US20190085368A1 - Manufacturing methods for production of rna transcripts - Google Patents

Manufacturing methods for production of rna transcripts Download PDF

Info

Publication number
US20190085368A1
US20190085368A1 US16/144,282 US201816144282A US2019085368A1 US 20190085368 A1 US20190085368 A1 US 20190085368A1 US 201816144282 A US201816144282 A US 201816144282A US 2019085368 A1 US2019085368 A1 US 2019085368A1
Authority
US
United States
Prior art keywords
optionally substituted
rna transcript
modified
independently
mmrna
Prior art date
Legal status (The legal status 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 status listed.)
Abandoned
Application number
US16/144,282
Inventor
Stephane Bancel
William Joseph ISSA
John Grant Aunins
Tirtha Chakraborty
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
ModernaTx Inc
Original Assignee
ModernaTx Inc
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
Application filed by ModernaTx Inc filed Critical ModernaTx Inc
Priority to US16/144,282 priority Critical patent/US20190085368A1/en
Assigned to MODERNA THERAPEUTICS, INC. reassignment MODERNA THERAPEUTICS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BANCEL, STEPHANE, CHAKRABORTY, TIRTHA, ISSA, WILLIAM JOSEPH
Assigned to MODERNA THERAPEUTICS, INC. reassignment MODERNA THERAPEUTICS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: AUNINS, JOHN GRANT
Assigned to MODERNATX, INC. reassignment MODERNATX, INC. CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: MODERNA THERAPEUTICS, INC.
Publication of US20190085368A1 publication Critical patent/US20190085368A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • 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
    • C12P19/00Preparation of compounds containing saccharide radicals
    • C12P19/26Preparation of nitrogen-containing carbohydrates
    • C12P19/28N-glycosides
    • C12P19/30Nucleotides
    • C12P19/34Polynucleotides, e.g. nucleic acids, oligoribonucleotides
    • 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
    • C12N15/1003Extracting or separating nucleic acids from biological samples, e.g. pure separation or isolation methods; Conditions, buffers or apparatuses therefor
    • C12N15/1006Extracting or separating nucleic acids from biological samples, e.g. pure separation or isolation methods; Conditions, buffers or apparatuses therefor by means of a solid support carrier, e.g. particles, polymers
    • C12N15/101Extracting or separating nucleic acids from biological samples, e.g. pure separation or isolation methods; Conditions, buffers or apparatuses therefor by means of a solid support carrier, e.g. particles, polymers by chromatography, e.g. electrophoresis, ion-exchange, reverse phase
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6844Nucleic acid amplification reactions
    • C12Q1/6865Promoter-based amplification, e.g. nucleic acid sequence amplification [NASBA], self-sustained sequence replication [3SR] or transcription-based amplification system [TAS]

