and DMG-PEG2K were mixed and diluted with ethanol to 3 mL final volume. Separately, an aqueous buffered solution (10 mM citrate/150 mM NaCl, pH 4.5) of ASS1 mRNA was prepared from a 1 mg/mL stock. The lipid solution was injected rapidly into the aqueous mRNA solution and shaken to yield a final suspension in 20% ethanol. The resulting nanoparticle suspension was filtered, diafiltrated with 1×PBS (pH 7.4), concentrated and stored at 2-8° C. Final concentration=0.20 mg/mL ASS1 mRNA (encapsulated). Zave=78 nm (Dv(50)=60 nm; Dv(90)=92 nm).


H. DODAP


Aliquots of 50 mg/mL ethanolic solutions of DODAP, DOPE, cholesterol and DMG-PEG2K were mixed and diluted with ethanol to 3 mL final volume. Separately, an aqueous buffered solution (10 mM citrate/150 mM NaCl, pH 4.5) of ASS1 mRNA was prepared from a 1 mg/mL stock. The lipid solution was injected rapidly into the aqueous mRNA solution and shaken to yield a final suspension in 20% ethanol. The resulting nanoparticle suspension was filtered, diafiltrated with 1×PBS (pH 7.4), concentrated and stored at 2-8° C. Final concentration=0.20 mg/mL ASS1 mRNA (encapsulated). Zave=84 nm (Dv(50)=62 nm; Dv(90)=92 nm).


I. DODMA


Aliquots of 50 mg/mL ethanolic solutions of DODMA, DOPE, cholesterol and DMG-PEG2K were mixed and diluted with ethanol to 3 mL final volume. Separately, an aqueous buffered solution (10 mM citrate/150 mM NaCl, pH 4.5) of ASS1 mRNA was prepared from a 1 mg/mL stock. The lipid solution was injected rapidly into the aqueous mRNA solution and shaken to yield a final suspension in 20% ethanol. The resulting nanoparticle suspension was filtered, diafiltrated with 1×PBS (pH 7.4), concentrated and stored at 2-8° C. Final concentration=0.20 mg/mL ASS1 mRNA (encapsulated). Zave=86 nm (Dv(50)=69 nm; Dv(90)=98 nm).


Example 2
Administration of ASS1 mRNA-Loaded Liposome Nanoparticles

This example illustrates exemplary methods of administering ASS1 mRNA-loaded liposome nanoparticles and methods for analyzing expressed protein in various target tissues in vivo.


All studies were performed using male CD-1 mice of approximately 6-8 weeks of age at the beginning of each experiment. Samples were introduced by a single bolus tail-vein injection of an equivalent total dose of 1.0 mg/kg (or otherwise specified) of encapsulated ASS1 mRNA. Mice were sacrificed and perfused with saline at the designated time points.


Tissues such as liver, spleen, kidney and heart of each mouse were harvested, apportioned into separate parts, and stored in either 10% neutral buffered formalin or snap-frozen and stored at −80° C. for analysis.


All animals were euthanized by CO2 asphyxiation at designated time points post dose administration (±5%) followed by thoracotomy and terminal cardiac blood collection. Whole blood (maximal obtainable volume) was collected via cardiac puncture on euthanized animals into serum separator tubes, allowed to clot at room temperature for at least 30 minutes, centrifuged at 22° C.±5° C. at 9300 g for 10 minutes, and the serum was extracted. For interim blood collections, approximately 40-50 μL of whole blood was collected via facial vein puncture or tail snip. Samples collected from non-treatment animals were used as baseline ASS1 levels for comparison to study animals.


Enzyme-Linked Immunosorbent Assay (ELISA) Analysis—Human ASS1 ELISA


Standard ELISA procedures were followed employing mouse anti-ASS1 2D1-2E12 IgG as the capture antibody with rabbit anti-ASS1 #3285 IgG as the secondary (detection) antibody (Shire Human Genetic Therapies). Horseradish peroxidase (HRP)-conjugated goat anti-rabbit IgG was used for activation of the 3,3′,5,5′-tetramethylbenzidine (TMB) substrate solution. The reaction was quenched using 2NH2SO4 after 20 minutes. Detection was monitored via absorption (450 nm) on a Molecular Device SpectraMax instrument. Untreated mouse serum and organs and human ASS1 protein were used as negative and positive controls, respectively.


Example 3
Efficient ASS1 Protein Expression In Vivo

This example demonstrates that administration of ASS1 mRNA results in successful protein production and clinical efficacy in vivo.


The production of human ASS1 protein via codon-optimized hASS1 mRNA-loaded lipid nanoparticles was tested in CD-1 mice as a single, bolus intravenous injection. FIG. 1 represents the amount of human ASS1 protein detected via ELISA when mice were treated with human ASS1 mRNA-loaded cKK-E12-based lipid nanoparticles at various doses. The mice were sacrificed twenty-four hours post-injection and organs were harvested (as described above).


As shown in FIG. 1, a clear dose response was achieved when measuring liver levels of human ASS1 protein. The dosing range was from 0.10-2.0 mg/kg of encapsulated human ASS1 mRNA. These data demonstrate the ability of the lipid nanoparticles to accumulate in the liver and release the mRNA payload and the liver to process this exogenous mRNA via translation to produce human ASS1 protein. Raw values of human ASS1 protein as measured via ELISA analysis (as depicted in FIG. 1) were shown in Table 1 below.












TABLE 1







Dose Encapsulated
Human



ASS1 mRNA (mg/kg)
ASS1 Protein (ng/mg total protein)



















0.10
BLD



0.30
BLD



0.60
546



1.0
1388



2.0
3371











Codon-optimized human ASS1 mRNA was delivered via cKK-E12-based lipid nanoparticles. Doses are based on encapsulated ASS1 mRNA. Values are depicted as nanogram of human ASS1 protein per milligram total protein in liver. BLD=Below Limit of Detection for ELISA.


While the sensitivity of the ELISA has limitations at lower values, western blot analysis allows for clear visualization of the human ASS1 protein at lower doses (0.30 mg/kg) (FIGS. 2A-2D).


To further understand the ability of ASS1 mRNA-encapsulated lipid nanoparticles to facilitate the delivery of mRNA to selected organs (liver), we pursued a pharmacokinetic study monitoring human ASS1 protein levels in the liver over a one week time period. FIG. 3 depicts the quantity of human ASS1 protein detected in the liver at various time points after administration of human ASS1-loaded lipid nanoparticles (cKK-E12). This was accomplished as a single dose (1.0 mg/kg encapsulated mRNA) given intravenously.


In this case we observed a maximum serum level of human ASS1 protein at approximately 24-48 hours post-administration. Measurable levels of protein were still observed 1 week post-administration as determined by both ELISA and western blot (FIGS. 3 and 4A-4E, respectively).


Direct detection of the active pharmaceutical ingredient (ASS1 mRNA) in the livers of the treated mice was achieved using in situ hybridization (ISH) based methods. As demonstrated in FIGS. 5A-5I, the exogenous human ASS1 messenger RNA could be detected in high levels at the earliest time point tested (30 minutes) and the signal remained strong for 48 hours after dosing. Further, human ASS1 mRNA was still detectable 7 days post-administration.


In addition to ISH, detection of the resulting human ASS1 protein was achieved using immunohistochemical (IHC) means. Using a mouse monoclonal antibody (02D2-2E12) for specific binding, we readily observed the target human ASS1 protein in the cytoplasm of hepatocytes of treated livers. The signal was first observed in treated livers faintly within 30 minutes but clearly within 3 hours post-administration. FIGS. 6A-6I show the staining of human ASS1 protein in treated mouse livers as a function of time after administration.


Further, one observes widespread distribution throughout the liver with strong detection of human ASS1 protein in both the sinusoidal cells as well as the target hepatocyte cells. FIGS. 7A-7B represent a low magnification representation of positive IHC staining for human ASS1 protein 24 hours post-administration.


The delivery of human ASS1 mRNA and subsequent protein production is not limited to a single lipid nanoparticle system. Several cationic lipid-based nanoparticle systems were explored for their ability to deliver mRNA and produce the desired protein. A screen of 10 different cationic lipid systems was investigated using human ASS1 mRNA as the analyte of choice. The cationic lipid component for each formulation is listed in Table 2 as well as depicted in FIG. 8. Single, intravenous injections were administered and liver samples were taken 24 hours post-administration.


Doses of formulations were all 1.0 mg/kg based on encapsulated mRNA Values are based on liver samples 24 hours post-administration.











TABLE 2







Human


Cationic/Ionizable

ASS1 Protein


Lipid
Dose
(ng/mg total


Component
(mg/kg)
protein)

















cKK-E12 (1)
1.0
2,028


cKK-E12 (2)
1.0
911


ICE
1.0
663


C12-200
1.0
385


HGT4003
1.0
100










Raw values of human ASS1 protein for various cationic lipid-based nanoparticle systems as measured via ELISA analysis (as depicted in FIG. 9). All doses were administered intravenously at 1.0 mg/kg. Values of protein are depicted as nanogram of human ASS1 protein per milligram of total liver protein. cKK-E12 (1) has a lower percentage of PEG lipid than cKK-E12 (2) (3% vs 5% PEG lipid).


While the production of protein via mRNA-loaded lipid nanoparticles can be detected, we further determined if the resulting protein is active and can function properly. To this end, in vitro activity studies were conducted which measured incorporation of 14C arginine into cellular proteins via supplementation of 14C citrulline. The radioactive citrulline was converted to 14C-argininosuccinate, and subsequently to 14C-arginine, in the presence of active ASS1 protein. By comparing human ASS1 mRNA transfected cells versus untreated cells, we could gauge the activity of the respective exogenous mRNA-derived ASS1 protein. FIG. 9 represents radioactive counts per minute of 14C arginine incorporation into cellular proteins. Transfection of SK (−) cells (ASS1 protein knockout cell line) with human ASS1 mRNA exposed to depleted media (no arginine or leucine) resulted in an increase in observed radioactivity as compared to untreated SK(−) cells. Activity measured in these transfected cells was comparable to a stably-transfected positive ASS1 cell line (SK (+)).


Example 4
Human ASS1 Protein Levels Following Treatment with ASS1 mRNA-Loaded Lipid Nanoparticles

This example demonstrates that administration of ASS1 mRNA results in successful production of ASS1 protein in the liver.


Male CD-1 mice were administered a single dose of 1.0 mg/kg of lipid nanoparticles (ASS1 mRNA-loaded cKK-E12-based lipid nanoparticle) intravenously, or untreated (i.e., control), as described above in Example 2. The mice were sacrificed and the organs were collected 24 hours post-administration. Human argininosuccinate synthetase (ASS1) protein levels in the liver were measured by ELISA. These data demonstrate increased levels of ASS1 protein were detected relative to the control and that the protein produced resulted from ASS1 mRNA delivered intravenously (FIG. 10).


Example 5
Plasma Ammonia Levels Following Treatment with ASS1 mRNA-Loaded Lipid Nanoparticles

This example demonstrates that administration of ASS1 mRNA results in successful reduction of plasma ammonia levels.


ASS1 knockout mice were administered 1.0 mg/kg of ASS1 mRNA lipid nanoparticles (ASS1 mRNA-loaded cKK-E12-based lipid nanoparticle) or empty lipid nanoparticles once every 14 days for 30 days as described above in Example 2. Mice which were administered empty lipid nanoparticles served as the vehicle control. Additional controls included untreated wild-type mice and untreated ASS1 knockout mice. Prior to each dose on days 1, 15 and 29, plasma samples were collected (i.e., pre-dose). Plasma samples were also collected within 24 hours following each dose on days 2, 16 and 30. Additional plasma samples were collected on days 8 and 22. Plasma ammonia levels were quantified in all samples and demonstrated that plasma ammonia levels were reproducibly reduced for at least 24 hours following treatment to levels near those observed in wild-type mice.


