WO2008016473A2 - Dinucleotide mrna cap analogs - Google Patents

Dinucleotide mrna cap analogs Download PDF

Info

Publication number
WO2008016473A2
WO2008016473A2 PCT/US2007/015896 US2007015896W WO2008016473A2 WO 2008016473 A2 WO2008016473 A2 WO 2008016473A2 US 2007015896 W US2007015896 W US 2007015896W WO 2008016473 A2 WO2008016473 A2 WO 2008016473A2
Authority
WO
WIPO (PCT)
Prior art keywords
composition
recited
och
halogen
cap
Prior art date
Application number
PCT/US2007/015896
Other languages
French (fr)
Other versions
WO2008016473A3 (en
Inventor
Anilkumar R. Kore
Muthian Shanmugasundaram
Original Assignee
Applera Corporation
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 Applera Corporation filed Critical Applera Corporation
Priority to EP07810384A priority Critical patent/EP2049665A2/en
Priority to JP2009522766A priority patent/JP2009544754A/en
Priority to US12/375,527 priority patent/US8304529B2/en
Priority to CA002659301A priority patent/CA2659301A1/en
Publication of WO2008016473A2 publication Critical patent/WO2008016473A2/en
Publication of WO2008016473A3 publication Critical patent/WO2008016473A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H21/00Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids
    • C07H21/02Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids with ribosyl as saccharide radical
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H21/00Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids
    • C07H21/04Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids with deoxyribosyl as saccharide radical
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/111General methods applicable to biologically active non-coding nucleic acids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/31Chemical structure of the backbone
    • C12N2310/317Chemical structure of the backbone with an inverted bond, e.g. a cap structure
    • 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
    • C12N2320/00Applications; Uses
    • C12N2320/50Methods for regulating/modulating their activity
    • C12N2320/51Methods for regulating/modulating their activity modulating the chemical stability, e.g. nuclease-resistance

Definitions

  • Eukaryotic mRNAs bear a "cap" structure at their 5'-termini that is well known to play an important role in translation.
  • Naturally occurring cap structures consist of a 7-methyl guanosine that is linked via a triphosphate bridge to the 5 '-end of the first transcribed nucleotide, resulting in m 7 G(5')ppp(5')N, where N is any nucleotide.
  • the mRNA cap plays an important role in gene expression. It protects the mRNAs from degradation by exonucleases, enables transport of RNAs from the nucleus to the cytoplasm, and participates in assembly of the translation initiation complex.
  • nlOCS ⁇ pppCS ⁇ G (mCAP) has been used as the primer in transcription with T7 or SP6 RNA polymerase in vitro to obtain RNAs having a cap structure in their 5 '-termini. In vivo, the cap is added en2ymatically. However, over the past 20 years or so, numerous studies have required the synthesis of proteins in an in vitro translation extract supplemented with in vitro synthesized mRNA. The prevailing method for the in vitro synthesis of capped mRNA employs a preformed dinucleotide of the form m'GOOpppCS ⁇ G as an initiator of transcription.
  • mCAP a pseudosymmetrical dinucleotide
  • G or m 7 G (m 7 Guo) moiety to serve as the initiating nucleophile for transcriptional elongation.
  • Unmethylated cap analog is a modified cap analog in which the methyl group on the guanosine is removed.
  • the selective procedure for methylation of guanosine at N7 and 3' Omethylation and 5' diphosphate synthesis was established (Kore,A. and Parmar,G. Nucleosides, Nucleotides, and Nucleic Acids, 25:337-340, 2006 and Kore A.R., et al. Nucleosides Nucleotides Nucleic Acids 200625(3): 307-14.
  • the Anti-Reverse Cap Analog is a modified cap analog in which the 3 1 OH group is replaced with OCH3. ARCA and triple-methylated cap analogs are incorporated in the forward orientation.
  • the cap is added in the nucleus and is catalyzed by the enzyme guanylyl transferase.
  • the addition of the cap to the 5' terminal end of RNA occurs after transcription but immediately after transcription initiation so that it is almost impossible to detect.
  • the terminal nucleoside is always a guanine, and is in the reverse orientation to all the other nucleotides, i.e., 5 'Gppp5 'GpNpNp... and the cap contains two nucleotides, connected by a 5 '-5' triphosphate linkage.
  • RNA Transcription of RNA usually starts with a nucleoside triphosphate (usually a purine, A or G). When transcription occurs in vitro, it typically includes a phage RNA polymerase such as T7, T3 or SP6, a DNA template containing a phage polymerase promoter, nucleotides (ATP, GTP, CTP and UTP) and a buffer containing magnesium salt.
  • a phage RNA polymerase such as T7, T3 or SP6
  • a DNA template containing a phage polymerase promoter a DNA template containing a phage polymerase promoter
  • nucleotides ATP, GTP, CTP and UTP
  • a buffer containing magnesium salt e.g., MCP, CTP and UTP
  • the synthesis of capped RNA includes the incorporation of a cap analog (e.g., N7 methyl GpppG or m7GpppG) in the transcription reaction. Excess m7
  • the mMESSAGE mMACMNE® kit from Ambion (Ambion, Inc., Austin, TX, a business of Applied Biosystems) recommends this ratio and will typically yield 80% capped RNA to 20% uncapped RNA, although total yields of total RNA are lower as GTP concentration becomes rate limiting as GTP is necessary for the elongation of the transcript.
  • the 5' cap structure enhances the translation of mRNA by helping to bind the eukaryotic ribosome and assuring recognition of the proper AUG initiator codon. This function may vary with the translation system and with the specific mRNA being synthesized.
  • the consensus sequence 5'-GCCACCAUGG-3' also known as the "Kozak" sequence, is considered to be the strongest ribosomal binding signal in eukaryotic mRNA.
  • the key elements are the G residue at the +1 position and the A residue at the -3 position.
  • Capped RNA encoding specific genes can be transfected into eukaryotic cells or microinjected into cells or embryos to study the effect of translated product in the cell or embryo. If uncapped RNA is used, the RNA in these experiments is rapidly degraded and the yield of translated protein is much reduced.
  • Isolated dendritic cells from a patient can be transfected with capped RNA encoding immunogen.
  • the dendritic cells translate the capped RNA into a protein that induces an immune response against this protein.
  • immunotherapy with dendritic cells loaded with CEA capped RNA was shown to be safe and feasible for pancreatic patients (Morse et al. Int. J. Gastroinstest. Cancer, 32:1-6, 2002). It was also noted that introducing a single capped RNA species into immature dendritic cells induced a specific T-cell response (Heiser et al. J. Clin. Invest., 109:409-417, 2002.
  • composition comprising:
  • A is selected from a halogen, OH, OCH 3 , H, ter/-butyldimethylsilyl and 2',3'-O- isopropylidene
  • B is selected from a halogen, OH, OCH 3 , NH 2 , N 3 , NO 2 , CF 3 , CHO, S, tert- butyldimethylsilyl, LNA, and 2',3'-0-isopropylidene
  • R] is CH 3
  • R 2 is selected from OH, OCH 3 and a halogen
  • n is 1, 2 or 3
  • R 1 is void
  • R 2 is OH 3 then A is neither OH nor OCH 3 , and when A, B and R 2 are OH, Rj is not CH 3 .
  • An anti-reverse cap analog composition is also provided which is represented by the formula:
  • A is selected from a halogen, OH, OCH 3 , H, terf-butyldimethylsilyl and 2 ',3'- O-isopropylidene
  • B is selected from a halogen, OH 5 OCH 3 , NH 2, N 3, NO 2, CHO, S, tert- butyldimethylsilyl, LNA, and 2',3'-0-isopropylidene
  • Ri is void
  • R 2 is selected from OH, OCH 3 and a halogen
  • n is 1, 2 or 3; and when A is either OH or OCH 3 , then R 2 is not OH.
  • a double-methylated cap analog composition is also provided as represented by the formula:
  • A is selected from a halogen, OH, OCH 3 , H,-and 2',3'-O-isopropylidene
  • B is selected from CF 3> OH, OCH 3 , NH 2 , N 3 , NO 2 , CHO 5 LNA 5 and 2',3'-0-isopropylidene
  • Ri is CH 3
  • R 2 is selected from OH 5 OCH 3 and a halogen
  • n is I 5 2 or 3 and when A is OH, R 2 is not OH.
  • a triple-methylated cap analog is provided as represented by the formula: wherein A is selected from OH and OCH3, B is selected from OH and OCH 3 , and n is, I 5 2 or 3.
  • the halogen can be fluorine, chlorine, bromine or iodine.
  • the phophodiester linkage between the guanosine molecules can be a triphosphate linkage, a tetraphosphate linkage or a pentaphosphate linkage.
  • RNA molecule is also provided which has incorporated at its 5' end one of the structures as described above.
  • kits for capping an RNA transcript comprising the formula: a) a cap analog have the structure as recited in claim 1 :
  • a is selected from a halogen, OH, OCH 3 , H, TBDMS and 2',3'-O- isopropylidene
  • B is selected from OH, OCH 3 , NH 2, N 3 , NO 2 , CF 3, CHO, halogen, S, TBDMS 5 LNA, and 2',3'-0-isopropylidene
  • Ri is CH 3 or void
  • R 2 is selected from OH, OCThand a halogen
  • n is, 1, 2 or 3; wherein when B is OH or OCH 3, and R 1 is void, A is neither OH nor OCH3 if R 2 is OH and b).
  • the kit may also comprise nucleotides, ribonuclease inhibitor, enzyme buffer, and nucleotide buffer.
  • a method of synthesizing a dinucleotide cap analog comprising: a) providing a first nucleoside comprising at least one of a 2' substituent and a 3' substituent on the ribose ring, b) phosphorylating the first nucleoside, forming a first nucleotide, c) methylating the first nucleotide, d) adding a phosphorylated second nucleotide optionally comprising a T ribose ring substituent, and e) linking said first nucleotide with said second nucleotide, forming a dinucleotide cap analog.
  • Figure 1 illustrates the synthesis scheme for novel double-methylated cap analogs.
  • Figure 2 illustrates the synthesis of a novel ARCA.
  • Figure 3 illustrates the basic RNA cap structure (mCAP) consisting of Pl- guanosine-5'-yl P3-7-methylguanosine-5'-yl triphosphate (m 7 G5'ppp5 ! G).
  • mCAP basic RNA cap structure
  • Figure 4 illustrates exemplary modified cap analogs. Modifications are at either or both G7 positions and/or the 3' ribose position of the m 7 Guo.
  • Figure 5 shows the results of digestion of AmbLuc poly A RNA with BIp 1
  • Figure 6 graphically depicts yields from transcription reactions by using double- and triple-methylated cap analogs in an AmbLuc poly A vector.
  • Figure 7 shows the high level of RNA integrity when capped as analyzed with an Agilent 2100 B ⁇ oanalyzer.
  • Figure 8 show the capping efficiency of RNA in a transcription reaction by a gel shift assay.
  • Figure 9 illustrates the comparison of protein expression between standard and ARCA, double-methylated, and triple-methylated capped luciferase RNAs with poly(A) tail at different time points after transfection.
  • Figure 10 illustrates transcript yield by using modified and normal cap analogs.
  • Figure 1 1 illustrates capping efficiency with fluorine-modified cap analogs
  • Figure 12 illustrates transfection efficiency of fluorine-modified cap analogs in a luciferase assay
  • ARCA anti-reverse cap analog refers to a modified cap analog in which the 3' OH group is replaced with OCH3.
  • the structure is represented as m 2 7 ' 3' °(5')Gppp(5')G.
  • cap refers to a non-extendible di-nucleotide that facilitates translation or localization, and/or prevents degradation of an RNAtranscript when incorporated at the 5' end of an RNA transcript, typically having an m7GpppG or m7GpppA structure. It consists in nature of the modified base 7-methylguanosine joined in the opposite orientation, 5' to 5' rather than 5' to 3', to the rest of the molecule via three phosphate groups i.e., Pl-guanosine-5'-yI P3-7-methylguanosine-5'-yl triphosphate (m 7 G5'ppp5'G).
  • cap analog refers to a structural derivative of an RNA cap that may differ by as little as a single element.
  • double-methylated cap refers to each guanosine molecule of a dinucleotide cap in which each N7 position contains a -CH 3 group. It is illustrated as m 7 (5')Gppp(5')m 7 G.
  • tm-CAP triple-methylated cap
  • tmCAPs are illustrated as ni2 7 ' 2 ° (5')Gppp(5')m 7 G, m 2 7 ' 3 ° (5')Gppp(5')m 7 G, m 7 (5')Gppp(5') m 2 7 ' 2'0 G,or m 7 (5')Gppp(5') m 2 7l3' °-G.
  • the term "enzymatically incorporatable” means that a nucleotide is capable of being enzymatically incorporated onto the terminus, e.g. 3 1 terminus, of a polynucleotide chain, or internally through nick-translation of a polynucleotide chain, through action of a template-dependent or template-independent polymerase enzyme.
  • a nucleotide-5 '-triphosphate is an example of an enzymatically incorporatable nucleotide.
  • the term "enzymatically extendable” or “3' extendable” means a nucleotide or polynuceotide that is capable of being appended to a nucleotide or polynucleotide by enzyme action.
  • a polynucleotide containing a 3' hydroxyl group is an example of an enzymatically extendable polynucleotide.
  • halogen refers to nonmetal elements of Group 7A of the Periodic Table of the Elemnts comprising fluorine, F, chlorine, Cl, bromine, Br, iodine, I, and astatine, At. Halogens are monovalent, readily form negative ions and occur as compounds or ions.
  • LNA locked nucleic acid
  • nucleobase refers to a nitrogen containing heterocyclic moiety nucleobase.
  • suitable nucleobases include: adenine, cytosine, guanine, thymine, uracil, 5-propynyl-uracil, 2-thio-5-propynyl -uracil, 5- methylcytosine, pseudoisocytosine, 2-thiouracil, 2-thiothymine, 2-aminopurine, N9-(2- amino-6-chloropurine), N9-(2,6-diaminopurine), hypoxanthine, N9-(7-deaza-guanine), N9- (7-deaza-8-aza-guanine) and N8-(8-aza-7-deazaadenine).
  • nucleoside refers to a compound consisting of a nucleobase linked to the C-I ' carbon of a ribose sugar or analog thereof.
  • the ribose or analog may be substituted or unsubstituted.
  • Substituted ribose sugars include, but are not limited to, those riboses in which one or more of the carbon atoms, preferably the 3'-carbon atom, is substituted with one or more of the same or different substituents such as -R, -OR, -NRR or halogen (e.g., fluoro, chloro, bromo, or iodo), where each R group is independently -H, Ci-C 6 alkyl or C3-C14 aryl.
  • substituents such as -R, -OR, -NRR or halogen (e.g., fluoro, chloro, bromo, or iodo)
  • riboses are ribose, T- deoxyribose, 2',3'-dideoxyribose, 3'-haloribose (such as 3'-fluororibose or 3'-chlororibose) and 3'-alkylribose.
  • the nucleobase is A or G
  • the ribose sugar is attached to the N 9 -position of the nucleobase.
  • the nucleobase is C, T or U
  • the pentose sugar is attached to the N'-position of the nucleobase (Kornberg and Baker, DNA Replication, 2 nd Ed., Freeman, San Francisco, CA, (1992)).
  • ribose analogs include arabinose, 2'-O-methyl ribose, and locked nucleoside analogs (e.g., WO 99/14226), for example, although many other analogs are also known in the art.
  • nucleotide refers to a phosphate ester of a nucleoside as a monomer unit or within a polynucleotide.
  • nucleotide triphosphate refers to a nucleotide with a triphosphate ester group at the 5' position.
