![]() compositions, mrna encoding a gland and its use, use of at least one mrna molecule and a vehicle for
专利摘要:
COMPOSITIONS, MRNA THAT CODES FOR A HGLA AND ITS USE, USE AT LEAST ONE MRNA MOLECULE AND A TRANSFER VEHICLE AND USE OF A MRNA THAT CODES FOR EXOGENOUS PROTEIN.Compositions and methods for modulating the production of a protein in a target cell are disclosed in this document. The compositions and methods disclosed in this document are capable of ameliorating diseases associated with protein or enzyme deficiencies. 公开号:BR112013031553A2 申请号:R112013031553-9 申请日:2012-06-08 公开日:2020-11-10 发明作者:Braydon Charles Guild;Frank DeRosa;Michael Heartlein 申请人:Shire Human Genetic Therapies, Inc.; IPC主号:
专利说明:
COMPOSITIONS, mRNA THAT CODES FOR A HGLA AND ITS USE, USE AT LEAST ONE MRNA MOLECULE AND ONE TRANSFER VEHICLE AND USE OF AN mRNA THAT CODES FOR PROTEIN EXÓGENA 5 New approaches and therapies are still needed for the treatment of protein and enzyme deficiencies. For example, lysosomal storage diseases are a group of approximately 50 rare inherited metabolic disorders that result from defects in lysosomal function, usually due to a deficiency of an enzyme required for metabolism. Fabry's disease is a lysosomal storage disease that results from a deficiency of the alpha enzyme galactosidase (GLA), which causes a glycolipid known as globotriaosylceramide to accumulate in blood vessels and other tissues, leading to various painful manifestations. For certain diseases, such as Fabry's disease, there is no need to replace a protein or enzyme that is normally secreted by cells in the bloodstream. Therapies, such as gene therapy, that increase the level or production of an affected protein or enzyme could provide a treatment or even a cure for these disorders. However, there are several limitations to the use of conventional gene therapy for this purpose. Conventional gene therapy involves the use of DNA to insert desired genetic information into host cells. The DNA introduced into the cell is normally integrated to some extent in the genome of one or more transfected cells, allowing long-term action of the genetic material introduced into the host. While there may be substantial benefits to this sustained action, the integration of exogenous DNA into a host genome can also have many deleterious effects. For example, it is possible for the introduced DNA to be inserted into an intact gene, resulting in a mutation that prevents or even completely eliminates the function of the endogenous gene. Thus, gene therapy with DNA can result in the compromise of a vital genetic function in the treated host, such as eliminating or deleteriously reducing the production of an essential enzyme or interrupting a gene essential for the regulation of cell growth, resulting in proliferation cancerous or unregulated cells. In addition, with conventional DNA-based gene therapy, it is necessary, for efficient expression of the desired gene product, to include a strong promoter sequence, which again can lead to undesirable changes in the regulation of normal gene expression in the cell. It is also possible that genetic material based on DNA results in the induction of undesirable antiDNA antibodies, which in turn can trigger a possibly fatal immune response. Gene therapy approaches using viral vectors can also result in an adverse immune response. In some circumstances, the viral vector may integrate into the host's genome. In addition, the production of clinical-grade viral vectors is also expensive and time-consuming. The target distribution of the genetic material introduced using viral vectors can also be difficult to control. Thus, while DNA-based gene therapy is evaluated for distribution of secreted proteins using viral vectors (US Patent 6,066,626; US2004 / 0110709), these approaches can be limited by these various criteria. Another apparent obstacle in these earlier approaches to the distribution of nucleic acids that encode secreted proteins is in the levels of protein that are produced in the end. It is difficult to achieve significant levels of the desired protein in the blood, and the amounts are not sustained over time. For example, the amount of protein produced by the distribution of nucleic acid does not reach normal physiological levels. See, for example, US2004 / 0110709. In contrast to DNA, the use of RNA as a gene therapy agent is substantially safer because (1) RNA does not involve the risk of being integrated stably into the genome of transfected cells, thus eliminating the concern that the genetic material introduced will impair the normal functioning of an essential gene, or cause a mutation that results in deleterious or oncogenic effects; (2) strange promoter sequences are not necessary for effective translation of the encoded protein, again avoiding harmful side effects; (3) in contrast to plasmid DNA (pDNA), messenger RNA (mRNA) is devoid of immunogenic CpG motifs so that antiRNA antibodies are not generated; and (4) any deleterious effects resulting from mRNA based on gene therapy would be of limited duration due to the relatively short half-life of the RNA. In addition, it is not necessary for mRNA to enter the nucleus to perform its function, while DNA must overcome this major obstacle. One reason that mRNA-based gene therapy has not been used in the past is that mRNA is much less stable than DNA, especially when it reaches a cell's cytoplasm and is exposed to degrading enzymes. The presence of a hydroxyl group on the second carbon of the sugar fraction in mRNA causes steric impediment that prevents the mRNA from forming the most stable double helix structure of DNA and, therefore, makes the mRNA more prone to hydrolytic degradation. As a result, until recently, mRNA was believed to be very unstable to support transfection protocols. Advances in RNA stabilization modifications have sparked more interest in the use of mRNA in place of plasmid DNA in gene therapy. Certain delivery vehicles, such as cationic lipid or polymer delivery vehicles, can also help protect the transfected mRNA from endogenous RlSlases. Yet, despite the increased stability of the modified mRNA, the distribution of mRNA to cells in vivo of a form that allows therapeutic levels of protein production is still a challenge, especially for mRNAs that encode whole proteins. Although the distribution of the mRNA encoding secreted proteins has been contemplated (US2009 / 0286852), the levels of an entire secreted protein that could be produced through the distribution of mRNA in vivo are not known and there is no reason to expect the levels to exceed those seen with DNA-based gene therapy. To date, significant progress using mRNA gene therapy has only been made in applications for which low levels of translation are not a limiting factor, such as immunization with mRNA that encodes antigens. Clinical trials involving vaccination against tumor antigens by intradermal injection of pure or protamine-complexed mRNA have demonstrated viability, absence of toxicity and promising results. X. Su et al., Mol. Pharmaceutics 8: 774-787 (2011). Unfortunately, low levels of translation have severely restricted the exploration of mRNA-based gene therapy in other applications that require high levels of sustained expression of the mRNA-encoded protein to exert a biological or therapeutic effect. The invention provides methods for delivering therapeutic mRNA gene agents that lead to the production of therapeutically effective levels of the secreted proteins through a "deposit effect". In embodiments of the invention, the mRNA encoding a secreted protein is loaded onto lipid nanoparticles and delivered to target cells in vivo. The target cells then act as a source of deposit for the production of soluble protein, secreted into the circulatory system at therapeutic levels. In some modalities, the levels of secreted protein produced are above normal physiological levels. The invention provides compositions and methods for intracellular delivery of mRNA in a liposomal transfer vehicle to one or more target cells for the production of therapeutic levels of the functional secreted protein. The compositions and methods of the invention are useful in the management and treatment of a large number of diseases, in particular, diseases resulting from protein and / or enzyme deficiencies, in which the protein or enzyme is normally secreted. Individuals suffering from these diseases may have underlying genetic defects that lead to impaired expression of a protein or enzyme, including, for example, non-synthesis of the secreted protein, reduced synthesis of the secreted protein or synthesis of a secreted protein lacking or having decreased biological activity. In particular, the methods and compositions of the invention are useful for treating lysosomal storage disorders and / or metabolic disorders of the urea cycle that occur as a result of one or more defects in the biosynthesis of the secreted enzymes involved in the urea cycle. The compositions of the invention comprise an mRNA, a transfer vehicle and, optionally, an agent to facilitate contact with and subsequent transfection of a target cell. The mRNA can encode a clinically useful secreted protein. For example, mRNA can encode a functional secreted urea cycle enzyme or a secreted enzyme implicated in lysosomal storage disorders. The mRNA can encode, for example, erythropoietin (e.g., human EPO) or α-galactosidase (e.g., human α-galactosidase (human GLA)). In some embodiments the mRNA may comprise one or more modifications that confer stability to the mRNA (for example, compared to a wild-type or native version of the mRNA) and may also comprise one or more modifications in relation to the wild-type that corrects a defect involved in the associated abnormal protein expression. For example, the nucleic acids of the present invention may comprise modifications for one or both 5 'and 3' untranslated regions. These modifications may include, but are not limited to, the inclusion of a partial sequence of an immediate-onset cytomegalovirus (CMV) gene 1 (IEI), a poly A tail, a Capl structure or a sequence encoding human growth hormone (hGH ). In some embodiments, mRNA is modified to decrease mRNA immunogenicity. Methods for treating a subject comprising administering a composition of the invention are also contemplated. For example, methods for treating or preventing conditions in which the production of a particular secreted protein and / or use of a particular secreted protein is inadequate or compromised are provided. In one embodiment, the methods provided in this document can be used to treat a subject having a deficiency in one or more enzymes in the urea cycle or in one or more enzymes deficient in a lysosomal storage disorder. In a preferred embodiment, the mRNA in the compositions of the invention is formulated in a liposomal transfer vehicle to facilitate delivery to the target cell. Predicted transfer vehicles may include one or more cationic lipids, non-cationic lipids, and / or PEG-modified lipids. For example, the transfer vehicle may comprise at least one of the following cationic lipids: C12-200, DLin-KC2-DMA, DODAP, HGT4003, ICE, HGT5000, or HGT5001. In the embodiments, the transfer vehicle comprises cholesterol (chol) and / or a PEG-modified lipid. In some embodiments, the transfer vehicles comprise DMG-PEG2K. In certain embodiments, the transfer vehicle comprises one of the following lipid formulations: Cl2-200, DOPE, chol, DMG-PEG2K; DODAP, DOPE, cholesterol, DMG-PEG2K; HGT5000, DOPE, chol, DMG-PEG2K, HGT5001, DOPE, chol, DMG-PEG2K. The invention also provides compositions and methods useful to facilitate the transfection and delivery of one or more mRNA molecules to target cells capable of exhibiting the "deposit effect". For example, the compositions and methods of the present invention contemplate the use of targeting ligands capable of improving the affinity of the composition for one or more target cells. In one embodiment, the targeting ligand is apolipoprotein-B or apolipoprotein-E and the corresponding target cells express low-density lipoprotein receptors, thus facilitating recognition of the targeting ligand. The methods and compositions of the present invention can be used to preferentially target a large number of target cells. For example, target cells contemplated include, but are not limited to, hepatocytes, epithelial cells, hematopoietic cells, epithelial cells, endothelial cells, lung cells, bone cells, stem cells, mesenchymal cells, neural cells, cardiac cells, adipocytes, vascular smooth muscle, cardiomyocytes, skeletal muscle cells, beta cells, pituitary cells, synovial lining cells, ovarian cells, testicular cells, fibroblasts, B cells, T cells, reticulocytes, leukocytes, granulocytes and tumor cells. In embodiments, the secreted protein is produced by the target cell for sustained amounts of time. For example, the secreted protein can be produced for more than an hour, more than four, more than six, more than 12, more than 24, more than 48 hours, or more than 72 hours after administration. In some embodiments, the polypeptide is expressed at a peak level of about six hours after administration. In some embodiments, expression of the polypeptide is sustained at least at a therapeutic level. In some embodiments, the polypeptide is expressed at at least one therapeutic level for more than one, more than four, more than six, more than 12, more than 24, more than 48 hours, or more than 72 hours after administration. In some embodiments, the polypeptide is detectable at level 5 in the patient's serum or tissue (eg, liver or lung). In some embodiments, the level of the detectable polypeptide is one of continuous expression of the mRNA composition over time periods of more than one, more than four, more than six, more than 12, more than 24, more than 48 hours, or more than 72 hours after administration. In certain embodiments, the secreted protein is produced at levels above normal physiological levels. The level of secreted protein can be increased compared to a control. In some embodiments, control is the baseline physiological level of the polypeptide in a normal individual or in a population of normal individuals. In other embodiments, control is the baseline physiological level of the polypeptide in an individual having a deficiency in the relevant protein or polypeptide or in a population of individuals having a deficiency in the relevant protein or polypeptide. In some embodiments, the control may be the normal level of the relevant protein or polypeptide in the individual to whom the composition is administered. In other modalities, control is the level of expression of the polypeptide after other therapeutic interventions, for example, after direct injection of the corresponding polypeptide, at one or more comparable time points. In certain embodiments, the polypeptide is expressed by the target cell at a level that is at least 1.5 times, at least 2 times, at least 5 times, at least 10 times, at least 20 times, 30 times, at least 100 times at least 500 times, at least 5000 times, at least 50,000 times, or at least 100,000 times greater than a control. In some modalities, the number of times of expression increase more than the control is sustained by more than one, more than four, more than six, more than 12, more than 24, or more than 48 hours or more than 72 hours after administration. For example, in one embodiment, levels of secreted protein are detected in the serum at least 1.5 times, at least 2 times, at least 5 times, at least 10 times, at least 20 times, 30 times, at least 100 times at least 500 times, at least 5000 times, at least 50,000 times or at least 100,000 times more than in a control for at least 48 hours or 2 days. In certain embodiments, the levels of secreted protein are detectable within 3 days, 4 days, 5 days, or 1 week or more after administration. Increased levels of secreted protein can be seen in serum and / or tissue (eg, liver, lung). In some embodiments, the method produces a sustained circulation half-life of the desired secreted protein. For example, the secreted protein can be detected for hours or days longer than the half-life observed through subcutaneous injection of the secreted protein. In the modalities, the half-life of the secreted protein is sustained for more than 1 day, 2 days, 3 days, 4 days, 5 days, or 1 week or more. In some embodiments, the administration comprises a single or repeated dose. In certain modalities, the dose is administered intravenously, or by pulmonary delivery. The polypeptide can be, for example, one or more of erythropoietin, cl-galactosidase, LDL receptor, Factor VIII, Factor IX, oL-iduronidase (for MPS I), iduronate sulfatase (for MPS II), heparin-N-sulfatase (for MPS IIIA), oc-N-acetylglucosaminidase (for MPS IIIB), galactose 6-sulfatase (for MPS IVA), lysosomal acid lipase, arylsulfatase. Certain embodiments refer to the compositions and methods that provide a cell or subject with mRNA, at least a portion that encodes a functional protein, in an amount that is substantially less than the amount of corresponding functional protein generated from the mRNA. In other words, in certain embodiments, the mRNA delivered to the cell can produce an amount of protein that is substantially greater than the amount of mRNA delivered to the cell. For example, over a given period of time, for example 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 15, 20 or 24 hours after mRNA administration to a cell or subject , the amount of corresponding protein generated by that mRNA can be at least 1.5, 2, 3, 5, 10, 15, 20, 25, 50, 100, 150, 200, 250, 300, 400, 500, or more times greater than the amount of mRNA actually delivered to the cell or subject. This can be measured on a mass-by-mass basis, on a mol-per-mol basis, and / or on a molecule-per-molecule basis. Protein can be measured in several ways. For example, for a cell, the measured protein can be measured as an intracellular protein, an extracellular protein, or a combination of the two. For a subject, the measured protein can be protein measured in the serum; in a specific tissue or tissues such as the liver, kidney, heart or brain; in a specific cell type such as one of several types of liver or brain cells; or in any combination of serum, tissue type, and / or cells. Besides that, a basal amount of the endogenous protein can be measured in the cell or subject prior to administration of the mRNA and then subtracted from the measured protein after administration of the mRNA to produce the corresponding amount of protein generated from the mRNA. In this way, mRNA can provide a reservoir or deposit source of a large amount of therapeutic material for the cell or subject, for example, compared to the amount of mRNA delivered to the cell or subject. The deposition source can act as a continuous source for expression of mRNA polypeptide for sustained periods of time. The above discussed, and many other features and associated advantages of the present invention will become better understood with reference to the following detailed description of the invention when taken in conjunction with the accompanying examples. The various modalities described in this document are complementary and can be combined or used together in a manner understood by the person skilled in the art taking into account the teachings contained in this document. BRIEF DESCRIPTION OF THE DRAWINGS FIG. 1 shows the nucleotide sequence of a 5 'CMV sequence (SEQ ID NO: 1), where X, if present, is GGA. FIG. 2 shows the nucleotide sequence of a 3 'hGH sequence (SEQ ID NO: 2). FIG. 3 shows the nucleotide sequence of human erythropoietin (EPO) mRNA (SEQ ID NO: 3). This sequence can be flanked at the 5 'end with SEQ ID NO: 1 and at the 3' end with SEQ ID NO: 2. FIG. 4 shows the nucleotide sequence of human alpha-galactosidase (GLA) rnRNA (SEQ ID NO: 4). This sequence can be flanked at the 5 'end with SEQ ID NO: 1 and at the 3' end with SEQ ID NO: 2. FIG. 5 shows the nucleotide sequence of human alpha-1 antitrypsin mRNA (AIAT) (SEQ ID NO: 5). This sequence can be flanked at the 5 'end with SEQ ID NO: 1 and at the 3' end with SEQ ID NO: 2. FIG. 6 shows the nucleotide sequence of human factor IX (FIX) mRNA (SEQ ID NO: 6). This sequence can be flanked at the 5 'end with SEQ ID NO: 1 and at the 3' end with SEQ ID NO: 2. FIG. 7 shows the quantification of secreted hEPO protein levels measured by ELISA. The detected protein is a result of its production from hEPO mRNA distributed intravenously through a single dose of several lipid nanoparticle formulations. The formulations C12-200 (30 µg), HGT4003 (150 µg), ICE (100 µg), DODAP (200 µg) are represented as the cationic / ionizable lipid component of each test article (For / nulls 1-4) . Values are 5 based on a blood sample four hours after administration. FIG. 8 shows the hematocrit measurement of mice treated with a single IV dose of lipid nanoparticles loaded with human EPO mRNA (Formulations 1-4). Whole blood samples were taken at 4 hours (Day 1), 24 hours (Day 2), 4 days, 7 days and 10 days after administration. FIG. 9 shows hematocrit measurements from mice treated with lipid nanoparticles loaded with human EPO mRNA with a single IV dose or three injections (day 1, day 3, day 5). Whole blood samples were taken before the injection (day -4), day 7 and day 15. Formulation 1 was administered: (30 µg, single dose) or (3 x 10 µg, dose day 1, day 3, day 5 ); Forniulation 2 was administered: (3 x 50 µg, dose day 1, day 3, day 5). FIG. 10 shows the quantification of levels of secreted human β-galactosidase (hGLA) protein measured by ELISA. The detected protein is a result of the production of hGLA mRNA distributed through lipid nanoparticles (Formulation 1; single intravenous dose of 30 µg, based on the encapsulated mRNA). The hGLA protein is detected within 48 hours. FIG. 11 shows hGLA activity in serum. HGLA activity was measured using the substrate 4-methylumbelliferyl-a-D-galactopyranoside (4-MU-a-gal) at 37 ° C. The data are the average of 6 to 9 individual measurements. FIG. 12 shows the quantification of serum hGLA protein levels measured by ELISA. The protein is produced from hGLA mRNA distributed through C12-200-based lipid nanoparticles (C12-200: DOPE: Chol: DMGPEG2K, 40: 30: 25: 5 (Formulation /); 30 ug mRNA-based mRNA encapsulated, single IV dose). The hGLA protein is monitored for 72 hours. single intravenous dose based on encapsulated mRNA). The hGLA protein is monitored for 72 hours. FIG. 13 shows the quantification of hGLA protein levels in the liver, kidneys and spleen, measured by ELISA. The protein is produced from hGLA mRNA distributed through Cl2-200-based lipid nanoparticles (Formulation 1; 30 µg mRNA based on encapsulated mRNA, single dose IV). The hGLA protein is monitored for 72 hours. FIG. 14 shows a dose-response study monitoring hGLA protein production as MRT-derived Hunan GLA protein secreted in serum (A) and liver (B). Samples were measured 24 hours after administration (Formulation 1; single dose 5, IV, N = 4 mice / group) and quantified by ELISA. FIG. 15 shows the pharmacokinetic profiles of ERT-based alpha-galactosidase in pure mice without thymus (40 µg / kg dose) and hGLA protein produced from MRT (Formulation /; 1.0 mg / kg dose of mRNA). FIG. 16 shows the quantification of levels of hGLA protein secreted in Fabry mice treated with MRT measured by ELISA. The hGLA protein is produced from hGLA mRNA distributed through C12-200-based lipid nanoparticles (Delivery /; 10 µg mRNA per single intravenous dose, based on encapsulated mRNA). the serum is monitored for 72 hours. FIG. 17 shows the quantification of hGLA protein levels in the liver, kidneys, spleen and heart of KO Fabry mice treated with MRT, measured by ELISA. The protein is produced from hGLA mRNA distributed through C12-200-based lipid nanoparticles (Formulation 1); 30 µg mRNA based on encapsulated mRNA, single dose IV). The hGLA protein is monitored for 72 hours. Literature values representing normal physiological levels are plotted as dashed lines. FIG. 18 shows the quantification of hGLA protein levels secreted in Fabry mice treated with Alpha-galactosity and MRT measured by ELISA. Both therapies were dosed as a single intravenous dose of 1.0 mg / kg. FIG. 19 shows the quantification of hGLA protein levels in the liver, kidneys, spleen and heart of KO Fabry mice treated with ERT (alpha-galactosity) and MRT, measured by ELISA. The protein is produced from hGLA rnRNA distributed through lipid nanoparticles (Formulation /; 1.0 mg / kg mRNA based on encapsulated mRNA, single dose IV). FIG. 20 shows the relative quantification of globotrioasilceramide (Gb3) and lyso-Gb3 in the kidneys of treated and untreated mice. Male KO Fabry mice were treated with a single dose of lipid nanoparticles loaded with 1.0 mg / kg GLA or Alpha-galactosidase mRNA. The amounts reflect the amount of Gb3 / smooth-Gb3 one week after administration. FIG. 21 shows the relative quantification of globotrioasilceramide (Gb3) and lyso- .. Gb3 in the heart of treated and untreated mice. Male KO Fabry mice were treated with a single dose of lipid nanoparticles loaded with 1.0 mg / kg GLA or Alpha-galactosidase mRNA. The amounts reflect the amount of Gb3 / smooth-Gb3 one week after administration. FIG. 22 shows a dose-response study monitoring the production of GLA protein as human GLA protein derived from MRT secreted in serum. Samples were measured 24 hours after administration (single dose, IV, N = 4 mice / group) of lipid nanoparticles based on HGT4003 (Formulation 3) or HGT5000 10 (Formu / action 5) and quantified by ELISA. FIG. 23 shows the production of hGLA protein measured in serum (A) or in the liver, kidneys and spleen (B). Samples were measured 6 hours and 24 hours after administration (single dose, IV, N = 4 mice / group) of lipid nanoparticles based on HGT5001 (Formulation 6) and quantified by ELISA. FIG. 24 shows the quantification of secreted human Factor IX protein levels measured by ELISA (mean ng / mL ± standard deviation). The FIX protein is produced from the FIX mRNA distributed through C12-200-based lipid nanoparticles (C12-200: DOPE: Chol: DMGPEG2K, 40: 30: 25: 5 (Formulation /); 30 ug mRNA per dose intravenous injection, based on encapsulated mRNA). The FIX protein is monitored 20 over 72 hours. (n = 24 mice) FIG. 25 shows the quantification of the levels of secreted human ot-1-antitrypsin protein (AIAT) measured by ELISA. The AIAT protein is produced from the AIAT mRNA distributed through C12-200-based lipid nanoparticles (C12-200: DOPE: Chol: DMGPEG2K, 40: 30: 25: 5 (Formulation 1); 30 ug mRNA per dose 25 single intravenous, based on encapsulated mRNA). The AIAT protein is monitored for 24 hours. FIG. 26 shows an ELISA-based quantification of hEPO protein detected in the lungs and serum of treated mice after intratracheal administration of nanoparticles loaded with hEPO mRNA (measured mIU) (lipid nanoparticles 30 based on C12-200, HGT5000 or HGT5001; Formulations 1, 5, 6 respectively). The animals were sacrificed 6 hours after administration (n = 4 mice per group). DESCRIPTION OF EXEMPLARY MODALITIES The invention provides compositions and methods for intracellular delivery of mRNA in a liposomal transfer vehicle to one or more target cells for the production of therapeutic levels of functional secreted protein. The term "functional", as used in this document to qualify a protein or enzyme, means that the protein or enzyme has biological activity, or alternatively is able to perform the same, or a similar function as the native or functioning protein or enzyme normal. The mRNA compositions of the invention are useful for the treatment of various metabolic or genetic disorders and, in particular those genetic or metabolic disorders that involve the non-expression, wrong expression or deficiency of a protein or enzyme. The term "therapeutic levels" refers to levels of protein detected in blood or tissues that are above control levels, where control can be normal physiological levels, or levels in the subject prior to administration of the mRNA composition. The term "secreted" refers to the protein that is detected outside the target cell, in the extracellular space. The protein can be detected in the blood or in the tissues. In the context of the present invention the term "produced" is used in its broadest sense to refer to the translation of at least one mRNA into a protein or enzyme. As provided herein, the compositions include a transfer vehicle. As used herein, the term "transfer vehicle" includes any of the carriers, diluents, standard pharmaceutical excipients and the like that are generally intended for use in connection with the administration of biologically active agents, including nucleic acids. The compositions and, in particular, the transfer vehicles described in this document are capable of delivering the mRNA to the target cell. In the modalities, the transfer vehicle is a lipid nanoparticle. mRNA The mRNA in the compositions of the invention can encode, for example, a hormone, enzyme, receptor, polypeptide, peptide or other secreted protein of interest that is normally secreted. In one embodiment of the invention, the rnRNA can optionally have chemical or biological modifications, which, for example, improve the stability and / or half-life of that mRNA or which improve or otherwise facilitate the production of protein. The methods of the invention provide optional codistribution of one or more unique mRNA to target cells, for example, combining two unique mRNAs in a single transfer vehicle. In one embodiment of the present invention, a first therapeutic mRNA