Definitions

  • the invention relates to methods useful for manufacture of RNA transcripts, e.g., mRNA.
  • RNA must be as homogeneous as possible; this includes obtaining a uniform cap structure/5′ terminus, correct sequence, correct poly A tail length and minimizing the formation of product related impurities.
  • PCR PCR to generate DNA templates containing a Poly A:T tract
  • the poly A:T tracts are introduced via PCR primers, introducing another step for creation of impurities.
  • RNA transcript e.g., mRNA
  • a method for production of an RNA transcript using a non-amplified, linearized DNA template in an in vitro transcription reaction to generate the RNA transcript.
  • the RNA transcript is capped via enzymatic capping.
  • the method results in production of homogeneous RNA transcripts with high purity and potency.
  • the RNA transcript is purified via chromatographic methods, e.g., use of an oligo dT substrate.
  • the method excludes the use of DNase.
  • FIG. 1 is a flow chart of one embodiment of the method.
  • FIG. 2 illustrates overhangs of linearized plasmid DNA template with different endonucleases, according to one embodiment.
  • FIG. 3 illustrates an mRNA cap structure, according to one embodiment.
  • FIG. 4 illustrates an examplary RNA trasncript diagram, according to one embodiment.
  • FIG. 5 is a map of plasmid pJ344:91543-TC-GCSF-pA-V1, according to one embodiment.
  • FIG. 6 is a DNA sequence of plasmid pJ344:91543-TC-GCSF-pA-V1, according to one embodiment.
  • FIGS. 7A and 7B are electropherograms of RNA transcripts, according to one embodiment.
  • FIG. 8 is an electropherogram of capped RNA, according to one embodiment.
  • FIG. 9 illustrates a bar graph showing results of an ELISA used to quantify expressed protein data, according to one embodiment.
  • FIG. 10 illustrates a bar graph showing results of an assay for determination of IFN- ⁇ induction following transfection of RNAs into PBMC, according to one embodiment.
  • FIG. 11A is a map of plasmid pJ204:109475-TC-FIX-Hs-3-pA140-Sap, according to one embodiment.
  • FIG. 11B is a DNA sequence of plasmid pJ204:109475-TC-FIX-Hs-3-pA140-Sap, according to one embodiment.
  • RNA transcripts useful for producing clinical grade mRNA of high purity and potency, consistently, reproducibly, and in compliance with current good manufacturing practices (cGMP).
  • the method uses a non-amplified, linearized DNA template in an in vitro transcription reaction to generate the RNA transcript.
  • the RNA transcript is purified via oligo dT substrate without the use of DNase. Enzymatic capping is used for 5′ capping of the RNA transcript.
  • Plasmid DNA template can be produced at microgram, milligram and gram scale in a cGMP compliant fashion. Large scale production of PCR generated templates is not commercially viable.
  • RNA transcripts of higher quality and purity mRNA transcripts than with PCR product templates: There is lower risk that no additional aberrant species are transcribed when using a linearized, non-amplified DNA template. Additionally, PCR amplification creates another intermediate in the manufacturing process where additional impurities may be introduced. When using PCR amplification, the gene of interest is amplified and typically does not contain a poly A tail. The poly A tail is introduced via the reverse primer, not encoded in the plasmid template. Utilizing linearized plasmid DNA template produces RNA transcripts of higher quality and purity
  • PCR additional unit operation
  • Whole plasmid allows for simple DNA/RNA separation: Use of whole plasmid DNA template is more amenable to SEC and IEX chromatographic separations compared to use of PCR product due to larger disparity in size and charge density between DNA and RNA.
  • Whole plasmid can also be removed using a poly dT based affinity purification in the flowthrough fraction.
  • RNA transcripts are typically performed using a chemical cap analog. This is performed co-transcriptionally where the cap analog: GTP molar ratio in the reaction is 4:1. This typically results in ⁇ 80% capping efficiency, as well as reduced RNA transcript yields due to consumption of GTP. This high abundance of uncapped species is undesirable when developing therapeutic RNA. Since only capped mRNA is translated into protein, the presence of a high abundance of uncapped species (ie 20%) is problematic as efficacy (protein production/mg RNA) is reduced by 20% and 20% of the final drug substance is an inert impurity, decreasing process productivity.
  • RNAs The presence of uncapped species is also potentially immunogenic: Presence of a 5′ triphosphate motif on uncapped RNAs can be potentially immunostimulatory (see Hornung et. al and Abbas et. al). Use of cap analogs requires additional phosphatase treatment to remove the 5′-triphosphate motif from the RNAs.
  • Polynucleotide is interchangeable with nucleic acid, and includes any compound and/or substance that comprise a polymer of nucleotides.
  • RNA transcripts produced by the method of the invention and DNA templates used in the methods of the invention are polynucleotides.
  • Exemplary polynucleotides include, but are not limited to, ribonucleic acids (RNAs), deoxyribonucleic acids (DNAs), threose nucleic acids (TNAs), glycol nucleic acids (GNAs), peptide nucleic acids (PNAs), locked nucleic acids (LNAs, including LNA having a ⁇ -D-ribo configuration, ⁇ -LNA having an ⁇ -L-ribo configuration (a diastereomer of LNA), 2′-amino-LNA having a 2′-amino functionalization, and 2′-amino- ⁇ -LNA having a 2′-amino functionalization) or hybrids thereof.
  • RNAs ribonucleic acids
  • DNAs deoxyribonucleic acids
  • TAAs threose nucleic acids
  • GNAs glycol nucleic acids
  • PNAs peptide nucleic acids
  • LNAs locked nucleic acids
  • RNA transcript refers to a ribonucleic acid produced by an in vitro transcription reaction using a DNA template and an RNA polymerase. As described in more detail below, an RNA transcript typically includes the coding sequence for a gene of interest and a poly A tail. RNA transcript includes an mRNA. The RNA transcript can include modifications, e.g., modified nucleotides. As used herein, the term RNA transcript includes and is interchangeable with mRNA, modified mRNA “mmRNA” or modified mRNA, and primary construct.
  • Gene of interest refers to a polynucleotide which encodes a polypeptide or protein of interest.
  • the gene of interest refers to a deoxyribonucleic acid, e.g., a gene of interest in a DNA template which can be transcribed to an RNA transcript, or a ribonucleic acid, e.g., a gene of interest in an RNA transcript which can be translated to produce the encoded polypeptide of interest in vitro, in vivo, in situ or ex vivo.
  • a polypeptide of interest includes but is not limited to, biologics, antibodies, vaccines, therapeutic proteins or peptides, etc.
  • DNA template refers to a polynucleotide template for RNA polymerase.
  • a DNA template includes the sequence for a gene of interest operably linked to a RNA polymerase promoter sequence.
  • operably linked refers to a functional connection between two or more molecules, constructs, transcripts, entities, moieties or the like.
  • a gene of interest operably linked to an RNA polymerase promoter allows transcription of the gene of interest.
  • Poly A tail refers to a chain of adenine nucleotides. The term can refer to poly A tail that is to be added to an RNA transcript, or can refer to the poly A tail that already exists at the 3′ end of an RNA transcript. As described in more detail below, a poly A tail is typically 5-300 nucleotides in length.
  • 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).
  • RNA transcript e.g., mRNA
  • a method for production of an RNA transcript using a non-amplified, linearized DNA template in an in vitro transcription reaction to generate the RNA transcript.
  • the RNA transcript is capped via enzymatic capping.
  • the method results in production of homogeneous RNA transcripts with high purity and potency.
  • the RNA transcript is purified via chromatographic methods, e.g., use of an oligo dT substrate.
  • the method excludes the use of DNase.
  • an RNA transcript is synthesized from a non-amplified, linear DNA template coding for the gene of interest via an enzymatic in vitro transcription reaction utilizing a T7 phage polymerase and nucleotide triphosphates of the desired chemistry.
  • the RNA transcript is enzymatically capped post transcriptionally at the 5′ end using Vaccinia guanylyltransferase, guanosine triphosphate and s-adenosyl-L-methionine to yield cap 0 structure, i.e., an inverted 7-methylguanosine cap is added via a 5′ to 5′ triphosphate bridge.
  • RNA transcript is chromatographically purified during the manufacturing process using affinity and/or anion exchange methods and diafiltered into the appropriate formulation buffer. This process is PCR free.
  • the method used a non amplified, linearized plasmid DNA as the template for in vitro transcription.
  • Cells e.g., bacterial cells, e.g., E. coli , e.g., DH-1 cells are transfected with the plasmid DNA template.
  • the transfected cells are cultured to replicate the plasmid DNA which is then isolated and purified.
  • the plasmid DNA template includes a gene of interest coding for, e.g., a polypeptide of interest.
  • a gene of interest coding for, e.g., a polypeptide of interest.
  • the gene of interest is a GCSF (granular colony stimulating factor) gene.
  • the gene of interest codes is a Factor IX gene.
  • the gene of interest is a EP (erythropoietin) gene.
  • the plasmid DNA template also includes a RNA polymerase promoter, e.g., a T7 promoter located 5′ to and operably linked to the gene of interest. Also included is a sequence coding for a poly A tail located 3′ to the gene of interest. Additional description of promoters and poly A tails is found below.
  • a RNA polymerase promoter e.g., a T7 promoter located 5′ to and operably linked to the gene of interest.
  • a sequence coding for a poly A tail located 3′ to the gene of interest. Additional description of promoters and poly A tails is found below.
  • the endonuclease produces a 5′ overhang on the linearized DNA template.
  • the endonuclease produces a blunt end on the linearized DNA template.
  • the endonuclease produces 3′ overhang on the linearized DNA template. Examples of endonucleases resulting in 5′ overhangs include XbaI, SapI, NotI, EcoRI, HindIII, and SPEI. FIG. 2 shows examples of such overhangs. Examples of endonucleases resulting in blunt ends include SPE1. Additional endonucleases are well known to one of skill in the art and can be used depending on the application.
  • XbaI is utilized as the restriction endonuclease with a recognition site of 5′ TCTAGA3′.
  • the five base overhang left on the DNA template sequence results in additional bases on the 3′ end of the RNA transcript post-transcription.
  • the restriction endonuclease SAP I is used. The resulting overhang on the linearized DNA template sequence does not generate additional bases on the 3′ end of the RNA transcript.
  • the plasmid into which the gene of interest, promoter, poly A tail sequence, and 5′ and 3′ UTR, and linearization restriction sites are inserted can be, e.g., one knows to one of skill in the art. Examples include but are not limited to pUC57, pJ204 (from DNA 2.0) and pJ344 (from DNA 2.0).
  • DNA templates to be used in the methods of the invention include, e.g., pJ344:91543 (including a GCSF gene and XbaI site) and pJ204:109475 (including a Factor IX gene and a SAPI site). Both plasmids are described in detail below.
  • the plasmid DNA template is filtered into an appropriate solvent, e.g., water, TE (Tris-EDTA), 10 mM Tris HCl pH 7.5, HEPES/phosphate and the like. Filtration occurs via, e.g., ultrafiltration, diafiltration, or, e.g., tangential flow ultrafiltration/diafiltration.
  • an appropriate solvent e.g., water, TE (Tris-EDTA), 10 mM Tris HCl pH 7.5, HEPES/phosphate and the like. Filtration occurs via, e.g., ultrafiltration, diafiltration, or, e.g., tangential flow ultrafiltration/diafiltration.
  • the linearized DNA template can be purified before use as a template for in vitro transcription.
  • the linearized DNA template can be purified chromatographically, or purified using a silica filter based DNA capture method.
  • the linearized DNA template is used in an in vitro transcription (IVT) system.
  • the system typically comprises a transcription buffer, nucleotide triphosphates (NTPs), an RNase inhibitor and an RNA polymerase.
  • NTPs may be manufactured in house, may be selected from a supplier, or may be synthesized as described herein.
  • the NTPs may be selected from, but are not limited to, those described herein including natural and unnatural (modified) NTPs. Additional description of modified nucleotides is found below.
  • RNA polymerases or variants may be used in the method of the present invention.
  • the polymerase may be selected from, but is not limited to, a phage RNA polymerase, e.g., a T7 RNA polymerase, a T3 RNA polymerase, an SP6 RNa polymerase, and/or mutant polymerases such as, but not limited to, polymerases able to incorporate modified nucleic acids.
  • RNA polymerases may be modified by inserting or deleting amino acids of the RNA polymerase sequence.
  • the RNA polymerase may be modified to exhibit an increased ability to incorporate a 2′-modified nucleotide triphosphate compared to an unmodified RNA polymerase (see International Publication WO2008078180 and U.S. Pat. No. 8,101,385; herein incorporated by reference in their entireties).
  • Variants may be obtained by evolving an RNA polymerase, optimizing the RNA polymerase amino acid and/or nucleic acid sequence and/or by using other methods known in the art.
  • T7 RNA polymerase variants may be evolved using the continuous directed evolution system set out by Esvelt et al.
  • T7 RNA polymerase may encode at least one mutation such as, but not limited to, lysine at position 93 substituted for threonine (K93T), I4M, A7T, E63V, V64D, A65E, D66Y, T76N, C125R, S128R, A136T, N165S, G175R, H176L, Y178H, F182L, L196F, G198V, D208Y, E222K, S228A, Q239R, T243N, G259D, M2671, G280C, H300R, D351A, A354S, E356D, L360P, A383V, Y385C, D388Y, S397R, M401T, N410S, K450R, P451T, G452V, E484A, H523
  • T7 RNA polymerase variants may encode at least mutation as described in U.S. Pub. Nos. 20100120024 and 20070117112; herein incorporated by reference in their entireties.
  • Variants of RNA polymerase may also include, but are not limited to, substitutional variants, conservative amino acid substitution, insertional variants, deletional variants and/or covalent derivatives.
  • the RNA transcript may be designed to be recognized by the wild type or variant RNA polymerases. In doing so, the RNA transcript may be modified to contain sites or regions of sequence changes from the wild type or parent primary construct.
  • Typical reaction conditions for in vitro transcription can be as follows.
  • parameters can be changed according to the application, e.g., the polymerase and nucleotides used.
  • the in vitro transcription reaction includes the following: an RNA polymerase, e.g., a T7 RNA polymerase at a final concentration of, e.g., 1000-12000 U/mL, e.g., 7000 U/mL; the DNA template at a final concentration of, e.g., 10-70 nM, e.g., 40 nM; nucleotides (NTPs) at a final concentration of e.g., 0.5-10 nM, e.g., 7.5 nM each; magnesisum at a final concentration of, e.g., 12-60 mM, e.g., magnesium acetate at 40 mM; a buffer such as, e.g., HEPES or Tris at a pH of, e.g., 7-8.5, e.g. 40 mM Tris HCl, pH 8
  • an RNase inhibitor is included in the in vitro transcription reaction to ensure no RNase induced degradation during the transcription reaction.
  • murine RNase inhibitor can be utilized at a final concentration of 1000 U/mL
  • a pyrophosphatase is included in the in vitro transcription reaction to cleave the inorganic pyrophosphate generated following each nucleotide incorporation into two units of inorganic phosphate. This ensures that magnesium, which is essential for transcription, remains in solution and does not precipitate as magnesium pyrophosphate.
  • an E. Coli inorganic pyrophosphatase can utilized at a final concentration of 1 U/mL.
  • the in vitro transcription reaction is allowed to proceed, for example, under constant mixing at 37° C. for 4 hours.
  • Typical yields can be, e.g., 5 mg of RNA per mL of transcription reaction.
  • a typical in vitro transcription reaction might have the following properties:
  • the in vitro transcription reaction includes nucleotides (NTPs).
  • the nucleotides can be unmodified NTPs, e.g., A, C, C, and U ribonucleotides, or modified nucleotides, or a combination. A more detailed description of modified nucleotides is found below. Examples of nucleotide combinations useful for in vitro transcription are found in the table below.
  • ATP Adenosine triphosphate
  • GTP Guanosine triphosphate
  • CTP Cytidine Triphosphate
  • UTP Uridine Triphosphate
  • 5mCTP 5-methylcytidine triphosphate
  • 2-thioUTP 2-thiouridine triphosphate
  • ⁇ TP pseudouridine triphosphate
  • N-1-me ⁇ TP 1-methylpseudouridine triphosphate.
  • the RNA transcript is enzymatically capped at the 5/end after in vitro transcription.
  • Capping can be performed either before or after further purification of the RNA transcript, e.g., oligo dT purification. If not performing purification prior to capping, an ultrafiltration/diafiltration step must be performed (e.g., buffer exchange).
  • Non-limiting examples of 5′ cap structures are those which, among other things, have enhanced binding of cap binding proteins, increased half life, reduced susceptibility to 5′ endonucleases and/or reduced 5′decapping, as compared to synthetic 5′cap structures known in the art (or to a wild-type, natural or physiological 5′cap structure).
  • recombinant Vaccinia Virus Capping Enzyme and recombinant 2′-O-methyltransferase enzyme can create a canonical 5′-5′-triphosphate linkage between the 5′-terminal nucleotide of an mRNA and a guanine cap nucleotide wherein the cap guanine contains an N7 methylation and the 5′-terminal nucleotide of the mRNA contains a 2′-O-methyl.
  • Cap1 structure is termed the Cap1 structure.
  • Cap structures include, but are not limited to, 7mG(5′)ppp(5′)N,pN2p (cap 0), 7mG(5′)ppp(5′)NlmpNp (cap 1), and 7mG(5′)-ppp(5′)NlmpN2mp (cap 2).
  • An example of a cap structure is illustrated in FIG. 3 .
  • the RNA transcript can be enzymatically capped at the 5′ end using Vaccinia guanylyltransferase, guanosine triphosphate and s-adenosyl-L-methionine to yield cap 0 structure.
  • An inverted 7-methylguanosine cap is added via a 5′ to 5′ triphosphate bridge.
  • use of a 2′O-methyltransferase with Vaccinia guanylyltransferase yields the cap 1 structure where in addition to the cap 0 structure, the 2′OH group is methylated on the penultimate nucleotide.
  • S-adenosyl-L-methionine (SAM) is a cofactor utilized as a methyl transfer reagent.
  • enzymatic 5′ capping is performed as follows. S-adenosylmethione chloride*2HCl is dissolved at 20 mM in 5 mM HCl 10/90 v/v % ethanol/water as a prepared stock. RNase inhibitor are utilized as a safeguard to ensure no RNase degradation is observed during the reaction. The final 1 ⁇ buffer conditions consist of the following: 50 mM Tris HCl pH 8, 5 mM KCl, 1 mM MgCl2, and 1 mM dithiothreitol. The reaction is run under constant mixing at 37° C. for 2 hours. Enzymatic capping is of considerably higher efficiency than performing co-transcription through the use of dinucleotide cap analogs.
  • the RNA transcript is co-transcriptionall capped.
  • the 5′ terminal caps may include endogenous caps or cap analogs.
  • a 5′ terminal cap may comprise a guanine analog.
  • Useful guanine analogs include, but are not limited to, inosine, N1-methyl-guanosine, 2′fluoro-guanosine, 7-deaza-guanosine, 8-oxo-guanosine, 2-amino-guanosine, LNA-guanosine, and 2-azido-guanosine.
  • RNA transcript is the polynucleotide product of the in vitro transcription reaction.
  • the RNA transcript includes a gene of interest and a poly-A tail.
  • the RNA transcript includes a 5′UTR and a 3′UTR.
  • the RNA transcript includes a 5′ cap, typically added post trasncriptionally.
  • FIG. 4 illustrates an exemplary RNA transcript.
  • the RNA transcript 100 here contains a first region of linked nucleotides 102 that is flanked by a first flanking region 104 and a second flaking region 106 .
  • the “first region” may be referred to as a “coding region” or “region encoding” or simply the “first region.” This first region may include, but is not limited to, the gene of interest.
  • the polypeptide of interest may comprise at its 5′ terminus one or more signal sequences which are encoded by a signal sequence region 103 .
  • the flanking region 104 may comprise a region of linked nucleotides comprising one or more complete or incomplete 5′ UTRs sequences.
  • the flanking region 104 may also comprise a 5′ terminal cap 108.
  • the second flanking region 106 may comprise a region of linked nucleotides comprising one or more complete or incomplete 3′ UTRs.
  • the flanking region 106 may also comprise a 3′ tailing sequence 110 , e.g., a poly A tail sequence.
  • first operational region 105 Bridging the 5′ terminus of the first region 102 and the first flanking region 104 is a first operational region 105 .
  • this operational region comprises a Start codon.
  • the operational region may alternatively comprise any translation initiation sequence or signal including a Start codon.
  • this operational region comprises a Stop codon.
  • the operational region may alternatively comprise any translation initiation sequence or signal including a Stop codon. According to the present invention, multiple serial stop codons may also be used.
  • the shortest length of the first region of the RNA transcript can be the length of a nucleic acid sequence that is sufficient to encode for a dipeptide, a tripeptide, a tetrapeptide, a pentapeptide, a hexapeptide, a heptapeptide, an octapeptide, a nonapeptide, or a decapeptide.
  • the length may be sufficient to encode a peptide of 2-30 amino acids, e.g. 5-30, 10-30, 2-25, 5-25, 10-25, or 10-20 amino acids.
  • the length may be sufficient to encode for a peptide of at least 11, 12, 13, 14, 15, 17, 20, 25 or 30 amino acids, or a peptide that is no longer than 40 amino acids, e.g. no longer than 35, 30, 25, 20, 17, 15, 14, 13, 12, 11 or 10 amino acids.
  • the length of the first region encoding the polypeptide of interest of the present invention is greater than about 30 nucleotides in length (e.g., at least or greater than about 35, 40, 45, 50, 55, 60, 70, 80, 90, 100, 120, 140, 160, 180, 200, 250, 300, 350, 400, 450, 500, 600, 700, 800, 900, 1,000, 1,100, 1,200, 1,300, 1,400, 1,500, 1,600, 1,700, 1,800, 1,900, 2,000, 2,500, and 3,000, 4,000, 5,000, 6,000, 7,000, 8,000, 9,000, 10,000, 20,000, 30,000, 40,000, 50,000, 60,000, 70,000, 80,000, 90,000 or up to and including 100,000 nucleotides).
  • the RNA transcript includes from about 30 to about 100,000 nucleotides (e.g., from 30 to 50, from 30 to 100, from 30 to 250, from 30 to 500, from 30 to 1,000, from 30 to 1,500, from 30 to 3,000, from 30 to 5,000, from 30 to 7,000, from 30 to 10,000, from 30 to 25,000, from 30 to 50,000, from 30 to 70,000, from 100 to 250, from 100 to 500, from 100 to 1,000, from 100 to 1,500, from 100 to 3,000, from 100 to 5,000, from 100 to 7,000, from 100 to 10,000, from 100 to 25,000, from 100 to 50,000, from 100 to 70,000, from 100 to 100,000, from 500 to 1,000, from 500 to 1,500, from 500 to 2,000, from 500 to 3,000, from 500 to 5,000, from 500 to 7,000, from 500 to 10,000, from 500 to 25,000, from 500 to 50,000, from 500 to 70,000, from 500 to 100,000, from 1,000 to 1,500, from 1,000, from 500 to 2,000, from 500 to 3,000, from 500 to 5,000, from 500 to
  • the first and second flanking regions may range independently from 15-1,000 nucleotides in length (e.g., greater than 30, 40, 45, 50, 55, 60, 70, 80, 90, 100, 120, 140, 160, 180, 200, 250, 300, 350, 400, 450, 500, 600, 700, 800, and 900 nucleotides or at least 30, 40, 45, 50, 55, 60, 70, 80, 90, 100, 120, 140, 160, 180, 200, 250, 300, 350, 400, 450, 500, 600, 700, 800, 900, and 1,000 nucleotides).
  • 15-1,000 nucleotides in length e.g., greater than 30, 40, 45, 50, 55, 60, 70, 80, 90, 100, 120, 140, 160, 180, 200, 250, 300, 350, 400, 450, 500, 600, 700, 800, 900, and 1,000 nucleotides.
  • the tailing sequence may range from 1 to 500 nucleotides in length (e.g., at least 60, 70, 80, 90, 120, 140, 160, 180, 200, 250, 300, 350, 400, 450, or 500 nucleotides).
  • the length may be determined in units of or as a function of polyA Binding Protein binding.
  • the polyA tail is long enough to bind at least 4 monomers of PolyA Binding Protein.
  • PolyA Binding Protein monomers bind to stretches of approximately 38 nucleotides. As such, it has been observed that polyA tails of about 80 nucleotides and 160 nucleotides are functional.
  • the capping region may comprise a single cap or a series of nucleotides forming the cap.
  • the capping region may be from 1 to 10, e.g. 2-9, 3-8, 4-7, 1-5, 5-10, or at least 2, or 10 or fewer nucleotides in length.
  • the cap is absent.
  • the first and second operational regions may range from 3 to 40, e.g., 5-30, 10-20, 15, or at least 4, or 30 or fewer nucleotides in length and may comprise, in addition to a Start and/or Stop codon, one or more signal and/or restriction sequences.
  • RNA transcripts can comprise one or more structural and/or chemical modifications or alterations which impart useful properties to the polynucleotide including, in some embodiments, the lack of a substantial induction of the innate immune response of a cell into which the polynucleotide is introduced.
  • a “structural” feature or modification is one in which two or more linked nucleotides are inserted, deleted, duplicated, inverted or randomized in an RNA transcript without significant chemical modification to the nucleotides themselves. Because chemical bonds will necessarily be broken and reformed to effect a structural modification, structural modifications are of a chemical nature and hence are chemical modifications. However, structural modifications will result in a different sequence of nucleotides.
  • the polynucleotide “ATCG” may be chemically modified to “AT-5meC-G”.
  • the same polynucleotide may be structurally modified from “ATCG” to “ATCCCG”.
  • the dinucleotide “CC” has been inserted, resulting in a structural modification to the polynucleotide.
  • the linearized plasmid DNA template is removed from the in vitro transcription, e.g., the DNA template is separated from the RNA transcript.
  • the DNA template is removed chromatographically using an poly A capture, e.g., oligo dT, based affinity purification step.
  • the RNA transcript binds affinity substrate while the DNA template flow through and is removed.
  • DNase I it is typical to utilize DNase I to enzymatically digest DNA template immediately following in vitro transcription. In the methods of the invention, DNase is not utilized. Whole plasmid removal is preferred to enzymatic digestion due to the fact that the risk of degraded DNA fragments hybridizing to the transcribed mRNA is mitigated.
  • the poly A capture based affinity purification is oligo dT purification.
  • a polythymidine ligand is immobilized to a derivatized chromatography resin.
  • the mechanism of purification involves hybridization of the poly A tail of the RNA transcript to the oligonucleotide ligand.
  • the DNA template will not bind.
  • RNA transcripts that do not contain Poly A stretches will not bind to the resin and will not form a duplex with the affinity ligand.
  • Poly Adenylated RNA can then be eluted from the resin utilizing a low ionic strength buffer or a competitive binding oligonucleotide solution.
  • a one pot purification method can yield highly purified poly A containing RNA with recoveries >80% actively removes endotoxin, DNA template, and enzymes utilized in the production of RNA using a simple capture and elute methodology with no subsequent fraction of captured poly A containing RNA. This purification increases mRNA product purity and in turn significantly increases target protein expression.
  • the method for production of an RNA transcript can include additional purification steps after the in vitro transcription, e.g., an ion exchange chromatography step.
  • RNA transcript produced by the methods of the invention can be analyzed and characterized using any number of methods. Analysis can be performed before or after capping. Alternatively, analysis can be performed before or after poly A capture based affinity purification. In another embodiment, analysis can be performed before or after additional purification steps, e.g., anion exchange chromatography and the like.
  • RNA template quality can be determined using Bioanalyzer chip based electrophoresis system.
  • RNA template purity is analysed using analytical reverse phase HPLC respectively.
  • Capping efficiency can be analyzed using, e.g., total nuclease digestion followed by MS/MS quantitation of the dinucleotide cap species vs. uncapped GTP species.
  • In vitro efficacy can be analyzed by, e.g., transfecting RNA transcript into a human cell line, e.g., HeLA, PBMC, BJ Fibroblasts, Hek 293). Protein expression of the polypeptide of interest can be quantified using methods such as ELISA or flow cytometry.
  • Immunogenicity can be analyzed by, e.g., transfecting RNA transcripts into cell lines that indicate innate immune stimulation, e.g., PBMCs.
  • Cytokine induction can be analyzed using, e.g., methods such as ELISA to quantify a cytokine, e.g., Interferon- ⁇ .
  • RNA transcript of the invention can produce RNA transcript that is at least 30% full length transcript, or at least 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, or at least 95% full length transcript. Purity can be determined as described herein, e.g., via reverse phase HPLC or Bioanalyzer chip based electrophoresis and measure by, e.g., peak area of full length RNA transcript relative to total peak.
  • the DNA template and resulting RNA transcript of the present invention include a gene of interest.
  • the gene of interest encodes a polypeptide of interest selected from, e.g., biologics, antibodies, vaccines, therapeutic proteins or peptides, cell penetrating peptides, secreted proteins, plasma membrane proteins, cytoplasmic or cytoskeletal proteins, intracellular membrane bound proteins, nuclear proteins, proteins associated with human disease, targeting moieties or those proteins encoded by the human genome for which no therapeutic indication has been identified but which nonetheless have utility in areas of research and discovery.
  • the sequence for a particular gene of interest is readily identified by one of skill in the art using public and private databases, e.g., GenBank.
  • the gene of interest may encode variant polypeptides which have a certain identity with a reference polypeptide sequence.
  • a “reference polypeptide sequence” refers to a starting polypeptide sequence. Reference sequences may be wild type sequences or any sequence to which reference is made in the design of another 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.
  • the polypeptide variant may have the same or a similar activity as the reference polypeptide.
  • the variant may have an altered activity (e.g., increased or decreased) relative to a reference polypeptide.
  • variants of a particular polynucleotide or polypeptide of the invention 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% but less than 100% 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.
  • Such tools for alignment include those of the BLAST suite (Stephen F. Altschul, Thomas L. Madden, Alejandro A. Schulffer, Jinghui Zhang, Zheng Zhang, Webb Miller, and David J. Lipman (1997), “Gapped BLAST and PSI-BLAST: a new generation of protein database search programs”, Nucleic Acids Res. 25:3389-3402.) Other tools are described herein, specifically in the definition of “Identity.”
  • BLAST algorithm Default parameters in the BLAST algorithm include, for example, an expect threshold of 10, Word size of 28, Match/Mismatch Scores 1, ⁇ 2, Gap costs Linear. Any filter can be applied as well as a selection for species specific repeats, e.g., Homo sapiens.
  • the gene of interest can encode one or more biologics.
  • a “biologic” is a polypeptide-based molecule produced by the methods provided herein and which may be used to treat, cure, mitigate, prevent, or diagnose a serious or life-threatening disease or medical condition.
  • Biologics include, but are not limited to, allergenic extracts (e.g. for allergy shots and tests), blood components, gene therapy products, human tissue or cellular products used in transplantation, vaccines, monoclonal antibodies, cytokines, growth factors, enzymes, thrombolytics, and immunomodulators, among others.
  • one or more biologics currently being marketed or in development may be encoded by the polynucleotides, primary constructs or mmRNA of the present invention. While not wishing to be bound by theory, it is believed that incorporation of the encoding polynucleotides of a known biologic into the primary constructs or mmRNA of the invention will result in improved therapeutic efficacy due at least in part to the specificity, purity and/or selectivity of the construct designs.
  • the gene of interest can encode one or more antibodies or fragments thereof.
  • antibody includes monoclonal antibodies (including full length antibodies which have an immunoglobulin Fc region), antibody compositions with polyepitopic specificity, multispecific antibodies (e.g., bispecific antibodies, diabodies, and single-chain molecules), as well as antibody fragments.
  • immunoglobulin Ig is used interchangeably with “antibody” herein.
  • the term “monoclonal antibody” refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible naturally occurring mutations and/or post-translation modifications (e.g., isomerizations, amidations) that may be present in minor amounts. Monoclonal antibodies are highly specific, being directed against a single antigenic site.
  • the monoclonal antibodies herein specifically include “chimeric” antibodies (immunoglobulins) in which a portion of the heavy and/or light chain is identical with or homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular antibody class or subclass, while the remainder of the chain(s) is(are) identical with or homologous to corresponding sequences in antibodies derived from another species or belonging to another antibody class or subclass, as well as fragments of such antibodies, so long as they exhibit the desired biological activity.
  • chimeric antibodies immunoglobulins in which a portion of the heavy and/or light chain is identical with or homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular antibody class or subclass, while the remainder of the chain(s) is(are) identical with or homologous to corresponding sequences in antibodies derived from another species or belonging to another antibody class or subclass, as well as fragments of such antibodies, so long as they exhibit the desired biological activity.
  • Chimeric antibodies of interest herein include, but are not limited to, “primatized” antibodies comprising variable domain antigen-binding sequences derived from a non-human primate (e.g., Old World Monkey, Ape etc.) and human constant region sequences.
  • a non-human primate e.g., Old World Monkey, Ape etc.
  • antibody fragment comprises a portion of an intact antibody, preferably the antigen binding and/or the variable region of the intact antibody.
  • antibody fragments include Fab, Fab′, F(ab′)2 and Fv fragments; diabodies; linear antibodies; nanobodies; single-chain antibody molecules and multispecific antibodies formed from antibody fragments.
  • any of the five classes of immunoglobulins, IgA, IgD, IgE, IgG and IgM, may be encoded by the mmRNA of the invention, including the heavy chains designated alpha, delta, epsilon, gamma and mu, respectively. Also included are polynucleotide sequences encoding the subclasses, gamma and mu. Hence any of the subclasses of antibodies may be encoded in part or in whole and include the following subclasses: IgG1, IgG2, IgG3, IgG4, IgA1 and IgA2.
  • the gene of interest can encode monoclonal antibodies and/or variants thereof. Variants of antibodies may also include, but are not limited to, substitutional variants, conservative amino acid substitution, insertional variants, deletional variants and/or covalent derivatives.
  • the primary construct and/or mmRNA disclosed herein may encode an immunoglobulin Fc region.
  • the primary constructs and/or mmRNA may encode a variant immunoglobulin Fc region.
  • the primary constructs and/or mmRNA may encode an antibody having a variant immunoglobulin Fc region as described in U.S. Pat. No. 8,217,147 herein incorporated by reference in its entirety.
  • the gene of interest can encode one or more vaccines.
  • a “vaccine” is a biological preparation that improves immunity to a particular disease or infectious agent.
  • one or more vaccines currently being marketed or in development may be encoded by the polynucleotides, primary constructs or mmRNA of the present invention. While not wishing to be bound by theory, it is believed that incorporation into the primary constructs or mmRNA of the invention will result in improved therapeutic efficacy due at least in part to the specificity, purity and selectivity of the construct designs.
  • Vaccines encoded in the polynucleotides, primary constructs or mmRNA of the invention may be utilized to treat conditions or diseases in many therapeutic areas such as, but not limited to, cardiovascular, CNS, dermatology, endocrinology, oncology, immunology, respiratory, and anti-infective.
  • the gene of interest can encode one or more validated or “in testing” therapeutic proteins or peptides.
  • “Therapeutic protein” refers to a protein that, when administered to a cell has a therapeutic, diagnostic, and/or prophylactic effect and/or elicits a desired biological and/or pharmacological effect.
  • Therapeutic proteins and peptides may be utilized to treat conditions or diseases in many therapeutic areas such as, but not limited to, blood, cardiovascular, CNS, poisoning (including antivenoms), dermatology, endocrinology, genetic, genitourinary, gastrointestinal, musculoskeletal, oncology, and immunology, respiratory, sensory and anti-infective.
  • the gene of interest can encode one or more cell-penetrating polypeptides.
  • “cell-penetrating polypeptide” or CPP refers to a polypeptide which may facilitate the cellular uptake of molecules.
  • a cell-penetrating polypeptide of the present invention may contain one or more detectable labels.
  • the polypeptides may be partially labeled or completely labeled throughout.
  • the gene of interest can encode the detectable label completely, partially or not at all.
  • the cell-penetrating peptide may also include a signal sequence.
  • a “signal sequence” refers to a sequence of amino acid residues bound at the amino terminus of a nascent protein during protein translation. The signal sequence may be used to signal the secretion of the cell-penetrating polypeptide.
  • the gene of interest can encode a fusion protein.
  • the fusion protein may be created by operably linking a charged protein to a therapeutic protein.
  • “operably linked” refers to the therapeutic protein and the charged protein being connected in such a way to permit the expression of the complex when introduced into the cell.
  • “charged protein” refers to a protein that carries a positive, negative or overall neutral electrical charge.
  • the therapeutic protein may be covalently linked to the charged protein in the formation of the fusion protein.
  • the ratio of surface charge to total or surface amino acids may be approximately 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8 or 0.9.
  • the cell-penetrating polypeptide may form a complex after being translated.
  • the complex may comprise a charged protein linked, e.g. covalently linked, to the cell-penetrating polypeptide.
  • the cell-penetrating polypeptide may comprise a first domain and a second domain.
  • the first domain may comprise a supercharged polypeptide.
  • the second domain may comprise a protein-binding partner.
  • protein-binding partner includes, but is not limited to, antibodies and functional fragments thereof, scaffold proteins, or peptides.
  • the cell-penetrating polypeptide may further comprise an intracellular binding partner for the protein-binding partner.
  • the cell-penetrating polypeptide may be capable of being secreted from a cell where the polynucleotide, primary construct or mmRNA may be introduced.
  • the cell-penetrating polypeptide may also be capable of penetrating the first cell.
  • the cell-penetrating polypeptide is capable of penetrating a second cell.
  • the second cell may be from the same area as the first cell, or it may be from a different area.
  • the area may include, but is not limited to, tissues and organs.
  • the second cell may also be proximal or distal to the first cell.
  • the cell-penetrating polypeptide which may comprise a protein-binding partner.
  • the protein binding partner may include, but is not limited to, an antibody, a supercharged antibody or a functional fragment.
  • the polynucleotides, primary constructs or mmRNA may be introduced into the cell where a cell-penetrating polypeptide comprising the protein-binding partner is introduced.
  • the gene of interest can encode a secreted protein.
  • the secreted proteins may be selected from, e.g., those in US Patent Publication, 20100255574, the contents of which are incorporated herein by reference in their entirety.
  • the gene of interest can encode a protein of the plasma membrane; a cytoplasmic or cytoskeletal proteins; a intracellular membrane bound proteins; or a nuclear protein; a targeting moiety (e.g., 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); antimicrobial peptides (AMP) or antiviral peptides (AVP).
  • AMP antimicrobial peptides
  • AVP antiviral peptides
  • UTRs Untranslated Regions
  • the DNA template and RNA transcript can include UTRs.
  • UTRs Untranslated regions (UTRs) of a gene are transcribed but not translated. The 5′UTR starts at the transcription start site and continues to the start codon but does not include the start codon; whereas, the 3′UTR starts immediately following the stop codon and continues until the transcriptional termination signal.
  • the regulatory features of a UTR can be incorporated into the polynucleotides, primary constructs and/or mmRNA of the present invention to enhance the stability of the molecule. The specific features can also be incorporated to ensure controlled down-regulation of the transcript in case they are misdirected to undesired organs sites.
  • Natural 5′UTRs bear features which play roles in translation initiation. They harbor signatures like Kozak sequences which are commonly known to be involved in the process by which the ribosome initiates translation of many genes. Kozak sequences have the consensus CCR(A/G)CCAUGG, where R is a purine (adenine or guanine) three bases upstream of the start codon (AUG), which is followed by another ‘G’. 5′UTR also have been known to form secondary structures which are involved in elongation factor binding.
  • mRNA such as albumin, serum amyloid A, Apolipoprotein AB/E, transferrin, alpha fetoprotein, erythropoietin, or Factor VIII
  • tissue-specific mRNA to improve expression in that tissue is possible for muscle (MyoD, Myosin, Myoglobin, Myogenin, Herculin), for endothelial cells (Tie-1, CD36), for myeloid cells (C/EBP, AML1, G-CSF, GM-CSF, CDIIb, MSR, Fr-1, i-NOS), for leukocytes (CD45, CD18), for adipose tissue (CD36, GLUT4, ACRP30, adiponectin) and for lung epithelial cells (SP-A/B/C/D).
  • non-UTR sequences may be incorporated into the 5′ (or 3′ UTR) UTRs.
  • introns or portions of introns sequences may be incorporated into the flanking regions of the polynucleotides, primary constructs or mmRNA of the invention. Incorporation of intronic sequences may increase protein production as well as mRNA levels.
  • 3′ UTRs are known to have stretches of Adenosines and Uridines embedded in them. These AU rich signatures are particularly prevalent in genes with high rates of turnover. Based on their sequence features and functional properties, the AU rich elements (AREs) can be separated into three classes (Chen et al, 1995): Class I AREs contain several dispersed copies of an AUUUA motif within U-rich regions. C-Myc and MyoD contain class I AREs. Class II AREs possess two or more overlapping UUAUUUA(U/A)(U/A) nonamers. Molecules containing this type of AREs include GM-CSF and TNF-a. Class III ARES are less well defined.
  • AREs 3′ UTR AU rich elements
  • Introduction, removal or modification of 3′ UTR AU rich elements can be used to modulate the stability of polynucleotides, primary constructs or mmRNA of the invention.
  • AREs can be identified and removed or mutated to increase the intracellular stability and thus increase translation and production of the resultant protein.
  • Transfection experiments can be conducted in relevant cell lines, using polynucleotides, primary constructs or mmRNA of the invention and protein production can be assayed at various time points post-transfection.
  • cells can be transfected with different ARE-engineering molecules and by using an ELISA kit to the relevant protein and assaying protein produced at 6 hour, 12 hour, 24 hour, 48 hour, and 7 days post-transfection.
  • microRNAs are 19-25 nucleotide long noncoding RNAs that bind to the 3′UTR of nucleic acid molecules and down-regulate gene expression either by reducing nucleic acid molecule stability or by inhibiting translation.
  • the polynucleotides, primary constructs or mmRNA of the invention may comprise one or more microRNA target sequences, microRNA sequences, or microRNA seeds. Such sequences may correspond to any known microRNA such as those taught in US Publication US2005/0261218 and US Publication US2005/0059005, the contents of which are incorporated herein by reference in their entirety.
  • a microRNA sequence comprises a “seed” region, i.e., a sequence in the region of positions 2-8 of the mature microRNA, which sequence has perfect Watson-Crick complementarity to the miRNA target sequence.
  • a microRNA seed may comprise positions 2-8 or 2-7 of the mature microRNA.
  • a microRNA seed may comprise 7 nucleotides (e.g., nucleotides 2-8 of the mature microRNA), wherein the seed-complementary site in the corresponding miRNA target is flanked by an adenine (A) opposed to microRNA position 1.
  • a microRNA seed may comprise 6 nucleotides (e.g., nucleotides 2-7 of the mature microRNA), wherein the seed-complementary site in the corresponding miRNA target is flanked byan adenine (A) opposed to microRNA position 1.
  • A an adenine
  • the bases of the microRNA seed have complete complementarity with the target sequence.
  • microRNA target sequences By engineering microRNA target sequences into the 3′UTR of RNA transcripts of the invention one can target the molecule for degradation or reduced translation, provided the microRNA in question is available. This process will reduce the hazard of off target effects upon nucleic acid molecule delivery. Identification of microRNA, microRNA target regions, and their expression patterns and role in biology have been reported (Bonauer et al., Curr Drug Targets 2010 11:943-949; Anand and Cheresh Curr Opin Hematol 2011 18:171-176; Contreras and Rao Leukemia 2012 26:404-413 (2011 Dec. 20. doi: 10.1038/1eu.2011.356); Bartel Cell 2009 136:215-233; Landgraf et al, Cell, 2007 129:1401-1414; each of which is herein incorporated by reference in its entirety).
  • miR-122 a microRNA abundant in liver, can inhibit the expression of the gene of interest if one or multiple target sites of miR-122 are engineered into the 3′ UTR of the RNA transcripts.
  • Introduction of one or multiple binding sites for different microRNA can be engineered to further decrease the longevity, stability, and protein translation of a RNA transcripts.
  • microRNA site refers to a microRNA target site or a microRNA recognition site, or any nucleotide sequence to which a microRNA binds or associates. It should be understood that “binding” may follow traditional Watson-Crick hybridization rules or may reflect any stable association of the microRNA with the target sequence at or adjacent to the microRNA site.
  • microRNA binding sites can be engineered out of (i.e. removed from) sequences in which they naturally occur in order to increase protein expression in specific tissues.
  • miR-122 binding sites may be removed to improve protein expression in the liver. Regulation of expression in multiple tissues can be accomplished through introduction or removal or one or several microRNA binding sites.
  • tissues where microRNA are known to regulate mRNA, and thereby protein expression include, but are not limited to, liver (miR-122), muscle (miR-133, miR-206, miR-208), endothelial cells (miR-17-92, miR-126), myeloid cells (miR-142-3p, miR-142-5p, miR-16, miR-21, miR-223, miR-24, miR-27), adipose tissue (let-7, miR-30c), heart (miR-1d, miR-149), kidney (miR-192, miR-194, miR-204), and lung epithelial cells (let-7, miR-133, miR-126).
  • liver miR-122
  • muscle miR-133, miR-206, miR-208
  • endothelial cells miR-17-92, miR-126
  • myeloid cells miR-142-3p, miR-142-5p, miR-16, miR-21, miR-223, miR
  • MicroRNA can also regulate complex biological processes such as angiogenesis (miR-132) (Anand and Cheresh Curr Opin Hematol 2011 18:171-176; herein incorporated by reference in its entirety).
  • binding sites for microRNAs that are involved in such processes may be removed or introduced, in order to tailor the expression of the RNA transcripts expression to biologically relevant cell types or to the context of relevant biological processes.
  • a listing of MicroRNA, miR sequences and miR binding sites is listed in Table 9 of U.S. Provisional Application No. 61/753,661 filed Jan. 17, 2013, in Table 9 of U.S. Provisional Application No. 61/754,159 filed Jan. 18, 2013, and in Table 7 of U.S. Provisional Application No. 61/758,921 filed Jan. 31, 2013, each of which are herein incorporated by reference in their entireties.g
  • RNA transcripts can be engineered for more targeted expression in specific cell types or only under specific biological conditions. Through introduction of tissue-specific microRNA binding sites, RNA transcripts could be designed that would be optimal for protein expression in a tissue or in the context of a biological condition.
  • Transfection experiments can be conducted in relevant cell lines, using engineered RNA transcripts and protein production can be assayed at various time points post-transfection.
  • cells can be transfected with different microRNA binding site-engineering RNA transcripts and by using an ELISA kit to the relevant protein and assaying protein produced at 6 hour, 12 hour, 24 hour, 48 hour, 72 hour and 7 days post-transfection.
  • In vivo experiments can also be conducted using microRNA-binding site-engineered molecules to examine changes in tissue-specific expression of formulated RNA transcripts.
  • Additional viral sequences such as, but not limited to, the translation enhancer sequence of the barley yellow dwarf virus (BYDV-PAV), the Jaagsiekte sheep retrovirus (JSRV) and/or the Enzootic nasal tumor virus (See e.g., International Pub. No. WO2012129648; herein incorporated by reference in its entirety) can be engineered and inserted in the 3′ UTR of the RNA transcripts of the invention and can stimulate the translation of the construct in vitro and in vivo. Transfection experiments can be conducted in relevant cell lines at and protein production can be assayed by ELISA at 12 hr, 24 hr, 48 hr, 72 hr and day 7 post-transfection.
  • BYDV-PAV barley yellow dwarf virus
  • JSRV Jaagsiekte sheep retrovirus
  • Enzootic nasal tumor virus See e.g., International Pub. No. WO2012129648; herein incorporated by reference in its entirety
  • Transfection experiments can be conducted in relevant cell lines at and protein
  • RNA transcripts which may contain an internal ribosome entry site (IRES).
  • IRES internal ribosome entry site
  • An IRES may act as the sole ribosome binding site, or may serve as one of multiple ribosome binding sites of an mRNA.
  • RNA transcripts containing more than one functional ribosome binding site may encode several peptides or polypeptides that are translated independently by the ribosomes (“multicistronic nucleic acid molecules”).
  • multicistronic nucleic acid molecules When RNA transcripts are provided with an IRES, further optionally provided is a second translatable region.
  • IRES sequences that can be used according to the invention include without limitation, those from picornaviruses (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).
  • picornaviruses e.g. FMDV
  • 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
  • a long chain of adenine nucleotides may be added to a polynucleotide such as an mRNA molecules in order to increase stability.
  • a polynucleotide such as an mRNA molecules
  • the 3′ end of the transcript may be cleaved to free a 3′ hydroxyl.
  • poly-A polymerase adds a chain of adenine nucleotides to the RNA.
  • the process called polyadenylation, adds a poly-A tail that can be between, for example, approximately 100 and 250 residues long.
  • poly A tail is 5-300 nucleotides in length. In another embodiment, the poly A tail is 60-160 nucleotides in length.
  • the poly-A tail is greater than 35 nucleotides in length (e.g., at least or greater than about 35, 40, 45, 50, 55, 60, 70, 80, 90, 100, 120, 140, 160, 180, 200, 250, 300, 350, 400, 450, 500, 600, 700, 800, 900, 1,000, 1,100, 1,200, 1,300, 1,400, 1,500, 1,600, 1,700, 1,800, 1,900, 2,000, 2,500, and 3,000 nucleotides).
  • the polynucleotide, primary construct, or mmRNA includes from about 30 to about 3,000 nucleotides (e.g., from 30 to 50, from 30 to 100, from 30 to 250, from 30 to 500, from 30 to 750, from 30 to 1,000, from 30 to 1,500, from 30 to 2,000, from 30 to 2,500, from 50 to 100, from 50 to 250, from 50 to 500, from 50 to 750, from 50 to 1,000, from 50 to 1,500, from 50 to 2,000, from 50 to 2,500, from 50 to 3,000, from 100 to 500, from 100 to 750, from 100 to 1,000, from 100 to 1,500, from 100 to 2,000, from 100 to 2,500, from 100 to 3,000, from 500 to 750, from 500 to 1,000, from 500 to 1,500, from 500 to 2,000, from 500 to 2,500, from 500 to 3,000, from 1,000 to 1,500, from 1,000 to 2,000, from 1,000 to 2,500, from 1,000 to 3,000, from 1,500 to 2,000, from 1,500 to 2,500, from 1,500 to 2,500,
  • the poly-A tail is designed relative to the length of the overall RNA transcripts. This design may be based on the length of the coding region, the length of a particular feature or region (such as the first or flanking regions), or based on the length of the ultimate product expressed from the RNA transcripts.
  • the poly-A tail may be 10, 20, 30, 40, 50, 60, 70, 80, 90, or 100% greater in length than the RNA transcripts or feature thereof.
  • the poly-A tail may also be designed as a fraction of RNA transcripts to which it belongs.
  • the poly-A tail may be 10, 20, 30, 40, 50, 60, 70, 80, or 90% or more of the total length of the construct or the total length of the construct minus the poly-A tail.
  • engineered binding sites and conjugation of RNA transcripts for Poly-A binding protein may enhance expression.
  • RNA transcripts may be linked together to the PABP (Poly-A binding protein) through the 3′-end using modified nucleotides at the 3′-terminus of the poly-A tail.
  • Transfection experiments can be conducted in relevant cell lines at and protein production can be assayed by ELISA at 12 hr, 24 hr, 48 hr, 72 hr and day 7 post-transfection.
  • the polynucleotide primary constructs of the present invention are designed to include a polyA-G quartet.
  • the G-quartet is a cyclic hydrogen bonded array of four guanine nucleotides that can be formed by G-rich sequences in both DNA and RNA.
  • the G-quartet is incorporated at the end of the poly-A tail.
  • the resultant mmRNA construct is assayed for stability, protein production and other parameters including half-life at various time points. It has been discovered that the polyA-G quartet results in protein production equivalent to at least 75% of that seen using a poly-A tail of 120 nucleotides alone.
  • modify refers to modification with respect to A, G, U or C ribonucleotides. Generally, herein, these terms are not intended to refer to the ribonucleotide modifications in naturally occurring 5′-terminal mRNA cap moieties.
  • modification refers to a modification as compared to the canonical set of 20 amino acids, moiety
  • the modifications may be various distinct modifications.
  • the coding region, the flanking regions and/or the terminal regions may contain one, two, or more (optionally different) nucleoside or nucleotide modifications.
  • a modified polynucleotide, primary construct, or mmRNA introduced to a cell may exhibit reduced degradation in the cell, as compared to an unmodified polynucleotide, primary construct, or mmRNA.
  • the polynucleotides, primary constructs, and mmRNA can include any useful modification, such as to the sugar, the nucleobase, or the internucleoside linkage (e.g. to a linking phosphate/to a phosphodiester linkage/to the phosphodiester backbone).
  • One or more atoms of a pyrimidine nucleobase may be replaced or substituted with optionally substituted amino, optionally substituted thiol, optionally substituted alkyl (e.g., methyl or ethyl), or halo (e.g., chloro or fluoro).
  • modifications e.g., one or more modifications
  • RNAs ribonucleic acids
  • DNAs deoxyribonucleic acids
  • TAAs threose nucleic acids
  • GNAs glycol nucleic acids
  • PNAs peptide nucleic acids
  • LNAs locked nucleic acids
  • the polynucleotides, primary constructs, and mmRNA of the invention do not substantially induce an innate immune response of a cell into which the mRNA is introduced.
  • an induced innate immune response include 1) increased expression of pro-inflammatory cytokines, 2) activation of intracellular PRRs (RIG-I, MDAS, etc, and/or 3) termination or reduction in protein translation.
  • the invention provides a modified nucleic acid molecule containing a degradation domain, which is capable of being acted on in a directed manner within a cell.
  • the present disclosure provides polynucleotides comprising a nucleoside or nucleotide that can disrupt the binding of a major groove interacting, e.g. binding, partner with the polynucleotide (e.g., where the modified nucleotide has decreased binding affinity to major groove interacting partner, as compared to an unmodified nucleotide).
  • the polynucleotides, primary constructs, and mmRNA can optionally include other agents (e.g., RNAi-inducing agents, RNAi agents, siRNAs, shRNAs, miRNAs, antisense RNAs, ribozymes, catalytic DNA, tRNA, RNAs that induce triple helix formation, aptamers, vectors, etc.).
  • the polynucleotides, primary constructs, or mmRNA may include one or more messenger RNAs (mRNAs) and one or more modified nucleoside or nucleotides (e.g., mmRNA molecules). Details for these polynucleotides, primary constructs, and mmRNA follow.
  • the polynucleotides, primary constructs, and mmRNA of the invention includes a first region of linked nucleosides encoding a polypeptide of interest, a first flanking region located at the 5′ terminus of the first region, and a second flanking region located at the 3′ terminus of the first region.
  • the polynucleotide, primary construct, or mmRNA (e.g., the first region, first flanking region, or second flanking region) includes n number of linked nucleosides having Formula (Ia) or Formula (Ia-1):
  • U is O, S, N(RU)nu, or C(RU)nu, wherein nu is an integer from 0 to 2 and each RU is, independently, H, halo, or optionally substituted alkyl;
  • each of R1′, R2′, R1′′, R2′′, R1, R2, R3, R4, and R5 is, independently, if present, H, halo, hydroxy, thiol, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, optionally substituted hydroxyalkoxy, optionally substituted amino, azido, optionally substituted aryl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, or absent; wherein the combination of R3 with one or more of R1′, R1′′, R2′, R2′′, or R5 (e.g., the combination of R1′ and R3, the combination of R1′′ and R3, the combination of R2′ and R3, the combination of R2′′ and R3, or the combination of R5 and R3) can join together
  • each of Y1, Y2, and Y3, is, independently, O, S, Se, —NRN1-, optionally substituted alkylene, or optionally substituted heteroalkylene, wherein RN1 is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, or absent;
  • each Y4 is, independently, H, hydroxy, thiol, boranyl, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted thioalkoxy, optionally substituted alkoxyalkoxy, or optionally substituted amino;
  • each Y5 is, independently, O, S, Se, optionally substituted alkylene (e.g., methylene), or optionally substituted heteroalkylene;
  • n is an integer from 1 to 100,000;
  • B is a nucleobase (e.g., a purine, a pyrimidine, or derivatives thereof), wherein the combination of B and R1′, the combination of B and R2′, the combination of B and R1′′, or the combination of B and R2′′ can, taken together with the carbons to which they are attached, optionally form a bicyclic group (e.g., a bicyclic heterocyclyl) or wherein the combination of B, R1′′, and R3 or the combination of B, R2′′, and R3 can optionally form a tricyclic or tetracyclic group (e.g., a tricyclic or tetracyclic heterocyclyl, such as in Formula (IIo)-(IIp) herein).
  • a nucleobase e.g., a purine, a pyrimidine, or derivatives thereof
  • the polynucleotide, primary construct, or mmRNA includes a modified ribose.
  • the polynucleotide, primary construct, or mmRNA e.g., the first region, the first flanking region, or the second flanking region
  • the polynucleotide, primary construct, or mmRNA includes n number of linked nucleosides having Formula (Ia-2)-(Ia-5) or a pharmaceutically acceptable salt or stereoisomer thereof.
  • the polynucleotide, primary construct, or mmRNA (e.g., the first region, the first flanking region, or the second flanking region) includes n number of linked nucleosides having Formula (Ib) or Formula (Ib-1):
  • U is O, S, N(RU)nu, or C(RU)nu, wherein nu is an integer from 0 to 2 and each RU is, independently, H, halo, or optionally substituted alkyl;
  • each of R1, R3′, R3′′, and R4 is, independently, H, halo, hydroxy, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, optionally substituted hydroxyalkoxy, optionally substituted amino, azido, optionally substituted aryl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, or absent; and wherein the combination of R1 and R3′ or the combination of R1 and R3′′ can be taken together to form optionally substituted alkylene or optionally substituted heteroalkylene (e.g., to produce a locked nucleic acid);
  • each R5 is, independently, H, halo, hydroxy, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, or absent;
  • each of Y1, Y2, and Y3 is, independently, O, S, Se, —NRN1-, optionally substituted alkylene, or optionally substituted heteroalkylene, wherein RN1 is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, or optionally substituted aryl;
  • each Y4 is, independently, H, hydroxy, thiol, boranyl, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted alkoxyalkoxy, or optionally substituted amino;
  • n is an integer from 1 to 100,000;
  • B is a nucleobase
  • the polynucleotide, primary construct, or mmRNA (e.g., the first region, first flanking region, or second flanking region) includes n number of linked nucleosides having Formula (Ic):
  • U is O, S, N(RU)nu, or C(RU)nu, wherein nu is an integer from 0 to 2 and each RU is, independently, H, halo, or optionally substituted alkyl;
  • each of B1, B2, and B3 is, independently, a nucleobase (e.g., a purine, a pyrimidine, or derivatives thereof, as described herein), H, halo, hydroxy, thiol, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, optionally substituted hydroxyalkoxy, optionally substituted amino, azido, optionally substituted aryl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, or optionally substituted aminoalkynyl, wherein one and only one of B1, B2, and B3 is a nucleobase;
  • a nucleobase e.g., a purine, a pyrimidine, or derivatives thereof, as described herein
  • H halo, hydroxy, thiol, optionally substituted al
  • each of Rb1, Rb2, Rb3, R3, and R5 is, independently, H, halo, hydroxy, thiol, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, optionally substituted hydroxyalkoxy, optionally substituted amino, azido, optionally substituted aryl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl or optionally substituted aminoalkynyl;
  • each of Y1, Y2, and Y3, is, independently, O, S, Se, —NRN1-, optionally substituted alkylene, or optionally substituted heteroalkylene, wherein RN1 is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, or optionally substituted aryl;
  • each Y4 is, independently, H, hydroxy, thiol, boranyl, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted thioalkoxy, optionally substituted alkoxyalkoxy, or optionally substituted amino;
  • each Y5 is, independently, O, S, Se, optionally substituted alkylene (e.g., methylene), or optionally substituted heteroalkylene;
  • n is an integer from 1 to 100,000;
  • ring including U can include one or more double bonds.
  • the ring including U does not have a double bond between U-CB3Rb3 or between CB3Rb3-CB2Rb2.
  • the polynucleotide, primary construct, or mmRNA (e.g., the first region, first flanking region, or second flanking region) includes n number of linked nucleosides having Formula (Id):
  • U is O, S, N(RU)nu, or C(RU)nu, wherein nu is an integer from 0 to 2 and each RU is, independently, H, halo, or optionally substituted alkyl;
  • each R3 is, independently, H, halo, hydroxy, thiol, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, optionally substituted hydroxyalkoxy, optionally substituted amino, azido, optionally substituted aryl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, or optionally substituted aminoalkynyl;
  • each of Y1, Y2, and Y3, is, independently, O, S, Se, —NRN1-, optionally substituted alkylene, or optionally substituted heteroalkylene, wherein RN1 is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, or optionally substituted aryl;
  • each Y4 is, independently, H, hydroxy, thiol, boranyl, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted thioalkoxy, optionally substituted alkoxyalkoxy, or optionally substituted amino;
  • each Y5 is, independently, O, S, optionally substituted alkylene (e.g., methylene), or optionally substituted heteroalkylene;
  • n is an integer from 1 to 100,000;
  • B is a nucleobase (e.g., a purine, a pyrimidine, or derivatives thereof).
  • the polynucleotide, primary construct, or mmRNA (e.g., the first region, first flanking region, or second flanking region) includes n number of linked nucleosides having Formula (Ie):
  • each of U′ and U′′ is, independently, O, S, N(RU)nu, or C(RU)nu, wherein nu is an integer from 0 to 2 and each RU is, independently, H, halo, or optionally substituted alkyl;
  • each R6 is, independently, H, halo, hydroxy, thiol, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, optionally substituted hydroxyalkoxy, optionally substituted amino, azido, optionally substituted aryl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, or optionally substituted aminoalkynyl;
  • each Y5′ is, independently, O, S, optionally substituted alkylene (e.g., methylene or ethylene), or optionally substituted heteroalkylene;
  • n is an integer from 1 to 100,000;
  • B is a nucleobase (e.g., a purine, a pyrimidine, or derivatives thereof).
  • the polynucleotide, primary construct, or mmRNA (e.g., the first region, first flanking region, or second flanking region) includes n number of linked nucleosides having Formula (If) or (If-1):
  • each of U′ and U′′ is, independently, O, S, N, N(RU)nu, or C(RU)nu, wherein nu is an integer from 0 to 2 and each RU is, independently, H, halo, or optionally substituted alkyl (e.g., U′ is 0 and U′′ is N);
  • each of R1′, R2′, R1′′, R2′′, R3, and R4 is, independently, H, halo, hydroxy, thiol, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, optionally substituted hydroxyalkoxy, optionally substituted amino, azido, optionally substituted aryl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, or absent; and wherein the combination of R1′ and R3, the combination of R1′′ and R3, the combination of R2′ and R3, or the combination of R2′′ and R3 can be taken together to form optionally substituted alkylene or optionally substituted heteroalkylene (e.g., to produce a locked nucleic acid); each of m′ and m′′ is, independently, an integer from
  • each of Y1, Y2, and Y3, is, independently, O, S, Se, —NRN1-, optionally substituted alkylene, or optionally substituted heteroalkylene, wherein RN1 is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, or absent;
  • each Y4 is, independently, H, hydroxy, thiol, boranyl, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted thioalkoxy, optionally substituted alkoxyalkoxy, or optionally substituted amino;
  • each Y5 is, independently, O, S, Se, optionally substituted alkylene (e.g., methylene), or optionally substituted heteroalkylene;
  • n is an integer from 1 to 100,000;
  • B is a nucleobase (e.g., a purine, a pyrimidine, or derivatives thereof).
