WO2012045075A1 - Modified nucleosides, nucleotides, and nucleic acids, and uses thereof - Google Patents

Modified nucleosides, nucleotides, and nucleic acids, and uses thereof Download PDF

Info

Publication number
WO2012045075A1
WO2012045075A1 PCT/US2011/054617 US2011054617W WO2012045075A1 WO 2012045075 A1 WO2012045075 A1 WO 2012045075A1 US 2011054617 W US2011054617 W US 2011054617W WO 2012045075 A1 WO2012045075 A1 WO 2012045075A1
Authority
WO
WIPO (PCT)
Prior art keywords
nucleic acid
alkyl
compound
formula
nucleotide
Prior art date
Application number
PCT/US2011/054617
Other languages
French (fr)
Inventor
Jason Schrum
Original Assignee
Jason Schrum
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=45893552&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO2012045075(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Jason Schrum filed Critical Jason Schrum
Priority to EP22173763.8A priority Critical patent/EP4108671A1/en
Priority to EP19177059.3A priority patent/EP3590949B1/en
Priority to CA2813466A priority patent/CA2813466A1/en
Priority to EP11830061.5A priority patent/EP2622064B1/en
Priority to ES11830061T priority patent/ES2737960T3/en
Publication of WO2012045075A1 publication Critical patent/WO2012045075A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/005Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'active' part of the composition delivered, i.e. the nucleic acid delivered
    • A61K48/0066Manipulation of the nucleic acid to modify its expression pattern, e.g. enhance its duration of expression, achieved by the presence of particular introns in the delivered nucleic acid
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H19/00Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof
    • C07H19/02Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof sharing nitrogen
    • C07H19/04Heterocyclic radicals containing only nitrogen atoms as ring hetero atom
    • C07H19/06Pyrimidine radicals
    • C07H19/10Pyrimidine radicals with the saccharide radical esterified by phosphoric or polyphosphoric acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H21/00Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids
    • C07H21/02Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids with ribosyl as saccharide radical
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2887Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against CD20
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/10Processes for the isolation, preparation or purification of DNA or RNA
    • C12N15/102Mutagenizing nucleic acids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • C12N15/1136Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against growth factors, growth regulators, cytokines, lymphokines or hormones
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • C12N15/1138Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against receptors or cell surface proteins
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/67General methods for enhancing the expression
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12PFERMENTATION OR ENZYME-USING PROCESSES TO SYNTHESISE A DESIRED CHEMICAL COMPOUND OR COMPOSITION OR TO SEPARATE OPTICAL ISOMERS FROM A RACEMIC MIXTURE
    • C12P21/00Preparation of peptides or proteins
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/558Immunoassay; Biospecific binding assay; Materials therefor using diffusion or migration of antigen or antibody
    • G01N33/559Immunoassay; Biospecific binding assay; Materials therefor using diffusion or migration of antigen or antibody through a gel, e.g. Ouchterlony technique
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/33Chemical structure of the base
    • C12N2310/334Modified C
    • C12N2310/33415-Methylcytosine
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/33Chemical structure of the base
    • C12N2310/335Modified T or U

Definitions

  • RNAs are synthesized from four basic ribonucleotides: ATP, CTP, UTP and GTP, but may contain post-transcriptionally modified nucleotides. Further, approximately one hundred different nucleoside modifications have been identified in RNA (Rozenski, J, Crain, P, and McCloskey, J. (1999). The RNA Modification Database: 1999 update. Nucl Acids Res 27: 196-197). The role of nucleoside modifications on the irnmuno- stimulatory potential, stability, and on the translation efficiency of RNA, and the consequent benefits to this for enhancing protein expression and producing therapeutics however, is unclear.
  • heterologous deoxyribonucleic acid (DNA) introduced into a cell can be inherited by daughter cells (whether or not the heterologous DNA has integrated into the chromosome) or by offspring. Introduced DNA can integrate into host cell genomic DNA at some frequency, resulting in alterations and/or damage to the host cell genomic DNA.
  • multiple steps must occur before a protein is made. Once inside the cell, DNA must be transported into the nucleus where it is transcribed into RNA. The RNA transcribed from DNA must then enter the cytoplasm where it is translated into protein. This need for multiple processing steps creates lag times before the generation of a protein of interest. Further, it is difficult to obtain DNA expression in cells; frequently DNA enters cells but is not expressed or not expressed at reasonable rates or concentrations. This can be a particular problem when DNA is introduced into cells such as primary cells or modified cell lines.
  • modified nucleosides modified nucleotides, and modified nucleic acids which can exhibit a reduced innate immune response when introduced into a population of cells, both in vivo and ex vivo.
  • these modified nucleosides, modified nucleotides, and modified nucleic acids described herein can disrupt binding of a major groove interacting partner with the nucleic acid. Because of the reduced immunogenicity and the decrease in major groove interactions, these modified nucleosides, modified nucleotides, and modified nucleic acids can be more efficient during protein production than, e.g., unmodified nucleic acids.
  • the present disclosure provides compounds comprising nucleotides that can disrupt binding of a major groove binding partner with a nucleic acid, wherein the nucleotide has decreased binding affinity to the major groove binding partner.
  • the present disclosure further provides nucleic acid sequences of at least two nucleotides comprising a compound of Formula I-d:
  • compositions comprising at least one compound of Formula I.
  • the present disclosure further provides pharmaceutical compositions comprising a compound of Formula I.
  • the present disclosure further provides methods of preparing nucleic acid sequences of at least two nucleotides of a compound of Formula I-d.
  • the present disclosure further provides methods of amplifying nucleic acid sequences of at least two nucleotides of a compound of Formula I-d.
  • kits comprising a compound of Formula I.
  • FIGs. 1A and IB depict images of non-denaturing agarose gels of each in vitro- transcribed modified RNA.
  • FIGs. 2A and 2B depict images of an Enzyme-linked immunosorbent assay (ELISA) for Human Granulocyte-Colony Stimulating Factor (G-CSF) of in vitro transfected Human
  • ELISA Enzyme-linked immunosorbent assay
  • G-CSF Human Granulocyte-Colony Stimulating Factor
  • Keratinocyte cells with each indicated modRNA encoding human G-CSF and the line indicates a saturating level of maximum detectable limit of secreted G-CSF in the assay.
  • FIGs. 3A-N depict line graphs of a series of Enzyme-linked immunosorbent assays (ELISA) for Human Granulocyte-Colony Stimulating Factor (G-CSF) secreted from in vitro- transfected Human Keratinocyte cells at different time points with each indicated human G-CSF- encoding modRNA at the indicated doses.
  • the line indicates a saturating level of maximum detectable limit of secreted G-CSF in the assay.
  • FIGs. 4A and 4B depict bar graphs of a series of Enzyme-linked immunosorbent assays (ELISA) for endogenous cellular human Tumor Necrosis Factor-a (TNF- a) secreted from in vzYro-transfected Human Keratinocyte cells at 24 hours with each indicated hu-G-CSF-encoding modRNA at increasing doses.
  • ELISA Enzyme-linked immunosorbent assays
  • FIGs. 4C and 4D depict bar graphs of a series of Enzyme-linked immunosorbent assays (ELISA) for endogenous cellular human Interferon- ⁇ (IFN- ⁇ ) secreted from in vzYro-transfected Human Keratinocyte cells at 24 hours with each indicated hu-G-CSF-encoding modRNA at increasing doses.
  • ELISA Enzyme-linked immunosorbent assays
  • FIGs. 4E and 4F depict bar graphs of a series of Enzyme-linked immunosorbent assays (ELISA) for human-G-CSF secreted from in vzYro-transfected Human Keratinocyte cells at 24 hours with each indicated hu-G-CSF-encoding modRNA at increasing doses.
  • ELISA Enzyme-linked immunosorbent assays
  • FIG. 5A is a table showing results from an Enzyme-linked immunosorbent assay
  • ELISA for human-G-CSF secreted from in vzYro-transfected Human Keratinocyte cells sampled from individual wells in a co-culture 24-well tissue culture plate 42 hours post-transfection with 750ng of each indicated hu-G-CSF-encoding modRNA.
  • FIG. 5B depicts an image of an agarose gel of RT-PCR hu-G-CSF modRNA products from co-culture cell extracts 42 hours post-transfection of the human keratinocyte feeder layer with hu-G-CSG modRNA and the un-transfected Kasumi-1 and KG-1 insert culture cells.
  • FIGs. 5C and 5D depict graphs of results from a hu-G-CSF-modRNA-induced cell proliferation assay of Kasumi-1 (FIG. 5C) and KG-1 (FIG. 5D) cells normalized to untransfected cells.
  • Kasumi-1 FIG. 5C
  • KG-1 FIG. 5D
  • FIGs. 6A-L depict graphs of the UV absorbance spectra for exemplary modRNA molecules that incorporate the indicated modified nucleotide.
  • the present disclosure provides, inter alia, modified nucleosides, modified nucleotides, and modified nucleic acids that exhibit a reduced innate immune response when introduced into a population of cells.
  • the modified nucleosides, modified nucleotides, and modified nucleic acids can be chemically modified on the major groove face, thereby disrupting major groove binding partner interactions, which cause innate immune responses.
  • exogenous unmodified nucleic acids particularly viral nucleic acids
  • IFN interferon
  • RNA ribonucleic acid
  • nucleic acids characterized by integration into a target cell are generally imprecise in their expression levels, deleteriously transferable to progeny and neighbor cells, and suffer from the substantial risk of causing mutation.
  • nucleic acids encoding useful polypeptides capable of modulating a cell's function and/or activity are provided herein in part, and methods of making and using these nucleic acids and polypeptides. As described herein, these nucleic acids are capable of reducing the innate immune activity of a population of cells into which they are introduced, thus increasing the efficiency of protein production in that cell population. Further, one or more additional advantageous activities and/or properties of the nucleic acids and proteins of the present disclosure are described.
  • modified nucleosides, modified nucleotides, and modified nucleic acids described herein can be modified on the major groove face. These major groove modifications can allow for alterations, e.g. a decrease, in the interaction of the modified nucleosides, modified nucleotides, and modified nucleic acids with a binding groove partner.
  • the present disclosure provides compounds comprising a nucleotide that can disrupts binding of a major groove interacting, e.g. binding, partner with a nucleic acid, wherein the nucleotide has decreased binding affinity to major groove interacting, e.g. binding, partners.
  • the present disclosure provides compounds comprising a nucleotide that contains chemical modifications, wherein the nucleotide can have altered binding to major groove interacting, e.g. binding, partners.
  • the chemical modifications are located on the major groove face of the nucleobase, and wherein the chemical modifications can include replacing or substituting an atom of a pyrimidine nucleobase with an amine, an SH, an alkyl (e.g., methyl or ethyl), or a halo (e.g., chloro or fluoro).
  • the chemical modifications can be located on the major groove face of the nucleobase, and wherein the chemical modification can include replacing or substituting an atom of a pyrimidine nucleobase with an amine, an SH, a methyl or ethyl, or a chloro or fluoro.
  • the chemical modifications can be located on the sugar moiety of the nucleotide.
  • the chemical modifications can be located on the phosphate backbone of the nucleotide.
  • the chemical modifications can alter the electrochemistry on the major groove face of the nucleotide.
  • the present disclosure provides nucleotides that contain chemical modifications, wherein the nucleotide reduces the cellular innate immune response, as compared to the cellular innate immune induced by a corresponding unmodified nucleic acid.
  • the present disclosure provides nucleic acid sequences comprising at least two nucleotides, the nucleic acid sequence comprising a nucleotide that disrupts binding of a major groove interacting partner with the nucleic acid sequence, wherein the nucleotide has decreased binding affinity to the major groove binding partner.
  • compositions comprising a compound as described herein.
  • the composition is a reaction mixture.
  • the composition is a pharmaceutical composition.
  • the composition is a cell culture.
  • compositions further comprise an RNA polymerase and a cDNA template.
  • compositions further comprise a nucleotide selected from the group consisting of adenosine, cytosine, guanosine, and uracil.
  • the present disclosure provides for methods of synthesizing a pharmaceutical nucleic acid, comprising providing a complementary deoxyribonucleic acid (cDNA) that encodes a pharmaceutical protein of interest; selecting a nucleotide that is known to disrupt a binding of a major groove binding partner with a nucleic acid, wherein the nucleotide has decreased binding affinity to the major groove binding partner; and contacting the provided cDNA and the selected nucleotide with an RNA polymerase, under conditions such that the pharmaceutical nucleic acid is synthesized.
  • cDNA complementary deoxyribonucleic acid
  • the pharmaceutical nucleic acid is a ribonucleic acid (RNA).
  • the present disclosure provides for methods of making a
  • composition comprising a physiologically active secreted protein, comprising transfecting a first population of human cells with a pharmaceutical nucleic acid made by the methods described herein, wherein the secreted protein is active upon a second population of human cells.
  • the secreted protein is capable of interacting, e.g. binding, with a receptor on the surface of at least one cell present in the second population.
  • the secreted protein is Granulocyte-Colony Stimulating Factor (G-1)
  • the second population contains myeloblast cells that express the G-CSF receptor.
  • the present disclosure provides for methods of making a
  • pharmaceutical formulation comprising human cells comprising a physiologically active secreted protein, comprising transfecting a first population of human cells with a pharmaceutical nucleic acid made by the methods described herein, wherein the secreted protein is active upon a second population of human cells.
  • substituents of compounds of the present disclosure are disclosed in groups or in ranges. It is specifically intended that the present disclosure include each and every individual subcombination of the members of such groups and ranges.
  • the term "Ci_6 alkyl” is specifically intended to individually disclose methyl, ethyl, C 3 alkyl, C 4 alkyl, C5 alkyl, and C 6 alkyl.
  • stable refers to a compound that is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture, and preferably capable of formulation into an efficacious therapeutic agent.
  • alkyl is meant to refer to a saturated hydrocarbon group which is straight-chained or branched.
  • Example alkyl groups include methyl (Me), ethyl (Et), propyl (e.g., n-propyl and isopropyl), butyl (e.g., n-butyl, isobutyl, t-butyl), pentyl (e.g., n-pentyl, isopentyl, neopentyl), and the like.
  • An alkyl group can contain from 1 to about 20, from 2 to about 20, from 1 to about 12, from 1 to about 8, from 1 to about 6, from 1 to about 4, or from 1 to about 3 carbon atoms.
  • alkenyl refers to an alkyl group having one or more double carbon- carbon bonds.
  • Example alkenyl groups include ethenyl, propenyl, and the like.
  • alkoxy refers to an -O-alkyl group.
  • Example alkoxy groups include methoxy, ethoxy, propoxy (e.g., n-propoxy and isopropoxy), t-butoxy, and the like.
  • alkynyl refers to an alkyl group having one or more triple carbon- carbon bonds.
  • Example alkynyl groups include ethynyl, propynyl, and the like.
  • aryl refers to monocyclic or polycyclic (e.g., having 2, 3 or 4 fused rings) aromatic hydrocarbons such as, for example, phenyl, naphthyl, anthracenyl,
  • aryl groups have from 6 to about 20 carbon atoms.
  • halo or halogen includes fluoro, chloro, bromo, and iodo.
  • therapeutic agent refers to any agent that, when administered to a subject, has a therapeutic, diagnostic, and/or prophylactic effect and/or elicits a desired biological and/or pharmacological effect.
  • animal refers to any member of the animal kingdom. In some embodiments, “animal” refers to humans at any stage of development. In some embodiments, “animal” refers to non-human animals at any stage of development. In certain embodiments, the non-human animal is a mammal (e.g. , a rodent, a mouse, a rat, a rabbit, a monkey, a dog, a cat, a sheep, cattle, a primate, or a pig). In some embodiments, animals include, but are not limited to, mammals, birds, reptiles, amphibians, fish, and worms. In some embodiments, the animal is a transgenic animal, genetically-engineered animal, or a clone.
  • the term “approximately” or “about” refers to a range of values that fall within 25%, 20%, 19%, 18%, 17%, 16%, 15%, 14%, 13%, 12%, 11%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, or less in either direction (greater than or less than) of the stated reference value unless otherwise stated or otherwise evident from the context (except where such number would exceed 100% of a possible value).
  • association with means that the moieties are physically associated or connected with one another, either directly or via one or more additional moieties that serves as a linking agent, to form a structure that is sufficiently stable so that the moieties remain physically associated under the conditions in which the structure is used, e.g., physiological conditions.
  • biologically active refers to a characteristic of any substance that has activity in a biological system and/or organism. For instance, a substance that, when
  • nucleic acid administered to an organism, has a biological effect on that organism, is considered to be biologically active.
  • a portion of that nucleic acid that shares at least one biological activity of the whole nucleic acid is typically referred to as a "biologically active" portion.
  • Nucleotides or amino acids that are relatively conserved are those that are conserved amongst more related sequences than nucleotides or amino acids appearing elsewhere in the sequences.
  • two or more sequences are said to be "completely conserved” if they are 100% identical to one another.
  • two or more sequences are said to be "highly conserved” if they are at least 70% identical, at least 80% identical, at least 90% identical, or at least 95% identical to one another.
  • two or more sequences are said to be "highly conserved” if they are about 70% identical, about 80%> identical, about 90% identical, about 95%, about 98%, or about 99% identical to one another. In some embodiments, two or more sequences are said to be "conserved” if they are at least 30% identical, at least 40% identical, at least 50% identical, at least 60% identical, at least 70% identical, at least 80% identical, at least 90% identical, or at least 95% identical to one another.
  • two or more sequences are said to be "conserved” if they are about 30% identical, about 40% identical, about 50% identical, about 60%> identical, about 70% identical, about 80%> identical, about 90%> identical, about 95% identical, about 98% identical, or about 99% identical to one another.
  • expression of a nucleic acid sequence refers to one or more of the following events: (1) production of an RNA template from a DNA sequence (e.g., by transcription); (2) processing of an RNA transcript (e.g., by splicing, editing, 5' cap formation, and/or 3' end processing); (3) translation of an RNA into a polypeptide or protein; and (4) post- translational modification of a polypeptide or protein.
  • a "functional" biological molecule is a biological molecule in a form in which it exhibits a property and/or activity by which it is characterized.
  • in vitro refers to events that occur in an artificial environment, e.g., in a test tube or reaction vessel, in cell culture, in a Petri dish, etc., rather than within an organism (e.g., animal, plant, or microbe).
  • an artificial environment e.g., in a test tube or reaction vessel, in cell culture, in a Petri dish, etc., rather than within an organism (e.g., animal, plant, or microbe).
  • in vivo refers to events that occur within an organism (e.g., animal, plant, or microbe).
  • isolated refers to a substance or entity that has been (1) separated from at least some of the components with which it was associated when initially produced (whether in nature or in an experimental setting), and/or (2) produced, prepared, and/or manufactured by the hand of man. Isolated substances and/or entities may be separated from at least about 10%, about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, or more of the other components with which they were initially associated. In some
  • isolated agents are more than about 80%>, about 85%, about 90%, about 91%, about 92%, about 93%, about 94%, about 95%, about 96%, about 97%, about 98%, about 99%, or more than about 99% pure.
  • a substance is "pure” if it is substantially free of other components.
  • subject or patient refers to any organism to which a composition in accordance with the present disclosure may be administered, e.g. , for experimental, diagnostic, prophylactic, and/or therapeutic purposes. Typical subjects include animals (e.g., mammals such as mice, rats, rabbits, non-human primates, and humans) and/or plants.
  • substantially refers to the qualitative condition of exhibiting total or near-total extent or degree of a characteristic or property of interest.
  • biological and chemical phenomena rarely, if ever, go to completion and/or proceed to completeness or achieve or avoid an absolute result.
  • the term “substantially” is therefore used herein to capture the potential lack of completeness inherent in many biological and chemical phenomena.
  • An individual who is "suffering from” a disease, disorder, and/or condition has been diagnosed with or displays one or more symptoms of a disease, disorder, and/or condition.
  • an individual who is "susceptible to" a disease, disorder, and/or condition has not been diagnosed with and/or may not exhibit symptoms of the disease, disorder, and/or condition.
  • an individual who is susceptible to a disease, disorder, and/or condition may be characterized by one or more of the following: (1) a genetic mutation associated with development of the disease, disorder, and/or condition; (2) a genetic variant associated with development of the disease, disorder, and/or condition; (2) a genetic
  • polymorphism associated with development of the disease, disorder, and/or condition (3) increased and/or decreased expression and/or activity of a protein and/or nucleic acid associated with the disease, disorder, and/or condition; (4) habits and/or lifestyles associated with development of the disease, disorder, and/or condition; (5) a family history of the disease, disorder, and/or condition; and (6) exposure to and/or infection with a microbe associated with development of the disease, disorder, and/or condition.
  • an individual who is susceptible to a disease, disorder, and/or condition will develop the disease, disorder, and/or condition.
  • an individual who is susceptible to a disease, disorder, and/or condition will not develop the disease, disorder, and/or condition.
  • terapéuticaally effective amount means an amount of an agent to be delivered (e.g., nucleic acid, drug, therapeutic agent, diagnostic agent, prophylactic agent, etc.) that is sufficient, when administered to a subject suffering from or susceptible to a disease, disorder, and/or condition, to treat, improve symptoms of, diagnose, prevent, and/or delay the onset of the disease, disorder, and/or condition.
  • transcription factor refers to a DNA-binding protein that regulates transcription of DNA into RNA, for example, by activation or repression of transcription. Some transcription factors effect regulation of transcription alone, while others act in concert with other proteins. Some transcription factor can both activate and repress transcription under certain conditions. In general, transcription factors bind a specific target sequence or sequences highly similar to a specific consensus sequence in a regulatory region of a target gene. Transcription factors may regulate transcription of a target gene alone or in a complex with other molecules.
  • treating refers to partially or completely alleviating, ameliorating, improving, relieving, delaying onset of, inhibiting progression of, reducing severity of, and/or reducing incidence of one or more symptoms or features of a particular disease, disorder, and/or condition.
  • treating cancer may refer to inhibiting survival, growth, and/or spread of a tumor.
  • Treatment may be administered to a subject who does not exhibit signs of a disease, disorder, and/or condition and/or to a subject who exhibits only early signs of a disease, disorder, and/or condition for the purpose of decreasing the risk of developing pathology associated with the disease, disorder, and/or condition.
  • treatment comprises delivery of a protein associated with a therapeutically active nucleic acid to a subject in need thereof.
  • unmodified refers to a nucleic acid prior to being modified, e.g.
  • the compounds described herein can be asymmetric (e.g., having one or more stereocenters). All stereoisomers, such as enantiomers and diastereomers, are intended unless otherwise indicated.
  • Compounds of the present disclosure that contain asymmetrically substituted carbon atoms can be isolated in optically active or racemic forms. Methods on how to prepare optically active forms from optically active starting materials are known in the art, such as by resolution of racemic mixtures or by stereoselective synthesis.
  • Tautomeric forms result from the swapping of a single bond with an adjacent double bond together with the concomitant migration of a proton.
  • Tautomeric forms include prototropic tautomers which are isomeric protonation states having the same empirical formula and total charge.
  • Example prototropic tautomers include ketone - enol pairs, amide - imidic acid pairs, lactam - lactim pairs, amide - imidic acid pairs, enamine - imine pairs, and annular forms where a proton can occupy two or more positions of a heterocyclic system, for example, IH- and 3H-imidazole, 1H-, 2H- and 4H- 1,2,4-triazole, IH- and 2H- isoindole, and IH- and 2H-pyrazole.
  • Tautomeric forms can be in equilibrium or sterically locked into one form by appropriate substitution.
  • Compounds of the present disclosure can also include all isotopes of atoms occurring in the intermediates or final compounds.
  • Isotopes include those atoms having the same atomic number but different mass numbers.
  • isotopes of hydrogen include tritium and deuterium.
  • compound as used herein, is meant to include all stereoisomers, geometric isomers, tautomers, and isotopes of the structures depicted.
  • the compounds of the present disclosure are substantially isolated.
  • substantially isolated is meant that the compound is at least partially or substantially separated from the environment in which it was formed or detected. Partial separation can include, for example, a composition enriched in the compound of the present disclosure.
  • Substantial separation can include compositions containing at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 95%, at least about 97%), or at least about 99% by weight of the compound of the present disclosure, or salt thereof. Methods for isolating compounds and their salts are routine in the art.
  • the compounds of the present disclosure, and salts thereof, can also be prepared in combination with solvent or water molecules to form solvates and hydrates by routine methods.
  • the present disclosure also includes pharmaceutically acceptable salts of the compounds described herein.
  • pharmaceutically acceptable salts refers to derivatives of the disclosed compounds wherein the parent compound is modified by converting an existing acid or base moiety to its salt form.
  • examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like.
  • the pharmaceutically acceptable salts of the present disclosure include the conventional non-toxic salts of the parent compound formed, for example, from non-toxic inorganic or organic acids.
  • the pharmaceutically acceptable salts of the present disclosure can be synthesized from the parent compound which contains a basic or acidic moiety by conventional chemical methods.
  • such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two; generally, nonaqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile are preferred. Lists of suitable salts are found in Remington 's Pharmaceutical Sciences, 17 th ed., Mack Publishing Company, Easton, Pa., 1985, p. 1418 and Journal of Pharmaceutical Science, 66, 2 (1977), each of which is incorporated herein by reference in its entirety.
  • phrases "pharmaceutically acceptable” is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • prodrugs refer to any carriers, typically covalently bonded, which release the active parent drug when administered to a mammalian subject.
  • Prodrugs can be prepared by modifying functional groups present in the compounds in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to the parent compounds.
  • Prodrugs include compounds wherein hydroxyl, amino, sulfhydryl, or carboxyl groups are bonded to any group that, when administered to a mammalian subject, cleaves to form a free hydroxyl, amino, sulfhydryl, or carboxyl group respectively.
  • prodrugs include, but are not limited to, acetate, formate and benzoate derivatives of alcohol and amine functional groups in the compounds of the present disclosure. Preparation and use of prodrugs is discussed in T. Higuchi and V. Stella, "Pro-drugs as Novel Delivery Systems," Vol. 14 of the A.C.S. Symposium Series, and in Bioreversible Carriers in Drug Design, ed. Edward B. Roche, American Pharmaceutical Association and Pergamon Press, 1987, both of which are hereby incorporated by reference in their entirety.
  • nucleoside is defined as a compound containing a five-carbon sugar molecule (a pentose or ribose) or derivative thereof, and an organic base, purine or pyrimidine, or a derivative thereof.
  • nucleotide is defined as a nucleoside consisting of a phosphate group.
  • the nucleosides and nucleotides described herein are generally chemically modified on the major groove face.
  • the major groove chemical modifications can include an amino group, a thiol group, an alkyl group, or a halo group.
  • Table 1 below identifies the chemical faces of each canonical nucleotide. Circles identify the atoms comprising the respective chemical regions.
  • modified nucleosides include pyridin-4-one ribonucleoside, 5-aza- uridine, 2-thio-5-aza-uridine, 2-thiouridine, 4-thio-pseudouridine, 2-thio-pseudouridine, 5- hydroxyuridine, 3-methyluridine, 5-carboxymethyl-uridine, 1-carboxymethyl-pseudouridine, 5- propynyl-uridine, 1-propynyl-pseudouridine, 5-taurinomethyluridine, 1-taurinomethyl- pseudouridine, 5-taurinomethyl-2-thio-uridine, 1 -taurinomethyl-4-thio-uridine, 5-methyl-uridine, 1-methyl-pseudouridine, 4-thio-l-methyl-pseudouridine, 2-thio-l-methyl-pseudouridine, 1- methyl- 1 -deaza-pseudouridine, 2-thio-l-
  • modified nucleosides include 5-aza-cytidine, pseudoisocytidme, 3-methyl-cytidine, N4-acetylcytidine, 5-formylcytidine, N4-methylcytidine, 5- hydroxymethylcytidine, 1-methyl-pseudoisocytidine, pyrrolo-cytidine, pyrrolo- pseudoisocytidine, 2-thio-cytidine, 2-thio-5-methyl-cytidine, 4-thio-pseudoisocytidine, 4-thio-l- methyl-pseudoisocytidine, 4-thio- 1 -methyl- 1 -deaza-pseudoisocytidine, 1 -methyl- 1 -deaza- pseudoisocytidine, zebularine, 5-aza-zebularine, 5-methyl-zebularine, 5-aza-2-thio-zebularine, 2-
  • modified nucleosides include 2-aminopurine, 2, 6-diaminopurine, 7-deaza-adenine, 7-deaza-8-aza-adenine, 7-deaza-2-aminopurine, 7-deaza-8-aza-2-aminopurine, 7-deaza-2, 6-diaminopurine, 7-deaza-8-aza-2, 6-diaminopurine, 1-methyladenosine, N6- methyladenosine, N6-isopentenyladenosine, N6-(cis-hydroxyisopentenyl)adenosine, 2- methylthio-N6-(cis-hydroxyisopentenyl) adenosine, N6-glycinylcarbamoyladenosine, N6- threonylcarbamoyladenosine, 2-methylthio-N6-threonyl carbamoyladenosine, N6,N6- dimethyl
  • modified nucleosides include inosine, 1-methyl-inosine, wyosine, wybutosine, 7-deaza-guanosine, 7-deaza-8-aza-guanosine, 6-thio-guanosine, 6-thio-7-deaza- guanosine, 6-thio-7-deaza-8-aza-guanosine, 7-methyl-guanosine, 6-thio-7-methyl-guanosine, 7- methylinosine, 6-methoxy-guanosine, 1-methylguanosine, N2-methylguanosine, N2,N2- dimethylguanosine, 8-oxo-guanosine, 7-methyl-8-oxo-guanosine, l-methyl-6-thio-guanosine, N2-methyl-6-thio-guanosine, and N2,N2-dimethyl-6-thio-guanosine.
  • the nucleotide can be modified on the major groove face and can include replacement of the hydrogen on C-5 of uracil with a methyl group or a halo group.
  • the nucleoside and nucleotide can be a compound of Formula I:
  • Z is O or S
  • each of Y 1 is independently selected from -OR al , -NR al R bl , and -SR al ;
  • each of Y 2 is independently selected from O, NR a , S or a linker comprising an atom selected from the group consisting of C, O, N, and S;
  • each of Y 3 is independently selected from O and S;
  • Y 4 is selected from H, -OR a , -SR a , and -NHR a ;
  • n 0, 1 , 2, or 3;
  • n 0, 1 , 2 or 3;
  • B is a nucleobase
  • R a is H, Ci-20 alkyl, C 2- 2o alkenyl, C 2-2 o alkynyl, or C 6-2 o aryl;
  • R al and R bl are each independently H or a counterion
  • -Y 3 -R cl is OH or SH at a pH of about 1 or -Y 3 -R cl is O " or S " at physiological pH; or -Y 3 -R cl is Ci-20 alkoxy, C 2-2 o -O-alkenyl, or Ci -2 o -O-alkynyl;
  • B is an unmodified nucleobase selected from cytosine, guanine, uracil and adenine, then at least one of Z, Y 1 or Y 2 is not O or OH.
  • B is a nucleobase of Formula Il-a, Il-b, or II-c:
  • X is O or S
  • U and W are each independently C or N;
  • V is O, S, C or N;
  • R 1 is H, C 1-6 alkyl, C 1-6 alkenyl, C 1-6 alkynyl, halo, or -OR c , wherein Ci-20 alkyl, C2-20 alkenyl, C2-20 alkynyl are each optionally substituted with -OH, -NR a R b , -SH, - C(0)R c , -C(0)OR c , -NHC(0)R c , or -NHC(0)OR c ;
  • R 2 is H, -OR c , -SR C , -NR a R b , or halo;
  • R 1 and R 2 together with the carbon atoms to which they are attached can form a 5- or 6-membered ring optionally substituted with 1-4 substituents selected from halo, - OH, -SH, -NR a R b , Ci-20 alkyl, C 2- 2o alkenyl, C 2- 2o alkynyl, C 1-20 alkoxy, or C 1-20 thioalkyl;
  • R 3 is H or Ci-20 alkyl
  • R 4 is H or C 1-20 alkyl; wherein when ⁇ denotes a double bond then R 4 is absent, or N- R 4 , taken together, forms a positively charged N substituted with C 1-20 alkyl;
  • R a and R b are each independently H, C 1-20 alkyl, C2-20 alkenyl, C2-20 alkynyl, or C6-20 aryl;
  • R c is H, Ci-20 alkyl, C 2- 2o alkenyl, phenyl, benzyl, a polyethylene glycol group, or an amino-polyethylene glycol group.
  • B is a nucleobase of Formula Il-al , II-a2, II-a3, II-a4, or II-a5:
  • B is a nucleobase selected from the group consisting of cytosine, guanine, adenine, and uracil.
  • B is a pyrimidine or derivative thereof.
  • the nucleotide is selected from the group consisting of:
  • the modified nucleotide can be:
  • the major groove chemical modification can include replacement of the C-H group at C-5 with an -NH- group or a -NH(CH 3 )- group.
  • the modified nucleotide can be:
  • the major groove chemical modification can include replacement of the hydrogen at C-5 of cytosine with a halo group or a methyl group.
  • the modified nucleotide can be:
  • the major groove chemical modification can include a fused ring that is formed by the NH 2 at the C-4 position and the carbon atom at the C-5 position.
  • the modified nucleotide can be:
  • a modified nucleotide is 5 '-0-(l -Thiophosphate)- Adenosine, 5'- 0-(l-Thiophosphate)-Cytidine, 5'-0-(l-Thiophosphate)-Guanosine, 5'-0-(l-Thiophosphate)- Uridine or 5'-0-(l-Thiophosphate)-Pseudouridine.
  • a-thio substituted phosphate moiety is provided to confer stability to RNA and DNA polymers through the unnatural phosphorothioate backbone linkages.
  • Phosphorothioate DNA and RNA have increased nuclease resistance and subsequently a longer half-life in a cellular environment.
  • Phosphorothioate linked nucleic acids are expected to also reduce the innate immune response through weaker binding/activation of cellular innate immune molecules.
  • modified nucleotides and modified nucleotide combinations are provided below in Table 2.
  • N6-methyl-adenosine 25% Nl-Methyl-pseudo-uridine/75%-pseudo-uridine a-thio-adenosine 5-methyl-uridine
  • modified nucleosides and nucleotides disclosed herein can be prepared from readily available starting materials using the following general methods and procedures. It is understood that where typical or preferred process conditions (i.e., reaction temperatures, times, mole ratios of reactants, solvents, pressures, etc.) are given; other process conditions can also be used unless otherwise stated. Optimum reaction conditions may vary with the particular reactants or solvent used, but such conditions can be determined by one skilled in the art by routine optimization procedures.
  • spectroscopic means such as nuclear magnetic resonance spectroscopy (e.g., 1H or 13 C) infrared spectroscopy, spectrophotometry (e.g., UV-visible), or mass spectrometry, or by chromatography such as high performance liquid chromatography (HPLC) or thin layer chromatography.
  • spectroscopic means such as nuclear magnetic resonance spectroscopy (e.g., 1H or 13 C) infrared spectroscopy, spectrophotometry (e.g., UV-visible), or mass spectrometry
  • HPLC high performance liquid chromatography
  • Preparation of modified nucleosides and nucleotides can involve the protection and deprotection of various chemical groups. The need for protection and deprotection, and the selection of appropriate protecting groups can be readily determined by one skilled in the art.
  • the chemistry of protecting groups can be found, for example, in Greene, et al., Protective Groups in Organic Synthesis, 2d. Ed., Wiley
  • Suitable solvents can be substantially nonreactive with the starting materials (reactants), the intermediates, or products at the temperatures at which the reactions are carried out, i.e., temperatures which can range from the solvent's freezing temperature to the solvent's boiling temperature.
  • a given reaction can be carried out in one solvent or a mixture of more than one solvent.
  • suitable solvents for a particular reaction step can be selected.
  • Resolution of racemic mixtures of modified nucleosides and nucleotides can be carried out by any of numerous methods known in the art.
  • An example method includes fractional
  • Suitable resolving agents for fractional recrystallization methods are, for example, optically active acids, such as the D and L forms of tartaric acid, diacetyltartaric acid, dibenzoyltartaric acid, mandelic acid, malic acid, lactic acid or the various optically active camphorsulfonic acids.
  • Resolution of racemic mixtures can also be carried out by elution on a column packed with an optically active resolving agent ⁇ e.g., dinitrobenzoylphenylglycine).
  • Suitable elution solvent composition can be determined by one skilled in the art.
  • Modified nucleosides and nucleotides can also be prepared according to the synthetic methods described in Ogata et al. Journal of Organic Chemistry 74:2585-2588, 2009; Purmal et al. Nucleic Acids Research 22(1): 72-78, 1994; Fukuhara et al. Biochemistry 1(4): 563-568, 1962; and Xu et al. Tetrahedron 48(9): 1729-1740, 1992, each of which are incorporated by reference in their entirety.
  • nucleic acids including RNAs such as mRNAs that contain one or more modified nucleosides (termed “modified nucleic acids”) or nucleotides as described herein, which have useful properties including the significant decreast or lack of a substantial induction of the innate immune response of a cell into which the mRNA is introduced, or the suppression thereof.
  • modified nucleic acids enhance the efficiency of protein production, intracellular retention of nucleic acids, and viability of contacted cells, as well as possess reduced immunogenicity, of these nucleic acids compared to unmodified nucleic acids, having these properties are termed “enhanced nucleic acids” herein.
  • nucleic acids which have decreased binding affinity to a major groove interacting, e.g. binding, partner.
  • the nucleic acids are comprised of at least one nucleotide that has been chemically modified on the major groove face as described herein.
  • nucleic acid in its broadest sense, includes any compound and/or substance that is or can be incorporated into an oligonucleotide chain.
  • exemplary nucleic acids for use in accordance with the present disclosure include, but are not limited to, one or more of DNA, RNA including messenger mRNA (mRNA), hybrids thereof, RNAi-inducing agents, RNAi agents, siRNAs, shRNAs, miRNAs, antisense RNAs, ribozymes, catalytic DNA, RNAs that induce triple helix formation, aptamers, vectors, etc., described in detail herein.
  • mRNA messenger mRNA
  • modified nucleic acids containing a translatable region and one, two, or more than two different nucleoside modifications.
  • the modified nucleic acid exhibits reduced degradation in a cell into which the nucleic acid is introduced, relative to a corresponding unmodified nucleic acid.
  • exemplary nucleic acids include ribonucleic acids (RNAs), deoxyribonucleic acids (DNAs), threose nucleic acids (TNAs), glycol nucleic acids (GNAs), locked nucleic acids (LNAs) or a hybrid thereof.
  • the modified nucleic acid includes messenger RNAs (mRNAs). As described herein, the nucleic acids of the present disclosure do not substantially induce an innate immune response of a cell into which the mRNA is introduced.
  • the present disclosure provides a modified nucleic acid containing a degradation domain, which is capable of being acted on in a directed manner within a cell.
  • nucleic acid is optional, and are beneficial in some embodiments.
  • a 5' untranslated region (UTR) and/or a 3'UTR are provided, wherein either or both may independently contain one or more different nucleoside modifications.
  • nucleoside modifications may also be present in the translatable region. Also provided are nucleic acids containing a Kozak sequence. Additionally, provided are nucleic acids containing one or more intronic nucleotide sequences capable of being excised from the nucleic acid.
  • nucleic acids containing an internal ribosome entry site may act as the sole ribosome binding site, or may serve as one of multiple ribosome binding sites of an mRNA.
  • An mRNA containing more than one functional ribosome binding site may encode several peptides or polypeptides that are translated independently by the ribosomes ("multicistronic mRNA").
  • multicistronic mRNA When nucleic acids are provided with an IRES, further optionally provided is a second translatable region. Examples of IRES sequences that can be used according to the present disclosure include without limitation, those from picomaviruses (e.g.
  • FMDV pest viruses
  • CFFV pest viruses
  • PV polio viruses
  • ECMV encephalomyocarditis viruses
  • FMDV foot-and-mouth disease viruses
  • HCV hepatitis C viruses
  • CSFV classical swine fever viruses
  • MLV murine leukemia virus
  • SIV simian immune deficiency viruses
  • CrPV cricket paralysis viruses
  • the nucleic acid sequences comprise a compound of Formula I-d:
  • Z is O or S
  • each of Y 1 is independently selected from -OR al , -NR al R bl , and -SR al ;
  • each of Y 2 is independently selected from O, NR a , S or a linker comprising
  • each of Y 3 is independently selected from O and S;
  • Y 4 is selected from H, -OR a , -SR a , and -NHR a ;
  • B is a nucleobase
  • R a is H, C 1-20 alkyl, C 2-2 o alkenyl, C 2-20 alkynyl, or C 6-20 aryl;
  • R al and R bl are each independently H or a counterion;
  • -Y 3 -R cl is OH or SH at a pH of about 1 or -Y 3 -R cl is O " or S " at physiological pH;
  • B is an unmodified nucleobase selected from cytosine, guanine, thymidine, uracil and adenine, then at least one of Z, Y 1 or Y 2 is not O or OH.
  • B is a nucleobase of Formula Il-a, Il-b, or II-c:
  • denotes a single or double bond
  • X is O or S
  • U and W are each independently C or N;
  • V is O, S, C or N;
  • R 1 is H, Ci -6 alkyl, Ci -6 alkenyl, Ci -6 alkynyl, halo, or -OR c , wherein Ci-20 alkyl, C 2-2 o alkenyl, C 2-2 o alkynyl are each optionally substituted with -OH, -NR a R b , -SH, - C(0)R c , -C(0)OR c , -NHC(0)R c , or -NHC(0)OR c ;
  • R 2 is H, -OR c , -SR C , -NR a R b , or halo;
  • R 1 and R 2 together with the carbon atoms to which they are attached can form a 5- or 6-membered ring optionally substituted with 1-4 substituents selected from halo, - OH, -SH, -NR a R b , Ci-20 alkyl, C 2-2 o alkenyl, C 2-2 o alkynyl, Ci -2 o alkoxy, or Ci -2 o thioalkyl;
  • R 3 is H or Ci-20 alkyl
  • R 4 is H or C 1-20 alkyl; wherein when ⁇ denotes a double bond then R 4 is absent, or N- R 4 , taken together, forms a positively charged N substituted with Ci -2 o alkyl;
  • R a and R b are each independently H, Ci -2 o alkyl, C 2-2 o alkenyl, C 2-2 o alkynyl, or C6 -2 o aryl;
  • R c is H, Ci-20 alkyl, C 2-2 o alkenyl, phenyl, benzyl, a polyethylene glycol group, or an amino-polyethylene glycol group.
  • B is a nucleobase of Formula Il-al, II-a2, II-a3, II-a4, or II-a5:
  • At least 25% of the cytosines are replaced by a compound of Formula I-a (e.g., at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, or about 100%).
  • a compound of Formula I-a e.g., at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, or about 100%).
  • At least 25% of the uracils are replaced by a compound of Formula I-a (e.g., at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, or about 100%).
  • a compound of Formula I-a e.g., at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, or about 100%).
  • At least 25% of the cytosines and 25% of the uracils are replaced by a compound of Formula I-a (e.g., at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, or about 100%).
  • a compound of Formula I-a e.g., at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, or about 100%).
  • the nucleic acid is translatable.
  • RNA recognition receptors that detect and respond to RNA ligands through interactions, e.g. binding, with the major groove face of a nucleotide or nucleic acid.
  • RNA ligands comprising modified nucleotides or nucleic acids as described herein decrease interactions with major groove binding partners, and therefore decrease an innate immune response, or expression and secretion of pro-inflammatory cytokines, or both.
  • Example major groove interacting, e.g. binding, partners include, but are not limited to the following nucleases and helicases. Within membranes, TLRs (Toll-like Receptors) 3, 7, and 8 can respond to single- and double-stranded RNAs.
  • helicases within the cytoplasm, members of the superfamily 2 class of DE (D/H) helicases and ATPases can sense RNAs to initiate antiviral responses.
  • D/H superfamily 2 class of DE
  • helicases include the RIG-I (retinoic acid-inducible gene I) and MDA5 (melanoma differentiation-associated gene 5).
  • RIG-I retinoic acid-inducible gene I
  • MDA5 melanoma differentiation-associated gene 5
  • Other examples include laboratory of genetics and physiology 2 (LGP2), ⁇ -200 domain containing proteins, or Helicase-domain containing proteins.
  • innate immune response includes a cellular response to exogenous nucleic acids, including single stranded nucleic acids, generally of viral or bacterial origin, which involves the induction of cytokine expression and release, particularly the interferons, and cell death. Protein synthesis is also reduced during the innate cellular immune response. While it is advantageous to eliminate the innate immune response in a cell, the present disclosure provides modified mRNAs that substantially reduce the immune response, including interferon signaling, without entirely eliminating such a response.
  • the immune response is reduced by 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99%, 99.9%, or greater than 99.9% as compared to the immune response induced by a corresponding unmodified nucleic acid.
  • a reduction can be measured by expression or activity level of Type 1 interferons or the expression of interferon-regulated genes such as the toll-like receptors (e.g., TLR7 and TLR8).
  • Reduction of innate immune response can also be measured by decreased cell death following one or more administrations of modified RNAs to a cell population; e.g., cell death is 10%, 25%, 50%, 75%, 85%, 90%, 95%, or over 95% less than the cell death frequency observed with a corresponding unmodified nucleic acid.
  • cell death may affect fewer than 50%, 40%, 30%, 20%, 10%, 5%, 1%, 0.1%, 0.01% or fewer than 0.01% of cells contacted with the modified nucleic acids.
  • the present disclosure provides for the repeated introduction (e.g., transfection) of modified nucleic acids into a target cell population, e.g., in vitro, ex vivo, or in vivo.
  • the step of contacting the cell population may be repeated one or more times (such as two, three, four, five or more than five times).
  • the step of contacting the cell population with the modified nucleic acids is repeated a number of times sufficient such that a predetermined efficiency of protein translation in the cell population is achieved. Given the reduced cytotoxicity of the target cell population provided by the nucleic acid modifications, such repeated transfections are achievable in a diverse array of cell types.
  • nucleic acids that encode variant polypeptides, which have a certain identity with a reference polypeptide sequence.
  • identity refers to a relationship between the sequences of two or more peptides, as determined by comparing the sequences. In the art, “identity” also means the degree of sequence relatedness between peptides, as determined by the number of matches between strings of two or more amino acid residues. “Identity” measures the percent of identical matches between the smaller of two or more sequences with gap alignments (if any) addressed by a particular mathematical model or computer program (i.e., "algorithms"). Identity of related peptides can be readily calculated by known methods.
  • Such methods include, but are not limited to, those described in Computational Molecular Biology, Lesk, A. M., ed., Oxford University Press, New York, 1988; Biocomputing: Informatics and Genome Projects, Smith, D. W., ed., Academic Press, New York, 1993;
  • the polypeptide variant has the same or a similar activity as the reference polypeptide.
  • the variant has an altered activity (e.g., increased or decreased) relative to a reference polypeptide.
  • variants of a particular polynucleotide or polypeptide of the present disclosure will have at least about 40%, 45%, 50%>, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity to that particular reference polynucleotide or polypeptide as determined by sequence alignment programs and parameters described herein and known to those skilled in the art.
  • protein fragments, functional protein domains, and homologous proteins are also considered to be within the scope of this present disclosure.
  • a reference protein meaning a polypeptide sequence at least one amino acid residue shorter than a reference polypeptide sequence but otherwise identical
  • any protein that includes a stretch of about 20, about 30, about 40, about 50, or about 100 amino acids which are about 40%, about 50%, about 60%>, about 70%), about 80%>, about 90%>, about 95%, or about 100%.
  • identical to any of the sequences described herein can be utilized in accordance with the present disclosure.
  • a protein sequence to be utilized in accordance with the present disclosure includes 2, 3, 4, 5, 6, 7, 8, 9, 10, or more mutations as shown in any of the sequences provided or referenced herein.
  • polynucleotide libraries containing nucleoside modifications wherein the polynucleotides individually contain a first nucleic acid sequence encoding a polypeptide, such as an antibody, protein binding partner, scaffold protein, and other polypeptides known in the art.
  • a polypeptide such as an antibody, protein binding partner, scaffold protein, and other polypeptides known in the art.
  • the polynucleotides are mRNA in a form suitable for direct introduction into a target cell host, which in turn synthesizes the encoded polypeptide.
  • multiple variants of a protein are produced and tested to determine the best variant in terms of
  • Such a library may contain 10, 10 , 10 , 10 , 10 , 10 , 10 , 10 , or over 10 9 possible variants (including substitutions, deletions of one or more residues, and insertion of one or more residues).
  • Proper protein translation involves the physical aggregation of a number of polypeptides and nucleic acids associated with the mRNA.
  • Provided by the present disclosure are protein- nucleic acid complexes, containing a translatable mRNA having one or more nucleoside modifications (e.g., at least two different nucleoside modifications) and one or more polypeptides bound to the mRNA.
  • the proteins are provided in an amount effective to prevent or reduce an innate immune response of a cell into which the complex is introduced.
  • mRNAs having sequences that are substantially not translatable. Such mRNA is effective as a vaccine when administered to a mammalian subject.
  • modified nucleic acids that contain one or more noncoding regions. Such modified nucleic acids are generally not translated, but are capable of binding to and sequestering one or more translational machinery component such as a ribosomal protein or a transfer RNA (tRNA), thereby effectively reducing protein expression in the cell.
  • the modified nucleic acid may contain a small nucleolar RNA (sno-RNA), micro RNA (miRNA), small interfering RNA (siRNA) or Piwi-interacting RNA (piRNA).
  • Nucleic acids for use in accordance with the present disclosure may be prepared according to any available technique including, but not limited to chemical synthesis, enzymatic synthesis, which is generally termed in vitro transcription, enzymatic or chemical cleavage of a longer precursor, etc.
  • Methods of synthesizing RNAs are known in the art (see, e.g., Gait, M.J. (ed.) Oligonucleotide synthesis: a practical approach, Oxford [Oxfordshire], Washington, DC: IRL Press, 1984; and Herdewijn, P. (ed.) Oligonucleotide synthesis: methods and applications, Methods in Molecular Biology, v. 288 (Clifton, N.J.) Totowa, N.J.: Humana Press, 2005; both of which are incorporated herein by reference).
  • modified nucleosides and nucleotides disclosed herein can be prepared from readily available starting materials using the following general methods and procedures. It is understood that where typical or preferred process conditions ⁇ i.e., reaction temperatures, times, mole ratios of reactants, solvents, pressures, etc.) are given; other process conditions can also be used unless otherwise stated. Optimum reaction conditions may vary with the particular reactants or solvent used, but such conditions can be determined by one skilled in the art by routine optimization procedures.
  • product formation can be monitored by spectroscopic means, such as nuclear magnetic resonance spectroscopy (e.g., 1H or 13 C) infrared spectroscopy, spectrophotometry (e.g., UV-visible), or mass spectrometry, or by chromatography such as high performance liquid chromatography (HPLC) or thin layer chromatography.
  • spectroscopic means such as nuclear magnetic resonance spectroscopy (e.g., 1H or 13 C) infrared spectroscopy, spectrophotometry (e.g., UV-visible), or mass spectrometry
  • chromatography such as high performance liquid chromatography (HPLC) or thin layer chromatography.
  • Preparation of modified nucleosides and nucleotides can involve the protection and deprotection of various chemical groups.
  • the need for protection and deprotection, and the selection of appropriate protecting groups can be readily determined by one skilled in the art.
  • the chemistry of protecting groups can be found, for example, in Greene, et al., Protective Groups in Organic Synthesis, 2d. Ed., Wiley & Sons, 1991, which is incorporated herein by reference in its entirety.
  • Suitable solvents can be substantially nonreactive with the starting materials (reactants), the intermediates, or products at the temperatures at which the reactions are carried out, i.e., temperatures which can range from the solvent's freezing temperature to the solvent's boiling temperature.
  • a given reaction can be carried out in one solvent or a mixture of more than one solvent.
  • suitable solvents for a particular reaction step can be selected.
  • An example method includes fractional recrystallization using a "chiral resolving acid" which is an optically active, salt-forming organic acid.
  • Suitable resolving agents for fractional recrystallization methods are, for example, optically active acids, such as the D and L forms of tartaric acid, diacetyltartaric acid, dibenzoyltartaric acid, mandelic acid, malic acid, lactic acid or the various optically active camphorsulfonic acids.
  • Resolution of racemic mixtures can also be carried out by elution on a column packed with an optically active resolving agent (e.g., dinitrobenzoylphenylglycine).
  • Suitable elution solvent composition can be determined by one skilled in the art.
  • Modified nucleic acids need not be uniformly modified along the entire length of the molecule. Different nucleotide modifications and/or backbone structures may exist at various positions in the nucleic acid.
  • nucleotide analogs or other modification(s) may be located at any position(s) of a nucleic acid such that the function of the nucleic acid is not substantially decreased.
  • a modification may also be a 5 Or 3' terminal modification.
  • the nucleic acids may contain at a minimum one and at maximum 100% modified nucleotides, or any intervening percentage, such as at least 5% modified nucleotides, at least 10% modified nucleotides, at least 25% modified nucleotides, at least 50%> modified nucleotides, at least 80%> modified nucleotides, or at least 90% modified nucleotides.
  • the nucleic acids may contain a modified pyrimidine such as uracil or cytosine.
  • at least 5%, at least 10%, at least 25%, at least 50%, at least 80%, at least 90% or 100% of the uracil in the nucleic acid is replaced with a modified uracil.
  • the modified uracil can be replaced by a compound having a single unique structure, or can be replaced by a plurality of compounds having different structures (e.g., 2, 3, 4 or more unique structures). In some embodiments, at least 5%, at least 10%, at least 25%, at least 50%, at least 80%, at least 90% or 100% of the cytosine in the nucleic acid is replaced with a modified cytosine.
  • the modified cytosine can be replaced by a compound having a single unique structure, or can be replaced by a plurality of compounds having different structures (e.g., 2, 3, 4 or more unique structures).
  • the shortest length of a modified mRNA of the present disclosure can be the length of an mRNA sequence that is sufficient to encode for a dipeptide.
  • the length of the mRNA sequence is sufficient to encode for a tripeptide. In another embodiment, the length of an mRNA sequence is sufficient to encode for a tetrapeptide. In another embodiment, the length of an mRNA sequence is sufficient to encode for a pentapeptide. In another embodiment, the length of an mRNA sequence is sufficient to encode for a hexapeptide. In another embodiment, the length of an mRNA sequence is sufficient to encode for a heptapeptide. In another embodiment, the length of an mRNA sequence is sufficient to encode for an octapeptide. In another embodiment, the length of an mRNA sequence is sufficient to encode for a nonapeptide. In another embodiment, the length of an mRNA sequence is sufficient to encode for a decapeptide.
  • dipeptides that the modified nucleic acid sequences can encode for include, but are not limited to, carnosine and anserine.
  • the mRNA is greater than 30 nucleotides in length. In another embodiment, the RNA molecule is greater than 35 nucleotides in length. In another embodiment, the length is at least 40 nucleotides. In another embodiment, the length is at least 45 nucleotides. In another embodiment, the length is at least 55 nucleotides. In another embodiment, the length is at least 60 nucleotides. In another embodiment, the length is at least 60 nucleotides. In another embodiment, the length is at least 80 nucleotides. In another embodiment, the length is at least 90 nucleotides. In another embodiment, the length is at least 100 nucleotides. In another embodiment, the length is at least 120 nucleotides.
  • the length is at least 140 nucleotides. In another embodiment, the length is at least 160 nucleotides. In another embodiment, the length is at least 180 nucleotides. In another embodiment, the length is at least 200 nucleotides. In another embodiment, the length is at least 250 nucleotides. In another embodiment, the length is at least 300 nucleotides. In another embodiment, the length is at least 350 nucleotides. In another embodiment, the length is at least 400 nucleotides. In another embodiment, the length is at least 450 nucleotides. In another embodiment, the length is at least 500 nucleotides. In another embodiment, the length is at least 600 nucleotides.
  • the length is at least 700 nucleotides. In another embodiment, the length is at least 800 nucleotides. In another embodiment, the length is at least 900 nucleotides. In another embodiment, the length is at least 1000 nucleotides. In another embodiment, the length is at least 1100 nucleotides. In another embodiment, the length is at least 1200 nucleotides. In another embodiment, the length is at least 1300 nucleotides. In another embodiment, the length is at least 1400 nucleotides. In another embodiment, the length is at least 1500 nucleotides. In another embodiment, the length is at least 1600 nucleotides. In another embodiment, the length is at least 1800 nucleotides.
  • the length is at least 2000 nucleotides. In another embodiment, the length is at least 2500 nucleotides. In another embodiment, the length is at least 3000 nucleotides. In another embodiment, the length is at least 4000 nucleotides. In another embodiment, the length is at least 5000 nucleotides, or greater than 5000 nucleotides.
  • the present disclosure provides methods of preparing a nucleic acid sequence comprising a nucleotide that disrupts binding of a major groove interacting partner with the nucleic acid sequence, wherein the nucleic acid sequence comprises a compound of Formula I-d:
  • Z is O or S
  • each of Y 1 is independently selected from -OR al , -NR al R bl , and -SR al ;
  • each of Y 2 is independently selected from O, NR a , S or a linker comprising an atom selected from the group consisting of C, O, N, and S;
  • each of Y 3 is independently selected from O and S;
  • Y 4 is selected from H, -OR a , -SR a , and -NHR a ;
  • B is a nucleobase
  • R a is H, Ci-20 alkyl, C 2- 2o alkenyl, C 2-2 o alkynyl, or C 6-2 o aryl;
  • R al and R bl are each independently H or a counterion
  • -Y 3 -R cl is OH or SH at a pH of about 1 or -Y 3 -R cl is O " or S " at physiological pH; or -Y 3 -R cl is Ci-20 alkoxy, C 2-2 o -O-alkenyl, or Ci -2 o -O-alkynyl;
  • B is an unmodified nucleobase selected from cytosine, guanine, uracil and adenine, then at least one of Z, Y 1 or Y 2 is not O or OH;
  • the reaction is repeated from 1 to about 7,000 times.
  • B is a nucleobase of Formula Il-a, Il-b, or II-c:
  • X is O or S
  • U and W are each independently C or N;
  • V is O, S, C or N;
  • R 1 is H, C 1-6 alkyl, C 1-6 alkenyl, C 1-6 alkynyl, halo, or -OR c , wherein Ci-20 alkyl, C 2-2 o alkenyl, C 2-2 o alkynyl are each optionally substituted with -OH, -NR a R b , -SH, - C(0)R c , -C(0)OR c , -NHC(0)R c , or -NHC(0)OR c ;
  • R 2 is H, -OR c , -SR C , -NR a R b , or halo;
  • R 1 and R 2 together with the carbon atoms to which they are attached can form a 5- or 6-membered ring optionally substituted with 1-4 substituents selected from halo, - OH, -SH, -NR a R b , Ci-20 alkyl, C 2-2 o alkenyl, C 2-2 o alkynyl, C 1-20 alkoxy, or C 1-20 thioalkyl;
  • R 3 is H or Ci-20 alkyl
  • R 4 is H or C 1-20 alkyl; wherein when ⁇ denotes a double bond then R 4 is absent, or N- R 4 , taken together, forms a positively charged N substituted with C 1-20 alkyl;
  • R a and R b are each independently H, C 1-20 alkyl, C 2-2 o alkenyl, C 2-2 o alkynyl, or C6 -2 o aryl;
  • R c is H, Ci-20 alkyl, C 2-2 o alkenyl, phenyl, benzyl, a polyethylene glycol group, or an amino-polyethylene glycol group.
  • B is a nucleobase of Formula Il-al , II-a2, II-a3, II-a4, or II-a5:
  • the methods further comprise a nucleotide selected from the group consisting of adenosine, cytosine, guanosine, and uracil.
  • the nucleobase is a pyrimidine or derivative thereof.
  • the present disclosure provides methods of amplifying a nucleic acid sequence comprising a nucleotide that disrupts binding of a major groove binding partner with the nucleic acid sequence, the method comprising:
  • Z is O or S
  • each of Y 1 is independently selected from -OR al , -NR al R bl , and -SR al ;
  • each of Y 2 is independently selected from O, NR a , S or a linker comprising an atom selected from the group consisting of C, O, N, and S;
  • each of Y 3 is independently selected from O and S;
  • Y 4 is selected from H, -OR a , -SR a , and -NHR a ;
  • B is a nucleobase
  • R a is H, Ci-20 alkyl, C 2- 2o alkenyl, C 2-2 o alkynyl, or C 6-2 o aryl;
  • R al and R bl are each independently H or a counterion
  • -Y 3 -R cl is OH or SH at a pH of about 1 or -Y 3 -R cl is O " or S " at physiological pH; or -Y 3 -R cl is Ci-20 alkoxy, C 2-2 o -O-alkenyl, or Ci -2 o -O-alkynyl;
  • B is an unmodified nucleobase selected from cytosine, guanine, uracil and adenine, then at least one of Z, Y 1 or Y 2 is not O or OH;
  • RNA polymerase a primer, a cDNA template, and an RNA polymerase.
  • B is a nucleobase of Formula Il-a, Il-b, or II-c:
  • denotes a single or double bond
  • X is O or S
  • U and W are each independently C or N;
  • V is O, S, C or N; wherein when V is C then R 1 is H, C 1-6 alkyl, C 1-6 alkenyl, C 1-6 alkynyl, halo, or -OR c , wherein Ci-20 alkyl, C 2- 2o alkenyl, C 2-2 o alkynyl are each optionally substituted with -OH, -NR a R b , -SH, - C(0)R c , -C(0)OR c , -NHC(0)R c , or -NHC(0)OR c ;
  • R 2 is H, -OR c , -SR C , -NR a R b , or halo;
  • R 1 and R 2 together with the carbon atoms to which they are attached can form a 5- or 6-membered ring optionally substituted with 1-4 substituents selected from halo, - OH, -SH, -NR a R b , Ci-20 alkyl, C 2-2 o alkenyl, C 2-2 o alkynyl, C 1-20 alkoxy, or C 1-20 thioalkyl;
  • R 3 is H or Ci-20 alkyl
  • R 4 is H or C 1-20 alkyl; wherein when ⁇ denotes a double bond then R 4 is absent, or N- R 4 , taken together, forms a positively charged N substituted with C 1-20 alkyl;
  • R a and R b are each independently H, C 1-20 alkyl, C 2-2 o alkenyl, C 2-2 o alkynyl, or C6 -2 o aryl;
  • R c is H, Ci-20 alkyl, C 2-2 o alkenyl, phenyl, benzyl, a polyethylene glycol group, or an amino-polyethylene glycol group.
  • B is a nucleobase of Formula Il-al, II-a2, II-a3, II-a4, or II-a5:
  • the methods further comprise a nucleotide selected from the group consisting of adenosine, cytosine, guanosine, and uracil.
  • the nucleobase is a pyrimidine or derivative thereof.
  • modified nucleic acids and the proteins translated from the modified nucleic acids described herein can be used as therapeutic agents.
  • a modified nucleic acid described herein can be administered to a subject, wherein the modified nucleic acid is translated in vivo to produce a therapeutic peptide in the subject.
  • compositions, methods, kits, and reagents for treatment or prevention of disease or conditions in humans and other mammals include modified nucleic acids, cells containing modified nucleic acids or polypeptides translated from the modified nucleic acids, polypeptides translated from modified nucleic acids, and cells contacted with cells containing modified nucleic acids or polypeptides translated from the modified nucleic acids.
  • combination therapeutics containing one or more modified nucleic acids containing translatable regions that encode for a protein or proteins that boost a mammalian subject's immunity along with a protein that induces antibody-dependent cellular toxitity.
  • G-CSF granulocyte-colony stimulating factor
  • such combination therapeutics are useful in Her2+ breast cancer patients who develop induced resistance to trastuzumab. (See, e.g., Albrecht, Immunotherapy. 2(6):795-8 (2010)).
  • Such translation can be in vivo, ex vivo, in culture, or in vitro.
  • the cell population is contacted with an effective amount of a composition containing a nucleic acid that has at least one nucleoside modification, and a translatable region encoding the recombinant polypeptide.
  • the population is contacted under conditions such that the nucleic acid is localized into one or more cells of the cell population and the recombinant polypeptide is translated in the cell from the nucleic acid.
  • an effective amount of the composition is provided based, at least in part, on the target tissue, target cell type, means of administration, physical characteristics of the nucleic acid ⁇ e.g., size, and extent of modified nucleosides), and other determinants.
  • an effective amount of the composition provides efficient protein production in the cell, preferably more efficient than a composition containing a corresponding unmodified nucleic acid. Increased efficiency may be demonstrated by increased cell transfection (i.e., the percentage of cells transfected with the nucleic acid), increased protein translation from the nucleic acid, decreased nucleic acid degradation (as demonstrated, e.g. , by increased duration of protein translation from a modified nucleic acid), or reduced innate immune response of the host cell.
  • aspects of the present disclosure are directed to methods of inducing in vivo translation of a recombinant polypeptide in a mammalian subject in need thereof.
  • an effective amount of a composition containing a nucleic acid that has at least one nucleoside modification and a translatable region encoding the recombinant polypeptide is administered to the subject using the delivery methods described herein.
  • the nucleic acid is provided in an amount and under other conditions such that the nucleic acid is localized into a cell of the subject and the recombinant polypeptide is translated in the cell from the nucleic acid.
  • the cell in which the nucleic acid is localized, or the tissue in which the cell is present, may be targeted with one or more than one rounds of nucleic acid administration.
  • compositions containing modified nucleic acids are formulated for administration intramuscularly,
  • the composition is formulated for extended release.
  • the subject to whom the therapeutic agent is administered suffers from or is at risk of developing a disease, disorder, or deleterious condition.
  • GWAS genome-wide association studies
  • the administered modified nucleic acid directs production of one or more recombinant polypeptides that provide a functional activity which is substantially absent in the cell in which the recombinant polypeptide is translated.
  • the missing functional activity may be enzymatic, structural, or gene regulatory in nature.
  • the administered modified nucleic acid directs production of one or more recombinant polypeptides that replace a polypeptide (or multiple polypeptides) that is substantially absent in the cell in which the recombinant polypeptide is translated. Such absence may be due to genetic mutation of the encoding gene or regulatory pathway thereof.
  • the recombinant polypeptide functions to antagonize the activity of an endogenous protein present in, on the surface of, or secreted from the cell. Usually, the activity of the endogenous protein is deleterious to the subject, for example, do to mutation of the endogenous protein resulting in altered activity or localization.
  • the recombinant polypeptide antagonizes, directly or indirectly, the activity of a biological moiety present in, on the surface of, or secreted from the cell.
  • antagonized biological moieties include lipids (e.g., cholesterol), a lipoprotein (e.g., low density lipoprotein), a nucleic acid, a carbohydrate, or a small molecule toxin.
  • the recombinant proteins described herein are engineered for localization within the cell, potentially within a specific compartment such as the nucleus, or are engineered for secretion from the cell or translocation to the plasma membrane of the cell.
  • a useful feature of the modified nucleic acids of the present disclosure is the capacity to reduce the innate immune response of a cell to an exogenous nucleic acid.
  • the cell is contacted with a first composition that contains a first dose of a first exogenous nucleic acid including a translatable region and at least one nucleoside modification, and the level of the innate immune response of the cell to the first exogenous nucleic acid is determined.
  • the cell is contacted with a second composition, which includes a second dose of the first exogenous nucleic acid, the second dose containing a lesser amount of the first exogenous nucleic acid as compared to the first dose.
  • the cell is contacted with a first dose of a second exogenous nucleic acid.
  • the second exogenous nucleic acid may contain one or more modified nucleosides, which may be the same or different from the first exogenous nucleic acid or, alternatively, the second exogenous nucleic acid may not contain modified nucleosides.
  • the steps of contacting the cell with the first composition and/or the second composition may be repeated one or more times. Additionally, efficiency of protein production (e.g., protein translation) in the cell is optionally determined, and the cell may be re-transfected with the first and/or second composition repeatedly until a target protein production efficiency is achieved.
  • Therapeutics for diseases and conditions Provided are methods for treating or preventing a symptom of diseases characterized by missing or aberrant protein activity, by replacing the missing protein activity or overcoming the aberrant protein activity. Because of the rapid initiation of protein production following introduction of modified mRNAs, as compared to viral DNA vectors, the compounds of the present disclosure are particularly advantageous in treating acute diseases such as sepsis, stroke, and myocardial infarction. Moreover, the lack of transcriptional regulation of the modified mRNAs of the present disclosure is advantageous in that accurate titration of protein production is achievable.
  • Diseases characterized by dysfunctional or aberrant protein activity include, but not limited to, cancer and proliferative diseases, genetic diseases (e.g., cystic fibrosis), autoimmune diseases, diabetes, neurodegenerative diseases, cardiovascular diseases, and metabolic diseases.
  • the present disclosure provides a method for treating such conditions or diseases in a subject by introducing nucleic acid or cell-based therapeutics containing the modified nucleic acids provided herein, wherein the modified nucleic acids encode for a protein that antagonizes or otherwise overcomes the aberrant protein activity present in the cell of the subject.
  • Specific examples of a dysfunctional protein are the missense mutation variants of the cystic fibrosis transmembrane conductance regulator (CFTR) gene, which produce a dysfunctional protein variant of CFTR protein, which causes cystic fibrosis.
  • CFTR cystic fibrosis transmembrane conductance regulator
  • the present disclosure provides a method for treating such conditions or diseases in a subject by introducing nucleic acid or cell-based therapeutics containing the modified nucleic acids provided herein, wherein the modified nucleic acids encode for a protein that replaces the protein activity missing from the target cells of the subject.
  • a dysfunctional protein are the nonsense mutation variants of the cystic fibrosis
  • CFTR transmembrane conductance regulator
  • RNA molecules are formulated for administration by inhalation.
  • the present disclosure provides a method for treating
  • hyperlipidemia in a subject by introducing into a cell population of the subject with a modified mRNA molecule encoding Sortilin, a protein recently characterized by genomic studies, thereby ameliorating the hyperlipidemia in a subject.
  • the SORTl gene encodes a trans-Golgi network (TGN) transmembrane protein called Sortilin.
  • TGN trans-Golgi network
  • Genetic studies have shown that one of five individuals has a single nucleotide polymorphism, rsl2740374, in the lpl3 locus of the SORTl gene that predisposes them to having low levels of low-density lipoprotein (LDL) and very-low- density lipoprotein (VLDL).
  • LDL low-density lipoprotein
  • VLDL very-low- density lipoprotein
  • Methods of the present disclosure enhance nucleic acid delivery into a cell population, in vivo, ex vivo, or in culture.
  • a cell culture containing a plurality of host cells ⁇ e.g., eukaryotic cells such as yeast or mammalian cells
  • the composition also generally contains a transfection reagent or other compound that increases the efficiency of enhanced nucleic acid uptake into the host cells.
  • the enhanced nucleic acid exhibits enhanced retention in the cell population, relative to a
  • the retention of the enhanced nucleic acid is greater than the retention of the unmodified nucleic acid. In some embodiments, it is at least about 50%, 75%, 90%, 95%, 100%, 150%, 200% or more than 200% greater than the retention of the unmodified nucleic acid. Such retention advantage may be achieved by one round of transfection with the enhanced nucleic acid, or may be obtained following repeated rounds of transfection.
  • the enhanced nucleic acid is delivered to a target cell population with one or more additional nucleic acids. Such delivery may be at the same time, or the enhanced nucleic acid is delivered prior to delivery of the one or more additional nucleic acids.
  • the additional one or more nucleic acids may be modified nucleic acids or unmodified nucleic acids. It is understood that the initial presence of the enhanced nucleic acids does not substantially induce an innate immune response of the cell population and, moreover, that the innate immune response will not be activated by the later presence of the unmodified nucleic acids. In this regard, the enhanced nucleic acid may not itself contain a translatable region, if the protein desired to be present in the target cell population is translated from the unmodified nucleic acids.
  • modified nucleic acids are provided to express a protein-binding partner or a receptor on the surface of the cell, which functions to target the cell to a specific tissue space or to interact with a specific moiety, either in vivo or in vitro.
  • Suitable protein- binding partners include antibodies and functional fragments thereof, scaffold proteins, or peptides.
  • modified nucleic acids can be employed to direct the synthesis and extracellular localization of lipids, carbohydrates, or other biological moieties.
  • a method for epigenetically silencing gene expression in a mammalian subject comprising a nucleic acid where the translatable region encodes a polypeptide or polypeptides capable of directing sequence-specific histone H3 methylation to initiate heterochromatin formation and reduce gene transcription around specific genes for the purpose of silencing the gene.
  • a gain-of-function mutation in the Janus Kinase 2 gene is responsible for the family of Myeloproliferative Diseases.
  • the present disclosure provides proteins generated from modified mRNAs.
  • compositions may optionally comprise one or more additional therapeutically active substances.
  • a method of administering pharmaceutical compositions comprising one or more proteins to be delivered to a subject in need thereof is provided.
  • compositions are administered to humans.
  • active ingredient generally refers to a protein or protein-containing complex as described herein.
  • compositions are principally directed to pharmaceutical compositions which are suitable for administration to humans, it will be understood by the skilled artisan that such compositions are generally suitable for
  • compositions suitable for administration to humans Modification of pharmaceutical compositions suitable for administration to humans in order to render the compositions suitable for administration to various animals is well understood, and the ordinarily skilled veterinary pharmacologist can design and/or perform such modification with merely ordinary, if any, experimentation.
  • Subjects to which administration of the pharmaceutical compositions is contemplated include, but are not limited to, humans and/or other primates; mammals, including commercially relevant mammals such as cattle, pigs, horses, sheep, cats, dogs, mice, and/or rats; and/or birds, including commercially relevant birds such as chickens, ducks, geese, and/or turkeys.
  • Formulations of the pharmaceutical compositions described herein may be prepared by any method known or hereafter developed in the art of pharmacology. In general, such preparatory methods include the step of bringing the active ingredient into association with an excipient and/or one or more other accessory ingredients, and then, if necessary and/or desirable, shaping and/or packaging the product into a desired single- or multi-dose unit.
  • a pharmaceutical composition in accordance with the present disclosure may be prepared, packaged, and/or sold in bulk, as a single unit dose, and/or as a plurality of single unit doses.
  • a "unit dose" is discrete amount of the pharmaceutical composition comprising a predetermined amount of the active ingredient.
  • the amount of the active ingredient is generally equal to the dosage of the active ingredient which would be administered to a subject and/or a convenient fraction of such a dosage such as, for example, one-half or one- third of such a dosage.
  • Relative amounts of the active ingredient, the pharmaceutically acceptable excipient, and/or any additional ingredients in a pharmaceutical composition in accordance with the present disclosure will vary, depending upon the identity, size, and/or condition of the subject treated and further depending upon the route by which the composition is to be administered.
  • the composition may comprise between 0.1% and 100% (w/w) active ingredient.
  • compositions may additionally comprise a pharmaceutically acceptable excipient, which, as used herein, includes any and all solvents, dispersion media, diluents, or other liquid vehicles, dispersion or suspension aids, surface active agents, isotonic agents, thickening or emulsifying agents, preservatives, solid binders, lubricants and the like, as suited to the particular dosage form desired.
  • a pharmaceutically acceptable excipient includes any and all solvents, dispersion media, diluents, or other liquid vehicles, dispersion or suspension aids, surface active agents, isotonic agents, thickening or emulsifying agents, preservatives, solid binders, lubricants and the like, as suited to the particular dosage form desired.
  • Remington's The Science and Practice of Pharmacy, 21 st Edition, A. R. Gennaro discloses various excipients used in formulating pharmaceutical
  • compositions and known techniques for the preparation thereof Except insofar as any conventional excipient medium is incompatible with a substance or its derivatives, such as by producing any undesirable biological effect or otherwise interacting in a deleterious manner with any other component(s) of the pharmaceutical composition, its use is contemplated to be within the scope of this present disclosure.
  • a pharmaceutically acceptable excipient is at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% pure.
  • an excipient is approved for use in humans and for veterinary use.
  • an excipient is approved by United States Food and Drug Administration.
  • an excipient is pharmaceutical grade.
  • an excipient meets the standards of the United States Pharmacopoeia (USP), the European Pharmacopoeia (EP), the British Pharmacopoeia, and/or the International Pharmacopoeia.
  • compositions used in the manufacture of pharmaceutical compositions include, but are not limited to, inert diluents, dispersing and/or granulating agents, surface active agents and/or emulsifiers, disintegrating agents, binding agents, preservatives, buffering agents, lubricating agents, and/or oils. Such excipients may optionally be included in pharmaceutical formulations. Excipients such as cocoa butter and suppository waxes, coloring agents, coating agents, sweetening, flavoring, and/or perfuming agents can be present in the composition, according to the judgment of the formulator.
  • Exemplary diluents include, but are not limited to, calcium carbonate, sodium carbonate, calcium phosphate, dicalcium phosphate, calcium sulfate, calcium hydrogen phosphate, sodium phosphate lactose, sucrose, cellulose, microcrystalline cellulose, kaolin, mannitol, sorbitol, inositol, sodium chloride, dry starch, cornstarch, powdered sugar, etc., and/or combinations thereof.
  • Exemplary granulating and/or dispersing agents include, but are not limited to, potato starch, corn starch, tapioca starch, sodium starch glycolate, clays, alginic acid, guar gum, citrus pulp, agar, bentonite, cellulose and wood products, natural sponge, cation-exchange resins, calcium carbonate, silicates, sodium carbonate, cross-linked poly(vinyl-pyrrolidone)
  • crospovidone sodium carboxymethyl starch (sodium starch glycolate), carboxymethyl cellulose, cross-linked sodium carboxymethyl cellulose (croscarmellose), methylcellulose, pregelatinized starch (starch 1500), microcrystalline starch, water insoluble starch, calcium carboxymethyl cellulose, magnesium aluminum silicate (Veegum), sodium lauryl sulfate, quaternary ammonium compounds, etc., and/or combinations thereof.
  • Exemplary surface active agents and/or emulsifiers include, but are not limited to, natural emulsifiers ⁇ e.g. acacia, agar, alginic acid, sodium alginate, tragacanth, chondrux, cholesterol, xanthan, pectin, gelatin, egg yolk, casein, wool fat, cholesterol, wax, and lecithin), colloidal clays (e.g. bentonite [aluminum silicate] and Veegum ® [magnesium aluminum silicate]), long chain amino acid derivatives, high molecular weight alcohols (e.g.
  • natural emulsifiers ⁇ e.g. acacia, agar, alginic acid, sodium alginate, tragacanth, chondrux, cholesterol, xanthan, pectin, gelatin, egg yolk, casein, wool fat, cholesterol, wax, and lecithin
  • colloidal clays e.g. bentonite [alumin
  • stearyl alcohol cetyl alcohol, oleyl alcohol, triacetin monostearate, ethylene glycol distearate, glyceryl monostearate, and propylene glycol monostearate, polyvinyl alcohol), carbomers (e.g. carboxy polymethylene, polyacrylic acid, acrylic acid polymer, and carboxyvinyl polymer), carrageenan, cellulosic derivatives (e.g. carboxymethylcellulose sodium, powdered cellulose, hydroxymethyl cellulose, hydroxypropyl cellulose, hydroxypropyl methylcellulose, methylcellulose), sorbitan fatty acid esters (e.g.
  • polyoxyethylene monostearate [Myrj ® 45], polyoxyethylene hydrogenated castor oil, polyethoxylated castor oil, polyoxymethylene stearate, and Solutol ® ), sucrose fatty acid esters, polyethylene glycol fatty acid esters (e.g. Cremophor ® ), polyoxyethylene ethers, (e.g.
  • polyoxyethylene lauryl ether [Brij ® 30]), poly(vinyl-pyrrolidone), diethylene glycol monolaurate, triethanolamine oleate, sodium oleate, potassium oleate, ethyl oleate, oleic acid, ethyl laurate, sodium lauryl sulfate, Pluronic ® F 68, Poloxamer ® 188, cetrimonium bromide, cetylpyridinium chloride, benzalkonium chloride, docusate sodium, etc. and/or combinations thereof.
  • Exemplary binding agents include, but are not limited to, starch ⁇ e.g. cornstarch and starch paste); gelatin; sugars ⁇ e.g. sucrose, glucose, dextrose, dextrin, molasses, lactose, lactitol, mannitol,); natural and synthetic gums ⁇ e.g.
  • acacia sodium alginate, extract of Irish moss, panwar gum, ghatti gum, mucilage of isapol husks, carboxymethylcellulose, methylcellulose, ethylcellulose, hydroxyethylcellulose, hydroxypropyl cellulose, hydroxypropyl methylcellulose, microcrystalline cellulose, cellulose acetate, poly(vinyl-pyrrolidone), magnesium aluminum silicate (Veegum ® ), and larch arabogalactan); alginates; polyethylene oxide; polyethylene glycol; inorganic calcium salts; silicic acid; polymethacrylates; waxes; water; alcohol; etc.; and combinations thereof.
  • Exemplary preservatives may include, but are not limited to, antioxidants, chelating agents, antimicrobial preservatives, antifungal preservatives, alcohol preservatives, acidic preservatives, and/or other preservatives.
  • Exemplary antioxidants include, but are not limited to, alpha tocopherol, ascorbic acid, acorbyl palmitate, butylated hydroxyanisole, butylated hydroxytoluene, monothioglycerol, potassium metabisulfite, propionic acid, propyl gallate, sodium ascorbate, sodium bisulfite, sodium metabisulfite, and/or sodium sulfite.
  • Exemplary chelating agents include ethylenediaminetetraacetic acid (EDTA), citric acid monohydrate, disodium edetate, dipotassium edetate, edetic acid, fumaric acid, malic acid, phosphoric acid, sodium edetate, tartaric acid, and/or trisodium edetate.
  • EDTA ethylenediaminetetraacetic acid
  • citric acid monohydrate disodium edetate
  • dipotassium edetate dipotassium edetate
  • edetic acid fumaric acid, malic acid, phosphoric acid, sodium edetate, tartaric acid, and/or trisodium edetate.
  • antimicrobial preservatives include, but are not limited to, benzalkonium chloride, benzethonium chloride, benzyl alcohol, bronopol, cetrimide, cetylpyridinium chloride, chlorhexidine, chlorobutanol, chlorocresol, chloroxylenol, cresol, ethyl alcohol, glycerin, hexetidine, imidurea, phenol, phenoxyethanol, phenylethyl alcohol, phenylmercuric nitrate, propylene glycol, and/or thimerosal.
  • antifungal preservatives include, but are not limited to, butyl paraben, methyl paraben, ethyl paraben, propyl paraben, benzoic acid, hydroxybenzoic acid, potassium benzoate, potassium sorbate, sodium benzoate, sodium propionate, and/or sorbic acid.
  • exemplary alcohol e.g., butyl paraben, methyl paraben, ethyl paraben, propyl paraben, benzoic acid, hydroxybenzoic acid, potassium benzoate, potassium sorbate, sodium benzoate, sodium propionate, and/or sorbic acid.
  • preservatives include, but are not limited to, ethanol, polyethylene glycol, phenol, phenolic compounds, bisphenol, chlorobutanol, hydroxybenzoate, and/or phenylethyl alcohol.
  • acidic preservatives include, but are not limited to, vitamin A, vitamin C, vitamin E, beta- carotene, citric acid, acetic acid, dehydroacetic acid, ascorbic acid, sorbic acid, and/or phytic acid.
  • preservatives include, but are not limited to, tocopherol, tocopherol acetate, deteroxime mesylate, cetrimide, butylated hydroxyanisol (BHA), butylated hydroxytoluened (BHT), ethylenediamine, sodium lauryl sulfate (SLS), sodium lauryl ether sulfate (SLES), sodium bisulfite, sodium metabisulfite, potassium sulfite, potassium metabisulfite, Glydant Plus ® , Phenonip ® , methylparaben, Germall ® 115, Germaben ® II, Neolone TM , Kathon TM , and/or Euxyl ® .
  • Exemplary buffering agents include, but are not limited to, citrate buffer solutions, acetate buffer solutions, phosphate buffer solutions, ammonium chloride, calcium carbonate, calcium chloride, calcium citrate, calcium glubionate, calcium gluceptate, calcium gluconate, d- gluconic acid, calcium glycerophosphate, calcium lactate, propanoic acid, calcium levulinate, pentanoic acid, dibasic calcium phosphate, phosphoric acid, tribasic calcium phosphate, calcium hydroxide phosphate, potassium acetate, potassium chloride, potassium gluconate, potassium mixtures, dibasic potassium phosphate, monobasic potassium phosphate, potassium phosphate mixtures, sodium acetate, sodium bicarbonate, sodium chloride, sodium citrate, sodium lactate, dibasic sodium phosphate, monobasic sodium phosphate, sodium phosphate mixtures, tromethamine, magnesium hydroxide, aluminum hydroxide, alginic acid, pyrogen-free water, iso
  • Exemplary lubricating agents include, but are not limited to, magnesium stearate, calcium stearate, stearic acid, silica, talc, malt, glyceryl behanate, hydrogenated vegetable oils, polyethylene glycol, sodium benzoate, sodium acetate, sodium chloride, leucine, magnesium lauryl sulfate, sodium lauryl sulfate, etc., and combinations thereof.
  • oils include, but are not limited to, almond, apricot kernel, avocado, babassu, bergamot, black current seed, borage, cade, camomile, canola, caraway, carnauba, castor, cinnamon, cocoa butter, coconut, cod liver, coffee, corn, cotton seed, emu, eucalyptus, evening primrose, fish, flaxseed, geraniol, gourd, grape seed, hazel nut, hyssop, isopropyl myristate, jojoba, kukui nut, lavandin, lavender, lemon, litsea cubeba, macademia nut, mallow, mango seed, meadowfoam seed, mink, nutmeg, olive, orange, orange roughy, palm, palm kernel, peach kernel, peanut, poppy seed, pumpkin seed, rapeseed, rice bran, rosemary, safflower, sandalwood, sasquana, savoury
  • oils include, but are not limited to, butyl stearate, caprylic triglyceride, capric triglyceride, cyclomethicone, diethyl sebacate, dimethicone 360, isopropyl myristate, mineral oil, octyldodecanol, oleyl alcohol, silicone oil, and/or combinations thereof.
  • Liquid dosage forms for oral and parenteral administration include, but are not limited to, pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups, and/or elixirs.
  • liquid dosage forms may comprise inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, dimethylformamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof.
  • inert diluents commonly used in the art such as, for example,
  • oral compositions can include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and/or perfuming agents.
  • adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and/or perfuming agents.
  • compositions are mixed with solubilizing agents such as Cremophor ® , alcohols, oils, modified oils, glycols, polysorbates, cyclodextrins, polymers, and/or combinations thereof.
  • Injectable preparations for example, sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing agents, wetting agents, and/or suspending agents.
  • Sterile injectable preparations may be sterile injectable solutions, suspensions, and/or emulsions in nontoxic parenterally acceptable diluents and/or solvents, for example, as a solution in 1,3-butanediol.
  • the acceptable vehicles and solvents that may be employed are water, Ringer's solution, U.S.P., and isotonic sodium chloride solution.
  • Sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil can be employed including synthetic mono- or diglycerides.
  • Fatty acids such as oleic acid can be used in the preparation of injectables.
  • Injectable formulations can be sterilized, for example, by filtration through a bacterial- retaining filter, and/or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved or dispersed in sterile water or other sterile injectable medium prior to use.
  • sterilizing agents in the form of sterile solid compositions which can be dissolved or dispersed in sterile water or other sterile injectable medium prior to use.
  • delayed absorption of a parenterally administered drug form is accomplished by dissolving or suspending the drug in an oil vehicle.
  • injectable depot forms are made by forming microencapsule matrices of the drug in biodegradable polymers such as polylactide-polyglycolide. Depending upon the ratio of drug to polymer and the nature of the particular polymer employed, the rate of drug release can be controlled. Examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides). Depot injectable formulations are prepared by entrapping the drug in liposomes or microemulsions which are compatible with body tissues.
  • compositions for rectal or vaginal administration are typically suppositories which can be prepared by mixing compositions with suitable non-irritating excipients such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active ingredient.
  • suitable non-irritating excipients such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active ingredient.
  • Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules.
  • an active ingredient is mixed with at least one inert, pharmaceutically acceptable excipient such as sodium citrate or dicalcium phosphate and/or fillers or extenders (e.g. starches, lactose, sucrose, glucose, mannitol, and silicic acid), binders (e.g. carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidinone, sucrose, and acacia), humectants (e.g. glycerol), disintegrating agents (e.g.
  • the dosage form may comprise buffering agents.
  • solution retarding agents e.g. paraffin
  • absorption accelerators e.g. quaternary ammonium compounds
  • wetting agents e.g. cetyl alcohol and glycerol monostearate
  • absorbents e.g. kaolin and bentonite clay
  • lubricants e.g. talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate
  • the dosage form may comprise buffering agents.
  • Solid compositions of a similar type may be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like.
  • Solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings and other coatings well known in the pharmaceutical formulating art. They may optionally comprise opacifying agents and can be of a composition that they release the active ingredient(s) only, or
  • embedding compositions which can be used include polymeric substances and waxes.
  • Solid compositions of a similar type may be employed as fillers in soft and hard- filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like.
  • Dosage forms for topical and/or transdermal administration of a composition may include ointments, pastes, creams, lotions, gels, powders, solutions, sprays, inhalants and/or patches.
  • an active ingredient is admixed under sterile conditions with a pharmaceutically acceptable excipient and/or any needed preservatives and/or buffers as may be required.
  • transdermal patches which often have the added advantage of providing controlled delivery of a compound to the body.
  • dosage forms may be prepared, for example, by dissolving and/or dispensing the compound in the proper medium.
  • rate may be controlled by either providing a rate controlling membrane and/or by dispersing the compound in a polymer matrix and/or gel.
  • Suitable devices for use in delivering intradermal pharmaceutical compositions described herein include short needle devices such as those described in U.S. Patents 4,886,499; 5,190,521; 5,328,483; 5,527,288; 4,270,537; 5,015,235; 5,141,496; and 5,417,662.
  • compositions may be administered by devices which limit the effective penetration length of a needle into the skin, such as those described in PCT publication WO 99/34850 and functional equivalents thereof.
  • Jet injection devices which deliver liquid compositions to the dermis via a liquid jet injector and/or via a needle which pierces the stratum corneum and produces a jet which reaches the dermis are suitable. Jet injection devices are described, for example, in U.S. Patents 5,480,381; 5,599,302; 5,334,144; 5,993,412; 5,649,912; 5,569,189; 5,704,911;
  • Ballistic powder/particle delivery devices which use compressed gas to accelerate vaccine in powder form through the outer layers of the skin to the dermis are suitable.
  • conventional syringes may be used in the classical mantoux method of intradermal administration.
  • Formulations suitable for topical administration include, but are not limited to, liquid and/or semi liquid preparations such as liniments, lotions, oil in water and/or water in oil emulsions such as creams, ointments and/or pastes, and/or solutions and/or suspensions.
  • liquid and/or semi liquid preparations such as liniments, lotions, oil in water and/or water in oil emulsions such as creams, ointments and/or pastes, and/or solutions and/or suspensions.
  • Topically-administrable formulations may, for example, comprise from about 1% to about 10% (w/w) active ingredient, although the concentration of active ingredient may be as high as the solubility limit of the active ingredient in the solvent.
  • Formulations for topical administration may further comprise one or more of the additional ingredients described herein.
  • a pharmaceutical composition may be prepared, packaged, and/or sold in a formulation suitable for pulmonary administration via the buccal cavity.
  • a formulation may comprise dry particles which comprise the active ingredient and which have a diameter in the range from about 0.5 nm to about 7 nm or from about 1 nm to about 6 nm.
  • Such compositions are conveniently in the form of dry powders for administration using a device comprising a dry powder reservoir to which a stream of propellant may be directed to disperse the powder and/or using a self propelling solvent/powder dispensing container such as a device comprising the active ingredient dissolved and/or suspended in a low-boiling propellant in a sealed container.
  • Such powders comprise particles wherein at least 98% of the particles by weight have a diameter greater than 0.5 nm and at least 95% of the particles by number have a diameter less than 7 nm. Alternatively, at least 95% of the particles by weight have a diameter greater than 1 nm and at least 90% of the particles by number have a diameter less than 6 nm.
  • Dry powder compositions may include a solid fine powder diluent such as sugar and are conveniently provided in a unit dose form.
  • Low boiling propellants generally include liquid propellants having a boiling point of below 65 °F at atmospheric pressure. Generally the propellant may constitute 50% to 99.9% (w/w) of the composition, and active ingredient may constitute 0.1 % to 20% (w/w) of the composition.
  • a propellant may further comprise additional ingredients such as a liquid non- ionic and/or solid anionic surfactant and/or a solid diluent (which may have a particle size of the same order as particles comprising the active ingredient).
  • compositions formulated for pulmonary delivery may provide an active ingredient in the form of droplets of a solution and/or suspension.
  • Such formulations may be prepared, packaged, and/or sold as aqueous and/or dilute alcoholic solutions and/or suspensions, optionally sterile, comprising active ingredient, and may conveniently be administered using any nebulization and/or atomization device.
  • Such formulations may further comprise one or more additional ingredients including, but not limited to, a flavoring agent such as saccharin sodium, a volatile oil, a buffering agent, a surface active agent, and/or a preservative such as
  • Droplets provided by this route of administration may have an average diameter in the range from about 0.1 nm to about 200 nm.
  • Formulations described herein as being useful for pulmonary delivery are useful for intranasal delivery of a pharmaceutical composition.
  • Another formulation suitable for intranasal administration is a coarse powder comprising the active ingredient and having an average particle from about 0.2 ⁇ to 500 ⁇ .
  • Such a formulation is administered in the manner in which snuff is taken, i.e. by rapid inhalation through the nasal passage from a container of the powder held close to the nose.
  • Formulations suitable for nasal administration may, for example, comprise from about as little as 0.1% (w/w) and as much as 100% (w/w) of active ingredient, and may comprise one or more of the additional ingredients described herein.
  • a pharmaceutical composition may be prepared, packaged, and/or sold in a formulation suitable for buccal administration. Such formulations may, for example, be in the form of tablets and/or lozenges made using
  • formulations suitable for buccal administration may comprise a powder and/or an aerosolized and/or atomized solution and/or suspension comprising active ingredient.
  • Such powdered, aerosolized, and/or aerosolized formulations, when dispersed, may have an average particle and/or droplet size in the range from about 0.1 nm to about 200 nm, and may further comprise one or more of any additional ingredients described herein.
  • a pharmaceutical composition may be prepared, packaged, and/or sold in a formulation suitable for ophthalmic administration.
  • Such formulations may, for example, be in the form of eye drops including, for example, a 0.1/1.0%) (w/w) solution and/or suspension of the active ingredient in an aqueous or oily liquid excipient.
  • Such drops may further comprise buffering agents, salts, and/or one or more other of any additional ingredients described herein.
  • Other opthalmically-administrable formulations which are useful include those which comprise the active ingredient in microcrystalline form and/or in a liposomal preparation. Ear drops and/or eye drops are contemplated as being within the scope of this present disclosure.
  • the present disclosure provides methods comprising administering proteins or complexes in accordance with the present disclosure to a subject in need thereof.
  • Proteins or complexes, or pharmaceutical, imaging, diagnostic, or prophylactic compositions thereof may be administered to a subject using any amount and any route of administration effective for preventing, treating, diagnosing, or imaging a disease, disorder, and/or condition ⁇ e.g., a disease, disorder, and/or condition relating to working memory deficits).
  • the exact amount required will vary from subject to subject, depending on the species, age, and general condition of the subject, the severity of the disease, the particular composition, its mode of administration, its mode of activity, and the like.
  • Compositions in accordance with the present disclosure are typically formulated in dosage unit form for ease of administration and uniformity of dosage.
  • compositions of the present disclosure will be decided by the attending physician within the scope of sound medical judgment.
  • the specific therapeutically effective, prophylactially effective, or appropriate imaging dose level for any particular patient will depend upon a variety of factors including the disorder being treated and the severity of the disorder; the activity of the specific compound employed; the specific composition employed; the age, body weight, general health, sex and diet of the patient; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidental with the specific compound employed; and like factors well known in the medical arts.
  • Proteins to be delivered and/or pharmaceutical, prophylactic, diagnostic, or imaging compositions thereof may be administered to animals, such as mammals ⁇ e.g., humans, domesticated animals, cats, dogs, mice, rats, etc.). In some embodiments, pharmaceutical, prophylactic, diagnostic, or imaging compositions thereof are administered to humans. Proteins to be delivered and/or pharmaceutical, prophylactic, diagnostic, or imaging compositions thereof in accordance with the present disclosure may be administered by any route.
  • proteins and/or pharmaceutical, prophylactic, diagnostic, or imaging compositions thereof are administered by one or more of a variety of routes, including oral, intravenous, intramuscular, intra-arterial, intramedullary, intrathecal, subcutaneous, intraventricular, transdermal, interdermal, rectal, intravaginal, intraperitoneal, topical (e.g.
  • proteins or complexes, and/or pharmaceutical, prophylactic, diagnostic, or imaging compositions thereof are administered by systemic intravenous injection.
  • proteins or complexes and/or pharmaceutical, prophylactic, diagnostic, or imaging compositions thereof may be administered intravenously and/or orally.
  • proteins or complexes, and/or pharmaceutical, prophylactic, diagnostic, or imaging compositions thereof may be administered in a way which allows the protein or complex to cross the blood- brain barrier, vascular barrier, or other epithelial barrier.
  • the present disclosure encompasses the delivery of proteins or complexes, and/or pharmaceutical, prophylactic, diagnostic, or imaging compositions thereof, by any appropriate route taking into consideration likely advances in the sciences of drug delivery.
  • the most appropriate route of administration will depend upon a variety of factors including the nature of the protein or complex comprising proteins associated with at least one agent to be delivered (e.g., its stability in the environment of the gastrointestinal tract, bloodstream, etc.), the condition of the patient (e.g., whether the patient is able to tolerate particular routes of administration), etc.
  • the present disclosure encompasses the delivery of the pharmaceutical, prophylactic, diagnostic, or imaging compositions by any appropriate route taking into consideration likely advances in the sciences of drug delivery.
  • compositions in accordance with the present disclosure may be administered at dosage levels sufficient to deliver from about 0.0001 mg/kg to about 100 mg/kg, from about 0.01 mg/kg to about 50 mg/kg, from about 0.1 mg/kg to about 40 mg/kg, from about 0.5 mg/kg to about 30 mg/kg, from about 0.01 mg/kg to about 10 mg/kg, from about 0.1 mg/kg to about 10 mg/kg, or from about 1 mg/kg to about 25 mg/kg, of subject body weight per day, one or more times a day, to obtain the desired therapeutic, diagnostic, prophylactic, or imaging effect.
  • the desired dosage may be delivered three times a day, two times a day, once a day, every other day, every third day, every week, every two weeks, every three weeks, or every four weeks. In certain embodiments, the desired dosage may be delivered using multiple
  • administrations e.g., two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, fourteen, or more administrations.
  • Proteins or complexes may be used in combination with one or more other therapeutic, prophylactic, diagnostic, or imaging agents.
  • combination with it is not intended to imply that the agents must be administered at the same time and/or formulated for delivery together, although these methods of delivery are within the scope of the present disclosure.
  • Compositions can be administered concurrently with, prior to, or subsequent to, one or more other desired therapeutics or medical procedures. In general, each agent will be administered at a dose and/or on a time schedule determined for that agent.
  • the present disclosure encompasses the delivery of pharmaceutical, prophylactic, diagnostic, or imaging compositions in combination with agents that improve their bioavailability, reduce and/or modify their metabolism, inhibit their excretion, and/or modify their distribution within the body.
  • therapeutically, prophylactically, diagnostically, or imaging active agents utilized in combination may be administered together in a single composition or administered separately in different compositions.
  • agents utilized in combination with be utilized at levels that do not exceed the levels at which they are utilized individually. In some embodiments, the levels utilized in combination will be lower than those utilized individually.
  • kits for example, a composition useful for treating cancer in accordance with the present disclosure may be administered concurrently with a chemotherapeutic agent), or they may achieve different effects (e.g., control of any adverse effects).
  • kits for conveniently and/or effectively carrying out methods of the present disclosure.
  • kits will comprise sufficient amounts and/or numbers of components to allow a user to perform multiple treatments of a subject(s) and/or to perform multiple experiments.
  • kits for protein production comprising a first isolated nucleic acid comprising a translatable region and a nucleic acid modification, wherein the nucleic acid is capable of evading an innate immune response of a cell into which the first isolated nucleic acid is introduced, and packaging and instructions.
  • kits for protein production comprising: a first isolated nucleic acid comprising a translatable region, provided in an amount effective to produce a desired amount of a protein encoded by the translatable region when introduced into a target cell; a second nucleic acid comprising an inhibitory nucleic acid, provided in an amount effective to substantially inhibit the innate immune response of the cell; and packaging and instructions.
  • kits for protein production comprising a first isolated nucleic acid comprising a translatable region and a nucleoside modification, wherein the nucleic acid exhibits reduced degradation by a cellular nuclease, and packaging and instructions.
  • kits for protein production comprising a first isolated nucleic acid comprising a translatable region and at least two different nucleoside modifications, wherein the nucleic acid exhibits reduced degradation by a cellular nuclease, and packaging and instructions.
  • kits for protein production comprising a first isolated nucleic acid comprising a translatable region and at least one nucleoside modification, wherein the nucleic acid exhibits reduced degradation by a cellular nuclease; a second nucleic acid comprising an inhibitory nucleic acid; and packaging and instructions.
  • the first isolated nucleic acid comprises messenger RNA
  • the mRNA comprises at least one nucleoside selected from the group consisting of pyridin-4-one ribonucleoside, 5-aza-uridine, 2-thio-5-aza-uridine, 2- thiouridine, 4-thio-pseudouridine, 2-thio-pseudouridine, 5-hydroxyuridine, 3-methyluridine, 5- carboxymethyl-uridine, 1-carboxymethyl-pseudouridine, 5-propynyl-uridine, 1-propynyl- pseudouridine, 5-taurinomethyluridine, 1-taurinomethyl-pseudouridine, 5-taurinomethyl-2-thio- uridine, l-taurinomethyl-4-thio-uridine, 5-methyl-uridine, 1-methyl-pseudouridine, 4-thio- 1- methyl-pseudouridine, 2-thio- 1-methyl-pseudouridine, 1 -methyl- 1-d
  • 2- thio-dihydropseudouridine 2-methoxyuridine, 2-methoxy-4-thio-uridine, 4-methoxy- pseudouridine, and 4-methoxy-2-thio-pseudouridine.
  • the mRNA comprises at least one nucleoside selected from the group consisting of 5-aza-cytidine, pseudoisocytidine, 3-methyl-cytidine, N4-acetylcytidine, 5- formylcytidine, N4-methylcytidine, 5-hydroxymethylcytidine, 1-methyl-pseudoisocytidine, pyrrolo-cytidine, pyrrolo-pseudoisocytidine, 2-thio-cytidine, 2-thio-5-methyl-cytidine, 4-thio- pseudoisocytidine, 4-thio- 1 -methyl-pseudoisocytidine, 4-thio- 1 -methyl- 1 -deaza- pseudoisocytidine, 1 -methyl- 1-deaza-pseudoisocytidine, zebularine, 5-aza-zebularine, 5-methyl- zebularine, 5-aza-2-thio
  • the mRNA comprises at least one nucleoside selected from the group consisting of 2-aminopurine, 2, 6-diaminopurine, 7-deaza-adenine, 7-deaza-8-aza-adenine, 7-deaza-2-aminopurine, 7-deaza-8-aza-2-aminopurine, 7-deaza-2, 6-diaminopurine, 7-deaza-8-aza-2, 6-diaminopurine, 1-methyladenosine, N6-methyladenosine, N6-isopentenyladenosine, N6- (cis-hydroxyisopentenyl)adenosine, 2-methylthio-N6-(cis-hydroxyisopentenyl) adenosine, N6- glycinylcarbamoyladenosine, N6-threonylcarbamoyladenosine, 2-methylthio-N6-threonyl carbamoyladen
  • the mRNA comprises at least one nucleoside selected from the group consisting of inosine, 1-methyl-inosine, wyosine, wybutosine, 7-deaza-guanosine, 7- deaza-8-aza-guanosine, 6-thio-guanosine, 6-thio-7-deaza-guanosine, 6-thio-7-deaza-8-aza- guanosine, 7-methyl-guanosine, 6-thio-7-methyl-guanosine, 7-methylinosine, 6-methoxy- guanosine, 1-methylguanosine, N2-methylguanosine, N2,N2-dimethylguanosine, 8-oxo- guanosine, 7-methyl-8-oxo-guanosine, l-methyl-6-thio-guanosine, N2-methyl-6-thio-guanosine, and N2,N2-dimethyl-6-thio-guanosine.
  • nucleoside selected from the
  • compositions for protein production comprising a first isolated nucleic acid comprising a translatable region and a nucleoside modification, wherein the nucleic acid exhibits reduced degradation by a cellular nuclease, and a mammalian cell suitable for translation of the translatable region of the first nucleic acid.
  • Modified mRNAs were made using standard laboratory methods and materials for in vitro transcription with the exception that the nucleotide mix contained modified nucleotides.
  • the open reading frame (ORF) of the gene of interest is flanked by a 5' untranslated region (UTR) containing a strong Kozak translational initiation signal and an alpha-globin 3' UTR terminating with an oligo(dT) sequence for templated addition of a polyA tail for modRNAs not incorporating Adenosine analogs.
  • Adenosine-containing modRNAs were synthesized without an oligo (dT) sequence to allow for post-transcription poly (A) polymerase poly-(A) tailing.
  • the modRNAs were modified by incorporating chemically modified nucleotides indicated in Table 3 (below) during the in vitro transcription with 100% replacement of the corresponding natural nucleotide or partial replacement of the corresponding natural nucleotide at the indicated percentage.
  • Table 3 indicates the chemical identity of each chemically-distinct modified nucleotide incorporated into a modified mRNA with the given chemistry designation number.
  • Chem 8 Pseudo-iso-cytidine/a-thio-uridine Chem 36 a-thio-uridine Chem 9 Pseudo-iso-cytidine/5-methyl-uridine Chem 37
  • Nanodrop modRNA quantification and UV spectral data modRNAs in TE buffer (1 ⁇ ) were used for Nanodrop UV absorbance readings to quantitate the yield of each modRNA from an in vitro transcription reaction (UV absorbance traces are shown in Figures 6A-6L). These data indicate which chemically modified nucleotides were transcribed into chemically- modified mRNAs. These data also demonstrate that nucleotides with chemical modifications on the major groove and minor groove face of the nucleotide were capable of being transcribed into a modRNA. These data further demonstrate that the nucleotides of the present invention are transcription-competent and compatible with incorporation into a modRNA, which may have altered UV spectra due to the presence of a given modified nucleotide.
  • Pyrrolo-C containing modRNAs have an increase in UV absorbance at a lower wavelength due to the presence of the pyrrolo ring of the modified C nucleotide.
  • 2-amino-adenine nucleotide-containing modRNAs have an increase in UV absorbance at a higher wavelength due to the presence of an exocyclic amine off the purine ring.
  • Nucleotides that are not transcription- competent and cannot be incorporated into a modRNA have a scrambled UV spectrum indicating no product from the transcription reaction.
  • RNAiMAX RNAiMAX was prepared as described and mixed with the cells in the multi-well plate within a period of time, e.g., 6 hours, of cell seeding before cells had adhered to the tissue culture plate.
  • Keratinocytes were seeded at a cell density of 0.7 x 10 5 .
  • Keratinocytes were seeded at a cell density of 0.3 x 10 5 .
  • RNAiMAX RNAiMAX was prepared as described and transfected onto the cells in the multi-well plate over 24 hours after cell seeding and adherence to the tissue culture plate.
  • FIGs. 2A and 2B show an Enzyme-linked immunosorbent assay (ELISA) for Human Granulocyte-Colony Stimulating Factor (hu-G-CSF) of in vitro transfected Human Keratinocyte cells. Keratinocytes were grown in EpiLife medium with Supplement S7 from Invitrogen at a confluence of >70%.
  • FIG. 2A keratinocytes were reverse transfected with 300 ng of the indicated chemically modified mRNA complexed with RNAiMAX from Invitrogen.
  • FIG. 2B keratinocytes were forward transfected with 300 ng modRNA complexed with RNAiMAX from Invitrogen.
  • RNA:RNAiMAX complex was formed by first incubating the RNA with Supplement- free EpiLife media in a 5X volumetric dilution for 10 minutes at room temperature. In a second vial, RNAiMAX reagent was incubated with Supplement- free EpiLife Media in a 10X volumetric dilution for 10 minutes at room temperature. The RNA vial was then mixed with the RNAiMAX vial and incubated for 20-30 at room temperature before being added to the cells in a drop-wise fashion. Secreted huG-CSF concentration in the culture medium was measured at 18 hours post-transfection for each of the chemically modified mRNAs in triplicate.
  • G-CSF Human Granulocyte-Colony Stimulating Factor
  • FIGs. 3A-N show Enzyme-linked immunosorbent assays (ELISA) for Human
  • G-CSF Granulocyte-Colony Stimulating Factor
  • G-CSF Stimulating Factor
  • modified RNA containing modified nucleotides with chemical modifications on the major groove face of pyrimidine analogs have the highest levels of secreted hu-G-CSF into the cell culture medium and that 750ng of modRNA elicits the highest level of secreted hu-G-CSF.
  • FIGs. 4A-F show an Enzyme-linked immunosorbent assay (ELISA) for Human Tumor Necrosis Factor-a (TNF-a) (FIGs. 4A and 4B); Human Interferon- ⁇ (IFN- ⁇ ) (FIGs. 4C and 4D); and Human Granulocyte-Colony Stimulating Factor (G-CSF) (FIGs. 4E and 4F) secreted from in vzYro-transfected Human Keratinocyte cells. Keratinocytes were grown in EpiLife medium with Human Keratinocyte Growth Supplement in the absence of hydrocortisone from Invitrogen at a confluence of >70%. In FIGs.
  • ELISA Enzyme-linked immunosorbent assay
  • keratinocytes were reverse transfected with Ong, 93.75ng, 187.5ng, 375ng, 750ng, 1500ng or 3000ng of the indicated chemically modified mRNA complexed with RNAiMAX from Invitrogen as described in triplicate.
  • Secreted TNF-a in the culture medium was measured 24 hours post-transfection for each of the chemically modified mRNAs using an ELISA kit from Invitrogen according to the manufacturer protocols.
  • G-CSF Human Granulocyte-Colony Stimulating Factor
  • modified RNAs containing modified nucleotides were capable of eliciting a reduced cellular innate immune response in comparison to natural and other chemically modified nucleotides by measuring exemplary type I cytokines TNF-a and IFN- ⁇ . These data show that modified RNAs containing modified nucleotides with chemical
  • modifications on the major groove face of pyrimidine analogs have the lowest levels of secreted TNF-a and IFN- ⁇ into the cell culture medium while maintaining high levels of modRNA- encoding hu-G-CSF secretion into the cell culture medium.
  • FIGs. 5A-D show modRNA-encoding hu-G-CSF produced by a human keratinocyte feeder cell layer induced the proliferation of both human myeloblast cells KG-1 and Kasumi-1 that express the G-CSF-receptor where the cell populations are separated by a semi-permeable membrane.
  • 5A is a table showing the results from an Enzyme-linked immunosorbent assay (ELISA) for human-G-CSF secreted from in vzYro-transfected Human Keratinocyte cells sampled from individual wells in a co-culture 24- well tissue culture plate 42 hours post-transfection with 750ng of each indicated hu-G-CSF- encoding modRNA.
  • ELISA Enzyme-linked immunosorbent assay
  • G-CSF Human Granulocyte-Colony Stimulating Factor
  • RNA from sample cells was extracted and lysed using RNeasy kit (Qiagen, Valencia, CA) according to the manufacturer instructions. Extracted total RNA was submitted to RT-PCR for specific amplification of modRNA-G-CSF using ProtoScript® M-MuLV Taq RT-PCR kit (New England BioLabs, Ipswich, MA) according to the manufacturer instructions with hu-G-CSF-specific primers (see below). RT-PCR products were visualized by 1.2% agarose gel electrophoresis (FIG. 5B). Table 6 below shows which modRNAs were run on the agarose gel.
  • Keratinocytes are grown in EpiLife medium with Human Keratinocyte Growth Supplement in the absence of hydrocortisone from Invitrogen at a confluence of >70%. Keratinocytes are reverse transfected with Ong, 46.875ng, 93.75ng, 187.5ng, 375ng, 750ng, 1500ng, 3000ng, or 6000ng of modRNA complexed with RNAiMAX from Invitrogen. The modRNA:RNAiMAX complex is formed. Secreted huG-CSF
  • G-CSF Human Granulocyte-Colony Stimulating Factor
  • the modified mRNA comprised of chemically-distinct modified nucleotides encoding human Granulocyte-Colony Stimulating Factor (G-CSF) may stimulate the cellular proliferation of a transfection incompetent cell in co-culture environment.
  • the co-culture includes a highly transfectable cell type such as a human keratinocyte and a transfection incompetent cell type such as a white blood cell (WBC).
  • WBC white blood cell
  • the modified mRNA encoding G-CSF may be transfected into the highly transfectable cell allowing for the production and secretion of G-CSF protein into the extracellular environment where G-CSF acts in a paracrine-like manner to stimulate the white blood cell expressing the G-CSF receptor to proliferate.
  • the expanded WBC population may be used to treat immune-compromised patients or partially reconstitute the WBC population of an immunosuppressed patient and thus reduce the risk of opportunistic infections.
  • a highly transfectable cell such as a fibroblast may be transfected with certain growth factors to support and simulate the growth, maintenance, or differentiation of poorly transfectable embryonic stem cells or induced pluripotent stem cells.
  • the cloning, gene synthesis and vector sequencing was performed by DNA2.0 Inc. (Menlo Park, CA). Sequence and insert sequence are set forth herein.
  • the ORF was restriction digested using Xbal and used for cDNA synthesis using tailed-or tail-less-PCR.
  • the tailed-PCR cDNA product was used as the template for the modified mRNA synthesis reaction using 25mM mixture each modified nucleotide (all modified nucleotides were custom synthesized or purchased from TriLink Biotech, San Diego, CA except pyrrolo-C triphosphate purchased from Glen Research, Sterling VA; unmodifed nucleotides were purchased from Epicenter
  • modRNAs were post-transcriptionally capped using recombinant Vaccinia Virus Capping Enzyme (New England BioLabs, Ipswich, MA) and a recombinant 2'-0-methyltransferase (Epicenter Biotechnologies, Madison, WI) to generate the 5'-guanosine Capl structure.
  • Cap 2 structure and Cap 3 structures may be generated using additional 2'-0-methyltransferases.
  • the in vitro transcribed mRNA product was run on an agarose gel and visualized. modRNA was purified with Ambion/ Applied Biosystems (Austin, TX) MEGAClear RNATM purification kit. PCR used PureLinkTM PCR purification kit
  • the product was quantified on NanodropTM UV Absorbance (ThermoFisher, Waltham, MA). Quality, UV absorbance quality and visualization of the product was performed on an 1.2% agarose gel. The product was resuspended in TE buffer.
  • 5 '-modRNA capping may be completed concomitantly during the in vzYro-transcription reaction using the following chemical RNA cap analogs to generate the 5'-guanosine cap structure according to manufacturer protocols: 3 ' -0-Me-m 7 G(5')ppp(5')G; G(5')ppp(5')A;
  • 5'- modRNA capping may be completed post-transcriptionally using a Vaccinia Virus Capping Enzyme to generate the "Cap 0" structure: m 7 G(5')ppp(5')G (New England BioLabs, Ipswich, MA).
  • Cap 1 structure may be generated using both Vaccinia Virus Capping Enzyme and a 2'-0 methyl-transferase to generate: m7G(5')ppp(5')G-2'-0-methyl.
  • Cap 2 structure may be generated from the Cap 1 structure followed by the 2'-0-methylation of the 5 '-antepenultimate nucleotide using a 2'-0 methyl-transferase.
  • Cap 3 structure may be generated from the Cap 2 structure followed by the 2'-0-methylation of the 5'-preantepenultimate nucleotide using a 2'-0 methyl-transferase.
  • Enzymes are preferably derived from a recombinant source.
  • G-CSF protein
  • G-CSF modRNA RT-PCR primers