  • nucleosides and/or nucleotides of the present teachings can comprise "natural sugars” (i.e., -ribose, 2'-deoxyribose, and the like) or sugar analogues.
  • sugar analog refers to analogs of the sugar ribose.
  • exemplary ribose sugar analogs include, but are not limited to, substituted or unsubstituted furanoses having more or fewer than 5 ring atoms, e.g., erythroses and hexoses and substituted or unsubstituted 3-6 carbon acyclic sugars.
  • Typical substituted furanoses and acyclic sugars are those in which one or more of the carbon atoms are substituted with one or more of the same or different -R, -OR, -NRR or halogen groups, where each R is independently -H, (Ci-C ⁇ ) alkyl or (C 1 -Ci 4 ) aryl.
  • substituted furanoses having 5 ring atoms include but are not limited to 2'-deoxyribose, 2'-(Ci-C 6 )alkylribose, 2' -(C 1 - C ⁇ Jalkoxyribose, 2'-(C 5 -Ci 4 )aryloxyribose, 2',3'-dideoxyribose, 2',3'-didehydroribose J 2'-deoxy-3'-haloribose, 2'-deoxy-3'-fluororibose, 2'-deoxy-3'-chlororibose, 2'-deoxy- 3'-aminoribose, 2'-deoxy-3'-(Ci-C 6 )alkylribose, 2'-deoxy-3'-(Ci-C 6 )alkoxyribose, 2'-deoxy-3'-(C 5 -Ci 4 )aryloxyribo
  • polynucleotide As used herein, the terms “polynucleotide”, “oligonucleotide” and “nucleic acid' are used interchangeably and refer to single stranded and double stranded polymers of nucleotide monomers, including ribonucleotides (RNA) and 2'-deoxyribonucleotides (DNA) linked by internucleotide phosphodiester bond linkages.
  • RNA ribonucleotides
  • DNA 2'-deoxyribonucleotides linked by internucleotide phosphodiester bond linkages.
  • a polynucleotide may be composed entirely of deoxyribonucleotides, entirely of ribonucleotides or chimeric mixtures thereof.
  • terminal means an enzymatically incorporatable nucleotide which prevents subsequent incorporation of nucleotides to the resulting polynucleotide chain and thereby halts polymerase-mediated extension.
  • Typical terminators lack a 3'-hydroxyl substituent and include 2',3'-dideoxyribose, 2 ⁇ ,3'-didehydroribose, and 2',3'- dideoxy-3'-haloribose 5 e.g.3'-deoxy-3'-fluoro-ribose or 2',3'-dideoxy-3 r -fluororibose, for example.
  • a ribofuranose analog can be used, such as 2',3'-dideoxy- ⁇ -D- ribofuranosyl, ⁇ -D-arabinofuranosyl, 3'-deoxy- ⁇ -D-arabinofuranosyl, 3'-amino-2',3'- dideoxy- ⁇ -D-ribofuranosyl, and 2',3'-dideoxy-3'-fluoro- ⁇ -D-ribofuranosyl (see, for example, Chidgeavadze et al., Nucleic Acids Res., 12: 1671-1686 (1984), and Chidgeavadze et al. FEB. Lett., 183: 275-278 (1985)). Nucleotide terminators also include reversible nucleotide terminators (Metzker et al. Nucleic Acids Res., 22(20):4259 (1994)).
  • nonextendable or “3 1 nonextendable” refers to the fact that a terminator is incapable, or substantially incapable, of being extended in the 3' direction by a template -dependent DNA or RNA polymerase.
  • TDMS refers to /ert-butyldimethylsilyl
  • the term "void" refers to the absence of a substituent group at the Ri position of the cap analog. The lack of a substituent group results in no positive chare o the imidazole ring.
  • the substituent group may be a CH 3 group. When the CH 3 group is present, there is a positive charge on the imidazole ring.
  • Cap analog is used for the synthesis of 5' capped RNA molecules in in vitro transcription reactions. Substitution of cap analog for a portion of the GTP in a transcription reaction results in the incorporation of the cap structure into a corresponding fraction of the transcripts. Capped mRNAs are generally translated more efficiently in reticulocyte lysate and wheat germ in vitro translation systems. It is important that in vitro transcripts be capped for microinjection experiments because uncapped mRNAs are rapidly degraded. Cap analogs are also used as a highly specific inhibitor of the initiation step of protein synthesis.
  • cap structures have been synthesized and designed to attach in the forward orientation, i.e., 5'Gppp5'GpNpNp....
  • the resulting novel cap analogs have been demonstrated to improve the yield and efficiency of transcription compared to the standard cap analog as shown in Figures 6, 8, and 10.
  • capped RNA which is not only efficiently capped and yields high levels of transcribed RNA is an area of unmet need.
  • One approach which is being used by Ambion is disclosed in US patent application 2005/0287539, incorporated herein by reference.
  • the synthesis of cap analogs which incorporate in only the forward orientation will improve transcription and translation efficiency.
  • RNA transcripts are used for applications requiring protein synthesis such as in vivo expression (e.g., microinjection, transfection and infection experiments) and in vitro translation.
  • caps include m27,2'OG[5 I ]ppp[5 I ]m7G, m27,2OG[5']ppp[5'] m2'OG 5 m27 5 3'OG[5 l ]ppp[5 I ]m7G, and m27 5 2OG[5']ppp[5'] m27,2'OG. Additionally, details of the compatibility of new, modified cap analogs with respect to transcription, capping and translation efficiency by using HeLa cells in comparison with the standard and conventional ARCA cap analog is presented.
  • AU of the reagents and solvents are used as such without further purification, unless otherwise stated.
  • Guanosine 5 '-diphosphate, Dimethyl sulfate, anhydrous dimethylformamide , 2,2'-dithiodipyridine (Aldrithiol), Triphenylphosphine, trimethylphosphate ((OMe) 3 P), phosphorous oxychloride, phosphorous pentoxide, orthophosphoric acid, anhydrous methylene chloride, dichloromethane, Tributylamine, anhydrous pyridine were purchased from Sigma-Aldrich Co. 3'-O-Me-Guanosine is available from Chemgene, Boston, MA.
  • Imidazolide GMP lmidazolide GDP, Imidazolide 2'F-GMP, Imidazolide 3'CF 3 -GDP, Imidazolide m 7 GMP, IM tris(triethylammonium) phosphate, and tributylammonium orthophosphate were made as taught herein or in A. Kore, and G. Parmar, Synthetic Comm., 36:3393-3399, 2006, incorporated herein by reference in its entirety.
  • the resulting aqueous layer was further evaporated on a rotary evaporator to remove any chloroform traces, and then further diluted to 1.5 L with water and loaded on an anion exchange resin, i.e., DEAE Sepharosa fast flow packed in a BPG 100 column (Amersham GE, Piscataway, NJ, USA).
  • the desired compound was eluted by using four bed volumes of gradient from 0 to 80% of 1 M TEAB buffer (triethyammonium bicarbonate), pH 7.5, at a flow rate of 100 mL/min, using AKTA purifier 100 FPLC (Amersham GE).
  • TEAB buffer triethyammonium bicarbonate
  • AKTA purifier 100 FPLC FPLC
  • m 7 GDP 7-methylguanosine 5 '-diphosphate
  • the residual bicarbonate was removed by co-evaporating with methanol, 3 X 600 mL.
  • the resulting residue was transferred to a centrifuge tube, and 8.9 g sodium perchlorate dissolved in 1.1 L acetone was added and cooled 2 h at 4 0 C. The resulting mixture was centrifuged and the supernatant liquid was discarded. The precipitate was ground with a new portion of acetone, cooled and centrifuged, repeating once. The precipitate was dried in a vacuum desiccator over P 2 O 5 . The resulting amorphous white powder was 7 methyl-guanosine 5'- diphosphate.
  • the reaction was allowed to go to completion as determined by HPLC and then poured slowly into a mixture of sodium perchlorate (7 g) in acetone (1500 mL), and then cooled for 30 min. at 4 0 C. The reaction mixture was centrifuged, discarding the supernatant. Traces of imidazole and triphenylphosphine were removed by grinding the solid with a new portion of acetone (400 mL), cooling and again centrifuged, repeating once. The precipitate was dried in a vacuum oven over P 2 O 5 at 24 0 C (30 mbar pressure).
  • the ribonucleoside-5'-phosphoroimidazolide thus obtained was dissolved in dimethylforamide (200 mL), and a 1 M solution of tributylammonium orthophosphate in dimethylformamide (80 mL) was added drop-wise to the vigorously stirred mixture over a period of 30 min.
  • Zinc chloride (2 g, 14.67 mmol) was added and the reaction mixture stirred at room temperature for 3 h. Completion of the reaction was monitored by HPLC.
  • the reaction mixture was quenched with water (50 mL) and extracted with chloroform (3 X 200 mL), concentrated in a rotary evaporator and then purified by application to an anion exchange resin.
  • nucleoside-5'-diphosphate-containing fractions were pooled and evaporated using a rotary evaporator to give triethylamine salt of the desired diphosphate compound.
  • the mixture was then subjected to centrifugation at 3000 rpm for 15 min and the supernatant was discarded.
  • the precipitate was ground with a new portion of acetone and centrifuged. The process was repeated once more and the precipitate was dried in a vacuum desiccator over phosphorous pentoxide, yielding 3'-0-Me-GMP Imidazolide.
  • Tris(triethylammonium) phosphate linker product was dissolved in 100 mL anhydrous DMF so as to have a final concentration of 1 M, and stored over 4A molecular sieves at 4 0 C.
  • anhydrous DMF 40 mL
  • Zinc chloride (2.0 g, 14.6 7 mmol) was added in small portions until the contents were dissolved. Thereafter, the tris(triethylammonium) phosphate linker andl M tributylammonium orthophosphate (40 mL) was added slowly to the reaction mixture under argon and the reaction was allowed to stir at room temperature for 5 h. The reaction when followed on HPLC showing complete conversion of the starting material, 3'-0-Me-GMP Imidazolide (3) to its corresponding diphosphate.
  • reaction was supplemented with water, 100 mL, and the resultant mixture was extracted with chloroform (3 X 250 mL), subjected to volume reduction ( ⁇ 100 mL) by evaporation and applied to DEAE Sephadex A25 column, eluting with a linear gradient of freshly prepared 1 M TEAB, pH 7.5.
  • the fractions containing the pure 3'O-Me-GDP TEA salt (compound 4) were eluted, combined and evaporated to dryness.
  • the aqueous layer was applied to a DEAE Sephadex column and the fractions containing the product were pooled, evaporated and dried in a vacuum desiccator over phosphorous pentoxide to give 3'-O-Me (N 7 -Me) GDP (compound 5) as a fine powder.
  • the aqueous solution was concentrated under a rotor evaporator to around 20 mL.
  • a solution of sodium perchlorate (3.0 g) in 100 mL acetone in a centrifuge tube at O 0 C this concentrated solution (20.0 mL) was added slowly for 2 minutes. The resulting mixture was centrif ⁇ ged and the supernatant liquid was removed. This precipitate was dissolved in 500 mL water and adjusted the pH to 5.5 with saturated NaHCCb and loaded on a DEAE Sephadex column.
  • the collected aqueous solution was adjusted to pH 5.5 with saturated NaHCCb and loaded on a DEAE Sephadex column.
  • the desired product was eluted using a linear gradient of 0 - IM TEAB buffer, pH 7.5 and the fractions containing the product were pooled, evaporated and dried in vacuum desiccator over phosphorous pentoxide to give 2'F GDP as a fine white powder (compound 16) (Yield 134 g, 72%).
  • the collected aqueous solution was adjusted to pH 5.5 with saturated NaHCC> 3 and applied onto a DEAE Sephadex column.
  • the final product was monitored for methylation by HPLC?? ⁇ n ⁇ i eluted using a linear gradient of 0 - IM TEAB buffer, pH 7.5 and the fractions containing the product were pooled, evaporated and dried in vacuum desiccator over phosphorous pentoxide to give m 7>2/r GDP as a fine white powder (compound 17) (Yield 0.64 g, 83%).
  • the desired product was eluted using a linear gradient of 0 - IM TEAB buffer, pH 7.5 and the fractions containing the product were pooled, evaporated and concentrated to 10.0 mL TEA salt of compound 18.
  • the resulting 10.0 mL was passed through a Strata-X-AW column and washed with 10.0 mL water followed by 10.0 mL MeOH.
  • compound 18 was eluted with 15.0 mL OfNH 4 OHTMeOHZH 2 O (2/25/73) and the collected solution was evaporated and dried to give m 7>2Vr GpppG as a fine white powder (compound 18) (Yield: 0.15 g, 58%).
  • the resulting 10.0 mL was passed through a Strata-X-AW column and washed with 10.0 mL water followed by 10.0 mL MeOH.
  • compound 20 was eluted with 15.0 mL OfNH 4 OHZMeOHZH 2 O (2Z25Z73) and the collected solution was evaporated and dried to give m 7> 2 "F Gp ⁇ pm 7 G as a fine white powder (compound 20) (Yield: 0.21 g, 64%).
  • the resulting aqueous solution was adjusted to pH 5.5 with saturated NaHCO 3 and loaded on a DEAE Sephadex column.
  • the desired product was eluted using a linear gradient of 0 - IM TEAB buffer, pH 7.5 and the fractions containing the product were pooled, evaporated and concentrated to 10.0 mL TEA salt of compound 23.
  • the resulting 10.0 mL was passed through a Strata-X-AW column and washed with 10.0 mL water followed by 10.0 mL MeOH.
  • the resulting aqueous solution was adjusted to pH 5.5 with saturated NaHCO 3 and loaded on a DEAE Sephadex column.
  • the desired product was eluted using a linear gradient of 0 - IM TEAB buffer, pH 7.5 and the fractions containing the product were pooled, evaporated and concentrated to 10.0 mL TEA salt of compound 25.
  • the resulting 10.0 mL was passed through a Strata-X-AW column and washed with 10.0 mL water followed by 10.0 mL MeOH.
  • the resulting aqueous solution was adjusted to pH 5.5 with saturated NaHCOsand loaded on a DEAE Sephadex column.
  • the desired product was eluted using a linear gradient of 0 - IM TEAB buffer, pH 7.5 and the fractions containing the product were pooled, evaporated and concentrated to 10.0 mL TEA salt of 26.
  • the resulting 10.0 mL was passed through a Strata-X-AW column and washed with 10.0 mL water followed by 10.0 mL MeOH.
  • T7 RNA polymerase transcription was performed by using mMESSAGE mMACHINE® T7 Kit (Ambion) in 20 ⁇ L final volume, and contains the following reagents at the final concentrations indicated: 1 ⁇ g linearized AmbLuc PoIyA DNA, IX reaction buffer,7.5mM of each ATP 5 UTP, and CTP 5 1.5mM GTP 5 6.5 raM of mCAP and 2' fluoro cap analogs, and 50 UZ ⁇ l of T7 RNA polymerase. The transcription reactions were incubated at 37 0 C for 2 hours.
  • Protein expression from mCAP, double methylated, triple methylated, and ARCA capped luciferase RNAs, at different time points after transfection with HeLa cells indicates that the transcripts containing triple methylated cap are more highly translated in transfected cells.
  • Comparison of protein expression with mCAP and double methylated cap suggest that the double methylated cap transcript produces one fold higher protein, while ARCA produces 3 fold higher, and triple methylated cap produces 4 fold higher protein in in vivo with HeLa cells. See Figure 9.
  • EXAMPLE 20 [0135] The coupling reaction of m ⁇ ' ⁇ GDP (N7Me-2'FGDP) with Imidazolide 27 OMP (lm2'FGMP), or Imidazolide m 7>2'F GMP (N7MeIm2TGMP) in the presence of ZnCI 2 as the catalyst affords the corresponding cap analogs A and B.
  • EXAMPLE 21 [0136] The coupling reaction of m 7>3WHj GDP (N7Me-3 'NH 2 GDP) with Imidazolide 277 GMP (lm2'FGMP), or Imidazolide m ⁇ GMP (N7MeIm2'FGMP) in the presence of ZnCl 2 as the catalyst affords the corresponding cap analogs E and F.
  • EXAMPLE 26 [0141] The coupling reaction of m 7>3Wj 2 ' dGDP (N7Me-3'N 3 2'dGDP) with Imidazole GMP (ImGMP) or Imidazolide m 7 GMP (m7-ImGMP) in the presence OfZnCl 2 as the catalyst affords the corresponding cap analogs Q and R.