EQUIVALENTS

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:

Claims
  • 1. A method of treating Argininosuccinate Synthetase Deficiency (ASD), the method comprising administering intravenously to a subject with the ASD a composition comprising an mRNA-loaded liposome at an effective dose and administration interval to treat the ASD;wherein the administering results in reduced ammonia levels in a blood sample from the treated subject as compared to a baseline ammonia level before treatment;wherein the mRNA encodes an human argininosuccinate synthetase (ASS1) and comprises a nucleotide sequence at least 90% identical to
  • 2. The method of claim 1, wherein the one or more cationic lipids comprise a cationic lipid selected from the group consisting of C12-200, MC3, DLinDMA, DLinkC2DMA, cKK-E12, ICE (Imidazol-based), HGT5000, HGT5001, DODAC, DDAB, DMRIE, DOSPA, DOGS, DODAP, DODMA and DMDMA, DODAC, DLenDMA, DMRIE, CLinDMA, CpLinDMA, DMOBA, DOcarbDAP, DLinDAP, DLincarbDAP, DLinCDAP, KLin-K-DMA, DLin-K-XTC2-DMA, HGT4003, and combination thereof.
  • 3. The method of claim 1, wherein the one or more cholesterol-based lipids are at least one of cholesterol and PEGylated cholesterol.
  • 4. The method of claim 1, wherein the one or more cationic lipids constitutes about 30-50% of the liposome by weight.
  • 5. The method of claim 1, wherein the ratio of the one or more cationic lipids: the one or more non-cationic lipids: the one or more cholesterol-based lipids: the one or more PEG-modified lipids is approximately 40:30:25:5 by molar ratio.
  • 6. The method of claim 1, wherein the liposome comprises a combination selected from: cKK-E12, DOPE, cholesterol and DMG-PEG2K;C12-200, DOPE, cholesterol and DMG-PEG2K;HGT4003, DOPE, cholesterol and DMG-PEG2K; or ICE, DOPE, cholesterol and DMG-PEG2K.
  • 7. The method of claim 1, wherein the effective dose ranges from about 0.01 to 5.0 mg/kg body weight.
  • 8. The method of claim 1, wherein the composition is administered once a week.
  • 9. The method of claim 1, wherein the administering of the composition results in a reduced citrulline level in the subject as compared to a baseline citrulline level before the treatment.
  • 10. The method of claim 1, wherein the mRNA is codon optimized.
  • 11. The method of claim 1, wherein the mRNA comprises one or more modified nucleotides.
  • 12. The method of claim 1, wherein the mRNA is unmodified.
  • 13. The method of claim 1, wherein the mRNA comprises a nucleotide sequence identical to
  • 14. A method of treating Argininosuccinate Synthetase Deficiency (ASD), the method comprising administering intravenously to a subject with the ASD a composition comprising an mRNA molecule encoding an human argininosuccinate synthetase (ASS1) encapsulated in a liposome at an effective dose and an administration interval such that the administering of the composition results in reduced ammonia levels in a blood sample from the treated subject as compared to a baseline ammonia level before treatment,wherein the liposome has a diameter of less than about 100 nm, andwherein the liposome comprises cKK-E12:
  • 15. The method of claim 14, wherein the mRNA comprises a nucleotide sequence at least 90% identical to
  • 16. The method of claim 14, wherein the mRNA comprises a nucleotide sequence identical to
RELATED APPLICATIONS

This application claims priority to U.S. Provisional Application Ser. No. 61/894,294, filed Oct. 22, 2013, the disclosure of which is hereby incorporated by reference. The instant application contains a Sequence Listing which has been submitted electronically in ASCII format and is hereby incorporated by reference in its entirety. Said ASCII copy, created on Dec. 22, 2014, is named “2006685-0691_SL.txt” and is 22,079 bytes in size. The entire contents of the Sequence Listing are herein incorporated by reference.