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Genetics & Genomics (AREA)
  • Organic Chemistry (AREA)
  • Molecular Biology (AREA)
  • Biotechnology (AREA)
  • Biomedical Technology (AREA)
  • Biochemistry (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • General Health & Medical Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Engineering & Computer Science (AREA)
  • Microbiology (AREA)
  • Biophysics (AREA)
  • Immunology (AREA)
  • Physics & Mathematics (AREA)
  • Plant Pathology (AREA)
  • Medicinal Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Cell Biology (AREA)
  • Hematology (AREA)
  • Urology & Nephrology (AREA)
  • Crystallography & Structural Chemistry (AREA)
  • Dispersion Chemistry (AREA)
  • Food Science & Technology (AREA)
  • Analytical Chemistry (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Endocrinology (AREA)
  • Public Health (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)

Abstract

The present disclosure provides modified nucleosides, nucleotides, and nucleic acids, and methods of using thereof.

Description

MODIFIED NUCLEOSIDES, NUCLEOTIDES, AND NUCLEIC ACIDS, AND USES
THEREOF
CLAIM OF PRIORITY
This application claims the benefit of U.S. Provisional Application Serial No.
61/404,413, filed on October 1, 2010, which is incorporated herein by reference in its entirety.
BACKGROUND
Naturally occurring RNAs are synthesized from four basic ribonucleotides: ATP, CTP, UTP and GTP, but may contain post-transcriptionally modified nucleotides. Further, approximately one hundred different nucleoside modifications have been identified in RNA (Rozenski, J, Crain, P, and McCloskey, J. (1999). The RNA Modification Database: 1999 update. Nucl Acids Res 27: 196-197). The role of nucleoside modifications on the irnmuno- stimulatory potential, stability, and on the translation efficiency of RNA, and the consequent benefits to this for enhancing protein expression and producing therapeutics however, is unclear.
There are multiple problems with prior methodologies of effecting protein expression. For example, heterologous deoxyribonucleic acid (DNA) introduced into a cell can be inherited by daughter cells (whether or not the heterologous DNA has integrated into the chromosome) or by offspring. Introduced DNA can integrate into host cell genomic DNA at some frequency, resulting in alterations and/or damage to the host cell genomic DNA. In addition, multiple steps must occur before a protein is made. Once inside the cell, DNA must be transported into the nucleus where it is transcribed into RNA. The RNA transcribed from DNA must then enter the cytoplasm where it is translated into protein. This need for multiple processing steps creates lag times before the generation of a protein of interest. Further, it is difficult to obtain DNA expression in cells; frequently DNA enters cells but is not expressed or not expressed at reasonable rates or concentrations. This can be a particular problem when DNA is introduced into cells such as primary cells or modified cell lines.
There is a need in the art for biological modalities to address the modulation of intracellular translation of nucleic acids. SUMMARY
The present disclosure provides, inter alia, modified nucleosides, modified nucleotides, and modified nucleic acids which can exhibit a reduced innate immune response when introduced into a population of cells, both in vivo and ex vivo. Further, these modified nucleosides, modified nucleotides, and modified nucleic acids described herein can disrupt binding of a major groove interacting partner with the nucleic acid. Because of the reduced immunogenicity and the decrease in major groove interactions, these modified nucleosides, modified nucleotides, and modified nucleic acids can be more efficient during protein production than, e.g., unmodified nucleic acids.
Thus, the present disclosure provides compounds comprising nucleotides that can disrupt binding of a major groove binding partner with a nucleic acid, wherein the nucleotide has decreased binding affinity to the major groove binding partner.
The present disclosure further provides compounds having Formula I:
Figure imgf000003_0001
I
wherein constituent variables are provided herein.
The present disclosure further provides nucleic acid sequences of at least two nucleotides comprising a compound of Formula I-d:
Figure imgf000003_0002
I-d
wherein constituent variables are provided herein.
The present disclosure further provides compositions comprising at least one compound of Formula I.
The present disclosure further provides pharmaceutical compositions comprising a compound of Formula I.
The present disclosure further provides methods of preparing nucleic acid sequences of at least two nucleotides of a compound of Formula I-d.
The present disclosure further provides methods of amplifying nucleic acid sequences of at least two nucleotides of a compound of Formula I-d.
The present disclosure further provides kits comprising a compound of Formula I.
Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Methods and materials are described herein for use in the present invention; other, suitable methods and materials known in the art can also be used. The materials, methods, and examples are illustrative only and not intended to be limiting. All publications, patent applications, patents, sequences, database entries, and other references mentioned herein are incorporated by reference in their entirety. In case of conflict, the present specification, including definitions, will control.
Other features and advantages of the invention will be apparent from the following detailed description and figures, and from the claims.
BRIEF DESCRIPTION OF DRAWINGS FIGs. 1A and IB depict images of non-denaturing agarose gels of each in vitro- transcribed modified RNA.
FIGs. 2A and 2B depict images of an Enzyme-linked immunosorbent assay (ELISA) for Human Granulocyte-Colony Stimulating Factor (G-CSF) of in vitro transfected Human
Keratinocyte cells with each indicated modRNA encoding human G-CSF and the line indicates a saturating level of maximum detectable limit of secreted G-CSF in the assay.
FIGs. 3A-N depict line graphs of a series of Enzyme-linked immunosorbent assays (ELISA) for Human Granulocyte-Colony Stimulating Factor (G-CSF) secreted from in vitro- transfected Human Keratinocyte cells at different time points with each indicated human G-CSF- encoding modRNA at the indicated doses. The line indicates a saturating level of maximum detectable limit of secreted G-CSF in the assay.
FIGs. 4A and 4B depict bar graphs of a series of Enzyme-linked immunosorbent assays (ELISA) for endogenous cellular human Tumor Necrosis Factor-a (TNF- a) secreted from in vzYro-transfected Human Keratinocyte cells at 24 hours with each indicated hu-G-CSF-encoding modRNA at increasing doses.
FIGs. 4C and 4D depict bar graphs of a series of Enzyme-linked immunosorbent assays (ELISA) for endogenous cellular human Interferon-β (IFN-β) secreted from in vzYro-transfected Human Keratinocyte cells at 24 hours with each indicated hu-G-CSF-encoding modRNA at increasing doses.
FIGs. 4E and 4F depict bar graphs of a series of Enzyme-linked immunosorbent assays (ELISA) for human-G-CSF secreted from in vzYro-transfected Human Keratinocyte cells at 24 hours with each indicated hu-G-CSF-encoding modRNA at increasing doses.
FIG. 5A is a table showing results from an Enzyme-linked immunosorbent assay
(ELISA) for human-G-CSF secreted from in vzYro-transfected Human Keratinocyte cells sampled from individual wells in a co-culture 24-well tissue culture plate 42 hours post-transfection with 750ng of each indicated hu-G-CSF-encoding modRNA.
FIG. 5B depicts an image of an agarose gel of RT-PCR hu-G-CSF modRNA products from co-culture cell extracts 42 hours post-transfection of the human keratinocyte feeder layer with hu-G-CSG modRNA and the un-transfected Kasumi-1 and KG-1 insert culture cells.
FIGs. 5C and 5D depict graphs of results from a hu-G-CSF-modRNA-induced cell proliferation assay of Kasumi-1 (FIG. 5C) and KG-1 (FIG. 5D) cells normalized to untransfected cells. Hu-G-CSF modRNA identity transfected into human keratinocyte feeder cells is indicated.
FIGs. 6A-L depict graphs of the UV absorbance spectra for exemplary modRNA molecules that incorporate the indicated modified nucleotide.
DETAILED DESCRIPTION
The present disclosure provides, inter alia, modified nucleosides, modified nucleotides, and modified nucleic acids that exhibit a reduced innate immune response when introduced into a population of cells. The modified nucleosides, modified nucleotides, and modified nucleic acids can be chemically modified on the major groove face, thereby disrupting major groove binding partner interactions, which cause innate immune responses.
In general, exogenous unmodified nucleic acids, particularly viral nucleic acids, introduced into cells induce an innate immune response, resulting in cytokine and interferon (IFN) production and cell death. However, it is of great interest for therapeutics, diagnostics, reagents and for biological assays to deliver a nucleic acid, e.g., a ribonucleic acid (RNA) inside a cell, either in vivo or ex vivo, such as to cause intracellular translation of the nucleic acid and production of the encoded protein. Of particular importance is the delivery and function of a non-integrative nucleic acid, as nucleic acids characterized by integration into a target cell are generally imprecise in their expression levels, deleteriously transferable to progeny and neighbor cells, and suffer from the substantial risk of causing mutation. Provided herein in part are nucleic acids encoding useful polypeptides capable of modulating a cell's function and/or activity, and methods of making and using these nucleic acids and polypeptides. As described herein, these nucleic acids are capable of reducing the innate immune activity of a population of cells into which they are introduced, thus increasing the efficiency of protein production in that cell population. Further, one or more additional advantageous activities and/or properties of the nucleic acids and proteins of the present disclosure are described.
Further, the modified nucleosides, modified nucleotides, and modified nucleic acids described herein can be modified on the major groove face. These major groove modifications can allow for alterations, e.g. a decrease, in the interaction of the modified nucleosides, modified nucleotides, and modified nucleic acids with a binding groove partner.
Accordingly, in a first aspect, the present disclosure provides compounds comprising a nucleotide that can disrupts binding of a major groove interacting, e.g. binding, partner with a nucleic acid, wherein the nucleotide has decreased binding affinity to major groove interacting, e.g. binding, partners.
In another aspect, the present disclosure provides compounds comprising a nucleotide that contains chemical modifications, wherein the nucleotide can have altered binding to major groove interacting, e.g. binding, partners.
In some embodiments, the chemical modifications are located on the major groove face of the nucleobase, and wherein the chemical modifications can include replacing or substituting an atom of a pyrimidine nucleobase with an amine, an SH, an alkyl (e.g., methyl or ethyl), or a halo (e.g., chloro or fluoro).
In some embodiments, the chemical modifications can be located on the major groove face of the nucleobase, and wherein the chemical modification can include replacing or substituting an atom of a pyrimidine nucleobase with an amine, an SH, a methyl or ethyl, or a chloro or fluoro.
In some embodiments, the chemical modifications can be located on the sugar moiety of the nucleotide.
In some embodiments, the chemical modifications can be located on the phosphate backbone of the nucleotide.
In some embodiments, the chemical modifications can alter the electrochemistry on the major groove face of the nucleotide.
In another aspect, the present disclosure provides nucleotides that contain chemical modifications, wherein the nucleotide reduces the cellular innate immune response, as compared to the cellular innate immune induced by a corresponding unmodified nucleic acid.
In another aspect, the present disclosure provides nucleic acid sequences comprising at least two nucleotides, the nucleic acid sequence comprising a nucleotide that disrupts binding of a major groove interacting partner with the nucleic acid sequence, wherein the nucleotide has decreased binding affinity to the major groove binding partner.
In another aspect, the present disclosure provides compositions comprising a compound as described herein.
In some embodiments, the composition is a reaction mixture.
In some embodiments, the composition is a pharmaceutical composition.
In some embodiments, the composition is a cell culture.
In some embodiments, the compositions further comprise an RNA polymerase and a cDNA template.
In some embodiments, the compositions further comprise a nucleotide selected from the group consisting of adenosine, cytosine, guanosine, and uracil.
In a further aspect, the present disclosure provides for methods of synthesizing a pharmaceutical nucleic acid, comprising providing a complementary deoxyribonucleic acid (cDNA) that encodes a pharmaceutical protein of interest; selecting a nucleotide that is known to disrupt a binding of a major groove binding partner with a nucleic acid, wherein the nucleotide has decreased binding affinity to the major groove binding partner; and contacting the provided cDNA and the selected nucleotide with an RNA polymerase, under conditions such that the pharmaceutical nucleic acid is synthesized.
In some embodiments, the pharmaceutical nucleic acid is a ribonucleic acid (RNA).
In a further aspect, the present disclosure provides for methods of making a
pharmaceutical formulation comprising a physiologically active secreted protein, comprising transfecting a first population of human cells with a pharmaceutical nucleic acid made by the methods described herein, wherein the secreted protein is active upon a second population of human cells.
In some embodiments, the secreted protein is capable of interacting, e.g. binding, with a receptor on the surface of at least one cell present in the second population.
In some embodiments, the secreted protein is Granulocyte-Colony Stimulating Factor (G-
CSF).
In some embodiments, the second population contains myeloblast cells that express the G-CSF receptor.
In a further aspect, the present disclosure provides for methods of making a
pharmaceutical formulation comprising human cells comprising a physiologically active secreted protein, comprising transfecting a first population of human cells with a pharmaceutical nucleic acid made by the methods described herein, wherein the secreted protein is active upon a second population of human cells.
Definitions
At various places in the present specification, substituents of compounds of the present disclosure are disclosed in groups or in ranges. It is specifically intended that the present disclosure include each and every individual subcombination of the members of such groups and ranges. For example, the term "Ci_6 alkyl" is specifically intended to individually disclose methyl, ethyl, C3 alkyl, C4 alkyl, C5 alkyl, and C6 alkyl.
It is further intended that the compounds of the present disclosure are stable. As used herein "stable" refers to a compound that is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture, and preferably capable of formulation into an efficacious therapeutic agent.
It is further appreciated that certain features of the present disclosure, which are, for clarity, described in the context of separate embodiments, can also be provided in combination in a single embodiment. Conversely, various features of the present disclosure which are, for brevity, described in the context of a single embodiment, can also be provided separately or in any suitable subcombination.
As used herein, the term "alkyl" is meant to refer to a saturated hydrocarbon group which is straight-chained or branched. Example alkyl groups include methyl (Me), ethyl (Et), propyl (e.g., n-propyl and isopropyl), butyl (e.g., n-butyl, isobutyl, t-butyl), pentyl (e.g., n-pentyl, isopentyl, neopentyl), and the like. An alkyl group can contain from 1 to about 20, from 2 to about 20, from 1 to about 12, from 1 to about 8, from 1 to about 6, from 1 to about 4, or from 1 to about 3 carbon atoms.
As used herein, "alkenyl" refers to an alkyl group having one or more double carbon- carbon bonds. Example alkenyl groups include ethenyl, propenyl, and the like.
As used herein, "alkoxy" refers to an -O-alkyl group. Example alkoxy groups include methoxy, ethoxy, propoxy (e.g., n-propoxy and isopropoxy), t-butoxy, and the like.
As used herein, "alkynyl" refers to an alkyl group having one or more triple carbon- carbon bonds. Example alkynyl groups include ethynyl, propynyl, and the like.
As used herein, "aryl" refers to monocyclic or polycyclic (e.g., having 2, 3 or 4 fused rings) aromatic hydrocarbons such as, for example, phenyl, naphthyl, anthracenyl,
phenanthrenyl, indanyl, indenyl, and the like. In some embodiments, aryl groups have from 6 to about 20 carbon atoms.
As used herein, "halo" or "halogen" includes fluoro, chloro, bromo, and iodo.
As used herein, "therapeutic agent" refers to any agent that, when administered to a subject, has a therapeutic, diagnostic, and/or prophylactic effect and/or elicits a desired biological and/or pharmacological effect.
As used herein, "animal" refers to any member of the animal kingdom. In some embodiments, "animal" refers to humans at any stage of development. In some embodiments, "animal" refers to non-human animals at any stage of development. In certain embodiments, the non-human animal is a mammal (e.g. , a rodent, a mouse, a rat, a rabbit, a monkey, a dog, a cat, a sheep, cattle, a primate, or a pig). In some embodiments, animals include, but are not limited to, mammals, birds, reptiles, amphibians, fish, and worms. In some embodiments, the animal is a transgenic animal, genetically-engineered animal, or a clone.
As used herein, "approximately" or "about," as applied to one or more values of interest, refers to a value that is similar to a stated reference value. In certain embodiments, the term "approximately" or "about" refers to a range of values that fall within 25%, 20%, 19%, 18%, 17%, 16%, 15%, 14%, 13%, 12%, 11%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, or less in either direction (greater than or less than) of the stated reference value unless otherwise stated or otherwise evident from the context (except where such number would exceed 100% of a possible value).
As used herein, "associated with," "conjugated," "linked," "attached," and "tethered," when used with respect to two or more moieties, means that the moieties are physically associated or connected with one another, either directly or via one or more additional moieties that serves as a linking agent, to form a structure that is sufficiently stable so that the moieties remain physically associated under the conditions in which the structure is used, e.g., physiological conditions.
As used herein, "biologically active" refers to a characteristic of any substance that has activity in a biological system and/or organism. For instance, a substance that, when
administered to an organism, has a biological effect on that organism, is considered to be biologically active. In particular embodiments, where a nucleic acid is biologically active, a portion of that nucleic acid that shares at least one biological activity of the whole nucleic acid is typically referred to as a "biologically active" portion.
As used herein, "conserved" refers to nucleotides or amino acid residues of a
polynucleotide sequence or amino acid sequence, respectively, that are those that occur unaltered in the same position of two or more related sequences being compared. Nucleotides or amino acids that are relatively conserved are those that are conserved amongst more related sequences than nucleotides or amino acids appearing elsewhere in the sequences. In some embodiments, two or more sequences are said to be "completely conserved" if they are 100% identical to one another. In some embodiments, two or more sequences are said to be "highly conserved" if they are at least 70% identical, at least 80% identical, at least 90% identical, or at least 95% identical to one another. In some embodiments, two or more sequences are said to be "highly conserved" if they are about 70% identical, about 80%> identical, about 90% identical, about 95%, about 98%, or about 99% identical to one another. In some embodiments, two or more sequences are said to be "conserved" if they are at least 30% identical, at least 40% identical, at least 50% identical, at least 60% identical, at least 70% identical, at least 80% identical, at least 90% identical, or at least 95% identical to one another. In some embodiments, two or more sequences are said to be "conserved" if they are about 30% identical, about 40% identical, about 50% identical, about 60%> identical, about 70% identical, about 80%> identical, about 90%> identical, about 95% identical, about 98% identical, or about 99% identical to one another.
As used herein, "expression" of a nucleic acid sequence refers to one or more of the following events: (1) production of an RNA template from a DNA sequence (e.g., by transcription); (2) processing of an RNA transcript (e.g., by splicing, editing, 5' cap formation, and/or 3' end processing); (3) translation of an RNA into a polypeptide or protein; and (4) post- translational modification of a polypeptide or protein.
As used herein, a "functional" biological molecule is a biological molecule in a form in which it exhibits a property and/or activity by which it is characterized.
As used herein, "in vitro'" refers to events that occur in an artificial environment, e.g., in a test tube or reaction vessel, in cell culture, in a Petri dish, etc., rather than within an organism (e.g., animal, plant, or microbe).
As used herein, "in vivo" refers to events that occur within an organism (e.g., animal, plant, or microbe).
As used herein, "isolated" refers to a substance or entity that has been (1) separated from at least some of the components with which it was associated when initially produced (whether in nature or in an experimental setting), and/or (2) produced, prepared, and/or manufactured by the hand of man. Isolated substances and/or entities may be separated from at least about 10%, about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, or more of the other components with which they were initially associated. In some
embodiments, isolated agents are more than about 80%>, about 85%, about 90%, about 91%, about 92%, about 93%, about 94%, about 95%, about 96%, about 97%, about 98%, about 99%, or more than about 99% pure. As used herein, a substance is "pure" if it is substantially free of other components. As used herein, "subject" or "patient" refers to any organism to which a composition in accordance with the present disclosure may be administered, e.g. , for experimental, diagnostic, prophylactic, and/or therapeutic purposes. Typical subjects include animals (e.g., mammals such as mice, rats, rabbits, non-human primates, and humans) and/or plants.
As used herein, "substantially" refers to the qualitative condition of exhibiting total or near-total extent or degree of a characteristic or property of interest. One of ordinary skill in the biological arts will understand that biological and chemical phenomena rarely, if ever, go to completion and/or proceed to completeness or achieve or avoid an absolute result. The term "substantially" is therefore used herein to capture the potential lack of completeness inherent in many biological and chemical phenomena.
An individual who is "suffering from" a disease, disorder, and/or condition has been diagnosed with or displays one or more symptoms of a disease, disorder, and/or condition.
An individual who is "susceptible to" a disease, disorder, and/or condition has not been diagnosed with and/or may not exhibit symptoms of the disease, disorder, and/or condition. In some embodiments, an individual who is susceptible to a disease, disorder, and/or condition (for example, cancer) may be characterized by one or more of the following: (1) a genetic mutation associated with development of the disease, disorder, and/or condition; (2) a genetic
polymorphism associated with development of the disease, disorder, and/or condition; (3) increased and/or decreased expression and/or activity of a protein and/or nucleic acid associated with the disease, disorder, and/or condition; (4) habits and/or lifestyles associated with development of the disease, disorder, and/or condition; (5) a family history of the disease, disorder, and/or condition; and (6) exposure to and/or infection with a microbe associated with development of the disease, disorder, and/or condition. In some embodiments, an individual who is susceptible to a disease, disorder, and/or condition will develop the disease, disorder, and/or condition. In some embodiments, an individual who is susceptible to a disease, disorder, and/or condition will not develop the disease, disorder, and/or condition.
As used herein, "therapeutically effective amount" means an amount of an agent to be delivered (e.g., nucleic acid, drug, therapeutic agent, diagnostic agent, prophylactic agent, etc.) that is sufficient, when administered to a subject suffering from or susceptible to a disease, disorder, and/or condition, to treat, improve symptoms of, diagnose, prevent, and/or delay the onset of the disease, disorder, and/or condition. As used herein, "transcription factor" refers to a DNA-binding protein that regulates transcription of DNA into RNA, for example, by activation or repression of transcription. Some transcription factors effect regulation of transcription alone, while others act in concert with other proteins. Some transcription factor can both activate and repress transcription under certain conditions. In general, transcription factors bind a specific target sequence or sequences highly similar to a specific consensus sequence in a regulatory region of a target gene. Transcription factors may regulate transcription of a target gene alone or in a complex with other molecules.
As used herein, "treating" refers to partially or completely alleviating, ameliorating, improving, relieving, delaying onset of, inhibiting progression of, reducing severity of, and/or reducing incidence of one or more symptoms or features of a particular disease, disorder, and/or condition. For example, "treating" cancer may refer to inhibiting survival, growth, and/or spread of a tumor. Treatment may be administered to a subject who does not exhibit signs of a disease, disorder, and/or condition and/or to a subject who exhibits only early signs of a disease, disorder, and/or condition for the purpose of decreasing the risk of developing pathology associated with the disease, disorder, and/or condition. In some embodiments, treatment comprises delivery of a protein associated with a therapeutically active nucleic acid to a subject in need thereof.
As used herein, "unmodified" refers to a nucleic acid prior to being modified, e.g.
adenosine, guanosine, cytosine, thymidine, and uracil, or a naturally occurring amino acid. The compounds described herein can be asymmetric (e.g., having one or more stereocenters). All stereoisomers, such as enantiomers and diastereomers, are intended unless otherwise indicated. Compounds of the present disclosure that contain asymmetrically substituted carbon atoms can be isolated in optically active or racemic forms. Methods on how to prepare optically active forms from optically active starting materials are known in the art, such as by resolution of racemic mixtures or by stereoselective synthesis. Many geometric isomers of olefins, C=N double bonds, and the like can also be present in the compounds described herein, and all such stable isomers are contemplated in the present disclosure. Cis and trans geometric isomers of the compounds of the present disclosure are described and may be isolated as a mixture of isomers or as separated isomeric forms.
Compounds of the present disclosure also include tautomeric forms. Tautomeric forms result from the swapping of a single bond with an adjacent double bond together with the concomitant migration of a proton. Tautomeric forms include prototropic tautomers which are isomeric protonation states having the same empirical formula and total charge. Example prototropic tautomers include ketone - enol pairs, amide - imidic acid pairs, lactam - lactim pairs, amide - imidic acid pairs, enamine - imine pairs, and annular forms where a proton can occupy two or more positions of a heterocyclic system, for example, IH- and 3H-imidazole, 1H-, 2H- and 4H- 1,2,4-triazole, IH- and 2H- isoindole, and IH- and 2H-pyrazole. Tautomeric forms can be in equilibrium or sterically locked into one form by appropriate substitution.
Compounds of the present disclosure can also include all isotopes of atoms occurring in the intermediates or final compounds. Isotopes include those atoms having the same atomic number but different mass numbers. For example, isotopes of hydrogen include tritium and deuterium.
The term "compound," as used herein, is meant to include all stereoisomers, geometric isomers, tautomers, and isotopes of the structures depicted.
In some embodiments, the compounds of the present disclosure are substantially isolated. By "substantially isolated" is meant that the compound is at least partially or substantially separated from the environment in which it was formed or detected. Partial separation can include, for example, a composition enriched in the compound of the present disclosure.
Substantial separation can include compositions containing at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 95%, at least about 97%), or at least about 99% by weight of the compound of the present disclosure, or salt thereof. Methods for isolating compounds and their salts are routine in the art.
The compounds of the present disclosure, and salts thereof, can also be prepared in combination with solvent or water molecules to form solvates and hydrates by routine methods.
The present disclosure also includes pharmaceutically acceptable salts of the compounds described herein. As used herein, "pharmaceutically acceptable salts" refers to derivatives of the disclosed compounds wherein the parent compound is modified by converting an existing acid or base moiety to its salt form. Examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like. The pharmaceutically acceptable salts of the present disclosure include the conventional non-toxic salts of the parent compound formed, for example, from non-toxic inorganic or organic acids. The pharmaceutically acceptable salts of the present disclosure can be synthesized from the parent compound which contains a basic or acidic moiety by conventional chemical methods. Generally, such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two; generally, nonaqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile are preferred. Lists of suitable salts are found in Remington 's Pharmaceutical Sciences, 17th ed., Mack Publishing Company, Easton, Pa., 1985, p. 1418 and Journal of Pharmaceutical Science, 66, 2 (1977), each of which is incorporated herein by reference in its entirety.
The phrase "pharmaceutically acceptable" is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
The present disclosure also includes prodrugs of the compounds described herein. As used herein, "prodrugs" refer to any carriers, typically covalently bonded, which release the active parent drug when administered to a mammalian subject. Prodrugs can be prepared by modifying functional groups present in the compounds in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to the parent compounds. Prodrugs include compounds wherein hydroxyl, amino, sulfhydryl, or carboxyl groups are bonded to any group that, when administered to a mammalian subject, cleaves to form a free hydroxyl, amino, sulfhydryl, or carboxyl group respectively. Examples of prodrugs include, but are not limited to, acetate, formate and benzoate derivatives of alcohol and amine functional groups in the compounds of the present disclosure. Preparation and use of prodrugs is discussed in T. Higuchi and V. Stella, "Pro-drugs as Novel Delivery Systems," Vol. 14 of the A.C.S. Symposium Series, and in Bioreversible Carriers in Drug Design, ed. Edward B. Roche, American Pharmaceutical Association and Pergamon Press, 1987, both of which are hereby incorporated by reference in their entirety.
Modified Nucleosides and Nucleotides
The present disclosure provides for modified nucleosides and nucleotides. As described herein "nucleoside" is defined as a compound containing a five-carbon sugar molecule (a pentose or ribose) or derivative thereof, and an organic base, purine or pyrimidine, or a derivative thereof. As described herein, "nucleotide" is defined as a nucleoside consisting of a phosphate group. The nucleosides and nucleotides described herein are generally chemically modified on the major groove face. In some embodiments, the major groove chemical modifications can include an amino group, a thiol group, an alkyl group, or a halo group.
Table 1 below identifies the chemical faces of each canonical nucleotide. Circles identify the atoms comprising the respective chemical regions.
Pyrimidines
Purines
Figure imgf000016_0001
In some embodiments, modified nucleosides include pyridin-4-one ribonucleoside, 5-aza- uridine, 2-thio-5-aza-uridine, 2-thiouridine, 4-thio-pseudouridine, 2-thio-pseudouridine, 5- hydroxyuridine, 3-methyluridine, 5-carboxymethyl-uridine, 1-carboxymethyl-pseudouridine, 5- propynyl-uridine, 1-propynyl-pseudouridine, 5-taurinomethyluridine, 1-taurinomethyl- pseudouridine, 5-taurinomethyl-2-thio-uridine, 1 -taurinomethyl-4-thio-uridine, 5-methyl-uridine, 1-methyl-pseudouridine, 4-thio-l-methyl-pseudouridine, 2-thio-l-methyl-pseudouridine, 1- methyl- 1 -deaza-pseudouridine, 2-thio- 1 -methyl- 1 -deaza-pseudouridine, dihydrouridine, dihydropseudouridine, 2-thio-dihydrouridine, 2-thio-dihydropseudouridine, 2-methoxyuridine, 2- methoxy-4-thio-uridine, 4-methoxy-pseudouridine, and 4-methoxy-2-thio-pseudouridine.
In some embodiments, modified nucleosides include 5-aza-cytidine, pseudoisocytidme, 3-methyl-cytidine, N4-acetylcytidine, 5-formylcytidine, N4-methylcytidine, 5- hydroxymethylcytidine, 1-methyl-pseudoisocytidine, pyrrolo-cytidine, pyrrolo- pseudoisocytidine, 2-thio-cytidine, 2-thio-5-methyl-cytidine, 4-thio-pseudoisocytidine, 4-thio-l- methyl-pseudoisocytidine, 4-thio- 1 -methyl- 1 -deaza-pseudoisocytidine, 1 -methyl- 1 -deaza- pseudoisocytidine, zebularine, 5-aza-zebularine, 5-methyl-zebularine, 5-aza-2-thio-zebularine, 2- thio-zebularine, 2-methoxy-cytidine, 2-methoxy-5-methyl-cytidine, 4-methoxy- pseudoisocytidine, and 4-methoxy- 1-methyl-pseudoisocytidine.
In other embodiments, modified nucleosides include 2-aminopurine, 2, 6-diaminopurine, 7-deaza-adenine, 7-deaza-8-aza-adenine, 7-deaza-2-aminopurine, 7-deaza-8-aza-2-aminopurine, 7-deaza-2, 6-diaminopurine, 7-deaza-8-aza-2, 6-diaminopurine, 1-methyladenosine, N6- methyladenosine, N6-isopentenyladenosine, N6-(cis-hydroxyisopentenyl)adenosine, 2- methylthio-N6-(cis-hydroxyisopentenyl) adenosine, N6-glycinylcarbamoyladenosine, N6- threonylcarbamoyladenosine, 2-methylthio-N6-threonyl carbamoyladenosine, N6,N6- dimethyladenosine, 7-methyladenine, 2-methylthio-adenine, and 2-methoxy-adenine.
In some embodiments, modified nucleosides include inosine, 1-methyl-inosine, wyosine, wybutosine, 7-deaza-guanosine, 7-deaza-8-aza-guanosine, 6-thio-guanosine, 6-thio-7-deaza- guanosine, 6-thio-7-deaza-8-aza-guanosine, 7-methyl-guanosine, 6-thio-7-methyl-guanosine, 7- methylinosine, 6-methoxy-guanosine, 1-methylguanosine, N2-methylguanosine, N2,N2- dimethylguanosine, 8-oxo-guanosine, 7-methyl-8-oxo-guanosine, l-methyl-6-thio-guanosine, N2-methyl-6-thio-guanosine, and N2,N2-dimethyl-6-thio-guanosine.
In some embodiments, the nucleotide can be modified on the major groove face and can include replacement of the hydrogen on C-5 of uracil with a methyl group or a halo group.
In some embodiments, the nucleoside and nucleotide can be a compound of Formula I:
Figure imgf000018_0001
I
wherein:
Z is O or S;
each of Y1 is independently selected from -ORal, -NRalRbl, and -SRal;
each of Y2 is independently selected from O, NRa, S or a linker comprising an atom selected from the group consisting of C, O, N, and S;
each of Y3 is independently selected from O and S;
Y4 is selected from H, -ORa, -SRa, and -NHRa;
n is 0, 1 , 2, or 3;
m is 0, 1 , 2 or 3;
B is a nucleobase;
Ra is H, Ci-20 alkyl, C2-2o alkenyl, C2-2o alkynyl, or C6-2o aryl;
Ral and Rbl are each independently H or a counterion; and
-Y3-Rcl is OH or SH at a pH of about 1 or -Y3-Rcl is O" or S" at physiological pH; or -Y3-Rcl is Ci-20 alkoxy, C2-2o -O-alkenyl, or Ci-2o -O-alkynyl;
wherein when B is an unmodified nucleobase selected from cytosine, guanine, uracil and adenine, then at least one of Z, Y1 or Y2 is not O or OH.
In some embodiments, B is a nucleobase of Formula Il-a, Il-b, or II-c:
Figure imgf000018_0002
Il-a Il-b II-c
wherein: denotes a single or double bond;
X is O or S;
U and W are each independently C or N;
V is O, S, C or N;
wherein when V is C then R1 is H, C1-6 alkyl, C1-6 alkenyl, C1-6 alkynyl, halo, or -ORc, wherein Ci-20 alkyl, C2-20 alkenyl, C2-20 alkynyl are each optionally substituted with -OH, -NRaRb, -SH, - C(0)Rc, -C(0)ORc, -NHC(0)Rc, or -NHC(0)ORc;
and wherein when V is O, S, or N then R1 is absent;
R2 is H, -ORc, -SRC, -NRaRb, or halo;
or when V is C then R1 and R2 together with the carbon atoms to which they are attached can form a 5- or 6-membered ring optionally substituted with 1-4 substituents selected from halo, - OH, -SH, -NRaRb, Ci-20 alkyl, C2-2o alkenyl, C2-2o alkynyl, C1-20 alkoxy, or C1-20 thioalkyl;
R3 is H or Ci-20 alkyl;
R4 is H or C 1-20 alkyl; wherein when ^ denotes a double bond then R4 is absent, or N- R4, taken together, forms a positively charged N substituted with C1-20 alkyl;
Ra and Rb are each independently H, C1-20 alkyl, C2-20 alkenyl, C2-20 alkynyl, or C6-20 aryl; and
Rc is H, Ci-20 alkyl, C2-2o alkenyl, phenyl, benzyl, a polyethylene glycol group, or an amino-polyethylene glycol group.
In some embodiments, B is a nucleobase of Formula Il-al , II-a2, II-a3, II-a4, or II-a5:
Il-al II-a2 II-a3 II-a4 II-a5..
In some embodiments, B is a nucleobase selected from the group consisting of cytosine, guanine, adenine, and uracil.
In some embodiments, B is a pyrimidine or derivative thereof.
In some embodiments the nucleotide is a compound of Formula I-a:
Figure imgf000020_0001
I-a.
In some embodiments the nucleotide is a compound of Formula I-b:
Figure imgf000020_0002
I-b.
In some embodiments the nucleotide is a compound of Formula I-c:
Figure imgf000020_0003
I-c.
In some embodiments, the nucleotide is selected from the group consisting of:
Figure imgf000021_0001
Figure imgf000021_0002
20 For example, the modified nucleotide can be:
Figure imgf000022_0001
In some embodiments, the major groove chemical modification can include replacement of the C-H group at C-5 with an -NH- group or a -NH(CH3)- group.
For example, the modified nucleotide can be:
Figure imgf000022_0002
In another embodiment, the major groove chemical modification can include replacement of the hydrogen at C-5 of cytosine with a halo group or a methyl group.
For example, the modified nucleotide can be:
Figure imgf000022_0003
In yet a further embodiment, the major groove chemical modification can include a fused ring that is formed by the NH2 at the C-4 position and the carbon atom at the C-5 position.
For example, the modified nucleotide can be:
Figure imgf000023_0001
In some embodiments, a modified nucleotide is 5 '-0-(l -Thiophosphate)- Adenosine, 5'- 0-(l-Thiophosphate)-Cytidine, 5'-0-(l-Thiophosphate)-Guanosine, 5'-0-(l-Thiophosphate)- Uridine or 5'-0-(l-Thiophosphate)-Pseudouridine.
Figure imgf000023_0002
5 '-0-( 1 -Thiophosphate)- Adenosine
Figure imgf000023_0003
5 '-0-( 1 -Thiophosphate)-Cytidine
Figure imgf000023_0004
5'-0-( 1 -Thiophosphate )-Guanosine
Figure imgf000024_0001
5'-0-(l-Thiophosphate)-Uridine
Figure imgf000024_0002
5'-0-(l-Thiophosphate)-Pseudouridine
The a-thio substituted phosphate moiety is provided to confer stability to RNA and DNA polymers through the unnatural phosphorothioate backbone linkages. Phosphorothioate DNA and RNA have increased nuclease resistance and subsequently a longer half-life in a cellular environment. Phosphorothioate linked nucleic acids are expected to also reduce the innate immune response through weaker binding/activation of cellular innate immune molecules.
Further examples of modified nucleotides and modified nucleotide combinations are provided below in Table 2.
Table 2
Figure imgf000024_0003
6-aza-uridine Pyrrolo-cytidine
5-hydroxy-uridine Pyrrolo-cytidine/5-iodo-uridine
Deoxy-thymidine Pyrrolo-cytidine/Nl-methyl-pseudo-uridine
Pseudo-uridine Pyrrolo-cytidine/a-thio-uridine
Inosine Pyrrolo-cytidine/5-methyl-uridine
a-thio-guanosine Pyrrolo-cytidine/Pseudo-uridine
8-oxo-guanosine 5-methyl-cytidine/5-iodo-uridine
06-methyl-guanosine 5-methyl-cytidine/Nl-methyl-pseudo-uridine
7-deaza-guanosine 5-methyl-cytidine/a-thio-uridine
No modification 5-methyl-cytidine/5-methyl-uridine
Nl-methyl-adenosine 5-methyl-cytidine/Pseudo-uridine
2-amino-6-Chloro- 5-methyl-cytidine
purine
N6-methyl-2-amino- 25% Pseudo-iso-cytidine
purine
6-Chloro-purine 25% Nl-methyl-pseudo-uridine
N6-methyl-adenosine 25% Nl-Methyl-pseudo-uridine/75%-pseudo-uridine a-thio-adenosine 5-methyl-uridine
8-azido-adenosine 5-iodo-cytidine
7-deaza-adenosine
Synthesis of Modified Nucleotides
The modified nucleosides and nucleotides disclosed herein can be prepared from readily available starting materials using the following general methods and procedures. It is understood that where typical or preferred process conditions (i.e., reaction temperatures, times, mole ratios of reactants, solvents, pressures, etc.) are given; other process conditions can also be used unless otherwise stated. Optimum reaction conditions may vary with the particular reactants or solvent used, but such conditions can be determined by one skilled in the art by routine optimization procedures.
The processes described herein can be monitored according to any suitable method known in the art. For example, product formation can be monitored by spectroscopic means, such as nuclear magnetic resonance spectroscopy (e.g., 1H or 13C) infrared spectroscopy, spectrophotometry (e.g., UV-visible), or mass spectrometry, or by chromatography such as high performance liquid chromatography (HPLC) or thin layer chromatography. Preparation of modified nucleosides and nucleotides can involve the protection and deprotection of various chemical groups. The need for protection and deprotection, and the selection of appropriate protecting groups can be readily determined by one skilled in the art. The chemistry of protecting groups can be found, for example, in Greene, et al., Protective Groups in Organic Synthesis, 2d. Ed., Wiley & Sons, 1991, which is incorporated herein by reference in its entirety.
The reactions of the processes described herein can be carried out in suitable solvents, which can be readily selected by one of skill in the art of organic synthesis. Suitable solvents can be substantially nonreactive with the starting materials (reactants), the intermediates, or products at the temperatures at which the reactions are carried out, i.e., temperatures which can range from the solvent's freezing temperature to the solvent's boiling temperature. A given reaction can be carried out in one solvent or a mixture of more than one solvent. Depending on the particular reaction step, suitable solvents for a particular reaction step can be selected.
Resolution of racemic mixtures of modified nucleosides and nucleotides can be carried out by any of numerous methods known in the art. An example method includes fractional
recrystallization using a "chiral resolving acid" which is an optically active, salt-forming organic acid. Suitable resolving agents for fractional recrystallization methods are, for example, optically active acids, such as the D and L forms of tartaric acid, diacetyltartaric acid, dibenzoyltartaric acid, mandelic acid, malic acid, lactic acid or the various optically active camphorsulfonic acids. Resolution of racemic mixtures can also be carried out by elution on a column packed with an optically active resolving agent {e.g., dinitrobenzoylphenylglycine). Suitable elution solvent composition can be determined by one skilled in the art.
Exemplary syntheses of modified nucleotides are provided below in Schemes 1 and 2.
Figure imgf000027_0001
Scheme 2
Figure imgf000028_0001
1) POCl3
2) Pyrophosphate
Figure imgf000028_0002
Modified nucleosides and nucleotides can also be prepared according to the synthetic methods described in Ogata et al. Journal of Organic Chemistry 74:2585-2588, 2009; Purmal et al. Nucleic Acids Research 22(1): 72-78, 1994; Fukuhara et al. Biochemistry 1(4): 563-568, 1962; and Xu et al. Tetrahedron 48(9): 1729-1740, 1992, each of which are incorporated by reference in their entirety.
Modified Nucleic Acids
The present disclosure provides nucleic acids, including RNAs such as mRNAs that contain one or more modified nucleosides (termed "modified nucleic acids") or nucleotides as described herein, which have useful properties including the significant decreast or lack of a substantial induction of the innate immune response of a cell into which the mRNA is introduced, or the suppression thereof. Because these modified nucleic acids enhance the efficiency of protein production, intracellular retention of nucleic acids, and viability of contacted cells, as well as possess reduced immunogenicity, of these nucleic acids compared to unmodified nucleic acids, having these properties are termed "enhanced nucleic acids" herein.
In addition, the present disclosure provides nucleic acids, which have decreased binding affinity to a major groove interacting, e.g. binding, partner. For example, the nucleic acids are comprised of at least one nucleotide that has been chemically modified on the major groove face as described herein.
The term "nucleic acid," in its broadest sense, includes any compound and/or substance that is or can be incorporated into an oligonucleotide chain. Exemplary nucleic acids for use in accordance with the present disclosure include, but are not limited to, one or more of DNA, RNA including messenger mRNA (mRNA), hybrids thereof, RNAi-inducing agents, RNAi agents, siRNAs, shRNAs, miRNAs, antisense RNAs, ribozymes, catalytic DNA, RNAs that induce triple helix formation, aptamers, vectors, etc., described in detail herein.
Provided are modified nucleic acids containing a translatable region and one, two, or more than two different nucleoside modifications. In some embodiments, the modified nucleic acid exhibits reduced degradation in a cell into which the nucleic acid is introduced, relative to a corresponding unmodified nucleic acid. Exemplary nucleic acids include ribonucleic acids (RNAs), deoxyribonucleic acids (DNAs), threose nucleic acids (TNAs), glycol nucleic acids (GNAs), locked nucleic acids (LNAs) or a hybrid thereof. In preferred embodiments, the modified nucleic acid includes messenger RNAs (mRNAs). As described herein, the nucleic acids of the present disclosure do not substantially induce an innate immune response of a cell into which the mRNA is introduced.
In certain embodiments, it is desirable to intracellularly degrade a modified nucleic acid introduced into the cell, for example if precise timing of protein production is desired. Thus, the present disclosure provides a modified nucleic acid containing a degradation domain, which is capable of being acted on in a directed manner within a cell.
Other components of nucleic acid are optional, and are beneficial in some embodiments. For example, a 5' untranslated region (UTR) and/or a 3'UTR are provided, wherein either or both may independently contain one or more different nucleoside modifications. In such
embodiments, nucleoside modifications may also be present in the translatable region. Also provided are nucleic acids containing a Kozak sequence. Additionally, provided are nucleic acids containing one or more intronic nucleotide sequences capable of being excised from the nucleic acid.
Further, provided are nucleic acids containing an internal ribosome entry site (IRES). An IRES may act as the sole ribosome binding site, or may serve as one of multiple ribosome binding sites of an mRNA. An mRNA containing more than one functional ribosome binding site may encode several peptides or polypeptides that are translated independently by the ribosomes ("multicistronic mRNA"). When nucleic acids are provided with an IRES, further optionally provided is a second translatable region. Examples of IRES sequences that can be used according to the present disclosure include without limitation, those from picomaviruses (e.g. FMDV), pest viruses (CFFV), polio viruses (PV), encephalomyocarditis viruses (ECMV), foot-and-mouth disease viruses (FMDV), hepatitis C viruses (HCV), classical swine fever viruses (CSFV), murine leukemia virus (MLV), simian immune deficiency viruses (SIV) or cricket paralysis viruses (CrPV).
In some embodiments, the nucleic acid sequences comprise a compound of Formula I-d:
Figure imgf000030_0001
I-d
wherein:
Z is O or S;
each of Y1 is independently selected from -ORal, -NRalRbl, and -SRal;
each of Y2 is independently selected from O, NRa, S or a linker comprising
selected from the group consisting of C, O, N, and S;
each of Y3 is independently selected from O and S;
Y4 is selected from H, -ORa, -SRa, and -NHRa;
B is a nucleobase;
Ra is H, C1-20 alkyl, C2-2o alkenyl, C2-20 alkynyl, or C6-20 aryl; Ral and Rbl are each independently H or a counterion; and
-Y3-Rcl is OH or SH at a pH of about 1 or -Y3-Rcl is O" or S" at physiological pH;
or -Y3-Rcl is Ci-20 alkoxy, C2-2o -O-alkenyl, or Ci-2o -O-alkynyl;
wherein when B is an unmodified nucleobase selected from cytosine, guanine, thymidine, uracil and adenine, then at least one of Z, Y1 or Y2 is not O or OH.
In some embodiments, B is a nucleobase of Formula Il-a, Il-b, or II-c:
Figure imgf000031_0001
II-a Il-b II-c
wherein:
^ denotes a single or double bond;
X is O or S;
U and W are each independently C or N;
V is O, S, C or N;
wherein when V is C then R1 is H, Ci-6 alkyl, Ci-6 alkenyl, Ci-6 alkynyl, halo, or -ORc, wherein Ci-20 alkyl, C2-2o alkenyl, C2-2o alkynyl are each optionally substituted with -OH, -NRaRb, -SH, - C(0)Rc, -C(0)ORc, -NHC(0)Rc, or -NHC(0)ORc;
and wherein when V is O, S, or N then R1 is absent;
R2 is H, -ORc, -SRC, -NRaRb, or halo;
or when V is C then R1 and R2 together with the carbon atoms to which they are attached can form a 5- or 6-membered ring optionally substituted with 1-4 substituents selected from halo, - OH, -SH, -NRaRb, Ci-20 alkyl, C2-2o alkenyl, C2-2o alkynyl, Ci-2o alkoxy, or Ci-2o thioalkyl;
R3 is H or Ci-20 alkyl;
R4 is H or C 1-20 alkyl; wherein when ^ denotes a double bond then R4 is absent, or N- R4, taken together, forms a positively charged N substituted with Ci-2o alkyl;
Ra and Rb are each independently H, Ci-2o alkyl, C2-2o alkenyl, C2-2o alkynyl, or C6-2o aryl; and
Rc is H, Ci-20 alkyl, C2-2o alkenyl, phenyl, benzyl, a polyethylene glycol group, or an amino-polyethylene glycol group. In some embodiments, B is a nucleobase of Formula Il-al, II-a2, II-a3, II-a4, or II-a5:
Figure imgf000032_0001
Il-al II-a2 II-a3 II-a4 II-a5.
In some embodiments, at least 25% of the cytosines are replaced by a compound of Formula I-a (e.g., at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, or about 100%).
In some embodiments, at least 25% of the uracils are replaced by a compound of Formula I-a (e.g., at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, or about 100%).
In some embodiments, at least 25% of the cytosines and 25% of the uracils are replaced by a compound of Formula I-a (e.g., at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, or about 100%).
In some embodiments, the nucleic acid is translatable.
Major Groove Interacting Partners
As described herein, the phrase "major groove interacting partner" refers RNA recognition receptors that detect and respond to RNA ligands through interactions, e.g. binding, with the major groove face of a nucleotide or nucleic acid. As such, RNA ligands comprising modified nucleotides or nucleic acids as described herein decrease interactions with major groove binding partners, and therefore decrease an innate immune response, or expression and secretion of pro-inflammatory cytokines, or both. Example major groove interacting, e.g. binding, partners include, but are not limited to the following nucleases and helicases. Within membranes, TLRs (Toll-like Receptors) 3, 7, and 8 can respond to single- and double-stranded RNAs. Within the cytoplasm, members of the superfamily 2 class of DE (D/H) helicases and ATPases can sense RNAs to initiate antiviral responses. These helicases include the RIG-I (retinoic acid-inducible gene I) and MDA5 (melanoma differentiation-associated gene 5). Other examples include laboratory of genetics and physiology 2 (LGP2), ΗΓΝ-200 domain containing proteins, or Helicase-domain containing proteins.
Prevention or reduction of innate cellular immune response activation using modified nucleic acids
The term "innate immune response" includes a cellular response to exogenous nucleic acids, including single stranded nucleic acids, generally of viral or bacterial origin, which involves the induction of cytokine expression and release, particularly the interferons, and cell death. Protein synthesis is also reduced during the innate cellular immune response. While it is advantageous to eliminate the innate immune response in a cell, the present disclosure provides modified mRNAs that substantially reduce the immune response, including interferon signaling, without entirely eliminating such a response. In some embodiments, the immune response is reduced by 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99%, 99.9%, or greater than 99.9% as compared to the immune response induced by a corresponding unmodified nucleic acid. Such a reduction can be measured by expression or activity level of Type 1 interferons or the expression of interferon-regulated genes such as the toll-like receptors (e.g., TLR7 and TLR8). Reduction of innate immune response can also be measured by decreased cell death following one or more administrations of modified RNAs to a cell population; e.g., cell death is 10%, 25%, 50%, 75%, 85%, 90%, 95%, or over 95% less than the cell death frequency observed with a corresponding unmodified nucleic acid. Moreover, cell death may affect fewer than 50%, 40%, 30%, 20%, 10%, 5%, 1%, 0.1%, 0.01% or fewer than 0.01% of cells contacted with the modified nucleic acids.
The present disclosure provides for the repeated introduction (e.g., transfection) of modified nucleic acids into a target cell population, e.g., in vitro, ex vivo, or in vivo. The step of contacting the cell population may be repeated one or more times (such as two, three, four, five or more than five times). In some embodiments, the step of contacting the cell population with the modified nucleic acids is repeated a number of times sufficient such that a predetermined efficiency of protein translation in the cell population is achieved. Given the reduced cytotoxicity of the target cell population provided by the nucleic acid modifications, such repeated transfections are achievable in a diverse array of cell types.
Polypeptide variants
Provided are nucleic acids that encode variant polypeptides, which have a certain identity with a reference polypeptide sequence. The term "identity" as known in the art, refers to a relationship between the sequences of two or more peptides, as determined by comparing the sequences. In the art, "identity" also means the degree of sequence relatedness between peptides, as determined by the number of matches between strings of two or more amino acid residues. "Identity" measures the percent of identical matches between the smaller of two or more sequences with gap alignments (if any) addressed by a particular mathematical model or computer program (i.e., "algorithms"). Identity of related peptides can be readily calculated by known methods. Such methods include, but are not limited to, those described in Computational Molecular Biology, Lesk, A. M., ed., Oxford University Press, New York, 1988; Biocomputing: Informatics and Genome Projects, Smith, D. W., ed., Academic Press, New York, 1993;
Computer Analysis of Sequence Data, Part 1 , Griffin, A. M., and Griffin, H. G., eds., Humana Press, New Jersey, 1994; Sequence Analysis in Molecular Biology, von Heinje, G., Academic Press, 1987; Sequence Analysis Primer, Gribskov, M. and Devereux, J., eds., M. Stockton Press, New York, 1991 ; and Carillo et al, SIAM J. Applied Math. 48, 1073 (1988).
In some embodiments, the polypeptide variant has the same or a similar activity as the reference polypeptide. Alternatively, the variant has an altered activity (e.g., increased or decreased) relative to a reference polypeptide. Generally, variants of a particular polynucleotide or polypeptide of the present disclosure will have at least about 40%, 45%, 50%>, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity to that particular reference polynucleotide or polypeptide as determined by sequence alignment programs and parameters described herein and known to those skilled in the art. As recognized by those skilled in the art, protein fragments, functional protein domains, and homologous proteins are also considered to be within the scope of this present disclosure. For example, provided herein is any protein fragment of a reference protein (meaning a polypeptide sequence at least one amino acid residue shorter than a reference polypeptide sequence but otherwise identical) 10, 20, 30, 40, 50, 60, 70, 80, 90, 100 or greater than 100 amino acids in length In another example, any protein that includes a stretch of about 20, about 30, about 40, about 50, or about 100 amino acids which are about 40%, about 50%, about 60%>, about 70%), about 80%>, about 90%>, about 95%, or about 100%. identical to any of the sequences described herein can be utilized in accordance with the present disclosure. In certain
embodiments, a protein sequence to be utilized in accordance with the present disclosure includes 2, 3, 4, 5, 6, 7, 8, 9, 10, or more mutations as shown in any of the sequences provided or referenced herein.
Polypeptide libraries
Also provided are polynucleotide libraries containing nucleoside modifications, wherein the polynucleotides individually contain a first nucleic acid sequence encoding a polypeptide, such as an antibody, protein binding partner, scaffold protein, and other polypeptides known in the art. Preferably, the polynucleotides are mRNA in a form suitable for direct introduction into a target cell host, which in turn synthesizes the encoded polypeptide.
In certain embodiments, multiple variants of a protein, each with different amino acid modification(s), are produced and tested to determine the best variant in terms of
pharmacokinetics, stability, biocompatibility, and/or biological activity, or a biophysical property
2 3 4 5 6 7 8 9 such as expression level. Such a library may contain 10, 10 , 10 , 10 , 10 , 10 , 10 , 10 , 10 , or over 109 possible variants (including substitutions, deletions of one or more residues, and insertion of one or more residues).
Polypeptide-nucleic acid complexes
Proper protein translation involves the physical aggregation of a number of polypeptides and nucleic acids associated with the mRNA. Provided by the present disclosure are protein- nucleic acid complexes, containing a translatable mRNA having one or more nucleoside modifications (e.g., at least two different nucleoside modifications) and one or more polypeptides bound to the mRNA. Generally, the proteins are provided in an amount effective to prevent or reduce an innate immune response of a cell into which the complex is introduced.
Untranslatable Modified Nucleic Acids
As described herein, provided are mRNAs having sequences that are substantially not translatable. Such mRNA is effective as a vaccine when administered to a mammalian subject.
Also provided are modified nucleic acids that contain one or more noncoding regions. Such modified nucleic acids are generally not translated, but are capable of binding to and sequestering one or more translational machinery component such as a ribosomal protein or a transfer RNA (tRNA), thereby effectively reducing protein expression in the cell. The modified nucleic acid may contain a small nucleolar RNA (sno-RNA), micro RNA (miRNA), small interfering RNA (siRNA) or Piwi-interacting RNA (piRNA).
Synthesis of Modified Nucleic Acids
Nucleic acids for use in accordance with the present disclosure may be prepared according to any available technique including, but not limited to chemical synthesis, enzymatic synthesis, which is generally termed in vitro transcription, enzymatic or chemical cleavage of a longer precursor, etc. Methods of synthesizing RNAs are known in the art (see, e.g., Gait, M.J. (ed.) Oligonucleotide synthesis: a practical approach, Oxford [Oxfordshire], Washington, DC: IRL Press, 1984; and Herdewijn, P. (ed.) Oligonucleotide synthesis: methods and applications, Methods in Molecular Biology, v. 288 (Clifton, N.J.) Totowa, N.J.: Humana Press, 2005; both of which are incorporated herein by reference).
The modified nucleosides and nucleotides disclosed herein can be prepared from readily available starting materials using the following general methods and procedures. It is understood that where typical or preferred process conditions {i.e., reaction temperatures, times, mole ratios of reactants, solvents, pressures, etc.) are given; other process conditions can also be used unless otherwise stated. Optimum reaction conditions may vary with the particular reactants or solvent used, but such conditions can be determined by one skilled in the art by routine optimization procedures.
The processes described herein can be monitored according to any suitable method known in the art. For example, product formation can be monitored by spectroscopic means, such as nuclear magnetic resonance spectroscopy (e.g., 1H or 13C) infrared spectroscopy, spectrophotometry (e.g., UV-visible), or mass spectrometry, or by chromatography such as high performance liquid chromatography (HPLC) or thin layer chromatography.
Preparation of modified nucleosides and nucleotides can involve the protection and deprotection of various chemical groups. The need for protection and deprotection, and the selection of appropriate protecting groups can be readily determined by one skilled in the art. The chemistry of protecting groups can be found, for example, in Greene, et al., Protective Groups in Organic Synthesis, 2d. Ed., Wiley & Sons, 1991, which is incorporated herein by reference in its entirety.
The reactions of the processes described herein can be carried out in suitable solvents, which can be readily selected by one of skill in the art of organic synthesis. Suitable solvents can be substantially nonreactive with the starting materials (reactants), the intermediates, or products at the temperatures at which the reactions are carried out, i.e., temperatures which can range from the solvent's freezing temperature to the solvent's boiling temperature. A given reaction can be carried out in one solvent or a mixture of more than one solvent. Depending on the particular reaction step, suitable solvents for a particular reaction step can be selected.
Resolution of racemic mixtures of modified nucleosides and nucleotides can be carried out by any of numerous methods known in the art. An example method includes fractional recrystallization using a "chiral resolving acid" which is an optically active, salt-forming organic acid. Suitable resolving agents for fractional recrystallization methods are, for example, optically active acids, such as the D and L forms of tartaric acid, diacetyltartaric acid, dibenzoyltartaric acid, mandelic acid, malic acid, lactic acid or the various optically active camphorsulfonic acids. Resolution of racemic mixtures can also be carried out by elution on a column packed with an optically active resolving agent (e.g., dinitrobenzoylphenylglycine). Suitable elution solvent composition can be determined by one skilled in the art.Modified nucleic acids need not be uniformly modified along the entire length of the molecule. Different nucleotide modifications and/or backbone structures may exist at various positions in the nucleic acid. One of ordinary skill in the art will appreciate that the nucleotide analogs or other modification(s) may be located at any position(s) of a nucleic acid such that the function of the nucleic acid is not substantially decreased. A modification may also be a 5 Or 3' terminal modification. The nucleic acids may contain at a minimum one and at maximum 100% modified nucleotides, or any intervening percentage, such as at least 5% modified nucleotides, at least 10% modified nucleotides, at least 25% modified nucleotides, at least 50%> modified nucleotides, at least 80%> modified nucleotides, or at least 90% modified nucleotides. For example, the nucleic acids may contain a modified pyrimidine such as uracil or cytosine. In some embodiments, at least 5%, at least 10%, at least 25%, at least 50%, at least 80%, at least 90% or 100% of the uracil in the nucleic acid is replaced with a modified uracil. The modified uracil can be replaced by a compound having a single unique structure, or can be replaced by a plurality of compounds having different structures (e.g., 2, 3, 4 or more unique structures). In some embodiments, at least 5%, at least 10%, at least 25%, at least 50%, at least 80%, at least 90% or 100% of the cytosine in the nucleic acid is replaced with a modified cytosine. The modified cytosine can be replaced by a compound having a single unique structure, or can be replaced by a plurality of compounds having different structures (e.g., 2, 3, 4 or more unique structures).
Generally, the shortest length of a modified mRNA of the present disclosure can be the length of an mRNA sequence that is sufficient to encode for a dipeptide. In another
embodiment, the length of the mRNA sequence is sufficient to encode for a tripeptide. In another embodiment, the length of an mRNA sequence is sufficient to encode for a tetrapeptide. In another embodiment, the length of an mRNA sequence is sufficient to encode for a pentapeptide. In another embodiment, the length of an mRNA sequence is sufficient to encode for a hexapeptide. In another embodiment, the length of an mRNA sequence is sufficient to encode for a heptapeptide. In another embodiment, the length of an mRNA sequence is sufficient to encode for an octapeptide. In another embodiment, the length of an mRNA sequence is sufficient to encode for a nonapeptide. In another embodiment, the length of an mRNA sequence is sufficient to encode for a decapeptide.
Examples of dipeptides that the modified nucleic acid sequences can encode for include, but are not limited to, carnosine and anserine.
In a further embodiment, the mRNA is greater than 30 nucleotides in length. In another embodiment, the RNA molecule is greater than 35 nucleotides in length. In another embodiment, the length is at least 40 nucleotides. In another embodiment, the length is at least 45 nucleotides. In another embodiment, the length is at least 55 nucleotides. In another embodiment, the length is at least 60 nucleotides. In another embodiment, the length is at least 60 nucleotides. In another embodiment, the length is at least 80 nucleotides. In another embodiment, the length is at least 90 nucleotides. In another embodiment, the length is at least 100 nucleotides. In another embodiment, the length is at least 120 nucleotides. In another embodiment, the length is at least 140 nucleotides. In another embodiment, the length is at least 160 nucleotides. In another embodiment, the length is at least 180 nucleotides. In another embodiment, the length is at least 200 nucleotides. In another embodiment, the length is at least 250 nucleotides. In another embodiment, the length is at least 300 nucleotides. In another embodiment, the length is at least 350 nucleotides. In another embodiment, the length is at least 400 nucleotides. In another embodiment, the length is at least 450 nucleotides. In another embodiment, the length is at least 500 nucleotides. In another embodiment, the length is at least 600 nucleotides. In another embodiment, the length is at least 700 nucleotides. In another embodiment, the length is at least 800 nucleotides. In another embodiment, the length is at least 900 nucleotides. In another embodiment, the length is at least 1000 nucleotides. In another embodiment, the length is at least 1100 nucleotides. In another embodiment, the length is at least 1200 nucleotides. In another embodiment, the length is at least 1300 nucleotides. In another embodiment, the length is at least 1400 nucleotides. In another embodiment, the length is at least 1500 nucleotides. In another embodiment, the length is at least 1600 nucleotides. In another embodiment, the length is at least 1800 nucleotides. In another embodiment, the length is at least 2000 nucleotides. In another embodiment, the length is at least 2500 nucleotides. In another embodiment, the length is at least 3000 nucleotides. In another embodiment, the length is at least 4000 nucleotides. In another embodiment, the length is at least 5000 nucleotides, or greater than 5000 nucleotides.
The present disclosure provides methods of preparing a nucleic acid sequence comprising a nucleotide that disrupts binding of a major groove interacting partner with the nucleic acid sequence, wherein the nucleic acid sequence comprises a compound of Formula I-d:
Figure imgf000039_0001
wherein:
Z is O or S;
each of Y1 is independently selected from -ORal, -NRalRbl, and -SRal;
each of Y2 is independently selected from O, NRa, S or a linker comprising an atom selected from the group consisting of C, O, N, and S;
each of Y3 is independently selected from O and S;
Y4 is selected from H, -ORa, -SRa, and -NHRa;
B is a nucleobase;
Ra is H, Ci-20 alkyl, C2-2o alkenyl, C2-2o alkynyl, or C6-2o aryl;
Ral and Rbl are each independently H or a counterion; and
-Y3-Rcl is OH or SH at a pH of about 1 or -Y3-Rcl is O" or S" at physiological pH; or -Y3-Rcl is Ci-20 alkoxy, C2-2o -O-alkenyl, or Ci-2o -O-alkynyl;
wherein when B is an unmodified nucleobase selected from cytosine, guanine, uracil and adenine, then at least one of Z, Y1 or Y2 is not O or OH;
the method comprising:
reacting a compound of Formula I-c:
Figure imgf000040_0001
I-c
with an RNA polymerase, and a cDNA template.
In some embodiments, the reaction is repeated from 1 to about 7,000 times.
In some embodiments, B is a nucleobase of Formula Il-a, Il-b, or II-c:
Figure imgf000040_0002
Il-a Il-b II-c
wherein:
denotes a single or double bond; X is O or S;
U and W are each independently C or N;
V is O, S, C or N;
wherein when V is C then R1 is H, C1-6 alkyl, C1-6 alkenyl, C1-6 alkynyl, halo, or -ORc, wherein Ci-20 alkyl, C2-2o alkenyl, C2-2o alkynyl are each optionally substituted with -OH, -NRaRb, -SH, - C(0)Rc, -C(0)ORc, -NHC(0)Rc, or -NHC(0)ORc;
and wherein when V is O, S, or N then R1 is absent;
R2 is H, -ORc, -SRC, -NRaRb, or halo;
or when V is C then R1 and R2 together with the carbon atoms to which they are attached can form a 5- or 6-membered ring optionally substituted with 1-4 substituents selected from halo, - OH, -SH, -NRaRb, Ci-20 alkyl, C2-2o alkenyl, C2-2o alkynyl, C1-20 alkoxy, or C1-20 thioalkyl;
R3 is H or Ci-20 alkyl;
R4 is H or C 1-20 alkyl; wherein when ^ denotes a double bond then R4 is absent, or N- R4, taken together, forms a positively charged N substituted with C1-20 alkyl;
Ra and Rb are each independently H, C1-20 alkyl, C2-2o alkenyl, C2-2o alkynyl, or C6-2o aryl; and
Rc is H, Ci-20 alkyl, C2-2o alkenyl, phenyl, benzyl, a polyethylene glycol group, or an amino-polyethylene glycol group.
In some embodiments, B is a nucleobase of Formula Il-al , II-a2, II-a3, II-a4, or II-a5:
Figure imgf000041_0001
Il-al II-a2 II-a3 II-a4 II-a5.
In some embodiments, the methods further comprise a nucleotide selected from the group consisting of adenosine, cytosine, guanosine, and uracil.
In some embodiments, the nucleobase is a pyrimidine or derivative thereof.
In a further aspect, the present disclosure provides methods of amplifying a nucleic acid sequence comprising a nucleotide that disrupts binding of a major groove binding partner with the nucleic acid sequence, the method comprising:
reacting a compound of Formula I-c:
Figure imgf000042_0001
I-c
Z is O or S;
each of Y1 is independently selected from -ORal, -NRalRbl, and -SRal;
each of Y2 is independently selected from O, NRa, S or a linker comprising an atom selected from the group consisting of C, O, N, and S;
each of Y3 is independently selected from O and S;
Y4 is selected from H, -ORa, -SRa, and -NHRa;
B is a nucleobase;
Ra is H, Ci-20 alkyl, C2-2o alkenyl, C2-2o alkynyl, or C6-2o aryl;
Ral and Rbl are each independently H or a counterion; and
-Y3-Rcl is OH or SH at a pH of about 1 or -Y3-Rcl is O" or S" at physiological pH; or -Y3-Rcl is Ci-20 alkoxy, C2-2o -O-alkenyl, or Ci-2o -O-alkynyl;
wherein when B is an unmodified nucleobase selected from cytosine, guanine, uracil and adenine, then at least one of Z, Y1 or Y2 is not O or OH;
with a primer, a cDNA template, and an RNA polymerase.
In some embodiments, B is a nucleobase of Formula Il-a, Il-b, or II-c:
Figure imgf000042_0002
Il-a Il-b II-c
wherein:
^ denotes a single or double bond;
X is O or S;
U and W are each independently C or N;
V is O, S, C or N; wherein when V is C then R1 is H, C1-6 alkyl, C1-6 alkenyl, C1-6 alkynyl, halo, or -ORc, wherein Ci-20 alkyl, C2-2o alkenyl, C2-2o alkynyl are each optionally substituted with -OH, -NRaRb, -SH, - C(0)Rc, -C(0)ORc, -NHC(0)Rc, or -NHC(0)ORc;
and wherein when V is O, S, or N then R1 is absent;
R2 is H, -ORc, -SRC, -NRaRb, or halo;
or when V is C then R1 and R2 together with the carbon atoms to which they are attached can form a 5- or 6-membered ring optionally substituted with 1-4 substituents selected from halo, - OH, -SH, -NRaRb, Ci-20 alkyl, C2-2o alkenyl, C2-2o alkynyl, C1-20 alkoxy, or C1-20 thioalkyl;
R3 is H or Ci-20 alkyl;
R4 is H or C 1-20 alkyl; wherein when ^ denotes a double bond then R4 is absent, or N- R4, taken together, forms a positively charged N substituted with C1-20 alkyl;
Ra and Rb are each independently H, C1-20 alkyl, C2-2o alkenyl, C2-2o alkynyl, or C6-2o aryl; and
Rc is H, Ci-20 alkyl, C2-2o alkenyl, phenyl, benzyl, a polyethylene glycol group, or an amino-polyethylene glycol group.
In some embodiments, B is a nucleobase of Formula Il-al, II-a2, II-a3, II-a4, or II-a5:
Figure imgf000043_0001
Il-al II-a2 II-a3 II-a4 II-a5.
In some embodiments, the methods further comprise a nucleotide selected from the group consisting of adenosine, cytosine, guanosine, and uracil.
In some embodiments, the nucleobase is a pyrimidine or derivative thereof.
Uses of Modified Nucleic Acids
Therapeutic Agents
The modified nucleic acids and the proteins translated from the modified nucleic acids described herein can be used as therapeutic agents. For example, a modified nucleic acid described herein can be administered to a subject, wherein the modified nucleic acid is translated in vivo to produce a therapeutic peptide in the subject. Accordingly, provided herein are compositions, methods, kits, and reagents for treatment or prevention of disease or conditions in humans and other mammals. The active therapeutic agents of the present disclosure include modified nucleic acids, cells containing modified nucleic acids or polypeptides translated from the modified nucleic acids, polypeptides translated from modified nucleic acids, and cells contacted with cells containing modified nucleic acids or polypeptides translated from the modified nucleic acids.
In certain embodiments, provided are combination therapeutics containing one or more modified nucleic acids containing translatable regions that encode for a protein or proteins that boost a mammalian subject's immunity along with a protein that induces antibody-dependent cellular toxitity. For example, provided are therapeutics containing one or more nucleic acids that encode trastuzumab and granulocyte-colony stimulating factor (G-CSF). In particular, such combination therapeutics are useful in Her2+ breast cancer patients who develop induced resistance to trastuzumab. (See, e.g., Albrecht, Immunotherapy. 2(6):795-8 (2010)).
Provided are methods of inducing translation of a recombinant polypeptide in a cell population using the modified nucleic acids described herein. Such translation can be in vivo, ex vivo, in culture, or in vitro. The cell population is contacted with an effective amount of a composition containing a nucleic acid that has at least one nucleoside modification, and a translatable region encoding the recombinant polypeptide. The population is contacted under conditions such that the nucleic acid is localized into one or more cells of the cell population and the recombinant polypeptide is translated in the cell from the nucleic acid.