  • the ring including U has one or two double bonds.
  • polynucleotides, primary constructs, or mmRNA e.g., Formulas (Ia)-(Ia-5), (Ib)-(If-1), (IIa)-(IIp), (IIb-1), (IIb-2), (IIc-1)-(IIc-2), (IIn-1), (IIn-2), (IVa)-(IVl), and (IXa)-(IXr)), each of R1, R1′, and R1′′, if present, is H.
  • Formulas (Ia)-(Ia-5), (Ib)-(If-1), (IIa)-(IIp), (IIb-1), (IIb-2), (IIc-1)-(IIc-2), (IIn-1), (IIn-2), (IVa)-(IVl), and (IXa)-(IXr) each of R1, R1′, and R1′′, if present, is H.
  • each of R2, R2′, and R2′′ is, independently, H, halo (e.g., fluoro), hydroxy, optionally substituted alkoxy (e.g., methoxy or ethoxy), or optionally substituted alkoxyalkoxy.
  • alkoxyalkoxy is —(CH2)s2(OCH2CH2)s1(CH2)s3OR′, wherein s1 is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R′ is H or C1-20 alkyl).
  • s2 is 0, s1 is 1 or 2
  • s3 is 0 or 1
  • R′ is C1-6 alkyl.
  • polynucleotides, primary constructs, or mmRNA e.g., Formulas (Ia)-(Ia-5), (Ib)-(If-1), (IIa)-(IIp), (IIb-1), (IIb-2), (IIc-1)-(IIc-2), (IIn-1), (IIn-2), (IVa)-(IVl), and (IXa)-(IXr)), each of R2, R2′, and R2′′, if present, is H.
  • Formulas (Ia)-(Ia-5), (Ib)-(If-1), (IIa)-(IIp), (IIb-1), (IIb-2), (IIc-1)-(IIc-2), (IIn-1), (IIn-2), (IVa)-(IVl), and (IXa)-(IXr) each of R2, R2′, and R2′′, if present, is H.
  • each of R1, R1′, and R1′′ is, independently, H, halo (e.g., fluoro), hydroxy, optionally substituted alkoxy (e.g., methoxy or ethoxy), or optionally substituted alkoxyalkoxy.
  • alkoxyalkoxy is —(CH2)s2(OCH2CH2)s1(CH2)s3OR′, wherein s1 is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R′ is H or C1-20 alkyl).
  • s2 is 0, s1 is 1 or 2
  • s3 is 0 or 1
  • R′ is C1-6 alkyl.
  • each of R3, R4, and R5 is, independently, H, halo (e.g., fluoro), hydroxy, optionally substituted alkyl, optionally substituted alkoxy (e.g., methoxy or ethoxy), or optionally substituted alkoxyalkoxy.
  • R3 is H, R4 is H, R5 is H, or R3, R4, and R5 are all H.
  • R3 is C1-6 alkyl
  • R4 is C1-6 alkyl
  • R5 is C1-6 alkyl
  • R3, R4, and R5 are all C1-6 alkyl.
  • R3 and R4 are both H, and R5 is C1-6 alkyl.
  • R3 and R5 join together to form optionally substituted alkylene or optionally substituted heteroalkylene and, taken together with the carbons to which they are attached, provide an optionally substituted heterocyclyl (e.g., a bicyclic, tricyclic, or tetracyclic heterocyclyl, such as trans-3′,4′ analogs, wherein R3 and R5 join together to form heteroalkylene (e.g., —(CH2)b1O(CH2)b2O(CH2)b3-, wherein each of b1,
  • polynucleotides, primary constructs, or mmRNA e.g., Formulas (Ia)-(Ia-5), (Ib)-(If-1), (IIa)-(IIp), (IIb-1), (IIb-2), (IIc-1)-(IIc-2), (IIn-1), (IIn-2), (IVa)-(IVl), and (IXa)-(IXr)
  • R3 and one or more of R1′, R1′′, R2′, R2′′, or R5 join together to form optionally substituted alkylene or optionally substituted heteroalkylene and, taken together with the carbons to which they are attached, provide an optionally substituted heterocyclyl (e.g., a bicyclic, tricyclic, or tetracyclic heterocyclyl, R3 and one or more of R1′, R1′′, R2′, R2′′, or R5 join together to form heteroalkylene (e.g.,
  • polynucleotides, primary constructs, or mmRNA e.g., Formulas (Ia)-(Ia-5), (Ib)-(If-1), (IIa)-(IIp), (IIb-1), (IIb-2), (IIc-1)-(IIc-2), (IIn-1), (IIn-2), (IVa)-(IV1), and (IXa)-(IXr)
  • R5 and one or more of R1′, R1′′, R2′, or R2′′ join together to form optionally substituted alkylene or optionally substituted heteroalkylene and, taken together with the carbons to which they are attached, provide an optionally substituted heterocyclyl (e.g., a bicyclic, tricyclic, or tetracyclic heterocyclyl, R5 and one or more of R1′, R1′′, R2′, or R2′′ join together to form heteroalkylene (e.g., —(CH2)b
  • each Y2 is, independently, O, S, or —NRN1-, wherein RN1 is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, or optionally substituted aryl.
  • Y2 is NRN1-, wherein RN1 is H or optionally substituted alkyl (e.g., C1-6 alkyl, such as methyl, ethyl, isopropyl, or n-propyl).
  • RN1 is H or optionally substituted alkyl (e.g., C1-6 alkyl, such as methyl, ethyl, isopropyl, or n-propyl).
  • each Y3 is, independently, O or S.
  • R1 is H; each R2 is, independently, H, halo (e.g., fluoro), hydroxy, optionally substituted alkoxy (e.g., methoxy or ethoxy), or optionally substituted alkoxyalkoxy (e.g., —(CH2)s2(OCH2CH2)s1(CH2)s3OR′, wherein s1 is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to
  • R3 is H, halo (e.g., fluoro), hydroxy, optionally substituted alkyl, optionally substituted alkoxy (e.g., methoxy or ethoxy), or optionally substituted alkoxyalkoxy.
  • halo e.g., fluoro
  • hydroxy optionally substituted alkyl
  • optionally substituted alkoxy e.g., methoxy or ethoxy
  • optionally substituted alkoxyalkoxy optionally substituted alkoxyalkoxy.
  • each Y1 is, independently, O or —NRN1-, wherein RN1 is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, or optionally substituted aryl (e.g., wherein RN1 is H or optionally substituted alkyl (e.g., C1-6 alkyl, such as methyl, ethyl, isopropyl, or n-propyl)); and each Y4 is, independently, H, hydroxy, thiol, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted thioalkoxy, optionally substituted alkoxyalkoxy, or optionally substituted amino.
  • RN1 is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, or optionally substituted aryl (e.g., wherein RN1 is H or optionally substituted alkyl (e.g., C
  • each R1 is, independently, H, halo (e.g., fluoro), hydroxy, optionally substituted alkoxy (e.g., methoxy or ethoxy), or optionally substituted alkoxyalkoxy (e.g., —(CH2)s2(OCH2CH2)s1(CH2)s3OR′, wherein s1 is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g.,
  • R3 is H, halo (e.g., fluoro), hydroxy, optionally substituted alkyl, optionally substituted alkoxy (e.g., methoxy or ethoxy), or optionally substituted alkoxyalkoxy.
  • halo e.g., fluoro
  • hydroxy optionally substituted alkyl
  • optionally substituted alkoxy e.g., methoxy or ethoxy
  • optionally substituted alkoxyalkoxy optionally substituted alkoxyalkoxy.
  • each Y1 is, independently, O or —NRN1-, wherein RN1 is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, or optionally substituted aryl (e.g., wherein RN1 is H or optionally substituted alkyl (e.g., C1-6 alkyl, such as methyl, ethyl, isopropyl, or n-propyl)); and each Y4 is, independently, H, hydroxy, thiol, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted thioalkoxy, optionally substituted alkoxyalkoxy, or optionally substituted amino.
  • RN1 is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, or optionally substituted aryl (e.g., wherein RN1 is H or optionally substituted alkyl (e.g., C
  • the ring including U is in the ⁇ -D (e.g., ⁇ -D-ribo) configuration.
  • the ring including U is in the ⁇ -L (e.g., ⁇ -L-ribo) configuration.
  • one or more B is not pseudouridine (w) or 5-methyl-cytidine (m5C).
  • about 10% to about 100% of n number of B nucleobases is not w or m5C (e.g., from 10% to 20%, from 10% to 35%, from 10% to 50%, from 10% to 60%, from 10% to 75%, from 10% to 90%, from 10% to 95%, from 10% to 98%, from 10% to 99%, from 20% to 35%, from 20% to 50%, from 20% to 60%, from 20% to 75%, from 20% to 90%, from 20% to 95%, from 20% to 98%, from 20% to 99%, from 20% to 100%, from 50% to 60%, from 50% to 75%, from 50% to 90%, from 50% to 95%, from 50% to 98%, from 50% to 99%, from 50% to 100%, from 75% to 90%, from 75% to 95%, from 75% to 98%, from 75% to 99%, and from 75% to 100% of n number of B is not ⁇ or m5C). In some embodiments, B is not ⁇ or m5C.
  • polynucleotides, primary constructs, or mmRNA e.g., Formulas (Ia)-(Ia-5), (Ib)-(If-1), (IIa)-(IIp), (IIb-1), (IIb-2), (IIn-1), (IVa)-(IVl), and (IXa)-(IXr)
  • B is an unmodified nucleobase selected from cytosine, guanine, uracil and adenine, then at least one of Y1, Y2, or Y3 is not 0.
  • the polynucleotide, primary construct, or mmRNA includes a modified ribose.
  • the polynucleotide, primary construct, or mmRNA e.g., the first region, the first flanking region, or the second flanking region
  • U is O or C(RU)nu, wherein nu is an integer from 0 to 2 and each RU is, independently, H, halo, or optionally substituted alkyl (e.g., U is —CH2- or —CH—).
  • each of R1, R2, R3, R4, and R5 is, independently, H, halo, hydroxy, thiol, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, optionally substituted hydroxyalkoxy, optionally substituted amino, azido, optionally substituted aryl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, or absent (e.g., each R1 and R2 is, independently, H, halo, hydroxy, optionally substituted alkyl, or optionally substituted alkoxy; each R3 and R4 is, independently, H or optionally substituted alkyl; and R5 is H or hydroxy), and - - - is a single bond or double bond.
  • the polynucleotides or mmRNA includes n number of linked nucleosides having Formula (IIb-1)-(IIb-2):
  • U is O or C(RU)nu, wherein nu is an integer from 0 to 2 and each RU is, independently, H, halo, or optionally substituted alkyl (e.g., U is —CH2- or —CH—).
  • each of R1 and R2 is, independently, H, halo, hydroxy, thiol, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, optionally substituted hydroxyalkoxy, optionally substituted amino, azido, optionally substituted aryl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, or absent (e.g., each R1 and R2 is, independently, H, halo, hydroxy, optionally substituted alkyl, or optionally substituted alkoxy, e.g., H, halo, hydroxy, alkyl, or alkoxy).
  • R2 is hydroxy or optionally substituted alkoxy (e.g., methoxy, ethoxy, or any described herein).
  • the polynucleotide, primary construct, or mmRNA includes n number of linked nucleosides having Formula (IIc-1)-(IIc-4):
  • U is O or C(RU)nu, wherein nu is an integer from 0 to 2 and each RU is, independently, H, halo, or optionally substituted alkyl (e.g., U is —CH2- or —CH—).
  • each of R1, R2, and R3 is, independently, H, halo, hydroxy, thiol, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, optionally substituted hydroxyalkoxy, optionally substituted amino, azido, optionally substituted aryl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, or absent (e.g., each R1 and R2 is, independently, H, halo, hydroxy, optionally substituted alkyl, or optionally substituted alkoxy, e.g., H, halo, hydroxy, alkyl, or alkoxy; and each R3 is, independently, H or optionally substituted alkyl)).
  • R2 is optionally substituted alkoxy (e.g., methoxy or ethoxy, or any described herein).
  • R1 is optionally substituted alkyl
  • R2 is hydroxy.
  • R1 is hydroxy
  • R2 is optionally substituted alkyl.
  • R3 is optionally substituted alkyl.
  • the polynucleotide, primary construct, or mmRNA includes an acyclic modified ribose.
  • the polynucleotide, primary construct, or mmRNA e.g., the first region, the first flanking region, or the second flanking region
  • the polynucleotide, primary construct, or mmRNA includes n number of linked nucleosides having Formula (IId)-(IIf):
  • the polynucleotide, primary construct, or mmRNA includes an acyclic modified hexitol.
  • the polynucleotide, primary construct, or mmRNA e.g., the first region, the first flanking region, or the second flanking region
  • the polynucleotide, primary construct, or mmRNA includes a sugar moiety having a contracted or an expanded ribose ring.
  • the polynucleotide, primary construct, or mmRNA e.g., the first region, the first flanking region, or the second flanking region
  • each of R1′, R1′′, R2′, and R2′′ is, independently, H, halo, hydroxy, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, or absent; and wherein the combination of R2′ and R3 or the combination of R2′′ and R3 can be taken together to form optionally substituted alkylene or optionally substituted heteroalkylene.
  • the polynucleotide, primary construct, or mmRNA includes a locked modified ribose.
  • the polynucleotide, primary construct, or mmRNA e.g., the first region, the first flanking region, or the second flanking region
  • the polynucleotide, primary construct, or mmRNA includes n number of linked nucleosides having Formula (IIn):
  • R3′ is O, S, or —NRN1-, wherein RN1 is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, or optionally substituted aryl and R3′′ is optionally substituted alkylene (e.g., —CH2-, —CH2CH2-, or —CH2CH2CH2-) or optionally substituted heteroalkylene (e.g., —CH2NH—, —CH2CH2NH—, —CH2OCH2-, or —CH2CH2OCH2-)(e.g., R3′ is O and R3′′ is optionally substituted alkylene (e.g., —CH2-, —CH2CH2-, or —CH2CH2CH2-)).
  • the polynucleotide, primary construct, or mmRNA includes n number of linked nucleosides having Formula (IIn-1)-(II-n2):
  • R3′ is O, S, or —NRN1-, wherein RN1 is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, or optionally substituted aryl and R3′′ is optionally substituted alkylene (e.g., —CH2-, —CH2CH2-, or —CH2CH2CH2-) or optionally substituted heteroalkylene (e.g., —CH2NH—, —CH2CH2NH—, —CH2OCH2-, or —CH2CH2OCH2-) (e.g., R3′ is O and R3′′ is optionally substituted alkylene (e.g., —CH2-, —CH2CH2-, or —CH2CH2CH2-)).
  • the polynucleotide, primary construct, or mmRNA includes a locked modified ribose that forms a tetracyclic heterocyclyl.
  • the polynucleotide, primary construct, or mmRNA e.g., the first region, the first flanking region, or the second flanking region
  • R12a, R12c, T1′, T1′′, T2′, T2′′, V1, and V3 are as described herein.
  • any of the formulas for the polynucleotides, primary constructs, or mmRNA can include one or more nucleobases described herein (e.g., Formulas (b1)-(b43)).
  • the present invention provides methods of preparing a polynucleotide, primary construct, or mmRNA, wherein the polynucleotide comprises n number of nucleosides having Formula (Ia), as defined herein:
  • the present invention provides methods of amplifying a polynucleotide, primary construct, or mmRNA comprising at least one nucleotide (e.g., mmRNA molecule), the method comprising: reacting a compound of Formula (IIIa), as defined herein, with a primer, a cDNA template, and an RNA polymerase.
  • a nucleotide e.g., mmRNA molecule
  • the present invention provides methods of amplifying a polynucleotide, primary construct, or mmRNA comprising at least one nucleotide (e.g., mmRNA molecule), the method comprising:
  • the present invention provides methods of amplifying a modified mRNA comprising at least one nucleotide (e.g., mmRNA molecule), the method comprising: reacting a compound of Formula (IIIa-2), as defined herein, with a primer, a cDNA template, and an RNA polymerase.
  • a modified mRNA comprising at least one nucleotide (e.g., mmRNA molecule)
  • the method comprising: reacting a compound of Formula (IIIa-2), as defined herein, with a primer, a cDNA template, and an RNA polymerase.
  • reaction may be repeated from 1 to about 7,000 times.
  • B may be a nucleobase of Formula (b1)-(b43).
  • polynucleotides, primary constructs, and mmRNA can optionally include 5′ and/or 3′ flanking regions, which are described herein.
  • RNA Modified RNA (mmRNA) Molecules
  • the present invention also includes building blocks, e.g., modified ribonucleosides, modified ribonucleotides, of modified RNA (mmRNA) molecules.
  • building blocks e.g., modified ribonucleosides, modified ribonucleotides, of modified RNA (mmRNA) molecules.
  • these building blocks can be useful for preparing the polynucleotides, primary constructs, or mmRNA of the invention.
  • the building block molecule has Formula (IIa) or (IIa-1):
  • the building block molecule which may be incorporated into a polynucleotide, primary construct, or mmRNA, has Formula (IVa)-(IVb):
  • Formula (IVa) or (IVb) is combined with a modified uracil (e.g., any one of formulas (b1)-(b9), (b21)-(b23), and (b28)-(b31), such as formula (b1), (b8), (b28), (b29), or (b30)).
  • a modified uracil e.g., any one of formulas (b1)-(b9), (b21)-(b23), and (b28)-(b31), such as formula (b1), (b8), (b28), (b29), or (b30)
  • Formula (IVa) or (IVb) is combined with a modified cytosine (e.g., any one of formulas (b10)-(b14), (b24), (b25), and (b32)-(b36), such as formula (b10) or (b32)).
  • a modified cytosine e.g., any one of formulas (b10)-(b14), (b24), (b25), and (b32)-(b36), such as formula (b10) or (b32)
  • Formula (IVa) or (IVb) is combined with a modified guanine (e.g., any one of formulas (b15)-(b17) and (b37)-(b40)).
  • Formula (IVa) or (IVb) is combined with a modified adenine (e.g., any one of formulas (b18)-(b20) and (b41)-(b43)).
  • the building block molecule which may be incorporated into a polynucleotide, primary construct, or mmRNA, has Formula (IVc)-(IVk):
  • one of Formulas (IVc)-(IVk) is combined with a modified uracil (e.g., any one of formulas (b1)-(b9), (b21)-(b23), and (b28)-(b31), such as formula (b1), (b8), (b28), (b29), or (b30)).
  • a modified uracil e.g., any one of formulas (b1)-(b9), (b21)-(b23), and (b28)-(b31), such as formula (b1), (b8), (b28), (b29), or (b30)
  • one of Formulas (IVc)-(IVk) is combined with a modified cytosine (e.g., any one of formulas (b10)-(b14), (b24), (b25), and (b32)-(b36), such as formula (b10) or (b32)).
  • one of Formulas (IVc)-(IVk) is combined with a modified guanine (e.g., any one of formulas (b15)-(b17) and (b37)-(b40)).
  • one of Formulas (IVc)-(IVk) is combined with a modified adenine (e.g., any one of formulas (b18)-(b20) and (b41)-(b43)).
  • the building block molecule which may be incorporated into a polynucleotide, primary construct, or mmRNA, has Formula (Va) or (Vb):
  • B is as described herein (e.g., any one of (b1)-(b43)).
  • the building block molecule which may be incorporated into a polynucleotide, primary construct, or mmRNA, has Formula (IXa)-(IXd):
  • one of Formulas (IXa)-(IXd) is combined with a modified uracil (e.g., any one of formulas (b1)-(b9), (b21)-(b23), and (b28)-(b31), such as formula (b1), (b8), (b28), (b29), or (b30)).
  • a modified uracil e.g., any one of formulas (b1)-(b9), (b21)-(b23), and (b28)-(b31), such as formula (b1), (b8), (b28), (b29), or (b30)
  • one of Formulas (IXa)-(IXd) is combined with a modified cytosine (e.g., any one of formulas (b10)-(b14), (b24), (b25), and (b32)-(b36), such as formula (b10) or (b32)).
  • one of Formulas (IXa)-(IXd) is combined with a modified guanine (e.g., any one of formulas (b15)-(b17) and (b37)-(b40)).
  • one of Formulas (IXa)-(IXd) is combined with a modified adenine (e.g., any one of formulas (b18)-(b20) and (b41)-(b43)).
  • the building block molecule which may be incorporated into a polynucleotide, primary construct, or mmRNA, has Formula (IXe)-(IXg):
  • one of Formulas (IXe)-(IXg) is combined with a modified uracil (e.g., any one of formulas (b1)-(b9), (b21)-(b23), and (b28)-(b31), such as formula (b1), (b8), (b28), (b29), or (b30)).
  • a modified uracil e.g., any one of formulas (b1)-(b9), (b21)-(b23), and (b28)-(b31), such as formula (b1), (b8), (b28), (b29), or (b30)
  • one of Formulas (IXe)-(IXg) is combined with a modified cytosine (e.g., any one of formulas (b10)-(b14), (b24), (b25), and (b32)-(b36), such as formula (b10) or (b32)).
  • one of Formulas (IXe)-(IXg) is combined with a modified guanine (e.g., any one of formulas (b15)-(b17) and (b37)-(b40)).
  • one of Formulas (IXe)-(IXg) is combined with a modified adenine (e.g., any one of formulas (b18)-(b20) and (b41)-(b43)).
  • the building block molecule which may be incorporated into a polynucleotide, primary construct, or mmRNA, has Formula (IXh)-(IXk):
  • one of Formulas (IXh)-(IXk) is combined with a modified uracil (e.g., any one of formulas (b1)-(b9), (b21)-(b23), and (b28)-(b31), such as formula (b1), (b8), (b28), (b29), or (b30)).
  • a modified uracil e.g., any one of formulas (b1)-(b9), (b21)-(b23), and (b28)-(b31), such as formula (b1), (b8), (b28), (b29), or (b30)
  • one of Formulas (IXh)-(IXk) is combined with a modified cytosine (e.g., any one of formulas (b10)-(b14), (b24), (b25), and (b32)-(b36), such as formula (b10) or (b32)).
  • one of Formulas (IXh)-(IXk) is combined with a modified guanine (e.g., any one of formulas (b15)-(b17) and (b37)-(b40)).
  • one of Formulas (IXh)-(IXk) is combined with a modified adenine (e.g., any one of formulas (b18)-(b20) and (b41)-(b43)).
  • the building block molecule which may be incorporated into a polynucleotide, primary construct, or mmRNA, has Formula (IXl)-(IXr):
  • each r1 and r2 is, independently, an integer from 0 to 5 (e.g., from 0 to 3, from 1 to 3, or from 1 to 5) and B is as described herein (e.g., any one of (b1)-(b43)).
  • one of Formulas (IXl)-(IXr) is combined with a modified uracil (e.g., any one of formulas (b1)-(b9), (b21)-(b23), and (b28)-(b31), such as formula (b1), (b8), (b28), (b29), or (b30)).
  • one of Formulas (IXl)-(IXr) is combined with a modified cytosine (e.g., any one of formulas (b10)-(b14), (b24), (b25), and (b32)-(b36), such as formula (b10) or (b32)).
  • one of Formulas (IXl)-(IXr) is combined with a modified guanine (e.g., any one of formulas (b15)-(b17) and (b37)-(b40)).
  • one of Formulas (IXl)-(IXr) is combined with a modified adenine (e.g., any one of formulas (b18)-(b20) and (b41)-(b43)).
  • the building block molecule which may be incorporated into a polynucleotide, primary construct, or mmRNA, can be selected from the group consisting of:
  • each r is, independently, an integer from 0 to 5 (e.g., from 0 to 3, from 1 to 3, or from 1 to 5).
  • each r is, independently, an integer from 0 to 5 (e.g., from 0 to 3, from 1 to 3, or from 1 to 5) and s1 is as described herein.
  • the building block molecule which may be incorporated into a nucleic acid (e.g., RNA, mRNA, polynucleotide, primary construct, or mmRNA), is a modified uridine (e.g., selected from the group consisting of:
  • Y1, Y3, Y4, Y6, and r are as described herein (e.g., each r is, independently, an integer from 0 to 5, such as from 0 to 3, from 1 to 3, or from 1 to 5)).
  • the building block molecule which may be incorporated into a polynucleotide, primary construct, or mmRNA, is a modified cytidine (e.g., selected from the group consisting of:
  • the building block molecule which may be incorporated into a polynucleotide, primary construct, or mmRNA, can be:
  • each r is, independently, an integer from 0 to 5 (e.g., from 0 to 3, from 1 to 3, or from 1 to 5).
  • the building block molecule which may be incorporated into a polynucleotide, primary construct, or mmRNA, is a modified adenosine (e.g., selected from the group consisting of:
  • Y1, Y3, Y4, Y6, and r are as described herein (e.g., each r is, independently, an integer from 0 to 5, such as from 0 to 3, from 1 to 3, or from 1 to 5)).
  • the building block molecule which may be incorporated into a polynucleotide, primary construct, or mmRNA, is a modified guanosine (e.g., selected from the group consisting of:
  • Y1, Y3, Y4, Y6, and r are as described herein (e.g., each r is, independently, an integer from 0 to 5, such as from 0 to 3, from 1 to 3, or from 1 to 5)).
  • the chemical modification can include replacement of C group at C-5 of the ring (e.g., for a pyrimidine nucleoside, such as cytosine or uracil) with N (e.g., replacement of the >CH group at C-5 with >NRN1 group, wherein RN1 is H or optionally substituted alkyl).
  • the building block molecule which may be incorporated into a polynucleotide, primary construct, or mmRNA, can be:
  • each r is, independently, an integer from 0 to 5 (e.g., from 0 to 3, from 1 to 3, or from 1 to 5).
  • the chemical modification can include replacement of the hydrogen at C-5 of cytosine with halo (e.g., Br, Cl, F, or I) or optionally substituted alkyl (e.g., methyl).
  • halo e.g., Br, Cl, F, or I
  • optionally substituted alkyl e.g., methyl
  • the building block molecule which may be incorporated into a polynucleotide, primary construct, or mmRNA, can be:
  • each r is, independently, an integer from 0 to 5 (e.g., from 0 to 3, from 1 to 3, or from 1 to 5).
  • the 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.
  • the building block molecule which may be incorporated into a polynucleotide, primary construct, or mmRNA, can be:
  • each r is, independently, an integer from 0 to 5 (e.g., from 0 to 3, from 1 to 3, or from 1 to 5).
  • modified nucleosides and nucleotides which may be incorporated into a polynucleotide, primary construct, or mmRNA (e.g., RNA or mRNA, as described herein), can be modified on the sugar of the ribonucleic acid.
  • the 2′ hydroxyl group (OH) can be modified or replaced with a number of different substituents.
  • substitutions at the 2′-position include, but are not limited to, H, halo, optionally substituted C1-6 alkyl; optionally substituted C1-6 alkoxy; optionally substituted C6-10 aryloxy; optionally substituted C3-8 cycloalkyl; optionally substituted C3-8 cycloalkoxy; optionally substituted C6-10 aryloxy; optionally substituted C6-10 aryl-C1-6 alkoxy, optionally substituted C1-12 (heterocyclyl)oxy; a sugar (e.g., ribose, pentose, or any described herein); a polyethyleneglycol (PEG), —O(CH2CH2O)nCH2CH2OR, where R is H or optionally substituted alkyl, and n is an integer from 0 to 20 (e.g., from 0 to 4, from 0 to 8, from 0 to 10, from 0 to 16, from 1 to 4, from 1 to 8, from 1 to 10, from 1 to 16, from 1 to 20, from
  • RNA includes the sugar group ribose, which is a 5-membered ring having an oxygen.
  • modified nucleotides include replacement of the oxygen in ribose (e.g., with S, Se, or alkylene, such as methylene or ethylene); addition of a double bond (e.g., to replace ribose with cyclopentenyl or cyclohexenyl); ring contraction of ribose (e.g., to form a 4-membered ring of cyclobutane or oxetane); ring expansion of ribose (e.g., to form a 6- or 7-membered ring having an additional carbon or heteroatom, such as for anhydrohexitol, altritol, mannitol, cyclohexanyl, cyclohexenyl, and morpholino that also has a phosphoramidate backbone); multicyclic forms (e.
  • the sugar group can also contain one or more carbons that possess the opposite stereochemical configuration than that of the corresponding carbon in ribose.
  • a polynucleotide, primary construct, or mmRNA molecule can include nucleotides containing, e.g., arabinose, as the sugar.
  • nucleoside is defined as a compound containing a sugar molecule (e.g., a pentose or ribose) or a derivative thereof in combination with an organic base (e.g., a purine or pyrimidine) or a derivative thereof (also referred to herein as “nucleobase”).
  • organic base e.g., a purine or pyrimidine
  • nucleotide is defined as a nucleoside including a phosphate group.
  • the modified nucleotides may by synthesized by any useful method, as described herein (e.g., chemically, enzymatically, or recombinantly to include one or more modified or non-natural nucleosides).
  • the modified nucleotide base pairing encompasses not only the standard adenosine-thymine, adenosine-uracil, or guanosine-cytosine base pairs, but also base pairs formed between nucleotides and/or modified nucleotides comprising non-standard or modified bases, wherein the arrangement of hydrogen bond donors and hydrogen bond acceptors permits hydrogen bonding between a non-standard base and a standard base or between two complementary non-standard base structures.
  • non-standard base pairing is the base pairing between the modified nucleotide inosine and adenine, cytosine or uracil.
  • the modified nucleosides and nucleotides can include a modified nucleobase.
  • nucleobases found in RNA include, but are not limited to, adenine, guanine, cytosine, and uracil.
  • nucleobase found in DNA include, but are not limited to, adenine, guanine, cytosine, and thymine.
  • These nucleobases can be modified or wholly replaced to provide polynucleotides, primary constructs, or mmRNA molecules having enhanced properties, e.g., resistance to nucleases through disruption of the binding of a major groove binding partner.
  • Table 8 below identifies the chemical faces of each canonical nucleotide. Circles identify the atoms comprising the respective chemical regions.
  • B is a modified uracil.
  • exemplary modified uracils include those having Formula (b1)-(b5):
  • each of T1′, T1′′, T2′, and T2′′ is, independently, H, optionally substituted alkyl, optionally substituted alkoxy, or optionally substituted thioalkoxy, or the combination of T1′ and T1′′ or the combination of T2′ and T2′′ join together (e.g., as in T2) to form O (oxo), S (thio), or Se (seleno);
  • each of V1 and V2 is, independently, O, S, N(RVb)nv, or C(RVb)nv, wherein nv is an integer from 0 to 2 and each RVb is, independently, H, halo, optionally substituted amino acid, optionally substituted alkyl, optionally substituted haloalkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted
  • R10 is H, halo, optionally substituted amino acid, hydroxy, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aminoalkyl, optionally substituted hydroxyalkyl, optionally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, optionally substituted alkoxy, optionally substituted alkoxycarbonylalkyl, optionally substituted alkoxycarbonylalkenyl, optionally substituted alkoxycarbonylalkynyl, optionally substituted alkoxycarbonylalkoxy, optionally substituted carboxyalkoxy, optionally substituted carboxyalkyl, or optionally substituted carbamoylalkyl;
  • R11 is H or optionally substituted alkyl
  • R12a is H, optionally substituted alkyl, optionally substituted hydroxyalkyl, optionally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, or optionally substituted aminoalkynyl, optionally substituted carboxyalkyl (e.g., optionally substituted with hydroxy), optionally substituted carboxyalkoxy, optionally substituted carboxyaminoalkyl, or optionally substituted carbamoylalkyl; and
  • R12c is H, halo, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted thioalkoxy, optionally substituted amino, optionally substituted hydroxyalkyl, optionally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, or optionally substituted aminoalkynyl.
  • Other exemplary modified uracils include those having Formula (b6)-(b9):
  • each of T1′, T1′′, T2′, and T2′′ is, independently, H, optionally substituted alkyl, optionally substituted alkoxy, or optionally substituted thioalkoxy, or the combination of T1′ and T1′′ join together (e.g., as in T1) or the combination of T2′ and T2′′ join together (e.g., as in T2) to form O (oxo), S (thio), or Se (seleno), or each T1 and T2 is, independently, O (oxo), S (thio), or Se (seleno);
  • each of W1 and W2 is, independently, N(RWa)nw or C(RWa)nw, wherein nw is an integer from 0 to 2 and each RWa is, independently, H, optionally substituted alkyl, or optionally substituted alkoxy;
  • each V3 is, independently, O, S, N(RVa)nv, or C(RVa)nv, wherein nv is an integer from 0 to 2 and each RVa is, independently, H, halo, optionally substituted amino acid, optionally substituted alkyl, optionally substituted hydroxyalkyl, optionally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted heterocyclyl, optionally substituted alkheterocyclyl, optionally substituted alkoxy, optionally substituted alkenyloxy, or optionally substituted alkynyloxy, optionally substituted aminoalkyl (e.g., substituted with an N-protecting group, such as any described herein, e.g., trifluoroacetyl, or sulfoalkyl), optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, optional
  • R12a is H, optionally substituted alkyl, optionally substituted hydroxyalkyl, optionally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, optionally substituted carboxyalkyl (e.g., optionally substituted with hydroxy and/or an O-protecting group), optionally substituted carboxyalkoxy, optionally substituted carboxyaminoalkyl, optionally substituted carbamoylalkyl, or absent;
  • R12b is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted hydroxyalkyl, optionally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, optionally substituted alkaryl, optionally substituted heterocyclyl, optionally substituted alkheterocyclyl, optionally substituted amino acid, optionally substituted alkoxycarbonylacyl, optionally substituted alkoxycarbonylalkoxy, optionally substituted alkoxycarbonylalkyl, optionally substituted alkoxycarbonylalkenyl, optionally substituted alkoxycarbonylalkynyl, optionally substituted alkoxycarbonylalkoxy, optionally substituted carboxyalkyl (e.g., optionally substituted with hydroxy and/or an O-protecting group), optionally substitute
  • R12c is H, halo, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted thioalkoxy, optionally substituted amino, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, or optionally substituted aminoalkynyl.
  • modified uracils include those having Formula (b28)-(b31):
  • R12a is H, optionally substituted alkyl, optionally substituted carboxyaminoalkyl, optionally substituted aminoalkyl (e.g., e.g., substituted with an N-protecting group, such as any described herein, e.g., trifluoroacetyl, or sulfoalkyl), optionally substituted aminoalkenyl, or optionally substituted aminoalkynyl; and
  • R12b is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted hydroxyalkyl, optionally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, optionally substituted aminoalkynyl (e.g., e.g., substituted with an N-protecting group, such as any described herein, e.g., trifluoroacetyl, or sulfoalkyl),
  • optionally substituted alkoxycarbonylacyl optionally substituted alkoxycarbonylalkoxy, optionally substituted alkoxycarbonylalkyl, optionally substituted alkoxycarbonylalkenyl, optionally substituted alkoxycarbonylalkynyl, optionally substituted alkoxycarbonylalkoxy, optionally substituted carboxyalkoxy, optionally substituted carboxyalkyl, or optionally substituted carbamoylalkyl.
  • T1 is O (oxo), and T2 is S (thio) or Se (seleno). In other embodiments, T1 is S (thio), and T2 is O (oxo) or Se (seleno).
  • RVb′ is H, optionally substituted alkyl, or optionally substituted alkoxy.
  • each R12a and R12b is, independently, H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, or optionally substituted hydroxyalkyl.
  • R12a is H.
  • both R12a and R12b are H.
  • each RVb′ of R12b is, independently, optionally substituted aminoalkyl (e.g., substituted with an N-protecting group, such as any described herein, e.g., trifluoroacetyl, or sulfoalkyl), optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, or optionally substituted acylaminoalkyl (e.g., substituted with an N-protecting group, such as any described herein, e.g., trifluoroacetyl).
  • an N-protecting group such as any described herein, e.g., trifluoroacetyl
  • the amino and/or alkyl of the optionally substituted aminoalkyl is substituted with one or more of optionally substituted alkyl, optionally substituted alkenyl, optionally substituted sulfoalkyl, optionally substituted carboxy (e.g., substituted with an O-protecting group), optionally substituted hydroxy (e.g., substituted with an O-protecting group), optionally substituted carboxyalkyl (e.g., substituted with an O-protecting group), optionally substituted alkoxycarbonylalkyl (e.g., substituted with an O-protecting group), or N-protecting group.
  • optionally substituted alkyl optionally substituted alkenyl, optionally substituted sulfoalkyl
  • optionally substituted carboxy e.g., substituted with an O-protecting group
  • optionally substituted hydroxy e.g., substituted with an O-protecting group
  • optionally substituted carboxyalkyl e.g.,
  • optionally substituted aminoalkyl is substituted with an optionally substituted sulfoalkyl or optionally substituted alkenyl.
  • R12a and RVb′′ are both H.
  • T1 is O (oxo)
  • T2 is S (thio) or Se (seleno).
  • RVb′ is optionally substituted alkoxycarbonylalkyl or optionally substituted carbamoylalkyl.
  • the optional substituent for R12a, R12b, R12c, or RVa is a polyethylene glycol group (e.g., —(CH2)s2(OCH2CH2)s1(CH2)s3OR′, wherein s1 is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R′ is H or C1-20 alkyl); or an amino-polyethylene glycol group (e.g., —NRN1(CH2)s2(CH2CH2O)s1(CH2)s3NRN1, wherein s1 is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from
  • B is a modified cytosine.
  • exemplary modified cytosines include compounds of Formula (b10)-(b14):
  • each of T3′ and T3′′ is, independently, H, optionally substituted alkyl, optionally substituted alkoxy, or optionally substituted thioalkoxy, or the combination of T3′ and T3′′ join together (e.g., as in T3) to form O (oxo), S (thio), or Se (seleno); each V4 is, independently, O, S, N(RVc)nv, or C(RVc)nv, wherein nv is an integer from 0 to 2 and each RVc is, independently, H, halo, optionally substituted amino acid, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted heterocyclyl, optionally substituted alkheterocyclyl, or optionally substituted alkynyloxy (e.g., optionally substituted
  • each V5 is, independently, N(RVd)nv, or C(RVd)nv, wherein nv is an integer from 0 to 2 and each RVd is, independently, H, halo, optionally substituted amino acid, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted heterocyclyl, optionally substituted alkheterocyclyl, or optionally substituted alkynyloxy (e.g., optionally substituted with any substituent described herein, such as those selected from (1)-(21) for alkyl) (e.g., V5 is —CH or N);
  • each of R13a and R13b is, independently, H, optionally substituted acyl, optionally substituted acyloxyalkyl, optionally substituted alkyl, or optionally substituted alkoxy, wherein the combination of R13b and R14 can be taken together to form optionally substituted heterocyclyl;
  • each R14 is, independently, H, halo, hydroxy, thiol, optionally substituted acyl, optionally substituted amino acid, optionally substituted alkyl, optionally substituted haloalkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted hydroxyalkyl (e.g., substituted with an O-protecting group), optionally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, optionally substituted acyloxyalkyl, optionally substituted amino (e.g., —NHR, wherein R is H, alkyl, aryl, or phosphoryl), azido, optionally substituted aryl, optionally substituted heterocyclyl, optionally substituted alkheterocyclyl, optionally
  • each of R15 and R16 is, independently, H, optionally substituted alkyl, optionally substituted alkenyl, or optionally substituted alkynyl.
  • modified cytosines include those having Formula (b32)-(b35):
  • each of T1 and T3 is, independently, O (oxo), S (thio), or Se (seleno); each of R13a and R13b is, independently, H, optionally substituted acyl, optionally substituted acyloxyalkyl, optionally substituted alkyl, or optionally substituted alkoxy, wherein the combination of R13b and R14 can be taken together to form optionally substituted heterocyclyl;
  • each R14 is, independently, H, halo, hydroxy, thiol, optionally substituted acyl, optionally substituted amino acid, optionally substituted alkyl, optionally substituted haloalkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted hydroxyalkyl (e.g., substituted with an O-protecting group), optionally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, optionally substituted acyloxyalkyl, optionally substituted amino (e.g., —NHR, wherein R is H, alkyl, aryl, or phosphoryl), azido, optionally substituted aryl, optionally substituted heterocyclyl, optionally substituted alkheterocyclyl, optionally
  • each of R15 and R16 is, independently, H, optionally substituted alkyl, optionally substituted alkenyl, or optionally substituted alkynyl (e.g., R15 is H, and R16 is H or optionally substituted alkyl).
  • R15 is H, and R16 is H or optionally substituted alkyl.
  • R14 is H, acyl, or hydroxyalkyl.
  • R14 is halo.
  • both R14 and R15 are H.
  • both R15 and R16 are H.
  • each of R14 and R15 and R16 is H.
  • each of R13a and R13b is independently, H or optionally substituted alkyl.
  • modified cytosines include compounds of Formula (b36):
  • each R13b is, independently, H, optionally substituted acyl, optionally substituted acyloxyalkyl, optionally substituted alkyl, or optionally substituted alkoxy, wherein the combination of R13b and R14b can be taken together to form optionally substituted heterocyclyl; each R14a and R14b is, independently, H, halo, hydroxy, thiol, optionally substituted acyl, optionally substituted amino acid, optionally substituted alkyl, optionally substituted haloalkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted hydroxyalkyl (e.g., substituted with an O-protecting group), optionally substituted hydroxyalkenyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, optionally substituted
  • each of R15 is, independently, H, optionally substituted alkyl, optionally substituted alkenyl, or optionally substituted alkynyl.
  • R14b is an optionally substituted amino acid (e.g., optionally substituted lysine). In some embodiments, R14a is H.
  • B is a modified guanine.
  • exemplary modified guanines include compounds of Formula (b15)-(b17):
  • each of T4′, T4′′, T5′, T5′′, T6′, and T6′′ is, independently, H, optionally substituted alkyl, or optionally substituted alkoxy, and wherein the combination of T4′ and T4′′ (e.g., as in T4) or the combination of T5′ and T5′′ (e.g., as in T5) or the combination of T6′ and T6′′ (e.g., as in T6) join together form O (oxo), S (thio), or Se (seleno);
  • each of V5 and V6 is, independently, O, S, N(RVd)nv, or C(RVd)nv, wherein nv is an integer from 0 to 2 and each RVd is, independently, H, halo, thiol, optionally substituted amino acid, cyano, amidine, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted alkoxy, optionally substituted alkenyloxy, or optionally substituted alkynyloxy (e.g., optionally substituted with any substituent described herein, such as those selected from (1)-(21) for alkyl), optionally substituted thioalkoxy, or optionally substituted amino; and
  • each of R17, R18, R19a, R19b, R21, R22, R23, and R24 is, independently, H, halo, thiol, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted thioalkoxy, optionally substituted amino, or optionally substituted amino acid.
  • Exemplary modified guanosines include compounds of Formula (b37)-(b40):
  • each of T4′ is, independently, H, optionally substituted alkyl, or optionally substituted alkoxy, and each T4 is, independently, O (oxo), S (thio), or Se (seleno); each of R18, R19a, R19b, and R21 is, independently, H, halo, thiol, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted thioalkoxy, optionally substituted amino, or optionally substituted amino acid.
  • R18 is H or optionally substituted alkyl.
  • T4 is oxo.
  • each of R19a and R19b is, independently, H or optionally substituted alkyl.
  • B is a modified adenine.
  • exemplary modified adenines include compounds of Formula (b18)-(b20):
  • each V7 is, independently, O, S, N(RVe)nv, or C(RVe)nv, wherein nv is an integer from 0 to 2 and each RVe is, independently, H, halo, optionally substituted amino acid, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted alkoxy, optionally substituted alkenyloxy, or optionally substituted alkynyloxy (e.g., optionally substituted with any substituent described herein, such as those selected from (1)-(21) for alkyl);
  • each R25 is, independently, H, halo, thiol, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted thioalkoxy, or optionally substituted amino;
  • each of R26a and R26b is, independently, H, optionally substituted acyl, optionally substituted amino acid, optionally substituted carbamoylalkyl, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted hydroxyalkyl, optionally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally substituted alkoxy, or polyethylene glycol group (e.g., —(CH2)s2(OCH2CH2)s1(CH2)s3OR′, wherein s1 is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R′ is H or C1-20 alkyl); or an amino-polyethylene glycol group (e.g., —
  • each R27 is, independently, H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted alkoxy, optionally substituted thioalkoxy or optionally substituted amino;
  • each R28 is, independently, H, optionally substituted alkyl, optionally substituted alkenyl, or optionally substituted alkynyl;
  • each R29 is, independently, H, optionally substituted acyl, optionally substituted amino acid, optionally substituted carbamoylalkyl, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted hydroxyalkyl, optionally substituted hydroxyalkenyl, optionally substituted alkoxy, or optionally substituted amino.
  • Exemplary modified adenines include compounds of Formula (b41)-(b43):
  • each R25 is, independently, H, halo, thiol, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted thioalkoxy, or optionally substituted amino; each of R26a and R26b is, independently, H, optionally substituted acyl, optionally substituted amino acid, optionally substituted carbamoylalkyl, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted hydroxyalkyl, optionally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally substituted alkoxy, or polyethylene glycol group (e.g., —(CH2)s2(OCH2CH2)s1(CH2)s3OR′, wherein s1 is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of
  • each R27 is, independently, H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted alkoxy, optionally substituted thioalkoxy, or optionally substituted amino.
  • R26a is H, and R26b is optionally substituted alkyl. In some embodiments, each of R26a and R26b is, independently, optionally substituted alkyl. In particular embodiments, R27 is optionally substituted alkyl, optionally substituted alkoxy, or optionally substituted thioalkoxy. In other embodiments, R25 is optionally substituted alkyl, optionally substituted alkoxy, or optionally substituted thioalkoxy.
  • the optional substituent for R26a, R26b, or R29 is a polyethylene glycol group (e.g., —(CH2)s2(OCH2CH2)s1(CH2)s3OR′, wherein s1 is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R′ is H or C1-20 alkyl); or an amino-polyethylene glycol group (e.g., —NRN1(CH2)s2(CH2CH2O)s1(CH2)s3NRN1, wherein s1 is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from
  • B may have Formula (b21):
  • X12 is, independently, O, S, optionally substituted alkylene (e.g., methylene), or optionally substituted heteroalkylene
  • xa is an integer from 0 to 3
  • R12a and T2 are as described herein.
  • B may have Formula (b22):
  • R10′ is, independently, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted heterocyclyl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, optionally substituted alkoxy, optionally substituted alkoxycarbonylalkyl, optionally substituted alkoxycarbonylalkenyl, optionally substituted alkoxycarbonylalkynyl, optionally substituted alkoxycarbonylalkoxy, optionally substituted carboxyalkoxy, optionally substituted carboxyalkyl, or optionally substituted carbamoylalkyl, and R11, R12a, T1, and T2 are as described herein.
  • B may have Formula (b23):
  • R10 is optionally substituted heterocyclyl (e.g., optionally substituted furyl, optionally substituted thienyl, or optionally substituted pyrrolyl), optionally substituted aryl (e.g., optionally substituted phenyl or optionally substituted naphthyl), or any substituent described herein (e.g., for R10); and wherein R11 (e.g., H or any substituent described herein), R12a (e.g., H or any substituent described herein), T1 (e.g., oxo or any substituent described herein), and T2 (e.g., oxo or any substituent described herein) are as described herein.
  • R11 e.g., H or any substituent described herein
  • R12a e.g., H or any substituent described herein
  • T1 e.g., oxo or any substituent described herein
  • T2 e.g., oxo or any substitu
  • B may have Formula (b24):
  • R14′ is, independently, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted heterocyclyl, optionally substituted alkaryl, optionally substituted alkheterocyclyl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, optionally substituted alkoxy, optionally substituted alkoxycarbonylalkenyl, optionally substituted alkoxycarbonylalkynyl, optionally substituted alkoxycarbonylalkyl, optionally substituted alkoxycarbonylalkoxy, optionally substituted carboxyalkoxy, optionally substituted carboxyalkyl, or optionally substituted carbamoylalkyl, and R13a, R13b, R15, and T3 are as described herein.
  • B may have Formula (b25):
  • R14′ is optionally substituted heterocyclyl (e.g., optionally substituted furyl, optionally substituted thienyl, or optionally substituted pyrrolyl), optionally substituted aryl (e.g., optionally substituted phenyl or optionally substituted naphthyl), or any substituent described herein (e.g., for R14 or R14′); and wherein R13a (e.g., H or any substituent described herein), R13b (e.g., H or any substituent described herein), R15 (e.g., H or any substituent described herein), and T3 (e.g., oxo or any substituent described herein) are as described herein.
  • R13a e.g., H or any substituent described herein
  • R13b e.g., H or any substituent described herein
  • R15 e.g., H or any substituent described herein
  • T3 e.g., oxo or any
  • B is a nucleobase selected from the group consisting of cytosine, guanine, adenine, and uracil. In some embodiments, B may be:
  • the modified nucleobase is a modified uracil.
  • exemplary nucleobases and nucleosides having a modified uracil include pseudouridine (w), pyridin-4-one ribonucleoside, 5-aza-uridine, 6-aza-uridine, 2-thio-5-aza-uridine, 2-thio-uridine (s2U), 4-thio-uridine (s4U), 4-thio-pseudouridine, 2-thio-pseudouridine, 5-hydroxy-uridine (ho5U), 5-aminoallyl-uridine, 5-halo-uridine (e.g., 5-iodo-uridine or 5-bromo-uridine), 3-methyl-uridine (m3U), 5-methoxy-uridine (mo5U), uridine 5-oxyacetic acid (cmo5U), uridine 5-oxyacetic acid methyl ester (mcmo5U), 5-carboxymethyl-uridine (cm5U), 1-car
  • the modified nucleobase is a modified cytosine.
  • exemplary nucleobases and nucleosides having a modified cytosine include 5-aza-cytidine, 6-aza-cytidine, pseudoisocytidine, 3-methyl-cytidine (m3C), N4-acetyl-cytidine (ac4C), 5-formyl-cytidine (f5C), N4-methyl-cytidine (m4C), 5-methyl-cytidine (m5C), 5-halo-cytidine (e.g., 5-iodo-cytidine), 5-hydroxymethyl-cytidine (hm5C), 1-methyl-pseudoisocytidine, pyrrolo-cytidine, pyrrolo-pseudoisocytidine, 2-thio-cytidine (s2C), 2-thio-5-methyl-cytidine, 4-thio-pseudoisocy
  • the modified nucleobase is a modified adenine.
  • exemplary nucleobases and nucleosides having a modified adenine include 2-amino-purine, 2, 6-diaminopurine, 2-amino-6-halo-purine (e.g., 2-amino-6-chloro-purine), 6-halo-purine (e.g., 6-chloro-purine), 2-amino-6-methyl-purine, 8-azido-adenosine, 7-deaza-adenine, 7-deaza-8-aza-adenine, 7-deaza-2-amino-purine, 7-deaza-8-aza-2-amino-purine, 7-deaza-2,6-diaminopurine, 7-deaza-8-aza-2,6-diaminopurine, 1-methyl-adenosine (m1A), 2-methyl-adenine (m2A), N6-methyl-adenosine (m1A
  • the modified nucleobase is a modified guanine.
  • exemplary nucleobases and nucleosides having a modified guanine include inosine (I), 1-methyl-inosine (m1I), wyosine (imG), methylwyosine (mimG), 4-demethyl-wyosine (imG-14), isowyosine (imG2), wybutosine (yW), peroxywybutosine (o2yW), hydroxywybutosine (OHyW), undermodified hydroxywybutosine (OHyW*), 7-deaza-guanosine, queuosine (Q), epoxyqueuosine (oQ), galactosyl-queuosine (galQ), mannosyl-queuosine (manQ), 7-cyano-7-deaza-guanosine (preQ0), 7-aminomethyl-7-deaza-guanosine (preQ0
  • the nucleobase of the nucleotide can be independently selected from a purine, a pyrimidine, a purine or pyrimidine analog.
  • the nucleobase can each be independently selected from adenine, cytosine, guanine, uracil, or hypoxanthine.
  • the nucleobase can also include, for example, naturally-occurring and synthetic derivatives of a base, including pyrazolo[3,4-d]pyrimidines, 5-methylcytosine (5-me-C), 5-hydroxymethyl cytosine, xanthine, hypoxanthine, 2-aminoadenine, 6-methyl and other alkyl derivatives of adenine and guanine, 2-propyl and other alkyl derivatives of adenine and guanine, 2-thiouracil, 2-thiothymine and 2-thiocytosine, 5-propynyl uracil and cytosine, 6-azo uracil, cytosine and thymine, 5-uracil (pseudouracil), 4-thiouracil, 8-halo (e.g., 8-bromo), 8-amino, 8-thiol, 8-thioalkyl, 8-hydroxyl and other 8-substituted adenines and
  • each letter refers to the representative base and/or derivatives thereof, e.g., A includes adenine or adenine analogs, e.g., 7-deaza adenine).
  • the modified nucleotides which may be incorporated into a polynucleotide, primary construct, or mmRNA molecule, can be modified on the internucleoside linkage (e.g., phosphate backbone).
  • internucleoside linkage e.g., phosphate backbone
  • the phrases “phosphate” and “phosphodiester” are used interchangeably.
  • Backbone phosphate groups can be modified by replacing one or more of the oxygen atoms with a different substituent.
  • the modified nucleosides and nucleotides can include the wholesale replacement of an unmodified phosphate moiety with another internucleoside linkage as described herein.
  • modified phosphate groups include, but are not limited to, phosphorothioate, phosphoroselenates, boranophosphates, boranophosphate esters, hydrogen phosphonates, phosphoramidates, phosphorodiamidates, alkyl or aryl phosphonates, and phosphotriesters.
  • Phosphorodithioates have both non-linking oxygens replaced by sulfur.
  • the phosphate linker can also be modified by the replacement of a linking oxygen with nitrogen (bridged phosphoramidates), sulfur (bridged phosphorothioates), and carbon (bridged methylene-phosphonates).
  • the ⁇ -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 polynucleotides, primary constructs, or mmRNA molecules are expected to also reduce the innate immune response through weaker binding/activation of cellular innate immune molecules.
  • a modified nucleoside includes an alpha-thio-nucleoside (e.g., 5′-O-(1-thiophosphate)-adenosine, 5′-O-(1-thiophosphate)-cytidine ( ⁇ -thio-cytidine), 5′-O-(1-thiophosphate)-guanosine, 5′-O-(1-thiophosphate)-uridine, or 5′-O-(1-thiophosphate)-pseudouridine).
  • alpha-thio-nucleoside e.g., 5′-O-(1-thiophosphate)-adenosine, 5′-O-(1-thiophosphate)-cytidine ( ⁇ -thio-cytidine), 5′-O-(1-thiophosphate)-guanosine, 5′-O-(1-thiophosphate)-uridine, or 5′-O-(1-thiophosphate)-p
  • internucleoside linkages that may be employed according to the present invention, including internucleoside linkages which do not contain a phosphorous atom, are described herein below.
  • the polynucleotides, primary constructs, and mmRNA of the invention can include a combination of modifications to the sugar, the nucleobase, and/or the internucleoside linkage. These combinations can include any one or more modifications described herein.
  • any of the nucleotides described herein in Formulas (Ia), (Ia-1)-(Ia-3), (Ib)-(If), (IIa)-(IIp), (IIb-1), (IIb-2), (IIc-1)-(IIc-2), (IIn-2), (IVa)-(IVl), and (IXa)-(IXr) can be combined with any of the nucleobases described herein (e.g., in Formulas (b1)-(b43) or any other described herein).
  • the polynucleotides, primary constructs or mmRNA of the present invention may incorporate one or more cytotoxic nucleosides.
  • cytotoxic nucleosides may be incorporated into polynucleotides, primary constructs or mmRNA such as bifunctional modified RNAs or mRNAs.
  • Cytotoxic nucleoside anti-cancer agents include, but are not limited to, adenosine arabinoside, cytarabine, cytosine arabinoside, 5-fluorouracil, fludarabine, floxuridine, FTORAFUR® (a combination of tegafur and uracil), tegafur ((RS)-5-fluoro-1-(tetrahydrofuran-2-yl)pyrimidine-2,4(1H,3H)-dione), and 6-mercaptopurine.
  • cytotoxic nucleoside analogues are in clinical use, or have been the subject of clinical trials, as anticancer agents.
  • examples of such analogues include, but are not limited to, cytarabine, gemcitabine, troxacitabine, decitabine, tezacitabine, 2′-deoxy-2′-methylidenecytidine (DMDC), cladribine, clofarabine, 5-azacytidine, 4′-thio-aracytidine, cyclopentenylcytosine and 1-(2-C-cyano-2-deoxy-beta-D-arabino-pentofuranosyl)-cytosine.
  • Another example of such a compound is fludarabine phosphate.
  • cytotoxic nucleoside analogues examples include, but are not limited to, N4-behenoyl-1-beta-D-arabinofuranosylcytosine, N4-octadecyl-1-beta-D-arabinofuranosylcytosine, N4-palmitoyl-1-(2-C-cyano-2-deoxy-beta-D-arabino-pentofuranosyl) cytosine, and P-4055 (cytarabine 5′-elaidic acid ester).
  • these prodrugs may be converted into the active drugs mainly in the liver and systemic circulation and display little or no selective release of active drug in the tumor tissue.
  • active drug for example, capecitabine, a prodrug of 5′-deoxy-5-fluorocytidine (and eventually of 5-fluorouracil), is metabolized both in the liver and in the tumor tissue.
  • capecitabine analogues containing “an easily hydrolysable radical under physiological conditions” has been claimed by Fujiu et al. (U.S. Pat. No. 4,966,891) and is herein incorporated by reference.
  • Cytotoxic nucleotides which may be chemotherapeutic also include, but are not limited to, pyrazolo [3,4-D]-pyrimidines, allopurinol, azathioprine, capecitabine, cytosine arabinoside, fluorouracil, mercaptopurine, 6-thioguanine, acyclovir, ara-adenosine, ribavirin, 7-deaza-adenosine, 7-deaza-guanosine, 6-aza-uracil, 6-aza-cytidine, thymidine ribonucleotide, 5-bromodeoxyuridine, 2-chloro-purine, and inosine, or combinations thereof.
  • modified nucleosides and nucleotides used in the synthesis of polynucleotides, primary constructs, and mmRNA molecules disclosed herein can be prepared from readily available starting materials using the following general methods and procedures. Where typical or preferred process conditions (e.g., reaction temperatures, times, mole ratios of reactants, solvents, pressures, etc.) are provided, a skilled artisan would be able to optimize and develop additional process conditions. 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.
  • process conditions e.g., reaction temperatures, times, mole ratios of reactants, solvents, pressures, etc.
  • 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.
  • spectroscopic means such as nuclear magnetic resonance spectroscopy (e.g., 1H or 13C) infrared spectroscopy, spectrophotometry (e.g., UV-visible), or mass spectrometry
  • chromatography such as high performance liquid chromatography (HPLC) or thin layer chromatography.
  • Preparation of polypeptides, primary constructs, and mmRNA molecules of the present invention 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).
  • 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 be prepared according to the synthetic methods described in Ogata et al., J. Org. Chem. 74:2585-2588 (2009); Purmal et al., Nucl. Acids Res. 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.
  • polypeptides, primary constructs, and mmRNA of the invention may or may not be uniformly modified along the entire length of the molecule.
  • nucleotide e.g., purine or pyrimidine, or any one or more or all of A, G, U, C
  • a polynucleotide of the invention or in a given predetermined sequence region thereof (e.g. one or more of the sequence regions represented in FIG. 1 ).
  • nucleotides X in a polynucleotide of the invention are modified, wherein X may any one of nucleotides A, G, U, C, or any one of the combinations A+G, A+U, A+C, G+U, G+C, U+C, A+G+U, A+G+C, G+U+C or A+G+C.
  • nucleotide modifications may exist at various positions in the polynucleotide, primary construct, or mmRNA.
  • nucleotide analogs or other modification(s) may be located at any position(s) of a polynucleotide, primary construct, or mmRNA such that the function of the polynucleotide, primary construct, or mmRNA is not substantially decreased.
  • a modification may also be a 5′ or 3′ terminal modification.
  • the polynucleotide, primary construct, or mmRNA may contain from about 1% to about 100% modified nucleotides (either in relation to overall nucleotide content, or in relation to one or more types of nucleotide, i.e.
  • any one or more of A, G, U or C) or any intervening percentage e.g., from 1% to 20%, from 1% to 25%, from 1% to 50%, from 1% to 60%, from 1% to 70%, from 1% to 80%, from 1% to 90%, from 1% to 95%, from 10% to 20%, from 10% to 25%, from 10% to 50%, from 10% to 60%, from 10% to 70%, from 10% to 80%, from 10% to 90%, from 10% to 95%, from 10% to 100%, from 20% to 25%, from 20% to 50%, from 20% to 60%, from 20% to 70%, from 20% to 80%, from 20% to 90%, from 20% to 95%, from 20% to 100%, from 50% to 60%, from 50% to 70%, from 50% to 80%, from 50% to 90%, from 50% to 95%, from 50% to 100%, from 70% to 80%, from 70% to 90%, from 70% to 95%, from 70% to 100%, from 80% to 90%, from 80% to 95%, from 90% to 100%, and from 95% to 100%).
  • any intervening percentage e.g.,
  • the polynucleotide, primary construct, or mmRNA includes a modified pyrimidine (e.g., a modified uracil/uridine/U or modified cytosine/cytidine/C).
  • the uracil or uridine (generally: U) in the polynucleotide, primary construct, or mmRNA molecule may be replaced with from about 1% to about 100% of a modified uracil or modified uridine (e.g., from 1% to 20%, from 1% to 25%, from 1% to 50%, from 1% to 60%, from 1% to 70%, from 1% to 80%, from 1% to 90%, from 1% to 95%, from 10% to 20%, from 10% to 25%, from 10% to 50%, from 10% to 60%, from 10% to 70%, from 10% to 80%, from 10% to 90%, from 10% to 95%, from 10% to 100%, from 20% to 25%, from 20% to 50%, from 20% to 60%, from 20% to 70%, from 20% to 80%, from 20% to 80%, from 20% to
  • the modified uracil or uridine can be replaced by a compound having a single unique structure or by a plurality of compounds having different structures (e.g., 2, 3, 4 or more unique structures, as described herein).
  • the cytosine or cytidine (generally: C) in the polynucleotide, primary construct, or mmRNA molecule may be replaced with from about 1% to about 100% of a modified cytosine or modified cytidine (e.g., from 1% to 20%, from 1% to 25%, from 1% to 50%, from 1% to 60%, from 1% to 70%, from 1% to 80%, from 1% to 90%, from 1% to 95%, from 10% to 20%, from 10% to 25%, from 10% to 50%, from 10% to 60%, from 10% to 70%, from 10% to 80%, from 10% to 90%, from 10% to 95%, from 10% to 100%, from 20% to 25%, from 20% to 50%, from 20% to 60%, from 20% to 70%, from 20% to 80%, from 20% to 90%, from from 10%
  • the present disclosure provides methods of synthesizing a polynucleotide, primary construct, or mmRNA (e.g., the first region, first flanking region, or second flanking region) including n number of linked nucleosides having Formula (Ia-1):
  • Y9 is H, hydroxy, phosphoryl, pyrophosphate, sulfate, amino, thiol, optionally substituted amino acid, or a peptide (e.g., including from 2 to 12 amino acids); and each P1, P2, and P3 is, independently, a suitable protecting group; and denotes a solid support;
  • steps a) and b) are repeated from 1 to about 10,000 times.
  • the methods further comprise a nucleotide (e.g., mmRNA molecule) selected from the group consisting of A, C, G and U adenosine, cytosine, guanosine, and uracil.
  • the nucleobase may be a pyrimidine or derivative thereof.
  • the polynucleotide, primary construct, or mmRNA is translatable.
  • polynucleotides, primary constructs, and mmRNA are 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 nucleotide modifications.
  • nucleotide modifications may also be present in the translatable region.
  • polynucleotides, primary constructs, and mmRNA containing a Kozak sequence are also be present in the translatable region.
  • modified nucleotides and modified nucleotide combinations are provided below in Table 9. These combinations of modified nucleotides can be used to form the polypeptides, primary constructs, or mmRNA of the invention. Unless otherwise noted, the modified nucleotides may be completely substituted for the natural nucleotides of the modified nucleic acids or mmRNA of the invention. As a non-limiting example, the natural nucleotide uridine may be substituted with a modified nucleoside described herein.
  • the natural nucleotide uridine may be partially substituted (e.g., about 0.1%, 1%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 99.9%) with at least one of the modified nucleoside disclosed herein.
  • modified nucleotide combinations are provided below in Table 10. These combinations of modified nucleotides can be used to form the polypeptides, primary constructs, or mmRNA of the invention.
  • At least 25% of the cytosines are replaced by a compound of Formula (b10)-(b14) (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 (b10)-(b14) 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 (b1)-(b9) (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 (b1)-(b9) 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 are replaced by a compound of Formula (b10)-(b14), and at least 25% of the uracils are replaced by a compound of Formula (b1)-(b9) (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 plasmid pJ344:91543-TC-GCSF includes the coding sequence for GCSF (granulyte colony stimulating factor) and 141 nucleotide sequence coding for Poly A Tail and an XBaI restriction endonuclease recognition site immediately downstream of the poly A tail sequence tract.
  • a plasmid map is shown in FIG. 5
  • a plasmid DNA sequence is shown in FIG. 6 .
  • An E. coli strain DH10B harboring above plasmid was grown in 250 ml animal-free broth with ampicillin. This process generated 1107 ⁇ g of plasmid DNA.
  • the plasmid DNA was isolated and purified according to DNA2.0 standard operating procedures. Plasmid DNA yield and DNA homogeneity was determined using agarose gel electrophoresis. The insert (but not vector backbone) was sequence verified in both orientations using a capillary electrophoresis DNA analyzer.
  • the plasmid DNA template was linearized using the restriction endonuclease XbaI.
  • the XbaI restriction digest reaction conditions were as follows:
  • the reaction proceeded at 37° C. overnight.
  • the linearized plasmid was diafiltered into 10 mM Tris HCl pH 7.5 and concentrated to 959 ng/uL prior to use using 100 kDa MWCO Amicon spin filters (EMD Millipore).
  • the non-amplified, linearized DNA plasmid was used as a template for in vitro transcription.
  • a 1 mL transcription reaction was performed utilizing 250 ⁇ g of plasmid template.
  • Nucleotides ATP, GTP, 5mCTP (5-methyl cytosine triphosphate) and N-1-methylpseudouridine triphosphate were added at 7.5 mM each.
  • RNase inhibitor (1000 U), inorganic pyrophosphatase (1U) and T7 RNA polymerase (7000U) were added.
  • the final buffer conditions (1 ⁇ ) were as follows: 40 mM Tris HCl pH8, 40 mM magnesium acetate, 5 mM dithiothreitol (DTT), and 1 mM spermidine.
  • the in vitro transcription reaction proceeded for 4 hours at 37° C. under constant mixing.
  • the total reaction yield was 5.4 mg RNA transcript.
  • a 400 uL (40%) portion of the reaction was diafiltered into water and concentrated to 622 ng/ ⁇ L using 100 kDa MWCO Amicon spin filters (EMD Millipore).
  • RNA transcript The full length poly A containing RNA transcript was purified from truncated RNA, DNA template, and residual enzymes using oligo dT chromatography.
  • a 20 mer polythymidine Sepharose (3 ml) was packed in a 5 mL SPE column on a solid phase extraction vacuum manifold.
  • the RNA transcript (2.18 mg) was applied to column, followed by washing and elution.
  • the oligo dT purified RNA transcript was diafiltered into water and concentrated to 1.22 mg/mL using 100 kDa MWCO Amicon spin filters (EMD Millipore). Approximately 1.82 mg of 2.18 mg was recovered (83%) as determined by Bioanalyzer gel elctrophoresis.
  • RNA transcript was analyzed using a Bioanalyzer gel electrohphoresis. Sample taken both before and after oligo dT purification were analyzed. The results are shown in the electopherograms in FIGS. 7A and 7B .
  • RNA transcript A 5′ cap was enzymatically added to the RNA transcript. Approximately 0.98 mg of oligo dT purified RNA transcript (1225 ng/uL) was capped to obtain a 5′ Cap 1 structure. GTP was added at a final reaction concentration of 0.9 mM. SAM (S-adenosyl-L-methionine) was added at a final reaction concentration of 0.4 mM. 907 units of RNase inhibitor, 3630 units of 2′O-methyltransferase, and 363 units of vaccinia guanylyltransferase were added to the reaction.
  • SAM S-adenosyl-L-methionine
  • the final 1 ⁇ buffer conditions consist of the following: 50 mM Tris HCl pH 8, 5 mM KCl, 1 mM MgCl2, and 1 mM dithiothreitol. Final reaction volume was 1070 ⁇ L. The reaction proceeded at 37° C. for 2 hours under constant mixing. Reaction setup for capped RNA (lot 12-04-155-C) is shown below.
  • RNA transcript The projected sequence of the RNA transcript is below:
  • RNA transcript was purified using 2 mL of 20mer polythymidine Sepharose packed in a 5 mL SPE column on a solid phase extraction vacuum manifold. 0.82 mg was recovered in the elution (84% recovery) The RNA was diafiltered into water and harvested at a concentration of 366 ng/uL using 100 kDa MWCO Amicon spin filters (EMD Millipore). The results are shown in FIG. 8 .
  • Example 8 Protein Expression Using Capped RNA Transcript Produced by the Method of the Invention
  • 12-04-155-C produced using a linearized Poly A:T tract DNA template and enzymatic capping yielded ⁇ 1,150 ng/mL GCSF expression, significantly higher than the four lots that utilized PCR product as a DNA template: 12-04-89-C, 12-04-106-C, 12-04-114-C, and 12-04-122-C.
  • Example 9 Interferon-a Induction Assessment Using Capped RNA Transcript Produced by the Method of the Invention
  • RNAs were transfected into Peripheral blood mononuclear cells (PBMC) to assay in vitro cytokine induction. The results are shown in FIG. 10 .
  • two of four samples that utilized PCR product as a DNA template 12-04-89-C, 12-04-106-C showed some level of interferon- ⁇ induction.
  • Poly IC Polyinosinic:polycytidylic acid
  • R-848 is an imidazoquinoline compound that is also used as a positive control.
  • the plasmid pJ204109475 includes the coding sequence for Factor IX and 141 nucleotide sequence coding for Poly A Tail and an SapI restriction endonuclease recognition site immediately downstream of the poly A tail sequence tract.
  • FIGS. 11A and 11B A plasmid map and DNA sequence are shown in FIGS. 11A and 11B .