US Referenced Citations (435)
Number Name Date Kind
2647121 Jacoby Jul 1953 A
2717909 Kosmin Sep 1955 A
2819718 Goldman Jan 1958 A
2844629 William et al. Jul 1958 A
3096560 Liebig Jul 1963 A
3535289 Yoshihara et al. Oct 1970 A
3614954 Mirowski et al. Oct 1971 A
3614955 Mirowski Oct 1971 A
3656185 Carpentier Apr 1972 A
3805301 Liebig Apr 1974 A
3945052 Liebig Mar 1976 A
3995623 Blake et al. Dec 1976 A
4013507 Rembaum Mar 1977 A
4072146 Howes Feb 1978 A
4096860 McLaughlin Jun 1978 A
4099528 Sorenson et al. Jul 1978 A
4106129 Carpentier et al. Aug 1978 A
4134402 Mahurkar Jan 1979 A
4140126 Choudhury Feb 1979 A
4180068 Jacobsen et al. Dec 1979 A
4182833 Hicks Jan 1980 A
4227533 Godfrey Oct 1980 A
4284459 Patel et al. Aug 1981 A
4308085 Horhold et al. Dec 1981 A
4323525 Bornat Apr 1982 A
4335723 Patel Jun 1982 A
4339369 Hicks et al. Jul 1982 A
4355426 MacGregor Oct 1982 A
4375817 Engle et al. Mar 1983 A
4385631 Uthmann May 1983 A
4401472 Gerber Aug 1983 A
4406656 Hattler et al. Sep 1983 A
4475972 Wong Oct 1984 A
4530113 Matterson Jul 1985 A
4550447 Seiler, Jr. et al. Nov 1985 A
4562596 Kornberg Jan 1986 A
4568329 Mahurkar Feb 1986 A
4571241 Christopher Feb 1986 A
4601718 Possis et al. Jul 1986 A
4647416 Seiler, Jr. et al. Mar 1987 A
4662382 Sluetz et al. May 1987 A
4701162 Rosenberg Oct 1987 A
4710169 Christopher Dec 1987 A
4720517 Ravichandran et al. Jan 1988 A
4737323 Martin et al. Apr 1988 A
4762915 Kung et al. Aug 1988 A
4782836 Alt Nov 1988 A
4856521 Irnich Aug 1989 A
4860751 Callaghan Aug 1989 A
4878908 Martin et al. Nov 1989 A
4892540 Vallana Jan 1990 A
4897355 Eppstein et al. Jan 1990 A
4920016 Allen et al. Apr 1990 A
4946683 Forssen Aug 1990 A
4946857 Kanehira et al. Aug 1990 A
4960409 Catalano Oct 1990 A
4966945 Drawert et al. Oct 1990 A
5024671 Tu et al. Jun 1991 A
5025005 Nomura et al. Jun 1991 A
5047540 Kamata et al. Sep 1991 A
5101824 Lekholm Apr 1992 A
5104399 Lazarus Apr 1992 A
5116360 Pinchuk et al. May 1992 A
5138067 Kamata et al. Aug 1992 A
5151105 Kwan-Gett Sep 1992 A
5171678 Behr et al. Dec 1992 A
5176661 Evard et al. Jan 1993 A
5194654 Hostetler et al. Mar 1993 A
5200395 Eto et al. Apr 1993 A
5223263 Hostetler et al. Jun 1993 A
5261419 Osypka Nov 1993 A
5264618 Felgner et al. Nov 1993 A
5279833 Rose Jan 1994 A
5282824 Gianturco Feb 1994 A
5284491 Sutton et al. Feb 1994 A
5300022 Klapper et al. Apr 1994 A
5314430 Bardy May 1994 A
5330768 Park et al. Jul 1994 A
5334761 Gebeyehu et al. Aug 1994 A
5395619 Zalipsky et al. Mar 1995 A
5405363 Kroll et al. Apr 1995 A
5405379 Lane Apr 1995 A
5455352 Huellmann et al. Oct 1995 A
5464924 Silvis et al. Nov 1995 A
5503852 Steiner et al. Apr 1996 A
5528023 Butturini et al. Jun 1996 A
5552155 Bailey et al. Sep 1996 A
5595756 Bally et al. Jan 1997 A
5607385 Francischelli et al. Mar 1997 A
5609624 Kalis Mar 1997 A
5610283 Buechler Mar 1997 A
5614548 Piantadosi et al. Mar 1997 A
5626869 Nyqvist et al. May 1997 A
5631018 Zalipsky et al. May 1997 A
5677124 DuBois et al. Oct 1997 A
5693088 Lazarus Dec 1997 A
5697953 Kroll et al. Dec 1997 A
5700437 Fujii et al. Dec 1997 A
5705188 Junichi et al. Jan 1998 A
5705385 Bally et al. Jan 1998 A
5736573 Galat Apr 1998 A
5744335 Wolff et al. Apr 1998 A
5772694 Bokros et al. Jun 1998 A
5776165 Ripart Jul 1998 A
5776747 Schinstine et al. Jul 1998 A
5783383 Kondo et al. Jul 1998 A
5844107 Hanson et al. Dec 1998 A
5874105 Watkins et al. Feb 1999 A
5885613 Holland et al. Mar 1999 A
5910168 Myers et al. Jun 1999 A
5916208 Luther et al. Jun 1999 A
5965434 Wolff et al. Oct 1999 A
5976567 Wheeler et al. Nov 1999 A
5976569 Milstein Nov 1999 A
5981501 Wheeler et al. Nov 1999 A
6055454 Heemels Apr 2000 A
6067471 Warren May 2000 A
6090384 Ra et al. Jul 2000 A
6096070 Ragheb et al. Aug 2000 A
6096075 Bokros et al. Aug 2000 A
6120799 McDonald et al. Sep 2000 A
6147055 Hobart et al. Nov 2000 A
6152955 KenKnight et al. Nov 2000 A
6165763 Brown et al. Dec 2000 A
6169923 Kroll Jan 2001 B1
6176877 Buchanan et al. Jan 2001 B1
6204297 Tracy et al. Mar 2001 B1
6210892 Bennett et al. Apr 2001 B1
6214804 Felgner et al. Apr 2001 B1
6271208 Bischoff Aug 2001 B1
6271209 Smith et al. Aug 2001 B1
6287591 Semple et al. Sep 2001 B1
6299604 Ragheb et al. Oct 2001 B1
6335199 Bischoff et al. Jan 2002 B1
6358278 Brendzel et al. Mar 2002 B1
6370434 Zhang et al. Apr 2002 B1
6371983 Lane Apr 2002 B1
6417326 Cullis et al. Jul 2002 B1
6485726 Blumberg et al. Nov 2002 B1
6534484 Wheeler et al. Mar 2003 B1
6585410 Ryan Jul 2003 B1
6586410 Wheeler et al. Jul 2003 B1
6670178 Selden et al. Dec 2003 B1
6696424 Wheeler Feb 2004 B1
6733777 Erbacher et al. May 2004 B2
6743823 Summar et al. Jun 2004 B1
6756055 McDonald et al. Jun 2004 B2
6790838 Alison et al. Sep 2004 B2
6815432 Wheeler et al. Nov 2004 B2
6821530 Koob et al. Nov 2004 B2
6835395 Semple et al. Dec 2004 B1
6858224 Wheeler et al. Feb 2005 B2
6858225 Semple et al. Feb 2005 B2
6887665 Trulson et al. May 2005 B2
6998115 Langer et al. Feb 2006 B2
7022214 Olech Apr 2006 B2
7067697 Gao Jun 2006 B2
7084303 Watanabe et al. Aug 2006 B2
7341738 Semple et al. Mar 2008 B2
7422902 Wheeler et al. Sep 2008 B1
7427394 Anderson et al. Sep 2008 B2
7507859 Grinstaff et al. Mar 2009 B2
7556684 Bury et al. Jul 2009 B2
7745651 Heyes et al. Jun 2010 B2
7799565 MacLachlan et al. Sep 2010 B2
7803397 Heyes et al. Sep 2010 B2
7901708 MacLachlan et al. Mar 2011 B2
7972435 Bury et al. Jul 2011 B2
8021686 Semple et al. Sep 2011 B2
8071082 Zugates et al. Dec 2011 B2
8101741 MacLachlan et al. Jan 2012 B2
8106022 Manoharan et al. Jan 2012 B2
8158601 Chen et al. Apr 2012 B2
8188263 MacLachlan et al. May 2012 B2
RE43612 Anderson et al. Aug 2012 E
8236943 Lee et al. Aug 2012 B2
8278036 Kariko et al. Oct 2012 B2
8287849 Langer et al. Oct 2012 B2
8329070 MacLachlan et al. Dec 2012 B2
8450298 Mahon et al. May 2013 B2
8450467 Manoharan et al. May 2013 B2
8513403 MacLachlan et al. Aug 2013 B2
8557231 Langer et al. Oct 2013 B2
8562966 Zugates et al. Oct 2013 B2
8569256 Heyes et al. Oct 2013 B2
8652512 Schmehl et al. Feb 2014 B2
8691966 Kariko et al. Apr 2014 B2
8710200 Schrum et al. Apr 2014 B2
8748089 Kariko et al. Jun 2014 B2
8802644 Chen et al. Aug 2014 B2
8808681 Anderson et al. Aug 2014 B2
8808982 Dahl et al. Aug 2014 B2
8822663 Schrum et al. Sep 2014 B2
8828956 Manoharan et al. Sep 2014 B2
8835108 Kariko et al. Sep 2014 B2
8846348 Jendrisak et al. Sep 2014 B2
8853377 Guild et al. Oct 2014 B2
8859229 Rabinovich et al. Oct 2014 B2
8883202 Manoharan et al. Nov 2014 B2
8936942 Heyes et al. Jan 2015 B2
8969353 Mahon et al. Mar 2015 B2
8980864 Hoge et al. Mar 2015 B2
8999351 Manoharan et al. Apr 2015 B2
8999950 MacLachlan et al. Apr 2015 B2
9005930 Jendrisak et al. Apr 2015 B2
9012219 Kariko et al. Apr 2015 B2
9012498 Manoharan et al. Apr 2015 B2
9018187 Heyes et al. Apr 2015 B2
9040256 Grunenwald et al. May 2015 B2
9051567 Fitzgerald et al. Jun 2015 B2
9061021 Guild et al. Jun 2015 B2
9061059 Chakraborty et al. Jun 2015 B2
9074208 MacLachlan et al. Jul 2015 B2
9080211 Grunenwald et al. Jul 2015 B2
9085801 Grunenwald et al. Jul 2015 B2
9089604 Chakraborty et al. Jul 2015 B2
9095552 Chakraborty et al. Aug 2015 B2
9107886 Chakraborty et al. Aug 2015 B2
9114113 Chakraborty et al. Aug 2015 B2
9192651 Chakraborty et al. Nov 2015 B2
9220755 Chakraborty et al. Dec 2015 B2
9220792 Chakraborty et al. Dec 2015 B2
9233141 Chakraborty et al. Jan 2016 B2
9254311 Bancel et al. Feb 2016 B2
9295689 de Fougerolles et al. Mar 2016 B2
9301993 Chakraborty et al. Apr 2016 B2
9303079 Chakraborty et al. Apr 2016 B2
9334328 Schrum et al. May 2016 B2
9345780 Manoharan et al. May 2016 B2
9352042 Heyes et al. May 2016 B2
9352048 Manoharan et al. May 2016 B2
9364435 Yaworski et al. Jun 2016 B2
9394234 Chen et al. Jul 2016 B2
9428751 MacDonald et al. Aug 2016 B2
9464124 Bancel et al. Oct 2016 B2
20020022721 Trulson et al. Feb 2002 A1
20020094528 Salafsky Jul 2002 A1
20020192651 Wheeler et al. Dec 2002 A1
20020192721 Rizzuto et al. Dec 2002 A1
20020193622 Watanabe et al. Dec 2002 A1
20030082154 Leamon May 2003 A1
20030083272 Wiederholt et al. May 2003 A1
20030104044 Semple et al. Jun 2003 A1
20030181410 Wheeler et al. Sep 2003 A1
20030215395 Yu et al. Nov 2003 A1
20040110709 Li et al. Jun 2004 A1
20040132683 Felgner et al. Jul 2004 A1
20040142025 MacLachlan et al. Jul 2004 A1
20040224912 Dobie et al. Nov 2004 A1
20040235982 Rabasco et al. Nov 2004 A1
20050004058 Benoit et al. Jan 2005 A1
20050008689 Semple et al. Jan 2005 A1
20050032730 Von Der Mulbe et al. Feb 2005 A1
20050054026 Atsushi et al. Mar 2005 A1
20050059005 Tuschl et al. Mar 2005 A1
20050059624 Hoerr et al. Mar 2005 A1
20050065107 Hobart et al. Mar 2005 A1
20050069590 Buehler et al. Mar 2005 A1
20050079212 Wheeler et al. Apr 2005 A1
20050143332 Monahan et al. Jun 2005 A1
20050148786 Ikeda et al. Jul 2005 A1
20050158302 Faustman et al. Jul 2005 A1
20050244961 Short et al. Nov 2005 A1
20050250723 Hoerr et al. Nov 2005 A1
20060008910 MacLachlan et al. Jan 2006 A1
20060059576 Pasinetti et al. Mar 2006 A1
20060069225 Wintermantel et al. Mar 2006 A1
20060083780 Heyes et al. Apr 2006 A1
20060172003 Meers et al. Aug 2006 A1
20060204566 Smyth-Templeton et al. Sep 2006 A1
20060216343 Panzner et al. Sep 2006 A1
20060223939 Lange et al. Oct 2006 A1
20060228404 Anderson et al. Oct 2006 A1
20060241071 Grinstaff et al. Oct 2006 A1
20070135372 MacLachlan et al. Jun 2007 A1
20070142628 Ghoshal et al. Jun 2007 A1
20070172950 Wheeler et al. Jul 2007 A1
20070252295 Panzner et al. Nov 2007 A1
20070275923 Chen et al. Nov 2007 A1
20070281336 Jendrisak et al. Dec 2007 A1
20080145338 Anderson et al. Jun 2008 A1
20080160048 Fuller Jul 2008 A1
20080242626 Zugates et al. Oct 2008 A1
20080260706 Rabinovich et al. Oct 2008 A1
20090023673 Manoharan et al. Jan 2009 A1
20090093433 Woolf et al. Apr 2009 A1
20090163705 Manoharan et al. Jun 2009 A1
20090186805 Tabor et al. Jul 2009 A1
20090221684 Grinstaff et al. Sep 2009 A1
20090263407 Dande et al. Oct 2009 A1
20090270481 MacLachlan et al. Oct 2009 A1
20090286852 Kariko et al. Nov 2009 A1
20090326051 Corey et al. Dec 2009 A1
20100028943 Thomas et al. Feb 2010 A1
20100035249 Hayashizaki et al. Feb 2010 A1
20100036084 Langer et al. Feb 2010 A1
20100041152 Wheeler et al. Feb 2010 A1
20100047261 Hoerr et al. Feb 2010 A1
20100120129 Amshey et al. May 2010 A1
20100178699 Gao et al. Jul 2010 A1
20100189729 Hoerr et al. Jul 2010 A1
20100267806 Bumcrot et al. Oct 2010 A1
20100331234 Mahon et al. Dec 2010 A1
20110009641 Anderson et al. Jan 2011 A1
20110038941 Lee et al. Feb 2011 A1
20110092739 Chen et al. Apr 2011 A1
20110143397 Kariko et al. Jun 2011 A1
20110200582 Baryza et al. Aug 2011 A1
20110244026 Guild et al. Oct 2011 A1
20110256175 Hope et al. Oct 2011 A1
20110287435 Grunenwald et al. Nov 2011 A1
20110293703 Mahon et al. Dec 2011 A1
20110311583 Manoharan et al. Dec 2011 A1
20120007803 Takatsuka Jan 2012 A1
20120009222 Nguyen et al. Jan 2012 A1
20120065252 Schrum et al. Mar 2012 A1
20120065358 Langer et al. Mar 2012 A1
20120114831 Semple et al. May 2012 A1
20120128760 Manoharan et al. May 2012 A1
20120129910 Thompson et al. May 2012 A1
20120142756 Guild et al. Jun 2012 A1
20120195936 Rudolph et al. Aug 2012 A1
20120202871 Heyes et al. Aug 2012 A1
20120237975 Schrum et al. Sep 2012 A1
20120251560 Dahlman et al. Oct 2012 A1
20120251618 Schrum et al. Oct 2012 A1
20120328668 MacLachlan et al. Dec 2012 A1
20130017223 Hope et al. Jan 2013 A1
20130158021 Dong et al. Jun 2013 A1
20130195967 Guild et al. Aug 2013 A1
20130237594 de Fougerolles et al. Sep 2013 A1