An effective amount of the composition is provided based, at least in part, on the target tissue, target cell type, means of administration, physical characteristics of the nucleic acid {e.g., size, and extent of modified nucleosides), and other determinants. In general, an effective amount of the composition provides efficient protein production in the cell, preferably more efficient than a composition containing a corresponding unmodified nucleic acid. Increased efficiency may be demonstrated by increased cell transfection (i.e., the percentage of cells transfected with the nucleic acid), increased protein translation from the nucleic acid, decreased nucleic acid degradation (as demonstrated, e.g. , by increased duration of protein translation from a modified nucleic acid), or reduced innate immune response of the host cell. Aspects of the present disclosure are directed to methods of inducing in vivo translation of a recombinant polypeptide in a mammalian subject in need thereof. Therein, an effective amount of a composition containing a nucleic acid that has at least one nucleoside modification and a translatable region encoding the recombinant polypeptide is administered to the subject using the delivery methods described herein. The nucleic acid is provided in an amount and under other conditions such that the nucleic acid is localized into a cell of the subject and the recombinant polypeptide is translated in the cell from the nucleic acid. The cell in which the nucleic acid is localized, or the tissue in which the cell is present, may be targeted with one or more than one rounds of nucleic acid administration.
Other aspects of the present disclosure relate to transplantation of cells containing modified nucleic acids to a mammalian subject. Administration of cells to mammalian subjects is known to those of ordinary skill in the art, such as local implantation {e.g., topical or subcutaneous administration), organ delivery or systemic injection {e.g., intravenous injection or inhalation), as is the formulation of cells in pharmaceutically acceptable carrier. Compositions containing modified nucleic acids are formulated for administration intramuscularly,
transarterially, intraperitoneally, intravenously, intranasally, subcutaneously, endoscopically, transdermally, or intrathecally. In some embodiments, the composition is formulated for extended release.
The subject to whom the therapeutic agent is administered suffers from or is at risk of developing a disease, disorder, or deleterious condition. Provided are methods of identifying, diagnosing, and classifying subjects on these bases, which may include clinical diagnosis, biomarker levels, genome-wide association studies (GWAS), and other methods known in the art.
In certain embodiments, the administered modified nucleic acid directs production of one or more recombinant polypeptides that provide a functional activity which is substantially absent in the cell in which the recombinant polypeptide is translated. For example, the missing functional activity may be enzymatic, structural, or gene regulatory in nature.
In other embodiments, the administered modified nucleic acid directs production of one or more recombinant polypeptides that replace a polypeptide (or multiple polypeptides) that is substantially absent in the cell in which the recombinant polypeptide is translated. Such absence may be due to genetic mutation of the encoding gene or regulatory pathway thereof. Alternatively, the recombinant polypeptide functions to antagonize the activity of an endogenous protein present in, on the surface of, or secreted from the cell. Usually, the activity of the endogenous protein is deleterious to the subject, for example, do to mutation of the endogenous protein resulting in altered activity or localization. Additionally, the recombinant polypeptide antagonizes, directly or indirectly, the activity of a biological moiety present in, on the surface of, or secreted from the cell. Examples of antagonized biological moieties include lipids (e.g., cholesterol), a lipoprotein (e.g., low density lipoprotein), a nucleic acid, a carbohydrate, or a small molecule toxin.
The recombinant proteins described herein are engineered for localization within the cell, potentially within a specific compartment such as the nucleus, or are engineered for secretion from the cell or translocation to the plasma membrane of the cell.
As described herein, a useful feature of the modified nucleic acids of the present disclosure is the capacity to reduce the innate immune response of a cell to an exogenous nucleic acid. Provided are methods for performing the titration, reduction or elimination of the immune response in a cell or a population of cells. In some embodiments, the cell is contacted with a first composition that contains a first dose of a first exogenous nucleic acid including a translatable region and at least one nucleoside modification, and the level of the innate immune response of the cell to the first exogenous nucleic acid is determined. Subsequently, the cell is contacted with a second composition, which includes a second dose of the first exogenous nucleic acid, the second dose containing a lesser amount of the first exogenous nucleic acid as compared to the first dose. Alternatively, the cell is contacted with a first dose of a second exogenous nucleic acid. The second exogenous nucleic acid may contain one or more modified nucleosides, which may be the same or different from the first exogenous nucleic acid or, alternatively, the second exogenous nucleic acid may not contain modified nucleosides. The steps of contacting the cell with the first composition and/or the second composition may be repeated one or more times. Additionally, efficiency of protein production (e.g., protein translation) in the cell is optionally determined, and the cell may be re-transfected with the first and/or second composition repeatedly until a target protein production efficiency is achieved.
Therapeutics for diseases and conditions Provided are methods for treating or preventing a symptom of diseases characterized by missing or aberrant protein activity, by replacing the missing protein activity or overcoming the aberrant protein activity. Because of the rapid initiation of protein production following introduction of modified mRNAs, as compared to viral DNA vectors, the compounds of the present disclosure are particularly advantageous in treating acute diseases such as sepsis, stroke, and myocardial infarction. Moreover, the lack of transcriptional regulation of the modified mRNAs of the present disclosure is advantageous in that accurate titration of protein production is achievable.
Diseases characterized by dysfunctional or aberrant protein activity include, but not limited to, cancer and proliferative diseases, genetic diseases (e.g., cystic fibrosis), autoimmune diseases, diabetes, neurodegenerative diseases, cardiovascular diseases, and metabolic diseases. The present disclosure provides a method for treating such conditions or diseases in a subject by introducing nucleic acid or cell-based therapeutics containing the modified nucleic acids provided herein, wherein the modified nucleic acids encode for a protein that antagonizes or otherwise overcomes the aberrant protein activity present in the cell of the subject. Specific examples of a dysfunctional protein are the missense mutation variants of the cystic fibrosis transmembrane conductance regulator (CFTR) gene, which produce a dysfunctional protein variant of CFTR protein, which causes cystic fibrosis.
Multiple diseases are characterized by missing (or substantially diminished such that proper protein function does not occur) protein activity. Such proteins may not be present, or are essentially non-functional. The present disclosure provides a method for treating such conditions or diseases in a subject by introducing nucleic acid or cell-based therapeutics containing the modified nucleic acids provided herein, wherein the modified nucleic acids encode for a protein that replaces the protein activity missing from the target cells of the subject. Specific examples of a dysfunctional protein are the nonsense mutation variants of the cystic fibrosis
transmembrane conductance regulator (CFTR) gene, which produce a nonfunctional protein variant of CFTR protein, which causes cystic fibrosis.
Thus, provided are methods of treating cystic fibrosis in a mammalian subject by contacting a cell of the subject with a modified nucleic acid having a translatable region that encodes a functional CFTR polypeptide, under conditions such that an effective amount of the CTFR polypeptide is present in the cell. Preferred target cells are epithelial cells, such as the lung, and methods of administration are determined in view of the target tissue; i.e., for lung delivery, the RNA molecules are formulated for administration by inhalation.
In another embodiment, the present disclosure provides a method for treating
hyperlipidemia in a subject, by introducing into a cell population of the subject with a modified mRNA molecule encoding Sortilin, a protein recently characterized by genomic studies, thereby ameliorating the hyperlipidemia in a subject. The SORTl gene encodes a trans-Golgi network (TGN) transmembrane protein called Sortilin. Genetic studies have shown that one of five individuals has a single nucleotide polymorphism, rsl2740374, in the lpl3 locus of the SORTl gene that predisposes them to having low levels of low-density lipoprotein (LDL) and very-low- density lipoprotein (VLDL). Each copy of the minor allele, present in about 30% of people, alters LDL cholesterol by 8 mg/dL, while two copies of the minor allele, present in about 5% of the population, lowers LDL cholesterol 16 mg/dL. Carriers of the minor allele have also been shown to have a 40% decreased risk of myocardial infarction. Functional in vivo studies in mice describes that overexpression of SORTl in mouse liver tissue led to significantly lower LDL- cholesterol levels, as much as 80% lower, and that silencing SORTl increased LDL cholesterol approximately 200% (Musunuru K et al. From noncoding variant to phenotype via SORTl at the lpl3 cholesterol locus. Nature 2010; 466: 714-721).
Methods of cellular nucleic acid delivery
Methods of the present disclosure enhance nucleic acid delivery into a cell population, in vivo, ex vivo, or in culture. For example, a cell culture containing a plurality of host cells {e.g., eukaryotic cells such as yeast or mammalian cells) is contacted with a composition that contains an enhanced nucleic acid having at least one nucleoside modification and, optionally, a translatable region. The composition also generally contains a transfection reagent or other compound that increases the efficiency of enhanced nucleic acid uptake into the host cells. The enhanced nucleic acid exhibits enhanced retention in the cell population, relative to a
corresponding unmodified nucleic acid. The retention of the enhanced nucleic acid is greater than the retention of the unmodified nucleic acid. In some embodiments, it is at least about 50%, 75%, 90%, 95%, 100%, 150%, 200% or more than 200% greater than the retention of the unmodified nucleic acid. Such retention advantage may be achieved by one round of transfection with the enhanced nucleic acid, or may be obtained following repeated rounds of transfection.
In some embodiments, the enhanced nucleic acid is delivered to a target cell population with one or more additional nucleic acids. Such delivery may be at the same time, or the enhanced nucleic acid is delivered prior to delivery of the one or more additional nucleic acids. The additional one or more nucleic acids may be modified nucleic acids or unmodified nucleic acids. It is understood that the initial presence of the enhanced nucleic acids does not substantially induce an innate immune response of the cell population and, moreover, that the innate immune response will not be activated by the later presence of the unmodified nucleic acids. In this regard, the enhanced nucleic acid may not itself contain a translatable region, if the protein desired to be present in the target cell population is translated from the unmodified nucleic acids.
Targeting Moieties
In some embodiments, modified nucleic acids are provided to express a protein-binding partner or a receptor on the surface of the cell, which functions to target the cell to a specific tissue space or to interact with a specific moiety, either in vivo or in vitro. Suitable protein- binding partners include antibodies and functional fragments thereof, scaffold proteins, or peptides. Additionally, modified nucleic acids can be employed to direct the synthesis and extracellular localization of lipids, carbohydrates, or other biological moieties.
Permanent Gene Expression Silencing
A method for epigenetically silencing gene expression in a mammalian subject, comprising a nucleic acid where the translatable region encodes a polypeptide or polypeptides capable of directing sequence-specific histone H3 methylation to initiate heterochromatin formation and reduce gene transcription around specific genes for the purpose of silencing the gene. For example, a gain-of-function mutation in the Janus Kinase 2 gene is responsible for the family of Myeloproliferative Diseases. Pharmaceutical Compositions
The present disclosure provides proteins generated from modified mRNAs.
Pharmaceutical compositions may optionally comprise one or more additional therapeutically active substances. In accordance with some embodiments, a method of administering pharmaceutical compositions comprising one or more proteins to be delivered to a subject in need thereof is provided. In some embodiments, compositions are administered to humans. For the purposes of the present disclosure, the phrase "active ingredient" generally refers to a protein or protein-containing complex as described herein.
Although the descriptions of pharmaceutical compositions provided herein are principally directed to pharmaceutical compositions which are suitable for administration to humans, it will be understood by the skilled artisan that such compositions are generally suitable for
administration to animals of all sorts. Modification of pharmaceutical compositions suitable for administration to humans in order to render the compositions suitable for administration to various animals is well understood, and the ordinarily skilled veterinary pharmacologist can design and/or perform such modification with merely ordinary, if any, experimentation. Subjects to which administration of the pharmaceutical compositions is contemplated include, but are not limited to, humans and/or other primates; mammals, including commercially relevant mammals such as cattle, pigs, horses, sheep, cats, dogs, mice, and/or rats; and/or birds, including commercially relevant birds such as chickens, ducks, geese, and/or turkeys.
Formulations of the pharmaceutical compositions described herein may be prepared by any method known or hereafter developed in the art of pharmacology. In general, such preparatory methods include the step of bringing the active ingredient into association with an excipient and/or one or more other accessory ingredients, and then, if necessary and/or desirable, shaping and/or packaging the product into a desired single- or multi-dose unit.
A pharmaceutical composition in accordance with the present disclosure may be prepared, packaged, and/or sold in bulk, as a single unit dose, and/or as a plurality of single unit doses. As used herein, a "unit dose" is discrete amount of the pharmaceutical composition comprising a predetermined amount of the active ingredient. The amount of the active ingredient is generally equal to the dosage of the active ingredient which would be administered to a subject and/or a convenient fraction of such a dosage such as, for example, one-half or one- third of such a dosage. Relative amounts of the active ingredient, the pharmaceutically acceptable excipient, and/or any additional ingredients in a pharmaceutical composition in accordance with the present disclosure will vary, depending upon the identity, size, and/or condition of the subject treated and further depending upon the route by which the composition is to be administered. By way of example, the composition may comprise between 0.1% and 100% (w/w) active ingredient.
Pharmaceutical formulations may additionally comprise a pharmaceutically acceptable excipient, which, as used herein, includes any and all solvents, dispersion media, diluents, or other liquid vehicles, dispersion or suspension aids, surface active agents, isotonic agents, thickening or emulsifying agents, preservatives, solid binders, lubricants and the like, as suited to the particular dosage form desired. Remington's The Science and Practice of Pharmacy, 21st Edition, A. R. Gennaro (Lippincott, Williams & Wilkins, Baltimore, MD, 2006; incorporated herein by reference) discloses various excipients used in formulating pharmaceutical
compositions and known techniques for the preparation thereof. Except insofar as any conventional excipient medium is incompatible with a substance or its derivatives, such as by producing any undesirable biological effect or otherwise interacting in a deleterious manner with any other component(s) of the pharmaceutical composition, its use is contemplated to be within the scope of this present disclosure.
In some embodiments, a pharmaceutically acceptable excipient is at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% pure. In some embodiments, an excipient is approved for use in humans and for veterinary use. In some embodiments, an excipient is approved by United States Food and Drug Administration. In some embodiments, an excipient is pharmaceutical grade. In some embodiments, an excipient meets the standards of the United States Pharmacopoeia (USP), the European Pharmacopoeia (EP), the British Pharmacopoeia, and/or the International Pharmacopoeia.
Pharmaceutically acceptable excipients used in the manufacture of pharmaceutical compositions include, but are not limited to, inert diluents, dispersing and/or granulating agents, surface active agents and/or emulsifiers, disintegrating agents, binding agents, preservatives, buffering agents, lubricating agents, and/or oils. Such excipients may optionally be included in pharmaceutical formulations. Excipients such as cocoa butter and suppository waxes, coloring agents, coating agents, sweetening, flavoring, and/or perfuming agents can be present in the composition, according to the judgment of the formulator. Exemplary diluents include, but are not limited to, calcium carbonate, sodium carbonate, calcium phosphate, dicalcium phosphate, calcium sulfate, calcium hydrogen phosphate, sodium phosphate lactose, sucrose, cellulose, microcrystalline cellulose, kaolin, mannitol, sorbitol, inositol, sodium chloride, dry starch, cornstarch, powdered sugar, etc., and/or combinations thereof.
Exemplary granulating and/or dispersing agents include, but are not limited to, potato starch, corn starch, tapioca starch, sodium starch glycolate, clays, alginic acid, guar gum, citrus pulp, agar, bentonite, cellulose and wood products, natural sponge, cation-exchange resins, calcium carbonate, silicates, sodium carbonate, cross-linked poly(vinyl-pyrrolidone)
(crospovidone), sodium carboxymethyl starch (sodium starch glycolate), carboxymethyl cellulose, cross-linked sodium carboxymethyl cellulose (croscarmellose), methylcellulose, pregelatinized starch (starch 1500), microcrystalline starch, water insoluble starch, calcium carboxymethyl cellulose, magnesium aluminum silicate (Veegum), sodium lauryl sulfate, quaternary ammonium compounds, etc., and/or combinations thereof.
Exemplary surface active agents and/or emulsifiers include, but are not limited to, natural emulsifiers {e.g. acacia, agar, alginic acid, sodium alginate, tragacanth, chondrux, cholesterol, xanthan, pectin, gelatin, egg yolk, casein, wool fat, cholesterol, wax, and lecithin), colloidal clays (e.g. bentonite [aluminum silicate] and Veegum® [magnesium aluminum silicate]), long chain amino acid derivatives, high molecular weight alcohols (e.g. stearyl alcohol, cetyl alcohol, oleyl alcohol, triacetin monostearate, ethylene glycol distearate, glyceryl monostearate, and propylene glycol monostearate, polyvinyl alcohol), carbomers (e.g. carboxy polymethylene, polyacrylic acid, acrylic acid polymer, and carboxyvinyl polymer), carrageenan, cellulosic derivatives (e.g. carboxymethylcellulose sodium, powdered cellulose, hydroxymethyl cellulose, hydroxypropyl cellulose, hydroxypropyl methylcellulose, methylcellulose), sorbitan fatty acid esters (e.g.
polyoxyethylene sorbitan monolaurate [Tween®20], polyoxyethylene sorbitan [T ween® 60], polyoxyethylene sorbitan monooleate [Tween®80], sorbitan monopalmitate [Span®40], sorbitan monostearate [Span®60], sorbitan tristearate [Span®65], glyceryl monooleate, sorbitan monooleate [Span®80]), polyoxyethylene esters (e.g. polyoxyethylene monostearate [Myrj®45], polyoxyethylene hydrogenated castor oil, polyethoxylated castor oil, polyoxymethylene stearate, and Solutol®), sucrose fatty acid esters, polyethylene glycol fatty acid esters (e.g. Cremophor®), polyoxyethylene ethers, (e.g. polyoxyethylene lauryl ether [Brij®30]), poly(vinyl-pyrrolidone), diethylene glycol monolaurate, triethanolamine oleate, sodium oleate, potassium oleate, ethyl oleate, oleic acid, ethyl laurate, sodium lauryl sulfate, Pluronic®F 68, Poloxamer®188, cetrimonium bromide, cetylpyridinium chloride, benzalkonium chloride, docusate sodium, etc. and/or combinations thereof.
Exemplary binding agents include, but are not limited to, starch {e.g. cornstarch and starch paste); gelatin; sugars {e.g. sucrose, glucose, dextrose, dextrin, molasses, lactose, lactitol, mannitol,); natural and synthetic gums {e.g. acacia, sodium alginate, extract of Irish moss, panwar gum, ghatti gum, mucilage of isapol husks, carboxymethylcellulose, methylcellulose, ethylcellulose, hydroxyethylcellulose, hydroxypropyl cellulose, hydroxypropyl methylcellulose, microcrystalline cellulose, cellulose acetate, poly(vinyl-pyrrolidone), magnesium aluminum silicate (Veegum®), and larch arabogalactan); alginates; polyethylene oxide; polyethylene glycol; inorganic calcium salts; silicic acid; polymethacrylates; waxes; water; alcohol; etc.; and combinations thereof.
Exemplary preservatives may include, but are not limited to, antioxidants, chelating agents, antimicrobial preservatives, antifungal preservatives, alcohol preservatives, acidic preservatives, and/or other preservatives. Exemplary antioxidants include, but are not limited to, alpha tocopherol, ascorbic acid, acorbyl palmitate, butylated hydroxyanisole, butylated hydroxytoluene, monothioglycerol, potassium metabisulfite, propionic acid, propyl gallate, sodium ascorbate, sodium bisulfite, sodium metabisulfite, and/or sodium sulfite. Exemplary chelating agents include ethylenediaminetetraacetic acid (EDTA), citric acid monohydrate, disodium edetate, dipotassium edetate, edetic acid, fumaric acid, malic acid, phosphoric acid, sodium edetate, tartaric acid, and/or trisodium edetate. Exemplary antimicrobial preservatives include, but are not limited to, benzalkonium chloride, benzethonium chloride, benzyl alcohol, bronopol, cetrimide, cetylpyridinium chloride, chlorhexidine, chlorobutanol, chlorocresol, chloroxylenol, cresol, ethyl alcohol, glycerin, hexetidine, imidurea, phenol, phenoxyethanol, phenylethyl alcohol, phenylmercuric nitrate, propylene glycol, and/or thimerosal. Exemplary antifungal preservatives include, but are not limited to, butyl paraben, methyl paraben, ethyl paraben, propyl paraben, benzoic acid, hydroxybenzoic acid, potassium benzoate, potassium sorbate, sodium benzoate, sodium propionate, and/or sorbic acid. Exemplary alcohol
preservatives include, but are not limited to, ethanol, polyethylene glycol, phenol, phenolic compounds, bisphenol, chlorobutanol, hydroxybenzoate, and/or phenylethyl alcohol. Exemplary acidic preservatives include, but are not limited to, vitamin A, vitamin C, vitamin E, beta- carotene, citric acid, acetic acid, dehydroacetic acid, ascorbic acid, sorbic acid, and/or phytic acid. Other preservatives include, but are not limited to, tocopherol, tocopherol acetate, deteroxime mesylate, cetrimide, butylated hydroxyanisol (BHA), butylated hydroxytoluened (BHT), ethylenediamine, sodium lauryl sulfate (SLS), sodium lauryl ether sulfate (SLES), sodium bisulfite, sodium metabisulfite, potassium sulfite, potassium metabisulfite, Glydant Plus®, Phenonip®, methylparaben, Germall®115, Germaben®II, Neolone, Kathon, and/or Euxyl®.
Exemplary buffering agents include, but are not limited to, citrate buffer solutions, acetate buffer solutions, phosphate buffer solutions, ammonium chloride, calcium carbonate, calcium chloride, calcium citrate, calcium glubionate, calcium gluceptate, calcium gluconate, d- gluconic acid, calcium glycerophosphate, calcium lactate, propanoic acid, calcium levulinate, pentanoic acid, dibasic calcium phosphate, phosphoric acid, tribasic calcium phosphate, calcium hydroxide phosphate, potassium acetate, potassium chloride, potassium gluconate, potassium mixtures, dibasic potassium phosphate, monobasic potassium phosphate, potassium phosphate mixtures, sodium acetate, sodium bicarbonate, sodium chloride, sodium citrate, sodium lactate, dibasic sodium phosphate, monobasic sodium phosphate, sodium phosphate mixtures, tromethamine, magnesium hydroxide, aluminum hydroxide, alginic acid, pyrogen-free water, isotonic saline, Ringer's solution, ethyl alcohol, etc., and/or combinations thereof.
Exemplary lubricating agents include, but are not limited to, magnesium stearate, calcium stearate, stearic acid, silica, talc, malt, glyceryl behanate, hydrogenated vegetable oils, polyethylene glycol, sodium benzoate, sodium acetate, sodium chloride, leucine, magnesium lauryl sulfate, sodium lauryl sulfate, etc., and combinations thereof.
Exemplary oils include, but are not limited to, almond, apricot kernel, avocado, babassu, bergamot, black current seed, borage, cade, camomile, canola, caraway, carnauba, castor, cinnamon, cocoa butter, coconut, cod liver, coffee, corn, cotton seed, emu, eucalyptus, evening primrose, fish, flaxseed, geraniol, gourd, grape seed, hazel nut, hyssop, isopropyl myristate, jojoba, kukui nut, lavandin, lavender, lemon, litsea cubeba, macademia nut, mallow, mango seed, meadowfoam seed, mink, nutmeg, olive, orange, orange roughy, palm, palm kernel, peach kernel, peanut, poppy seed, pumpkin seed, rapeseed, rice bran, rosemary, safflower, sandalwood, sasquana, savoury, sea buckthorn, sesame, shea butter, silicone, soybean, sunflower, tea tree, thistle, tsubaki, vetiver, walnut, and wheat germ oils. Exemplary oils include, but are not limited to, butyl stearate, caprylic triglyceride, capric triglyceride, cyclomethicone, diethyl sebacate, dimethicone 360, isopropyl myristate, mineral oil, octyldodecanol, oleyl alcohol, silicone oil, and/or combinations thereof.
Liquid dosage forms for oral and parenteral administration include, but are not limited to, pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups, and/or elixirs. In addition to active ingredients, liquid dosage forms may comprise inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, dimethylformamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof. Besides inert diluents, oral compositions can include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and/or perfuming agents. In certain embodiments for parenteral administration, compositions are mixed with solubilizing agents such as Cremophor®, alcohols, oils, modified oils, glycols, polysorbates, cyclodextrins, polymers, and/or combinations thereof.
Injectable preparations, for example, sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing agents, wetting agents, and/or suspending agents. Sterile injectable preparations may be sterile injectable solutions, suspensions, and/or emulsions in nontoxic parenterally acceptable diluents and/or solvents, for example, as a solution in 1,3-butanediol. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution, U.S.P., and isotonic sodium chloride solution. Sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose any bland fixed oil can be employed including synthetic mono- or diglycerides. Fatty acids such as oleic acid can be used in the preparation of injectables.
Injectable formulations can be sterilized, for example, by filtration through a bacterial- retaining filter, and/or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved or dispersed in sterile water or other sterile injectable medium prior to use. In order to prolong the effect of an active ingredient, it is often desirable to slow the absorption of the active ingredient from subcutaneous or intramuscular injection. This may be accomplished by the use of a liquid suspension of crystalline or amorphous material with poor water solubility. The rate of absorption of the drug then depends upon its rate of dissolution which, in turn, may depend upon crystal size and crystalline form. Alternatively, delayed absorption of a parenterally administered drug form is accomplished by dissolving or suspending the drug in an oil vehicle. Injectable depot forms are made by forming microencapsule matrices of the drug in biodegradable polymers such as polylactide-polyglycolide. Depending upon the ratio of drug to polymer and the nature of the particular polymer employed, the rate of drug release can be controlled. Examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides). Depot injectable formulations are prepared by entrapping the drug in liposomes or microemulsions which are compatible with body tissues.
Compositions for rectal or vaginal administration are typically suppositories which can be prepared by mixing compositions with suitable non-irritating excipients such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active ingredient.
Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules. In such solid dosage forms, an active ingredient is mixed with at least one inert, pharmaceutically acceptable excipient such as sodium citrate or dicalcium phosphate and/or fillers or extenders (e.g. starches, lactose, sucrose, glucose, mannitol, and silicic acid), binders (e.g. carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidinone, sucrose, and acacia), humectants (e.g. glycerol), disintegrating agents (e.g. agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate), solution retarding agents (e.g. paraffin), absorption accelerators (e.g. quaternary ammonium compounds), wetting agents (e.g. cetyl alcohol and glycerol monostearate), absorbents (e.g. kaolin and bentonite clay), and lubricants (e.g. talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate), and mixtures thereof. In the case of capsules, tablets and pills, the dosage form may comprise buffering agents.
Solid compositions of a similar type may be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like. Solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings and other coatings well known in the pharmaceutical formulating art. They may optionally comprise opacifying agents and can be of a composition that they release the active ingredient(s) only, or
preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Examples of embedding compositions which can be used include polymeric substances and waxes. Solid compositions of a similar type may be employed as fillers in soft and hard- filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like.
Dosage forms for topical and/or transdermal administration of a composition may include ointments, pastes, creams, lotions, gels, powders, solutions, sprays, inhalants and/or patches. Generally, an active ingredient is admixed under sterile conditions with a pharmaceutically acceptable excipient and/or any needed preservatives and/or buffers as may be required.
Additionally, the present disclosure contemplates the use of transdermal patches, which often have the added advantage of providing controlled delivery of a compound to the body. Such dosage forms may be prepared, for example, by dissolving and/or dispensing the compound in the proper medium. Alternatively or additionally, rate may be controlled by either providing a rate controlling membrane and/or by dispersing the compound in a polymer matrix and/or gel.
Suitable devices for use in delivering intradermal pharmaceutical compositions described herein include short needle devices such as those described in U.S. Patents 4,886,499; 5,190,521; 5,328,483; 5,527,288; 4,270,537; 5,015,235; 5,141,496; and 5,417,662. Intradermal
compositions may be administered by devices which limit the effective penetration length of a needle into the skin, such as those described in PCT publication WO 99/34850 and functional equivalents thereof. Jet injection devices which deliver liquid compositions to the dermis via a liquid jet injector and/or via a needle which pierces the stratum corneum and produces a jet which reaches the dermis are suitable. Jet injection devices are described, for example, in U.S. Patents 5,480,381; 5,599,302; 5,334,144; 5,993,412; 5,649,912; 5,569,189; 5,704,911;
5,383,851; 5,893,397; 5,466,220; 5,339,163; 5,312,335; 5,503,627; 5,064,413; 5,520,639;
4,596,556; 4,790,824; 4,941,880; 4,940,460; and PCT publications WO 97/37705 and WO 97/13537. Ballistic powder/particle delivery devices which use compressed gas to accelerate vaccine in powder form through the outer layers of the skin to the dermis are suitable. Alternatively or additionally, conventional syringes may be used in the classical mantoux method of intradermal administration.
Formulations suitable for topical administration include, but are not limited to, liquid and/or semi liquid preparations such as liniments, lotions, oil in water and/or water in oil emulsions such as creams, ointments and/or pastes, and/or solutions and/or suspensions.
Topically-administrable formulations may, for example, comprise from about 1% to about 10% (w/w) active ingredient, although the concentration of active ingredient may be as high as the solubility limit of the active ingredient in the solvent. Formulations for topical administration may further comprise one or more of the additional ingredients described herein.
A pharmaceutical composition may be prepared, packaged, and/or sold in a formulation suitable for pulmonary administration via the buccal cavity. Such a formulation may comprise dry particles which comprise the active ingredient and which have a diameter in the range from about 0.5 nm to about 7 nm or from about 1 nm to about 6 nm. Such compositions are conveniently in the form of dry powders for administration using a device comprising a dry powder reservoir to which a stream of propellant may be directed to disperse the powder and/or using a self propelling solvent/powder dispensing container such as a device comprising the active ingredient dissolved and/or suspended in a low-boiling propellant in a sealed container. Such powders comprise particles wherein at least 98% of the particles by weight have a diameter greater than 0.5 nm and at least 95% of the particles by number have a diameter less than 7 nm. Alternatively, at least 95% of the particles by weight have a diameter greater than 1 nm and at least 90% of the particles by number have a diameter less than 6 nm. Dry powder compositions may include a solid fine powder diluent such as sugar and are conveniently provided in a unit dose form.
Low boiling propellants generally include liquid propellants having a boiling point of below 65 °F at atmospheric pressure. Generally the propellant may constitute 50% to 99.9% (w/w) of the composition, and active ingredient may constitute 0.1 % to 20% (w/w) of the composition. A propellant may further comprise additional ingredients such as a liquid non- ionic and/or solid anionic surfactant and/or a solid diluent (which may have a particle size of the same order as particles comprising the active ingredient).
Pharmaceutical compositions formulated for pulmonary delivery may provide an active ingredient in the form of droplets of a solution and/or suspension. Such formulations may be prepared, packaged, and/or sold as aqueous and/or dilute alcoholic solutions and/or suspensions, optionally sterile, comprising active ingredient, and may conveniently be administered using any nebulization and/or atomization device. Such formulations may further comprise one or more additional ingredients including, but not limited to, a flavoring agent such as saccharin sodium, a volatile oil, a buffering agent, a surface active agent, and/or a preservative such as
methylhydroxybenzoate. Droplets provided by this route of administration may have an average diameter in the range from about 0.1 nm to about 200 nm.
Formulations described herein as being useful for pulmonary delivery are useful for intranasal delivery of a pharmaceutical composition. Another formulation suitable for intranasal administration is a coarse powder comprising the active ingredient and having an average particle from about 0.2 μιη to 500 μιη. Such a formulation is administered in the manner in which snuff is taken, i.e. by rapid inhalation through the nasal passage from a container of the powder held close to the nose.
Formulations suitable for nasal administration may, for example, comprise from about as little as 0.1% (w/w) and as much as 100% (w/w) of active ingredient, and may comprise one or more of the additional ingredients described herein. A pharmaceutical composition may be prepared, packaged, and/or sold in a formulation suitable for buccal administration. Such formulations may, for example, be in the form of tablets and/or lozenges made using
conventional methods, and may, for example, 0.1% to 20% (w/w) active ingredient, the balance comprising an orally dissolvable and/or degradable composition and, optionally, one or more of the additional ingredients described herein. Alternately, formulations suitable for buccal administration may comprise a powder and/or an aerosolized and/or atomized solution and/or suspension comprising active ingredient. Such powdered, aerosolized, and/or aerosolized formulations, when dispersed, may have an average particle and/or droplet size in the range from about 0.1 nm to about 200 nm, and may further comprise one or more of any additional ingredients described herein.
A pharmaceutical composition may be prepared, packaged, and/or sold in a formulation suitable for ophthalmic administration. Such formulations may, for example, be in the form of eye drops including, for example, a 0.1/1.0%) (w/w) solution and/or suspension of the active ingredient in an aqueous or oily liquid excipient. Such drops may further comprise buffering agents, salts, and/or one or more other of any additional ingredients described herein. Other opthalmically-administrable formulations which are useful include those which comprise the active ingredient in microcrystalline form and/or in a liposomal preparation. Ear drops and/or eye drops are contemplated as being within the scope of this present disclosure.
General considerations in the formulation and/or manufacture of pharmaceutical agents may be found, for example, in Remington: The Science and Practice of Pharmacy 21st ed., Lippincott Williams & Wilkins, 2005 (incorporated herein by reference).
Administration
The present disclosure provides methods comprising administering proteins or complexes in accordance with the present disclosure to a subject in need thereof. Proteins or complexes, or pharmaceutical, imaging, diagnostic, or prophylactic compositions thereof, may be administered to a subject using any amount and any route of administration effective for preventing, treating, diagnosing, or imaging a disease, disorder, and/or condition {e.g., a disease, disorder, and/or condition relating to working memory deficits). The exact amount required will vary from subject to subject, depending on the species, age, and general condition of the subject, the severity of the disease, the particular composition, its mode of administration, its mode of activity, and the like. Compositions in accordance with the present disclosure are typically formulated in dosage unit form for ease of administration and uniformity of dosage. It will be understood, however, that the total daily usage of the compositions of the present disclosure will be decided by the attending physician within the scope of sound medical judgment. The specific therapeutically effective, prophylactially effective, or appropriate imaging dose level for any particular patient will depend upon a variety of factors including the disorder being treated and the severity of the disorder; the activity of the specific compound employed; the specific composition employed; the age, body weight, general health, sex and diet of the patient; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidental with the specific compound employed; and like factors well known in the medical arts.
Proteins to be delivered and/or pharmaceutical, prophylactic, diagnostic, or imaging compositions thereof may be administered to animals, such as mammals {e.g., humans, domesticated animals, cats, dogs, mice, rats, etc.). In some embodiments, pharmaceutical, prophylactic, diagnostic, or imaging compositions thereof are administered to humans. Proteins to be delivered and/or pharmaceutical, prophylactic, diagnostic, or imaging compositions thereof in accordance with the present disclosure may be administered by any route. In some embodiments, proteins and/or pharmaceutical, prophylactic, diagnostic, or imaging compositions thereof, are administered by one or more of a variety of routes, including oral, intravenous, intramuscular, intra-arterial, intramedullary, intrathecal, subcutaneous, intraventricular, transdermal, interdermal, rectal, intravaginal, intraperitoneal, topical (e.g. by powders, ointments, creams, gels, lotions, and/or drops), mucosal, nasal, buccal, enteral, vitreal, intratumoral, sublingual; by intratracheal instillation, bronchial instillation, and/or inhalation; as an oral spray, nasal spray, and/or aerosol, and/or through a portal vein catheter. In some embodiments, proteins or complexes, and/or pharmaceutical, prophylactic, diagnostic, or imaging compositions thereof, are administered by systemic intravenous injection. In specific embodiments, proteins or complexes and/or pharmaceutical, prophylactic, diagnostic, or imaging compositions thereof may be administered intravenously and/or orally. In specific embodiments, proteins or complexes, and/or pharmaceutical, prophylactic, diagnostic, or imaging compositions thereof, may be administered in a way which allows the protein or complex to cross the blood- brain barrier, vascular barrier, or other epithelial barrier.
However, the present disclosure encompasses the delivery of proteins or complexes, and/or pharmaceutical, prophylactic, diagnostic, or imaging compositions thereof, by any appropriate route taking into consideration likely advances in the sciences of drug delivery.
In general the most appropriate route of administration will depend upon a variety of factors including the nature of the protein or complex comprising proteins associated with at least one agent to be delivered (e.g., its stability in the environment of the gastrointestinal tract, bloodstream, etc.), the condition of the patient (e.g., whether the patient is able to tolerate particular routes of administration), etc. The present disclosure encompasses the delivery of the pharmaceutical, prophylactic, diagnostic, or imaging compositions by any appropriate route taking into consideration likely advances in the sciences of drug delivery.
In certain embodiments, compositions in accordance with the present disclosure may be administered at dosage levels sufficient to deliver from about 0.0001 mg/kg to about 100 mg/kg, from about 0.01 mg/kg to about 50 mg/kg, from about 0.1 mg/kg to about 40 mg/kg, from about 0.5 mg/kg to about 30 mg/kg, from about 0.01 mg/kg to about 10 mg/kg, from about 0.1 mg/kg to about 10 mg/kg, or from about 1 mg/kg to about 25 mg/kg, of subject body weight per day, one or more times a day, to obtain the desired therapeutic, diagnostic, prophylactic, or imaging effect. The desired dosage may be delivered three times a day, two times a day, once a day, every other day, every third day, every week, every two weeks, every three weeks, or every four weeks. In certain embodiments, the desired dosage may be delivered using multiple
administrations (e.g., two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, fourteen, or more administrations).
Proteins or complexes may be used in combination with one or more other therapeutic, prophylactic, diagnostic, or imaging agents. By "in combination with," it is not intended to imply that the agents must be administered at the same time and/or formulated for delivery together, although these methods of delivery are within the scope of the present disclosure. Compositions can be administered concurrently with, prior to, or subsequent to, one or more other desired therapeutics or medical procedures. In general, each agent will be administered at a dose and/or on a time schedule determined for that agent. In some embodiments, the present disclosure encompasses the delivery of pharmaceutical, prophylactic, diagnostic, or imaging compositions in combination with agents that improve their bioavailability, reduce and/or modify their metabolism, inhibit their excretion, and/or modify their distribution within the body.
It will further be appreciated that therapeutically, prophylactically, diagnostically, or imaging active agents utilized in combination may be administered together in a single composition or administered separately in different compositions. In general, it is expected that agents utilized in combination with be utilized at levels that do not exceed the levels at which they are utilized individually. In some embodiments, the levels utilized in combination will be lower than those utilized individually.
The particular combination of therapies (therapeutics or procedures) to employ in a combination regimen will take into account compatibility of the desired therapeutics and/or procedures and the desired therapeutic effect to be achieved. It will also be appreciated that the therapies employed may achieve a desired effect for the same disorder (for example, a composition useful for treating cancer in accordance with the present disclosure may be administered concurrently with a chemotherapeutic agent), or they may achieve different effects (e.g., control of any adverse effects). Kits
The present disclosure provides a variety of kits for conveniently and/or effectively carrying out methods of the present disclosure. Typically kits will comprise sufficient amounts and/or numbers of components to allow a user to perform multiple treatments of a subject(s) and/or to perform multiple experiments.
In one aspect, the disclosure provides kits for protein production, comprising a first isolated nucleic acid comprising a translatable region and a nucleic acid modification, wherein the nucleic acid is capable of evading an innate immune response of a cell into which the first isolated nucleic acid is introduced, and packaging and instructions.
In one aspect, the disclosure provides kits for protein production, comprising: a first isolated nucleic acid comprising a translatable region, provided in an amount effective to produce a desired amount of a protein encoded by the translatable region when introduced into a target cell; a second nucleic acid comprising an inhibitory nucleic acid, provided in an amount effective to substantially inhibit the innate immune response of the cell; and packaging and instructions.
In one aspect, the disclosure provides kits for protein production, comprising a first isolated nucleic acid comprising a translatable region and a nucleoside modification, wherein the nucleic acid exhibits reduced degradation by a cellular nuclease, and packaging and instructions.
In one aspect, the disclosure provides kits for protein production, comprising a first isolated nucleic acid comprising a translatable region and at least two different nucleoside modifications, wherein the nucleic acid exhibits reduced degradation by a cellular nuclease, and packaging and instructions.
In one aspect, the disclosure provides kits for protein production, comprising a first isolated nucleic acid comprising a translatable region and at least one nucleoside modification, wherein the nucleic acid exhibits reduced degradation by a cellular nuclease; a second nucleic acid comprising an inhibitory nucleic acid; and packaging and instructions.
In some embodiments, the first isolated nucleic acid comprises messenger RNA
(mRNA). In some embodiments the mRNA comprises at least one nucleoside selected from the group consisting of pyridin-4-one ribonucleoside, 5-aza-uridine, 2-thio-5-aza-uridine, 2- thiouridine, 4-thio-pseudouridine, 2-thio-pseudouridine, 5-hydroxyuridine, 3-methyluridine, 5- carboxymethyl-uridine, 1-carboxymethyl-pseudouridine, 5-propynyl-uridine, 1-propynyl- pseudouridine, 5-taurinomethyluridine, 1-taurinomethyl-pseudouridine, 5-taurinomethyl-2-thio- uridine, l-taurinomethyl-4-thio-uridine, 5-methyl-uridine, 1-methyl-pseudouridine, 4-thio- 1- methyl-pseudouridine, 2-thio- 1-methyl-pseudouridine, 1 -methyl- 1-deaza-pseudouridine, 2-thio-
1 - methyl- 1-deaza-pseudouridine, dihydrouridine, dihydropseudouridine, 2-thio-dihydrouridine,
2- thio-dihydropseudouridine, 2-methoxyuridine, 2-methoxy-4-thio-uridine, 4-methoxy- pseudouridine, and 4-methoxy-2-thio-pseudouridine.
In some embodiments, the mRNA comprises at least one nucleoside selected from the group consisting of 5-aza-cytidine, pseudoisocytidine, 3-methyl-cytidine, N4-acetylcytidine, 5- formylcytidine, N4-methylcytidine, 5-hydroxymethylcytidine, 1-methyl-pseudoisocytidine, pyrrolo-cytidine, pyrrolo-pseudoisocytidine, 2-thio-cytidine, 2-thio-5-methyl-cytidine, 4-thio- pseudoisocytidine, 4-thio- 1 -methyl-pseudoisocytidine, 4-thio- 1 -methyl- 1 -deaza- pseudoisocytidine, 1 -methyl- 1-deaza-pseudoisocytidine, zebularine, 5-aza-zebularine, 5-methyl- zebularine, 5-aza-2-thio-zebularine, 2-thio-zebularine, 2-methoxy-cytidine, 2-methoxy-5-methyl- cytidine, 4-methoxy-pseudoisocytidine, and 4-methoxy- 1-methyl-pseudoisocytidine.
In some embodiments, the mRNA comprises at least one nucleoside selected from the group consisting of 2-aminopurine, 2, 6-diaminopurine, 7-deaza-adenine, 7-deaza-8-aza-adenine, 7-deaza-2-aminopurine, 7-deaza-8-aza-2-aminopurine, 7-deaza-2, 6-diaminopurine, 7-deaza-8- aza-2, 6-diaminopurine, 1-methyladenosine, N6-methyladenosine, N6-isopentenyladenosine, N6- (cis-hydroxyisopentenyl)adenosine, 2-methylthio-N6-(cis-hydroxyisopentenyl) adenosine, N6- glycinylcarbamoyladenosine, N6-threonylcarbamoyladenosine, 2-methylthio-N6-threonyl carbamoyladenosine, N6,N6-dimethyladenosine, 7-methyladenine, 2-methylthio-adenine, and 2- methoxy-adenine.
In some embodiments, the mRNA comprises at least one nucleoside selected from the group consisting of inosine, 1-methyl-inosine, wyosine, wybutosine, 7-deaza-guanosine, 7- deaza-8-aza-guanosine, 6-thio-guanosine, 6-thio-7-deaza-guanosine, 6-thio-7-deaza-8-aza- guanosine, 7-methyl-guanosine, 6-thio-7-methyl-guanosine, 7-methylinosine, 6-methoxy- guanosine, 1-methylguanosine, N2-methylguanosine, N2,N2-dimethylguanosine, 8-oxo- guanosine, 7-methyl-8-oxo-guanosine, l-methyl-6-thio-guanosine, N2-methyl-6-thio-guanosine, and N2,N2-dimethyl-6-thio-guanosine.
In another aspect, the disclosure provides compositions for protein production, comprising a first isolated nucleic acid comprising a translatable region and a nucleoside modification, wherein the nucleic acid exhibits reduced degradation by a cellular nuclease, and a mammalian cell suitable for translation of the translatable region of the first nucleic acid.
EXAMPLES
The invention is further described in the following examples, which do not limit the scope of the invention described in the claims.
Example 1. Modified mRNA In Vitro Transcription
Materials and Methods
Modified mRNAs (modRNAs) were made using standard laboratory methods and materials for in vitro transcription with the exception that the nucleotide mix contained modified nucleotides. The open reading frame (ORF) of the gene of interest is flanked by a 5' untranslated region (UTR) containing a strong Kozak translational initiation signal and an alpha-globin 3' UTR terminating with an oligo(dT) sequence for templated addition of a polyA tail for modRNAs not incorporating Adenosine analogs. Adenosine-containing modRNAs were synthesized without an oligo (dT) sequence to allow for post-transcription poly (A) polymerase poly-(A) tailing. The modRNAs were modified by incorporating chemically modified nucleotides indicated in Table 3 (below) during the in vitro transcription with 100% replacement of the corresponding natural nucleotide or partial replacement of the corresponding natural nucleotide at the indicated percentage.
Table 3 indicates the chemical identity of each chemically-distinct modified nucleotide incorporated into a modified mRNA with the given chemistry designation number.
Table 3
Figure imgf000065_0001
5-iodo-uridine Chem 6 Pseudo-iso-cytidine/5-iodo-uridine Chem 34
N l-methyl-pseudouridine Chem 7 Pseudo-iso-cytidine/N l-methyl-pseudo-uridine Chem 35
5,6-dihydrouridine Chem 8 Pseudo-iso-cytidine/a-thio-uridine Chem 36 a-thio-uridine Chem 9 Pseudo-iso-cytidine/5-methyl-uridine Chem 37
4-thio-uridine Chem 10 Pseudo-iso-cytidine/Pseudo-uridine Chem 38
6-aza-uridine Chem 11 Pyrrolo-cytidine Chem 39
5-hydroxy-uridine Chem 12 Pyrrolo-cytidine/5-iodo-uridine Chem 40
Deoxy-thymidine Chem 13 Pyrrolo-cytidine/N l-methyl-pseudo-uridine Chem 41
Pseudo-uridine Chem 14 Pyrrolo-cytidine/a-thio-uridine Chem 42
Inosine Chem 15 Pyrrolo-cytidine/5-methyl-uridine Chem 43 a-thio-guanosine Chem 16 Pyrrolo-cytidine/Pseudo-uridine Chem 44
8-oxo-guanosine Chem 17 5-methyl-cytidine/5-iodo-uridine Chem 45
06-methyl-guanosine Chem 18 5-methyl-cytidine/N l-methyl-pseudo-uridine Chem 46
7-deaza-guanosine Chem 19 5-methyl-cytidine/a-thio-uridine Chem 47
No modification Chem 20 5-methyl-cytidine/5-methyl-uridine Chem 48
N 1-methyl-adenosine Chem 21 5-methyl-cytidine/Pseudo-uridine Chem 49
2-amino-6-Chloro-purine Chem 22 5-methyl-cytidine Chem 50
N6-methyl-2-amino- Chem 23 25% Pseudo-iso-cytidine Chem 51 purine
6-Chloro-purine Chem 24 25% N l-methyl-pseudo-uridine Chem 52
N6-methyl-adenosine Chem 25 25% N l-Methyl-pseudo-uridine/75%-pseudo- Chem 53 uridine
a-thio-adenosine Chem 26 5-methyl-uridine Chem 54
8-azido-adenosine Chem 27 5-iodo-cytidine Chem 55
7-deaza-adenosine Chem 28 Agarose Gel Electrophoresis of modRNA: Individual modRNAs (200-400 ng in a 20 μΐ volume) were loaded into a well on a non-denaturing 1.2% Agarose E-Gel (Invitrogen, Carlsbad, CA) and run for 12-15 minutes according to the manufacturer protocol (FIG. 1A). Tables 4 and 5 below indicate the modified nucleotide (Table 4) or nucleic acid (Table 5) loaded in each lane. These data indicate which chemically modified nucleotides were transcribed into chemically-modified mRNAs and the quality of each individual modRNA. These data demonstrate that nucleotides with chemical modifications on the major groove and minor groove face of the nucleotide were capable of being transcribed into a modRNA.
Table 4
Lane Modified NTP
1 a-thio-cytidine
2 Pseudo-iso-cytidine
3 5-aminoallyl-uridine
4 5-iodo-uridine
5 Nl-methyl-pseudo-uridine
6 a-thio-uridine
7 4-t io-uridine
8 5-hydroxy-uridine
9 Deoxy-thymidine
10 Pseudo-uridine
11 Inosine
12 a-thio-guanosine
13 8-oxo-guanosine
14 Nl-methyl-guanosine
15 06-methyl-guanosine
16 No modification
17 Nl-methyl-adenosine
18 2-amino-6-Chloro-purine
19 N6-methyl-2-amino-purine
20 6-Chloro-purine
21 ct-thio-adenosine
22 8-azido-adenosine
23 7-deaza-adenosine
24 6-aza-cytidine
25 2-thio-cytidine
26 5,6-dihydro-uridine
27 6-aza-uridine
28 7-deaza-guanosine
29 N6-methyl-adenosine Table 5
Lane Modified NTP combination
1 a-thio-cytidine/5-iodo-uridine
2 a-thio-cytidine/Nl-methyl-pseudouridine
3 a-thio-cytidine/a-thio-uridine
4 a-thio-cytidine/5-methyl-uridine
5 a-thio-cytidine/pseudouridine
6 5-iodo-cytidine/5-iodo-uridine
7 5-iodo-cytidine/Nl-methyl-pseudouridine
8 5-iodo-cytidine/a-thio-uridine
9 5-iodo-cytidine/5-methyl-uridine
10 5-iodo-cytidine/pseudouridine
11 Pseudo-iso-cytidine/5-iodo-uridine
12 Pyrrolo-cytidine
13 Pyrrolo-cytidine/5-iodo-uridine
14 Pyrrolo-cytidine/Nl-methyl-pseudouridine
15 Py rro 1 o-cyti d i n e/a-t hio- uri d i n e
16 Pyrrolo-cytidine/5-methyl-uridine
17 Py rro 1 o-cyti d i n e/pseudo uri d i n e
18 5-methyl-cytidine/5-iodo-uridine
19 5-methyl-cytidine/Nl-methyl-uridine
20 5-methyl-cytidine/a-thio-uridine
21 5-methyl-cytidine/5-methyl-uridine
22 5-methyl-cytidine/pseudouridine
23 Pseudo-iso-cytidine/Nl-methyl-pseudouridine
24 Pseudo-iso-cytidine/a-thio-uridine
25 Pseudo-iso-cytidine/5-methyl-uridine
26 Pseudo-iso-cytidine/pseudouridine
27 5-methyl-cytidine
28 25% pseudo-iso-cytidine
29 25% Nl-methyl-pseudouridine
30 25% Nl-methyl-pseudouridine/75% pseudouridine
Agarose Gel Electrophoresis of RT-PCR products: Individual reverse transcribed-PCR products (200-400ng) were loaded into a well of a non-denaturing 1.2% Agarose E-Gel
(Invitrogen, Carlsbad, CA) and run for 12-15 minutes according to the manufacturer protocol (FIG. IB). Table 5 below indicates the modified nucleotide loaded in each lane.
Nanodrop modRNA quantification and UV spectral data: modRNAs in TE buffer (1 μΐ) were used for Nanodrop UV absorbance readings to quantitate the yield of each modRNA from an in vitro transcription reaction (UV absorbance traces are shown in Figures 6A-6L). These data indicate which chemically modified nucleotides were transcribed into chemically- modified mRNAs. These data also demonstrate that nucleotides with chemical modifications on the major groove and minor groove face of the nucleotide were capable of being transcribed into a modRNA. These data further demonstrate that the nucleotides of the present invention are transcription-competent and compatible with incorporation into a modRNA, which may have altered UV spectra due to the presence of a given modified nucleotide. For example, Pyrrolo-C containing modRNAs have an increase in UV absorbance at a lower wavelength due to the presence of the pyrrolo ring of the modified C nucleotide. In another example, 2-amino-adenine nucleotide-containing modRNAs have an increase in UV absorbance at a higher wavelength due to the presence of an exocyclic amine off the purine ring. Nucleotides that are not transcription- competent and cannot be incorporated into a modRNA have a scrambled UV spectrum indicating no product from the transcription reaction.
Example 2. Modified RNA Transfection
Reverse Transfection: For experiments performed in a 24-well collagen-coated tissue culture plate, Keratinocytes were seeded at a cell density of 1 x 105. For experiments performed in a 96-well collagen-coated tissue culture plate, Keratinocytes were seeded at a cell density of 0.5 x 105. For each modRNA to be transfected, modRNA: RNAiMAX was prepared as described and mixed with the cells in the multi-well plate within a period of time, e.g., 6 hours, of cell seeding before cells had adhered to the tissue culture plate.
Forward Transfection: In a 24-well collagen-coated tissue culture plate, Keratinocytes were seeded at a cell density of 0.7 x 105. For experiments performed in a 96-well collagen- coated tissue culture plate, Keratinocytes were seeded at a cell density of 0.3 x 105.
Keratinocytes were then grown to a confluency of >70% for over 24 hours. For each modRNA to be transfected, modRNA: RNAiMAX was prepared as described and transfected onto the cells in the multi-well plate over 24 hours after cell seeding and adherence to the tissue culture plate.
modRNA Translation Screen: G-CSF ELISA
FIGs. 2A and 2B show an Enzyme-linked immunosorbent assay (ELISA) for Human Granulocyte-Colony Stimulating Factor (hu-G-CSF) of in vitro transfected Human Keratinocyte cells. Keratinocytes were grown in EpiLife medium with Supplement S7 from Invitrogen at a confluence of >70%. FIG. 2A keratinocytes were reverse transfected with 300 ng of the indicated chemically modified mRNA complexed with RNAiMAX from Invitrogen. FIG. 2B keratinocytes were forward transfected with 300 ng modRNA complexed with RNAiMAX from Invitrogen. The RNA:RNAiMAX complex was formed by first incubating the RNA with Supplement- free EpiLife media in a 5X volumetric dilution for 10 minutes at room temperature. In a second vial, RNAiMAX reagent was incubated with Supplement- free EpiLife Media in a 10X volumetric dilution for 10 minutes at room temperature. The RNA vial was then mixed with the RNAiMAX vial and incubated for 20-30 at room temperature before being added to the cells in a drop-wise fashion. Secreted huG-CSF concentration in the culture medium was measured at 18 hours post-transfection for each of the chemically modified mRNAs in triplicate. Secretion of Human Granulocyte-Colony Stimulating Factor (G-CSF) from transfected human keratinocytes was quantified using an ELISA kit from Invitrogen or R&D Systems (Minneapolis, MN) following the manufacturers recommended instructions. These data show that huG-CSF modRNAs comprised of chemically distinct nucleotide analogs (SEQ ID NO: 2) is capable of being translated in Human Keratinocyte cells and that huG-CSF is transported out of the cells and released into the extracellular environment. These data indicate which modified nucleotides were translated into protein when incorporated into a chemically modified mRNA. These data show that modified RNA containing nucleotides with chemical modifications on the major groove face of pyrimidine analogs have the highest levels of secreted hu-G-CSF into the cell culture medium.
modRNA Dose and Duration: G-CSF ELISA
FIGs. 3A-N show Enzyme-linked immunosorbent assays (ELISA) for Human
Granulocyte-Colony Stimulating Factor (G-CSF) of in vitro transfected Human Keratinocyte cells. Keratinocytes were grown in EpiLife medium with Supplement S7 from Invitrogen at a confluence of >70%. Keratinocytes were reverse transfected with Ong, 46.875ng, 93.75ng, 187.5ng, 375ng, 750ng, or 1500ng modRNA complexed with RNAiMAX from Invitrogen. The modRNA:RNAiMAX complex was formed as described. Secreted huG-CSF concentration in the culture medium was measured at 0, 6, 12, 24, and 48 hours post-transfection for each concentration of each modRNA in triplicate. Secretion of Human Granulocyte-Colony
Stimulating Factor (G-CSF) from transfected human keratinocytes was quantified using an ELISA kit from Invitrogen or R&D Systems following the manufacturers recommended instructions. These data show that huG-CSF modRNAs comprised of chemically distinct nucleotide analogs (SEQ ID NO: X and Table 6) secreted hu-G-CSF protein in a modRNA dose- dependent manner from Human Keratinocyte cells and that huG-CSF is transported out of the cells and released into the extracellular environment. These data indicate which modified RNAs containing modified nucleotide analogs sustain hu-G-CSF expression for the longest and at the highest levels. These data show that modified RNA containing modified nucleotides with chemical modifications on the major groove face of pyrimidine analogs have the highest levels of secreted hu-G-CSF into the cell culture medium and that 750ng of modRNA elicits the highest level of secreted hu-G-CSF.
Example 3. Cellular Innate Immune Response to modRNA
IFN-β ELISA and TNF-a ELISA:
FIGs. 4A-F show an Enzyme-linked immunosorbent assay (ELISA) for Human Tumor Necrosis Factor-a (TNF-a) (FIGs. 4A and 4B); Human Interferon-β (IFN-β) (FIGs. 4C and 4D); and Human Granulocyte-Colony Stimulating Factor (G-CSF) (FIGs. 4E and 4F) secreted from in vzYro-transfected Human Keratinocyte cells. Keratinocytes were grown in EpiLife medium with Human Keratinocyte Growth Supplement in the absence of hydrocortisone from Invitrogen at a confluence of >70%. In FIGs. 4A and 4B, keratinocytes were reverse transfected with Ong, 93.75ng, 187.5ng, 375ng, 750ng, 1500ng or 3000ng of the indicated chemically modified mRNA complexed with RNAiMAX from Invitrogen as described in triplicate. Secreted TNF-a in the culture medium was measured 24 hours post-transfection for each of the chemically modified mRNAs using an ELISA kit from Invitrogen according to the manufacturer protocols.
In FIGs. 4C and 4D, secreted IFN-β in the same culture medium was measured 24 hours post-transfection for each of the chemically modified mRNAs using an ELISA kit from
Invitrogen according to the manufacturer protocols. In FIGs. 4E and 4F, secreted hu-G-CSF concentration in the same culture medium was measured at 24 hours post-transfection for each of the chemically modified mRNAs. Secretion of Human Granulocyte-Colony Stimulating Factor (G-CSF) from transfected human keratinocytes was quantified using an ELISA kit from
Invitrogen or R&D Systems (Minneapolis, MN) following the manufacturers recommended instructions. These data indicate which modified RNAs containing modified nucleotides were capable of eliciting a reduced cellular innate immune response in comparison to natural and other chemically modified nucleotides by measuring exemplary type I cytokines TNF-a and IFN-β. These data show that modified RNAs containing modified nucleotides with chemical
modifications on the major groove face of pyrimidine analogs have the lowest levels of secreted TNF-a and IFN-β into the cell culture medium while maintaining high levels of modRNA- encoding hu-G-CSF secretion into the cell culture medium.
Example 4. Human Granulocyte-Colony Stimulating Factor-modified RNA-induced Cell Proliferation Assay
FIGs. 5A-D show modRNA-encoding hu-G-CSF produced by a human keratinocyte feeder cell layer induced the proliferation of both human myeloblast cells KG-1 and Kasumi-1 that express the G-CSF-receptor where the cell populations are separated by a semi-permeable membrane.
Human keratinocytes were grown in EpiLife medium with Supplement S7 from
Invitrogen at a confluence of >70% in a 24-well collagen-coated Transwell® (Corning, Lowell, MA) co-culture tissue culture plate. Keratinocytes were reverse transfected with 750ng of the indicated chemically modified mRNA complexed with RNAiMAX from Invitrogen as described in triplicate. The modRNA:RNAiMAX complex was formed as described. Keratinocyte media was exchanged 6-8 hours post-transfection. 42-hours post-transfection, the 24-well Transwell® plate insert with a Ο μιη-ροΓε semi-permeable polyester membrane was placed into the hu-G- CSF modRNA-transfected keratinocyte containing culture plate. FIG. 5A is a table showing the results from an Enzyme-linked immunosorbent assay (ELISA) for human-G-CSF secreted from in vzYro-transfected Human Keratinocyte cells sampled from individual wells in a co-culture 24- well tissue culture plate 42 hours post-transfection with 750ng of each indicated hu-G-CSF- encoding modRNA.
Human myeloblast cells, Kasumi-1 cells (FIG. 5C) or KG-1 (FIG. 5D) (0.2 x 105 cells), were seeded into the insert well and cell proliferation was quantified 42 hours post-co-culture initiation using the CyQuant Direct Cell Proliferation Assay (Invitrogen) in a 100-120 μΐ volume in a 96-well plate. modRNA-encoding hu-G-CSF-induced myeloblast cell proliferation was expressed as a percent cell proliferation normalized to untransfected keratinocyte/myeloblast co- culture control wells. Secreted hu-G-CSF concentration in both the keratinocyte and myeloblast insert co-culture wells was measured at 42 hours post-co-culture initiation for each modRNA in duplicate. Secretion of Human Granulocyte-Colony Stimulating Factor (G-CSF) was quantified using an ELISA kit from Invitrogen following the manufacturers recommended instructions.
Transfected hu-G-CSF modRNA in human keratinocyte feeder cells and untransfected human myeloblast cells were detected by RT-PCR. Total RNA from sample cells was extracted and lysed using RNeasy kit (Qiagen, Valencia, CA) according to the manufacturer instructions. Extracted total RNA was submitted to RT-PCR for specific amplification of modRNA-G-CSF using ProtoScript® M-MuLV Taq RT-PCR kit (New England BioLabs, Ipswich, MA) according to the manufacturer instructions with hu-G-CSF-specific primers (see below). RT-PCR products were visualized by 1.2% agarose gel electrophoresis (FIG. 5B). Table 6 below shows which modRNAs were run on the agarose gel.
Table 6
Lane Cell tvDe RT-PCR hu-G-CSF modRNA Tareet
1 Keratinocyte KG-1 Feeder Vehicle
2 Keratinocyte KG-1 Feeder Scramble RNA
3 Keratinocyte KG-1 Feeder No Modification
4 Keratinocyte KG-1 Feeder Chem 7
5 Keratinocyte KG-1 Feeder Ch m 6
6 Keratinocyte KG-1 Feeder Chem 37
7 Keratinocyte Kasumi-1 Feeder Vehicle
8 Keratinocyte Kasumi-1 Feeder Scramble RNA
9 Keratinocyte Kasumi-1 Feeder No Modification
10 Keratinocyte Kasumi-1 Feeder Chem 7
11 Keratinocyte Kasumi-1 Feeder Chem 6
12 Keratinocyte Kasumi-1 Feeder Chem 37
13 Keratinocyte KG-1 Feeder Chem 46
14 Keratinocyte KG-1 Feeder Chem 48
15 Keratinocyte KG-1 Feeder Chem 49
16 Keratinocyte KG-1 Feeder Chem 53
17 Keratinocyte Kasumi-1 Feeder Chem 46
18 Keratinocyte Kasumi-1 Feeder Chem 48
19 Keratinocyte Kasumi-1 Feeder Chem 49
20 Keratinocyte Kasumi-1 Feeder Chem 53
21 Kasumi-1 Vehicle
22 KG-1 Vehicle
23 Kasumi-1 Vehicle
24 Kasumi-1 Scramble RNA
25 Kasumi-1 No Modification
26 Kasumi-1 Chem 7
27 Kasumi-1 Chem 6
28 Kasumi-1 Chem 37
29 Kasumi-1 Chem 46 30 Kasumi-1 Chem 48
3i Kasumi-1 Chem 49
32 Kasumi-1 Chem 53
33 KG-1 Vehicle
34 KG-1 Scramble RNA
35 KG-1 No Modification
36 KG-1 Chem 7
37 Empty Empty
38 Empty Empty
39 Empty Empty
40 Empty Empty
41 Empty Empty
42 Empty Empty
Empty Empty
44 Empty Empty
These data show that human keratinocyte cells containing hu-G-CSF modRNAs comprised of chemically distinct nucleotide analogs secreted hu-G-CSF protein and that the secreted hu-G-CSF was physiologically-active in inducing the proliferation of human myeloblast cells expressing the G-CSF receptor. These data also show the secreted hu-G-CSF protein was permeable across a semi-permeable membrane and acted on a different non-G-CSF-producing cell population. Additionally, these data show that hu-G-CSF modRNA-transfected into human keratinocyte cells in a co-culture environment was present in only the transfected keratinocyte cells and not the un-transfected myeloblast cells. Further, these data show that the modified nucleotide chemical composition of hu-G-CSF modRNA did not affect resultant protein activity.
Example 5. The Effect of modRNA on Cellular Viability
Cytotoxicity and Apoptosis:
This experiment demonstrates cellular viability, cytotoxity and apoptosis for distinct modRNA- in vitro transfected Human Keratinocyte cells. Keratinocytes are grown in EpiLife medium with Human Keratinocyte Growth Supplement in the absence of hydrocortisone from Invitrogen at a confluence of >70%. Keratinocytes are reverse transfected with Ong, 46.875ng, 93.75ng, 187.5ng, 375ng, 750ng, 1500ng, 3000ng, or 6000ng of modRNA complexed with RNAiMAX from Invitrogen. The modRNA:RNAiMAX complex is formed. Secreted huG-CSF
concentration in the culture medium is measured at 0, 6, 12, 24, and 48 hours post-transfection for each concentration of each modRNA in triplicate. Secretion of Human Granulocyte-Colony Stimulating Factor (G-CSF) from transfected human keratinocytes is quantified using an ELISA kit from Invitrogen or R&D Systems following the manufacturers recommended instructions. Cellular viability, cytotoxicity and apoptosis is measured at 0, 12, 48, 96, and 192 hours post- transfection using the ApoToxGlo kit from Promega (Madison, WI) according to manufacturer instructions.
Example 6. Co-culture
The modified mRNA comprised of chemically-distinct modified nucleotides encoding human Granulocyte-Colony Stimulating Factor (G-CSF) may stimulate the cellular proliferation of a transfection incompetent cell in co-culture environment. The co-culture includes a highly transfectable cell type such as a human keratinocyte and a transfection incompetent cell type such as a white blood cell (WBC). The modified mRNA encoding G-CSF may be transfected into the highly transfectable cell allowing for the production and secretion of G-CSF protein into the extracellular environment where G-CSF acts in a paracrine-like manner to stimulate the white blood cell expressing the G-CSF receptor to proliferate. The expanded WBC population may be used to treat immune-compromised patients or partially reconstitute the WBC population of an immunosuppressed patient and thus reduce the risk of opportunistic infections.
Another example, a highly transfectable cell such as a fibroblast may be transfected with certain growth factors to support and simulate the growth, maintenance, or differentiation of poorly transfectable embryonic stem cells or induced pluripotent stem cells.
Example 7. 5'-Guanosine Capping on Modified Nucleic Acids (modRNAs)
The cloning, gene synthesis and vector sequencing was performed by DNA2.0 Inc. (Menlo Park, CA). Sequence and insert sequence are set forth herein. The ORF was restriction digested using Xbal and used for cDNA synthesis using tailed-or tail-less-PCR. The tailed-PCR cDNA product was used as the template for the modified mRNA synthesis reaction using 25mM mixture each modified nucleotide (all modified nucleotides were custom synthesized or purchased from TriLink Biotech, San Diego, CA except pyrrolo-C triphosphate purchased from Glen Research, Sterling VA; unmodifed nucleotides were purchased from Epicenter
Biotechnologies, Madison, WI) and CellScript MegaScript™ (Epicenter Biotechnologies, Madison, WI) complete mRNA synthesis kit. The in vitro transcription reaction was run for 4 hours at 37°C. modRNAs incorporating adenosine analogs were poly (A) tailed using yeast Poly (A) Polymerase (Affymetrix, Santa Clara, CA). PCR reaction used HiFi PCR 2X Master Mix™ (Kapa Biosystems, Woburn, MA). modRNAs were post-transcriptionally capped using recombinant Vaccinia Virus Capping Enzyme (New England BioLabs, Ipswich, MA) and a recombinant 2'-0-methyltransferase (Epicenter Biotechnologies, Madison, WI) to generate the 5'-guanosine Capl structure. Cap 2 structure and Cap 3 structures may be generated using additional 2'-0-methyltransferases. The in vitro transcribed mRNA product was run on an agarose gel and visualized. modRNA was purified with Ambion/ Applied Biosystems (Austin, TX) MEGAClear RNA™ purification kit. PCR used PureLink™ PCR purification kit
(Invitrogen, Carlsbad, CA). The product was quantified on Nanodrop™ UV Absorbance (ThermoFisher, Waltham, MA). Quality, UV absorbance quality and visualization of the product was performed on an 1.2% agarose gel. The product was resuspended in TE buffer.
5' Capping Modified Nucleic Acid (mRNA) Structure:
5 '-modRNA capping may be completed concomitantly during the in vzYro-transcription reaction using the following chemical RNA cap analogs to generate the 5'-guanosine cap structure according to manufacturer protocols: 3 '-0-Me-m7G(5')ppp(5')G; G(5')ppp(5')A;
G(5*)ppp(5*)G; m7G(5*)ppp(5*)A; m7G(5*)ppp(5*)G (New England BioLabs, Ipswich, MA). 5'- modRNA capping may be completed post-transcriptionally using a Vaccinia Virus Capping Enzyme to generate the "Cap 0" structure: m7G(5')ppp(5')G (New England BioLabs, Ipswich, MA). Cap 1 structure may be generated using both Vaccinia Virus Capping Enzyme and a 2'-0 methyl-transferase to generate: m7G(5')ppp(5')G-2'-0-methyl. Cap 2 structure may be generated from the Cap 1 structure followed by the 2'-0-methylation of the 5 '-antepenultimate nucleotide using a 2'-0 methyl-transferase. Cap 3 structure may be generated from the Cap 2 structure followed by the 2'-0-methylation of the 5'-preantepenultimate nucleotide using a 2'-0 methyl-transferase. Enzymes are preferably derived from a recombinant source.
Sequences:
G-CSF cDNA:
agcttttggaccctcgtacagaagctaatacgactcactatagggaaataagagagaaaagaagagtaagaagaaatataagag ccaccatggccggtcccgcgacccaaagccccatgaaacttatggccctgcagttgctgctttggcactcggccctctggacagtccaaga agcgactcctctcggacctgcctcatcgttgccgcagtcattccttttgaagtgtctggagcaggtgcgaaagattcagggcgatggagccg cactccaagagaagctctgcgcgacatacaaactttgccatcccgaggagctcgtactgctcgggcacagcttggggattccctgggctcc tctctcgtcctgtccgtcgcaggctttgcagttggcagggtgcctttcccagctccactccggtttgttcttgtatcagggactgctgcaagccc ttgagggaatctcgccagaattgggcccgacgctggacacgttgcagctcgacgtggcggatttcgcaacaaccatctggcagcagatgg aggaactggggatggcacccgcgctgcagcccacgcagggggcaatgccggcctttgcgtccgcgtttcagcgcagggcgggtggagt cctcgtagcgagccaccttcaatcatttttggaagtctcgtaccgggtgctgagacatcttgcgcagccgtgaagcgctgccttctgcggggc ttgccttctggccatgcccttcttctctcccttgcacctgtacctcttggtctttgaataaagcctgagtaggaaggcggccgctcgagcatgca tctagagggcccaattcgccctattcgaagtcg (SEQ ID NO: 1)
G-CSF mRNA:
agcuuuuggacccucguacagaagcuaauacgacucacuauagggaaauaagagagaaaagaagaguaagaagaaau auaagagccaccauggccggucccgcgacccaaagccccaugaaacuuauggcccugcaguugcugcuuuggcacucggcccu cuggacaguccaagaagcgacuccucucggaccugccucaucguugccgcagucauuccuuuugaagugucuggagcaggug cgaaagauucagggcgauggagccgcacuccaagagaagcucugcgcgacauacaaacuuugccaucccgaggagcucguacu gcucgggcacagcuuggggauucccugggcuccucucucguccuguccgucgcaggcuuugcaguuggcagggugccuuuc ccagcuccacuccgguuuguucuuguaucagggacugcugcaagcccuugagggaaucucgccagaauugggcccgacgcug gacacguugcagcucgacguggcggauuucgcaacaaccaucuggcagcagauggaggaacuggggauggcacccgcgcugc agcccacgcagggggcaaugccggccuuugcguccgcguuucagcgcagggcggguggaguccucguagcgagccaccuuca aucauuuuuggaagucucguaccgggugcugagacaucuugcgcagccgugaagcgcugccuucugcggggcuugccuucu ggccaugcccuucuucucucccuugcaccuguaccucuuggucuuugaauaaagccugaguaggaaggcggccgcucgagca ugcaucuagagggcccaauucgcccuauucgaagucg (SEQ ID NO: 2)
G-CSF protein:
MAGPATQSPMKLMALQLLLWHSALWTVQEATPLGPASSLPQSFLLKCLEQVRKI QGDGAALQEKLVSECATYKLCHPEELVLLGHSLGIPWAPLSSCPSQALQLAGCLSQLHS GLFLYQGLLQALEGISPELGPTLDTLQLDVADFATTIWQQMEELGMAPALQPTQGAMPA FASAFQRRAGGVLVASHLQSFLEVSYPvVLRHLAQP (SEQ ID NO: 3) cDNA synthesis primers:
Forward Primer: 5*- TTG GAC CCT CGT ACA GAA GCT AAT ACG (SEQ ID NO: 4) Reverse Primer for template Poly (A) tailing: 5*- T(i20)CT TCC TAC TCA GGC TTT ATT CAA AGA CCA (SEQ ID NO: 5)
Reverse Primer for post-transcriptional Poly (A) Polymerase tailing: 5'- CTT CCT ACT CAG GCT TTA TTC AAA GAC CA (SEQ ID NO: 6)
G-CSF modRNA RT-PCR primers:
Forward Primer: 5*- TGG CCG GTC CCG CGA CCC AA (SEQ ID NO: 7)
Reverse Primer: 5*- GCT TCA CGG CTG CGC AAG AT (SEQ ID NO: 8)
OTHER EMBODIMENTS
It is to be understood that while the invention has been described in conjunction with the detailed description thereof, the foregoing description is intended to illustrate and not limit the scope of the invention, which is defined by the scope of the appended claims. Other aspects, advantages, and modifications are within the scope of the following claims.