Abstract

Novel cap analogs which are easily synthesized, resulting in high levels of capping efficiency and transcription and improved translation efficiencies are provided. Such caps are methylated at the N7 position of one or both guanosines of the dinucleotide cap as well as at the 3' position on the ribose ring. Substituent groups on the ribose ring also result in the cap being incorporated in the forward orientation. Also provided are methods useful for preparing capped analogs and using mRNA species containing such analogs are also contemplated herein, as well as kits containing the novel cap analogs.

Description

DINUCLEOTIDE MRNA CAP ANALOGS
Cross-Reference to Related Applications
[0001] This application claims a priority benefit under 35 U.S.C. § 119(e) from U.S. Patent Application No. 60/820,771, filed July 28, 2006, which is incorporated herein by reference. Statement Regarding Federally Funded Sponsored Research or Development
[0002] Work described herein was funded at least in part by an SBIR grant (SBIR Phase II, No. R44GM070156-02) awarded by the National Institutes of Health. The U.S. Government may therefore have certain rights therein.
[0003] The section headings used herein are for organizational purposes only and should not be construed as limiting the subject matter described herein in any way.
Introduction
[0004] Eukaryotic mRNAs bear a "cap" structure at their 5'-termini that is well known to play an important role in translation. Naturally occurring cap structures consist of a 7-methyl guanosine that is linked via a triphosphate bridge to the 5 '-end of the first transcribed nucleotide, resulting in m7G(5')ppp(5')N, where N is any nucleotide. The mRNA cap plays an important role in gene expression. It protects the mRNAs from degradation by exonucleases, enables transport of RNAs from the nucleus to the cytoplasm, and participates in assembly of the translation initiation complex. nlOCS^pppCS^G (mCAP) has been used as the primer in transcription with T7 or SP6 RNA polymerase in vitro to obtain RNAs having a cap structure in their 5 '-termini. In vivo, the cap is added en2ymatically. However, over the past 20 years or so, numerous studies have required the synthesis of proteins in an in vitro translation extract supplemented with in vitro synthesized mRNA. The prevailing method for the in vitro synthesis of capped mRNA employs a preformed dinucleotide of the form m'GOOpppCS^G as an initiator of transcription. A disadvantage of using mCAP, a pseudosymmetrical dinucleotide, has always been the propensity of the 3'-OH of either the G or m7G (m7Guo) moiety to serve as the initiating nucleophile for transcriptional elongation. This leads to the synthesis of two isomeric RNAs of the form m7G(5')pppG(pN)n and G(5')ppp7G(pN)n, in approximately equal proportions, depending upon the ionic conditions of the transcription reaction. This may be problematic for various downstream processes, such as in vitro translation or crystallization studies. [0005] Unmethylated cap analog is a modified cap analog in which the methyl group on the guanosine is removed. The selective procedure for methylation of guanosine at N7 and 3' Omethylation and 5' diphosphate synthesis was established (Kore,A. and Parmar,G. Nucleosides, Nucleotides, and Nucleic Acids, 25:337-340, 2006 and Kore A.R., et al. Nucleosides Nucleotides Nucleic Acids 200625(3): 307-14. The Anti-Reverse Cap Analog (ARCA) is a modified cap analog in which the 31 OH group is replaced with OCH3. ARCA and triple-methylated cap analogs are incorporated in the forward orientation.
[0006] In the cell, the cap is added in the nucleus and is catalyzed by the enzyme guanylyl transferase. The addition of the cap to the 5' terminal end of RNA occurs after transcription but immediately after transcription initiation so that it is almost impossible to detect. The terminal nucleoside is always a guanine, and is in the reverse orientation to all the other nucleotides, i.e., 5 'Gppp5 'GpNpNp... and the cap contains two nucleotides, connected by a 5 '-5' triphosphate linkage.
[0007] Transcription of RNA usually starts with a nucleoside triphosphate (usually a purine, A or G). When transcription occurs in vitro, it typically includes a phage RNA polymerase such as T7, T3 or SP6, a DNA template containing a phage polymerase promoter, nucleotides (ATP, GTP, CTP and UTP) and a buffer containing magnesium salt. The synthesis of capped RNA includes the incorporation of a cap analog (e.g., N7 methyl GpppG or m7GpppG) in the transcription reaction. Excess m7GppρG to GTP (4:1) favors to increase the opportunity that each transcript will have a 5' cap. The mMESSAGE mMACMNE® kit from Ambion (Ambion, Inc., Austin, TX, a business of Applied Biosystems) recommends this ratio and will typically yield 80% capped RNA to 20% uncapped RNA, although total yields of total RNA are lower as GTP concentration becomes rate limiting as GTP is necessary for the elongation of the transcript.
[0008] The 5' cap structure enhances the translation of mRNA by helping to bind the eukaryotic ribosome and assuring recognition of the proper AUG initiator codon. This function may vary with the translation system and with the specific mRNA being synthesized. The consensus sequence 5'-GCCACCAUGG-3', also known as the "Kozak" sequence, is considered to be the strongest ribosomal binding signal in eukaryotic mRNA. For efficient translation initiation, the key elements are the G residue at the +1 position and the A residue at the -3 position.
[0009] During translation the cap is bound by translation initiation factor eEF-4E and the CBC recruits additional initiation factors. Decapping is catalyzed by proteins dcpl and dcp2 which compete with eIF-4E to bind to the cap. Translation results in amino acids as encoded by the mRNA to join together to form a peptide and occurs as three processes, initiation, elongation and termination. Initiation in eukaryotes involves attachment of a ribosome which scans the mRNA for the first methionine codon. Elongation proceeds with the successive addition of amino acids until a stop codon is reached, terminating translation.
[0010] Capped RNA encoding specific genes can be transfected into eukaryotic cells or microinjected into cells or embryos to study the effect of translated product in the cell or embryo. If uncapped RNA is used, the RNA in these experiments is rapidly degraded and the yield of translated protein is much reduced.
[0011] Isolated dendritic cells from a patient can be transfected with capped RNA encoding immunogen. The dendritic cells translate the capped RNA into a protein that induces an immune response against this protein. In a small human study, immunotherapy with dendritic cells loaded with CEA capped RNA was shown to be safe and feasible for pancreatic patients (Morse et al. Int. J. Gastroinstest. Cancer, 32:1-6, 2002). It was also noted that introducing a single capped RNA species into immature dendritic cells induced a specific T-cell response (Heiser et al. J. Clin. Invest., 109:409-417, 2002.
[0012] The recent literature reveals that chemical modification of m7Guo at either the 2' or 3' OH group results in the cap being incorporated solely in the forward orientation, even though the 2' OH group does not participate in the phosphodiester bond. This observation has prompted investigation of 2' and 3' OH modifications of m7Guo as well as modifications of m7Guo to create double- and triple-methylated cap analogs.
SUMMARY
[0013] A composition is provided comprising:
Figure imgf000004_0001
wherein A is selected from a halogen, OH, OCH3, H, ter/-butyldimethylsilyl and 2',3'-O- isopropylidene, B is selected from a halogen, OH, OCH3, NH2, N3, NO2, CF3, CHO, S, tert- butyldimethylsilyl, LNA, and 2',3'-0-isopropylidene; R] is CH3 Or void, R2 is selected from OH, OCH3 and a halogen, n is 1, 2 or 3, and when B is OH or OCH3> R1 is void, and R2 is OH3 then A is neither OH nor OCH3, and when A, B and R2 are OH, Rj is not CH3.
[0014] An anti-reverse cap analog composition is also provided which is represented by the formula:
Figure imgf000005_0001
wherein A is selected from a halogen, OH, OCH3, H, terf-butyldimethylsilyl and 2 ',3'- O-isopropylidene, B is selected from a halogen, OH5 OCH3, NH2, N3, NO2, CHO, S, tert- butyldimethylsilyl, LNA, and 2',3'-0-isopropylidene, Ri is void, R2 is selected from OH, OCH3 and a halogen, n is 1, 2 or 3; and when A is either OH or OCH3, then R2 is not OH.
[0015] A double-methylated cap analog composition is also provided as represented by the formula:
Figure imgf000005_0002
wherein A is selected from a halogen, OH, OCH3, H,-and 2',3'-O-isopropylidene, B is selected from CF3>OH, OCH3, NH2, N3, NO2, CHO5 LNA5 and 2',3'-0-isopropylidene, Ri is CH3, R2 is selected from OH5 OCH3 and a halogen, n is I5 2 or 3, and when A is OH, R2 is not OH.
[0016] A triple-methylated cap analog is provided as represented by the formula:
Figure imgf000006_0001
wherein A is selected from OH and OCH3, B is selected from OH and OCH3, and n is, I5 2 or 3.
[0017] When A or B are a halogen, the halogen can be fluorine, chlorine, bromine or iodine. The phophodiester linkage between the guanosine molecules can be a triphosphate linkage, a tetraphosphate linkage or a pentaphosphate linkage.
[0018] An RNA molecule is also provided which has incorporated at its 5' end one of the structures as described above.
[0019] Also provided is a kit for capping an RNA transcript comprising the formula: a) a cap analog have the structure as recited in claim 1 :
Figure imgf000006_0002
wherein a is selected from a halogen, OH, OCH3, H, TBDMS and 2',3'-O- isopropylidene; B is selected from OH, OCH3, NH2, N3, NO2, CF3, CHO, halogen, S, TBDMS5 LNA, and 2',3'-0-isopropylidene; Ri is CH3 or void; R2 is selected from OH, OCThand a halogen; and n is, 1, 2 or 3; wherein when B is OH or OCH3, and R1 is void, A is neither OH nor OCH3 if R2 is OH and b). RNA polymerase. The kit may also comprise nucleotides, ribonuclease inhibitor, enzyme buffer, and nucleotide buffer.
[0020] Also provided is a method of synthesizing a dinucleotide cap analog comprising: a) providing a first nucleoside comprising at least one of a 2' substituent and a 3' substituent on the ribose ring, b) phosphorylating the first nucleoside, forming a first nucleotide, c) methylating the first nucleotide, d) adding a phosphorylated second nucleotide optionally comprising a T ribose ring substituent, and e) linking said first nucleotide with said second nucleotide, forming a dinucleotide cap analog. .
[0021] These and other features of the present teachings are set forth herein.
BRIEF DESCRIPTION OF THE FIGURES
[0022] The skilled artisan will understand that the drawings, described below, are for illustration purposes only. The drawings are not intended to limit the scope of the present teachings in any way.
[0023] Figure 1 illustrates the synthesis scheme for novel double-methylated cap analogs.
[0024] Figure 2 illustrates the synthesis of a novel ARCA.
[0025] Figure 3 illustrates the basic RNA cap structure (mCAP) consisting of Pl- guanosine-5'-yl P3-7-methylguanosine-5'-yl triphosphate (m7G5'ppp5!G).
[0026] Figure 4 illustrates exemplary modified cap analogs. Modifications are at either or both G7 positions and/or the 3' ribose position of the m7 Guo.
[0027] Figure 5 shows the results of digestion of AmbLuc poly A RNA with BIp 1
[0028] Figure 6 graphically depicts yields from transcription reactions by using double- and triple-methylated cap analogs in an AmbLuc poly A vector.
[0029] Figure 7 shows the high level of RNA integrity when capped as analyzed with an Agilent 2100 Bϊoanalyzer.
[0030] Figure 8 show the capping efficiency of RNA in a transcription reaction by a gel shift assay.
[0031] Figure 9 illustrates the comparison of protein expression between standard and ARCA, double-methylated, and triple-methylated capped luciferase RNAs with poly(A) tail at different time points after transfection.
[0032] Figure 10 illustrates transcript yield by using modified and normal cap analogs.
[0033] Figure 1 1 illustrates capping efficiency with fluorine-modified cap analogs
[0034] Figure 12 illustrates transfection efficiency of fluorine-modified cap analogs in a luciferase assay
DESCRIPTION OF VARIOUS EMBODIMENTS
[0035] For the purposes of interpreting this specification, the following definitions will apply and whenever appropriate, terms used in the singular will also include the plural and vice versa. In the event that any definition set forth below conflicts with the usage of that word in any other document, including any document incorporated herein by reference, the definition set forth below shall always control for purposes of interpreting this specification and its associated claims unless a contrary meaning is clearly intended (for example in interpreting the document where the term is originally used). The use of "or" herein means "and/or" unless stated otherwise or where the use of "and/or" is clearly inappropriate. The use of "a" herein means "one or more" unless stated otherwise or where the use of "one or more" is clearly inappropriate. The use of "comprise," "comprises," "comprising," "include," "includes," and "including" are interchangeable and not intended to be limiting.
[0036] As used throughout, "Me" is equivalent to "CH3", "OCH3" or "OMe" denotes an oxygen atom bound to a methyl group, "CHO" denotes a carbon atom, C, bonded to a hydrogen atom, H, and double-bonded to an oxygen atom, O, (O=CH-) and "Et" denotes "C2H5".
[0037] As used herein, the term "ARCA" or anti-reverse cap analog refers to a modified cap analog in which the 3' OH group is replaced with OCH3. The structure is represented as m2 7'3'°(5')Gppp(5')G.
[0038] As used herein, the term "cap" refers to a non-extendible di-nucleotide that facilitates translation or localization, and/or prevents degradation of an RNAtranscript when incorporated at the 5' end of an RNA transcript, typically having an m7GpppG or m7GpppA structure. It consists in nature of the modified base 7-methylguanosine joined in the opposite orientation, 5' to 5' rather than 5' to 3', to the rest of the molecule via three phosphate groups i.e., Pl-guanosine-5'-yI P3-7-methylguanosine-5'-yl triphosphate (m7G5'ppp5'G).
[0039] As used herein, the term "cap analog" refers to a structural derivative of an RNA cap that may differ by as little as a single element.
[0040] As used herein, the term "double-methylated" cap (dm-CAP) refers to each guanosine molecule of a dinucleotide cap in which each N7 position contains a -CH3 group. It is illustrated as m7 (5')Gppp(5')m7G.
[0041] As used herein, the term "triple-methylated" cap (tm-CAP) refers to the presence of a -CH3 group on each N7 position of the guanosine molecules of a dinucleotide cap and an additional OCH3 group in the 2' or 3' position of the ribose ring of one of the guanosine molecules. tmCAPs are illustrated as ni27'2 ° (5')Gppp(5')m7G, m2 7'3 ° (5')Gppp(5')m7G, m7(5')Gppp(5') m2 7'2'0 G,or m7(5')Gppp(5') m2 7l3'°-G.
[0042] As used herein, the term "enzymatically incorporatable" means that a nucleotide is capable of being enzymatically incorporated onto the terminus, e.g. 31 terminus, of a polynucleotide chain, or internally through nick-translation of a polynucleotide chain, through action of a template-dependent or template-independent polymerase enzyme. A nucleotide-5 '-triphosphate is an example of an enzymatically incorporatable nucleotide.
[0043] As used herein, the term "enzymatically extendable" or "3' extendable" means a nucleotide or polynuceotide that is capable of being appended to a nucleotide or polynucleotide by enzyme action. A polynucleotide containing a 3' hydroxyl group is an example of an enzymatically extendable polynucleotide.
[0044] As used herein, the term "halogen" refers to nonmetal elements of Group 7A of the Periodic Table of the Elemnts comprising fluorine, F, chlorine, Cl, bromine, Br, iodine, I, and astatine, At. Halogens are monovalent, readily form negative ions and occur as compounds or ions.
[0045] As used herein, the term "locked nucleic acid" (LNA) refers to a bridge between the 2'0 and 4'C methylene bicyclonucleotide monomers.
[0046] As used herein, the term "nucleobase" refers to a nitrogen containing heterocyclic moiety nucleobase. Non-limiting examples of suitable nucleobases include: adenine, cytosine, guanine, thymine, uracil, 5-propynyl-uracil, 2-thio-5-propynyl -uracil, 5- methylcytosine, pseudoisocytosine, 2-thiouracil, 2-thiothymine, 2-aminopurine, N9-(2- amino-6-chloropurine), N9-(2,6-diaminopurine), hypoxanthine, N9-(7-deaza-guanine), N9- (7-deaza-8-aza-guanine) and N8-(8-aza-7-deazaadenine).
[0047] As used herein, the term "nucleoside" refers to a compound consisting of a nucleobase linked to the C-I ' carbon of a ribose sugar or analog thereof. The ribose or analog may be substituted or unsubstituted. Substituted ribose sugars include, but are not limited to, those riboses in which one or more of the carbon atoms, preferably the 3'-carbon atom, is substituted with one or more of the same or different substituents such as -R, -OR, -NRR or halogen (e.g., fluoro, chloro, bromo, or iodo), where each R group is independently -H, Ci-C6 alkyl or C3-C14 aryl. Particularly, riboses are ribose, T- deoxyribose, 2',3'-dideoxyribose, 3'-haloribose (such as 3'-fluororibose or 3'-chlororibose) and 3'-alkylribose. Typically, when the nucleobase is A or G, the ribose sugar is attached to the N9-position of the nucleobase. When the nucleobase is C, T or U, the pentose sugar is attached to the N'-position of the nucleobase (Kornberg and Baker, DNA Replication, 2nd Ed., Freeman, San Francisco, CA, (1992)). Examples of ribose analogs include arabinose, 2'-O-methyl ribose, and locked nucleoside analogs (e.g., WO 99/14226), for example, although many other analogs are also known in the art.
[0048] As used herein, the term "nucleotide" refers to a phosphate ester of a nucleoside as a monomer unit or within a polynucleotide.
[0049] As used herein, the term "nucleotide triphosphate" refers to a nucleotide with a triphosphate ester group at the 5' position.
[0050] As used herein, nucleosides and/or nucleotides of the present teachings can comprise "natural sugars" (i.e., -ribose, 2'-deoxyribose, and the like) or sugar analogues.
[0051] As used herein, the term "sugar analog" refers to analogs of the sugar ribose. Exemplary ribose sugar analogs include, but are not limited to, substituted or unsubstituted furanoses having more or fewer than 5 ring atoms, e.g., erythroses and hexoses and substituted or unsubstituted 3-6 carbon acyclic sugars. Typical substituted furanoses and acyclic sugars are those in which one or more of the carbon atoms are substituted with one or more of the same or different -R, -OR, -NRR or halogen groups, where each R is independently -H, (Ci-Cβ) alkyl or (C1-Ci4) aryl. Examples of substituted furanoses having 5 ring atoms include but are not limited to 2'-deoxyribose, 2'-(Ci-C6)alkylribose, 2' -(C1- CόJalkoxyribose, 2'-(C5-Ci4)aryloxyribose, 2',3'-dideoxyribose, 2',3'-didehydroriboseJ 2'-deoxy-3'-haloribose, 2'-deoxy-3'-fluororibose, 2'-deoxy-3'-chlororibose, 2'-deoxy- 3'-aminoribose, 2'-deoxy-3'-(Ci-C6)alkylribose, 2'-deoxy-3'-(Ci-C6)alkoxyribose, 2'-deoxy-3'-(C5-Ci4)aryloxyribose, 3'-(Ci-C6)alkylribose-5'-triphosphate, 2'-deoxy-3'-(Ci- C6)alkylribose-5'-triphosphate.2'-deoxy-3'-(Ci -C6)alkoxyribose-5 '-triphosphate, T- deoxy~3'-(Cs-C]4)aryloxyribose-5'-triphosphate, 2'-deoxy-3'-haloribose-5'-rriphosphate, 2'-deoxy-3'-aminoribose-5'-triphosphate, 2',3'-dideoxyribose-5'-triphosphate or 2',3'-didehydroribose-5'-triphosphate. Further sugar analogs also include so called locked nucleic acids (LNAs) having the structure