20130259923 Bancel et al. Oct 2013 A1
20130259924 Bancel et al. Oct 2013 A1
20130266640 de Fougerolles et al. Oct 2013 A1
20130302401 Ma et al. Nov 2013 A1
20140010861 Bancel et al. Jan 2014 A1
20140044772 MacLachlan et al. Feb 2014 A1
20140094399 Langer et al. Apr 2014 A1
20140105964 Bancel et al. Apr 2014 A1
20140105965 Bancel et al. Apr 2014 A1
20140147432 Bancel et al. May 2014 A1
20140147454 Chakraborty et al. May 2014 A1
20140148502 Bancel et al. May 2014 A1
20140155472 Bancel et al. Jun 2014 A1
20140155473 Bancel et al. Jun 2014 A1
20140155474 Bancel et al. Jun 2014 A1
20140155475 Bancel et al. Jun 2014 A1
20140161830 Anderson et al. Jun 2014 A1
20140162897 Grunenwald et al. Jun 2014 A1
20140171485 Bancel et al. Jun 2014 A1
20140179771 Bancel et al. Jun 2014 A1
20140186432 Bancel et al. Jul 2014 A1
20140193482 Bancel et al. Jul 2014 A1
20140194494 Bancel et al. Jul 2014 A1
20140199371 Bancel et al. Jul 2014 A1
20140200163 Mikkelsen et al. Jul 2014 A1
20140200261 Hoge et al. Jul 2014 A1
20140200262 Bancel et al. Jul 2014 A1
20140200263 Bancel et al. Jul 2014 A1
20140200264 Bancel et al. Jul 2014 A1
20140206752 Afeyan et al. Jul 2014 A1
20140206753 Guild et al. Jul 2014 A1
20140206755 Bancel et al. Jul 2014 A1
20140206852 Hoge et al. Jul 2014 A1
20140221248 Jendrisak et al. Aug 2014 A1
20140221465 Bancel et al. Aug 2014 A1
20140227300 Chin et al. Aug 2014 A1
20140243399 Schrum et al. Aug 2014 A1
20140249208 Bancel et al. Sep 2014 A1
20140255467 Bancel et al. Sep 2014 A1
20140255468 Bancel et al. Sep 2014 A1
20140275227 Hoge et al. Sep 2014 A1
20140275229 Bancel et al. Sep 2014 A1
20140288160 Guild et al. Sep 2014 A1
20140294937 MacLachlan et al. Oct 2014 A1
20140294938 Guild et al. Oct 2014 A1
20140294939 Guild et al. Oct 2014 A1
20140294940 Guild et al. Oct 2014 A1
20140329884 Dong et al. Nov 2014 A1
20140343129 de Fougerolles et al. Nov 2014 A1
20140363876 Jendrisak et al. Dec 2014 A1
20150004217 Guild et al. Jan 2015 A1
20150005372 Hoge et al. Jan 2015 A1
20150011615 Manoharan et al. Jan 2015 A1
20150011633 Shorr et al. Jan 2015 A1
20150017211 de Fougerolles et al. Jan 2015 A1
20150038556 Heartlein et al. Feb 2015 A1
20150038558 Kariko et al. Feb 2015 A1
20150044277 Bancel et al. Feb 2015 A1
20150050354 Bouchon et al. Feb 2015 A1
20150051268 Bancel et al. Feb 2015 A1
20150056253 Bancel et al. Feb 2015 A1
20150064235 Bancel et al. Mar 2015 A1
20150064236 Bancel et al. Mar 2015 A1
20150064242 Heyes et al. Mar 2015 A1
20150064725 Schrum et al. Mar 2015 A1
20150086614 Bancel et al. Mar 2015 A1
20150110857 DeRosa et al. Apr 2015 A1
20150110858 DeRosa et al. Apr 2015 A1
20150111248 Bancel et al. Apr 2015 A1
20150111945 Geisbert et al. Apr 2015 A1
20150119444 Manoharan et al. Apr 2015 A1
20150119445 Manoharan et al. Apr 2015 A1
20150157565 Heartlein et al. Jun 2015 A1
20150166465 Chen et al. Jun 2015 A1
20150190515 Manoharan et al. Jul 2015 A1
20150191760 Jendrisak et al. Jul 2015 A1
20150265708 Manoharan et al. Sep 2015 A1
20150267192 Heartlein et al. Sep 2015 A1
20150315541 Bancel et al. Nov 2015 A1
20150315584 MacDonald et al. Nov 2015 A1
20150366997 Guild et al. Dec 2015 A1
20160095924 Hope et al. Apr 2016 A1
20160114011 Bancel et al. Apr 2016 A1
20160115477 MacLachlan et al. Apr 2016 A1
20160115483 MacLachlan et al. Apr 2016 A1
20160136236 Hoge et al. May 2016 A1
20160151284 Heyes et al. Jun 2016 A1
20160158385 Bancel et al. Jun 2016 A1
20160193299 de Fougerolles et al. Jul 2016 A1
20160194368 Hoge et al. Jul 2016 A1
20160194625 Hoge et al. Jul 2016 A1
20160199485 Manoharan et al. Jul 2016 A1
20160213785 Manoharan et al. Jul 2016 A1
20160237108 Fraley et al. Aug 2016 A1
20160237134 Hoge et al. Aug 2016 A1
20160250354 Manoharan et al. Sep 2016 A1
20160251681 Yaworski et al. Sep 2016 A1
20160256567 Heyes et al. Sep 2016 A1
20160256568 Heyes et al. Sep 2016 A1
20160264971 Geisbert et al. Sep 2016 A1
20160274089 Ciufolini et al. Sep 2016 A1
20160304552 Roy et al. Oct 2016 A1
20160317647 Ciaramella et al. Nov 2016 A1
20160317676 Hope et al. Nov 2016 A1
Foreign Referenced Citations (193)
Number Date Country
2518132 Mar 2006 CA
2807552 Feb 2012 CA
2807 552 Sep 2012 CA
100569877 Dec 2009 CN
101863544 Oct 2010 CN
24 30 998 Jan 1975 DE
2520814 Nov 1976 DE
3728917 Mar 1989 DE
6 73 637 Sep 1995 EP
0783297 Jul 1997 EP
0853123 Jul 1998 EP
0959092 Nov 1999 EP
2449106 Nov 1999 EP
1519714 Apr 2005 EP
2045251 Apr 2009 EP
1519 714 Oct 2010 EP
2338478 Jun 2011 EP
2449 106 May 2012 EP
2532649 Dec 2012 EP
2578685 Apr 2013 EP
2823 809 Jan 2015 EP
2823809 Jan 2015 EP
2 235 112 Jan 1975 FR
1072118 Jun 1967 GB
1602085 Nov 1981 GB
H07-053535 Feb 1955 JP
S48-022365 Mar 1973 JP
S49-127908 Dec 1974 JP
S51-023537 Feb 1976 JP
51-125144 Nov 1976 JP
S63125144 May 1988 JP
63-154788 Jun 1988 JP
H09-505593 Jun 1997 JP
H10-197978 Jul 1998 JP
11-005786 Jan 1999 JP
11-080142 Mar 1999 JP
2001-523215 Nov 2001 JP
2002-167368 Jun 2002 JP
2003-519199 Jun 2003 JP
4-108173 Jun 2008 JP
2008-247749 Oct 2008 JP
50-24216 Sep 2012 JP
WO-9011092 Oct 1990 WO
WO-9318229 Sep 1993 WO
WO-9318754 Sep 1993 WO
WO-9511004 Apr 1995 WO
WO-9514651 Jun 1995 WO
WO-9527478 Oct 1995 WO
WO-9618372 Jun 1996 WO
WO-9626179 Aug 1996 WO
WO-9637211 Nov 1996 WO
WO-9640964 Dec 1996 WO
WO-9746223 Dec 1997 WO
WO-1998010748 Mar 1998 WO
WO-9816202 Apr 1998 WO
WO-9851278 Nov 1998 WO
WO-9914346 Mar 1999 WO
WO-0003044 Jan 2000 WO
WO-00062813 Oct 2000 WO
WO-0064484 Nov 2000 WO
WO-0069913 Nov 2000 WO
WO-0105375 Jan 2001 WO
WO-0107599 Feb 2001 WO
WO-0222709 Mar 2002 WO
WO-0231025 Apr 2002 WO
WO-0242317 May 2002 WO
WO-200234236 May 2002 WO
WO-03040288 May 2003 WO
WO-03070735 Aug 2003 WO
WO-2004043588 May 2004 WO
WO-2004048345 Jun 2004 WO
WO-2004106411 Dec 2004 WO
WO2005026372 Mar 2005 WO
WO-2005026372 Mar 2005 WO
WO-2005028619 Mar 2005 WO
WO-2005037226 Apr 2005 WO
WO-2005121348 Dec 2005 WO
WO-2006000448 Jan 2006 WO
WO-2006016097 Feb 2006 WO
WO-2006082088 Aug 2006 WO
WO-2006105043 Oct 2006 WO
WO-2006138380 Dec 2006 WO
WO-2007024708 Mar 2007 WO
WO-2007031091 Mar 2007 WO
WO-2007120863 Oct 2007 WO
WO-2007126386 Nov 2007 WO
WO-2007143659 Dec 2007 WO
WO-2008011561 Jan 2008 WO
WO-2008042973 Apr 2008 WO
WO-2008045548 Apr 2008 WO
WO-2008083949 Jul 2008 WO
WO-2008113364 Sep 2008 WO
WO-2009046220 Apr 2009 WO
WO-2009127060 Oct 2009 WO
WO-2009127230 Oct 2009 WO
WO-2010042877 Apr 2010 WO
WO-2010045512 Apr 2010 WO
WO-2010053572 May 2010 WO
WO-2010054401 May 2010 WO
WO-2010054405 May 2010 WO
WO-2010056403 May 2010 WO
WO-2010099387 Sep 2010 WO
WO-2010114789 Oct 2010 WO
WO-2010119256 Oct 2010 WO
WO-2010129709 Nov 2010 WO
WO-2010144740 Dec 2010 WO
WO-2010147992 Dec 2010 WO
WO-2010148013 Dec 2010 WO
WO-2011012316 Feb 2011 WO
WO-2011012746 Feb 2011 WO
WO-2011039144 Apr 2011 WO
WO-2011068810 Jun 2011 WO
WO-2011075656 Jun 2011 WO
WO-2011141705 Nov 2011 WO
WO-2012019168 Feb 2012 WO
WO-2012019630 Feb 2012 WO
WO-2012019780 Feb 2012 WO
WO-2012027675 Mar 2012 WO
WO-2012045075 Apr 2012 WO
WO-2012045082 Apr 2012 WO
WO-2012075040 Jun 2012 WO
WO-2012133737 Oct 2012 WO
WO-2012135025 Oct 2012 WO
WO-2012135805 Oct 2012 WO
WO-2012170889 Dec 2012 WO
WO-2012170930 Dec 2012 WO
WO-2013039857 Mar 2013 WO
WO-2013039861 Mar 2013 WO
WO-2013063468 May 2013 WO
WO2013090186 Jun 2013 WO
WO-2013101690 Jul 2013 WO
WO-2013102203 Jul 2013 WO
WO-2013126803 Aug 2013 WO
WO-2013149140 Oct 2013 WO
WO-2013149141 Oct 2013 WO
WO-2013151663 Oct 2013 WO
WO-2013151664 Oct 2013 WO
WO-2013151666 Oct 2013 WO
WO-2013151667 Oct 2013 WO
WO-2013151668 Oct 2013 WO
WO-2013151670 Oct 2013 WO
WO-2013151671 Oct 2013 WO
WO-2013151672 Oct 2013 WO
WO-2013151736 Oct 2013 WO
WO-2013182683 Dec 2013 WO
WO-2013185067 Dec 2013 WO
WO-2013185069 Dec 2013 WO
WO-2014028487 Feb 2014 WO
WO-2014089486 Jun 2014 WO
WO2014113089 Jul 2014 WO
WO-2014113089 Jul 2014 WO
WO-2014144039 Sep 2014 WO
WO-2014144196 Sep 2014 WO
WO-2014144711 Sep 2014 WO
WO-2014144767 Sep 2014 WO
WO-2014152027 Sep 2014 WO
WO-2014152030 Sep 2014 WO
WO-2014152031 Sep 2014 WO
WO-2014152211 Sep 2014 WO
WO-2014152513 Sep 2014 WO
WO-2014152540 Sep 2014 WO
WO-2014152659 Sep 2014 WO
WO-2014152673 Sep 2014 WO
WO-2014152774 Sep 2014 WO
WO-2014152940 Sep 2014 WO
WO-2014152966 Sep 2014 WO
WO-2014153052 Sep 2014 WO
WO-2014158795 Oct 2014 WO
WO-2014159813 Oct 2014 WO
WO-2014179562 Nov 2014 WO
WO-2014210356 Dec 2014 WO
2015006747 Jan 2015 WO
WO-2015006747 Jan 2015 WO
WO-2015011633 Jan 2015 WO
WO-2015048744 Apr 2015 WO
WO-2015051169 Apr 2015 WO
WO-2015051173 Apr 2015 WO
WO-2015058069 Apr 2015 WO
WO2015085318 Jun 2015 WO
WO2015089511 Jun 2015 WO
WO2015011633 Jan 2016 WO
2016054421 Apr 2016 WO
WO2016054421 Apr 2016 WO
2016071857 May 2016 WO
2016077123 May 2016 WO
2016077125 May 2016 WO
WO2016071857 May 2016 WO
WO2016077123 May 2016 WO
WO2016077125 May 2016 WO
WO2016118724 Jul 2016 WO
WO2016118725 Jul 2016 WO
WO2016154127 Sep 2016 WO
WO2016164762 Oct 2016 WO
Non-Patent Literature Citations (308)
Entry
Chandler et al, Liver-directed adeno-associated virus serotype 8 gene transfer rescues a lethal murine model of citrullinemia type 1, Gene Therapy (2013) 20, 1188-1191.
McIvor, Therapeutic Delivery of mRNA: The Medium Is the Message, Molecular Therapy, 2011, pp. 822-823.
Fath et al, Multiparameter RNA and Codon Optimization: A Standardized Tool to Assess and Enhance Autologous Mammalian Gene Expression, PLoS One, 2011, vol. 6(3), pp. 1-14.
Sahin et al, mRNA-based therapeutics—developing a new class of drugs, Nature reviews, 2014, pp. 759-780.
Balachandran S. et al., Essential role for the dsRNA-dependent protein kinase PKR in innate immunity to viral infection, Immunity 13, 129-141 (2000).
Sercombe et al, Advances and Challenges of Liposome Assisted Drug Delivery, Front. Pharmacol. 6:286, 2015, pp. 1-13.
Saffari et al, Barriers to Liposomal Gene Delivery: from Application Site to the Target, Iranian Journal of Pharmaceutical Research (2016), 15 (Special issue): 3-17.
Hackman, DG, Translating animal research into clinical benefit, BMJ, 2007, pp. 163-168.
Martinez, M.N., Factors Influencing the Use and Interpretation of Animal Models in the Development of Parenteral Drug Delivery Systems, The AAPS Journal, vol. 13, No. 4, Dec. 2011, p. 632-649.
Kaczmarek et al., Advances in the delivery of RNA therapeutics: from concept to clinical reality, Genome Medicine (2017) 9:60, pp. 1-16.
Carlsson et al, Biocompatible, Purified VEGF-A mRNA Improves Cardiac Function after Intracardiac Injection 1 Week Post-myocardial Infarction in Swine, Molecular Therapy: Methods & Clinical Development vol. 9 Jun. 2018, pp. 330-346.
U.S. Appl. No. 60/083,294.
Alton et al., Cationic Lipid-Mediated CFTR Gene Transfer to the Lungs and Nose of Patients with Cystic Fibrosis: a Double-Blind Placebo-Controlled Trial, Lancet, 353: 947-954 (1999).
Anderson, D.M. et al., Stability of mRNA/Cationic Lipid Lipoplexes in Human and Rat Cerebrospinal Fluid: Methods and Evidence for Nonviral mRNA Gene Delivery to the Central Nervous System, Human Gene Therapy, 14:191-202 (2003).
Andries et al., Comparison of the Gene Transfer Efficiency of mRNA/GL67 and pDNA/GL67 Complexes in Respiratory Cells, Mol. Pharmaceut., 9: 2136-2145 (2012).
Auffray, C. et al., Purification of Mouse Immunoglubulin Heavy-Chain Messenger RNAs from Total Myeloma Tumor RNA, European Journal of Biochemistry, 107(2):303-314 (1980).
Author Not Known, Oligotex Handbook, Qiagen (2002).
Author Not Known, PolyATtract mRNA Iosaltion Systems, Promega (2012).
Barreau, C. et al., Liposome-mediated RNA transfection should be used with caution, RNA, 12:1790-1793 (2006).