Claims

WHAT IS CLAIMED IS:
1. A compound comprising a nucleotide that disrupts binding of a major groove binding partner with a nucleic acid comprising the nucleotide, wherein the nucleotide has decreased binding affinity to the major groove binding partner.
2. The compound of claim 1, wherein the nucleotide comprises a chemical modification located on the major groove face of a nucleobase portion of the nucleotide.
3. The compound of claim 2, wherein the nucleobase portion comprises a pyrimidine nucleobase, and wherein the chemical modification comprises replacing or substituting an atom of the major groove face of the pyrimidine nucleobase with an amine, an SH, a methyl, an ethyl, a chloro or a fluoro group.
4. The compound of claim 2, wherein the chemical modification is located on a sugar portion of the nucleotide.
5. The compound of claim 2, wherein the chemical modification is located on a phosphate backbone of the nucleotide.
6. The compound of claim 1, having Formula I:
Figure imgf000079_0001
wherein:
Z is O or S; each of Y1 is independently selected from -ORal, -NRalRbl, and -SRal;
each of Y2 is independently selected from O, NRa, S or a linker comprising an atom selected from the group consisting of C, O, N, and S;
each of Y3 is independently selected from O and S;
Y4 is selected from H, -ORa, -SRa, and -NHRa;
n is 0, 1, 2, or 3;
m is 0, 1, 2 or 3;
B is a nucleobase;
Ra is H, Ci-20 alkyl, C2-2o alkenyl, C2-2o alkynyl, or C6-2o aryl;
Ral and Rbl are each independently H or a counterion; and
-Y3-Rcl is OH or SH at a pH of about 1 or -Y3-Rcl is O" or S" at physiological pH; or -Y3-Rcl is Ci-20 alkoxy, C2-2o -O-alkenyl, or Ci-2o -O-alkynyl;
wherein when B is an unmodified nucleobase selected from cytosine, guanine, uracil and adenine, then at least one of Z, Y1 or Y2 is not O or OH.
The compound of claim 6, wherein B is a nucleobase of Formula Il-a, Il-b, or II-c:
R
Figure imgf000080_0001
Il-a Il-b II-c
wherein:
^ denotes a single or double bond;
X is O or S;
V, U and W are each independently C or N;
wherein when V is C then R1 is H, Ci_6 alkyl, Ci_6 alkenyl, Ci_6 alkynyl, halo, or -ORc, wherein Ci_6 alkyl, Ci_6 alkenyl, Ci_6 alkynyl are each optionally substituted with -OH, -NRaRb, SH, -C(0)Rc, -C(0)ORc, -NHC(0)Rc, or -NHC(0)ORc;
and wherein when V is N then R1 is absent;
R2 is H, -ORc, -SRC, -NRaRb, or halo; or when V is C then R1 and R2 together with the carbon atoms to which they are attached can form a 5- or 6-membered ring optionally substituted with 1-4 substituents selected from halo, -OH, -SH, -NRaRb, Ci-6 alkyl, Ci-6 alkenyl, Ci-6 alkynyl, Ci-6 alkoxy, or Ci-6 thioalkyl;
R3 is H or Ci-6 alkyl;
R4 is H or Ci-6 alkyl; wherein when ^ denotes a double bond then R4 is absent, or N-R4, taken together, forms a positively charged N substituted with Ci_6 alkyl;
Ra and Rb are each independently H, Ci_6 alkyl, Ci_6 alkenyl, Ci_6 alkynyl, or C6-io aryl; and
Rc is H, Ci-6 alkyl, Ci_6 alkenyl, phenyl, benzyl, a polyethylene glycol group, or an amino-polyethylene glycol group.
8. The compound of claim 7, wherein B is a nucleobase of Formula Il-al , II-a2, II-a3, II-a4, or II-a5:
Figure imgf000081_0001
Il-al II-a2 II-a3 II-a4 II-a5.
9. The compound of claim 6, wherein B is a nucleobase selected from the group consisting of cytosine, guanine, adenine, and uracil.
The compound of claim 6, having Formula I-
Figure imgf000081_0002
I-a.
11. The compound of claim 6, having Formula I-b:
Figure imgf000082_0001
I-b.
12. The compound of claim 6, having Formula I-c:
Figure imgf000082_0002
I-c.
Figure imgf000082_0003
Figure imgf000083_0001
15. A nucleic acid sequence comprising at least two nucleotides, the nucleic acid sequence comprising a nucleotide that disrupts binding of a major groove binding partner with the nucleic acid sequence, wherein the nucleotide has decreased binding affinity to the major groove binding partner.
The nucleic acid sequence of claim 15 comprising a compound of Formula I-d:
Figure imgf000083_0002
wherein:
Z is O or S;
each of Y1 is independently selected from -ORal, -NRalRbl, and -SRal; each of Y2 is independently selected from O, NRa, S or a linker comprising an atom selected from the group consisting of C, O, N, and S;
B is a nucleobase;
Ra is H, Ci-6 alkyl, C1-6 alkenyl, C1-6 alkynyl, or C6-10 aryl; and
Ral and Rbl are each independently H or a counterion; and
-ORcl is OH at a pH of about 1 or -ORcl is 0~ at physiological pH;
wherein when B is an unmodified nucleobase selected from cytosine, guanine, uracil and adenine, then at least one of Z, Y1 or Y2 is not O or OH.
17. The nucleic acid sequence of claim 16, wherein B is a nucleobase of Formula Il-a, Il-b, or II-c:
Figure imgf000084_0001
Il-a Il-b II-c
wherein:
^ denotes a single or double bond;
X is O or S;
V, U and W are each independently C or N;
wherein when V is C then R1 is H, C1-6 alkyl, C1-6 alkenyl, C1-6 alkynyl, halo, or -ORc, wherein C1-6 alkyl, C1-6 alkenyl, C1-6 alkynyl are each optionally substituted with -OH, -NRaRb, - SH, -C(0)Rc, -C(0)ORc, -NHC(0)Rc, or -NHC(0)ORc;
and wherein when V is N then R1 is absent;
R2 is H, -ORc, -SRC, -NRaRb, or halo;
or when V is C then R1 and R2 together with the carbon atoms to which they are attached can form a 5- or 6-membered ring optionally substituted with 1-4 substituents selected from halo, -OH, -SH, -NRaRb, Ci-6 alkyl, Ci-6 alkenyl, Ci-6 alkynyl, Ci-6 alkoxy, or Ci-6 thioalkyl;
R3 is H or Ci_6 alkyl;
R4 is H or C1-6 alkyl; wherein when ^ denotes a double bond then R4 is absent, or N-R4, taken together, forms a positively charged N substituted with C1-6 alkyl; Ra and Rb are each independently H, C1-6 alkyl, C1-6 alkenyl, C1-6 alkynyl, or C6-10 aryl; and
Rc is H, Ci-6 alkyl, C1-6 alkenyl, phenyl, benzyl, a polyethylene glycol group, or an amino-polyethylene glycol group.
18. The nucleic acid sequence of claim 17, wherein B is a nucleobase of Formula Il-al, II- II-a3, II-a4, -a5:
Figure imgf000085_0001
Il-al II-a2 II-a3 II-a4 II-a5.
19. The nucleic acid sequence of claim 16, wherein B is a nucleobase selected from the group consisting of cytosine, guanine, adenine, and uracil.
20. The nucleic acid sequence of claim 16, wherein the nucleic acid sequence contains a plurality of structurally unique compounds of Formula I-d.
21. The nucleic acid sequence of claim 16, wherein at least 25% of the cytosines are replaced by a compound of Formula I-d and/or wherein at least 25% of the uracils are replaced by a compound of Formula I-d.
22. The compound of claim 1, wherein the major groove interacting partners are selected from the group consisting of: TLRs (Toll-like Receptors) 3, 7, and 8; RIG-I (retinoic acid- inducible gene I); MDA5 (melanoma differentiation-associated gene 5); laboratory of genetics and physiology 2 (LGP2); ΗΓΝ-200 domain containing proteins; and Helicase-domain containing proteins.
23. A non-naturally occurring nucleotide comprising one or more chemical modifications of a naturally occurring nucleotide, wherein the nucleotide reduces the induction of the cellular innate immune response of a cell to a modified nucleic acid comprising the non-naturally occurring nucleotide when the modified nucleic acid is introduced into the cell, as compared to the induction of the cellular innate immune in a cell induced by a corresponding unmodified nucleic acid.
24. The compound of claim 23, wherein the nucleotide reduces the innate immune response or the secretion of pro-inflammatory cytokines or both by at least about 10%.
25. The compound of claim 23, wherein the nucleotide reduces the innate immune response or the secretion of pro-inflammatory cytokines or both by about 75%.
26. The compound of claim 23, wherein the nucleotide reduces the innate immune response or the secretion of pro-inflammatory cytokines or both by at least 90%.
27. The compound of claim 23, further comprising a translateable region encoding a protein of interest.
28. A composition comprising the compound of claim 27, in an amount sufficient to increase the production of the protein of interest when introduced into a target cell, as compared to the amount of protein produced in a cell containing a corresponding unmodified nucleic acid encoding the protein of interest.
29. The composition of claim 28, wherein the increase is at least about 10%.
30. The composition of claim 28, wherein the increase is at least about 50%.
31. The composition of claim 28, wherein the increase is at least about 100%.
32. The composition of claim 28, wherein at least 25% of the cytosines in the nucleic acid are replaced by a compound of Formula I-d.
33. The composition of claim 28, wherein at least 90% of the cytosines in the nucleic acid are replaced by a compound of Formula I-d.
34. The composition of claim 28, wherein about 100% of the cytosines in the nucleic acid are replaced by a compound of Formula I-d.
35. The composition of claim 28, wherein at least 25% of the uracils in the nucleic acid are replaced by a compound of Formula I-d.
36. The composition of claim 28, wherein at least 90% of the uracils in the nucleic acid are replaced by a compound of Formula I-d.
37. The composition of claim 28, wherein about 100% of the uracils in the nucleic acid are replaced by a compound of Formula I-d.
38. The composition of claim 28, wherein at least 25% of the cytosines in the nucleic acid are replaced by a first compound of Formula I-d and wherein at least 25% of the uracils in the nucleic acid are replaced by a second compound of Formula I-d.
39. The composition of claim 28, wherein about 100% of the cytosines in the nucleic acid are replaced by a first compound of Formula I-d and wherein about 100% of the uracils in the nucleic acid are replaced by a second compound of Formula I-d.
40. The composition of claim 28, further comprising an RNA polymerase, a cDNA template, or a combination thereof.
41. The composition of claim 40, further comprising a nucleotide selected from the group consisting of adenosine, cytosine, guanosine, and uracil.
42. A method of preparing a nucleic acid sequence comprising a nucleotide that disrupts binding of a major groove binding partner with the nucleic acid sequence, wherein the nucleic acid sequence comprises a compoun f Formula I-d:
Figure imgf000088_0001
I-d
wherein:
the nucleotide has decreased binding affinity to the major groove binding partner;
Z is O or S;
each of Y1 is independently selected from -ORal, -NRalRbl, and -SRal;
each of Y2 is independently selected from O, NRa, S or a linker comprising an atom selected from the group consisting of C, O, N, and S;
B is a nucleobase; and
Ral and Rbl are each independently H or a counterion; and
-ORcl is OH at a pH of about 1 or -ORcl is 0~ at physiological pH;
wherein when B is an unmodified nucleobase selected from cytosine, guanine, uracil and adenine, then at least one of Z, Y1 or Y2 is not O or OH;
the method comprising:
reacting a compound of Formula I-c:
Figure imgf000088_0002
I-c
with an RNA polymerase, and a cDNA template.
43. The method of claim 42, wherein the reaction is repeated from 1 to about 7,000 times.
44. The method of claim 42, wherein B is a nucleobase of Formula Il-a, Il-b, or II-c:
R
Figure imgf000089_0001
Il-a Il-b II-c
wherein:
^ denotes a single or double bond;
X is O or S;
V, U and W are each independently C or N;
wherein when V is C then R1 is H, Ci_6 alkyl, Ci_6 alkenyl, Ci_6 alkynyl, halo, or -ORc, wherein Ci-6 alkyl, Ci-6 alkenyl, Ci-6 alkynyl are each optionally substituted with -OH, -NRaRb, - SH, -C(0)Rc, -C(0)ORc, -NHC(0)Rc, or -NHC(0)ORc;
and wherein when V is N then R1 is absent;
R2 is H, -ORc, -SRC, -NRaRb, or halo;
or when V is C then R1 and R2 together with the carbon atoms to which they are attached can form a 5- or 6-membered ring optionally substituted with 1-4 substituents selected from halo, -OH, -SH, -NRaRb, Ci-6 alkyl, Ci-6 alkenyl, Ci-6 alkynyl, Ci-6 alkoxy, or Ci-6 thioalkyl;
R3 is H or Ci-6 alkyl;
R4 is H or Ci-6 alkyl; wherein when ^ denotes a double bond then R4 is absent, or N-R4, taken together, forms a positively charged N substituted with C1-6 alkyl;
Ra and Rb are each independently H, C1-6 alkyl, C1-6 alkenyl, C1-6 alkynyl, or C6-10 aryl; and
Rc is H, Ci-6 alkyl, C1-6 alkenyl, phenyl, benzyl, a polyethylene glycol group, or an amino-polyethylene glycol group.
45. The method of claim 44, wherein B is a nucleobase of Formula Il-al, II-a2, II-a3, II-a4, or II-a5:
Figure imgf000089_0002
II-al II-a2 II-a3 II-a4 II-a5.
46. A method of amplifying a nucleic acid sequence comprising a nucleotide that disrupts binding of a major groove binding partner with the nucleic acid sequence, the method comprising:
reacting a compound of Formula I-c:
Figure imgf000090_0001
I-c
wherein:
the nucleotide has decreased binding affinity to the major groove binding partner;
Z is O or S;
each of Y1 is independently selected from -ORal, -NRalRbl, and -SRal;
each of Y2 is independently selected from O, NRa, S or a linker comprising an atom selected from the group consisting of C, O, N, and S;
B is a nucleobase; and
Ral and Rbl are each independently H or a counterion; and
-ORcl is OH at a pH of about 1 or -ORcl is 0~ at physiological pH;
wherein when B is an unmodified nucleobase selected from cytosine, guanine, uracil and adenine, then at least one of Z, Y1 or Y2 is not O or OH;
with a primer, a cDNA template, and an RNA polymerase.
47. The method of claim 46, wherein B is a nucleobase of Formula Il-a, Il-b, or II-c:
Rl
Figure imgf000090_0002
wherein: denotes a single or double bond;
X is O or S;
V, U and W are each independently C or N;
wherein when V is C then R1 is H, C1-6 alkyl, C1-6 alkenyl, C1-6 alkynyl, halo, or -ORc, wherein C1-6 alkyl, C1-6 alkenyl, C1-6 alkynyl are each optionally substituted with -OH, -NRaRb, - SH, -C(0)Rc, -C(0)ORc, -NHC(0)Rc, or -NHC(0)ORc;
and wherein when V is N then R1 is absent;
R2 is H, -ORc, -SRC, -NRaRb, or halo;
or when V is C then R1 and R2 together with the carbon atoms to which they are attached can form a 5- or 6-membered ring optionally substituted with 1-4 substituents selected from halo, -OH, -SH, -NRaRb, Ci-6 alkyl, Ci-6 alkenyl, Ci-6 alkynyl, Ci-6 alkoxy, or Ci-6 thioalkyl;
R3 is H or Ci_6 alkyl;
R4 is H or C1-6 alkyl; wherein when ^ denotes a double bond then R4 is absent, or N-R4, taken together, forms a positively charged N substituted with C1-6 alkyl;
Ra and Rb are each independently H, C1-6 alkyl, C1-6 alkenyl, C1-6 alkynyl, or C6-10 aryl; and
Rc is H, Ci-6 alkyl, C1-6 alkenyl, phenyl, benzyl, a polyethylene glycol group, or an amino-polyethylene glycol group.
48. The method of claim 47, wherein B is a nucleobase of Formula Il-al, II-a2, II-a3, II-a4, or II-a5:
Figure imgf000091_0001
Il-al II-a2 II-a3 II-a4 II-a5.
49. A method of synthesizing a pharmaceutical nucleic acid, comprising the steps of:
a) providing a complementary deoxyribonucleic acid (cDNA) that encodes a pharmaceutical protein of interest; b) selecting a nucleotide that is known to disrupt a binding of a major groove binding partner with a nucleic acid, wherein the nucleotide has decreased binding affinity to the major groove binding partner; and
c) contacting the provided cDNA and the selected nucleotide with an RNA polymerase, under conditions such that the pharmaceutical nucleic is synthesized.
50. The method of claim 49, wherein the pharmaceutical nucleic acid is a ribonucleic acid (RNA).
51. A method of inducing a physiological change in a target cell population, comprising the steps of:
a) providing a first nucleic acid comprising i) a translatable region encoding a protein of interest and ii) a nucleic acid modification, wherein the first nucleic acid is substantially resistant to cellular degradation; and
b) contacting an effective amount of the first nucleic acid to a producer cell under conditions such that the protein of interest is produced in the producer cell and secreted therefrom, wherein the secreted protein of interest contacts the target cell population and induces a physiological change therein.
52. The method of claim 51 , wherein the protein of interest is capable of binding to a receptor on the surface of at least one cell present in the target cell population.
53. The method of claim 51 , wherein the secreted protein is capable of interacting with a receptor on the surface of at least one cell present in the target cell population.
54. The method of claim 53, wherein the secreted protein is Granulocyte-Colony Stimulating Factor (G-CSF).
55. The method of claim 53, wherein the target cell population comprises one or more cells that express the G-CSF receptor.
56. A compound comprising a nucleic acid comprising one or more nucleotides having Formula I:
Figure imgf000093_0001
I
wherein:
Z is O or S;
each of Y1 is independently selected from -ORal, -NRalRbl, and -SRal;
each of Y2 is independently selected from O, NRa, S or a linker comprising an atom selected from the group consisting of C, O, N, and S;
each of Y3 is independently selected from O and S;
Y4 is selected from H, -ORa, -SRa, and -NHRa;
n is 0, 1 , 2, or 3;
m is 0, 1 , 2 or 3;
B is a nucleobase;
Ra is H, Ci-20 alkyl, C2-2o alkenyl, C2-2o alkynyl, or C6-2o aryl;
Ral and Rbl are each independently H or a counterion; and
-Y3-Rcl is OH or SH at a pH of about 1 or -Y3-Rcl is O" or S" at physiological pH; or -Y3-Rcl is Ci-20 alkoxy, C2-2o -O-alkenyl, or Ci-2o -O-alkynyl;
wherein when B is an unmodified nucleobase selected from cytosine, guanine, uracil and adenine, then at least one of Z, Y1 or Y2 is not O or OH; and
wherein a cell comprising the nucleic acid is characterized by:
i) decreased cellular secretion of s pro-inflammatory cytokine;
ii) decreased activation of a cellular innate immune responder;
iii) decreased suspectibility to a cellular nuclease;
iv) decreased binding to a negative regulator of gene expression; decreased binding to a nucleic acid; increased protein translation efficiency; increased half-life;
or a combination thereof.
PCT/US2011/054617 2010-10-01 2011-10-03 Modified nucleosides, nucleotides, and nucleic acids, and uses thereof WO2012045075A1 (en)

Priority Applications (5)

Application Number Priority Date Filing Date Title
EP22173763.8A EP4108671A1 (en) 2010-10-01 2011-10-03 Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
EP19177059.3A EP3590949B1 (en) 2010-10-01 2011-10-03 Ribonucleic acids containing n1-methyl-pseudouracils and uses thereof
CA2813466A CA2813466A1 (en) 2010-10-01 2011-10-03 Modified nucleosides, nucleotides, and nucleic acids that disrupt major groove binding partner interactions
EP11830061.5A EP2622064B1 (en) 2010-10-01 2011-10-03 Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
ES11830061T ES2737960T3 (en) 2010-10-01 2011-10-03 Nucleosides, nucleotides and modified nucleic acids and their uses

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US40441310P 2010-10-01 2010-10-01
US61/404,413 2010-10-01

Publications (1)

Publication Number Publication Date
WO2012045075A1 true WO2012045075A1 (en) 2012-04-05

Family

ID=45893552

Family Applications (2)

Application Number Title Priority Date Filing Date
PCT/US2011/054636 WO2012045082A2 (en) 2010-10-01 2011-10-03 Engineered nucleic acids and methods of use thereof
PCT/US2011/054617 WO2012045075A1 (en) 2010-10-01 2011-10-03 Modified nucleosides, nucleotides, and nucleic acids, and uses thereof

Family Applications Before (1)

Application Number Title Priority Date Filing Date
PCT/US2011/054636 WO2012045082A2 (en) 2010-10-01 2011-10-03 Engineered nucleic acids and methods of use thereof

Country Status (24)

Country Link
US (11) US20120237975A1 (en)
EP (7) EP2625189B1 (en)
JP (1) JP2013543381A (en)
CN (3) CN104531671A (en)
AU (2) AU2011308496A1 (en)
BR (1) BR112013007862A2 (en)
CA (3) CA3162352A1 (en)
DE (1) DE19177059T1 (en)
DK (1) DK3590949T3 (en)
ES (3) ES2737960T3 (en)
HR (1) HRP20220796T1 (en)
HU (1) HUE058896T2 (en)
IL (1) IL225493A0 (en)
LT (1) LT3590949T (en)
MX (1) MX2013003681A (en)
NZ (1) NZ608972A (en)
PL (1) PL3590949T3 (en)
PT (1) PT3590949T (en)
RS (1) RS63430B1 (en)
RU (1) RU2013120302A (en)
SG (2) SG190679A1 (en)
SI (1) SI3590949T1 (en)
WO (2) WO2012045082A2 (en)
ZA (2) ZA201303161B (en)

Cited By (97)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8664194B2 (en) 2011-12-16 2014-03-04 Moderna Therapeutics, Inc. Method for producing a protein of interest in a primate
US8710200B2 (en) 2011-03-31 2014-04-29 Moderna Therapeutics, Inc. Engineered nucleic acids encoding a modified erythropoietin and their expression
WO2014081507A1 (en) 2012-11-26 2014-05-30 Moderna Therapeutics, Inc. Terminally modified rna
WO2014093924A1 (en) 2012-12-13 2014-06-19 Moderna Therapeutics, Inc. Modified nucleic acid molecules and uses thereof
WO2014113089A2 (en) 2013-01-17 2014-07-24 Moderna Therapeutics, Inc. Signal-sensor polynucleotides for the alteration of cellular phenotypes
US8822663B2 (en) 2010-08-06 2014-09-02 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
DE102013005361A1 (en) 2013-03-28 2014-10-02 Eberhard Karls Universität Tübingen Medizinische Fakultät polyribonucleotide
WO2014159813A1 (en) 2013-03-13 2014-10-02 Moderna Therapeutics, Inc. Long-lived polynucleotide molecules
US8853377B2 (en) 2010-11-30 2014-10-07 Shire Human Genetic Therapies, Inc. mRNA for use in treatment of human genetic diseases
WO2015034925A1 (en) 2013-09-03 2015-03-12 Moderna Therapeutics, Inc. Circular polynucleotides
US8980864B2 (en) 2013-03-15 2015-03-17 Moderna Therapeutics, Inc. Compositions and methods of altering cholesterol levels
US8999380B2 (en) 2012-04-02 2015-04-07 Moderna Therapeutics, Inc. Modified polynucleotides for the production of biologics and proteins associated with human disease
US9107886B2 (en) 2012-04-02 2015-08-18 Moderna Therapeutics, Inc. Modified polynucleotides encoding basic helix-loop-helix family member E41
US9181321B2 (en) 2013-03-14 2015-11-10 Shire Human Genetic Therapies, Inc. CFTR mRNA compositions and related methods and uses
US9283287B2 (en) 2012-04-02 2016-03-15 Moderna Therapeutics, Inc. Modified polynucleotides for the production of nuclear proteins
US9308281B2 (en) 2011-06-08 2016-04-12 Shire Human Genetic Therapies, Inc. MRNA therapy for Fabry disease
US9334328B2 (en) 2010-10-01 2016-05-10 Moderna Therapeutics, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US9404127B2 (en) 2010-06-30 2016-08-02 Protiva Biotherapeutics, Inc. Non-liposomal systems for nucleic acid delivery
US9428535B2 (en) 2011-10-03 2016-08-30 Moderna Therapeutics, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US9464124B2 (en) 2011-09-12 2016-10-11 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US9512456B2 (en) 2012-08-14 2016-12-06 Modernatx, Inc. Enzymes and polymerases for the synthesis of RNA
US9522176B2 (en) 2013-10-22 2016-12-20 Shire Human Genetic Therapies, Inc. MRNA therapy for phenylketonuria
US9572897B2 (en) 2012-04-02 2017-02-21 Modernatx, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
EP3052106A4 (en) * 2013-09-30 2017-07-19 ModernaTX, Inc. Polynucleotides encoding immune modulating polypeptides
WO2017127750A1 (en) 2016-01-22 2017-07-27 Modernatx, Inc. Messenger ribonucleic acids for the production of intracellular binding polypeptides and methods of use thereof
US9751925B2 (en) 2014-11-10 2017-09-05 Modernatx, Inc. Alternative nucleic acid molecules containing reduced uracil content and uses thereof
US9850269B2 (en) 2014-04-25 2017-12-26 Translate Bio, Inc. Methods for purification of messenger RNA
WO2018009838A1 (en) 2016-07-07 2018-01-11 Rubius Therapeutics, Inc. Compositions and methods related to therapeutic cell systems expressing exogenous rna
US9872900B2 (en) 2014-04-23 2018-01-23 Modernatx, Inc. Nucleic acid vaccines
US9957499B2 (en) 2013-03-14 2018-05-01 Translate Bio, Inc. Methods for purification of messenger RNA
WO2018081459A1 (en) 2016-10-26 2018-05-03 Modernatx, Inc. Messenger ribonucleic acids for enhancing immune responses and methods of use thereof
WO2018144775A1 (en) 2017-02-01 2018-08-09 Modernatx, Inc. Immunomodulatory therapeutic mrna compositions encoding activating oncogene mutation peptides
US10093706B2 (en) 2017-01-30 2018-10-09 Indiana University Research And Technology Corporation Dominant positive hnRNP-E1 polypeptide compositions and methods
US10106800B2 (en) 2005-09-28 2018-10-23 Biontech Ag Modification of RNA, producing an increased transcript stability and translation efficiency
WO2018213789A1 (en) 2017-05-18 2018-11-22 Modernatx, Inc. Modified messenger rna comprising functional rna elements
US10143758B2 (en) 2009-12-01 2018-12-04 Translate Bio, Inc. Liver specific delivery of messenger RNA
US10155031B2 (en) 2012-11-28 2018-12-18 Biontech Rna Pharmaceuticals Gmbh Individualized vaccines for cancer
US10258698B2 (en) 2013-03-14 2019-04-16 Modernatx, Inc. Formulation and delivery of modified nucleoside, nucleotide, and nucleic acid compositions
US10323076B2 (en) 2013-10-03 2019-06-18 Modernatx, Inc. Polynucleotides encoding low density lipoprotein receptor
WO2019152557A1 (en) 2018-01-30 2019-08-08 Modernatx, Inc. Compositions and methods for delivery of agents to immune cells
WO2019200171A1 (en) 2018-04-11 2019-10-17 Modernatx, Inc. Messenger rna comprising functional rna elements
WO2019204743A1 (en) 2018-04-19 2019-10-24 Checkmate Pharmaceuticals, Inc. Synthetic rig-i-like receptor agonists
US10485884B2 (en) 2012-03-26 2019-11-26 Biontech Rna Pharmaceuticals Gmbh RNA formulation for immunotherapy
WO2020056304A1 (en) 2018-09-14 2020-03-19 Modernatx, Inc. Methods and compositions for treating cancer using mrna therapeutics
WO2020097409A2 (en) 2018-11-08 2020-05-14 Modernatx, Inc. Use of mrna encoding ox40l to treat cancer in human patients
US10738355B2 (en) 2011-05-24 2020-08-11 Tron-Translationale Onkologie An Der Universitätsmedizin Der Johannes Gutenberg-Universität Mainz Ggmbh Individualized vaccines for cancer
WO2020227510A1 (en) 2019-05-07 2020-11-12 Modernatx, Inc. Polynucleotides for disrupting immune cell activity and methods of use thereof
WO2020227537A1 (en) 2019-05-07 2020-11-12 Modernatx, Inc Differentially expressed immune cell micrornas for regulation of protein expression
US10849920B2 (en) 2015-10-05 2020-12-01 Modernatx, Inc. Methods for therapeutic administration of messenger ribonucleic acid drugs
WO2020263985A1 (en) 2019-06-24 2020-12-30 Modernatx, Inc. Messenger rna comprising functional rna elements and uses thereof
WO2020263883A1 (en) 2019-06-24 2020-12-30 Modernatx, Inc. Endonuclease-resistant messenger rna and uses thereof
WO2021007515A1 (en) 2019-07-11 2021-01-14 Tenaya Therapeutics, Inc. Cardiac cell reprogramming with micrornas and other factors
WO2021050986A1 (en) 2019-09-11 2021-03-18 Modernatx, Inc. Lnp-formulated mrna therapeutics and use thereof for treating human subjects
WO2021081353A1 (en) 2019-10-23 2021-04-29 Checkmate Pharmaceuticals, Inc. Synthetic rig-i-like receptor agonists
US11015211B2 (en) 2018-08-30 2021-05-25 Tenaya Therapeutics, Inc. Cardiac cell reprogramming with myocardin and ASCL1
WO2021178246A1 (en) 2020-03-02 2021-09-10 Tenaya Therapeutics, Inc. Gene vector control by cardiomyocyte-expressed micrornas
US11156617B2 (en) 2015-02-12 2021-10-26 BioNTech RNA Pharmaceuticals GbmH Predicting T cell epitopes useful for vaccination
US11174500B2 (en) 2018-08-24 2021-11-16 Translate Bio, Inc. Methods for purification of messenger RNA
US11173120B2 (en) 2014-09-25 2021-11-16 Biontech Rna Pharmaceuticals Gmbh Stable formulations of lipids and liposomes
US11173190B2 (en) 2017-05-16 2021-11-16 Translate Bio, Inc. Treatment of cystic fibrosis by delivery of codon-optimized mRNA encoding CFTR
WO2021243207A1 (en) 2020-05-28 2021-12-02 Modernatx, Inc. Use of mrnas encoding ox40l, il-23 and il-36gamma for treating cancer
US11222711B2 (en) 2013-05-10 2022-01-11 BioNTech SE Predicting immunogenicity of T cell epitopes
US11224642B2 (en) 2013-10-22 2022-01-18 Translate Bio, Inc. MRNA therapy for argininosuccinate synthetase deficiency
WO2022032154A2 (en) 2020-08-06 2022-02-10 Modernatx, Inc. Compositions for the delivery of payload molecules to airway epithelium
US11254936B2 (en) 2012-06-08 2022-02-22 Translate Bio, Inc. Nuclease resistant polynucleotides and uses thereof
US11253605B2 (en) 2017-02-27 2022-02-22 Translate Bio, Inc. Codon-optimized CFTR MRNA
EP3971287A1 (en) 2013-07-11 2022-03-23 ModernaTX, Inc. Compositions comprising synthetic polynucleotides encoding crispr related proteins and synthetic sgrnas and methods of use
US11298426B2 (en) 2003-10-14 2022-04-12 BioNTech SE Recombinant vaccines and use thereof
EP3718565B1 (en) 2015-10-22 2022-04-27 ModernaTX, Inc. Respiratory virus vaccines
US20220125723A1 (en) 2010-07-06 2022-04-28 Glaxosmithkline Biologicals Sa Lipid formulations with viral immunogens
US11492628B2 (en) 2015-10-07 2022-11-08 BioNTech SE 3′-UTR sequences for stabilization of RNA
US11547673B1 (en) 2020-04-22 2023-01-10 BioNTech SE Coronavirus vaccine
WO2023009421A1 (en) 2021-07-26 2023-02-02 Modernatx, Inc. Processes for preparing lipid nanoparticle compositions
WO2023009422A1 (en) 2021-07-26 2023-02-02 Modernatx, Inc. Processes for preparing lipid nanoparticle compositions for the delivery of payload molecules to airway epithelium
US11596645B2 (en) 2010-07-06 2023-03-07 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11597744B2 (en) 2017-06-30 2023-03-07 Sirius Therapeutics, Inc. Chiral phosphoramidite auxiliaries and methods of their use
WO2023064469A1 (en) 2021-10-13 2023-04-20 Modernatx, Inc. Compositions of mrna-encoded il15 fusion proteins and methods of use thereof
WO2023069498A1 (en) 2021-10-22 2023-04-27 Senda Biosciences, Inc. Mrna vaccine composition
US11639370B2 (en) 2010-10-11 2023-05-02 Glaxosmithkline Biologicals Sa Antigen delivery platforms
WO2023086465A1 (en) 2021-11-12 2023-05-19 Modernatx, Inc. Compositions for the delivery of payload molecules to airway epithelium
US11655475B2 (en) 2010-07-06 2023-05-23 Glaxosmithkline Biologicals Sa Immunisation of large mammals with low doses of RNA
WO2023096858A1 (en) 2021-11-23 2023-06-01 Senda Biosciences, Inc. A bacteria-derived lipid composition and use thereof
WO2023122080A1 (en) 2021-12-20 2023-06-29 Senda Biosciences, Inc. Compositions comprising mrna and lipid reconstructed plant messenger packs
WO2023154818A1 (en) 2022-02-09 2023-08-17 Modernatx, Inc. Mucosal administration methods and formulations
US11744801B2 (en) 2017-08-31 2023-09-05 Modernatx, Inc. Methods of making lipid nanoparticles
US11759422B2 (en) 2010-08-31 2023-09-19 Glaxosmithkline Biologicals Sa Pegylated liposomes for delivery of immunogen-encoding RNA
WO2023196988A1 (en) 2022-04-07 2023-10-12 Modernatx, Inc. Methods of use of mrnas encoding il-12
US11786607B2 (en) 2017-06-15 2023-10-17 Modernatx, Inc. RNA formulations
WO2023199113A1 (en) 2022-04-15 2023-10-19 Smartcella Solutions Ab COMPOSITIONS AND METHODS FOR EXOSOME-MEDIATED DELIVERY OF mRNA AGENTS
US11865159B2 (en) 2017-02-28 2024-01-09 Sanofi Therapeutic RNA
US11878055B1 (en) 2022-06-26 2024-01-23 BioNTech SE Coronavirus vaccine
US11896636B2 (en) 2011-07-06 2024-02-13 Glaxosmithkline Biologicals Sa Immunogenic combination compositions and uses thereof
US11958891B2 (en) 2017-01-26 2024-04-16 Surrozen Operating, Inc. Tissue-specific Wnt signal enhancing molecules and uses thereof
WO2024044741A3 (en) * 2022-08-26 2024-04-18 Trilink Biotechnologies, Llc Efficient method for making highly purified 5'-capped oligonucleotides
US11981703B2 (en) 2016-08-17 2024-05-14 Sirius Therapeutics, Inc. Polynucleotide constructs
WO2024102434A1 (en) 2022-11-10 2024-05-16 Senda Biosciences, Inc. Rna compositions comprising lipid nanoparticles or lipid reconstructed natural messenger packs
US11993645B2 (en) 2018-01-11 2024-05-28 The Board Of Trustees Of The Leland Stanford Junior University Compositions comprising R-Spondin (RSPO) surrogate molecules

Families Citing this family (92)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9012219B2 (en) 2005-08-23 2015-04-21 The Trustees Of The University Of Pennsylvania RNA preparations comprising purified modified RNA for reprogramming cells
KR102171849B1 (en) 2009-12-07 2020-10-30 더 트러스티스 오브 더 유니버시티 오브 펜실바니아 Rna preparations comprising purified modified rna for reprogramming cells
EP3235508B1 (en) * 2011-03-16 2020-12-30 Sanofi Compositions comprising a dual v region antibody-like protein
EP2788033B1 (en) 2011-12-05 2017-05-31 Factor Bioscience Inc. Methods and products for transfecting cells
KR20140116095A (en) 2011-12-12 2014-10-01 더 트러스티스 오브 더 유니버시티 오브 펜실바니아 Proteins comprising mrsa pbp2a and fragments thereof, nucleic acids encoding the same, and compositions and their use to prevent and treat mrsa infections
WO2013185069A1 (en) 2012-06-08 2013-12-12 Shire Human Genetic Therapies, Inc. Pulmonary delivery of mrna to non-lung target cells
JP6510416B2 (en) 2012-11-01 2019-05-08 ファクター バイオサイエンス インコーポレイテッド Methods and products for expressing proteins in cells
WO2014124457A1 (en) * 2013-02-11 2014-08-14 University Of Louisville Research Foundation, Inc. Methods for producing antibodies
LT2970456T (en) * 2013-03-14 2021-08-10 Translate Bio, Inc. Methods and compositions for delivering mrna coded antibodies
WO2014152031A1 (en) 2013-03-15 2014-09-25 Moderna Therapeutics, Inc. Ribonucleic acid purification
WO2014144196A1 (en) 2013-03-15 2014-09-18 Shire Human Genetic Therapies, Inc. Synergistic enhancement of the delivery of nucleic acids via blended formulations
WO2014152030A1 (en) 2013-03-15 2014-09-25 Moderna Therapeutics, Inc. Removal of dna fragments in mrna production process
WO2014152027A1 (en) 2013-03-15 2014-09-25 Moderna Therapeutics, Inc. Manufacturing methods for production of rna transcripts
WO2014144767A1 (en) 2013-03-15 2014-09-18 Moderna Therapeutics, Inc. Ion exchange purification of mrna
US10385088B2 (en) 2013-10-02 2019-08-20 Modernatx, Inc. Polynucleotide molecules and uses thereof
CN105658800A (en) 2013-10-22 2016-06-08 夏尔人类遗传性治疗公司 CNS delivery of MRNA and uses thereof
CN112618732A (en) 2013-10-22 2021-04-09 夏尔人类遗传性治疗公司 Lipid formulations for delivery of messenger RNA
JP2017500865A (en) 2013-12-19 2017-01-12 ノバルティス アーゲー Compositions and formulations of leptin mRNA
MX2016009771A (en) 2014-01-31 2016-11-14 Factor Bioscience Inc Methods and products for nucleic acid production and delivery.
JP6557722B2 (en) 2014-05-30 2019-08-07 シャイアー ヒューマン ジェネティック セラピーズ インコーポレイテッド Biodegradable lipids for delivery of nucleic acids
US10286086B2 (en) 2014-06-19 2019-05-14 Modernatx, Inc. Alternative nucleic acid molecules and uses thereof
CN106795142B (en) 2014-06-24 2022-11-04 川斯勒佰尔公司 Stereochemically enriched compositions for delivery of nucleic acids
AU2015283954B2 (en) 2014-07-02 2020-11-12 Translate Bio, Inc. Encapsulation of messenger RNA
CA2955238A1 (en) 2014-07-16 2016-01-21 Moderna Therapeutics, Inc. Circular polynucleotides
GB201418965D0 (en) * 2014-10-24 2014-12-10 Ospedale San Raffaele And Fond Telethon
JP6767976B2 (en) 2014-12-05 2020-10-14 トランスレイト バイオ, インコーポレイテッド Messenger RNA therapy for the treatment of joint diseases
CN109477102A (en) 2015-02-13 2019-03-15 菲克特生物科学股份有限公司 Nucleic acid product and its method of administration
WO2016130943A1 (en) 2015-02-13 2016-08-18 Rana Therapeutics, Inc. Hybrid oligonucleotides and uses thereof
EP3900702A1 (en) 2015-03-19 2021-10-27 Translate Bio, Inc. Mrna therapy for pompe disease
DK3294885T3 (en) 2015-05-08 2020-08-10 Curevac Real Estate Gmbh METHOD OF PREPARING RNA
WO2016193206A1 (en) 2015-05-29 2016-12-08 Curevac Ag A method for producing and purifying rna, comprising at least one step of tangential flow filtration
EP3324979B1 (en) 2015-07-21 2022-10-12 ModernaTX, Inc. Infectious disease vaccines
US11364292B2 (en) 2015-07-21 2022-06-21 Modernatx, Inc. CHIKV RNA vaccines
WO2017031232A1 (en) 2015-08-17 2017-02-23 Modernatx, Inc. Methods for preparing particles and related compositions
WO2017049286A1 (en) 2015-09-17 2017-03-23 Moderna Therapeutics, Inc. Polynucleotides containing a morpholino linker
CN108473969B (en) 2015-10-14 2022-09-13 川斯勒佰尔公司 Modification of RNA-associated enzymes for enhanced production
JP2018531290A (en) 2015-10-22 2018-10-25 モデルナティーエックス, インコーポレイテッド Sexually transmitted disease vaccine
JP6925688B2 (en) 2015-10-22 2021-08-25 モデルナティーエックス, インコーポレイテッド Nucleic acid vaccine for varicella-zoster virus (VZV)
JP6921833B2 (en) 2015-10-22 2021-08-18 モデルナティーエックス, インコーポレイテッド Human cytomegalovirus vaccine
WO2017070624A1 (en) 2015-10-22 2017-04-27 Modernatx, Inc. Tropical disease vaccines
ES2763822T3 (en) 2015-12-09 2020-06-01 Novartis Ag Mark-free analysis of the efficiency of adding caps to the RNA using rnasa h, probes and liquid chromatography / mass spectrometry
SI3386484T1 (en) 2015-12-10 2022-06-30 Modernatx, Inc. Compositions and methods for delivery of therapeutic agents
US10465190B1 (en) 2015-12-23 2019-11-05 Modernatx, Inc. In vitro transcription methods and constructs
TW201738256A (en) * 2016-04-04 2017-11-01 日產化學工業股份有限公司 Production method of protein
CN109072223B (en) 2016-04-08 2022-10-21 川斯勒佰尔公司 Multimeric coding nucleic acids and uses thereof
US20180126003A1 (en) * 2016-05-04 2018-05-10 Curevac Ag New targets for rna therapeutics
KR20230074598A (en) 2016-05-18 2023-05-30 모더나티엑스, 인크. Polynucleotides encoding relaxin
WO2017218524A1 (en) 2016-06-13 2017-12-21 Rana Therapeutics, Inc. Messenger rna therapy for the treatment of ornithine transcarbamylase deficiency
JP2019528284A (en) 2016-08-17 2019-10-10 ファクター バイオサイエンス インコーポレイテッド Nucleic acid product and method of administration thereof
SG11201901941YA (en) 2016-09-14 2019-04-29 Modernatx Inc High purity rna compositions and methods for preparation thereof
US11390899B2 (en) * 2016-09-26 2022-07-19 SOLA Biosciences, LLC Cell-associated secretion-enhancing fusion proteins
JP6980780B2 (en) 2016-10-21 2021-12-15 モデルナティーエックス, インコーポレイテッド Human cytomegalovirus vaccine
US10925958B2 (en) 2016-11-11 2021-02-23 Modernatx, Inc. Influenza vaccine
US11103578B2 (en) 2016-12-08 2021-08-31 Modernatx, Inc. Respiratory virus nucleic acid vaccines
US11384352B2 (en) 2016-12-13 2022-07-12 Modernatx, Inc. RNA affinity purification
EP3582790A4 (en) 2017-02-16 2020-11-25 ModernaTX, Inc. High potency immunogenic compositions
WO2018170260A1 (en) 2017-03-15 2018-09-20 Modernatx, Inc. Respiratory syncytial virus vaccine
WO2018170270A1 (en) 2017-03-15 2018-09-20 Modernatx, Inc. Varicella zoster virus (vzv) vaccine
EP3609534A4 (en) 2017-03-15 2021-01-13 ModernaTX, Inc. Broad spectrum influenza virus vaccine
WO2018170256A1 (en) 2017-03-15 2018-09-20 Modernatx, Inc. Herpes simplex virus vaccine
WO2018170347A1 (en) 2017-03-17 2018-09-20 Modernatx, Inc. Zoonotic disease rna vaccines
US11905525B2 (en) 2017-04-05 2024-02-20 Modernatx, Inc. Reduction of elimination of immune responses to non-intravenous, e.g., subcutaneously administered therapeutic proteins
CN106929513A (en) * 2017-04-07 2017-07-07 东南大学 The nano antibody of mRNA codings and its application
WO2018217897A1 (en) * 2017-05-23 2018-11-29 David Weiner Compositions and method for inducing an immune response
EP3648786A4 (en) * 2017-07-03 2021-12-15 Torque Therapeutics, Inc. Fusion molecules targeting immune regulatory cells and uses thereof
MA49913A (en) 2017-08-18 2021-05-05 Modernatx Inc RNA POLYMERASE VARIANTS
WO2019036683A1 (en) 2017-08-18 2019-02-21 Modernatx, Inc. Analytical hplc methods
WO2019036685A1 (en) 2017-08-18 2019-02-21 Modernatx, Inc. Methods for hplc analysis
EP3681514A4 (en) 2017-09-14 2021-07-14 ModernaTX, Inc. Zika virus rna vaccines
US20210180053A1 (en) 2017-11-01 2021-06-17 Novartis Ag Synthetic rnas and methods of use
AU2018392716A1 (en) 2017-12-20 2020-06-18 Translate Bio, Inc. Improved composition and methods for treatment of ornithine transcarbamylase deficiency
EP3735270A1 (en) 2018-01-05 2020-11-11 Modernatx, Inc. Polynucleotides encoding anti-chikungunya virus antibodies
MA54676A (en) 2018-01-29 2021-11-17 Modernatx Inc RSV RNA VACCINES
US11351242B1 (en) 2019-02-12 2022-06-07 Modernatx, Inc. HMPV/hPIV3 mRNA vaccine composition
US11851694B1 (en) 2019-02-20 2023-12-26 Modernatx, Inc. High fidelity in vitro transcription
CA3130888A1 (en) 2019-02-20 2020-08-27 Modernatx, Inc. Rna polymerase variants for co-transcriptional capping
US10501404B1 (en) 2019-07-30 2019-12-10 Factor Bioscience Inc. Cationic lipids and transfection methods
US11576966B2 (en) 2020-02-04 2023-02-14 CureVac SE Coronavirus vaccine
US11241493B2 (en) 2020-02-04 2022-02-08 Curevac Ag Coronavirus vaccine
AU2021252164A1 (en) 2020-04-09 2022-12-15 Finncure Oy Mimetic nanoparticles for preventing the spreading and lowering the infection rate of novel coronaviruses
US11406703B2 (en) 2020-08-25 2022-08-09 Modernatx, Inc. Human cytomegalovirus vaccine
US11591544B2 (en) 2020-11-25 2023-02-28 Akagera Medicines, Inc. Ionizable cationic lipids
TW202237844A (en) 2020-12-09 2022-10-01 德商拜恩技術股份公司 Rna manufacturing
KR20230164648A (en) 2020-12-22 2023-12-04 큐어백 에스이 RNA vaccines against SARS-CoV-2 variants
WO2022155404A1 (en) * 2021-01-14 2022-07-21 Translate Bio, Inc. Methods and compositions for delivering mrna coded antibodies
EP4281034A1 (en) 2021-01-24 2023-11-29 Forrest, Michael, David Inhibitors of atp synthase - cosmetic and therapeutic uses
US11524023B2 (en) 2021-02-19 2022-12-13 Modernatx, Inc. Lipid nanoparticle compositions and methods of formulating the same
CN113736768B (en) * 2021-08-18 2023-06-23 新发药业有限公司 Pseudo uridine synthetase mutant, mutant gene and application thereof in preparation of vitamin B2
WO2023025404A1 (en) 2021-08-24 2023-03-02 BioNTech SE In vitro transcription technologies
WO2023068931A1 (en) 2021-10-21 2023-04-27 Curevac Netherlands B.V. Cancer neoantigens
WO2023196898A1 (en) 2022-04-07 2023-10-12 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Beta globin mimetic peptides and their use
WO2024083345A1 (en) 2022-10-21 2024-04-25 BioNTech SE Methods and uses associated with liquid compositions

Citations (30)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4270537A (en) 1979-11-19 1981-06-02 Romaine Richard A Automatic hypodermic syringe
US4596556A (en) 1985-03-25 1986-06-24 Bioject, Inc. Hypodermic injection apparatus
US4790824A (en) 1987-06-19 1988-12-13 Bioject, Inc. Non-invasive hypodermic injection device
US4886499A (en) 1986-12-18 1989-12-12 Hoffmann-La Roche Inc. Portable injection appliance
US4940460A (en) 1987-06-19 1990-07-10 Bioject, Inc. Patient-fillable and non-invasive hypodermic injection device assembly
US4941880A (en) 1987-06-19 1990-07-17 Bioject, Inc. Pre-filled ampule and non-invasive hypodermic injection device assembly
US5015235A (en) 1987-02-20 1991-05-14 National Carpet Equipment, Inc. Syringe needle combination
US5064413A (en) 1989-11-09 1991-11-12 Bioject, Inc. Needleless hypodermic injection device
US5141496A (en) 1988-11-03 1992-08-25 Tino Dalto Spring impelled syringe guide with skin penetration depth adjustment
US5190521A (en) 1990-08-22 1993-03-02 Tecnol Medical Products, Inc. Apparatus and method for raising a skin wheal and anesthetizing skin
US5312335A (en) 1989-11-09 1994-05-17 Bioject Inc. Needleless hypodermic injection device
US5328483A (en) 1992-02-27 1994-07-12 Jacoby Richard M Intradermal injection device with medication and needle guard
US5334144A (en) 1992-10-30 1994-08-02 Becton, Dickinson And Company Single use disposable needleless injector
US5339163A (en) 1988-03-16 1994-08-16 Canon Kabushiki Kaisha Automatic exposure control device using plural image plane detection areas
US5383851A (en) 1992-07-24 1995-01-24 Bioject Inc. Needleless hypodermic injection device
US5417662A (en) 1991-09-13 1995-05-23 Pharmacia Ab Injection needle arrangement
US5466220A (en) 1994-03-08 1995-11-14 Bioject, Inc. Drug vial mixing and transfer device
US5480381A (en) 1991-08-23 1996-01-02 Weston Medical Limited Needle-less injector
US5527288A (en) 1990-12-13 1996-06-18 Elan Medical Technologies Limited Intradermal drug delivery device and method for intradermal delivery of drugs
US5569189A (en) 1992-09-28 1996-10-29 Equidyne Systems, Inc. hypodermic jet injector
US5599302A (en) 1995-01-09 1997-02-04 Medi-Ject Corporation Medical injection system and method, gas spring thereof and launching device using gas spring
WO1997013537A1 (en) 1995-10-10 1997-04-17 Visionary Medical Products Corporation Gas pressured needle-less injection device
US5649912A (en) 1994-03-07 1997-07-22 Bioject, Inc. Ampule filling device
WO1997037705A1 (en) 1996-04-11 1997-10-16 Weston Medical Limited Spring-powered dispensing device for medical purposes
US5893397A (en) 1996-01-12 1999-04-13 Bioject Inc. Medication vial/syringe liquid-transfer apparatus
WO1999034850A1 (en) 1998-01-08 1999-07-15 Fiderm S.R.L. Device for controlling the penetration depth of a needle, for application to an injection syringe
US5993412A (en) 1997-05-19 1999-11-30 Bioject, Inc. Injection apparatus
WO2007024708A2 (en) 2005-08-23 2007-03-01 The Trustees Of The University Of Pennsylvania Rna containing modified nucleosides and methods of use thereof
WO2009127230A1 (en) * 2008-04-16 2009-10-22 Curevac Gmbh MODIFIED (m)RNA FOR SUPPRESSING OR AVOIDING AN IMMUNOSTIMULATORY RESPONSE AND IMMUNOSUPPRESSIVE COMPOSITION
WO2010111290A1 (en) * 2009-03-23 2010-09-30 University Of Utah Research Foundation Methods and compositions related to modified guanine bases for controlling off-target effects in rna interference