Abstract

Described are methods for production of RNA transcripts using a non-amplified, linearized DNA tempate in an in vitro transcription reaction. Enzymatic 5′ capping and oligo dT purification can also be included in the methods.

Description

    CROSS REFERENCE TO RELATED APPLICATIONS
  • Not applicable.
  • SEQUENCE LISTING
  • Not applicable.
  • STATEMENT REGARDING FEDERALLY SPONSORED RESEARCH OR DEVELOPMENT
  • Not applicable.
  • BACKGROUND OF THE INVENTION Field of the Invention
  • The invention relates to methods useful for manufacture of RNA transcripts, e.g., mRNA.
  • Description of the Related Art
  • The manufacturing process used to produce clinical grade chemically modified mRNA therapeutics must be able to generate mRNA of high purity and potency, consistently, reproducibly, and in compliance with current good manufacturing practices (cGMP). The RNA must be as homogeneous as possible; this includes obtaining a uniform cap structure/5′ terminus, correct sequence, correct poly A tail length and minimizing the formation of product related impurities.
  • To generate poly A tail containing mRNAs, several approaches have been taken utilizing in vitro transcription. The first is transcription using a “tailless” DNA template. A 3′ Poly A tail is added, post-transcriptionally using a Poly A polymerase. Tails greater than 100 bases are typically generated. The downfall of this approach is the difficulty of controlling tail length and the evolution of tail length distribution in the RNA.
  • The use of PCR to generate DNA templates containing a Poly A:T tract is another approach. The poly A:T tracts are introduced via PCR primers, introducing another step for creation of impurities.
  • SUMMARY OF THE INVENTION
  • Disclosed is a method for production of an RNA transcript, e.g., mRNA, using a non-amplified, linearized DNA template in an in vitro transcription reaction to generate the RNA transcript. In some embodiments the RNA transcript is capped via enzymatic capping. The method results in production of homogeneous RNA transcripts with high purity and potency. In some embodiments the RNA transcript is purified via chromatographic methods, e.g., use of an oligo dT substrate. In one embodiment the method excludes the use of DNase.
  • BRIEF DESCRIPTION OF THE SEVERAL VIEWS OF THE DRAWINGS
  • FIG. 1 is a flow chart of one embodiment of the method.
  • FIG. 2 illustrates overhangs of linearized plasmid DNA template with different endonucleases, according to one embodiment.
  • FIG. 3 illustrates an mRNA cap structure, according to one embodiment.
  • FIG. 4 illustrates an examplary RNA trasncript diagram, according to one embodiment.
  • FIG. 5 is a map of plasmid pJ344:91543-TC-GCSF-pA-V1, according to one embodiment.
  • FIG. 6 is a DNA sequence of plasmid pJ344:91543-TC-GCSF-pA-V1, according to one embodiment.
  • FIGS. 7A and 7B are electropherograms of RNA transcripts, according to one embodiment.
  • FIG. 8 is an electropherogram of capped RNA, according to one embodiment.
  • FIG. 9 illustrates a bar graph showing results of an ELISA used to quantify expressed protein data, according to one embodiment.
  • FIG. 10 illustrates a bar graph showing results of an assay for determination of IFN-α induction following transfection of RNAs into PBMC, according to one embodiment.
  • FIG. 11A is a map of plasmid pJ204:109475-TC-FIX-Hs-3-pA140-Sap, according to one embodiment.
  • FIG. 11B is a DNA sequence of plasmid pJ204:109475-TC-FIX-Hs-3-pA140-Sap, according to one embodiment.
  • DETAILED DESCRIPTION OF THE INVENTION
  • Disclosed are methods for production of RNA transcripts, e.g., mRNA, useful for producing clinical grade mRNA of high purity and potency, consistently, reproducibly, and in compliance with current good manufacturing practices (cGMP). The method uses a non-amplified, linearized DNA template in an in vitro transcription reaction to generate the RNA transcript. In some embodiments the RNA transcript is purified via oligo dT substrate without the use of DNase. Enzymatic capping is used for 5′ capping of the RNA transcript.
  • PCR generated DNA templates have disadvantages that can be mitigated using linearized whole plasmid DNA templates A PCR-free process has the following advantages:
  • Scalability: Plasmid DNA template can be produced at microgram, milligram and gram scale in a cGMP compliant fashion. Large scale production of PCR generated templates is not commercially viable.
  • Higher purity mRNA transcripts than with PCR product templates: There is lower risk that no additional aberrant species are transcribed when using a linearized, non-amplified DNA template. Additionally, PCR amplification creates another intermediate in the manufacturing process where additional impurities may be introduced. When using PCR amplification, the gene of interest is amplified and typically does not contain a poly A tail. The poly A tail is introduced via the reverse primer, not encoded in the plasmid template. Utilizing linearized plasmid DNA template produces RNA transcripts of higher quality and purity
  • Increased process efficiency: An additional unit operation (PCR) is removed from the process increasing throughput and reducing production time.
  • Whole plasmid allows for simple DNA/RNA separation: Use of whole plasmid DNA template is more amenable to SEC and IEX chromatographic separations compared to use of PCR product due to larger disparity in size and charge density between DNA and RNA. Whole plasmid can also be removed using a poly dT based affinity purification in the flowthrough fraction.
  • In addition, there are advantages to the methods use of enzymatic capping. For large scale manufacturing, 5′ capping of RNA transcripts is typically performed using a chemical cap analog. This is performed co-transcriptionally where the cap analog: GTP molar ratio in the reaction is 4:1. This typically results in ˜80% capping efficiency, as well as reduced RNA transcript yields due to consumption of GTP. This high abundance of uncapped species is undesirable when developing therapeutic RNA. Since only capped mRNA is translated into protein, the presence of a high abundance of uncapped species (ie 20%) is problematic as efficacy (protein production/mg RNA) is reduced by 20% and 20% of the final drug substance is an inert impurity, decreasing process productivity.
  • The presence of uncapped species is also potentially immunogenic: Presence of a 5′ triphosphate motif on uncapped RNAs can be potentially immunostimulatory (see Hornung et. al and Abbas et. al). Use of cap analogs requires additional phosphatase treatment to remove the 5′-triphosphate motif from the RNAs.
  • In contrast, the use of enzymatic capping for clinical grade mRNA production is a nearly quantitative capping process that is much more efficient than co-transcriptional capping using cap analogs. This increases potency, process productivity, as well as reduces the potential for immunogenicity.
  • Definitions
  • Terms used in the claims and specification are defined as set forth below unless otherwise specified.
  • Polynucleotide. The term “polynucleotide” is interchangeable with nucleic acid, and includes any compound and/or substance that comprise a polymer of nucleotides. RNA transcripts produced by the method of the invention and DNA templates used in the methods of the invention are polynucleotides. Exemplary polynucleotides include, but are not limited to, ribonucleic acids (RNAs), deoxyribonucleic acids (DNAs), threose nucleic acids (TNAs), glycol nucleic acids (GNAs), peptide nucleic acids (PNAs), locked nucleic acids (LNAs, including LNA having a β-D-ribo configuration, α-LNA having an α-L-ribo configuration (a diastereomer of LNA), 2′-amino-LNA having a 2′-amino functionalization, and 2′-amino-α-LNA having a 2′-amino functionalization) or hybrids thereof.
  • RNA transcript. As used herein, an “RNA transcript” refers to a ribonucleic acid produced by an in vitro transcription reaction using a DNA template and an RNA polymerase. As described in more detail below, an RNA transcript typically includes the coding sequence for a gene of interest and a poly A tail. RNA transcript includes an mRNA. The RNA transcript can include modifications, e.g., modified nucleotides. As used herein, the term RNA transcript includes and is interchangeable with mRNA, modified mRNA “mmRNA” or modified mRNA, and primary construct.
  • Gene of interest. As used herein, “gene of interest” refers to a polynucleotide which encodes a polypeptide or protein of interest. Depending on the context, the gene of interest refers to a deoxyribonucleic acid, e.g., a gene of interest in a DNA template which can be transcribed to an RNA transcript, or a ribonucleic acid, e.g., a gene of interest in an RNA transcript which can be translated to produce the encoded polypeptide of interest in vitro, in vivo, in situ or ex vivo. As described in more detail below, a polypeptide of interest includes but is not limited to, biologics, antibodies, vaccines, therapeutic proteins or peptides, etc.
  • DNA template. As used herein, a DNA template refers to a polynucleotide template for RNA polymerase. Typically a DNA template includes the sequence for a gene of interest operably linked to a RNA polymerase promoter sequence.
  • Operably linked: As used herein, the phrase “operably linked” refers to a functional connection between two or more molecules, constructs, transcripts, entities, moieties or the like. For example, a gene of interest operably linked to an RNA polymerase promoter allows transcription of the gene of interest.
  • Poly A tail. As used herein, “poly A tail” refers to a chain of adenine nucleotides. The term can refer to poly A tail that is to be added to an RNA transcript, or can refer to the poly A tail that already exists at the 3′ end of an RNA transcript. As described in more detail below, a poly A tail is typically 5-300 nucleotides in length.
  • In vitro: As used herein, the term “in vitro” refers to events that occur in an artificial environment, e.g., in a test tube or reaction vessel, in cell culture, in a Petri dish, etc., rather than within an organism (e.g., animal, plant, or microbe).
  • It must be noted that, as used in the specification and the appended claims, the singular forms “a,” “an” and “the” include plural referents unless the context clearly dictates otherwise.
  • METHODS OF THE INVENTION
  • Disclosed is a method for production of an RNA transcript, e.g., mRNA, using a non-amplified, linearized DNA template in an in vitro transcription reaction to generate the RNA transcript. In some embodiments the RNA transcript is capped via enzymatic capping. The method results in production of homogeneous RNA transcripts with high purity and potency. In some embodiments the RNA transcript is purified via chromatographic methods, e.g., use of an oligo dT substrate. In one embodiment the method excludes the use of DNase.
  • In one embodiment, an RNA transcript is synthesized from a non-amplified, linear DNA template coding for the gene of interest via an enzymatic in vitro transcription reaction utilizing a T7 phage polymerase and nucleotide triphosphates of the desired chemistry. The RNA transcript is enzymatically capped post transcriptionally at the 5′ end using Vaccinia guanylyltransferase, guanosine triphosphate and s-adenosyl-L-methionine to yield cap 0 structure, i.e., an inverted 7-methylguanosine cap is added via a 5′ to 5′ triphosphate bridge. Alternatively, use of an 2′O-methyltransferase with the Vaccinia guanylyltransferase yields a cap 1 structure where in addition to the cap 0 structure, the 2′OH group is methylated on the penultimate nucleotide. In some embodiments the RNA transcript is chromatographically purified during the manufacturing process using affinity and/or anion exchange methods and diafiltered into the appropriate formulation buffer. This process is PCR free.
  • DNA Templates
  • The method used a non amplified, linearized plasmid DNA as the template for in vitro transcription. Cells, e.g., bacterial cells, e.g., E. coli, e.g., DH-1 cells are transfected with the plasmid DNA template. The transfected cells are cultured to replicate the plasmid DNA which is then isolated and purified.
  • The plasmid DNA template includes a gene of interest coding for, e.g., a polypeptide of interest. A detailed description of genes of interest is found below. In one embodiment, the gene of interest is a GCSF (granular colony stimulating factor) gene. In another embodiment, the gene of interest codes is a Factor IX gene. In another embodiment the gene of interest is a EP (erythropoietin) gene.
  • The plasmid DNA template also includes a RNA polymerase promoter, e.g., a T7 promoter located 5′ to and operably linked to the gene of interest. Also included is a sequence coding for a poly A tail located 3′ to the gene of interest. Additional description of promoters and poly A tails is found below.
  • Immediately downstream of the poly A tail coding sequence on the plasmid DNA template is a recognition site for a restriction endonuclease to linearize the plasmid. Linearization of the plasmid mitigates transcriptional readthrough. In one embodiment, the endonuclease produces a 5′ overhang on the linearized DNA template. In another embodiment, the endonuclease produces a blunt end on the linearized DNA template. In another embodiment, the endonuclease produces 3′ overhang on the linearized DNA template. Examples of endonucleases resulting in 5′ overhangs include XbaI, SapI, NotI, EcoRI, HindIII, and SPEI. FIG. 2 shows examples of such overhangs. Examples of endonucleases resulting in blunt ends include SPE1. Additional endonucleases are well known to one of skill in the art and can be used depending on the application.
  • In one embodiment, XbaI is utilized as the restriction endonuclease with a recognition site of 5′ TCTAGA3′. The five base overhang left on the DNA template sequence results in additional bases on the 3′ end of the RNA transcript post-transcription. In other embodiments, the restriction endonuclease SAP I is used. The resulting overhang on the linearized DNA template sequence does not generate additional bases on the 3′ end of the RNA transcript.
  • The plasmid into which the gene of interest, promoter, poly A tail sequence, and 5′ and 3′ UTR, and linearization restriction sites are inserted can be, e.g., one knows to one of skill in the art. Examples include but are not limited to pUC57, pJ204 (from DNA 2.0) and pJ344 (from DNA 2.0).
  • Examples of DNA templates to be used in the methods of the invention include, e.g., pJ344:91543 (including a GCSF gene and XbaI site) and pJ204:109475 (including a Factor IX gene and a SAPI site). Both plasmids are described in detail below.
  • In some embodiments, following linearization, the plasmid DNA template is filtered into an appropriate solvent, e.g., water, TE (Tris-EDTA), 10 mM Tris HCl pH 7.5, HEPES/phosphate and the like. Filtration occurs via, e.g., ultrafiltration, diafiltration, or, e.g., tangential flow ultrafiltration/diafiltration.
  • The linearized DNA template can be purified before use as a template for in vitro transcription. For example, the linearized DNA template can be purified chromatographically, or purified using a silica filter based DNA capture method.
  • In Vitro Transcription
  • The linearized DNA template is used in an in vitro transcription (IVT) system. The system typically comprises a transcription buffer, nucleotide triphosphates (NTPs), an RNase inhibitor and an RNA polymerase. The NTPs may be manufactured in house, may be selected from a supplier, or may be synthesized as described herein. The NTPs may be selected from, but are not limited to, those described herein including natural and unnatural (modified) NTPs. Additional description of modified nucleotides is found below.
  • RNA Polymerases
  • Any number of RNA polymerases or variants may be used in the method of the present invention. The polymerase may be selected from, but is not limited to, a phage RNA polymerase, e.g., a T7 RNA polymerase, a T3 RNA polymerase, an SP6 RNa polymerase, and/or mutant polymerases such as, but not limited to, polymerases able to incorporate modified nucleic acids.
  • RNA polymerases may be modified by inserting or deleting amino acids of the RNA polymerase sequence. As a non-limiting example, the RNA polymerase may be modified to exhibit an increased ability to incorporate a 2′-modified nucleotide triphosphate compared to an unmodified RNA polymerase (see International Publication WO2008078180 and U.S. Pat. No. 8,101,385; herein incorporated by reference in their entireties).
  • Variants may be obtained by evolving an RNA polymerase, optimizing the RNA polymerase amino acid and/or nucleic acid sequence and/or by using other methods known in the art. As a non-limiting example, T7 RNA polymerase variants may be evolved using the continuous directed evolution system set out by Esvelt et al. (Nature (2011) 472(7344):499-503; herein incorporated by reference in its entirety) where clones of T7 RNA polymerase may encode at least one mutation such as, but not limited to, lysine at position 93 substituted for threonine (K93T), I4M, A7T, E63V, V64D, A65E, D66Y, T76N, C125R, S128R, A136T, N165S, G175R, H176L, Y178H, F182L, L196F, G198V, D208Y, E222K, S228A, Q239R, T243N, G259D, M2671, G280C, H300R, D351A, A354S, E356D, L360P, A383V, Y385C, D388Y, S397R, M401T, N410S, K450R, P451T, G452V, E484A, H523L, H524N, G542V, E565K, K577E, K577M, N601S, S684Y, L6991, K713E, N748D, Q754R, E775K, A827V, D851N or L864F. As another non-limiting example, T7 RNA polymerase variants may encode at least mutation as described in U.S. Pub. Nos. 20100120024 and 20070117112; herein incorporated by reference in their entireties. Variants of RNA polymerase may also include, but are not limited to, substitutional variants, conservative amino acid substitution, insertional variants, deletional variants and/or covalent derivatives.
  • In one embodiment, the RNA transcript may be designed to be recognized by the wild type or variant RNA polymerases. In doing so, the RNA transcript may be modified to contain sites or regions of sequence changes from the wild type or parent primary construct.
  • Transcription Reaction Conditions
  • Typical reaction conditions for in vitro transcription can be as follows. One of skill in the art understands that parameters can be changed according to the application, e.g., the polymerase and nucleotides used.
  • The in vitro transcription reaction includes the following: an RNA polymerase, e.g., a T7 RNA polymerase at a final concentration of, e.g., 1000-12000 U/mL, e.g., 7000 U/mL; the DNA template at a final concentration of, e.g., 10-70 nM, e.g., 40 nM; nucleotides (NTPs) at a final concentration of e.g., 0.5-10 nM, e.g., 7.5 nM each; magnesisum at a final concentration of, e.g., 12-60 mM, e.g., magnesium acetate at 40 mM; a buffer such as, e.g., HEPES or Tris at a pH of, e.g., 7-8.5, e.g. 40 mM Tris HCl, pH 8
  • In some embodiments 5 mM dithiothreitol (DTT) and/or 1 mM spermidine is included. In some embodiments, an RNase inhibitor is included in the in vitro transcription reaction to ensure no RNase induced degradation during the transcription reaction. For example, murine RNase inhibitor can be utilized at a final concentration of 1000 U/mL, In some embodiments a pyrophosphatase is included in the in vitro transcription reaction to cleave the inorganic pyrophosphate generated following each nucleotide incorporation into two units of inorganic phosphate. This ensures that magnesium, which is essential for transcription, remains in solution and does not precipitate as magnesium pyrophosphate. For example, an E. Coli inorganic pyrophosphatase can utilized at a final concentration of 1 U/mL.
  • The in vitro transcription reaction is allowed to proceed, for example, under constant mixing at 37° C. for 4 hours. Typical yields can be, e.g., 5 mg of RNA per mL of transcription reaction.
  • A typical in vitro transcription reaction might have the following properties:
  • Nucelotides
  • The in vitro transcription reaction includes nucleotides (NTPs). The nucleotides can be unmodified NTPs, e.g., A, C, C, and U ribonucleotides, or modified nucleotides, or a combination. A more detailed description of modified nucleotides is found below. Examples of nucleotide combinations useful for in vitro transcription are found in the table below.
  • Chemistry A G C U
    Gen
    0 ATP GTP CTP UTP
    Gen
    1 ATP GTP 5mCTP ψTP
    Gen
    2 ATP GTP 5mCTP N-1-meψTP
    Gen
    3 ATP GTP 5mCTP 2-thio UTP
    Gen 4 ATP GTP CTP ψTP
    Gen
    5 ATP GTP CTP N-1-meψTP
  • Example Combinations of Nucleotides for In Vitro Transcription.
  • Key: ATP=Adenosine triphosphate; GTP=Guanosine triphosphate; CTP=Cytidine Triphosphate; UTP=Uridine Triphosphate; 5mCTP=5-methylcytidine triphosphate; 2-thioUTP=2-thiouridine triphosphate; ψTP=pseudouridine triphosphate; N-1-me ψTP=1-methylpseudouridine triphosphate.
  • Capping of RNA Transcripts
  • In some embodiments the RNA transcript is enzymatically capped at the 5/end after in vitro transcription. Capping can be performed either before or after further purification of the RNA transcript, e.g., oligo dT purification. If not performing purification prior to capping, an ultrafiltration/diafiltration step must be performed (e.g., buffer exchange).
  • Non-limiting examples of 5′ cap structures are those which, among other things, have enhanced binding of cap binding proteins, increased half life, reduced susceptibility to 5′ endonucleases and/or reduced 5′decapping, as compared to synthetic 5′cap structures known in the art (or to a wild-type, natural or physiological 5′cap structure). For example, recombinant Vaccinia Virus Capping Enzyme and recombinant 2′-O-methyltransferase enzyme can create a canonical 5′-5′-triphosphate linkage between the 5′-terminal nucleotide of an mRNA and a guanine cap nucleotide wherein the cap guanine contains an N7 methylation and the 5′-terminal nucleotide of the mRNA contains a 2′-O-methyl. Such a structure is termed the Cap1 structure. This cap results in a higher translational-competency and cellular stability and a reduced activation of cellular pro-inflammatory cytokines, as compared, e.g., to other 5′cap analog structures known in the art. Cap structures include, but are not limited to, 7mG(5′)ppp(5′)N,pN2p (cap 0), 7mG(5′)ppp(5′)NlmpNp (cap 1), and 7mG(5′)-ppp(5′)NlmpN2mp (cap 2). An example of a cap structure is illustrated in FIG. 3.
  • For example, the RNA transcript can be enzymatically capped at the 5′ end using Vaccinia guanylyltransferase, guanosine triphosphate and s-adenosyl-L-methionine to yield cap 0 structure. An inverted 7-methylguanosine cap is added via a 5′ to 5′ triphosphate bridge. Alternatively, use of a 2′O-methyltransferase with Vaccinia guanylyltransferase yields the cap 1 structure where in addition to the cap 0 structure, the 2′OH group is methylated on the penultimate nucleotide. S-adenosyl-L-methionine (SAM) is a cofactor utilized as a methyl transfer reagent.
  • In one embodiment enzymatic 5′ capping is performed as follows. S-adenosylmethione chloride*2HCl is dissolved at 20 mM in 5 mM HCl 10/90 v/v % ethanol/water as a prepared stock. RNase inhibitor are utilized as a safeguard to ensure no RNase degradation is observed during the reaction. The final 1× buffer conditions consist of the following: 50 mM Tris HCl pH 8, 5 mM KCl, 1 mM MgCl2, and 1 mM dithiothreitol. The reaction is run under constant mixing at 37° C. for 2 hours. Enzymatic capping is of considerably higher efficiency than performing co-transcription through the use of dinucleotide cap analogs.
  • In another embodiment, the RNA transcript is co-transcriptionall capped. The 5′ terminal caps may include endogenous caps or cap analogs. A 5′ terminal cap may comprise a guanine analog. Useful guanine analogs include, but are not limited to, inosine, N1-methyl-guanosine, 2′fluoro-guanosine, 7-deaza-guanosine, 8-oxo-guanosine, 2-amino-guanosine, LNA-guanosine, and 2-azido-guanosine.
  • RNA Transcripts
  • An RNA transcript is the polynucleotide product of the in vitro transcription reaction. Typically the RNA transcript includes a gene of interest and a poly-A tail. In some embodiments, the RNA transcript includes a 5′UTR and a 3′UTR. In one embodiment the RNA transcript includes a 5′ cap, typically added post trasncriptionally.
  • FIG. 4 illustrates an exemplary RNA transcript. The RNA transcript 100 here contains a first region of linked nucleotides 102 that is flanked by a first flanking region 104 and a second flaking region 106. As used herein, the “first region” may be referred to as a “coding region” or “region encoding” or simply the “first region.” This first region may include, but is not limited to, the gene of interest. The polypeptide of interest may comprise at its 5′ terminus one or more signal sequences which are encoded by a signal sequence region 103. The flanking region 104 may comprise a region of linked nucleotides comprising one or more complete or incomplete 5′ UTRs sequences. The flanking region 104 may also comprise a 5′ terminal cap 108. The second flanking region 106 may comprise a region of linked nucleotides comprising one or more complete or incomplete 3′ UTRs. The flanking region 106 may also comprise a 3′ tailing sequence 110, e.g., a poly A tail sequence.
  • Bridging the 5′ terminus of the first region 102 and the first flanking region 104 is a first operational region 105. Traditionally this operational region comprises a Start codon. The operational region may alternatively comprise any translation initiation sequence or signal including a Start codon.
  • Bridging the 3′ terminus of the first region 102 and the second flanking region 106 is a second operational region 107. Traditionally this operational region comprises a Stop codon. The operational region may alternatively comprise any translation initiation sequence or signal including a Stop codon. According to the present invention, multiple serial stop codons may also be used.
  • Generally, the shortest length of the first region of the RNA transcript, e.g., the gene of interest can be the length of a nucleic acid sequence that is sufficient to encode for a dipeptide, a tripeptide, a tetrapeptide, a pentapeptide, a hexapeptide, a heptapeptide, an octapeptide, a nonapeptide, or a decapeptide. In another embodiment, the length may be sufficient to encode a peptide of 2-30 amino acids, e.g. 5-30, 10-30, 2-25, 5-25, 10-25, or 10-20 amino acids. The length may be sufficient to encode for a peptide of at least 11, 12, 13, 14, 15, 17, 20, 25 or 30 amino acids, or a peptide that is no longer than 40 amino acids, e.g. no longer than 35, 30, 25, 20, 17, 15, 14, 13, 12, 11 or 10 amino acids. Examples of dipeptides that the polynucleotide sequences can encode or include, but are not limited to, carnosine and anserine.
  • Generally, the length of the first region encoding the polypeptide of interest of the present invention is greater than about 30 nucleotides in length (e.g., at least or greater than about 35, 40, 45, 50, 55, 60, 70, 80, 90, 100, 120, 140, 160, 180, 200, 250, 300, 350, 400, 450, 500, 600, 700, 800, 900, 1,000, 1,100, 1,200, 1,300, 1,400, 1,500, 1,600, 1,700, 1,800, 1,900, 2,000, 2,500, and 3,000, 4,000, 5,000, 6,000, 7,000, 8,000, 9,000, 10,000, 20,000, 30,000, 40,000, 50,000, 60,000, 70,000, 80,000, 90,000 or up to and including 100,000 nucleotides).
  • In some embodiments, the RNA transcript includes from about 30 to about 100,000 nucleotides (e.g., from 30 to 50, from 30 to 100, from 30 to 250, from 30 to 500, from 30 to 1,000, from 30 to 1,500, from 30 to 3,000, from 30 to 5,000, from 30 to 7,000, from 30 to 10,000, from 30 to 25,000, from 30 to 50,000, from 30 to 70,000, from 100 to 250, from 100 to 500, from 100 to 1,000, from 100 to 1,500, from 100 to 3,000, from 100 to 5,000, from 100 to 7,000, from 100 to 10,000, from 100 to 25,000, from 100 to 50,000, from 100 to 70,000, from 100 to 100,000, from 500 to 1,000, from 500 to 1,500, from 500 to 2,000, from 500 to 3,000, from 500 to 5,000, from 500 to 7,000, from 500 to 10,000, from 500 to 25,000, from 500 to 50,000, from 500 to 70,000, from 500 to 100,000, from 1,000 to 1,500, from 1,000 to 2,000, from 1,000 to 3,000, from 1,000 to 5,000, from 1,000 to 7,000, from 1,000 to 10,000, from 1,000 to 25,000, from 1,000 to 50,000, from 1,000 to 70,000, from 1,000 to 100,000, from 1,500 to 3,000, from 1,500 to 5,000, from 1,500 to 7,000, from 1,500 to 10,000, from 1,500 to 25,000, from 1,500 to 50,000, from 1,500 to 70,000, from 1,500 to 100,000, from 2,000 to 3,000, from 2,000 to 5,000, from 2,000 to 7,000, from 2,000 to 10,000, from 2,000 to 25,000, from 2,000 to 50,000, from 2,000 to 70,000, and from 2,000 to 100,000).
  • In some embodiments, the first and second flanking regions may range independently from 15-1,000 nucleotides in length (e.g., greater than 30, 40, 45, 50, 55, 60, 70, 80, 90, 100, 120, 140, 160, 180, 200, 250, 300, 350, 400, 450, 500, 600, 700, 800, and 900 nucleotides or at least 30, 40, 45, 50, 55, 60, 70, 80, 90, 100, 120, 140, 160, 180, 200, 250, 300, 350, 400, 450, 500, 600, 700, 800, 900, and 1,000 nucleotides).
  • The tailing sequence may range from 1 to 500 nucleotides in length (e.g., at least 60, 70, 80, 90, 120, 140, 160, 180, 200, 250, 300, 350, 400, 450, or 500 nucleotides). Where the tailing region is a polyA tail, the length may be determined in units of or as a function of polyA Binding Protein binding. In this embodiment, the polyA tail is long enough to bind at least 4 monomers of PolyA Binding Protein. PolyA Binding Protein monomers bind to stretches of approximately 38 nucleotides. As such, it has been observed that polyA tails of about 80 nucleotides and 160 nucleotides are functional.
  • According to the present invention, the capping region may comprise a single cap or a series of nucleotides forming the cap. In this embodiment the capping region may be from 1 to 10, e.g. 2-9, 3-8, 4-7, 1-5, 5-10, or at least 2, or 10 or fewer nucleotides in length. In some embodiments, the cap is absent.
  • According to the present invention, the first and second operational regions may range from 3 to 40, e.g., 5-30, 10-20, 15, or at least 4, or 30 or fewer nucleotides in length and may comprise, in addition to a Start and/or Stop codon, one or more signal and/or restriction sequences.
  • The RNA transcripts can comprise one or more structural and/or chemical modifications or alterations which impart useful properties to the polynucleotide including, in some embodiments, the lack of a substantial induction of the innate immune response of a cell into which the polynucleotide is introduced. As used herein, a “structural” feature or modification is one in which two or more linked nucleotides are inserted, deleted, duplicated, inverted or randomized in an RNA transcript without significant chemical modification to the nucleotides themselves. Because chemical bonds will necessarily be broken and reformed to effect a structural modification, structural modifications are of a chemical nature and hence are chemical modifications. However, structural modifications will result in a different sequence of nucleotides. For example, the polynucleotide “ATCG” may be chemically modified to “AT-5meC-G”. The same polynucleotide may be structurally modified from “ATCG” to “ATCCCG”. Here, the dinucleotide “CC” has been inserted, resulting in a structural modification to the polynucleotide.
  • Removal of DNA Template from RNA Transcript
  • The linearized plasmid DNA template is removed from the in vitro transcription, e.g., the DNA template is separated from the RNA transcript. In one embodiment, the DNA template is removed chromatographically using an poly A capture, e.g., oligo dT, based affinity purification step. The RNA transcript binds affinity substrate while the DNA template flow through and is removed.
  • It is typical to utilize DNase I to enzymatically digest DNA template immediately following in vitro transcription. In the methods of the invention, DNase is not utilized. Whole plasmid removal is preferred to enzymatic digestion due to the fact that the risk of degraded DNA fragments hybridizing to the transcribed mRNA is mitigated.
  • In one embodiment the poly A capture based affinity purification is oligo dT purification. For example, a polythymidine ligand is immobilized to a derivatized chromatography resin. The mechanism of purification involves hybridization of the poly A tail of the RNA transcript to the oligonucleotide ligand. The DNA template will not bind. In addition, RNA transcripts that do not contain Poly A stretches (short aborts and other truncates formed during in vitro transcription) will not bind to the resin and will not form a duplex with the affinity ligand. Poly Adenylated RNA can then be eluted from the resin utilizing a low ionic strength buffer or a competitive binding oligonucleotide solution. A one pot purification method can yield highly purified poly A containing RNA with recoveries >80% actively removes endotoxin, DNA template, and enzymes utilized in the production of RNA using a simple capture and elute methodology with no subsequent fraction of captured poly A containing RNA. This purification increases mRNA product purity and in turn significantly increases target protein expression.
  • Additional Purification Steps
  • The method for production of an RNA transcript can include additional purification steps after the in vitro transcription, e.g., an ion exchange chromatography step.
  • Characterization and Analysis of RNA Transcript
  • The RNA transcript produced by the methods of the invention can be analyzed and characterized using any number of methods. Analysis can be performed before or after capping. Alternatively, analysis can be performed before or after poly A capture based affinity purification. In another embodiment, analysis can be performed before or after additional purification steps, e.g., anion exchange chromatography and the like.
  • For example, RNA template quality can be determined using Bioanalyzer chip based electrophoresis system. In other embodiments, RNA template purity is analysed using analytical reverse phase HPLC respectively. Capping efficiency can be analyzed using, e.g., total nuclease digestion followed by MS/MS quantitation of the dinucleotide cap species vs. uncapped GTP species. In vitro efficacy can be analyzed by, e.g., transfecting RNA transcript into a human cell line, e.g., HeLA, PBMC, BJ Fibroblasts, Hek 293). Protein expression of the polypeptide of interest can be quantified using methods such as ELISA or flow cytometry. Immunogenicity can be analyzed by, e.g., transfecting RNA transcripts into cell lines that indicate innate immune stimulation, e.g., PBMCs. Cytokine induction can be analyzed using, e.g., methods such as ELISA to quantify a cytokine, e.g., Interferon-α.
  • The method of producing RNA transcript of the invention can produce RNA transcript that is at least 30% full length transcript, or at least 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, or at least 95% full length transcript. Purity can be determined as described herein, e.g., via reverse phase HPLC or Bioanalyzer chip based electrophoresis and measure by, e.g., peak area of full length RNA transcript relative to total peak.
  • Genes of Interest
  • The DNA template and resulting RNA transcript of the present invention include a gene of interest. The gene of interest encodes a polypeptide of interest selected from, e.g., biologics, antibodies, vaccines, therapeutic proteins or peptides, cell penetrating peptides, secreted proteins, plasma membrane proteins, cytoplasmic or cytoskeletal proteins, intracellular membrane bound proteins, nuclear proteins, proteins associated with human disease, targeting moieties or those proteins encoded by the human genome for which no therapeutic indication has been identified but which nonetheless have utility in areas of research and discovery. The sequence for a particular gene of interest is readily identified by one of skill in the art using public and private databases, e.g., GenBank.
  • In one embodiment the gene of interest may encode variant polypeptides which have a certain identity with a reference polypeptide sequence. As used herein, a “reference polypeptide sequence” refers to a starting polypeptide sequence. Reference sequences may be wild type sequences or any sequence to which reference is made in the design of another 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 may have the same or a similar activity as the reference polypeptide. Alternatively, the variant may have an altered activity (e.g., increased or decreased) relative to a reference polypeptide. Generally, variants of a particular polynucleotide or polypeptide of the invention 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% but less than 100% 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. Such tools for alignment include those of the BLAST suite (Stephen F. Altschul, Thomas L. Madden, Alejandro A. Schäffer, Jinghui Zhang, Zheng Zhang, Webb Miller, and David J. Lipman (1997), “Gapped BLAST and PSI-BLAST: a new generation of protein database search programs”, Nucleic Acids Res. 25:3389-3402.) Other tools are described herein, specifically in the definition of “Identity.”
  • Default parameters in the BLAST algorithm include, for example, an expect threshold of 10, Word size of 28, Match/Mismatch Scores 1, −2, Gap costs Linear. Any filter can be applied as well as a selection for species specific repeats, e.g., Homo sapiens.
  • Biologics
  • The gene of interest can encode one or more biologics. As used herein, a “biologic” is a polypeptide-based molecule produced by the methods provided herein and which may be used to treat, cure, mitigate, prevent, or diagnose a serious or life-threatening disease or medical condition. Biologics, according to the present invention include, but are not limited to, allergenic extracts (e.g. for allergy shots and tests), blood components, gene therapy products, human tissue or cellular products used in transplantation, vaccines, monoclonal antibodies, cytokines, growth factors, enzymes, thrombolytics, and immunomodulators, among others.
  • According to the present invention, one or more biologics currently being marketed or in development may be encoded by the polynucleotides, primary constructs or mmRNA of the present invention. While not wishing to be bound by theory, it is believed that incorporation of the encoding polynucleotides of a known biologic into the primary constructs or mmRNA of the invention will result in improved therapeutic efficacy due at least in part to the specificity, purity and/or selectivity of the construct designs.
  • Antibodies
  • The gene of interest can encode one or more antibodies or fragments thereof. The term “antibody” includes monoclonal antibodies (including full length antibodies which have an immunoglobulin Fc region), antibody compositions with polyepitopic specificity, multispecific antibodies (e.g., bispecific antibodies, diabodies, and single-chain molecules), as well as antibody fragments. The term “immunoglobulin” (Ig) is used interchangeably with “antibody” herein. As used herein, the term “monoclonal antibody” refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible naturally occurring mutations and/or post-translation modifications (e.g., isomerizations, amidations) that may be present in minor amounts. Monoclonal antibodies are highly specific, being directed against a single antigenic site.
  • The monoclonal antibodies herein specifically include “chimeric” antibodies (immunoglobulins) in which a portion of the heavy and/or light chain is identical with or homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular antibody class or subclass, while the remainder of the chain(s) is(are) identical with or homologous to corresponding sequences in antibodies derived from another species or belonging to another antibody class or subclass, as well as fragments of such antibodies, so long as they exhibit the desired biological activity. Chimeric antibodies of interest herein include, but are not limited to, “primatized” antibodies comprising variable domain antigen-binding sequences derived from a non-human primate (e.g., Old World Monkey, Ape etc.) and human constant region sequences.
  • An “antibody fragment” comprises a portion of an intact antibody, preferably the antigen binding and/or the variable region of the intact antibody. Examples of antibody fragments include Fab, Fab′, F(ab′)2 and Fv fragments; diabodies; linear antibodies; nanobodies; single-chain antibody molecules and multispecific antibodies formed from antibody fragments.
  • Any of the five classes of immunoglobulins, IgA, IgD, IgE, IgG and IgM, may be encoded by the mmRNA of the invention, including the heavy chains designated alpha, delta, epsilon, gamma and mu, respectively. Also included are polynucleotide sequences encoding the subclasses, gamma and mu. Hence any of the subclasses of antibodies may be encoded in part or in whole and include the following subclasses: IgG1, IgG2, IgG3, IgG4, IgA1 and IgA2.
  • In one embodiment, the gene of interest can encode monoclonal antibodies and/or variants thereof. Variants of antibodies may also include, but are not limited to, substitutional variants, conservative amino acid substitution, insertional variants, deletional variants and/or covalent derivatives. In one embodiment, the primary construct and/or mmRNA disclosed herein may encode an immunoglobulin Fc region. In another embodiment, the primary constructs and/or mmRNA may encode a variant immunoglobulin Fc region. As a non-limiting example, the primary constructs and/or mmRNA may encode an antibody having a variant immunoglobulin Fc region as described in U.S. Pat. No. 8,217,147 herein incorporated by reference in its entirety.
  • Vaccines
  • The gene of interest can encode one or more vaccines. As used herein, a “vaccine” is a biological preparation that improves immunity to a particular disease or infectious agent. According to the present invention, one or more vaccines currently being marketed or in development may be encoded by the polynucleotides, primary constructs or mmRNA of the present invention. While not wishing to be bound by theory, it is believed that incorporation into the primary constructs or mmRNA of the invention will result in improved therapeutic efficacy due at least in part to the specificity, purity and selectivity of the construct designs.
  • Vaccines encoded in the polynucleotides, primary constructs or mmRNA of the invention may be utilized to treat conditions or diseases in many therapeutic areas such as, but not limited to, cardiovascular, CNS, dermatology, endocrinology, oncology, immunology, respiratory, and anti-infective.
  • Therapeutic Proteins or Peptides
  • The gene of interest can encode one or more validated or “in testing” therapeutic proteins or peptides. “Therapeutic protein” refers to a protein that, when administered to a cell has a therapeutic, diagnostic, and/or prophylactic effect and/or elicits a desired biological and/or pharmacological effect.
  • Therapeutic proteins and peptides may be utilized to treat conditions or diseases in many therapeutic areas such as, but not limited to, blood, cardiovascular, CNS, poisoning (including antivenoms), dermatology, endocrinology, genetic, genitourinary, gastrointestinal, musculoskeletal, oncology, and immunology, respiratory, sensory and anti-infective.
  • Cell-Penetrating Polypeptides
  • The gene of interest can encode one or more cell-penetrating polypeptides. As used herein, “cell-penetrating polypeptide” or CPP refers to a polypeptide which may facilitate the cellular uptake of molecules. A cell-penetrating polypeptide of the present invention may contain one or more detectable labels. The polypeptides may be partially labeled or completely labeled throughout. The gene of interest can encode the detectable label completely, partially or not at all. The cell-penetrating peptide may also include a signal sequence. As used herein, a “signal sequence” refers to a sequence of amino acid residues bound at the amino terminus of a nascent protein during protein translation. The signal sequence may be used to signal the secretion of the cell-penetrating polypeptide.
  • In one embodiment, the gene of interest can encode a fusion protein. The fusion protein may be created by operably linking a charged protein to a therapeutic protein. As used herein, “operably linked” refers to the therapeutic protein and the charged protein being connected in such a way to permit the expression of the complex when introduced into the cell. As used herein, “charged protein” refers to a protein that carries a positive, negative or overall neutral electrical charge. Preferably, the therapeutic protein may be covalently linked to the charged protein in the formation of the fusion protein. The ratio of surface charge to total or surface amino acids may be approximately 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8 or 0.9.
  • The cell-penetrating polypeptide may form a complex after being translated. The complex may comprise a charged protein linked, e.g. covalently linked, to the cell-penetrating polypeptide.
  • In one embodiment, the cell-penetrating polypeptide may comprise a first domain and a second domain. The first domain may comprise a supercharged polypeptide. The second domain may comprise a protein-binding partner. As used herein, “protein-binding partner” includes, but is not limited to, antibodies and functional fragments thereof, scaffold proteins, or peptides. The cell-penetrating polypeptide may further comprise an intracellular binding partner for the protein-binding partner. The cell-penetrating polypeptide may be capable of being secreted from a cell where the polynucleotide, primary construct or mmRNA may be introduced. The cell-penetrating polypeptide may also be capable of penetrating the first cell.
  • In a further embodiment, the cell-penetrating polypeptide is capable of penetrating a second cell. The second cell may be from the same area as the first cell, or it may be from a different area. The area may include, but is not limited to, tissues and organs. The second cell may also be proximal or distal to the first cell.
  • In one embodiment, the cell-penetrating polypeptide which may comprise a protein-binding partner. The protein binding partner may include, but is not limited to, an antibody, a supercharged antibody or a functional fragment. The polynucleotides, primary constructs or mmRNA may be introduced into the cell where a cell-penetrating polypeptide comprising the protein-binding partner is introduced.
  • Additional Proteins
  • The gene of interest can encode a secreted protein. The secreted proteins may be selected from, e.g., those in US Patent Publication, 20100255574, the contents of which are incorporated herein by reference in their entirety.
  • Alternatively, the gene of interest can encode a protein of the plasma membrane; a cytoplasmic or cytoskeletal proteins; a intracellular membrane bound proteins; or a nuclear protein; a targeting moiety (e.g., 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); antimicrobial peptides (AMP) or antiviral peptides (AVP).
  • Flanking Regions: Untranslated Regions (UTRs)
  • The DNA template and RNA transcript can include UTRs. Untranslated regions (UTRs) of a gene are transcribed but not translated. The 5′UTR starts at the transcription start site and continues to the start codon but does not include the start codon; whereas, the 3′UTR starts immediately following the stop codon and continues until the transcriptional termination signal. There is growing body of evidence about the regulatory roles played by the UTRs in terms of stability of the nucleic acid molecule and translation. The regulatory features of a UTR can be incorporated into the polynucleotides, primary constructs and/or mmRNA of the present invention to enhance the stability of the molecule. The specific features can also be incorporated to ensure controlled down-regulation of the transcript in case they are misdirected to undesired organs sites.
  • 5′ UTR and Translation Initiation
  • Natural 5′UTRs bear features which play roles in translation initiation. They harbor signatures like Kozak sequences which are commonly known to be involved in the process by which the ribosome initiates translation of many genes. Kozak sequences have the consensus CCR(A/G)CCAUGG, where R is a purine (adenine or guanine) three bases upstream of the start codon (AUG), which is followed by another ‘G’. 5′UTR also have been known to form secondary structures which are involved in elongation factor binding.
  • By engineering the features typically found in abundantly expressed genes of specific target organs, one can enhance the stability and protein production of the polynucleotides, primary constructs or mmRNA of the invention. For example, introduction of 5′ UTR of liver-expressed mRNA, such as albumin, serum amyloid A, Apolipoprotein AB/E, transferrin, alpha fetoprotein, erythropoietin, or Factor VIII, could be used to enhance expression of a nucleic acid molecule, such as a mmRNA, in hepatic cell lines or liver. Likewise, use of 5′ UTR from other tissue-specific mRNA to improve expression in that tissue is possible for muscle (MyoD, Myosin, Myoglobin, Myogenin, Herculin), for endothelial cells (Tie-1, CD36), for myeloid cells (C/EBP, AML1, G-CSF, GM-CSF, CDIIb, MSR, Fr-1, i-NOS), for leukocytes (CD45, CD18), for adipose tissue (CD36, GLUT4, ACRP30, adiponectin) and for lung epithelial cells (SP-A/B/C/D).
  • Other non-UTR sequences may be incorporated into the 5′ (or 3′ UTR) UTRs. For example, introns or portions of introns sequences may be incorporated into the flanking regions of the polynucleotides, primary constructs or mmRNA of the invention. Incorporation of intronic sequences may increase protein production as well as mRNA levels.
  • 3′ UTR and the AU Rich Elements
  • 3′ UTRs are known to have stretches of Adenosines and Uridines embedded in them. These AU rich signatures are particularly prevalent in genes with high rates of turnover. Based on their sequence features and functional properties, the AU rich elements (AREs) can be separated into three classes (Chen et al, 1995): Class I AREs contain several dispersed copies of an AUUUA motif within U-rich regions. C-Myc and MyoD contain class I AREs. Class II AREs possess two or more overlapping UUAUUUA(U/A)(U/A) nonamers. Molecules containing this type of AREs include GM-CSF and TNF-a. Class III ARES are less well defined. These U rich regions do not contain an AUUUA motif. c-Jun and Myogenin are two well-studied examples of this class. Most proteins binding to the AREs are known to destabilize the messenger, whereas members of the ELAV family, most notably HuR, have been documented to increase the stability of mRNA. HuR binds to AREs of all the three classes. Engineering the HuR specific binding sites into the 3′ UTR of nucleic acid molecules will lead to HuR binding and thus, stabilization of the message in vivo.
  • Introduction, removal or modification of 3′ UTR AU rich elements (AREs) can be used to modulate the stability of polynucleotides, primary constructs or mmRNA of the invention. When engineering specific polynucleotides, primary constructs or mmRNA, one or more copies of an ARE can be introduced to make polynucleotides, primary constructs or mmRNA of the invention less stable and thereby curtail translation and decrease production of the resultant protein. Likewise, AREs can be identified and removed or mutated to increase the intracellular stability and thus increase translation and production of the resultant protein. Transfection experiments can be conducted in relevant cell lines, using polynucleotides, primary constructs or mmRNA of the invention and protein production can be assayed at various time points post-transfection. For example, cells can be transfected with different ARE-engineering molecules and by using an ELISA kit to the relevant protein and assaying protein produced at 6 hour, 12 hour, 24 hour, 48 hour, and 7 days post-transfection.
  • Incorporating microRNA Binding Sites
  • microRNAs (or miRNA) are 19-25 nucleotide long noncoding RNAs that bind to the 3′UTR of nucleic acid molecules and down-regulate gene expression either by reducing nucleic acid molecule stability or by inhibiting translation. The polynucleotides, primary constructs or mmRNA of the invention may comprise one or more microRNA target sequences, microRNA sequences, or microRNA seeds. Such sequences may correspond to any known microRNA such as those taught in US Publication US2005/0261218 and US Publication US2005/0059005, the contents of which are incorporated herein by reference in their entirety.
  • A microRNA sequence comprises a “seed” region, i.e., a sequence in the region of positions 2-8 of the mature microRNA, which sequence has perfect Watson-Crick complementarity to the miRNA target sequence. A microRNA seed may comprise positions 2-8 or 2-7 of the mature microRNA. In some embodiments, a microRNA seed may comprise 7 nucleotides (e.g., nucleotides 2-8 of the mature microRNA), wherein the seed-complementary site in the corresponding miRNA target is flanked by an adenine (A) opposed to microRNA position 1. In some embodiments, a microRNA seed may comprise 6 nucleotides (e.g., nucleotides 2-7 of the mature microRNA), wherein the seed-complementary site in the corresponding miRNA target is flanked byan adenine (A) opposed to microRNA position 1. See for example, Grimson A, Farh K K, Johnston W K, Garrett-Engele P, Lim L P, Bartel D P; Mol Cell. 2007 Jul. 6; 27(1):91-105; each of which is herein incorporated by reference in their entirety. The bases of the microRNA seed have complete complementarity with the target sequence. By engineering microRNA target sequences into the 3′UTR of RNA transcripts of the invention one can target the molecule for degradation or reduced translation, provided the microRNA in question is available. This process will reduce the hazard of off target effects upon nucleic acid molecule delivery. Identification of microRNA, microRNA target regions, and their expression patterns and role in biology have been reported (Bonauer et al., Curr Drug Targets 2010 11:943-949; Anand and Cheresh Curr Opin Hematol 2011 18:171-176; Contreras and Rao Leukemia 2012 26:404-413 (2011 Dec. 20. doi: 10.1038/1eu.2011.356); Bartel Cell 2009 136:215-233; Landgraf et al, Cell, 2007 129:1401-1414; each of which is herein incorporated by reference in its entirety).
  • For example, if the nucleic acid molecule is an mRNA and is not intended to be delivered to the liver but ends up there, then miR-122, a microRNA abundant in liver, can inhibit the expression of the gene of interest if one or multiple target sites of miR-122 are engineered into the 3′ UTR of the RNA transcripts. Introduction of one or multiple binding sites for different microRNA can be engineered to further decrease the longevity, stability, and protein translation of a RNA transcripts.
  • As used herein, the term “microRNA site” refers to a microRNA target site or a microRNA recognition site, or any nucleotide sequence to which a microRNA binds or associates. It should be understood that “binding” may follow traditional Watson-Crick hybridization rules or may reflect any stable association of the microRNA with the target sequence at or adjacent to the microRNA site.
  • Conversely, for the purposes of the RNA transcripts of the present invention, microRNA binding sites can be engineered out of (i.e. removed from) sequences in which they naturally occur in order to increase protein expression in specific tissues. For example, miR-122 binding sites may be removed to improve protein expression in the liver. Regulation of expression in multiple tissues can be accomplished through introduction or removal or one or several microRNA binding sites.
  • Examples of tissues where microRNA are known to regulate mRNA, and thereby protein expression, include, but are not limited to, liver (miR-122), muscle (miR-133, miR-206, miR-208), endothelial cells (miR-17-92, miR-126), myeloid cells (miR-142-3p, miR-142-5p, miR-16, miR-21, miR-223, miR-24, miR-27), adipose tissue (let-7, miR-30c), heart (miR-1d, miR-149), kidney (miR-192, miR-194, miR-204), and lung epithelial cells (let-7, miR-133, miR-126). MicroRNA can also regulate complex biological processes such as angiogenesis (miR-132) (Anand and Cheresh Curr Opin Hematol 2011 18:171-176; herein incorporated by reference in its entirety). In the RNA transcripts of the present invention, binding sites for microRNAs that are involved in such processes may be removed or introduced, in order to tailor the expression of the RNA transcripts expression to biologically relevant cell types or to the context of relevant biological processes. A listing of MicroRNA, miR sequences and miR binding sites is listed in Table 9 of U.S. Provisional Application No. 61/753,661 filed Jan. 17, 2013, in Table 9 of U.S. Provisional Application No. 61/754,159 filed Jan. 18, 2013, and in Table 7 of U.S. Provisional Application No. 61/758,921 filed Jan. 31, 2013, each of which are herein incorporated by reference in their entireties.g
  • Lastly, through an understanding of the expression patterns of microRNA in different cell types, RNA transcripts can be engineered for more targeted expression in specific cell types or only under specific biological conditions. Through introduction of tissue-specific microRNA binding sites, RNA transcripts could be designed that would be optimal for protein expression in a tissue or in the context of a biological condition.
  • Transfection experiments can be conducted in relevant cell lines, using engineered RNA transcripts and protein production can be assayed at various time points post-transfection. For example, cells can be transfected with different microRNA binding site-engineering RNA transcripts and by using an ELISA kit to the relevant protein and assaying protein produced at 6 hour, 12 hour, 24 hour, 48 hour, 72 hour and 7 days post-transfection. In vivo experiments can also be conducted using microRNA-binding site-engineered molecules to examine changes in tissue-specific expression of formulated RNA transcripts.
  • Viral Sequences
  • Additional viral sequences such as, but not limited to, the translation enhancer sequence of the barley yellow dwarf virus (BYDV-PAV), the Jaagsiekte sheep retrovirus (JSRV) and/or the Enzootic nasal tumor virus (See e.g., International Pub. No. WO2012129648; herein incorporated by reference in its entirety) can be engineered and inserted in the 3′ UTR of the RNA transcripts of the invention and can stimulate the translation of the construct in vitro and in vivo. Transfection experiments can be conducted in relevant cell lines at and protein production can be assayed by ELISA at 12 hr, 24 hr, 48 hr, 72 hr and day 7 post-transfection.
  • IRES Sequences
  • Further, provided are RNA transcripts which may contain an internal ribosome entry site (IRES). First identified as a feature Picorna virus RNA, IRES plays an important role in initiating protein synthesis in absence of the 5′ cap structure. An IRES may act as the sole ribosome binding site, or may serve as one of multiple ribosome binding sites of an mRNA. RNA transcripts containing more than one functional ribosome binding site may encode several peptides or polypeptides that are translated independently by the ribosomes (“multicistronic nucleic acid molecules”). When RNA transcripts are provided with an IRES, further optionally provided is a second translatable region. Examples of IRES sequences that can be used according to the invention include without limitation, those from picornaviruses (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).
  • Poly-A Tails
  • During RNA processing, a long chain of adenine nucleotides (poly-A tail) may be added to a polynucleotide such as an mRNA molecules in order to increase stability. Immediately after transcription, the 3′ end of the transcript may be cleaved to free a 3′ hydroxyl. Then poly-A polymerase adds a chain of adenine nucleotides to the RNA. The process, called polyadenylation, adds a poly-A tail that can be between, for example, approximately 100 and 250 residues long.
  • It has been discovered that unique poly-A tail lengths provide certain advantages to the RNA transcripts of the present invention.
  • In one embodiment the poly A tail is 5-300 nucleotides in length. In another embodiment, the poly A tail is 60-160 nucleotides in length.