Behr et al.. Efficient Gene Transfer into Mammalian Primary Endocrine Cells with Lipo Polyamine-Coated DNA, Proc. Nat.'l Acad. Sci., 86: 6982-6986 (1989).
Bloomfield, V.A., Quasi-Elastic Light Scattering Applications in Biochemistry and Biology, Ann. Rev. Biophys. Bioeng. 10:421-450 (1981).
Caplen, N.J. et al., In vitro liposome-mediated DNA transfection of epithelial cell lines using the cationic liposome DC-Chol/DOPE, Gene Therapy, 2:603-613 (1995).
Cassiman, D. Gene transfer for inborn errors of metabolism of the liver: the clinical perspective, Current Pharmaceutical Design, 17(24):2550-2557 (2011).
Chiou, H.C. et al., Enhanced resistance to nuclease degradation of nucleic acids complexed to; asialoglycoprotein-polylysine carriers, Nucleic Acids Research, 22(24):5439-46 (1994).
Conese et al., Gene and Cell Therapy for Cystic Fibrosis: From Bench to Bedside, J. Cyst. Fibros., 10 Suppl 2: S114-s128 (2011).
Cowling, V.H., Regulation of mRNA cap methylation, Biochemical Journal, 425:295-302 (2010).
Dande, P. et al., Improving RNA interference in mammalian cells by 4′-thio-modified small interfering RNA (siRNA): effect on siRNA activity and muclease stability when used in combination with 2′-0-alkyl modifications, Journal of Medicinal Chemistry, 49(5): 1624-1634 (2006).
Debus et al., Delivery of Messenger RNA Using Poly(ethylene imine)-poly(ethylene glycol)-Copolymer Blends for Polyplex Formation: Biophysical Characterization and In Vitro Transfection Properties, J. Control. Rel., 148: 334-343 (2010).
Demeshkina, N. et al., Interactions of the ribosome with mRNA and tRNA, Current Opinion in Structural Biology, 20(3):325-332 (2010).
Dong, Y. et al., Lipopeptide nanoparticles for potent and selective siRNA delivery in rodents and nonhuman primates, Proceedings of the National Academy of Sciences, 111(11): 3955-3960 (2014).
Drummond, D.C. et al., Optimizing Liposomes for Delivery of Chemotherapeutic Agents to Solid Tumors, Pharmacological Reviews, 51(4): 691-743 (1999).
Elton, C., The Next Next Big Thing, Boston Magazine, pp. 106-118 (Mar. 2013).
Emlen, W. et al., Effect of DNA size and strandedness on the in vivo clearance and organ localization of DNA, Clinical & Experimental Immunology, 56:185-192 (1984).
Eon-Duval, A. et al., Removal of RNA impurities by tangential flow filtration in an RNase-free plasmid DNA purification process, Analytical Biochemistry, 316(1):66-73 (2003).
Ernst et al., Interaction of Liposomal and Polycationic Transfection Complexes with Pulmonary Surfactant, J. Gene. Med., 1: 331-340 (1999).
Estimated No. of Animal and Plant Species on Earth, http://www.factmonster.com/ipka/A0934288.html, 2000-2014, 3 pages, (Retreived Aug. 2, 2014).
Fechter, P. et al., Recognition of mRNA cap structures by viral and cellular proteins, Journal of General Virology, 86:1239-1249 (2005).
Felgner et al., Lipofection: A Highly Efficient, Lipid-Mediated DNA-Transfection Procedure, Proc. Natl. Acad., 84:7413-7417 (1987).
Felgner, P.L. and Ringold, G.M., Cationic liposome-mediated transfection, Nature, 337(6205):387-388 (1989).
Fernandez, V. et al., Cross Flow Filtration of RNA Extracts by Hollow Fiber Membrane, Acta Biotechnologica, 12(1):49-56 (1992).
Fischer, D. et al., Effect of poly(ethylene imine) molecular weight and pegylation on organ distribution and pharmacokinetics; of polyplexes with oligodeoxynucleotides in mice, Drug Metabolism and Disposition, 32(9):983-92 (2004).
Fumoto et al., Targeted Gene Delivery: Importance of Administration Routes, Novel Gene Therapy Approaches, Mar. 31, 2013.
Galipon, J. et al., Stress-induced 1ncRNAs evade nuclear degradation and enter the translational machinery, Genes to Cells, 18(5):353-368 (2013).
Gao, X. et al., A novel cationic liposome reagent for efficient transfection of mammalian cells, Biochemical and Biophysical Research Communications, 179(1):280-285 (1991).
Garbuzenko, O.B. et al., Intratracheal Versus Intravenous Liposomal Delivery of siRNA, Antisense Oligonucleotides and Anticancer Drug, Pharmaceutical Research, 26(2):382-394 (2009).
Geraerts, M. et al., Upscaling of lentiviral vector production by tangential flow filtration, Journal of Gene Medicine, 7(10):1299-1310 (2005).
Gonzalez-Aseguinolaza, G. et al., Gene therapy of liver diseases: A 2011 perspective, Clinics and Research in Hepatology and Gastroenterology, 35(11):699-708 (2011).
Gordon, N. Ornithine transcarbamylase deficiency: a urea cycle defect, European Journal of Paediatric Neurology, 7:115-121 (2003).
Grudzien, E. et al., Novel cap analogs for in vitro synthesis of mRNAs with high translational efficiency, RNA Biology, 10(9):1479-1487 (2004).
Guttman, M. et al., Chromatin signature reveals over a thousand highly conserved large non-coding RNAs in mammals, Nature, 458:223-227 (2009).
Harada-Shiba, M. et al., Polyion complex micelles as vectors in gene therapy—pharmacokinetics and in vivo; gene transfer, Gene Therapy, 9(6):407-14 (2002).
Hata, A. et al., Isolation and Characterization of the Human Ornithine Transcarbamylase Gene: Structure of the 5′-End Region, Journal of Biochemistry, 100:717-725 (1986).
Hecker, J. et al., Advances in Self-Limited Gene Expression of Protective Intracellular Proteins In-Vivo in Rat Brain Using mRNA / Cationic Lipid Complexes, Anesthesia and Analgesia, 86(2S):346S (1994).
Heidenreich, O. et al., High Activity and Stability of Hammerhead Ribozymes Containing 2′-Modified Pyrimidine Nucleosides and Phosphorothioates, The Journal of Biological Chemistry, 269(3):2131-2138 (1994).
Hess et al., Vaccination with mRNAs Encoding Tumor-Associated Antigens and Granulocyte-Macrophage Colony-Stimulating Factor Efficiently Primes CTL Responses, but is Insufficient to Overcome Tolerance to a Model Tumor/Self Antigen, Cancer Immunology, Immunotherapy: CII, 55(6): 672-83 (2006).
Heyes et al., Cationic Lipid Saturation Influences Intracellular Delivery of Encapsulated Nucleic Acids, J. Controlled Release, 107: 276-287 (2005).
Higman, M.A. et al., The mRNA (Guanine-7-)methyltransferase Domain of the Vaccinia Virus mRNA Capping Enzyme, The Journal of Biological Chemistry, 269(21):14974-14981 (1994).
Hillery, A.M. et al., Drug Delivery and Targeting for Pharmacists and Pharmaceutical Scientists, Taylor and Francis (2005).
Hoerr et al., In Vivo Application of RNA Leads to Induction of Specific Cytotoxic T Lymphocytes and Antibodies, European Journal of Immunology, 30(1): 1-7 (2000).
Hope, M.J. et al., Reduction of Liposome Size and Preparation of Unilamellar Vesicles by Extrusion Techniques, In: Liposome Technology, 1:123-139 (1993).
Horwich, A.L. et al., Structure and Expression of a Complementary DNA for the Nuclear Coded Precursor of Human Mitochondrial Ornithine Transcarbamylase, Science, 224(4653):1068-1074 (1984).
Horwich, A.L. et al., Targeting of Pre-Ornithine Transcarbamylase to Mitochondria: Definition of Critical Regions and Residues in the Leader Peptide, Cell, 44:451-459 (1986).
Huang, Z. et al., Thiocholesterol-based lipids for ordered assembly of bioresponsive gene carriers, Molecular Therapy, 11(3):409-417 (2005).
Huttenhofer, A. and Noller, H., Footprinting mRNA-ribosome complexes with chemical probes, The EMBO Journal, 13(16):3892-3901 (1994).
Jakobsen, K. et al., Purification of MRNA Directly From Crude Plant Tissues in 15 Minutes Using Magnetic Oligo DT Microsheres, Nucleic Acids Research, 18(12):3669 (1990).
Jeffs, L.B. et al., A scalable, extrusion-free method for efficient liposomal encapsulation of plasmid DNA, Pharmaceutical Research, 22(3):362-372 (2005).
Jones, G. et al., Duplex- and Triplex-Forming Properties of 4′-Thio-Modified Oligodeoxynucleotides, Bioorganic & Medicinal Chemistry Letters, 7(10):1275-1278 (1997).
Kariko, K. et al., In vivo protein expression from mRNA delivered into adult rat brain, Journal of Neuroscience Methods, 105(1):77-86 (2001).
Kariko, K. et al., Incorporation of Pseudouridine Into mRNA Yields Superior Nonimmunogenic Vector With Increased Translational Capacity and Biological Stability, Molecular Therapy, 16(11): 1833-1840 (2008).
Kasuya, T. et al., In Vivo Delivery of Bionanocapsules Displaying Phaseolus vulgaris Agglutinin-L4 Isolectin to Malignant Tumors Overexpressing N-Acetylglucosaminyltransferase V, Human Gene Therapy, 19:887-895 (2008).
Kiew, L.V. et al., Effect of antisense oligodeoxynucleotides for ICAM-1 on renal ischaemia-reperfusion injury in the anaesthetised rat, The Journal of Physiology, 557(3):981-989 (2004).
Klibanov, A.L. et al., Amphipathic polyethyleneglycols effectively prolong the circulation time of liposomes, FEBS, 268(1): 235-237 (1990).
Kober et al., Optimized Signal Peptides for the Development of High Expressing CHO Cell Lines, Biotechnol. Bioeng., 110: 1164-1173 (2012).
Kormann, M.S.D. et al., Expression of therapeutic proteins after delivery of chemically modified mRNA in mice, Nature Biotechnology, 29(2):154-157 (2011).
Kozak, M. An analysis of 5′-noncoding sequences from 699 vertebrate messenger RNAs, Nucleic Acid Research, 15(20):8125-8148 (1987).
Krieg, P.A. et al., In vitro RNA synthesis with SP6 RNA polymerase, Methods in Enzymology, 155:397-415(1987).
Kvasnica, M. et al., Platinum(II) complexes with steroidal esters of L-methionine and L-histidine: Synthesis, characterization and cytotoxic activity, Bioorganic & Medicinal Chemistry, 16:3704-3713 (2008).
Lam, J.K.W et al., Pulmonary delivery of therapeutic siRNA, Advanced Drug Delivery Reviews (2011).
Lasic, D.D. et al., Gelation of liposome interior: A novel method for drug encapsulation, FEBS, 312(2,3):255-258 (1992).
Lasic, D.D. Novel applications of liposomes, Trends in Biotechnology, 16:307-321 (1998).
Li, L. et al., Preparation and Gene Delivery of Alkaline Amino Acids-Based Cationic Liposomes, Archives of Pharmaceutical Research, 31(7):924-931 (2008).
Li, S. et al., In vivo gene transfer via intravenous administration of cationic lipid-protamine-DNA (LPD) complexes, Gene Therapy, 4:891-900 (1997).
Li, W. et al., Lipid-based Nanoparticles for Nucleic Acid Delivery, Pharmaceutical Research, 24(3):438-449 (2007).
Liebhaber, S.A. et al., Translation inhibition by an mRNA coding region secondary structure is determined by its proximity to the AUG initiation codon, Journal of Molecular Biology, 226(3):609-621 (1992).
Lindgren, V. et al., Human Ornithine Transcarbamylase Locus Mapped to Band Xp21.1 Near the Duchenne Muscular Dystrophy Locus, Science, 226(2675):698-700 (1984).
Liu et al., COStar: a D-star Lite-based Dynamic Search Algorithm for Codon Optimization, Journal of Theoretical Biology, 344: 19-30 (2014).
Liu, Y. et al., Designer Lipids Advance Systemic siRNA Delivery, Molecular Therapy, 18(4):669-670 (2010).
Lo, K-M et al., High level expression and secretion of Fc-X fusion proteins in mammalian cells, Protein Engineering, 11(6)495-500 (1998).
Love, K.T. et al., Lipid-like materials for low-dose, in vivo gene silencing, PNAS, 107(5):1864-1869 (2010).
Lu, D. et al., Optimization of methods to achieve mRNA-mediated transfection of tumor cells in vitro and in vivo employing cationic liposome vectors, Cancer Gene Therapy, 1(4):245-52 (1994).
Maclachlan, I. et al., Lipid nanoparticle-mediated delivery of messenger RNA, 1st International mRNA health conference, Tubingen, Germarny (2013).
Maeda-Mamiya, R. et al.,. In vivo gene delivery by cationic tetraamino; fullerene. Proceedings of National Academy of Sciences U S A, 107(12):5339-44 (2010).