Family Cites Families (1131)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2008526A (en) 1932-11-03 1935-07-16 Wappler Frederick Charles Method and means for treating living tissue
US3467096A (en) 1966-04-12 1969-09-16 Ferrell S Horn Multiple hypodermic syringe arrangement
BE757653A (en) 1969-10-21 1971-04-16 Ugine Kuhlmann NEW DRUGS DERIVED FROM NUCLEIC ACIDS AND METHODS FOR THEIR PREPARATION
BE786542A (en) 1971-07-22 1973-01-22 Dow Corning SUCTION DEVICE ALLOWING TO OBTAIN CELL SAMPLES
US3906092A (en) 1971-11-26 1975-09-16 Merck & Co Inc Stimulation of antibody response
US4399216A (en) 1980-02-25 1983-08-16 The Trustees Of Columbia University Processes for inserting DNA into eucaryotic cells and for producing proteinaceous materials
US4500707A (en) 1980-02-29 1985-02-19 University Patents, Inc. Nucleosides useful in the preparation of polynucleotides
US5132418A (en) 1980-02-29 1992-07-21 University Patents, Inc. Process for preparing polynucleotides
US4458066A (en) 1980-02-29 1984-07-03 University Patents, Inc. Process for preparing polynucleotides
US4411657A (en) 1980-05-19 1983-10-25 Anibal Galindo Hypodermic needle
US4668777A (en) 1981-03-27 1987-05-26 University Patents, Inc. Phosphoramidite nucleoside compounds
US4973679A (en) 1981-03-27 1990-11-27 University Patents, Inc. Process for oligonucleo tide synthesis using phosphormidite intermediates
US4415732A (en) 1981-03-27 1983-11-15 University Patents, Inc. Phosphoramidite compounds and processes
US4373071A (en) 1981-04-30 1983-02-08 City Of Hope Research Institute Solid-phase synthesis of polynucleotides
US4401796A (en) 1981-04-30 1983-08-30 City Of Hope Research Institute Solid-phase synthesis of polynucleotides
US4474569A (en) 1982-06-28 1984-10-02 Denver Surgical Developments, Inc. Antenatal shunt
US4737462A (en) 1982-10-19 1988-04-12 Cetus Corporation Structural genes, plasmids and transformed cells for producing cysteine depleted muteins of interferon-β
US4588585A (en) 1982-10-19 1986-05-13 Cetus Corporation Human recombinant cysteine depleted interferon-β muteins
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US4579849A (en) 1984-04-06 1986-04-01 Merck & Co., Inc. N-alkylguanine acyclonucleosides as antiviral agents
US4957735A (en) 1984-06-12 1990-09-18 The University Of Tennessee Research Corporation Target-sensitive immunoliposomes- preparation and characterization
US4959314A (en) 1984-11-09 1990-09-25 Cetus Corporation Cysteine-depleted muteins of biologically active proteins
US5036006A (en) 1984-11-13 1991-07-30 Cornell Research Foundation, Inc. Method for transporting substances into living cells and tissues and apparatus therefor
US5116943A (en) 1985-01-18 1992-05-26 Cetus Corporation Oxidation-resistant muteins of Il-2 and other protein
CA1288073C (en) 1985-03-07 1991-08-27 Paul G. Ahlquist Rna transformation vector
EP0204401A1 (en) 1985-04-09 1986-12-10 Biogen, Inc. Method of improving the yield of polypeptides produced in a host cell by stabilizing mRNA
US5017691A (en) 1986-07-03 1991-05-21 Schering Corporation Mammalian interleukin-4
US5153319A (en) 1986-03-31 1992-10-06 University Patents, Inc. Process for preparing polynucleotides
US4879111A (en) 1986-04-17 1989-11-07 Cetus Corporation Treatment of infections with lymphokines
JP2774121B2 (en) 1987-07-31 1998-07-09 ザ ボード オブ トラスティーズ オブ ザ リーランド スタンフォード ジュニア ユニバーシティ Selective amplification of target polynucleotide sequence
US6090591A (en) 1987-07-31 2000-07-18 The Board Of Trustees Of The Leland Stanford Junior University Selective amplification of target polynucleotide sequences
DE68908054T2 (en) 1988-01-21 1994-03-10 Genentech Inc REINFORCEMENT AND DETECTION OF NUCLEIC ACID SEQUENCES.
CA1340807C (en) 1988-02-24 1999-11-02 Lawrence T. Malek Nucleic acid amplification process
JP2650159B2 (en) 1988-02-24 1997-09-03 アクゾ・ノベル・エヌ・ベー Nucleic acid amplification method
WO1989007947A1 (en) 1988-03-04 1989-09-08 Cancer Research Campaign Technology Limited Improvements relating to antigens
WO1989009622A1 (en) 1988-04-15 1989-10-19 Protein Design Labs, Inc. Il-2 receptor-specific chimeric antibodies
US5168038A (en) 1988-06-17 1992-12-01 The Board Of Trustees Of The Leland Stanford Junior University In situ transcription in cells and tissues
US5021335A (en) 1988-06-17 1991-06-04 The Board Of Trustees Of The Leland Stanford Junior University In situ transcription in cells and tissues
US5130238A (en) 1988-06-24 1992-07-14 Cangene Corporation Enhanced nucleic acid amplification process
US5759802A (en) 1988-10-26 1998-06-02 Tonen Corporation Production of human serum alubumin A
US5047524A (en) 1988-12-21 1991-09-10 Applied Biosystems, Inc. Automated system for polynucleotide synthesis and purification
US5262530A (en) 1988-12-21 1993-11-16 Applied Biosystems, Inc. Automated system for polynucleotide synthesis and purification
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
AU5344190A (en) 1989-03-21 1990-10-22 Vical, Inc. Expression of exogenous polynucleotide sequences in a vertebrate
US6214804B1 (en) 1989-03-21 2001-04-10 Vical Incorporated Induction of a protective immune response in a mammal by injecting a DNA sequence
US5693622A (en) 1989-03-21 1997-12-02 Vical Incorporated Expression of exogenous polynucleotide sequences cardiac muscle of a mammal
US6673776B1 (en) 1989-03-21 2004-01-06 Vical Incorporated Expression of exogenous polynucleotide sequences in a vertebrate, mammal, fish, bird or human
US6867195B1 (en) 1989-03-21 2005-03-15 Vical Incorporated Lipid-mediated polynucleotide administration to reduce likelihood of subject's becoming infected
US5703055A (en) 1989-03-21 1997-12-30 Wisconsin Alumni Research Foundation Generation of antibodies through lipid mediated DNA delivery
US5012818A (en) 1989-05-04 1991-05-07 Joishy Suresh K Two in one bone marrow surgical needle
IE66597B1 (en) 1989-05-10 1996-01-24 Akzo Nv Method for the synthesis of ribonucleic acid (RNA)
US5240855A (en) 1989-05-12 1993-08-31 Pioneer Hi-Bred International, Inc. Particle gun
US5332671A (en) 1989-05-12 1994-07-26 Genetech, Inc. Production of vascular endothelial cell growth factor and DNA encoding same
CA2020958C (en) 1989-07-11 2005-01-11 Daniel L. Kacian Nucleic acid sequence amplification methods
US5545522A (en) 1989-09-22 1996-08-13 Van Gelder; Russell N. Process for amplifying a target polynucleotide sequence using a single primer-promoter complex
FR2740360B1 (en) 1995-10-25 1997-12-26 Rhone Poulenc Chimie WATER REDISPERSABLE GRANULES COMPRISING AN ACTIVE MATERIAL IN LIQUID FORM
NO904633L (en) 1989-11-09 1991-05-10 Molecular Diagnostics Inc AMPLIFICATION OF NUCLEIC ACIDS BY TRANSCRIPABLE HAIRNEL PROBE.
US5215899A (en) 1989-11-09 1993-06-01 Miles Inc. Nucleic acid amplification employing ligatable hairpin probe and transcription
US5633076A (en) 1989-12-01 1997-05-27 Pharming Bv Method of producing a transgenic bovine or transgenic bovine embryo
US5697901A (en) 1989-12-14 1997-12-16 Elof Eriksson Gene delivery by microneedle injection
US5194370A (en) 1990-05-16 1993-03-16 Life Technologies, Inc. Promoter ligation activated transcription amplification of nucleic acid sequences
AU649066B2 (en) 1990-07-25 1994-05-12 Syngene, Inc. Circular extension for generating multiple nucleic acid complements
US5489677A (en) 1990-07-27 1996-02-06 Isis Pharmaceuticals, Inc. Oligonucleoside linkages containing adjacent oxygen and nitrogen atoms
US6140496A (en) 1990-10-09 2000-10-31 Benner; Steven Albert Precursors for deoxyribonucleotides containing non-standard nucleosides
US6100024A (en) 1991-02-08 2000-08-08 Promega Corporation Methods and compositions for nucleic acid detection by target extension and probe amplification
DE69231828T3 (en) 1991-03-18 2008-06-19 New York University Specific anti-human tumor necrosis factor monoclonal and chimeric antibodies
US5426180A (en) 1991-03-27 1995-06-20 Research Corporation Technologies, Inc. Methods of making single-stranded circular oligonucleotides
JP3370324B2 (en) 1991-04-25 2003-01-27 中外製薬株式会社 Reshaped human antibodies to human interleukin-6 receptor
US5169766A (en) 1991-06-14 1992-12-08 Life Technologies, Inc. Amplification of nucleic acid molecules
US5199441A (en) 1991-08-20 1993-04-06 Hogle Hugh H Fine needle aspiration biopsy apparatus and method
US5298422A (en) 1991-11-06 1994-03-29 Baylor College Of Medicine Myogenic vector systems
US5824307A (en) 1991-12-23 1998-10-20 Medimmune, Inc. Human-murine chimeric antibodies against respiratory syncytial virus
WO1993014778A1 (en) 1992-01-23 1993-08-05 Vical, Inc. Ex vivo gene transfer
JP3368603B2 (en) 1992-02-28 2003-01-20 オリンパス光学工業株式会社 Gene therapy treatment device
US6174666B1 (en) 1992-03-27 2001-01-16 The United States Of America As Represented By The Department Of Health And Human Services Method of eliminating inhibitory/instability regions from mRNA
US6132419A (en) 1992-05-22 2000-10-17 Genetronics, Inc. Electroporetic gene and drug therapy
US5514545A (en) 1992-06-11 1996-05-07 Trustees Of The University Of Pennsylvania Method for characterizing single cells based on RNA amplification for diagnostics and therapeutics
US6670178B1 (en) 1992-07-10 2003-12-30 Transkaryotic Therapies, Inc. In Vivo production and delivery of insulinotropin for gene therapy
WO1994003637A1 (en) 1992-07-31 1994-02-17 Syntex (Usa) Inc. Method for introducing defined sequences at the 3' end of polynucleotides
US5273525A (en) 1992-08-13 1993-12-28 Btx Inc. Injection and electroporation apparatus for drug and gene delivery
US5240885A (en) 1992-09-21 1993-08-31 Corning Incorporated Rare earth-doped, stabilized cadmium halide glasses
AU5665694A (en) 1992-11-04 1994-05-24 Denver Biomaterials Inc. Apparatus for removal of pleural effusion fluid
US5736137A (en) 1992-11-13 1998-04-07 Idec Pharmaceuticals Corporation Therapeutic application of chimeric and radiolabeled antibodies to human B lymphocyte restricted differentiation antigen for treatment of B cell lymphoma
JP3095175B2 (en) 1992-11-13 2000-10-03 アイデック ファーマシューティカルズ コーポレイション Therapeutic use of chimeric and radiolabeled antibodies against human B lymphocyte restricted differentiation antigen for the treatment of B cell lymphoma
DE69419721T2 (en) 1993-01-12 2000-04-27 Biogen Inc RECOMBINANT ANTI-VLA4 ANTIBODY MOLECULES
FR2703253B1 (en) 1993-03-30 1995-06-23 Centre Nat Rech Scient APPLICATOR OF ELECTRIC PULSES FOR TREATING BIOLOGICAL TISSUES.
US7135312B2 (en) 1993-04-15 2006-11-14 University Of Rochester Circular DNA vectors for synthesis of RNA and DNA
US5773244A (en) 1993-05-19 1998-06-30 Regents Of The University Of California Methods of making circular RNA
US5851829A (en) 1993-07-16 1998-12-22 Dana-Farber Cancer Institute Method of intracellular binding of target molecules
US5672491A (en) 1993-09-20 1997-09-30 The Leland Stanford Junior University Recombinant production of novel polyketides
US6432711B1 (en) 1993-11-03 2002-08-13 Diacrin, Inc. Embryonic stem cells capable of differentiating into desired cell lines
US6096503A (en) 1993-11-12 2000-08-01 The Scripps Research Institute Method for simultaneous identification of differentially expresses mRNAs and measurement of relative concentrations
US7435802B2 (en) 1994-01-25 2008-10-14 Elan Pharaceuticals, Inc. Humanized anti-VLA4 immunoglobulins
US5840299A (en) 1994-01-25 1998-11-24 Athena Neurosciences, Inc. Humanized antibodies against leukocyte adhesion molecule VLA-4
DE69533295T3 (en) 1994-02-16 2009-07-16 The Government Of The United States Of America, As Represented By The Secretary, The Department Of Health And Human Services Melanoma-associated antigens, epitopes thereof and melanoma-containing vaccines
IL112820A0 (en) 1994-03-07 1995-05-26 Merck & Co Inc Coordinate in vivo gene expression
WO1995025814A1 (en) 1994-03-18 1995-09-28 Lynx Therapeutics, Inc. Oligonucleotide n3'→p5' phosphoramidates: synthesis and compounds; hybridization and nuclease resistance properties
US5457041A (en) 1994-03-25 1995-10-10 Science Applications International Corporation Needle array and method of introducing biological substances into living cells using the needle array
WO1995026204A1 (en) 1994-03-25 1995-10-05 Isis Pharmaceuticals, Inc. Immune stimulation by phosphorothioate oligonucleotide analogs
US6074642A (en) 1994-05-02 2000-06-13 Alexion Pharmaceuticals, Inc. Use of antibodies specific to human complement component C5 for the treatment of glomerulonephritis
IL113776A (en) 1994-05-18 2008-12-29 Bayer Bioscience Gmbh Dna sequences coding for enzymes which catalyze the synthesis of linear alpha 1,4 - glucans in plants, fungi and microorganisms
AU2656295A (en) 1994-06-02 1996-01-04 Chiron Corporation Nucleic acid immunization using a virus-based infection/transfection system
GB9412230D0 (en) 1994-06-17 1994-08-10 Celltech Ltd Interleukin-5 specific recombiant antibodies
US6239116B1 (en) 1994-07-15 2001-05-29 University Of Iowa Research Foundation Immunostimulatory nucleic acid molecules
WO1996004925A1 (en) 1994-08-12 1996-02-22 Immunomedics, Inc. Immunoconjugates and humanized antibodies specific for b-cell lymphoma and leukemia cells
US5665545A (en) 1994-11-28 1997-09-09 Akzo Nobel N.V. Terminal repeat amplification method
US5641665A (en) 1994-11-28 1997-06-24 Vical Incorporated Plasmids suitable for IL-2 expression
US5588960A (en) 1994-12-01 1996-12-31 Vidamed, Inc. Transurethral needle delivery device with cystoscope and method for treatment of urinary incontinence
US5807718A (en) 1994-12-02 1998-09-15 The Scripps Research Institute Enzymatic DNA molecules
PT795018E (en) 1995-01-06 2007-12-21 Plant Res Int Bv Dna sequences encoding carbohydrate polymer synthesizing enzymes and method for producing transgenic plants
US5795587A (en) 1995-01-23 1998-08-18 University Of Pittsburgh Stable lipid-comprising drug delivery complexes and methods for their production
US5824497A (en) 1995-02-10 1998-10-20 Mcmaster University High efficiency translation of mRNA molecules
DE69629326D1 (en) 1995-02-15 2003-09-11 Joseph Eldor Spinal needle with several holes
DE69621507T2 (en) 1995-03-28 2003-01-09 Japan Science & Tech Corp Method for molecular indexing of genes using restriction enzymes
US5869230A (en) 1995-03-30 1999-02-09 Beth Israel Hospital Association Gene transfer into the kidney
US5986054A (en) 1995-04-28 1999-11-16 The Hospital For Sick Children, Hsc Research And Development Limited Partnership Genetic sequences and proteins related to alzheimer's disease
FR2733762B1 (en) 1995-05-02 1997-08-01 Genset Sa METHOD FOR THE SPECIFIC COUPLING OF THE HAIR OF THE 5 'END OF A RNAM FRAGMENT AND PREPARATION OF RNAM AND COMPLETE DNA
US5700642A (en) 1995-05-22 1997-12-23 Sri International Oligonucleotide sizing using immobilized cleavable primers
US6051429A (en) 1995-06-07 2000-04-18 Life Technologies, Inc. Peptide-enhanced cationic lipid transfections
US6111095A (en) 1995-06-07 2000-08-29 Merck & Co., Inc. Capped synthetic RNA, analogs, and aptamers
US5889136A (en) 1995-06-09 1999-03-30 The Regents Of The University Of Colorado Orthoester protecting groups in RNA synthesis
US5766903A (en) 1995-08-23 1998-06-16 University Technology Corporation Circular RNA and uses thereof
US6265389B1 (en) 1995-08-31 2001-07-24 Alkermes Controlled Therapeutics, Inc. Microencapsulation and sustained release of oligonucleotides
AU7073096A (en) 1995-09-19 1997-04-09 University Of Massachusetts Inhibited biological degradation of oligodeoxynucleotides
US5830879A (en) 1995-10-02 1998-11-03 St. Elizabeth's Medical Center Of Boston, Inc. Treatment of vascular injury using vascular endothelial growth factor
US6265387B1 (en) 1995-10-11 2001-07-24 Mirus, Inc. Process of delivering naked DNA into a hepatocyte via bile duct
EP0771873A3 (en) 1995-10-27 1998-03-04 Takeda Chemical Industries, Ltd. Neuronal cell-specific receptor protein
CU22584A1 (en) 1995-11-17 1999-11-03 Centro Inmunologia Molecular PHARMACEUTICAL COMPOSITIONS CONTAINING A MONOCLONAL ANTIBODY THAT RECOGNIZES THE CD6 HUMAN LEUKOCYTARY DIFFERENTIATION ANTIGEN AND ITS USES FOR THE DIAGNOSIS AND TREATMENT OF PSORIASIS
US6090382A (en) 1996-02-09 2000-07-18 Basf Aktiengesellschaft Human antibodies that bind human TNFα
US5962271A (en) 1996-01-03 1999-10-05 Cloutech Laboratories, Inc. Methods and compositions for generating full-length cDNA having arbitrary nucleotide sequence at the 3'-end
US6261584B1 (en) 1996-02-02 2001-07-17 Alza Corporation Sustained delivery of an active agent using an implantable system
US6395292B2 (en) 1996-02-02 2002-05-28 Alza Corporation Sustained delivery of an active agent using an implantable system
AU1874397A (en) 1996-02-16 1997-09-02 Stichting Rega Vzw Hexitol containing oligonucleotides and their use in antisense strategies
US6534312B1 (en) 1996-02-22 2003-03-18 Merck & Co., Inc. Vaccines comprising synthetic genes
US6090391A (en) 1996-02-23 2000-07-18 Aviron Recombinant tryptophan mutants of influenza
US6300487B1 (en) 1996-03-19 2001-10-09 Cell Therapuetics, Inc. Mammalian lysophosphatidic acid acyltransferase
SE9601245D0 (en) 1996-03-29 1996-03-29 Pharmacia Ab Chimeric superantigens and their use
TW517061B (en) 1996-03-29 2003-01-11 Pharmacia & Amp Upjohn Ab Modified/chimeric superantigens and their use
US5712127A (en) 1996-04-29 1998-01-27 Genescape Inc. Subtractive amplification
US5853719A (en) 1996-04-30 1998-12-29 Duke University Methods for treating cancers and pathogen infections using antigen-presenting cells loaded with RNA
ATE438717T1 (en) 1996-06-05 2009-08-15 Novartis Vaccines & Diagnostic DP-75 CODING DNA AND METHOD OF USE THEREOF
US7329741B2 (en) 1996-06-05 2008-02-12 Chiron Corporation Polynucleotides that hybridize to DP-75 and their use
WO1997048370A2 (en) 1996-06-21 1997-12-24 Merck & Co., Inc. Vaccines comprising synthetic genes
US6234990B1 (en) 1996-06-28 2001-05-22 Sontra Medical, Inc. Ultrasound enhancement of transdermal transport
US5939262A (en) 1996-07-03 1999-08-17 Ambion, Inc. Ribonuclease resistant RNA preparation and utilization
US5677124A (en) 1996-07-03 1997-10-14 Ambion, Inc. Ribonuclease resistant viral RNA standards
US7288266B2 (en) 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
US5849546A (en) 1996-09-13 1998-12-15 Epicentre Technologies Corporation Methods for using mutant RNA polymerases with reduced discrimination between non-canonical and canonical nucleoside triphosphates
US6114148C1 (en) 1996-09-20 2012-05-01 Gen Hospital Corp High level expression of proteins
AU737323B2 (en) 1996-09-24 2001-08-16 Genentech, Inc. A family of genes encoding apoptosis-related peptides, peptides encoded thereby and methods of use thereof
US6214966B1 (en) 1996-09-26 2001-04-10 Shearwater Corporation Soluble, degradable poly(ethylene glycol) derivatives for controllable release of bound molecules into solution
AU4992197A (en) 1996-10-11 1998-05-11 Regents Of The University Of California, The Immunostimulatory polynucleotide/immunomodulatory molecule conjugates
EP0839912A1 (en) 1996-10-30 1998-05-06 Instituut Voor Dierhouderij En Diergezondheid (Id-Dlo) Infectious clones of RNA viruses and vaccines and diagnostic assays derived thereof
GB9623051D0 (en) 1996-11-06 1997-01-08 Schacht Etienne H Delivery of DNA to target cells in biological systems
US5980887A (en) 1996-11-08 1999-11-09 St. Elizabeth's Medical Center Of Boston Methods for enhancing angiogenesis with endothelial progenitor cells
US6143559A (en) * 1996-11-18 2000-11-07 Arch Development Corporation Methods for the production of chicken monoclonal antibodies
US5759179A (en) 1996-12-31 1998-06-02 Johnson & Johnson Medical, Inc. Needle and valve assembly for use with a catheter
ATE332368T1 (en) 1997-01-21 2006-07-15 Gen Hospital Corp SELECTION OF PROTEINS USING RNA-PROTEIN FUSIONS
EP0855184A1 (en) 1997-01-23 1998-07-29 Grayson B. Dr. Lipford Pharmaceutical composition comprising a polynucleotide and an antigen especially for vaccination
US6696291B2 (en) 1997-02-07 2004-02-24 Merck & Co., Inc. Synthetic HIV gag genes
JP2001512308A (en) 1997-02-07 2001-08-21 メルク エンド カンパニー インコーポレーテッド Synthetic HIV GAG gene
US6251665B1 (en) 1997-02-07 2001-06-26 Cem Cezayirli Directed maturation of stem cells and production of programmable antigen presenting dentritic cells therefrom
US6228640B1 (en) 1997-02-07 2001-05-08 Cem Cezayirli Programmable antigen presenting cell of CD34 lineage
US6406705B1 (en) 1997-03-10 2002-06-18 University Of Iowa Research Foundation Use of nucleic acids containing unmethylated CpG dinucleotide as an adjuvant
US6306393B1 (en) 1997-03-24 2001-10-23 Immunomedics, Inc. Immunotherapy of B-cell malignancies using anti-CD22 antibodies
US6261281B1 (en) 1997-04-03 2001-07-17 Electrofect As Method for genetic immunization and introduction of molecules into skeletal muscle and immune cells
US5914269A (en) 1997-04-04 1999-06-22 Isis Pharmaceuticals, Inc. Oligonucleotide inhibition of epidermal growth factor receptor expression
WO1998047913A2 (en) 1997-04-18 1998-10-29 The University Of Medicine And Dentistry Of New Jersey Inhibition of hiv-1 replication by a tat rna-binding domain peptide analog
US5958688A (en) 1997-04-28 1999-09-28 The Trustees Of The University Of Pennsylvania Characterization of mRNA patterns in neurites and single cells for medical diagnosis and therapeutics
US6235883B1 (en) 1997-05-05 2001-05-22 Abgenix, Inc. Human monoclonal antibodies to epidermal growth factor receptor
US5989911A (en) 1997-05-09 1999-11-23 University Of Massachusetts Site-specific synthesis of pseudouridine in RNA
US6124091A (en) 1997-05-30 2000-09-26 Research Corporation Technologies, Inc. Cell growth-controlling oligonucleotides
US6589940B1 (en) 1997-06-06 2003-07-08 Dynavax Technologies Corporation Immunostimulatory oligonucleotides, compositions thereof and methods of use thereof
EP0986572B2 (en) 1997-06-06 2007-06-13 Dynavax Technologies Corporation Immunostimulatory oligonucleotides, compositions thereof and methods of use thereof
CA2294988C (en) 1997-07-01 2015-11-24 Isis Pharmaceuticals Inc. Compositions and methods for the delivery of oligonucleotides via the alimentary canal
US5994511A (en) 1997-07-02 1999-11-30 Genentech, Inc. Anti-IgE antibodies and methods of improving polypeptides
ATE361102T1 (en) 1997-07-21 2007-05-15 Active Biotech Ab CYTOLYSIS OF APPROPRIATE TARGET CELLS WITH SUPERANTIGEN CONJUGATES BY MEANS OF THE INDUCTION OF T-CELL ACTIVATION
WO1999006073A1 (en) 1997-07-31 1999-02-11 St. Elizabeth's Medical Center Of Boston, Inc. Method for the treatment of grafts
WO1999013896A1 (en) 1997-09-18 1999-03-25 The Trustees Of The University Of Pennsylvania Attenuated vif dna immunization cassettes for genetic vaccines
WO1999014346A2 (en) * 1997-09-19 1999-03-25 Sequitur, Inc. SENSE mRNA THERAPY
US6004573A (en) 1997-10-03 1999-12-21 Macromed, Inc. Biodegradable low molecular weight triblock poly(lactide-co-glycolide) polyethylene glycol copolymers having reverse thermal gelation properties
WO1999018221A1 (en) 1997-10-07 1999-04-15 University Of Maryland Biotechnology Institute Method for introducing and expressing rna in animal cells
CN1179048C (en) 1997-10-20 2004-12-08 Gtc生物治疗学公司 Modified nucleic acid sequences and method for increasing mRNA level and protein expression in cell system
US6019747A (en) 1997-10-21 2000-02-01 I-Flow Corporation Spring-actuated infusion syringe
JP2001520889A (en) 1997-10-24 2001-11-06 バレンティス,インコーポレイティド Methods for preparing polynucleotide transfection complexes
CA2309766C (en) 1997-11-20 2008-09-30 Vical Incorporated Treatment of cancer using cytokine-expressing polynucleotides and compositions therefor
US7655777B2 (en) 1997-11-24 2010-02-02 Monsanto Technology Llc Nucleic acid molecules associated with the tocopherol pathway
AU1819499A (en) 1997-12-12 1999-06-28 Samyang Corporation Positively-charged poly{alpha-(omega-aminoalkyl)glycolic acid} for the delivery of a bioactive agent via tissue and cellular uptake
US6517869B1 (en) 1997-12-12 2003-02-11 Expression Genetics, Inc. Positively charged poly(alpha-(omega-aminoalkyl)lycolic acid) for the delivery of a bioactive agent via tissue and cellular uptake
JP2002500010A (en) 1997-12-23 2002-01-08 カイロン コーポレイション Human genes and gene expression products I
US6383811B2 (en) 1997-12-30 2002-05-07 Mirus Corporation Polyampholytes for delivering polyions to a cell
WO1999034831A1 (en) 1998-01-05 1999-07-15 University Of Washington Enhanced transport using membrane disruptive agents
US8287483B2 (en) 1998-01-08 2012-10-16 Echo Therapeutics, Inc. Method and apparatus for enhancement of transdermal transport
JP2002500075A (en) 1998-01-08 2002-01-08 ソントラ メディカル, インコーポレイテッド Transdermal transport enhanced by ultrasound transmission
US6365346B1 (en) 1998-02-18 2002-04-02 Dade Behring Inc. Quantitative determination of nucleic acid amplification products
US5955310A (en) 1998-02-26 1999-09-21 Novo Nordisk Biotech, Inc. Methods for producing a polypeptide in a bacillus cell
US6432925B1 (en) 1998-04-16 2002-08-13 John Wayne Cancer Institute RNA cancer vaccine and methods for its use
US6429301B1 (en) 1998-04-17 2002-08-06 Whitehead Institute For Biomedical Research Use of a ribozyme to join nucleic acids and peptides
GB9808327D0 (en) 1998-04-20 1998-06-17 Chiron Spa Antidiotypic compounds
US6395253B2 (en) 1998-04-23 2002-05-28 The Regents Of The University Of Michigan Microspheres containing condensed polyanionic bioactive agents and methods for their production
AU3534199A (en) 1998-04-23 1999-11-08 Takara Shuzo Co., Ltd. Method For Synthesizing DNA
US20020064517A1 (en) 1998-04-30 2002-05-30 Stewart A. Cederholm-Williams Fibrin sealant as a transfection/transformation vehicle for gene therapy
US20090208418A1 (en) 2005-04-29 2009-08-20 Innexus Biotechnology Internaltional Ltd. Superantibody synthesis and use in detection, prevention and treatment of disease
CN1333678A (en) 1998-05-20 2002-01-30 表达遗传学公司 Hepatocyte targeting polyethylene glyco-grafted poly-lysine polymeric gene carrier
US6503231B1 (en) 1998-06-10 2003-01-07 Georgia Tech Research Corporation Microneedle device for transport of molecules across tissue
US7091192B1 (en) 1998-07-01 2006-08-15 California Institute Of Technology Linear cyclodextrin copolymers
KR20010079526A (en) 1998-07-13 2001-08-22 추후제출 Polyester analogue of poly-l-lysine as a soluble, biodegradable gene delivery carrier
US6222030B1 (en) 1998-08-03 2001-04-24 Agilent Technologies, Inc. Solid phase synthesis of oligonucleotides using carbonate protecting groups and alpha-effect nucleophile deprotection
ES2388893T3 (en) 1998-08-11 2012-10-19 Biogen Idec Inc. Combination therapies for B-cell lymphomas comprising administration of anti-CD20 antibodies
GB9817662D0 (en) 1998-08-13 1998-10-07 Crocker Peter J Substance delivery
US6924365B1 (en) 1998-09-29 2005-08-02 Transkaryotic Therapies, Inc. Optimized messenger RNA
US20090042283A1 (en) 1998-09-29 2009-02-12 Shire Human Genetic Therapies, Inc., A Delaware Corporation Optimized messenger rna
WO2000026226A1 (en) 1998-11-03 2000-05-11 Yale University Multidomain polynucleotide molecular sensors
ES2351149T3 (en) 1998-11-09 2011-02-01 Biogen Idec Inc. CHEMICAL ANTIBODY DIRECTED AGAINST CD20, RITUXAN, FOR USE IN THE TREATMENT OF CHRONIC LYMPHOCYTIC LEUKEMIA.
MXPA01004649A (en) 1998-11-09 2002-05-06 Idec Pharma Corp Chimeric anti-cd20 antibody treatment of patients receiving bmt or pbsc transplants.
AU2023400A (en) 1998-11-12 2000-05-29 Children's Medical Center Corporation Compositions and methods for inhibiting angiogenesis using trna and fragments thereof
US6210931B1 (en) 1998-11-30 2001-04-03 The United States Of America As Represented By The Secretary Of Agriculture Ribozyme-mediated synthesis of circular RNA
US20040171980A1 (en) 1998-12-18 2004-09-02 Sontra Medical, Inc. Method and apparatus for enhancement of transdermal transport
CA2356542A1 (en) 1998-12-23 2000-07-06 Human Genome Sciences, Inc. Peptidoglycan recognition proteins
WO2000050586A2 (en) 1999-02-22 2000-08-31 European Molecular Biology Laboratory In vitro translation system
US6255476B1 (en) 1999-02-22 2001-07-03 Pe Corporation (Ny) Methods and compositions for synthesis of labelled oligonucleotides and analogs on solid-supports
US7629311B2 (en) 1999-02-24 2009-12-08 Edward Lewis Tobinick Methods to facilitate transmission of large molecules across the blood-brain, blood-eye, and blood-nerve barriers
EP1574210B1 (en) 1999-02-26 2016-04-06 Novartis Vaccines and Diagnostics, Inc. Microemulsions with adsorbed macromolecules
CA2369119A1 (en) 1999-03-29 2000-05-25 Statens Serum Institut Nucleotide construct with optimised codons for an hiv genetic vaccine based on a primary, early hiv isolate and synthetic envelope
US7217762B1 (en) 1999-04-09 2007-05-15 Invitrogen Corporation Process for the preparation of monodisperse polymer particles
KR20020011985A (en) 1999-05-07 2002-02-09 파르마솔 게엠베하 Lipid particles on the basis of mixtures of liquid and solid lipids and method for producing same
KR20080039550A (en) 1999-05-07 2008-05-07 제넨테크, 인크. Treatment of autoimmune diseases with antagonists which bind to b cell surface markers
US7074403B1 (en) 1999-06-09 2006-07-11 Immunomedics, Inc. Immunotherapy of autoimmune disorders using antibodies which target B-cells
US6346382B1 (en) 1999-06-01 2002-02-12 Vanderbilt University Human carbamyl phosphate synthetase I polymorphism and diagnostic methods related thereto
US6743211B1 (en) 1999-11-23 2004-06-01 Georgia Tech Research Corporation Devices and methods for enhanced microneedle penetration of biological barriers
US6611707B1 (en) 1999-06-04 2003-08-26 Georgia Tech Research Corporation Microneedle drug delivery device
AU4332399A (en) 1999-06-04 2000-12-28 Cheng-Ming Chuong Rna polymerase chain reaction
US6303573B1 (en) 1999-06-07 2001-10-16 The Burnham Institute Heart homing peptides and methods of using same
CA2376634A1 (en) 1999-06-08 2000-12-14 Aventis Pasteur Immunostimulant oligonucleotide
US6949245B1 (en) 1999-06-25 2005-09-27 Genentech, Inc. Humanized anti-ErbB2 antibodies and treatment with anti-ErbB2 antibodies
JP2003503071A (en) 1999-06-30 2003-01-28 アドバンスド セル テクノロジー、インコーポレイテッド Cytoplasmic transfer to dedifferentiated recipient cells
US6514948B1 (en) 1999-07-02 2003-02-04 The Regents Of The University Of California Method for enhancing an immune response
BR0012325A (en) 1999-07-09 2002-05-21 American Home Prod Methods and compositions for preventing the formation of abnormal RNA during the transcription of a plasmid sequence
US8557244B1 (en) 1999-08-11 2013-10-15 Biogen Idec Inc. Treatment of aggressive non-Hodgkins lymphoma with anti-CD20 antibody
CN1371416B (en) 1999-08-24 2012-10-10 梅达里克斯公司 Human CTLA-4 antibodies and their uses
US20050112141A1 (en) 2000-08-30 2005-05-26 Terman David S. Compositions and methods for treatment of neoplastic disease
US20040106567A1 (en) 1999-09-07 2004-06-03 Hagstrom James E. Intravascular delivery of non-viral nucleic acid
DE69943068D1 (en) 1999-09-09 2011-02-03 Curevac Gmbh Transfer of mRNA using polycationic compounds
WO2001021810A1 (en) 1999-09-17 2001-03-29 Aventis Pasteur Limited Chlamydia antigens and corresponding dna fragments and uses thereof
US6623457B1 (en) 1999-09-22 2003-09-23 Becton, Dickinson And Company Method and apparatus for the transdermal administration of a substance
WO2002064799A2 (en) 1999-09-28 2002-08-22 Transkaryotic Therapies, Inc. Optimized messenger rna
WO2001025488A2 (en) 1999-10-06 2001-04-12 Quark Biotech, Inc. Method for enrichment of natural antisense messenger rna
US7060291B1 (en) 1999-11-24 2006-06-13 Transave, Inc. Modular targeted liposomal delivery system
US6613026B1 (en) 1999-12-08 2003-09-02 Scimed Life Systems, Inc. Lateral needle-less injection apparatus and method
US6277974B1 (en) 1999-12-14 2001-08-21 Cogent Neuroscience, Inc. Compositions and methods for diagnosing and treating conditions, disorders, or diseases involving cell death
US6245929B1 (en) 1999-12-20 2001-06-12 General Electric Company Catalyst composition and method for producing diaryl carbonates, using bisphosphines
ES2230173T3 (en) 1999-12-22 2005-05-01 Basell Poliolefine Italia S.P.A. CATALYSTING SYSTEM FOR THE POLYMERIZATION OF ALFA OLEFINS, WHICH CONTAINS AN AROMATIC COMPOUND OF SILANO.
AU2764801A (en) 2000-01-07 2001-07-24 University Of Washington Enhanced transport of agents using membrane disruptive agents
AU2001232485A1 (en) 2000-01-13 2001-07-24 Amsterdam Support Diagnostics B.V. A universal nucleic acid amplification system for nucleic acids in a sample
WO2001055341A2 (en) 2000-01-31 2001-08-02 The Regents Of The University Of California Immunomodulatory polynucleotides in treatment of an infection by an intracellular pathogen
WO2001055306A2 (en) 2000-01-31 2001-08-02 Human Genome Sciences, Inc. Nucleic acids, proteins, and antibodies
AU2001238595A1 (en) 2000-02-22 2001-09-03 Shearwater Corporation N-maleimidyl polymer derivatives
IL151378A0 (en) 2000-02-24 2003-04-10 Univ Washington Humanized antibodies that sequester amyloid beta peptide
BR0108962A (en) 2000-03-03 2002-12-24 Valentis Inc Nucleic acid formulations for gene distribution and methods of use
AU8729101A (en) 2000-03-31 2001-10-15 Idec Pharma Corp Combined use of anti-cytokine antibodies or antagonists and anti-CD20 for the treatment of B cell lymphoma
WO2001075166A2 (en) 2000-03-31 2001-10-11 Genentech, Inc. Compositions and methods for detecting and quantifying gene expression
US6565572B2 (en) 2000-04-10 2003-05-20 Sdgi Holdings, Inc. Fenestrated surgical screw and method
US6368801B1 (en) 2000-04-12 2002-04-09 Molecular Staging, Inc. Detection and amplification of RNA using target-mediated ligation of DNA by RNA ligase
CA2405563A1 (en) 2000-04-12 2001-10-25 Human Genome Sciences, Inc. Albumin fusion proteins
US20010046496A1 (en) 2000-04-14 2001-11-29 Brettman Lee R. Method of administering an antibody
US6375972B1 (en) 2000-04-26 2002-04-23 Control Delivery Systems, Inc. Sustained release drug delivery devices, methods of use, and methods of manufacturing thereof
US7871598B1 (en) 2000-05-10 2011-01-18 Novartis Ag Stable metal ion-lipid powdered pharmaceutical compositions for drug delivery and methods of use
US20040229271A1 (en) 2000-05-19 2004-11-18 Williams Richard B. Compositions and methods for the identification and selection of nucleic acids and polypeptides
WO2001092523A2 (en) 2000-05-30 2001-12-06 Curagen Corporation Human polynucleotides and polypeptides encoded thereby
EP1292331A2 (en) 2000-06-07 2003-03-19 Biosynexus Incorporated Immunostimulatory rna/dna hybrid molecules
ES2273852T3 (en) 2000-06-23 2007-05-16 Wyeth Holdings Corporation MODIFIED MORBILLIVIRUS PROTEINS V.
DE60139690D1 (en) 2000-07-03 2009-10-08 Novartis Vaccines & Diagnostic IMMUNIZATION AGAINST CHLAMYDIA PNEUMONIAE
US6440096B1 (en) 2000-07-14 2002-08-27 Becton, Dickinson And Co. Microdevice and method of manufacturing a microdevice
DE60124918T2 (en) 2000-07-21 2007-08-02 Glaxo Group Ltd., Greenford CODON-OPTIMIZED PAPILLOMA VIRUS SEQUENCES
US6902734B2 (en) 2000-08-07 2005-06-07 Centocor, Inc. Anti-IL-12 antibodies and compositions thereof
US20040142474A1 (en) 2000-09-14 2004-07-22 Expression Genetics, Inc. Novel cationic lipopolymer as a biocompatible gene delivery agent
US6696038B1 (en) 2000-09-14 2004-02-24 Expression Genetics, Inc. Cationic lipopolymer as biocompatible gene delivery agent
AU2001290078A1 (en) 2000-09-20 2002-04-02 Ruggero Della Bitta Stem cell therapy
CA2424216A1 (en) 2000-10-04 2002-04-11 The Trustees Of The University Of Pennsylvania Compositions and methods of using capsid protein from flaviviruses and pestiviruses
US6998115B2 (en) 2000-10-10 2006-02-14 Massachusetts Institute Of Technology Biodegradable poly(β-amino esters) and uses thereof
US7202226B2 (en) 2000-10-23 2007-04-10 Detroit R & D Augmentation of wound healing by elF-4E mRNA and EGF mRNA
US20030077604A1 (en) 2000-10-27 2003-04-24 Yongming Sun Compositions and methods relating to breast specific genes and proteins
US20020132788A1 (en) 2000-11-06 2002-09-19 David Lewis Inhibition of gene expression by delivery of small interfering RNA to post-embryonic animal cells in vivo
WO2002040545A2 (en) 2000-11-17 2002-05-23 The Government Of The United States, As Represented By The Secretary Of The Department Of Health And Human Services Reduction of the nonspecific animal toxicity of immunotoxins by mutating the framework regions of the fv to lower the isoelectric point
EP1415005B1 (en) 2000-12-07 2012-11-21 Novartis Vaccines and Diagnostics, Inc. Endogenous retroviruses up-regulated in prostate cancer
US20020130430A1 (en) 2000-12-29 2002-09-19 Castor Trevor Percival Methods for making polymer microspheres/nanospheres and encapsulating therapeutic proteins and other products
US7708915B2 (en) 2004-05-06 2010-05-04 Castor Trevor P Polymer microspheres/nanospheres and encapsulating therapeutic proteins therein
KR20040002853A (en) 2001-01-19 2004-01-07 비로노바티브 비.브이. A virus causing respiratory tract illness in susceptible mammals
EP1224943A1 (en) 2001-01-19 2002-07-24 Crucell Holland B.V. Fibronectin as a tumor marker detected by phage antibodies
US20040110191A1 (en) 2001-01-31 2004-06-10 Winkler Matthew M. Comparative analysis of nucleic acids using population tagging
CA2437737A1 (en) 2001-02-14 2002-08-22 Stephen D. Ginsberg Methods and compositions of amplifying rna
US6652886B2 (en) 2001-02-16 2003-11-25 Expression Genetics Biodegradable cationic copolymers of poly (alkylenimine) and poly (ethylene glycol) for the delivery of bioactive agents
DE10109897A1 (en) 2001-02-21 2002-11-07 Novosom Ag Optional cationic liposomes and their use
US7232425B2 (en) 2001-03-02 2007-06-19 Sorenson Development, Inc. Apparatus and method for specific interstitial or subcutaneous diffusion and dispersion of medication
DE60228212D1 (en) 2001-03-09 2008-09-25 Gene Stream Pty Ltd NEW EXPRESSION VECTORS
JP2002262882A (en) 2001-03-12 2002-09-17 Nisshinbo Ind Inc Method for amplifying rna
FR2822164B1 (en) 2001-03-19 2004-06-18 Centre Nat Rech Scient POLYPEPTIDES DERIVED FROM POLYMERASE RNAS, AND USES THEREOF
US6520949B2 (en) 2001-04-02 2003-02-18 Martin St. Germain Method and apparatus for administering fluid to animals subcutaneously
DE10119005A1 (en) 2001-04-18 2002-10-24 Roche Diagnostics Gmbh Process for protein expression starting from stabilized linear short DNA in cell-free in vitro transcription / translation systems with exonuclease-containing lysates or in a cellular system containing exonucleases
US20030171253A1 (en) 2001-04-19 2003-09-11 Averil Ma Methods and compositions relating to modulation of A20
WO2002086134A2 (en) 2001-04-23 2002-10-31 Amaxa Gmbh Buffer solution for electroporation and a method comprising the use of the same
US7560424B2 (en) 2001-04-30 2009-07-14 Zystor Therapeutics, Inc. Targeted therapeutic proteins
US6777187B2 (en) 2001-05-02 2004-08-17 Rubicon Genomics, Inc. Genome walking by selective amplification of nick-translate DNA library and amplification from complex mixtures of templates
AU2002308623A1 (en) 2001-05-08 2002-11-18 Magnatech International, L.P. Electronic length control wire pay-off system and method
US20050137155A1 (en) 2001-05-18 2005-06-23 Sirna Therapeutics, Inc. RNA interference mediated treatment of Parkinson disease using short interfering nucleic acid (siNA)
US8137911B2 (en) 2001-05-22 2012-03-20 Cellscript, Inc. Preparation and use of single-stranded transcription substrates for synthesis of transcription products corresponding to target sequences
US7514098B2 (en) 2001-05-30 2009-04-07 The Board Of Trustees Of The Leland Stanford Junior University Use of targeted cross-linked nanoparticles for in vivo gene delivery
EP1832603B1 (en) 2001-06-05 2010-02-03 CureVac GmbH Stabilised mRNA with increased G/C-content encoding a bacterial antigen and its use
US20040175787A1 (en) 2001-06-18 2004-09-09 Klemens Kaupmann Novel g-protein coupled receptors and dna sequences thereof
US7785610B2 (en) 2001-06-21 2010-08-31 Dynavax Technologies Corporation Chimeric immunomodulatory compounds and methods of using the same—III
US7547551B2 (en) 2001-06-21 2009-06-16 University Of Antwerp. Transfection of eukaryontic cells with linear polynucleotides by electroporation
JP2005503137A (en) 2001-06-26 2005-02-03 ノバルティス アクチエンゲゼルシャフト G protein coupled receptor and DNA sequence thereof
SE0102327D0 (en) 2001-06-28 2001-06-28 Active Biotech Ab A novel engineered superantigen for human therapy
US20040236092A1 (en) 2001-07-13 2004-11-25 Roman Dziarski Peptidologlycan recognition protein encoding nucleic acids and methods of use thereof
US6586524B2 (en) 2001-07-19 2003-07-01 Expression Genetics, Inc. Cellular targeting poly(ethylene glycol)-grafted polymeric gene carrier
AU2002312543A1 (en) 2001-08-01 2003-02-17 University Of Utah, Technology Transfer Office Isoform-selective inhibitors and activators of pde3 cyclic
AU2002331181A1 (en) 2001-08-27 2003-03-10 Novartis Ag G-protein coupled receptor and dna sequences thereof
US20040142325A1 (en) 2001-09-14 2004-07-22 Liat Mintz Methods and systems for annotating biomolecular sequences
AR045702A1 (en) 2001-10-03 2005-11-09 Chiron Corp COMPOSITIONS OF ASSISTANTS.
DE10148886A1 (en) 2001-10-04 2003-04-30 Avontec Gmbh Inhibition of STAT-1
US7276489B2 (en) 2002-10-24 2007-10-02 Idera Pharmaceuticals, Inc. Modulation of immunostimulatory properties of oligonucleotide-based compounds by optimal presentation of 5′ ends
EP1452593B1 (en) 2001-11-14 2009-04-08 Toyo Boseki Kabushiki Kaisha Dna synthesis promoters, dna polymerase-associated factors and utilization thereof
US20030138419A1 (en) 2001-11-16 2003-07-24 The University Of Tennessee Research Corporation Recombinant antibody fusion proteins and methods for detection of apoptotic cells
WO2003046578A2 (en) 2001-11-29 2003-06-05 Novartis Ag Method for the assessment and prognosis of sarcoidosis
CA2409775C (en) 2001-12-03 2010-07-13 F. Hoffmann-La Roche Ag Reversibly modified thermostable enzymes for dna synthesis and amplification in vitro
US20060275747A1 (en) 2001-12-07 2006-12-07 Hardy Stephen F Endogenous retrovirus up-regulated in prostate cancer
JP4646315B2 (en) 2001-12-07 2011-03-09 ノバルティス バクシンズ アンド ダイアグノスティックス,インコーポレーテッド Endogenous retroviruses up-regulated in prostate cancer
WO2003050258A2 (en) 2001-12-07 2003-06-19 Chiron Corporation Endogenous retrovirus polypeptides linked to oncogenic transformation
EP1458755A2 (en) 2001-12-17 2004-09-22 Novartis AG Novel g-protein coupled receptors and dna sequences thereof
DE10162480A1 (en) 2001-12-19 2003-08-07 Ingmar Hoerr The application of mRNA for use as a therapeutic agent against tumor diseases
PL211494B1 (en) 2001-12-21 2012-05-31 Alcon Use of synthetic inorganic nanoparticles as carriers for ophthalmic and otic drugs
WO2003059381A2 (en) 2002-01-18 2003-07-24 Curevac Gmbh Immunogenic preparations and vaccines on the basis of mrna
JP2005526497A (en) 2002-02-04 2005-09-08 ビオミラ,インコーポレーテッド Immunostimulatory, covalently lipidated oligonucleotide
AU2003210802B2 (en) 2002-02-05 2009-09-10 Genentech Inc. Protein purification
FR2835749B1 (en) 2002-02-08 2006-04-14 Inst Nat Sante Rech Med PHARMACEUTICAL COMPOSITION IMPROVING IN VIVO GENE TRANSFER
DE10207178A1 (en) 2002-02-19 2003-09-04 Novosom Ag Components for the production of amphoteric liposomes
AR038568A1 (en) 2002-02-20 2005-01-19 Hoffmann La Roche ANTI-A BETA ANTIBODIES AND ITS USE
US7354742B2 (en) 2002-02-22 2008-04-08 Ortho-Mcneil Pharmaceutical, Inc. Method for generating amplified RNA
NZ535690A (en) 2002-02-26 2009-04-30 Maxygen Inc Novel flavivirus antigens
PT1916001E (en) 2002-03-04 2011-07-18 Imclone Llc Human antibodies specific to kdr and uses thereof
AU2003221497A1 (en) 2002-03-13 2003-09-22 Novartis Ag Pharmaceutical microparticles
US7074596B2 (en) 2002-03-25 2006-07-11 Board Of Supervisors Of Louisiana State University And Agricultural And Mechanical College Synthesis and use of anti-reverse mRNA cap analogues
US8153141B2 (en) 2002-04-04 2012-04-10 Coley Pharmaceutical Gmbh Immunostimulatory G, U-containing oligoribonucleotides
EP1497327A2 (en) 2002-04-17 2005-01-19 Novartis AG Method for the identification of inhibitors of the binding of are-containing mrna and an hur protein
GB0209539D0 (en) 2002-04-26 2002-06-05 Avecia Ltd Monomer Polymer and process
EP1361277A1 (en) 2002-04-30 2003-11-12 Centre National De La Recherche Scientifique (Cnrs) Optimization of transgene expression in mammalian cells
NZ573563A (en) 2002-05-02 2010-10-29 Wyeth Corp Calicheamicin derivative-carrier conjugates with reduced low conjugated fraction (LCF)
US7374930B2 (en) 2002-05-21 2008-05-20 Expression Genetics, Inc. GLP-1 gene delivery for the treatment of type 2 diabetes
US20040018525A1 (en) 2002-05-21 2004-01-29 Bayer Aktiengesellschaft Methods and compositions for the prediction, diagnosis, prognosis, prevention and treatment of malignant neoplasma
DE10224200C1 (en) 2002-05-31 2003-08-21 Artus Ges Fuer Molekularbiolog Replicating RNA, useful, after reverse transcription, for analysis on microarrays, comprises conversion to cDNA then reverse transcription of this to form antisense sequences
AU2003237367A1 (en) 2002-06-03 2003-12-19 Chiron Corporation Use of nrg4, or inhibitors thereof, in the treatment of colon and pancreatic cancer
SE0201907D0 (en) 2002-06-19 2002-06-19 Atos Medical Ab Patches for tracheostoma valves
ES2354607T3 (en) 2002-06-28 2011-03-16 Protiva Biotherapeutics Inc. PROCEDURE AND APPLIANCE TO PRODUCE LIPOSOMES.
AU2003251770B9 (en) 2002-07-01 2009-06-04 H. Lundbeck A/S Recombinant tissue protective cytokines and encoding nucleic acids thereof for protection, restoration, and enhancement of responsive cells, tissues, and organs
DE10229872A1 (en) 2002-07-03 2004-01-29 Curevac Gmbh Immune stimulation through chemically modified RNA
GB0215509D0 (en) 2002-07-04 2002-08-14 Novartis Ag Marker genes
AR040575A1 (en) 2002-07-16 2005-04-13 Advisys Inc OPTIMIZED SYNTHETIC PLASMIDS IN EXPRESSION CODONS IN MAMMERS
AU2003252136A1 (en) 2002-07-24 2004-02-09 Ptc Therapeutics, Inc. METHODS FOR IDENTIFYING SMALL MOLEDULES THAT MODULATE PREMATURE TRANSLATION TERMINATION AND NONSENSE MEDIATED mRNA DECAY
EP1393745A1 (en) 2002-07-29 2004-03-03 Hybridon, Inc. Modulation of immunostimulatory properties of oligonucleotide-based compounds by optimal presentation of 5'ends
US6653468B1 (en) 2002-07-31 2003-11-25 Isis Pharmaceuticals, Inc. Universal support media for synthesis of oligomeric compounds
EP1386925A1 (en) 2002-07-31 2004-02-04 Girindus AG Method for preparing oligonucleotides
EP1873180B1 (en) 2002-08-14 2014-05-07 Novartis AG Ophthalmic device made from a radiation-curable prepolymer
ES2377318T3 (en) 2002-09-06 2012-03-26 Cerulean Pharma Inc. Cyclodextrin-based polymers for the delivery of covalently bound therapeutic agents to them
ATE412684T1 (en) 2002-09-09 2008-11-15 Nektar Therapeutics Al Corp METHOD FOR PRODUCING WATER-SOLUBLE POLYMER DERIVATIVES WITH TERMINAL CARBOXYL GROUPS
US7534872B2 (en) 2002-09-27 2009-05-19 Syngen, Inc. Compositions and methods for the use of FMOC derivatives in DNA/RNA synthesis
CN101928344B (en) 2002-10-17 2014-08-13 根马布股份公司 Human monoclonal antibodies against cd20
WO2004038018A1 (en) 2002-10-22 2004-05-06 Eisai Co., Ltd. Gene expressed specifically in dopamine-producing neuron precursor cells after termination of division
EP1567675A4 (en) 2002-11-21 2006-05-10 Epict Technologies Methods for using primers that encode one strand of a double-stranded promoter
US7491234B2 (en) 2002-12-03 2009-02-17 Boston Scientific Scimed, Inc. Medical devices for delivery of therapeutic agents
HU227217B1 (en) 2002-12-16 2010-11-29 Genentech Inc Immunoglobulin variants and uses thereof
WO2004058159A2 (en) 2002-12-23 2004-07-15 Dynavax Technologies Corporation Branched immunomodulatory compounds and methods of using the same
US7169892B2 (en) 2003-01-10 2007-01-30 Astellas Pharma Inc. Lipid-peptide-polymer conjugates for long blood circulation and tumor specific drug delivery systems
WO2004067728A2 (en) 2003-01-17 2004-08-12 Ptc Therapeutics Methods and systems for the identification of rna regulatory sequences and compounds that modulate their function
US8460864B2 (en) 2003-01-21 2013-06-11 Ptc Therapeutics, Inc. Methods for identifying compounds that modulate untranslated region-dependent gene expression and methods of using same
US9068234B2 (en) 2003-01-21 2015-06-30 Ptc Therapeutics, Inc. Methods and agents for screening for compounds capable of modulating gene expression
US8426194B2 (en) 2003-01-21 2013-04-23 Ptc Therapeutics, Inc. Methods and agents for screening for compounds capable of modulating VEGF expression
US20040147027A1 (en) 2003-01-28 2004-07-29 Troy Carol M. Complex for facilitating delivery of dsRNA into a cell and uses thereof
EP1592440A4 (en) 2003-02-10 2007-07-11 Elan Pharm Inc Immunoglobulin formulation and method of preparation thereof
US20040167090A1 (en) 2003-02-21 2004-08-26 Monahan Sean D. Covalent modification of RNA for in vitro and in vivo delivery
CA2450289A1 (en) 2003-03-20 2005-05-19 Imclone Systems Incorporated Method of producing an antibody to epidermal growth factor receptor
US7320961B2 (en) 2003-03-24 2008-01-22 Abbott Laboratories Method for treating a disease, disorder or adverse effect caused by an elevated serum concentration of an UGT1A1 substrate
JP4722035B2 (en) 2003-03-25 2011-07-13 アジレント・テクノロジーズ・インク DNA polymerase fusion and use thereof
US20040242502A1 (en) 2003-04-08 2004-12-02 Galenica Pharmaceuticals, Inc. Semi-synthetic saponin analogs with carrier and immune stimulatory activities for DNA and RNA vaccines
MXPA05010778A (en) 2003-04-09 2005-12-12 Genentech Inc Therapy of autoimmune disease in a patient with an inadequate response to a tnf-alpha inhibitor.
EP1620140B1 (en) 2003-05-05 2013-10-09 Ben-Gurion University Of The Negev Research And Development Authority Injectable cross-linked polymeric preparations and uses thereof
US7348004B2 (en) 2003-05-06 2008-03-25 Syntonix Pharmaceuticals, Inc. Immunoglobulin chimeric monomer-dimer hybrids
EP2298347B1 (en) 2003-05-06 2015-09-30 Biogen Hemophilia Inc. Clotting factor chimeric proteins for treatment of a hemostatic disorder
TWI353991B (en) 2003-05-06 2011-12-11 Syntonix Pharmaceuticals Inc Immunoglobulin chimeric monomer-dimer hybrids
US9567591B2 (en) 2003-05-15 2017-02-14 Mello Biotechnology, Inc. Generation of human embryonic stem-like cells using intronic RNA
GB0313132D0 (en) 2003-06-06 2003-07-09 Ich Productions Ltd Peptide ligands
WO2005009346A2 (en) 2003-06-24 2005-02-03 Mirus Corporation Inhibition of gene function by delivery of polynucleotide-based gene expression inhibitors to mammalian cells in vivo
GB0316089D0 (en) 2003-07-09 2003-08-13 Xo Bioscience Ltd Differentiation method
US8592197B2 (en) 2003-07-11 2013-11-26 Novavax, Inc. Functional influenza virus-like particles (VLPs)
US7575572B2 (en) 2003-07-15 2009-08-18 Spinal Generations, Llc Method and device for delivering medicine to bone
US20050013870A1 (en) 2003-07-17 2005-01-20 Toby Freyman Decellularized extracellular matrix of conditioned body tissues and uses thereof
CN101124240B (en) 2003-07-18 2013-12-18 安姆根有限公司 Specific binding agents to hepatocyte growth factor
DE10335833A1 (en) 2003-08-05 2005-03-03 Curevac Gmbh Transfection of blood cells with mRNA for immune stimulation and gene therapy
US8668926B1 (en) 2003-09-15 2014-03-11 Shaker A. Mousa Nanoparticle and polymer formulations for thyroid hormone analogs, antagonists, and formulations thereof
US7135010B2 (en) 2003-09-30 2006-11-14 Damage Control Surgical Technologies, Inc. Method and apparatus for rapid deployment chest drainage
BRPI0415302A (en) 2003-10-06 2006-12-05 Novartis Ag use of genetic polymorphisms that are associated with the efficacy of treating inflammatory disease
US20050130201A1 (en) 2003-10-14 2005-06-16 Dharmacon, Inc. Splint-assisted enzymatic synthesis of polyribounucleotides
DE10347710B4 (en) 2003-10-14 2006-03-30 Johannes-Gutenberg-Universität Mainz Recombinant vaccines and their use
RS54450B1 (en) 2003-11-05 2016-06-30 Roche Glycart Ag Antigen binding molecules with increased fc receptor binding affinity and effector function
WO2005047536A2 (en) 2003-11-13 2005-05-26 Novartis Ag Detection of genomic amplification and deletion in cancer
US20070054278A1 (en) 2003-11-18 2007-03-08 Applera Corporation Polymorphisms in nucleic acid molecules encoding human enzyme proteins, methods of detection and uses thereof
US7699852B2 (en) 2003-11-19 2010-04-20 Zimmer Spine, Inc. Fenestrated bone tap and method
US20050153333A1 (en) 2003-12-02 2005-07-14 Sooknanan Roy R. Selective terminal tagging of nucleic acids
EP1691746B1 (en) 2003-12-08 2015-05-27 Gel-Del Technologies, Inc. Mucoadhesive drug delivery devices and methods of making and using thereof
US7674884B2 (en) 2003-12-10 2010-03-09 Novimmune S.A. Neutralizing antibodies and methods of use thereof
WO2005062854A2 (en) 2003-12-19 2005-07-14 University Of Cincinnati Polyamides for nucleic acid delivery
SI1711528T1 (en) 2003-12-23 2012-10-30 Genentech Inc Treatment of cancer with novel anti-il 13 monoclonal antibodies
WO2005072710A2 (en) 2004-01-28 2005-08-11 Johns Hopkins University Drugs and gene carrier particles that rapidly move through mucous barriers
EP1716233B1 (en) 2004-01-30 2009-08-26 Maxygen Holdings Ltd. Regulated stop codon readthrough
US7309487B2 (en) 2004-02-09 2007-12-18 George Inana Methods and compositions for detecting and treating retinal diseases
CA2556027C (en) 2004-02-12 2015-11-24 Morphotek, Inc. Monoclonal antibodies that specifically block biological activity of a tumor antigen
US20070265220A1 (en) * 2004-03-15 2007-11-15 City Of Hope Methods and compositions for the specific inhibition of gene expression by double-stranded RNA
EP3269738A1 (en) 2004-03-24 2018-01-17 Chugai Seiyaku Kabushiki Kaisha Subtypes of humanized antibody against interleukin-6 receptor
WO2005098433A2 (en) 2004-04-01 2005-10-20 Novartis Ag Diagnostic assays for alzheimer’s disease
JP5848861B2 (en) 2004-04-20 2016-01-27 ジェンマブ エー/エスGenmab A/S Human monoclonal antibody against CD20
ES2246694B1 (en) 2004-04-29 2007-05-01 Instituto Cientifico Y Tecnologico De Navarra, S.A. PEGILATED NANOPARTICLES.
US20080119645A1 (en) 2004-05-05 2008-05-22 Isis Pharmaceuticals, Inc. Amidites and Methods of Rna Synthesis
WO2005112894A1 (en) 2004-05-12 2005-12-01 Baxter International Inc. Nucleic acid microspheres, production and delivery thereof
CA2568772A1 (en) 2004-06-01 2005-12-15 San Diego State University Foundation Expression system based on recombinant vesicular stomatitis virus synthesizing t7 rna polymerase
US7745651B2 (en) 2004-06-07 2010-06-29 Protiva Biotherapeutics, Inc. Cationic lipids and methods of use
EP1766035B1 (en) 2004-06-07 2011-12-07 Protiva Biotherapeutics Inc. Lipid encapsulated interfering rna
EP1773240B1 (en) 2004-06-11 2019-11-20 Trustees of the Tufts College Silk-based drug delivery system
WO2006046978A2 (en) 2004-06-28 2006-05-04 Argos Therapeutics, Inc. Cationic peptide-mediated transformation
KR101100059B1 (en) 2004-06-30 2011-12-29 넥타르 테라퓨틱스 Polymer-factor ix moiety conjugates
US7579451B2 (en) 2004-07-21 2009-08-25 Alnylam Pharmaceuticals, Inc. Oligonucleotides comprising a modified or non-natural nucleobase
DE102004035227A1 (en) 2004-07-21 2006-02-16 Curevac Gmbh mRNA mixture for vaccination against tumor diseases
US7603349B1 (en) 2004-07-29 2009-10-13 Yahoo! Inc. User interfaces for search systems using in-line contextual queries
GB0417487D0 (en) 2004-08-05 2004-09-08 Novartis Ag Organic compound
SE0402025D0 (en) 2004-08-13 2004-08-13 Active Biotech Ab Treatment of hyperproliferative disease with superantigens in combination with another anticancer agent
US7291208B2 (en) 2004-08-13 2007-11-06 Gore Enterprise Holdings, Inc. Grooved active and passive adsorbent filters
CA2478458A1 (en) 2004-08-20 2006-02-20 Michael Panzara Treatment of pediatric multiple sclerosis
HUE024999T2 (en) 2004-08-26 2016-02-29 Engeneic Molecular Delivery Pty Ltd Delivering functional nucleic acids to mammalian cells via bacterially-derived, intact minicells
DE102004042546A1 (en) 2004-09-02 2006-03-09 Curevac Gmbh Combination therapy for immune stimulation
US7501486B2 (en) 2004-09-07 2009-03-10 Burnham Institute For Medical Research Peptides that selectively home to heart vasculature and related conjugates and methods
US8663599B1 (en) 2004-10-05 2014-03-04 Gp Medical, Inc. Pharmaceutical composition of nanoparticles
EP1811018A4 (en) 2004-10-12 2007-11-28 Tissue Targeting Japan Inc Brain disposition marrow progenitor
WO2006044682A1 (en) 2004-10-13 2006-04-27 Ptc Therapeutics, Inc. Compounds for nonsense suppression, and methods for their use
US8057821B2 (en) 2004-11-03 2011-11-15 Egen, Inc. Biodegradable cross-linked cationic multi-block copolymers for gene delivery and methods of making thereof
US20080261905A1 (en) 2004-11-08 2008-10-23 K.U. Leuven Research And Development Modified Nucleosides for Rna Interference
US8946444B2 (en) 2004-11-23 2015-02-03 Ptc Therapeutics, Inc. Tetrahydrocarbazoles as active agents for inhibiting VEGF production by translational control
US7964571B2 (en) 2004-12-09 2011-06-21 Egen, Inc. Combination of immuno gene therapy and chemotherapy for treatment of cancer and hyperproliferative diseases
US8354476B2 (en) 2004-12-10 2013-01-15 Kala Pharmaceuticals, Inc. Functionalized poly(ether-anhydride) block copolymers
US9068969B2 (en) 2004-12-28 2015-06-30 Ptc Therapeutics, Inc. Cell based methods and systems for the identification of RNA regulatory sequences and compounds that modulate their functions
US8535702B2 (en) 2005-02-01 2013-09-17 Boston Scientific Scimed, Inc. Medical devices having porous polymeric regions for controlled drug delivery and regulated biocompatibility
US20100221186A1 (en) 2005-03-11 2010-09-02 Hueseyin Firat Biomarkers for cardiovascular side-effects induced by cox-2 inhibitory compounds
US8415325B2 (en) 2005-03-31 2013-04-09 University Of Delaware Cell-mediated delivery and targeted erosion of noncovalently crosslinked hydrogels
JP2008535857A (en) 2005-04-07 2008-09-04 ノバルティス ヴァクシンズ アンド ダイアグノスティクス インコーポレイテッド CACNA1E in cancer diagnosis, detection and treatment
CA2604844A1 (en) 2005-04-07 2006-10-19 Novartis Vaccines And Diagnostics, Inc. Cancer-related genes
WO2006110776A2 (en) 2005-04-12 2006-10-19 Nektar Therapeutics Al, Corporation Polyethylene glycol cojugates of antimicrobial agents
JP2008539241A (en) 2005-04-25 2008-11-13 ファイザー インコーポレイティッド Antibody to myostatin
WO2006116458A2 (en) 2005-04-26 2006-11-02 Coley Pharmaceutical Gmbh Modified oligoribonucleotide analogs with enhances immunostimulatory activity
EP2418278A3 (en) 2005-05-09 2012-07-04 Ono Pharmaceutical Co., Ltd. Human monoclonal antibodies to programmed death 1(PD-1) and methods for treating cancer using anti-PD-1 antibodies alone or in combination with other immunotherapeutics
US20070072175A1 (en) 2005-05-13 2007-03-29 Biogen Idec Ma Inc. Nucleotide array containing polynucleotide probes complementary to, or fragments of, cynomolgus monkey genes and the use thereof
US20060265771A1 (en) 2005-05-17 2006-11-23 Lewis David L Monitoring microrna expression and function
DE102005023170A1 (en) 2005-05-19 2006-11-23 Curevac Gmbh Optimized formulation for mRNA
JP2008541770A (en) 2005-06-03 2008-11-27 ジェネンテック・インコーポレーテッド Methods for producing antibodies having altered fucosylation levels
US7550264B2 (en) 2005-06-10 2009-06-23 Datascope Investment Corporation Methods and kits for sense RNA synthesis
CN101242858B (en) 2005-06-16 2012-12-19 尼克塔治疗公司 Conjugates having a degradable linkage and polymeric reagents useful in preparing such conjugates
US8202835B2 (en) 2005-06-17 2012-06-19 Yitzchak Hillman Disease treatment via antimicrobial peptides or their inhibitors
US20110182805A1 (en) 2005-06-17 2011-07-28 Desimone Joseph M Nanoparticle fabrication methods, systems, and materials
RU2008103346A (en) 2005-06-30 2009-08-10 Аркемикс Корп. (Us) MATERIALS AND METHODS FOR PRODUCING FULLY 2-MODIFIED NUCLEIC ACIDS TRANSCRIPTS
US8101385B2 (en) 2005-06-30 2012-01-24 Archemix Corp. Materials and methods for the generation of transcripts comprising modified nucleotides
US20080220471A1 (en) 2005-07-27 2008-09-11 Genentech, Inc. Vectors and Methods Using Same
US7612181B2 (en) 2005-08-19 2009-11-03 Abbott Laboratories Dual variable domain immunoglobulin and uses thereof
US9012219B2 (en) 2005-08-23 2015-04-21 The Trustees Of The University Of Pennsylvania RNA preparations comprising purified modified RNA for reprogramming cells
US20070048741A1 (en) 2005-08-24 2007-03-01 Getts Robert C Methods and kits for sense RNA synthesis
WO2007026249A2 (en) 2005-09-01 2007-03-08 Novartis Vaccines And Diagnostics Gmbh & Co Kg Multiple vaccination including serogroup c meningococcus
ATE499112T1 (en) 2005-09-01 2011-03-15 Celgene Corp IMMUNOLOGICAL USES OF IMMUNOMODULATORY COMPOUNDS FOR A VACCINE AND THERAPY AGAINST INFECTIOUS DISEASES
US8420605B2 (en) 2005-09-07 2013-04-16 The University Of Strathclyde Hydrogel compositions
US20120021042A1 (en) 2005-09-15 2012-01-26 Steffen Panzner Efficient Method For Loading Amphoteric Liposomes With Nucleic Acid Active Substances
DE102005046490A1 (en) 2005-09-28 2007-03-29 Johannes-Gutenberg-Universität Mainz New nucleic acid molecule comprising promoter, a transcriptable nucleic acid sequence, a first and second nucleic acid sequence for producing modified RNA with transcriptional stability and translational efficiency
US20070087437A1 (en) 2005-10-14 2007-04-19 Jifan Hu Methods for rejuvenating cells in vitro and in vivo
AU2006310339B2 (en) 2005-11-04 2013-01-10 Novartis Ag Influenza vaccines including combinations of particulate adjuvants and immunopotentiators
US20070105124A1 (en) 2005-11-08 2007-05-10 Getts Robert C Methods and kits for nucleic acid amplification
US20090170090A1 (en) 2005-11-18 2009-07-02 Bioline Limited Method for Enhancing Enzymatic DNA Polymerase Reactions
JP5525729B2 (en) 2005-11-28 2014-06-18 ゲンマブ エー/エス Recombinant monovalent antibody and production method thereof
AU2005338632B2 (en) 2005-11-30 2010-05-20 Epicentre Technologies Corporation Selective terminal tagging of nucleic acids
TWI389709B (en) 2005-12-01 2013-03-21 Novartis Ag Transdermal therapeutic system
US9393215B2 (en) 2005-12-02 2016-07-19 Novartis Ag Nanoparticles for use in immunogenic compositions
US8603457B2 (en) 2005-12-02 2013-12-10 University Of Rochester Nonsense suppression and genetic codon alteration by targeted modification
US7579318B2 (en) 2005-12-06 2009-08-25 Centre De La Recherche De La Scientifique Cell penetrating peptides for intracellular delivery of molecules
WO2007069090A2 (en) 2005-12-06 2007-06-21 Centre National De La Recherche Scientifique Cell penetrating peptides for intracellular delivery of molecules
AU2006321602B2 (en) 2005-12-08 2012-03-08 Novartis Ag Effects of inhibitors of FGFR3 on gene transcription
CN101864392B (en) 2005-12-13 2016-03-23 国立大学法人京都大学 Nuclear reprogramming factor
EP1968643A2 (en) 2005-12-16 2008-09-17 Diatos Cell penetrating peptide conjugates for delivering of nucleic acids into a cell
WO2007077042A1 (en) 2006-01-06 2007-07-12 Topotarget Switzerland Sa New method for the treatment of gout or pseudogout
KR101421745B1 (en) 2006-01-13 2014-07-22 더 트러스티스 오브 더 유니버시티 오브 펜실바니아 Vaccines and immunotherapeutics using codon optimized il-15 and methods for using the same
US20070178103A1 (en) 2006-01-30 2007-08-02 Fey Georg H CD19-specific immunotoxin and treatment method
US8476234B2 (en) 2006-02-03 2013-07-02 Prolor Biotech Inc. Long-acting coagulation factors and methods of producing same
US9458444B2 (en) 2006-02-03 2016-10-04 Opko Biologics Ltd. Long-acting coagulation factors and methods of producing same
US8946155B2 (en) 2006-02-03 2015-02-03 Opko Biologics Ltd. Long-acting polypeptides and methods of producing and administering same
DE102006007433A1 (en) 2006-02-17 2007-08-23 Curevac Gmbh Immunostimulant adjuvant useful in vaccines against cancer or infectious diseases comprises a lipid-modified nucleic acid
KR100859972B1 (en) 2006-02-20 2008-09-25 이화여자대학교 산학협력단 Cell membrane-penetrating peptide
US8309680B2 (en) 2006-02-21 2012-11-13 Nektar Therapeutics Segmented degradable polymers and conjugates made therefrom
AU2007221154A1 (en) 2006-02-24 2007-09-07 Novartis Ag Microparticles containing biodegradable polymer and cationic polysaccharide for use in immunogenic compositions
US20080038278A1 (en) 2006-02-24 2008-02-14 Jingsong Cao GPAT3 encodes a mammalian, microsomal acyl-coa:glycerol 3- phosphate acyltransferase
US7910152B2 (en) 2006-02-28 2011-03-22 Advanced Cardiovascular Systems, Inc. Poly(ester amide)-based drug delivery systems with controlled release rate and morphology
JP2009528359A (en) 2006-02-28 2009-08-06 エラン ファーマシューティカルズ,インコーポレイテッド Methods of treating inflammatory and autoimmune diseases with natalizumab
GB0605217D0 (en) 2006-03-15 2006-04-26 Novartis Ag Method and compositions for assessing acute rejection
US8231907B2 (en) 2006-03-21 2012-07-31 Morehouse School Of Medicine Nanoparticles for delivery of active agents
WO2008105773A2 (en) 2006-03-31 2008-09-04 Massachusetts Institute Of Technology System for targeted delivery of therapeutic agents
US8257685B2 (en) 2006-04-04 2012-09-04 Stc.Unm Swellable particles for drug delivery
WO2007120863A2 (en) 2006-04-14 2007-10-25 Epicentre Technologies Kits and methods for generating 5' capped rna
EP1852127A1 (en) 2006-05-02 2007-11-07 Charité - Universitätsmedizin Berlin Use of a B-cell-depleting antibody for treatment of polyoma virus infections
CN101573141B (en) 2006-05-15 2016-05-04 麻省理工学院 For the polymer of functional particles
JP2009537605A (en) 2006-05-24 2009-10-29 メルク セローノ ソシエテ アノニム Cladribine regimen for the treatment of multiple sclerosis
CN104356230A (en) 2006-06-02 2015-02-18 哈佛大学校长及研究员协会 Protein surface remodeling
US9506056B2 (en) 2006-06-08 2016-11-29 Northwestern University Nucleic acid functionalized nanoparticles for therapeutic applications
EP2046383B1 (en) 2006-07-04 2014-11-19 Genmab A/S Cd20 binding molecules for the treatment of copd
EP2397123A1 (en) 2006-07-07 2011-12-21 Aarhus Universitet Nanoparticles for nucleic acid delivery
TWI441835B (en) 2006-07-12 2014-06-21 Novartis Ag Novel polymers
JP2009544287A (en) 2006-07-20 2009-12-17 ノバルティス アーゲー AMIGO-2 inhibitor for treating, diagnosing, or detecting cancer
JP5571380B2 (en) 2006-07-24 2014-08-13 ルミナス バイオサイエンシズ,インコーポレイテッド Solid nanoparticle formulations of water-insoluble pharmaceutical substances with reduced Ostwald ripening
CA2659301A1 (en) 2006-07-28 2008-02-07 Applera Corporation Dinucleotide mrna cap analogs
AU2007280690C1 (en) 2006-07-31 2012-08-23 Curevac Gmbh Nucleic acid of formula (I): GIXmGn, or (II): CIXmCn, in particular as an immune-stimulating agent/adjuvant
DE102006035618A1 (en) 2006-07-31 2008-02-07 Curevac Gmbh New nucleic acid useful as immuno-stimulating adjuvant for manufacture of a composition for treatment of cancer diseases e.g. colon carcinomas and infectious diseases e.g. influenza and malaria
RU2009108289A (en) 2006-08-07 2010-09-20 Джензим Корпорейшн (Us) COMBINED THERAPY
AU2007286059A1 (en) 2006-08-18 2008-02-21 Mdrna, Inc. Dicer substrate RNA peptide conjugates and methods for RNA therapeutics
US8658211B2 (en) 2006-08-18 2014-02-25 Arrowhead Madison Inc. Polyconjugates for in vivo delivery of polynucleotides
AU2007292221B2 (en) 2006-09-06 2013-08-29 The Regents Of The University Of California Selectively targeted antimicrobial peptides and the use thereof
KR101485197B1 (en) 2006-09-07 2015-01-23 크루셀 홀란드 비.브이. Human binding molecules capable of neutralizing influenza virus h5n1 and uses thereof
ES2360538T3 (en) 2006-09-08 2011-06-06 Johns Hopkins University COMPOSITIONS TO INCREASE TRANSPORTATION THROUGH MOCO.
US8454948B2 (en) 2006-09-14 2013-06-04 Medgenics Medical Israel Ltd. Long lasting drug formulations
GB0619182D0 (en) 2006-09-29 2006-11-08 Leuven K U Res & Dev Oligonucleotide arrays
KR101129509B1 (en) 2006-10-03 2012-04-13 알닐람 파마슈티칼스 인코포레이티드 Lipid containing formulations
CN101583379B (en) 2006-10-05 2013-04-03 约翰斯霍普金斯大学 Water-dispersible oral, parenteral, and topical formulations for poorly water soluble drugs using smart polymeric nanoparticles
DE102006051516A1 (en) * 2006-10-31 2008-05-08 Curevac Gmbh (Base) modified RNA to increase the expression of a protein
US8414927B2 (en) 2006-11-03 2013-04-09 Boston Scientific Scimed, Inc. Cross-linked polymer particles
US7999087B2 (en) 2006-11-15 2011-08-16 Agilent Technologies, Inc. 2′-silyl containing thiocarbonate protecting groups for RNA synthesis
US8242258B2 (en) 2006-12-03 2012-08-14 Agilent Technologies, Inc. Protecting groups for RNA synthesis
US8399007B2 (en) 2006-12-05 2013-03-19 Landec Corporation Method for formulating a controlled-release pharmaceutical formulation
CN101553252A (en) 2006-12-06 2009-10-07 诺华有限公司 Vaccines including antigen from four strains of influenza virus
US9034348B2 (en) 2006-12-11 2015-05-19 Chi2Gel Ltd. Injectable chitosan mixtures forming hydrogels
KR102223231B1 (en) 2006-12-18 2021-03-08 악셀레론 파마 인코포레이티드 Activin-actrii antagonists and uses for increasing red blood cell levels
EP2104739B1 (en) 2006-12-21 2013-06-19 Novozymes Inc. Modified messenger rna stabilizing sequences for expressing genes in bacterial cells
CA2671925A1 (en) 2006-12-21 2008-07-10 Stryker Corporation Sustained-release formulations comprising crystals, macromolecular gels, and particulate suspensions of biologic agents
WO2008078180A2 (en) 2006-12-22 2008-07-03 Archemix Corp. Materials and methods for the generation of transcripts comprising modified nucleotides
DE102006061015A1 (en) 2006-12-22 2008-06-26 Curevac Gmbh Process for the purification of RNA on a preparative scale by HPLC
US8338166B2 (en) 2007-01-04 2012-12-25 Lawrence Livermore National Security, Llc Sorting, amplification, detection, and identification of nucleic acid subsequences in a complex mixture
DE102007001370A1 (en) * 2007-01-09 2008-07-10 Curevac Gmbh RNA-encoded antibodies
WO2008091799A2 (en) 2007-01-22 2008-07-31 The Trustees Of Columbia University In The City Of New York Cell-based methods for identifying inhibitors of parkinson's disease-associated lrrk2 mutants
CN103755789B (en) 2007-01-30 2016-12-07 埃皮瓦克斯公司 Regulatory t cell epitopes, compositions and application thereof
TWI782836B (en) 2007-02-02 2022-11-01 美商艾瑟勒朗法瑪公司 Variants derived from actriib and uses therefor
WO2008097926A2 (en) 2007-02-02 2008-08-14 Yale University Transient transfection with rna
WO2008096370A2 (en) 2007-02-05 2008-08-14 Natco Pharma Limited An efficient and novel purification method of recombinant hg-csf
US8333799B2 (en) 2007-02-12 2012-12-18 C. R. Bard, Inc. Highly flexible stent and method of manufacture
US8242087B2 (en) 2007-02-27 2012-08-14 K.U.Leuven Research & Development Phosphate modified nucleosides useful as substrates for polymerases and as antiviral agents
EP1964922A1 (en) * 2007-03-02 2008-09-03 Boehringer Ingelheim Pharma GmbH & Co. KG Improvement of protein production
EP2522729A1 (en) * 2007-03-02 2012-11-14 Boehringer Ingelheim Pharma GmbH & Co. KG Improvement of protein production
JP5249248B2 (en) 2007-03-05 2013-07-31 ワシントン ユニバーシティー Nanoparticle transport system for membrane-embedded peptides
WO2008115504A2 (en) 2007-03-20 2008-09-25 Millennium Pharmaceuticals, Inc. Nucleic acids encoding humanized immunoglobulin that binds a4b7 integrin
SI2152358T1 (en) 2007-04-27 2011-08-31 Echo Therapeutics Inc Skin permeation device for analyte sensing or transdermal drug delivery
DK2152290T3 (en) 2007-04-30 2014-08-18 Glaxosmithkline Llc PROCEDURES FOR ADMINISTRATION OF ANTI-IL-5 ANTIBODIES
US8703204B2 (en) 2007-05-03 2014-04-22 Bend Research, Inc. Nanoparticles comprising a cholesteryl ester transfer protein inhibitor and anon-ionizable polymer
CA2686735A1 (en) 2007-05-04 2008-11-13 Mdrna, Inc. Amino acid lipids and uses thereof
US7682789B2 (en) 2007-05-04 2010-03-23 Ventana Medical Systems, Inc. Method for quantifying biomolecules conjugated to a nanoparticle
US8728491B2 (en) 2007-05-07 2014-05-20 Alba Therapeutics Corporation Transcutaneous delivery of therapeutic agents
JP5296328B2 (en) 2007-05-09 2013-09-25 独立行政法人理化学研究所 Single-stranded circular RNA and method for producing the same
DK2476689T3 (en) 2007-05-10 2016-01-18 Agilent Technologies Inc Thiocarbonbeskyttende groups for RNA synthesis
RU2519227C2 (en) 2007-05-14 2014-06-10 Медиммун, Ллк Method for reducing eosinophil count
WO2008144365A2 (en) 2007-05-17 2008-11-27 Novartis Ag Method for making dry powder compositions containing ds-rna based on supercritical fluid technology
BRPI0812384A2 (en) 2007-05-22 2014-12-02 Novartis Ag TREATMENT, DIAGNOSTIC AND DISEASE TREATMENT METHODS ASSOCIATED WITH FGF21