Figure imgf000010_0001
and those described in Wengel, et al. WO 99/14226, incorporated herein by reference.
[0052] As used herein, the terms "polynucleotide", "oligonucleotide" and "nucleic acid' are used interchangeably and refer to single stranded and double stranded polymers of nucleotide monomers, including ribonucleotides (RNA) and 2'-deoxyribonucleotides (DNA) linked by internucleotide phosphodiester bond linkages. A polynucleotide may be composed entirely of deoxyribonucleotides, entirely of ribonucleotides or chimeric mixtures thereof.
[0053] As used herein, the term "terminator" means an enzymatically incorporatable nucleotide which prevents subsequent incorporation of nucleotides to the resulting polynucleotide chain and thereby halts polymerase-mediated extension. Typical terminators lack a 3'-hydroxyl substituent and include 2',3'-dideoxyribose, 2\,3'-didehydroribose, and 2',3'- dideoxy-3'-haloribose5 e.g.3'-deoxy-3'-fluoro-ribose or 2',3'-dideoxy-3r-fluororibose, for example. Alternatively, a ribofuranose analog can be used, such as 2',3'-dideoxy-β-D- ribofuranosyl, β-D-arabinofuranosyl, 3'-deoxy-β-D-arabinofuranosyl, 3'-amino-2',3'- dideoxy-β-D-ribofuranosyl, and 2',3'-dideoxy-3'-fluoro-β-D-ribofuranosyl (see, for example, Chidgeavadze et al., Nucleic Acids Res., 12: 1671-1686 (1984), and Chidgeavadze et al. FEB. Lett., 183: 275-278 (1985)). Nucleotide terminators also include reversible nucleotide terminators (Metzker et al. Nucleic Acids Res., 22(20):4259 (1994)).
[0054] As used herein, the term "nonextendable" or "31 nonextendable" refers to the fact that a terminator is incapable, or substantially incapable, of being extended in the 3' direction by a template -dependent DNA or RNA polymerase.
[0055] As used herein, the term "TBDMS refers to /ert-butyldimethylsilyl.
[0056] As used herein, the term "void" refers to the absence of a substituent group at the Ri position of the cap analog. The lack of a substituent group results in no positive chare o the imidazole ring. In one embodiment the substituent group may be a CH3 group. When the CH3 group is present, there is a positive charge on the imidazole ring.
[0057] Cap analog is used for the synthesis of 5' capped RNA molecules in in vitro transcription reactions. Substitution of cap analog for a portion of the GTP in a transcription reaction results in the incorporation of the cap structure into a corresponding fraction of the transcripts. Capped mRNAs are generally translated more efficiently in reticulocyte lysate and wheat germ in vitro translation systems. It is important that in vitro transcripts be capped for microinjection experiments because uncapped mRNAs are rapidly degraded. Cap analogs are also used as a highly specific inhibitor of the initiation step of protein synthesis.
[0058] In one embodiement novel ARCA, double- and triple-methylated cap analogs are disclosed. The cap structures have been synthesized and designed to attach in the forward orientation, i.e., 5'Gppp5'GpNpNp.... The resulting novel cap analogs have been demonstrated to improve the yield and efficiency of transcription compared to the standard cap analog as shown in Figures 6, 8, and 10.
[0059] These analogs also have novel substituent groups at the 2' and/or 3' positions of the ribose ring which also resulting in attaching of the cap in the forward orientation. In one embodiement, fluorine attached at the 2' or 3' position of the ribose.ring has been shown to improve both capping efficiency and translation efficiency as shown in Figures 11 and 12.
[0060] The synthesis of capped RNA which is not only efficiently capped and yields high levels of transcribed RNA is an area of unmet need. One approach which is being used by Ambion is disclosed in US patent application 2005/0287539, incorporated herein by reference. The synthesis of cap analogs which incorporate in only the forward orientation will improve transcription and translation efficiency.
[0061] The teachings of Darynkiewicz et al. in US Patent No. 7,074,596 have attemped to present synthesis methods and methylation methods for anti-reverse cap analogs. The method of methylation as taught was not reproducible in our hands, neither was the synthesis of a linker and methods for linking the nucleotides.
[0062] Thus, there exists in the art an unmet need for high yield transcription reactions that efficiently synthesize RNA. The resulting RNA finds use in a variety of applications, including ribozyme, antisense and biophysical studies, and gene array analysis. Additionally, capped RNA transcripts are used for applications requiring protein synthesis such as in vivo expression (e.g., microinjection, transfection and infection experiments) and in vitro translation.
[0063] In order to overcome the problem of synthetic mCAP attaching to RNA in the reverse orientation around 50% of the time, when NTPs and cap RNA are present in comparable concentrations, the present application teaches chemically convenient and reproducible methods for the synthesis of modified cap analog, anti-reverse cap analog (ARCA), and double- and triple- methylated cap analogues (Figures 1 and 2). [0064] The design and synthesis of novel cap analogs such as m7,2'FG[5']ppp[5']G and m7,2TG[5']ppp[5']m7G, a double-methylated cap, in which various moieties at the 2' and 3' positions on the ribose ring have been substituted are presented (Figure 2).
[0065] Also discovered and synthesized are triple-methylated caps. Such caps include m27,2'OG[5I]ppp[5I]m7G, m27,2OG[5']ppp[5'] m2'OG5 m2753'OG[5l]ppp[5I]m7G, and m2752OG[5']ppp[5'] m27,2'OG. Additionally, details of the compatibility of new, modified cap analogs with respect to transcription, capping and translation efficiency by using HeLa cells in comparison with the standard and conventional ARCA cap analog is presented.
[0066] Structures were confirmed by 1H NMR and 31P NMR. Transcripts produced with T7 RNA polymerases using "anti-reverse" cap analogs (ARCAs) were of the predicted length and indistinguishable in size and homogeneity from those produced with m7GpppG. Transfection assays were performed by using standard cap analog-capped and ARCA- capped luciferase in in vitro transcribed RNA with HeLa cells. Finally, luciferase activity was measured and revealed that ARCA-capped transcripts were 2.2 to 2.5 fold higher than that of m7GpppG-capped transcripts. This finding also suggest that the presence of reverse caps in conventional in v/ϊro-synthesized mRNA reduces their translation efficiency.
[0067] Those having ordinary skill in the art will understand that many modifications, alternatives, and equivalents are possible.
[0068] All such modifications,- alternatives, and equivalents are intended to be encompassed herein.
MATERIALS AND METHODS Reagents
[0100] AU of the reagents and solvents are used as such without further purification, unless otherwise stated. Guanosine 5 '-diphosphate, Dimethyl sulfate, anhydrous dimethylformamide , 2,2'-dithiodipyridine (Aldrithiol), Triphenylphosphine, trimethylphosphate ((OMe)3P), phosphorous oxychloride, phosphorous pentoxide, orthophosphoric acid, anhydrous methylene chloride, dichloromethane, Tributylamine, anhydrous pyridine were purchased from Sigma-Aldrich Co. 3'-O-Me-Guanosine is available from Chemgene, Boston, MA. Imidazolide GMP, lmidazolide GDP, Imidazolide 2'F-GMP, Imidazolide 3'CF3-GDP, Imidazolide m7GMP, IM tris(triethylammonium) phosphate, and tributylammonium orthophosphate were made as taught herein or in A. Kore, and G. Parmar, Synthetic Comm., 36:3393-3399, 2006, incorporated herein by reference in its entirety.
[0101] The cap analogs were analyzed by 1H NMR and 31P NMR (Broker Avance), 400 MHz. 1H was collected at 400.1446006 MHz and the 31P was collected at 161.9968531, both using a QNP probe. Mass Spectroscopy (i.e., Applied Biosystems/Sciex MDX API 150 model) and MALDI-TOF (Applied Biosystems Voyager DE-PRO model), and analytical HPLC (Aliance, Water's) was performed using Hypersil SAX columns, 5 μm, 250 X 4.6 mm (Altech).
EXAMPLES
[0102] Aspects of the present teachings may be further understood in light of the following examples, which should not be construed as limiting the scope of the present teachings in any way.
[0103] While the present teachings are described in conjunction with various embodiments, it is not intended that the present teachings be limited to such embiodiements. On the contrary, the present teachings encompass various alternatives, modifications, and equivalents, as will be appreciated by those of skill in the art.
EXAMPLE l
[0104] Synthesis of 7-Methyl Guanosine 5'-diphosphate In a clean, dry 2000 mL round bottom flask equipped with a stirring bar and under a stream of argon slowly dissolve dry and finely powdered guanosine 5 '-diphosphate (1), (10.0 g, 20.5 mmol), either in free acid or sodium as a counter ion form, in 200 mL water, adjusting pH to 4.0 with glacial acetic acid. Dimethyl sulfate (20 mL, 119.04 mmol) was then added over a period of one hour with constant stirring at room temperature and the reaction continued for an addition hour during which time a decrease in pH was observed but pH was kept between pH 3.8 to 4.0 by drop-wise addition of 10 mM NaOH and methylations was monitored by analytical HPLC for progress. Methylation was determined to be 98% complete within 2 h. After 2 h, the reaction mixture was extracted with CHCl3(3 X 200 mL) to remove unreacted excess dimethyl sulfate.
[0105] The resulting aqueous layer was further evaporated on a rotary evaporator to remove any chloroform traces, and then further diluted to 1.5 L with water and loaded on an anion exchange resin, i.e., DEAE Sepharosa fast flow packed in a BPG 100 column (Amersham GE, Piscataway, NJ, USA). BPG (biological process glass) 100 specification: 100/500 column (100 mm in diameter and 50 cm in height), packed with DEAE Sepharosa fast flow resin to the bed volume of 400 mm. The desired compound was eluted by using four bed volumes of gradient from 0 to 80% of 1 M TEAB buffer (triethyammonium bicarbonate), pH 7.5, at a flow rate of 100 mL/min, using AKTA purifier 100 FPLC (Amersham GE). At 45% TEAB buffer, 7-methylguanosine 5 '-diphosphate (m7GDP) was eluted as a large broad peak, with a strong ultraviolet absorbance at 254 run. The residual bicarbonate was removed by co-evaporating with methanol, 3 X 600 mL. The resulting residue was transferred to a centrifuge tube, and 8.9 g sodium perchlorate dissolved in 1.1 L acetone was added and cooled 2 h at 40C. The resulting mixture was centrifuged and the supernatant liquid was discarded. The precipitate was ground with a new portion of acetone, cooled and centrifuged, repeating once. The precipitate was dried in a vacuum desiccator over P2O5. The resulting amorphous white powder was 7 methyl-guanosine 5'- diphosphate. Taken from Kore,A. and Parmar,G. Nucleosides, Nucleotides, and Nucleic Acids, 25:337-340, 2006, incorporated herein by reference in its entirety.
EXAMPLE 2 Synthesis of NucIeoside-5'-diphosphates
[0106] Although the following procedure illustrates synthesis of guanosine-5'- diphosphate, one of skill in the art would be able to use the procedure for the synthesis of adenosine-5' -diphosphate, uridine-5'-diphosphate, and cytidine-5'-diphosphate and analogs thereof. In a clean, dry 500 mL round bottom flask equipped with a stirring bar the triethylammonium salt of guanosine 5 '-monophosphate (10.0 g, 21.5 mmol) in anhydrous dimethylformamide (200 mL) was stirred together, triethylamine was added (2.4 mL, 142.8 mmol) and allowed to stir for 5 min, followed by the addition of Imidazole (5.86 g, 86.1 mmol), 2,2'-dithiodipyridine (7.4 g, 33.58 mmol), and triphenylphosphine (8.9 g, 33.9 mmol). Stirring was continued for 2 h at room temperature. The reaction was allowed to go to completion as determined by HPLC and then poured slowly into a mixture of sodium perchlorate (7 g) in acetone (1500 mL), and then cooled for 30 min. at 40C. The reaction mixture was centrifuged, discarding the supernatant. Traces of imidazole and triphenylphosphine were removed by grinding the solid with a new portion of acetone (400 mL), cooling and again centrifuged, repeating once. The precipitate was dried in a vacuum oven over P2O5 at 240C (30 mbar pressure). The ribonucleoside-5'-phosphoroimidazolide thus obtained was dissolved in dimethylforamide (200 mL), and a 1 M solution of tributylammonium orthophosphate in dimethylformamide (80 mL) was added drop-wise to the vigorously stirred mixture over a period of 30 min. Zinc chloride (2 g, 14.67 mmol) was added and the reaction mixture stirred at room temperature for 3 h. Completion of the reaction was monitored by HPLC. The reaction mixture was quenched with water (50 mL) and extracted with chloroform (3 X 200 mL), concentrated in a rotary evaporator and then purified by application to an anion exchange resin.
[0107] Purification by column chromatography was accomplished with a DEAE Sepharose fast flow resin packed in an XK 50/60 column (50 mm diameter and 60 cm long) (Amersham GE). The desired compound was eluted by using four bed volumes of gradient from 0 to 80% of 1 M TEAB buffer pH 7.5 (triethylammonium bicarbonate) at a 'flow rate of 20 mL/min, using an AKTA purifier 100 FPLC (Amersham GE). At 55% TEAB buffer, the desired product (nucleoside-5'-diphosphate) was eluted as a large broad peak, with a strong ultraviolet absorbance at 254 nm. The nucleoside-5'-diphosphate-containing fractions were pooled and evaporated using a rotary evaporator to give triethylamine salt of the desired diphosphate compound. Taken from A. Kore, and G. Parmar, Synthetic Comm., (2006) supra.
EXAMPLE 3
Synthesis of 3'-O-Methyl Guanosine Monophosphate (3'-0-Me-GMP) TEA Salt (Compound 2)
[0108] In a clean, dry 500 mL round bottom flask equipped with a stirring bar and under a stream of argon slowly add dry and finely powdered 3'-O-Me-Guanosine (1), (6 g, 20 mmol) to a mixture of trimethylphosphate ((OMe)3P) (50 mL) and phosphorous oxychloride (POCI3) (6 mL, 60 mmol) at O0C in small portions with continuous stirring under argon. The mixture was kept at 0 — 40C and allowed to stir at least 19 hrs. Diethyl ether (200 mL) was added to extract the excess phosphorous oxychloride and to simultaneously precipitate the 3'-O-methylguanosine-5'-phosphodichloridate, which was then pelleted by centrifugation and dissolved in 100 mL ice-cold 5% NaHCO3 in water. The resulting aqueous solution was adjusted to pH ~1.5 using 1 N NaOH. After stirring at 0 - 4 0C for an additional 20 h, the pH was adjusted to 7.0 and the resulting mixture was applied to a column of DEAE Sephadex A25. The column was washed with 5 mM TEAB buffer, pH 7.5 and then eluted with freshly prepared IM Triethylammonium bicarbonate (TEAB) buffer, pH 7.5. Fractions containing the 3'-O-Me GMP TEA salt (2) were pooled, concentrated to dryness.
EXAMPLE 4 Synthesis of 3'-0-Me-GMP Imidazolide (Compound 3)
[0109] In a clean, dry 1 L round bottom flask equipped with a stirring bar and under a stream of argon slowly add anhydrous DMF (144 mL) and the triethylamine (0.933 mL, 9.23 mmol) allow to stir for at least 5 min. To this slowly was add the dry and finely powdered 3'-0-Me-GMP TEA salt (2), (3.5 g, 7.34 mmol) in small portions with continuous stirring under argon. Thereafter the Imidazole (2.05 g, 30.1 mmol), Aldrithiol (2.65 g, 12.02 mmol), and triphenylphosphine (3.13 g, 11.9 mmol) were added and the reaction allowed to stir at room temperature for at least 2-3 h, during which the reactants became soluble making the reaction appear clear yellow colored. Upon completion, sodium perchlorate (3.0 g, 24.5 mmol) dissolved in acetone with continuous stirring and to this mixture, slowly added, was the reaction mixture. This mixture was then poured in two IL nalgene bottles and cooled in a refrigerator at -8O0C for 30 minutes. The mixture was then subjected to centrifugation at 3000 rpm for 15 min and the supernatant was discarded. The precipitate was ground with a new portion of acetone and centrifuged. The process was repeated once more and the precipitate was dried in a vacuum desiccator over phosphorous pentoxide, yielding 3'-0-Me-GMP Imidazolide.
EXAMPLE 5 Synthesis of 3'0-Me-GDP TEA salt (Compound 4)
[0110] Synthesis of Tris(triethylammonium) phosphate linker:
Anhydrous orthophosphoric acid (22.5 g, 229.59 mmol) was added to 50 mL of anhydrous methylene chloride in a clean, oven dried 250 mL flask equipped with a stirring bar. Tributylamine (54.6 mL, 229.6 mmol) was then added into the solution drop wise through an addition funnel over a period of 30 min. The mixture was left stirring for 1 h. CH2Cl2 was then evaporated and the reaction residue was co-evaporated with 3 X 30 mL of anhydrous pyridine and then 2 X 30 mL of anhydrous DMF. The Tris(triethylammonium) phosphate linker product was dissolved in 100 mL anhydrous DMF so as to have a final concentration of 1 M, and stored over 4A molecular sieves at 40C. [0111] In a clean, dry IL round bottom flask equipped with a stirring bar and under a stream of argon anhydrous DMF (40 mL) was slowly added and stirred for at least 5 minutes. To this was slowly added finely powdered 3'-0-Me-GMP Imidazolide (3), (3.0 g, 7.04 mmol) in small portions with continuous stirring under argon. Zinc chloride (2.0 g, 14.6 7 mmol) was added in small portions until the contents were dissolved. Thereafter, the tris(triethylammonium) phosphate linker andl M tributylammonium orthophosphate (40 mL) was added slowly to the reaction mixture under argon and the reaction was allowed to stir at room temperature for 5 h. The reaction when followed on HPLC showing complete conversion of the starting material, 3'-0-Me-GMP Imidazolide (3) to its corresponding diphosphate. Upon completion, the reaction was supplemented with water, 100 mL, and the resultant mixture was extracted with chloroform (3 X 250 mL), subjected to volume reduction (~100 mL) by evaporation and applied to DEAE Sephadex A25 column, eluting with a linear gradient of freshly prepared 1 M TEAB, pH 7.5. The fractions containing the pure 3'O-Me-GDP TEA salt (compound 4) were eluted, combined and evaporated to dryness.
EXAMPLE 6 Synthesis of m2 7>3'0GDP (Compound 5)
[0112] To a stirred solution of 3'-0-Me-GDP TEA salt (4), (4.0 g, 6.1 mmol) in 100 mL of nuclease free water, concentrated glacial acetic acid was slowly added to adjust the pH of the solution to 4.0; dimethyl sulfate ((Me)2SO4) (20 mL, 210 mmol) was slowly added drop wise over a period of 60 min, while maintaining the pH~4.0-4.5 with 50 mM NaOH. The reaction was allowed to stir at room temperature for 2 h and methylation was > monitored by HPLC. After 2 h, the reaction mixture was extracted with CHCl3 (3 X 250 mL) to remove unreacted dimethyl sulfate. The aqueous layer was applied to a DEAE Sephadex column and the fractions containing the product were pooled, evaporated and dried in a vacuum desiccator over phosphorous pentoxide to give 3'-O-Me (N7-Me) GDP (compound 5) as a fine powder.
EXAMPLE 7 Synthesis of m2 7l3 OG[5']ppp[5']G (Compound 7)