Malone, R.W., et al., Cationic liposome-mediated RNA transfection, PNAS, 86:6077-6081 (1989).
Mammal, http://en.wikipedia.org/wiki/Mammal, 2007, Pearson Education, NY, NY, Author unkown (Source: The international union for conservation of nature and natural resources), 2 pages, (Retreived Aug. 2, 2014).
Mansour, H.M. et al., Nanomedicine in pulmonary delivery, International Journal of Nanomedicine, 4:299-319 (2009).
Martinon et al., Induction of Virus-Specific Cytotoxic T Lymphocytes in Vivo by Liposome-Entrapped mRNA, European Journal of Immunology, 23(7): 1719-22 (1993).
Mccracken, S. et al., 5′-Capping Enzymes are Targeted to Pre-MRNA by Binding to the Phosphorylated Carboxy-Terminal Domain of RNA Polymerase II, Genes and Development, 22(24):3306-3318 (1997).
Melton, D.A. et al., Efficient in vitro synthesis of biologically active RNA and RNA hybridization probes from; plasmids containing a bacteriophage SP6 promoter, Nucleic Acids Research, 12(18):7035-56 (1984).
Merkel, O.M. et al., Nonviral Pulmonary Delivery of siRNA, Accounts of Chemical Research, 10 pages (2011).
Merten, O. et al., Large-Scale Manufacture and Characterizationof a Lentiviral Vector Produced for Clinical Ex Vivo Gene Therapy Application, Human Gene Therapy, 22(3):343-356 (2011).
Monia, B.P. et al., Evaluation of 2′-Modified Oligonucleotides Containing 2′-Deoxy Gaps as Antisense Inhibitors of Gene Epression, The Journal of Biological Chemistry, 268(19):14514-14522 (1993).
Morrissey et al., Potent and Persistent in vivo Anti-HBV Activity of Chemically Modified siRNAs, Nat. Biotechnol., 23(8): 1003-1007 (2005).
Ng, J. et al., LincRNAs join the pluripotency alliance, Nature Genetics, 42:1035-1036 (2010).
Nojima, T. et al., The Interaction between Cap-binding Complex and RNA Export Factor is Required for Intronless mRNA Export, Journal of Biological Chemistry, 282(21):15645-15651 (2007).
Okumura, K. et al., Bax mRNA therapy using cationic liposomes for human malignant melanoma, The Journal of Gene Medicine, 10:910-917 (2008).
Otsuka, Y. et al., Identification of a Cytoplasmic Complex That Adds a Cap onto 5′-Monophosphate RNA, Molecular and Cellular Biology, 29(8):2155-2167 (2009).
Painter et al, Topical Delivery of mRNA to the Murine Lung and Nasal Epithelium, Gene Medicine Group and the Medical Informatics Unit, Nuffield Department of Clinical Laboratory Sciences, University of Oxford, 1 page.
Painter et al., Topical Delivery of mRNA to the Murine Lung and Nasal Epithelium, Molecular Therapy, 9: S187 (2004).
Painter, An Investigation of mRNA as a Gene Transfer Agent, Oxford University GeneMedicine, Abstract Only, 1 page (2007).
Probst, J. et al., Spontaneous cellular uptake of exogenous messenger RNA in vivo is nucleic acid-specific, saturable and ion dependent, Gene Therapy, 14:1175-1180 (2007).
Rabinovich, P.M. et al., Synthetic Messenger RNA as a Tool for Gene Therapy, Human Gene Therapy, 17:1027-1035 (2006).
Raper, S.E. et al., Developing adenoviral-mediated in vivo gene therapy for ornithine transcarbamylase deficiency, Journal of Inherited Metabolic Disease, 21:119-137 (1998).
Ratajczak, J. et al., Membrane-derived microvesicles: important and underappreciated mediators of cell-to-cell communication, Leukemia, 20:1487-1495 (2006).
Reddy, A. et al., The Effect of Labour and Placental Separation on the Shedding of Syncytiotrophoblast Microparticles, Cell-free DNA and mRNA in Normal Pregnancy and Preeclampsia, Placenta, 29:942-949.
Rejman, J. et al., Characterization and transfection properties of lipoplexes stabilized with novel exchangeable polyethylene glycol-lipid conjugates, Biochimica et Biophysica Acta, 1660:41-52 (2004).
Rosenecker et al., Gene Therapy for Cystic Fibrosis Lung Disease: Current Status and Future Perspectives, Curr. Opin. Mol. Ther., 8: 439-445 (2006).
Rosenecker et al., Interaction of Bronchoalveolar Lavage Fluid with Polyplexes and Lipoplexes: Analysing the Role of Proteins and Glycoproteins, J. Gene. Med., 5: 49-60 (2003).
Rowe et al., Cystic Fibrosis, New Engl. J. Med. 352: 1992-2001 (2005).
Schnierle, B.S. et al., Cap-specific mRNA (nucleoside-O2′-)-methyltransferase and poly(A) polymerase stimulatory activities of vaccinia virus are mediated by a single protein, Proceedings of the National Academy of Sciences, 89:2897-2901 (1992).
Schreier, Han, The new frontier: gene and oligonucleotide therapy, Pharmaceutica Acta Helvetiae, 68(3):145-159 (1994).
Semple, S.C. et al., Rational design of cationic lipids for siRNA delivery, Nature Biotechnology, 28(2): 172-176 (2010).
Shimada, A. et al., Translocation Pathway of the Intratracheally Instilled Ultrafine Particles from the Lung into the Blood Circulation in the Mouse, Toxicologic Pathology, 34:949-957 (2006).
Smisterova, J. et al., Molecular Shape of the Cationic Lipid Controls the Structure of Cationic Lipid/Dioleylphosphatidylethanolamine-DNA Complexes and the Efficiency of Gene Delivery, The Journal of Biological Chemistry, 276(50):47615-47622 (2001).
Su, X. et al., In Vitro and in Vivo mRNA Delivery Using Lipid-Enveloped pH-Responsive Polymer Nanopartides, Molecular Pharmaceutics, 8(3):774-787 (2011).
Tagawa, M. et al., Gene expression and active virus replication in the liver after injection of duck hepatitis B virus DNA into the peripheral vein of ducklings, Journal of Hepatology, 24:328-334 (1996).
Tang, F. and Hughes, J. et al., Introduction of a Disulfide Bond into a Cationic Lipid Enhances Transgene Expression of Plasmid DNA, Biochemical and Biophysical Research Communications, 242(1):141-145 (1998).
Tavernier, G. et al., mRNA as gene therapeutic: How to control protein expression, Journal of Controlled Release, 150:238-247 (2011).
Tsui, N.B. et al., Stability of endogenous and added RNA in blood specimens, serum, and plasma, Clinical Chemistry, 48(10): 1647-53 (2002).
Tuschl, T. et al., Targeted mRNA degradation by double-stranded RNA in vitro, Genes and Development, 13(24):3191-7 (1999).
Van Der Gun, B.T.F et al., Serum insensitive, intranuclear protein delivery by the multipurpose cationic lipid Saint-2, Journal of Controlled Release, 123:228-238 (2007).
Van Tendeloo, V.F.I et al., mRNA-based gene transfer as a tool for gene and cell therapy, Current Opinion in Molecular Therapeutics, 9(5):423-431 (2007).
Varambally, S. et al., Genomic Loss of microRNA-101 Leads to Overexpression of Histone Methyltransferase EZH2 in Cancer, Science, 322:1695-1699 (2008).
Viecelli, H. et al., Gene Therapy for Hepatic Diseases Using Non-Viral Minicircle-DNA Vector, Journal of Inherited Metabolic Disease, 35(1):S144 (2012).
Viecelli, H. et al., Gene therapy for liver diseases using non-viral minicirde-DNA vector, Human Gene Therapy, 23(10):A145 (2012).
Viecelli, H. et al., Gene therapy for liver diseases using non-viral minicircle-DNA vector, Molecular Therapy, 21 (1):S136 (2013).
Vomelova, I. et al., Methods of RNA Purification. All Ways (Should) Lead to Rome, Folia Biologica, 55(6):242-251 (2009).
Wang, H. et al., N-acetylgalactosamine functionalized mixed micellar nanoparticles for targeted delivery of siRNA to liver, Journal of Controlled Release, 166(2):106-114 (2012).
Wang, Y. et al., Systemic delivery of modified mRNA encoding herpes simplex virus 1 thymidine kinase for targeted camcer gene therapy, Molecular Therapy, 21(2):358-367 (2013).
Webb, M. et al., Sphinogomyeline-cholesterol liposomes significantly enhance the pharmacokinetic and therapeutic properties of vincristine in murine and human tumour models, British Journal of Cancer, 72(4):896-904 (1995).
Wetzer, B. et al., Reducible cationic lipids for gene transfer, Journal of Biochemistry, 356:745-756 (2001).
Wiehe, J.M. et al., mRNA-mediated gene delivery into human progenitor cells promotes highly efficient protein expression, Journal of Cellular and Molecular Medicine, 11(3):521-530 (2007).
Williams, D. et al., A Simple, Highly Efficient Method for Heterologous Expression in Mammalian Primary Neurons Using Cationic Lipid-mediated mRNA Transfection, Frontiers in Neuroscience, 4:181 (2010).
Wolf, J.A. et al., Protein/Amphipathic Polyamine Complexes Enable Highly Efficient Transfection with Minimal Toxicity, BioTechniques, 23:139-147 (1997).
Wu, J. and Zern, M., Modification of liposomes for liver targeting, Journal of Hepatology, 24(6):757-763 (1996).
Yamamoto et al., Important Role of the Proline Residue in the Signal Sequence that Directs the Secretion of Human Lysozyme in Saccharomyces cerevisiae, Biochemistry, 28:2728-2732 (1989).
Yamamoto, A. et al., Current prospects for mRNA gene delivery, European Journal of Pharmaceutics and Biopharmaceutics, 71:484-489 (2009).
Yasuda et al., Fabry Disease: Novel [alpha]-Galactosidase A 3-terminal Mutations Result in Multiple Transcripts Due to Aberrant 3-End Formation, American Journal of Human Genetics, 73: 162-73 (2003).
Ye, X. et al., Nucleic Acids, Protein Synthesis, and Molecular Genetics: Prolonged Metabolic Correction in Adult Ornithine Transcarbamylase-deficient Mice with Adenoviral Vectors, The Journal of Biological Chemistry, 271:3639-3646 (1996).
Yokoe, H. et al., Spatial dynamics of GFP-tagged proteins investigated by local fluorescence enhancement, Nature Biotechnology, 14(10):1252-1256 (1996).
Zou, S. et al., Lipid-mediated delivery of RNA is more efficient than delivery of DNA in non-dividing cells, International Journal of Pharmaceutics, 389(1-2):232-243 (2010).
U.S. Appl. No. 61/494,714, filed Jun. 8, 2011.
Adami, R.C. et al., An amino acid-based amphoteric liposomal delivery system for systemic administration of siRNA. Molecular Therapy 19(6):1141-1151 (2011).
Akinc, A. et al., A combinatorial library of lipid-like materials for delivery of RNAi therapeutics. Nature Biotechnology 26(5):561-569 (2008).
Akinc, A. et al., Development of lipidoid-siRNA formulations for systemic delivery to the liver. Molecular Therapy 17(5):872-879 (2009).
Anderson, D.G. et al., Structure/property studies of polymeric gene delivery using a library of poly(beta-amino esters). Molecular Therapy 11(3):426-434 (2005).
Anderson, J. Biological Responses to Materials. Annual Review of Materials Research 31:81-110 (2001).
Anderson, W. French, Human gene therapy, Nature, 392, 25-30 (1998).
Author Unknown, Blood Proteins, published by WikiPedia, San Francisco, CA, 2 pages, <http://en.wikipedia.org/wiki/Biood_proteins> downloaded May 17, 2015.
Bahlke, M. A. et al., Progress towards in vivo use of siRNAs, Molecular Therapy, 13:644-670 (2006).
Bajaj, A. et al., Synthesis and gene transfection efficacies of PEI-cholesterol-based lipopolymers. Bioconjugate Chemistry 19(8): 1640-516511 (2008).
Bennett, J. Immune response following intraocular delivery of recombinant viral vectors, Gene Therapy, 10: 977-982 (2003).
Bhaduri, S. et al., Procedure for the preparation of milligram quantities of adenovirus messenger ribonucleic acid, J. Virol., 10(6): 1126-1129(1972).
Boussif, O. et al., A versatile vector for gene and oligonucleotide transfer into cells in culture and in vivo: polyethylenimine. Proceedings of the National Academy of Sciences of the USA. 92(16):7297-7301 (1995).
Braun, C.S. et al., Structure/function relationships of polyamidoamine/DNA dendrimers as gene delivery vehicles. Journal of Pharmaceutical Sciences 94(2):423-436 (2005).
Breunig, M. et al., Breaking up the correlation between efficacy and toxicity for nonviral gene delivery. Proceedings of the National Academy of Sciences of the U S A. 104(36):14454-14459 (2007).