and a second therapeutic mRNA can be formulated in a single, administered delivery vehicle. The present invention also contemplates the co-distribution and / or co-administration of a first therapeutic mRNA and a second nucleic acid to facilitate and / or improve the function or distribution of the first therapeutic mRNA. For example, that second nucleic acid (for example, synthetic or exogenous mRNA) can encode a membrane transport protein that after expression (for example, translation of exogenous or synthetic mRNA) facilitates the distribution or improves the biological activity of the first mRNA . Alternatively, the first therapeutic mRNA can be administered with a second nucleic acid that functions as a "chaperone", for example, to target the folding of any first therapeutic mRNA. The methods of the invention also provide for the delivery of one or more therapeutic nucleic acids to treat a unique disorder or deficiency, each of which nucleic acids functions by a different mechanism of action. For example, the compositions of the present invention can comprise a first therapeutic mRNA which, for example, is administered to correct an endogenous protein or enzyme deficiency, and which is accompanied by a second nucleic acid, which is administered to disable or "silence" a defective endogenous nucleic acid and its protein or enzyme product. This "second" nucleic acid can encode, for example, mRNA or s1RNA. After transfection, a natural mRNA in the compositions of the invention can decay with a half-life of between 30 minutes and several days. The mRNA in the compositions of the invention preferably maintains at least some ability to be translated, thereby producing a functional secreted protein or enzyme. Accordingly, the invention provides compositions comprising and methods for administering a stabilized mRNA. In some embodiments of the invention, mRNA activity is prolonged over an extended period of time. For example, mRNA activity can be prolonged so that the compositions of the present invention are administered to a subject on a bi-weekly Oll bi-weekly basis, or more preferably on a monthly, bimonthly, quarterly or annual basis. The prolonged or extended activity of the mRNA of the present invention is directly related to the amount of secreted functional protein or enzyme produced from that mRNA. Likewise, the activity of the compositions of the present invention can be further extended or prolonged by modifications made to improve or enhance mRNA translation. In addition, the amount of protein or functional enzyme produced by the target cell is a function of the amount of mRNA distributed to the target cells and the stability of that mRNA. As the stability of the mRNA of the present invention can be improved or improved, the half-life, the activity of the secreted protein or enzyme produced and the dosing frequency of the composition can be extended again. Accordingly, in some embodiments, the mRNA in the compositions of the invention comprises at least one modification that provides increased or improved stability for the nucleic acid, including, for example, improved resistance to nuclease digestion in vivo. As used in this document, the terms "modification" and "modified" as these terms relate to the nucleic acids presented in this document, include at least one change that preferably improves stability and makes nial mRNA stable (for example, resistant to digestion nuclease) than the wild-type or naturally occurring version of the mRNA. As used herein, the terms "stable" and "stability" as these terms relate to the nucleic acids of the present invention and particularly with respect to mRNA, refer to increased or improved resistance to degradation by, for example, nucleases (ie, endonucleases or exonucleases) that are normally able to degrade that mRNA. Increased stability may include, for example, less sensitivity to hydrolysis or other destruction by endogenous enzymes (e.g., endonucleases or exonucleases) or conditions within the target cell or tissue, thereby increasing or improving the residence of that mRNA in the cell, tissue, subject and / or target cytoplasm. The stabilized mRNA molecules provided in this document demonstrate a longer half-life compared to naturally occurring, unmodified homologs (for example, the wild-type version of the mRNA). Also contemplated by the terms "modification" and "modified" as those terms relate to the mRNA of the present invention are changes that improve or enhance the translation of mRNA nucleic acids, including, for example, the inclusion of sequences that work in the initiation of translation of proteins (for example, the Kozac consensus sequence). (Kozak, M., Nucleic Acids Res 15 (20): 8125-48 (1987)). In some embodiments, the invention's mRNA has undergone chemical or biological modification to make it more stable. Exemplary modifications of an mRNA include the depletion of a base (for example, by deletion or replacement of one nucleotide with another) or modification of a base, for example, the chemical modification of a base. The phrase "chemical modifications" as used in this document, includes modifications that introduce chemical substances that differ from those seen in naturally occurring mRNA, for example, covalent modifications, such as the introduction of modified nucleotides, (for example, nucleotide analogs, or the inclusion of groups 5 ligands that are not found naturally in these mRNA molecules). In addition, appropriate modifications include changes in one or more nucleotides in a codon so that the codon encodes the same amino acid, but is more stable than the codon found in the wild-type version of the mRNA. For example, an inverse relationship between RNA stability and a greater number of cytidine (Cs) and / or uridine (Us) residues has been demonstrated, and RNA devoid of C and U residues has been shown to be stable for most RNases ( Heidenreich, et al. J Biol Chem 269, 213 1-8 (1994). In some embodiments, the number of C and / or U residues in an mRNA sequence is reduced. In another embodiment, the number of C and / or U residues is reduced by replacing a codon that encodes a particular amino acid with another codon that encodes the same or a related amino acid. The modifications contemplated for the mRNA nucleic acids of the present invention also include the incorporation of pseudouridines. The incorporation of pseudouridines into the mRNA nucleic acids of the present invention can improve the stability and translucency, as well as decrease immunogenicity in vivo. See, for example., Karikó, K., et al., Molecular Therapy 16 (11): 1833-1840 (2008). Substitutions and modifications to the mRNA of the present invention can be carried out by methods readily known to a person skilled in the art. The restrictions on reducing the number of C and U residues in a sequence are likely to be greater within the coding region of an mRNA, compared to an untranslated region, (that is, it will probably not be possible to eliminate all C and U residues present in the message while still maintaining the message's ability to encode the desired amino acid sequence). The degeneration of the genetic code, however, presents an opportunity to allow the number of C and / or U residues that is present in the sequence to be reduced, while maintaining the same coding capacity (that is, depending on which amino acid is encoded by a codon, several different possibilities for modifying RNA sequences may be possible). For example, codons for Gly can be changed to GGA or GGG instead of GGU or GGC. The term modification also includes, for example, the incorporation of non-nucleotide bonds or modified nucleotides into the mRNA sequences of the present invention (for example, modifications to one or both 3 'and 5' ends of an mRNA molecule encoding a protein or functional secreted enzyme). These modifications 5 include the addition of bases to an mRNA sequence (for example, the inclusion of a longer poly A tail or longer poly A tail), the alteration of 3 'UTR or 5' UTR, complexation of the mRNA with an agent ( for example, a protein or a complementary nucleic acid molecule) and the inclusion of elements that alter the structure of an mRNA molecule (for example, that form secondary structures). The poly A tail appears to stabilize natural messengers. Therefore, in one embodiment, a long poly A tail can be added to an mRNA molecule, making the mRNA more stable. Poly A tails can be added using a variety of recognized techniques. For example, long poly A tails can be added to synthetic mRNA or transcribed in vitro 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. In one embodiment, the length of the poly A tail is at least about 90, 200, 300, 400, at least 500 nucleotides. In one embodiment, the length of the poly A tail is adjusted to control the stability of a modified mRNA molecule of the invention and thus the transcription of the protein. For example, since the length of the poly A tail can influence the half-life of an mRNA molecule, the length of the poly A tail can be adjusted to modify the level of resistance of the mRNA to nucleases and thus control the course of the time of protein expression in a cell. In one embodiment, the stabilized mRNA molecules are sufficiently resistant to degradation in vivo (for example, by nucleases), so that they can be delivered to the target cell without a transfer vehicle. In one embodiment, an mRNA can be modified by untranslated sequences (RTU) 3 'and / or 5' of incorporation that are not found naturally in wild-type mRNA. In one embodiment, the 3 'and / or 5' flanking sequences that naturally flank one mRNA and encode a second unrelated protein can be incorporated into the nucleotide sequences of an mRNA molecule that encodes a functional or therapeutic protein in order to modify them. For example, the 3 'or 5' sequences of mRNA molecules that are stable (for example, globin, actin, GAPDH, tubulin, histone or enzymes in the citric acid cycle) can be incorporated into the 3 'and / or 5 region 'of a sense mRNA nucleic acid molecule to increase the stability of the sense mRNA molecule. See, for example, 5 US2003 / 0083272. In some embodiments, the mRNA in the compositions of the invention include modifications of the 5 'end of the mRNA to include a partial sequence of a CMV immediate start gene (IEI), or a fragment thereof (for example, SEQ ID NO: 1) to improve nuclease resistance and / or improve mRNA half-life. In addition to increasing the stability of the mRNA nucleic acid sequence, it was surprisingly found that the inclusion of a partial sequence of a CMV immediate-onset gene (IEI) improves mRNA translation and expression of the functional enzyme or protein. Also contemplated is the inclusion of a human growth hormone (hGH) gene sequence, or a fragment thereof (eg, SEQ ID NO: 2) for the 15 3 'ends of the nucleic acid (eg "mRNA) to further stabilize the mRNA. Generally, preferred modifications improve the stability and / or pharmacokinetic properties (e.g., half-life) of the mRNA relative to its unmodified counterparts and include, for example, modifications made to improve the resistance of that mRNA to in vivo nuclease digestion. 20 Variants of the nucleic acid sequence of SEQ ID NO: 1 and / or SEQ ID NO: 2 are also contemplated, where the variants retain the functional properties of the nucleic acids including mRNA stabilization and / or pharmacokinetic properties (for example, half -life). Variants can have more than 9 ° / 0, more than 95 ° 4, more than 98 ° 4, or more and 99 ° 4 of sequence identity for SEQ ID NO: 1 or SEQ ID NO: 2. In some embodiments, the composition may comprise a stabilizing reagent. The compositions can include one or more formulation reagents that bind directly or indirectly and stabilize the mRNA, improving the residence time in the target cell. These reagents preferentially lead to an improved mRNA half-life in the target cells. For example, the stability of an mRNA and the efficiency of translation can be increased by the incotporation of "stabilizing reagents" that form complexes with naturally occurring niRNA within a cell (see, for example, US Patent 5,677,124) . The incorporation of a stabilizing reagent can be carried out, for example, by combining poly A and a protein with the mRNA to be stabilized in vitro before loading or encapsulating the mRNA within a transfer vehicle. Exemplary stabilizing reagents include one or more of proteins, peptides, aptamers, translational accessory protein, mRNA binding proteins, and / or translation initiation factors. 5 The stabilization of the compositions can also be improved by the use of opsonization inhibiting fractions, which are typically large hydrophilic polymers that are chemically or physically attached to the transfer vehicle (for example, by the intercalation of a lipid-soluble anchor in the membrane itself, or linking directly to active groups of membrane lipids). These hydrophilic polymers 10 opsonization inhibitors form a protective surface layer that significantly decreases the absorption of liposomes by the monocyte-macrophage system and reticuloendothelial system, (for example, as described in US Patent 4,920,016, the entire disclosure of which is incorporated in this document. as a reference.) Transfer vehicles with opsonization inhibiting fractions thus remain in circulation for 15 much longer than their unmodified counterparts. When RNA is hybridized to a complementary nucleic acid molecule (for example, DNA or RNA) it can be protected from nucleases. (Krieg, et al. Melton. Methods in Enzymology. 1987; 155, 397-415). The stability of the hybridized mRNA is probably due to the inherent single-strand specificity of most RNases. 20 In some embodiments, the stabilizing reagent selected to complex an mRNA is a eukaryotic protein, (for example, a mammalian protein). In yet another mode, the mRNA can be modified by hybridization to a second nucleic acid molecule. If an entire mRNA molecule is hybridized to a complementary nucleic acid molecule, the initiation of translation can be reduced. In some embodiments, the 5 'untranslated region and the AUG start region of the mRNA molecule can optionally be kept unhybridized. After the initiation of the translation, the unwinding activity of the ribosome complex can work even in high-affinity duplex so that you can continue the translation. (Liebhaber. J. Mol. Biol. 1992; 226: 2-13; Monia, el al. J Biol Chem. 1993; 268: 14514-22.) 30 It will be understood that any of the methods described above to improve mRNA stability can be used alone or in combination with one or more of any of the other methods and / or compositions described above. The mRNA of the present invention can optionally be combined with a reporter gene (for example, upstream or downstream of the mRNA coding region) which, for example, facilitates the determination of the distribution of mRNA to target cells or tissues. Appropriate reporter genes may include, for example, Fluorescent Green Protein inRNA (GFP mRNA), Renilla Luciferase mRNA (Luciferase mRNA), Vaginal Luciferase 5 mRNA, or any combination thereof. For example, GFP mRNA can be fused with an mRNA that encodes a protein that can be secreted to facilitate confirmation of the location of mRNA in target cells that will act as a deposit for protein production. As used herein, the terms "transfect" or "transfection" mean the intracellular introduction of an mRNA into a cell, or preferably into a target cell. The introduced mRNA can be stable or transiently maintained in the target cell. The term "transfection efficiency" refers to the relative amount of mRNA taken by the target cell that is subject to transfection. In practice, transfection efficiency is estimated by the amount of a reporter nucleic acid product expressed by the target cells after transfection. Preferred embodiments include compositions with high transfection efficiencies and, in particular, those compositions that minimize the adverse effects that are mediated by transfection of non-target cells. Compositions of the present invention that demonstrate high transfection efficacy improve the likelihood that appropriate dosages of mRNA will be delivered to the target cell, while minimizing potential systemic adverse effects. In one embodiment of the present invention, the transfer vehicles of the present invention are capable of delivering large sequences of rnRNA (for example, mRNA of at least lkDa, 1.5kDa, 2 kDa, 2.5kDa, 5klja, IOkDa, 12kDa, 15kDa , 20klja, 25kDa, 30kDa, or more). The mRNA can be formulated with one or more acceptable reagents, which provide a vehicle for delivering that mRNA to target cells. Appropriate reagents are generally selected for a number of factors, which include, but are not limited to, the biological or chemical properties of the mRNA, the desired route of administration, the anticipated biological environment to which that mRNA will be exposed, and the specific properties of desired target cells. In some embodiments, transfer vehicles, such as liposomes, encapsulate the rnRNA without compromising biological activity. In some embodiments, the transfer vehicle demonstrates preferential and / or substantial binding to a target cell over non-target cells. In a preferred embodiment, the transfer vehicle distributes its content to the target cell so that the mRNA is distributed to the appropriate subcellular compartment, such as the cytoplasm. Transfer Vehicle In the embodiments, the transfer vehicle in the compositions of the invention is a liposomal transfer vehicle, for example, a lipid nanoparticle. In one embodiment, the transfer vehicle can be selected and / or prepared to optimize the distribution of mRNA to a target cell. For example, if the target cell is a hepatocyte, the properties of the transfer vehicle (for example, size, charge and / or pH) can be optimized to effectively deliver that transfer vehicle to the target cell, reduce immune clearance and / or promote retention in the target cell. Alternatively, if the target cell is the central nervous system (for example, the mRNA administered to treat neurodegenerative diseases can be targeted specifically to the brain or spinal tissue), the selection and preparation of the transfer vehicle should consider penetration and the retention within the blood-brain barrier and / or the use of alternative means of distribution directly from that transfer vehicle to that target cell. In one embodiment, the compositions of the present invention can be combined with agents that facilitate the transfer of exogenous mRNA (for example, agents that interrupt or improve the permeability of the blood-brain barrier and thereby improve the transfer of exogenous mRNA to the target cells). The use of liposomal transfer vehicles to facilitate the delivery of nucleic acids to target cells is contemplated by the present invention. Liposomes (for example, liposomal lipid nanoparticles) are generally useful in a variety of applications in research, industry and medicine, particularly for use as vehicles for transferring in vivo diagnostic or therapeutic compounds (Lasic, Trends Biotechnol., 16: 307-321, 1998; Drummond et al., Pharmacol. Rev., 51: 691-743, 1999) and are generally characterized as microscopic vesicles having an aqueous interior space isolated from the external medium by a membrane of one or more bilayers. Liposome bilayer membranes are usually formed by amphiphilic molecules, such as lipids of natural or synthetic origin that comprise spatially separate hydrophilic and hydrophobic domains (Lasic, Trends Biotechnol., 16: 307- 321, 1998). Liposome bilayer membranes can also be formed by amphiphilic polymers and surfactants (for example, polymerosomes, niosomes, etc.). In the context of the present invention, a liposomal transfer vehicle is normally used to transport mRNA to the target cell. For the purposes of the present invention, liposomal transfer vehicles are prepared to contain the desired nucleic acids. The process of incorporating a desired entity (e.g., a nucleic acid) into a liposome is often referred to as "loading" (Lasic, et al., FEBS Lett., 3 12: 255-258, 1992). The nucleic acids incorporated in the liposomes can be completely or partially located in the interior space of the liposome, within the liposome bilayer membrane, or associated with the outer surface of the liposome membrane. The incorporation of a nucleic acid into the liposomes is also referred to herein as "encapsulation", where the nucleic acid is entirely contained within the interior space of the liposome. The purpose of incorporating an mRNA into a transfer vehicle, such as a liposome, is often to protect nucleic acid from an environment that may contain enzymes or chemicals that degrade nucleic acids and / or systems or receptors that cause rapid excretion of nucleic acids. Accordingly, in a preferred embodiment of the present invention, the transfer vehicle selected is capable of improving the stability of the mRNA contained therein. The liposome can allow the encapsulated mRNA to reach the target cell and / or it can preferably allow the encapsulated mRNA to reach the target cell or alternatively limit the distribution of that mRNA to other sites or cells where the presence of the administered mRNA may be useless or undesirable. In addition, the incorporation of mRNA into a transfer vehicle, such as a cationic liposome, also facilitates the distribution of that mRNA in a target cell. Ideally, liposomal transfer vehicles are prepared to encapsulate one or more desired mRNA so that the compositions demonstrate high transfection efficiency and improved stability. While liposomes can facilitate the introduction of nucleic acids into target cells, the addition of polycations (eg, poly L-lysine and protamine) as a copolymer can facilitate, and in some cases, markedly improve the transfection efficiency of various types of cationic liposomes by 2—28 times in a number of cell lines in vitro and in vivo. (See N.J. Caplen, et al., Gene Ther. 1995; 2: 603; S. Li, et al., Gene Ther. 1997; 4, 891.) Lipid nanoparticles In a preferred embodiment of the present invention, the transfer vehicle is formulated as a lipid nanoparticle. As used in this document, the phrase WAI "lipid nanoparticle" refers to a transfer vehicle comprising one or more lipids (for example, cationic lipids, non-cationic lipids and PEG-modified lipids). Preferably, lipid nanoparticles are formulated to deliver one or more mRNA to one or more target cells. Examples of suitable lipids include, for example, phosphatidyl compounds (e.g., phosphatidylglycerol, phosphatidylcholine, phosphatidylserine, phosphatidylethanolamine, sphingolipids, cerebrosides and gangliosides). Also contemplated is the use of polymers as transfer vehicles, alone or in combination with other transfer vehicles. Suitable polymers can include, for example, polyacrylates, polyalkylacrylates, polylactide, polylactide-polyglycolide copolymers, polycaprolactones, dextran, albumin, gelatin, alginate, collagen, chitosan, cyclodextrins, dendrimers and polyethylene. In one embodiment, the transfer vehicle is selected based on its ability to facilitate the transfection of an mRNA into a target cell. The invention contemplates the use of lipid nanoparticles as transfer vehicles comprising a cationic lipid to encapsulate and / or improve the distribution of mRNA in the target cell that will act as a deposit for the production of protein. As used in this document, the phrase "cationic lipid" refers to a series of lipid species that carry a positive net charge at a selected pH, such as the physiological pH. The contemplated lipid nanoparticles can be prepared including mixtures of multicomponent lipids for different reasons, employing one or more cationic lipids, non-cationic lipids and PEG-modified lipids. Several cationic lipids have been described in the literature, many of which are available cornercially. Cationic lipids particularly suitable for use in the compositions and methods of the invention include those described in the international patent publication WO 2010/053572, incorporated herein by reference, and more particularly, C12-200 described in paragraph [00225] of WO 2010/053572. In certain embodiments, the compositions and methods of the invention employ lipid nanoparticles comprising an ionizable cationic lipid described in provisional patent application US 61 / 617,468, filed March 29, 2012 (incorporated herein by reference), such as, for example , (15Z, 18Z) -N, N-dimethyl-6- (9Z, 12Z) -octadeca-9,12-dien-1-yl) tetracosa-1 5,1 B-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) tetl "acosa-5,15,18-trien-1-amine (HGT5002). - In some embodiments, the cationic lipid N- [1- (2,3-dioleyloxy) propyl] -N, N, N-trimethylammonium chloride or "DOTMA" is used. (Felgner et al. (Proc. Nat'l Acad. Sci. 84, 7413 (1987); US Patent 4,897,355). DOTMA can be formulated 5 alone or can be combined with neutral lipid, dioleoylphosphatidyl-ethanolamine or "DOPE" or other cationic or non-cationic lipids in a liposomal transfer vehicle or a lipid nanoparticle and these liposomes can be used to improve distribution of nucleic acids in target cells. Other suitable cationic lipids include, for example, S-carboxispermylglycinadioctadecylamide or "DOGS," 10 2,3-dioleyloxy-N- [2 (spermina-carboxamido) ethyl] -N, N-dimethyl-1-propanamine or "DOSPA" ( Behr et al. Proc. Nat.'l Acad. Sci. 86, 6982 (1989); US Patent 5,171,678; US Patent 5,334,761), 1,2-Dioleoyl-3-Dimethylammonium-Propane or "DODAP" , 1,2- Dioleoyl-3-Trimethylammonium-Propane or "DOTAP". Cationic lipids contemplated also include 1,2-distearyloxy-N, N-dimethyl-3-aminopropane or "DSDMA", 1,2-15 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 or" DDAB ", N- (1,2-dimyristyloxyprop-3-yl) -N, N-dimethyl-N-hydroxyethyl ammonium bromide or" DMRIE " , 3-dimethylamino-2- (colest-5-en-3-beta-20 oxybutan-4-oxy) -1- (cys, cis-9,12-octadecadienoxy) propane or "CLinDMA", 2- [5 '- (colest-5-en-3-beta-oxy) -3' -oxapentoxy) -3-dimethyl ll- (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 25 or "DLincarbDAP", 1,2-Dilinoleoylcarbamyl-3-dimethylaminopropane or "DL inCDAP ", 2,2-dilinoleyl-4-dimethylaminomethyl- [1,3] -dioxolane or" DLin-K-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-l-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 30. (Heyes, J., et al., J Controlled Release 107: 276-287 (2005); Morrissey, DV., Et al., Nat. Biotechnol. 23 (8): 1003-1007 (2005); PCT Publication WO2005 / 12! 