  • In another embodiment, the poly-A tail is greater than 35 nucleotides in length (e.g., at least or greater than about 35, 40, 45, 50, 55, 60, 70, 80, 90, 100, 120, 140, 160, 180, 200, 250, 300, 350, 400, 450, 500, 600, 700, 800, 900, 1,000, 1,100, 1,200, 1,300, 1,400, 1,500, 1,600, 1,700, 1,800, 1,900, 2,000, 2,500, and 3,000 nucleotides). In some embodiments, the polynucleotide, primary construct, or mmRNA includes from about 30 to about 3,000 nucleotides (e.g., from 30 to 50, from 30 to 100, from 30 to 250, from 30 to 500, from 30 to 750, from 30 to 1,000, from 30 to 1,500, from 30 to 2,000, from 30 to 2,500, from 50 to 100, from 50 to 250, from 50 to 500, from 50 to 750, from 50 to 1,000, from 50 to 1,500, from 50 to 2,000, from 50 to 2,500, from 50 to 3,000, from 100 to 500, from 100 to 750, from 100 to 1,000, from 100 to 1,500, from 100 to 2,000, from 100 to 2,500, from 100 to 3,000, from 500 to 750, from 500 to 1,000, from 500 to 1,500, from 500 to 2,000, from 500 to 2,500, from 500 to 3,000, from 1,000 to 1,500, from 1,000 to 2,000, from 1,000 to 2,500, from 1,000 to 3,000, from 1,500 to 2,000, from 1,500 to 2,500, from 1,500 to 3,000, from 2,000 to 3,000, from 2,000 to 2,500, and from 2,500 to 3,000).
  • In one embodiment, the poly-A tail is designed relative to the length of the overall RNA transcripts. This design may be based on the length of the coding region, the length of a particular feature or region (such as the first or flanking regions), or based on the length of the ultimate product expressed from the RNA transcripts.
  • In this context the poly-A tail may be 10, 20, 30, 40, 50, 60, 70, 80, 90, or 100% greater in length than the RNA transcripts or feature thereof. The poly-A tail may also be designed as a fraction of RNA transcripts to which it belongs. In this context, the poly-A tail may be 10, 20, 30, 40, 50, 60, 70, 80, or 90% or more of the total length of the construct or the total length of the construct minus the poly-A tail. Further, engineered binding sites and conjugation of RNA transcripts for Poly-A binding protein may enhance expression.
  • Additionally, multiple distinct RNA transcripts may be linked together to the PABP (Poly-A binding protein) through the 3′-end using modified nucleotides at the 3′-terminus of the poly-A tail. Transfection experiments can be conducted in relevant cell lines at and protein production can be assayed by ELISA at 12 hr, 24 hr, 48 hr, 72 hr and day 7 post-transfection.
  • In one embodiment, the polynucleotide primary constructs of the present invention are designed to include a polyA-G Quartet. The G-quartet is a cyclic hydrogen bonded array of four guanine nucleotides that can be formed by G-rich sequences in both DNA and RNA. In this embodiment, the G-quartet is incorporated at the end of the poly-A tail. The resultant mmRNA construct is assayed for stability, protein production and other parameters including half-life at various time points. It has been discovered that the polyA-G quartet results in protein production equivalent to at least 75% of that seen using a poly-A tail of 120 nucleotides alone.
  • Modified Nucleotides
  • Herein, in a polynucleotide (such as a primary construct or an mRNA molecule), the terms “modification” or, as appropriate, “modified” refer to modification with respect to A, G, U or C ribonucleotides. Generally, herein, these terms are not intended to refer to the ribonucleotide modifications in naturally occurring 5′-terminal mRNA cap moieties. In a polypeptide, the term “modification” refers to a modification as compared to the canonical set of 20 amino acids, moiety)
  • The modifications may be various distinct modifications. In some embodiments, the coding region, the flanking regions and/or the terminal regions may contain one, two, or more (optionally different) nucleoside or nucleotide modifications. In some embodiments, a modified polynucleotide, primary construct, or mmRNA introduced to a cell may exhibit reduced degradation in the cell, as compared to an unmodified polynucleotide, primary construct, or mmRNA.
  • The polynucleotides, primary constructs, and mmRNA can include any useful modification, such as to the sugar, the nucleobase, or the internucleoside linkage (e.g. to a linking phosphate/to a phosphodiester linkage/to the phosphodiester backbone). One or more atoms of a pyrimidine nucleobase may be replaced or substituted with optionally substituted amino, optionally substituted thiol, optionally substituted alkyl (e.g., methyl or ethyl), or halo (e.g., chloro or fluoro). In certain embodiments, modifications (e.g., one or more modifications) are present in each of the sugar and the internucleoside linkage. Modifications according to the present invention may be modifications of ribonucleic acids (RNAs) to deoxyribonucleic acids (DNAs), threose nucleic acids (TNAs), glycol nucleic acids (GNAs), peptide nucleic acids (PNAs), locked nucleic acids (LNAs) or hybrids thereof). Additional modifications are described herein.
  • As described herein, the polynucleotides, primary constructs, and mmRNA of the invention do not substantially induce an innate immune response of a cell into which the mRNA is introduced. Features of an induced innate immune response include 1) increased expression of pro-inflammatory cytokines, 2) activation of intracellular PRRs (RIG-I, MDAS, etc, and/or 3) termination or reduction in protein translation.
  • In certain embodiments, it may desirable to intracellularly degrade a modified nucleic acid molecule introduced into the cell. For example, degradation of a modified nucleic acid molecule may be preferable if precise timing of protein production is desired. Thus, in some embodiments, the invention provides a modified nucleic acid molecule containing a degradation domain, which is capable of being acted on in a directed manner within a cell. In another aspect, the present disclosure provides polynucleotides comprising a nucleoside or nucleotide that can disrupt the binding of a major groove interacting, e.g. binding, partner with the polynucleotide (e.g., where the modified nucleotide has decreased binding affinity to major groove interacting partner, as compared to an unmodified nucleotide).
  • The polynucleotides, primary constructs, and mmRNA can optionally include other agents (e.g., RNAi-inducing agents, RNAi agents, siRNAs, shRNAs, miRNAs, antisense RNAs, ribozymes, catalytic DNA, tRNA, RNAs that induce triple helix formation, aptamers, vectors, etc.). In some embodiments, the polynucleotides, primary constructs, or mmRNA may include one or more messenger RNAs (mRNAs) and one or more modified nucleoside or nucleotides (e.g., mmRNA molecules). Details for these polynucleotides, primary constructs, and mmRNA follow.
  • Polynucleotides and Primary Constructs
  • The polynucleotides, primary constructs, and mmRNA of the invention includes a first region of linked nucleosides encoding a polypeptide of interest, a first flanking region located at the 5′ terminus of the first region, and a second flanking region located at the 3′ terminus of the first region.
  • In some embodiments, the polynucleotide, primary construct, or mmRNA (e.g., the first region, first flanking region, or second flanking region) includes n number of linked nucleosides having Formula (Ia) or Formula (Ia-1):
  • Figure US20190085368A1-20190321-C00001
  • or a pharmaceutically acceptable salt or stereoisomer thereof,
  • wherein
  • U is O, S, N(RU)nu, or C(RU)nu, wherein nu is an integer from 0 to 2 and each RU is, independently, H, halo, or optionally substituted alkyl;
  • --- is a single bond or absent;
  • each of R1′, R2′, R1″, R2″, R1, R2, R3, R4, and R5 is, independently, if present, H, halo, hydroxy, thiol, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, optionally substituted hydroxyalkoxy, optionally substituted amino, azido, optionally substituted aryl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, or absent; wherein the combination of R3 with one or more of R1′, R1″, R2′, R2″, or R5 (e.g., the combination of R1′ and R3, the combination of R1″ and R3, the combination of R2′ and R3, the combination of R2″ and R3, or the combination of R5 and R3) can join together to form optionally substituted alkylene or optionally substituted heteroalkylene and, taken together with the carbons to which they are attached, provide an optionally substituted heterocyclyl (e.g., a bicyclic, tricyclic, or tetracyclic heterocyclyl); wherein the combination of R5 with one or more of R1′, R1″, R2′, or R2″ (e.g., the combination of R1′ and R5, the combination of R1″ and R5, the combination of R2′ and R5, or the combination of R2″ and R5) can join together to form optionally substituted alkylene or optionally substituted heteroalkylene and, taken together with the carbons to which they are attached, provide an optionally substituted heterocyclyl (e.g., a bicyclic, tricyclic, or tetracyclic heterocyclyl); and wherein the combination of R4 and one or more of R1′, R1″, R2′, R2″, R3, or R5 can join together to form optionally substituted alkylene or optionally substituted heteroalkylene and, taken together with the carbons to which they are attached, provide an optionally substituted heterocyclyl (e.g., a bicyclic, tricyclic, or tetracyclic heterocyclyl); each of m′ and m″ is, independently, an integer from 0 to 3 (e.g., from 0 to 2, from 0 to 1, from 1 to 3, or from 1 to 2);
  • each of Y1, Y2, and Y3, is, independently, O, S, Se, —NRN1-, optionally substituted alkylene, or optionally substituted heteroalkylene, wherein RN1 is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, or absent;
  • each Y4 is, independently, H, hydroxy, thiol, boranyl, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted thioalkoxy, optionally substituted alkoxyalkoxy, or optionally substituted amino;
  • each Y5 is, independently, O, S, Se, optionally substituted alkylene (e.g., methylene), or optionally substituted heteroalkylene;
  • n is an integer from 1 to 100,000; and
  • B is a nucleobase (e.g., a purine, a pyrimidine, or derivatives thereof), wherein the combination of B and R1′, the combination of B and R2′, the combination of B and R1″, or the combination of B and R2″ can, taken together with the carbons to which they are attached, optionally form a bicyclic group (e.g., a bicyclic heterocyclyl) or wherein the combination of B, R1″, and R3 or the combination of B, R2″, and R3 can optionally form a tricyclic or tetracyclic group (e.g., a tricyclic or tetracyclic heterocyclyl, such as in Formula (IIo)-(IIp) herein). In some embodiments, the polynucleotide, primary construct, or mmRNA includes a modified ribose. In some embodiments, the polynucleotide, primary construct, or mmRNA (e.g., the first region, the first flanking region, or the second flanking region) includes n number of linked nucleosides having Formula (Ia-2)-(Ia-5) or a pharmaceutically acceptable salt or stereoisomer thereof.
  • Figure US20190085368A1-20190321-C00002
  • In some embodiments, the polynucleotide, primary construct, or mmRNA (e.g., the first region, the first flanking region, or the second flanking region) includes n number of linked nucleosides having Formula (Ib) or Formula (Ib-1):
  • Figure US20190085368A1-20190321-C00003
  • or a pharmaceutically acceptable salt or stereoisomer thereof,
  • wherein
  • U is O, S, N(RU)nu, or C(RU)nu, wherein nu is an integer from 0 to 2 and each RU is, independently, H, halo, or optionally substituted alkyl;
  • --- is a single bond or absent;
  • each of R1, R3′, R3″, and R4 is, independently, H, halo, hydroxy, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, optionally substituted hydroxyalkoxy, optionally substituted amino, azido, optionally substituted aryl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, or absent; and wherein the combination of R1 and R3′ or the combination of R1 and R3″ can be taken together to form optionally substituted alkylene or optionally substituted heteroalkylene (e.g., to produce a locked nucleic acid);
  • each R5 is, independently, H, halo, hydroxy, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, or absent;
  • each of Y1, Y2, and Y3 is, independently, O, S, Se, —NRN1-, optionally substituted alkylene, or optionally substituted heteroalkylene, wherein RN1 is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, or optionally substituted aryl;
  • each Y4 is, independently, H, hydroxy, thiol, boranyl, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted alkoxyalkoxy, or optionally substituted amino;
  • n is an integer from 1 to 100,000; and
  • B is a nucleobase.
  • In some embodiments, the polynucleotide, primary construct, or mmRNA (e.g., the first region, first flanking region, or second flanking region) includes n number of linked nucleosides having Formula (Ic):
  • Figure US20190085368A1-20190321-C00004
  • or a pharmaceutically acceptable salt or stereoisomer thereof,
  • wherein
  • U is O, S, N(RU)nu, or C(RU)nu, wherein nu is an integer from 0 to 2 and each RU is, independently, H, halo, or optionally substituted alkyl;
  • --- is a single bond or absent;
  • each of B1, B2, and B3 is, independently, a nucleobase (e.g., a purine, a pyrimidine, or derivatives thereof, as described herein), H, halo, hydroxy, thiol, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, optionally substituted hydroxyalkoxy, optionally substituted amino, azido, optionally substituted aryl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, or optionally substituted aminoalkynyl, wherein one and only one of B1, B2, and B3 is a nucleobase;
  • each of Rb1, Rb2, Rb3, R3, and R5 is, independently, H, halo, hydroxy, thiol, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, optionally substituted hydroxyalkoxy, optionally substituted amino, azido, optionally substituted aryl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl or optionally substituted aminoalkynyl;
  • each of Y1, Y2, and Y3, is, independently, O, S, Se, —NRN1-, optionally substituted alkylene, or optionally substituted heteroalkylene, wherein RN1 is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, or optionally substituted aryl;
  • each Y4 is, independently, H, hydroxy, thiol, boranyl, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted thioalkoxy, optionally substituted alkoxyalkoxy, or optionally substituted amino;
  • each Y5 is, independently, O, S, Se, optionally substituted alkylene (e.g., methylene), or optionally substituted heteroalkylene;
  • n is an integer from 1 to 100,000; and
  • wherein the ring including U can include one or more double bonds.
  • In particular embodiments, the ring including U does not have a double bond between U-CB3Rb3 or between CB3Rb3-CB2Rb2.
  • In some embodiments, the polynucleotide, primary construct, or mmRNA (e.g., the first region, first flanking region, or second flanking region) includes n number of linked nucleosides having Formula (Id):
  • Figure US20190085368A1-20190321-C00005
  • or a pharmaceutically acceptable salt or stereoisomer thereof,
  • wherein
  • U is O, S, N(RU)nu, or C(RU)nu, wherein nu is an integer from 0 to 2 and each RU is, independently, H, halo, or optionally substituted alkyl;
  • each R3 is, independently, H, halo, hydroxy, thiol, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, optionally substituted hydroxyalkoxy, optionally substituted amino, azido, optionally substituted aryl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, or optionally substituted aminoalkynyl;
  • each of Y1, Y2, and Y3, is, independently, O, S, Se, —NRN1-, optionally substituted alkylene, or optionally substituted heteroalkylene, wherein RN1 is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, or optionally substituted aryl;
  • each Y4 is, independently, H, hydroxy, thiol, boranyl, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted thioalkoxy, optionally substituted alkoxyalkoxy, or optionally substituted amino;
  • each Y5 is, independently, O, S, optionally substituted alkylene (e.g., methylene), or optionally substituted heteroalkylene;
  • n is an integer from 1 to 100,000; and
  • B is a nucleobase (e.g., a purine, a pyrimidine, or derivatives thereof).
  • In some embodiments, the polynucleotide, primary construct, or mmRNA (e.g., the first region, first flanking region, or second flanking region) includes n number of linked nucleosides having Formula (Ie):
  • Figure US20190085368A1-20190321-C00006
  • or a pharmaceutically acceptable salt or stereoisomer thereof,
  • wherein
  • each of U′ and U″ is, independently, O, S, N(RU)nu, or C(RU)nu, wherein nu is an integer from 0 to 2 and each RU is, independently, H, halo, or optionally substituted alkyl;
  • each R6 is, independently, H, halo, hydroxy, thiol, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, optionally substituted hydroxyalkoxy, optionally substituted amino, azido, optionally substituted aryl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, or optionally substituted aminoalkynyl;
  • each Y5′ is, independently, O, S, optionally substituted alkylene (e.g., methylene or ethylene), or optionally substituted heteroalkylene;
  • n is an integer from 1 to 100,000; and
  • B is a nucleobase (e.g., a purine, a pyrimidine, or derivatives thereof).
  • In some embodiments, the polynucleotide, primary construct, or mmRNA (e.g., the first region, first flanking region, or second flanking region) includes n number of linked nucleosides having Formula (If) or (If-1):
  • Figure US20190085368A1-20190321-C00007
  • or a pharmaceutically acceptable salt or stereoisomer thereof,
  • wherein
  • each of U′ and U″ is, independently, O, S, N, N(RU)nu, or C(RU)nu, wherein nu is an integer from 0 to 2 and each RU is, independently, H, halo, or optionally substituted alkyl (e.g., U′ is 0 and U″ is N);
  • --- is a single bond or absent;
  • each of R1′, R2′, R1″, R2″, R3, and R4 is, independently, H, halo, hydroxy, thiol, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, optionally substituted hydroxyalkoxy, optionally substituted amino, azido, optionally substituted aryl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, or absent; and wherein the combination of R1′ and R3, the combination of R1″ and R3, the combination of R2′ and R3, or the combination of R2″ and R3 can be taken together to form optionally substituted alkylene or optionally substituted heteroalkylene (e.g., to produce a locked nucleic acid); each of m′ and m″ is, independently, an integer from 0 to 3 (e.g., from 0 to 2, from 0 to 1, from 1 to 3, or from 1 to 2);
  • each of Y1, Y2, and Y3, is, independently, O, S, Se, —NRN1-, optionally substituted alkylene, or optionally substituted heteroalkylene, wherein RN1 is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, or absent;
  • each Y4 is, independently, H, hydroxy, thiol, boranyl, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted thioalkoxy, optionally substituted alkoxyalkoxy, or optionally substituted amino;
  • each Y5 is, independently, O, S, Se, optionally substituted alkylene (e.g., methylene), or optionally substituted heteroalkylene;
  • n is an integer from 1 to 100,000; and
  • B is a nucleobase (e.g., a purine, a pyrimidine, or derivatives thereof).
  • In some embodiments of the polynucleotides, primary constructs, or mmRNA (e.g., Formulas (Ia), (Ia-1)-(Ia-3), (Ib)-(If), and (IIa)-(IIp)), the ring including U has one or two double bonds.
  • In some embodiments of the polynucleotides, primary constructs, or mmRNA (e.g., Formulas (Ia)-(Ia-5), (Ib)-(If-1), (IIa)-(IIp), (IIb-1), (IIb-2), (IIc-1)-(IIc-2), (IIn-1), (IIn-2), (IVa)-(IVl), and (IXa)-(IXr)), each of R1, R1′, and R1″, if present, is H. In further embodiments, each of R2, R2′, and R2″, if present, is, independently, H, halo (e.g., fluoro), hydroxy, optionally substituted alkoxy (e.g., methoxy or ethoxy), or optionally substituted alkoxyalkoxy. In particular embodiments, alkoxyalkoxy is —(CH2)s2(OCH2CH2)s1(CH2)s3OR′, wherein s1 is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R′ is H or C1-20 alkyl). In some embodiments, s2 is 0, s1 is 1 or 2, s3 is 0 or 1, and R′ is C1-6 alkyl.
  • In some embodiments of the polynucleotides, primary constructs, or mmRNA (e.g., Formulas (Ia)-(Ia-5), (Ib)-(If-1), (IIa)-(IIp), (IIb-1), (IIb-2), (IIc-1)-(IIc-2), (IIn-1), (IIn-2), (IVa)-(IVl), and (IXa)-(IXr)), each of R2, R2′, and R2″, if present, is H. In further embodiments, each of R1, R1′, and R1″, if present, is, independently, H, halo (e.g., fluoro), hydroxy, optionally substituted alkoxy (e.g., methoxy or ethoxy), or optionally substituted alkoxyalkoxy. In particular embodiments, alkoxyalkoxy is —(CH2)s2(OCH2CH2)s1(CH2)s3OR′, wherein s1 is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R′ is H or C1-20 alkyl). In some embodiments, s2 is 0, s1 is 1 or 2, s3 is 0 or 1, and R′ is C1-6 alkyl.
  • In some embodiments of the polynucleotides, primary constructs, or mmRNA (e.g., Formulas (Ia)-(Ia-5), (Ib)-(If-1), (IIa)-(IIp), (IIb-1), (IIb-2), (IIc-1)-(IIc-2), (IIn-1), (IIn-2), (IVa)-(IVl), and (IXa)-(IXr)), each of R3, R4, and R5 is, independently, H, halo (e.g., fluoro), hydroxy, optionally substituted alkyl, optionally substituted alkoxy (e.g., methoxy or ethoxy), or optionally substituted alkoxyalkoxy. In particular embodiments, R3 is H, R4 is H, R5 is H, or R3, R4, and R5 are all H. In particular embodiments, R3 is C1-6 alkyl, R4 is C1-6 alkyl, R5 is C1-6 alkyl, or R3, R4, and R5 are all C1-6 alkyl. In particular embodiments, R3 and R4 are both H, and R5 is C1-6 alkyl.
  • In some embodiments of the polynucleotides, primary constructs, or mmRNA (e.g., Formulas (Ia)-(Ia-5), (Ib)-(If-1), (IIa)-(IIp), (IIb-1), (IIb-2), (IIc-1)-(IIc-2), (IIn-1), (IIn-2), (IVa)-(IVl), and (IXa)-(IXr)), R3 and R5 join together to form optionally substituted alkylene or optionally substituted heteroalkylene and, taken together with the carbons to which they are attached, provide an optionally substituted heterocyclyl (e.g., a bicyclic, tricyclic, or tetracyclic heterocyclyl, such as trans-3′,4′ analogs, wherein R3 and R5 join together to form heteroalkylene (e.g., —(CH2)b1O(CH2)b2O(CH2)b3-, wherein each of b1, b2, and b3 are, independently, an integer from 0 to 3).
  • In some embodiments of the polynucleotides, primary constructs, or mmRNA (e.g., Formulas (Ia)-(Ia-5), (Ib)-(If-1), (IIa)-(IIp), (IIb-1), (IIb-2), (IIc-1)-(IIc-2), (IIn-1), (IIn-2), (IVa)-(IVl), and (IXa)-(IXr)), R3 and one or more of R1′, R1″, R2′, R2″, or R5 join together to form optionally substituted alkylene or optionally substituted heteroalkylene and, taken together with the carbons to which they are attached, provide an optionally substituted heterocyclyl (e.g., a bicyclic, tricyclic, or tetracyclic heterocyclyl, R3 and one or more of R1′, R1″, R2′, R2″, or R5 join together to form heteroalkylene (e.g., —(CH2)b1O(CH2)b2O(CH2)b3-, wherein each of b1, b2, and b3 are, independently, an integer from 0 to 3).
  • In some embodiments of the polynucleotides, primary constructs, or mmRNA (e.g., Formulas (Ia)-(Ia-5), (Ib)-(If-1), (IIa)-(IIp), (IIb-1), (IIb-2), (IIc-1)-(IIc-2), (IIn-1), (IIn-2), (IVa)-(IV1), and (IXa)-(IXr)), R5 and one or more of R1′, R1″, R2′, or R2″ join together to form optionally substituted alkylene or optionally substituted heteroalkylene and, taken together with the carbons to which they are attached, provide an optionally substituted heterocyclyl (e.g., a bicyclic, tricyclic, or tetracyclic heterocyclyl, R5 and one or more of R1′, R1″, R2′, or R2″ join together to form heteroalkylene (e.g., —(CH2)b1O(CH2)b2O(CH2)b3-, wherein each of b1, b2, and b3 are, independently, an integer from 0 to 3).
  • In some embodiments of the polynucleotides, primary constructs, or mmRNA (e.g., Formulas (Ia)-(Ia-5), (Ib)-(If-1), (IIa)-(IIp), (IIb-1), (IIb-2), (IIc-1)-(IIc-2), (IIn-1), (IIn-2), (IVa)-(IVl), and (IXa)-(IXr)), each Y2 is, independently, O, S, or —NRN1-, wherein RN1 is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, or optionally substituted aryl. In particular embodiments, Y2 is NRN1-, wherein RN1 is H or optionally substituted alkyl (e.g., C1-6 alkyl, such as methyl, ethyl, isopropyl, or n-propyl).
  • In some embodiments of the polynucleotides, primary constructs, or mmRNA (e.g., Formulas (Ia)-(Ia-5), (Ib)-(If-1), (IIa)-(IIp), (IIb-1), (IIb-2), (IIc-1)-(IIc-2), (IIn-1), (IIn-2), (IVa)-(IVl), and (IXa)-(IXr)), each Y3 is, independently, O or S.
  • In some embodiments of the polynucleotides, primary constructs, or mmRNA (e.g., Formulas (Ia)-(Ia-5), (Ib)-(If-1), (IIa)-(IIp), (IIb-1), (IIb-2), (IIc-1)-(IIc-2), (IIn-1), (IIn-2), (IVa)-(IVl), and (IXa)-(IXr)), R1 is H; each R2 is, independently, H, halo (e.g., fluoro), hydroxy, optionally substituted alkoxy (e.g., methoxy or ethoxy), or optionally substituted alkoxyalkoxy (e.g., —(CH2)s2(OCH2CH2)s1(CH2)s3OR′, wherein s1 is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R′ is H or C1-20 alkyl, such as wherein s2 is 0, s1 is 1 or 2, s3 is 0 or 1, and R′ is C1-6 alkyl); each Y2 is, independently, O or —NRN1-, wherein RN1 is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, or optionally substituted aryl (e.g., wherein RN1 is H or optionally substituted alkyl (e.g., C1-6 alkyl, such as methyl, ethyl, isopropyl, or n-propyl)); and each Y3 is, independently, O or S (e.g., S). In further embodiments, R3 is H, halo (e.g., fluoro), hydroxy, optionally substituted alkyl, optionally substituted alkoxy (e.g., methoxy or ethoxy), or optionally substituted alkoxyalkoxy. In yet further embodiments, each Y1 is, independently, O or —NRN1-, wherein RN1 is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, or optionally substituted aryl (e.g., wherein RN1 is H or optionally substituted alkyl (e.g., C1-6 alkyl, such as methyl, ethyl, isopropyl, or n-propyl)); and each Y4 is, independently, H, hydroxy, thiol, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted thioalkoxy, optionally substituted alkoxyalkoxy, or optionally substituted amino.
  • In some embodiments of the polynucleotides, primary constructs, or mmRNA (e.g., Formulas (Ia)-(Ia-5), (Ib)-(If-1), (IIa)-(IIp), (IIb-1), (IIb-2), (IIc-1)-(IIc-2), (IIn-1), (IIn-2), (IVa)-(IVl), and (IXa)-(IXr)), each R1 is, independently, H, halo (e.g., fluoro), hydroxy, optionally substituted alkoxy (e.g., methoxy or ethoxy), or optionally substituted alkoxyalkoxy (e.g., —(CH2)s2(OCH2CH2)s1(CH2)s3OR′, wherein s1 is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R′ is H or C1-20 alkyl, such as wherein s2 is 0, s1 is 1 or 2, s3 is 0 or 1, and R′ is C1-6 alkyl); R2 is H; each Y2 is, independently, O or —NRN1-, wherein RN1 is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, or optionally substituted aryl (e.g., wherein RN1 is H or optionally substituted alkyl (e.g., C1-6 alkyl, such as methyl, ethyl, isopropyl, or n-propyl)); and each Y3 is, independently, O or S (e.g., S). In further embodiments, R3 is H, halo (e.g., fluoro), hydroxy, optionally substituted alkyl, optionally substituted alkoxy (e.g., methoxy or ethoxy), or optionally substituted alkoxyalkoxy. In yet further embodiments, each Y1 is, independently, O or —NRN1-, wherein RN1 is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, or optionally substituted aryl (e.g., wherein RN1 is H or optionally substituted alkyl (e.g., C1-6 alkyl, such as methyl, ethyl, isopropyl, or n-propyl)); and each Y4 is, independently, H, hydroxy, thiol, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted thioalkoxy, optionally substituted alkoxyalkoxy, or optionally substituted amino.
  • In some embodiments of the polynucleotides, primary constructs, or mmRNA (e.g., Formulas (Ia)-(Ia-5), (Ib)-(If-1), (IIa)-(IIp), (IIb-1), (IIb-2), (IIc-1)-(IIc-2), (IIn-1), (IIn-2), (IVa)-(IVl), and (IXa)-(IXr)), the ring including U is in the β-D (e.g., β-D-ribo) configuration.
  • In some embodiments of the polynucleotides, primary constructs, or mmRNA (e.g., Formulas (Ia)-(Ia-5), (Ib)-(If-1), (IIa)-(IIp), (IIb-2), (IIn-1), (IVa)-(IVl), and (IXa)-(IXr)), the ring including U is in the α-L (e.g., α-L-ribo) configuration.
  • In some embodiments of the polynucleotides, primary constructs, or mmRNA (e.g., Formulas (Ia)-(Ia-5), (Ib)-(If-1), (IIa)-(IIp), (IIb-2), (IIn-1), (IVa)-(IVl), and (IXa)-(IXr)), one or more B is not pseudouridine (w) or 5-methyl-cytidine (m5C). In some embodiments, about 10% to about 100% of n number of B nucleobases is not w or m5C (e.g., from 10% to 20%, from 10% to 35%, from 10% to 50%, from 10% to 60%, from 10% to 75%, from 10% to 90%, from 10% to 95%, from 10% to 98%, from 10% to 99%, from 20% to 35%, from 20% to 50%, from 20% to 60%, from 20% to 75%, from 20% to 90%, from 20% to 95%, from 20% to 98%, from 20% to 99%, from 20% to 100%, from 50% to 60%, from 50% to 75%, from 50% to 90%, from 50% to 95%, from 50% to 98%, from 50% to 99%, from 50% to 100%, from 75% to 90%, from 75% to 95%, from 75% to 98%, from 75% to 99%, and from 75% to 100% of n number of B is not ψ or m5C). In some embodiments, B is not ψ or m5C.
  • In some embodiments of the polynucleotides, primary constructs, or mmRNA (e.g., Formulas (Ia)-(Ia-5), (Ib)-(If-1), (IIa)-(IIp), (IIb-1), (IIb-2), (IIn-1), (IVa)-(IVl), and (IXa)-(IXr)), when B is an unmodified nucleobase selected from cytosine, guanine, uracil and adenine, then at least one of Y1, Y2, or Y3 is not 0.
  • In some embodiments, the polynucleotide, primary construct, or mmRNA includes a modified ribose. In some embodiments, the polynucleotide, primary construct, or mmRNA (e.g., the first region, the first flanking region, or the second flanking region) includes n number of linked nucleosides having Formula (IIa)-(IIc):
  • Figure US20190085368A1-20190321-C00008
  • or a pharmaceutically acceptable salt or stereoisomer thereof. In particular embodiments, U is O or C(RU)nu, wherein nu is an integer from 0 to 2 and each RU is, independently, H, halo, or optionally substituted alkyl (e.g., U is —CH2- or —CH—). In other embodiments, each of R1, R2, R3, R4, and R5 is, independently, H, halo, hydroxy, thiol, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, optionally substituted hydroxyalkoxy, optionally substituted amino, azido, optionally substituted aryl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, or absent (e.g., each R1 and R2 is, independently, H, halo, hydroxy, optionally substituted alkyl, or optionally substituted alkoxy; each R3 and R4 is, independently, H or optionally substituted alkyl; and R5 is H or hydroxy), and - - - is a single bond or double bond.
  • In particular embodiments, the polynucleotides or mmRNA includes n number of linked nucleosides having Formula (IIb-1)-(IIb-2):
  • Figure US20190085368A1-20190321-C00009
  • or a pharmaceutically acceptable salt or stereoisomer thereof. In some embodiments, U is O or C(RU)nu, wherein nu is an integer from 0 to 2 and each RU is, independently, H, halo, or optionally substituted alkyl (e.g., U is —CH2- or —CH—). In other embodiments, each of R1 and R2 is, independently, H, halo, hydroxy, thiol, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, optionally substituted hydroxyalkoxy, optionally substituted amino, azido, optionally substituted aryl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, or absent (e.g., each R1 and R2 is, independently, H, halo, hydroxy, optionally substituted alkyl, or optionally substituted alkoxy, e.g., H, halo, hydroxy, alkyl, or alkoxy). In particular embodiments, R2 is hydroxy or optionally substituted alkoxy (e.g., methoxy, ethoxy, or any described herein).
  • In particular embodiments, the polynucleotide, primary construct, or mmRNA includes n number of linked nucleosides having Formula (IIc-1)-(IIc-4):
  • Figure US20190085368A1-20190321-C00010
  • or a pharmaceutically acceptable salt or stereoisomer thereof. In some embodiments, U is O or C(RU)nu, wherein nu is an integer from 0 to 2 and each RU is, independently, H, halo, or optionally substituted alkyl (e.g., U is —CH2- or —CH—). In some embodiments, each of R1, R2, and R3 is, independently, H, halo, hydroxy, thiol, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, optionally substituted hydroxyalkoxy, optionally substituted amino, azido, optionally substituted aryl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, or absent (e.g., each R1 and R2 is, independently, H, halo, hydroxy, optionally substituted alkyl, or optionally substituted alkoxy, e.g., H, halo, hydroxy, alkyl, or alkoxy; and each R3 is, independently, H or optionally substituted alkyl)). In particular embodiments, R2 is optionally substituted alkoxy (e.g., methoxy or ethoxy, or any described herein). In particular embodiments, R1 is optionally substituted alkyl, and R2 is hydroxy. In other embodiments, R1 is hydroxy, and R2 is optionally substituted alkyl. In further embodiments, R3 is optionally substituted alkyl.
  • In some embodiments, the polynucleotide, primary construct, or mmRNA includes an acyclic modified ribose. In some embodiments, the polynucleotide, primary construct, or mmRNA (e.g., the first region, the first flanking region, or the second flanking region) includes n number of linked nucleosides having Formula (IId)-(IIf):
  • Figure US20190085368A1-20190321-C00011
  • or a pharmaceutically acceptable salt or stereoisomer thereof.
  • In some embodiments, the polynucleotide, primary construct, or mmRNA includes an acyclic modified hexitol. In some embodiments, the polynucleotide, primary construct, or mmRNA (e.g., the first region, the first flanking region, or the second flanking region) includes n number of linked nucleosides Formula (IIg)-(IIj):
  • Figure US20190085368A1-20190321-C00012
  • or a pharmaceutically acceptable salt or stereoisomer thereof.
  • In some embodiments, the polynucleotide, primary construct, or mmRNA includes a sugar moiety having a contracted or an expanded ribose ring. In some embodiments, the polynucleotide, primary construct, or mmRNA (e.g., the first region, the first flanking region, or the second flanking region) includes n number of linked nucleosides having Formula (IIk)-(IIm):
  • Figure US20190085368A1-20190321-C00013
  • or a pharmaceutically acceptable salt or stereoisomer thereof, wherein each of R1′, R1″, R2′, and R2″ is, independently, H, halo, hydroxy, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, or absent; and wherein the combination of R2′ and R3 or the combination of R2″ and R3 can be taken together to form optionally substituted alkylene or optionally substituted heteroalkylene.
  • In some embodiments, the polynucleotide, primary construct, or mmRNA includes a locked modified ribose. In some embodiments, the polynucleotide, primary construct, or mmRNA (e.g., the first region, the first flanking region, or the second flanking region) includes n number of linked nucleosides having Formula (IIn):
  • Figure US20190085368A1-20190321-C00014
  • or a pharmaceutically acceptable salt or stereoisomer thereof, wherein R3′ is O, S, or —NRN1-, wherein RN1 is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, or optionally substituted aryl and R3″ is optionally substituted alkylene (e.g., —CH2-, —CH2CH2-, or —CH2CH2CH2-) or optionally substituted heteroalkylene (e.g., —CH2NH—, —CH2CH2NH—, —CH2OCH2-, or —CH2CH2OCH2-)(e.g., R3′ is O and R3″ is optionally substituted alkylene (e.g., —CH2-, —CH2CH2-, or —CH2CH2CH2-)).
  • In some embodiments, the polynucleotide, primary construct, or mmRNA includes n number of linked nucleosides having Formula (IIn-1)-(II-n2):
  • Figure US20190085368A1-20190321-C00015
  • or a pharmaceutically acceptable salt or stereoisomer thereof, wherein R3′ is O, S, or —NRN1-, wherein RN1 is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, or optionally substituted aryl and R3″ is optionally substituted alkylene (e.g., —CH2-, —CH2CH2-, or —CH2CH2CH2-) or optionally substituted heteroalkylene (e.g., —CH2NH—, —CH2CH2NH—, —CH2OCH2-, or —CH2CH2OCH2-) (e.g., R3′ is O and R3″ is optionally substituted alkylene (e.g., —CH2-, —CH2CH2-, or —CH2CH2CH2-)).
  • In some embodiments, the polynucleotide, primary construct, or mmRNA includes a locked modified ribose that forms a tetracyclic heterocyclyl. In some embodiments, the polynucleotide, primary construct, or mmRNA (e.g., the first region, the first flanking region, or the second flanking region) includes n number of linked nucleosides having Formula (IIo):
  • Figure US20190085368A1-20190321-C00016
  • or a pharmaceutically acceptable salt or stereoisomer thereof, wherein R12a, R12c, T1′, T1″, T2′, T2″, V1, and V3 are as described herein.
  • Any of the formulas for the polynucleotides, primary constructs, or mmRNA can include one or more nucleobases described herein (e.g., Formulas (b1)-(b43)).
  • In one embodiment, the present invention provides methods of preparing a polynucleotide, primary construct, or mmRNA, wherein the polynucleotide comprises n number of nucleosides having Formula (Ia), as defined herein:
  • Figure US20190085368A1-20190321-C00017
  • the method comprising reacting a compound of Formula (IIIa), as defined herein:
  • Figure US20190085368A1-20190321-C00018
  • with an RNA polymerase, and a cDNA template.
  • In a further embodiment, the present invention provides methods of amplifying a polynucleotide, primary construct, or mmRNA comprising at least one nucleotide (e.g., mmRNA molecule), the method comprising: reacting a compound of Formula (IIIa), as defined herein, with a primer, a cDNA template, and an RNA polymerase.
  • In one embodiment, the present invention provides methods of preparing a polynucleotide, primary construct, or mmRNA comprising at least one nucleotide (e.g., mmRNA molecule), wherein the polynucleotide comprises n number of nucleosides having Formula (Ia), as defined herein:
  • Figure US20190085368A1-20190321-C00019
  • the method comprising reacting a compound of Formula (IIIa-1), as defined herein:
  • Figure US20190085368A1-20190321-C00020
  • with an RNA polymerase, and a cDNA template.
  • In a further embodiment, the present invention provides methods of amplifying a polynucleotide, primary construct, or mmRNA comprising at least one nucleotide (e.g., mmRNA molecule), the method comprising:
  • reacting a compound of Formula (IIIa-1), as defined herein, with a primer, a cDNA template, and an RNA polymerase.
  • In one embodiment, the present invention provides methods of preparing a modified mRNA comprising at least one nucleotide (e.g., mmRNA molecule), wherein the polynucleotide comprises n number of nucleosides having Formula (Ia-2), as defined herein:
  • Figure US20190085368A1-20190321-C00021
  • the method comprising reacting a compound of Formula (IIIa-2), as defined herein:
  • Figure US20190085368A1-20190321-C00022
  • with an RNA polymerase, and a cDNA template.
  • In a further embodiment, the present invention provides methods of amplifying a modified mRNA comprising at least one nucleotide (e.g., mmRNA molecule), the method comprising: reacting a compound of Formula (IIIa-2), as defined herein, with a primer, a cDNA template, and an RNA polymerase.
  • In some embodiments, the reaction may be repeated from 1 to about 7,000 times. In any of the embodiments herein, B may be a nucleobase of Formula (b1)-(b43).
  • The polynucleotides, primary constructs, and mmRNA can optionally include 5′ and/or 3′ flanking regions, which are described herein.
  • Modified RNA (mmRNA) Molecules
  • The present invention also includes building blocks, e.g., modified ribonucleosides, modified ribonucleotides, of modified RNA (mmRNA) molecules. For example, these building blocks can be useful for preparing the polynucleotides, primary constructs, or mmRNA of the invention.
  • In some embodiments, the building block molecule has Formula (IIa) or (IIa-1):
  • Figure US20190085368A1-20190321-C00023
  • or a pharmaceutically acceptable salt or stereoisomer thereof, wherein the substituents are as described herein (e.g., for Formula (Ia) and (Ia-1)), and wherein when B is an unmodified nucleobase selected from cytosine, guanine, uracil and adenine, then at least one of Y1, Y2, or Y3 is not O.
  • In some embodiments, the building block molecule, which may be incorporated into a polynucleotide, primary construct, or mmRNA, has Formula (IVa)-(IVb):
  • Figure US20190085368A1-20190321-C00024
  • or a pharmaceutically acceptable salt or stereoisomer thereof, wherein B is as described herein (e.g., any one of (b1)-(b43)). In particular embodiments, Formula (IVa) or (IVb) is combined with a modified uracil (e.g., any one of formulas (b1)-(b9), (b21)-(b23), and (b28)-(b31), such as formula (b1), (b8), (b28), (b29), or (b30)). In particular embodiments, Formula (IVa) or (IVb) is combined with a modified cytosine (e.g., any one of formulas (b10)-(b14), (b24), (b25), and (b32)-(b36), such as formula (b10) or (b32)). In particular embodiments, Formula (IVa) or (IVb) is combined with a modified guanine (e.g., any one of formulas (b15)-(b17) and (b37)-(b40)). In particular embodiments, Formula (IVa) or (IVb) is combined with a modified adenine (e.g., any one of formulas (b18)-(b20) and (b41)-(b43)).
  • In some embodiments, the building block molecule, which may be incorporated into a polynucleotide, primary construct, or mmRNA, has Formula (IVc)-(IVk):
  • Figure US20190085368A1-20190321-C00025
    Figure US20190085368A1-20190321-C00026
  • or a pharmaceutically acceptable salt or stereoisomer thereof, wherein B is as described herein (e.g., any one of (b1)-(b43)). In particular embodiments, one of Formulas (IVc)-(IVk) is combined with a modified uracil (e.g., any one of formulas (b1)-(b9), (b21)-(b23), and (b28)-(b31), such as formula (b1), (b8), (b28), (b29), or (b30)). In particular embodiments, one of Formulas (IVc)-(IVk) is combined with a modified cytosine (e.g., any one of formulas (b10)-(b14), (b24), (b25), and (b32)-(b36), such as formula (b10) or (b32)). In particular embodiments, one of Formulas (IVc)-(IVk) is combined with a modified guanine (e.g., any one of formulas (b15)-(b17) and (b37)-(b40)). In particular embodiments, one of Formulas (IVc)-(IVk) is combined with a modified adenine (e.g., any one of formulas (b18)-(b20) and (b41)-(b43)).
  • In other embodiments, the building block molecule, which may be incorporated into a polynucleotide, primary construct, or mmRNA, has Formula (Va) or (Vb):
  • Figure US20190085368A1-20190321-C00027
  • or a pharmaceutically acceptable salt or stereoisomer thereof, wherein B is as described herein (e.g., any one of (b1)-(b43)).
  • In other embodiments, the building block molecule, which may be incorporated into a polynucleotide, primary construct, or mmRNA, has Formula (IXa)-(IXd):
  • Figure US20190085368A1-20190321-C00028
  • or a pharmaceutically acceptable salt or stereoisomer thereof, wherein B is as described herein (e.g., any one of (b1)-(b43)). In particular embodiments, one of Formulas (IXa)-(IXd) is combined with a modified uracil (e.g., any one of formulas (b1)-(b9), (b21)-(b23), and (b28)-(b31), such as formula (b1), (b8), (b28), (b29), or (b30)). In particular embodiments, one of Formulas (IXa)-(IXd) is combined with a modified cytosine (e.g., any one of formulas (b10)-(b14), (b24), (b25), and (b32)-(b36), such as formula (b10) or (b32)). In particular embodiments, one of Formulas (IXa)-(IXd) is combined with a modified guanine (e.g., any one of formulas (b15)-(b17) and (b37)-(b40)). In particular embodiments, one of Formulas (IXa)-(IXd) is combined with a modified adenine (e.g., any one of formulas (b18)-(b20) and (b41)-(b43)).
  • In other embodiments, the building block molecule, which may be incorporated into a polynucleotide, primary construct, or mmRNA, has Formula (IXe)-(IXg):
  • Figure US20190085368A1-20190321-C00029
  • or a pharmaceutically acceptable salt or stereoisomer thereof, wherein B is as described herein (e.g., any one of (b1)-(b43)). In particular embodiments, one of Formulas (IXe)-(IXg) is combined with a modified uracil (e.g., any one of formulas (b1)-(b9), (b21)-(b23), and (b28)-(b31), such as formula (b1), (b8), (b28), (b29), or (b30)). In particular embodiments, one of Formulas (IXe)-(IXg) is combined with a modified cytosine (e.g., any one of formulas (b10)-(b14), (b24), (b25), and (b32)-(b36), such as formula (b10) or (b32)). In particular embodiments, one of Formulas (IXe)-(IXg) is combined with a modified guanine (e.g., any one of formulas (b15)-(b17) and (b37)-(b40)). In particular embodiments, one of Formulas (IXe)-(IXg) is combined with a modified adenine (e.g., any one of formulas (b18)-(b20) and (b41)-(b43)).
  • In other embodiments, the building block molecule, which may be incorporated into a polynucleotide, primary construct, or mmRNA, has Formula (IXh)-(IXk):
  • Figure US20190085368A1-20190321-C00030
  • or a pharmaceutically acceptable salt or stereoisomer thereof, wherein B is as described herein (e.g., any one of (b1)-(b43)). In particular embodiments, one of Formulas (IXh)-(IXk) is combined with a modified uracil (e.g., any one of formulas (b1)-(b9), (b21)-(b23), and (b28)-(b31), such as formula (b1), (b8), (b28), (b29), or (b30)). In particular embodiments, one of Formulas (IXh)-(IXk) is combined with a modified cytosine (e.g., any one of formulas (b10)-(b14), (b24), (b25), and (b32)-(b36), such as formula (b10) or (b32)). In particular embodiments, one of Formulas (IXh)-(IXk) is combined with a modified guanine (e.g., any one of formulas (b15)-(b17) and (b37)-(b40)). In particular embodiments, one of Formulas (IXh)-(IXk) is combined with a modified adenine (e.g., any one of formulas (b18)-(b20) and (b41)-(b43)).
  • In other embodiments, the building block molecule, which may be incorporated into a polynucleotide, primary construct, or mmRNA, has Formula (IXl)-(IXr):
  • Figure US20190085368A1-20190321-C00031
    Figure US20190085368A1-20190321-C00032
  • or a pharmaceutically acceptable salt or stereoisomer thereof, wherein each r1 and r2 is, independently, an integer from 0 to 5 (e.g., from 0 to 3, from 1 to 3, or from 1 to 5) and B is as described herein (e.g., any one of (b1)-(b43)). In particular embodiments, one of Formulas (IXl)-(IXr) is combined with a modified uracil (e.g., any one of formulas (b1)-(b9), (b21)-(b23), and (b28)-(b31), such as formula (b1), (b8), (b28), (b29), or (b30)). In particular embodiments, one of Formulas (IXl)-(IXr) is combined with a modified cytosine (e.g., any one of formulas (b10)-(b14), (b24), (b25), and (b32)-(b36), such as formula (b10) or (b32)). In particular embodiments, one of Formulas (IXl)-(IXr) is combined with a modified guanine (e.g., any one of formulas (b15)-(b17) and (b37)-(b40)). In particular embodiments, one of Formulas (IXl)-(IXr) is combined with a modified adenine (e.g., any one of formulas (b18)-(b20) and (b41)-(b43)).
  • In some embodiments, the building block molecule, which may be incorporated into a polynucleotide, primary construct, or mmRNA, can be selected from the group consisting of:
  • Figure US20190085368A1-20190321-C00033
    Figure US20190085368A1-20190321-C00034
    Figure US20190085368A1-20190321-C00035
  • or a pharmaceutically acceptable salt or stereoisomer thereof, wherein each r is, independently, an integer from 0 to 5 (e.g., from 0 to 3, from 1 to 3, or from 1 to 5).
  • In some embodiments, the building block molecule, which may be incorporated into a polynucleotide, primary construct, or mmRNA, can be selected from the group consisting of:
  • Figure US20190085368A1-20190321-C00036
    Figure US20190085368A1-20190321-C00037
  • or a pharmaceutically acceptable salt or stereoisomer thereof, wherein each r is, independently, an integer from 0 to 5 (e.g., from 0 to 3, from 1 to 3, or from 1 to 5) and s1 is as described herein.
  • In some embodiments, the building block molecule, which may be incorporated into a nucleic acid (e.g., RNA, mRNA, polynucleotide, primary construct, or mmRNA), is a modified uridine (e.g., selected from the group consisting of:
  • Figure US20190085368A1-20190321-C00038
    Figure US20190085368A1-20190321-C00039
    Figure US20190085368A1-20190321-C00040
    Figure US20190085368A1-20190321-C00041
    Figure US20190085368A1-20190321-C00042
    Figure US20190085368A1-20190321-C00043
    Figure US20190085368A1-20190321-C00044
    Figure US20190085368A1-20190321-C00045
    Figure US20190085368A1-20190321-C00046
    Figure US20190085368A1-20190321-C00047
    Figure US20190085368A1-20190321-C00048
    Figure US20190085368A1-20190321-C00049
    Figure US20190085368A1-20190321-C00050
    Figure US20190085368A1-20190321-C00051
    Figure US20190085368A1-20190321-C00052
    Figure US20190085368A1-20190321-C00053
    Figure US20190085368A1-20190321-C00054
    Figure US20190085368A1-20190321-C00055
    Figure US20190085368A1-20190321-C00056
    Figure US20190085368A1-20190321-C00057
    Figure US20190085368A1-20190321-C00058
    Figure US20190085368A1-20190321-C00059
  • or a pharmaceutically acceptable salt or stereoisomer thereof, wherein Y1, Y3, Y4, Y6, and r are as described herein (e.g., each r is, independently, an integer from 0 to 5, such as from 0 to 3, from 1 to 3, or from 1 to 5)).
  • In some embodiments, the building block molecule, which may be incorporated into a polynucleotide, primary construct, or mmRNA, is a modified cytidine (e.g., selected from the group consisting of:
  • Figure US20190085368A1-20190321-C00060
    Figure US20190085368A1-20190321-C00061
    Figure US20190085368A1-20190321-C00062
    Figure US20190085368A1-20190321-C00063
    Figure US20190085368A1-20190321-C00064
    Figure US20190085368A1-20190321-C00065
    Figure US20190085368A1-20190321-C00066
  • or a pharmaceutically acceptable salt or stereoisomer thereof, wherein Y1, Y3, Y4, Y6, and r are as described herein (e.g., each r is, independently, an integer from 0 to 5, such as from 0 to 3, from 1 to 3, or from 1 to 5)). For example, the building block molecule, which may be incorporated into a polynucleotide, primary construct, or mmRNA, can be:
  • Figure US20190085368A1-20190321-C00067
  • or a pharmaceutically acceptable salt or stereoisomer thereof, wherein each r is, independently, an integer from 0 to 5 (e.g., from 0 to 3, from 1 to 3, or from 1 to 5).
  • In some embodiments, the building block molecule, which may be incorporated into a polynucleotide, primary construct, or mmRNA, is a modified adenosine (e.g., selected from the group consisting of:
  • Figure US20190085368A1-20190321-C00068
    Figure US20190085368A1-20190321-C00069
    Figure US20190085368A1-20190321-C00070
    Figure US20190085368A1-20190321-C00071
    Figure US20190085368A1-20190321-C00072
    Figure US20190085368A1-20190321-C00073
    Figure US20190085368A1-20190321-C00074
    Figure US20190085368A1-20190321-C00075
    Figure US20190085368A1-20190321-C00076
  • or a pharmaceutically acceptable salt or stereoisomer thereof, wherein Y1, Y3, Y4, Y6, and r are as described herein (e.g., each r is, independently, an integer from 0 to 5, such as from 0 to 3, from 1 to 3, or from 1 to 5)).
  • In some embodiments, the building block molecule, which may be incorporated into a polynucleotide, primary construct, or mmRNA, is a modified guanosine (e.g., selected from the group consisting of:
  • Figure US20190085368A1-20190321-C00077
    Figure US20190085368A1-20190321-C00078
    Figure US20190085368A1-20190321-C00079
    Figure US20190085368A1-20190321-C00080
    Figure US20190085368A1-20190321-C00081
    Figure US20190085368A1-20190321-C00082
    Figure US20190085368A1-20190321-C00083
    Figure US20190085368A1-20190321-C00084
  • or a pharmaceutically acceptable salt or stereoisomer thereof, wherein Y1, Y3, Y4, Y6, and r are as described herein (e.g., each r is, independently, an integer from 0 to 5, such as from 0 to 3, from 1 to 3, or from 1 to 5)).
  • In some embodiments, the chemical modification can include replacement of C group at C-5 of the ring (e.g., for a pyrimidine nucleoside, such as cytosine or uracil) with N (e.g., replacement of the >CH group at C-5 with >NRN1 group, wherein RN1 is H or optionally substituted alkyl). For example, the building block molecule, which may be incorporated into a polynucleotide, primary construct, or mmRNA, can be:
  • Figure US20190085368A1-20190321-C00085
  • or a pharmaceutically acceptable salt or stereoisomer thereof, wherein each r is, independently, an integer from 0 to 5 (e.g., from 0 to 3, from 1 to 3, or from 1 to 5).
  • In another embodiment, the chemical modification can include replacement of the hydrogen at C-5 of cytosine with halo (e.g., Br, Cl, F, or I) or optionally substituted alkyl (e.g., methyl). For example, the building block molecule, which may be incorporated into a polynucleotide, primary construct, or mmRNA, can be:
  • Figure US20190085368A1-20190321-C00086
  • or a pharmaceutically acceptable salt or stereoisomer thereof, wherein each r is, independently, an integer from 0 to 5 (e.g., from 0 to 3, from 1 to 3, or from 1 to 5).
  • In yet a further embodiment, the 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 building block molecule, which may be incorporated into a polynucleotide, primary construct, or mmRNA, can be:
  • Figure US20190085368A1-20190321-C00087
  • or a pharmaceutically acceptable salt or stereoisomer thereof, wherein each r is, independently, an integer from 0 to 5 (e.g., from 0 to 3, from 1 to 3, or from 1 to 5).
  • Modifications on the Sugar
  • The modified nucleosides and nucleotides (e.g., building block molecules), which may be incorporated into a polynucleotide, primary construct, or mmRNA (e.g., RNA or mRNA, as described herein), can be modified on the sugar of the ribonucleic acid. For example, the 2′ hydroxyl group (OH) can be modified or replaced with a number of different substituents. Exemplary substitutions at the 2′-position include, but are not limited to, H, halo, optionally substituted C1-6 alkyl; optionally substituted C1-6 alkoxy; optionally substituted C6-10 aryloxy; optionally substituted C3-8 cycloalkyl; optionally substituted C3-8 cycloalkoxy; optionally substituted C6-10 aryloxy; optionally substituted C6-10 aryl-C1-6 alkoxy, optionally substituted C1-12 (heterocyclyl)oxy; a sugar (e.g., ribose, pentose, or any described herein); a polyethyleneglycol (PEG), —O(CH2CH2O)nCH2CH2OR, where R is H or optionally substituted alkyl, and n is an integer from 0 to 20 (e.g., from 0 to 4, from 0 to 8, from 0 to 10, from 0 to 16, from 1 to 4, from 1 to 8, from 1 to 10, from 1 to 16, from 1 to 20, from 2 to 4, from 2 to 8, from 2 to 10, from 2 to 16, from 2 to 20, from 4 to 8, from 4 to 10, from 4 to 16, and from 4 to 20); “locked” nucleic acids (LNA) in which the 2′-hydroxyl is connected by a C1-6 alkylene or C1-6 heteroalkylene bridge to the 4′-carbon of the same ribose sugar, where exemplary bridges included methylene, propylene, ether, or amino bridges; aminoalkyl, as defined herein; aminoalkoxy, as defined herein; amino as defined herein; and amino acid, as defined herein
  • Generally, RNA includes the sugar group ribose, which is a 5-membered ring having an oxygen. Exemplary, non-limiting modified nucleotides include replacement of the oxygen in ribose (e.g., with S, Se, or alkylene, such as methylene or ethylene); addition of a double bond (e.g., to replace ribose with cyclopentenyl or cyclohexenyl); ring contraction of ribose (e.g., to form a 4-membered ring of cyclobutane or oxetane); ring expansion of ribose (e.g., to form a 6- or 7-membered ring having an additional carbon or heteroatom, such as for anhydrohexitol, altritol, mannitol, cyclohexanyl, cyclohexenyl, and morpholino that also has a phosphoramidate backbone); multicyclic forms (e.g., tricyclo; and “unlocked” forms, such as glycol nucleic acid (GNA) (e.g., R-GNA or S-GNA, where ribose is replaced by glycol units attached to phosphodiester bonds), threose nucleic acid (TNA, where ribose is replace with α-L-threofuranosyl-(3′→2)), and peptide nucleic acid (PNA, where 2-amino-ethyl-glycine linkages replace the ribose and phosphodiester backbone). The sugar group can also contain one or more carbons that possess the opposite stereochemical configuration than that of the corresponding carbon in ribose. Thus, a polynucleotide, primary construct, or mmRNA molecule can include nucleotides containing, e.g., arabinose, as the sugar.
  • Modifications on the Nucleobase
  • The present disclosure provides for modified nucleosides and nucleotides. As described herein “nucleoside” is defined as a compound containing a sugar molecule (e.g., a pentose or ribose) or a derivative thereof in combination with an organic base (e.g., a purine or pyrimidine) or a derivative thereof (also referred to herein as “nucleobase”). As described herein, “nucleotide” is defined as a nucleoside including a phosphate group. The modified nucleotides may by synthesized by any useful method, as described herein (e.g., chemically, enzymatically, or recombinantly to include one or more modified or non-natural nucleosides).
  • The modified nucleotide base pairing encompasses not only the standard adenosine-thymine, adenosine-uracil, or guanosine-cytosine base pairs, but also base pairs formed between nucleotides and/or modified nucleotides comprising non-standard or modified bases, wherein the arrangement of hydrogen bond donors and hydrogen bond acceptors permits hydrogen bonding between a non-standard base and a standard base or between two complementary non-standard base structures. One example of such non-standard base pairing is the base pairing between the modified nucleotide inosine and adenine, cytosine or uracil.
  • The modified nucleosides and nucleotides can include a modified nucleobase. Examples of nucleobases found in RNA include, but are not limited to, adenine, guanine, cytosine, and uracil. Examples of nucleobase found in DNA include, but are not limited to, adenine, guanine, cytosine, and thymine. These nucleobases can be modified or wholly replaced to provide polynucleotides, primary constructs, or mmRNA molecules having enhanced properties, e.g., resistance to nucleases through disruption of the binding of a major groove binding partner. Table 8 below identifies the chemical faces of each canonical nucleotide. Circles identify the atoms comprising the respective chemical regions.
  • TABLE 8
    Watson-Crick
    Major Groove Minor Groove Base-pairing
    Face Face Face
    Pyrimidines Cytidine:
    Figure US20190085368A1-20190321-C00088
    Figure US20190085368A1-20190321-C00089
    Figure US20190085368A1-20190321-C00090
    Uridine:
    Figure US20190085368A1-20190321-C00091
    Figure US20190085368A1-20190321-C00092
    Figure US20190085368A1-20190321-C00093
    Purines Adenosine:
    Figure US20190085368A1-20190321-C00094
    Figure US20190085368A1-20190321-C00095
    Figure US20190085368A1-20190321-C00096
    Guanosine:
    Figure US20190085368A1-20190321-C00097
    Figure US20190085368A1-20190321-C00098
    Figure US20190085368A1-20190321-C00099
  • In some embodiments, B is a modified uracil. Exemplary modified uracils include those having Formula (b1)-(b5):
  • Figure US20190085368A1-20190321-C00100
  • or a pharmaceutically acceptable salt or stereoisomer thereof,
  • Wherein
    Figure US20190085368A1-20190321-P00002
    is a single or double bond; each of T1′, T1″, T2′, and T2″ is, independently, H, optionally substituted alkyl, optionally substituted alkoxy, or optionally substituted thioalkoxy, or the combination of T1′ and T1″ or the combination of T2′ and T2″ join together (e.g., as in T2) to form O (oxo), S (thio), or Se (seleno); each of V1 and V2 is, independently, O, S, N(RVb)nv, or C(RVb)nv, wherein nv is an integer from 0 to 2 and each RVb is, independently, H, halo, optionally substituted amino acid, optionally substituted alkyl, optionally substituted haloalkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted hydroxyalkyl, optionally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally substituted aminoalkyl (e.g., substituted with an N-protecting group, such as any described herein, e.g., trifluoroacetyl), optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, optionally substituted acylaminoalkyl (e.g., substituted with an N-protecting group, such as any described herein, e.g., trifluoroacetyl), optionally substituted alkoxycarbonylalkyl, optionally substituted alkoxycarbonylalkenyl, optionally substituted alkoxycarbonylalkynyl, or optionally substituted alkynyloxy (e.g., optionally substituted with any substituent described herein, such as those selected from (1)-(21) for alkyl);
  • R10 is H, halo, optionally substituted amino acid, hydroxy, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aminoalkyl, optionally substituted hydroxyalkyl, optionally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, optionally substituted alkoxy, optionally substituted alkoxycarbonylalkyl, optionally substituted alkoxycarbonylalkenyl, optionally substituted alkoxycarbonylalkynyl, optionally substituted alkoxycarbonylalkoxy, optionally substituted carboxyalkoxy, optionally substituted carboxyalkyl, or optionally substituted carbamoylalkyl;
  • R11 is H or optionally substituted alkyl;
  • R12a is H, optionally substituted alkyl, optionally substituted hydroxyalkyl, optionally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, or optionally substituted aminoalkynyl, optionally substituted carboxyalkyl (e.g., optionally substituted with hydroxy), optionally substituted carboxyalkoxy, optionally substituted carboxyaminoalkyl, or optionally substituted carbamoylalkyl; and
  • R12c is H, halo, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted thioalkoxy, optionally substituted amino, optionally substituted hydroxyalkyl, optionally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, or optionally substituted aminoalkynyl. Other exemplary modified uracils include those having Formula (b6)-(b9):
  • Figure US20190085368A1-20190321-C00101
  • or a pharmaceutically acceptable salt or stereoisomer thereof,
  • wherein