[0113] In a clean, dry IL round bottom flask equipped with a stirring bar and under a stream of argon anhydrous DMF (165 mL) was slowly added and allowed to stir. To this was slowly add finely powdered 3'-0-Me-GDP TEA salt (5), (4.0 g, 5.84 mmol) in small portions with continuous stirring under argon. Zinc chloride (2.0 g, 14.67 mmol) was added in small portions until the contents were dissolved. Thereafter, the Imidazolide GMP (compound 6) (6.00 g, 14.08 mmol) was slowly added to the reaction mixture under argon. The reaction was allowed to stir at room temperature and methylation was monitored by HPLC. Upon complete disappearance of starting material, 3'-0-Me-GDP TEA salt, the reaction was poured into a solution of EDTA (3.0 g) in water (200 mL) and pH adjusted to 7.0 with saturated NaHCOs. This was then applied to a DEAE Sephadex column, eluted with a linear gradient of freshly prepared 1 M TEAB, pH 7.5 and the fractions containing the product were pooled, evaporated and dried to give pure 3'-O-Me-N7-Me-G[5']ppp[5']G (compound 7), stored at -2O0C.
EXAMPLE 8 Synthesis of m2 7>3'oG[5']ppp[5']in7G (Compound 8)
,[0114] To a stirred solution of compound 7 (500 mg, 0.612 mmol) in 5.0 mL water, concentrated glacial acetic acid was added slowly to adjust the pH of the solution to 4.0 and to this mixture dimethyl sulfate (2.0 mL, 21.1 mmol) was slowly added drop-wise over a period of 60 min, while maintaining the at pH~4.0-4.5 with 50 mM NaOH. The reaction was allowed to stir at room temperature for 2 h and methylation was monitored by HPLC. After 2 h, the reaction mixture was extracted with CHC13 (3 X 50 mL) to remove unreacted dimethyl sulfate. The aqueous layer was applied to a DEAE Sephadex column and the fractions containing the product were pooled, evaporated and dried to give pure trimethylated CAP (m27,3OG[5']ppp[5']m7G ). (compound 8) which was then passed through a Strata AW column to remove the salt and make it a free acid form.
EXAMPLE 9 Synthesis of 2'F GMP (Compound 14):
[0115] To a stirred solution OfPOCl3 (3.97 g, 26.3 mmol) and (OMe)3P (20.0 mL) at O0C under nitrogen atmosphere, 2'-fluro guanosine (2.5 g, 8.77 mmol) was added and the reaction mixture was stirred for 2 h at O0C. After 2 h, 50.0 mL water was added to the reaction mixture. The resulting reaction mixture was washed with ethyl acetate (2 X 50 mL) to remove the phosphorylating agent. The collected aqueous solution was adjusted to pH 1.5 drop-wise with IN NaOH and allowed to stir at 40C for 12 h. After 12 h, the aqueous solution was concentrated under a rotor evaporator to around 20 mL. To a solution of sodium perchlorate (3.0 g) in 100 mL acetone in a centrifuge tube at O0C this concentrated solution (20.0 mL) was added slowly for 2 minutes. The resulting mixture was centrifυged and the supernatant liquid was removed. This precipitate was dissolved in 500 mL water and adjusted the pH to 5.5 with saturated NaHCCb and loaded on a DEAE Sephadex column. The desired product was eluted using a linear gradient of 0-lM TEAB buffer, pH 7.5 and the fractions containing the product were pooled, evaporated and dried in a vacuum desiccator over phosphorous pentoxide (P2O5) to give 2'F GMP as a fine white powder (compound 14) (Yield: 3.54 g, 87%).
EXAMPLE 10 Synthesis of Imidazolide 2'F GMP (Im2'F GMP) (Compound 15):
[0116] To a stirred solution of 21F GMP TEA salt 14 (2.0 g, 4.3 mmol) in a 50 mL dry DMF5 imidazole (1.50 g, 21.5 mmol), triphenyl phosphine (PPh3) (2.26 g, 8.6 mmol), aldrithiol (1.90 g, 8.6 mmol) and triethylamine (0.43 g, 4.3 mmol) were added. The reaction mixture was stirred under nitrogen atmosphere at room temperature for 5 h. To a solution of sodium perchlorate (2.0 g) in 100 mL acetone in a centrifuge tube at 00C, the above reaction mixture was added slowly for 5 minutes. The resulting mixture was centrifuged and the supernatant liquid was removed. The solid was ground with a new portion of acetone (100 mL), cooled, and centrifuged again. This process was repeated twice, and the resulting solid was dried in a vacuum desiccator over P2O5 to give Imidazolide 21F GMP as a white powder (compound 15) (Yield: 1.44 g, 81%).
EXAMPLE 11 Synthesis of 2'F GDP (Compound 16):
[0117] To a stirred solution of Imidazolide 21F GMP 15 (1.2 g, 2.89 mmol) and Zinc chloride (0.38 g, 2.89 mmol) in 10.0 mL dry DMF, 18 mL of IM tris(triethylammonium) phosphate (tris(Et3NH)3PO4) in DMF was added under nitrogen atmosphere. The reaction mixture was stirred at room temperature for 3 h. After 3 h, the reaction mixture was diluted with 50.0 mL of water. The resulting reaction mixture was washed with ethyl acetate (2 X 50 mL) to remove the phosphorylating agent. The collected aqueous solution was adjusted to pH 5.5 with saturated NaHCCb and loaded on a DEAE Sephadex column. The desired product was eluted using a linear gradient of 0 - IM TEAB buffer, pH 7.5 and the fractions containing the product were pooled, evaporated and dried in vacuum desiccator over phosphorous pentoxide to give 2'F GDP as a fine white powder (compound 16) (Yield 134 g, 72%).
EXAMPLE 12 Synthesis of m7'2 ;rGDP (Compound 17):
[0118] To a stirred solution of 2'F GDP 16 (0.75 g, 1.16 mmol) in 20.0 mL of water, acetic acid was added slowly to adjust the pH of the solution to 4.0. To this mixture, dimethyl sulfate (3.0 mL) was added drop-wise over a period of 30 min. and the reaction mixture was allowed to stir at room temperature for 4 h. As the methylation proceeds, the pH decreased to around pH 2.0 and was re-adjusted back to pH 4.0 using a IM NaOH solution. After 4h, the reaction mixture was extracted with CHC13 (3 X 50 mL) to remove unreacted, excess dimethyl sulfate. The collected aqueous solution was adjusted to pH 5.5 with saturated NaHCC>3 and applied onto a DEAE Sephadex column. The final product was monitored for methylation by HPLC?? αn<i eluted using a linear gradient of 0 - IM TEAB buffer, pH 7.5 and the fractions containing the product were pooled, evaporated and dried in vacuum desiccator over phosphorous pentoxide to give m7>2/rGDP as a fine white powder (compound 17) (Yield 0.64 g, 83%).
EXAMPLE 13 Synthesis of m7>2 fGpppG (Compound 18):
[0119] To a stirred solution of m^GDP 17 (0.2 g, 0.3 mmol) and Imidazolide GMP (ImGMP) (0.19g, 0.45 mmol) in 10.0 mL dry DMF, Zinc chloride (81 mg, 0.6 mmol) was added under nitrogen atmosphere and the reaction mixture was stirred at room temperature for 1 h. After 1 h, the reaction mixture was added to a solution of EDTA disodium (0.45 g, 1.2 mmol) in 100.0 mL of water at 0°C. The resulting aqueous solution was adjusted to pH 5.5 with saturated NaHCO3 and loaded on a DEAE Sephadex column. The desired product was eluted using a linear gradient of 0 - IM TEAB buffer, pH 7.5 and the fractions containing the product were pooled, evaporated and concentrated to 10.0 mL TEA salt of compound 18. The resulting 10.0 mL was passed through a Strata-X-AW column and washed with 10.0 mL water followed by 10.0 mL MeOH. Then, compound 18 was eluted with 15.0 mL OfNH4OHTMeOHZH2O (2/25/73) and the collected solution was evaporated and dried to give m7>2VrGpppG as a fine white powder (compound 18) (Yield: 0.15 g, 58%).
[0120] HPLC Analysis: The starting imidzaolide gave a peak at 3.49 min. and the GDP peak was at 5.95 min. The new product peak was at 7.13 min. The crude reaction mixture after 1 h gave 97% of the product peak (7.14 min.) and 3% of the starting material peak (5.95 min.).
[0121] Data for 18. 1H NMR (D20, 400 MHz) δ 8.02 (s, IH)5 6.14 (d, J= 14.4 Hz, IH), 5.80 (d, J= 5.6 Hz, IH), 5.41 - 5.27 (m, IH), 4.63 - 4.47 (m, 3H), 4.39 -4.24 (m, 6H), 4.03 (s, 3H); 31P NMR (D20, 162 MHz) δ -10.4 (d, J = 16.8 Hz), -10.54 (d, J= 18.6 Hz), -21.95 (t, J= 19.3 Hz); 19F NMR (D2O, 376 MHz) δ -20.47 (m); MS (m/z): 803
EXAMPLE 14
Figure imgf000022_0001
Synthesis of m7l 2'"FG[S']ppp[5']m7G (Compound 20):
[0122] To a stirred solution of m^GDP 17 (0.25 g, 0.38 mmol) and Imidazolide m7GMP (m7-ImGMP) (0.24g, 0.57 mmol) in 10.0 mL dry DMF, Zinc chloride (0.10 g, 0.76 mmol) was added under nitrogen atmosphere and the reaction mixture was stirred at room temperature for 6 h. After 6 h, the reaction mixture was added to a solution of EDTA disodium (0.57 g, 1.52 mmol) in 100.0 mL of water at 00C. The resulting aqueous solution was adjusted to pH 5.5 with saturated NaHCOsand loaded on a DEAE Sephadex column. The desired product was eluted using a linear gradient of 0 - IM TEAB buffer, pH 7.5 and the fraction OCLs=- containing the product were pooled, evaporated and concentrated to 10.0 mL TEA salt of compound 20. The resulting 10.0 mL was passed through a Strata-X-AW column and washed with 10.0 mL water followed by 10.0 mL MeOH. Then, compound 20 was eluted with 15.0 mL OfNH4OHZMeOHZH2O (2Z25Z73) and the collected solution was evaporated and dried to give m7> 2 "FGpρpm7G as a fine white powder (compound 20) (Yield: 0.21 g, 64%).
[0123] Data for 20: 1HNMR (D2O, 400 MHz) δ 6.31 (d, J= 14.8 Hz, IH), 6.02 (d, J- 3.2 Hz, IH), 5.53 - 5.37 (m, IH), 4.75 - 4.62 (m, 2H), 4.52 - 4.38 (m, 5H)54.30 - 4.21 (m, 2H), 4.1 1 (s, 6H); 31P NMR (D20, 162 MHz) δ -10.35 (d, J= 11.8 Hz), -10.50 (d, J= 10.0 Hz), -21.80 (t, J= 19.1 Hz); 19FNMR (D20, 376 MHz) δ -20.51 (m); MS (mZz): 817 [M]+.
EXAMPLE 15 rt
Figure imgf000023_0001
Synthesis of m2 7'2'0G[5'lppp[5']m2'0G (Compound 23):
[0124] To a stirred solution of m2 7'2'°GDP (N7Me2'O-MeGDP) 21 (0.22 g, 0.33 mmol) and Imidazolide m2'°[GMP (Im2'O-MeGMP) 22 (0.14g, 0.33 mmol) in 10.0 mL dry DMF5 Zinc chloride (0.09 g, 0.66 mmol) was added under nitrogen atmosphere and the reaction mixture was stirred at room temperaturefor 2 h. After 2 h, the reaction mixture was added to a solution of EDTA disodium (0.49 g, 1.32 mmol) in 100.0 mL of water at 00C. The resulting aqueous solution was adjusted to pH 5.5 with saturated NaHCO3 and loaded on a DEAE Sephadex column. The desired product was eluted using a linear gradient of 0 - IM TEAB buffer, pH 7.5 and the fractions containing the product were pooled, evaporated and concentrated to 10.0 mL TEA salt of compound 23. The resulting 10.0 mL was passed through a Strata-X-AW column and washed with 10.0 mL water followed by 10.0 mL MeOH. Then, compound 23 was eluted with 15.0 mL OfNH4OHMeOHyH2O (2/25/73) and the collected solution was evaporated and dried to give mj'1 °G[5']ppp[5']m2 0G as a fine white powder (compound 23) (Yield: 0.18 g, 63%).
[0125] Data for 23: 1H NMR (D20, 400 MHz) δ 8.00 (s, 2H), 5.90 (d, J= 2.0 Hz, IH), 5.83 (d, J= 5.2 Hz, IH), 4.59 (t, J= 4.4 Hz, IH), 4.53 - 4.41 (m, 2H), 4.33 - 4.22 (m, 6H), 4.16 (m, IH), 4.06 (s, 3H), 3.60 (s, 3H), 3.45 (s, 3H); 31P NMR (D2O, 162 MHz) δ - 10.35 (t, J= 17.8 Hz), -21.83 (t, J= 18.0 Hz); MS (m/z): 831 [M]+-
EXAMPLE 16
rt
Figure imgf000024_0001
21 24
Figure imgf000024_0002
Synthesis of TO2 712 0G[S5IpPp[S9Im2 712 0G (Compound 25):
[0126] To a stirred solution of m2 7> 2'°GDP (N7Me2 '-0-MeGDP) 21 (0.18 g, 0.27 mmol) and Imidazolide m2 7>2'0GMP (Im N7Me-2'-O-MeGMP) 24 (0.18g, 0.41 mmol) in 10.0 mL dry DMF, Zinc chloride (0.07 g, 0.54 mmol) was added under nitrogen atmosphere and the reaction mixture was stirred at room temperaturefor 6 h. After 6 h, the reaction mixture was added to a solution of EDTA disodium (0.40 g, 1.08 mmol) in 100.0 mL of water at O0C. The resulting aqueous solution was adjusted to pH 5.5 with saturated NaHCO3 and loaded on a DEAE Sephadex column. The desired product was eluted using a linear gradient of 0 - IM TEAB buffer, pH 7.5 and the fractions containing the product were pooled, evaporated and concentrated to 10.0 mL TEA salt of compound 25. The resulting 10.0 mL was passed through a Strata-X-AW column and washed with 10.0 mL water followed by 10.0 mL MeOH. Then, compound 25 was eluted with 15.0 mL of NH4OHyMeOHZH2O (2/25/73) and the collected solution was evaporated and dried to give m2 7'2'°G[5']ppp[5']m2 7l2'oG as a fine white powder (compound 25) (Yield: 0.13 g, 54%).
[0127] Data for 25: 1H NMR (D2O5400 MHz) δ 6.10 (d, J= 2.8 Hz52H)54.59 (t, J = 5.2 Hz, 2H), 4.43 - 4.23 (m5 8H), 4.11 (s, 6H), 3.61 (s, 6H); 31P NMR (D20, 162 MHz) δ -10.32 (d, J= 18.] Hz), -21.72 (t, J= 20.7 Hz); MS (m/z): 845 [M-H]+.
EXAMPLE 17
rt
Figure imgf000025_0001
21
Figure imgf000025_0002
Synthesis of m2 7 roG[5']ppp[5']in7G (Compound 26):
[0128] To a stirred solution of m2 7'2'°GDP (N7Me25 -0-MeGDP) 21 (0.20 g, 0.3 mmol) and Imidazolide m7GDP QmN7MeGMP) (0.19 g, 0.45 mmol) in 10.0 mL dry DMF, Zinc chloride (0.08 g, 0.6 mmol) was added under nitrogen atmosphere and the reaction mixture was stirred at room temperature for 6 h. After 6 h, the reaction mixture was added tb a solution of EDTA disodium (0.45 g, 1.2 mmol) in 100.0 mL of water at O0C. The resulting aqueous solution was adjusted to pH 5.5 with saturated NaHCOsand loaded on a DEAE Sephadex column. The desired product was eluted using a linear gradient of 0 - IM TEAB buffer, pH 7.5 and the fractions containing the product were pooled, evaporated and concentrated to 10.0 mL TEA salt of 26. The resulting 10.0 mL was passed through a Strata-X-AW column and washed with 10.0 mL water followed by 10.0 mL MeOH. Then, compound 26 was eluted with 15.0 mL OfNH4OHZMeOHZH2O (2Z25Z73) and the collected solution was evaporated and dried to give a fine white powder (compound 26) (Yield: 0.16 g, 62%)
[0129] Data for 26: 1H NMR (D20, 400 MHz) δ 6.10 (d, J= 2.8 Hz, IH)5 6.01 (d, J = 3.6 Hz, IH), 4.63 (t, J= 5.6 Hz, IH), 4.58 (t, J= 5.6 Hz, IH)5 4.49 (t, J= 5.6 Hz5 IH), 4.41 - 4.31 (m, 5H), 4.27 - 4.23 (m, 2H), 4.12 (s, 6H), 3.60 (s, 3H); 31P NMR (D2O5 162 MHz) δ -10.36 (d, J= 19.0 Hz)5 -21.77 (t, J= 19.3 Hz); MS (mZz): 829 [M]+.
EXAMPLE 18 T7 RNA Transcription:
[0130] T7 RNA polymerase transcription was performed by using mMESSAGE mMACHINE® T7 Kit (Ambion) in 20 μL final volume, and contains the following reagents at the final concentrations indicated: 1 μg linearized AmbLuc PoIyA DNA, IX reaction buffer,7.5mM of each ATP5 UTP, and CTP5 1.5mM GTP5 6.5 raM of mCAP and 2' fluoro cap analogs, and 50 UZμl of T7 RNA polymerase. The transcription reactions were incubated at 370C for 2 hours. In order to hydrolyze the remaining plasmid DNA5 1 μL of turbo DNAse was added to the reaction mixture, and further incubated at 370C for 15 minutes. The transcribed capped and uncapped mRNAs were purified by using the MEGAclear™ Kit (Ambion).
[0131] AmbLuc PoIy(A) (Ambion), transcription produced comparable yields for modified and standard cap analogs (see Figures 5 and 6). The Bioanalyzer assay indicates that all the mRNAs are not degraded and retain great integrity (see Figure T).
EXAMPLE 19 Translation Assay:
[0132] Protein expression from mCAP, double methylated, triple methylated, and ARCA capped luciferase RNAs, at different time points after transfection with HeLa cells indicates that the transcripts containing triple methylated cap are more highly translated in transfected cells. Comparison of protein expression with mCAP and double methylated cap suggest that the double methylated cap transcript produces one fold higher protein, while ARCA produces 3 fold higher, and triple methylated cap produces 4 fold higher protein in in vivo with HeLa cells. See Figure 9.
[0133] All modified cap compounds tested are substrates for T7 polymerase, as determined by gel shift assays that clearly revealed that capped RNAs were formed, based on slower migration relative to uncapped RNAs (see Figures 8 and 10).
[0134] The examples above described particular cap analogs, however, there are additional cap analogs conceived which will also function in practicing the invention as illustrated below:
Figure imgf000027_0001
General Structure of CAP
Figure imgf000027_0002
Figure imgf000028_0002
EXAMPLE 20
Figure imgf000028_0001
[0135] The coupling reaction of m^'^GDP (N7Me-2'FGDP) with Imidazolide 27OMP (lm2'FGMP), or Imidazolide m7>2'FGMP (N7MeIm2TGMP) in the presence of ZnCI2 as the catalyst affords the corresponding cap analogs A and B.
EXAMPLE 21
Figure imgf000029_0002
[0136] The coupling reaction of m7>3WHjGDP (N7Me-3 'NH2GDP) with Imidazolide 277GMP (lm2'FGMP), or Imidazolide m^GMP (N7MeIm2'FGMP) in the presence of ZnCl2 as the catalyst affords the corresponding cap analogs E and F.
EXAMPLE 22
Figure imgf000029_0001
The coupling reaction of m7'3VWj2dGDP (N7Me-3'NH22'dGDP) with the corresponding substituted Imidazolide GMP (ImGMP), Imidazolide m7GMP (m7-ImGMP), Imidazolide 2F GMP (lm2'FGMP), or Imidazolide m7"2"^ GMP (N7MeIm2 'FGMP)5 respectively, in the presence OfZnCl2 as the catalyst affords the corresponding cap analogs K, L5 M, and N.
Figure imgf000030_0001
Scheme to Make C, D, G, H, I, J, O, P, Q, and R:
EXAMPLE 23
Scheme to Make m7l3'CFjG[5 ]ppp[5']G and πT^'Gβ'lpppβ ] m7G: [0138] The coupling reaction of m7-ycFjGO? (N7Me-3 'CF3GDP) with Imidazolide
GMP (ImGMP) or Imidazolide m7GMP (m7-ImGMP) in the presence of ZnCl2 as the catalyst affords the corresponding cap analogs C and D.
EXAMPLE 24
Figure imgf000030_0002
[0139] T he coupling reaction of m7'3WO'GDP (N7Me-3 'NO2GDP) or m7-3w^GDP (N7Me-3'NH3GDP) with Imidazolide GMP (ImGMP) or Imidazolide m7GMP (m7- ImGMP) in the presence Of ZnCl2 as the catalyst affords the corresponding cap analogs G, H, I, and J.
EXAMPLE 25
Figure imgf000030_0003
[0140] T he coupling reaction of m7'3W<jDP (N7Me-3 'N3GDP) with Imidazolide
GMP (ImGMP) or Imidazolide m7GMP (m7-ImGMP) in the presence OfZnCl2 as the catalyst affords the corresponding cap analogs O and P.
EXAMPLE 26
Figure imgf000030_0004
[0141] The coupling reaction of m7>3Wj2'dGDP (N7Me-3'N32'dGDP) with Imidazole GMP (ImGMP) or Imidazolide m7GMP (m7-ImGMP) in the presence OfZnCl2 as the catalyst affords the corresponding cap analogs Q and R.
EXAMPLE 27
Figure imgf000031_0001
The coupling reaction of m2 7>3 OGDP (N7Me-3 OMe-GDP) with Imidazolide m2'°GMP (Imm2'°GMP) or Imidazolide m2 7a'oGMP (N7Me-2 'OMeImGMP) in the presence OfZnCl2 as the catalyst affords the corresponding cap analogs S and T.
EXAMPLE 28
Figure imgf000031_0002
The coupling reaction of m7'3'c"°GDP (N7Me-3 'CHOGDP) with Imidazolide GMP (ImGMP) or Imidazolide m7GMP (m7-ImGMP) in the presence OfZnCl2 as the catalyst affords the corresponding cap analogs U and V. EXAMPLE 29
Figure imgf000032_0001
The coupling reaction of m2 7'3'°GDP (N7Me-3 'OMeGDP) with Imidazolide ZFGMP (lm2'FGMP) or Imidazolide m7'2'FGMP (N7MeIm2'FGMP) in the presence of ZnCl2 as the catalyst affords the corresponding cap analogs W and X.
EXAMPLE 30
Figure imgf000032_0002
The coupling reaction of m7'3'F2'dGDP (N7Me-3'F-2'dGDP) or m7'3's2'dGDP (N7Me-3'S-2'dGDP) with Imidazolide GMP (ImGMP) in the presence OfZnCl2 as the catalyst affords the corresponding cap analogs Y and Z.
EXAMPLE 31
Figure imgf000032_0003
Figure imgf000033_0001
m 7,2TMsGMp (N7Me-2'TBDMS-GMP) with Imidazolide GMP (ImGMP) in the presence OfZnCl2 as the catalyst affords the corresponding cap analogs Aa and Bb.
EXAMPLE 32
Figure imgf000033_0002
The coupling reaction of m7>LNAGDP (N 7Me-LNA-GDP) with Imidazolide GDP (ImGDP) or Imidazolide m7GDP (m7-ImGDP) in the presence of ZnCl2 as the catalyst affords the corresponding cap analogs Cc and Dd.
EXAMPLE 33
Figure imgf000033_0003
Figure imgf000034_0001
[0148] The coupling reaction of m7-2'3' b°Pr°py'idineGDP (N7Me- 2'3'isopropylideneGDP) with Imidazolide GDP (ImGDP) or Imidazolide m7GDP (m7- ImGDP) in the presence OfZnCl2 as the catalyst affords the corresponding cap analogs Ee and Ff.
[01491 Although the present disclosure is described with respect to certain embodiments and examples, various modifications may be made without departing from the spirit and scope of the invention.