348A1). The use of cholesterol-based cationic lipids is also contemplated by the present invention. These cholesterol-based cationic lipids can be used , alone or in combination with other cationic or non-cationic lipids. Suitable cholesterol-based cationic lipids include, for example, DC-Chol (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 5 (1997); US Patent 5,744,335), or ICE. In addition, several reagents are commercially available to improve the effectiveness of transfection. Suitable examples include LIPOFECTIN (DOTMA: DOPE) (Invitrogen, Carlsbad, Calif.), LIPOFECTAMINE (DOSPA: DOPE) (Invitrogen), LIPOFECTAMINE2000. (Invitrogen), FUGENE, TRANSFECT (DOGS), and 10 EFFECTENE. Cationic lipids such as dialkylamino-based, imidazole-based, and guanidinium-based lipids are also contemplated. For example, certain embodiments are directed to a composition comprising one or more cationic lipids based on imidazole, for example, the cholesterol ester of imidazole 15 or lipid "ICE" (3S, IOR, 13R, 17R) -1O, 13-dimethyl -17 - ((R) -6-methylheptan-2-yl) -2, 3, 4, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17-tetradecahydro-1H-cyclopenta [a] phenantren-3-yl 3- (1H-imidazol-4-yl) propanoate, as represented by structure (I) below. In a preferred embodiment, a transfer vehicle for mRNA delivery may comprise one or more imidazole-based cationic lipids, for example, imidazole cholesterol ester 20 or "ICE" lipid (3S, IOR, 13R, 17R) -1O , 13-dimethyl-17 - ((R) -6- methylheptan-2-yl) -2, 3, 4, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17-tetradecahydro -lH- cyclopenta [a] phenantren-3-yl 3- (1H-imidazol-4-yl) propanoate, as represented by structure (1) · , 5cÂo'jir "(I) 25 Without wishing to be bound by a particular theory, it is believed that the ability to fuse the cationic lipid ICE based on imidazole is related to endosomal disruption that is facilitated by the imidazole group, which has a lower pKa compared to . traditional cationic lipids. Endosomal disruption, in turn, promotes osmotic edema and disruption of the liposomal membrane, followed by transfection or intracellular release of the nucleic acid content loaded into it in the target cell. Imidazole-based cationic lipids are also characterized by their reduced toxicity compared to other cationic lipids. Imidazole-based cationic lipids (eg, ICE) can be used as the only cationic lipid in the lipid nanoparticle, or alternatively, they can be combined with cationic lipids, non-cationic lipids and traditional PEG-modified lipids. The cationic lipid may comprise a molar ratio of about 1 ° / 0 to about 9 ° / 0, 10 about 2 ° / 0 to about 7 ° / 0, about 5 ° / 6 to about 5 ° / o about 10 ° 4 to about 40 ° 4 of the total lipid present in the transfer vehicle, or preferably about 20 ° 4 to about 70 ° 4 of the total lipid present in the transfer vehicle. Likewise, certain modalities are directed to lipid nanoparticles comprising the cationic lipid HGT4003 2 - ((2,3-Bis ((9Z, 12Z) -octadeca-9,12-15 dien-l-yloxy) propi!) Disulfanil ) -N, N-dimethylethanamine, as represented by structure (II) below, and as further described in US Provisional Application No: 61 / 494,745, deposited on June 8, 2011, the entirety of which is incorporated in this document as a reference in its entirety: ! iN ~~ S- s "Y" o " The "" (I) 20 In other embodiments, the compositions and methods described in this document are directed to lipid nanoparticles comprising one or more cleavable lipids, such as, for example, one or more cationic lipids or compounds that comprise a cleavable functional group disulfide (SS) (for example, HGT4001, HGT4002, HGT4003, HGT4004 and HGT4005) as further described in Provisional Order 25 US 61 / 494,745, the complete teachings of which are incorporated into this document as a reference in their entirety. The use of phospholipids modified with polyethylene glycol (PEG) and lipids derived as derived ceramides (PEG-CER), including N-Octanoyl-Sphingosine-l- [Succinyl (Methoxy Polyethylene Glycol) -2000] (C8 PEG-2000 ceramide) is also contemplated by the present invention, alone or preferably in combination with other lipids together which comprise the transfer vehicle (for example, a lipid nanoparticle). PEG-modified lipids contemplated include, among others, a polyethylene glycol chain of up to 5 kDa in length covalently linked to a lipid with C6-C20 long alkyl chains. The addition of these components can prevent complex aggregation and can also provide a means to increase circulation life and increase the distribution of the lipid-nucleic acid composition to the target cell, (Klibanov et al. (1990) FEBS Letters, 268 (1): 235-237), or can be selected to quickly change the formulation in vivo (see US Patent 5,885,613). Particularly useful exchangeable lipids are PEG-ceramides having shorter acyl chains (for example, C14 or C18). The PEG-modified phospholipids and lipids derived from the present invention may comprise a molar ratio of about 0 ° / 0 to about 20 ° 4, about 0.5 ° 4 to about 20 ° 4, about i ° / oa about 15 ° 4, about 4 ° 4 to about 10 ° 4, or about 2 ° '6 of the total lipids present in the liposomal transfer vehicle. The present invention also contemplates the use of non-cationic lipids. As used in this document, the phrase "non-cationic lipid" refers to any neutral, zwitterionic or anionic lipid. As used in this document, the phrase "anionic lipid" refers to a series of lipid species that carry a negative net charge at a selected pH, such as the physiological pH. non-cationic lipids include, among others, distearoylphosphatidylcholine (DSPC), dioleoyl phosphatidylcholine (DOPC), dipalmitoylphosphatidylcholine (DPPC), dioleoylphosphatidylglycerol (DOPG), dipalmitoylphosphatidylglycerol (DPPG), dioleoyl phosphatidylethanolamine (DOPE), palmitoyloleoylphosphatidylcholine (POPC), palmitoiloleoil phosphatidylethanolamine (POPE) dioleoyl-phosphatidylethanolamine 4 {N-maleimidomethyl) -cyclohexane-1-carboxylate (DOPE-mal), dipalmitoyl phosphatidyl ethanolamine (DPPE), dimyristoylphosphethanolamine (DMPE), distearoyl-phosphatidyl-ethanolamine (16PE-16, 16PE-16, 16 -O-dimethyl PE, 18-1- trans PE, 1-stearoyl-2-oleoyl-phosphatidiethanolamine (SOPE), cholesterol, or a mixture thereof. These non-cationic lipids can be used alone, but are preferably used in combination with other excipients, for example, cationic lipids. When used in combination with a cationic lipid, the non-cationic lipid may comprise a molar ratio of 5 ° to 6 ° to about 90 ° 4, or preferably about 10 ° / 0 to about 70 ° 4 of the total lipid present in the transfer vehicle. Preferably, the transfer vehicle (e.g., a lipid nanoparticle) is prepared by combining various lipid and / or polymer components. For example, a transfer vehicle can be prepared using C12-200, DOPE, chol, DMG-PEG2K in a 40: 30: 25: 5 molar ratio, or DODAP, DOPE, cholesterol, DMG-5 PEG2K in a molar ratio of 18: 56: 20: 6, or HGT5000, DOPE, chol, DMG-PEG2K in a 40: 20: 35: 5 molar ratio, or HGT5001, DOPE, chol, DMG-PEG2K in a 40:20 molar ratio : 35: 5. The selection of cationic lipids, non-cationic lipids and / or PEG-modified lipids that comprise the lipid nanoparticle, as well as the relative molar ratio of these lipids to the other, is based on the characteristics of the selected lipids, the nature of the desired target cells, characteristics of the mRNA to be distributed. Additional considerations include, for example, the saturation of the alkyl chain, as well as the size, charge, pH, pKa, spindle capacity and toxicity of the selected lipids. Thus, the molar ratios can be adjusted accordingly. For example, in modalities, the percentage of cationic lipids in the lipid nanoparticle may be greater than greater than 10 ° / o, greater than 20 ° 4, greater than 30%, greater than 40 ° 4, greater than 50 ° 4, greater than 60 ° 4, or greater than 70%. The percentage of non-cationic lipid in the lipid nanoparticle can be greater than 5 ° / 0, greater than 10 ° / o, greater than 20 ° 4, greater than 30 ° 4, or greater than 4 ° ° / o. The percentage of cholesterol in the lipid nanoparticle can be greater than 10%, greater than 20 ° 4, greater than 3 ° ° / o, or greater than 4 ° ° / o. The percentage of PEG-modified lipid in the lipid nanoparticle can be greater than 1 ° / 0, greater than 2 ° / 0, greater than 5%, greater than 10 ° / ,, or greater than 20 ° 4, in certain preferred embodiments , the lipid nanoparticles of the invention comprise at least one of the following cationic lipids: Cl2-200, DLin-KC2-DMA, DODAP, HGT4003, ICE, HGT5000, or HGT5001. In the embodiments, the transfer vehicle comprises cholesterol and / or a PEG-modified lipid. In some cases, transfer vehicles comprise DMG-PEG2K. In certain fashion, the transfer vehicle comprises one of the following lipid formulations: C12-200, DOPE, chol, DMG-PEG2K; DODAP, DOPE, cholesterol, DMG-PEG2K; HGT5000, DOPE, chol, DMG-PEG2K, HGT5001, DOPE, chol, DMG-PEG2K. Liposomal transfer vehicles for use in the compositions of the invention can be prepared by various techniques that are currently known in the art. Multilamellar vesicles (MLV) can be prepared by conventional techniques, for example, by depositing a selected lipid on the inner wall of an appropriate container or bottle by dissolving the lipids in an appropriate solvent and then evaporating the solvent to leave a thin film inside the bottle or by spray drying. An aqueous phase can then be added to the flask with a vortex motion, which results in the formation of MLVS. Unilamellar vesicles 5 (ULV) can be formed by homogenization, sonication or extrusion of multilamellar vesicles. In addition, unilamellar vesicles can be formed by detergent removal techniques. In certain embodiments of this invention, the compositions of the present invention comprise a transfer vehicle in which the mRNA is associated on the surface of the transfer vehicle and encapsulated within the same transfer vehicle. For example, during the preparation of the compositions of the present invention, cationic liposomal transfer vehicles can associate the mRNA through electrostatic interactions. In certain embodiments, the compositions of the invention can be loaded with diagnostic radionuclides, fluorescent materials or other materials that are detectable in in vitro and in vivo applications. For example, diagnostic materials suitable for use in the present invention may include Rhodamine-dioleoylphosphatidylethanolamin (Rh-PE), Green Fluorescent Protein mRNA (GFP mRNA), Renilla Luciferase mRNA and Firefly Luciferase mRNA. The selection of the appropriate size of a liposomal transfer vehicle should take into account the location of the target cell or tissue and, to some extent, the application for which the liposome is being prepared. In some embodiments, it may be desirable to limit the transfection of mRNA to certain cells or tissues. For example, to target hepatocytes, a liposomal transfer vehicle can be prepared in size so that its dimensions are smaller than the fenestrations of the hepatic sinusoids of the endothelial lining layer in the liver; in this sense, the liposomal transfer vehicle can quickly penetrate these endothelial fenestrations to reach the target hepatocytes. Alternatively, a liposomal transfer vehicle can be prepared in size so that the dimensions of the liposome are of sufficient diameter to limit or expressly prevent delivery to certain cells or tissues. For example, a liposomal transfer vehicle can be prepared in size so that its dimensions are larger than the fenestrations of the hepatic sinusoids of the endothelial lining layer, thus limit the distribution of the liposomal transfer vehicle to hepatocytes. Generally, the size of the transfer vehicle is within the range of about 25 to 250 nm, preferably less than about 250nm, 175nm, 150nm, 125nm, 100nm, 75nm, 50nm, 25nm or 1Onm. 5 A variety of alternative methods known in the art are available for sizing a population of liposomal transfer vehicles. One of these design methods is described in US Patent 4,737,323, which is not included in this document as a reference. The sonication of a liposome suspension by bath or probe sonication produces a progressive reduction in size for small ULV less than about 0.05 microns in diameter. Homogenization is another method that relies on shear energy to fragment large liposomes into smaller ones. In a typical homogenization procedure, MLV are recirculated through a standard emulsion homogenizer until selected sizes of liposomes, usually between about 0.1 and 0.5 microns, are observed. The size of the liposome vesicles can be determined by quasi-electric light scattering (QELS) as described in Bloomfield, Ann. Rev. Biophys. Bioeng., 10: 421-450 (1981), incorporated herein by reference. The diameter of the average liposome can be reduced by sonication of formed liposomes. Intermittent sonication cycles can be alternated with QELS assessment to guide efficient liposome synthesis. Target Cell As used herein, the term "target cell" refers to a cell or tissue to which a composition of the invention is directed. In some embodiments, the target cells are deficient in a protein or enzyme of interest. For example, where it is desired to deliver a nucleic acid to a hepatocyte, the hepatocyte represents the target cell. In some embodiments, the compositions of the invention transfect target cells in a discriminatory manner (i.e., they do not transfect non-target cells). The compositions of the invention can also be prepared to preferentially target a variety of target cells, which include, but are not limited to, hepatocytes, epithelial cells, hematopoietic cells, epithelial cells, endothelial cells, lung cells, bone cells, stem cells, mesenchymal cells, neural cells (eg, meninges, astrocytes, motor neurons, dorsal root ganglion cells and the anterior horn motor neurons), photoreceptor cells (eg cones and rods), retinal pigment epithelial cells, secretory cells , cardiac cells, adipocytes, vascular smooth muscle cells, cardiomyocytes, skeletal muscle cells, beta cells, pituitary cells, synovial lining cells, ovarian cells, testicular cells, fibroblasts, B cells, T cells, reticulocytes, leukocytes, granulocytes and cells tumoral. The compositions of the invention can be prepared to deliver 5 preferentially to target cells such as the heart, lungs, kidneys, liver and spleen. In some embodiments, the compositions of the invention deliver to liver cells to facilitate delivery and subsequent expression of the mRNA comprised therein by liver cells (e.g., hepatocytes). Target hepatocytes can function as a biological "reservoir" or "deposit" capable of producing and systemically excreting a functional protein or enzyme. Accordingly, in one embodiment of the invention, the liposomal transfer vehicle can be targeted to hepatocytes and / or preferentially deliver to liver cells after delivery. After transfection of the target hepatocytes, the mRNA loaded into the liposomal vehicle is translated and a functional protein product is produced, excreted and systemically distributed. In other embodiments, cells other than hepatocytes (eg, lung, spleen, heart, eye, or central nervous system cells) can serve as a storage site for protein production. In one embodiment, the compositions of the invention facilitate the endogenous production in one subject of one or more functional proteins and / or enzymes and, in particular, the production of proteins and / or enzymes that demonstrate less immunogenicity in relation to their homologously prepared counterparts. recombinant. In a preferred mode! of the present invention, transfer vehicles comprise mRNA that encodes a deficient protein or enzyme. By distributing these compositions to target tissues and subsequently transfecting these target cells, the exogenous mRNA loaded into the liposomal transfer vehicle (for example, a lipid nanoparticle) can be translated in vivo to produce a functional protein or enzyme encoded by the mRNA exogenously administered (for example, a protein or enzyme in which the subject is deficient). Accordingly, the compositions of the present invention exploit a subject's ability to translate the mRNA prepared exogenously or recombinantly to produce an endogenously translated protein or enzyme and thereby produce (and eventually excrete) a functional protein or enzyme. Expressed or translated proteins or enzymes can also be characterized by the inclusion in vivo of native post-translational modifications, which can often be absent in recombinantly prepared proteins or enzymes, thus further reducing the immunogenicity of the translated enzyme or protein. Administration of mRNA encoding a deficient protein or enzyme avoids the need to deliver nucleic acids to specific organelles within a 5 target cell (for example, mitochondria). Instead, after transfection of a target cell and distribution of the nucleic acids into the cytoplasm of the target cell, the mRNA content of a transfer vehicle can be translated and a protein or functional enzyme expressed. The present invention also contemplates the targeting of target cells and tissues by means of active and passive targeting. The passive targeting phenomenon explores the natural distribution patterns of an in vivo transfer vehicle without resorting to the use of excipients or additional means to improve the recognition of the transfer vehicle by target cells. For example, transfer vehicles that are subject to phagocytosis by cells of the reticulo-system endothelial cells are likely to accumulate in the liver or spleen and, consequently, can provide means to passively target the distribution of the compositions to these target cells. Alternatively, the present invention contemplates active targeting, which involves the use of additional excipients, referred to as "targeting binders" that can be attached (covalently or not covalently) to the transfer vehicle to encourage the location of that transfer vehicle in certain cells target or target tissues. For example, targeting can be mediated by the inclusion of one or more endogenous targeting ligands (for example, apolipoprotein E) in or on the transfer vehicle to encourage delivery to target cells or tissues. Recognition of the targeting ligand by target tissues actively facilitates tissue distribution and cell uptake of the transfer vehicle and / or its contents in target cells and tissues (for example, the inclusion of an apolipoprotein-E targeting ligand in or on the transfer vehicle encourages the recognition and attachment of the transfer vehicle to the endogenous low-density lipoprotein receptors expressed by hepatocytes). As provided in this document, the composition can comprise a linker capable of improving the affinity of the composition for the target cell. Targeting ligands can be attached to the outer lipid layer of the lipid during formulation or after formulation. Such methods are well known in the art. In addition, some lipid particle formulations may employ fusogenic polymers such as PEAA, hemagglutinin, other lipopeptides (see US Patent Application 08 / 835,281 and 60 / 083,294, which are incorporated herein by reference) and other features useful for in vivo distribution and / or intracellular. In some other embodiments, the compositions of the present invention demonstrate improved transfection efficiencies, and / or demonstrate better selectivity for cells or tissues of interest. Therefore, compositions are contemplated that comprise one or more ligands (for example, peptides, aptamers, oligonucleotides, a vitamin or other molecules) that are capable of improving the affinity of the compositions and their nucleic acid content for the target cells or tissues. Suitable binders can optionally be attached to the surface of the transfer vehicle. In some embodiments, the targeting binder may cover the surface of a transfer vehicle or be encapsulated within the transfer vehicle. Appropriate ligands are selected on the basis of their physical, chemical or biological properties (for example, selective affinity and / or recognition of target cell surface markers or characteristics) Specific target sites in the cell and their corresponding targeting ligands can vary widely. Appropriate targeting ligands are selected so that the unique characteristics of a target cell are explored, allowing the composition to discriminate between target and non-target cells. For example, compositions of the invention may include surface markers (for example, apolipoprotein-B or apolipoprotein-E) that selectively improve the recognition of, or affinity for, hepatocytes (for example, by receptor-mediated recognition, and binding to those markers of surface). In addition, the use of galactose as a targeting ligand would be expected to target the compositions of the present invention to parenchymal hepatocytes, or, alternatively, the use of mannose containing sugar residues as a targeting ligand would be expected to target the compositions of the present invention for liver endothelial cells (e.g., mannose containing sugar residues that can preferentially bind to the asialoglycoprotein receptor present in hepatocytes). (See Hillery AM, et al. "Drug Delivery and Targeting: For Pharmacists and Pharmaceutical Scientists" (2002) Taylor & Francis, Inc.). The presentation of such targeting ligands that have been conjugated to fractions present in the transfer vehicle (for example, a lipid nanoparticle), therefore, facilitates the recognition and absorption of the compositions of the present invention in target cells and tissues. Examples of suitable targeting linkers include one or more peptides, proteins, aptamers, vitamins and oligonucleotides. Application and Administration 5 As used in this document, the term "subject" refers to any animal (for example, a mammal), including, but not limited to, humans, non-human primates, rodents and the like, for which compositions and niethodes of the present invention are administered. Typically, the terms "subject" and "patient" are used interchangeably in this document in reference to a human subject. The compositions and methods of the invention provide mRNA delivery to treat a variety of disorders. In particular, the compositions and methods of the present invention are suitable for treating diseases or disorders related to deficiency of proteins and / or enzymes that are excreted or secreted by the target cell in the surrounding extracellular fluid (for example, inRNA encoding hormones and neurotransmitters ). In embodiments, the disease may involve a defect or deficiency in a secreted protein (for example, Fabry's disease, or ALS). In certain embodiments, the disease may not be caused by a defect or deficit in a secreted protein, but it may benefit from providing a secreted protein. For example, the symptoms of a disease can be ameliorated by providing the compositions of the invention (for example, cystic fibrosis). Disorders for which the present invention is useful include, among others, disorders, such as Huntington's disease; Parkinson's disease; muscular dystrophies (such as Duchenne and Becker); hemophilic diseases (such as, for example, hemophilia B (FIX), hemophilia A (FVIII); SMNI-related spinal muscular atrophy (SMA); amyotrophic lateral sclerosis (ALS); GALT-related galactosemia; cystic fibrosis (CF); related disorders with SLC3A1, including cystinuria; COL4A5-related disorders, including Alport's syndrome; galactocerebrosidase deficiencies; X-linked adrenoleukodystrophy; Friedreich's ataxia; Pelizaeus-Merzbacher disease; TSCL and TSC2-related tuberous sclerosis; (MPS IIIB); CTNS-related cystinosis; FMRI-related disorders including fragile X syndrome, fragile X-associated tremor / ataxia syndrome and premature fragile X-ovarian failure syndrome; Prader-Willi syndrome; hemorrhagic telangiectasia hereditary disease (TA); Niemann-Pick disease type Cl; diseases related to lipofuscinosis . neuronal ceroid, including juvenile neuronal ceroid lipofuscinosis (JNCL), juvenile Batten disease, Santavuori-Haltia disease, elschowsky Jansky-Bi disease and PTT-I and TPPl deficiencies; ataxia of EIF2B1, EIF2B2, EIF2B3, EIF2B and EIF2B5 with hypomyelination / white matter evanescence of the central nervous system; Episodic ataxia Type 2 related to CACNAIA and CACNB4; MECP2-related disorders including Classical Rett Syndrome, MECP2-related Severe Neonatal Encephalopathy and PPM-X syndrome; Atypical Rett Syndrome related to CDKL5; Kennedy Disease (SBMA); Autosomal dominant cerebral arteriopathy related to Notch-3 with subcortical infarctions and 10 leukoencephalopathy (CADASIL); seizure disorders related to SCNIA and SCNIB; Polymerase G related disorders which include Alpers-Huttenlocher syndrome, POLG-related sensory ataxic neuropathy, dysarthria and ophthalmoplegia and ophthalmoplegia and progressive autosomal dominant and recessive internal with deletions of mitochondrial DNA; X-linked adrenal hypoplasia; X-linked agammaglobulinemia; Wilson's disease; and Fabry's disease. In one embodiment, the nucleic acids and, in particular, the mRNA of the invention can encode proteins or functional enzymes that are secreted in the extracellular space. For example, secreted proteins include clotting factors, components of the co-complement pathway, cytokines, chemokines, chemoattracts, protein honnons (eg, EGF, PDF), serum protein components 20, antibodies, secretible toll receptors, and others. In some embodiments, the compositions of the present invention may include mRNA encoding erythropoietin, al-antitrypsin, N carboxypeptidase or human growth hormone. In terms of niodalities, the invention encodes a secreted protein that is composed of sub-elevations that are encoded by more than one gene. For example, the secreted protein 25 can be a heterodinier, where each chain or subunit is encoded by a separate gene. It is possible that more than one mRNA molecule is distributed in the transfer vehicle and the mRNA encodes the separate subunit of the secreted protein. Alternatively, a single mRNA can be designed to encode the ends of a subunit (for example, in the case of a single chain Fv antibody). In certain embodiments, separate mRNA molecules that encode the individual subunits can be administered in separate transfer vehicles. In an innodality, niRNA can encode whole antibodies (both heavy and light chains of the variable and constant regions) or antibody groups (for example, Fab, Fv or a single chain Fv (ScFv) to confer immunity to a subject. While one embodiment of the present invention relates to methods and compositions useful for imparting immunity to a subject (for example, by translating the mRNA that encodes functional anti-antibodies), the inventions disclosed in this document and discussed here are widely applicable. In an alternative embodiment, the compositions of the present invention encode antibodies that can be used to transiently or chronically affect a functional response in subjects. For example, the mRNA of the present invention can encode a functional monoclonal or polyclonal antibody, which after translation and secretion of the target cell can be useful for targeting and / or inactivating a biological target (for example, a stimulating cytokine as a tumor necrosis factor). Likewise, the mRNA nucleic acids of the present invention can encode, for example, functional antinephritis factor antibodies useful for treating type II membranoproliferative glomerulonehitis or acute haemolytic uremic syndrome, or alternatively they can encode the antivascular endothelial growth factor (VEGF) antibodies ) useful for treating VEGF-mediated diseases, such as cancer. In other embodiments, the secreted protein is a cytokine or other secreted protein composed of more than one subunit (for example, IL-12, or IL-23). The compositions of the invention can be administered to a subject. In some embodiments, the composition is formulated in combination with one or more nucleic acids, 20 additional carriers, targeting binders or stabilizing reagents, or in pharmacological compositions, where it is mixed with appropriate excipients. For example, in one embodiment, the compositions of the invention can be prepared to deliver mRNA coke that encodes two or more distinct proteins or enzymes. Techniques for drug formulation and administration can be found in "Remington's 25 Pharmaceutical Sciences," Mack Publishing Co., Easton, Pa., Latest edition. A wide variety of molecules that can have therapeutic or pharmaceutical effects can be delivered to target cells using compositions and methods of the invention. The molecules can be organic or inorganic. Organic molecules can be peptides, proteins, carbohydrates, lipids, sterols, nucleic acids (including 30 peptide nucleic acids) or any combination thereof. A formulation for delivery to target cells can comprise more than one type of molecule, for example, two different nucleotide sequences, or a protein, an enzyme or a steroid. The compositions of the present invention can be administered and dosed in accordance with current medical practice, taking into account the subject's clinical condition, the location and mode of administration, the administration schedule, the age, sex, body weight of the subject and others relevant factors for clinical specialists in the technique. The "efficient amount" for the purposes of this document can be determined by these 5 relevant considerations as they are known to those skilled in the art of experimental clinical research, pharmacological, clinical and medical techniques. In some embodiments, the amount administered is efficient to achieve at least some stabilization, improvement or elimination of symptoms and other indicators that are selected as appropriate measures of progress, regression or improvement of the disease by those skilled in the art. For example, an appropriate amount and dosage schedule is what causes at least the production of transient protein. Suitable routes of administration include, for example, oral, rectal, vaginal, transmucosal, pulmonary including intratracheal or inhaled or intestinal administration; parenteral distribution, including intramuscular, subcutaneous, intramedullary injections, as well as direct intrathecal, intraventricular, intravenous, intraperitoneal, intranasal or intraocular injections. Alternatively, the compositions of the invention can be administered locally, rather than systemically, for example, by injecting the pharmaceutical composition directly into a target tissue, preferably in a sustained release formulation. Local distribution can be affected in several 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); the compositions of the present invention can be injected into the site of injury, disease or pain, for example; the compositions can be supplied in tablets for oral, tracheal, or esophageal application; they can be supplied as a liquid, tablet or capsule for administration to the stomach or intestine, they can be supplied as a suppository for rectal or vaginal application; or they can be distributed to the eye through the use of creams, eye drops or even injection. Formulations containing compositions of the present invention complexed with molecules or therapeutic binders can even be surgically administered, for example in combination with a polymer or other structure or substance that can allow the compositions to diffuse from the implantation site to the surrounding cells. Alternatively, they can be applied surgically without the use of polymers or supports. In one embodiment, the compositions of the invention are formulated in such a way that they are suitable for prolonged release of the mRNA contained therein. Such extended-release compositions can be conveniently administered to a subject at extended dosage 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, every ten days, every two weeks, every three weeks, or more preferably every four weeks, once. per month, every six weeks, every eight weeks, every two months, every three months, every four months, every six months, every eight months, every nine months or annually. Liposomal compositions and vehicles that are formulated for depot administration (for example, intramuscularly, subcutaneously, intravitreally) to deliver or release an mRNA over long periods of time are also contemplated. Preferably, the extended release media used are combined with modifications made to the mRNA to improve stability. Also included in this document are lyophilized pharmaceutical compositions comprising one or more of the liposome nanoparticles disclosed in this document and related methods for using these lyophilized compositions as disclosed, for example, in Provisional Application US 61 / 494,882, filed on June 8, 2011, whose teachings are incorporated in this document as a reference in their entirety. For example, lyophilized pharmaceutical compositions according to the invention can 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 (for example, an intradermal