Breunig, M. et al., Mechanistic investigation of poly(ethylene imine)-based siRNA delivery: disulfide bonds boost intracellular release of the cargo. Journal of Controlled Release 130(1):57-63 (2008).
Brey, D.M. et al., Controlling poly(beta-amino ester) network properties through macromer branching. Acta Biomaterialia 4(2):207-217 (2008).
Brey, D.M. et al., Influence of macromer molecular weight and chemistry on poly(beta-amino ester) network properties and initial cell interactions. Journal of Biomedical Materials Research Part A 85(3):731-741 (2007).
Burger, G. et al., Sequencing complete mitochondrial and plastid genomes, Nature Protocols, 2: 603-614 (2007).
Burnett, J.C. et al., Current progress of siRNA/shRNA therapeutics in clinical trials. Biotechnology Journal 6(9):1130-1146 (2011).
Byk, G. et al., Synthesis, activity, and structure—activity relationship studies of novel cationic lipids for DNA transfer. Journal of Medical Chemistry 41(2):224-235 (1998).
Castanotto, D. et al., The promises and pitfalls of RNA-interference-based therapeutics. Nature 457(7228):426-433 (2009).
Chakraborty, C. Potentiality of Small Interfering RNAs (siRNA) as Recent Therapeutic Targets for Gene-Silencing. Current Drug Targets 8(3):469-82 (2007).
Chau, Y. et al., Investigation of targeting mechanism of new dextran-peptide-methotrexate conjugates using biodistribution study in matrix-metalloproteinase-overexpressing tumor xenograft model, J. Pharm. Sci., 95(3): 542-551 (2006).
Chen, D. et al., Rapid discovery of potent siRNA-containing lipid nanoparticles enabled by controlled microfluidic formulation. Journal of the American Chemical Society 134(16):6948-6951 (2012).
Chen, Y. and Huang, L., Tumor-targeted delivery of siRNA by non-viral vector: safe and effective cancer therapy. Expert Opinion on Drug Delivery 5(12):1301-1311 (2008).
Christensen, U.B. et al., Intercalating nucleic acids containing insertions of 1-O-(1-pyrenylmethyl)glycerol: stabilisation of dsDNA and discrimination of DNA over RNA, Nucl. Acids. Res., 30(22): 4918-4925 (2002).
Cotten, M. et al., Receptor-mediated transport of DNA into eukaryotic cells. Methods in Enzymology 217 (H):618-644 (1993).
Creusat, G. et al., Proton sponge trick for pH-sensitive disassembly of polyethylenimine-based siRNA delivery systems. Bioconjugate Chemistry 21(5):994-1002 (2010).
Crooke, S.T. Molecular mechanisms of action of antisense drugs. Biochimica et Biophysica Acta 1489(1):31-44. Review (1999).
Crystal, R.G. Transfer of genes to humans: early lessons and obstacles to success. Science 270(5235)404-410. Review (1995).
Damen, M. et al., Delivery of DNA and siRNA by novel gemini-like amphiphilic peptides. Journal of Controlled Release 145(1):33-39 (2010).
Davis, M. E., The first targeted delivery of siRNA in humans via a self-assembling, cyclodextrin polymer-based nanoparticle: from concept to clinic. Molecular Pharmacuetics 6(3):659-668 (2009).
Davis, M.E. et al., Evidence of RNAi in humans from systemically administered siRNA via targeted nanoparticles. Nature 464(7291):1067-1070 (2010).
Decher, G. Fuzzy Nanoassemblies: Toward Layered Polymeric Multicomposites. Science 277: 1232-1237 (1997).
Denardo, S.J. et al., Enhanced Therapeutic Index of Radioimmunotherapy (RIT) in Prostate Cancer Patients Comparison of Radiation Dosimetry for 1,4,7,10-Tetraazacyclododecane-N,N′,N″,N″-Tetraacetic Acid (DOTA)-Peptide versus 2IT-DOTA Monoclonal Antibody Linkage for RIT1, Clin. Cancer Res., 9: 3665s (2003).
Dern, R.J. et al., Toxicity studies of pyrimethamine (daraprim). The American Journal of Tropical Medicine and Hygiene 4(2):217-220 (1955).
Deshmukh, H. M and Huang, L., Liposome and polylysine mediated gene therapy. New Journal of Chemistry 21:113-124 (1997).
Discher, B.M. et al., Polymersomes: tough vesicles made from diblock copolymers. Science 284(5417):1143-1146(1999).
Discher, D.E. and Eisenberg, A., Polymer vesicles. Science 297(5583):967-973. Review (2002).
Driscoll, K.E. et al., Intratracheal instillation as an exposure technique for the evaluation of respiratory tract toxicity: uses and limitations, Toxicol. Sci., 55(1): 24-35 (2000).
Dwarki, V. et al.. Cationic liposome-mediated RNA transfection, Methods in Enzymology, 217:644-654 (1993).
Elbashir, S.M. et al., RNA interference is mediated by 21- and 22-nucleotide RNAs. Genes & Development 15: 188-200 (2001).
Ewert, K. et al., Cationic lipid-DNA complexes for gene therapy: understanding the relationship between complex structure and gene delivery pathways at the molecular level. Current Medicinal Chemistry 11(2): 133-149 (2004).
Fenske, D.B. and Cullis, P., Liposomal nanomedicines. Expert Opinion on Drug Delivery 5(1):25-44 (2008).
Ferruti, P.F. and Barbucci, R. , Linear amino polymers: Synthesis, protonation and complex formation. Advances in Polymer Science 58:55-92 (1984).
Ferruti, P.F. et al., A novel modification of poly(l-lysine) leading to a soluble cationic polymer with reduced toxicity and with potential as a transfection agent. Macromolecular Chemistry and Physics 199:2565-2575 (1998).
Fire, A. et al., Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans. Nature 391(6669):806-811 (1998).
Furgeson, D.Y. et al., Modified linear polyethylenimine-cholesterol conjugates for DNA complexation. Bioconjugate Chemistry 14(4):840-847 (2003).
Furgeson, D.Y. et al., Novel water insoluble lipoparticulates for gene delivery. Pharmaceutical Research 19(4): 382-390 (2002).
Godbey, W.T. et al., Size matters: molecular weight affects the efficiency of poly(ethylenimine) as a gene delivery vehicle. Journal of Biomedical Materials Research 45(3):268-275 (1998).
Gonzalez, H. et al., New class of polymers for the delivery of macromolecular therapeutics. Bioconjugate Chemistry 10(6): 1068-1074 (1999).
Grayson, A.C.R. et al., Biophysical and structural characterization of polyethylenimine-mediated siRNA delivery in vitro. Pharmaceutical Research 23(8): 1868-1876 (2006).
Grunlan, M.A. et al., Synthesis of 1,9-bis[glycidyloxypropyl]penta(1′H, 1′H, 2′H, 2′H-perfluoroalkylmethylsiloxane)s and copolymerization with piperazine. Polymer 45:2517-2523 (2004).
Gupta, U. et al., A review of in vitro-in vivo investigations on dendrimers: the novel nanoscopic drug carriers. Nanomedicine: Nanotechnology, Biology, and Medicine 2(2):66-73 (2006).
Haensler, J. and Szoka, F., Polyamidoamine cascade polymers mediate efficient transfection of cells in culture. Bioconjugate Chemistry 4(5):372-379 (1993).
Haskins M., Gene Therapy for Lysosomal Storage Disorders (LDSs) in Large Animal Models, ILAR J., 50(2):112-121 (2009).
Henkin, R. I. et al., Inhaled Insulin—Intrapulmonary, intranasal, and other routes of administration: Mechanisms of action, Nutrition, 26: 33-39 (2010).
Hill, I.R.C. et al., In vitro cytotoxicity of poly(amidoamine)s: relevance to DNA delivery. Biochimica et Biophysica Acta 1427: 161-174 (1999).
Hill, J.G. et al., Enantioselective Epoxidation of Allylic Alcohols: (2S,3S)-3-Propyloxiranemethanol. Organic Syntheses Collection 7: 461 (1990) and 63: 66 (1985) (8 pages).
Hofland, H.E.J et al., Formation of stable cationic lipid/DNA complexes for gene transfer. Proceedings of the National Academy of Sciences of the USA 93 (14): 7305-7309 (1996).
Homo sapiens galactosidase, alpha (GLA) mRNA, NCBI Reference Sequence NM_000169.1, Modification Date: Nov. 17, 2006.
Hope, M.J. et al., Cationic Lipids, Phosphatidylethanolamine and the Intracellular Delivery of Polymeric, Nucleic Acid-Based Drugs. Molecular Membrane Technology 15:1-14 (1998).
Hornung, V. et al., Quantitative expression of toll-like receptor 1-10 mRNA in cellular subsets of human peripheral blood mononuclear cells and sensitivity to CpG oligodeoxynucleotides. The Journal of Immunology 168: 4531-4537 (2002).
Howard, K.A. Delivery of RNA interference therapeutics using polycation-based nanoparticles. Advanced Drug Delivery Reviews 61: 710-720 (2009).
Incani, V. et al., Lipid and hydrophobic modification of cationic carriers on route to superior gene vectors. Soft Matter 6: 2124-2138 (2010).
International Preliminary Report on Patentability for PCT/US2010/058457, 12 pages (dated Jun. 14, 2012).
Jiang, G. et al., Hyaluronic acid-polyethyleneimine conjugate for target specific intracellular delivery of siRNA. Biopolymers 89 (7): 635-642 (2008).
Jiang, M. et al., Electrochemically controlled release of lipid/DNA complexes: a new tool for synthetic gene delivery system. Electrochemistry Communications (6): 576-582 (2004).
Jiang, S. and Cao, Z., Ultralow-fouling, functionalizable, and hydrolyzable zwitterionic materials and their derivatives for biological applications. Advanced Materials 22(9):920-932 (2010).
Jolck, R.I. et al., Solid-phase synthesis of PEGylated lipopeptides using click chemistry. Bioconjugate Chemistry 21 (5):807-810 (2010).
Jon, S. et al., Degradable poly(amino alcohol esters) as potential DNA vectors with low cytotoxicity. Biomacromolecules 4(6): 1759-1762 (2003).
Kabanov, A.V. and Kabanov, V.A., DNA complexes with polycations for the delivery of genetic material into cells. Bioconjugate Chemistry 6(1): 7-20 (1995).
Kamath, S. et al., Surface chemistry influences implant-mediated host tissue responses. Journal of Biomedical Materials Research A 86(3):617-626 (2007).
Kaur, N. et al., A delineation of diketopiperazine self-assembly processes: understanding the molecular events involved in Nepsilon-(fumaroyl)diketopiperazine of L-Lys (FDKP) interactions. Molecular Pharmaceutics 5(2):294-315 (2007).
Kaur, T. et al., Addressing the Challenge: Current and Future Directions in Ovarian Cancer THerapy, Current Gene THerapy, 9: 434-458 (2009).
Kim, S.H. et al., Comparative evaluation of target-specific GFP gene silencing efficiencies for antisense ODN, synthetic siRNA, and siRNA plasmid complexed with PEI-PEG-FOL conjugate. Bioconjugate Chemistry 17(1): 241-244 (2006).
Kim, T. et al., Synthesis of biodegradable cross-linked poly(beta-amino ester) for gene delivery and its modification, inducing enhanced transfection efficiency and stepwise degradation. Bioconjugate Chemistry 16(5): 1140-1148 (2005).
Kodama, K. et al., The Features and Shortcomings for Gene Delivery of Current Non-Viral Carriers, Current Medicinal Chemistry, 13: 2155-2161 (2006).
Lee, S. et al., Stability and cellular uptake of polymerized siRNA (poly-siRNA)/polyethylenimine (PEI) complexes for efficient gene silencing. Journal of Controlled Release 141: 339-346 (2010).
Lim, Y. et al., A self-destroying polycationic polymer: biodegradable poly(4-hydroxy-l-proline ester). Journal of American Chemical Society 121: 5633-5639 (1999).
Liu, Y. et al., Factors influencing the efficiency of cationic liposome-mediated intravenous gene delivery, Nature Biotechnology, 15:167-173 (1997).
Lorenzi, J. C. C. et al., Intranasal Vaccination with Messenger RNA as a New Approach in Gene Therapy: Use Against Tuberculosis, BMC Biotechnology, 10(77):1-11 (2010).
Lukyanov, A.N. and Torchilin, V.P., Micelles from lipid derivatives of water-soluble polymers as delivery systems for poorly soluble drugs. Advanced Drug Delivery Reviews 56: 1273-1289 (2004).
Luo, D. and Saltzman, M., Synthetic DNA delivery systems. Nature Biotechnology 18: 33-37. Review (2000).
Lynn, D.M. and Langer, R., Degradable Poly(ß-amino esters):? Synthesis, Characterization, and Self-Assembly with Plasmid DNA. Journal of American Chemical Society 122(44): 10761-10768 (2000).