Claims

What is claimed: 1. A composition comprising
Figure imgf000035_0001
wherein:
A is selected from a halogen, OH, OCH3, H, tert-butyldimethylsilyl and 2',3 '-O-isopropylidene;
B is selected from a halogen, OH5 OCH3, NH2, N3, NO2, CF3, CHO, S5 ferf-butyldimethylsilyl, LNA, and 2',3 '-O-isopropylidene;
R1 is CH3 or void;
R2 is selected from OH, OCH3 and a halogen; n is 1, 2 or 3; and when B is OH or OCH3, R1 is void, and R2 is OH5 then A is neither OH nor OCH3, and when A5 B and R2 are OH, R1 is not CH3.
2. The composition as recited in claim 1, wherein A is fluorine.
3. The composition as recited in claim I5 wherein B is fluorine.
4. The composition as recited in claim 1 , wherein R2 is fluorine.
5. The composition as recited in claim 1 attached to the 5' end of an RNA molecule.
6. The composition as recited in claim 2 attached to the 5' end of an RNA molecule.
7. The composition as recited in claim 3 attached to the 5' end of an RNA molecule.
8. The composition as recited in claim 4 attached to the 5' end of an RNA molecule.
9. The composition as recited in claim 1 , wherein:
A is selected from a halogen, OH5 OCH3, H, *er/-butyldimethylsilyl and 2'53'-O- isopropylidene; B is selected from a halogen, OH, OCH3, NH2, N3, NO2, CHO, S5 tert- butyldimethylsilyl, LNA, and 2',3'-0-isopropylidene;
Ri is void;
R2 is selected from OH, OCH3 and a halogen; n is 1, 2 or 3; and when A is either OH or OCH3, then R2 is not OH.
10. The composition as recited in claim 9, wherein A is fluorine.
11. The composition as recited in claim 9, wherein B is fluorine.
12. The composition as recited in claim 9, wherein R2 is fluorine.
13. The composition as recited in claim 9 attached to the 5' end of an RNA molecule.
14. The composition as recited in claim 10 attached to the 5' end of an RNA molecule.
15. The composition as recited in claim 11 attached to the 5' end of an RNA molecule.
16. The composition as recited in claim 12 attached to the 5' end of an RNA molecule.
17. The composition as recited in claim 1, wherein:
A is selected from a halogen, OH, OCH3, H,-and 2',3'-O-isopropylidene;
B is selected from CF3, OH, OCH3, NH2, N3, NO2, CHO, LNA, and 2',3'- O-isopropylidene;
Ri is CH3;
R2 is selected from OH, OCH3 and a halogen; n is I52 or 3, and when A is OH, R2 is not OH.
18. The composition as recited in claim 17, wherein A is fluorine.
19. The composition as recited in claim 17, wherein R2 is fluorine.
20. The composition as recited in claim 17 attached to the 5' end of an RNA molecule.
21. The composition as recited in claim 18 attached to the 5' end of an RNA molecule.
22. The composition as recited in claim 19 attached to the 5' end of an RNA molecule.
23. The composition as recited in claim 1, comprising the structure wherein:
A is selected from OH and OCH3;
B is selected from OH and OCH3;
R1 is CH3;
R2 is OCH3; and n is 1, 2 or 3.
24. The composition as recited in claim 23, wherein A is OH and B is OCH3.
25. The composition as recited in claim 23, wherein A is OCH3 and B is OH
26. The composition as recited in claim 23 attached to the 5' end of an RNA molecule.
27. The composition as recited in claim 24 attached to the 5' end of an RNA molecule.
28. The composition as recited in claim 25 attached to the 5' end of an RNA molecule.
29. A kit for capping an RNA transcript comprising: a) an cap analog having the structure:
Figure imgf000037_0001
wherein:
A is selected from a halogen, OH, OCH3, H, tert-butyldimethylsilyl and 2',3'-O-isopropylidene;
B is selected from a halogen, OH, OCH3, NH2,N3iNO2,CF3>CHO, S, /er/-butyldimethylsilyl, LNA, and 2',3'-O-isopropylidene; Ri is CH3 or void;
R2 is selected from OH, OCH3 and a halogen; n is 1, 2 or 3; and when B is OH or 0CH3> R] is void, and R2 is OH, then A is neither OH nor OCH3, and when A, B and R2 are OH, Ri is not CH3, and b). RNA polymerase.
30. A method of synthesizing a dinucleotide cap analog comprising: a) providing a first nucleoside comprising at least one of a 2' substituent and a 3' substituent on the ribose ring, b) phosphorylating the first nucleoside, forming a first nucleotide, c) methylating the first nucleotide, d) adding a phosphorylated second nucleotide optionally comprising a T ribose ring substituent, ,and e) linking said first nucleotide with said second nucleotide, forming a dinucleotide cap analog.
31. The method of claim 30 wherein said ribose ring comprises a substituent at the 2' position.
32. The method of claim 31 wherein the 2' substituent is selected from a halogen, OH, OCH3, H, ter/-butyldimethylsilyl and 2',3'-O-isopropylidene.
33. The method of claim 32 wherein the halogen is fluorine.
34. The method of claim 30 wherein said ribose ring comprises a substituent at the 3' position.
35. The method of claim 34 wherein the 3' substituent is selected from a halogen, OH, OCH3, NH2, N3, NO2, CF3, CHO, S, tert-butyldimethylsilyl, LNA, and 2',3'- O-isopropylidene.
36. The method of claim 35 wherein the halogen is fluorine.
37. The method of claim 30 wherein said step e) is followed by f) methylating said second nucleotide.
38. The method of claim 30 wherein said first nucleoside is guanosine.
39. The method of claim 38 wherein step c) comprises methylating the N7 position on the nucleotide.
40. The method of claim 30 wherein said second nucleotide is guanylic acid.
41. The method of claim 30 wherein the 2' substituent of said second nucleotide is a halogen, OH or CH3.
42. The method of claim 41 wherein the halogen is fluorine.
43. The method of claim 30 further comprising f) methylating said second nucleotide.
44. The method of claim 43 wherein step f) comprises methylating position N7.
45. The method of claim 30 wherein said linking is catalyzed by ZnCl2.
46. The kit of claim 29 further comprising nucleotides.
47. The kit of claim 29 further comprising ribonuclease inhibitor.
48. The kit of claim 29 further comprising enzyme buffer.
49. The kit of claim 29 further comprising nucleotide buffer.
PCT/US2007/015896 2006-07-28 2007-07-10 Dinucleotide mrna cap analogs WO2008016473A2 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
EP07810384A EP2049665A2 (en) 2006-07-28 2007-07-10 Dinucleotide mrna cap analogs
JP2009522766A JP2009544754A (en) 2006-07-28 2007-07-10 Dinucleotide MRNA cap analog
US12/375,527 US8304529B2 (en) 2006-07-28 2007-07-10 Dinucleotide MRNA cap analogs
CA002659301A CA2659301A1 (en) 2006-07-28 2007-07-10 Dinucleotide mrna cap analogs

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US82077106P 2006-07-28 2006-07-28
US60/820,771 2006-07-28

Publications (2)

Publication Number Publication Date
WO2008016473A2 true WO2008016473A2 (en) 2008-02-07
WO2008016473A3 WO2008016473A3 (en) 2008-04-10

Family

ID=38903097

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2007/015896 WO2008016473A2 (en) 2006-07-28 2007-07-10 Dinucleotide mrna cap analogs

Country Status (5)

Country Link
US (1) US8304529B2 (en)
EP (1) EP2049665A2 (en)
JP (1) JP2009544754A (en)
CA (1) CA2659301A1 (en)
WO (1) WO2008016473A2 (en)

Cited By (100)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009058911A2 (en) * 2007-10-31 2009-05-07 Applied Biosystems Inc. Preparation and isolation of 5' capped mrna
WO2010071587A1 (en) * 2008-12-18 2010-06-24 Avaris Ab A complex and method for enhancing nuclear delivery
US8304529B2 (en) 2006-07-28 2012-11-06 Life Technologies Corporation Dinucleotide MRNA cap analogs
WO2013059475A1 (en) * 2011-10-18 2013-04-25 Life Technologies Corporation Alkynyl-derivatized cap analogs, preparation and uses thereof
WO2014093924A1 (en) 2012-12-13 2014-06-19 Moderna Therapeutics, Inc. Modified nucleic acid molecules and uses thereof
WO2015101416A1 (en) 2013-12-30 2015-07-09 Curevac Gmbh Methods for rna analysis
WO2015188933A1 (en) 2014-06-10 2015-12-17 Curevac Ag Methods and means for enhancing rna production
WO2016170176A1 (en) 2015-04-22 2016-10-27 Curevac Ag Rna containing composition for treatment of tumor diseases
WO2017049245A2 (en) 2015-09-17 2017-03-23 Modernatx, Inc. Compounds and compositions for intracellular delivery of therapeutic agents
WO2017053297A1 (en) 2015-09-21 2017-03-30 Trilink Biotechnologies, Inc. Compositions and methods for synthesizing 5'-capped rnas
WO2017066791A1 (en) * 2015-10-16 2017-04-20 Modernatx, Inc. Sugar substituted mrna cap analogs
WO2017112865A1 (en) 2015-12-22 2017-06-29 Modernatx, Inc. Compounds and compositions for intracellular delivery of agents
WO2017109161A1 (en) 2015-12-23 2017-06-29 Curevac Ag Method of rna in vitro transcription using a buffer containing a dicarboxylic acid or tricarboxylic acid or a salt thereof
WO2017140345A1 (en) 2016-02-15 2017-08-24 Curevac Ag Method for analyzing by-products of rna in vitro transcription
WO2017149139A1 (en) 2016-03-03 2017-09-08 Curevac Ag Rna analysis by total hydrolysis
WO2017191264A1 (en) 2016-05-04 2017-11-09 Curevac Ag Nucleic acid molecules and uses thereof
WO2017218704A1 (en) 2016-06-14 2017-12-21 Modernatx, Inc. Stabilized formulations of lipid nanoparticles
WO2018033254A2 (en) 2016-08-19 2018-02-22 Curevac Ag Rna for cancer therapy
WO2018078053A1 (en) 2016-10-26 2018-05-03 Curevac Ag Lipid nanoparticle mrna vaccines
WO2018089540A1 (en) 2016-11-08 2018-05-17 Modernatx, Inc. Stabilized formulations of lipid nanoparticles
WO2018115527A2 (en) 2016-12-23 2018-06-28 Curevac Ag Mers coronavirus vaccine
WO2018115507A2 (en) 2016-12-23 2018-06-28 Curevac Ag Henipavirus vaccine
WO2018170336A1 (en) 2017-03-15 2018-09-20 Modernatx, Inc. Lipid nanoparticle formulation
WO2018170306A1 (en) 2017-03-15 2018-09-20 Modernatx, Inc. Compounds and compositions for intracellular delivery of therapeutic agents
US10131686B2 (en) 2013-04-29 2018-11-20 Memorial Sloan Kettering Cancer Center Compositions and methods for altering second messenger signaling
WO2018232120A1 (en) 2017-06-14 2018-12-20 Modernatx, Inc. Compounds and compositions for intracellular delivery of agents
EP3424524A2 (en) 2017-07-04 2019-01-09 CureVac AG Cancer rna-vaccine
WO2019036638A1 (en) 2017-08-18 2019-02-21 Modernatx, Inc. Methods of preparing modified rna
WO2019038332A1 (en) 2017-08-22 2019-02-28 Curevac Ag Bunyavirales vaccine
WO2019046809A1 (en) 2017-08-31 2019-03-07 Modernatx, Inc. Methods of making lipid nanoparticles
EP3461904A1 (en) 2014-11-10 2019-04-03 ModernaTX, Inc. Alternative nucleic acid molecules containing reduced uracil content and uses thereof
WO2019092153A1 (en) 2017-11-08 2019-05-16 Curevac Ag Rna sequence adaptation
WO2019115635A1 (en) 2017-12-13 2019-06-20 Curevac Ag Flavivirus vaccine
WO2019122371A1 (en) 2017-12-21 2019-06-27 Curevac Ag Linear double stranded dna coupled to a single support or a tag and methods for producing said linear double stranded dna
US10414789B2 (en) 2012-12-13 2019-09-17 Aduro Biotech, Inc. Compositions comprising cyclic purine dinucleotides having defined stereochemistries and methods for their preparation and use
EP3540060A1 (en) 2013-12-30 2019-09-18 CureVac AG Methods for rna analysis
US10428106B2 (en) 2015-10-16 2019-10-01 Modernatx, Inc. Phosphate replacement mRNA cap analogs
WO2019193183A2 (en) 2018-04-05 2019-10-10 Curevac Ag Novel yellow fever nucleic acid molecules for vaccination
WO2020002525A1 (en) 2018-06-27 2020-01-02 Curevac Ag Novel lassa virus rna molecules and compositions for vaccination
WO2020061457A1 (en) 2018-09-20 2020-03-26 Modernatx, Inc. Preparation of lipid nanoparticles and methods of administration thereof
WO2020061367A1 (en) 2018-09-19 2020-03-26 Modernatx, Inc. Compounds and compositions for intracellular delivery of therapeutic agents
US10653774B2 (en) 2013-05-18 2020-05-19 Aduro Biotech, Inc. Compositions and methods for activating “stimulator of interferon gene”-dependent signalling
WO2020127959A1 (en) 2018-12-21 2020-06-25 Curevac Ag Methods for rna analysis
WO2020128031A2 (en) 2018-12-21 2020-06-25 Curevac Ag Rna for malaria vaccines
WO2020160397A1 (en) 2019-01-31 2020-08-06 Modernatx, Inc. Methods of preparing lipid nanoparticles
WO2020160430A1 (en) 2019-01-31 2020-08-06 Modernatx, Inc. Vortex mixers and associated methods, systems, and apparatuses thereof
WO2020161342A1 (en) 2019-02-08 2020-08-13 Curevac Ag Coding rna administered into the suprachoroidal space in the treatment of ophtalmic diseases
WO2020254535A1 (en) 2019-06-18 2020-12-24 Curevac Ag Rotavirus mrna vaccine
WO2021028439A1 (en) 2019-08-14 2021-02-18 Curevac Ag Rna combinations and compositions with decreased immunostimulatory properties
WO2021123332A1 (en) 2019-12-20 2021-06-24 Curevac Ag Lipid nanoparticles for delivery of nucleic acids
WO2021156267A1 (en) 2020-02-04 2021-08-12 Curevac Ag Coronavirus vaccine
WO2021204179A1 (en) 2020-04-09 2021-10-14 Suzhou Abogen Biosciences Co., Ltd. Nucleic acid vaccines for coronavirus
WO2021204175A1 (en) 2020-04-09 2021-10-14 Suzhou Abogen Biosciences Co., Ltd. Lipid nanoparticle composition
WO2021239880A1 (en) 2020-05-29 2021-12-02 Curevac Ag Nucleic acid based combination vaccines
WO2022002040A1 (en) 2020-06-30 2022-01-06 Suzhou Abogen Biosciences Co., Ltd. Lipid compounds and lipid nanoparticle compositions
WO2022023559A1 (en) 2020-07-31 2022-02-03 Curevac Ag Nucleic acid encoded antibody mixtures
KR102366490B1 (en) 2020-10-20 2022-02-23 에스티팜 주식회사 Oligonucleotides for synthesizing 5'-capped RNA
WO2022037652A1 (en) 2020-08-20 2022-02-24 Suzhou Abogen Biosciences Co., Ltd. Lipid compounds and lipid nanoparticle compositions
WO2022043551A2 (en) 2020-08-31 2022-03-03 Curevac Ag Multivalent nucleic acid based coronavirus vaccines
EP3970742B1 (en) 2010-08-31 2022-05-25 GlaxoSmithKline Biologicals S.A. Pegylated liposomes for delivery of immunogen-encoding rna
WO2022122689A1 (en) 2020-12-09 2022-06-16 BioNTech SE Rna manufacturing
WO2022135993A2 (en) 2020-12-22 2022-06-30 Curevac Ag Pharmaceutical composition comprising lipid-based carriers encapsulating rna for multidose administration
WO2022137133A1 (en) 2020-12-22 2022-06-30 Curevac Ag Rna vaccine against sars-cov-2 variants
WO2022152141A2 (en) 2021-01-14 2022-07-21 Suzhou Abogen Biosciences Co., Ltd. Polymer conjugated lipid compounds and lipid nanoparticle compositions
WO2022152109A2 (en) 2021-01-14 2022-07-21 Suzhou Abogen Biosciences Co., Ltd. Lipid compounds and lipid nanoparticle compositions
WO2022162027A2 (en) 2021-01-27 2022-08-04 Curevac Ag Method of reducing the immunostimulatory properties of in vitro transcribed rna
CN114853836A (en) * 2022-06-24 2022-08-05 江苏申基生物科技有限公司 Initial capped oligonucleotide primer containing GNA structure and preparation method and application thereof
WO2022200575A1 (en) 2021-03-26 2022-09-29 Glaxosmithkline Biologicals Sa Immunogenic compositions
WO2022207862A2 (en) 2021-03-31 2022-10-06 Curevac Ag Syringes containing pharmaceutical compositions comprising rna
US11471525B2 (en) 2020-02-04 2022-10-18 Curevac Ag Coronavirus vaccine
WO2022233880A1 (en) 2021-05-03 2022-11-10 Curevac Ag Improved nucleic acid sequence for cell type specific expression
WO2022247755A1 (en) 2021-05-24 2022-12-01 Suzhou Abogen Biosciences Co., Ltd. Lipid compounds and lipid nanoparticle compositions
EP4101930A1 (en) 2015-09-17 2022-12-14 ModernaTX, Inc. Polynucleotides containing a stabilizing tail region
WO2023006999A2 (en) 2021-07-30 2023-02-02 CureVac SE Mrnas for treatment or prophylaxis of liver diseases
WO2023031394A1 (en) 2021-09-03 2023-03-09 CureVac SE Novel lipid nanoparticles for delivery of nucleic acids
WO2023031392A2 (en) 2021-09-03 2023-03-09 CureVac SE Novel lipid nanoparticles for delivery of nucleic acids comprising phosphatidylserine
EP4162950A1 (en) 2021-10-08 2023-04-12 Suzhou Abogen Biosciences Co., Ltd. Nucleic acid vaccines for coronavirus
WO2023056917A1 (en) 2021-10-08 2023-04-13 Suzhou Abogen Biosciences Co., Ltd. Lipid compounds and lipid nanoparticle compositions
WO2023056914A1 (en) 2021-10-08 2023-04-13 Suzhou Abogen Biosciences Co., Ltd. Lipid compounds and lipid nanoparticle compositions
EP3131914B1 (en) * 2014-04-16 2023-05-10 Idenix Pharmaceuticals LLC 3'-substituted methyl or alkynyl nucleosides for the treatment of hcv
KR20230083197A (en) 2021-12-01 2023-06-09 에스티팜 주식회사 Oligonucleotides for synthesizing 5'-capped RNA
WO2023144330A1 (en) 2022-01-28 2023-08-03 CureVac SE Nucleic acid encoded transcription factor inhibitors
EP4227319A1 (en) 2018-04-17 2023-08-16 CureVac SE Novel rsv rna molecules and compositions for vaccination
KR20230123318A (en) 2022-02-16 2023-08-23 에스티팜 주식회사 Oligonucleotides for synthesizing 5'-capped RNA
EP4233898A2 (en) 2016-05-04 2023-08-30 CureVac SE Influenza mrna vaccines
EP4239080A2 (en) 2015-07-01 2023-09-06 CureVac Manufacturing GmbH Method for analysis of an rna molecule
WO2023166425A1 (en) 2022-03-01 2023-09-07 Crispr Therapeutics Ag Methods and compositions for treating angiopoietin-like 3 (angptl3) related conditions
WO2023180904A1 (en) 2022-03-21 2023-09-28 Crispr Therapeutics Ag Methods and compositions for treating lipoprotein-related diseases
US11779659B2 (en) 2020-04-22 2023-10-10 BioNTech SE RNA constructs and uses thereof
WO2023227608A1 (en) 2022-05-25 2023-11-30 Glaxosmithkline Biologicals Sa Nucleic acid based vaccine encoding an escherichia coli fimh antigenic polypeptide
US11834651B2 (en) 2015-05-29 2023-12-05 CureVac Manufacturing GmbH Method for producing and purifying RNA, comprising at least one step of tangential flow filtration
CN117362369A (en) * 2023-10-09 2024-01-09 长沙晨辰医药科技有限公司 One-pot synthesis of nucleoside diphosphate
US11866754B2 (en) 2015-10-16 2024-01-09 Modernatx, Inc. Trinucleotide mRNA cap analogs
US11872280B2 (en) 2020-12-22 2024-01-16 CureVac SE RNA vaccine against SARS-CoV-2 variants
US11878055B1 (en) 2022-06-26 2024-01-23 BioNTech SE Coronavirus vaccine
WO2024037578A1 (en) 2022-08-18 2024-02-22 Suzhou Abogen Biosciences Co., Ltd. Composition of lipid nanoparticles
US11920148B2 (en) 2017-02-22 2024-03-05 Crispr Therapeutics Ag Compositions and methods for gene editing
WO2024068545A1 (en) 2022-09-26 2024-04-04 Glaxosmithkline Biologicals Sa Influenza virus vaccines
WO2024075022A2 (en) 2022-10-04 2024-04-11 BioNTech SE Rna constructs and uses thereof
CN117362369B (en) * 2023-10-09 2024-04-19 长沙晨辰医药科技有限公司 One-pot synthesis of nucleoside diphosphate