dosage form such as a disc, rod or membrane) and administered in such a way that the dosage form is rehydrated over time in vivo by colloidal fluids. of the individual. While certain compounds, compositions and methods of the present invention have been specifically described 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. Each of the publications, reference materials, membership numbers and the like referenced in this document to describe the background of the invention and provide additional details about its practice are hereby incorporated by reference in their entirety. The articles "one" and "one" as used in this document in the specification and in the claims, unless clearly indicated to the contrary, must be understood to include the referring plurals. Claims or descriptions that include "or" between one or more members of a group are considered satisfied if one, more than one or all 5 members of the group are present in, employed in, or otherwise relevant to a particular product or process unless otherwise indicated or otherwise evident in the context. The invention includes modalities in which exactly one member of the group is present in, employed in or otherwise relevant to a particular product or process. The invention also includes modalities in which more than one, or the entire group is present in, employed in or otherwise relevant to a particular product or process. In addition, it should be understood that the invention includes all variations, combinations, and permutations in which one or more limitations, elements, clauses, descriptive terms, etc., of one or more of the listed claims is introduced in another claim depending on it basic claim (or, as relevant, any other claim) unless otherwise indicated or unless it is evident to a person skilled in the art that a contradiction or inconsistency arises. When elements are presented as lists, (for example, in the Markush group or similar format) it must be understood that each subgroup of the elements is also revealed, and any elements can be removed from the group. It is to be understood that, in general, where the invention, or aspects of the invention, are / are said to comprise elements, particular features, etc., certain embodiments of the invention or aspects of the invention consist, or consist essentially of, such elements, characteristics , etc. For the sake of simplicity, these modalities have not in all cases been specifically defined in so many words in this document. It should also be understood that any form or aspect of the invention can be explicitly excluded from the claims, regardless of whether the specific exclusion is recited in the specification. Publications and other reference materials referenced in this document to describe the background of the invention and provide additional details about its practice are hereby incorporated by reference. EXAMPLES Example 1: Protein Production Deposit through Intravenous Distribution of the RNA Polynucleotide Compositions Human erythropoietin (EPO) (SEQ ID NO: 3; FIG. 3), human alpha-galactosidase (GLA) (SEQ ID NO: 4; FIG. 4), human alpha-1 antitrypsin (AIAT) (SEQ ID NO: 5 ; FIG. 5), and human Factor IX (FIX) (SEQ ID NO: 6; FIG. 6) were synthesized by in vitro transcription from a plasmid DNA model encoding the gene, which was followed 5 by the addition of a 5 'cap structure (Capl) (Fechter & Brownlee, J Gen. Viroio © '86: 1239-1249 (2005)) and a po11 (A) 3' tail of approximately 200 nucleotides in length as determined by gel electrophoresis. The 5 'and 3' untranslated regions were present in each mRNA product in the following examples and are defined by SEQ ID NOs: 1 and 2 (FIG. 1 and FIG. 2) respectively. Nanoparticle Formulations of Llí'ídeo Formulation 1. ' Aliquots of 50 mg / mL ethanol solutions of C12-200, DOPE, Chol and DMG-PEG2K (40: 30: 25: 5) were mixed and diluted with ethanol to a final volume of 3 mL. Separately, a buffered aqueous solution (10 mM citrate / 150 mM NaCl, pH 4.5) of the mRNA was prepared from a 1 mg / mL stock. The lipid solution was injected quickly into the aqueous mRNA solution and stirred to generate a final suspension in 20% ethanol. The resulting nanoparticle suspension was filtered, diafiltered with 1x PBS (pH 7.4), concentrated and stored at 2-8 ° C. Formulation 2. ' Aliquots of the 50 mg / inL ethanol solutions of DODAP, DOPE, cholesterol and DMG-PEG2K (18: 56: 20: 6) were mixed and diluted with ethanol to a final volume of 3 mL. Separately, a buffered aqueous solution (10 mM citrate / 150 inM NaCl, pH 4.5) of EPO mRNA was prepared from a stock of 1 mg / ml. The lipid solution was injected quickly into the aqueous mRNA solution and stirred to generate a final suspension in 20 ° 4 of ethanol. The resulting nanoparticle suspension was filtered, diafiltered with 1x PBS (pH 7.4), concentrated and stored at 2-8 ° C. Final concentration = 1.35 mg / ml EPO mRNA (encapsulated). Za ,, = 75.9 nm (Dv (50) = 57.3 nni; Dv (90) = 92.1 nm). Formulation 3: Aliquots of the 50 mg / mL ethanol solutions of HGT4003, DOPE, cholesterol and DMG-PEG2K (50: 25: 20: 5) were mixed and diluted with ethanol to a final volume of 3 mL. Separately, a buffered aqueous solution (10 mM citrate / 150 mM NaCl, pH 4.5) of the mRNA was prepared from a 1 mg / mL stock. The lipid solution was injected quickly into the aqueous mRNA solution and stirred to generate a final suspension in 20 ° 4 of ethanol. The resulting nanoparticle suspension was filtered, diafiltered with 1x PBS (pH 7.4), concentrated and stored at 2- 8 ° C. Formulation 4. ' Aliquots of the 50 mg / mL ethanol solutions of ICE, DOPE and DMG-PEG2K (70: 25: 5) were mixed and diluted with ethanol to a final volume of 3 mL. Separately, a buffered aqueous solution (10 mM citrate / 150 mM NaCl, 5 pH 4.5) of the mRNA was prepared from a stock of 1 ing / ml. The lipid solution was injected quickly into the aqueous mRNA solution and stirred to generate a final suspension in 20 ° 4 of ethanol. The resulting nanoparticle suspension was filtered, diafiltered with 1x PBS (pH 7.4), concentrated and stored at 2-8 ° C. Formulation 5. ' Aliquots of the 50 mg / mL ethanol solutions of HGT5000, DOPE, cholesterol and DMG-PEG2K (40: 20: 35: 5) were mixed and diluted with ethanol to a final volume of 3 ml. Separately, a buffered aqueous solution (10 mM citrate / 150 mM NaCl, pH 4.5) of the EPO mRNA was prepared from a stock of 1 mg / ml. The lipid solution was injected quickly into the aqueous mRNA solution and stirred to generate a final suspension in 2 ° / 0 ethanol. The resulting nanoparticle suspension was filtered, diafiltered with 1x PBS (pH 7.4), concentrated and stored at 2-8 ° C. Final concentration = 1.82 mg / ml EPO mRNA (encapsulated). Z, v, = 105.6 nm (Dv (50) = 53.7 nm; Dv (90) = 157 nm). Formulation 6. "Aliquots of the 50 mg / mL ethanol solutions of HGT5001, DOPE, cholesterol and DMG-PEG2K (40: 20: 35: 5) were mixed and diluted with ethanol to a final volume of 3 mL. Separately, a buffered aqueous solution (10 mM citrate / 150 mM NaCl, pH 4.5) of the EPO mRNA was prepared from a stock of 1 mg / ml. The lipid solution was injected quickly into the aqueous mRNA solution and stirred to generate a final suspension in 20 ° 4 of ethanol. The resulting nanoparticle suspension was filtered, diafiltered with 1x PBS (pH 7.4), concentrated and stored at 2-8 ° C. Analysis of proteins produced using nanoparticles loaded with mRNA distributed intravenously Injection Protocol Studies were performed using male CD-1 mice approximately 6-8 weeks old, at the beginning of each experiment, unless otherwise indicated. The samples were introduced by a single bolus tail-vein injection of a total equivalent dose of 30-200 micrograms of encapsulated mRNA. The mice were sacrificed and perfused with saline at the time points , designated. l Organ tissue isolation for analysis The liver and spleen of each mouse was collected, divided into three parts and stored in 10 ° 4 of neutral buffered formalin or quickly frozen and 5 stored at -80 ° C for analysis. Serum isolation for analysis All animals were sacrificed by CO2 asphyxiation 48 hours after dose administration (± 5 ° / 0) followed by thoracotomy and collection of terminal cardiac blood. Whole blood (maximum obtainable volume) was collected by cardiac puncture in animals sacrificed in serum separator tubes, coagulated at room temperature for at least 30 minutes, centrifuged at 22 ° C ± 5 ° C at 9300 g for 10 minutes and the extracted serum. For interim blood collections, approximately 40- 50µL of whole blood was collected through facial vein puncture or tail cut. The samples collected from untreated animals were used as a basis for comparison to study the animals. Analysis of the Enzyme-Linked Immunoabsorbent Assay (ELISA) EPO ELISA. ' Quantification of the EPO protein was performed following the procedures reported for the human EPO ELISA kit (Quantikine IVD, R&D Systems, Catalog # Dep-OO). The positive controls employed consisted of ultrapure recombinant human erythropoietin 20 protein and tissue culture grade (R&D Systems, Catalog # 286-EP and 287-TC, respectively). Detection was monitored through absorption (450 nm) on a Molecular Device Flex Station instrument. GLA ELISA. ' Standard ELISA procedures were followed employing sheep IgG G-188 anti-alpha-galactosidase as the capture antibody with rabbit IgG TK-88 25 anti-alpha-galactosidase as the second antibody (detection) (Shire Human Genetic Therapies). Goat anti-rabbit IgG conjugated to horseradish peroxidase (HRP) was used to activate the 3,3 ', 5,5'-tetrwnetylbenzidine (TMB) substrate solution. The reaction was stopped with 2N H2SO4 after 20 minutes. Detection was monitored through absorption (450 nm) on a Molecular Device Flex Station instrument. Serum from 30 untreated mice and human alpha-galactosidase protein were used as negative and positive controls, respectively. FLX ELISA. ' Quantification of the FIX protein was performed following the procedures reported for the human FIX ELISA kit (Assayhtlax, Assay Pro, Catalog # . EF1009-1). AIAT ELISA. ' Quantification of the AIAT protein was performed following the procedures reported for the human AIAT ELISA kit (Innovative Research, Catalog # IRAPKTO15). 5 Western Blot Analysis (EPO): Western blot analyzes were performed using an anti-hEPO antibody (R&D Systems # MAB2871) and ultrapure human EPO protein (R&D Systems # 286-EP) as the control. Results 10 The work described in this example demonstrates the use of lipid nanoparticles encapsulated with mRNA as a source of deposit for the production of protein. This deposit effect can be achieved at various locations within the body (ie, liver, kidneys, spleen and muscle). Measurement of the desired exogenous base protein derived from messenger RNA distributed through liposome nanoparticles was achieved and quantified, and protein secretion from a deposit using human erythropoietin inRNA (hEPO), human alpha-galactosidase (hGLA), antitrypsin human alpha-1 (hA1AT) and human Factor IX (hFIX) has been demonstrated. 1A. Results of the Production of EPO Protein Hyunana In Vivo The production of hEPO protein was demonstrated with the different formulations of 20 nanoparticles of lipid. Of the four different cationic lipid systems, the C12-200-based lipid nanoparticles produced the highest amount of hEPO protein after four hours after intravenous administration as measured by ELISA (FIG. 7). This formulation (Formulation 1) resulted in 18.3 µg / ml of hEPO protein secreted into the bloodstream. Normal HEPO protein levels in human serum are 25 3.3-16.6 mIU / mL (NCCLS Document C28-P; Vol. 12, No. 2). Based on a specific activity of 120,000 1U / mg of EPO protein, this generates an amount of 27.5-138 pg / mL of hEPO protein in normal human subjects. Therefore, a single 30 µg dose of a C12-200-based cationic lipid formulation encapsulating hEPO mRNA generated an increase in the respective protein of more than 100,000 times the physiological levels. Of the tested lipid systems, the DODAP-based lipid nanoparticle formulation was the least efficient. However, the observed amount of human EPO protein derived from distribution through a lipid nanoparticle based on DODAP encapsulating the EPO mRNA was 4.1 ng / mL, which is still 30 times greater than the normal physiological levels of the EPO protein (Table 1). Lipid Component EPO Protein Dose Increase in Cationic / soluble Human encapsulated Hematocrit mRNA (° / 0) (ug) (ng / mL) C12-200 30 18.306 15.0 HGT4003 150 164 0.0 ICE 100 56.2 0 , 0 DODAP 200 4.1 0.0 Table 1. Gross values of secreted IEPE protein for various cationic lipid-based nanoparticle systems as measured by 5 ELISA analysis (as described in FIG. 8). The doses are based on the encapsulated hEPO mRNA. The protein values are depicted as a nanogram of human EPO protein per milliliter of serum. Hematocrit changes are based on a comparison of pre-bleed (Day -1) and Day 10. In addition, the resulting protein was tested to determine whether it was active and functioning correctly. In the case of mRNA replacement therapy (MRT) employing hEPO mRNA, changes in hematocrit were monitored over a period of ten days for five different lipid nanoparticle formulations (FIG. 8, Table 1) to assess protein activity . During this time, two of the five formulations demonstrated an increase in hematocrit (> 15 ° 4), which is indicative of active hEPO protein being produced from these systems. In another experiment, changes in hematocrit were monitored over a period of 15 days (FIG. 9, Table 2). The lipid nanoparticle formulation (Formulation 1) was administered 'as a single dose of 30 µg, or as three smaller doses of 10 µg injected on day 1, day 3 and day 5. Likewise, Formulation 2 was administered as 3 doses of 50 µg on day 1, day 3 and day 5. C12-200 produced a significant increase in hematocrit. In general, an increase of up to - 25 ° 4 of change was observed, which is indicative of active hEPO protein being produced from these systems. Dose "I Average Hct Levels (° / 0) ± SEM Test Article (µg / animal) I Day -4 I Day 7 I DialO I Day 15" 30 (dose II 50.8 ± 1.8 I 58.3 ± 3.3 I 62.8 ± 1.3 I 59.9 ± 3.3 C12-200 single) 30 (in 3 C12-200 doses) I 52.2 ± 0.5 I 55.3 ± 2.3 I 63.3 ± 1.6 I 62.3 ± 1.9 150 (in 3 DODAP doses) 54.8 ± 1.7 53.5 ± 1.6 54.2 ± 3.3 54.0 ± 0.3 Hct = hematocrit; SEM = standard error of the mean. 'Blood samples were collected in hematocrit tubes without heparin. Table 2. Levels of hematocrit for each group during an observation period of 15 days (FIG. 9). The mice were treated with a single injection, or three injections, on alternate days. N = 4 mice per group. , IB. Results of Human GLA Protein Production In Vivo A second exogenous-based protein system was explored to demonstrate the "deposit effect" when employing mRNA-loaded lipid nanoparticles. The animals were injected intravenously with a single dose of 30 micrograms of human alpha-galactosidase mRNA (hGLA) encapsulated using a C12-200-based lipid nanoparticle system and sacrificed after six hours (Formulation /). The quantification of the secreted hGLA protein was performed by ELISA. Untreated mouse serum and human alpha-galactosidase protein were used as controls. The detection of human alpha-galactosidase protein was monitored over a period of 48 hours. Measurable levels of hGLA protein were observed over the course of the experiment with a maximum level of 2.0 µg / ml hGLA protein in six hours (FIG. 10). Table 3 lists the specific amounts of hGLA found in the serum. Normal activity in healthy human males has been reported to be approximately 3.05 nanomol / h / mL. Activity for alpha-galactosidase, a recombinant human alpha-galactosidase protein, 3.56 x 106 nanomol / hhhg. The analysis of these values generated an amount of approximately 856 pg / mL of hGLA protein in normal healthy male males. The amount of 2.0 ug / mL of hGLA protein observed after six hours when measuring the lipid nanoparticle loaded with hGLA mRNA is more than 2300 times greater than physiological levels = normal. In addition, after 48 hours, appreciable levels of hGLA proteins (86.2 ng / mL) can still be detected. This level is representative of almost 100 times greater amounts of hGLA protein compared to physiological amounts still present in 48 hours. Human GLA Protein Time I) After Administration (h) I Secreted (ng / mL)) 6 I 2,038 l 12) 1,815 i 24 l 414 I 48 I 86.2 Table 3. Gross values of hGLA protein secreted over time measured 5 by ELISA analysis (as depicted in FIG. 10). Values are depicted as nanogram of human hGLA protein per milliliter of serum. N = 4 mice per group. In addition, the half-life of alpha-galactosidase when administered at 0.2 mg / kg is approximately 108 minutes. The production of GLA protein through the "deposit effect" when administering lipid nanoparticles loaded with GLA mRNA shows a substantial increase in residence time in the blood compared to direct injection of the pure recombinant protein. As described above, significant amounts of protein are present after 48 hours. The activity profile of the α-galactosidase protein produced from lipid nanoparticles loaded with GLA mRNA was measured as a function of the metabolism of 4-methylumbelliferyl-a-D-galactopyranoside (4-MU-a-gal). As shown in FIG. 11, the protein produced from these nanoparticle systems is very active and reflective of the levels of available protein (FIG. 12, Table 3). AUC comparisons of hGLA production based on 'mRNA therapy versus enzyme replacement therapy (ERT) in mice and humans show an increase of 182 times and 30 times, respectively (Table 4). Article by! I Dose AUCinf Test I Description I (mg / kg) Cmax (U / niL) (hU / mL) n Transplant 0.2 3478 3683 11 Patient Protein cl- II Dialysis I 0.2 3887 3600 6 Fabry 'gai No ESRDb I 0.2 3710 4283 18 Protein ol- I GAL l Pure without I (MMI) I Thymus! 0.04 I 3807 I 797 I3! Mouse Protein o- I GAL I Pure without) (MM2)! Thymus I 0.04 I 3705 I 602 I3 a-GAL) Mouse mRNA I mouse I 0.95 I 5885 (Cen16h) 'I 109428 I6 Table 4. Comparison of C ,,,,, and AUCinf values in Fabry patients after IV dosage of 0.2 mg / kg of alpha-galactosidase (pharmacological dose) with the post-IV mice with alpha-galactosity and GLA mRNA. 'Data 5 was from a published article (Gregory M. Pastores et al. Safety and Pharmacokinetics of hGLA in patients with Fabry disease and end-stage renal disease. Nephrol Dial Transplant (2007) 22: 1920-1925. B Kidney disease in non-terminal stage 'A-galactosidase activity 6 hours after dosing (the shortest time point tested in the study). The ability of lipid nanoparticles with encapsulated mRNA to target Organs that can act as a deposit for the production of a desired protein has been demonstrated. The levels of secreted protein observed were several orders of magnitude above normal physiological levels. This "deposit effect" is repeatable. FIG. 12 again shows that robust protein production is observed after dosing wild-type mice (CD-1) with a single dose of 30 µg of hGLA mRNA loaded on C12-200-based lipid nanoparticles (Formulation 1). In this experiment, hGLA levels were assessed over a 72 hour period. A maximum average of 4.0 µg of human hGLA protein / mL of serum is detected six hours after administration. Based on a value of - 1 ng / mL of hGLA protein for normal physiological levels, hGLA MRT provides protein levels about 4000 times higher. As before, the hGLA protein could be detected within 48 hours of administration (FIG. 12). An analysis of the tissues isolated from this same experiment provided insight into the distribution of hGLA protein in mice treated with hGLA MRT (FIG. 13). Supraphysiological levels of hGLA protein were detected in the liver, spleen and kidneys of 5 all cannunds treated with a maximum observed between 12 and 24 hours after administration. Detectable levels of MRT-derived protein could be observed three days after a single injection of hGLA-loaded lipid nanoparticles. In addition, the production of hGLA after administration of C12-200 nanoparticles loaded with hGLA mRNA has been shown to exhibit a dose response in serum (FIG. 14A) as well as in the liver (FIG. 14B). An inherent feature of lipid nanoparticle-mediated mRNA replacement therapy would be the pharmacokinetic profile of the respective protein produced. For example, ERT-based treatment of mice using alpha-galactosidase results in a plasma half-life of approximately 100 minutes. In contrast, MRT-derived alpha-galactosidase has a residence time in the blood of approximately 72 hours with a peak time of 6 hours. This allows for much greater exposure for the organs to participate in the possible continuous uptake of the desired protein. A comparison of the PK profiles is shown in FIG. 15 and demonstrates the marked difference in clearance rates and, finally, a major change in the area under the curve (AUC) can be achieved through MRT-based treatment. In a separate experiment, hGLA MRT was applied to a mouse disease model, hGLA KO mouse (Fabry mice). A dose of 0.33 mg / kg of C12-200-based lipid nanoparticles loaded with hGLA mRNA (Formu / action 1) was administered to female KO mice as a single intravenous injection. Substantial amounts of MRT-derived hGLA protein were produced with a peak at 6 h (-560 ng / mL of serum) that is approximately 600 times greater than normal physiological levels. In addition, the hGLA protein was still detectable 72 hours after administration (FIG. 16). Quantification of MRT-derived GLA protein in vital Organs demonstrated substantial accumulation, as shown in FIG. 17. A comparison of the MRT-derived hGLA protein observed with the reported normal physiological levels that are found in the main Organs is plotted (normal levels plotted as dashed lines). While protein levels at 24 hours are greater than 72 hours after administration, the levels of hGLA protein detected in the liver, kidneys, spleen and heart of treated Fabry mice are equivalent to wild-type levels. For example, 3.1 ng hGLA protein / mg tissue was found in the kidneys of mice treated 3 days after a single MRT treatment. 5 In a later experiment, a comparison of ERT-based alpha-galactosidase treatment versus hGLA MRT-based treatment of male Fabry KO mice was performed. A single, intravenous dose of 1.0 mg / kg was administered for each therapy and the mice were sacrificed one week after administration. Serum levels of hGLA protein were monitored at 6 hr and 1 week after injection. Liver, kidneys, spleen and heart were analyzed for hGLA protein accumulation one week after administration. In addition to the biodistribution analyzes, a measure of effectiveness was determined by measuring reductions in globotrioasilceramide (Gb3) and smooth-Gb3 in the kidneys and heart. FIG. 18 shows serum levels of hGLA protein after treatment of lipid nanoparticles loaded with GLA mRNA and alpha-galactosidase (Forniulation 1) in male Fabry mice. Serum samples were analyzed at 6hr and 1 week after administration. A robust signal was detected for MRT-treated mice after 6 hours, with serum hGLA protein levels of -4.0 µg / mL. In contrast, there was no detectable alpha-galactosidase remaining in the bloodstream at this time. Fabry's mice in this experiment were sacrificed one week after the initial injection and the organs were collected and analyzed (liver, kidneys, spleen, heart). FIG. 19 shows a comparison of the human GLA protein found in each respective organ after the hGLA MRT or alpha-galactosidase ERT treatment. The levels correspond to the hGLA present one week after administration. The hGLA protein was detected in all organs analyzed. For example, MRT-treated mice resulted in hGLA protein accumulation in the kidney of 2.42 ng hGLA protein / mg protein, whereas alpha-galactosidase-treated mice had only residual levels (0.37 ng / mg protein) . This corresponds to a —6.5 times higher level of hGLA protein when treated with hGLA MRT. After heart analysis, 11.5 ng hGLA protein / mg protein was found for the MRT-treated cohort compared to just 1.0 ng / mg alpha-galactosidase protein. This corresponds to a greater than 11-fold accumulation in the heart of hGLA MRT treated mice compared to ERT-based therapies. In addition to the biodistribution analyzes conducted, efficacy assessments were determined by measuring the levels of globotrioasilceramide (Gb3) and smooth-Gb3 in the main organs. A direct comparison of the reduction of Gb3 after a single, intravenous treatment of 1.0 mg / kg GLA MRT compared to an equivalent dose alpha-5 galactosidase ERT-based therapy generated a considerable difference in Gb3 levels in the kidneys, as well as in the heart. For example, Gb3 levels for GLA MRT versus alpha-galactosidase generated reductions of 60.2 ° 4 vs 26.8 ° 4, respectively (FIG. 20). In addition, Gb3 levels in the heart were reduced by 92.1 ° 4 vs 66.9 ° 4 for MRT and alpha-galactosidase, respectively (FIG. 21). A second biomarker relevant for measuring effectiveness is smooth-Gb3. GLA MRT reduced lyso-Gb3 more efficiently than alpha-galactosidase also in the kidneys and heart (FIG. 20 and FIG. 21, respectively). In particular, the MRT-treated Fabry mice demonstrated reductions in smooth-Gb3 of 86.1 ° 4 and 87.9% in the kidneys and heart compared to mice treated with alpha-galactosidase generating a decrease of 47.8% and 61, 3 ° 4, respectively. The results for hGLA in C12-200-based lipid nanoparticles extend to other lipid nanoparticle formulations. For example, hGLA mRNA loaded on lipid nanoparticles based on HGT4003 (Source 3) or HGT5000 (Formulation 5) administered as a single dose IV results in the production of hGLA within 24 hours after administration (FIG. 22). The production of hGLA exhibited a dose response. Likewise, hGLA production was observed at 6 hours and 24 hours after administration of hGLA loaded mRNA on lipid nanoparticles based on HGT5001 (Formulation 6) administered as a single dose IV. The production of hGLA was observed in the serum (FIG. 23A), as well as in organs (FIG. 23B). In general, nRNA replacement therapy applied as a deposit for protein production produces large amounts of active protein, functionally therapeutic at supraphysiological levels. This method has been shown to generate a sustained circulation half-life of the desired protein and this MRT-derived protein is highly effective for therapy, as demonstrated with the enzyme alpha-galactosidase in Fabry mice. lC. Results of the Production of Human F / X Protein In Vivo The studies were performed by administering lipid nanoparticles loaded with Factor IX rnRNA (FIX) in wild type mice (CD-1) and determining the FIX protein that is secreted in the bloodstream. . After intravenous injection of a single dose of 30 µg of lipid nanoparticles loaded with F12 mRNA based on C12-200 (C12-200: DOPE: Chol: PEG in a ratio of 40: 30: 25: 5) (dose based on encapsulated mRNA) (Formulation 1), a production of 5 robust protein was observed (FIG. 24). A pharmacokinetic analysis over the 72 hour period showed that the MRT-derived FIX protein could be detected at all times tested (FIG. 24). The peak serum concentration was observed 24h after injection with a value of -3 ug (2995 ± 738 ng / mL) of protein FlX / mL of serum. This represents another successful example of the deposit effect. 1D. Results of Production of Human AIA T Protein In Vivo The studies were carried out by administering lipid nanoparticles loaded with alpha-l antitrypsin mRNA (AIAT) in wild-type mice (CD-l) and determining the AIAT protein that is secreted in the blood flow. After intravenous injection of a single dose of 30 µg of lipid nanoparticles loaded with C12-200-based AIAT mRNA (dose based on encapsulated mRNA) (Formulation 1), a robust protein production was observed (FIG. 25). As pictured in FIG. 25, detectable levels of human AIAT protein derived from AIAT MRT could be observed for a period of 24 hours after administration. A maximum serum level of -48 µg AIAT protein / mL of serum was detected! 2 hours after injection. Example 2: Deposit of Protein Production through Pulmonary Distribution of Polynucleotide Compositions Injection Protocol All studies were performed using female CD-1 or BALB / C mice approximately 7-10 weeks old at the beginning of each experiment. Test articles were introduced through a single intratracheal aerosol administration. The mice were sacrificed and perfused with saline at the designated time points. The lungs of each mouse were collected, divided into three parts, and stored in 10 ° 4 of neutral buffered formalin or quickly frozen and stored at -80 ° C for analysis. The serum was isolated as described in Example 1. EPO ELISA. ' as described in Example 1. Results The deposit effect can be achieved through pulmonary distribution (for example, intranasal, intratracheal, nebulization). The measurement of the desired exogenous base protein derived from the messenger RNA distributed through nanoparticle systems was achieved and quantified. The production of human EPO protein through lipid nanoparticles loaded with hEPO mRNA was tested in CD-1 mice through a single intratracheal administration (MicroSprayer®). Several formulations have been tested using various cationic lipids (Formulations /, 5, 6). All formulations resulted in high encapsulation of human EPO mRNA. After administration, animals were sacrificed six hours after administration and lungs, as well as serum, were collected. The human EPO protein was detected in the administration site (lungs) after treatment through aerosol delivery. Serum analysis six hours after administration showed detectable amounts of circulating hEPO protein. These data (shown in FIG. 26) demonstrate the ability of the lung to act as a "deposit" for the production (and secretion) of the IÍEPO protein.