EP2164951A2 (en) 2007-05-30 2010-03-24 The General Hospital Corporation Methods of generating pluripotent cells from somatic cells
SI2167523T1 (en) 2007-06-19 2014-09-30 Board Of Supervisors Of Louisiana State University And Agricultural And Mechanical College Synthesis and use of anti-reverse phosphorothioate analogs of the messenger rna cap
US8039214B2 (en) 2007-06-29 2011-10-18 Cellscript, Inc. Synthesis of tagged nucleic acids
WO2009015071A1 (en) 2007-07-23 2009-01-29 Dharmacon, Inc. Screening of micro-rna cluster inhibitor pools
US9144546B2 (en) 2007-08-06 2015-09-29 Clsn Laboratories, Inc. Nucleic acid-lipopolymer compositions
US20090042825A1 (en) 2007-08-06 2009-02-12 Majed Matar Composition, method of preparation & application of concentrated formulations of condensed nucleic acids with a cationic lipopolymer
MX2010002113A (en) 2007-08-23 2010-03-26 Novartis Ag Methods for detecting oligonucleotides.
WO2009030254A1 (en) 2007-09-04 2009-03-12 Curevac Gmbh Complexes of rna and cationic peptides for transfection and for immunostimulation
CA2697482C (en) 2007-09-05 2016-05-31 F. Hoffmann-La Roche Ag Combination therapy with type i and type ii anti-cd20 antibodies
US8506928B2 (en) 2007-09-07 2013-08-13 The Regents Of The University Of California Methods and compounds for targeting tissues
WO2009039198A2 (en) 2007-09-17 2009-03-26 The Trustees Of The University Of Pennsylvania Generation of hyperstable mrnas
EP3156414B1 (en) 2007-09-26 2019-12-04 Intrexon Corporation Synthetic 5'utrs, expression vectors, and methods for increasing transgene expression
CA2700536A1 (en) 2007-09-26 2009-04-02 Oregon Health & Science University Cyclic undecapeptides and derivatives as multiple sclerosis therapies
EP2042193A1 (en) 2007-09-28 2009-04-01 Biomay AG RNA Vaccines
ES2647538T3 (en) 2007-09-28 2017-12-22 Pfizer Inc. Addressing to cancer cells using nanoparticles
US8470560B2 (en) 2007-10-03 2013-06-25 The United States Of America As Represented By The Secretary Of The Army CR-2 binding peptide P28 as molecular adjuvant for DNA vaccines
WO2009046739A1 (en) 2007-10-09 2009-04-16 Curevac Gmbh Composition for treating prostate cancer (pca)
WO2009046738A1 (en) 2007-10-09 2009-04-16 Curevac Gmbh Composition for treating lung cancer, particularly of non-small lung cancers (nsclc)
BRPI0817664A2 (en) 2007-10-12 2015-03-24 Massachusetts Inst Technology Nanoparticles, method for preparing nanoparticles and method for therapeutically or prophylactically treating an individual
US20090098118A1 (en) 2007-10-15 2009-04-16 Thomas Friess Combination therapy of a type ii anti-cd20 antibody with an anti-bcl-2 active agent
WO2009052830A1 (en) 2007-10-22 2009-04-30 Genmab A/S Novel antibody therapies
AU2008319183B2 (en) 2007-11-01 2014-09-04 University Of Rochester Recombinant factor VIII having increased stability
WO2009062054A1 (en) 2007-11-09 2009-05-14 Novartis Ag Uses of anti-cd40 antibodies
US8470771B2 (en) 2007-11-14 2013-06-25 Institute Of Microbiology, Chinese Academy Of Sciences Method and medicament for inhibiting the infection of influenza virus
CN101932608A (en) 2007-11-30 2010-12-29 葛兰素集团有限公司 Antigen-binding constructs
EP2617828B1 (en) 2007-12-10 2014-09-24 Alnylam Pharmaceuticals Inc. Compositions and methods for inhibiting expression of factor VII gene
CA2904904A1 (en) 2007-12-11 2009-06-18 The Scripps Research Institute Compositions and methods related to mrna translational enhancer elements
EP2229459B1 (en) 2007-12-13 2014-08-27 Alnylam Pharmaceuticals, Inc. Methods and compositions for prevention or treatment of RSV infection
EP2072618A1 (en) 2007-12-14 2009-06-24 Johannes Gutenberg-Universität Mainz Use of RNA for reprogramming somatic cells
AU2008342956A1 (en) 2007-12-21 2009-07-09 Genentech, Inc. Therapy of rituximab-refractory rheumatoid arthritis patients
WO2009093703A1 (en) 2008-01-23 2009-07-30 Ajinomoto Co., Inc. Method of producing l-amino acid
JPWO2009093384A1 (en) 2008-01-24 2011-05-26 独立行政法人産業技術総合研究所 Polynucleotide, polynucleotide analogue and gene expression control method using the same
EP2548960B1 (en) 2008-01-31 2018-01-31 CureVac AG Nucleic acids comprising formula (nugixmgnv)a and derivatives thereof as an immunostimulating agents/adjuvant
EP2250252A2 (en) 2008-02-11 2010-11-17 Cambridge Enterprise Limited Improved reprogramming of mammalian cells, and the cells obtained
DK2240155T3 (en) 2008-02-13 2012-09-17 Intarcia Therapeutics Inc Devices, formulations and methods for the delivery of several beneficial agents
DE102008009920A1 (en) 2008-02-15 2009-08-20 Aj Innuscreen Gmbh Mobile device for nucleic acid isolation
US20120027813A1 (en) 2008-02-22 2012-02-02 Novartis Vaccines And Diagnostics Srl Adjuvanted influenza vaccines for pediatric use
US8506966B2 (en) 2008-02-22 2013-08-13 Novartis Ag Adjuvanted influenza vaccines for pediatric use
WO2009108891A2 (en) 2008-02-29 2009-09-03 Egen, Inc. Modified poloxamers for gene expression and associated methods
US20100004315A1 (en) 2008-03-14 2010-01-07 Gregory Slobodkin Biodegradable Cross-Linked Branched Poly(Alkylene Imines)
CA2716919C (en) 2008-03-14 2015-01-20 Biocon Limited An anti-cd6 monoclonal antibody and use thereof
WO2009120927A2 (en) 2008-03-28 2009-10-01 Smithkline Beecham Corporation Methods of treatment
PT2279254T (en) 2008-04-15 2017-09-04 Protiva Biotherapeutics Inc Novel lipid formulations for nucleic acid delivery
US8323686B2 (en) 2008-04-25 2012-12-04 Northwestern University Nanostructures suitable for sequestering cholesterol and other molecules
WO2009134808A2 (en) 2008-04-28 2009-11-05 President And Fellows Of Harvard College Supercharged proteins for cell penetration
US8715689B2 (en) 2008-04-30 2014-05-06 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services, Centers For Disease Control And Prevention Chimeric west nile/dengue viruses
US9394538B2 (en) 2008-05-07 2016-07-19 Shi-Lung Lin Development of universal cancer drugs and vaccines
US8486278B2 (en) 2008-05-08 2013-07-16 Minipumps, Llc Drug-delivery pumps and methods of manufacture
US8697098B2 (en) 2011-02-25 2014-04-15 South Dakota State University Polymer conjugated protein micelles
CA2724105C (en) 2008-05-13 2017-09-05 University Of Washington Diblock copolymers and polynucleotide complexes thereof for delivery into cells
EP2297323A1 (en) 2008-05-21 2011-03-23 Hartmann, Gunther 5' triphosphate oligonucleotide with blunt end and uses thereof
FR2931824B1 (en) 2008-05-29 2014-11-28 Centre Nat Rech Scient PROCESS FOR RNA SYNTHESIS THROUGH CHEMICAL.
ATE548381T1 (en) 2008-05-29 2012-03-15 Hanall Biopharma Co Ltd MODIFIED ERYTHROPOIETIN (EPO) POLYPEPTIDES WITH INCREASED PROTEASE RESISTANCE AND PHARMACEUTICAL COMPOSITIONS THEREOF
WO2009148528A2 (en) 2008-05-30 2009-12-10 Millennium Pharmaceuticals, Inc. Assessment of chromosomal alterations to predict clinical outcome of bortezomib treatment
PL215513B1 (en) 2008-06-06 2013-12-31 Univ Warszawski New borane phosphate analogs of dinucleotides, their application, RNA particle, method of obtaining RNA and method of obtaining peptides or protein
TWI451876B (en) 2008-06-13 2014-09-11 Lilly Co Eli Pegylated insulin lispro compounds
HUE035770T2 (en) 2008-06-16 2018-05-28 Pfizer Methods for the preparation of targeting agent functionalized diblock copolymers for use in fabrication of therapeutic nanoparticles
US8613951B2 (en) 2008-06-16 2013-12-24 Bind Therapeutics, Inc. Therapeutic polymeric nanoparticles with mTor inhibitors and methods of making and using same
PT2774608T (en) 2008-06-16 2020-01-17 Pfizer Drug loaded polymeric nanoparticles and methods of making and using same
US20100009424A1 (en) 2008-07-14 2010-01-14 Natasha Forde Sonoporation systems and methods
WO2010009065A2 (en) 2008-07-15 2010-01-21 Novartis Ag Amphipathic peptide compositions
EP2313085A2 (en) 2008-07-15 2011-04-27 Novartis AG Immunogenic amphipathic peptide compositions
AU2009290137A1 (en) 2008-09-03 2010-03-11 Xenome Ltd Libraries of peptide conjugates and methods for making them
JP5714490B2 (en) 2008-09-06 2015-05-07 ケムジーンズ コーポレーション Methods for preparing oligonucleotides by RNA synthesis, phosphoramidites for synthetic RNA in the reverse direction, and bond formation in the 5 'to 3' direction for synthesis of RNA oligomers
US20120100558A1 (en) 2008-09-08 2012-04-26 Hanash Samir M Lung cancer diagnosis
US20100087337A1 (en) 2008-09-10 2010-04-08 Bind Biosciences, Inc. High Throughput Fabrication of Nanoparticles
TW201438738A (en) 2008-09-16 2014-10-16 Genentech Inc Methods for treating progressive multiple sclerosis
WO2010033906A2 (en) 2008-09-19 2010-03-25 President And Fellows Of Harvard College Efficient induction of pluripotent stem cells using small molecule compounds
WO2010037408A1 (en) 2008-09-30 2010-04-08 Curevac Gmbh Composition comprising a complexed (m)rna and a naked mrna for providing or enhancing an immunostimulatory response in a mammal and uses thereof
WO2010042490A1 (en) 2008-10-06 2010-04-15 Boston Medical Center Corporation A single lentiviral vector system for induced pluripotent (ips) stem cells derivation
PL2350043T3 (en) 2008-10-09 2014-09-30 Tekmira Pharmaceuticals Corp Improved amino lipids and methods for the delivery of nucleic acids
US8535655B2 (en) 2008-10-10 2013-09-17 Polyactiva Pty Ltd. Biodegradable polymer—bioactive moiety conjugates
US8343498B2 (en) 2008-10-12 2013-01-01 Massachusetts Institute Of Technology Adjuvant incorporation in immunonanotherapeutics
US8603532B2 (en) 2008-10-20 2013-12-10 Massachusetts Institute Of Technology Nanostructures for drug delivery
WO2010047839A1 (en) 2008-10-25 2010-04-29 Aura Biosciences Modified plant virus particles and uses therefor
MX353900B (en) 2008-11-07 2018-02-01 Massachusetts Inst Technology Aminoalcohol lipidoids and uses thereof.
JP5832898B2 (en) 2008-11-10 2015-12-16 テクミラ ファーマシューティカルズ コーポレイションTekmira Pharmaceuticals Corporation Novel lipids and compositions for delivering therapeutic agents
WO2010057203A2 (en) 2008-11-17 2010-05-20 The Board Of Regents Of The University Of Texas System Hdl particles for delivery of nucleic acids
EP2191840A1 (en) 2008-11-28 2010-06-02 Sanofi-Aventis Antitumor combinations containing antibodies recognizing specifically CD38 and melphalan
EP2196476A1 (en) 2008-12-10 2010-06-16 Novartis Ag Antibody formulation
JP5855462B2 (en) 2008-12-10 2016-02-09 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. DsRNA compositions targeting GNAQ and methods for inhibiting expression
US8512964B2 (en) 2008-12-12 2013-08-20 The Regents Of The University Of California Targets for treatment of hypercholesterolemia
WO2010068866A2 (en) 2008-12-12 2010-06-17 Bind Biosciences Therapeutic particles suitable for parenteral administration and methods of making and using same
JP2012512175A (en) 2008-12-15 2012-05-31 バインド バイオサイエンシズ インコーポレイテッド Long-circulating nanoparticles for sustained release of therapeutic agents
BRPI1006829A2 (en) 2009-01-16 2016-10-25 Glaxosmithkline Llc cancer treatment employing a combination of bendamustine and an anti-cd20 antibody
WO2010084371A1 (en) 2009-01-26 2010-07-29 Mitoprod Novel circular interfering rna molecules
US20120101148A1 (en) 2009-01-29 2012-04-26 Alnylam Pharmaceuticals, Inc. lipid formulation
WO2010088927A1 (en) 2009-02-09 2010-08-12 Curevac Gmbh Use of pei for the improvement of endosomal release and expression of transfected nucleic acids, complexed with cationic or polycationic compounds
US20140141089A1 (en) 2009-02-11 2014-05-22 Colorado School Of Mines Nanoparticles, Compositions Thereof, and Methods of Use, and Methods of Making the Same
JP5735927B2 (en) 2009-02-24 2015-06-17 ザ スクリプス リサーチ インスティテュート Re-engineering the primary structure of mRNA to enhance protein production
WO2010102065A1 (en) 2009-03-05 2010-09-10 Bend Research, Inc. Pharmaceutical compositions of dextran polymer derivatives
WO2010141135A2 (en) 2009-03-05 2010-12-09 Trustees Of Boston University Bacteriophages expressing antimicrobial peptides and uses thereof
EP3235509A1 (en) 2009-03-05 2017-10-25 AbbVie Inc. Il-17 binding proteins
AU2010223888A1 (en) 2009-03-13 2011-10-06 Egen, Inc. Compositions and methods for the delivery of biologically active RNAs
WO2010108108A2 (en) 2009-03-20 2010-09-23 Egen, Inc. Polyamine derivatives
JP5622254B2 (en) 2009-03-31 2014-11-12 国立大学法人東京大学 Double-stranded ribonucleic acid polyion complex
SI3281947T1 (en) 2009-04-03 2020-07-31 The University Of Chicago Compositions and methods related to protein a (spa) variants
WO2011127032A1 (en) 2010-04-05 2011-10-13 University Of Chicago Compositions and methods related to protein a (spa) antibodies as an enhancer of immune response
US9724404B2 (en) 2009-04-13 2017-08-08 INSERM (Institut National de la Santé et de la Recherche Médicale) HPV particles and uses thereof
EP2419137A4 (en) 2009-04-17 2013-01-09 Biogen Idec Inc Compositions and methods to treat acute myelogenous leukemia
US20100273220A1 (en) 2009-04-22 2010-10-28 Massachusetts Institute Of Technology Innate immune suppression enables repeated delivery of long rna molecules
EA027071B1 (en) 2009-04-27 2017-06-30 Новартис Аг ANTI-ActRIIB ANTIBODY AND COMPOSITION COMPRISING SAME
US20110033389A1 (en) 2009-04-29 2011-02-10 Zhifeng Chen Modified antibodies for passive immunotherapy
WO2010127159A2 (en) 2009-04-30 2010-11-04 Intezyne Technologies, Incorporated Polymeric micelles for polynucleotide encapsulation
US8715736B2 (en) 2009-04-30 2014-05-06 Florida Agricultural And Mechanical University Nanoparticle formulations for skin delivery
NZ711583A (en) 2009-05-05 2017-03-31 Arbutus Biopharma Corp Lipid compositions
DE202009007116U1 (en) 2009-05-18 2010-10-14 Amoena Medizin-Orthopädie-Technik GmbH Anti decubitus cushions
US8574835B2 (en) 2009-05-29 2013-11-05 Life Technologies Corporation Scaffolded nucleic acid polymer particles and methods of making and using
CN102625696B (en) 2009-06-10 2015-06-03 阿尔尼拉姆医药品有限公司 Improved lipid formulation
EP2440556A1 (en) 2009-06-10 2012-04-18 Vertex Pharmaceuticals Incorporated Inhibitors of phosphatidylinositol 3-kinase
MX2011013421A (en) 2009-06-15 2012-03-16 Alnylam Pharmaceuticals Inc Lipid formulated dsrna targeting the pcsk9 gene.
US20110097329A1 (en) 2009-06-26 2011-04-28 Massachusetts Institute Of Technology Compositions and methods for treating cancer and modulating stress granule formation
JP5766188B2 (en) 2009-07-01 2015-08-19 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 Lipid formulations for delivering therapeutic agents to solid tumors
US8569256B2 (en) 2009-07-01 2013-10-29 Protiva Biotherapeutics, Inc. Cationic lipids and methods for the delivery of therapeutic agents
US20110300205A1 (en) 2009-07-06 2011-12-08 Novartis Ag Self replicating rna molecules and uses thereof
CN105255881A (en) 2009-07-31 2016-01-20 埃泽瑞斯公司 Rna with a combination of unmodified and modified nucleotides for protein expression
EP2281579A1 (en) 2009-08-05 2011-02-09 BioNTech AG Vaccine composition comprising 5'-Cap modified RNA
US20110053829A1 (en) 2009-09-03 2011-03-03 Curevac Gmbh Disulfide-linked polyethyleneglycol/peptide conjugates for the transfection of nucleic acids
US20110070227A1 (en) 2009-09-18 2011-03-24 Anna-Marie Novotney-Barry Treatment of Autoimmune and Inflammatory Diseases
US8859284B2 (en) 2009-10-22 2014-10-14 The United States Of America, As Represented By The Secretary Of The Navy Delivery of nanoparticles to neurons
US8449916B1 (en) 2009-11-06 2013-05-28 Iowa State University Research Foundation, Inc. Antimicrobial compositions and methods
WO2011060250A1 (en) 2009-11-13 2011-05-19 Bend Research, Inc. Cationic dextran polymer derivatives
WO2011062965A2 (en) 2009-11-18 2011-05-26 University Of Washington Through Its Center For Commercialization Targeting monomers and polymers having targeting blocks
US8530429B2 (en) 2009-11-24 2013-09-10 Arch Cancer Therapeutics, Inc. Brain tumor targeting peptides and methods
DK3338765T3 (en) 2009-12-01 2019-03-04 Translate Bio Inc STEROID DERIVATIVE FOR THE SUPPLY OF MRNA IN HUMANGENETIC DISEASES
DE102009056884B4 (en) 2009-12-03 2021-03-18 Novartis Ag Vaccine Adjuvants and Improved Methods for Making Same
US20110245756A1 (en) 2009-12-03 2011-10-06 Rishi Arora Devices for material delivery, electroporation, sonoporation, and/or monitoring electrophysiological activity
CA3121739A1 (en) 2009-12-06 2011-06-09 Bioverativ Therapeutics Inc. Factor viii-fc chimeric and hybrid polypeptides, and methods of use thereof
CA3044884A1 (en) 2009-12-07 2011-06-16 Arbutus Biopharma Corporation Compositions for nucleic acid delivery
KR102171849B1 (en) 2009-12-07 2020-10-30 더 트러스티스 오브 더 유니버시티 오브 펜실바니아 Rna preparations comprising purified modified rna for reprogramming cells
US20130189741A1 (en) 2009-12-07 2013-07-25 Cellscript, Inc. Compositions and methods for reprogramming mammalian cells
WO2011069529A1 (en) 2009-12-09 2011-06-16 Curevac Gmbh Mannose-containing solution for lyophilization, transfection and/or injection of nucleic acids
WO2011069528A1 (en) 2009-12-09 2011-06-16 Curevac Gmbh Lyophilization of nucleic acids in lactate-containing solutions
JP5891175B2 (en) 2009-12-11 2016-03-22 バインド セラピューティックス インコーポレイテッド Stable formulation against lyophilization of therapeutic particles
EP2512487A4 (en) 2009-12-15 2013-08-07 Therapeutic polymeric nanoparticles comprising corticosteroids and methods of making and using same
US9295649B2 (en) 2009-12-15 2016-03-29 Bind Therapeutics, Inc. Therapeutic polymeric nanoparticle compositions with high glass transition temperature or high molecular weight copolymers
EA201290506A1 (en) 2009-12-16 2013-03-29 Брихэм Энд Уимен'З Хоспитал, Инк. PARTICLES FOR DELIVERY OF A SET OF AGENTS
DE102009058769A1 (en) 2009-12-16 2011-06-22 MagForce Nanotechnologies AG, 10589 Temperature-dependent activation of catalytic nucleic acids for controlled drug release
CA2784568A1 (en) 2009-12-18 2011-06-23 Martin A. Maier Lipid particles for delivery of nucleic acids
EP2338520A1 (en) 2009-12-21 2011-06-29 Ludwig Maximilians Universität Conjugate with targeting ligand and use of same
US20110200582A1 (en) 2009-12-23 2011-08-18 Novartis Ag Lipids, lipid compositions, and methods of using them
WO2011088309A1 (en) 2010-01-14 2011-07-21 Regulus Therapeutics Inc. Microrna compositions and methods
DK2539451T3 (en) 2010-02-24 2016-04-04 Arrowhead Res Corp Compositions for targeted delivery of siRNA
CA2791278C (en) 2010-02-25 2015-11-24 The Johns Hopkins University Sustained delivery of therapeutic agents to an eye compartment
US20130133483A1 (en) 2010-03-08 2013-05-30 University Of Rochester Synthesis of Nanoparticles Using Reducing Gases
AU2011227264A1 (en) 2010-03-16 2012-10-04 University Of Utah Research Foundation Cleavable modifications to reducible poly (amido ethylenimines)s to enhance nucleotide delivery
WO2011116072A1 (en) 2010-03-16 2011-09-22 Escape Therapeutics, Inc. Hybrid hydrogel scaffold compositions and methods of use
US20110230816A1 (en) 2010-03-18 2011-09-22 Tyco Healthcare Group Lp Gels for Transdermal Delivery
US9149432B2 (en) 2010-03-19 2015-10-06 Massachusetts Institute Of Technology Lipid vesicle compositions and methods of use
GB201005005D0 (en) 2010-03-25 2010-05-12 Angeletti P Ist Richerche Bio New vaccine
US8207290B2 (en) 2010-03-26 2012-06-26 Cerulean Pharma Inc. Methods and systems for generating nanoparticles
US8349308B2 (en) 2010-03-26 2013-01-08 Mersana Therapeutics, Inc. Modified polymers for delivery of polynucleotides, method of manufacture, and methods of use thereof
US20110247090A1 (en) 2010-04-02 2011-10-06 Intrexon Corporation Synthetic 5'UTRs, Expression Vectors, and Methods for Increasing Transgene Expression
WO2011127316A1 (en) 2010-04-07 2011-10-13 Novartis Ag Method for generating a parvovirus b19 virus-like particle
WO2011127255A1 (en) 2010-04-08 2011-10-13 Merck Sharp & Dohme Corp. Preparation of lipid nanoparticles
EP2555752B1 (en) 2010-04-09 2019-06-26 Pacira Pharmaceuticals, Inc. Method for formulating multivesicular liposomes
CA2795695A1 (en) 2010-04-09 2011-10-13 The University Of Tokyo Microrna-controlled recombinant vaccinia virus and use thereof
KR101196667B1 (en) 2010-04-15 2012-11-02 포항공과대학교 산학협력단 A DELEVERY SYSTEM OF ANTI-CANCER AGENT USING pH SENSITIVE METAL NANOPARTICLE
EP2377938A1 (en) 2010-04-16 2011-10-19 Eukarys Capping-prone RNA polymerase enzymes and their applications
EP2558571A4 (en) 2010-04-16 2014-09-24 Immune Disease Inst Inc Sustained polypeptide expression from synthetic, modified rnas and uses thereof
SI2558577T1 (en) 2010-04-16 2019-05-31 Nuevolution A/S Bi-functional complexes and methods for making and using such complexes
WO2011133868A2 (en) 2010-04-22 2011-10-27 Alnylam Pharmaceuticals, Inc. Conformationally restricted dinucleotide monomers and oligonucleotides
EP2563455A4 (en) 2010-04-28 2014-02-19 Kimberly Clark Co Method for increasing permeability of an epithelial barrier
US20130156845A1 (en) 2010-04-29 2013-06-20 Alnylam Pharmaceuticals, Inc. Lipid formulated single stranded rna
US20130052644A1 (en) 2010-04-30 2013-02-28 Baltazar Gomez-Mancilla Predictive Markers Useful in the Treatment of Fragile X Syndrome (FXS)
US9254327B2 (en) 2010-05-10 2016-02-09 Alnylam Pharmaceuticals, Inc. Methods and compositions for delivery of active agents
CA2799091A1 (en) 2010-05-12 2011-11-17 Protiva Biotherapeutics, Inc. Cationic lipids and methods of use thereof
WO2011141704A1 (en) 2010-05-12 2011-11-17 Protiva Biotherapeutics, Inc Novel cyclic cationic lipids and methods of use
EP2387999A1 (en) 2010-05-21 2011-11-23 CureVac GmbH Histidine-containing solution for transfection and/or injection of nucleic acids and uses thereof
JP2013531634A (en) 2010-05-24 2013-08-08 メルク・シャープ・エンド・ドーム・コーポレイション Novel aminoalcohol cationic lipids for oligonucleotide delivery
JP5957646B2 (en) 2010-06-04 2016-07-27 サーナ・セラピューティクス・インコーポレイテッドSirna Therapeutics,Inc. Novel low molecular weight cationic lipids for oligonucleotide delivery
AU2011267078B2 (en) 2010-06-14 2014-09-25 F. Hoffmann-La Roche Ag Cell-penetrating peptides and uses therof
US20130236968A1 (en) 2010-06-21 2013-09-12 Alnylam Pharmaceuticals, Inc. Multifunctional copolymers for nucleic acid delivery
US10478483B2 (en) 2010-06-25 2019-11-19 Glaxosmithkline Biologicals Sa Combinations of meningococcal factor H binding proteins
US9066971B2 (en) 2010-07-01 2015-06-30 Postech Academy-Industry Foundation Method for treating and diagnosing cancer by using cell-derived microvesicles
EP2588120B1 (en) 2010-07-02 2017-11-15 The University of Chicago Compositions and methods related to protein a (spa) variants
ES2649896T3 (en) 2010-07-06 2018-01-16 Glaxosmithkline Biologicals Sa Cationic emulsions of oil in water
WO2012006369A2 (en) 2010-07-06 2012-01-12 Novartis Ag Immunisation of large mammals with low doses of rna
RS63817B1 (en) 2010-07-06 2023-01-31 Glaxosmithkline Biologicals Sa Virion-like delivery particles for self-replicating rna molecules
DK3243526T3 (en) 2010-07-06 2020-02-17 Glaxosmithkline Biologicals Sa SUPPLY OF RNA TO TRACT MULTIPLE IMMUNE SIGNAL WAYS
US9192661B2 (en) 2010-07-06 2015-11-24 Novartis Ag Delivery of self-replicating RNA using biodegradable polymer particles
US9770463B2 (en) 2010-07-06 2017-09-26 Glaxosmithkline Biologicals Sa Delivery of RNA to different cell types
CN103153284B (en) 2010-07-06 2015-11-25 诺华股份有限公司 The liposome of the lipid of RNA delivery is conducive to containing its pKa value
MX356527B (en) 2010-07-09 2018-06-01 Bioverativ Therapeutics Inc Factor ix polypeptides and methods of use thereof.
US9611310B2 (en) 2010-07-09 2017-04-04 Bioverativ Therapeutics Inc. Systems for factor VIII processing and methods thereof
US20130177523A1 (en) 2010-07-13 2013-07-11 University Of Utah Research Foundation Gold particles and methods of making and using the same in cancer treatment
GB201012410D0 (en) 2010-07-23 2010-09-08 Medical Res Council Intracellular immunity
CA2801523C (en) 2010-07-30 2021-08-03 Curevac Gmbh Complexation of nucleic acids with disulfide-crosslinked cationic components for transfection and immunostimulation
US8822663B2 (en) 2010-08-06 2014-09-02 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US9121065B2 (en) 2010-08-09 2015-09-01 The Trustees Of The University Of Pennsylvania Nanoparticle-oligonucleotide hybrid structures and methods of use thereof
WO2012019630A1 (en) 2010-08-13 2012-02-16 Curevac Gmbh Nucleic acid comprising or coding for a histone stem-loop and a poly(a) sequence or a polyadenylation signal for increasing the expression of an encoded protein
EP2605792B1 (en) 2010-08-20 2014-12-10 Novartis AG Soluble needle arrays for delivery of influenza vaccines
WO2012024595A2 (en) 2010-08-20 2012-02-23 University Of Washington Circumferential aerosol device for delivering drugs to olfactory epithelium and brain
AU2011291582A1 (en) 2010-08-20 2013-03-07 Cerulean Pharma Inc. Conjugates, particles, compositions, and related methods
EP2611420B1 (en) 2010-08-31 2019-03-27 GlaxoSmithKline Biologicals SA Lipids suitable for liposomal delivery of protein-coding rna
BR112013004879A2 (en) 2010-08-31 2018-04-24 Novartis Ag small liposomes for delivery of immunogen encoding RNA
HUE058667T2 (en) 2010-08-31 2022-09-28 Glaxosmithkline Biologicals Sa Pegylated liposomes for delivery of immunogen-encoding rna
CA3109036C (en) 2010-08-31 2023-08-01 Theraclone Sciences, Inc. Human immunodeficiency virus (hiv)-neutralizing antibodies
JP2013541510A (en) 2010-08-31 2013-11-14 メルク・シャープ・エンド・ドーム・コーポレイション Novel single chemical and oligonucleotide delivery method
WO2012031205A2 (en) 2010-09-03 2012-03-08 The Brigham And Women's Hospital, Inc. Lipid-polymer hybrid particles
WO2012034067A1 (en) 2010-09-09 2012-03-15 The University Of Chicago Methods and compositions involving protective staphylococcal antigens
US20130236419A1 (en) 2010-09-09 2013-09-12 The University Of Chicago Compositions and methods related to attenuated staphylococcal strains
US10307372B2 (en) 2010-09-10 2019-06-04 The Johns Hopkins University Rapid diffusion of large polymeric nanoparticles in the mammalian brain
US8466122B2 (en) 2010-09-17 2013-06-18 Protiva Biotherapeutics, Inc. Trialkyl cationic lipids and methods of use thereof
MX349088B (en) 2010-09-20 2017-07-10 Merck Sharp & Dohme Novel low molecular weight cationic lipids for oligonucleotide delivery.
US20130183718A1 (en) 2010-09-21 2013-07-18 RibpxX GmbH Method for Synthesizing RNA using DNA Template
BR112013008697A2 (en) 2010-09-24 2016-06-21 Massachusetts Inst Technology nanostructured gels capable of controlled release of encapsulated agents
WO2012050975A2 (en) 2010-09-29 2012-04-19 The University Of North Carolina At Chapel Hill Novel circular mammalian rna molecules and uses thereof
CN103260611A (en) 2010-09-30 2013-08-21 默沙东公司 Low molecular weight cationic lipids for oligonucleotide delivery
EP2625189B1 (en) 2010-10-01 2018-06-27 ModernaTX, Inc. Engineered nucleic acids and methods of use thereof
WO2013086505A1 (en) 2011-12-09 2013-06-13 Vanderbilt University Integrated organ-on-chip system and applications of the same
EP2627351B1 (en) 2010-10-11 2018-12-26 GlaxoSmithKline Biologicals SA Antigen delivery platforms
WO2012053297A1 (en) 2010-10-19 2012-04-26 三菱電機株式会社 Laser processing machine control device and laser processing machine control method
US9029590B2 (en) 2010-10-21 2015-05-12 Sirna Therapeutics, Inc. Low molecular weight cationic lipids for oligonucleotide delivery
JP6018575B2 (en) 2010-10-29 2016-11-02 メルク・シャープ・アンド・ドーム・コーポレーションMerck Sharp & Dohme Corp. Dengue virus recombinant subunit vaccine
AU2011323250B2 (en) 2010-11-05 2015-11-19 The Johns Hopkins University Compositions and methods relating to reduced mucoadhesion
AU2011326732B2 (en) 2010-11-09 2016-07-21 The Regents Of The University Of California Skin permeating and cell entering (space) peptides and methods of use thereof
PL3536703T3 (en) 2010-11-12 2021-10-25 The Trustees Of The University Of Pennsylvania Consensus prostate antigens, nucleic acid molecules encoding the same and uses thereof
AU2011329850B2 (en) 2010-11-16 2017-03-02 Selecta Biosciences, Inc. Immunostimulatory oligonucleotides
LT2640842T (en) 2010-11-17 2018-09-10 Aduro Biotech, Inc. Methods and compositions for inducing an immune response to egfrviii
KR102100110B1 (en) 2010-11-19 2020-04-14 이데라 파마슈티칼즈, 인코포레이티드 Immune regulatory oligonucleotide(iro) compounds to modulate toll-like receptor based immune response
WO2012075040A2 (en) 2010-11-30 2012-06-07 Shire Human Genetic Therapies, Inc. mRNA FOR USE IN TREATMENT OF HUMAN GENETIC DISEASES
WO2012072096A1 (en) 2010-12-03 2012-06-07 Biontech Ag Method for cellular rna expression
CA2821622C (en) 2010-12-16 2019-09-03 Sprna Gmbh Pharmaceutical composition consisting of rna having alkali metal as counter ion and formulated with dications
US8501930B2 (en) 2010-12-17 2013-08-06 Arrowhead Madison Inc. Peptide-based in vivo siRNA delivery system
AU2011348204B2 (en) 2010-12-22 2017-03-02 President And Fellows Of Harvard College Continuous directed evolution
CN104328121A (en) 2010-12-29 2015-02-04 弗·哈夫曼-拉罗切有限公司 Small molecule conjugates for intracellular delivery of nucleic acids
WO2012089225A1 (en) 2010-12-29 2012-07-05 Curevac Gmbh Combination of vaccination and inhibition of mhc class i restricted antigen presentation
US10364440B2 (en) 2011-01-04 2019-07-30 Brown University Nanotubes as carriers of nucleic acids into cells
WO2012094574A2 (en) 2011-01-06 2012-07-12 The Johns Hopkins University Stabilized polyribonucleotide nanoparticles
US20140080766A1 (en) 2011-01-07 2014-03-20 Massachusetts Institute Of Technology Compositions and methods for macromolecular drug delivery
DK2663548T3 (en) 2011-01-11 2017-07-24 Alnylam Pharmaceuticals Inc PEGYLED LIPIDS AND THEIR USE FOR PHARMACEUTICAL SUPPLY
US20120189700A1 (en) 2011-01-19 2012-07-26 Zoraida Aguilar Nanoparticle Based Immunological Stimulation
WO2012101235A1 (en) 2011-01-26 2012-08-02 Cenix Bioscience Gmbh Delivery system and conjugates for compound delivery via naturally occurring intracellular transport routes
US10363309B2 (en) 2011-02-04 2019-07-30 Case Western Reserve University Targeted nanoparticle conjugates
WO2012109121A1 (en) 2011-02-07 2012-08-16 Purdue Research Foundation Carbohydrate nanoparticles for prolonged efficacy of antimicrobial peptide
WO2012116715A1 (en) 2011-03-02 2012-09-07 Curevac Gmbh Vaccination in newborns and infants
US20120207840A1 (en) 2011-02-10 2012-08-16 Aura Biosciences, Inc. Virion Derived Protein Nanoparticles For Delivering Diagnostic Or Therapeutic Agents For The Treatment Of Non-Melanoma Skin Cancer
CN103703013A (en) 2011-02-14 2014-04-02 斯威夫特生物科学公司 Polynucleotide primers and probes
CA2827118A1 (en) 2011-02-15 2012-08-23 Merrimack Pharmaceuticals, Inc. Compositions and methods for delivering nucleic acid to a cell
US20140081012A1 (en) 2011-02-15 2014-03-20 The University Of North Carolina At Chapel Hill Nanoparticle, liposomes, polymers, agents and proteins modified with reversible linkers
EP2489371A1 (en) 2011-02-18 2012-08-22 Instituto Nacional de Investigacion y Tecnologia Agraria y Alimentaria Carrier peptides for drug delivery
WO2012113413A1 (en) 2011-02-21 2012-08-30 Curevac Gmbh Vaccine composition comprising complexed immunostimulatory nucleic acids and antigens packaged with disulfide-linked polyethyleneglycol/peptide conjugates
JP6091435B2 (en) 2011-02-22 2017-03-08 カリフォルニア インスティチュート オブ テクノロジー Protein delivery using adeno-associated virus (AAV) vectors
US8696637B2 (en) 2011-02-28 2014-04-15 Kimberly-Clark Worldwide Transdermal patch containing microneedles
US20140112950A1 (en) 2011-03-02 2014-04-24 Manmohan Singh Combination vaccines with lower doses of antigen and/or adjuvant
WO2012116714A1 (en) 2011-03-02 2012-09-07 Curevac Gmbh Vaccination in elderly patients
CA2832807A1 (en) 2011-03-07 2012-09-13 Massachusetts Institute Of Technology Methods for transfecting cells with nucleic acids
WO2012125680A1 (en) 2011-03-16 2012-09-20 Novartis Ag Methods of treating vasculitis using an il-17 binding molecule
WO2012125812A1 (en) 2011-03-17 2012-09-20 Novartis Ag Fgfr and ligands thereof as biomarkers for breast cancer in hr positive subjects
WO2012125987A2 (en) 2011-03-17 2012-09-20 Massachusetts Institute Of Technology Delivery system
US10357568B2 (en) 2011-03-24 2019-07-23 Glaxosmithkline Biologicals S.A. Adjuvant nanoemulsions with phospholipids
JP2014510533A (en) 2011-03-28 2014-05-01 ノバルティス アーゲー Markers associated with cyclin-dependent kinase inhibitors
WO2012135025A2 (en) 2011-03-28 2012-10-04 Massachusetts Institute Of Technology Conjugated lipomers and uses thereof
JP2014511687A (en) 2011-03-31 2014-05-19 モデルナ セラピューティクス インコーポレイテッド Engineered nucleic acid delivery and formulation
EP3460064B8 (en) 2011-04-03 2024-03-20 The General Hospital Corporation d/b/a Massachusetts General Hospital Efficient protein expression in vivo using modified rna (mod-rna)
WO2012138530A1 (en) 2011-04-04 2012-10-11 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services 2'-o-aminooxymethyl nucleoside derivatives for use in the synthesis and modification of nucleosides, nucleotides and oligonucleotides
WO2012142132A1 (en) 2011-04-11 2012-10-18 Life Technologies Corporation Polymer particles and methods of making and using same
WO2012142240A1 (en) 2011-04-13 2012-10-18 The Trustees Of The University Of Pennsylvania Coated mesoporous nanoparticles
WO2013158127A1 (en) 2012-04-16 2013-10-24 Molecular Transfer, Inc. Agents for improved delivery of nucleic acids to eukaryotic cells
US20140178894A1 (en) 2011-04-20 2014-06-26 Novartis Forschungsstiftung, Zweigniederlassung Culture medium suitable for the culture of undifferentiated cells
CA2834349A1 (en) 2011-04-26 2012-11-01 Molecular Express, Inc. Liposomal formulations
WO2012149536A1 (en) 2011-04-28 2012-11-01 The Henry M. Jackson Foundation For The Advancement Of Military Medicine, Inc. Neutralizing antibodies to nipah and hendra virus
CN103687590A (en) 2011-04-28 2014-03-26 Stc·Unm公司 Porous nanoparticle-supported lipid bilayers (protocells) for targeted delivery and methods of using same
US9265815B2 (en) 2011-04-29 2016-02-23 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers
US20140056912A1 (en) 2011-04-29 2014-02-27 Novartis Ag Methods of treating squamous cell carcinoma
UA116189C2 (en) 2011-05-02 2018-02-26 Мілленніум Фармасьютікалз, Інк. FORMULATION FOR ANTI-α4β7 ANTIBODY
ES2957478T3 (en) 2011-05-02 2024-01-19 Univ Wayne State A protein-induced pluripotent stem cell technology and uses thereof
US8945588B2 (en) 2011-05-06 2015-02-03 The University Of Chicago Methods and compositions involving protective staphylococcal antigens, such as EBH polypeptides
CN103547350A (en) 2011-05-10 2014-01-29 巴斯夫欧洲公司 Oil-in-water emulsions
WO2012153297A1 (en) 2011-05-11 2012-11-15 Ramot At Tel-Aviv University Ltd. Targeted polymeric conjugates and uses thereof
US20140072657A1 (en) 2011-05-12 2014-03-13 Helmut Vockner Novel pharmaceutical formulation
WO2012153338A2 (en) 2011-05-12 2012-11-15 Yissum Research Development Company Of The Hebrew University Of Jerusalem Ltd. Liposomes comprising polymer-conjugated lipids and related uses
SG194755A1 (en) 2011-05-13 2013-12-30 Novartis Ag Pre-fusion rsv f antigens
US8691750B2 (en) 2011-05-17 2014-04-08 Axolabs Gmbh Lipids and compositions for intracellular delivery of biologically active compounds
BR112013029490A2 (en) 2011-05-17 2019-09-24 Moderna Therapeutics Inc engineered nucleic acids and methods of use for non-human vertebrates
CN103052757B (en) 2011-05-20 2015-02-25 科勒公司 Toilet installation system and method
NZ730355A (en) 2011-05-24 2022-10-28 Tron Translationale Onkologie An Der Univ Der Johannes Gutenberg Univ Mainz Gemeinnuetzige Gmbh Individualized vaccines for cancer
KR20140024907A (en) 2011-05-25 2014-03-03 노파르티스 아게 Biomarkers for lung cancer
US20140308363A1 (en) 2011-05-31 2014-10-16 Bind Therapeutics, Inc. Drug loaded polymeric nanoparticles and methods of making and using same
WO2013052167A2 (en) 2011-06-02 2013-04-11 The Regents Of The University Of California Membrane encapsulated nanoparticles and method of use
JP2014516549A (en) 2011-06-02 2014-07-17 ノバルティス アーゲー Biomarkers for hedgehog inhibitor therapy
WO2012168259A1 (en) 2011-06-06 2012-12-13 Novartis Forschungsstiftung, Zweigniederlassung Protein tyrosine phosphatase, non-receptor type 11 (ptpn11) and triple-negative breast cancer
WO2012170889A1 (en) 2011-06-08 2012-12-13 Shire Human Genetic Therapies, Inc. Cleavable lipids
ME03491B (en) 2011-06-08 2020-01-20 Translate Bio Inc Lipid nanoparticle compositions and methods for mrna delivery
US8636696B2 (en) 2011-06-10 2014-01-28 Kimberly-Clark Worldwide, Inc. Transdermal device containing microneedles
WO2012170607A2 (en) 2011-06-10 2012-12-13 Novartis Ag Use of pcsk9 antagonists
WO2012168491A1 (en) 2011-06-10 2012-12-13 Novartis Ag Pharmaceutical formulations of pcsk9 antagonists
PE20140576A1 (en) 2011-06-10 2014-05-24 Novartis Ag BOVINE VACCINES AND METHODS
US8916696B2 (en) 2011-06-12 2014-12-23 City Of Hope Aptamer-mRNA conjugates for targeted protein or peptide expression and methods for their use
WO2012172495A1 (en) 2011-06-14 2012-12-20 Novartis Ag Compositions and methods for antibodies targeting tem8
WO2012172424A1 (en) 2011-06-15 2012-12-20 Crontech Pharma Ab Injection needle and device
MX2013014789A (en) 2011-06-16 2014-01-20 Novartis Ag Soluble proteins for use as therapeutics.
EP3199545B1 (en) 2011-06-20 2019-10-30 University of Pittsburgh - Of the Commonwealth System of Higher Education Computationally optimized broadly reactive antigens for h1n1 influenza
US9862926B2 (en) 2011-06-27 2018-01-09 Cellscript, Llc. Inhibition of innate immune response
CN103687644B (en) 2011-06-28 2016-11-09 艾诺奥医药品有限公司 Invasive skin electroporation device
PT2726099T (en) 2011-07-01 2018-11-13 Novartis Ag Method for treating metabolic disorders
US20150024488A1 (en) 2011-07-04 2015-01-22 Commonwealth Scientific And Industrial Research Organisation Nucleic acid complex
CA2840913C (en) 2011-07-06 2020-01-21 Novartis Ag Oil-in-water emulsions that contain nucleic acids
EP4115875A1 (en) 2011-07-06 2023-01-11 GlaxoSmithKline Biologicals S.A. Liposomes having useful n:p ratio for delivery of rna molecules
WO2013006837A1 (en) 2011-07-06 2013-01-10 Novartis Ag Cationic oil-in-water emulsions
US11896636B2 (en) 2011-07-06 2024-02-13 Glaxosmithkline Biologicals Sa Immunogenic combination compositions and uses thereof
EP2729168A2 (en) 2011-07-06 2014-05-14 Novartis AG Immunogenic compositions and uses thereof
EP2729501A2 (en) 2011-07-07 2014-05-14 Life Technologies Corporation Polymer particles, nucleic acid polymer particles and methods of making and using the same
WO2013009736A2 (en) 2011-07-10 2013-01-17 President And Fellows Of Harvard College Compositions and methods for self-assembly of polymers with complementary macroscopic and microscopic scale units
WO2013009717A1 (en) 2011-07-10 2013-01-17 Elisabet De Los Pinos Virion derived protein nanoparticles for delivering diagnostic or therapeutic agents for the treatment of skin-related diseases
US20130012566A1 (en) 2011-07-10 2013-01-10 Aura Biosciences, Inc. Virion Derived Protein Nanoparticles For Delivering Diagnostic Or Therapeutic Agents For The Treatment of Alopecia
WO2013012921A2 (en) 2011-07-20 2013-01-24 University Of Iowa Research Foundation Nucleic acid aptamers
GB2492999A (en) 2011-07-20 2013-01-23 Univ Central Lancashire Neutron detector
ES2670944T3 (en) 2011-07-21 2018-06-04 Croda International Plc Branched polyether polyamide block copolymers and methods of preparing and using them
EP2736921B1 (en) 2011-07-25 2018-06-27 GlaxoSmithKline Biologicals SA Compositions and methods for assessing functional immunogenicity of parvovirus vaccines
US9493549B2 (en) 2011-07-25 2016-11-15 The Rockefeller University Antibodies directed toward the HIV-1 GP120 CD4 binding site with increased potency and breadth
US20130028857A1 (en) 2011-07-29 2013-01-31 Selecta Biosciences, Inc. Synthetic nanocarriers comprising polymers comprising multiple immunomodulatory agents
US9556281B2 (en) 2011-08-15 2017-01-31 The University Of Chicago Compositions and methods related to antibodies to staphylococcal protein A
BR112014004585A2 (en) 2011-08-26 2017-06-13 Arrowhead Res Corp polyvinyl ester polymers for in vivo nucleic acid release
CN103974719A (en) 2011-08-31 2014-08-06 诺华股份有限公司 Pegylated liposomes for delivery of immunogen-encoding RNA
KR20140067070A (en) 2011-08-31 2014-06-03 말린크로트 엘엘씨 Nanoparticle peg modification with h-phosphonates
US9126966B2 (en) 2011-08-31 2015-09-08 Protiva Biotherapeutics, Inc. Cationic lipids and methods of use thereof
KR20140105433A (en) 2011-09-01 2014-09-01 아이알엠 엘엘씨 Compounds and compositions as pdgfr kinase inhibitors
JP2014525435A (en) 2011-09-02 2014-09-29 ノバルティス アーゲー Organic composition for treating HSF1-related diseases
EP2755986A4 (en) 2011-09-12 2015-05-20 Moderna Therapeutics Inc Engineered nucleic acids and methods of use thereof
US9464124B2 (en) 2011-09-12 2016-10-11 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
EP3384938A1 (en) 2011-09-12 2018-10-10 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
TR201909110T4 (en) 2011-09-14 2019-07-22 Glaxosmithkline Biologicals Sa Methods for making saccharide-protein glycoconjugates.
EA201490636A1 (en) 2011-09-16 2014-08-29 Дзе Трастиз Оф Дзе Юниверсити Оф Пенсильвания T-CELLS DESIGNED WITH THE HELP OF RNA FOR THE TREATMENT OF MALIGNOUS NON-FORMATIONS
JP2014531456A (en) 2011-09-22 2014-11-27 バインド セラピューティックス インコーポレイテッド Therapeutic nanoparticles and methods of treating cancer
US9375388B2 (en) 2011-09-23 2016-06-28 Indian Institute Of Technology, Bombay Nanoparticle based cosmetic composition
UY34346A (en) 2011-09-26 2013-04-30 Novartis Ag FUSION PROTEINS TO TREAT METABOLIC DISORDERS
TW201315742A (en) 2011-09-26 2013-04-16 Novartis Ag Dual fuction proteins for treating metabolic disorders
WO2013049328A1 (en) 2011-09-27 2013-04-04 Alnylam Pharmaceuticals, Inc. Di-aliphatic substituted pegylated lipids
WO2013045505A1 (en) 2011-09-28 2013-04-04 Novartis Ag Biomarkers for raas combination therapy
HUE057725T2 (en) 2011-10-03 2022-06-28 Modernatx Inc Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
WO2013055971A1 (en) 2011-10-11 2013-04-18 Arizona Board Of Regents For And On Behalf Of Arizona State University Polymers for delivering a substance into a cell
BR112014008694A2 (en) 2011-10-11 2017-06-20 Novartis Ag recombinant polycistronic nucleic acid molecules
WO2013055331A1 (en) 2011-10-12 2013-04-18 The Curators Of The University Of Missouri Pentablock polymers
WO2014066811A1 (en) 2012-10-25 2014-05-01 The Johns Hopkins University Bioreducible poly (b-amino ester)s for sirna delivery
MX2014004415A (en) 2011-10-14 2015-06-05 Stc Unm Porous nanoparticle-supported lipid bilayers (protocells) for targeted delivery including transdermal delivery of cargo and methods thereof.
ES2769786T3 (en) 2011-10-14 2020-06-29 Recordati Ag Antibodies and methods for diseases related to the Wnt pathway
EP2768571B1 (en) 2011-10-18 2023-02-22 Micell Technologies, Inc. Drug delivery medical device
KR102011048B1 (en) 2011-10-18 2019-08-14 다이서나 파마수이티컬, 인크. Amine cationic lipids and uses thereof
CA2852857A1 (en) 2011-10-20 2013-04-25 Novartis Ag Adjuvanted influenza b virus vaccines for pediatric priming
BR112014007485B1 (en) 2011-10-20 2022-05-31 Novartis Ag Methods for predicting therapeutic responsiveness of an individual to treatment with an activator of the alpha 7 nicotinic acetylcholine receptor, and uses of said activator
JP2015502337A (en) 2011-10-25 2015-01-22 ザ ユニバーシティ オブ ブリティッシュ コロンビア Limit size lipid nanoparticles and related methods
WO2013061290A1 (en) 2011-10-26 2013-05-02 Nanopass Technologies Ltd. Microneedle intradermal drug delivery device with auto-disable functionality
JP6151707B2 (en) 2011-10-27 2017-06-21 マサチューセッツ インスティテュート オブ テクノロジー Amino acid derivative functionalized at the n-terminus capable of forming drug-encapsulating microspheres
MX343262B (en) 2011-10-27 2016-10-28 Kimberly Clark Co Transdermal delivery of high viscosity bioactive agents.
AU2012328457A1 (en) 2011-10-28 2014-04-24 Presage Biosciences, Inc. Methods for drug delivery
CN104023748B (en) 2011-10-28 2018-03-02 诚信生物公司 Protein formulation containing amino acid
WO2013062140A1 (en) 2011-10-28 2013-05-02 Kyoto University Method for efficiently inducing differentiation of pluripotent stem cells into hepatic lineage cells
EP3091029B1 (en) 2011-10-31 2022-12-28 F. Hoffmann-La Roche AG Anti-il13 antibody formulations
ES2694154T3 (en) 2011-10-31 2018-12-18 Mallinckrodt Llc Compositions of combinatorial liposomes for the treatment of cancer
WO2013067537A1 (en) 2011-11-04 2013-05-10 Univertiy Of Notre Dame Du Lac Nanoparticle-based drug delivery
US9579338B2 (en) 2011-11-04 2017-02-28 Nitto Denko Corporation Method of producing lipid nanoparticles for drug delivery
WO2013067355A1 (en) 2011-11-04 2013-05-10 Novartis Ag Low density lipoprotein-related protein 6 (lrp6) - half life extender constructs
JP6143269B2 (en) 2011-11-04 2017-06-07 エージェンシー フォー サイエンス,テクノロジー アンド リサーチ Self-assembled composite ultra-small peptide polymer hydrogel
RS58562B1 (en) 2011-11-04 2019-05-31 Nitto Denko Corp Method for sterilely producing lipid-nucleic acid particles
US20130116408A1 (en) 2011-11-05 2013-05-09 Aura Biosciences, Inc. Virion Derived Protein Nanoparticles For Delivering Radioisotopes For The Diagnosis And Treatment Of Malignant And Systemic Disease And The Monitoring Of Therapy
US20130115247A1 (en) 2011-11-05 2013-05-09 Aura Biosciences, Inc. Virion Derived Protein Nanoparticles For Delivering Radioisotopes For The Diagnosis And Treatment Of Malignant And Systemic Disease And The Monitoring Of Therapy
EP2776013B8 (en) 2011-11-08 2023-08-30 The Board of Trustees of the University of Arkansas Methods and compositions for x-ray induced release from ph sensitive liposomes
US20140287510A1 (en) 2011-11-08 2014-09-25 Novartis Forschungsstiftung, Zweigniederlassung Friedrich Miescher Institute Rod cell-specific promoter
EP2776022A1 (en) 2011-11-08 2014-09-17 Novartis Forschungsstiftung, Zweigniederlassung Friedrich Miescher Institute For Biomedical Research New treatment for neurodegenerative diseases
US20140294732A1 (en) 2011-11-08 2014-10-02 Novartis Forschungsstiftung, Zweigniederlassung, Friedrich Miescher Institute Early diagnostic of neurodegenerative diseases
US9581590B2 (en) 2011-11-09 2017-02-28 Board Of Trustees Of Michigan State University Metallic nanoparticle synthesis with carbohydrate capping agent
WO2013071047A1 (en) 2011-11-11 2013-05-16 Children's Medical Center Corporation Compositions and methods for in vitro transcription of rna
CA3166278A1 (en) 2011-11-11 2013-05-16 Variation Biotechnologies Inc. Compositions and methods for treatment of cytomegalovirus
EP2780035A1 (en) 2011-11-14 2014-09-24 Novartis AG Immunogenic complexes of polyanionic carbomers and env polypeptides and methods of manufacture and use thereof
CA2855619A1 (en) 2011-11-15 2013-05-23 Novartis Ag Combination of a phosphoinositide 3-kinase inhibitor and a modulator of the janus kinase 2 - signal transducer and activator of transcription 5 pathway
PT3384903T (en) 2011-11-18 2020-06-25 Regeneron Pharma Micro-particle containing therapeutic protein coated with biodegradable polymer for medical use
WO2013077907A1 (en) 2011-11-21 2013-05-30 Novartis Ag Methods of treating psoriatic arthritis (psa) using il-17 antagonists and psa response or non- response alleles
WO2013078199A2 (en) 2011-11-23 2013-05-30 Children's Medical Center Corporation Methods for enhanced in vivo delivery of synthetic, modified rnas
WO2013082111A2 (en) 2011-11-29 2013-06-06 The University Of North Carolina At Chapel Hill Geometrically engineered particles and methods for modulating macrophage or immune responses
US9364549B2 (en) 2011-11-30 2016-06-14 Andreas Voigt Hydrophobic drug-delivery material, method for manufacturing thereof and methods for delivery of a drug-delivery composition
CA2857502C (en) 2011-11-30 2019-08-13 3M Innovative Properties Company Microneedle device including a peptide therapeutic agent and an amino acid and methods of making and using the same
JP2015500241A (en) 2011-12-02 2015-01-05 ペガサス ラボラトリーズ インコーポレイテッド Sustained release composition based on amphiphilic lipids
US20130142781A1 (en) 2011-12-02 2013-06-06 Invivo Therapeutics Corporation Peg based hydrogel for peripheral nerve injury applications and compositions and method of use of synthetic hydrogel sealants
WO2013082529A1 (en) 2011-12-02 2013-06-06 Yale University Enzymatic synthesis of poly(amine-co-esters) and methods of use thereof for gene delivery
US8497124B2 (en) 2011-12-05 2013-07-30 Factor Bioscience Inc. Methods and products for reprogramming cells to a less differentiated state
EP2788033B1 (en) 2011-12-05 2017-05-31 Factor Bioscience Inc. Methods and products for transfecting cells
EP2788439A4 (en) 2011-12-05 2015-09-02 Nano Prec Medical Inc Device having titania nanotube membrane for drug delivery
EP2788316B1 (en) 2011-12-07 2019-04-24 Alnylam Pharmaceuticals, Inc. Branched alkyl and cycloalkyl terminated biodegradable lipids for the delivery of active agents
WO2013086373A1 (en) 2011-12-07 2013-06-13 Alnylam Pharmaceuticals, Inc. Lipids for the delivery of active agents
GB201121070D0 (en) 2011-12-07 2012-01-18 Isis Innovation composition for delivery of biotherapeutics
HUE21212055T1 (en) 2011-12-07 2022-11-28 Alnylam Pharmaceuticals Inc Biodegradable lipids for the delivery of active agents
EP2787977A4 (en) 2011-12-09 2015-05-06 Univ California Liposomal drug encapsulation
US9725687B2 (en) 2011-12-09 2017-08-08 President And Fellows Of Harvard College Integrated human organ-on-chip microphysiological systems
US10087422B2 (en) 2011-12-09 2018-10-02 President And Fellows Of Harvard College Organ chips and uses thereof
JP6182457B2 (en) 2011-12-12 2017-08-16 協和発酵キリン株式会社 Lipid nanoparticles for drug delivery systems containing cationic lipids
KR20140116095A (en) 2011-12-12 2014-10-01 더 트러스티스 오브 더 유니버시티 오브 펜실바니아 Proteins comprising mrsa pbp2a and fragments thereof, nucleic acids encoding the same, and compositions and their use to prevent and treat mrsa infections
US20140045913A1 (en) 2011-12-12 2014-02-13 Kyowa Hakko Kirin Co., Ltd. Lipid nano particles comprising combination of cationic lipid
US20150000936A1 (en) 2011-12-13 2015-01-01 Schlumberger Technology Corporation Energization of an element with a thermally expandable material
KR102046042B1 (en) 2011-12-13 2019-11-18 엔진아이씨 몰레큘러 딜리버리 피티와이 리미티드 Bacterially derived, intact minicells for delivery of therapeutic agents to brain tumors
EP2604253A1 (en) 2011-12-13 2013-06-19 Otto Glatter Water-in-oil emulsions and methods for their preparation
EP2791159A4 (en) 2011-12-14 2015-10-14 Moderna Therapeutics Inc Modified nucleic acids, and acute care uses thereof
US20140378538A1 (en) 2011-12-14 2014-12-25 Moderma Therapeutics, Inc. Methods of responding to a biothreat
WO2013090897A1 (en) 2011-12-15 2013-06-20 The Trustees Of The University Of Pennsylvania Using adaptive immunity to detect drug resistance
US9636414B2 (en) 2011-12-15 2017-05-02 Biontech Ag Particles comprising single stranded RNA and double stranded RNA for immunomodulation
WO2013090601A2 (en) 2011-12-16 2013-06-20 Massachusetts Institute Of Technology Compact nanoparticles for biological applications
HRP20220717T1 (en) 2011-12-16 2022-07-22 Modernatx, Inc. Modified mrna compositions
US20130157963A1 (en) 2011-12-16 2013-06-20 Allergan, Inc. Ophthalmic compositions comprising polyvinyl capralactam-polyvinyl acetate-polyethylene glycol graft copolymers
KR20140103939A (en) 2011-12-16 2014-08-27 노파르티스 아게 Aerosolization apparatus for inhalation profile-independent drug delivery
EP2791172B1 (en) 2011-12-16 2017-07-19 Synthon Biopharmaceuticals B.V. Compounds and methods for treating inflammatory diseases
CN104245794A (en) 2011-12-16 2014-12-24 麻省理工学院 Alpha-aminoamidine polymers and uses thereof
US9546235B2 (en) 2011-12-19 2017-01-17 The University Of Sydney Peptide-hydrogel composite
US9241829B2 (en) 2011-12-20 2016-01-26 Abbott Medical Optics Inc. Implantable intraocular drug delivery apparatus, system and method
JP2015510495A (en) 2011-12-21 2015-04-09 モデルナ セラピューティクス インコーポレイテッドModerna Therapeutics,Inc. Methods for extending the viability or longevity of an organ or organ graft
US20130195851A1 (en) 2011-12-23 2013-08-01 Genentech, Inc. Articles of manufacture and methods for co-administration of antibodies
KR101963230B1 (en) 2011-12-26 2019-03-29 삼성전자주식회사 Protein complex comprising multi-specific monoclonal antibodies
US10814115B2 (en) 2011-12-27 2020-10-27 Massachusetts Institute Of Technology Microneedle devices and uses thereof
WO2013101690A1 (en) 2011-12-29 2013-07-04 modeRNA Therapeutics Modified mrnas encoding cell-penetrating polypeptides
JP2015505309A (en) 2011-12-29 2015-02-19 ノバルティス アーゲー Adjuvanted combination of meningococcal factor H binding protein
SI3421601T1 (en) 2011-12-30 2020-03-31 Cellscript, Llc Making and using in vitro-synthesized ssrna for introducing into mammalian cells to induce a biological or biochemical effect
CN104136023A (en) 2012-01-06 2014-11-05 密执安生命治疗有限责任公司 Methods of reducing risk of cardiovascular disease
US20150030576A1 (en) 2012-01-10 2015-01-29 Moderna Therapeutics, Inc. Methods and compositions for targeting agents into and across the blood-brain barrier
AU2013212066B2 (en) 2012-01-26 2018-11-08 Life Technologies Corporation Methods for increasing the infectivity of viruses
SG11201404361UA (en) 2012-01-26 2014-09-26 Life Technologies Corp Methods for increasing the infectivity of viruses
WO2013113326A1 (en) 2012-01-31 2013-08-08 Curevac Gmbh Pharmaceutical composition comprising a polymeric carrier cargo complex and at least one protein or peptide antigen
WO2013113325A1 (en) 2012-01-31 2013-08-08 Curevac Gmbh Negatively charged nucleic acid comprising complexes for immunostimulation
EP2623121A1 (en) 2012-01-31 2013-08-07 Bayer Innovation GmbH Pharmaceutical composition comprising a polymeric carrier cargo complex and an antigen
CN106883334B (en) 2012-02-09 2019-06-18 生命技术公司 Hydrophilic polymer particle and preparation method thereof
EP2817345A1 (en) 2012-02-22 2014-12-31 Cerulean Pharma Inc. Conjugates, particles, compositions, and related methods
US20130243867A1 (en) 2012-02-23 2013-09-19 University Of South Florida (A Florida Non-Profit Corporation) Micelle compositions and methods for their use
US20130224268A1 (en) 2012-02-27 2013-08-29 Newgen Biopharma Corp. Topical delivery of hormonal and non hormonal nano formulations, methods of making and using the same
US20150037334A1 (en) 2012-03-01 2015-02-05 Amgen Research (Munich) Gmbh Long life polypeptide binding molecules
CN104271149B (en) 2012-03-13 2016-03-16 夸祖鲁-纳塔尔大学 Transdermal delivery device
US10322089B2 (en) 2012-03-14 2019-06-18 The Board Of Trustees Of The Leland Stanford Junior University Nanoparticles, nanoparticle delivery methods, and systems of delivery
JP6138904B2 (en) 2012-03-16 2017-05-31 ザ・ジョンズ・ホプキンス・ユニバーシティー Nonlinear multiblock copolymer drug conjugates for delivery of active agents
US8962577B2 (en) 2012-03-16 2015-02-24 The Johns Hopkins University Controlled release formulations for the delivery of HIF-1 inhibitors
AU2013231638B2 (en) 2012-03-16 2017-10-12 Merck Patent Gmbh Targeting aminoacid lipids
US9610346B2 (en) 2012-03-23 2017-04-04 International Aids Vaccine Initiative Recombinant viral vectors
WO2013142349A1 (en) 2012-03-23 2013-09-26 University Of Chicago Compositions and methods related to staphylococcal sbi
WO2013143555A1 (en) 2012-03-26 2013-10-03 Biontech Ag Rna formulation for immunotherapy
US9717685B2 (en) 2012-03-26 2017-08-01 President And Fellows Of Harvard College Lipid-coated nucleic acid nanostructures of defined shape
EP2830594B1 (en) 2012-03-27 2018-05-09 Sirna Therapeutics, Inc. DIETHER BASED BIODEGRADABLE CATIONIC LIPIDS FOR siRNA DELIVERY
CA2859452C (en) 2012-03-27 2021-12-21 Curevac Gmbh Artificial nucleic acid molecules for improved protein or peptide expression
CA2866955A1 (en) 2012-03-27 2013-10-03 Curevac Gmbh Artificial nucleic acid molecules
CN108929880A (en) 2012-03-27 2018-12-04 库瑞瓦格股份公司 Artificial nucleic acid molecule comprising 5 ' TOPUTR
ES2858523T3 (en) 2012-03-29 2021-09-30 Translate Bio Inc Neutral lipid-derived nanoparticles
US10501512B2 (en) 2012-04-02 2019-12-10 Modernatx, Inc. Modified polynucleotides
AU2013243947A1 (en) 2012-04-02 2014-10-30 Moderna Therapeutics, Inc. Modified polynucleotides for the production of proteins
CA2868391A1 (en) 2012-04-02 2013-10-10 Stephane Bancel Polynucleotides comprising n1-methyl-pseudouridine and methods for preparing the same
US20140275229A1 (en) 2012-04-02 2014-09-18 Moderna Therapeutics, Inc. Modified polynucleotides encoding udp glucuronosyltransferase 1 family, polypeptide a1
US20150050354A1 (en) 2012-04-02 2015-02-19 Moderna Therapeutics, Inc. Modified polynucleotides for the treatment of otic diseases and conditions
EP2833918B1 (en) 2012-04-05 2019-05-08 Massachusetts Institute of Technology Immunostimulatory compositions and methods of use thereof
WO2013151650A1 (en) 2012-04-05 2013-10-10 University Of Florida Research Foundation, Inc. Neurophilic nanoparticles
WO2013152351A2 (en) 2012-04-06 2013-10-10 The Trustees Of Columbia University In The City Of New York Fusion polypeptides and methods of use thereof
DK2836200T3 (en) 2012-04-08 2020-09-21 Urogen Pharma Ltd THERMOREVERSIBLE HYDROGEL PREPARATIONS FOR USE IN THE TREATMENT OF UROTHELIUM DISORDERS
ES2560235T3 (en) 2012-04-11 2016-02-18 Intezyne Technologies Inc. Block copolymers for stable micelles
US9603800B2 (en) 2012-04-12 2017-03-28 Yale University Methods of treating inflammatory and autoimmune diseases and disorders using nanolipogels
WO2013154766A1 (en) 2012-04-13 2013-10-17 New York University Microrna control of ldl receptor pathway
WO2013155513A1 (en) 2012-04-13 2013-10-17 President And Fellows Of Harvard College Devices and methods for in vitro aerosol delivery
KR20150001710A (en) 2012-04-18 2015-01-06 애로우헤드 리서치 코오포레이션 Poly(acrylate) Polymers for In Vivo Nucleic Acid Delivery
EP2838877B1 (en) 2012-04-19 2018-09-12 Sirna Therapeutics, Inc. Novel diester and triester based low molecular weight, biodegradable cationic lipids for oligonucleotide delivery
WO2013163234A1 (en) 2012-04-23 2013-10-31 Massachusetts Institute Of Technology Stable layer-by-layer coated particles
EP2841579B1 (en) 2012-04-25 2018-10-10 Regulus Therapeutics Inc. Microrna compounds and methods for modulating mir-21 activity
KR102310775B1 (en) 2012-05-03 2021-10-07 칼라 파마슈티컬스, 인크. Pharmaceutical nanoparticles showing improved mucosal transport
EP2849728A1 (en) 2012-05-04 2015-03-25 The Johns Hopkins University Lipid-based drug carriers for rapid penetration through mucus linings
US20150087671A1 (en) 2012-05-16 2015-03-26 Micell Technologies, Inc. Low burst sustained release lipophilic and biologic agent compositions
WO2013173582A1 (en) 2012-05-17 2013-11-21 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Hepatitis c virus neutralizing antibody
WO2013173693A1 (en) 2012-05-18 2013-11-21 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Nanoparticles with enhanced entry into cancer cells
JP6228191B2 (en) 2012-05-23 2017-11-08 ジ・オハイオ・ステート・ユニバーシティ Lipid-coated albumin nanoparticle composition, method of making the same, and method of using the same
US20150306249A1 (en) 2012-05-25 2015-10-29 Curevac Gmbh Reversible immobilization and/or controlled release of nucleic acid containing nanoparticles by (biodegradable) polymer coatings
ES2811798T3 (en) 2012-06-06 2021-03-15 Loma Vista Medical Inc Inflatable medical devices
DK2858677T3 (en) 2012-06-08 2020-08-31 Ethris Gmbh PULMONARY DELIVERY OF MESSENGER RNA
DK3489220T3 (en) 2012-06-08 2021-09-06 Nitto Denko Corp LIPIDES FOR THERAPEUTIC MEDIUM DELIVERY FORMULATIONS
WO2013185069A1 (en) 2012-06-08 2013-12-12 Shire Human Genetic Therapies, Inc. Pulmonary delivery of mrna to non-lung target cells
US20140005379A1 (en) 2012-06-20 2014-01-02 Frank GU Nanoparticle delivery system and components thereof
WO2014014613A2 (en) 2012-06-20 2014-01-23 President And Fellows Of Harvard College Self-assembling peptides, peptide nanostructures and uses thereof
EP2866833B1 (en) 2012-06-27 2019-05-15 Merck Sharp & Dohme Corp. Crystalline anti-human il-23 antibodies
US9150841B2 (en) 2012-06-29 2015-10-06 Shire Human Genetic Therapies, Inc. Cells for producing recombinant iduronate-2-sulfatase
US9415109B2 (en) 2012-07-06 2016-08-16 Alnylam Pharmaceuticals, Inc. Stable non-aggregating nucleic acid lipid particle formulations
US9956291B2 (en) 2012-07-10 2018-05-01 Shaker A. Mousa Nanoformulation and methods of use of thyroid receptor beta1 agonists for liver targeting
EP2872120B1 (en) 2012-07-16 2017-05-03 Nanoderm Sciences, Inc. Therapeutic nanoparticles with a polymyxin b as targeting agent
EP2687251A1 (en) 2012-07-17 2014-01-22 Sanofi-Aventis Deutschland GmbH Drug delivery device
EP2687252A1 (en) 2012-07-17 2014-01-22 Sanofi-Aventis Deutschland GmbH Drug delivery device
CN103566377A (en) 2012-07-18 2014-02-12 上海博笛生物科技有限公司 Targeted immunotherapy for cancer
WO2014015334A1 (en) 2012-07-20 2014-01-23 Brown University System and methods for nanostructure protected delivery of treatment agent and selective release thereof
EP2877600B1 (en) 2012-07-24 2019-04-10 President and Fellows of Harvard College Self-assembly of nucleic acid nanostructures
WO2014015422A1 (en) 2012-07-27 2014-01-30 Ontario Institute For Cancer Research Cellulose-based nanoparticles for drug delivery
GB201213624D0 (en) 2012-07-27 2012-09-12 Univ Ulster The Method and system for production of conjugated nanoparticles
WO2014024193A1 (en) 2012-08-07 2014-02-13 Prodel Pharma Ltd. Compositions and methods for rapid transmucosal delivery of pharmaceutical ingredients
WO2014025795A1 (en) 2012-08-07 2014-02-13 Northeastern University Compositions for the delivery of rna and drugs into cells
CN104582747B (en) 2012-08-08 2016-12-21 南洋理工大学 For manufacturing the method for the hydrogel fines with living cells and for manufacturing the compositions of tissue engineering bracket
US20140155861A1 (en) 2012-08-08 2014-06-05 Presage Biosciences, Inc. Extrusion methods and devices for drug delivery
CA2881440C (en) 2012-08-10 2022-05-17 University Of North Texas Health Science Center Drug delivery vehicle comprising conjugates between targeting polyamino acids and fatty acids
WO2014027006A1 (en) 2012-08-13 2014-02-20 Edko Pazarlama Tanitim Ticaret Limited Sirketi Bioadhesive formulations for use in drug delivery
WO2014028487A1 (en) 2012-08-13 2014-02-20 Massachusetts Institute Of Technology Amine-containing lipidoids and uses thereof
US9827321B2 (en) 2012-08-14 2017-11-28 The Trustees Of The University Of Pennsylvania Stabilizing shear-thinning hydrogels
WO2014026284A1 (en) 2012-08-14 2014-02-20 Froese Aaron Internal structured self assembling liposomes
WO2014028429A2 (en) 2012-08-14 2014-02-20 Moderna Therapeutics, Inc. Enzymes and polymerases for the synthesis of rna
EP2885414B1 (en) 2012-08-15 2020-09-23 The University of Chicago Exosome-based therapeutics against neurodegenerative disorders
WO2014039185A1 (en) 2012-09-05 2014-03-13 Creighton University Polymeric nanoparticles in a thermosensitive gel for coital-independent vaginal prophylaxis of hiv
US8703197B2 (en) 2012-09-13 2014-04-22 International Business Machines Corporation Branched polyamines for delivery of biologically active materials
EP2895156B1 (en) 2012-09-17 2019-05-08 Pfizer Inc. Process for preparing therapeutic nanoparticles
WO2014047649A1 (en) 2012-09-24 2014-03-27 The Regents Of The University Of California Methods for arranging and packing nucleic acids for unusual resistance to nucleases and targeted delivery for gene therapy
WO2014052634A1 (en) 2012-09-27 2014-04-03 The University Of North Carolina At Chapel Hill Lipid coated nanoparticles containing agents having low aqueous and lipid solubilities and methods thereof
US20150307542A1 (en) 2012-10-03 2015-10-29 Moderna Therapeutics, Inc. Modified nucleic acid molecules and uses thereof
WO2014053879A1 (en) 2012-10-04 2014-04-10 Centre National De La Recherche Scientifique Cell penetrating peptides for intracellular delivery of molecules
WO2014053882A1 (en) 2012-10-04 2014-04-10 Centre National De La Recherche Scientifique Cell penetrating peptides for intracellular delivery of molecules
EP2903595A1 (en) 2012-10-04 2015-08-12 University Of The Witwatersrand, Johannesburg Liposomal drug delivery system
EP2716655A1 (en) 2012-10-04 2014-04-09 Institut Pasteur Neutralizing antibodies directed against Hepatitis C virus ectodomain glycoprotein E2
US20140100178A1 (en) 2012-10-04 2014-04-10 Aslam Ansari Composition and methods for site-specific drug delivery to treat malaria and other liver diseases
WO2014053880A1 (en) 2012-10-04 2014-04-10 Centre National De La Recherche Scientifique Cell penetrating peptides for intracellular delivery of molecules
WO2014053881A1 (en) 2012-10-04 2014-04-10 Centre National De La Recherche Scientifique Cell penetrating peptides for intracellular delivery of molecules
WO2014064534A2 (en) 2012-10-05 2014-05-01 Chrontech Pharma Ab Injection needle, device, immunogenic compositions and method of use
EP2716689A1 (en) 2012-10-05 2014-04-09 National University of Ireland, Galway Polymer comprising a plurality of branches having at least one disulfide group and/or at least one vinyl group
WO2014059022A1 (en) 2012-10-09 2014-04-17 The Brigham And Women's Hospital, Inc. Nanoparticles for targeted delivery of multiple therapeutic agents and methods of use
US20140106260A1 (en) 2012-10-11 2014-04-17 The Trustees Of The University Of Pennsylvania Core-shell nanoparticulate compositions and methods
EP2908818A4 (en) 2012-10-16 2016-07-13 Endocyte Inc Drug delivery conjugates containing unnatural amino acids and methods for using
ES2822135T3 (en) 2012-10-18 2021-04-29 Univ Rockefeller Broadly neutralizing anti-HIV antibodies
CN104918639B (en) 2012-10-22 2018-01-26 萨拜格Rfa公司 System for being delivered to therapeutic agent in living cells and nucleus
US10172956B2 (en) 2012-10-26 2019-01-08 Vanderbilt University Polymeric nanoparticles
US20140315795A1 (en) 2012-10-26 2014-10-23 Nlife Therapeutics, S.L. Compositions and Methods for Selective Delivery of Oligonucleotide Molecules to Cell Types
US20150272900A1 (en) 2012-10-26 2015-10-01 The Johns Hopkins University Layer-By-Layer Approach to Co-Deliver DNA and siRNA via AuNPs: A Potential Platform for Modifying Release Kinetics
US20150376581A1 (en) 2012-10-29 2015-12-31 Technische Universitaet Dortmund T7 rna polymerase variants and methods of using the same
JP6510416B2 (en) 2012-11-01 2019-05-08 ファクター バイオサイエンス インコーポレイテッド Methods and products for expressing proteins in cells
WO2014071072A2 (en) 2012-11-02 2014-05-08 Pungente Michael D Novel cationic carotenoid-based lipids for cellular nucleic acid uptake
EP2914723B1 (en) 2012-11-05 2018-06-13 Fondazione Centro San Raffaele Novel targets in multiple myeloma and other disorders
US8852648B2 (en) 2012-11-06 2014-10-07 Rochal Industries, Llp Delivery of biologically-active agents using volatile, hydrophobic solvents
US9975916B2 (en) 2012-11-06 2018-05-22 President And Fellows Of Harvard College Compositions and methods relating to complex nucleic acid nanostructures
EP2916873B1 (en) 2012-11-07 2017-07-26 Council of Scientific & Industrial Research Nanocomplex containing amphipathic peptide useful for efficient transfection of biomolecules
US9572893B2 (en) 2012-11-07 2017-02-21 Council Of Scientific And Industrial Research Nanocomplex containing cationic peptide for biomolecule delivery
TW201920677A (en) 2012-11-08 2019-06-01 美商武田疫苗股份有限公司 Compositions, methods and uses for dengue virus serotype-4 constructs
MX2015005839A (en) 2012-11-08 2015-12-17 Clearside Biomedical Inc Methods and devices for the treatment of ocular diseases in human subjects.
MX2015005831A (en) 2012-11-08 2015-09-24 Eleven Biotherapeutics Inc Il-6 antagonists and uses thereof.
EP2917233A1 (en) 2012-11-08 2015-09-16 Novozymes Biopharma DK A/S Albumin variants
AU2013343864B2 (en) 2012-11-09 2019-04-04 BioNTech SE Method for cellular RNA expression
EP2916853B1 (en) 2012-11-09 2020-05-06 Velin-Pharma A/S Compositions for pulmonary delivery
WO2014071963A1 (en) 2012-11-09 2014-05-15 Biontech Ag Method for cellular rna expression
US9200119B2 (en) 2012-11-09 2015-12-01 Momentive Performance Materials Inc. Silicon-containing zwitterionic linear copolymer composition
WO2014075047A2 (en) 2012-11-12 2014-05-15 Genvec, Inc. Malaria antigens and methods of use
WO2014074218A1 (en) 2012-11-12 2014-05-15 Redwood Bioscience, Inc. Compounds and methods for producing a conjugate
GB201220354D0 (en) 2012-11-12 2012-12-26 Medpharm Ltd Dermal compositions
US9943608B2 (en) 2012-11-13 2018-04-17 Baylor College Of Medicine Multi-arm biodegradable polymers for nucleic acid delivery
US9310374B2 (en) 2012-11-16 2016-04-12 Redwood Bioscience, Inc. Hydrazinyl-indole compounds and methods for producing a conjugate
WO2014078636A1 (en) 2012-11-16 2014-05-22 President And Fellows Of Harvard College Nucleic acid hydrogel self-assembly
EP2732825B1 (en) 2012-11-19 2015-07-01 Invivogen Conjugates of a TLR7 and/or TLR8 agonist and a TLR2 agonist
WO2014076709A1 (en) 2012-11-19 2014-05-22 Technion Research And Development Foundation Ltd. Liposomes for in-vivo delivery
US20140141037A1 (en) 2012-11-20 2014-05-22 Novartis Ag Rsv f prefusion trimers
US20150290328A1 (en) 2012-11-20 2015-10-15 Phasebio Pharmaceuticals, Inc. Formulations of active agents for sustained release
WO2014081303A1 (en) 2012-11-22 2014-05-30 Tagworks Pharmaceuticals B.V. Chemically cleavable group
WO2014081299A1 (en) 2012-11-22 2014-05-30 Tagworks Pharmaceuticals B.V. Activatable liposomes
WO2014081300A1 (en) 2012-11-22 2014-05-30 Tagworks Pharmaceuticals B.V. Channel protein activatable liposomes
ES2921623T3 (en) 2012-11-26 2022-08-30 Modernatx Inc terminally modified RNA
WO2014093574A1 (en) 2012-12-13 2014-06-19 Moderna Therapeutics, Inc. Modified polynucleotides for altering cell phenotype
WO2014108515A1 (en) 2013-01-10 2014-07-17 Novartis Ag Influenza virus immunogenic compositions and uses thereof
US20140200261A1 (en) 2013-01-17 2014-07-17 Moderna Therapeutics, Inc. Signal-sensor polynucleotides for the alteration of cellular phenotypes
US20160022840A1 (en) 2013-03-09 2016-01-28 Moderna Therapeutics, Inc. Heterologous untranslated regions for mrna
EP2968397A4 (en) 2013-03-12 2016-12-28 Moderna Therapeutics Inc Diagnosis and treatment of fibrosis
WO2014159813A1 (en) 2013-03-13 2014-10-02 Moderna Therapeutics, Inc. Long-lived polynucleotide molecules
EP2971010B1 (en) 2013-03-14 2020-06-10 ModernaTX, Inc. Formulation and delivery of modified nucleoside, nucleotide, and nucleic acid compositions
WO2014152027A1 (en) 2013-03-15 2014-09-25 Moderna Therapeutics, Inc. Manufacturing methods for production of rna transcripts
WO2014152030A1 (en) 2013-03-15 2014-09-25 Moderna Therapeutics, Inc. Removal of dna fragments in mrna production process
US20160032273A1 (en) 2013-03-15 2016-02-04 Moderna Therapeutics, Inc. Characterization of mrna molecules
WO2014152031A1 (en) 2013-03-15 2014-09-25 Moderna Therapeutics, Inc. Ribonucleic acid purification
US20160017313A1 (en) 2013-03-15 2016-01-21 Moderna Therapeutics, Inc. Analysis of mrna heterogeneity and stability
WO2014144767A1 (en) 2013-03-15 2014-09-18 Moderna Therapeutics, Inc. Ion exchange purification of mrna
US8980864B2 (en) 2013-03-15 2015-03-17 Moderna Therapeutics, Inc. Compositions and methods of altering cholesterol levels
PL3019619T3 (en) 2013-07-11 2022-01-10 Modernatx, Inc. Compositions comprising synthetic polynucleotides encoding crispr related proteins and synthetic sgrnas and methods of use
WO2015034928A1 (en) 2013-09-03 2015-03-12 Moderna Therapeutics, Inc. Chimeric polynucleotides
WO2015034925A1 (en) 2013-09-03 2015-03-12 Moderna Therapeutics, Inc. Circular polynucleotides
EP3043826A4 (en) 2013-09-13 2017-05-24 Moderna Therapeutics, Inc. Polynucleotide compositions containing amino acids
EP3052106A4 (en) 2013-09-30 2017-07-19 ModernaTX, Inc. Polynucleotides encoding immune modulating polypeptides
EP3052521A1 (en) 2013-10-03 2016-08-10 Moderna Therapeutics, Inc. Polynucleotides encoding low density lipoprotein receptor
EP3058082A4 (en) 2013-10-18 2017-04-26 ModernaTX, Inc. Compositions and methods for tolerizing cellular systems
EP3092250A4 (en) 2014-01-08 2017-05-24 Moderna Therapeutics, Inc. Polynucleotides for the in vivo production of antibodies