Lynn, D.M. et al., Accelerated discovery of synthetic transfection vectors: parallel synthesis and screening of a degradable polymer library. Journal of American Chemical Society 123 (33): 8155-8156 (2001).
Lynn, D.M. et al., pH-Responsive Polymer Microspheres: Rapid Release of Encapsulated Material within the Range of Intracellular pH. Angewandte Chemie International Edition 40(9): 1707-1710 (2001).
Ma, M. et al., Developlment of Cationic Polymer Coatings to Regulate Foreign Body Responses. Advanced Healthcare Materials 23: H189-H194. Reviews (2011).
Margus, H. et al., Cell-penetrating peptides as versatile vehicles for oligonucleotide delivery. Molecular Therapy 20 (3): 525-533 (2012).
Martell, A.E. and Chaberek, S., The Preparation and the Properties of Some N,N′-Disubstituted-ethylenediaminedipropionic Acids. Journal of the American Chemical Society 72: 5357-5361 (1950).
Mathiowitz, E. and Langer, R., Polyanhydride microspheres as drug carriers I. Hot-melt microencapsulation. Journal of Controlled Release 5: 13-22 (1987).
Mathiowitz, E. et al., Novel microcapsules for delivery systems. Reactive Polymers 6: 275-283 (1987).
Mathiowitz, E. et al., Polyanhydride microspheres as drug carriers II. Microencapsulation by solvent removal. Journal of Applied Polymer Sciences 35: 755-774 (1988).
Mendelsohn, J.D. et al., Rational design of cytophilic and cytophobic polyelectrolyte multilayer thin films. Biomacromolecules 4(1): 96-106 (2003).
Miller, A. Cationic Liposomes for Gene Therapy. Angewandte Chemie International Edition 37: 1768-1785 (1998).
Narang, A.S. et al., Cationic lipids with increased DNA binding affinity for nonviral gene transfer in dividing and nondividing cells. Bioconjugate Chemistry 16(1): 156-168 (2005).
Navarro, G. et al., Phospholipid-polyethylenimine conjugate-based micelle-like nanoparticles for siRNA delivery. Drug Delivery and Translational Research 1:25-33 (2011).
Neamnark, A. et al., Aliphatic lipid substitution on 2 kDa polyethylenimine improves plasmid delivery and transgene expression. Molecular Pharmaceutics 6(6): 1798-1815 (2009).
Nguyen, D.N. et al., A novel high-throughput cell-based method for integrated quantification of type I interferons and in vitro screening of immunostimulatory RNA drug delivery. Biotechnology and Bioengineering 103(4): 664-675 (2009).
Nguyen, D.N. et al., Drug delivery-mediated control of RNA immunostimulation. Molecular Therapy 17(9): 1555-1562 (2009).
Nori, A. et al., Tat-conjugated synthetic macromolecules facilitate cytoplasmic drug delivery to human ovarian carcinoma cells, Bioconj. Chem., 14(1): 44-50 (2003).
Ozer, A., Alternative applications for drug delivery: nasal and pulmonary routes, Nanomaterials and Nanosystems for Biomedical Applications, M.R. Mozafari (ed.): 99-112 (2007).
Painter, H., An Investigation of mRNA as a Gene Transfer Agent, Gene Medicine Research Group Nuffield Department of Clinical Laboratory Sciences and Merton College, University of Oxford, 1-282 (2007).
Parrish, D.A. and Mathias, L.J., Five- and six-membered ring opening of pyroglutamic diketopiperazine. Journal of Organic Chemistry 67(6): 1820-1826 (2002).
Patton, J., Market Trends in Pulmonary Therapies, Trends and Opportunities, VI: 372-377.
Paulus, C. and Nevels, M., The Human Cytomegalovirus Major Immediate-Early Proteins as Antagonists of Intrinsic and Innate Antiviral Host Responses, Viruses, 1:760-779 (2009).
Peppas, N.A. et al., Hydrogels in Biology and Medicine: From Molecular Principles to Bionanotechnology. Advanced Materials 18: 1345-1360 (2006).
Philipp, A. et al., Hydrophobically modified oligoethylenimines as highly efficient transfection agents for siRNA delivery. Bioconjugate Chemistry 20(11): 2055-2061 (2009).
Pons, M. et al., Liposomes obtained by the ethanol injection method, Int. J. Pharm., 95: 51-56. (1993).
Prata, C.A. et al., Lipophilic peptides for gene delivery. Bioconjugate Chemistry 19(2): 418-420 (2008).
Putnam, D. Polymers for gene delivery across length scales. Nature Materials 5: 439-451 (2006).
Putnam, D. and Langer, R., Poly(4-hydroxy-l-proline ester): Low-Temperature Polycondensation and Plasmid DNA Complexation. Macromolecules 32(11): 3658-3662 (1999).
Ratner, B.D. and Bryant, S., Biomaterials: where we have been and where we are going. Annual Review of Biomedical Engineering 6: 41-75 (2004).
Remington: The Science and Practice of Pharmacy, 21st Edition, Philadelphia, PA. Lippincott Williams & Wilkins (2005).
Rudolph, C. et al., Aerosolized Nanogram Quantities of Plasmid DNA Mediate Highly Efficient Gene Delivery to Mouse Airway Epithelium, Molecular Therapy, 12(3): 493-501 (2005).
Rudolph, C. et al., Methodological optimization of polyethylenimine (PEI)-based gene delivery to the lungs of mice via aerosol application, Journal of Gene Medicine, 7(1): 59-66 (2005).
Ryng, S. et al., Synthesis and structure elucidation of 5-aminomethinimino-3-methy1-4-isoxazolecarboxylic acid phenylamides and their immunological activity. Arch. Pharm. Pharm. Med. Chem 330(11):319-26 (1997).
Sahay, G. et al., Endocytosis of nanomedicines. Journal of Controlled Release 145: 182-195 (2010).
Sakiyama-Elbert, S.E. and Hubbell, J.A., Functional Biomaterials: Design of Novel Biomaterials. Annual Review of Materials Research 31: 183-201 (2001).
Shchori E., Poly(secondary Amine)s from Diacrylates and Diamines. Journal of Polymer Science 21(6):413-15 (1983).
Sherwood, R.F. Advanced drug delivery reviews: enzyme prodrug therapy, Adv. Drug Del. Rev., 22: 269-288 (1996).
Siegwart, D.J. et al., Combinatorial synthesis of chemically diverse core-shell nanoparticles for intracellular delivery. Proceedings of the National Academy of the Sciences of the USA 108(32):12996-123001 (2011).
Stern, L. et al., A novel antitumor prodrug platform designed to be cleaved by the endoprotease legumain, Bioconj. Chem., 20: 500-510 (2009).
Su, X. et al., Cytosolic Delivery Mediated Via Electrostatic Surface Binding of mRNA to Degradable Lipid-Coated Polymeric Nanoparticles, Polymer Preprints, 51(2):668-669 (2010).
Suri, M. et al., Genetics for Pediatricians, Remedica Publishing, (2005).
Szoka, F. and Papahadjopoulos, D., Comparative properties and methods of preparation of lipid vesicles (liposomes). Annual Review of Biophysics Bioengineering 9: 467-508 (1980).
Takahashi, Y. et al., Development of safe and effective nonviral gene therapy by eliminating CpG motifs from plasmid DNA vector, Frontiers in Bioscience, S4: 133-141 (2012).
Tan, S. et al., Engineering Nanocarriers for siRNA Delivery. Small 7(7): 841-856 (2011).
Tang, M.X. et al., In vitro gene delivery by degraded polyamidoamine dendrimers. Bioconjugate Chemistry 7(6): 703-714 (1996).
Tarcha, P.J. et al., Synthesis and characterization of chemically condensed oligoethylenimine containing beta-aminopropionamide linkages for siRNA delivery. Biomaterials 28: 3731-3740 (2007).
Thomas, C. E. et al., Progress and problems with the use of viral vectors for gene therapy, Nature Reviews/Genetics, 4: 346-358 (2003).
Thompson, P.E. et al., Antiamebic action of 5-chloro-7-diethylaminomethyl-8-quinolinol and of other substituted 8-quinolinols in vitro and in experimental animals. American Journal of Tropical Medicine and Hygiene 2(4): 224-248 (1955).
Toki, B.E. et al., Protease-mediated fragmentation of p-amidobenzyl ethers: a new strategy for the activation of anticancer prodrugs, J. Org. Chem., 67(6): 1866-1872 (2002).
Tranchant, I. et al., Physicochemical optimisation of plasmid delivery by cationic lipids. Journal of Gene Medicine 6: S24-S35 (2004).
Tsvetkov, D.E. et al., Neoglycoconjugates based on dendrimeric poly(aminoamides). Russian Journal of Bioorganic Chemistry 28(6): 470-486 (2002).
Urban-Klein, B. et al., RNAi-mediated gene-targeting through systemic application of polyethylenimine (PEI)-complexed siRNA in vivo. Gene Therapy 12(5): 461-466 (2005).
Van Balen, G.P. et al., Liposome/water lipophilicity: methods, information content, and pharmaceutical applications. Medicinal Research Reviews 24(3): 299-324 (2004).
Van De Wetering, P. et al., Structure-activity relationships of water-soluble cationic methacrylate/methacrylamide polymers for nonviral gene delivery. Bioconjugate Chemistry 10(4): 589-597 (1999).
Vandenbroucke, R.E. et al., Prolonged gene silencing in hepatoma cells and primary hepatocytes after small interfering RNA delivery with biodegradable poly(beta-amino esters). Journal of Gene Medicine 10: 783-794 (2008).
Veronese, F.M. et al., PEG-doxorubicin conjugates: influence of polymer structure on drug release, in vitro cytotoxicity, biodistribution, and antitumor activity, Bioconj. Chem., 16(4): 775-784 (2005).
Von Harpe et al., Characterization of commercially available and synthesized polyethylenimines for gene delivery. Journal of Controlled Release 69(2):309-322 (2000).
Walde, P. et al.. Preparation of Vesicles (Liposomes). Encyclopedia of Nanoscience and Nanotechnology. Nalwa, ed. American Scientific Publishers, Los Angeles 9:43-79 (2004).
Werth, S. et al., A low molecular weight fraction of polyethylenimine (PEI) displays increased transfection efficiency of DNA and siRNA in fresh or lyophilized complexes. Journal of Controlled Release 112: 257-270 (2006).
White, J.E. et al., Poly(hydroxyaminoethers): A New Family of Epoxy-Based Thermoplastics. Advanced Materials 12(23): 1791-1800 (2000).
White, J.E. et al., Step-growth polymerization of 10,11-epoxyundecanoic acid. Synthesis and properties of a new hydroxy-functionalized thermopastic polyester. Advanced Materials 48: 3990-3998 (2007).
Whitehead, K.A. et al., Knocking down barriers: advances in siRNA delivery. Nature Reviews Drug Discovery 8(2): 129-139 (2009).
Wu, J. et al., Cationic lipid polymerization as a novel approach for constructing new DNA delivery agents. Bioconjugate Chemistry 12(2): 251-257 (2001).
Wurdinger, T. et al., A secreted luciferase for ex-vivo monitoring of in vivo processes, Nat. Methods, 5(2):171-173 (2008).
Yoneda et al., A cell-penetrating peptidic GRP78 ligand for tumor cell-specific prodrug therapy, Bioorg. Med. Chern. Lett., 18(5): 1632-1636 (2008).
Yoshioka, Y. and Calvert, P., Epoxy-based Electroactive Polymer Gels. Experimental Mechanics 42(4): 404-408 (2002).
Zagridullin, P.H. et al., Monobasic amines. II. Cycloalkylation and hydroxyalkylation of cyclic and acyclic di-and polyamines. Journal of Organic Chemistry26(1):184-88. Russian (1990).
Zaugg, H.E. et al., 3-Carboxy-2,5-piperazinedione and Derivatives. Journal of American Chemical Society 78(11):2626-2631 (1956).
Zauner, W.et al., Polylysine-based transfection systems utilizing receptor-mediated delivery. Advanced Drug Delivery Reviews 30(1-3):97-113(1998).
Zintchenko, A. et al., Simple modifications of branched PEI lead to highly efficient siRNA carriers with low toxicity. Bioconjugate Chemistry 19(7): 1448-1455 (2008).
T. Wajima, et al., “Prediction of Human Pharmacokinetic Profile in Animal Scale Up Based on Normalizing Time Course Profiles”, Journal of Pharmaceutical Sciences, vol. 93, No. 7, Jul. 2004, p. 1890-1900.
J. Ra, et al., Safety of Intravenous Infusion of Human Adipose Tissue-Derived Mesenchymal Stem Cells in Animals and Humans, Stem Cells and Development, vol. 20, No. 8, 2011, p. 1297-1308.
Patton, J., Market Trends in Pulmonary Therapies, Trends and Opportunities, VI: 372-377 (2007).
Related Publications (1)
Number Date Country
20150110859 A1 Apr 2015 US
Provisional Applications (1)
Number Date Country
61894294 Oct 2013 US