  • Figure US20190085368A1-20190321-P00002
    is a single or double bond;
  • each of T1′, T1″, T2′, and T2″ is, independently, H, optionally substituted alkyl, optionally substituted alkoxy, or optionally substituted thioalkoxy, or the combination of T1′ and T1″ join together (e.g., as in T1) or the combination of T2′ and T2″ join together (e.g., as in T2) to form O (oxo), S (thio), or Se (seleno), or each T1 and T2 is, independently, O (oxo), S (thio), or Se (seleno);
  • each of W1 and W2 is, independently, N(RWa)nw or C(RWa)nw, wherein nw is an integer from 0 to 2 and each RWa is, independently, H, optionally substituted alkyl, or optionally substituted alkoxy;
  • each V3 is, independently, O, S, N(RVa)nv, or C(RVa)nv, wherein nv is an integer from 0 to 2 and each RVa is, independently, H, halo, optionally substituted amino acid, optionally substituted alkyl, optionally substituted hydroxyalkyl, optionally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted heterocyclyl, optionally substituted alkheterocyclyl, optionally substituted alkoxy, optionally substituted alkenyloxy, or optionally substituted alkynyloxy, optionally substituted aminoalkyl (e.g., substituted with an N-protecting group, such as any described herein, e.g., trifluoroacetyl, or sulfoalkyl), optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, optionally substituted acylaminoalkyl (e.g., substituted with an N-protecting group, such as any described herein, e.g., trifluoroacetyl), optionally substituted alkoxycarbonylalkyl, optionally substituted alkoxycarbonylalkenyl, optionally substituted alkoxycarbonylalkynyl, optionally substituted alkoxycarbonylacyl, optionally substituted alkoxycarbonylalkoxy, optionally substituted carboxyalkyl (e.g., optionally substituted with hydroxy and/or an O-protecting group), optionally substituted carboxyalkoxy, optionally substituted carboxyaminoalkyl, or optionally substituted carbamoylalkyl (e.g., optionally substituted with any substituent described herein, such as those selected from (1)-(21) for alkyl), and wherein RVa and R12c taken together with the carbon atoms to which they are attached can form optionally substituted cycloalkyl, optionally substituted aryl, or optionally substituted heterocyclyl (e.g., a 5- or 6-membered ring);
  • R12a is H, optionally substituted alkyl, optionally substituted hydroxyalkyl, optionally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, optionally substituted carboxyalkyl (e.g., optionally substituted with hydroxy and/or an O-protecting group), optionally substituted carboxyalkoxy, optionally substituted carboxyaminoalkyl, optionally substituted carbamoylalkyl, or absent;
  • R12b is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted hydroxyalkyl, optionally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, optionally substituted alkaryl, optionally substituted heterocyclyl, optionally substituted alkheterocyclyl, optionally substituted amino acid, optionally substituted alkoxycarbonylacyl, optionally substituted alkoxycarbonylalkoxy, optionally substituted alkoxycarbonylalkyl, optionally substituted alkoxycarbonylalkenyl, optionally substituted alkoxycarbonylalkynyl, optionally substituted alkoxycarbonylalkoxy, optionally substituted carboxyalkyl (e.g., optionally substituted with hydroxy and/or an O-protecting group), optionally substituted carboxyalkoxy, optionally substituted carboxyaminoalkyl, or optionally substituted carbamoylalkyl,
  • wherein the combination of R12b and T1′ or the combination of R12b and R12c can join together to form optionally substituted heterocyclyl; and
  • R12c is H, halo, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted thioalkoxy, optionally substituted amino, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, or optionally substituted aminoalkynyl.
  • Further exemplary modified uracils include those having Formula (b28)-(b31):
  • Figure US20190085368A1-20190321-C00102
  • or a pharmaceutically acceptable salt or stereoisomer thereof, wherein each of T1 and T2 is, independently, O (oxo), S (thio), or Se (seleno); each RVb′ and RVb″ is, independently, H, halo, optionally substituted amino acid, optionally substituted alkyl, optionally substituted haloalkyl, optionally substituted hydroxyalkyl, optionally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkyl (e.g., substituted with an N-protecting group, such as any described herein, e.g., trifluoroacetyl, or sulfoalkyl), optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, optionally substituted acylaminoalkyl (e.g., substituted with an N-protecting group, such as any described herein, e.g., trifluoroacetyl), optionally substituted alkoxycarbonylalkyl, optionally substituted alkoxycarbonylalkenyl, optionally substituted alkoxycarbonylalkynyl, optionally substituted alkoxycarbonylacyl, optionally substituted alkoxycarbonylalkoxy, optionally substituted carboxyalkyl (e.g., optionally substituted with hydroxy and/or an O-protecting group), optionally substituted carboxyalkoxy, optionally substituted carboxyaminoalkyl, or optionally substituted carbamoylalkyl (e.g., optionally substituted with any substituent described herein, such as those selected from (1)-(21) for alkyl) (e.g., RVb′ is optionally substituted alkyl, optionally substituted alkenyl, or optionally substituted aminoalkyl, e.g., substituted with an N-protecting group, such as any described herein, e.g., trifluoroacetyl, or sulfoalkyl);
  • R12a is H, optionally substituted alkyl, optionally substituted carboxyaminoalkyl, optionally substituted aminoalkyl (e.g., e.g., substituted with an N-protecting group, such as any described herein, e.g., trifluoroacetyl, or sulfoalkyl), optionally substituted aminoalkenyl, or optionally substituted aminoalkynyl; and
  • R12b is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted hydroxyalkyl, optionally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, optionally substituted aminoalkynyl (e.g., e.g., substituted with an N-protecting group, such as any described herein, e.g., trifluoroacetyl, or sulfoalkyl),
  • optionally substituted alkoxycarbonylacyl, optionally substituted alkoxycarbonylalkoxy, optionally substituted alkoxycarbonylalkyl, optionally substituted alkoxycarbonylalkenyl, optionally substituted alkoxycarbonylalkynyl, optionally substituted alkoxycarbonylalkoxy, optionally substituted carboxyalkoxy, optionally substituted carboxyalkyl, or optionally substituted carbamoylalkyl.
  • In particular embodiments, T1 is O (oxo), and T2 is S (thio) or Se (seleno). In other embodiments, T1 is S (thio), and T2 is O (oxo) or Se (seleno). In some embodiments, RVb′ is H, optionally substituted alkyl, or optionally substituted alkoxy.
  • In other embodiments, each R12a and R12b is, independently, H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, or optionally substituted hydroxyalkyl. In particular embodiments, R12a is H. In other embodiments, both R12a and R12b are H.
  • In some embodiments, each RVb′ of R12b is, independently, optionally substituted aminoalkyl (e.g., substituted with an N-protecting group, such as any described herein, e.g., trifluoroacetyl, or sulfoalkyl), optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, or optionally substituted acylaminoalkyl (e.g., substituted with an N-protecting group, such as any described herein, e.g., trifluoroacetyl). In some embodiments, the amino and/or alkyl of the optionally substituted aminoalkyl is substituted with one or more of optionally substituted alkyl, optionally substituted alkenyl, optionally substituted sulfoalkyl, optionally substituted carboxy (e.g., substituted with an O-protecting group), optionally substituted hydroxy (e.g., substituted with an O-protecting group), optionally substituted carboxyalkyl (e.g., substituted with an O-protecting group), optionally substituted alkoxycarbonylalkyl (e.g., substituted with an O-protecting group), or N-protecting group. In some embodiments, optionally substituted aminoalkyl is substituted with an optionally substituted sulfoalkyl or optionally substituted alkenyl. In particular embodiments, R12a and RVb″ are both H. In particular embodiments, T1 is O (oxo), and T2 is S (thio) or Se (seleno).
  • In some embodiments, RVb′ is optionally substituted alkoxycarbonylalkyl or optionally substituted carbamoylalkyl.
  • In particular embodiments, the optional substituent for R12a, R12b, R12c, or RVa is a polyethylene glycol group (e.g., —(CH2)s2(OCH2CH2)s1(CH2)s3OR′, wherein s1 is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R′ is H or C1-20 alkyl); or an amino-polyethylene glycol group (e.g., —NRN1(CH2)s2(CH2CH2O)s1(CH2)s3NRN1, wherein s1 is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and each RN1 is, independently, hydrogen or optionally substituted C1-6 alkyl).
  • In some embodiments, B is a modified cytosine. Exemplary modified cytosines include compounds of Formula (b10)-(b14):
  • Figure US20190085368A1-20190321-C00103
  • or a pharmaceutically acceptable salt or stereoisomer thereof, wherein each of T3′ and T3″ is, independently, H, optionally substituted alkyl, optionally substituted alkoxy, or optionally substituted thioalkoxy, or the combination of T3′ and T3″ join together (e.g., as in T3) to form O (oxo), S (thio), or Se (seleno); each V4 is, independently, O, S, N(RVc)nv, or C(RVc)nv, wherein nv is an integer from 0 to 2 and each RVc is, independently, H, halo, optionally substituted amino acid, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted heterocyclyl, optionally substituted alkheterocyclyl, or optionally substituted alkynyloxy (e.g., optionally substituted with any substituent described herein, such as those selected from (1)-(21) for alkyl), wherein the combination of R13b and RVc can be taken together to form optionally substituted heterocyclyl;
  • each V5 is, independently, N(RVd)nv, or C(RVd)nv, wherein nv is an integer from 0 to 2 and each RVd is, independently, H, halo, optionally substituted amino acid, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted heterocyclyl, optionally substituted alkheterocyclyl, or optionally substituted alkynyloxy (e.g., optionally substituted with any substituent described herein, such as those selected from (1)-(21) for alkyl) (e.g., V5 is —CH or N);
  • each of R13a and R13b is, independently, H, optionally substituted acyl, optionally substituted acyloxyalkyl, optionally substituted alkyl, or optionally substituted alkoxy, wherein the combination of R13b and R14 can be taken together to form optionally substituted heterocyclyl;
  • each R14 is, independently, H, halo, hydroxy, thiol, optionally substituted acyl, optionally substituted amino acid, optionally substituted alkyl, optionally substituted haloalkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted hydroxyalkyl (e.g., substituted with an O-protecting group), optionally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, optionally substituted acyloxyalkyl, optionally substituted amino (e.g., —NHR, wherein R is H, alkyl, aryl, or phosphoryl), azido, optionally substituted aryl, optionally substituted heterocyclyl, optionally substituted alkheterocyclyl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, or optionally substituted aminoalkyl; and
  • each of R15 and R16 is, independently, H, optionally substituted alkyl, optionally substituted alkenyl, or optionally substituted alkynyl.
  • Further exemplary modified cytosines include those having Formula (b32)-(b35):
  • Figure US20190085368A1-20190321-C00104
  • or a pharmaceutically acceptable salt or stereoisomer thereof, wherein each of T1 and T3 is, independently, O (oxo), S (thio), or Se (seleno); each of R13a and R13b is, independently, H, optionally substituted acyl, optionally substituted acyloxyalkyl, optionally substituted alkyl, or optionally substituted alkoxy, wherein the combination of R13b and R14 can be taken together to form optionally substituted heterocyclyl;
  • each R14 is, independently, H, halo, hydroxy, thiol, optionally substituted acyl, optionally substituted amino acid, optionally substituted alkyl, optionally substituted haloalkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted hydroxyalkyl (e.g., substituted with an O-protecting group), optionally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, optionally substituted acyloxyalkyl, optionally substituted amino (e.g., —NHR, wherein R is H, alkyl, aryl, or phosphoryl), azido, optionally substituted aryl, optionally substituted heterocyclyl, optionally substituted alkheterocyclyl, optionally substituted aminoalkyl (e.g., hydroxyalkyl, alkyl, alkenyl, or alkynyl), optionally substituted aminoalkenyl, or optionally substituted aminoalkynyl; and
  • each of R15 and R16 is, independently, H, optionally substituted alkyl, optionally substituted alkenyl, or optionally substituted alkynyl (e.g., R15 is H, and R16 is H or optionally substituted alkyl).
  • In some embodiments, R15 is H, and R16 is H or optionally substituted alkyl. In particular embodiments, R14 is H, acyl, or hydroxyalkyl. In some embodiments, R14 is halo. In some embodiments, both R14 and R15 are H. In some embodiments, both R15 and R16 are H. In some embodiments, each of R14 and R15 and R16 is H. In further embodiments, each of R13a and R13b is independently, H or optionally substituted alkyl.
  • Further non-limiting examples of modified cytosines include compounds of Formula (b36):
  • Figure US20190085368A1-20190321-C00105
  • or a pharmaceutically acceptable salt or stereoisomer thereof, wherein each R13b is, independently, H, optionally substituted acyl, optionally substituted acyloxyalkyl, optionally substituted alkyl, or optionally substituted alkoxy, wherein the combination of R13b and R14b can be taken together to form optionally substituted heterocyclyl; each R14a and R14b is, independently, H, halo, hydroxy, thiol, optionally substituted acyl, optionally substituted amino acid, optionally substituted alkyl, optionally substituted haloalkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted hydroxyalkyl (e.g., substituted with an O-protecting group), optionally substituted hydroxyalkenyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, optionally substituted acyloxyalkyl, optionally substituted amino (e.g., —NHR, wherein R is H, alkyl, aryl, phosphoryl, optionally substituted aminoalkyl, or optionally substituted carboxyaminoalkyl), azido, optionally substituted aryl, optionally substituted heterocyclyl, optionally substituted alkheterocyclyl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, or optionally substituted aminoalkynyl; and
  • each of R15 is, independently, H, optionally substituted alkyl, optionally substituted alkenyl, or optionally substituted alkynyl.
  • In particular embodiments, R14b is an optionally substituted amino acid (e.g., optionally substituted lysine). In some embodiments, R14a is H.
  • In some embodiments, B is a modified guanine. Exemplary modified guanines include compounds of Formula (b15)-(b17):
  • Figure US20190085368A1-20190321-C00106
  • or a pharmaceutically acceptable salt or stereoisomer thereof,
  • wherein
  • each of T4′, T4″, T5′, T5″, T6′, and T6″ is, independently, H, optionally substituted alkyl, or optionally substituted alkoxy, and wherein the combination of T4′ and T4″ (e.g., as in T4) or the combination of T5′ and T5″ (e.g., as in T5) or the combination of T6′ and T6″ (e.g., as in T6) join together form O (oxo), S (thio), or Se (seleno);
  • each of V5 and V6 is, independently, O, S, N(RVd)nv, or C(RVd)nv, wherein nv is an integer from 0 to 2 and each RVd is, independently, H, halo, thiol, optionally substituted amino acid, cyano, amidine, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted alkoxy, optionally substituted alkenyloxy, or optionally substituted alkynyloxy (e.g., optionally substituted with any substituent described herein, such as those selected from (1)-(21) for alkyl), optionally substituted thioalkoxy, or optionally substituted amino; and
  • each of R17, R18, R19a, R19b, R21, R22, R23, and R24 is, independently, H, halo, thiol, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted thioalkoxy, optionally substituted amino, or optionally substituted amino acid.
  • Exemplary modified guanosines include compounds of Formula (b37)-(b40):
  • Figure US20190085368A1-20190321-C00107
  • or a pharmaceutically acceptable salt or stereoisomer thereof, wherein each of T4′ is, independently, H, optionally substituted alkyl, or optionally substituted alkoxy, and each T4 is, independently, O (oxo), S (thio), or Se (seleno); each of R18, R19a, R19b, and R21 is, independently, H, halo, thiol, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted thioalkoxy, optionally substituted amino, or optionally substituted amino acid.
  • In some embodiments, R18 is H or optionally substituted alkyl. In further embodiments, T4 is oxo. In some embodiments, each of R19a and R19b is, independently, H or optionally substituted alkyl.
  • In some embodiments, B is a modified adenine. Exemplary modified adenines include compounds of Formula (b18)-(b20):
  • Figure US20190085368A1-20190321-C00108
  • or a pharmaceutically acceptable salt or stereoisomer thereof, wherein each V7 is, independently, O, S, N(RVe)nv, or C(RVe)nv, wherein nv is an integer from 0 to 2 and each RVe is, independently, H, halo, optionally substituted amino acid, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted alkoxy, optionally substituted alkenyloxy, or optionally substituted alkynyloxy (e.g., optionally substituted with any substituent described herein, such as those selected from (1)-(21) for alkyl);
  • each R25 is, independently, H, halo, thiol, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted thioalkoxy, or optionally substituted amino;
  • each of R26a and R26b is, independently, H, optionally substituted acyl, optionally substituted amino acid, optionally substituted carbamoylalkyl, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted hydroxyalkyl, optionally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally substituted alkoxy, or polyethylene glycol group (e.g., —(CH2)s2(OCH2CH2)s1(CH2)s3OR′, wherein s1 is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R′ is H or C1-20 alkyl); or an amino-polyethylene glycol group (e.g., —NRN1(CH2)s2(CH2CH2O)s1(CH2)s3NRN1, wherein s1 is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and each RN1 is, independently, hydrogen or optionally substituted C1-6 alkyl);
  • each R27 is, independently, H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted alkoxy, optionally substituted thioalkoxy or optionally substituted amino;
  • each R28 is, independently, H, optionally substituted alkyl, optionally substituted alkenyl, or optionally substituted alkynyl; and
  • each R29 is, independently, H, optionally substituted acyl, optionally substituted amino acid, optionally substituted carbamoylalkyl, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted hydroxyalkyl, optionally substituted hydroxyalkenyl, optionally substituted alkoxy, or optionally substituted amino.
  • Exemplary modified adenines include compounds of Formula (b41)-(b43):
  • Figure US20190085368A1-20190321-C00109
  • or a pharmaceutically acceptable salt or stereoisomer thereof, wherein each R25 is, independently, H, halo, thiol, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted thioalkoxy, or optionally substituted amino; each of R26a and R26b is, independently, H, optionally substituted acyl, optionally substituted amino acid, optionally substituted carbamoylalkyl, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted hydroxyalkyl, optionally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally substituted alkoxy, or polyethylene glycol group (e.g., —(CH2)s2(OCH2CH2)s1(CH2)s3OR′, wherein s1 is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R′ is H or C1-20 alkyl); or an amino-polyethylene glycol group (e.g., —NRN1(CH2)s2(CH2CH2O)s1(CH2)s3NRN1, wherein s1 is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and each RN1 is, independently, hydrogen or optionally substituted C1-6 alkyl); and
  • each R27 is, independently, H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted alkoxy, optionally substituted thioalkoxy, or optionally substituted amino.
  • In some embodiments, R26a is H, and R26b is optionally substituted alkyl. In some embodiments, each of R26a and R26b is, independently, optionally substituted alkyl. In particular embodiments, R27 is optionally substituted alkyl, optionally substituted alkoxy, or optionally substituted thioalkoxy. In other embodiments, R25 is optionally substituted alkyl, optionally substituted alkoxy, or optionally substituted thioalkoxy.
  • In particular embodiments, the optional substituent for R26a, R26b, or R29 is a polyethylene glycol group (e.g., —(CH2)s2(OCH2CH2)s1(CH2)s3OR′, wherein s1 is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R′ is H or C1-20 alkyl); or an amino-polyethylene glycol group (e.g., —NRN1(CH2)s2(CH2CH2O)s1(CH2)s3NRN1, wherein s1 is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and each RN1 is, independently, hydrogen or optionally substituted C1-6 alkyl).
  • In some embodiments, B may have Formula (b21):
  • Figure US20190085368A1-20190321-C00110
  • wherein X12 is, independently, O, S, optionally substituted alkylene (e.g., methylene), or optionally substituted heteroalkylene, xa is an integer from 0 to 3, and R12a and T2 are as described herein.
  • In some embodiments, B may have Formula (b22):
  • Figure US20190085368A1-20190321-C00111
  • wherein R10′ is, independently, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted heterocyclyl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, optionally substituted alkoxy, optionally substituted alkoxycarbonylalkyl, optionally substituted alkoxycarbonylalkenyl, optionally substituted alkoxycarbonylalkynyl, optionally substituted alkoxycarbonylalkoxy, optionally substituted carboxyalkoxy, optionally substituted carboxyalkyl, or optionally substituted carbamoylalkyl, and R11, R12a, T1, and T2 are as described herein.
  • In some embodiments, B may have Formula (b23):
  • Figure US20190085368A1-20190321-C00112
  • wherein R10 is optionally substituted heterocyclyl (e.g., optionally substituted furyl, optionally substituted thienyl, or optionally substituted pyrrolyl), optionally substituted aryl (e.g., optionally substituted phenyl or optionally substituted naphthyl), or any substituent described herein (e.g., for R10); and wherein R11 (e.g., H or any substituent described herein), R12a (e.g., H or any substituent described herein), T1 (e.g., oxo or any substituent described herein), and T2 (e.g., oxo or any substituent described herein) are as described herein.
  • In some embodiments, B may have Formula (b24):
  • Figure US20190085368A1-20190321-C00113
  • wherein R14′ is, independently, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted heterocyclyl, optionally substituted alkaryl, optionally substituted alkheterocyclyl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, optionally substituted alkoxy, optionally substituted alkoxycarbonylalkenyl, optionally substituted alkoxycarbonylalkynyl, optionally substituted alkoxycarbonylalkyl, optionally substituted alkoxycarbonylalkoxy, optionally substituted carboxyalkoxy, optionally substituted carboxyalkyl, or optionally substituted carbamoylalkyl, and R13a, R13b, R15, and T3 are as described herein.
  • In some embodiments, B may have Formula (b25):
  • Figure US20190085368A1-20190321-C00114
  • wherein R14′ is optionally substituted heterocyclyl (e.g., optionally substituted furyl, optionally substituted thienyl, or optionally substituted pyrrolyl), optionally substituted aryl (e.g., optionally substituted phenyl or optionally substituted naphthyl), or any substituent described herein (e.g., for R14 or R14′); and wherein R13a (e.g., H or any substituent described herein), R13b (e.g., H or any substituent described herein), R15 (e.g., H or any substituent described herein), and T3 (e.g., oxo or any substituent described herein) are as described herein.
  • In some embodiments, B is a nucleobase selected from the group consisting of cytosine, guanine, adenine, and uracil. In some embodiments, B may be:
  • Figure US20190085368A1-20190321-C00115
  • In some embodiments, the modified nucleobase is a modified uracil. Exemplary nucleobases and nucleosides having a modified uracil include pseudouridine (w), pyridin-4-one ribonucleoside, 5-aza-uridine, 6-aza-uridine, 2-thio-5-aza-uridine, 2-thio-uridine (s2U), 4-thio-uridine (s4U), 4-thio-pseudouridine, 2-thio-pseudouridine, 5-hydroxy-uridine (ho5U), 5-aminoallyl-uridine, 5-halo-uridine (e.g., 5-iodo-uridine or 5-bromo-uridine), 3-methyl-uridine (m3U), 5-methoxy-uridine (mo5U), uridine 5-oxyacetic acid (cmo5U), uridine 5-oxyacetic acid methyl ester (mcmo5U), 5-carboxymethyl-uridine (cm5U), 1-carboxymethyl-pseudouridine, 5-carboxyhydroxymethyl-uridine (chm5U), 5-carboxyhydroxymethyl-uridine methyl ester (mchm5U), 5-methoxycarbonylmethyl-uridine (mcm5U), 5-methoxycarbonylmethyl-2-thio-uridine (mcm5s2U), 5-aminomethyl-2-thio-uridine (nm5s2U), 5-methylaminomethyl-uridine (mnm5U), 5-methylaminomethyl-2-thio-uridine (mnm5s2U), 5-methylaminomethyl-2-seleno-uridine (mnm5se2U), 5-carbamoylmethyl-uridine (ncm5U), 5-carboxymethylaminomethyl-uridine (cmnm5U), 5-carboxymethylaminomethyl-2-thio-uridine (cmnm5s2U), 5-propynyl-uridine, 1-propynyl-pseudouridine, 5-taurinomethyl-uridine (τm5U), 1-taurinomethyl-pseudouridine, 5-taurinomethyl-2-thio-uridine(τm5s2U), 1-taurinomethyl-4-thio-pseudouridine, 5-methyl-uridine (m5U, i.e., having the nucleobase deoxythymine), 1-methylpseudouridine (m1ψ), 5-methyl-2-thio-uridine (m5s2U), 1-methyl-4-thio-pseudouridine (m1s4ψ), 4-thio-1-methyl-pseudouridine, 3-methyl-pseudouridine (m3ψ), 2-thio-1-methyl-pseudouridine, 1-methyl-1-deaza-pseudouridine, 2-thio-1-methyl-1-deaza-pseudouridine, dihydrouridine (D), dihydropseudouridine, 5,6-dihydrouridine, 5-methyl-dihydrouridine (m5D), 2-thio-dihydrouridine, 2-thio-dihydropseudouridine, 2-methoxy-uridine, 2-methoxy-4-thio-uridine, 4-methoxy-pseudouridine, 4-methoxy-2-thio-pseudouridine, N1-methyl-pseudouridine (also known as 1-methylpseudouridine (m1ψ)), 3-(3-amino-3-carboxypropyl)uridine (acp3U), 1-methyl-3-(3-amino-3-carboxypropyl)pseudouridine (acp3 ψ), 5-(isopentenylaminomethyl)uridine (inm5U), 5-(isopentenylaminomethyl)-2-thio-uridine (inm5s2U), α-thio-uridine, 2′-O-methyl-uridine (Um), 5,2′-O-dimethyl-uridine (m5Um), 2′-O-methyl-pseudouridine (ψm), 2-thio-2′-O-methyl-uridine (s2Um), 5-methoxycarbonylmethyl-2′-O-methyl-uridine (mcm5Um), 5-carbamoylmethyl-2′-O-methyl-uridine (ncm5Um), 5-carboxymethylaminomethyl-2′-O-methyl-uridine (cmnm5Um), 3,2′-O-dimethyl-uridine (m3Um), 5-(isopentenylaminomethyl)-2′-O-methyl-uridine (inm5Um), 1-thio-uridine, deoxythymidine, 2′-F-ara-uridine, 2′-F-uridine, 2′-OH-ara-uridine, 5-(2-carbomethoxyvinyl) uridine, and 5-[3-(1-E-propenylamino)uridine.
  • In some embodiments, the modified nucleobase is a modified cytosine. Exemplary nucleobases and nucleosides having a modified cytosine include 5-aza-cytidine, 6-aza-cytidine, pseudoisocytidine, 3-methyl-cytidine (m3C), N4-acetyl-cytidine (ac4C), 5-formyl-cytidine (f5C), N4-methyl-cytidine (m4C), 5-methyl-cytidine (m5C), 5-halo-cytidine (e.g., 5-iodo-cytidine), 5-hydroxymethyl-cytidine (hm5C), 1-methyl-pseudoisocytidine, pyrrolo-cytidine, pyrrolo-pseudoisocytidine, 2-thio-cytidine (s2C), 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, 4-methoxy-1-methyl-pseudoisocytidine, lysidine (k2C), α-thio-cytidine, 2′-O-methyl-cytidine (Cm), 5,2′-O-dimethyl-cytidine (m5Cm), N4-acetyl-2′-O-methyl-cytidine (ac4Cm), N4,2′-O-dimethyl-cytidine (m4Cm), 5-formyl-2′-O-methyl-cytidine (f5Cm), N4,N4,2′-O-trimethyl-cytidine (m42Cm), 1-thio-cytidine, 2′-F-ara-cytidine, 2′-F-cytidine, and 2′-OH-ara-cytidine.
  • In some embodiments, the modified nucleobase is a modified adenine. Exemplary nucleobases and nucleosides having a modified adenine include 2-amino-purine, 2, 6-diaminopurine, 2-amino-6-halo-purine (e.g., 2-amino-6-chloro-purine), 6-halo-purine (e.g., 6-chloro-purine), 2-amino-6-methyl-purine, 8-azido-adenosine, 7-deaza-adenine, 7-deaza-8-aza-adenine, 7-deaza-2-amino-purine, 7-deaza-8-aza-2-amino-purine, 7-deaza-2,6-diaminopurine, 7-deaza-8-aza-2,6-diaminopurine, 1-methyl-adenosine (m1A), 2-methyl-adenine (m2A), N6-methyl-adenosine (m6A), 2-methylthio-N6-methyl-adenosine (ms2m6A), N6-isopentenyl-adenosine (i6A), 2-methylthio-N6-isopentenyl-adenosine (ms2i6A), N6-(cis-hydroxyisopentenyl)adenosine (io6A), 2-methylthio-N6-(cis-hydroxyisopentenyl)adenosine (ms2io6A), N6-glycinylcarbamoyl-adenosine (g6A), N6-threonylcarbamoyl-adenosine (t6A), N6-methyl-N6-threonylcarbamoyl-adenosine (m6t6A), 2-methylthio-N6-threonylcarbamoyl-adenosine (ms2g6A), N6,N6-dimethyl-adenosine (m62A), N6-hydroxynorvalylcarbamoyl-adenosine (hn6A), 2-methylthio-N6-hydroxynorvalylcarbamoyl-adenosine (ms2hn6A), N6-acetyl-adenosine (ac6A), 7-methyl-adenine, 2-methylthio-adenine, 2-methoxy-adenine, α-thio-adenosine, 2′-O-methyl-adenosine (Am), N6,2′-O-dimethyl-adenosine (m6Am), N6,N6,2′-O-trimethyl-adenosine (m62Am), 1,2′-O-dimethyl-adenosine (m1Am), 2′-O-ribosyladenosine (phosphate) (Ar(p)), 2-amino-N6-methyl-purine, 1-thio-adenosine, 8-azido-adenosine, 2′-F-ara-adenosine, 2′-F-adenosine, 2′-OH-ara-adenosine, and N6-(19-amino-pentaoxanonadecyl)-adenosine.
  • In some embodiments, the modified nucleobase is a modified guanine. Exemplary nucleobases and nucleosides having a modified guanine include inosine (I), 1-methyl-inosine (m1I), wyosine (imG), methylwyosine (mimG), 4-demethyl-wyosine (imG-14), isowyosine (imG2), wybutosine (yW), peroxywybutosine (o2yW), hydroxywybutosine (OHyW), undermodified hydroxywybutosine (OHyW*), 7-deaza-guanosine, queuosine (Q), epoxyqueuosine (oQ), galactosyl-queuosine (galQ), mannosyl-queuosine (manQ), 7-cyano-7-deaza-guanosine (preQ0), 7-aminomethyl-7-deaza-guanosine (preQ1), archaeosine (G+), 7-deaza-8-aza-guanosine, 6-thio-guanosine, 6-thio-7-deaza-guanosine, 6-thio-7-deaza-8-aza-guanosine, 7-methyl-guanosine (m7G), 6-thio-7-methyl-guanosine, 7-methyl-inosine, 6-methoxy-guanosine, 1-methyl-guanosine (m1G), N2-methyl-guanosine (m2G), N2,N2-dimethyl-guanosine (m22G), N2,7-dimethyl-guanosine (m2,7G), N2, N2,7-dimethyl-guanosine (m2,2,7G), 8-oxo-guanosine, 7-methyl-8-oxo-guanosine, 1-methyl-6-thio-guanosine, N2-methyl-6-thio-guanosine, N2,N2-dimethyl-6-thio-guanosine, α-thio-guanosine, 2′-O-methyl-guanosine (Gm), N2-methyl-2′-O-methyl-guanosine (m2Gm), N2,N2-dimethyl-2′-O-methyl-guanosine (m22Gm), 1-methyl-2′-O-methyl-guanosine (m1Gm), N2,7-dimethyl-2′-O-methyl-guanosine (m2,7Gm), 2′-O-methyl-inosine (Im), 1,2′-O-dimethyl-inosine (m1Im), and 2′-O-ribosylguanosine (phosphate) (Gr(p)).
  • The nucleobase of the nucleotide can be independently selected from a purine, a pyrimidine, a purine or pyrimidine analog. For example, the nucleobase can each be independently selected from adenine, cytosine, guanine, uracil, or hypoxanthine. In another embodiment, the nucleobase can also include, for example, naturally-occurring and synthetic derivatives of a base, including pyrazolo[3,4-d]pyrimidines, 5-methylcytosine (5-me-C), 5-hydroxymethyl cytosine, xanthine, hypoxanthine, 2-aminoadenine, 6-methyl and other alkyl derivatives of adenine and guanine, 2-propyl and other alkyl derivatives of adenine and guanine, 2-thiouracil, 2-thiothymine and 2-thiocytosine, 5-propynyl uracil and cytosine, 6-azo uracil, cytosine and thymine, 5-uracil (pseudouracil), 4-thiouracil, 8-halo (e.g., 8-bromo), 8-amino, 8-thiol, 8-thioalkyl, 8-hydroxyl and other 8-substituted adenines and guanines, 5-halo particularly 5-bromo, 5-trifluoromethyl and other 5-substituted uracils and cytosines, 7-methylguanine and 7-methyladenine, 8-azaguanine and 8-azaadenine, deazaguanine, 7-deazaguanine, 3-deazaguanine, deazaadenine, 7-deazaadenine, 3-deazaadenine, pyrazolo[3,4-d]pyrimidine, imidazo[1,5-a]1,3,5 triazinones, 9-deazapurines, imidazo[4,5-d]pyrazines, thiazolo[4,5-d]pyrimidines, pyrazin-2-ones, 1,2,4-triazine, pyridazine; and 1,3,5 triazine. When the nucleotides are depicted using the shorthand A, G, C, T or U, each letter refers to the representative base and/or derivatives thereof, e.g., A includes adenine or adenine analogs, e.g., 7-deaza adenine).
  • Modifications on the Internucleoside Linkage
  • The modified nucleotides, which may be incorporated into a polynucleotide, primary construct, or mmRNA molecule, can be modified on the internucleoside linkage (e.g., phosphate backbone). Herein, in the context of the polynucleotide backbone, the phrases “phosphate” and “phosphodiester” are used interchangeably. Backbone phosphate groups can be modified by replacing one or more of the oxygen atoms with a different substituent. Further, the modified nucleosides and nucleotides can include the wholesale replacement of an unmodified phosphate moiety with another internucleoside linkage as described herein. Examples of modified phosphate groups include, but are not limited to, phosphorothioate, phosphoroselenates, boranophosphates, boranophosphate esters, hydrogen phosphonates, phosphoramidates, phosphorodiamidates, alkyl or aryl phosphonates, and phosphotriesters. Phosphorodithioates have both non-linking oxygens replaced by sulfur. The phosphate linker can also be modified by the replacement of a linking oxygen with nitrogen (bridged phosphoramidates), sulfur (bridged phosphorothioates), and carbon (bridged methylene-phosphonates).
  • The α-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 polynucleotides, primary constructs, or mmRNA molecules are expected to also reduce the innate immune response through weaker binding/activation of cellular innate immune molecules.
  • In specific embodiments, a modified nucleoside includes an alpha-thio-nucleoside (e.g., 5′-O-(1-thiophosphate)-adenosine, 5′-O-(1-thiophosphate)-cytidine (α-thio-cytidine), 5′-O-(1-thiophosphate)-guanosine, 5′-O-(1-thiophosphate)-uridine, or 5′-O-(1-thiophosphate)-pseudouridine).
  • Other internucleoside linkages that may be employed according to the present invention, including internucleoside linkages which do not contain a phosphorous atom, are described herein below.
  • Combinations of Modified Sugars, Nucleobases, and Internucleoside Linkages
  • The polynucleotides, primary constructs, and mmRNA of the invention can include a combination of modifications to the sugar, the nucleobase, and/or the internucleoside linkage. These combinations can include any one or more modifications described herein. For examples, any of the nucleotides described herein in Formulas (Ia), (Ia-1)-(Ia-3), (Ib)-(If), (IIa)-(IIp), (IIb-1), (IIb-2), (IIc-1)-(IIc-2), (IIn-2), (IVa)-(IVl), and (IXa)-(IXr) can be combined with any of the nucleobases described herein (e.g., in Formulas (b1)-(b43) or any other described herein).
  • Cytotoxic Nucleosides
  • In one embodiment, the polynucleotides, primary constructs or mmRNA of the present invention may incorporate one or more cytotoxic nucleosides. For example, cytotoxic nucleosides may be incorporated into polynucleotides, primary constructs or mmRNA such as bifunctional modified RNAs or mRNAs. Cytotoxic nucleoside anti-cancer agents include, but are not limited to, adenosine arabinoside, cytarabine, cytosine arabinoside, 5-fluorouracil, fludarabine, floxuridine, FTORAFUR® (a combination of tegafur and uracil), tegafur ((RS)-5-fluoro-1-(tetrahydrofuran-2-yl)pyrimidine-2,4(1H,3H)-dione), and 6-mercaptopurine.
  • A number of cytotoxic nucleoside analogues are in clinical use, or have been the subject of clinical trials, as anticancer agents. Examples of such analogues include, but are not limited to, cytarabine, gemcitabine, troxacitabine, decitabine, tezacitabine, 2′-deoxy-2′-methylidenecytidine (DMDC), cladribine, clofarabine, 5-azacytidine, 4′-thio-aracytidine, cyclopentenylcytosine and 1-(2-C-cyano-2-deoxy-beta-D-arabino-pentofuranosyl)-cytosine. Another example of such a compound is fludarabine phosphate. These compounds may be administered systemically and may have side effects which are typical of cytotoxic agents such as, but not limited to, little or no specificity for tumor cells over proliferating normal cells.
  • A number of prodrugs of cytotoxic nucleoside analogues are also reported in the art. Examples include, but are not limited to, N4-behenoyl-1-beta-D-arabinofuranosylcytosine, N4-octadecyl-1-beta-D-arabinofuranosylcytosine, N4-palmitoyl-1-(2-C-cyano-2-deoxy-beta-D-arabino-pentofuranosyl) cytosine, and P-4055 (cytarabine 5′-elaidic acid ester). In general, these prodrugs may be converted into the active drugs mainly in the liver and systemic circulation and display little or no selective release of active drug in the tumor tissue. For example, capecitabine, a prodrug of 5′-deoxy-5-fluorocytidine (and eventually of 5-fluorouracil), is metabolized both in the liver and in the tumor tissue. A series of capecitabine analogues containing “an easily hydrolysable radical under physiological conditions” has been claimed by Fujiu et al. (U.S. Pat. No. 4,966,891) and is herein incorporated by reference. The series described by Fujiu includes N4 alkyl and aralkyl carbamates of 5′-deoxy-5-fluorocytidine and the implication that these compounds will be activated by hydrolysis under normal physiological conditions to provide 5′-deoxy-5-fluorocytidine.
  • A series of cytarabine N4-carbamates has been by reported by Fadl et al (Pharmazie. 1995, 50, 382-7, herein incorporated by reference) in which compounds were designed to convert into cytarabine in the liver and plasma. WO 2004/041203, herein incorporated by reference, discloses prodrugs of gemcitabine, where some of the prodrugs are N4-carbamates. These compounds were designed to overcome the gastrointestinal toxicity of gemcitabine and were intended to provide gemcitabine by hydrolytic release in the liver and plasma after absorption of the intact prodrug from the gastrointestinal tract. Nomura et al (Bioorg Med. Chem. 2003, 11, 2453-61, herein incorporated by reference) have described acetal derivatives of 1-(3-C-ethynyl-β-D-ribo-pentofaranosyl) cytosine which, on bioreduction, produced an intermediate that required further hydrolysis under acidic conditions to produce a cytotoxic nucleoside compound.
  • Cytotoxic nucleotides which may be chemotherapeutic also include, but are not limited to, pyrazolo [3,4-D]-pyrimidines, allopurinol, azathioprine, capecitabine, cytosine arabinoside, fluorouracil, mercaptopurine, 6-thioguanine, acyclovir, ara-adenosine, ribavirin, 7-deaza-adenosine, 7-deaza-guanosine, 6-aza-uracil, 6-aza-cytidine, thymidine ribonucleotide, 5-bromodeoxyuridine, 2-chloro-purine, and inosine, or combinations thereof.
  • The The modified nucleosides and nucleotides used in the synthesis of polynucleotides, primary constructs, and mmRNA molecules disclosed herein can be prepared from readily available starting materials using the following general methods and procedures. Where typical or preferred process conditions (e.g., reaction temperatures, times, mole ratios of reactants, solvents, pressures, etc.) are provided, a skilled artisan would be able to optimize and develop additional process conditions. 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 polypeptides, primary constructs, and mmRNA molecules of the present invention 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 nucleosides and nucleotides (e.g., building block molecules) can be prepared according to the synthetic methods described in Ogata et al., J. Org. Chem. 74:2585-2588 (2009); Purmal et al., Nucl. Acids Res. 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.
  • The polypeptides, primary constructs, and mmRNA of the invention may or may not be uniformly modified along the entire length of the molecule. For example, one or more or all types of nucleotide (e.g., purine or pyrimidine, or any one or more or all of A, G, U, C) may or may not be uniformly modified in a polynucleotide of the invention, or in a given predetermined sequence region thereof (e.g. one or more of the sequence regions represented in FIG. 1). In some embodiments, all nucleotides X in a polynucleotide of the invention (or in a given sequence region thereof) are modified, wherein X may any one of nucleotides A, G, U, C, or any one of the combinations A+G, A+U, A+C, G+U, G+C, U+C, A+G+U, A+G+C, G+U+C or A+G+C.
  • Different sugar modifications, nucleotide modifications, and/or internucleoside linkages (e.g., backbone structures) may exist at various positions in the polynucleotide, primary construct, or mmRNA. 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 polynucleotide, primary construct, or mmRNA such that the function of the polynucleotide, primary construct, or mmRNA is not substantially decreased. A modification may also be a 5′ or 3′ terminal modification. The polynucleotide, primary construct, or mmRNA may contain from about 1% to about 100% modified nucleotides (either in relation to overall nucleotide content, or in relation to one or more types of nucleotide, i.e. any one or more of A, G, U or C) or any intervening percentage (e.g., from 1% to 20%, from 1% to 25%, from 1% to 50%, from 1% to 60%, from 1% to 70%, from 1% to 80%, from 1% to 90%, from 1% to 95%, from 10% to 20%, from 10% to 25%, from 10% to 50%, from 10% to 60%, from 10% to 70%, from 10% to 80%, from 10% to 90%, from 10% to 95%, from 10% to 100%, from 20% to 25%, from 20% to 50%, from 20% to 60%, from 20% to 70%, from 20% to 80%, from 20% to 90%, from 20% to 95%, from 20% to 100%, from 50% to 60%, from 50% to 70%, from 50% to 80%, from 50% to 90%, from 50% to 95%, from 50% to 100%, from 70% to 80%, from 70% to 90%, from 70% to 95%, from 70% to 100%, from 80% to 90%, from 80% to 95%, from 80% to 100%, from 90% to 95%, from 90% to 100%, and from 95% to 100%).
  • In some embodiments, the polynucleotide, primary construct, or mmRNA includes a modified pyrimidine (e.g., a modified uracil/uridine/U or modified cytosine/cytidine/C). In some embodiments, the uracil or uridine (generally: U) in the polynucleotide, primary construct, or mmRNA molecule may be replaced with from about 1% to about 100% of a modified uracil or modified uridine (e.g., from 1% to 20%, from 1% to 25%, from 1% to 50%, from 1% to 60%, from 1% to 70%, from 1% to 80%, from 1% to 90%, from 1% to 95%, from 10% to 20%, from 10% to 25%, from 10% to 50%, from 10% to 60%, from 10% to 70%, from 10% to 80%, from 10% to 90%, from 10% to 95%, from 10% to 100%, from 20% to 25%, from 20% to 50%, from 20% to 60%, from 20% to 70%, from 20% to 80%, from 20% to 90%, from 20% to 95%, from 20% to 100%, from 50% to 60%, from 50% to 70%, from 50% to 80%, from 50% to 90%, from 50% to 95%, from 50% to 100%, from 70% to 80%, from 70% to 90%, from 70% to 95%, from 70% to 100%, from 80% to 90%, from 80% to 95%, from 80% to 100%, from 90% to 95%, from 90% to 100%, and from 95% to 100% of a modified uracil or modified uridine). The modified uracil or uridine can be replaced by a compound having a single unique structure or by a plurality of compounds having different structures (e.g., 2, 3, 4 or more unique structures, as described herein). In some embodiments, the cytosine or cytidine (generally: C) in the polynucleotide, primary construct, or mmRNA molecule may be replaced with from about 1% to about 100% of a modified cytosine or modified cytidine (e.g., from 1% to 20%, from 1% to 25%, from 1% to 50%, from 1% to 60%, from 1% to 70%, from 1% to 80%, from 1% to 90%, from 1% to 95%, from 10% to 20%, from 10% to 25%, from 10% to 50%, from 10% to 60%, from 10% to 70%, from 10% to 80%, from 10% to 90%, from 10% to 95%, from 10% to 100%, from 20% to 25%, from 20% to 50%, from 20% to 60%, from 20% to 70%, from 20% to 80%, from 20% to 90%, from 20% to 95%, from 20% to 100%, from 50% to 60%, from 50% to 70%, from 50% to 80%, from 50% to 90%, from 50% to 95%, from 50% to 100%, from 70% to 80%, from 70% to 90%, from 70% to 95%, from 70% to 100%, from 80% to 90%, from 80% to 95%, from 80% to 100%, from 90% to 95%, from 90% to 100%, and from 95% to 100% of a modified cytosine or modified cytidine). The modified cytosine or cytidine can be replaced by a compound having a single unique structure or by a plurality of compounds having different structures (e.g., 2, 3, 4 or more unique structures, as described herein).
  • In some embodiments, the present disclosure provides methods of synthesizing a polynucleotide, primary construct, or mmRNA (e.g., the first region, first flanking region, or second flanking region) including n number of linked nucleosides having Formula (Ia-1):
  • Figure US20190085368A1-20190321-C00116
  • comprising:
  • a) reacting a nucleotide of Formula (IV-1):
  • Figure US20190085368A1-20190321-C00117
  • with a phosphoramidite compound of Formula (V-1):
  • Figure US20190085368A1-20190321-C00118
  • wherein Y9 is H, hydroxy, phosphoryl, pyrophosphate, sulfate, amino, thiol, optionally substituted amino acid, or a peptide (e.g., including from 2 to 12 amino acids); and each P1, P2, and P3 is, independently, a suitable protecting group; and
    Figure US20190085368A1-20190321-P00003
    denotes a solid support;
  • to provide a polynucleotide, primary construct, or mmRNA of Formula (VI-1):
  • Figure US20190085368A1-20190321-C00119
  • and
  • b) oxidizing or sulfurizing the polynucleotide, primary construct, or mmRNA of Formula (V) to yield a polynucleotide, primary construct, or mmRNA of Formula (VII-1):
  • Figure US20190085368A1-20190321-C00120
  • and
  • c) removing the protecting groups to yield the polynucleotide, primary construct, or mmRNA of Formula (Ia).
  • In some embodiments, steps a) and b) are repeated from 1 to about 10,000 times. In some embodiments, the methods further comprise a nucleotide (e.g., mmRNA molecule) selected from the group consisting of A, C, G and U adenosine, cytosine, guanosine, and uracil. In some embodiments, the nucleobase may be a pyrimidine or derivative thereof. In some embodiments, the polynucleotide, primary construct, or mmRNA is translatable.
  • Other components of polynucleotides, primary constructs, and mmRNA 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 nucleotide modifications. In such embodiments, nucleotide modifications may also be present in the translatable region. Also provided are polynucleotides, primary constructs, and mmRNA containing a Kozak sequence.
  • Combinations of Nucleotides in mmRNA
  • Further examples of modified nucleotides and modified nucleotide combinations are provided below in Table 9. These combinations of modified nucleotides can be used to form the polypeptides, primary constructs, or mmRNA of the invention. Unless otherwise noted, the modified nucleotides may be completely substituted for the natural nucleotides of the modified nucleic acids or mmRNA of the invention. As a non-limiting example, the natural nucleotide uridine may be substituted with a modified nucleoside described herein. In another non-limiting example, the natural nucleotide uridine may be partially substituted (e.g., about 0.1%, 1%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 99.9%) with at least one of the modified nucleoside disclosed herein.
  • TABLE 9
    Modified Nucleotide Modified Nucleotide Combination
    α-thio- α-thio-cytidine/5-iodo-uridine
    cytidine α-thio-cytidine/N1-methyl-pseudouridine
    α-thio-cytidine/α-thio-uridine
    α-thio-cytidine/5-methyl-uridine
    α-thio-cytidine/pseudo-uridine
    about 50% of the cytosines are α-thio-cytidine
    pseudo- pseudoisocytidine/5-iodo-uridine
    isocytidine pseudoisocytidine/N1-methyl-pseudouridine
    pseudoisocytidine/α-thio-uridine
    pseudoisocytidine/5-methyl-uridine
    pseudoisocytidine/pseudouridine
    about 25% of cytosines are pseudoisocytidine
    pseudoisocytidine/about 50% of uridines are N1-methyl-
    pseudouridine and about 50% of uridines are pseudouridine
    pseudoisocytidine/about 25% of uridines are N1-methyl-
    pseudouridine and about 25% of uridines are pseudouridine
    pyrrolo- pyrrolo-cytidine/5-iodo-uridine
    cytidine pyrrolo-cytidine/N1-methyl-pseudouridine
    pyrrolo-cytidine/α-thio-uridine
    pyrrolo-cytidine/5-methyl-uridine
    pyrrolo-cytidine/pseudouridine
    about 50% of the cytosines are pyrrolo-cytidine
    5-methyl- 5-methyl-cytidine/5-iodo-uridine
    cytidine 5-methyl-cytidine/N1-methyl-pseudouridine
    5-methyl-cytidine/α-thio-uridine
    5-methyl-cytidine/5-methyl-uridine
    5-methyl-cytidine/pseudouridine
    about 25% of cytosines are 5-methyl-cytidine
    about 50% of cytosines are 5-methyl-cytidine
    5-methyl-cytidine/5-methoxy-uridine
    5-methyl-cytidine/5-bromo-uridine
    5-methyl-cytidine/2-thio-uridine
    5-methyl-cytidine/about 50% of uridines are 2-thio-uridine
    about 50% of uridines are 5-methyl-cytidine/about 50% of
    uridines are 2-thio-uridine
    N4-acetyl- N4-acetyl-cytidine/5-iodo-uridine
    cytidine N4-acetyl-cytidine/N1-methyl-pseudouridine
    N4-acetyl-cytidine/α-thio-uridine
    N4-acetyl-cytidine/5-methyl-uridine
    N4-acetyl-cytidine/pseudouridine
    about 50% of cytosines are N4-acetyl-cytidine
    about 25% of cytosines are N4-acetyl-cytidine
    N4-acetyl-cytidine/5-methoxy-uridine
    N4-acetyl-cytidine/5-bromo-uridine
    N4-acetyl-cytidine/2-thio-uridine
    about 50% of cytosines are N4-acetyl-cytidine/about 50%
    of uridines are 2-thio-uridine
  • Further examples of modified nucleotide combinations are provided below in Table 10. These combinations of modified nucleotides can be used to form the polypeptides, primary constructs, or mmRNA of the invention.
  • TABLE 10
    Modified Nucleotide Modified Nucleotide Combination
    modified cytidine having modified cytidine with (b10)/pseudouridine
    one or more nucleobases modified cytidine with (b10)/N1-methyl-pseudouridine
    of Formula (b10) modified cytidine with (b10)/5-methoxy-uridine
    modified cytidine with (b10)/5-methyl-uridine
    modified cytidine with (b10)/5-bromo-uridine
    modified cytidine with (b10)/2-thio-uridine
    about 50% of cytidine substituted with modified cytidine (b10)/
    about 50% of uridines are 2-thio-uridine
    modified cytidine having modified cytidine with (b32)/pseudouridine
    one or more nucleobases modified cytidine with (b32)/N1-methyl-pseudouridine
    of Formula (b32) modified cytidine with (b32)/5-methoxy-uridine
    modified cytidine with (b32)/5-methyl-uridine
    modified cytidine with (b32)/5-bromo-uridine
    modified cytidine with (b32)/2-thio-uridine
    about 50% of cytidine substituted with modified cytidine (b32)/
    about 50% of uridines are 2-thio-uridine
    modified uridine having modified uridine with (b1)/N4-acetyl-cytidine
    one or more nucleobases modified uridine with (b1)/5-methyl-cytidine
    of Formula (b1)
    modified uridine having modified uridine with (b8)/N4-acetyl-cytidine
    one or more nucleobases modified uridine with (b8)/5-methyl-cytidine
    of Formula (b8)
    modified uridine having modified uridine with (b28)/N4-acetyl-cytidine
    one or more nucleobases modified uridine with (b28)/5-methyl-cytidine
    of Formula (b28)
    modified uridine having modified uridine with (b29)/N4-acetyl-cytidine
    one or more nucleobases modified uridine with (b29)/5-methyl-cytidine
    of Formula (b29)
    modified uridine having modified uridine with (b30)/N4-acetyl-cytidine
    one or more nucleobases modified uridine with (b30)/5-methyl-cytidine
    of Formula (b30)
  • In some embodiments, at least 25% of the cytosines are replaced by a compound of Formula (b10)-(b14) (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 (b1)-(b9) (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 are replaced by a compound of Formula (b10)-(b14), and at least 25% of the uracils are replaced by a compound of Formula (b1)-(b9) (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%).
  • EXAMPLES
  • Below are examples of specific embodiments for carrying out the present invention. The examples are offered for illustrative purposes only, and are not intended to limit the scope of the present invention in any way. Efforts have been made to ensure accuracy with respect to numbers used (e.g., amounts, temperatures, etc.), but some experimental error and deviation should, of course, be allowed for.
  • The practice of the present invention will employ, unless otherwise indicated, conventional methods of protein chemistry, biochemistry, recombinant DNA techniques and pharmacology, within the skill of the art. Such techniques are explained fully in the literature. See, e.g., T.E. Creighton, Proteins: Structures and Molecular Properties (W.H. Freeman and Company, 1993); A. L. Lehninger, Biochemistry (Worth Publishers, Inc., current addition); Sambrook, et al., Molecular Cloning: A Laboratory Manual (2nd Edition, 1989); Methods In Enzymology (S. Colowick and N. Kaplan eds., Academic Press, Inc.); Remington's Pharmaceutical Sciences, 18th Edition (Easton, Pa.: Mack Publishing Company, 1990); Carey and Sundberg Advanced Organic Chemistry 3rd Ed. (Plenum Press) Vols A and B (1992).
  • Example 1: DNA Template with GCSF Gene
  • The plasmid pJ344:91543-TC-GCSF includes the coding sequence for GCSF (granulyte colony stimulating factor) and 141 nucleotide sequence coding for Poly A Tail and an XBaI restriction endonuclease recognition site immediately downstream of the poly A tail sequence tract. A plasmid map is shown in FIG. 5, and a plasmid DNA sequence is shown in FIG. 6.
  • An E. coli strain DH10B harboring above plasmid was grown in 250 ml animal-free broth with ampicillin. This process generated 1107 μg of plasmid DNA. The plasmid DNA was isolated and purified according to DNA2.0 standard operating procedures. Plasmid DNA yield and DNA homogeneity was determined using agarose gel electrophoresis. The insert (but not vector backbone) was sequence verified in both orientations using a capillary electrophoresis DNA analyzer.
  • Example 2: Linearization of DNA Template
  • The plasmid DNA template was linearized using the restriction endonuclease XbaI. The XbaI restriction digest reaction conditions were as follows:
  • Water 25573.6 μL
    Plasmid DNA (1145 μg/μL  1879.9 μL
    BSA (100X)  430.5 μL
    Buffer 4 (10X)  4305.0 μL
    XbaI (20000 U/ml    861 μL
    Total vol   43050 μL
  • The reaction proceeded at 37° C. overnight. The linearized plasmid was diafiltered into 10 mM Tris HCl pH 7.5 and concentrated to 959 ng/uL prior to use using 100 kDa MWCO Amicon spin filters (EMD Millipore).
  • Example 3: In Vitro Transcription
  • The non-amplified, linearized DNA plasmid was used as a template for in vitro transcription. A 1 mL transcription reaction was performed utilizing 250 μg of plasmid template. Nucleotides ATP, GTP, 5mCTP (5-methyl cytosine triphosphate) and N-1-methylpseudouridine triphosphate were added at 7.5 mM each. RNase inhibitor (1000 U), inorganic pyrophosphatase (1U) and T7 RNA polymerase (7000U) were added. The final buffer conditions (1×) were as follows: 40 mM Tris HCl pH8, 40 mM magnesium acetate, 5 mM dithiothreitol (DTT), and 1 mM spermidine.
  • The in vitro transcription reaction proceeded for 4 hours at 37° C. under constant mixing. The total reaction yield was 5.4 mg RNA transcript. A 400 uL (40%) portion of the reaction was diafiltered into water and concentrated to 622 ng/μL using 100 kDa MWCO Amicon spin filters (EMD Millipore).
  • The results demonstrate successful, milligram scale production of RNA transcript with in vitro transcription using a non-PCT amplified, linearized plasmid DNA template.
  • Example 4: Oligo dT Removal of DNA Template
  • The full length poly A containing RNA transcript was purified from truncated RNA, DNA template, and residual enzymes using oligo dT chromatography. A 20 mer polythymidine Sepharose (3 ml) was packed in a 5 mL SPE column on a solid phase extraction vacuum manifold. The RNA transcript (2.18 mg) was applied to column, followed by washing and elution. The oligo dT purified RNA transcript was diafiltered into water and concentrated to 1.22 mg/mL using 100 kDa MWCO Amicon spin filters (EMD Millipore). Approximately 1.82 mg of 2.18 mg was recovered (83%) as determined by Bioanalyzer gel elctrophoresis.
  • Example 5: Characterization of Uncapped RNA Transcript, Pre and Post Oligo dT
  • The RNA transcript was analyzed using a Bioanalyzer gel electrohphoresis. Sample taken both before and after oligo dT purification were analyzed. The results are shown in the electopherograms in FIGS. 7A and 7B.
  • The results demonstrate recovery of >80% RNA transcript using oligo dT purification and virtual elimination of impurities.
  • Example 6: Capping of RNA Transcript
  • A 5′ cap was enzymatically added to the RNA transcript. Approximately 0.98 mg of oligo dT purified RNA transcript (1225 ng/uL) was capped to obtain a 5′ Cap 1 structure. GTP was added at a final reaction concentration of 0.9 mM. SAM (S-adenosyl-L-methionine) was added at a final reaction concentration of 0.4 mM. 907 units of RNase inhibitor, 3630 units of 2′O-methyltransferase, and 363 units of vaccinia guanylyltransferase were added to the reaction. The final 1× buffer conditions consist of the following: 50 mM Tris HCl pH 8, 5 mM KCl, 1 mM MgCl2, and 1 mM dithiothreitol. Final reaction volume was 1070 μL. The reaction proceeded at 37° C. for 2 hours under constant mixing. Reaction setup for capped RNA (lot 12-04-155-C) is shown below.
  • The projected sequence of the RNA transcript is below:
  • mRNA Sequence: 12-04-101-I and 12-04-111-I
    (Capped: 12-04-154-C and 12-04-155-C)
    Parent Gene ID 91543 and 103294
    5′-G*GGGAAAUAAG AGAGAAAAGA AGAGUAAGAA
    GAAAUAUAAG AGCCACCAUG GCCGGUCCCG CGACCCAAAG
    CCCAUGAAA CUUAUGGCCC UGCAGUUGCU GCUUUGGCAC
    UCGGCCCUCU GGACAGUCCA AGAAGCGACU CCUCUCGGAC
    CUGCCUCAUC GUUGCCGCAG UCAUUCCUUU UGAAGUGUCU
    GGAGCAGGUG CGAAAGAUUC AGGGCGAUGG AGCCGCACUC
    CAAGAGAAGC UCUGCGCGAC AUACAAACUU UGCCAUCCCG
    AGGAGCUCGU ACUGCUCGGG CACAGCUUGG GGAUUCCCUG
    GGCUCCUCUC UCGUCCUGUC CGUCGCAGGC UUUGCAGUUG
    GCAGGGUGCC UUUCCCAGCU CCACUCCGGU UUGUUCUUGU
    AUCAGGGACU GCUGCAAGCC CUUGAGGGAA UCUCGCCAGA
    AUUGGGCCCG ACGCUGGACA CGUUGCAGCU CGACGUGGCG
    GAUUUCGCAA CAACCAUCUG GCAGCAGAUG GAGGAACUGG
    GGAUGGCACC CGCGCUGCAG CCCACGCAGG GGGCAAUGCC
    GGCCUUUGCG UCCGCGUUUC AGCGCAGGGC GGGUGGAGUC
    CUCGUAGCGA GCCACCUUCA AUCAUUUUUG GAAGUCUCGU
    ACCGGGUGCU GAGACAUCUU GCGCAGCCGU GAUAAGCUGC
    CUUCUGCGGG GCUUGCCUUC UGGCCAUGCC CUUCUUCUCU
    CCCUUGCACC UGUACCUCUU GGUCUUUGAA UAAAGCCUGA
    GUAGGAAGGC GGCCGCUCGA GCAUGCAAAA AAAAAAAAAA
    AAAAAAAAAA AAAAAAAAAA AAAAAAAAAA AAAAAAAAAA
    AAAAAAAAAA AAAAAAAAAA AAAAAAAAAA AAAAAAAAAA
    AAAAAAAAAA AAAAAAAAAA AAAAAAAAAA AAAAAAAAAA
    AAAAAAAUCU AG
     3′
    G* = cap
  • Example 7: Oligo dT Purification of Capped RNA Transcript
  • The capped RNA transcript was purified using 2 mL of 20mer polythymidine Sepharose packed in a 5 mL SPE column on a solid phase extraction vacuum manifold. 0.82 mg was recovered in the elution (84% recovery) The RNA was diafiltered into water and harvested at a concentration of 366 ng/uL using 100 kDa MWCO Amicon spin filters (EMD Millipore). The results are shown in FIG. 8.
  • Example 8: Protein Expression Using Capped RNA Transcript Produced by the Method of the Invention
  • Assessment of protein expression was conducted via forward transfection of RNA into HeLa cells. GCSF ELISA was used to quantify expressed protein ELISA data; data are shown in FIG. 9.
  • 12-04-155-C produced using a linearized Poly A:T tract DNA template and enzymatic capping yielded ˜1,150 ng/mL GCSF expression, significantly higher than the four lots that utilized PCR product as a DNA template: 12-04-89-C, 12-04-106-C, 12-04-114-C, and 12-04-122-C.
  • Example 9: Interferon-a Induction Assessment Using Capped RNA Transcript Produced by the Method of the Invention
  • RNAs were transfected into Peripheral blood mononuclear cells (PBMC) to assay in vitro cytokine induction. The results are shown in FIG. 10. The RNA transcript 12-04-155-C produced using a linearized Poly A:T tract DNA template and enzymatic capping resulted in undetectable levels of interferon-α as determined by IFN-α ELISA. In contrast, two of four samples that utilized PCR product as a DNA template: 12-04-89-C, 12-04-106-C showed some level of interferon-α induction. Poly IC (Polyinosinic:polycytidylic acid) is an immune stimulatory positive control and R-848 is an imidazoquinoline compound that is also used as a positive control.
  • Example 10: DNA Template with Factor IX Gene
  • The plasmid pJ204109475 includes the coding sequence for Factor IX and 141 nucleotide sequence coding for Poly A Tail and an SapI restriction endonuclease recognition site immediately downstream of the poly A tail sequence tract.
  • A plasmid map and DNA sequence are shown in FIGS. 11A and 11B.
  • While the invention has been particularly shown and described with reference to a preferred embodiment and various alternate embodiments, it will be understood by persons skilled in the relevant art that various changes in form and details can be made therein without departing from the spirit and scope of the invention.
  • All references, issued patents and patent applications cited within the body of the instant specification are hereby incorporated by reference in their entirety, for all purposes.