Families Citing this family (86)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2807552A1 (en) 2010-08-06 2012-02-09 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
ES2862955T3 (en) 2010-10-01 2021-10-08 Modernatx Inc Manipulated nucleic acids and methods of using them
US8710200B2 (en) 2011-03-31 2014-04-29 Moderna Therapeutics, Inc. Engineered nucleic acids encoding a modified erythropoietin and their expression
US9464124B2 (en) 2011-09-12 2016-10-11 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
WO2013052523A1 (en) 2011-10-03 2013-04-11 modeRNA Therapeutics Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
CN104114572A (en) 2011-12-16 2014-10-22 现代治疗公司 Modified nucleoside, nucleotide, and nucleic acid compositions
US10501513B2 (en) 2012-04-02 2019-12-10 Modernatx, Inc. Modified polynucleotides for the production of oncology-related proteins and peptides
US9283287B2 (en) 2012-04-02 2016-03-15 Moderna Therapeutics, Inc. Modified polynucleotides for the production of nuclear proteins
EP2833920A2 (en) 2012-04-02 2015-02-11 Moderna Therapeutics, Inc. Modified polynucleotides for the production of biologics and proteins associated with human disease
US9878056B2 (en) 2012-04-02 2018-01-30 Modernatx, Inc. Modified polynucleotides for the production of cosmetic proteins and peptides
US9572897B2 (en) 2012-04-02 2017-02-21 Modernatx, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
PL2922554T3 (en) 2012-11-26 2022-06-20 Modernatx, Inc. Terminally modified rna
EP2970940B1 (en) 2013-03-14 2018-07-25 Translate Bio, Inc. Mrna therapeutic compositions and use to treat diseases and disorders
AU2014239250A1 (en) 2013-03-14 2015-08-27 Shire Human Genetic Therapies, Inc. Quantitative assessment for cap efficiency of messenger RNA
WO2014152673A1 (en) 2013-03-14 2014-09-25 Shire Human Genetic Therapies, Inc. Quantitative assessment for cap efficiency of messenger rna
US10590161B2 (en) 2013-03-15 2020-03-17 Modernatx, Inc. Ion exchange purification of mRNA
US10138507B2 (en) 2013-03-15 2018-11-27 Modernatx, Inc. Manufacturing methods for production of RNA transcripts
US10077439B2 (en) 2013-03-15 2018-09-18 Modernatx, Inc. Removal of DNA fragments in mRNA production process
US8980864B2 (en) 2013-03-15 2015-03-17 Moderna Therapeutics, Inc. Compositions and methods of altering cholesterol levels
EP4279610A3 (en) 2013-03-15 2024-01-03 ModernaTX, Inc. Ribonucleic acid purification
AU2014287009B2 (en) 2013-07-11 2020-10-29 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
EP3052521A1 (en) 2013-10-03 2016-08-10 Moderna Therapeutics, Inc. Polynucleotides encoding low density lipoprotein receptor
KR101988552B1 (en) 2013-10-22 2019-09-25 샤이어 휴먼 지네틱 테라피즈 인크. Lipid formulations for delivery of messenger rna
CN106413811A (en) 2013-10-22 2017-02-15 夏尔人类遗传性治疗公司 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
MX2016005239A (en) 2013-10-22 2016-08-12 Shire Human Genetic Therapies Mrna therapy for phenylketonuria.
EP3122878B1 (en) 2014-03-24 2018-10-24 Translate Bio, Inc. Mrna therapy for the treatment of ocular diseases
ES2750686T3 (en) 2014-05-30 2020-03-26 Translate Bio Inc Biodegradable lipids for nucleic acid administration
US10286086B2 (en) 2014-06-19 2019-05-14 Modernatx, Inc. Alternative nucleic acid molecules and uses thereof
ES2964588T3 (en) 2014-06-24 2024-04-08 Translate Bio Inc Stereochemically enriched compositions for nucleic acid delivery
AU2015289656A1 (en) 2014-07-16 2017-02-16 Modernatx, Inc. Circular polynucleotides
WO2016090262A1 (en) 2014-12-05 2016-06-09 Shire Human Genetic Therapies, Inc. Messenger rna therapy for treatment of articular disease
AU2016233135B2 (en) 2015-03-19 2021-07-08 Translate Bio, Inc. mRNA therapy for pompe disease
WO2016154596A1 (en) 2015-03-25 2016-09-29 Editas Medicine, Inc. Crispr/cas-related methods, compositions and components
EP3294888A1 (en) 2015-05-11 2018-03-21 Editas Medicine, Inc. Crispr/cas-related methods and compositions for treating hiv infection and aids
AU2016261358B2 (en) 2015-05-11 2021-09-16 Editas Medicine, Inc. Optimized CRISPR/Cas9 systems and methods for gene editing in stem cells
CN116334142A (en) 2015-06-09 2023-06-27 爱迪塔斯医药公司 CRISPR/CAS related methods and compositions for improving transplantation
US11434486B2 (en) 2015-09-17 2022-09-06 Modernatx, Inc. Polynucleotides containing a morpholino linker
WO2017066793A1 (en) * 2015-10-16 2017-04-20 Modernatx, Inc. Mrna cap analogs and methods of mrna capping
CA3001351A1 (en) 2015-10-21 2017-04-27 Editas Medicine, Inc. Crispr/cas-related methods and compositions for treating hepatitis b virus
WO2017075475A1 (en) 2015-10-30 2017-05-04 Editas Medicine, Inc. Crispr/cas-related methods and compositions for treating herpes simplex virus
WO2017177169A1 (en) 2016-04-08 2017-10-12 Rana Therapeutics, Inc. Multimeric coding nucleic acid and uses thereof
CA3022611A1 (en) 2016-05-06 2017-11-09 Juno Therapeutics, Inc. Genetically engineered cells and methods of making the same
JP2019522047A (en) 2016-06-13 2019-08-08 トランスレイト バイオ, インコーポレイテッド Messenger RNA therapy for the treatment of ornithine transcarbamylase deficiency
AU2017356190A1 (en) 2016-11-10 2019-05-16 Translate Bio, Inc. Subcutaneous delivery of messenger RNA
US11407800B2 (en) 2017-02-28 2022-08-09 Arcturus Therapeutics, Inc. Translatable molecules and synthesis thereof
MA47824A (en) 2017-03-07 2020-01-15 Translate Bio Inc POLYANIONIC ADMINISTRATION OF NUCLEIC ACIDS
WO2018222890A1 (en) 2017-05-31 2018-12-06 Arcturus Therapeutics, Inc. Synthesis and structure of high potency rna therapeutics
WO2018222926A1 (en) 2017-05-31 2018-12-06 Ultragenyx Pharmaceutical Inc. Therapeutics for glycogen storage disease type iii
EP3630200A4 (en) 2017-05-31 2021-02-24 Arcturus Therapeutics, Inc. Therapeutics for phenylketonuria
WO2018231709A1 (en) 2017-06-12 2018-12-20 Translate Bio, Inc. Poly(phosphoesters) for delivery of nucleic acids
EP3641834B1 (en) 2017-06-19 2023-10-04 Translate Bio, Inc. Messenger rna therapy for the treatment of friedreich's ataxia
WO2019126593A1 (en) 2017-12-20 2019-06-27 Translate Bio, Inc. Improved composition and methods for treatment of ornithine transcarbamylase deficiency
US20210113483A1 (en) 2018-02-02 2021-04-22 Translate Bio, Inc. Cationic Polymers
EP3793689A1 (en) 2018-05-15 2021-03-24 Translate Bio, Inc. Subcutaneous delivery of messenger rna
CA3100218A1 (en) 2018-05-16 2019-11-21 Translate Bio, Inc. Ribose cationic lipids
JP2021525240A (en) 2018-05-24 2021-09-24 トランスレイト バイオ, インコーポレイテッド Thioester cationic lipid
JP2021525725A (en) 2018-05-30 2021-09-27 トランスレイト バイオ, インコーポレイテッド Messenger RNA vaccine and its use
EP3802507A1 (en) 2018-05-30 2021-04-14 Translate Bio, Inc. Vitamin cationic lipids
WO2019232097A1 (en) 2018-05-30 2019-12-05 Translate Bio, Inc. Phosphoester cationic lipids
CN112424214A (en) 2018-05-30 2021-02-26 川斯勒佰尔公司 Cationic lipids comprising steroidal moieties
EP3849617A1 (en) 2018-09-14 2021-07-21 Translate Bio, Inc. Composition and methods for treatment of methylmalonic acidemia
WO2020097384A1 (en) 2018-11-09 2020-05-14 Translate Bio, Inc. 2,5-dioxopiperazine lipids with intercalated ester, thioester, disulfide and anhydride moieities
EP3876914A2 (en) 2018-11-09 2021-09-15 Translate Bio, Inc. Messenger rna therapy for treatment of ocular diseases
CA3117866A1 (en) 2018-11-09 2020-05-14 Translate Bio, Inc. Multi-peg lipid compounds
AU2019374871A1 (en) 2018-11-09 2021-05-27 Translate Bio, Inc. PEG lipidoid compounds
CA3119449A1 (en) 2018-11-12 2020-05-22 Translate Bio, Inc. Methods for inducing immune tolerance
AU2019383413A1 (en) 2018-11-21 2021-05-27 Translate Bio, Inc. Cationic lipid compounds and compositions thereof for use in the delivery of messenger RNA
HUE064076T2 (en) 2018-12-06 2024-02-28 Arcturus Therapeutics Inc Compositions and methods for treating ornithine transcarbamylase deficiency
WO2020146344A1 (en) 2019-01-07 2020-07-16 Translate Bio, Inc. Composition and methods for treatment of primary ciliary dyskinesia
EP3956303A1 (en) 2019-04-18 2022-02-23 Translate Bio, Inc. Cystine cationic lipids
EP3959195B1 (en) 2019-04-22 2023-11-08 Translate Bio, Inc. Thioester cationic lipids
US20220257724A1 (en) 2019-05-03 2022-08-18 Translate Bio, Inc. Di-thioester cationic lipids
US20220226244A1 (en) 2019-05-31 2022-07-21 Translate Bio, Inc. Macrocyclic lipids
CN114401942B (en) 2019-06-21 2024-01-26 川斯勒佰尔公司 Tris (hydroxymethyl) methylglycine and citrate lipids
WO2020257611A1 (en) 2019-06-21 2020-12-24 Translate Bio, Inc. Cationic lipids comprising an hydroxy moiety
AU2021236068A1 (en) 2020-03-09 2022-10-06 Arcturus Therapeutics, Inc. Compositions and methods for inducing immune responses
EP4149556A1 (en) 2020-05-14 2023-03-22 Translate Bio, Inc. Peg lipidoid compounds
WO2022006527A1 (en) 2020-07-02 2022-01-06 Maritime Therapeutics, Inc. Compositions and methods for reverse gene therapy
AU2021412833A1 (en) 2020-12-28 2023-07-06 Arcturus Therapeutics, Inc. Transcription activator-like effector nucleases (talens) targeting hbv
CN114250268A (en) * 2021-12-23 2022-03-29 北京毅新博创生物科技有限公司 Product for detecting capping efficiency of mRNA sample and application
WO2023183889A1 (en) 2022-03-24 2023-09-28 Excepgen Inc. Compositions and methods for protein expression with rna
CN114685588B (en) * 2022-05-05 2024-03-29 江苏申基生物科技有限公司 Initial capping oligonucleotide primer containing open-loop nucleoside structure
CN115057903B (en) * 2022-06-22 2024-03-29 江苏申基生物科技有限公司 Initial capping oligonucleotide primer containing morpholine ring structure and preparation method and application thereof
CN114941018B (en) * 2022-06-28 2023-09-22 翌圣生物科技(上海)股份有限公司 Synthesis method of cap1 cap analogue

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999014226A2 (en) 1997-09-12 1999-03-25 Exiqon A/S Bi- and tri-cyclic nucleoside, nucleotide and oligonucleotide analogues
US20050287539A1 (en) 2004-06-29 2005-12-29 Emmanuel Labourier Methods and compositions for preparing capped RNA

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU738907C (en) * 1997-02-06 2002-05-16 Merck Sharp & Dohme Corp. Dinucleotides and their use
US7074596B2 (en) 2002-03-25 2006-07-11 Board Of Supervisors Of Louisiana State University And Agricultural And Mechanical College Synthesis and use of anti-reverse mRNA cap analogues
WO2006078821A2 (en) * 2005-01-21 2006-07-27 Invitrogen Corporation Products and processes for in vitro synthesis of biomolecules
CA2659301A1 (en) 2006-07-28 2008-02-07 Applera Corporation Dinucleotide mrna cap analogs
AU2008265683B2 (en) 2007-06-19 2013-08-29 Board Of Supervisors Of Louisiana State University And Agricultural And Mechanical College Synthesis and use of anti-reverse phosphorothioate analogs of the messenger RNA cap
WO2009058911A2 (en) 2007-10-31 2009-05-07 Applied Biosystems Inc. Preparation and isolation of 5' capped mrna

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999014226A2 (en) 1997-09-12 1999-03-25 Exiqon A/S Bi- and tri-cyclic nucleoside, nucleotide and oligonucleotide analogues
US20050287539A1 (en) 2004-06-29 2005-12-29 Emmanuel Labourier Methods and compositions for preparing capped RNA

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
CHIDGEAVADZE ET AL., FEB. LETT., vol. 183, 1985, pages 275 - 278
CHIDGEAVADZE ET AL., NUCLEIC ACIDS RES., vol. 12, 1984, pages 1671 - 1686
METZKER ET AL., NUCLEIC ACIDS RES., vol. 22, no. 20, 1994, pages 4259