权利要求:
Claims (34) [1] 1. Composition for use in the treatment of an individual having a defect or deficiency in a secreted polypeptide, characterized by the fact that it comprises (a) 5 at least one mRNA molecule, at least a portion of which encodes said functional secreted polypeptide ; and (b) a transfer vehicle comprising a lipid nanoparticle. [2] 2. Composition for use according to claim 1, characterized by the fact that the mRNA molecule encodes an enzyme that is abnormally deficient in an individual with a lysosomal storage disease. [3] Composition for use according to claim 1, characterized by the fact that the polypeptide encoded by the mRNA molecule is erythropoietin, α-galactosidase polypeptide, LDL receptor, Factor VIII, Factor IX, α-L-iduronidase, iduronate sulfatase, heparin-N-sulfatase, α-N-acetylglucosaminidase, galactose-6-sulfatase, β-galactosidase, lysosomal acid lipase or aryl sulfatase-A peptide. [4] Composition for use according to any one of claims 1 to 3, characterized by the fact that the mRNA molecule is unmodified or the mRNA molecule comprises: at least one modification that confers stability on the mRNA molecule; at least one modification that improves the stability of the mRNA with respect to its unmodified counterpart, optionally, wherein the modification comprises a modified nucleotide, optionally, where the modified nucleotide is pseudouridine; a modification of the 5 'untranslated region of said mRNA molecule, optionally, wherein said modification comprises the inclusion of a Cap1 structure; or a modification of the 3 'untranslated region of said mRNA molecule, optionally, wherein said modification comprises the insertion of a poly A tail; and / or where the mRNA molecule is at least 30 kDa. [5] Composition for use according to any one of claims 1 to 4, characterized in that it further comprises an agent to facilitate the transfer of the mRNA molecule to an intracellular compartment of a target cell, optionally, wherein said target cell is selected from the group consisting of hepatocytes, epithelial cells, hematopoietic cells, epithelial cells, endothelial cells, lung cells, bone cells, stem cells, mesenchymal cells, neuronal cells, cardiac cells, adipocytes, vascular smooth muscle cells, cardiomyocytes, skeletal muscle cells, beta cells, pituitary cells, synovial lining cells, ovary cells, testis cells, fibroblasts, B cells, T cells, reticulocytes, granulocytes, leukocytes and tumor cells. [6] Composition for use according to any one of claims 1 to 5, characterized by the fact that the lipid nanoparticle comprises: one or more cationic lipids; 5 one or more non-cationic lipids; one or more cholesterol-based lipids; and / or one or more PEG-modified lipids. [7] Composition for use according to any one of claims 1 to 6, characterized in that the lipid nanoparticle comprises C12-200; DLinKC2DMA, CHOL, DOPE, and DMG-PEG-2000; or C12-200, DOPE, CHOL, and DMGPEG2K. [8] Composition for use according to any one of claims 1 to 7, characterized in that the lipid comprises a cleavable lipid nanoparticle. [9] Composition for use according to any one of claims 1 to 8, characterized in that said composition is lyophilized. [10] Composition for use according to any one of claims 1 to 9, characterized in that said composition is a reconstituted lyophilized composition. [11] Composition for use according to any one of claims 1 to 10, characterized in that the composition is for intravenous or pulmonary administration. [12] Composition for use according to any one of claims 1 to 11, characterized in that the composition is for administration to the individual twice a week, once a week, every ten days, every two weeks, or triweekly. [13] Composition for use according to any one of claims 1 to 12, characterized in that the size of the transfer vehicle is within the range of about 25 to 250 nm. [14] Composition for use according to any one of claims 1 to 12, wherein the size of the transfer vehicle is less than 250 nm, 175 nm, 150 nm, 125 nm, 100 nm, 75 nm, 50 nm, 25 nm or 10 nm, optionally, where the lipid nanoparticle is less than about 100 nm in size. [15] Composition for use according to any one of claims 1 to 14, characterized by the fact that the polypeptide is expressed in at least one therapeutic level for more than one, more than four, more than six, more than 12, more than 24, more than 48 hours, or more than 72 hours after administration, optionally, where the level of secreted protein is detectable in 3 days, 4 days, 5 days, or a week or more after administration. [16] Composition for use according to any one of claims 1 to 15, characterized in that after administration of said composition to a subject, said mRNA is translated in vivo to produce said secreted functional polypeptide, and in which the secreted functional polypeptide is present at levels that are detectable in the serum at least one week after administration. [17] 17. Composition for use according to any of claims 1 to 16, characterized in that the administration of a composition is obtained by a route of administration selected from a group consisting of oral, rectal, vaginal, transmucosal, intestinal, intramuscular, subcutaneous, intramedullary, intrathecal, direct intraventricular, intravenous, intraperitoneal, intranasal and intraocular. [18] 18. Composition for use according to any one of claims 1 to 17, characterized in that the mRNA is administered to a target cell in vivo, optionally in which (i) the target cell is selected from the group consisting of hepatocytes, epithelial cells, hematopoietic cells, epithelial cells, endothelial cells, lung cells, bone cells, stem cells, mesenchymal cells, neuronal cells, cardiac cells, adipocytes, vascular smooth muscle cells, cardiomyocytes, skeletal muscle cells, beta cells, pituitary cells, synovial lining cells, ovary cells, testicular cells, fibroblasts, B cells, T cells, reticulocytes, granulocytes, leukocytes and tumor cells; (ii) the target cell is deficient in a protein or enzyme of interest; and / or (iii) the composition comprises a targeting ligand capable of increasing the affinity of the composition for one or more target cells, optionally, in which the targeting ligand is selected from the group consisting of apolipoprotein-B and apolipoprotein- E 5 and one or more target cells that express low density lipoprotein receptors. [19] 19. Composition for use in the treatment of an individual who has a defect or deficiency in an exogenous protein or is in need of said exogenous protein to target and / or inactivate a biological target, characterized by the fact that it comprises an mRNA that encodes said protein exogenous [20] 20. Composition for use according to claim 19, characterized in that it is administered via pulmonary administration to the individual so that administration of the composition results in the in vivo expression of the exogenous protein encoded by the mRNA in the individual's lung cells , in which pulmonary administration is achieved through intratracheal administration or nebulization. [21] 21. Composition for use according to claim 19 or 20, characterized in that the mRNA is: (i) encapsulated within a liposome, optionally, in which the liposome comprises one or more cationic lipids, one or more non-lipids cationic, one or more cholesterol-based lipids and one or more lipids modified with PEG; or (ii) complexed with a polymer. [22] 22. Composition for use according to claim 19 or 20, characterized in that the mRNA comprises a non-naturally occurring nucleotide, optionally, in which the non-naturally occurring nucleotide is pseudouridine. [23] 23. Composition for use according to claim 19 or 20, characterized in that the exogenous protein encoded by the mRNA is an enzyme, a hormone, a receptor or an antibody. [24] 24. Composition for use according to claim 19 or 20, characterized in that the exogenous protein is: (i) retained within the cytosol of the lung cells after expression; or (ii) secreted extracellularly after expression, optionally, in which the exogenous protein is distributed systemically. [25] 25. Composition for use according to claim 19 or 20, characterized in that the in vivo expression of the exogenous protein encoded by the mRNA is detectable at least 6 hours after administration. [26] 26. mRNA encoding a human alpha-galactosidase (hGLA) protein, characterized by the fact that it is for use in the treatment of Fabry disease, where the mRNA is administered intravenously or pulmonary administration in such a way that the level of hGLA protein in serum is increased for at least 72 hours, compared to the base level of hGLA in serum before treatment. [27] 27. mRNA for use according to claim 26, characterized in that the mRNA comprises SEQ ID 5 NO: 4. [28] 28. mRNA for use according to claim 26, characterized in that the mRNA is encapsulated within a liposome, optionally, wherein the 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. [29] 29. mRNA for use according to claim 26, characterized in that the mRNA comprises a non-naturally occurring nucleotide, optionally, in which the non-natural nucleotide is pseudouridine. [30] 30. mRNA for use according to claim 26, characterized in that the hGLA protein is: (i) secreted extracellularly after expression; (ii) systemically distributed; and / or (iii) detectable at least 6 hours after administration. [31] 31. mRNA for use according to claim 26, characterized by the fact that administration of the mRNA results in a reduction in the level of globotrioasilceramide (Gb3) in a human subject, compared to the level of Gb3 before treatment. [32] 32. Use of (a) at least one mRNA molecule, at least a portion of which encodes a secreted functional polypeptide; and (b) a transfer vehicle comprising a lipid nanoparticle, characterized in that it is for the preparation of a composition to treat a subject who has a defect or deficiency in said secreted polypeptide. [33] 33. Use of an mRNA that encodes for exogenous protein, characterized by the fact that it is for the preparation of a composition to treat a subject who has a defect or deficiency in said exogenous protein or is in need of said exogenous protein to target and / or inactivate a biological target. [34] 34. Use of an mRNA encoding a human alpha-galactosidase (hGLA) protein, characterized by the fact that it is for the preparation of a composition to treat Fabry's disease, in which the mRNA is administered intravenously or pulmonary administration in such a way that the level of hGLA protein in serum is increased for at least 72 hours, compared to the base level of hGLA in serum before treatment.
类似技术:
公开号 | 公开日 | 专利标题 US10507249B2|2019-12-17|Lipid nanoparticle compositions and methods for mRNA delivery
同族专利:
公开号 | 公开日 CA2838069A1|2012-12-13| US20190192689A1|2019-06-27| EP3586861A1|2020-01-01| SI2717893T1|2019-10-30| PE20140797A1|2014-07-11| NZ618275A|2016-11-25| US10507249B2|2019-12-17| CN111671918A|2020-09-18| US10238754B2|2019-03-26| CA3107288A1|2012-12-13| CN103906527A|2014-07-02| AU2017204509B2|2019-04-11| MX2013014419A|2014-01-23| CL2013003478A1|2014-07-04| AU2019201924A1|2019-04-11| ME03491B|2020-01-20| JP2017014278A|2017-01-19| LT2717893T|2019-08-12| HUE044277T2|2019-10-28| PL2717893T3|2019-12-31| JP6463810B2|2019-02-06| US20140294939A1|2014-10-02| WO2012170930A9|2014-01-03| EP2717893A1|2014-04-16| US20210060177A1|2021-03-04| IL280771D0|2021-04-29| MX367605B|2019-08-28| US20170239371A1|2017-08-24| RU2013154295A|2015-07-20| CA2838069C|2021-10-12| AU2012267531B2|2017-06-22| AU2019201924B2|2021-04-08| JP2014523411A|2014-09-11| RS59037B1|2019-08-30| CN111671919A|2020-09-18| US20190321489A1|2019-10-24| US9597413B2|2017-03-21| US10350303B1|2019-07-16| US20210300755A1|2021-09-30| US20200338214A1|2020-10-29| ES2740248T3|2020-02-05| US20220040330A1|2022-02-10| HRP20191338T1|2019-10-18| US11052159B2|2021-07-06| JP2017203045A|2017-11-16| US9308281B2|2016-04-12| EP2717893B1|2019-05-08| JP2020186266A|2020-11-19| JP6752299B2|2020-09-09| HK1199206A1|2015-06-26| US11185595B2|2021-11-30| MX2019010226A|2019-10-15| JP6372042B2|2018-08-15| IL229699A|2021-02-28| CN103906527B|2020-07-10| US20140294938A1|2014-10-02| US20200085973A1|2020-03-19| DK2717893T3|2019-07-22| WO2012170930A1|2012-12-13| IL229699D0|2014-01-30| AU2012267531A1|2013-12-19| US10413618B2|2019-09-17| TR201910686T4|2019-08-21| US10888626B2|2021-01-12| JP2019048900A|2019-03-28| PT2717893T|2019-08-20| JP6184945B2|2017-08-23| EP3586861B1|2022-02-09| KR102128248B1|2020-07-01| US20140206753A1|2014-07-24| KR20140044356A|2014-04-14| AU2017204509C1|2019-07-25| AU2017204509A1|2017-07-20| AU2021204597A1|2021-07-29| US20190175761A1|2019-06-13| KR20200084048A|2020-07-09|
引用文献:
公开号 | 申请日 | 公开日 | 申请人 | 专利标题 US2647121A|1951-02-02|1953-07-28|Ruth P Jacoby|Diamine-bis-acetamides| US2819718A|1953-07-16|1958-01-14|Isidore H Goldman|Drainage tube| US2717909A|1953-09-24|1955-09-13|Monsanto Chemicals|Hydroxyethyl-keryl-alkylene-ammonium compounds| US2844629A|1956-04-25|1958-07-22|American Home Prod|Fatty acid amides and derivatives thereof| US3096560A|1958-11-21|1963-07-09|William J Liebig|Process for synthetic vascular implants| GB1072118A|1962-12-01|1967-06-14|Sandoz Ag|Amides of aminopropionic acid| FR1378382A|1962-12-01|1964-11-13|Sandoz Sa|Amides of amino-propionic acid, usable in particular for the treatment of textile fibers| JPS5141663B1|1966-03-12|1976-11-11| JPS4822365B1|1968-10-25|1973-07-05| NL143127B|1969-02-04|1974-09-16|Rhone Poulenc Sa|REINFORCEMENT DEVICE FOR A DEFECTIVE HEART VALVE.| JPS4822365Y1|1969-11-28|1973-06-29| US3614954A|1970-02-09|1971-10-26|Medtronic Inc|Electronic standby defibrillator| US3614955A|1970-02-09|1971-10-26|Medtronic Inc|Standby defibrillator and method of operation| JPS5024216B1|1970-12-29|1975-08-14| JPS5024216Y1|1970-12-29|1975-07-21| JPS5012146Y2|1971-07-27|1975-04-15| JPS5123537Y2|1972-01-17|1976-06-17| US3945052A|1972-05-01|1976-03-23|Meadox Medicals, Inc.|Synthetic vascular graft and method for manufacturing the same| US3805301A|1972-07-28|1974-04-23|Meadox Medicals Inc|Tubular grafts having indicia thereon| JPS5210847Y2|1972-10-11|1977-03-09| JPS49127908A|1973-04-20|1974-12-07| JPS5624664B2|1973-06-28|1981-06-08| US4013507A|1973-09-18|1977-03-22|California Institute Of Technology|Ionene polymers for selectively inhibiting the vitro growth of malignant cells| JPS5123537A|1974-04-26|1976-02-25|Adeka Argus Chemical Co Ltd| Kasozaisoseibutsu | GB1527592A|1974-08-05|1978-10-04|Ici Ltd|Wound dressing| US3995623A|1974-12-23|1976-12-07|American Hospital Supply Corporation|Multipurpose flow-directed catheter| JPS5813576B2|1974-12-27|1983-03-14|Adeka Argus Chemical Co Ltd| JPS5524302Y2|1975-03-31|1980-06-10| DE2520814A1|1975-05-09|1976-11-18|Bayer Ag|Light stabilisation of polyurethanes - using polymeric tert. amines from aliphatic diamines and acrylic esters or amides| US4281669A|1975-05-09|1981-08-04|Macgregor David C|Pacemaker electrode with porous system| JPS5210847A|1975-07-16|1977-01-27|Nippon Steel Corp|Pinch roll| US4096860A|1975-10-08|1978-06-27|Mclaughlin William F|Dual flow encatheter| CA1069652A|1976-01-09|1980-01-15|Alain F. Carpentier|Supported bioprosthetic heart valve with compliant orifice ring| US4134402B1|1976-02-11|1989-07-25| US4072146A|1976-09-08|1978-02-07|Howes Randolph M|Venous catheter device| US4335723A|1976-11-26|1982-06-22|The Kendall Company|Catheter having inflatable retention means| US4099528A|1977-02-17|1978-07-11|Sorenson Research Co., Inc.|Double lumen cannula| US4140126A|1977-02-18|1979-02-20|Choudhury M Hasan|Method for performing aneurysm repair| US4265745A|1977-05-25|1981-05-05|Teijin Limited|Permselective membrane| US4182833A|1977-12-07|1980-01-08|Celanese Polymer Specialties Company|Cationic epoxide-amine reaction products| US4180068A|1978-04-13|1979-12-25|Motion Control, Incorporated|Bi-directional flow catheter with retractable trocar/valve structure| DE2960875D1|1978-04-19|1981-12-10|Ici Plc|A method of preparing a tubular product by electrostatic spinning| US4284459A|1978-07-03|1981-08-18|The Kendall Company|Method for making a molded catheter| US4227533A|1978-11-03|1980-10-14|Bristol-Myers Company|Flushable urinary catheter| US4375817A|1979-07-19|1983-03-08|Medtronic, Inc.|Implantable cardioverter| DE3010841A1|1980-03-21|1981-10-08|Ulrich Dr.med. 6936 Haag Uthmann|CATHEDER| US4308085A|1980-07-28|1981-12-29|Jenoptik Jena Gmbh|Process for the preparation of high molecular thermoplastic epoxide-amine-polyadducts| US4475972A|1981-10-01|1984-10-09|Ontario Research Foundation|Implantable material| US4339369A|1981-04-23|1982-07-13|Celanese Corporation|Cationic epoxide-amine reaction products| US4406656A|1981-06-01|1983-09-27|Brack Gillium Hattler|Venous catheter having collapsible multi-lumens| US4401472A|1982-02-26|1983-08-30|Martin Marietta Corporation|Hydraulic cement mixes and processes for improving hydraulic cement mixes| US4568329A|1982-03-08|1986-02-04|Mahurkar Sakharam D|Double lumen catheter| US4546499A|1982-12-13|1985-10-15|Possis Medical, Inc.|Method of supplying blood to blood receiving vessels| US4530113A|1983-05-20|1985-07-23|Intervascular, Inc.|Vascular grafts with cross-weave patterns| US4647416A|1983-08-03|1987-03-03|Shiley Incorporated|Method of preparing a vascular graft prosthesis| US4550447A|1983-08-03|1985-11-05|Shiley Incorporated|Vascular graft prosthesis| US5104399A|1986-12-10|1992-04-14|Endovascular Technologies, Inc.|Artificial graft and implantation method| US4710169A|1983-12-16|1987-12-01|Christopher T Graham|Urinary catheter with collapsible urethral tube| US4571241A|1983-12-16|1986-02-18|Christopher T Graham|Urinary catheter with collapsible urethral tube| US4737518A|1984-04-03|1988-04-12|Takeda Chemical Industries, Ltd.|Lipid derivatives, their production and use| US4562596A|1984-04-25|1986-01-07|Elliot Kornberg|Aortic graft, device and method for performing an intraluminal abdominal aortic aneurysm repair| US4782836A|1984-05-24|1988-11-08|Intermedics, Inc.|Rate adaptive cardiac pacemaker responsive to patient activity and temperature| US4897355A|1985-01-07|1990-01-30|Syntex Inc.|N[ω,-dialkyloxy]- and N-[ω,-dialkenyloxy]-alk-1-yl-N,N,N-tetrasubstituted ammonium lipids and uses therefor| US4662382A|1985-01-16|1987-05-05|Intermedics, Inc.|Pacemaker lead with enhanced sensitivity| US4762915A|1985-01-18|1988-08-09|Liposome Technology, Inc.|Protein-liposome conjugates| US4860751A|1985-02-04|1989-08-29|Cordis Corporation|Activity sensor for pacemaker control| CA1320724C|1985-07-19|1993-07-27|Koichi Kanehira|Terpene amino alcohols and medicinal uses thereof| US4701162A|1985-09-24|1987-10-20|The Kendall Company|Foley catheter assembly| US4737323A|1986-02-13|1988-04-12|Liposome Technology, Inc.|Liposome extrusion method| DE3616824A1|1986-05-17|1987-11-19|Schering Ag|USE OF CURABLE RESIN MIXTURES FOR SURFACE COATINGS AND PRINTING INKS AND METHOD FOR THE PRODUCTION THEREOF| EP0255899B1|1986-07-31|1992-07-15|Werner Prof. Dr.-Ing. Irnich|Rate adaptive pacemaker| US4960409A|1986-09-11|1990-10-02|Catalano Marc L|Method of using bilumen peripheral venous catheter with adapter| JPH0829776B2|1986-10-29|1996-03-27|東燃化学株式会社|Synthetic resin container and mold for manufacturing the same| US4720517A|1986-11-24|1988-01-19|Ciba-Geigy Corporation|Compositions stabilized with N-hydroxyiminodiacetic and dipropionic acids and esters thereof| US4920016A|1986-12-24|1990-04-24|Linear Technology, Inc.|Liposomes with enhanced circulation time| JPS63125144U|1987-02-09|1988-08-16| JPS63154788U|1987-03-31|1988-10-11| JPH0524216B2|1987-06-16|1993-04-07|Hitachi Cable| DE3728917A1|1987-08-29|1989-03-09|Roth Hermann J|Novel lipids containing an asymmetrically substituted disulphide bridge, processes for their preparation, and their use as medicaments| US4946683A|1987-11-18|1990-08-07|Vestar, Inc.|Multiple step entrapment/loading procedure for preparing lipophilic drug-containing liposomes| US5138067A|1987-12-17|1992-08-11|Shionogi & Co. Ltd.|Lipid derivatives| US5047540A|1987-12-17|1991-09-10|Shionogi & Co., Ltd.|Lipid derivatives| US4892540A|1988-04-21|1990-01-09|Sorin Biomedica S.P.A.|Two-leaflet prosthetic heart valve| US5223263A|1988-07-07|1993-06-29|Vical, Inc.|Liponucleotide-containing liposomes| US5176661A|1988-09-06|1993-01-05|Advanced Cardiovascular Systems, Inc.|Composite vascular catheter| US5024671A|1988-09-19|1991-06-18|Baxter International Inc.|Microporous vascular graft| US5200395A|1988-10-18|1993-04-06|Ajinomoto Company, Inc.|Pharmaceutical composition of BUF-5 for treating anemia| CA2001401A1|1988-10-25|1990-04-25|Claude Piantadosi|Quaternary amine containing ether or ester lipid derivatives and therapeutic compositions| DE69032284T2|1989-03-21|1998-10-08|Vical Inc|EXPRESSION OF EXOGENOUS POLYNUCLEOTIDE SEQUENCES IN VERTEBLE| US6214804B1|1989-03-21|2001-04-10|Vical Incorporated|Induction of a protective immune response in a mammal by injecting a DNA sequence| US5703055A|1989-03-21|1997-12-30|Wisconsin Alumni Research Foundation|Generation of antibodies through lipid mediated DNA delivery| FR2645866B1|1989-04-17|1991-07-05|Centre Nat Rech Scient|NEW LIPOPOLYAMINES, THEIR PREPARATION AND THEIR USE| US5194654A|1989-11-22|1993-03-16|Vical, Inc.|Lipid derivatives of phosphonoacids for liposomal incorporation and method of use| US5279833A|1990-04-04|1994-01-18|Yale University|Liposomal transfection of nucleic acids into animal cells| US5101824A|1990-04-16|1992-04-07|Siemens-Pacesetter, Inc.|Rate-responsive pacemaker with circuitry for processing multiple sensor inputs| US5264618A|1990-04-19|1993-11-23|Vical, Inc.|Cationic lipids for intracellular delivery of biologically active molecules| EP0549590A4|1990-07-26|1994-04-13|Rodney James Lane| JPH0765267B2|1990-08-22|1995-07-12|花王株式会社|Softening agent| DK0480667T3|1990-10-09|1996-06-10|Cook Inc|Percutaneous stent construction| DE59008908D1|1990-12-19|1995-05-18|Osypka Peter|Pacemaker lead with an inner channel and with an electrode head.