Patent Citations (33)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4270537A (en) 1979-11-19 1981-06-02 Romaine Richard A Automatic hypodermic syringe
US4596556A (en) 1985-03-25 1986-06-24 Bioject, Inc. Hypodermic injection apparatus
US4886499A (en) 1986-12-18 1989-12-12 Hoffmann-La Roche Inc. Portable injection appliance
US5015235A (en) 1987-02-20 1991-05-14 National Carpet Equipment, Inc. Syringe needle combination
US4790824A (en) 1987-06-19 1988-12-13 Bioject, Inc. Non-invasive hypodermic injection device
US4940460A (en) 1987-06-19 1990-07-10 Bioject, Inc. Patient-fillable and non-invasive hypodermic injection device assembly
US4941880A (en) 1987-06-19 1990-07-17 Bioject, Inc. Pre-filled ampule and non-invasive hypodermic injection device assembly
US5339163A (en) 1988-03-16 1994-08-16 Canon Kabushiki Kaisha Automatic exposure control device using plural image plane detection areas
US5141496A (en) 1988-11-03 1992-08-25 Tino Dalto Spring impelled syringe guide with skin penetration depth adjustment
US5312335A (en) 1989-11-09 1994-05-17 Bioject Inc. Needleless hypodermic injection device
US5064413A (en) 1989-11-09 1991-11-12 Bioject, Inc. Needleless hypodermic injection device
US5503627A (en) 1989-11-09 1996-04-02 Bioject, Inc. Ampule for needleless injection
US5190521A (en) 1990-08-22 1993-03-02 Tecnol Medical Products, Inc. Apparatus and method for raising a skin wheal and anesthetizing skin
US5527288A (en) 1990-12-13 1996-06-18 Elan Medical Technologies Limited Intradermal drug delivery device and method for intradermal delivery of drugs
US5480381A (en) 1991-08-23 1996-01-02 Weston Medical Limited Needle-less injector
US5417662A (en) 1991-09-13 1995-05-23 Pharmacia Ab Injection needle arrangement
US5328483A (en) 1992-02-27 1994-07-12 Jacoby Richard M Intradermal injection device with medication and needle guard
US5383851A (en) 1992-07-24 1995-01-24 Bioject Inc. Needleless hypodermic injection device
US5520639A (en) 1992-07-24 1996-05-28 Bioject, Inc. Needleless hypodermic injection methods and device
US5704911A (en) 1992-09-28 1998-01-06 Equidyne Systems, Inc. Needleless hypodermic jet injector
US5569189A (en) 1992-09-28 1996-10-29 Equidyne Systems, Inc. hypodermic jet injector
US5334144A (en) 1992-10-30 1994-08-02 Becton, Dickinson And Company Single use disposable needleless injector
US5649912A (en) 1994-03-07 1997-07-22 Bioject, Inc. Ampule filling device
US5466220A (en) 1994-03-08 1995-11-14 Bioject, Inc. Drug vial mixing and transfer device
US5599302A (en) 1995-01-09 1997-02-04 Medi-Ject Corporation Medical injection system and method, gas spring thereof and launching device using gas spring
WO1997013537A1 (en) 1995-10-10 1997-04-17 Visionary Medical Products Corporation Gas pressured needle-less injection device
US5893397A (en) 1996-01-12 1999-04-13 Bioject Inc. Medication vial/syringe liquid-transfer apparatus
WO1997037705A1 (en) 1996-04-11 1997-10-16 Weston Medical Limited Spring-powered dispensing device for medical purposes
US5993412A (en) 1997-05-19 1999-11-30 Bioject, Inc. Injection apparatus
WO1999034850A1 (en) 1998-01-08 1999-07-15 Fiderm S.R.L. Device for controlling the penetration depth of a needle, for application to an injection syringe
WO2007024708A2 (en) 2005-08-23 2007-03-01 The Trustees Of The University Of Pennsylvania Rna containing modified nucleosides and methods of use thereof
WO2009127230A1 (en) * 2008-04-16 2009-10-22 Curevac Gmbh MODIFIED (m)RNA FOR SUPPRESSING OR AVOIDING AN IMMUNOSTIMULATORY RESPONSE AND IMMUNOSUPPRESSIVE COMPOSITION
WO2010111290A1 (en) * 2009-03-23 2010-09-30 University Of Utah Research Foundation Methods and compositions related to modified guanine bases for controlling off-target effects in rna interference