Cited By (153)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8304529B2 (en) 2006-07-28 2012-11-06 Life Technologies Corporation Dinucleotide MRNA cap analogs
WO2009058911A3 (en) * 2007-10-31 2009-06-25 Applied Biosystems Preparation and isolation of 5' capped mrna
US8093367B2 (en) 2007-10-31 2012-01-10 Applied Biosystems, Llc Preparation and isolation of 5′ capped mRNA
WO2009058911A2 (en) * 2007-10-31 2009-05-07 Applied Biosystems Inc. Preparation and isolation of 5' capped mrna
US9067964B2 (en) 2008-12-18 2015-06-30 Oligomer Sciences Ab Complex and method for enhancing nuclear delivery
WO2010071587A1 (en) * 2008-12-18 2010-06-24 Avaris Ab A complex and method for enhancing nuclear delivery
EP3970742B1 (en) 2010-08-31 2022-05-25 GlaxoSmithKline Biologicals S.A. Pegylated liposomes for delivery of immunogen-encoding rna
US9752153B2 (en) 2011-10-18 2017-09-05 Life Technologies Corporation Alkynyl-derivatized cap analogs, preparation and uses thereof
US8969545B2 (en) 2011-10-18 2015-03-03 Life Technologies Corporation Alkynyl-derivatized cap analogs, preparation and uses thereof
US9388420B2 (en) 2011-10-18 2016-07-12 Life Technologies Corporation Alkynyl-derivatized cap analogs, preparation and uses thereof
WO2013059475A1 (en) * 2011-10-18 2013-04-25 Life Technologies Corporation Alkynyl-derivatized cap analogs, preparation and uses thereof
WO2014093924A1 (en) 2012-12-13 2014-06-19 Moderna Therapeutics, Inc. Modified nucleic acid molecules and uses thereof
US10414789B2 (en) 2012-12-13 2019-09-17 Aduro Biotech, Inc. Compositions comprising cyclic purine dinucleotides having defined stereochemistries and methods for their preparation and use
US11014956B2 (en) 2013-04-29 2021-05-25 Memorial Sloan Kettering Cancer Center; The Rockefeller Compositions and methods for altering second messenger signaling
US10385091B2 (en) 2013-04-29 2019-08-20 Memorial Sloan Kettering Cancer Center Compositions and methods for altering second messenger signaling
US10131686B2 (en) 2013-04-29 2018-11-20 Memorial Sloan Kettering Cancer Center Compositions and methods for altering second messenger signaling
US10653774B2 (en) 2013-05-18 2020-05-19 Aduro Biotech, Inc. Compositions and methods for activating “stimulator of interferon gene”-dependent signalling
EP3540060A1 (en) 2013-12-30 2019-09-18 CureVac AG Methods for rna analysis
WO2015101416A1 (en) 2013-12-30 2015-07-09 Curevac Gmbh Methods for rna analysis
EP3131914B1 (en) * 2014-04-16 2023-05-10 Idenix Pharmaceuticals LLC 3'-substituted methyl or alkynyl nucleosides for the treatment of hcv
WO2015188933A1 (en) 2014-06-10 2015-12-17 Curevac Ag Methods and means for enhancing rna production
EP3521456A1 (en) 2014-06-10 2019-08-07 CureVac AG Methods and means for enhancing rna production
EP3461904A1 (en) 2014-11-10 2019-04-03 ModernaTX, Inc. Alternative nucleic acid molecules containing reduced uracil content and uses thereof
EP3603661A2 (en) 2015-04-22 2020-02-05 CureVac AG Rna containing composition for treatment of tumor diseases
EP3173092A2 (en) 2015-04-22 2017-05-31 CureVac AG Rna containing composition for treatment of tumor diseases
EP3326641A1 (en) 2015-04-22 2018-05-30 CureVac AG Rna containing composition for treatment of tumor diseases
WO2016170176A1 (en) 2015-04-22 2016-10-27 Curevac Ag Rna containing composition for treatment of tumor diseases
US11834651B2 (en) 2015-05-29 2023-12-05 CureVac Manufacturing GmbH Method for producing and purifying RNA, comprising at least one step of tangential flow filtration
EP4239080A2 (en) 2015-07-01 2023-09-06 CureVac Manufacturing GmbH Method for analysis of an rna molecule
EP4101930A1 (en) 2015-09-17 2022-12-14 ModernaTX, Inc. Polynucleotides containing a stabilizing tail region
EP4286012A2 (en) 2015-09-17 2023-12-06 ModernaTX, Inc. Compounds and compositions for intracellular delivery of therapeutic agents
WO2017049245A2 (en) 2015-09-17 2017-03-23 Modernatx, Inc. Compounds and compositions for intracellular delivery of therapeutic agents
EP3736261A1 (en) 2015-09-17 2020-11-11 ModernaTX, Inc. Compounds and compositions for intracellular delivery of therapeutic agents
CN108366604A (en) * 2015-09-21 2018-08-03 垂林克生物技术公司 For synthesizing the 5 '-compositions and method for capping RNA
US10494399B2 (en) 2015-09-21 2019-12-03 Trilink Biotechnologies, Inc. Compositions and methods for synthesizing 5′-Capped RNAS
EP3954225A1 (en) * 2015-09-21 2022-02-16 TriLink BioTechnologies, LLC Compositions and methods for synthesizing 5'-capped rnas
US10913768B2 (en) 2015-09-21 2021-02-09 Trilink Biotechnologies, Inc. Compositions and methods for synthesizing 5′-capped RNAs
EP3352584A1 (en) * 2015-09-21 2018-08-01 Trilink Biotechnologies, Inc. Compositions and methods for synthesizing 5'-capped rnas
AU2021206780B2 (en) * 2015-09-21 2023-04-06 Trilink Biotechnologies, Llc Compositions and methods for synthesizing 5'-Capped RNAs
EP4140491A1 (en) * 2015-09-21 2023-03-01 TriLink BioTechnologies, LLC Compositions and methods for synthesizing 5'-capped rnas
US11414453B2 (en) 2015-09-21 2022-08-16 Trilink Biotechnologies, Llc Compositions and methods for synthesizing 5′-capped RNAs
EP3352584A4 (en) * 2015-09-21 2019-03-27 Trilink Biotechnologies, Inc. Compositions and methods for synthesizing 5'-capped rnas
US11578095B2 (en) 2015-09-21 2023-02-14 Trilink Biotechnologies, Llc Compositions and methods for synthesizing 5'-Capped RNAs
EP4276189A3 (en) * 2015-09-21 2024-01-17 TriLink BioTechnologies, LLC Initiating capped oligonucleotide primers for synthesizing 5'-capped rnas
EP3906789A1 (en) * 2015-09-21 2021-11-10 TriLink BioTechnologies, LLC Compositions and methods for synthesizing 5 -capped rnas
CN113584020A (en) * 2015-09-21 2021-11-02 垂林克生物技术公司 Compositions and methods for synthesizing 5' -capped RNA
EP4104687A1 (en) * 2015-09-21 2022-12-21 TriLink BioTechnologies, LLC Compositions and methods for synthesizing 5 -capped rnas
EP3352584B1 (en) 2015-09-21 2021-05-12 TriLink BioTechnologies, LLC Compositions and methods for synthesizing 5'-capped rnas
AU2016328645B2 (en) * 2015-09-21 2021-05-13 Trilink Biotechnologies, Llc Compositions and methods for synthesizing 5'-Capped RNAs
WO2017053297A1 (en) 2015-09-21 2017-03-30 Trilink Biotechnologies, Inc. Compositions and methods for synthesizing 5'-capped rnas
EP3954224A1 (en) * 2015-09-21 2022-02-16 TriLink BioTechnologies, LLC Compositions and methods for synthesizing 5'-capped rnas
AU2016328645C1 (en) * 2015-09-21 2021-10-28 Trilink Biotechnologies, Llc Compositions and methods for synthesizing 5'-Capped RNAs
US11878991B2 (en) 2015-09-21 2024-01-23 Trilink Biotechnologies, Llc Compositions and methods for synthesizing 5′-Capped RNAs
US10519189B2 (en) 2015-09-21 2019-12-31 Trilink Biotechnologies, Inc. Compositions and methods for synthesizing 5′-Capped RNAs
WO2017066791A1 (en) * 2015-10-16 2017-04-20 Modernatx, Inc. Sugar substituted mrna cap analogs
US10428106B2 (en) 2015-10-16 2019-10-01 Modernatx, Inc. Phosphate replacement mRNA cap analogs
US10563195B2 (en) 2015-10-16 2020-02-18 Modernatx, Inc. Phosphate replacement mRNA cap analogs
US10570388B2 (en) 2015-10-16 2020-02-25 Modernatx, Inc. Phosphate replacement MRNA cap analogs
US11866754B2 (en) 2015-10-16 2024-01-09 Modernatx, Inc. Trinucleotide mRNA cap analogs
WO2017112865A1 (en) 2015-12-22 2017-06-29 Modernatx, Inc. Compounds and compositions for intracellular delivery of agents
EP4036079A2 (en) 2015-12-22 2022-08-03 ModernaTX, Inc. Compounds and compositions for intracellular delivery of agents
WO2017109161A1 (en) 2015-12-23 2017-06-29 Curevac Ag Method of rna in vitro transcription using a buffer containing a dicarboxylic acid or tricarboxylic acid or a salt thereof
US11248223B2 (en) 2015-12-23 2022-02-15 Curevac Ag Method of RNA in vitro transcription using a buffer containing a dicarboxylic acid or tricarboxylic acid or a salt thereof
WO2017140345A1 (en) 2016-02-15 2017-08-24 Curevac Ag Method for analyzing by-products of rna in vitro transcription
US11920174B2 (en) 2016-03-03 2024-03-05 CureVac SE RNA analysis by total hydrolysis and quantification of released nucleosides
WO2017149139A1 (en) 2016-03-03 2017-09-08 Curevac Ag Rna analysis by total hydrolysis
EP4233898A2 (en) 2016-05-04 2023-08-30 CureVac SE Influenza mrna vaccines
WO2017191264A1 (en) 2016-05-04 2017-11-09 Curevac Ag Nucleic acid molecules and uses thereof
WO2017218704A1 (en) 2016-06-14 2017-12-21 Modernatx, Inc. Stabilized formulations of lipid nanoparticles
WO2018033254A2 (en) 2016-08-19 2018-02-22 Curevac Ag Rna for cancer therapy
WO2018078053A1 (en) 2016-10-26 2018-05-03 Curevac Ag Lipid nanoparticle mrna vaccines
WO2018089540A1 (en) 2016-11-08 2018-05-17 Modernatx, Inc. Stabilized formulations of lipid nanoparticles
WO2018115527A2 (en) 2016-12-23 2018-06-28 Curevac Ag Mers coronavirus vaccine
WO2018115507A2 (en) 2016-12-23 2018-06-28 Curevac Ag Henipavirus vaccine
US11920148B2 (en) 2017-02-22 2024-03-05 Crispr Therapeutics Ag Compositions and methods for gene editing
WO2018170336A1 (en) 2017-03-15 2018-09-20 Modernatx, Inc. Lipid nanoparticle formulation
EP4186888A1 (en) 2017-03-15 2023-05-31 ModernaTX, Inc. Compound and compositions for intracellular delivery of therapeutic agents
WO2018170306A1 (en) 2017-03-15 2018-09-20 Modernatx, Inc. Compounds and compositions for intracellular delivery of therapeutic agents
WO2018232120A1 (en) 2017-06-14 2018-12-20 Modernatx, Inc. Compounds and compositions for intracellular delivery of agents
WO2019008001A1 (en) 2017-07-04 2019-01-10 Curevac Ag Novel nucleic acid molecules
EP3424524A2 (en) 2017-07-04 2019-01-09 CureVac AG Cancer rna-vaccine
WO2019036638A1 (en) 2017-08-18 2019-02-21 Modernatx, Inc. Methods of preparing modified rna
WO2019038332A1 (en) 2017-08-22 2019-02-28 Curevac Ag Bunyavirales vaccine
WO2019046809A1 (en) 2017-08-31 2019-03-07 Modernatx, Inc. Methods of making lipid nanoparticles
WO2019092153A1 (en) 2017-11-08 2019-05-16 Curevac Ag Rna sequence adaptation
WO2019115635A1 (en) 2017-12-13 2019-06-20 Curevac Ag Flavivirus vaccine
WO2019122371A1 (en) 2017-12-21 2019-06-27 Curevac Ag Linear double stranded dna coupled to a single support or a tag and methods for producing said linear double stranded dna
WO2019193183A2 (en) 2018-04-05 2019-10-10 Curevac Ag Novel yellow fever nucleic acid molecules for vaccination
EP4227319A1 (en) 2018-04-17 2023-08-16 CureVac SE Novel rsv rna molecules and compositions for vaccination
WO2020002525A1 (en) 2018-06-27 2020-01-02 Curevac Ag Novel lassa virus rna molecules and compositions for vaccination
WO2020061367A1 (en) 2018-09-19 2020-03-26 Modernatx, Inc. Compounds and compositions for intracellular delivery of therapeutic agents
WO2020061457A1 (en) 2018-09-20 2020-03-26 Modernatx, Inc. Preparation of lipid nanoparticles and methods of administration thereof
WO2020127959A1 (en) 2018-12-21 2020-06-25 Curevac Ag Methods for rna analysis
WO2020128031A2 (en) 2018-12-21 2020-06-25 Curevac Ag Rna for malaria vaccines
WO2020160397A1 (en) 2019-01-31 2020-08-06 Modernatx, Inc. Methods of preparing lipid nanoparticles
WO2020160430A1 (en) 2019-01-31 2020-08-06 Modernatx, Inc. Vortex mixers and associated methods, systems, and apparatuses thereof
WO2020161342A1 (en) 2019-02-08 2020-08-13 Curevac Ag Coding rna administered into the suprachoroidal space in the treatment of ophtalmic diseases
WO2020254535A1 (en) 2019-06-18 2020-12-24 Curevac Ag Rotavirus mrna vaccine
WO2021028439A1 (en) 2019-08-14 2021-02-18 Curevac Ag Rna combinations and compositions with decreased immunostimulatory properties
WO2021123332A1 (en) 2019-12-20 2021-06-24 Curevac Ag Lipid nanoparticles for delivery of nucleic acids
US11576966B2 (en) 2020-02-04 2023-02-14 CureVac SE Coronavirus vaccine
DE112021000012T5 (en) 2020-02-04 2021-11-18 Curevac Ag Coronavirus vaccine
US11964012B2 (en) 2020-02-04 2024-04-23 CureVac SE Coronavirus vaccine
US11964011B2 (en) 2020-02-04 2024-04-23 CureVac SE Coronavirus vaccine
US11471525B2 (en) 2020-02-04 2022-10-18 Curevac Ag Coronavirus vaccine
DE202021004130U1 (en) 2020-02-04 2022-10-26 Curevac Ag Coronavirus Vaccine
DE202021004123U1 (en) 2020-02-04 2022-10-26 Curevac Ag Coronavirus Vaccine
EP4147717A1 (en) 2020-02-04 2023-03-15 CureVac SE Coronavirus vaccine
WO2021156267A1 (en) 2020-02-04 2021-08-12 Curevac Ag Coronavirus vaccine
US11596686B2 (en) 2020-02-04 2023-03-07 CureVac SE Coronavirus vaccine
DE202021003575U1 (en) 2020-02-04 2022-01-17 Curevac Ag Coronavirus Vaccine
WO2021204175A1 (en) 2020-04-09 2021-10-14 Suzhou Abogen Biosciences Co., Ltd. Lipid nanoparticle composition
WO2021204179A1 (en) 2020-04-09 2021-10-14 Suzhou Abogen Biosciences Co., Ltd. Nucleic acid vaccines for coronavirus
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
WO2021239880A1 (en) 2020-05-29 2021-12-02 Curevac Ag Nucleic acid based combination vaccines
WO2022002040A1 (en) 2020-06-30 2022-01-06 Suzhou Abogen Biosciences Co., Ltd. Lipid compounds and lipid nanoparticle compositions
WO2022023559A1 (en) 2020-07-31 2022-02-03 Curevac Ag Nucleic acid encoded antibody mixtures
WO2022037652A1 (en) 2020-08-20 2022-02-24 Suzhou Abogen Biosciences Co., Ltd. Lipid compounds and lipid nanoparticle compositions
WO2022043551A2 (en) 2020-08-31 2022-03-03 Curevac Ag Multivalent nucleic acid based coronavirus vaccines
WO2022086140A1 (en) 2020-10-20 2022-04-28 에스티팜 주식회사 Oligonucleotide for 5'-capped rna synthesis
KR102366490B1 (en) 2020-10-20 2022-02-23 에스티팜 주식회사 Oligonucleotides for synthesizing 5'-capped RNA
WO2022122689A1 (en) 2020-12-09 2022-06-16 BioNTech SE Rna manufacturing
WO2022137133A1 (en) 2020-12-22 2022-06-30 Curevac Ag Rna vaccine against sars-cov-2 variants
WO2022135993A2 (en) 2020-12-22 2022-06-30 Curevac Ag Pharmaceutical composition comprising lipid-based carriers encapsulating rna for multidose administration
US11918643B2 (en) 2020-12-22 2024-03-05 CureVac SE RNA vaccine against SARS-CoV-2 variants
US11872280B2 (en) 2020-12-22 2024-01-16 CureVac SE RNA vaccine against SARS-CoV-2 variants
WO2022152109A2 (en) 2021-01-14 2022-07-21 Suzhou Abogen Biosciences Co., Ltd. Lipid compounds and lipid nanoparticle compositions
WO2022152141A2 (en) 2021-01-14 2022-07-21 Suzhou Abogen Biosciences Co., Ltd. Polymer conjugated lipid compounds and lipid nanoparticle compositions
WO2022162027A2 (en) 2021-01-27 2022-08-04 Curevac Ag Method of reducing the immunostimulatory properties of in vitro transcribed rna
WO2022200575A1 (en) 2021-03-26 2022-09-29 Glaxosmithkline Biologicals Sa Immunogenic compositions
WO2022207862A2 (en) 2021-03-31 2022-10-06 Curevac Ag Syringes containing pharmaceutical compositions comprising rna
WO2022233880A1 (en) 2021-05-03 2022-11-10 Curevac Ag Improved nucleic acid sequence for cell type specific expression
WO2022247755A1 (en) 2021-05-24 2022-12-01 Suzhou Abogen Biosciences Co., Ltd. Lipid compounds and lipid nanoparticle compositions
WO2023006999A2 (en) 2021-07-30 2023-02-02 CureVac SE Mrnas for treatment or prophylaxis of liver diseases
WO2023031394A1 (en) 2021-09-03 2023-03-09 CureVac SE Novel lipid nanoparticles for delivery of nucleic acids
WO2023031392A2 (en) 2021-09-03 2023-03-09 CureVac SE Novel lipid nanoparticles for delivery of nucleic acids comprising phosphatidylserine
EP4162950A1 (en) 2021-10-08 2023-04-12 Suzhou Abogen Biosciences Co., Ltd. Nucleic acid vaccines for coronavirus
WO2023056917A1 (en) 2021-10-08 2023-04-13 Suzhou Abogen Biosciences Co., Ltd. Lipid compounds and lipid nanoparticle compositions
WO2023056914A1 (en) 2021-10-08 2023-04-13 Suzhou Abogen Biosciences Co., Ltd. Lipid compounds and lipid nanoparticle compositions
KR20230083197A (en) 2021-12-01 2023-06-09 에스티팜 주식회사 Oligonucleotides for synthesizing 5'-capped RNA
WO2023144330A1 (en) 2022-01-28 2023-08-03 CureVac SE Nucleic acid encoded transcription factor inhibitors
KR20230123318A (en) 2022-02-16 2023-08-23 에스티팜 주식회사 Oligonucleotides for synthesizing 5'-capped RNA
WO2023166425A1 (en) 2022-03-01 2023-09-07 Crispr Therapeutics Ag Methods and compositions for treating angiopoietin-like 3 (angptl3) related conditions
WO2023180904A1 (en) 2022-03-21 2023-09-28 Crispr Therapeutics Ag Methods and compositions for treating lipoprotein-related diseases
WO2023227608A1 (en) 2022-05-25 2023-11-30 Glaxosmithkline Biologicals Sa Nucleic acid based vaccine encoding an escherichia coli fimh antigenic polypeptide
CN114853836A (en) * 2022-06-24 2022-08-05 江苏申基生物科技有限公司 Initial capped oligonucleotide primer containing GNA structure and preparation method and application thereof
US11878055B1 (en) 2022-06-26 2024-01-23 BioNTech SE Coronavirus vaccine
WO2024037578A1 (en) 2022-08-18 2024-02-22 Suzhou Abogen Biosciences Co., Ltd. Composition of lipid nanoparticles
WO2024068545A1 (en) 2022-09-26 2024-04-04 Glaxosmithkline Biologicals Sa Influenza virus vaccines
WO2024075022A2 (en) 2022-10-04 2024-04-11 BioNTech SE Rna constructs and uses thereof
CN117362369A (en) * 2023-10-09 2024-01-09 长沙晨辰医药科技有限公司 One-pot synthesis of nucleoside diphosphate
CN117362369B (en) * 2023-10-09 2024-04-19 长沙晨辰医药科技有限公司 One-pot synthesis of nucleoside diphosphate

Also Published As

Publication number Publication date
WO2008016473A3 (en) 2008-04-10
JP2009544754A (en) 2009-12-17
CA2659301A1 (en) 2008-02-07
US8304529B2 (en) 2012-11-06
US20100261231A1 (en) 2010-10-14
EP2049665A2 (en) 2009-04-22

Similar Documents

Publication Publication Date Title
EP2049665A2 (en) Dinucleotide mrna cap analogs
Flamme et al. Chemical methods for the modification of RNA
EP2217612B1 (en) Preparation of ribonucleotide oligomer
US8093367B2 (en) Preparation and isolation of 5′ capped mRNA
EP1954707B1 (en) Polynucleotide containing a phosphate mimetic
KR101718406B1 (en) - 3- rna synthesis phosphoramidites for synthetic rna in the reverse direction and application in convenient introduction of ligands chromophores and modifications of synthetic rna at the 3-end
CN105658658B (en) Cytidine-5-formyl amine-modified polynucleotide composition and its correlation technique
KR101995521B1 (en) Phosphoramidites for synthetic rna in the reverse direction
EP1795536A1 (en) Phosphoramidite compound and method for producing oligo-rna
WO2010110775A1 (en) Reagents for reversibly terminating primer extension
JPH11510790A (en) Improved method for oligonucleotide synthesis
CN113412268A (en) 5&#39; -modified nucleoside and nucleotide using the same
JP2002536381A5 (en)
CN104011209A (en) Inhibition Of Viral Gene Expression
WO2002077002A2 (en) Synthesis of selenium-derivatized nucleosides, nucleotides, phosphoramidites, triphosphates and nucleic acids
EP3484907B1 (en) Novel phosphotriazole mrna 5&#39;-end cap analogs, composition comprising the same, rna molecule incorporating the same, uses thereof and method of synthesizing rna molecule, protein or peptide
WO1995031470A2 (en) Antisense inhibitors of gene expression
WO2023282245A1 (en) Method for purifying nucleotides, device for purifying nucleotides, hydrophobic reagent, and hydrophobic substrate
CN117940441A (en) MRNA cap analogues and uses thereof
JP2005046073A (en) Method of rna transcription and/or amplification using modified nucleic acid

Legal Events

Date Code Title Description
DPE1 Request for preliminary examination filed after expiration of 19th month from priority date (pct application filed from 20040101)
WWE Wipo information: entry into national phase

Ref document number: 2659301

Country of ref document: CA

Ref document number: 2009522766

Country of ref document: JP

Ref document number: 2007810384

Country of ref document: EP

NENP Non-entry into the national phase

Ref country code: DE

NENP Non-entry into the national phase

Ref country code: RU

WWE Wipo information: entry into national phase

Ref document number: 12375527

Country of ref document: US