| US5116360A|1990-12-27|1992-05-26|Corvita Corporation|Mesh composite graft| JP2547524Y2|1991-01-22|1997-09-10|東洋ラジエーター株式会社|Oil cooler| US5405363A|1991-03-15|1995-04-11|Angelon Corporation|Implantable cardioverter defibrillator having a smaller displacement volume| US5697953A|1993-03-13|1997-12-16|Angeion Corporation|Implantable cardioverter defibrillator having a smaller displacement volume| US5330768A|1991-07-05|1994-07-19|Massachusetts Institute Of Technology|Controlled drug delivery using polymer/pluronic blends| US5545449A|1991-10-02|1996-08-13|Weyerhaeuser Company|Polyether-reinforced fiber-based materials| US5151105A|1991-10-07|1992-09-29|Kwan Gett Clifford|Collapsible vessel sleeve implant| US6670178B1|1992-07-10|2003-12-30|Transkaryotic Therapies, Inc.|In Vivo production and delivery of insulinotropin for gene therapy| US5284491A|1992-02-27|1994-02-08|Medtronic, Inc.|Cardiac pacemaker with hysteresis behavior| US5352461A|1992-03-11|1994-10-04|Pharmaceutical Discovery Corporation|Self assembling diketopiperazine drug delivery system| SE9200951D0|1992-03-27|1992-03-27|Kabi Pharmacia Ab|PHARMACEUTICAL COMPOSITION CONTAINING A DEFINED LIPID SYSTEM| CA2132342A1|1992-04-06|1993-10-14|Kenneth F. Buechler|Novel opiate derivatives and protein and polypeptide opiate derivative conjugates and labels| WO1994003598A1|1992-08-04|1994-02-17|The Green Cross Corporation|Antiallergic agent| US5334761A|1992-08-28|1994-08-02|Life Technologies, Inc.|Cationic lipids| US5461223A|1992-10-09|1995-10-24|Eastman Kodak Company|Bar code detecting circuitry| US5300022A|1992-11-12|1994-04-05|Martin Klapper|Urinary catheter and bladder irrigation system| US5496362A|1992-11-24|1996-03-05|Cardiac Pacemakers, Inc.|Implantable conformal coil patch electrode with multiple conductive elements for cardioversion and defibrillation| US5552155A|1992-12-04|1996-09-03|The Liposome Company, Inc.|Fusogenic lipsomes and methods for making and using same| US5716395A|1992-12-11|1998-02-10|W.L. Gore & Associates, Inc.|Prosthetic vascular graft| AT187713T|1993-02-19|2000-01-15|Nippon Shinyaku Co Ltd|GLYCEROL DERIVATIVE, DEVICE AND PHARMACEUTICAL COMPOSITION| US5395619A|1993-03-03|1995-03-07|Liposome Technology, Inc.|Lipid-polymer conjugates and liposomes| JPH0753535Y2|1993-03-16|1995-12-13|株式会社ハンズ|rucksack| US5314430A|1993-06-24|1994-05-24|Medtronic, Inc.|Atrial defibrillator employing transvenous and subcutaneous electrodes and method of use| DE4325848A1|1993-07-31|1995-02-02|Basf Ag|Process for the preparation of N- piperazine| EP1062999A3|1993-10-06|2001-03-14|The Kansai Electric Power Co., Inc.|Method for removing carbon dioxide from combustion exhaust gas| US5609624A|1993-10-08|1997-03-11|Impra, Inc.|Reinforced vascular graft and method of making same| SE9303481L|1993-10-22|1995-04-23|Berol Nobel Ab|hygiene composition| AU1091095A|1993-11-08|1995-05-29|Harrison M. Lazarus|Intraluminal vascular graft and method| CA2176714A1|1993-11-24|1995-06-01|Timothy D. Heath|Amphiphilic derivatives of piperazine| US5595756A|1993-12-22|1997-01-21|Inex Pharmaceuticals Corporation|Liposomal compositions for enhanced retention of bioactive agents| US5464924A|1994-01-07|1995-11-07|The Dow Chemical Company|Flexible poly for barrier packaging| JP2001523215A|1995-04-17|2001-11-20|イマークスファーマシューティカルコーポレーション|Hybrid magnetic resonance contrast agent| US6077835A|1994-03-23|2000-06-20|Case Western Reserve University|Delivery of compacted nucleic acid to cells| US5844107A|1994-03-23|1998-12-01|Case Western Reserve University|Compacted nucleic acids and their delivery to cells| US5624976A|1994-03-25|1997-04-29|Dentsply Gmbh|Dental filling composition and method| AU689479B2|1994-04-12|1998-04-02|Transave, Inc.|Fusogenic liposomes and methods of making and using same| US5840576A|1994-07-20|1998-11-24|Cytotherapeutics, Inc.|Methods and compositions of growth control for cells encapsulated within bioartificial organs| US5693338A|1994-09-29|1997-12-02|Emisphere Technologies, Inc.|Diketopiperazine-based delivery systems| US5885613A|1994-09-30|1999-03-23|The University Of British Columbia|Bilayer stabilizing components and their use in forming programmable fusogenic liposomes| US5820873A|1994-09-30|1998-10-13|The University Of British Columbia|Polyethylene glycol modified ceramide lipids and liposome uses thereof| US5641665A|1994-11-28|1997-06-24|Vical Incorporated|Plasmids suitable for IL-2 expression| US6071890A|1994-12-09|2000-06-06|Genzyme Corporation|Organ-specific targeting of cationic amphiphile/DNA complexes for gene therapy| US5965434A|1994-12-29|1999-10-12|Wolff; Jon A.|Amphipathic PH sensitive compounds and delivery systems for delivering biologically active compounds| US6485726B1|1995-01-17|2002-11-26|The Brigham And Women's Hospital, Inc.|Receptor specific transepithelial transport of therapeutics| US5830430A|1995-02-21|1998-11-03|Imarx Pharmaceutical Corp.|Cationic lipids and the use thereof| US5772694A|1995-05-16|1998-06-30|Medical Carbon Research Institute L.L.C.|Prosthetic heart valve with improved blood flow| US5783383A|1995-05-23|1998-07-21|The Board Of Trustees Of The Leland Stanford Junior University|Method of detecting cytomegalovirus | US7422902B1|1995-06-07|2008-09-09|The University Of British Columbia|Lipid-nucleic acid particles prepared via a hydrophobic lipid-nucleic acid complex intermediate and use for gene transfer| US5609629A|1995-06-07|1997-03-11|Med Institute, Inc.|Coated implantable medical device| US5705385A|1995-06-07|1998-01-06|Inex Pharmaceuticals Corporation|Lipid-nucleic acid particles prepared via a hydrophobic lipid-nucleic acid complex intermediate and use for gene transfer| US5981501A|1995-06-07|1999-11-09|Inex Pharmaceuticals Corp.|Methods for encapsulating plasmids in lipid bilayers| US5976567A|1995-06-07|1999-11-02|Inex Pharmaceuticals Corp.|Lipid-nucleic acid particles prepared via a hydrophobic lipid-nucleic acid complex intermediate and use for gene transfer| US5607385A|1995-08-17|1997-03-04|Medtronic, Inc.|Device and algorithm for a combined cardiomyostimulator and a cardiac pacer-carioverter-defibrillator| US5744335A|1995-09-19|1998-04-28|Mirus Corporation|Process of transfecting a cell with a polynucleotide mixed with an amphipathic compound and a DNA-binding protein| FR2740978B1|1995-11-10|1998-01-02|Ela Medical Sa|IMPLANTABLE DEFIBRILLATOR / CARDIOVERVER ACTIVE MEDICAL DEVICE| US5874105A|1996-01-31|1999-02-23|Collaborative Laboratories, Inc.|Lipid vesicles formed with alkylammonium fatty acid salts| WO1997038010A2|1996-04-11|1997-10-16|The University Of British Columbia|Fusogenic liposomes| US5783566A|1996-05-10|1998-07-21|California Institute Of Technology|Method for increasing or decreasing transfection efficiency| US5935936A|1996-06-03|1999-08-10|Genzyme Corporation|Compositions comprising cationic amphiphiles and co-lipids for intracellular delivery of therapeutic molecules| US5913848A|1996-06-06|1999-06-22|Luther Medical Products, Inc.|Hard tip over-the-needle catheter and method of manufacturing the same| US5677124A|1996-07-03|1997-10-14|Ambion, Inc.|Ribonuclease resistant viral RNA standards| US5736573A|1996-07-31|1998-04-07|Galat; Alexander|Lipid and water soluble derivatives of drugs| US7288266B2|1996-08-19|2007-10-30|United States Of America As Represented By The Secretary, Department Of Health And Human Services|Liposome complexes for increased systemic delivery| ES2208936T3|1996-08-26|2004-06-16|Transgene S.A.|CATIONIC LIPID COMPLEX-NUCLEIC ACIDS.| US6458574B1|1996-09-12|2002-10-01|Transkaryotic Therapies, Inc.|Treatment of a α-galactosidase a deficiency| AT395904T|1996-09-13|2008-06-15|Lipoxen Technologies Ltd|LIPOSOME| TW520297B|1996-10-11|2003-02-11|Sequus Pharm Inc|Fusogenic liposome composition and method| DE69735382T2|1996-11-04|2006-11-30|Qiagen Gmbh|CATIONIC REAGENTS FOR TRANSFECTION| US6887665B2|1996-11-14|2005-05-03|Affymetrix, Inc.|Methods of array synthesis| US5985930A|1996-11-21|1999-11-16|Pasinetti; Giulio M.|Treatment of neurodegenerative conditions with nimesulide| US6204297B1|1996-11-26|2001-03-20|Rhodia Inc.|Nonionic gemini surfactants| JPH10197978A|1997-01-09|1998-07-31|Mitsubishi Paper Mills Ltd|Silver halide photographic sensitive material| EP0853123A1|1997-01-10|1998-07-15|Roche Diagnostics GmbH|Purification of DNA by 'cross-flow-filtration'| FR2760193B1|1997-02-28|1999-05-28|Transgene Sa|LIPIDS AND COMPLEXES OF CATIONIC LIPIDS AND ACTIVE SUBSTANCES, IN PARTICULAR FOR THE TRANSFECTION OF CELLS| US5837283A|1997-03-12|1998-11-17|The Regents Of The University Of California|Cationic lipid compositions targeting angiogenic endothelial cells| US6093392A|1997-03-14|2000-07-25|Childrens Hospital Of Phildelphia|Methods and compositions for use in gene therapy for treatment of hemophilia| US5945326A|1997-03-20|1999-08-31|New England Biolabs, Inc.|Method for cloning and producing the Spel restriction endonuclease| EP1027033B1|1997-05-14|2009-07-22|The University Of British Columbia|High efficiency encapsulation of nucleic acids in lipid vesicles| US6835395B1|1997-05-14|2004-12-28|The University Of British Columbia|Composition containing small multilamellar oligodeoxynucleotide-containing lipid vesicles| US20030104044A1|1997-05-14|2003-06-05|Semple Sean C.|Compositions for stimulating cytokine secretion and inducing an immune response| JPH115786A|1997-06-13|1999-01-12|Pola Chem Ind Inc|Novel aminohydroxypropylpiperazine derivative| JPH1180142A|1997-09-05|1999-03-26|Pola Chem Ind Inc|Production of diphenylalkyl compound| EP1021549A2|1997-09-19|2000-07-26|Sequitur, Inc.|SENSE mRNA THERAPY| DE69837274T2|1997-10-10|2007-11-08|Inex Pharmaceuticals Corp., Burnaby|METHOD FOR THE SECULATION OF NUCLEIC ACIDS IN LIPID DOUBLE LAYERS| US6734171B1|1997-10-10|2004-05-11|Inex Pharmaceuticals Corp.|Methods for encapsulating nucleic acids in lipid bilayers| JP2002522509A|1997-10-29|2002-07-23|ジェンザイム・コーポレイション|Lysosomal storage disease therapeutic compositions and methods| US6165763A|1997-10-30|2000-12-26|Smithkline Beecham Corporation|Ornithine carbamoyltransferase| US6096075A|1998-01-22|2000-08-01|Medical Carbon Research Institute, Llc|Prosthetic heart valve| US6617171B2|1998-02-27|2003-09-09|The General Hospital Corporation|Methods for diagnosing and treating autoimmune disease| US6271209B1|1998-04-03|2001-08-07|Valentis, Inc.|Cationic lipid formulation delivering nucleic acid to peritoneal tumors| US6176877B1|1998-04-20|2001-01-23|St. Jude Medical, Inc.|Two piece prosthetic heart valve| DE19822602A1|1998-05-20|1999-11-25|Goldschmidt Ag Th|Process for the preparation of polyamino acid esters by esterification of acidic polyamino acids or transesterification of polyamino acid esters| NO313244B1|1998-07-08|2002-09-02|Crew Dev Corp|Process for the isolation and production of magnesite or magnesium chloride| US6055454A|1998-07-27|2000-04-25|Cardiac Pacemakers, Inc.|Cardiac pacemaker with automatic response optimization of a physiologic sensor based on a second sensor| US6067471A|1998-08-07|2000-05-23|Cardiac Pacemakers, Inc.|Atrial and ventricular implantable cardioverter-defibrillator and lead system| WO2000010622A1|1998-08-20|2000-03-02|Cook Incorporated|Coated implantable medical device| US6210892B1|1998-10-07|2001-04-03|Isis Pharmaceuticals, Inc.|Alteration of cellular behavior by antisense modulation of mRNA processing| AU1830200A|1998-11-25|2000-06-13|Vanderbilt University|Cationic liposomes for gene transfer| US6248725B1|1999-02-23|2001-06-19|Amgen, Inc.|Combinations and methods for promoting in vivo liver cell proliferation and enhancing in vivo liver-directed gene transduction| US6379698B1|1999-04-06|2002-04-30|Isis Pharmaceuticals, Inc.|Fusogenic lipids and vesicles| JP5117648B2|1999-04-20|2013-01-16|ザ・ユニバーシティ・オブ・ブリティッシュ・コロンビア|Cationic PEG lipids and methods of use.| AT340592T|1999-04-23|2006-10-15|Alza Corp|CONJUGATE CONTAINS A SPLICABLE BINDING FOR USE IN A LIPOSOM| US6169923B1|1999-04-23|2001-01-02|Pacesetter, Inc.|Implantable cardioverter-defibrillator with automatic arrhythmia detection criteria adjustment| PL352332A1|1999-05-19|2003-08-11|Lexigen Pharm Corp|Expression and export of interferon-alpha proteins as fc fusion proteins| US6696424B1|1999-05-28|2004-02-24|Vical Incorporated|Cytofectin dimers and methods of use thereof| US6346382B1|1999-06-01|2002-02-12|Vanderbilt University|Human carbamyl phosphate synthetase I polymorphism and diagnostic methods related thereto| WO2001005375A1|1999-07-16|2001-01-25|Purdue Research Foundation|Vinyl ether lipids with cleavable hydrophilic headgroups| JP2003505077A|1999-07-23|2003-02-12|ジェネンテック・インコーポレーテッド|Purification of RNAse- and organic solvent-free plasmid DNA using parallel flow filtration| US6358278B1|1999-09-24|2002-03-19|St. Jude Medical, Inc.|Heart valve prosthesis with rotatable cuff| US6371983B1|1999-10-04|2002-04-16|Ernest Lane|Bioprosthetic heart valve| US7060291B1|1999-11-24|2006-06-13|Transave, Inc.|Modular targeted liposomal delivery system| AU3366901A|1999-12-30|2001-07-16|Novartis Ag|Novel colloid synthetic vectors for gene therapy| EP1261379A2|2000-02-17|2002-12-04|Genzyme Corporation|Genetic modification of the lung as a portal for gene delivery| US6370434B1|2000-02-28|2002-04-09|Cardiac Pacemakers, Inc.|Cardiac lead and method for lead implantation| US6565960B2|2000-06-01|2003-05-20|Shriners Hospital Of Children|Polymer composite compositions| WO2002000870A2|2000-06-26|2002-01-03|Christian Plank|Method for transfecting cells using a magnetic field| IL138474D0|2000-09-14|2001-10-31|Epox Ltd|Highly branched water-soluble polyamine oligomers, process for their preparation and applications thereof| US6998115B2|2000-10-10|2006-02-14|Massachusetts Institute Of Technology|Biodegradable poly and uses thereof| US7427394B2|2000-10-10|2008-09-23|Massachusetts Institute Of Technology|Biodegradable poly and uses thereof| USRE43612E1|2000-10-10|2012-08-28|Massachusetts Institute Of Technology|Biodegradable poly and uses thereof| AU1485402A|2000-10-25|2002-05-06|Univ British Columbia|Lipid formulations for target delivery| GB0028361D0|2000-11-21|2001-01-03|Glaxo Group Ltd|Method of separating extra chromosomal dna from other cellular components| US20020094528A1|2000-11-29|2002-07-18|Salafsky Joshua S.|Method and apparatus using a surface-selective nonlinear optical technique for detection of probe-target interations| JP2002167368A|2000-12-01|2002-06-11|Nitto Denko Corp|Alkyl group-substituted dendrimer and method for preparing the same| US20050004058A1|2000-12-07|2005-01-06|Patrick Benoit|Sequences upstream of the carp gene, vectors containing them and uses thereof| DE10109897A1|2001-02-21|2002-11-07|Novosom Ag|Optional cationic liposomes and their use| US20020192721A1|2001-03-28|2002-12-19|Engeneos, Inc.|Modular molecular clasps and uses thereof| KR100823815B1|2001-04-23|2008-04-21|신에쓰 가가꾸 고교 가부시끼가이샤|Novel Tertiary Amine Compounds Having an Ester Structure and Processes for Preparing Same| US6585410B1|2001-05-03|2003-07-01|The United States Of America As Represented By The Administrator Of The National Aeronautics And Space Administration|Radiant temperature nulling radiometer| DE50214379D1|2001-06-05|2010-05-27|Curevac Gmbh|Stabilized mRNA with elevated G / C content for gene therapy| EP1601767B1|2003-03-05|2012-04-25|Senesco Technologies, Inc.|USE OF siRNA TO SUPPRESS EXPRESSION OF EIF-5A1 IN THE TREATMENT OF GLAUCOMA| US7601367B2|2002-05-28|2009-10-13|Mirus Bio Llc|Compositions and processes using siRNA, amphipathic compounds and polycations| EP2428571B1|2001-09-28|2018-07-18|Max-Planck-Gesellschaft zur Förderung der Wissenschaften e.V.|MicroRNA molecules| WO2003040288A2|2001-11-09|2003-05-15|Bayer Healthcare Ag|Isotopically coded affinity markers 3| DE10162480A1|2001-12-19|2003-08-07|Ingmar Hoerr|The application of mRNA for use as a therapeutic agent against tumor diseases| DE10207178A1|2002-02-19|2003-09-04|Novosom Ag|Components for the production of amphoteric liposomes| DE10214983A1|2002-04-04|2004-04-08|TransMIT Gesellschaft für Technologietransfer mbH|Nebulisable liposomes and their use for pulmonary application of active substances| US20030215395A1|2002-05-14|2003-11-20|Lei Yu|Controllably degradable polymeric biomolecule or drug carrier and method of synthesizing said carrier| DE60334618D1|2002-06-28|2010-12-02|Protiva Biotherapeutics Inc|METHOD AND DEVICE FOR PREPARING LIPOSOMES| DE10229872A1|2002-07-03|2004-01-29|Curevac Gmbh|Immune stimulation through chemically modified RNA| WO2005028619A2|2003-09-15|2005-03-31|Massachusetts Institute Of Technology|Nanoliter-scale synthesis of arrayed biomaterials and screening thereof| US20040028804A1|2002-08-07|2004-02-12|Anderson Daniel G.|Production of polymeric microarrays| JP2005536214A|2002-08-22|2005-12-02|セルトランリミテッド|Cell culture surface| CN100457804C|2002-11-04|2009-02-04|Ge拜尔硅股份有限公司|Linear polyamino and/or polyammonium polysiloxane copolymers I| JP2006515574A|2002-11-22|2006-06-01|ノボ・ノルデイスク・エー/エス|Compounds used in the treatment of obesity| US7169892B2|2003-01-10|2007-01-30|Astellas Pharma Inc.|Lipid-peptide-polymer conjugates for long blood circulation and tumor specific drug delivery systems| JP4408862B2|2003-01-20|2010-02-03|旭化成エレクトロニクス株式会社|pointing device| US20040224912A1|2003-05-07|2004-11-11|Isis Pharmaceuticals Inc.|Modulation of PAI-1 mRNA-binding protein expression| US7619017B2|2003-05-19|2009-11-17|Wacker Chemical Corporation|Polymer emulsions resistant to biodeterioration| EP1644479A4|2003-06-16|2008-04-23|Mark W Grinstaff|Functional synthetic molecules and macromolecules for gene delivery| NZ581166A|2003-09-15|2011-06-30|Protiva Biotherapeutics Inc|Polyethyleneglycol-modified lipid compounds and uses thereof| US20050069590A1|2003-09-30|2005-03-31|Buehler Gail K.|Stable suspensions for medicinal dosages| JP4814099B2|2003-10-10|2011-11-09|パウダージェクトバクシーンズ,インコーポレイテッド|Nucleic acid construct| WO2005037226A2|2003-10-17|2005-04-28|Georgia Tech Research Corporation|Genetically engineered enteroendocrine cells for treating glucose-related metabolic disorders| JP4800769B2|2003-11-10|2011-10-26|日本化薬株式会社|Diimonium salt compounds and uses thereof| US7022214B2|2004-01-21|2006-04-04|Bio-Rad Laboratories, Inc.|Carrier ampholytes of high pH range| US7556684B2|2004-02-26|2009-07-07|Construction Research & Technology Gmbh|Amine containing strength improvement admixture| US20060228404A1|2004-03-04|2006-10-12|Anderson Daniel G|Compositions and methods for treatment of hypertrophic tissues| NZ550085A|2004-05-19|2009-06-26|Clariant Finance Bvi Ltd|Monoazo dyes| CA2569664C|2004-06-07|2013-07-16|Protiva Biotherapeutics, Inc.|Lipid encapsulated interfering rna| EP1781593B1|2004-06-07|2011-12-14|Protiva Biotherapeutics Inc.|Cationic lipids and methods of use| US7670595B2|2004-06-28|2010-03-02|Merck Patent Gmbh|Fc-interferon-beta fusion proteins| GB0418172D0|2004-08-13|2004-09-15|Ic Vec Ltd|Vector| DE102004042546A1|2004-09-02|2006-03-09|Curevac Gmbh|Combination therapy for immune stimulation| DE102004043342A1|2004-09-08|2006-03-09|Bayer Materialscience Ag|Blocked polyurethane prepolymers as adhesives| US20060216343A1|2004-11-05|2006-09-28|Steffen Panzner|Pharmaceutical compositions comprising an oligonucleotide as an active agent| GB0502482D0|2005-02-07|2005-03-16|Glaxo Group Ltd|Novel compounds| US7514099B2|2005-02-14|2009-04-07|Sirna Therapeutics, Inc.|Lipid nanoparticle based compositions and methods for the delivery of biologically active molecules| US7977452B2|2005-03-28|2011-07-12|Dendritic Nanotechnologies, Inc.|Janus dendrimers and dendrons| AU2006259415B2|2005-06-15|2012-08-30|Massachusetts Institute Of Technology|Amine-containing lipids and uses thereof| PT2578685T|2005-08-23|2019-07-10|Univ Pennsylvania|Rna containing modified nucleosides and methods of use thereof| US9012219B2|2005-08-23|2015-04-21|The Trustees Of The University Of Pennsylvania|RNA preparations comprising purified modified RNA for reprogramming cells| WO2007031091A2|2005-09-15|2007-03-22|Santaris Pharma A/S|Rna antagonist compounds for the modulation of p21 ras expression| JP5336853B2|2005-11-02|2013-11-06|プロチババイオセラピューティクスインコーポレイティッド|Modified siRNA molecules and methods of use thereof| US7238791B1|2005-12-16|2007-07-03|Roche Diagnostics Operations, Inc.|6-monoacetylmorphine derivatives useful in immunoassay| WO2007073489A2|2005-12-22|2007-06-28|Trustees Of Boston University|Molecules for gene delivery and gene therapy, and methods of use thereof| CN100569877C|2005-12-30|2009-12-16|财团法人工业技术研究院|Contain the dendritic structural compounds of branch and the application thereof of many UV crosslinking reactive group| CA2649325C|2006-04-14|2019-01-08|Epicentre Technologies Corporation|Kits and methods for generating 5' capped rna| US9085778B2|2006-05-03|2015-07-21|VL27, Inc.|Exosome transfer of nucleic acids to cells| US20070275923A1|2006-05-25|2007-11-29|Nastech Pharmaceutical Company Inc.|CATIONIC PEPTIDES FOR siRNA INTRACELLULAR DELIVERY| WO2007143659A2|2006-06-05|2007-12-13|Massachusetts Institute Of Technology|Crosslinked, degradable polymers and uses thereof| US20090186805A1|2006-07-06|2009-07-23|Aaron Thomas Tabor|Compositions and Methods for Genetic Modification of Cells Having Cosmetic Function to Enhance Cosmetic Appearance| FR2904144A1|2006-07-19|2008-01-25|St Microelectronics Rousset|METHOD FOR MANUFACTURING A SEMICONDUCTOR WAFER COMPRISING AN INTEGRATED OPTICAL FILTER| ES2293834B1|2006-07-20|2009-02-16|Consejo Superior Investig. Cientificas|COMPOSED WITH INHIBITING ACTIVITY OF UBC13-UEV INTERACTIONS, PHARMACEUTICAL COMPOSITIONS THAT INCLUDE IT AND ITS THERAPEUTIC APPLICATIONS.| CA2658768C|2006-07-21|2016-05-17|Massachusetts Institute Of Technology|End-modified poly and uses thereof| CA2665225C|2006-10-03|2015-06-30|Alnylam Pharmaceuticals, Inc.|Lipid containing formulations| US20110035819A1|2006-10-12|2011-02-10|Copernicus Therapeutics Inc.|Codon optimized cftr| DE102006051516A1|2006-10-31|2008-05-08|Curevac Gmbh| modified RNA to increase the expression of a protein| US8268586B2|2006-12-21|2012-09-18|Novozymes, Inc.