Non-Patent Citations (24)

* Cited by examiner, † Cited by third party
Title
"Computational Molecular Biology", 1988, OXFORD UNIVERSITY PRESS
"Remington: The Science and Practice of Pharmacy", 2005, LIPPINCOTT WILLIAMS & WILKINS
"Remington's Pharmaceutical Sciences", 1985, MACK PUBLISHING COMPANY, pages: 1418
A. R. GENNARO: "Remington's The Science and Practice of Pharmacy", 2006, LIPPINCOTT, WILLIAMS & WILKINS
ALBRECHT, IMMUNOTHERAPY, vol. 2, no. 6, 2010, pages 795 - 798
CARILLO ET AL., SIAM J. APPLIED MATH., vol. 48, 1988, pages 1073
EDWARD B. ROCHE: "Bioreversible Carriers in Drug Design", 1987, AMERICAN PHARMACEUTICAL ASSOCIATION AND PERGAMON PRESS
FUKUHARA ET AL., BIOCHEMISTRY, vol. 1, no. 4, 1962, pages 563 - 568
GAIT, M.J.: "Oligonucleotide synthesis: a practical approach", 1984, IRL PRESS
GREENE ET AL.: "Protective Groups in Organic Synthesis", 1991, WILEY & SONS
GRIBSKOV, M. AND DEVEREUX, J.: "Sequence Analysis Primer", 1991, M. STOCKTON PRESS
GRIFFIN, A. M., AND GRIFFIN: "Computer Analysis of Sequence Data", 1994, HUMANA PRESS
HEINJE, G.: "Sequence Analysis in Molecular Biology", 1987, ACADEMIC PRESS
HERDEWIJN, P.: "Oligonucleotide synthesis: methods and applications, Methods in Molecular Biology", vol. 288, 2005, HUMANA PRESS
JOURNAL OF PHARMACEUTICAL SCIENCE, vol. 66, no. 2, 1977
KAIRO KATALIN ET AL.: "Molecular Therapy", vol. 16, 1 November 2008, NATURE PUBLISHING GROUP, pages: 1833 - 1840
LESK, A. M.: "Biocomputing: Informatics and Genome Projects", 1993, ACADEMIC PRESS
MUSUNURU K ET AL.: "From noncoding variant to phenotype via SORT] at the lpl3 cholesterol locus", NATURE, vol. 466, 2010, pages 714 - 721
OGATA ET AL., JOURNAL OF ORGANIC CHEMISTRY, vol. 74, 2009, pages 2585 - 2588
PURMAL ET AL., NUCLEIC ACIDS RESEARCH, vol. 22, no. 1, 1994, pages 72 - 78
ROZENSKI, J; CRAIN, P; MCCLOSKEY, J: "The RNA Modification Database: 1999 update", NUCL ACIDS RES, vol. 27, 1999, pages 196 - 197
T. HIGUCHI; V. STELLA: "Pro-drugs as Novel Delivery Systems", vol. 14, A.C.S. SYMPOSIUM SERIES
VERMA S. ET AL.: "Modified Oligonucleotides: Synthesis and Strategy for Users", ANNU. REV. BIOCHEM., vol. 67, 1998, pages 99 - 134, XP002412536 *
XU ET AL., TETRAHEDRON, vol. 48, no. 9, 1992, pages 1729 - 1740

Cited By (226)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11298426B2 (en) 2003-10-14 2022-04-12 BioNTech SE Recombinant vaccines and use thereof
US10106800B2 (en) 2005-09-28 2018-10-23 Biontech Ag Modification of RNA, producing an increased transcript stability and translation efficiency
US10576166B2 (en) 2009-12-01 2020-03-03 Translate Bio, Inc. Liver specific delivery of messenger RNA
US10143758B2 (en) 2009-12-01 2018-12-04 Translate Bio, Inc. Liver specific delivery of messenger RNA
US9404127B2 (en) 2010-06-30 2016-08-02 Protiva Biotherapeutics, Inc. Non-liposomal systems for nucleic acid delivery
US11718852B2 (en) 2010-06-30 2023-08-08 Arbutus Biopharma Corporation Non-liposomal systems for nucleic acid delivery
US9518272B2 (en) 2010-06-30 2016-12-13 Protiva Biotherapeutics, Inc. Non-liposomal systems for nucleic acid delivery
US11717529B2 (en) 2010-07-06 2023-08-08 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11690865B2 (en) 2010-07-06 2023-07-04 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11786467B2 (en) 2010-07-06 2023-10-17 Glaxosmithkline Biologicals Sa Lipid formulations with immunogens
US11773395B1 (en) 2010-07-06 2023-10-03 Glaxosmithkline Biologicals Sa Immunization of large mammals with low doses of RNA
US11766401B2 (en) 2010-07-06 2023-09-26 Glaxosmithkline Biologicals Sa Methods of administering lipid formulations with immunogens
US11839686B2 (en) 2010-07-06 2023-12-12 Glaxosmithkline Biologicals Sa Lipid formulations with viral immunogens
US20220125723A1 (en) 2010-07-06 2022-04-28 Glaxosmithkline Biologicals Sa Lipid formulations with viral immunogens
US11845925B2 (en) 2010-07-06 2023-12-19 Glaxosmithkline Biologicals Sa Immunisation of large mammals with low doses of RNA
US11851660B2 (en) 2010-07-06 2023-12-26 Glaxosmithkline Biologicals Sa Immunisation of large mammals with low doses of RNA
US11850305B2 (en) 2010-07-06 2023-12-26 Glaxosmithkline Biologicals Sa Method of making lipid formulations with RNA encoding immunogens
US11759475B2 (en) 2010-07-06 2023-09-19 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11857562B2 (en) 2010-07-06 2024-01-02 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11865080B2 (en) 2010-07-06 2024-01-09 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11883534B2 (en) 2010-07-06 2024-01-30 Glaxosmithkline Biologicals Sa Immunisation with lipid formulations with RNA encoding immunogens
US11891608B2 (en) 2010-07-06 2024-02-06 Glaxosmithkline Biologicals Sa Immunization of large mammals with low doses of RNA
US11690861B2 (en) 2010-07-06 2023-07-04 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11596645B2 (en) 2010-07-06 2023-03-07 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11690862B1 (en) 2010-07-06 2023-07-04 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11638693B2 (en) 2010-07-06 2023-05-02 Glaxosmithkline Biologicals Sa Vaccine for eliciting immune response comprising RNA encoding an immunogen and lipid formulations comprising mole percentage of lipids
US11739334B2 (en) 2010-07-06 2023-08-29 Glaxosmithkline Biologicals Sa Immunisation of large mammals with low doses of RNA
US11905514B2 (en) 2010-07-06 2024-02-20 Glaxosmithkline Biological Sa Immunisation of large mammals with low doses of RNA
US11913001B2 (en) 2010-07-06 2024-02-27 Glaxosmithkline Biologicals Sa Immunisation of large mammals with low doses of RNA
US11638694B2 (en) 2010-07-06 2023-05-02 Glaxosmithkline Biologicals Sa Vaccine for eliciting immune response comprising lipid formulations and RNA encoding multiple immunogens
US11655475B2 (en) 2010-07-06 2023-05-23 Glaxosmithkline Biologicals Sa Immunisation of large mammals with low doses of RNA
US11666534B2 (en) 2010-07-06 2023-06-06 Glaxosmithkline Biologicals Sa Methods of administering lipid formulations with viral immunogens
US11696923B2 (en) 2010-07-06 2023-07-11 Glaxosmithkline Biologicals, Sa Delivery of RNA to trigger multiple immune pathways
US11730754B2 (en) 2010-07-06 2023-08-22 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11857681B2 (en) 2010-07-06 2024-01-02 Glaxosmithkline Biologicals Sa Lipid formulations with RNA encoding immunogens
US11707482B2 (en) 2010-07-06 2023-07-25 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11690864B2 (en) 2010-07-06 2023-07-04 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11690863B2 (en) 2010-07-06 2023-07-04 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US9447164B2 (en) 2010-08-06 2016-09-20 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US9937233B2 (en) 2010-08-06 2018-04-10 Modernatx, Inc. Engineered nucleic acids and methods of use thereof
US9181319B2 (en) 2010-08-06 2015-11-10 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US8822663B2 (en) 2010-08-06 2014-09-02 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US11759422B2 (en) 2010-08-31 2023-09-19 Glaxosmithkline Biologicals Sa Pegylated liposomes for delivery of immunogen-encoding RNA
US10064959B2 (en) 2010-10-01 2018-09-04 Modernatx, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
EP3590949B1 (en) 2010-10-01 2022-05-18 ModernaTX, Inc. Ribonucleic acids containing n1-methyl-pseudouracils and uses thereof
US9334328B2 (en) 2010-10-01 2016-05-10 Moderna Therapeutics, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US9657295B2 (en) 2010-10-01 2017-05-23 Modernatx, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US11639370B2 (en) 2010-10-11 2023-05-02 Glaxosmithkline Biologicals Sa Antigen delivery platforms
US9956271B2 (en) 2010-11-30 2018-05-01 Translate Bio, Inc. mRNA for use in treatment of human genetic diseases
US9061021B2 (en) 2010-11-30 2015-06-23 Shire Human Genetic Therapies, Inc. mRNA for use in treatment of human genetic diseases
US11135274B2 (en) 2010-11-30 2021-10-05 Translate Bio, Inc. MRNA for use in treatment of human genetic diseases
US8853377B2 (en) 2010-11-30 2014-10-07 Shire Human Genetic Therapies, Inc. mRNA for use in treatment of human genetic diseases
US8710200B2 (en) 2011-03-31 2014-04-29 Moderna Therapeutics, Inc. Engineered nucleic acids encoding a modified erythropoietin and their expression
US9533047B2 (en) 2011-03-31 2017-01-03 Modernatx, Inc. Delivery and formulation of engineered nucleic acids
US9950068B2 (en) 2011-03-31 2018-04-24 Modernatx, Inc. Delivery and formulation of engineered nucleic acids
US11248264B2 (en) 2011-05-24 2022-02-15 Tron-Translationale Onkologie An Der Universitätsmedizin Der Johannes Gutenberg-Universität Mainz Ggmbh Individualized vaccines for cancer
US10738355B2 (en) 2011-05-24 2020-08-11 Tron-Translationale Onkologie An Der Universitätsmedizin Der Johannes Gutenberg-Universität Mainz Ggmbh Individualized vaccines for cancer
US11730825B2 (en) 2011-06-08 2023-08-22 Translate Bio, Inc. Lipid nanoparticle compositions and methods for mRNA delivery
US10413618B2 (en) 2011-06-08 2019-09-17 Translate Bio, Inc. Lipid nanoparticle compositions and methods for mRNA delivery
US10350303B1 (en) 2011-06-08 2019-07-16 Translate Bio, Inc. Lipid nanoparticle compositions and methods for mRNA delivery
US10888626B2 (en) 2011-06-08 2021-01-12 Translate Bio, Inc. Lipid nanoparticle compositions and methods for mRNA delivery
US11951181B2 (en) 2011-06-08 2024-04-09 Translate Bio, Inc. Lipid nanoparticle compositions and methods for mRNA delivery
US11291734B2 (en) 2011-06-08 2022-04-05 Translate Bio, Inc. Lipid nanoparticle compositions and methods for mRNA delivery
US11185595B2 (en) 2011-06-08 2021-11-30 Translate Bio, Inc. Lipid nanoparticle compositions and methods for mRNA delivery
US11338044B2 (en) 2011-06-08 2022-05-24 Translate Bio, Inc. Lipid nanoparticle compositions and methods for mRNA delivery
US11951180B2 (en) 2011-06-08 2024-04-09 Translate Bio, Inc. Lipid nanoparticle compositions and methods for MRNA delivery
US11951179B2 (en) 2011-06-08 2024-04-09 Translate Bio, Inc. Lipid nanoparticle compositions and methods for MRNA delivery
US10238754B2 (en) 2011-06-08 2019-03-26 Translate Bio, Inc. Lipid nanoparticle compositions and methods for MRNA delivery
US11052159B2 (en) 2011-06-08 2021-07-06 Translate Bio, Inc. Lipid nanoparticle compositions and methods for mRNA delivery
US11547764B2 (en) 2011-06-08 2023-01-10 Translate Bio, Inc. Lipid nanoparticle compositions and methods for MRNA delivery
US9308281B2 (en) 2011-06-08 2016-04-12 Shire Human Genetic Therapies, Inc. MRNA therapy for Fabry disease
US9597413B2 (en) 2011-06-08 2017-03-21 Shire Human Genetic Therapies, Inc. Pulmonary delivery of mRNA
US10507249B2 (en) 2011-06-08 2019-12-17 Translate Bio, Inc. Lipid nanoparticle compositions and methods for mRNA delivery
US11896636B2 (en) 2011-07-06 2024-02-13 Glaxosmithkline Biologicals Sa Immunogenic combination compositions and uses thereof
US10022425B2 (en) 2011-09-12 2018-07-17 Modernatx, Inc. Engineered nucleic acids and methods of use thereof
US9464124B2 (en) 2011-09-12 2016-10-11 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US10751386B2 (en) 2011-09-12 2020-08-25 Modernatx, Inc. Engineered nucleic acids and methods of use thereof
US9428535B2 (en) 2011-10-03 2016-08-30 Moderna Therapeutics, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
CN110511939A (en) * 2011-10-03 2019-11-29 现代泰克斯公司 Nucleosides, nucleotide and nucleic acid of modification and application thereof
EP3492109B1 (en) 2011-10-03 2020-03-04 ModernaTX, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
EP4015005A1 (en) 2011-10-03 2022-06-22 ModernaTX, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
EP3682905A1 (en) 2011-10-03 2020-07-22 ModernaTX, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
EP3492109A1 (en) 2011-10-03 2019-06-05 Moderna Therapeutics, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
EP3682905B1 (en) 2011-10-03 2021-12-01 ModernaTX, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US8754062B2 (en) 2011-12-16 2014-06-17 Moderna Therapeutics, Inc. DLIN-KC2-DMA lipid nanoparticle delivery of modified polynucleotides
US8664194B2 (en) 2011-12-16 2014-03-04 Moderna Therapeutics, Inc. Method for producing a protein of interest in a primate
US9186372B2 (en) 2011-12-16 2015-11-17 Moderna Therapeutics, Inc. Split dose administration
US9295689B2 (en) 2011-12-16 2016-03-29 Moderna Therapeutics, Inc. Formulation and delivery of PLGA microspheres
US8680069B2 (en) 2011-12-16 2014-03-25 Moderna Therapeutics, Inc. Modified polynucleotides for the production of G-CSF
US9271996B2 (en) 2011-12-16 2016-03-01 Moderna Therapeutics, Inc. Formulation and delivery of PLGA microspheres
EP2791160B1 (en) 2011-12-16 2022-03-02 ModernaTX, Inc. Modified mrna compositions
US10485884B2 (en) 2012-03-26 2019-11-26 Biontech Rna Pharmaceuticals Gmbh RNA formulation for immunotherapy
US11559587B2 (en) 2012-03-26 2023-01-24 Tron-Translationale Onkologie An Der Universitätsmedizin Der Johannes Gutenberg-Universität Mainz Ggmbh RNA formulation for immunotherapy
US9061059B2 (en) 2012-04-02 2015-06-23 Moderna Therapeutics, Inc. Modified polynucleotides for treating protein deficiency
US9149506B2 (en) 2012-04-02 2015-10-06 Moderna Therapeutics, Inc. Modified polynucleotides encoding septin-4
US8999380B2 (en) 2012-04-02 2015-04-07 Moderna Therapeutics, Inc. Modified polynucleotides for the production of biologics and proteins associated with human disease
US9050297B2 (en) 2012-04-02 2015-06-09 Moderna Therapeutics, Inc. Modified polynucleotides encoding aryl hydrocarbon receptor nuclear translocator
US9089604B2 (en) 2012-04-02 2015-07-28 Moderna Therapeutics, Inc. Modified polynucleotides for treating galactosylceramidase protein deficiency
US9095552B2 (en) 2012-04-02 2015-08-04 Moderna Therapeutics, Inc. Modified polynucleotides encoding copper metabolism (MURR1) domain containing 1
US10501512B2 (en) 2012-04-02 2019-12-10 Modernatx, Inc. Modified polynucleotides
US9107886B2 (en) 2012-04-02 2015-08-18 Moderna Therapeutics, Inc. Modified polynucleotides encoding basic helix-loop-helix family member E41
US9114113B2 (en) 2012-04-02 2015-08-25 Moderna Therapeutics, Inc. Modified polynucleotides encoding citeD4
US9814760B2 (en) 2012-04-02 2017-11-14 Modernatx, Inc. Modified polynucleotides for the production of biologics and proteins associated with human disease
US11564998B2 (en) 2012-04-02 2023-01-31 Modernatx, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
US9192651B2 (en) 2012-04-02 2015-11-24 Moderna Therapeutics, Inc. Modified polynucleotides for the production of secreted proteins
US9878056B2 (en) 2012-04-02 2018-01-30 Modernatx, Inc. Modified polynucleotides for the production of cosmetic proteins and peptides
US9827332B2 (en) 2012-04-02 2017-11-28 Modernatx, Inc. Modified polynucleotides for the production of proteins
US9828416B2 (en) 2012-04-02 2017-11-28 Modernatx, Inc. Modified polynucleotides for the production of secreted proteins
US9216205B2 (en) 2012-04-02 2015-12-22 Moderna Therapeutics, Inc. Modified polynucleotides encoding granulysin
US9782462B2 (en) 2012-04-02 2017-10-10 Modernatx, Inc. Modified polynucleotides for the production of proteins associated with human disease
US9221891B2 (en) 2012-04-02 2015-12-29 Moderna Therapeutics, Inc. In vivo production of proteins
US9675668B2 (en) 2012-04-02 2017-06-13 Moderna Therapeutics, Inc. Modified polynucleotides encoding hepatitis A virus cellular receptor 2
US9220755B2 (en) 2012-04-02 2015-12-29 Moderna Therapeutics, Inc. Modified polynucleotides for the production of proteins associated with blood and lymphatic disorders
US9220792B2 (en) 2012-04-02 2015-12-29 Moderna Therapeutics, Inc. Modified polynucleotides encoding aquaporin-5
US9233141B2 (en) 2012-04-02 2016-01-12 Moderna Therapeutics, Inc. Modified polynucleotides for the production of proteins associated with blood and lymphatic disorders
US9255129B2 (en) 2012-04-02 2016-02-09 Moderna Therapeutics, Inc. Modified polynucleotides encoding SIAH E3 ubiquitin protein ligase 1
US9587003B2 (en) 2012-04-02 2017-03-07 Modernatx, Inc. Modified polynucleotides for the production of oncology-related proteins and peptides
US9572897B2 (en) 2012-04-02 2017-02-21 Modernatx, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
US9254311B2 (en) 2012-04-02 2016-02-09 Moderna Therapeutics, Inc. Modified polynucleotides for the production of proteins
US9283287B2 (en) 2012-04-02 2016-03-15 Moderna Therapeutics, Inc. Modified polynucleotides for the production of nuclear proteins
US9303079B2 (en) 2012-04-02 2016-04-05 Moderna Therapeutics, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
US9301993B2 (en) 2012-04-02 2016-04-05 Moderna Therapeutics, Inc. Modified polynucleotides encoding apoptosis inducing factor 1
US11254936B2 (en) 2012-06-08 2022-02-22 Translate Bio, Inc. Nuclease resistant polynucleotides and uses thereof
US9512456B2 (en) 2012-08-14 2016-12-06 Modernatx, Inc. Enzymes and polymerases for the synthesis of RNA
EP4074834A1 (en) 2012-11-26 2022-10-19 ModernaTX, Inc. Terminally modified rna
US10925935B2 (en) 2012-11-26 2021-02-23 Modernatx, Inc. Terminally Modified RNA
US9597380B2 (en) 2012-11-26 2017-03-21 Modernatx, Inc. Terminally modified RNA
WO2014081507A1 (en) 2012-11-26 2014-05-30 Moderna Therapeutics, Inc. Terminally modified rna
US10155029B2 (en) 2012-11-26 2018-12-18 Modernatx, Inc. Terminally modified RNA
AU2017202228B2 (en) * 2012-11-26 2019-03-14 Modernatx, Inc. Terminally modified RNA
US10155031B2 (en) 2012-11-28 2018-12-18 Biontech Rna Pharmaceuticals Gmbh Individualized vaccines for cancer
US11504419B2 (en) 2012-11-28 2022-11-22 BioNTech SE Individualized vaccines for cancer
WO2014093924A1 (en) 2012-12-13 2014-06-19 Moderna Therapeutics, Inc. Modified nucleic acid molecules and uses thereof
WO2014113089A2 (en) 2013-01-17 2014-07-24 Moderna Therapeutics, Inc. Signal-sensor polynucleotides for the alteration of cellular phenotypes
US11708396B2 (en) 2013-01-17 2023-07-25 Modernatx, Inc. Signal-sensor polynucleotides for the alteration of cellular phenotypes
WO2014159813A1 (en) 2013-03-13 2014-10-02 Moderna Therapeutics, Inc. Long-lived polynucleotide molecules
US10876104B2 (en) 2013-03-14 2020-12-29 Translate Bio, Inc. Methods for purification of messenger RNA
US11820977B2 (en) 2013-03-14 2023-11-21 Translate Bio, Inc. Methods for purification of messenger RNA
US11692189B2 (en) 2013-03-14 2023-07-04 Translate Bio, Inc. Methods for purification of messenger RNA
US9181321B2 (en) 2013-03-14 2015-11-10 Shire Human Genetic Therapies, Inc. CFTR mRNA compositions and related methods and uses
US11510937B2 (en) 2013-03-14 2022-11-29 Translate Bio, Inc. CFTR MRNA compositions and related methods and uses
US10420791B2 (en) 2013-03-14 2019-09-24 Translate Bio, Inc. CFTR MRNA compositions and related methods and uses
US9957499B2 (en) 2013-03-14 2018-05-01 Translate Bio, Inc. Methods for purification of messenger RNA
US9713626B2 (en) 2013-03-14 2017-07-25 Rana Therapeutics, Inc. CFTR mRNA compositions and related methods and uses
US10258698B2 (en) 2013-03-14 2019-04-16 Modernatx, Inc. Formulation and delivery of modified nucleoside, nucleotide, and nucleic acid compositions
US8980864B2 (en) 2013-03-15 2015-03-17 Moderna Therapeutics, Inc. Compositions and methods of altering cholesterol levels
DE102013005361A1 (en) 2013-03-28 2014-10-02 Eberhard Karls Universität Tübingen Medizinische Fakultät polyribonucleotide
US11222711B2 (en) 2013-05-10 2022-01-11 BioNTech SE Predicting immunogenicity of T cell epitopes
EP3971287A1 (en) 2013-07-11 2022-03-23 ModernaTX, Inc. Compositions comprising synthetic polynucleotides encoding crispr related proteins and synthetic sgrnas and methods of use
WO2015034925A1 (en) 2013-09-03 2015-03-12 Moderna Therapeutics, Inc. Circular polynucleotides
EP3041938A1 (en) * 2013-09-03 2016-07-13 Moderna Therapeutics, Inc. Circular polynucleotides
EP3052106A4 (en) * 2013-09-30 2017-07-19 ModernaTX, Inc. Polynucleotides encoding immune modulating polypeptides
US10815291B2 (en) 2013-09-30 2020-10-27 Modernatx, Inc. Polynucleotides encoding immune modulating polypeptides
US10023626B2 (en) 2013-09-30 2018-07-17 Modernatx, Inc. Polynucleotides encoding immune modulating polypeptides
US10323076B2 (en) 2013-10-03 2019-06-18 Modernatx, Inc. Polynucleotides encoding low density lipoprotein receptor
US10208295B2 (en) 2013-10-22 2019-02-19 Translate Bio, Inc. MRNA therapy for phenylketonuria
US11224642B2 (en) 2013-10-22 2022-01-18 Translate Bio, Inc. MRNA therapy for argininosuccinate synthetase deficiency
US11377642B2 (en) 2013-10-22 2022-07-05 Translate Bio, Inc. mRNA therapy for phenylketonuria
US9522176B2 (en) 2013-10-22 2016-12-20 Shire Human Genetic Therapies, Inc. MRNA therapy for phenylketonuria
US10022435B2 (en) 2014-04-23 2018-07-17 Modernatx, Inc. Nucleic acid vaccines
EP3134131B1 (en) 2014-04-23 2021-12-22 ModernaTX, Inc. Nucleic acid vaccines
US10709779B2 (en) 2014-04-23 2020-07-14 Modernatx, Inc. Nucleic acid vaccines
US9872900B2 (en) 2014-04-23 2018-01-23 Modernatx, Inc. Nucleic acid vaccines
US9850269B2 (en) 2014-04-25 2017-12-26 Translate Bio, Inc. Methods for purification of messenger RNA
US10155785B2 (en) 2014-04-25 2018-12-18 Translate Bio, Inc. Methods for purification of messenger RNA
US11059841B2 (en) 2014-04-25 2021-07-13 Translate Bio, Inc. Methods for purification of messenger RNA
US11884692B2 (en) 2014-04-25 2024-01-30 Translate Bio, Inc. Methods for purification of messenger RNA
US11173120B2 (en) 2014-09-25 2021-11-16 Biontech Rna Pharmaceuticals Gmbh Stable formulations of lipids and liposomes
US10072057B2 (en) 2014-11-10 2018-09-11 Modernatx, Inc. Alternative nucleic acid molecules containing reduced uracil content and uses thereof
US9751925B2 (en) 2014-11-10 2017-09-05 Modernatx, Inc. Alternative nucleic acid molecules containing reduced uracil content and uses thereof
US11156617B2 (en) 2015-02-12 2021-10-26 BioNTech RNA Pharmaceuticals GbmH Predicting T cell epitopes useful for vaccination
US11590157B2 (en) 2015-10-05 2023-02-28 Modernatx, Inc. Methods for therapeutic administration of messenger ribonucleic acid drugs
US10849920B2 (en) 2015-10-05 2020-12-01 Modernatx, Inc. Methods for therapeutic administration of messenger ribonucleic acid drugs
US11492628B2 (en) 2015-10-07 2022-11-08 BioNTech SE 3′-UTR sequences for stabilization of RNA
EP3718565B1 (en) 2015-10-22 2022-04-27 ModernaTX, Inc. Respiratory virus vaccines
WO2017127750A1 (en) 2016-01-22 2017-07-27 Modernatx, Inc. Messenger ribonucleic acids for the production of intracellular binding polypeptides and methods of use thereof
WO2018009838A1 (en) 2016-07-07 2018-01-11 Rubius Therapeutics, Inc. Compositions and methods related to therapeutic cell systems expressing exogenous rna
US11981703B2 (en) 2016-08-17 2024-05-14 Sirius Therapeutics, Inc. Polynucleotide constructs
WO2018081459A1 (en) 2016-10-26 2018-05-03 Modernatx, Inc. Messenger ribonucleic acids for enhancing immune responses and methods of use thereof
US11958891B2 (en) 2017-01-26 2024-04-16 Surrozen Operating, Inc. Tissue-specific Wnt signal enhancing molecules and uses thereof
US10093706B2 (en) 2017-01-30 2018-10-09 Indiana University Research And Technology Corporation Dominant positive hnRNP-E1 polypeptide compositions and methods
WO2018144775A1 (en) 2017-02-01 2018-08-09 Modernatx, Inc. Immunomodulatory therapeutic mrna compositions encoding activating oncogene mutation peptides
US11253605B2 (en) 2017-02-27 2022-02-22 Translate Bio, Inc. Codon-optimized CFTR MRNA
US11865159B2 (en) 2017-02-28 2024-01-09 Sanofi Therapeutic RNA
US11173190B2 (en) 2017-05-16 2021-11-16 Translate Bio, Inc. Treatment of cystic fibrosis by delivery of codon-optimized mRNA encoding CFTR
WO2018213789A1 (en) 2017-05-18 2018-11-22 Modernatx, Inc. Modified messenger rna comprising functional rna elements
EP4253544A2 (en) 2017-05-18 2023-10-04 ModernaTX, Inc. Modified messenger rna comprising functional rna elements
US11786607B2 (en) 2017-06-15 2023-10-17 Modernatx, Inc. RNA formulations
US11597744B2 (en) 2017-06-30 2023-03-07 Sirius Therapeutics, Inc. Chiral phosphoramidite auxiliaries and methods of their use
US11744801B2 (en) 2017-08-31 2023-09-05 Modernatx, Inc. Methods of making lipid nanoparticles
US11993645B2 (en) 2018-01-11 2024-05-28 The Board Of Trustees Of The Leland Stanford Junior University Compositions comprising R-Spondin (RSPO) surrogate molecules
WO2019152557A1 (en) 2018-01-30 2019-08-08 Modernatx, Inc. Compositions and methods for delivery of agents to immune cells
WO2019200171A1 (en) 2018-04-11 2019-10-17 Modernatx, Inc. Messenger rna comprising functional rna elements
WO2019204743A1 (en) 2018-04-19 2019-10-24 Checkmate Pharmaceuticals, Inc. Synthetic rig-i-like receptor agonists
US11174500B2 (en) 2018-08-24 2021-11-16 Translate Bio, Inc. Methods for purification of messenger RNA
US11015211B2 (en) 2018-08-30 2021-05-25 Tenaya Therapeutics, Inc. Cardiac cell reprogramming with myocardin and ASCL1
US11913012B2 (en) 2018-08-30 2024-02-27 Tenaya Therapeutics, Inc. Cardiac cell reprogramming with myocardin and ASCL1
WO2020056304A1 (en) 2018-09-14 2020-03-19 Modernatx, Inc. Methods and compositions for treating cancer using mrna therapeutics
WO2020097409A2 (en) 2018-11-08 2020-05-14 Modernatx, Inc. Use of mrna encoding ox40l to treat cancer in human patients
WO2020227510A1 (en) 2019-05-07 2020-11-12 Modernatx, Inc. Polynucleotides for disrupting immune cell activity and methods of use thereof
WO2020227537A1 (en) 2019-05-07 2020-11-12 Modernatx, Inc Differentially expressed immune cell micrornas for regulation of protein expression
WO2020263985A1 (en) 2019-06-24 2020-12-30 Modernatx, Inc. Messenger rna comprising functional rna elements and uses thereof
WO2020263883A1 (en) 2019-06-24 2020-12-30 Modernatx, Inc. Endonuclease-resistant messenger rna and uses thereof
WO2021007515A1 (en) 2019-07-11 2021-01-14 Tenaya Therapeutics, Inc. Cardiac cell reprogramming with micrornas and other factors
WO2021050986A1 (en) 2019-09-11 2021-03-18 Modernatx, Inc. Lnp-formulated mrna therapeutics and use thereof for treating human subjects
WO2021081353A1 (en) 2019-10-23 2021-04-29 Checkmate Pharmaceuticals, Inc. Synthetic rig-i-like receptor agonists
WO2021178246A1 (en) 2020-03-02 2021-09-10 Tenaya Therapeutics, Inc. Gene vector control by cardiomyocyte-expressed micrornas
US11951185B2 (en) 2020-04-22 2024-04-09 BioNTech SE RNA constructs and uses thereof
US11925694B2 (en) 2020-04-22 2024-03-12 BioNTech SE Coronavirus vaccine
US11547673B1 (en) 2020-04-22 2023-01-10 BioNTech SE Coronavirus vaccine
US11779659B2 (en) 2020-04-22 2023-10-10 BioNTech SE RNA constructs and uses thereof
WO2021243207A1 (en) 2020-05-28 2021-12-02 Modernatx, Inc. Use of mrnas encoding ox40l, il-23 and il-36gamma for treating cancer
WO2022032154A2 (en) 2020-08-06 2022-02-10 Modernatx, Inc. Compositions for the delivery of payload molecules to airway epithelium
WO2023009422A1 (en) 2021-07-26 2023-02-02 Modernatx, Inc. Processes for preparing lipid nanoparticle compositions for the delivery of payload molecules to airway epithelium
WO2023009421A1 (en) 2021-07-26 2023-02-02 Modernatx, Inc. Processes for preparing lipid nanoparticle compositions
WO2023064469A1 (en) 2021-10-13 2023-04-20 Modernatx, Inc. Compositions of mrna-encoded il15 fusion proteins and methods of use thereof
WO2023069498A1 (en) 2021-10-22 2023-04-27 Senda Biosciences, Inc. Mrna vaccine composition
WO2023086465A1 (en) 2021-11-12 2023-05-19 Modernatx, Inc. Compositions for the delivery of payload molecules to airway epithelium
WO2023096858A1 (en) 2021-11-23 2023-06-01 Senda Biosciences, Inc. A bacteria-derived lipid composition and use thereof
WO2023122080A1 (en) 2021-12-20 2023-06-29 Senda Biosciences, Inc. Compositions comprising mrna and lipid reconstructed plant messenger packs
WO2023154818A1 (en) 2022-02-09 2023-08-17 Modernatx, Inc. Mucosal administration methods and formulations
WO2023196988A1 (en) 2022-04-07 2023-10-12 Modernatx, Inc. Methods of use of mrnas encoding il-12
WO2023199113A1 (en) 2022-04-15 2023-10-19 Smartcella Solutions Ab COMPOSITIONS AND METHODS FOR EXOSOME-MEDIATED DELIVERY OF mRNA AGENTS
US11878055B1 (en) 2022-06-26 2024-01-23 BioNTech SE Coronavirus vaccine
WO2024044741A3 (en) * 2022-08-26 2024-04-18 Trilink Biotechnologies, Llc Efficient method for making highly purified 5'-capped oligonucleotides
WO2024102434A1 (en) 2022-11-10 2024-05-16 Senda Biosciences, Inc. Rna compositions comprising lipid nanoparticles or lipid reconstructed natural messenger packs

Also Published As

Publication number Publication date
US9334328B2 (en) 2016-05-10
JP2013543381A (en) 2013-12-05
EP2622064A4 (en) 2014-04-16
EP3431485A1 (en) 2019-01-23
EP2857413A1 (en) 2015-04-08
EP3431485B1 (en) 2020-12-30
PT3590949T (en) 2022-08-02
US20120237975A1 (en) 2012-09-20
US20190160185A1 (en) 2019-05-30
CN104531812A (en) 2015-04-22
BR112013007862A2 (en) 2019-09-24
US20160264975A1 (en) 2016-09-15
EP2625189B1 (en) 2018-06-27
US9701965B2 (en) 2017-07-11
DK3590949T3 (en) 2022-07-11
AU2011308496A2 (en) 2014-10-02
HRP20220796T1 (en) 2022-10-14
PL3590949T3 (en) 2022-08-29
US20210236655A1 (en) 2021-08-05
US20130102034A1 (en) 2013-04-25
ZA201403666B (en) 2016-03-30
US20170239374A1 (en) 2017-08-24
EP3590949A1 (en) 2020-01-08
HUE058896T2 (en) 2022-09-28
SI3590949T1 (en) 2022-09-30
RU2013120302A (en) 2014-11-20
EP2857499A1 (en) 2015-04-08
EP3590949B1 (en) 2022-05-18
US10064959B2 (en) 2018-09-04
ES2862955T3 (en) 2021-10-08
EP2625189A2 (en) 2013-08-14
US20130203115A1 (en) 2013-08-08
NZ608972A (en) 2015-09-25
SG10201508149TA (en) 2015-10-29
US20240033379A1 (en) 2024-02-01
US20130244282A1 (en) 2013-09-19
ES2925251T3 (en) 2022-10-14
US20180112221A1 (en) 2018-04-26
AU2011308496A1 (en) 2013-05-02
EP2622064B1 (en) 2019-05-29
WO2012045082A2 (en) 2012-04-05
RS63430B1 (en) 2022-08-31
ZA201303161B (en) 2014-10-29
IL225493A0 (en) 2013-06-27
US20150064725A1 (en) 2015-03-05
MX2013003681A (en) 2013-11-20
EP2625189A4 (en) 2014-04-30
LT3590949T (en) 2022-07-25
AU2017202958A1 (en) 2017-05-25
CA2821992A1 (en) 2012-04-05
SG190679A1 (en) 2013-07-31
CN104531671A (en) 2015-04-22
EP2622064A1 (en) 2013-08-07
US9657295B2 (en) 2017-05-23
CA2813466A1 (en) 2012-04-05
CA3162352A1 (en) 2012-04-05
ES2737960T3 (en) 2020-01-17
EP4108671A1 (en) 2022-12-28
DE19177059T1 (en) 2021-10-07
WO2012045082A3 (en) 2012-06-07
CN103429606A (en) 2013-12-04

Similar Documents

Publication Publication Date Title
US20240033379A1 (en) Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
JP7019639B2 (en) Modified nucleosides, nucleotides and nucleic acids, and how to use them
US20230416324A1 (en) Alternative nucleic acid molecules containing reduced uracil content and uses thereof
US20140371302A1 (en) Modified mrnas encoding cell-penetrating polypeptides
US20160022774A1 (en) Diagnosis and treatment of fibrosis

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 11830061

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2813466

Country of ref document: CA

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2011830061

Country of ref document: EP