Claims (18)

1. A method for producing a purified composition comprising a capped RNA transcript for a gene of interest, the method comprising:
(a) providing a sample comprising a linear, non-amplified DNA template, the DNA template comprising an RNA polymerase promoter sequence operatively linked to a target sequence coding for the gene of interest, a poly A tail sequence, an endonuclease recognition site sequence immediately downstream of the poly A tail sequence, and optionally a 5′ untranslated region (UTR) and/or a 3′ UTR;
(b) contacting the sample with a RNA polymerase and ribonucleotides
under conditions sufficient for vitro transcription to produce a first composition comprising an uncapped RNA transcript, wherein at least 80% of the RNA transcript is full-length uncapped RNA transcript;
(c) purifying the uncapped RNA transcript using oligo dT affinity purification;
(d) capping the uncapped RNA transcript to produce a second composition comprising a capped RNA transcript; and
(e) purifying the capped RNA transcript from the second composition by anion exchange chromatography,
thereby producing the purified composition comprising a capped RNA transcript.
2. The method of claim 1, wherein at least 95% of the uncapped RNA transcript in the first composition is full-length uncapped RNA transcript.
3. The method of claim 1, wherein the percent full length uncapped RNA transcript is determined using reverse phase HPLC and measured by peak area of full length relative to total peak area.
4. The method of claim 1, wherein the method does not comprising treating the composition with DNase.
5. (canceled)
6. The method of claim 1, wherein the method does not comprise a polymerase chain reaction (PCR) amplification step.
7. The method of claim 1, wherein the DNA template comprises 500-25000 or 3000-7000 basepairs.
8. (canceled)
9. The method of claim 1, wherein the poly A tail is 5-300 nucleotides or 60-160 nucleotides in length
10. The method of claim 1, wherein the endonuclease recognition site sequence is recognized by XbaI or SapI.
11. The method of claim 1, wherein the method further comprises producing the DNA template, wherein producing the DNA template comprises contacting a circular plasmid DNA template with an endonuclease that recognizes the endonuclease recognition site sequence.
12. The method of claim 11, wherein the method further comprises producing the circular plasmid DNA template at a microgram scale or a milligram scale or a gram scale.
13. The method of claim 1, wherein the RNA polymerase is a T7 polymerase.
14. The method of claim 1, wherein at least one nucleotide is a modified nucleotide.
15. The method of claim 1, wherein capping the uncapped RNA transcript comprises contacting the RNA transcript with Vaccinia guanylyltransferase, s-adenosyl-L-methionine (SAM), and guanosine triphosphate (GTP) in a reaction and maintaining the reaction under conditions sufficient to cap the RNA transcript, wherein the RNA transcript is capped with a Cap0 structure.
16. The method of claim 15, wherein the reaction further comprises 2′-O-methyltransferase wherein the RNA transcript is capped with a Cap1 structure.
17-22. (canceled)
23. The method of claim 1, wherein the DNA template comprises a poly A tail sequence of 60-160 nucleotides; the RNA polymerase is a T7 RNA polymerase; and capping the uncapped RNA transcript comprises contacting the uncapped RNA transcript with guanylyltransferase, s-adenosyl-L-methionine (SAM), guanosine triphosphate (GTP), and optionally 2′-O-methyltransferase.
US16/144,282 2013-03-15 2018-09-27 Manufacturing methods for production of rna transcripts Abandoned US20190085368A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US16/144,282 US20190085368A1 (en) 2013-03-15 2018-09-27 Manufacturing methods for production of rna transcripts

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201361800049P 2013-03-15 2013-03-15
PCT/US2014/026835 WO2014152027A1 (en) 2013-03-15 2014-03-13 Manufacturing methods for production of rna transcripts
US201514777190A 2015-09-15 2015-09-15
US16/144,282 US20190085368A1 (en) 2013-03-15 2018-09-27 Manufacturing methods for production of rna transcripts

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
US14/777,190 Continuation US10138507B2 (en) 2013-03-15 2014-03-13 Manufacturing methods for production of RNA transcripts
PCT/US2014/026835 Continuation WO2014152027A1 (en) 2013-03-15 2014-03-13 Manufacturing methods for production of rna transcripts

Publications (1)

Publication Number Publication Date
US20190085368A1 true US20190085368A1 (en) 2019-03-21

Family

ID=51581099

Family Applications (2)

Application Number Title Priority Date Filing Date
US14/777,190 Active US10138507B2 (en) 2013-03-15 2014-03-13 Manufacturing methods for production of RNA transcripts
US16/144,282 Abandoned US20190085368A1 (en) 2013-03-15 2018-09-27 Manufacturing methods for production of rna transcripts

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US14/777,190 Active US10138507B2 (en) 2013-03-15 2014-03-13 Manufacturing methods for production of RNA transcripts

Country Status (3)

Country Link
US (2) US10138507B2 (en)
EP (2) EP3578663A1 (en)
WO (1) WO2014152027A1 (en)

Cited By (31)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10925958B2 (en) 2016-11-11 2021-02-23 Modernatx, Inc. Influenza vaccine
US10933127B2 (en) 2015-10-22 2021-03-02 Modernatx, Inc. Betacoronavirus mRNA vaccine
US11045540B2 (en) 2017-03-15 2021-06-29 Modernatx, Inc. Varicella zoster virus (VZV) vaccine
US11103578B2 (en) 2016-12-08 2021-08-31 Modernatx, Inc. Respiratory virus nucleic acid vaccines
WO2021212034A1 (en) * 2020-04-16 2021-10-21 Nature's Toolbox, Inc. In vitro manufacturing and purification of therapeutic mrna
US11197927B2 (en) 2016-10-21 2021-12-14 Modernatx, Inc. Human cytomegalovirus vaccine
US11202793B2 (en) 2016-09-14 2021-12-21 Modernatx, Inc. High purity RNA compositions and methods for preparation thereof
US11207398B2 (en) 2017-09-14 2021-12-28 Modernatx, Inc. Zika virus mRNA vaccines
US11235052B2 (en) 2015-10-22 2022-02-01 Modernatx, Inc. Chikungunya virus RNA vaccines
US11285222B2 (en) 2015-12-10 2022-03-29 Modernatx, Inc. Compositions and methods for delivery of agents
US11351242B1 (en) 2019-02-12 2022-06-07 Modernatx, Inc. HMPV/hPIV3 mRNA vaccine composition
US11364292B2 (en) 2015-07-21 2022-06-21 Modernatx, Inc. CHIKV RNA vaccines
US11384352B2 (en) 2016-12-13 2022-07-12 Modernatx, Inc. RNA affinity purification
US11406703B2 (en) 2020-08-25 2022-08-09 Modernatx, Inc. Human cytomegalovirus vaccine
US11464848B2 (en) 2017-03-15 2022-10-11 Modernatx, Inc. Respiratory syncytial virus vaccine
US11484590B2 (en) 2015-10-22 2022-11-01 Modernatx, Inc. Human cytomegalovirus RNA vaccines
US11497807B2 (en) 2017-03-17 2022-11-15 Modernatx, Inc. Zoonotic disease RNA vaccines
US11547673B1 (en) 2020-04-22 2023-01-10 BioNTech SE Coronavirus vaccine
US11564893B2 (en) 2015-08-17 2023-01-31 Modernatx, Inc. Methods for preparing particles and related compositions
US11576961B2 (en) 2017-03-15 2023-02-14 Modernatx, Inc. Broad spectrum influenza virus vaccine
US11643441B1 (en) 2015-10-22 2023-05-09 Modernatx, Inc. Nucleic acid vaccines for varicella zoster virus (VZV)
US11744801B2 (en) 2017-08-31 2023-09-05 Modernatx, Inc. Methods of making lipid nanoparticles
US11752206B2 (en) 2017-03-15 2023-09-12 Modernatx, Inc. Herpes simplex virus vaccine
US11767548B2 (en) 2017-08-18 2023-09-26 Modernatx, Inc. RNA polymerase variants
US11786607B2 (en) 2017-06-15 2023-10-17 Modernatx, Inc. RNA formulations
US11851694B1 (en) 2019-02-20 2023-12-26 Modernatx, Inc. High fidelity in vitro transcription
US11866696B2 (en) 2017-08-18 2024-01-09 Modernatx, Inc. Analytical HPLC methods
US11878055B1 (en) 2022-06-26 2024-01-23 BioNTech SE Coronavirus vaccine
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
US11912982B2 (en) 2017-08-18 2024-02-27 Modernatx, Inc. Methods for HPLC analysis
US11911453B2 (en) 2018-01-29 2024-02-27 Modernatx, Inc. RSV RNA vaccines