|Modified messenger RNA stabilizing sequences for expressing genes in bacterial cells| DE102007001370A1|2007-01-09|2008-07-10|Curevac Gmbh|RNA-encoded antibodies| US7700734B2|2007-01-09|2010-04-20|Shu-Wha Lin|Recombinant human factor IX and use thereof| US20100189729A1|2007-01-09|2010-07-29|Curvac Gmbh|Rna-coded antibody| US8859229B2|2007-02-02|2014-10-14|Yale University|Transient transfection with RNA| EP2139461A2|2007-03-20|2010-01-06|Recepticon Aps|Amino derivatives to prevent nephrotoxicity and cancer| JP5186126B2|2007-03-29|2013-04-17|公益財団法人地球環境産業技術研究機構|Novel triazine derivatives, their production and their use as gas separation membranes| PL2144609T3|2007-04-18|2015-04-30|Cornerstone Pharmaceuticals Inc|Pharmaceutical formulations containing lipoic acid derivatives| WO2008137470A1|2007-05-01|2008-11-13|Pgr-Solutions|Multi-chain lipophilic polyamines| US20090163705A1|2007-05-21|2009-06-25|Alnylam Pharmaceuticals, Inc.|Cationic lipids| WO2009030254A1|2007-09-04|2009-03-12|Curevac Gmbh|Complexes of rna and cationic peptides for transfection and for immunostimulation| CA2701274A1|2007-10-02|2009-04-09|Mdrna, Inc.|Lipopeptides for delivery of nucleic acids| WO2009046739A1|2007-10-09|2009-04-16|Curevac Gmbh|Composition for treating prostate cancer | WO2009066758A1|2007-11-22|2009-05-28|Japan Science And Technology Agency|Translation regulation system in cell or artificial cell model by using low-molecular-weight rna| JP5519523B2|2007-12-04|2014-06-11|アルニラムファーマスーティカルズインコーポレイテッド|Carbohydrate conjugates as oligonucleotide delivery agents| WO2009120247A2|2007-12-27|2009-10-01|The Ohio State University Research Foundation|Lipid nanoparticle compositions and methods of making and using the same| ES2535419T3|2007-12-27|2015-05-11|Protiva Biotherapeutics Inc.|Polo kinase expression silencing using interfering RNA| WO2009088891A1|2008-01-02|2009-07-16|Alnylam Pharmaceuticals, Inc.|Screening method for selected amino lipid-containing compositions| JP5749494B2|2008-01-02|2015-07-15|テクミラ ファーマシューティカルズ コーポレイション|Improved compositions and methods for delivery of nucleic acids| US8575123B2|2008-04-11|2013-11-05|Tekmira Pharmaceuticals Corporation|Site-specific delivery of nucleic acids by combining targeting ligands with endosomolytic components| US8058069B2|2008-04-15|2011-11-15|Protiva Biotherapeutics, Inc.|Lipid formulations for nucleic acid delivery| US20090263407A1|2008-04-16|2009-10-22|Abbott Laboratories|Cationic Lipids and Uses Thereof| WO2009127230A1|2008-04-16|2009-10-22|Curevac Gmbh|MODIFIED RNA FOR SUPPRESSING OR AVOIDING AN IMMUNOSTIMULATORY RESPONSE AND IMMUNOSUPPRESSIVE COMPOSITION| US8222221B2|2008-06-04|2012-07-17|The Board Of Regents Of The University Of Texas System|Modulation of gene expression through endogenous small RNA targeting of gene promoters| JP5024216B2|2008-07-23|2012-09-12|トヨタ自動車株式会社|Ignition timing control device and ignition timing control method for internal combustion engine| US20100035249A1|2008-08-05|2010-02-11|Kabushiki Kaisha Dnaform|Rna sequencing and analysis using solid support| EP2331954B1|2008-08-27|2020-03-25|Life Technologies Corporation|Apparatus for and method of processing biological samples| WO2010037408A1|2008-09-30|2010-04-08|Curevac Gmbh|Composition comprising a complexed rna and a naked mrna for providing or enhancing an immunostimulatory response in a mammal and uses thereof| EP2350043B9|2008-10-09|2014-08-20|TEKMIRA Pharmaceuticals Corporation|Improved amino lipids and methods for the delivery of nucleic acids| AU2009305639B2|2008-10-16|2016-06-23|Marina Biotech, Inc.|Processes and compositions for liposomal and efficient delivery of gene silencing therapeutics| US9080211B2|2008-10-24|2015-07-14|Epicentre Technologies Corporation|Transposon end compositions and methods for modifying nucleic acids| US20120009222A1|2008-10-27|2012-01-12|Massachusetts Institute Of Technology|Modulation of the immune response| WO2010053572A2|2008-11-07|2010-05-14|Massachusetts Institute Of Technology|Aminoalcohol lipidoids and uses thereof| WO2010054401A1|2008-11-10|2010-05-14|Alnylam Pharmaceuticals, Inc.|Novel lipids and compositions for the delivery of therapeutics| EP2350296A4|2008-11-17|2013-04-03|Enzon Pharmaceuticals Inc|Branched cationic lipids for nucleic acids delivery system| US9023820B2|2009-01-26|2015-05-05|Protiva Biotherapeutics, Inc.|Compositions and methods for silencing apolipoprotein C-III expression| CA2751342C|2009-01-29|2019-05-07|Alnylam Pharmaceuticals, Inc.|Lipid formulations comprising cationic lipid and a targeting lipid comprising n-acetyl galactosamine for delivery of nucleic acid| US20100222489A1|2009-02-27|2010-09-02|Jiang Dayue D|Copolymer composition, membrane article, and methods thereof| JP6032724B2|2009-03-12|2016-11-30|アルナイラム ファーマシューティカルズ, インコーポレイテッドAlnylam Pharmaceuticals, Inc.|Lipid preparation composition and method for inhibiting expression of Eg5 gene and VEGF gene| WO2010114789A1|2009-04-02|2010-10-07|The Siemon Company|Telecommunications patch panel| CN102497887A|2009-04-17|2012-06-13|Isis创新公司|Composition for delivery of genetic material| US9220682B2|2009-04-22|2015-12-29|Emory University|Nanocarrier therapy for treating invasive tumors| CA2760776C|2009-05-05|2019-07-09|Alnylam Pharmaceuticals, Inc.|Lipid compositions for the delivery of therapeutic agents| LT3431076T|2009-06-10|2021-10-25|Arbutus Biopharma Corporation|Improved lipid formulation| WO2010147992A1|2009-06-15|2010-12-23|Alnylam Pharmaceuticals, Inc.|Methods for increasing efficacy of lipid formulated sirna| EA201270019A1|2009-06-15|2012-06-29|Элнилэм Фармасьютикалз, Инк.|BENTROVAL RNA INCLUDED IN LIPID COMPOSITION AND WHICH IS THE PCSK9 GENE| US8236943B2|2009-07-01|2012-08-07|Protiva Biotherapeutics, Inc.|Compositions and methods for silencing apolipoprotein B| US9018187B2|2009-07-01|2015-04-28|Protiva Biotherapeutics, Inc.|Cationic lipids and methods for the delivery of therapeutic agents| WO2011000106A1|2009-07-01|2011-01-06|Protiva Biotherapeutics, Inc.|Improved cationic lipids and methods for the delivery of therapeutic agents| US8716464B2|2009-07-20|2014-05-06|Thomas W. Geisbert|Compositions and methods for silencing Ebola virus gene expression| CA2769408C|2009-07-30|2017-09-05|Laboratorios Salvat, S.A.|Apaf-1 inhibitor compounds| US20120195936A1|2009-07-31|2012-08-02|Ethris Gmbh|Rna with a combination of unmodified and modified nucleotides for protein expression| DE102009043342A1|2009-09-29|2011-03-31|Bayer Technology Services Gmbh|Substances for self-organized carriers for the controlled release of an active substance| ES2734973T3|2009-12-01|2019-12-13|Translate Bio Inc|Administration of mRNA for the increase of proteins and enzymes in human genetic diseases| JP2013512690A|2009-12-07|2013-04-18|ザトラスティースオブザユニバーシティオブペンシルベニア|RNA preparation comprising purified modified RNA for reprogramming cells| WO2011069529A1|2009-12-09|2011-06-16|Curevac Gmbh|Mannose-containing solution for lyophilization, transfection and/or injection of nucleic acids| US20130017223A1|2009-12-18|2013-01-17|The University Of British Columbia|Methods and compositions for delivery of nucleic acids| EP2338520A1|2009-12-21|2011-06-29|Ludwig Maximilians Universität|Conjugate with targeting ligand and use of same| WO2011076807A2|2009-12-23|2011-06-30|Novartis Ag|Lipids, lipid compositions, and methods of using them| US20130123338A1|2010-05-12|2013-05-16|Protiva Biotherapeutics, Inc.|Novel cationic lipids and methods of use thereof| KR101967411B1|2010-06-03|2019-04-10|알닐람 파마슈티칼스 인코포레이티드|Biodegradable lipids for the delivery of active agents| CN101863544B|2010-06-29|2011-09-28|湖南科技大学|Cyanuric acid-based heavy metal chelating flocculant and preparation method thereof| US9006417B2|2010-06-30|2015-04-14|Protiva Biotherapeutics, Inc.|Non-liposomal systems for nucleic acid delivery| US20130323269A1|2010-07-30|2013-12-05|Muthiah Manoharan|Methods and compositions for delivery of active agents| US8822663B2|2010-08-06|2014-09-02|Moderna Therapeutics, Inc.|Engineered nucleic acids and methods of use thereof| WO2012019630A1|2010-08-13|2012-02-16|Curevac Gmbh|Nucleic acid comprising or coding for a histone stem-loop and a poly sequence or a polyadenylation signal for increasing the expression of an encoded protein| WO2012027675A2|2010-08-26|2012-03-01|Massachusetts Institute Of Technology|Poly, their preparation, and uses thereof| EP2625189B1|2010-10-01|2018-06-27|ModernaTX, Inc.|Engineered nucleic acids and methods of use thereof| US8853377B2|2010-11-30|2014-10-07|Shire Human Genetic Therapies, Inc.|mRNA for use in treatment of human genetic diseases| WO2012113413A1|2011-02-21|2012-08-30|Curevac Gmbh|Vaccine composition comprising complexed immunostimulatory nucleic acids and antigens packaged with disulfide-linked polyethyleneglycol/peptide conjugates| WO2012116714A1|2011-03-02|2012-09-07|Curevac Gmbh|Vaccination in elderly patients| WO2012116715A1|2011-03-02|2012-09-07|Curevac Gmbh|Vaccination in newborns and infants| WO2012135025A2|2011-03-28|2012-10-04|Massachusetts Institute Of Technology|Conjugated lipomers and uses thereof| WO2012135805A2|2011-03-31|2012-10-04|modeRNA Therapeutics|Delivery and formulation of engineered nucleic acids| WO2012133737A1|2011-03-31|2012-10-04|公益財団法人地球環境産業技術研究機構|Crosslinkable amine compound, polymer membrane using crosslinkable amine compound, and method for producing polymer membrane| CN103687957A|2011-05-17|2014-03-26|现代治疗公司|Engineered nucleic acids and methods of use thereof for non-human vertebrates| EP2532649B1|2011-06-07|2015-04-08|Incella GmbH|Amino lipids, their synthesis and uses thereof| RU2013154295A|2011-06-08|2015-07-20|Шир Хьюман Дженетик Терапис, Инк.|COMPOSITIONS OF LIPID NANOPARTICLES AND METHODS FOR DELIVERY OF mRNA| WO2012170889A1|2011-06-08|2012-12-13|Shire Human Genetic Therapies, Inc.|Cleavable lipids| US9862926B2|2011-06-27|2018-01-09|Cellscript, Llc.|Inhibition of innate immune response| US9464124B2|2011-09-12|2016-10-11|Moderna Therapeutics, Inc.|Engineered nucleic acids and methods of use thereof| EP2755986A4|2011-09-12|2015-05-20|Moderna Therapeutics Inc|Engineered nucleic acids and methods of use thereof| EP2755693A4|2011-09-12|2015-05-20|Moderna Therapeutics Inc|Engineered nucleic acids and methods of use thereof| CN110511939A|2011-10-03|2019-11-29|现代泰克斯公司|Nucleosides, nucleotide and nucleic acid of modification and application thereof| AU2012352180A1|2011-12-16|2014-07-31|Moderna Therapeutics, Inc.|Modified nucleoside, nucleotide, and nucleic acid compositions| RU2014117690A|2011-10-05|2015-11-10|Протива Байотерапьютикс Инк.|COMPOSITIONS AND METHODS FOR ALDEHYDE-DEHYDROGENASE SILENCING| KR102272498B1|2011-10-27|2021-07-06|메사추세츠 인스티튜트 오브 테크놀로지|Amino acid derivatives functionalized on the n-terminal capable of forming drug incapsulating microspheres| EP2791364A4|2011-12-14|2015-11-11|Moderna Therapeutics Inc|Methods of responding to a biothreat| EP2791159A4|2011-12-14|2015-10-14|Moderna Therapeutics Inc|Modified nucleic acids, and acute care uses thereof| US20140275229A1|2012-04-02|2014-09-18|Moderna Therapeutics, Inc.|Modified polynucleotides encoding udp glucuronosyltransferase 1 family, polypeptide a1| US9283287B2|2012-04-02|2016-03-15|Moderna Therapeutics, Inc.|Modified polynucleotides for the production of nuclear proteins| WO2013096709A2|2011-12-21|2013-06-27|modeRNA Therapeutics|Methods of increasing the viability or longevity of an organ or organ explant| EP2797634A4|2011-12-29|2015-08-05|Moderna Therapeutics Inc|Modified mrnas encoding cell-penetrating polypeptides| LT3144389T|2011-12-30|2018-08-10|Cellscript, Llc|Making and using in vitro-synthesized ssrna for introducing into mammalian cells to induce a biological or biochemical effect| WO2013120497A1|2012-02-15|2013-08-22|Curevac Gmbh|Nucleic acid comprising or coding for a histone stem-loop and a poly sequence or a polyadenylation signal for increasing the expression of an encoded therapeutic protein| WO2013120499A1|2012-02-15|2013-08-22|Curevac Gmbh|Nucleic acid comprising or coding for a histone stem-loop and a poly sequence or a polyadenylation signal for increasing the expression of an encoded pathogenic antigen| KR102075810B1|2012-02-24|2020-02-10|아뷰터스 바이오파마 코포레이션|Trialkyl cationic lipids and methods of use thereof| SG10201607968WA|2012-03-27|2016-12-29|Curevac Ag|Artificial nucleic acid molecules for improved protein or peptide expression| ES2762873T3|2012-03-29|2020-05-26|Translate Bio Inc|Ionizable Cationic Lipids| CA2868030C|2012-03-29|2021-05-25|Shire Human Genetic Therapies, Inc.|Lipid-derived neutral nanoparticles| US9878056B2|2012-04-02|2018-01-30|Modernatx, Inc.|Modified polynucleotides for the production of cosmetic proteins and peptides| AU2013243949A1|2012-04-02|2014-10-30|Moderna Therapeutics, Inc.|Modified polynucleotides for the production of biologics and proteins associated with human disease| US20150050354A1|2012-04-02|2015-02-19|Moderna Therapeutics, Inc.|Modified polynucleotides for the treatment of otic diseases and conditions| US9572897B2|2012-04-02|2017-02-21|Modernatx, Inc.|Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins| AU2013243955B2|2012-04-02|2018-02-22|Modernatx, Inc.|Modified polynucleotides for the production of oncology-related proteins and peptides| ES2864878T3|2012-06-08|2021-10-14|Translate Bio Inc|Pulmonary RNA Delivery to Non-pulmonary Target Cells| WO2013185067A1|2012-06-08|2013-12-12|Shire Human Genetic Therapies, Inc.|Nuclease resistant polynucleotides and uses thereof| ES2826203T3|2012-06-08|2021-05-17|Ethris Gmbh|Pulmonary supply of messenger RNA| RU2488732C1|2012-07-26|2013-07-27|Общество с ограниченной ответственностью "Новые композитные технологии"|Method of making combined pressure pipe| EP2882706A1|2012-08-13|2015-06-17|Massachusetts Institute of Technology|Amine-containing lipidoids and uses thereof| EP2922554B1|2012-11-26|2022-02-23|ModernaTX, Inc.|Terminally modified rna| US9636301B2|2012-12-04|2017-05-02|Arbutus Biopharma Corporation|In vitro release assay for liposome encapsulated vincristine| EP3628335A1|2012-12-07|2020-04-01|Translate Bio, Inc.|Lipidic nanoparticles for mrna delivery in the lungs| EP2931319B1|2012-12-13|2019-08-21|ModernaTX, Inc.|Modified nucleic acid molecules and uses thereof| US20150315541A1|2012-12-13|2015-11-05|Moderna Therapeutics, Inc.|Modified polynucleotides for altering cell phenotype| AU2013374345A1|2013-01-17|2015-08-06|Moderna Therapeutics, Inc.|Signal-sensor polynucleotides for the alteration of cellular phenotypes| WO2014158795A1|2013-03-12|2014-10-02|Moderna Therapeutics, Inc.|Diagnosis and treatment of fibrosis| WO2014159813A1|2013-03-13|2014-10-02|Moderna Therapeutics, Inc.|Long-lived polynucleotide molecules| EP3431592A1|2013-03-14|2019-01-23|Translate Bio, Inc.|Mrna therapeutic compositions and use to treat diseases and disorders| CA2903487A1|2013-03-14|2014-09-25|Shire Human Genetic Therapies, Inc.|Quantitative assessment for cap efficiency of messenger rna| PT2968586T|2013-03-14|2018-11-13|Ethris Gmbh|Cftr mrna compositions and related methods and uses| US10626439B2|2013-03-14|2020-04-21|Translate Bio, Inc.|Quantitative assessment for cap efficiency of messenger RNA| BR112015022855A2|2013-03-14|2017-11-07|Shire Human Genetic Therapies|compositions and method for producing an in vitro antibody| EA201591229A1|2013-03-14|2016-01-29|Шир Хьюман Дженетик Терапис, Инк.|METHODS OF CLEANING MATRIX RNA| EP3750903A1|2013-03-14|2020-12-16|Translate Bio, Inc.|Ribonucleic acids with 4'-thio-modified nucleotides and related methods| US10258698B2|2013-03-14|2019-04-16|Modernatx, Inc.|Formulation and delivery of modified nucleoside, nucleotide, and nucleic acid compositions| US10077439B2|2013-03-15|2018-09-18|Modernatx, Inc.|Removal of DNA fragments in mRNA production process| US10138507B2|2013-03-15|2018-11-27|Modernatx, Inc.|Manufacturing methods for production of RNA transcripts| US10590161B2|2013-03-15|2020-03-17|Modernatx, Inc.|Ion exchange purification of mRNA| WO2014144039A1|2013-03-15|2014-09-18|Moderna Therapeutics, Inc.|Characterization of mrna molecules| EP2971161B1|2013-03-15|2018-12-26|ModernaTX, Inc.|Ribonucleic acid purification| US10130649B2|2013-03-15|2018-11-20|Translate Bio, Inc.|Synergistic enhancement of the delivery of nucleic acids via blended formulations| US20160017313A1|2013-03-15|2016-01-21|Moderna Therapeutics, Inc.|Analysis of mrna heterogeneity and stability| US8980864B2|2013-03-15|2015-03-17|Moderna Therapeutics, Inc.|Compositions and methods of altering cholesterol levels| WO2014179562A1|2013-05-01|2014-11-06|Massachusetts Institute Of Technology|1,3,5-triazinane-2,4,6-trione derivatives and uses thereof| US9895443B2|2013-06-26|2018-02-20|Massachusetts Institute Of Technology|Multi-tailed lipids and uses thereof| PL3019619T3|2013-07-11|2022-01-10|Modernatx, Inc.|Compositions comprising synthetic polynucleotides encoding crispr related proteins and synthetic sgrnas and methods of use| WO2015011633A1|2013-07-23|2015-01-29|Protiva Biotherapeutics, Inc.|Compositions and methods for delivering messenger rna| RU2723328C2|2013-08-21|2020-06-09|Куревак Аг|Vaccine against respiratory syncytial virus | EP3041938A1|2013-09-03|2016-07-13|Moderna Therapeutics, Inc.|Circular polynucleotides| AU2014315287A1|2013-09-03|2015-03-12|Moderna Therapeutics, Inc.|Chimeric polynucleotides| WO2015048744A2|2013-09-30|2015-04-02|Moderna Therapeutics, Inc.|Polynucleotides encoding immune modulating polypeptides| US20160264614A1|2013-10-02|2016-09-15|Moderna Therapeutics, Inc.|Polynucleotide molecules and uses thereof| US10385088B2|2013-10-02|2019-08-20|Modernatx, Inc.|Polynucleotide molecules and uses thereof| US20160243221A1|2013-10-18|2016-08-25|Moderna Therapeutics, Inc.|Compositions and methods for tolerizing cellular systems| EA201690588A1|2013-10-22|2016-09-30|Шир Хьюман Дженетик Терапис, Инк.|DELIVERY OF MRNA IN THE CNS AND ITS APPLICATION| EP3501605A1|2013-10-22|2019-06-26|Translate Bio, Inc.|Mrna therapy for argininosuccinate synthetase deficiency| EA201992208A1|2013-10-22|2020-07-31|Транслейт Био, Инк.|TREATMENT OF PHENYLKETONURIA USING mRNA| WO2015085318A2|2013-12-06|2015-06-11|Moderna Therapeutics, Inc.|Targeted adaptive vaccines| EP3053585A1|2013-12-13|2016-08-10|Moderna Therapeutics, Inc.|Alternative nucleic acid molecules and uses thereof| JP6704850B2|2013-12-30|2020-06-03|キュアバック アーゲー|Artificial nucleic acid molecule| US20170002060A1|2014-01-08|2017-01-05|Moderna Therapeutics, Inc.|Polynucleotides for the in vivo production of antibodies| EP3116535B1|2014-03-12|2019-08-07|CureVac AG|Combination of vaccination and ox40 agonists| US10405937B2|2014-04-09|2019-09-10|Arbutus Medical Inc.|Drill cover and chuck mechanism| BR112016024644A2|2014-04-23|2017-10-10|Modernatx Inc|nucleic acid vaccines| PL3155129T3|2014-06-10|2019-09-30|Curevac Ag|Method for enhancing rna production| US20170175129A1|2014-06-19|2017-06-22|Moderna Therapeutics, Inc.|Alternative nucleic acid molecules and uses thereof| WO2015196128A2|2014-06-19|2015-12-23|Moderna Therapeutics, Inc.|Alternative nucleic acid molecules and uses thereof| CN111454165A|2014-06-25|2020-07-28|爱康泰生治疗公司|Novel lipid and lipid nanoparticle formulations for delivery of nucleic acids| WO2016005004A1|2014-07-11|2016-01-14|Biontech Rna Pharmaceuticals Gmbh|Stabilization of poly sequence encoding dna sequences| DK3169334T3|2014-07-16|2021-07-05|Ethris Gmbh|RNA FOR USE FOR TREATMENT OF LINK OR TENSE INJURIES| AU2015328012A1|2014-10-02|2017-05-11|Arbutus Biopharma Corporation|Compositions and methods for silencing Hepatitis B virus gene expression| WO2016071857A1|2014-11-07|2016-05-12|Protiva Biotherapeutics, Inc.|Compositions and methods for silencing ebola virus expression| EP3461904A1|2014-11-10|2019-04-03|ModernaTX, Inc.|Alternative nucleic acid molecules containing reduced uracil content and uses thereof| US20170362627A1|2014-11-10|2017-12-21|Modernatx, Inc.|Multiparametric nucleic acid optimization| EP3247363A4|2015-01-21|2018-10-03|Moderna Therapeutics, Inc.|Lipid nanoparticle compositions| WO2016118725A1|2015-01-23|2016-07-28|Moderna Therapeutics, Inc.|Lipid nanoparticle compositions| CA2979998A1|2015-03-20|2016-09-29|Protiva Biotherapeutics, Inc.|Compositions and methods for treating hypertriglyceridemia| WO2016164762A1|2015-04-08|2016-10-13|Moderna Therapeutics, Inc.|Polynucleotides encoding low density lipoprotein receptor egf-a and intracellular domain mutants and methods of using the same| WO2016183366A2|2015-05-12|2016-11-17|Protiva Biotherapeutics, Inc.|Compositions and methods for silencing expression of hepatitis d virus rna| US20180245074A1|2015-06-04|2018-08-30|Protiva Biotherapeutics, Inc.|Treating hepatitis b virus infection using crispr| US10626393B2|2015-06-04|2020-04-21|Arbutus Biopharma Corporation|Delivering CRISPR therapeutics with lipid nanoparticles| WO2016201377A1|2015-06-10|2016-12-15|Moderna Therapeutics, Inc.|Targeted adaptive vaccines| WO2017019891A2|2015-07-29|2017-02-02|Protiva Biotherapeutics, Inc.|Compositions and methods for silencing hepatitis b virus gene expression| CA2998370A1|2015-09-17|2017-03-23|Moderna Therapeutics, Inc.|Polynucleotides containing a stabilizing tail region| US20190078087A1|2015-09-17|2019-03-14|Moderna Therapeutics, Inc.|Polynucleotides containing a morpholino linker| WO2017049074A1|2015-09-18|2017-03-23|Moderna Therapeutics, Inc.|Polynucleotide formulations for use in the treatment of renal diseases| WO2017102010A1|2015-12-17|2017-06-22|Biontech Rna Pharmaceuticals Gmbh|Novel cytokine fusion proteins| US20180371392A1|2015-12-21|2018-12-27|Curevac Ag|Inlay for a culture plate and corresponding method for preparing a culture plate system with such inlay| KR20180107109A|2015-12-22|2018-10-01|큐어백 아게|Method for producing RNA molecular composition| EP3394280A1|2015-12-23|2018-10-31|CureVac AG|Method of rna in vitro transcription using a buffer containing a dicarboxylic acid or tricarboxylic acid or a salt thereof| EP3397613A1|2015-12-30|2018-11-07|Acuitas Therapeutics Inc.|Lipids and lipid nanoparticle formulations for delivery of nucleic acids| US9834510B2|2015-12-30|2017-12-05|Arcturus Therapeutics, Inc.|Aromatic ionizable cationic lipid| KR20210022535A|2018-04-25|2021-03-03|에트리스 게엠베하|Lipid-based formulation for RNA delivery|WO2010053572A2|2008-11-07|2010-05-14|Massachusetts Institute Of Technology|Aminoalcohol lipidoids and uses thereof| ES2734973T3|2009-12-01|2019-12-13|Translate Bio Inc|Administration of mRNA for the increase of proteins and enzymes in human genetic diseases| US8822663B2|2010-08-06|2014-09-02|Moderna Therapeutics, Inc.|Engineered nucleic acids and methods of use thereof| WO2012027675A2|2010-08-26|2012-03-01|Massachusetts Institute Of Technology|Poly, their preparation, and uses thereof| EP2625189B1|2010-10-01|2018-06-27|ModernaTX, Inc.|Engineered nucleic acids and methods of use thereof| US8853377B2|2010-11-30|2014-10-07|Shire Human Genetic Therapies, Inc.|mRNA for use in treatment of human genetic diseases| WO2012135025A2|2011-03-28|2012-10-04|Massachusetts Institute Of Technology|Conjugated lipomers and uses thereof| WO2012135805A2|2011-03-31|2012-10-04|modeRNA Therapeutics|Delivery and formulation of engineered nucleic acids| AU2012253373A1|2011-05-11|2013-11-28|Nanovalent Pharmaceuticals, Inc.|Enhanced growth inhibition of osteosarcoma by cytotoxic polymerized liposomal nanoparticles targeting the alcam cell surface receptor| RU2013154295A|2011-06-08|2015-07-20|Шир Хьюман Дженетик Терапис, Инк.|COMPOSITIONS OF LIPID NANOPARTICLES AND METHODS FOR DELIVERY OF mRNA| WO2012170889A1|2011-06-08|2012-12-13|Shire Human Genetic Therapies, Inc.|Cleavable lipids| US9464124B2|2011-09-12|2016-10-11|Moderna Therapeutics, Inc.|Engineered nucleic acids and methods of use thereof| CN110511939A|2011-10-03|2019-11-29|现代泰克斯公司|Nucleosides, nucleotide and nucleic acid of modification and application thereof| AU2012352180A1|2011-12-16|2014-07-31|Moderna Therapeutics, Inc.|Modified nucleoside, nucleotide, and nucleic acid compositions| KR102272498B1|2011-10-27|2021-07-06|메사추세츠 인스티튜트 오브 테크놀로지|Amino acid derivatives functionalized on the n-terminal capable of forming drug incapsulating microspheres| US10501513B2|2012-04-02|2019-12-10|Modernatx, Inc.|Modified polynucleotides for the production of oncology-related proteins and peptides| US9283287B2|2012-04-02|2016-03-15|Moderna Therapeutics, Inc.|Modified polynucleotides for the production of nuclear proteins| CA2868030C|2012-03-29|2021-05-25|Shire Human Genetic Therapies, Inc.|Lipid-derived neutral nanoparticles| AU2013243949A1|2012-04-02|2014-10-30|Moderna Therapeutics, Inc.|Modified polynucleotides for the production of biologics and proteins associated with human disease| US9878056B2|2012-04-02|2018-01-30|Modernatx, Inc.|Modified polynucleotides for the production of cosmetic proteins and peptides| US9572897B2|2012-04-02|2017-02-21|Modernatx, Inc.|Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins| AU2013243955B2|2012-04-02|2018-02-22|Modernatx, Inc.|Modified polynucleotides for the production of oncology-related proteins and peptides| ES2864878T3|2012-06-08|2021-10-14|Translate Bio Inc|Pulmonary RNA Delivery to Non-pulmonary Target Cells| WO2013185067A1|2012-06-08|2013-12-12|Shire Human Genetic Therapies, Inc.|Nuclease resistant polynucleotides and uses thereof| EP2882706A1|2012-08-13|2015-06-17|Massachusetts Institute of Technology|Amine-containing lipidoids and uses thereof| EP2885419A4|2012-08-14|2016-05-25|Moderna Therapeutics Inc|Enzymes and polymerases for the synthesis of rna| EP2922554B1|2012-11-26|2022-02-23|ModernaTX, Inc.|Terminally modified rna| EP3628335A1|2012-12-07|2020-04-01|Translate Bio, Inc.|Lipidic nanoparticles for mrna delivery in the lungs| EP3750903A1|2013-03-14|2020-12-16|Translate Bio, Inc.|Ribonucleic acids with 4'-thio-modified nucleotides and related methods| PT2968586T|2013-03-14|2018-11-13|Ethris Gmbh|Cftr mrna compositions and related methods and uses| US10258698B2|2013-03-14|2019-04-16|Modernatx, Inc.|Formulation and delivery of modified nucleoside, nucleotide, and nucleic acid compositions| EA201591229A1|2013-03-14|2016-01-29|Шир Хьюман Дженетик Терапис, Инк.|METHODS OF CLEANING MATRIX RNA| BR112015022855A2|2013-03-14|2017-11-07|Shire Human Genetic Therapies|compositions and method for producing an in vitro antibody| US8980864B2|2013-03-15|2015-03-17|Moderna Therapeutics, Inc.|Compositions and methods of altering cholesterol levels| US10130649B2|2013-03-15|2018-11-20|Translate Bio, Inc.|Synergistic enhancement of the delivery of nucleic acids via blended formulations| WO2014179562A1|2013-05-01|2014-11-06|Massachusetts Institute Of Technology|1,3,5-triazinane-2,4,6-trione derivatives and uses thereof| WO2015011633A1|2013-07-23|2015-01-29|Protiva Biotherapeutics, Inc.|Compositions and methods for delivering messenger rna| EP3041938A1|2013-09-03|2016-07-13|Moderna Therapeutics, Inc.|Circular polynucleotides| AU2014315287A1|2013-09-03|2015-03-12|Moderna Therapeutics, Inc.|Chimeric polynucleotides| WO2015048744A2|2013-09-30|2015-04-02|Moderna Therapeutics, Inc.|Polynucleotides encoding immune modulating polypeptides| US10323076B2|2013-10-03|2019-06-18|Modernatx, Inc.|Polynucleotides encoding low density lipoprotein receptor| EA201992208A1|2013-10-22|2020-07-31|Транслейт Био, Инк.|TREATMENT OF PHENYLKETONURIA USING mRNA| KR102096796B1|2013-10-22|2020-05-27|샤이어 휴먼 지네틱 테라피즈 인크.|Lipid formulations for delivery of messenger rna| EP3501605A1|2013-10-22|2019-06-26|Translate Bio, Inc.|Mrna therapy for argininosuccinate synthetase deficiency| EA201690588A1|2013-10-22|2016-09-30|Шир Хьюман Дженетик Терапис, Инк.|DELIVERY OF MRNA IN THE CNS AND ITS APPLICATION| US10202601B2|2013-11-22|2019-02-12|Mina Therapeutics Limited|C/EBPα short activating RNA compositions and methods of use| MX2016011686A|2014-03-09|2016-11-07|Univ Pennsylvania|Compositions useful in treatment of ornithine transcarbamylasedeficiency.| ES2774552T3|2014-03-24|2020-07-21|Translate Bio Inc|MRNA therapy for treating eye diseases| BR112016024644A2|2014-04-23|2017-10-10|Modernatx Inc|nucleic acid vaccines| JP6571679B2|2014-04-25|2019-09-04|トランスレイト バイオ, インコーポレイテッド|Method for purifying messenger RNA| US10392438B2|2014-05-16|2019-08-27|Pfizer Inc.|Bispecific antibodies| EP3587409A1|2014-05-30|2020-01-01|Translate Bio, Inc.|Biodegradable lipids for delivery of nucleic acids| EP3160959A4|2014-06-24|2018-02-28|Translate Bio, Inc.|Stereochemically enriched compositions for delivery of nucleic acids| CN114146063A|2014-07-02|2022-03-08|川斯勒佰尔公司|Encapsulation of messenger RNA| EP3164379A1|2014-07-02|2017-05-10|Massachusetts Institute of Technology|Polyamine-fatty acid derived lipidoids and uses thereof| EP3171895A1|2014-07-23|2017-05-31|Modernatx, Inc.|Modified polynucleotides for the production of intrabodies| EP3884964A1|2014-12-05|2021-09-29|Translate Bio, Inc.|Messenger rna therapy for treatment of articular disease| JP6650943B2|2014-12-22|2020-02-19|コデクシス, インコーポレイテッド|Human α-galactosidase variant| JP2018509387A|2015-01-21|2018-04-05|フェーズアールエックス インコーポレイテッド|Methods, compositions, and systems for delivering therapeutic and diagnostic agents to cells| JP6929791B2|2015-02-09|2021-09-01|デューク ユニバーシティ|Compositions and methods for epigenome editing| US10172924B2|2015-03-19|2019-01-08|Translate Bio, Inc.|MRNA therapy for pompe disease| JP6851319B2|2015-04-27|2021-03-31|ザ・トラステイーズ・オブ・ザ・ユニバーシテイ・オブ・ペンシルベニア|Dual AAV vector system for CRISPR / Cas9 mediated modification of human disease| GB201508025D0|2015-05-11|2015-06-24|Ucl Business Plc|Fabry disease gene therapy| EP3310764A1|2015-06-19|2018-04-25|Massachusetts Institute of Technology|Alkenyl substituted 2,5-piperazinediones and their use in compositions for delivering an agent to a subject or cell| WO2017015463A2|2015-07-21|2017-01-26|Modernatx, Inc.|Infectious disease vaccines| WO2017015637A1|2015-07-22|2017-01-26|Duke University|High-throughput screening of regulatory element function with epigenome editing technologies| CA2998810A1|2015-09-17|2017-03-23|Modernatx, Inc.|Compounds and compositions for intracellular delivery of therapeutic agents| US10849920B2|2015-10-05|2020-12-01|Modernatx, Inc.|Methods for therapeutic administration of messenger ribonucleic acid drugs| WO2017066573A1|2015-10-14|2017-04-20|Shire Human Genetic Therapies, Inc.|Modification of rna-related enzymes for enhanced production| BR112018008078A2|2015-10-22|2018-11-13|Modernatx Inc|broad spectrum influenza virus vaccine| EP3364950A4|2015-10-22|2019-10-23|ModernaTX, Inc.|Tropical disease vaccines| CA3002922A1|2015-10-22|2017-04-27|Modernatx, Inc.|Human cytomegalovirus vaccine| CN108513575A|2015-10-23|2018-09-07|哈佛大学的校长及成员们|Nucleobase editing machine and application thereof| EP3386484A4|2015-12-10|2019-07-31|ModernaTX, Inc.|Compositions and methods for delivery of therapeutic agents| JP2019508371A|2015-12-22|2019-03-28|モデルナティエックス インコーポレイテッドModernaTX,Inc.|Compounds and compositions for intracellular delivery of drugs| EP3394093B1|2015-12-23|2022-01-26|Modernatx, Inc.|Methods of using ox40 ligand encoding polynucleotides| US20190241658A1|2016-01-10|2019-08-08|Modernatx, Inc.|Therapeutic mRNAs encoding anti CTLA-4 antibodies| US20210206818A1|2016-01-22|2021-07-08|Modernatx, Inc.|Messenger ribonucleic acids for the production of intracellular binding polypeptides and methods of use thereof| DK3436589T3|2016-03-31|2020-11-23|Ethris Gmbh|NEW MINIMAL UTR SEQUENCES| WO2017177169A1|2016-04-08|2017-10-12|Rana Therapeutics, Inc.|Multimeric coding nucleic acid and uses thereof| WO2017180917A2|2016-04-13|2017-10-19|Modernatx, Inc.|Lipid compositions and their uses for intratumoral polynucleotide delivery| US20180126003A1|2016-05-04|2018-05-10|Curevac Ag|New targets for rna therapeutics| US20190298658A1|2016-05-18|2019-10-03|Modernatx, Inc.|Polynucleotides Encoding Cystic Fibrosis Transmembrane Conductance Regulator for the Treatment of Cystic Fibrosis| RU2018142589A3|2016-05-18|2020-09-18| WO2017201328A1|2016-05-18|2017-11-23|Modernatx, Inc.|POLYNUCLEOTIDES ENCODING α-GALACTOSIDASE A FOR THE TREATMENT OF FABRY DISEASE| JP2019516710A|2016-05-18|2019-06-20|モデルナティーエックス, インコーポレイテッド|Polynucleotide encoding interleukin-12and use thereof| CN109312313A|2016-06-13|2019-02-05|川斯勒佰尔公司|For treating the mRNA therapy of ornithine transcarbamylase deficiency disease| JP2019519568A|2016-06-30|2019-07-11|アルブータス・バイオファーマー・コーポレイション|Compositions and methods for delivering messenger RNA| WO2018033454A1|2016-08-19|2018-02-22|Fundación Para La Investigación Biomédica Del Hospital Universitario Ramón Y Cajal|Mir-127 agents for use in the treatment of renal fibrosis| CA3041307A1|2016-10-21|2018-04-26|Giuseppe Ciaramella|Human cytomegalovirus vaccine| AU2017356190A1|2016-11-10|2019-05-16|Translate Bio, Inc.|Subcutaneous delivery of messenger RNA| US20180153822A1|2016-11-10|2018-06-07|Translate Bio, Inc.|Process of Preparing mRNA-Loaded Lipid Nanoparticles| JP2020500857A|2016-11-23|2020-01-16|メイヨ・ファウンデーション・フォー・メディカル・エデュケーション・アンド・リサーチ|Particle-mediated delivery of biologics| EP3565605A1|2017-01-03|2019-11-13|ethris GmbH|Ornithine transcarbamylase coding polyribonucleotides and formulations thereof| EP3565891A4|2017-01-09|2020-07-29|Whitehead Institute for Biomedical Research|Methods of altering gene expression by perturbing transcription factor multimers that structure regulatory loops| WO2018151816A1|2017-02-16|2018-08-23|Modernatx, Inc.|High potency immunogenic compositions| WO2018157153A1|2017-02-27|2018-08-30|Translate Bio, Inc.|Large scale synthesis of messenger rna| CA3054323A1|2017-02-27|2018-08-30|Translate Bio, Inc.|Methods for purification of messenger rna| AU2018224843A1|2017-02-27|2019-09-19|Translate Bio, Inc.|Methods for purification of messenger RNA| AU2018224326A1|2017-02-27|2019-09-19|Translate Bio, Inc.|Novel codon-optimized CFTR mRNA| MA47688A|2017-02-28|2020-01-08|Univ Pennsylvania|USEFUL COMPOSITIONS IN THE TREATMENT OF SPINAL AMYOTROPHY| US10961184B2|2017-03-07|2021-03-30|Translate Bio, Inc.|Polyanionic delivery of nucleic acids| WO2018170322A1|2017-03-15|2018-09-20|Modernatx, Inc.|Crystal forms of amino lipids| CA3056132A1|2017-03-15|2018-09-20|Modernatx, Inc.|Compounds and compositions for intracellular delivery of therapeutic agents| US11268082B2|2017-03-23|2022-03-08|President And Fellows Of Harvard College|Nucleobase editors comprising nucleic acid programmable DNA binding proteins| CA3063531A1|2017-05-16|2018-11-22|Translate Bio, Inc.|Treatment of cystic fibrosis by delivery of codon-optimized mrna encoding cftr| US20200208145A1|2017-05-18|2020-07-02|Modernatx, Inc.|Modified messenger rna comprising functional rna elements| JP2020520640A|2017-05-18|2020-07-16|モデルナティーエックス, インコーポレイテッド|Polynucleotides encoding linked interleukin-12polypeptides and uses thereof| MA49395A|2017-06-14|2020-04-22|Modernatx Inc|POLYNUCLEOTIDES COAGULATION FACTOR VIII CODING| US20200131498A1|2017-06-14|2020-04-30|Modernatx, Inc.|Polynucleotides encoding methylmalonyl-coa mutase| US10034951B1|2017-06-21|2018-07-31|New England Biolabs, Inc.|Use of thermostable RNA polymerases to produce RNAs having reduced immunogenicity| CA3075205A1|2017-09-08|2019-03-14|Mina Therapeutics Limited|Stabilized hnf4a sarna compositions and methods of use| WO2019048645A1|2017-09-08|2019-03-14|Mina Therapeutics Limited|Stabilized cebpa sarna compositions and methods of use| EP3681514A4|2017-09-14|2021-07-14|ModernaTX, Inc.|Zika virus rna vaccines| WO2019067999A1|2017-09-29|2019-04-04|Intellia Therapeutics, Inc.|In vitro method of mrna delivery using lipid nanoparticles| JP2021504335A|2017-11-22|2021-02-15|モダーナティエックス・インコーポレイテッドModernaTX, Inc.|A polynucleotide encoding phenylalanine hydroxylase for the treatment of phenylketonuria| WO2019104195A1|2017-11-22|2019-05-31|Modernatx, Inc.|Polynucleotides encoding propionyl-coa carboxylase alpha and beta subunits for the treatment of propionic acidemia| MA50803A|2017-11-22|2020-09-30|Modernatx Inc|POLYNUCLEOTIDES CODING ORNITHINE TRANSCARBAMYLASE FOR THE TREATMENT OF UREA CYCLE DISORDERS| EP3727428A1|2017-12-20|2020-10-28|Translate Bio, Inc.|Improved composition and methods for treatment of ornithine transcarbamylase deficiency| MA51523A|2018-01-05|2020-11-11|Modernatx Inc|POLYNUCLEOTIDES CODING ANTI-BODY ANTI-CHIKUNGUNYA VIRUS| WO2019152802A1|2018-02-02|2019-08-08|Translate Bio, Inc.|Cationic polymers| EP3773745A1|2018-04-11|2021-02-17|ModernaTX, Inc.|Messenger rna comprising functional rna elements| WO2019197845A1|2018-04-12|2019-10-17|Mina Therapeutics Limited|Sirt1-sarna compositions and methods of use| US20210220449A1|2018-05-15|2021-07-22|Translate Bio, Inc.|Subcutaneous Delivery of Messenger RNA| CN112384523A|2018-05-16|2021-02-19|川斯勒佰尔公司|Cationic lipid of ribose| EP3796893A1|2018-05-23|2021-03-31|Modernatx, Inc.|Delivery of dna| WO2020237227A1|2019-05-22|2020-11-26|Massachusetts Institute Of Technology|Circular rna compositions and methods| WO2020023390A1|2018-07-25|2020-01-30|Modernatx, Inc.|Mrna based enzyme replacement therapy combined with a pharmacological chaperone for the treatment of lysosomal storage disorders| IL267923D0|2018-08-02|2020-02-27|Grifols Worldwide Operations Ltd|Composition comprising highly-concentrated alpha-1 proteinase inhibitor and method for obtaining thereof| US10842885B2|2018-08-20|2020-11-24|Ucl Business Ltd|Factor IX encoding nucleotides| KR20210060480A|2018-08-24|2021-05-26|트랜슬레이트 바이오 인코포레이티드|Method for purifying messenger RNA| WO2020047201A1|2018-09-02|2020-03-05|Modernatx, Inc.|Polynucleotides encoding very long-chain acyl-coa dehydrogenase for the treatment of very long-chain acyl-coa dehydrogenase deficiency| GB2592505A|2018-09-04|2021-09-01|Univ Texas|Compositions and methods for organ specific delivery of nucleic acids| JP2021535226A|2018-09-04|2021-12-16|ザ ボード オブ リージェンツ オブ ザ ユニバーシティー オブ テキサス システム|Compositions and Methods for Organ-Specific Delivery of Nucleic Acids| EP3849594A2|2018-09-13|2021-07-21|Modernatx, Inc.|Polynucleotides encoding branched-chain alpha-ketoacid dehydrogenase complex e1-alpha, e1-beta, and e2 subunits for the treatment of maple syrup urine disease| CA3112208A1|2018-09-13|2020-03-19|Modernatx, Inc.|Polynucleotides encoding glucose-6-phosphatase for the treatment of glycogen storage disease| WO2020056239A1|2018-09-14|2020-03-19|Modernatx, Inc.|Polynucleotides encoding uridine diphosphate glycosyltransferase 1 family, polypeptide a1 for the treatment of crigler-najjar syndrome| EP3856233A1|2018-09-27|2021-08-04|Modernatx, Inc.|Polynucleotides encoding arginase 1 for the treatment of arginase deficiency| US11072808B2|2018-10-04|2021-07-27|New England Biolabs, Inc.|Methods and compositions for increasing capping efficiency of transcribed RNA| CA3114892A1|2018-10-04|2020-04-09|New England Biolabs, Inc.|Methods and compositions for increasing capping efficiency of transcribed rna| US20210388338A1|2018-11-08|2021-12-16|Translate Bio, Inc.|Methods and Compositions for Messenger RNA Purification| WO2020097409A2|2018-11-08|2020-05-14|Modernatx, Inc.|Use of mrna encoding ox40l to treat cancer in human patients| WO2020102172A2|2018-11-12|2020-05-22|Translate Bio, Inc.|Methods for inducing immune tolerance| AU2019384557A1|2018-11-21|2021-06-10|Translate Bio, Inc.|Treatment of cystic fibrosis by delivery of nebulized mRNA encoding CFTR| CA3118034A1|2018-12-21|2020-06-25|Curevac Ag|Rna for malaria vaccines| EP3920950A1|2019-02-08|2021-12-15|CureVac AG|Coding rna administered into the suprachoroidal space in the treatment of ophtalmic diseases| CN114127285A|2019-03-19|2022-03-01|布罗德研究所股份有限公司|Methods and compositions for editing nucleotide sequences| WO2020208361A1|2019-04-12|2020-10-15|Mina Therapeutics Limited|Sirt1-sarna compositions and methods of use| EP3965797A1|2019-05-08|2022-03-16|AstraZeneca AB|Compositions for skin and wounds and methods of use thereof| AU2020274758A1|2019-05-14|2021-12-23|Translate Bio, Inc.|Improved process of preparing mRNA-loaded lipid nanoparticles| WO2020254535A1|2019-06-18|2020-12-24|Curevac Ag|Rotavirus mrna vaccine| AU2020297594A1|2019-06-21|2022-02-03|Kernal Biologics, Inc.|Engineered oncoselective protein expression| WO2020263883A1|2019-06-24|2020-12-30|Modernatx, Inc.|Endonuclease-resistant messenger rna and uses thereof| WO2020263985A1|2019-06-24|2020-12-30|Modernatx, Inc.|Messenger rna comprising functional rna elements and uses thereof| WO2020262150A1|2019-06-24|2020-12-30|国立大学法人北海道大学|Lipid nanoparticle| WO2021009336A1|2019-07-18|2021-01-21|INSERM |Methods for inducing full ablation of hematopoiesis| AU2020316335A1|2019-07-19|2022-02-17|University Of Florida Research Foundation, Incorporated|Multilamellar RNA nanoparticles| WO2021021988A1|2019-07-30|2021-02-04|Translate Bio, Inc.|Treatment of cystic fibrosis by delivery of nebulized mrna encoding cftr| WO2021028439A1|2019-08-14|2021-02-18|Curevac Ag|Rna combinations and compositions with decreased immunostimulatory properties| US11066355B2|2019-09-19|2021-07-20|Modernatx, Inc.|Branched tail lipid compounds and compositions for intracellular delivery of therapeutic agents| US20210106527A1|2019-10-09|2021-04-15|Translate Bio, Inc.|Compositions, methods and uses of messenger rna| EP3920976A2|2019-12-04|2021-12-15|Orna Therapeutics, Inc.|Circular rna compositions and methods| WO2021142355A1|2020-01-10|2021-07-15|Andranik Andrew Aprikyan|Nanoparticles for expression of genes of interest and/or regulation of signaling pathways| WO2021154763A1|2020-01-28|2021-08-05|Modernatx, Inc.|Coronavirus rna vaccines| WO2021156267A1|2020-02-04|2021-08-12|Curevac Ag|Coronavirus vaccine| WO2021159040A2|2020-02-07|2021-08-12|Modernatx, Inc.|Sars-cov-2 mrna domain vaccines| US20210275689A1|2020-02-25|2021-09-09|Translate Bio, Inc.|Processes of preparing mrna-loaded lipid nanoparticles| WO2021222304A1|2020-04-27|2021-11-04|Modernatx, Inc.|Sars-cov-2 rna vaccines| WO2021236855A1|2020-05-19|2021-11-25|Orna Therapeutics, Inc.|Circular rna compositions and methods| WO2021231579A1|2020-05-12|2021-11-18|The Trustees Of The University Of Pennsylvania|Compositions for drg-specific reduction of transgene expression| WO2021159130A2|2020-05-15|2021-08-12|Modernatx, Inc.|Coronavirus rna vaccines and methods of use| WO2021239880A1|2020-05-29|2021-12-02|Curevac Ag|Nucleic acid based combination vaccines| WO2021247507A1|2020-06-01|2021-12-09|Modernatx, Inc.|Phenylalanine hydroxylase variants and uses thereof| WO2021252354A1|2020-06-12|2021-12-16|University Of Rochester|ENCODING AND EXPRESSION OF ACE-tRNAs| WO2021257668A1|2020-06-17|2021-12-23|The Trustees Of The University Of Pennsylvania|Compositions and methods for treatment of gene therapy patients| WO2022015715A1|2020-07-13|2022-01-20|The Trustees Of The University Of Pennsylvania|Compositions useful for treatment of charcot-marie-tooth disease| WO2022023559A1|2020-07-31|2022-02-03|Curevac Ag|Nucleic acid encoded antibody mixtures| WO2022043551A2|2020-08-31|2022-03-03|Curevac Ag|Multivalent nucleic acid based coronavirus vaccines|
法律状态:
2020-11-17| B08F| Application dismissed because of non-payment of annual fees [chapter 8.6 patent gazette]|Free format text: REFERENTE AS 6A, 7A E 8A ANUIDADES. | 2021-03-09| B08K| Patent lapsed as no evidence of payment of the annual fee has been furnished to inpi [chapter 8.11 patent gazette]|Free format text: EM VIRTUDE DO ARQUIVAMENTO PUBLICADO NA RPI 2602 DE 17-11-2020 E CONSIDERANDO AUSENCIA DE MANIFESTACAO DENTRO DOS PRAZOS LEGAIS, INFORMO QUE CABE SER MANTIDO O ARQUIVAMENTO DO PEDIDO DE PATENTE, CONFORME O DISPOSTO NO ARTIGO 12, DA RESOLUCAO 113/2013. | 2021-12-07| B350| Update of information on the portal [chapter 15.35 patent gazette]|
优先权:
[返回顶部]
申请号 | 申请日 | 专利标题 US201161494881P| true| 2011-06-08|2011-06-08| US61/494,881|2011-06-08| PCT/US2012/041724|WO2012170930A1|2011-06-08|2012-06-08|Lipid nanoparticle compositions and methods for mrna delivery| 相关专利
Sulfonates, polymers, resist compositions and patterning process
Washing machine
Washing machine
Device for fixture finishing and tension adjusting of membrane
Structure for Equipping Band in a Plane Cathode Ray Tube
Process for preparation of 7 alpha-carboxyl 9, 11-epoxy steroids and intermediates useful therein an
国家/地区
|