Families Citing this family (103)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE10347710B4 (en) 2003-10-14 2006-03-30 Johannes-Gutenberg-Universität Mainz Recombinant vaccines and their use
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
PT2506857T (en) 2009-12-01 2018-05-14 Translate Bio Inc Delivery of mrna for the augmentation of proteins and enzymes in human genetic diseases
WO2012019168A2 (en) 2010-08-06 2012-02-09 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US20120237975A1 (en) 2010-10-01 2012-09-20 Jason Schrum Engineered nucleic acids and methods of use thereof
WO2012075040A2 (en) 2010-11-30 2012-06-07 Shire Human Genetic Therapies, Inc. mRNA FOR USE IN TREATMENT OF HUMAN GENETIC DISEASES
AU2012236099A1 (en) 2011-03-31 2013-10-03 Moderna Therapeutics, Inc. Delivery and formulation of engineered nucleic acids
HUE062102T2 (en) 2011-05-24 2023-09-28 BioNTech SE Individualized vaccines for cancer
SI2717893T1 (en) 2011-06-08 2019-10-30 Translate Bio Inc Lipid nanoparticle compositions and methods for mrna delivery
US9464124B2 (en) 2011-09-12 2016-10-11 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
RS63244B1 (en) 2011-12-16 2022-06-30 Modernatx Inc Modified mrna compositions
WO2013143555A1 (en) 2012-03-26 2013-10-03 Biontech Ag Rna formulation for immunotherapy
US9283287B2 (en) 2012-04-02 2016-03-15 Moderna Therapeutics, Inc. Modified polynucleotides for the production of nuclear proteins
US10501512B2 (en) 2012-04-02 2019-12-10 Modernatx, Inc. Modified polynucleotides
US9572897B2 (en) 2012-04-02 2017-02-21 Modernatx, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
EP2833892A4 (en) 2012-04-02 2016-07-20 Moderna Therapeutics Inc Modified polynucleotides for the production of oncology-related proteins and peptides
JP6561378B2 (en) 2012-06-08 2019-08-21 トランスレイト バイオ, インコーポレイテッド Transpulmonary delivery of mRNA to non-pulmonary target cells
EP3536787A1 (en) 2012-06-08 2019-09-11 Translate Bio, Inc. Nuclease resistant polynucleotides and uses thereof
EP2885419A4 (en) 2012-08-14 2016-05-25 Moderna Therapeutics Inc Enzymes and polymerases for the synthesis of rna
ES2921623T3 (en) 2012-11-26 2022-08-30 Modernatx Inc terminally modified RNA
AU2013351542B2 (en) 2012-11-28 2018-08-09 BioNTech SE Individualized vaccines for cancer
WO2014159813A1 (en) 2013-03-13 2014-10-02 Moderna Therapeutics, Inc. Long-lived polynucleotide molecules
KR102311614B1 (en) 2013-03-14 2021-10-08 샤이어 휴먼 지네틱 테라피즈 인크. Cftr mrna compositions and related methods and uses
AU2014239184B2 (en) 2013-03-14 2018-11-08 Translate Bio, Inc. Methods and compositions for delivering mRNA coded antibodies
US10258698B2 (en) 2013-03-14 2019-04-16 Modernatx, Inc. Formulation and delivery of modified nucleoside, nucleotide, and nucleic acid compositions
AU2014236396A1 (en) 2013-03-14 2015-08-13 Shire Human Genetic Therapies, Inc. Methods for purification of messenger RNA
WO2014144767A1 (en) 2013-03-15 2014-09-18 Moderna Therapeutics, Inc. Ion exchange purification of mrna
WO2014152030A1 (en) 2013-03-15 2014-09-25 Moderna Therapeutics, Inc. Removal of dna fragments in mrna production process
US11377470B2 (en) 2013-03-15 2022-07-05 Modernatx, Inc. Ribonucleic acid purification
EP3578663A1 (en) 2013-03-15 2019-12-11 ModernaTX, Inc. Manufacturing methods for production of rna transcripts
EP2972360B1 (en) 2013-03-15 2018-03-07 Translate Bio, Inc. Synergistic enhancement of the delivery of nucleic acids via blended formulations
WO2014180490A1 (en) 2013-05-10 2014-11-13 Biontech Ag Predicting immunogenicity of t cell epitopes
HRP20211563T1 (en) 2013-07-11 2022-01-07 Modernatx, Inc. Compositions comprising synthetic polynucleotides encoding crispr related proteins and synthetic sgrnas and methods of use
WO2015048744A2 (en) 2013-09-30 2015-04-02 Moderna Therapeutics, Inc. Polynucleotides encoding immune modulating polypeptides
WO2015051169A2 (en) 2013-10-02 2015-04-09 Moderna Therapeutics, Inc. Polynucleotide molecules and uses thereof
CA2928078A1 (en) 2013-10-22 2015-04-30 Shire Human Genetic Therapies, Inc. Lipid formulations for delivery of messenger rna
CN105658242A (en) 2013-10-22 2016-06-08 夏尔人类遗传性治疗公司 MRNA therapy for phenylketonuria
AU2014340092B2 (en) 2013-10-22 2019-09-19 Translate Bio, Inc. mRNA therapy for Argininosuccinate Synthetase Deficiency
EA201690588A1 (en) 2013-10-22 2016-09-30 Шир Хьюман Дженетик Терапис, Инк. DELIVERY OF MRNA IN THE CNS AND ITS APPLICATION
SG11201608798YA (en) 2014-04-23 2016-11-29 Modernatx Inc Nucleic acid vaccines
CN110511927A (en) 2014-04-25 2019-11-29 川斯勒佰尔公司 The purification process of mRNA
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
WO2015196128A2 (en) 2014-06-19 2015-12-23 Moderna Therapeutics, Inc. Alternative nucleic acid molecules and uses thereof
CN111588695A (en) 2014-06-24 2020-08-28 川斯勒佰尔公司 Stereochemically enriched compositions for delivery of nucleic acids
CN114146063A (en) 2014-07-02 2022-03-08 川斯勒佰尔公司 Encapsulation of messenger RNA
EP3169335B8 (en) 2014-07-16 2019-10-09 ModernaTX, Inc. Circular polynucleotides
WO2016014846A1 (en) 2014-07-23 2016-01-28 Moderna Therapeutics, Inc. Modified polynucleotides for the production of intrabodies
WO2016045732A1 (en) 2014-09-25 2016-03-31 Biontech Rna Pharmaceuticals Gmbh Stable formulations of lipids and liposomes
MA56412A (en) 2014-12-05 2022-05-04 Translate Bio Inc MESSENGER RNA THERAPY FOR THE TREATMENT OF JOINT DISEASES
WO2016128060A1 (en) 2015-02-12 2016-08-18 Biontech Ag Predicting t cell epitopes useful for vaccination
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
DE202016009003U1 (en) 2015-05-29 2021-05-28 Curevac Real Estate Gmbh Composition comprising in vitro transcribed RNA obtainable by a method for the production and purification of RNA with at least one step with a tangential flow filtration
WO2017009376A1 (en) * 2015-07-13 2017-01-19 Curevac Ag Method of producing rna from circular dna and corresponding template dna
US11007260B2 (en) 2015-07-21 2021-05-18 Modernatx, Inc. Infectious disease vaccines
ES2837100T3 (en) * 2015-08-10 2021-06-29 Curevac Real Estate Gmbh Method of increasing the replication of a circular DNA molecule
WO2017049286A1 (en) 2015-09-17 2017-03-23 Moderna Therapeutics, Inc. Polynucleotides containing a morpholino linker
CA3001003A1 (en) 2015-10-05 2017-04-13 Modernatx, Inc. Methods for therapeutic administration of messenger ribonucleic acid drugs
WO2017059902A1 (en) 2015-10-07 2017-04-13 Biontech Rna Pharmaceuticals Gmbh 3' utr sequences for stabilization of rna
MA56219A (en) 2015-10-14 2022-04-20 Translate Bio Inc MODIFICATION OF RNA-RELATED ENZYMES FOR ENHANCED PRODUCTION
JP2018536023A (en) 2015-10-22 2018-12-06 モデルナティーエックス, インコーポレイテッド Herpes simplex virus vaccine
AU2016342048B2 (en) 2015-10-22 2022-09-08 Modernatx, Inc. Broad spectrum influenza virus vaccine
SG11201803363YA (en) 2015-10-22 2018-05-30 Modernatx Inc Respiratory syncytial virus vaccine
EP3417069A1 (en) * 2016-02-15 2018-12-26 CureVac AG Method for analyzing by-products of rna in vitro transcription
EP3825400A1 (en) 2016-04-08 2021-05-26 Translate Bio Ma, Inc. Multimeric coding nucleic acid and uses thereof
CN109312313A (en) 2016-06-13 2019-02-05 川斯勒佰尔公司 For treating the mRNA therapy of ornithine transcarbamylase deficiency disease
WO2018009838A1 (en) 2016-07-07 2018-01-11 Rubius Therapeutics, Inc. Compositions and methods related to therapeutic cell systems expressing exogenous rna
EP3577221A4 (en) 2017-02-01 2020-12-23 ModernaTX, Inc. Polynucleotide secondary structure
MA47603A (en) 2017-02-27 2020-01-01 Translate Bio Inc NEW ARNM CFTR WITH OPTIMIZED CODONS
IL270631B2 (en) 2017-05-16 2024-03-01 Translate Bio Inc Treatment of cystic fibrosis by delivery of codon-optimized mrna encoding cftr
US10793841B2 (en) 2017-06-30 2020-10-06 Codexis, Inc. T7 RNA polymerase variants
KR20200023454A (en) 2017-06-30 2020-03-04 코덱시스, 인코포레이티드 T7 RNA Polymerase Variants
MA51306A (en) 2017-12-20 2020-10-28 Translate Bio Inc IMPROVED COMPOSITIONS AND METHODS FOR THE TREATMENT OF ORNITHINE TRANSCARBAMYLASE DEFICIENCY
CA3095125A1 (en) * 2018-03-27 2019-10-03 Factor Bioscience Inc. Nucleic acid-based therapeutics
CN112930396A (en) 2018-08-24 2021-06-08 川斯勒佰尔公司 Method for purifying messenger RNA
JP2022521094A (en) 2019-02-20 2022-04-05 モデルナティエックス インコーポレイテッド RNA polymerase variant for co-transcription capping
EP4055169A1 (en) 2019-11-04 2022-09-14 Flagship Pioneering, Inc. Methods of modifying a nucleic acid sequence
WO2021154763A1 (en) 2020-01-28 2021-08-05 Modernatx, Inc. Coronavirus rna vaccines
US20230346914A1 (en) 2020-02-07 2023-11-02 Modernatx, Inc. Sars-cov-2 mrna domain vaccines
WO2021222304A1 (en) 2020-04-27 2021-11-04 Modernatx, Inc. Sars-cov-2 rna vaccines
WO2021159130A2 (en) 2020-05-15 2021-08-12 Modernatx, Inc. Coronavirus rna vaccines and methods of use
WO2022067010A1 (en) 2020-09-25 2022-03-31 Modernatx, Inc. Multi-proline-substituted coronavirus spike protein vaccines
EP4274607A1 (en) 2021-01-11 2023-11-15 ModernaTX, Inc. Seasonal rna influenza virus vaccines
EP4277653A1 (en) 2021-01-15 2023-11-22 ModernaTX, Inc. Variant strain-based coronavirus vaccines
WO2022155524A1 (en) 2021-01-15 2022-07-21 Modernatx, Inc. Variant strain-based coronavirus vaccines
EP4277654A1 (en) 2021-01-18 2023-11-22 Conserv Bioscience Limited Coronavirus immunogenic compositions, methods and uses thereof
AU2022237382A1 (en) 2021-03-15 2023-09-28 Modernatx, Inc. Therapeutic use of sars-cov-2 mrna domain vaccines
WO2022245888A1 (en) 2021-05-19 2022-11-24 Modernatx, Inc. Seasonal flu rna vaccines and methods of use
CA3225545A1 (en) * 2021-09-16 2023-03-23 Immunitybio, Inc. Enzyme based system for production of messenger rna with increased transfection efficiency
CN113862235A (en) * 2021-09-24 2021-12-31 苏州近岸蛋白质科技股份有限公司 Chimeric enzyme and application and method thereof in synthesis of Cap0mRNA by in vitro one-step reaction
WO2023069498A1 (en) 2021-10-22 2023-04-27 Senda Biosciences, Inc. Mrna vaccine composition
WO2023092069A1 (en) 2021-11-18 2023-05-25 Modernatx, Inc. Sars-cov-2 mrna domain vaccines and methods of use
WO2023096858A1 (en) 2021-11-23 2023-06-01 Senda Biosciences, Inc. A bacteria-derived lipid composition and use thereof
WO2023104114A2 (en) 2021-12-07 2023-06-15 Immorna (hangzhou) Biotechnology Co., Ltd. Rna formulations and lipids
WO2023107999A2 (en) 2021-12-08 2023-06-15 Modernatx, Inc. Herpes simplex virus mrna vaccines
WO2023109961A1 (en) 2021-12-17 2023-06-22 Immorna (hangzhou) Biotechnology Co., Ltd. Interleukin-12 self-replicating rna and methods
WO2023122080A1 (en) 2021-12-20 2023-06-29 Senda Biosciences, Inc. Compositions comprising mrna and lipid reconstructed plant messenger packs
WO2023161350A1 (en) 2022-02-24 2023-08-31 Io Biotech Aps Nucleotide delivery of cancer therapy
WO2023196914A1 (en) 2022-04-08 2023-10-12 Modernatx, Inc. Influenza nucleic acid compositions and uses thereof
CN117730157A (en) * 2022-04-15 2024-03-19 萨默费尼根有限公司 Method for quantitatively monitoring mRNA capping efficiency
WO2024015890A1 (en) 2022-07-13 2024-01-18 Modernatx, Inc. Norovirus mrna vaccines
WO2024025815A1 (en) * 2022-07-25 2024-02-01 Modernatx, Inc. Use of imac to improve rna purity
WO2024050483A1 (en) 2022-08-31 2024-03-07 Modernatx, Inc. Variant strain-based coronavirus vaccines and uses thereof

Family Cites Families (222)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US354491A (en) 1886-12-14 Cigar-machine
DE3211309A1 (en) 1982-03-26 1983-09-29 Metin Dipl.-Ing. 6100 Darmstadt Colpan CHROMATOGRAPHIC METHOD FOR INSULATING MACROMOLECULES
DE3639949A1 (en) 1986-11-22 1988-06-09 Diagen Inst Molekularbio METHOD FOR SEPARATING LONG CHAIN NUCLEIC ACIDS
US5599667A (en) 1987-03-02 1997-02-04 Gen-Probe Incorporated Polycationic supports and nucleic acid purification separation and hybridization
CA1327358C (en) 1987-11-17 1994-03-01 Morio Fujiu Fluoro cytidine derivatives
US5759802A (en) 1988-10-26 1998-06-02 Tonen Corporation Production of human serum alubumin A
US5512439A (en) 1988-11-21 1996-04-30 Dynal As Oligonucleotide-linked magnetic particles and uses thereof
US5591722A (en) 1989-09-15 1997-01-07 Southern Research Institute 2'-deoxy-4'-thioribonucleosides and their antiviral activity
WO1991005058A1 (en) * 1989-10-05 1991-04-18 Glenn Kawasaki Cell-free synthesis and isolation of novel genes and polypeptides
CA2028849A1 (en) 1990-03-08 1991-09-09 Michinao Mizugaki Monoclonal antibodies, assay method, reagent kit, sarching method and drug missiles using them
US5637459A (en) 1990-06-11 1997-06-10 Nexstar Pharmaceuticals, Inc. Systematic evolution of ligands by exponential enrichment: chimeric selex
US5489677A (en) 1990-07-27 1996-02-06 Isis Pharmaceuticals, Inc. Oligonucleoside linkages containing adjacent oxygen and nitrogen atoms
US5426180A (en) 1991-03-27 1995-06-20 Research Corporation Technologies, Inc. Methods of making single-stranded circular oligonucleotides
US5437976A (en) 1991-08-08 1995-08-01 Arizona Board Of Regents, The University Of Arizona Multi-domain DNA ligands bound to a solid matrix for protein and nucleic acid affinity chromatography and processing of solid-phase DNA
EP1695979B1 (en) 1991-12-24 2011-07-06 Isis Pharmaceuticals, Inc. Gapped modified oligonucleotides
FR2687679B1 (en) 1992-02-05 1994-10-28 Centre Nat Rech Scient OLIGOTHIONUCLEOTIDES.
US6391542B1 (en) 1992-09-10 2002-05-21 Isis Pharmaceuticals, Inc. Compositions and methods for treatment of Hepatitis C virus-associated diseases
US5338448A (en) 1992-10-16 1994-08-16 Sarasep, Inc. Method of preventing contamination of a chromatography column
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
US5766903A (en) 1995-08-23 1998-06-16 University Technology Corporation Circular RNA and uses thereof
US5808039A (en) 1995-10-18 1998-09-15 Beckman Instruments, Inc. 2'-OMe CAC phosphoramidite and methods for preparation and use thereof
US5871697A (en) 1995-10-24 1999-02-16 Curagen Corporation Method and apparatus for identifying, classifying, or quantifying DNA sequences in a sample without sequencing
US6022737A (en) 1995-11-02 2000-02-08 Amgen Inc. Formulations for non-viral in vivo transfection in the lungs
US5789578A (en) 1996-01-11 1998-08-04 Massey University Methods for the preparation of resins with ligands attached thereto through a linking group comprising sulfide, sulfoxide or sulfone functionality
US7291463B2 (en) 1996-01-23 2007-11-06 Affymetrix, Inc. Nucleic acid labeling compounds
US6011148A (en) 1996-08-01 2000-01-04 Megabios Corporation Methods for purifying nucleic acids
US6447998B1 (en) 1996-08-09 2002-09-10 Isis Pharmaceuticals, Inc. 2-Aminopyridine and 2-pyridone C-nucleosides, oligonucleotides comprising, and tests using the same oligonucleotides
US6475388B1 (en) 1996-11-13 2002-11-05 Transgenomic, Inc. Method and system for RNA analysis by matched ion polynucleotide chromatography
GB2324370B (en) 1997-04-14 1999-03-03 Stuart Harbron Detection of hybrid double-stranded DNA with antibody after enzyme degradation of excess single-standed DNA
US6177559B1 (en) 1998-04-24 2001-01-23 Transgenomic, Inc. Process for separation of polynucleotide fragments
US5989911A (en) 1997-05-09 1999-11-23 University Of Massachusetts Site-specific synthesis of pseudouridine in RNA
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
US6111096A (en) 1997-10-31 2000-08-29 Bbi Bioseq, Inc. Nucleic acid isolation and purification
US5955310A (en) 1998-02-26 1999-09-21 Novo Nordisk Biotech, Inc. Methods for producing a polypeptide in a bacillus cell
US6248268B1 (en) 1998-11-16 2001-06-19 Xc Corporation Process of making microparticles of a thermally-gelled polysaccharide
CA2361201A1 (en) 1999-01-28 2000-08-03 Medical College Of Georgia Research Institute, Inc. Composition and method for in vivo and in vitro attenuation of gene expression using double stranded rna
US8410248B2 (en) 1999-03-12 2013-04-02 Human Genome Sciences Inc. HWBAO62 polypeptides
US6270970B1 (en) 1999-05-14 2001-08-07 Promega Corporation Mixed-bed solid phase and its use in the isolation of nucleic acids
EP1818409A1 (en) 1999-09-09 2007-08-15 CureVac GmbH Transfer of mRNAusing polycationic compounds
CN1317291C (en) 1999-09-10 2007-05-23 杰龙公司 Oligonucleotide N3'-P5' thiophosphoramidates, their synthesis and use
US6511832B1 (en) 1999-10-06 2003-01-28 Texas A&M University System In vitro synthesis of capped and polyadenylated mRNAs using baculovirus RNA polymerase
CA2325169A1 (en) 1999-12-03 2001-06-03 Ndsu Research Foundation Somatostatins and method
WO2001055306A2 (en) 2000-01-31 2001-08-02 Human Genome Sciences, Inc. Nucleic acids, proteins, and antibodies
JP2003529774A (en) 2000-03-31 2003-10-07 ジェネンテック・インコーポレーテッド Compositions and methods for detecting and quantifying gene expression
EP1274837A2 (en) 2000-04-21 2003-01-15 TRANSGENOMIC, Inc. Apparatus and method for separating and purifying polynucleotides
GB0013658D0 (en) 2000-06-05 2000-07-26 Dynal Asa Nucleic acid isolation
CN100495030C (en) 2000-09-30 2009-06-03 清华大学 Multi-force operator and use thereof
AU2002236524A1 (en) * 2000-11-28 2002-06-11 Rosetta Inpharmatics, Inc. In vitro transcription method for rna amplification
US20020130430A1 (en) 2000-12-29 2002-09-19 Castor Trevor Percival Methods for making polymer microspheres/nanospheres and encapsulating therapeutic proteins and other products
US20040142433A1 (en) 2001-02-02 2004-07-22 Padgett Hal S. Polynucleotide sequence variants
US7211654B2 (en) 2001-03-14 2007-05-01 Regents Of The University Of Michigan Linkers and co-coupling agents for optimization of oligonucleotide synthesis and purification on solid supports
EP1383556B9 (en) 2001-04-21 2008-03-19 Curevac GmbH INJECTION DEVICE FOR ADMINISTERING mRNA
US6812341B1 (en) 2001-05-11 2004-11-02 Ambion, Inc. High efficiency mRNA isolation methods and compositions
US20030170891A1 (en) 2001-06-06 2003-09-11 Mcswiggen James A. RNA interference mediated inhibition of epidermal growth factor receptor gene expression using short interfering nucleic acid (siNA)
US7235358B2 (en) 2001-06-08 2007-06-26 Expression Diagnostics, Inc. Methods and compositions for diagnosing and monitoring transplant rejection
CA2460819A1 (en) 2001-09-18 2003-03-27 Affinium Pharmaceuticals, Inc. Methods and apparatuses for purification
WO2003029459A2 (en) 2001-09-28 2003-04-10 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. Micro-rna molecules
IL161664A0 (en) * 2001-11-05 2004-09-27 Janssen Pharmaceutica Nv METHOD FOR THE IN VITRO SYNTHESIS OF SHORT DOUBLE STRANDED RNAs
WO2003039523A2 (en) 2001-11-05 2003-05-15 Exiqon A/S OLIGONUCLEOTIDES MODIFIED WITH NOVEL α-L-RNA ANALOGUES
US20040076978A1 (en) 2001-11-14 2004-04-22 Catherine Verfaillie Method to identify genes associated with chronic myelogenous leukemia
AU2002359732A1 (en) 2001-12-17 2003-06-30 Ribapharm Inc. Substituted purine nucleoside libraries and compounds by solid-phase combinatorial strategies
DE10162480A1 (en) 2001-12-19 2003-08-07 Ingmar Hoerr The application of mRNA for use as a therapeutic agent against tumor diseases
EP1487999B1 (en) 2002-03-15 2006-12-27 Epigenomics AG Discovery and diagnostic methods using 5-methylcytosine dna glycosylase
EP1527175B1 (en) 2002-06-24 2009-05-27 Exiqon A/S Methods and systems for detection and isolation of a nucleotide sequence
AU2003264041A1 (en) 2002-08-09 2004-02-25 California Institute Of Technology Method and compositions relating to 5'-chimeric ribonucleic acids
JP4339852B2 (en) 2002-08-12 2009-10-07 ニュー・イングランド・バイオラブズ・インコーポレイティッド Methods and compositions for gene silencing
AU2003264844A1 (en) 2002-09-24 2004-04-19 Yissum Research Development Company Of The Hebrew University Of Jerusalem Methods of determining the effect of an agent on diploid cells and/or on the pattern of expression of polypeptides expressed therewith
CA2504443A1 (en) 2002-10-30 2004-05-13 Pointilliste, Inc. Methods for producing polypeptide-tagged collections and capture systems containing the tagged polypeptides
WO2004041203A2 (en) 2002-11-04 2004-05-21 Xenoport, Inc. Gemcitabine prodrugs, pharmaceutical compositions and uses thereof
AU2004206820B2 (en) 2003-01-16 2010-06-10 Idera Pharmaceuticals, Inc. Modulation of immunostimulatory properties of oligonucleotide-based compounds by utilizing modified immunostimulatory dinucleotides
US20040224425A1 (en) 2003-05-08 2004-11-11 Gjerde Douglas T. Biomolecule open channel solid phase extraction systems and methods
US8158354B2 (en) 2003-05-13 2012-04-17 Ibis Biosciences, Inc. Methods for rapid purification of nucleic acids for subsequent analysis by mass spectrometry by solution capture
CA2533701A1 (en) 2003-07-31 2005-02-17 Isis Pharmaceuticals, Inc. Oligomeric compounds and compositions for use in modulation of small non-coding rnas
US7615539B2 (en) 2003-09-25 2009-11-10 Coley Pharmaceutical Group, Inc. Nucleic acid-lipophilic conjugates
WO2005042716A2 (en) 2003-10-31 2005-05-12 President And Fellows Of Harvard College Nucleic acid binding oligonucleotides
US8075780B2 (en) 2003-11-24 2011-12-13 Millipore Corporation Purification and concentration of synthetic biological molecules
SG187381A1 (en) 2003-12-03 2013-02-28 Coda Therapeutics Nz Ltd Antisense compounds targeted to connexins and methods of use thereof
CA2549985C (en) 2003-12-16 2011-05-24 Gentra Systems, Inc. Formulations and methods for denaturing proteins
US9790539B2 (en) 2004-06-30 2017-10-17 Russell Biotech, Inc. Methods and reagents for improved selection of biological molecules
DE102004035227A1 (en) 2004-07-21 2006-02-16 Curevac Gmbh mRNA mixture for vaccination against tumor diseases
EP2990410A1 (en) 2004-08-10 2016-03-02 Alnylam Pharmaceuticals Inc. Chemically modified oligonucleotides
CA2576280A1 (en) 2004-08-13 2006-02-16 Barry J. Marshall Helicobacter pylori-based delivery system
ATE540110T1 (en) 2004-11-11 2012-01-15 Modular Genetics Inc OLIGONUCLEOTIDE LADDER CONSTRUCTION AND SYSTEM FOR GENERATING MOLECULAR DIVERSITY
WO2007005645A2 (en) 2005-06-30 2007-01-11 Archemix Corp. Materials and methods for the generation of fully 2'-modified nucleic acid transcripts
US8101385B2 (en) 2005-06-30 2012-01-24 Archemix Corp. Materials and methods for the generation of transcripts comprising modified nucleotides
DK1919503T3 (en) 2005-08-10 2014-12-15 Macrogenics Inc Identification and preparation of antibodies with variant fc regions and methods of use 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
LT2578685T (en) 2005-08-23 2019-06-10 The Trustees Of The University Of Pennsylvania Rna containing modified nucleosides and methods of use thereof
US9157066B2 (en) 2005-12-13 2015-10-13 The Trustees Of The University Of Pennsylvania Transcriptome transfer produces cellular phenotype conversion
JP2009524430A (en) 2006-01-26 2009-07-02 ユニバーシティ オブ マサチューセッツ RNA interference agents for therapeutic use
WO2007092538A2 (en) 2006-02-07 2007-08-16 President And Fellows Of Harvard College Methods for making nucleotide probes for sequencing and synthesis
WO2007120863A2 (en) * 2006-04-14 2007-10-25 Epicentre Technologies Kits and methods for generating 5' capped rna
JP2007304525A (en) 2006-05-15 2007-11-22 Ricoh Co Ltd Image input device, electronic equipment, and image input method
US20080153078A1 (en) 2006-06-15 2008-06-26 Braman Jeffrey C System for isolating biomolecules from a sample
EP1882739A1 (en) 2006-06-30 2008-01-30 Qiagen GmbH Nucleic acid extraction method
EP2295045A1 (en) 2006-07-07 2011-03-16 Aarhus Universitet Nanoparticles for nucleic acid delivery
US8304529B2 (en) 2006-07-28 2012-11-06 Life Technologies Corporation Dinucleotide MRNA cap analogs
LT2056845T (en) 2006-08-08 2017-11-27 Rheinische Friedrich-Wilhelms-Universität Bonn Structure and use of 5` phosphate oligonucleotides
GB2445441B (en) 2006-09-26 2010-06-30 Ge Healthcare Bio Sciences Nucleic acid purification method
GB2445442A (en) 2006-09-26 2008-07-09 Ge Healthcare Bio Sciences Nucleic acid purification using anion exchange
US9163229B2 (en) 2006-10-10 2015-10-20 Trovagene, Inc. Compositions, methods and kits for isolating nucleic acids from body fluids using anion exchange media
DE102006051516A1 (en) 2006-10-31 2008-05-08 Curevac Gmbh (Base) modified RNA to increase the expression of a protein
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
DE102007001370A1 (en) 2007-01-09 2008-07-10 Curevac Gmbh RNA-encoded antibodies
GB0706243D0 (en) 2007-03-30 2007-05-09 Univ Southampton Modified nucleic acids
US7682789B2 (en) 2007-05-04 2010-03-23 Ventana Medical Systems, Inc. Method for quantifying biomolecules conjugated to a nanoparticle
US20080311140A1 (en) 2007-05-29 2008-12-18 Baylor College Of Medicine Antigen specific immunosuppression by dendritic cell therapy
JP2010531142A (en) 2007-06-22 2010-09-24 サークル バイオロジクス、 エルエルシー. Liquid concentrator, autologous concentrated body fluid, and methods of use thereof
WO2009016431A1 (en) 2007-08-01 2009-02-05 Digilab, Inc. Sample preparation method and apparatus
WO2009030254A1 (en) 2007-09-04 2009-03-12 Curevac Gmbh Complexes of rna and cationic peptides for transfection and for immunostimulation
KR101541935B1 (en) 2007-09-26 2015-08-05 인트렉손 코포레이션 Synthetic 5'UTRs, expression vectors, and methods for increasing transgene expression
US20100297242A1 (en) 2007-10-17 2010-11-25 Tae-Gwan Park Ldl-like cationic nanoparticles for deliverying nucleic acid gene, method for preparing thereof and method for deliverying nucleic acid gene using the same
WO2009058911A2 (en) * 2007-10-31 2009-05-07 Applied Biosystems Inc. Preparation and isolation of 5' capped mrna
US20090270278A1 (en) 2007-11-06 2009-10-29 Ambergen, Inc. Methods and compounds for making arrays
JP5530933B2 (en) 2007-12-10 2014-06-25 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 Compositions and methods for inhibiting factor VII gene expression
WO2009117167A1 (en) 2008-01-02 2009-09-24 Blood Cell Storage, Inc. Devices and processes for nucleic acid extraction
SG188104A1 (en) 2008-01-31 2013-03-28 Curevac Gmbh Nucleic acids comprising formula (nuglxmgnnv)a and derivatives thereof as an immunostimulating agents /adjuvants
NZ588583A (en) 2008-04-15 2012-08-31 Protiva Biotherapeutics Inc Novel lipid formulations for nucleic acid delivery
WO2009127230A1 (en) 2008-04-16 2009-10-22 Curevac Gmbh MODIFIED (m)RNA FOR SUPPRESSING OR AVOIDING AN IMMUNOSTIMULATORY RESPONSE AND IMMUNOSUPPRESSIVE COMPOSITION
WO2009147519A1 (en) 2008-06-06 2009-12-10 Centre National De La Recherche Scientifique - Cnrs- Use of endo-lysosomal system and secreted vesicles (exosome-like) in treatments and diagnostics based on small rna and experimental study of small rna
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
CA2731760C (en) 2008-07-24 2019-09-24 Meiji Seika Kaisha, Ltd. Pyripyropene a biosynthetic gene
WO2010017510A1 (en) 2008-08-07 2010-02-11 University Of Southern California A system for synergistic expression of multiple small functional rna elements
WO2010025302A2 (en) 2008-08-27 2010-03-04 Life Technologies Corporation Apparatus for and method of processing biological samples
DE102008063001A1 (en) 2008-12-23 2010-06-24 Qiagen Gmbh Nucleic acid purification method
CA2756520C (en) 2009-03-24 2017-07-11 Council Of Scientific & Industrial Research Process for the preparation of agarose polymer from seaweed extractive
EP3165606A1 (en) 2009-05-01 2017-05-10 Ophthotech Corporation Methods for treating or preventing ophthalmological diseases
US8765370B2 (en) 2009-06-11 2014-07-01 Scinopharm Taiwan, Ltd Inhibition-based high-throughput screen strategy for cell clones
EP3058953A1 (en) 2009-07-07 2016-08-24 The Research Foundation Of State University Of New York Lipidic compositions for induction of immune tolerance
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
US9574977B2 (en) 2013-02-26 2017-02-21 Innova Prep Liquid to liquid biological particle concentrator with disposable fluid path
PT2506857T (en) 2009-12-01 2018-05-14 Translate Bio Inc Delivery of mrna for the augmentation of proteins and enzymes in human genetic diseases
HUE036684T2 (en) 2009-12-07 2018-07-30 Univ Pennsylvania Rna preparations comprising purified modified rna for reprogramming cells
JP2011130725A (en) 2009-12-25 2011-07-07 Contig I:Kk Lna oligonucleotide and cosmetic containing the same
US10273470B2 (en) 2010-02-26 2019-04-30 Qiagen Gmbh Method for isolating RNA from a RNA and DNA containing sample
US20110218170A1 (en) 2010-03-02 2011-09-08 Southern Research Institute Use of 2'-deoxy-4'-thiocytidine and its analogues as dna hypomethylating anticancer agents
WO2011119711A1 (en) 2010-03-23 2011-09-29 Arcxis Biotechnologies Inc. Automated cellular material preparation
EP2377938A1 (en) 2010-04-16 2011-10-19 Eukarys Capping-prone RNA polymerase enzymes and their applications
WO2011127933A1 (en) 2010-04-16 2011-10-20 Nuevolution A/S Bi-functional complexes and methods for making and using such complexes
EP2558571A4 (en) 2010-04-16 2014-09-24 Immune Disease Inst Inc Sustained polypeptide expression from synthetic, modified rnas and uses thereof
WO2011133868A2 (en) 2010-04-22 2011-10-27 Alnylam Pharmaceuticals, Inc. Conformationally restricted dinucleotide monomers and oligonucleotides
WO2012019168A2 (en) 2010-08-06 2012-02-09 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US20120237975A1 (en) 2010-10-01 2012-09-20 Jason Schrum Engineered nucleic acids and methods of use thereof
US8898864B1 (en) 2010-10-08 2014-12-02 David Porter Integrated rockably released leverage snap fastening system
SG191089A1 (en) 2010-12-10 2013-07-31 Tracy Thompson Compositions for separation methods
WO2012129648A1 (en) 2011-03-25 2012-10-04 University Of Guelph Enhancing protein expression of adeno-associated virus vectors
AU2012236099A1 (en) 2011-03-31 2013-10-03 Moderna Therapeutics, Inc. Delivery and formulation of engineered nucleic acids
ES2587512T3 (en) 2011-04-04 2016-10-25 The U.S.A. 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
EP2710136A4 (en) 2011-05-17 2015-01-21 Moderna Therapeutics Inc Engineered nucleic acids and methods of use thereof for non-human vertebrates
WO2012164565A1 (en) 2011-06-01 2012-12-06 Yeda Research And Development Co. Ltd. Compositions and methods for downregulating prokaryotic genes
US8846883B2 (en) 2011-08-16 2014-09-30 University Of Southhampton Oligonucleotide ligation
US20130058902A1 (en) 2011-09-06 2013-03-07 Selecta Biosciences, Inc. Dendritic cell subsets for generating induced tolerogenic dendritic cells and related compositions and methods
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
WO2013039857A1 (en) 2011-09-12 2013-03-21 modeRNA Therapeutics Engineered nucleic acids and methods of use thereof
EP2760999A1 (en) 2011-09-26 2014-08-06 Qiagen GmbH Methods for separating nucleic acids by size
EP3682905B1 (en) 2011-10-03 2021-12-01 ModernaTX, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US9750795B2 (en) 2011-12-12 2017-09-05 The Trustees Of The University Of Pennsylvania Proteins comprising MRSA PBP2a and fragments thereof, nucleic acids encoding the same, and compositions and their use to prevent and treat MRSA infections
EP2791159A4 (en) 2011-12-14 2015-10-14 Moderna Therapeutics Inc Modified nucleic acids, and acute care uses thereof
EP2791364A4 (en) 2011-12-14 2015-11-11 Moderna 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
RS63244B1 (en) 2011-12-16 2022-06-30 Modernatx Inc Modified mrna compositions
CA2859691A1 (en) 2011-12-21 2013-06-27 Moderna Therapeutics, Inc. Methods of increasing the viability or longevity of an organ or organ explant
WO2013101690A1 (en) 2011-12-29 2013-07-04 modeRNA Therapeutics Modified mrnas encoding cell-penetrating polypeptides
US20150030576A1 (en) 2012-01-10 2015-01-29 Moderna Therapeutics, Inc. Methods and compositions for targeting agents into and across the blood-brain barrier
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
CN103325662B (en) 2012-03-21 2016-03-30 清华大学 The preparation method of semi-conductive single-walled carbon nanotubes
US20150050354A1 (en) 2012-04-02 2015-02-19 Moderna Therapeutics, Inc. Modified polynucleotides for the treatment of otic diseases and conditions
CN108949772A (en) 2012-04-02 2018-12-07 现代泰克斯公司 For generating the modification polynucleotides of biological agent relevant to human diseases and protein
EP2833892A4 (en) 2012-04-02 2016-07-20 Moderna Therapeutics Inc Modified polynucleotides for the production of oncology-related proteins and peptides
US10501512B2 (en) 2012-04-02 2019-12-10 Modernatx, Inc. Modified polynucleotides
EP2647426A1 (en) 2012-04-03 2013-10-09 Max-Planck-Gesellschaft zur Förderung der Wissenschaften e.V. Replication of distributed nucleic acid molecules with preservation of their relative distribution through hybridization-based binding
WO2013184976A2 (en) 2012-06-06 2013-12-12 Northwestern University Compositions and methods for antigen-specific tolerance
EP2885419A4 (en) 2012-08-14 2016-05-25 Moderna Therapeutics Inc Enzymes and polymerases for the synthesis of rna
US9416405B2 (en) 2012-11-02 2016-08-16 Life Technologies Corporation Compositions, methods and kits for enhancing PCR specificity
ES2921623T3 (en) 2012-11-26 2022-08-30 Modernatx Inc terminally modified RNA
US20150315541A1 (en) 2012-12-13 2015-11-05 Moderna Therapeutics, Inc. Modified polynucleotides for altering cell phenotype
EP2931319B1 (en) 2012-12-13 2019-08-21 ModernaTX, 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
EP2964234A4 (en) 2013-03-09 2016-12-07 Moderna Therapeutics Inc Heterologous untranslated regions for mrna
EP3495505A1 (en) 2013-03-14 2019-06-12 Translate Bio, Inc. Quantitative assessment for cap efficiency of messenger rna
WO2014152513A1 (en) 2013-03-14 2014-09-25 Shire Human Genetic Therapies, Inc. RIBONUCLEIC ACIDs WITH 4'-THIO-MODIFIED NUCLEOTIDES AND RELATED METHODS
AU2014236396A1 (en) * 2013-03-14 2015-08-13 Shire Human Genetic Therapies, Inc. Methods for purification of messenger RNA
US10258698B2 (en) 2013-03-14 2019-04-16 Modernatx, Inc. Formulation and delivery of modified nucleoside, nucleotide, and nucleic acid compositions
BR112015022505A2 (en) 2013-03-14 2017-10-24 Shire Human Genetic Therapies quantitative evaluation for messenger rna cap efficiency
WO2014144767A1 (en) 2013-03-15 2014-09-18 Moderna Therapeutics, Inc. Ion exchange purification of mrna
US20160017313A1 (en) 2013-03-15 2016-01-21 Moderna Therapeutics, Inc. Analysis of mrna heterogeneity and stability
WO2014144039A1 (en) 2013-03-15 2014-09-18 Moderna Therapeutics, Inc. Characterization of mrna molecules
WO2014144835A1 (en) 2013-03-15 2014-09-18 NVS Technologies, Inc. Analytical instrument systems
US11377470B2 (en) 2013-03-15 2022-07-05 Modernatx, Inc. Ribonucleic acid purification
EP3578663A1 (en) 2013-03-15 2019-12-11 ModernaTX, Inc. Manufacturing methods for production of rna transcripts
US20140273230A1 (en) 2013-03-15 2014-09-18 Sigma-Aldrich Co., Llc Crispr-based genome modification and regulation
US8980864B2 (en) 2013-03-15 2015-03-17 Moderna Therapeutics, Inc. Compositions and methods of altering cholesterol levels
WO2014152030A1 (en) 2013-03-15 2014-09-25 Moderna Therapeutics, Inc. Removal of dna fragments in mrna production process
HRP20211563T1 (en) 2013-07-11 2022-01-07 Modernatx, Inc. Compositions comprising synthetic polynucleotides encoding crispr related proteins and synthetic sgrnas and methods of use
CA2923029A1 (en) 2013-09-03 2015-03-12 Moderna Therapeutics, Inc. Chimeric polynucleotides
EP3041938A1 (en) 2013-09-03 2016-07-13 Moderna Therapeutics, Inc. Circular polynucleotides
EP3043826A4 (en) 2013-09-13 2017-05-24 Moderna Therapeutics, Inc. Polynucleotide compositions containing amino acids
GB201317301D0 (en) 2013-09-30 2013-11-13 Linnarsson Sten Method for capturing and encoding nucleic acid from a plurality of single cells
WO2015048744A2 (en) 2013-09-30 2015-04-02 Moderna Therapeutics, Inc. Polynucleotides encoding immune modulating polypeptides
EP3052479A4 (en) 2013-10-02 2017-10-25 Moderna Therapeutics, Inc. Polynucleotide molecules and uses thereof
WO2015051169A2 (en) 2013-10-02 2015-04-09 Moderna Therapeutics, Inc. Polynucleotide molecules and uses thereof
AU2014329452B2 (en) 2013-10-03 2019-06-20 Moderna Therapeutics, Inc. Polynucleotides encoding low density lipoprotein receptor
AU2014337156A1 (en) 2013-10-18 2016-05-12 Modernatx, Inc. Compositions and methods for tolerizing cellular systems
WO2015070413A1 (en) 2013-11-14 2015-05-21 深圳智慧能源技术有限公司 Ceramic thermal shielding piece and heat-resistant structure
US20170173128A1 (en) 2013-12-06 2017-06-22 Moderna TX, Inc. Targeted adaptive vaccines
US20150167017A1 (en) 2013-12-13 2015-06-18 Moderna Therapeutics, Inc. Alternative nucleic acid molecules and uses thereof
SG10201805660WA (en) 2013-12-30 2018-08-30 Curevac Ag Methods for rna analysis
EP3092250A4 (en) 2014-01-08 2017-05-24 Moderna Therapeutics, Inc. Polynucleotides for the in vivo production of antibodies
CN114797474A (en) 2014-05-21 2022-07-29 非链实验室 System and method for buffer solution exchange
US20170136132A1 (en) 2014-06-19 2017-05-18 Moderna Therapeutics, Inc. Alternative nucleic acid molecules and uses thereof
WO2015196128A2 (en) 2014-06-19 2015-12-23 Moderna Therapeutics, Inc. Alternative nucleic acid molecules and uses thereof
US20170175129A1 (en) 2014-06-19 2017-06-22 Moderna Therapeutics, Inc. Alternative nucleic acid molecules and uses thereof
CA2955250A1 (en) 2014-07-16 2016-01-21 Moderna Therapeutics, Inc. Chimeric polynucleotides
EP3169335B8 (en) 2014-07-16 2019-10-09 ModernaTX, Inc. Circular polynucleotides
WO2016036902A1 (en) 2014-09-03 2016-03-10 Moderna Therapeutics, Inc. Tolerogenic compositions and methods
EP2992958A1 (en) 2014-09-03 2016-03-09 STAT-Diagnostica D Innovation SL Nucleic acid purification cartridge
US20180000953A1 (en) 2015-01-21 2018-01-04 Moderna Therapeutics, Inc. Lipid nanoparticle compositions
WO2016118725A1 (en) 2015-01-23 2016-07-28 Moderna Therapeutics, Inc. Lipid nanoparticle compositions
DK3294885T3 (en) 2015-05-08 2020-08-10 Curevac Real Estate Gmbh METHOD OF PREPARING RNA

Cited By (39)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11364292B2 (en) 2015-07-21 2022-06-21 Modernatx, Inc. CHIKV RNA vaccines
US11564893B2 (en) 2015-08-17 2023-01-31 Modernatx, Inc. Methods for preparing particles and related compositions
US10933127B2 (en) 2015-10-22 2021-03-02 Modernatx, Inc. Betacoronavirus mRNA vaccine
US11872278B2 (en) 2015-10-22 2024-01-16 Modernatx, Inc. Combination HMPV/RSV RNA vaccines
US11643441B1 (en) 2015-10-22 2023-05-09 Modernatx, Inc. Nucleic acid vaccines for varicella zoster virus (VZV)
US11484590B2 (en) 2015-10-22 2022-11-01 Modernatx, Inc. Human cytomegalovirus RNA vaccines
US11235052B2 (en) 2015-10-22 2022-02-01 Modernatx, Inc. Chikungunya virus RNA vaccines
US11278611B2 (en) 2015-10-22 2022-03-22 Modernatx, Inc. Zika virus RNA vaccines
US11285222B2 (en) 2015-12-10 2022-03-29 Modernatx, Inc. Compositions and methods for delivery of agents
US11202793B2 (en) 2016-09-14 2021-12-21 Modernatx, Inc. High purity RNA compositions and methods for preparation thereof
US11541113B2 (en) 2016-10-21 2023-01-03 Modernatx, Inc. Human cytomegalovirus vaccine
US11197927B2 (en) 2016-10-21 2021-12-14 Modernatx, Inc. Human cytomegalovirus vaccine
US11696946B2 (en) 2016-11-11 2023-07-11 Modernatx, Inc. Influenza 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
US11752206B2 (en) 2017-03-15 2023-09-12 Modernatx, Inc. Herpes simplex virus vaccine
US11464848B2 (en) 2017-03-15 2022-10-11 Modernatx, Inc. Respiratory syncytial virus vaccine
US11918644B2 (en) 2017-03-15 2024-03-05 Modernatx, Inc. Varicella zoster virus (VZV) vaccine
US11045540B2 (en) 2017-03-15 2021-06-29 Modernatx, Inc. Varicella zoster virus (VZV) vaccine
US11576961B2 (en) 2017-03-15 2023-02-14 Modernatx, Inc. Broad spectrum influenza virus vaccine
US11497807B2 (en) 2017-03-17 2022-11-15 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
US11786607B2 (en) 2017-06-15 2023-10-17 Modernatx, Inc. RNA formulations
US11767548B2 (en) 2017-08-18 2023-09-26 Modernatx, Inc. RNA polymerase variants
US11866696B2 (en) 2017-08-18 2024-01-09 Modernatx, Inc. Analytical HPLC methods
US11912982B2 (en) 2017-08-18 2024-02-27 Modernatx, Inc. Methods for HPLC analysis
US11744801B2 (en) 2017-08-31 2023-09-05 Modernatx, Inc. Methods of making lipid nanoparticles
US11207398B2 (en) 2017-09-14 2021-12-28 Modernatx, Inc. Zika virus mRNA vaccines
US11911453B2 (en) 2018-01-29 2024-02-27 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
WO2021212034A1 (en) * 2020-04-16 2021-10-21 Nature's Toolbox, Inc. In vitro manufacturing and purification of therapeutic mrna
US11779659B2 (en) 2020-04-22 2023-10-10 BioNTech SE RNA constructs and uses thereof
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
US11406703B2 (en) 2020-08-25 2022-08-09 Modernatx, Inc. Human cytomegalovirus vaccine
US11878055B1 (en) 2022-06-26 2024-01-23 BioNTech SE Coronavirus vaccine

Also Published As

Publication number Publication date
EP2971033B8 (en) 2019-07-10
WO2014152027A1 (en) 2014-09-25
EP3578663A1 (en) 2019-12-11
EP2971033B1 (en) 2019-05-29
US10138507B2 (en) 2018-11-27
US20160024547A1 (en) 2016-01-28
EP2971033A1 (en) 2016-01-20
EP2971033A4 (en) 2016-11-16

Similar Documents

Publication Publication Date Title
US20190085368A1 (en) Manufacturing methods for production of rna transcripts
EP3041948B1 (en) Alternative nucleic acid molecules containing reduced uracil content and uses thereof
EP3350333B1 (en) Polynucleotides containing a stabilizing tail region
CA2892529C (en) Terminally modified rna
EP3682905B1 (en) Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
CA2869005A1 (en) Lipid nanoparticle compositions including polynucleotides encoding proteins
US20190017100A1 (en) Method for analysis of an rna molecule
CN111819185A (en) Compositions comprising cyclic polyribonucleotides and uses thereof
US20180201967A1 (en) Method of producing rna from circular dna and corresponding template dna
WO2015051173A2 (en) Polynucleotide molecules and uses thereof
EP2885419A2 (en) Enzymes and polymerases for the synthesis of rna
CN113661242A (en) Compositions comprising modified cyclic polyribonucleotides and uses thereof
KR20180131577A (en) New minimal UTR sequence
WO2020061397A1 (en) Compositions and methods for delivery of nucleic acids
KR20200051648A (en) RNA or protein expression in vivo using double-stranded concatemer DNA containing phosphorothioated nucleotides
US11898186B1 (en) Compositions and methods for preparing capped mRNA
WO2024010993A1 (en) Primer design for cell-free dna production

Legal Events

Date Code Title Description
AS Assignment

Owner name: MODERNA THERAPEUTICS, INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:AUNINS, JOHN GRANT;REEL/FRAME:047131/0502

Effective date: 20150813

Owner name: MODERNA THERAPEUTICS, INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:BANCEL, STEPHANE;ISSA, WILLIAM JOSEPH;CHAKRABORTY, TIRTHA;SIGNING DATES FROM 20141201 TO 20141208;REEL/FRAME:047131/0457

Owner name: MODERNATX, INC., MASSACHUSETTS

Free format text: CHANGE OF NAME;ASSIGNOR:MODERNA THERAPEUTICS, INC.;REEL/FRAME:047214/0526

Effective date: 20160808

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION