WO2024250923A1 - Chemically modified nucleoside, chemically modified nucleoside triphosphate and use thereof - Google Patents
Chemically modified nucleoside, chemically modified nucleoside triphosphate and use thereof Download PDFInfo
- Publication number
- WO2024250923A1 WO2024250923A1 PCT/CN2024/093272 CN2024093272W WO2024250923A1 WO 2024250923 A1 WO2024250923 A1 WO 2024250923A1 CN 2024093272 W CN2024093272 W CN 2024093272W WO 2024250923 A1 WO2024250923 A1 WO 2024250923A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- chemically modified
- triphosphate
- compound
- formula
- mrna
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Pending
Links
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K48/00—Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P31/00—Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
- A61P31/12—Antivirals
- A61P31/14—Antivirals for RNA viruses
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07H—SUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
- C07H1/00—Processes for the preparation of sugar derivatives
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07H—SUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
- C07H19/00—Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof
- C07H19/02—Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof sharing nitrogen
- C07H19/04—Heterocyclic radicals containing only nitrogen atoms as ring hetero atom
- C07H19/06—Pyrimidine radicals
- C07H19/067—Pyrimidine radicals with ribosyl as the saccharide radical
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07H—SUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
- C07H19/00—Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof
- C07H19/02—Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof sharing nitrogen
- C07H19/04—Heterocyclic radicals containing only nitrogen atoms as ring hetero atom
- C07H19/06—Pyrimidine radicals
- C07H19/10—Pyrimidine radicals with the saccharide radical esterified by phosphoric or polyphosphoric acids
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07H—SUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
- C07H21/00—Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids
- C07H21/02—Compounds 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
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07H—SUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
- C07H21/00—Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids
- C07H21/04—Compounds 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
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
- C12N15/11—DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
Definitions
- the present application belongs to the field of nucleic acid modification technology, and specifically relates to a chemically modified nucleoside, a chemically modified nucleoside triphosphate and applications thereof.
- mRNA also known as messenger RNA
- messenger RNA is transcribed from DNA as a template and is responsible for guiding the synthesis of intracellular proteins.
- mRNA technology uses this rule to introduce in vitro artificially synthesized mRNA into specific cells, making cells into small factories for the production of protein drugs, and exerting therapeutic effects through the proteins produced by these cells.
- mRNA technology has great application potential in infectious disease prevention and control, tumor vaccines, tumor immunotherapy, protein replacement therapy, etc., but natural mRNA has poor stability and is prone to immunogenicity, which seriously hinders the application of mRNA.
- naturally occurring modified nucleosides are used to replace natural nucleosides, for example, pseudouridine and methyl pseudouridine are used to replace uridine to solve the problems of stability and immunogenicity of mRNA drugs.
- the number of naturally occurring modified nucleosides is scarce, which cannot meet the needs of improving the properties of mRNA, nor is it conducive to establishing structure-activity relationships to reveal its mechanism of action at the molecular level. Therefore, it is of great research and application significance to prepare more chemically modified nucleosides using organic synthesis methods to improve the drugability of mRNA from different levels.
- the poly A tail structure is also an important control factor for translation initiation, and it can also regulate the stability and turnover rate of mRNA, thereby affecting the translation efficiency of mRNA, therefore, the development of a method using new chemically modified nucleotides and modifying the poly A tail structure of mRNA is of great significance and has broad application prospects in the relevant applications of mRNA.
- RNA modification methods used in existing patents mainly use modified nucleosides existing in nature to replace conventional nucleosides, such as using pseudouridine and methyl pseudouridine to replace uridine.
- CN107090436B discloses a non-therapeutic method or in vitro method for reducing the immunogenicity of an in vitro synthesized RNA molecule obtained by in vitro transcription or a preparation of a gene therapy vector containing the RNA molecule to mammalian cells, wherein the replacement uridine contained in the RNA molecule is selected from a modified nucleoside of pseudouridine, 1-methyl pseudouridine, 5-methyluridine, 5-methoxyuridine and 2-thiouridine.
- the types of naturally occurring modified bases are very limited, so the means available for improving the properties of mRNA are limited.
- non-natural modified nucleosides can greatly enrich the types and range of non-natural nucleosides and is also suitable for establishing structure-activity relationships, which is not available with naturally occurring modified bases.
- CN112673106A discloses a click-modified mRNA, which relates to alkyne and/or azide-modified mRNA, and stabilizes RNA by introducing alkyne and/or azide-modified nucleotides. This modification can not only stabilize the target mRNA for in vitro application and subsequent administration to human patients, animals or plants, but also can easily attach detectable markers or functional groups.
- CN114807154A discloses a modified nucleic acid and its application, the modified nucleic acid includes uridine, cytosine, adenine, guanosine and chemically modified nucleosides; the chemically modified Nucleosides include one or more of chemically modified uridine nucleosides, chemically modified cytosine nucleosides, chemically modified adenine nucleosides, and chemically modified guanine nucleosides; the modified nucleic acids described in this application have high stability, low immunogenicity, and long in vivo half-life. However, there are still relatively few types of modified nucleic acids.
- the present application provides a chemically modified nucleoside, a chemically modified nucleoside triphosphate and an application thereof.
- the present application uses chemical synthesis means to chemically modify natural nucleosides, wherein the chemically modified nucleosides include adenine nucleosides and cytosine nucleosides; the chemically modified nucleosides are triphosphorylated to obtain chemically modified adenine nucleoside triphosphates and chemically modified cytosine nucleoside triphosphates, and the chemically modified nucleoside triphosphates are used to globally replace the natural nucleoside triphosphates to obtain modified mRNA, and the modified mRNA exhibits enhanced translation efficiency and higher stability both in vitro and in cells.
- the present application provides a chemically modified nucleoside, wherein the chemically modified nucleoside is selected from a compound having a structure as shown in Formula 1, a salt thereof, or an isomer thereof:
- R 3 is selected from hydroxyl, hydrogen, fluorine, methoxy, ethoxy or methoxymethyl ether;
- Y is selected from a cytosine ring containing a substituent or an adenine ring containing a substituent, wherein the cytosine ring containing a substituent is as shown in Formula 2, and the adenine ring containing a substituent is as shown in Formula 3. Indicates the connection location;
- R 1 and R 1′ are each independently selected from hydrogen, acetyl, halogen-substituted acetyl, propionyl, halogen-substituted propionyl, butyryl, halogen-substituted butyryl, valeryl, halogen-substituted valeryl, hexanoyl, halogen-substituted hexanoyl, octanoyl, decanoyl, benzoyl, 4-methoxybenzoyl or 4-nitrobenzoyl;
- R 2 and R 2' are each independently selected from hydrogen, methyl, methoxy, fluoro, chloro, bromo, iodo or nitrile;
- X is selected from nitrogen or carbon.
- the chemically modified nucleoside is a chemically modified cytosine nucleoside
- the chemically modified cytosine nucleoside is selected from a compound having a structure as shown in Formula 4, a salt thereof, or an isomer thereof:
- R 1 is selected from hydrogen, acetyl, halogen-substituted acetyl, propionyl, halogen-substituted propionyl, butyryl, halogen-substituted butyryl, valeryl, halogen-substituted valeryl, hexanoyl, halogen-substituted hexanoyl, octanoyl, decanoyl, benzoyl, 4-methoxybenzoyl or 4-nitrobenzoyl;
- R2 is selected from hydrogen, methyl, methoxy, fluoro, chloro, bromo, iodo or nitrile;
- R3 is selected from hydroxy, hydrogen, fluorine, methoxy, ethoxy or methoxymethyl ether.
- the chemically modified cytosine nucleoside is selected from: N4-propionyl cytidine, N4-butyryl cytidine, N4-pentanoyl cytidine, N4-hexanoyl cytidine, N4-octanoyl cytidine, N4-decanoyl cytidine, N4-p-nitrobenzoyl cytidine, 5-methyl-N4-butyryl cytidine, 5-methyl-N4-hexanoyl cytidine, 5-methyl-N4-benzoyl cytidine, 2'-fluoro-N4-acetyl cytidine, 2'-fluoro-N4-butyryl cytidine, 2'-fluoro-N4-hexanoyl cytidine, 2'-methoxy cytidine or 2'-ethoxy cytidine
- the chemically modified nucleoside is a chemically modified adenine nucleoside
- the chemically modified adenine nucleoside is selected from a compound having a structure as shown in Formula 5, a salt thereof, or an isomer thereof:
- R 1′ is selected from acetyl, propionyl, butyryl, valeryl, hexanoyl or benzoyl;
- R 2' is hydrogen
- R3 is selected from hydrogen, hydroxy, fluoro or methoxy
- X is selected from nitrogen or carbon.
- Rn is selected from hydroxy, hydrogen, fluorine, methoxy, ethoxy or methoxymethyl ether
- W is selected from a cytosine ring containing a substituent or an adenine ring containing a substituent, wherein the cytosine ring containing a substituent is as shown in Formula 7, and the adenine ring containing a substituent is as shown in Formula 8. Indicates the connection location;
- R 5 and R 5' are each independently selected from hydrogen, methyl, methoxy, fluoro, chloro, bromo, iodo or nitrile;
- R4 is selected from hydrogen, acetyl, halogen-substituted acetyl, propionyl, halogen-substituted propionyl, butyryl, halogen-substituted butyryl, valeryl, halogen-substituted valeryl, hexanoyl, halogen-substituted hexanoyl, octanoyl, decanoyl, benzoyl, 4-methoxybenzoyl or 4-nitrobenzoyl;
- R 5 is selected from hydrogen, methyl, methoxy, fluoro, chloro, bromo, iodo, nitrile;
- R n is selected from hydroxy, hydrogen, fluorine, methoxy, ethoxy or methoxymethyl ether.
- the chemically modified cytosine triphosphate is selected from: N4-acetyl cytidine triphosphate, N4-propionyl cytidine triphosphate, N4-butyryl cytidine triphosphate, N4-pentanoyl cytidine triphosphate, N4-hexanoyl cytidine triphosphate, N4-octanoyl cytidine triphosphate, cytidine triphosphate, 5-methyl-N4-acetyl cytidine triphosphate, 5-methyl-N4-butyryl cytidine triphosphate, 5-methyl-N4-hexanoyl cytidine triphosphate, 5-methyl-N4-benzoyl cytidine triphosphate, 2'-fluoro-N4-acetyl cytidine triphosphate, 2'-fluoro-N4-butyryl cytidine triphosphate, 2'-fluoro
- the present application provides a new chemically modified cytosine triphosphate, which provides more available cytosine triphosphates for mRNA.
- the chemically modified cytosine triphosphates synthesized in the present application can be used for the synthesis of mRNA, can replace natural cytosine triphosphate, alone or in combination with other modified nucleotides, and can play a corresponding translation function, can enhance the translation efficiency and stability of the target mRNA, and reduce immunogenicity, providing a supplement and new choice for mRNA technology beyond the current patented technology.
- the chemically modified nucleoside triphosphate is a chemically modified adenine nucleoside triphosphate
- the chemically modified adenine nucleoside triphosphate is selected from a compound having a structure as shown in Formula 10, a salt thereof, or an isomer thereof:
- R 4′ is selected from acetyl, propionyl, butyryl, valeryl, hexanoyl or benzoyl;
- R 5' is selected from hydrogen
- Rn is selected from hydrogen, hydroxy, fluoro or methoxy
- M is selected from nitrogen or carbon.
- the present application provides a new chemically modified adenine nucleoside and a chemically modified adenine nucleoside triphosphate, which provide more optional raw materials for mRNA technology.
- the natural adenine nucleoside triphosphate in mRNA is completely replaced by the new chemically modified adenine nucleoside triphosphate in the present application, or the mRNA prepared by in vitro transcription is polyadenylated with the chemically modified adenine nucleoside triphosphate, which can enhance the stability and translation efficiency of the target mRNA.
- the chemically modified nucleoside triphosphate is prepared by the following preparation method:
- step (1) tert-butyldimethylsilyl chloride is used to carry out the hydroxyl protection reaction.
- the acid anhydride is selected from acetic anhydride, propionic anhydride, butyric anhydride, valeric anhydride, caproic anhydride, caprylic anhydride, capric anhydride or benzoic anhydride.
- the acyl chloride is selected from 4-methoxybenzoyl chloride or 4-nitrobenzoyl chloride.
- step (3) tetrabutylammonium fluoride is used to carry out a complete deprotection reaction.
- the chlorination agent is selected from phosphorus oxychloride.
- step (1") tert-butyldimethylsilyl chloride is used to carry out the hydroxyl protection reaction.
- the acid anhydride is selected from acetic anhydride, propionic anhydride, butyric anhydride, valeric anhydride, caproic anhydride or benzoic anhydride.
- the acyl chloride is selected from acetyl chloride, propionyl chloride, butyryl chloride, valeryl chloride, hexanoyl chloride or benzoyl chloride.
- the chlorinating agent is selected from phosphorus oxychloride.
- the present application provides a nucleic acid containing chemically modified cytosine nucleosides, wherein during the preparation process of the nucleic acid, the chemically modified cytosine nucleoside triphosphates described in the second aspect are used to completely replace the natural cytosine nucleoside triphosphates in the mRNA, or the chemically modified cytosine nucleoside triphosphates and N1-methylpseudouridine triphosphate are used in combination to replace the natural cytosine nucleoside triphosphates in the mRNA.
- the nucleic acid further contains pseudouridine, N-methylpseudouridine, 5-methoxyuridine or 2-thiouridine.
- the present application synthesizes non-natural modified nucleotides with novel structures and introduces them into mRNA, thereby improving the stability of mRNA and enhancing its translation activity, achieving the goal of replacing, enriching or supplementing existing mRNA chemical modifications, thereby providing assistance for novel chemical modifications and applications of mRNA.
- the nucleic acid comprises at least:
- the present application uses chemical synthesis to synthesize chemically modified cytosine triphosphates, and then uses transcriptase and/or tailing enzyme to synthesize modified mRNA with global replacement of modified cytosine.
- the modified mRNA exhibits enhanced translation efficiency and higher stability both in vitro and in cells.
- the present application provides a nucleic acid containing chemically modified adenine nucleosides, wherein during the preparation process of the nucleic acid, the chemically modified adenine nucleoside triphosphate described in the second aspect is used to completely replace the natural adenine nucleoside triphosphate in the mRNA, or the chemically modified adenine nucleoside triphosphate described in the second aspect is used to perform polyadenylation modification on the mRNA prepared by in vitro transcription.
- natural adenine is chemically modified by chemical synthesis, and these modified adenines are triphosphorylated, and then in vitro transcription is used to synthesize mRNA in which the modified adenines replace the natural adenines globally, or tailing enzymes are used to modify adenosines to perform polyadenylation modification on mRNA.
- modified mRNAs show enhanced translation efficiency and higher stability both in vitro and in cells.
- the chemically modified nucleic acid comprises at least:
- A' a promoter sequence
- the present application provides a method for preparing a nucleic acid containing a chemically modified cytosine nucleoside according to the third aspect, the preparation method comprising:
- PCR reaction is carried out with DNA as a template under the catalysis of RNA polymerase to synthesize chemically modified nucleic acids.
- the RNA polymerase is T7 RNA polymerase.
- the PCR reaction system includes: a DNA template, an RNase inhibitor, T7 RNA polymerase, an RNA polymerase buffer, adenosine triphosphate, guanosine triphosphate, uridine triphosphate, chemically modified cytosine triphosphate and a 5' cap structure.
- the chemically modified nucleic acid is a modified mRNA in which chemically modified cytidine triphosphate is used to globally replace natural cytidine, and the mRNA has higher translation efficiency and is more stable both in a cell-free system and in cells.
- the technical route for the synthesis and translation of mRNA containing chemically modified pyrimidine nucleosides is shown in Figure 1.
- the present application provides a method for preparing a nucleic acid containing chemically modified adenine nucleosides according to the fourth aspect, the preparation method comprising:
- PCR reaction is carried out with DNA as template under the catalysis of RNA polymerase to synthesize chemically modified nucleic acids;
- the mRNA is tailed using E. coli poly (A) polymerase or Yeast poly (A) polymerase to obtain chemically modified nucleic acid.
- the RNA polymerase is T7 RNA polymerase.
- the PCR reaction system includes: a DNA template, an RNase inhibitor, T7 RNA polymerase, an RNA polymerase buffer, chemically modified adenosine triphosphate, guanosine triphosphate, uridine triphosphate, cytosine triphosphate and a 5' cap structure.
- the chemically modified nucleic acid is a modified mRNA in which chemically modified adenosine triphosphate is used to replace natural adenosine triphosphate globally, and the mRNA has higher translation efficiency and is more stable in both cell-free systems and cells.
- the technical route for the synthesis and translation of mRNA containing chemically modified adenosine nucleosides is shown in Figure 1.
- the present application provides the use of any one or a combination of at least two of the chemically modified nucleosides described in the first aspect, the chemically modified nucleoside triphosphates described in the second aspect, the nucleic acids containing chemically modified cytosine nucleosides described in the third aspect, the nucleic acids containing chemically modified adenine nucleosides described in the fourth aspect, the method for preparing nucleic acids containing chemically modified cytosine nucleosides described in the fifth aspect, or the method for preparing nucleic acids containing chemically modified adenine nucleosides described in the sixth aspect in the preparation of mRNA drugs.
- the present application provides completely new chemically modified nucleosides and chemically modified nucleoside triphosphates, including chemically modified cytosine nucleosides, chemically modified cytosine nucleoside triphosphates, chemically modified adenine nucleosides and chemically modified adenine nucleoside triphosphates; more available cytosine nucleoside triphosphates are provided for mRNA; the chemically modified nucleoside triphosphates synthesized in the present application can all be used for the synthesis of mRNA, can replace the corresponding natural nucleosides, and can exert corresponding translation functions, can enhance the translation efficiency and stability of the target mRNA, reduce immunogenicity, and provide a supplement and new option for mRNA technology.
- the mRNA replaced with the chemically modified cytosine triphosphate provided in the present application showed that the translation effect of the mRNA replaced with modified cytosine reached or exceeded the modification method of replacing uridine with pseudouridine and methylpseudouridine used in the currently reported technology for mRNA modification, and at the same time had good biological stability.
- the mRNA replaced with the chemically modified adenine nucleoside triphosphate provided in the present application has good translation effects at the in vitro and cellular levels, which is better than the modification method using pseudouridine to replace uridine.
- the mRNA replaced with the chemically modified adenine nucleoside and pseudouridine provided in the present application has a better translation effect at the in vitro and cellular levels, which is also better than the modification method using pseudouridine to replace uridine.
- the mRNA polyadenylated and tailed with chemically modified adenosine has a better translation effect at the cellular level than the mRNA with a poly-natural adenosine tail, and at the same time has higher stability at the single nucleoside level.
- FIG1 is a technical roadmap for the synthesis and translation of mRNA containing chemically modified nucleosides.
- FIG. 2 is a technical roadmap for the preparation of chemically modified cytidine triphosphate.
- FIG3 is a 1 H NMR spectrum of N4-propionylcytidine triphosphate.
- FIG4 is a 31 P NMR spectrum of N4-propionylcytidine triphosphate.
- FIG5 is a mass spectrum of N4-propionylcytidine triphosphate.
- FIG6 is a 1 H NMR spectrum of N4-butyrylcytidine triphosphate.
- FIG8 is a 1 H NMR spectrum of N4-pentanoyl cytidine triphosphate.
- FIG. 10 is a 1 H NMR spectrum of N4-hexanoyl cytidine triphosphate.
- FIG. 11 is a 31 P NMR spectrum of N4-hexanoyl cytidine triphosphate.
- FIG12 is a 1 H NMR spectrum of 5-methyl-N4-acetylcytidine triphosphate.
- FIG13 is a 31 P NMR spectrum of 5-methyl-N4-acetylcytidine triphosphate.
- FIG14 is a 1 H NMR spectrum of 5-methyl-N4-butyryl cytidine triphosphate.
- FIG. 15 is a 31 P NMR spectrum of 5-methyl-N4-butyryl cytidine triphosphate.
- FIG. 16 is a 1 H NMR spectrum of N4-benzoylcytidine triphosphate.
- FIG. 17 is a 31 P NMR spectrum of N4-benzoylcytidine triphosphate.
- FIG. 18 is a 1 H NMR spectrum of N4-acetylcytidine triphosphate.
- FIG. 19 is a 31 P NMR spectrum of N4-acetylcytidine triphosphate.
- FIG. 20 is a 1 H NMR spectrum of 5-methylcytidine triphosphate.
- FIG. 21 is a 31 P NMR spectrum of 5-methylcytidine triphosphate.
- FIG. 22 is a 1 H NMR spectrum of 5-methyl-N4-hexanoyl cytidine triphosphate.
- FIG. 23 is a 31 P NMR spectrum of 5-methyl-N4-hexanoyl cytidine triphosphate.
- FIG. 24 is a 1 H NMR spectrum of N4-octanoylcytidine triphosphate.
- FIG. 25 is a 1 H NMR spectrum of N4-decanoylcytidine triphosphate.
- FIG26 is a 1 H NMR spectrum of 5-methyl-N4-benzoylcytidine triphosphate.
- FIG27 is a 1 H NMR spectrum of 2'-fluoro-N4-acetylcytidine triphosphate.
- FIG. 28 is a 1 H NMR spectrum of 2'-fluoro-N4-butyrylcytidine triphosphate.
- FIG. 29 is a 1 H NMR spectrum of 2'-fluoro-N4-hexanoyl cytidine triphosphate.
- FIG30 is a mass spectrum of N4-butyrylcytidine triphosphate.
- FIG31 is a mass spectrum of N4-pentanoylcytidine triphosphate.
- FIG32 is a mass spectrum of N4-hexanoyl cytidine triphosphate.
- FIG33 is a mass spectrum of 5-methyl-N4-acetylcytidine triphosphate.
- FIG34 is a mass spectrum of 5-methyl-N4-butyryl cytidine triphosphate.
- FIG35 is a mass spectrum of N4-acetylcytidine triphosphate.
- FIG36 is a mass spectrum of 5-methylcytidine triphosphate.
- FIG37 is a mass spectrum of 5-methyl-N4-hexanoyl cytidine triphosphate.
- FIG38 is a schematic diagram of the in vitro transcription results of chemically modified C-series nucleoside triphosphates.
- Figure 39 shows the expression efficiency of transcribed mRNA after transfection into HEK 293T cells for 24 hours.
- Figure 40 shows the expression efficiency of mRNA transcribed using N1-methylpseudouridine and modified cytidine 24 hours after transfection into HEK 293T cells.
- Figure 41 shows the expression efficiency of transcribed mRNA after transfection into HEK 293T cells for 24 hours.
- Figure 42 is a graph showing the expression efficiency of mRNA transcribed using a combination of N1-methylpseudouridine and modified cytidine 24 hours after transfection into HeLa cells.
- Figure 43 is the qPCR results of the stability of chemically modified mRNA in HEK 293T cells.
- FIG. 44 is a graph showing the qPCR results of the stability test of chemically modified mRNA in HeLa cells.
- Figure 45 shows the protein blotting results of the immunogenicity test of chemically modified mRNA in RAW264.7 cells.
- FIG. 46 is a 1 H NMR characterization chart of N6-propionyl adenosine.
- FIG. 47 is a 13 C NMR characterization chart of N6-propionyl adenosine.
- FIG. 48 is a HRMS characterization chart of N6-propionyl adenosine.
- FIG. 50 is a 13 C NMR characterization chart of N6-propionyl adenosine triphosphate.
- FIG. 51 is a HRMS characterization chart of N6-propionyl ATP.
- FIG. 52 is a 1 H NMR characterization chart of N6-butyryladenosine.
- FIG. 53 is a 13 C NMR characterization chart of N6-butyryladenosine.
- Figure 54 is a HRMS characterization of N6-butyryladenosine.
- FIG. 55 is a 1 H NMR characterization chart of N6-butyryl adenosine triphosphate.
- FIG. 56 is a 13 C NMR characterization chart of N6-butyryl adenosine triphosphate.
- Figure 57 is a HRMS characterization of N6-butyryl adenosine triphosphate.
- FIG. 58 is a 1 H NMR characterization chart of N6-pentanoyl adenosine.
- FIG. 59 is a 13 C NMR characterization chart of N6-pentanoyl adenosine.
- Figure 60 is a HRMS characterization chart of N6-pentanoyl adenosine.
- FIG. 61 is a 1 H NMR characterization chart of N6-pentanoyl adenosine triphosphate.
- FIG. 62 is a 13 C NMR characterization chart of N6-pentanoyl adenosine triphosphate.
- FIG. 63 is a HRMS characterization chart of N6-pentanoyl adenosine triphosphate.
- FIG. 64 is a 1 H NMR characterization chart of N6-hexanoyl adenosine.
- FIG. 65 is a 13 C NMR characterization chart of N6-hexanoyl adenosine.
- Figure 66 is a HRMS characterization chart of N6-hexanoyl adenosine.
- FIG. 67 is a 1 H NMR characterization chart of N6-hexanoyl adenosine triphosphate.
- FIG. 68 is a 13 C NMR characterization chart of N6-hexanoyl adenosine triphosphate.
- FIG. 69 is a HRMS characterization chart of N6-hexanoyl adenosine triphosphate.
- FIG. 70 is a HRMS characterization chart of N6-benzoyl adenosine triphosphate.
- Figure 71 is the HRMS characterization chart of N6-methyladenosine triphosphate.
- Figure 72 is a HRMS characterization chart of N6-acetyl adenosine triphosphate.
- Figure 75 is a diagram showing the effect of polyadenylation modification of eGFP mRNA by E. coli polyA polymerase.
- Figure 76 shows the effect of polyadenylation modification of eGFP mRNA by Yeast polyA polymerase.
- Figure 77 is a graph showing the expression of luciferase mRNA in combination with chemically modified adenosine and pseudouridine after incubation in rabbit reticulocyte lysate for 90 minutes.
- FIG78 is a graph showing the expression effect of luciferase mRNA in which chemically modified adenosine globally replaces natural adenosine after incubation in rabbit reticulocyte lysate for 90 minutes.
- Figure 80 is a graph showing the expression effect of luciferase mRNA combined with chemically modified adenosine and pseudouridine 24 hours after transfection into HeLa cells.
- Figure 81 is a fluorescence imaging image of the expression effect of chemically modified adenosine polyadenylation-modified eGFP mRNA in HEK 293T cells.
- This embodiment provides a method for preparing chemically modified cytidine triphosphate, and the preparation technology route of chemically modified cytidine triphosphate is shown in Figure 2.
- the reaction conditions of each step are as follows, and the structure of the compound is characterized by nuclear magnetic resonance and mass spectrometry:
- step a In the method for preparing chemically modified cytidine triphosphate in this embodiment, only step a, step b and step c are performed to obtain the method for preparing chemically modified cytidine.
- This embodiment provides a chemically modified cytidine triphosphate, wherein the chemically modified cytidine triphosphate is N4-propionyl cytidine triphosphate.
- the reaction conditions of each step of the preparation are as follows, and the structure of the compound is characterized by nuclear magnetic resonance and mass spectrometry:
- Figure 3 is the 1 H NMR spectrum of N4-propionylcytidine triphosphate. It can be seen from the figure that the peak positions (unit: ppm) are: 1.07, 1.09, 1.11; 2.44, 2.46, 2.48, 2.49; 4.27, 4.28, 4.30, 4.31, 4.32, 4.38, 4.40, 4.41; 5.92, 5.93; 7.29, 7.31; 8.39, 8.40.
- Figure 4 is a 31 P NMR spectrum of N4-propionyl cytidine triphosphate, from which it can be seen that the peak positions (unit: ppm) are: -22.70, -22.57, -22.45; -11.48, -11.46, -11.35, -11.34; -6.53, -6.40; 19.45, 19.51, 19.57.
- Figure 5 is a mass spectrum of N4-propionyl cytidine triphosphate.
- This embodiment provides a chemically modified cytidine triphosphate, wherein the chemically modified cytidine triphosphate is N4-butyryl cytidine triphosphate.
- the reaction conditions of each step of the preparation are as follows, and the structure of the compound is characterized by nuclear magnetic resonance and mass spectrometry:
- Figure 6 is the 1 H NMR spectrum of N4-butyrylcytidine triphosphate. It can be seen from the figure that the peak positions (unit: ppm) are: 0.87, 0.89, 0.91; 1.59, 1.61, 1.63, 1.65; 2.41, 2.42, 2.44; 4.24, 4.27, 4.28, 4.30, 4.31, 4.32, 4.39, 4.40, 4.42; 5.92, 5.93; 7.28, 7.30; 8.39, 8.41.
- Figure 7 is a 31 P NMR spectrum of N4-butyryl cytidine triphosphate, from which it can be seen that the peak positions (unit: ppm) are: -22.68, -22.56, -22.43; -11.47, -11.45, -11.34, -11.33; -6.46, -6.33; 19.52, 19.58.
- Figure 30 is a mass spectrum of N4-butyryl cytidine triphosphate.
- This embodiment provides a chemically modified cytidine triphosphate, wherein the chemically modified cytidine triphosphate is N4-pentanoyl cytidine triphosphate.
- the reaction conditions of each step of the preparation are as described in Example 1.
- the anhydride used in step b is valeric anhydride.
- Figure 8 is the 1 H NMR spectrum of N4-pentanoylcytidine triphosphate. It can be seen from the figure that the peak positions (unit: ppm) are: 0.80, 0.82, 0.84; 1.26, 1.28; 1.52, 1.53, 1.55, 1.57, 1.59; 2.40, 2.42, 2.44; 4.21, 4.24, 4.25, 4.27, 4.28, 4.29, 4.35, 4.37, 4.38; 5.89, 5.90; 7.25, 7.27; 8.36, 8.38.
- Figure 9 is a 31 P NMR spectrum of N4-pentanoyl cytidine triphosphate, from which it can be seen that the peak positions (unit: ppm) are: -22.69, -22.57, -22.44; -11.51, -11.49, -11.39, -11.36, -11.35, -11.32; -6.54, -6.41.
- Figure 31 is a mass spectrum of N4-pentanoyl cytidine triphosphate.
- This embodiment provides a chemically modified cytidine triphosphate, wherein the chemically modified cytidine triphosphate is N4-hexanoyl cytidine triphosphate.
- the reaction conditions of each step of the preparation are referred to Example 1.
- the anhydride used in step b is hexanoic anhydride.
- Figure 10 is a 1 H NMR spectrum of N4-hexanoyl cytidine triphosphate. It can be seen from the figure that the peak positions (unit: ppm) are: 0.77, 0.79, 0.80; 1.23, 1.24; 1.56, 1.58, 1.60; 2.39, 2.41, 2.43; 4.24, 4.25, 4.28, 4.35, 4.37, 4.38; 5.90; 7.26, 7.28; 8.36, 8.38.
- Figure 11 is a 31 P NMR spectrum of N4-hexanoyl cytidine triphosphate.
- FIG32 is a mass spectrum of N4-hexanoyl cytidine triphosphate.
- This embodiment provides a chemically modified cytidine triphosphate, wherein the chemically modified cytidine triphosphate is 5-methyl-N4-acetyl cytidine triphosphate.
- the reaction conditions of each step of the preparation are referred to Example 1, except that the reaction starting material used is 5-methyl-N4-acetyl cytidine triphosphate.
- Cytidine, the acid anhydride used in step b is acetic anhydride.
- FIG33 is a mass spectrum of 5-methyl-N4-acetylcytidine triphosphate.
- This embodiment provides a chemically modified cytidine triphosphate, wherein the chemically modified cytidine triphosphate is 5-methyl-N4-butyryl cytidine triphosphate.
- the reaction conditions of each step of the preparation refer to those of Example 1, except that the reaction starting material used is 5-methylcytidine, and the anhydride used in step b is butyric anhydride.
- Figure 14 is the 1 H NMR spectrum of 5-methyl-N4-butyryl cytidine triphosphate. It can be seen from the figure that the peak positions (unit: ppm) are: 0.84, 0.86, 0.88; 1.56, 1.57, 1.59, 1.61; 2.03; 2.43, 2.45, 2.47; 4.19, 4.21, 4.24, 4.25, 4.25, 4.36, 4.37, 4.39; 5.87, 5.88; 8.22.
- Figure 15 is a 31 P NMR spectrum of 5-methyl-N4-butyryl cytidine triphosphate.
- Figure 34 is a mass spectrum of 5-methyl-N4-butyryl cytidine triphosphate.
- Figure 16 is the 1 H NMR spectrum of N4-benzoylcytidine triphosphate; it can be seen from the figure that the peak positions (unit: ppm) are: 4.28, 4.28, 4.29, 4.30, 4.31, 4.33, 4.34, 4.34, 4.35, 4.39, 4.41, 4.42; 5.96, 5.97; 7.43, 7.45, 7.51, 7.53, 7.55, 7.63, 7.65, 7.67; 7.87, 7.87, 7.89; 8.45, 8.47.
- Figure 17 is the 31 P NMR spectrum of N4-benzoylcytidine triphosphate.
- the peak positions are: -22.88, -22.75, -22.63; -11.51, -11.48, -11.46, -11.43, -11.38, -11.35, -11.34, -11.31; -7.88, -7.77.
- This embodiment provides a chemically modified cytidine triphosphate, wherein the chemically modified cytidine triphosphate is N4-acetyl cytidine triphosphate.
- the reaction conditions of each step of the preparation are as described in Example 1.
- the anhydride used in step b is acetic anhydride.
- Figure 18 is a 1 H NMR spectrum of N4-acetylcytidine triphosphate. It can be seen from the figure that the peak positions (unit: ppm) are: 2.18; 4.24, 4.27; 4.28; 4.31, 4.31, 4.32, 4.39, 4.40, 4.41; 5.92, 5.93; 7.26, 7.27; 8.39, 8.41.
- Figure 19 is a 31 P NMR spectrum of N4-acetylcytidine triphosphate. It can be seen from the figure that the peak positions (unit: ppm) are: -22.68, -22.56, -22.43; -11.46, -11.34; -6.46, -6.33.
- Figure 35 is a mass spectrum of N4-acetylcytidine triphosphate.
- This embodiment provides a chemically modified cytidine triphosphate, wherein the chemically modified cytidine triphosphate is N4-p-methoxybenzoyl cytidine triphosphate.
- the reaction conditions of each step of the preparation are referred to Example 1.
- the acyl chloride used in step b is p-methoxybenzoyl chloride (4-methoxybenzoyl chloride).
- Figure 20 is a 1 H NMR spectrum of 5-methylcytidine triphosphate. It can be seen from the figure that the peak positions (unit: ppm) are: 1.91; 4.15, 4.16, 4.18; 4.23, 4.25, 4.26; 4.36, 4.37, 4.38; 5.91, 5.92; 7.73.
- Figure 21 is a 31 P NMR spectrum of 5-methylcytidine triphosphate. It can be seen from the figure that the peak positions (unit: ppm) are: -23.09, -22.66, -22.54, -22.41; -11.68, -11.56; -7.53, -6.49, -6.36.
- Figure 36 is a mass spectrum of 5-methylcytidine triphosphate.
- This embodiment provides a chemically modified cytidine triphosphate, wherein the chemically modified cytidine triphosphate is 5-methyl-N4-hexanoyl cytidine triphosphate.
- the reaction conditions of each step of the preparation are referred to Example 1, except that the reaction starting material used is 5-methylcytidine, and the acid anhydride used in step b is hexanoic anhydride.
- Figure 22 is the 1 H NMR spectrum of 5-methyl-N4-hexanoyl cytidine triphosphate. It can be seen from the figure that the peak positions (unit: ppm) are: 0.79, 0.80, 0.80, 0.82; 1.24, 1.25, 1.26; 1.60, 1.62; 2.06, 2.06; 2.49, 2.51, 2.52; 4.24, 4.25, 4.28, 4.28, 4.34, 4.35, 4.37; 5.90, 5.91; 8.22.
- Figure 23 is a 31 P NMR spectrum of 5-methyl-N4-hexanoyl cytidine triphosphate, from which it can be seen that the peak positions (unit: ppm) are: -22.68, -22.56, -22.43; -11.73, -11.61; -6.49, -6.36.
- Figure 37 is a mass spectrum of 5-methyl-N4-hexanoyl cytidine triphosphate.
- This example provides a chemically modified cytidine triphosphate, wherein the chemically modified cytidine triphosphate is N4-octanoyl cytidine triphosphate, and the reaction conditions of each step of the preparation are referred to Example 1.
- the anhydride used in step b is octanoic anhydride.
- Figure 24 is the 1 H NMR spectrum of N4-octanoylcytidine triphosphate. It can be seen from the figure that the peak positions (unit: ppm) are: 0.74, 0.76, 0.78; 1.23, 1.32, 1.34, 1.56, 1.57, 1.59; 2.39, 2.41, 2.43; 4.24, 4.25, 4.26, 4.28, 4.29, 4.30; 5.89, 5.90; 7.24, 7.26, 7.27; 8.32, 8.34.
- This example provides a chemically modified cytidine triphosphate, wherein the chemically modified cytidine triphosphate is N4-decanoyl cytidine triphosphate, and the reaction conditions of each step of the preparation are as described in Example 1.
- the anhydride used in step b is decanoic anhydride.
- Figure 25 is the 1 H NMR spectrum of N4-decanoylcytidine triphosphate. It can be seen from the figure that the peak positions (unit: ppm) are: 0.73, 0.75, 0.76; 1.20, 1.22; 1.56, 1.57, 1.59; 2.40, 2.42, 2.44; 4.24, 4.26, 4.27, 4.28, 4.30; 5.89, 5.90; 7.19, 7.21; 8.36, 8.38.
- This embodiment provides a chemically modified cytidine triphosphate, wherein the chemically modified cytidine triphosphate is 5-methyl-N4-benzoyl
- the reaction conditions of each step of the preparation are referred to Example 1, except that the reaction starting material used is 5-methylcytidine, and the acid anhydride used in step b is benzoic anhydride.
- Figure 26 is the 1 H NMR spectrum of 5-methyl-N4-benzoylcytidine triphosphate. It can be seen from the figure that the peak positions (unit: ppm) are: 2.10; 4.24, 4.25, 4.27, 4.28, 4.31, 4.32, 4.33, 4.41, 4.42, 4.43; 5.92, 5.93; 7.47, 7.49, 7.51, 7.59, 7.61, 7.63; 7.86, 7.88; 8.24.
- This embodiment provides a chemically modified cytidine triphosphate, wherein the chemically modified cytidine triphosphate is 2'-fluoro-N4-acetyl cytidine triphosphate, and the reaction conditions of each step of the preparation are referred to Example 1, except that the reaction starting material used is 2'-fluorocytidine, and the acid anhydride used in step b is acetic anhydride.
- Figure 27 is the 1 H NMR spectrum of 2'-fluoro-N4-acetylcytidine triphosphate. It can be seen from the figure that the peak positions (unit: ppm) are: 2.14; 4.22, 4.24, 4.25, 4.28, 4.30, 4.33, 4.36, 4.40, 4.41, 4.42, 4.44, 4.47, 4.48, 4.49, 4.50; 4.98, 4.99; 5.11, 5.12; 5.98, 6.02; 7.23, 7.25; 8.35, 8.37.
- This embodiment provides a chemically modified cytidine triphosphate, wherein the chemically modified cytidine triphosphate is 2'-fluoro-N4-butyryl cytidine triphosphate.
- the reaction conditions of each step of the preparation are referred to Example 1, except that the reaction starting material used is 2'-fluorocytidine, and the anhydride used in step b is butyric anhydride.
- Figure 28 is the 1 H NMR spectrum of 2'-fluoro-N4-butyryl cytidine triphosphate. It can be seen from the figure that the peak positions (unit: ppm) are: 0.83, 0.84, 0.86; 1.55, 1.56, 1.58, 1.60; 2.36, 2.38, 2.40; 4.22, 4.24, 4.27, 4.28, 4.30, 4.33, 4.36, 4.40, 4.41, 4.43, 4.43, 4.47, 4.48, 4.49, 4.50; 4.98, 4.99; 5.11, 5.12; 5.98, 6.02; 7.25, 7.27; 8.34, 8.36.
- This embodiment provides a chemically modified cytidine triphosphate, wherein the chemically modified cytidine triphosphate is 2'-fluoro-N4-hexanoyl cytidine triphosphate.
- the reaction conditions of each step of the preparation are referred to Example 1, except that the reaction starting material used is 2'-fluorocytidine, and the acid anhydride used in step b is hexanoic anhydride.
- Figure 29 is the 1 H NMR spectrum of 2'-fluoro-N4-hexanoyl cytidine triphosphate. It can be seen from the figure that the peak positions (unit: ppm) are: 0.76, 0.77, 0.79; 1.22, 1.22; 1.54, 1.56, 1.58; 2.38, 2.40, 2.41; 4.22, 4.25, 4.27, 4.34, 4.37, 4.39, 4.40, 4.41, 4.42, 4.46, 4.47, 4.48, 4.49; 4.98, 4.99; 5.11, 5.12; 5.98, 6.02; 7.26, 7.27; 8.33, 8.36.
- This example provides a chemically modified cytidine triphosphate, wherein the chemically modified cytidine triphosphate is 2'-methoxycytidine triphosphate.
- the reaction conditions of each step of the preparation are referred to Example 1, except that the reaction starting material used is 2'-methoxycytidine.
- This example provides a chemically modified cytidine triphosphate, wherein the chemically modified cytidine triphosphate is 2'-ethoxycytidine triphosphate.
- the reaction conditions of each step of the preparation are referred to Example 1, except that the reaction starting material used is 2'-ethoxycytidine.
- the plasmid encoding luciferase is used as a template and DNA polymerase is used in a PCR tube.
- the specific steps are as follows:
- the luciferase DNA template was prepared by PCR and carried out in a 96-well PCR instrument.
- the PCR product includes at least: A) a promoter sequence (T7 promoter); B) a 5'UTR containing at least one Kozak sequence; C) a 3'UTR; D) a luciferase coding sequence; E) a poly A tail.
- a promoter sequence T7 promoter
- E) a poly A tail The specific sequences involved in this embodiment are shown in Table 1.
- the DNA template was amplified according to the following reaction system: reaction volume, 50 ⁇ L (reaction volume of a single tube, multiple tubes were reacted at one time), and the specific reaction system is shown in Table 2.
- the reaction program was as follows: pre-denaturation at 95°C for 1 min, denaturation at 95°C for 15 s, annealing at 52°C for 5 s, extension at 72°C for 2 min, for a total of 32 cycles; and final extension at 72°C for 5 min.
- reaction solutions are combined in a 1.5 mL tube, and 2 ⁇ L is taken for DNA agarose gel electrophoresis to confirm the success of the reaction (agarose gel electrophoresis detection conditions: 2% agarose, 5 V/min, 30 min).
- the reaction product was purified using HiPure Gel Pure DNA Mini Kit.
- the purification process is as follows:
- the prepared chemically modified nucleosides are used to globally replace natural nucleosides to synthesize chemically modified mRNA.
- the chemically modified cytidine triphosphate includes modifications as follows: N4-acetyl, N4-propionyl, N4-butyryl, N4-pentanoyl, N4-hexanoyl, N4-benzoyl, 5-methyl-N4-acetyl, 5-methyl-N4-butyryl, 5-methyl-N4-hexanoyl, 5-methyl.
- the synthesis of chemically modified mRNA was achieved by in vitro transcription using T7 RNA polymerase. Taking a 50 ⁇ L reaction system as an example, the components listed in Table 3 were added sequentially into a 0.2 mL PCR reaction tube.
- CTP* is cytosine triphosphate or chemically modified cytosine triphosphate.
- Cap Analogue# is a commercially available cap structure analog; in this example, the one used is 3'-O-Me-m 7 -G(5')ppp(5')G RNA Cap Structure Analog provided by New England Biolabs, with the catalog number S1411S.
- the reaction procedure is as follows: Turn on the thermostat's hot cover and set it to 60°C. Click the thermostat reaction system, 37°C, 2-4h. After the incubation is completed, the mRNA purification method is as follows:
- the obtained mRNA product was dissolved in 20-50 ⁇ L of nuclease-free water, and the concentration of the purified mRNA was measured using an ultra-micro spectrophotometer, and the ratios of 260/230 and 260/280 were also measured simultaneously.
- Figure 38 is a schematic diagram of the in vitro transcription results of chemically modified C-series nucleoside triphosphates.
- lane 1 is natural cytosine
- lane 2 is 5-methylcytosine
- lane 3 is 5-methyl-N4-acetylcytosine
- lane 4 is 5-methyl-N4-butyrylcytosine
- lane 5 is 5-methyl-N4-hexanoyl
- lane 6 is 5-methyl-N4-benzoylcytosine
- lane 7 is N4-acetylcytosine
- lane 8 is N4-propionylcytosine
- lane 9 is N4-butyrylcytosine
- lane 10 is the mRNA of N4-hexanoylcytosine
- lane M is the RNA molecular weight standard.
- a tailing reaction is required here.
- the specific steps of the tailing reaction are as follows: prepare the reaction system as shown in Table 7 in a 0.2mL PCR Tube at room temperature; Table 4 is the tailing reaction system, and the total volume is 20 ⁇ L.
- the reaction procedure is as follows: set the thermostatic reactor with a lid temperature of 60°C and incubate at 37°C for 30 minutes. After the incubation is completed, the tailed mRNA is purified by the cellulose method described above, and then the mRNA tail length and the integrity of the tailed mRNA fragment are detected by agarose gel electrophoresis.
- Example 24 In vitro transcription synthesis of mRNA using chemically modified nucleosides and N1-methylpseudouridine to globally replace natural nucleosides
- Example 23 The same method as in Example 23 was used, except that in this example, chemically modified cytosine triphosphate and N1-methyl pseudouridine triphosphate were mixed in a 50/50 ratio, and natural cytosine triphosphate was replaced globally to perform in vitro transcription synthesis of luciferase mRNA.
- Example 25 Expression efficiency of chemically modified mRNA in cells
- the chemically modified mRNA is transfected into HEK 293T cells and HeLa cells, respectively, and the luminescence intensity is detected.
- Figure 39 is a graph showing the expression efficiency of transcribed mRNA transfected into HEK 293T cells 24 hours after transfection.
- the horizontal axis in the figure is the sample type. In the figure:
- the translation efficiency of luciferase mRNA with global replacement of N4-propionylcytosine, N4-butyrylcytosine, N4-pentanoylcytosine and N4-hexanoylcytosine in HEK 293T cells is higher than that of luciferase mRNA transcribed from natural cytosine, among which the translation efficiency of luciferase mRNA with global replacement of N4-butyrylcytosine can be enhanced to 2.91 times, proving that chemically modified cytosine can effectively enhance the translation efficiency of transcribed mRNA.
- the translation efficiency of luciferase mRNA with global replacement of N4-propionylcytosine, N4-butyrylcytosine, N4-pentanoylcytosine and N4-hexanoylcytosine in HeLa cells is higher than that of luciferase mRNA transcribed from natural cytosine, among which the translation efficiency of luciferase mRNA with global replacement of N4-propionylcytosine, N4-butyrylcytosine and N4-pentanoylcytosine can be enhanced by more than 10 times, proving that chemically modified cytosine can effectively enhance the translation efficiency of transcribed mRNA.
- Figure 40 is a graph showing the expression efficiency of mRNA transcribed by N1-methyl pseudouridine and modified cytidine after transfection into HEK 293T cells for 24 hours. In the figure:
- the translation efficiency of luciferase mRNA transcribed by combining N4-acetylcytidine triphosphate with N1-methyl pseudouridine triphosphate, N4-propionylcytidine triphosphate with N1-methyl pseudouridine triphosphate in HEK 293T cells was enhanced by 2.63-3.32 times.
- the mRNA with global replacement of N1-methyl pseudouridine triphosphate alone can achieve an enhanced translation efficiency of 1.87 times.
- FIG42 is a graph showing the expression efficiency of mRNA transcribed by N1-methyl pseudouridine and modified cytidine after transfection into HeLa cells for 24 hours, in which:
- the translation efficiency of luciferase mRNA transcribed by N4-acetylcytidine triphosphate, N4-propionylcytidine triphosphate, N4-butyrylcytidine triphosphate, and N4-pentanoylcytidine triphosphate in HeLa cells was enhanced by 1.66 to 10.82 times after the combination of N4-acetylcytidine triphosphate, N4-propionylcytidine triphosphate, N4-butyrylcytidine triphosphate and N1-methyl pseudouridine.
- the translation efficiency of mRNA replaced by N1-methyl pseudouridine alone was enhanced by 9.5 times.
- mRNA encoding luciferase containing a single type of cytidine modification was synthesized and its stability in cells was tested; its stability refers to the relative amount of residual intact mRNA extracted after the mRNA was transfected into the cells for a certain period of time.
- the chemically modified mRNA was transfected into HEK 293T cells and HeLa cells, and qPCR detection was performed.
- the specific steps of chemically modified mRNA transfection into HEK 293T cells and qPCR detection are as follows:
- Example 25 Cell inoculation and cell transfection steps are described in Example 25. 3h after transfection, the culture medium was discarded, the cells were rinsed once with 1 ⁇ PBS, and then the PBS washing solution was aspirated. 200 ⁇ L TransZol Up lysis solution (Quanshi Jin Bio, ET1111) was added, and it was placed horizontally for a moment to evenly distribute the lysis solution on the cell surface and lyse the cells, and then the cells were blown off with a pipette. After that, the lysis solution was transferred to a centrifuge tube, and it was blown repeatedly with a pipette until there was no obvious precipitation in the lysate, and it was left to stand at room temperature for 5 minutes.
- TransZol Up lysis solution Quanshi Jin Bio, ET1111
- FIG43 is a graph showing the qPCR results of the stability of chemically modified mRNA in HEK 293T cells, in which:
- the stability of luciferase mRNA with global replacement of N4-propionylcytosine, N4-butyrylcytosine, N4-pentanoylcytosine, and N4-hexanoylcytosine in HEK 293T cells is higher than that of luciferase mRNA transcribed from natural cytosine, proving that global replacement of chemically modified cytosine can enhance the stability of transcribed mRNA in cells.
- Figure 44 is a graph showing the qPCR results of the stability test of chemically modified mRNA in HeLa cells, in which:
- the stability of luciferase mRNA with global replacement of N4-propionylcytosine, N4-pentanoylcytosine and N4-hexanoylcytosine in HEK 293T cells is higher than that of luciferase mRNA transcribed from natural cytosine, proving that global replacement of chemically modified cytosine can enhance the stability of transcribed mRNA in cells.
- the cell inoculation and cell transfection steps refer to those described in Example 25.
- the cells used in this example are macrophages RAW264.7. 24 hours after cell transfection, digest the cells with 200 ⁇ L 1 ⁇ Cell Lysis Buffer (Abcam, ab152163) for 5 minutes, then blow down the cells with a pipette and collect them in a 1.5 mL centrifuge tube. Centrifuge at 14000g for 10 minutes at 4°C, and then take 170 ⁇ L of the supernatant solution in a new 1.5 mL centrifuge tube. Take 50 ⁇ L of the extracted protein solution, add 100 ⁇ L of BCA protein quantification kit solution (Yishen Bio, 20201ES76), and then blow down with a pipette.
- BCA protein quantification kit solution Yishen Bio, 20201ES76
- the immunogenicity of luciferase mRMA is expressed by the amount of cytokines secreted by it after stimulating RAW264.7 cells, including tumor necrosis factor ⁇ and interleukin 6.
- the amount of cytokines was quantified by western blotting, and the specific steps were as follows: 20 ⁇ g of total protein sample was taken, 1/5 volume of 6 ⁇ Protein Loading Buffer (All-Gold Bio, DL101) was added, and then boiled at 95°C for 10 minutes, and then cooled to 4°C on ice. After a brief centrifugation, all samples were separated by 12% SDS-PAGE gel. The sample was then transferred to a PVDF membrane, and the membrane was blocked with 5% skim milk at room temperature for 2 hours, and then rinsed with 1 ⁇ TBST Buffer 3 times, 20 mL each time.
- Protein Loading Buffer All-Gold Bio, DL101
- the corresponding primary antibodies are the primary antibodies of the target protein and the primary antibodies of the internal reference protein, so there are two types, including: recombinant Anti-TNF alpha antibody, EPR19147, Abcam and Antib-beta Actin antibody, AC-15, Abcam) diluted 1:3000 were incubated at 4°C overnight. Then, the membrane was rinsed 3 times with 1 ⁇ TBST Buffer, 20 mL each time.
- the membrane was incubated with the corresponding secondary antibody (Goat Anti-Rabbit IgG H&L HRP, ab6721, Abcam and Goat Anti-Mouse IgG H&L HRP, ab6728, Abcam) diluted 1:5000 for 1 hour, and then the membrane was rinsed 3 times with 1 ⁇ TBST Buffer, 10 mL each time. Then, the enhanced ECL chemiluminescence exposure reagent (Yisheng Bio, 36222ES60) was prepared for chemical exposure. The relative content of the produced cytokines was expressed as the intensity of the extracted bands.
- the corresponding secondary antibody Goat Anti-Rabbit IgG H&L HRP, ab6721, Abcam and Goat Anti-Mouse IgG H&L HRP, ab6728, Abcam
- Figure 45 is a Western blot result of the immunogenicity test of chemically modified mRNA in RAW264.7 cells, in which:
- the luciferase mRNA with global replacement of chemically modified cytosine did not show weakened immunogenicity in RAW264.7 cells.
- This embodiment provides a method for preparing chemically modified adenosine triphosphate, and the preparation technology route is as follows.
- R 5' of the chemically modified adenosine triphosphate is hydrogen
- R n is hydroxyl
- M is nitrogen.
- the structure of the compound is characterized by nuclear magnetic resonance and liquid chromatography-mass spectrometry.
- step a In the method for preparing chemically modified adenosine triphosphate in this embodiment, only step a, step b and step c are performed to form the method for preparing chemically modified adenosine, which will not be described in detail here.
- This embodiment provides a method for preparing chemically modified adenosine and chemically modified adenosine triphosphate, wherein the chemically modified adenosine is N6-propionyl adenosine, and the chemically modified adenosine triphosphate is N6-propionyl adenosine triphosphate, and the structures are shown below.
- the 1 H NMR characterization of N6-propionyl adenosine is shown in FIG46
- the 13 C NMR characterization of N6-propionyl adenosine is shown in FIG47
- the HRMS characterization of N6-propionyl adenosine is shown in FIG48 .
- This embodiment provides a method for preparing chemically modified adenosine and chemically modified adenosine triphosphate, wherein the chemically modified adenosine is N6-butyryladenosine, and the chemically modified adenosine triphosphate is N6-butyryladenosine triphosphate, and the structures are shown below.
- Example 29 The difference between the preparation method and Example 29 is that butyryl chloride is used for the reaction in step b, the reaction conditions of each preparation step refer to Example 29, and the structure of the compound is characterized by nuclear magnetic resonance and mass spectrometry.
- the 1 H NMR characterization chart of N6-butyryladenosine is shown in Figure 52
- the 13 C NMR characterization chart of N6-butyryladenosine is shown in Figure 53
- the HRMS characterization chart of N6-butyryladenosine is shown in Figure 54
- the 1 H NMR characterization chart of N6-butyryladenosine triphosphate is shown in Figure 55
- the 13 C NMR characterization chart of N6-butyryladenosine triphosphate is shown in Figure 56
- the HRMS characterization chart of N6-butyryladenosine triphosphate is shown in Figure 57.
- This embodiment provides a method for preparing chemically modified adenosine and chemically modified adenosine triphosphate, wherein the chemically modified adenosine is N6-pentanoyl adenosine, and the chemically modified adenosine triphosphate is N6-pentanoyl adenosine triphosphate, and the structures are shown below.
- Example 29 The difference between the preparation method and Example 29 is that valeryl chloride is used for the reaction in step b, the reaction conditions of each step of the preparation refer to Example 29, and the structure of the compound is characterized by nuclear magnetic resonance and mass spectrometry.
- the 1 H NMR characterization chart of N6-pentanoyl adenosine is shown in Figure 58
- the 13 C NMR characterization chart of N6-pentanoyl adenosine is shown in Figure 59
- the HRMS characterization chart of N6-pentanoyl adenosine is shown in Figure 60.
- the 1 H NMR characterization chart of N6-pentanoyl adenosine triphosphate is shown in Figure 61
- the 13 C NMR characterization chart of N6-pentanoyl adenosine triphosphate is shown in Figure 62
- the HRMS characterization chart of N6-pentanoyl adenosine triphosphate is shown in Figure 63.
- This embodiment provides a method for preparing chemically modified adenosine and chemically modified adenosine triphosphate, wherein the chemically modified adenosine is N6-hexanoyl adenosine, and the chemically modified adenosine triphosphate is N6-hexanoyl adenosine triphosphate, and the structures are shown below.
- Example 29 The difference between the preparation method and Example 29 is that hexanoyl chloride is used for the reaction in step b, the reaction conditions of each preparation step refer to Example 29, and the structure of the compound is characterized by nuclear magnetic resonance and mass spectrometry.
- the 1 H NMR characterization chart of N6-hexanoyl adenosine is shown in Figure 64
- the 13 C NMR characterization chart of N6-hexanoyl adenosine is shown in Figure 65
- the HRMS characterization chart of N6-hexanoyl adenosine is shown in Figure 66.
- the 1 H NMR characterization chart of N6-hexanoyl adenosine triphosphate is shown in Figure 67
- the 13 C NMR characterization chart of N6-hexanoyl adenosine triphosphate is shown in Figure 68
- the HRMS characterization chart of N6-hexanoyl adenosine triphosphate is shown in Figure 69.
- This embodiment provides a method for preparing chemically modified adenosine triphosphate, wherein the chemically modified adenosine triphosphate is N6-benzoyl adenosine triphosphate, and the structure is shown below.
- Example 29 The difference between the preparation method and Example 29 is that benzoic anhydride is used for the reaction in step b, the reaction conditions of each step of the preparation are referred to Example 29, and the structure of the compound is characterized by nuclear magnetic resonance and mass spectrometry.
- the HRMS characterization chart of N6-benzoyl adenosine triphosphate is shown in Figure 70.
- This embodiment provides a method for preparing chemically modified adenosine triphosphate, wherein the chemically modified adenosine triphosphate is N6-methyladenosine triphosphate, and the structure is shown below.
- Example 29 The difference between the preparation method and Example 29 is that commercial N6-methyladenosine (CAS No.: 1867-73-8) is used as a raw material for triphosphorylation reaction, the reaction conditions of each step of the preparation are referred to Example 29, and the structure of the compound is characterized by nuclear magnetic resonance and mass spectrometry.
- the HRMS characterization chart of N6-methyladenosine triphosphate is shown in Figure 71.
- This embodiment provides a method for preparing chemically modified adenosine triphosphate, wherein the chemically modified adenosine triphosphate is N6-acetyl adenosine triphosphate, and the structure is shown below.
- Example 29 The difference between the preparation method and Example 29 is that acetyl chloride is used for reaction in step b, and the reaction of each step of the preparation is The conditions were similar to those in Example 29, and the structure of the compound was characterized by nuclear magnetic resonance and mass spectrometry.
- the HRMS characterization graph of N6-acetyl adenosine triphosphate is shown in FIG72 .
- This embodiment provides a method for preparing chemically modified adenosine triphosphate, wherein the chemically modified adenosine triphosphate is 2-aminoadenosine triphosphate, and the structure is shown below.
- Example 29 The difference between the preparation method and Example 29 is that commercial 2-aminoadenosine (CAS No.: 2096-10-8) is directly used as a raw material for the reaction, the reaction conditions of each step of the preparation are referred to Example 29, and the structure of the compound is characterized by nuclear magnetic resonance and mass spectrometry.
- the HRMS characterization chart of 2-aminoadenosine triphosphate is shown in Figure 73.
- This embodiment provides a method for preparing chemically modified adenosine triphosphate, wherein the chemically modified adenosine triphosphate is 7-deazaadenosine triphosphate, and the structure is shown below.
- Example 29 The difference between the preparation method and Example 29 is that commercial 7-deazaadenosine (CAS No.: 69-33-0) is directly used as a raw material for the reaction, the reaction conditions of each step of the preparation are referred to Example 29, and the structure of the compound is characterized by nuclear magnetic resonance and mass spectrometry.
- the HRMS characterization chart of 7-deazaadenosine triphosphate is shown in Figure 74.
- the DNA template for compiling enhanced green fluorescent protein is prepared by linearizing the plasmid containing the target gene with an endonuclease.
- Enzyme digestion was used to prepare an enhanced green fluorescent protein linear DNA template in a 96-well PCR instrument.
- the linearized product at least includes: A) a promoter sequence (T7 promoter); B) a 5'UTR containing at least one Kozak sequence; C) a 3'UTR; D) an eGFP coding sequence.
- T7 promoter a promoter sequence
- C a 3'UTR
- D an eGFP coding sequence.
- reaction volume 20 ⁇ L (reaction volume of a single tube, multiple tubes were reacted at one time), and the specific reaction system is shown in Table 6.
- the enzyme digestion product was purified using HiPure Gel Pure DNA Mini Kit (Guangzhou Meiji Biotechnology Co., Ltd., catalog number: D2111). The purification process is as follows:
- DNA amplification was performed in a PCR tube using DNA polymerase. The specific steps are as follows:
- the luciferase DNA template was prepared by PCR and carried out in a 96-well PCR instrument.
- the PCR product contains at least: A) a promoter sequence (T7 promoter); B) at least one Kozak sequence 5'UTR; C) 3'UTR; D) luciferase coding sequence; E) poly A tail (polyA tail).
- a promoter sequence T7 promoter
- D) luciferase coding sequence E) poly A tail (polyA tail).
- reaction volume reaction volume of a single tube, multiple tubes were reacted at one time
- reaction system is shown in Table 8.
- the reaction program was as follows: pre-denaturation at 95°C for 1 minute, denaturation at 95°C for 15 seconds, annealing at 52°C for 5 seconds, extension at 72°C for 2 minutes, for a total of 32 cycles; and final extension at 72°C for 5 minutes.
- reaction solution is combined in a 1.5 mL tube. 2 ⁇ L is taken for DNA agarose gel electrophoresis to confirm the success of the reaction (agarose gel electrophoresis detection conditions: 2% agarose, 5 V/min, electrophoresis for 30 minutes).
- the reaction product was purified using HiPure Gel Pure DNA Mini Kit.
- the purification process is as follows:
- Example 40 In vitro transcription synthesis of mRNA in which chemically modified adenosine globally replaces natural adenosine
- RNA polymerase The synthesis of mRNA in which chemically modified adenosine globally replaces natural adenosine is achieved by in vitro transcription using T7 RNA polymerase. Taking a 50 ⁇ L reaction system as an example, the components listed in Table 9 are added sequentially into a 0.2 mL PCR reaction tube.
- Cap Analogue# is a commercially available cap structure analog; in this example, the one used is 3’-O-Me-m7-G(5’)ppp(5’)G RNA Cap Structure Analog provided by New England Biolabs, with the catalog number S1411S.
- the chemically modified adenine nucleosides include the following: N6-methyladenosine, N6-acetyladenosine, N6-propionyladenosine, N6-butyryladenosine, N6-pentanoyladenosine, N6-hexanoyladenosine, N6-benzoyladenosine, 2-aminoadenosine or 7-deazaadenosine, etc.
- Example 41 In vitro transcription and synthesis of mRNA using chemically modified adenosine and pseudouridine to globally replace natural adenosine and natural uridine
- Example 40 The conditions and operation methods of this example are exactly the same as those of Example 40, except that natural uridine is completely replaced by pseudouridine in the substrate for in vitro transcription. The rest of the purification and characterization methods are exactly the same.
- Cap Analogue# is a commercially available cap structure analog; in this example, 3’-O-Me-m7-G(5’)ppp(5’)G RNA Cap Structure Analog provided by New England Biolabs was used, with the catalog number S1411S.
- Chemically modified adenosine is used to perform polyadenylation modification on in vitro transcribed eGFP mRNA using E. coli polyA polymerase or Yeast polyA polymerase. Taking a 20 ⁇ L reaction system as an example, add the components shown in Tables 11 and 12 in a 0.2 mL PCR tube.
- buffer A * is adenosine triphosphate or chemically modified adenosine triphosphate.
- the components of buffer A (5 ⁇ ) are as follows: 500 mM potassium acetate, 100 mM Tris-HCl (pH 8.0), 10 mM magnesium acetate, 0.25% NP-40.
- buffer B is adenosine triphosphate or chemically modified adenosine triphosphate.
- the components of buffer B (5 ⁇ ) are as follows: 50% glycerol, 125 mM Tris-HCl (pH 7.0), 0.5 mg/mL BSA, 3.5 mM manganese chloride, 250 mM potassium chloride, 0.05 mM EDTA, 2.5 mM dithiothreitol.
- agarose gel electrophoresis detection conditions 2% agarose, 5 V/cm, electrophoresis for 30 minutes.
- Figure 75 is a diagram showing the effect of polyadenylation modification of eGFP mRNA by E. coli polyA polymerase.
- lanes 1-13 are: lane 1: eGFP mRNA template; lane 2: natural adenosine tailing product of eGFP mRNA; lane 3: 2'-methoxyadenosine tailing product of eGFP mRNA; lane 4: 2'-fluoroadenosine tailing product of eGFP mRNA; lane 5: N6-methyladenosine tailing product of eGFP mRNA; lane 6: N6-acetyladenosine tailing product of eGFP mRNA.
- Lane 7 N6-propionyladenosine tailing products of eGFP mRNA
- Lane 8 N6-butyryladenosine tailing products of eGFP mRNA
- Lane 9 N6-pentanoyladenosine tailing products of eGFP mRNA
- Lane 10 N6-hexanoyladenosine tailing products of eGFP mRNA
- Lane 11 N6-benzoyladenosine tailing products of eGFP mRNA
- Lane 12 2-aminoadenosine tailing products of eGFP mRNA
- Lane 13 7-deazaadenosine tailing products of eGFP mRNA.
- Figure 76 is a diagram showing the effect of polyadenylation modification of eGFP mRNA by Yeast polyA polymerase.
- lanes 1-13 are respectively: lane 1: eGFP mRNA template; lane 2: natural adenosine tailing product of eGFP mRNA; lane 3: 2'-methoxyadenosine tailing product of eGFP mRNA; lane 4: 2'-fluoroadenosine tailing product of eGFP mRNA; lane 5: N6-methyladenosine tailing product of eGFP mRNA; lane 6: N6-acetyladenosine tailing product of eGFP mRNA.
- Lane 10 N6-hexanoyladenosine tailing product of eGFP mRNA
- Lane 11 N6-benzoyladenosine tailing product of eGFP mRNA
- Lane 12 2-aminoadenosine tailing product of eGFP mRNA
- Lane 13 7-deazaadenosine tailing product of eGFP mRNA.
- Example 43 Translation of luciferase mRNA in which chemically modified adenosine globally replaces natural adenosine or luciferase mRNA in which chemically modified adenosine and pseudouridine are combined in rabbit reticulocyte lysate system
- the rabbit reticulocyte lysate system is shown in Table 13.
- thermostat cover Turn on the thermostat cover and set it to 60°C. Click the thermostat reaction system, 30°C, 90min. The incubated product is directly used for the luminescence experiment of luciferin.
- the experimental steps for detecting luminescence by microplate reader are as follows: 2.5 ⁇ L of the incubation product in Example 43 was added to a 96-well plate, and then incubated at room temperature for 10 minutes, followed by adding 50 ⁇ L of luciferase detection reagent (Promega, E1483), pipetting evenly to remove bubbles in the system, and completing the luminescence intensity detection on the microplate reader within 5 minutes. Two replicate wells were set for each sample.
- Figure 77 is a graph showing the expression of luciferase mRNA in combination with chemically modified adenosine and pseudouridine after incubation in rabbit reticulocyte lysate for 90 minutes.
- Figures 1-6 respectively represent: 1: expression effect graph of unmodified luciferase mRNA; 2: expression effect graph of luciferase mRNA replaced with pseudouridine; 3: expression effect graph of luciferase mRNA replaced with N6-acetyladenosine and pseudouridine; 4: expression effect graph of luciferase mRNA replaced with N6-propionyladenosine and pseudouridine; 5: expression effect graph of luciferase mRNA replaced with N6-butyryladenosine and pseudouridine; 6: expression effect graph of luciferase mRNA replaced with 2-aminoadenosine and pseudouridine.
- FIG78 is a graph showing the expression effect of luciferase mRNA in which chemically modified adenosine globally replaces natural adenosine after incubation in rabbit reticulocyte lysate for 90 minutes.
- 1-10 in the figure respectively represent, 1: the expression effect diagram of unmodified luciferase mRNA; 2: the expression effect diagram of luciferase mRNA replaced by N6-methyladenosine; 3: the expression effect diagram of luciferase mRNA replaced by N6-acetyladenosine; 4: the expression effect diagram of luciferase mRNA replaced by N6-propionyladenosine; 5: the expression effect diagram of luciferase mRNA replaced by N6-butyryladenosine; 6: the expression effect diagram of luciferase mRNA replaced by N6-pentanoyladenosine; 7: the expression effect diagram of luciferase mRNA replaced by N
- luciferase mRNA in which chemically modified adenosine globally replaced natural adenosine had higher expression efficiency in rabbit reticulocyte lysate than unmodified mRNA, where the modification types included N6-acetyl modification, N6-propionyl modification, N6-butyryl modification and N6-benzoyl modification.
- the experimental steps for extracting the target protein luciferase from cells are as follows: cell culture and mRNA transfection are performed in a 24-well plate. 24 hours after the luciferase mRNA transfection cells, the 24-well plate is placed on ice, the culture medium is removed, and the plate is washed twice with cold PBS solution, and then 200 ⁇ L RIPA lysis buffer (Yisheng, catalog number 20115ES60, 1 mM PMSF is added before use). The cells were fully lysed by blowing with a pipette for 5 minutes, and the cell lysate was collected and transferred to a 1.5 mL EP tube. The tube was centrifuged at 15,000 rpm for 10 minutes at 4°C, and 150 ⁇ L of the supernatant was taken for subsequent detection.
- the experimental steps of detecting luminescence by an ELISA instrument are as follows: The experimental steps of detecting luminescence by an ELISA instrument are as in Example 44, except that 50 ⁇ L of the protein solution extracted in this example is added to the 96-well plate, and then 50 ⁇ L of luciferase detection reagent (Promega, E1483) is added for detection. Two replicate wells are set for each sample.
- Figure 79 shows the expression effect of luciferase mRNA in which chemically modified adenosine globally replaces natural adenosine 24 hours after transfection into HEK 293T cells.
- 1-9 respectively represent, 1: expression effect of unmodified luciferase mRNA; 2: expression effect of luciferase mRNA replaced by N6-methyladenosine; 3: expression effect of luciferase mRNA replaced by N6-acetyladenosine; 4: expression effect of luciferase mRNA replaced by N6-propionyladenosine; 5: expression effect of luciferase mRNA replaced by N6-butyryladenosine; 6: expression effect of luciferase mRNA replaced by N6-pentanoyladenosine; 7: expression effect of luciferase mRNA replaced by N6-hexanoyladenosine; 8: expression effect of luciferase
- Figure 80 is a graph showing the expression of chemically modified adenosine and pseudouridine combined luciferase mRNA 24h after transfection into HeLa cells.
- 1-6 respectively represent, 1: expression effect graph of unmodified luciferase mRNA; 2: expression effect graph of luciferase mRNA replaced by pseudouridine; 3: expression effect graph of luciferase mRNA replaced by N6-acetyladenosine and pseudouridine; 4: expression effect graph of luciferase mRNA replaced by N6-propionyladenosine and pseudouridine; 5: expression effect graph of luciferase mRNA replaced by N6-butyryladenosine and pseudouridine; 6: expression effect graph of luciferase mRNA replaced by 2-aminoadenosine and pseudouridine.
- Example 46 Expression efficiency of eGFP mRNA containing a chemically modified polyadenosine tail in cells
- eGFP mRNA containing a chemically modified polyadenosine tail was synthesized and its expression efficiency in cells was tested. After being transfected into cells, it was translated into the target protein enhanced green fluorescent protein, and its fluorescence intensity was detected using a laser confocal fluorescence microscope to determine the translation efficiency of eGFP mRNA containing natural and chemically modified polyadenosine tails.
- Opti-MEM Take 50 ⁇ L Opti-MEM, add 800ng mRNA, and mix well. Also prepare a mixed solution of 2 ⁇ L Lipofectamine 2000 (liposome 2000) and 50 ⁇ L Opti-MEM, and incubate at room temperature for 10 minutes. Then add the liposome 2000 mixture to the mRNA mixture, mix well, and incubate for 20 minutes. Add the mixed solution to the well plate. Change the medium 6h after transfection, aspirate the old culture medium, and replace each well with 3mL of fresh culture medium, that is, 90% (volume percentage) DMEM + 10% (volume percentage) FBS culture medium. Replace the fresh culture medium 24h/48h after transfection, and detect the fluorescence intensity of the cells with a laser confocal fluorescence microscope.
- Figure 81 is a fluorescence imaging diagram of the expression effect of chemically modified adenosine polyadenylation-modified eGFP mRNA in HEK 293T cells.
- 1 represents the fluorescence imaging diagram of the expression effect of eGFP mRNA without poly (A) tail
- 2 represents the fluorescence imaging diagram of the expression effect of eGFP mRNA with poly natural adenosine tail
- 3 represents the fluorescence imaging diagram of the expression effect of eGFP mRNA with poly N6-acetyl adenosine tail
- 4 represents the fluorescence imaging diagram of the expression effect of eGFP mRNA with poly N6-propionyl adenosine tail.
- the scale bar in the figure is 50 ⁇ m.
- the eGFP mRNA with a poly-N6-acetyladenosine tail has a higher fluorescence intensity after being transfected into HEK 293T cells, and the protein expression efficiency exceeds that of the mRNA with a poly-natural adenosine tail.
- the present application provides a chemically modified nucleoside and a chemically modified nucleoside triphosphate, which provide supplements and new options for mRNA modification technology.
- the chemically modified cytosine triphosphate is used to globally replace the natural cytosine triphosphate to obtain a chemically modified mRNA, which has a higher translation efficiency in both the cell-free system and the cell, and is more stable.
- the natural adenine nucleosides in the mRNA are globally replaced with the chemically modified adenine triphosphate, or the mRNA is polyadenylated and tailed, which can enhance the translation efficiency and stability of the target mRNA, reduce immunogenicity, and provide supplements and new options for mRNA modification technology.
Landscapes
- Chemical & Material Sciences (AREA)
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Organic Chemistry (AREA)
- Molecular Biology (AREA)
- Engineering & Computer Science (AREA)
- Genetics & Genomics (AREA)
- Biochemistry (AREA)
- General Health & Medical Sciences (AREA)
- Biotechnology (AREA)
- Biomedical Technology (AREA)
- Pharmacology & Pharmacy (AREA)
- Animal Behavior & Ethology (AREA)
- Wood Science & Technology (AREA)
- Zoology (AREA)
- Veterinary Medicine (AREA)
- Public Health (AREA)
- Bioinformatics & Cheminformatics (AREA)
- General Engineering & Computer Science (AREA)
- Virology (AREA)
- Medicinal Chemistry (AREA)
- Oncology (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Communicable Diseases (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Plant Pathology (AREA)
- Microbiology (AREA)
- Physics & Mathematics (AREA)
- Biophysics (AREA)
- Epidemiology (AREA)
- Saccharide Compounds (AREA)
Abstract
Description
本申请属于核酸修饰技术领域,具体涉及一种化学修饰核苷、化学修饰核苷三磷酸及其应用。The present application belongs to the field of nucleic acid modification technology, and specifically relates to a chemically modified nucleoside, a chemically modified nucleoside triphosphate and applications thereof.
mRNA又称信使RNA,由DNA为模板转录而来,负责指导胞内蛋白质的合成。mRNA技术就是利用这一规则,将体外人工合成的mRNA导入到特定细胞,让细胞成为蛋白药物生产的小型工厂,通过这些细胞生产的蛋白质发挥治疗作用。mRNA技术在传染病防控、肿瘤疫苗、肿瘤免疫治疗、蛋白替代疗法等方面均有巨大应用潜力,但是天然mRNA稳定性差且易引发免疫原性,严重阻碍了mRNA的应用。主流使用的mRNA技术中,是利用将天然存在的修饰核苷替换天然核苷,例如,利用假尿苷和甲基假尿苷替换尿苷,以解决mRNA药物的稳定性和免疫原性的问题。然而,天然发生的修饰核苷种类稀少,无法满足改善mRNA性质的需求,也不利于建立构效关系从分子层面揭示其作用机理。因此,利用有机合成的方法制备更多的化学修饰核苷,从而从不同层面改善mRNA的成药性,具有重大的研究和应用意义。mRNA, also known as messenger RNA, is transcribed from DNA as a template and is responsible for guiding the synthesis of intracellular proteins. mRNA technology uses this rule to introduce in vitro artificially synthesized mRNA into specific cells, making cells into small factories for the production of protein drugs, and exerting therapeutic effects through the proteins produced by these cells. mRNA technology has great application potential in infectious disease prevention and control, tumor vaccines, tumor immunotherapy, protein replacement therapy, etc., but natural mRNA has poor stability and is prone to immunogenicity, which seriously hinders the application of mRNA. In the mainstream mRNA technology, naturally occurring modified nucleosides are used to replace natural nucleosides, for example, pseudouridine and methyl pseudouridine are used to replace uridine to solve the problems of stability and immunogenicity of mRNA drugs. However, the number of naturally occurring modified nucleosides is scarce, which cannot meet the needs of improving the properties of mRNA, nor is it conducive to establishing structure-activity relationships to reveal its mechanism of action at the molecular level. Therefore, it is of great research and application significance to prepare more chemically modified nucleosides using organic synthesis methods to improve the drugability of mRNA from different levels.
研究发现,真核生物mRNA结构中不同区域的修饰,对mRNA的翻译活性和免疫原性等有着截然不同的影响。因此,除了mRNA整体修饰策略之外,对于mRNA特定区域进行化学修饰的研究十分有必要。例如对3’尾结构进行修饰,可以实现对mRNA进行单一修饰而不影响到mRNA其他区域。目前对mRNA加尾的研究还主要集中在利用天然的腺苷对mRNA的poly A尾结构进行长度调节,而将化学改造的非天然腺苷引入poly A尾(聚A尾)结构,同时控制其长度,在该方面的研究还较为稀少。由于poly A尾结构也是翻译启动的重要控制因子,并且它还可以调节mRNA的稳定性和周转率,进而影响mRNA的翻译效率,因此,开发一种采用新型化学修饰核苷酸,并对mRNA的poly A尾结构进行修饰,在mRNA的相关应用方面具有十分重要的意义和广阔的应用前景。Studies have found that modifications in different regions of the eukaryotic mRNA structure have completely different effects on the translation activity and immunogenicity of mRNA. Therefore, in addition to the overall modification strategy of mRNA, it is very necessary to study the chemical modification of specific regions of mRNA. For example, by modifying the 3' tail structure, a single modification of mRNA can be achieved without affecting other regions of mRNA. At present, the research on mRNA tailing is still mainly focused on using natural adenosine to adjust the length of the poly A tail structure of mRNA, while the introduction of chemically modified non-natural adenosine into the poly A tail (poly A tail) structure and the control of its length at the same time are relatively scarce. Since the poly A tail structure is also an important control factor for translation initiation, and it can also regulate the stability and turnover rate of mRNA, thereby affecting the translation efficiency of mRNA, therefore, the development of a method using new chemically modified nucleotides and modifying the poly A tail structure of mRNA is of great significance and has broad application prospects in the relevant applications of mRNA.
现有专利使用的修饰mRNA方法主要利用自然界存在的修饰核苷来替换常规核苷,比如利用假尿苷和甲基假尿苷替换尿苷。CN107090436B公开了一种降低通过体外转录获得的体外合成的RNA分子或包含所述RNA分子的基因治疗载体的制剂对哺乳动物细胞的免疫原性的非治疗方法或体外方法,所述RNA分子中包含的替代尿苷选自假尿苷、1-甲基假尿苷、5-甲基尿苷、5-甲氧基尿苷和2-硫尿苷的修饰核苷。天然存在的修饰碱基的种类是十分有限的,因此可用于改善mRNA性质的手段有限。The mRNA modification methods used in existing patents mainly use modified nucleosides existing in nature to replace conventional nucleosides, such as using pseudouridine and methyl pseudouridine to replace uridine. CN107090436B discloses a non-therapeutic method or in vitro method for reducing the immunogenicity of an in vitro synthesized RNA molecule obtained by in vitro transcription or a preparation of a gene therapy vector containing the RNA molecule to mammalian cells, wherein the replacement uridine contained in the RNA molecule is selected from a modified nucleoside of pseudouridine, 1-methyl pseudouridine, 5-methyluridine, 5-methoxyuridine and 2-thiouridine. The types of naturally occurring modified bases are very limited, so the means available for improving the properties of mRNA are limited.
利用化学手段合成非天然修饰的核苷,可以极大丰富非天然核苷的种类和范围,也适合建立构效关系,这是天然存在的修饰碱基所不具备的。The use of chemical means to synthesize non-natural modified nucleosides can greatly enrich the types and range of non-natural nucleosides and is also suitable for establishing structure-activity relationships, which is not available with naturally occurring modified bases.
CN112673106A公开了一种点击修饰的mRNA,该发明涉及炔烃和/或叠氮化物修饰的mRNA,通过引入炔烃和/或叠氮化物-修饰的核苷酸来稳定RNA,这种修饰不仅能够稳定目标mRNA以用于离体应用和随后向人患者、动物或植物施用,而且能够容易的附接可检测的标记物或官能基团。CN114807154A公开了一种修饰的核酸及其应用,所述修饰的核酸,包括尿嘧啶核苷、胞嘧啶核苷、腺嘌呤核苷、鸟嘌呤核苷和化学修饰的核苷;所述化学修饰的 核苷包括化学修饰的尿嘧啶核苷、化学修饰的胞嘧啶核苷、化学修饰的腺嘌呤核苷和化学修饰的鸟嘌呤核苷中的一种或几种;该申请所述修饰的核酸稳定性高、免疫原性低、体内半衰期长。但是目前的修饰核酸的类型仍然较少,因此,需要开发新的具有新型结构的非天然修饰核苷酸,并将其引入到mRNA中,从而提高mRNA的稳定性,增强其翻译活性,达到替代、丰富或者补充现有mRNA化学修饰的目标,从而为mRNA新型化学修饰和应用提供助力。CN112673106A discloses a click-modified mRNA, which relates to alkyne and/or azide-modified mRNA, and stabilizes RNA by introducing alkyne and/or azide-modified nucleotides. This modification can not only stabilize the target mRNA for in vitro application and subsequent administration to human patients, animals or plants, but also can easily attach detectable markers or functional groups. CN114807154A discloses a modified nucleic acid and its application, the modified nucleic acid includes uridine, cytosine, adenine, guanosine and chemically modified nucleosides; the chemically modified Nucleosides include one or more of chemically modified uridine nucleosides, chemically modified cytosine nucleosides, chemically modified adenine nucleosides, and chemically modified guanine nucleosides; the modified nucleic acids described in this application have high stability, low immunogenicity, and long in vivo half-life. However, there are still relatively few types of modified nucleic acids. Therefore, it is necessary to develop new non-natural modified nucleotides with novel structures and introduce them into mRNA to improve the stability of mRNA and enhance its translation activity, so as to achieve the goal of replacing, enriching, or supplementing existing mRNA chemical modifications, thereby providing support for new chemical modifications and applications of mRNA.
发明内容Summary of the invention
本申请提供了一种化学修饰核苷、化学修饰核苷三磷酸及其应用。本申请利用化学合成手段,对天然的核苷进行化学修饰,其中,化学修饰的核苷包括腺嘌呤核苷和胞嘧啶核苷;将化学修饰核苷三磷酸化,得到化学修饰腺嘌呤核苷核苷三磷酸、化学修饰胞嘧啶核苷三磷酸,采用上述化学修饰的核苷三磷酸全局替换天然核苷三磷酸,得到修饰的mRNA,上述修饰的mRNA在体外水平和细胞内均表现出增强的翻译效率以及更高的稳定性。The present application provides a chemically modified nucleoside, a chemically modified nucleoside triphosphate and an application thereof. The present application uses chemical synthesis means to chemically modify natural nucleosides, wherein the chemically modified nucleosides include adenine nucleosides and cytosine nucleosides; the chemically modified nucleosides are triphosphorylated to obtain chemically modified adenine nucleoside triphosphates and chemically modified cytosine nucleoside triphosphates, and the chemically modified nucleoside triphosphates are used to globally replace the natural nucleoside triphosphates to obtain modified mRNA, and the modified mRNA exhibits enhanced translation efficiency and higher stability both in vitro and in cells.
第一方面,本申请提供一种化学修饰核苷,所述化学修饰核苷选自具有如式1所示结构的化合物、其盐或其异构体:
In a first aspect, the present application provides a chemically modified nucleoside, wherein the chemically modified nucleoside is selected from a compound having a structure as shown in Formula 1, a salt thereof, or an isomer thereof:
其中,R3选自羟基、氢、氟、甲氧基、乙氧基或甲氧基甲基醚基;Wherein, R 3 is selected from hydroxyl, hydrogen, fluorine, methoxy, ethoxy or methoxymethyl ether;
Y选自含有取代基的胞嘧啶环或含有取代基的腺嘌呤环,所述含有取代基的胞嘧啶环如式2所示,所述含有取代基的腺嘌呤环如式3所示,式2和式3中表示连接位置;
Y is selected from a cytosine ring containing a substituent or an adenine ring containing a substituent, wherein the cytosine ring containing a substituent is as shown in Formula 2, and the adenine ring containing a substituent is as shown in Formula 3. Indicates the connection location;
其中,R1和R1’各自独立地选自氢、乙酰基、卤素取代乙酰基、丙酰基、卤素取代丙酰基、丁酰基、卤素取代丁酰基、戊酰基、卤素取代戊酰基、己酰基、卤素取代己酰基、辛酰基、癸酰基、苯甲酰基、4-甲氧基苯甲酰基或4-硝基苯甲酰基;wherein R 1 and R 1′ are each independently selected from hydrogen, acetyl, halogen-substituted acetyl, propionyl, halogen-substituted propionyl, butyryl, halogen-substituted butyryl, valeryl, halogen-substituted valeryl, hexanoyl, halogen-substituted hexanoyl, octanoyl, decanoyl, benzoyl, 4-methoxybenzoyl or 4-nitrobenzoyl;
R2和R2’各自独立地选自氢、甲基、甲氧基、氟代、氯代、溴代、碘代或腈基;以及R 2 and R 2' are each independently selected from hydrogen, methyl, methoxy, fluoro, chloro, bromo, iodo or nitrile; and
X选自氮或碳。X is selected from nitrogen or carbon.
优选地,所述化学修饰核苷为化学修饰胞嘧啶核苷,所述化学修饰胞嘧啶核苷选自具有如式4所示结构的化合物、其盐或其异构体:
Preferably, the chemically modified nucleoside is a chemically modified cytosine nucleoside, and the chemically modified cytosine nucleoside is selected from a compound having a structure as shown in Formula 4, a salt thereof, or an isomer thereof:
其中,R1选自氢、乙酰基、卤素取代乙酰基、丙酰基、卤素取代丙酰基、丁酰基、卤素取代丁酰基、戊酰基、卤素取代戊酰基、己酰基、卤素取代己酰基、辛酰基、癸酰基、苯甲酰基、4-甲氧基苯甲酰基或4-硝基苯甲酰基;wherein R 1 is selected from hydrogen, acetyl, halogen-substituted acetyl, propionyl, halogen-substituted propionyl, butyryl, halogen-substituted butyryl, valeryl, halogen-substituted valeryl, hexanoyl, halogen-substituted hexanoyl, octanoyl, decanoyl, benzoyl, 4-methoxybenzoyl or 4-nitrobenzoyl;
R2选自氢、甲基、甲氧基、氟代、氯代、溴代、碘代或腈基;以及 R2 is selected from hydrogen, methyl, methoxy, fluoro, chloro, bromo, iodo or nitrile; and
R3选自羟基、氢、氟、甲氧基、乙氧基或甲氧基甲基醚基。 R3 is selected from hydroxy, hydrogen, fluorine, methoxy, ethoxy or methoxymethyl ether.
本申请中,所述化学修饰胞嘧啶核苷选自:N4-丙酰基胞苷、N4-丁酰基胞苷、N4-戊酰基胞苷、N4-己酰基胞苷、N4-辛酰基胞苷、N4-癸酰基胞苷、N4-对硝基苯甲酰基胞苷、5-甲基-N4-丁酰基胞苷、5-甲基-N4-己酰基胞苷、5-甲基-N4-苯甲酰基胞苷、2’-氟-N4-乙酰基胞苷、2’-氟-N4-丁酰基胞苷、2’-氟-N4-己酰基胞苷、2’-甲氧基胞苷或2’-乙氧基胞苷。In the present application, the chemically modified cytosine nucleoside is selected from: N4-propionyl cytidine, N4-butyryl cytidine, N4-pentanoyl cytidine, N4-hexanoyl cytidine, N4-octanoyl cytidine, N4-decanoyl cytidine, N4-p-nitrobenzoyl cytidine, 5-methyl-N4-butyryl cytidine, 5-methyl-N4-hexanoyl cytidine, 5-methyl-N4-benzoyl cytidine, 2'-fluoro-N4-acetyl cytidine, 2'-fluoro-N4-butyryl cytidine, 2'-fluoro-N4-hexanoyl cytidine, 2'-methoxy cytidine or 2'-ethoxy cytidine.
优选地,所述化学修饰核苷为化学修饰腺嘌呤核苷,所述化学修饰腺嘌呤核苷选自具有如式5所示结构的化合物、其盐或其异构体:
Preferably, the chemically modified nucleoside is a chemically modified adenine nucleoside, and the chemically modified adenine nucleoside is selected from a compound having a structure as shown in Formula 5, a salt thereof, or an isomer thereof:
其中,R1’选自乙酰基、丙酰基、丁酰基、戊酰基、己酰基或苯甲酰基;wherein R 1′ is selected from acetyl, propionyl, butyryl, valeryl, hexanoyl or benzoyl;
R2’为氢;R 2' is hydrogen;
R3选自氢、羟基、氟或甲氧基;以及 R3 is selected from hydrogen, hydroxy, fluoro or methoxy; and
X选自氮或碳。X is selected from nitrogen or carbon.
本申请中,利用化学手段合成非天然修饰的核苷,可以极大丰富非天然核苷的种类和范围,也利于建立构效关系,还可对mRNA进行区域修饰,这些都是当前利用天然存在的修饰碱基来改善mRNA所不具备的。In the present application, the use of chemical means to synthesize non-natural modified nucleosides can greatly enrich the types and ranges of non-natural nucleosides, is also conducive to the establishment of structure-activity relationships, and can also perform regional modifications on mRNA, which are all not available in the current use of naturally occurring modified bases to improve mRNA.
第二方面,本申请提供一种化学修饰核苷三磷酸,所述化学修饰核苷三磷酸选自具有如式6所示结构的化合物、其盐或其异构体:
In a second aspect, the present application provides a chemically modified nucleoside triphosphate, wherein the chemically modified nucleoside triphosphate is selected from a compound having a structure as shown in Formula 6, a salt thereof, or an isomer thereof:
其中,Rn选自羟基、氢、氟、甲氧基、乙氧基或甲氧基甲基醚基;wherein Rn is selected from hydroxy, hydrogen, fluorine, methoxy, ethoxy or methoxymethyl ether;
W选自含有取代基的胞嘧啶环或含有取代基的腺嘌呤环,所述含有取代基的胞嘧啶环如式7所示,所述含有取代基的腺嘌呤环如式8所示,式7和式8中表示连接位置;
W is selected from a cytosine ring containing a substituent or an adenine ring containing a substituent, wherein the cytosine ring containing a substituent is as shown in Formula 7, and the adenine ring containing a substituent is as shown in Formula 8. Indicates the connection location;
其中,R4和R4’各自独立地选自氢、乙酰基、卤素取代乙酰基、丙酰基、卤素取代丙酰基、丁酰基、卤素取代丁酰基、戊酰基、卤素取代戊酰基、己酰基、卤素取代己酰基、辛酰基、癸酰基、苯甲酰基、4-甲氧基苯甲酰基或4-硝基苯甲酰基;wherein R 4 and R 4′ are each independently selected from hydrogen, acetyl, halogen-substituted acetyl, propionyl, halogen-substituted propionyl, butyryl, halogen-substituted butyryl, valeryl, halogen-substituted valeryl, hexanoyl, halogen-substituted hexanoyl, octanoyl, decanoyl, benzoyl, 4-methoxybenzoyl or 4-nitrobenzoyl;
R5和R5’各自独立地选自氢、甲基、甲氧基、氟代、氯代、溴代、碘代或腈基;以及R 5 and R 5' are each independently selected from hydrogen, methyl, methoxy, fluoro, chloro, bromo, iodo or nitrile; and
M选自氮或碳。M is selected from nitrogen or carbon.
优选地,所述化学修饰核苷三磷酸为化学修饰胞嘧啶核苷三磷酸,所述化学修饰胞嘧啶核苷三磷酸选自具有如式9所示结构的化合物、其盐或其异构体:
Preferably, the chemically modified nucleoside triphosphate is a chemically modified cytosine triphosphate, and the chemically modified cytosine triphosphate is selected from a compound having a structure as shown in Formula 9, a salt thereof, or an isomer thereof:
其中,R4选自氢、乙酰基、卤素取代乙酰基、丙酰基、卤素取代丙酰基、丁酰基、卤素取代丁酰基、戊酰基、卤素取代戊酰基、己酰基、卤素取代己酰基、辛酰基、癸酰基、苯甲酰基、4-甲氧基苯甲酰基或4-硝基苯甲酰基;wherein R4 is selected from hydrogen, acetyl, halogen-substituted acetyl, propionyl, halogen-substituted propionyl, butyryl, halogen-substituted butyryl, valeryl, halogen-substituted valeryl, hexanoyl, halogen-substituted hexanoyl, octanoyl, decanoyl, benzoyl, 4-methoxybenzoyl or 4-nitrobenzoyl;
R5选自氢、甲基、甲氧基、氟代、氯代、溴代、碘代、腈基;以及R 5 is selected from hydrogen, methyl, methoxy, fluoro, chloro, bromo, iodo, nitrile; and
Rn选自羟基、氢、氟、甲氧基、乙氧基或甲氧基甲基醚基。R n is selected from hydroxy, hydrogen, fluorine, methoxy, ethoxy or methoxymethyl ether.
本申请中,所述化学修饰胞嘧啶核苷三磷酸选自:N4-乙酰基胞苷三磷酸、N4-丙酰基胞苷三磷酸、N4-丁酰基胞苷三磷酸、N4-戊酰基胞苷三磷酸、N4-己酰基胞苷三磷酸、N4-辛酰 基胞苷三磷酸、N4-癸酰基胞苷三磷酸、N4-苯甲酰基胞苷三磷酸、N4-对甲氧基苯甲酰基胞苷三磷酸、N4-对硝基苯甲酰基胞苷三磷酸、5-甲基胞苷三磷酸、5-甲基-N4-乙酰基胞苷三磷酸、5-甲基-N4-丁酰基胞苷三磷酸、5-甲基-N4-己酰基胞苷三磷酸、5-甲基-N4-苯甲酰基胞苷三磷酸、2’-氟-N4-乙酰基胞苷三磷酸、2’-氟-N4-丁酰基胞苷三磷酸、2’-氟-N4-己酰基胞苷三磷酸、2’-甲氧基胞苷三磷酸、2’-乙氧基胞苷三磷酸。In the present application, the chemically modified cytosine triphosphate is selected from: N4-acetyl cytidine triphosphate, N4-propionyl cytidine triphosphate, N4-butyryl cytidine triphosphate, N4-pentanoyl cytidine triphosphate, N4-hexanoyl cytidine triphosphate, N4-octanoyl cytidine triphosphate, cytidine triphosphate, 5-methyl-N4-acetyl cytidine triphosphate, 5-methyl-N4-butyryl cytidine triphosphate, 5-methyl-N4-hexanoyl cytidine triphosphate, 5-methyl-N4-benzoyl cytidine triphosphate, 2'-fluoro-N4-acetyl cytidine triphosphate, 2'-fluoro-N4-butyryl cytidine triphosphate, 2'-fluoro-N4-hexanoyl cytidine triphosphate, 2'-methoxy cytidine triphosphate, 2'-ethoxy cytidine triphosphate.
本申请提供了全新的化学修饰胞嘧啶核苷三磷酸,为mRNA提供了更多可用的胞嘧啶核苷三磷酸,本申请中合成的化学修饰的胞嘧啶核苷三磷酸均可以用于mRNA的合成,可以替代天然胞嘧啶核苷三磷酸,单独或联合其他修饰核苷酸,并且能够发挥相应的翻译功能,能够增强目标mRNA的翻译效率和稳定性,降低免疫原性,为mRNA技术在目前的专利技术之外提供补充和新选择。The present application provides a new chemically modified cytosine triphosphate, which provides more available cytosine triphosphates for mRNA. The chemically modified cytosine triphosphates synthesized in the present application can be used for the synthesis of mRNA, can replace natural cytosine triphosphate, alone or in combination with other modified nucleotides, and can play a corresponding translation function, can enhance the translation efficiency and stability of the target mRNA, and reduce immunogenicity, providing a supplement and new choice for mRNA technology beyond the current patented technology.
优选地,所述化学修饰核苷三磷酸为化学修饰腺嘌呤核苷三磷酸,所述化学修饰腺嘌呤核苷三磷酸选自具有如式10所示结构的化合物、其盐或其异构体:
Preferably, the chemically modified nucleoside triphosphate is a chemically modified adenine nucleoside triphosphate, and the chemically modified adenine nucleoside triphosphate is selected from a compound having a structure as shown in Formula 10, a salt thereof, or an isomer thereof:
其中,R4’选自乙酰基、丙酰基、丁酰基、戊酰基、己酰基或苯甲酰基;wherein R 4′ is selected from acetyl, propionyl, butyryl, valeryl, hexanoyl or benzoyl;
R5’选自氢;R 5' is selected from hydrogen;
Rn选自氢、羟基、氟或甲氧基;以及 Rn is selected from hydrogen, hydroxy, fluoro or methoxy; and
M选自氮或碳。M is selected from nitrogen or carbon.
本申请提供了一种新的化学修饰腺嘌呤核苷和化学修饰腺嘌呤核苷三磷酸,为mRNA技术提供更多可选的原料,以本申请中全新的化学修饰的腺嘌呤核苷三磷酸整体替换mRNA中的天然腺嘌呤核苷三磷酸,或者以化学修饰的腺嘌呤核苷三磷酸对体外转录制备的mRNA进行多聚腺苷化修饰,能够增强目标mRNA的稳定性和翻译效率。The present application provides a new chemically modified adenine nucleoside and a chemically modified adenine nucleoside triphosphate, which provide more optional raw materials for mRNA technology. The natural adenine nucleoside triphosphate in mRNA is completely replaced by the new chemically modified adenine nucleoside triphosphate in the present application, or the mRNA prepared by in vitro transcription is polyadenylated with the chemically modified adenine nucleoside triphosphate, which can enhance the stability and translation efficiency of the target mRNA.
优选地,所述化学修饰核苷三磷酸由如下制备方法制得:
Preferably, the chemically modified nucleoside triphosphate is prepared by the following preparation method:
(1)将式11化合物的羟基进行羟基保护反应,得到式12化合物;(1) subjecting the hydroxyl group of the compound of formula 11 to a hydroxyl protection reaction to obtain a compound of formula 12;
(2)将式12化合物与酸酐或酰氯反应,得到式13化合物;(2) reacting the compound of formula 12 with an acid anhydride or an acid chloride to obtain a compound of formula 13;
(3)将式13化合物的保护基进行完全脱保护反应,得到式14化合物;(3) performing a complete deprotection reaction on the protecting group of the compound of formula 13 to obtain a compound of formula 14;
(4)将式14化合物与氯代试剂反应,得到式15化合物;(4) reacting the compound of formula 14 with a chlorinating agent to obtain a compound of formula 15;
(5)将式15化合物与三丁基焦磷酸铵反应,得到式16化合物;(5) reacting the compound of formula 15 with tributylammonium pyrophosphate to obtain a compound of formula 16;
(6)将式16化合物进行淬灭反应,得到式9化合物。(6) subjecting the compound of formula 16 to a quenching reaction to obtain the compound of formula 9.
优选地,步骤(1)中,采用叔丁基二甲基氯硅烷进行羟基保护反应。Preferably, in step (1), tert-butyldimethylsilyl chloride is used to carry out the hydroxyl protection reaction.
优选地,步骤(2)中,所述酸酐选自乙酸酐、丙酸酐、丁酸酐、戊酸酐、己酸酐、辛酸酐、癸酸酐或苯甲酸酐。Preferably, in step (2), the acid anhydride is selected from acetic anhydride, propionic anhydride, butyric anhydride, valeric anhydride, caproic anhydride, caprylic anhydride, capric anhydride or benzoic anhydride.
优选地,步骤(2)中,所述酰氯选自4-甲氧基苯甲酰氯或4-硝基苯甲酰氯。Preferably, in step (2), the acyl chloride is selected from 4-methoxybenzoyl chloride or 4-nitrobenzoyl chloride.
优选地,步骤(3)中,采用四丁基氟化铵进行完全脱保护反应。Preferably, in step (3), tetrabutylammonium fluoride is used to carry out a complete deprotection reaction.
优选地,步骤(4)中,所述氯代试剂选自三氯氧磷。Preferably, in step (4), the chlorination agent is selected from phosphorus oxychloride.
优选地,所述化学修饰核苷三磷酸由如下制备方法制得:
Preferably, the chemically modified nucleoside triphosphate is prepared by the following preparation method:
(1”)将式17化合物的羟基进行羟基保护反应,得到式18化合物;(1") subjecting the hydroxyl group of the compound of formula 17 to a hydroxyl protection reaction to obtain a compound of formula 18;
(2”)将式18化合物与酸酐或酰氯反应,得到式19化合物;(2") reacting the compound of formula 18 with an acid anhydride or an acid chloride to obtain a compound of formula 19;
(3”)将式19化合物的保护基进行完全脱保护反应,得到式20化合物;(3") performing a complete deprotection reaction on the protecting group of the compound of formula 19 to obtain a compound of formula 20;
(4”)将式20化合物与氯代试剂反应,得到式21化合物;(4") reacting the compound of formula 20 with a chlorinating agent to obtain a compound of formula 21;
(5”)将式21化合物与三丁基焦磷酸铵反应,得到式22化合物;(5") reacting the compound of formula 21 with tributylammonium pyrophosphate to obtain a compound of formula 22;
(6”)将式22化合物进行淬灭反应,得到式23化合物。(6") The compound of formula 22 is subjected to a quenching reaction to obtain a compound of formula 23.
优选地,步骤(1”)中,采用叔丁基二甲基氯硅烷进行羟基保护反应。Preferably, in step (1"), tert-butyldimethylsilyl chloride is used to carry out the hydroxyl protection reaction.
优选地,步骤(2”)中,所述酸酐选自乙酸酐、丙酸酐、丁酸酐、戊酸酐、己酸酐或苯甲酸酐。Preferably, in step (2"), the acid anhydride is selected from acetic anhydride, propionic anhydride, butyric anhydride, valeric anhydride, caproic anhydride or benzoic anhydride.
优选地,步骤(2”)中,所述酰氯选自乙酰氯、丙酰氯、丁酰氯、戊酰氯、己酰氯或苯甲酰氯。Preferably, in step (2"), the acyl chloride is selected from acetyl chloride, propionyl chloride, butyryl chloride, valeryl chloride, hexanoyl chloride or benzoyl chloride.
本申请中,相比于酸酐,酰氯的反应效果更好。In the present application, compared with acid anhydride, the reaction effect of acid chloride is better.
优选地,步骤(3”)中,采用四丁基氟化铵进行完全脱保护反应。Preferably, in step (3"), tetrabutylammonium fluoride is used to carry out a complete deprotection reaction.
优选地,步骤(4”)中,所述氯代试剂选自三氯氧磷。Preferably, in step (4"), the chlorinating agent is selected from phosphorus oxychloride.
第三方面,本申请提供一种含有化学修饰胞嘧啶核苷的核酸,所述核酸在制备过程中采用第二方面所述的化学修饰胞嘧啶核苷三磷酸全部替换mRNA中的天然胞嘧啶核苷三磷酸,或者将化学修饰胞嘧啶核苷三磷酸和N1-甲基假尿苷三磷酸联用替换mRNA中的天然胞嘧啶核苷三磷酸。In a third aspect, the present application provides a nucleic acid containing chemically modified cytosine nucleosides, wherein during the preparation process of the nucleic acid, the chemically modified cytosine nucleoside triphosphates described in the second aspect are used to completely replace the natural cytosine nucleoside triphosphates in the mRNA, or the chemically modified cytosine nucleoside triphosphates and N1-methylpseudouridine triphosphate are used in combination to replace the natural cytosine nucleoside triphosphates in the mRNA.
优选地,所述核酸中还含有假尿苷、N-甲基假尿苷、5-甲氧基尿苷或2-硫代尿苷。Preferably, the nucleic acid further contains pseudouridine, N-methylpseudouridine, 5-methoxyuridine or 2-thiouridine.
本申请合成了具有新型结构的非天然修饰核苷酸,并将其引入到mRNA中,从而提高mRNA的稳定性,增强其翻译活性,达到替代、丰富或者补充现有mRNA化学修饰的目标,从而为mRNA新型化学修饰和应用提供助。 The present application synthesizes non-natural modified nucleotides with novel structures and introduces them into mRNA, thereby improving the stability of mRNA and enhancing its translation activity, achieving the goal of replacing, enriching or supplementing existing mRNA chemical modifications, thereby providing assistance for novel chemical modifications and applications of mRNA.
优选地,所述核酸中至少包括:Preferably, the nucleic acid comprises at least:
A)一个启动子序列;A) a promoter sequence;
B)包含至少一个Kozak序列的5’UTR;B) a 5’UTR containing at least one Kozak sequence;
C)3’UTR;C) 3’UTR;
D)连接的核苷组成的编码序列;D) a coding sequence consisting of linked nucleotides;
E)聚A尾。E) Poly A tail.
本申请利用化学合成手段,合成化学修饰胞嘧啶核苷三磷酸,然后利用转录酶和/或加尾酶合成修饰胞嘧啶全局替换的修饰mRNA。修饰mRNA在体外水平和细胞内均表现出增强的翻译效率以及更高的稳定性。The present application uses chemical synthesis to synthesize chemically modified cytosine triphosphates, and then uses transcriptase and/or tailing enzyme to synthesize modified mRNA with global replacement of modified cytosine. The modified mRNA exhibits enhanced translation efficiency and higher stability both in vitro and in cells.
第四方面,本申请提供一种含有化学修饰腺嘌呤核苷的核酸,所述核酸在制备过程中采用第二方面所述的化学修饰腺嘌呤核苷三磷酸全部替换mRNA中的天然腺嘌呤核苷三磷酸,或采用第二方面所述的化学修饰腺嘌呤核苷三磷酸对体外转录制备的mRNA进行多聚腺苷化修饰。In a fourth aspect, the present application provides a nucleic acid containing chemically modified adenine nucleosides, wherein during the preparation process of the nucleic acid, the chemically modified adenine nucleoside triphosphate described in the second aspect is used to completely replace the natural adenine nucleoside triphosphate in the mRNA, or the chemically modified adenine nucleoside triphosphate described in the second aspect is used to perform polyadenylation modification on the mRNA prepared by in vitro transcription.
本申请中,利用化学合成手段,对天然的腺嘌呤进行化学修饰,将这些修饰腺嘌呤三磷酸化,然后利用体外转录合成修饰腺嘌呤全局替换天然腺嘌呤的mRNA,或者利用加尾酶以修饰腺苷对mRNA进行多聚腺苷化修饰。这些修饰的mRNA在体外水平和细胞内均表现出增强的翻译效率以及更高的稳定性。In the present application, natural adenine is chemically modified by chemical synthesis, and these modified adenines are triphosphorylated, and then in vitro transcription is used to synthesize mRNA in which the modified adenines replace the natural adenines globally, or tailing enzymes are used to modify adenosines to perform polyadenylation modification on mRNA. These modified mRNAs show enhanced translation efficiency and higher stability both in vitro and in cells.
优选地,所述化学修饰的核酸中至少包括:Preferably, the chemically modified nucleic acid comprises at least:
A’)一个启动子序列;A') a promoter sequence;
B’)包含至少一个Kozak序列的5’UTR;B’) 5’UTR containing at least one Kozak sequence;
C’)3’UTR;C’) 3’UTR;
D’)连接的核苷组成的编码序列;D') a coding sequence consisting of linked nucleosides;
E’)聚A尾。E') Poly A tail.
第五方面,本申请提供第三方面所述的含有化学修饰胞嘧啶核苷的核酸的制备方法,所述制备方法包括:In a fifth aspect, the present application provides a method for preparing a nucleic acid containing a chemically modified cytosine nucleoside according to the third aspect, the preparation method comprising:
以腺嘌呤核苷三磷酸、鸟嘌呤核苷三磷酸、尿嘧啶核苷三磷酸和化学修饰的胞嘧啶核苷三磷酸四种核糖核苷酸为原料,在RNA聚合酶催化下,以DNA为模板进行PCR反应,合成化学修饰的核酸。Using four ribonucleotides, adenosine triphosphate, guanosine triphosphate, uridine triphosphate and chemically modified cytosine triphosphate, as raw materials, PCR reaction is carried out with DNA as a template under the catalysis of RNA polymerase to synthesize chemically modified nucleic acids.
优选地,所述RNA聚合酶为T7 RNA聚合酶。Preferably, the RNA polymerase is T7 RNA polymerase.
优选地,所述PCR反应的体系中包括:DNA模板、RNA酶抑制剂、T7 RNA聚合酶、RNA聚合酶缓冲液、腺嘌呤核苷三磷酸、鸟嘌呤核苷三磷酸、尿嘧啶核苷三磷酸、化学修饰胞嘧啶核苷三磷酸以及5’帽子结构。Preferably, the PCR reaction system includes: a DNA template, an RNase inhibitor, T7 RNA polymerase, an RNA polymerase buffer, adenosine triphosphate, guanosine triphosphate, uridine triphosphate, chemically modified cytosine triphosphate and a 5' cap structure.
本申请中,所述化学修饰的核酸为采用化学修饰胞苷三磷酸全局替换天然胞苷的修饰mRNA,所述mRNA在无细胞体系和细胞内均具有更高的翻译效率,且更稳定。含有化学修饰嘧啶核苷的mRNA的合成和翻译的技术路线如图1所示。In the present application, the chemically modified nucleic acid is a modified mRNA in which chemically modified cytidine triphosphate is used to globally replace natural cytidine, and the mRNA has higher translation efficiency and is more stable both in a cell-free system and in cells. The technical route for the synthesis and translation of mRNA containing chemically modified pyrimidine nucleosides is shown in Figure 1.
第六方面,本申请提供第四方面所述的含有化学修饰腺嘌呤核苷的核酸的制备方法,所述制备方法包括:In a sixth aspect, the present application provides a method for preparing a nucleic acid containing chemically modified adenine nucleosides according to the fourth aspect, the preparation method comprising:
以胞嘧啶核苷三磷酸、尿嘧啶核苷三磷酸、鸟嘌呤核苷三磷酸和化学修饰腺嘌呤核苷三磷酸四种核糖核苷酸为原料,在RNA聚合酶催化下,以DNA为模板进行PCR反应,合成化学修饰的核酸; Using cytosine triphosphate, uridine triphosphate, guanine triphosphate and chemically modified adenine triphosphate as raw materials, PCR reaction is carried out with DNA as template under the catalysis of RNA polymerase to synthesize chemically modified nucleic acids;
或,以体外转录得到的mRNA为模板,以化学修饰的腺嘌呤核苷三磷酸为底物,用E.Coli poly(A)聚合酶或者Yeast poly(A)聚合酶对mRNA进行加尾反应,得到化学修饰的核酸。Alternatively, using in vitro transcribed mRNA as a template and chemically modified adenine nucleoside triphosphate as a substrate, the mRNA is tailed using E. coli poly (A) polymerase or Yeast poly (A) polymerase to obtain chemically modified nucleic acid.
优选地,所述RNA聚合酶为T7 RNA聚合酶。Preferably, the RNA polymerase is T7 RNA polymerase.
优选地,所述PCR反应的体系中包括:DNA模板、RNA酶抑制剂、T7 RNA聚合酶、RNA聚合酶缓冲液、化学修饰腺嘌呤核苷三磷酸、鸟嘌呤核苷三磷酸、尿嘧啶核苷三磷酸、胞嘧啶核苷三磷酸以及5’帽子结构。Preferably, the PCR reaction system includes: a DNA template, an RNase inhibitor, T7 RNA polymerase, an RNA polymerase buffer, chemically modified adenosine triphosphate, guanosine triphosphate, uridine triphosphate, cytosine triphosphate and a 5' cap structure.
本申请中,所述化学修饰的核酸为采用化学修饰腺嘌呤核苷三磷酸全局替换天然腺嘌呤核苷三磷酸的修饰mRNA,所述mRNA在无细胞体系和细胞内均具有更高的翻译效率,且更稳定。含有化学修饰腺嘌呤核苷的mRNA的合成和翻译的技术路线如图1所示。In the present application, the chemically modified nucleic acid is a modified mRNA in which chemically modified adenosine triphosphate is used to replace natural adenosine triphosphate globally, and the mRNA has higher translation efficiency and is more stable in both cell-free systems and cells. The technical route for the synthesis and translation of mRNA containing chemically modified adenosine nucleosides is shown in Figure 1.
第七方面,本申请提供第一方面所述的化学修饰核苷、第二方面所述的化学修饰核苷三磷酸、第三方面所述的含有化学修饰胞嘧啶核苷的核酸、第四方面所述的含有化学修饰腺嘌呤核苷的核酸、第五方面所述的含有化学修饰胞嘧啶核苷的核酸的制备方法或第六方面所述的含有化学修饰腺嘌呤核苷的核酸的制备方法中任意一种或至少两种的组合在制备mRNA药物中的应用。In the seventh aspect, the present application provides the use of any one or a combination of at least two of the chemically modified nucleosides described in the first aspect, the chemically modified nucleoside triphosphates described in the second aspect, the nucleic acids containing chemically modified cytosine nucleosides described in the third aspect, the nucleic acids containing chemically modified adenine nucleosides described in the fourth aspect, the method for preparing nucleic acids containing chemically modified cytosine nucleosides described in the fifth aspect, or the method for preparing nucleic acids containing chemically modified adenine nucleosides described in the sixth aspect in the preparation of mRNA drugs.
相对于现有技术,本申请具有以下有益效果:Compared with the prior art, this application has the following beneficial effects:
本申请提供了全新的化学修饰核苷和化学修饰核苷三磷酸,包括化学修饰胞嘧啶核苷、化学修饰胞嘧啶核苷三磷酸、化学修饰腺嘌呤核苷和化学修饰腺嘌呤核苷三磷酸;为mRNA提供了更多可用的胞嘧啶核苷三磷酸,本申请中合成的化学修饰核苷三磷酸均可以用于mRNA的合成,可以替代相应的天然核苷,并且能够发挥相应的翻译功能,能够增强目标mRNA的翻译效率和稳定性,降低免疫原性,为mRNA技术提供补充和新选择。The present application provides completely new chemically modified nucleosides and chemically modified nucleoside triphosphates, including chemically modified cytosine nucleosides, chemically modified cytosine nucleoside triphosphates, chemically modified adenine nucleosides and chemically modified adenine nucleoside triphosphates; more available cytosine nucleoside triphosphates are provided for mRNA; the chemically modified nucleoside triphosphates synthesized in the present application can all be used for the synthesis of mRNA, can replace the corresponding natural nucleosides, and can exert corresponding translation functions, can enhance the translation efficiency and stability of the target mRNA, reduce immunogenicity, and provide a supplement and new option for mRNA technology.
以本申请提供的化学修饰胞嘧啶核苷三磷酸进行替换的mRNA在无细胞体系及细胞水平反应实验中,显示修饰胞嘧啶替换的mRNA在翻译效果达到或超过了目前已报到技术中mRNA修饰所用的假尿苷和甲基假尿苷替换尿苷的修饰方法,同时具有良好的生物稳定性。In cell-free system and cell-level reaction experiments, the mRNA replaced with the chemically modified cytosine triphosphate provided in the present application showed that the translation effect of the mRNA replaced with modified cytosine reached or exceeded the modification method of replacing uridine with pseudouridine and methylpseudouridine used in the currently reported technology for mRNA modification, and at the same time had good biological stability.
以本申请提供的化学修饰腺嘌呤核苷三磷酸进行替换的mRNA在体外和细胞水平上翻译效果好,优于采用假尿苷替换尿苷的修饰方法,以本申请提供的化学修饰腺嘌呤核苷与假尿苷联用进行替换的mRNA在体外和细胞水平上的翻译效果,也优于采用假尿苷替换尿苷的修饰方法,以化学修饰腺苷进行多聚腺苷化加尾的mRNA在细胞水平上的翻译效果,也优于具有多聚天然腺苷尾的mRNA,同时在单核苷水平上具有更高的稳定性。The mRNA replaced with the chemically modified adenine nucleoside triphosphate provided in the present application has good translation effects at the in vitro and cellular levels, which is better than the modification method using pseudouridine to replace uridine. The mRNA replaced with the chemically modified adenine nucleoside and pseudouridine provided in the present application has a better translation effect at the in vitro and cellular levels, which is also better than the modification method using pseudouridine to replace uridine. The mRNA polyadenylated and tailed with chemically modified adenosine has a better translation effect at the cellular level than the mRNA with a poly-natural adenosine tail, and at the same time has higher stability at the single nucleoside level.
图1是含有化学修饰核苷的mRNA的合成和翻译的技术路线图。FIG1 is a technical roadmap for the synthesis and translation of mRNA containing chemically modified nucleosides.
图2是化学修饰的胞苷三磷酸的制备技术路线图。FIG. 2 is a technical roadmap for the preparation of chemically modified cytidine triphosphate.
图3为N4-丙酰基胞苷三磷酸的1H NMR谱图。FIG3 is a 1 H NMR spectrum of N4-propionylcytidine triphosphate.
图4为N4-丙酰基胞苷三磷酸的31P NMR谱图。FIG4 is a 31 P NMR spectrum of N4-propionylcytidine triphosphate.
图5为N4-丙酰基胞苷三磷酸的质谱图。FIG5 is a mass spectrum of N4-propionylcytidine triphosphate.
图6为N4-丁酰基胞苷三磷酸的1H NMR谱图。FIG6 is a 1 H NMR spectrum of N4-butyrylcytidine triphosphate.
图7为N4-丁酰基胞苷三磷酸的31P NMR谱图。FIG. 7 is a 31 P NMR spectrum of N4-butyrylcytidine triphosphate.
图8为N4-戊酰基胞苷三磷酸的1H NMR谱图。FIG8 is a 1 H NMR spectrum of N4-pentanoyl cytidine triphosphate.
图9为N4-戊酰基胞苷三磷酸的31P NMR谱图。FIG. 9 is a 31 P NMR spectrum of N4-pentanoyl cytidine triphosphate.
图10为N4-己酰基胞苷三磷酸的1H NMR谱图。 FIG. 10 is a 1 H NMR spectrum of N4-hexanoyl cytidine triphosphate.
图11为N4-己酰基胞苷三磷酸的31P NMR谱图。FIG. 11 is a 31 P NMR spectrum of N4-hexanoyl cytidine triphosphate.
图12为5-甲基-N4-乙酰基胞苷三磷酸的1H NMR谱图。FIG12 is a 1 H NMR spectrum of 5-methyl-N4-acetylcytidine triphosphate.
图13为5-甲基-N4-乙酰基胞苷三磷酸的31P NMR谱图。FIG13 is a 31 P NMR spectrum of 5-methyl-N4-acetylcytidine triphosphate.
图14为5-甲基-N4-丁酰基胞苷三磷酸的1H NMR谱图。FIG14 is a 1 H NMR spectrum of 5-methyl-N4-butyryl cytidine triphosphate.
图15为5-甲基-N4-丁酰基胞苷三磷酸的31P NMR谱图。FIG. 15 is a 31 P NMR spectrum of 5-methyl-N4-butyryl cytidine triphosphate.
图16为N4-苯甲酰基胞苷三磷酸的1H NMR谱图。FIG. 16 is a 1 H NMR spectrum of N4-benzoylcytidine triphosphate.
图17为N4-苯甲酰基胞苷三磷酸的31P NMR谱图。FIG. 17 is a 31 P NMR spectrum of N4-benzoylcytidine triphosphate.
图18为N4-乙酰基胞苷三磷酸的1H NMR谱图。FIG. 18 is a 1 H NMR spectrum of N4-acetylcytidine triphosphate.
图19为N4-乙酰基胞苷三磷酸的31P NMR谱图。FIG. 19 is a 31 P NMR spectrum of N4-acetylcytidine triphosphate.
图20为5-甲基胞苷三磷酸的1H NMR谱图。FIG. 20 is a 1 H NMR spectrum of 5-methylcytidine triphosphate.
图21为5-甲基胞苷三磷酸的31P NMR谱图。FIG. 21 is a 31 P NMR spectrum of 5-methylcytidine triphosphate.
图22为5-甲基-N4-己酰基胞苷三磷酸的1H NMR谱图。FIG. 22 is a 1 H NMR spectrum of 5-methyl-N4-hexanoyl cytidine triphosphate.
图23为5-甲基-N4-己酰基胞苷三磷酸的31P NMR谱图。FIG. 23 is a 31 P NMR spectrum of 5-methyl-N4-hexanoyl cytidine triphosphate.
图24为N4-辛酰基胞苷三磷酸的1H NMR谱图。FIG. 24 is a 1 H NMR spectrum of N4-octanoylcytidine triphosphate.
图25为N4-癸酰基胞苷三磷酸的1H NMR谱图。FIG. 25 is a 1 H NMR spectrum of N4-decanoylcytidine triphosphate.
图26为5-甲基-N4-苯甲酰基胞苷三磷酸的1H NMR谱图。FIG26 is a 1 H NMR spectrum of 5-methyl-N4-benzoylcytidine triphosphate.
图27为2’-氟-N4-乙酰基胞苷三磷酸的1H NMR谱图。FIG27 is a 1 H NMR spectrum of 2'-fluoro-N4-acetylcytidine triphosphate.
图28为2’-氟-N4-丁酰基胞苷三磷酸的1H NMR谱图。FIG. 28 is a 1 H NMR spectrum of 2'-fluoro-N4-butyrylcytidine triphosphate.
图29为2’-氟-N4-己酰基胞苷三磷酸的1H NMR谱图。FIG. 29 is a 1 H NMR spectrum of 2'-fluoro-N4-hexanoyl cytidine triphosphate.
图30为N4-丁酰基胞苷三磷酸的质谱图。FIG30 is a mass spectrum of N4-butyrylcytidine triphosphate.
图31为N4-戊酰基胞苷三磷酸的质谱图。FIG31 is a mass spectrum of N4-pentanoylcytidine triphosphate.
图32为N4-己酰基胞苷三磷酸的质谱图。FIG32 is a mass spectrum of N4-hexanoyl cytidine triphosphate.
图33为5-甲基-N4-乙酰基胞苷三磷酸的质谱图。FIG33 is a mass spectrum of 5-methyl-N4-acetylcytidine triphosphate.
图34为5-甲基-N4-丁酰基胞苷三磷酸的质谱图。FIG34 is a mass spectrum of 5-methyl-N4-butyryl cytidine triphosphate.
图35为N4-乙酰基胞苷三磷酸的质谱图。FIG35 is a mass spectrum of N4-acetylcytidine triphosphate.
图36为5-甲基胞苷三磷酸的质谱图。FIG36 is a mass spectrum of 5-methylcytidine triphosphate.
图37为5-甲基-N4-己酰基胞苷三磷酸的质谱图。FIG37 is a mass spectrum of 5-methyl-N4-hexanoyl cytidine triphosphate.
图38为化学修饰的C系列核苷三磷酸的体外转录结果示意图。FIG38 is a schematic diagram of the in vitro transcription results of chemically modified C-series nucleoside triphosphates.
图39为转录的mRNA转染HEK 293T细胞24h后的表达效率图。Figure 39 shows the expression efficiency of transcribed mRNA after transfection into HEK 293T cells for 24 hours.
图40为N1-甲基假尿苷和修饰胞苷联用转录的mRNA转染HEK 293T细胞24h后的表达效率图。Figure 40 shows the expression efficiency of mRNA transcribed using N1-methylpseudouridine and modified cytidine 24 hours after transfection into HEK 293T cells.
图41为转录的mRNA转染HEK 293T细胞24h后的表达效率图。Figure 41 shows the expression efficiency of transcribed mRNA after transfection into HEK 293T cells for 24 hours.
图42为N1-甲基假尿苷和修饰胞苷联用转录的mRNA转染HeLa细胞24h后的表达效率图。Figure 42 is a graph showing the expression efficiency of mRNA transcribed using a combination of N1-methylpseudouridine and modified cytidine 24 hours after transfection into HeLa cells.
图43为化学修饰的mRNA在HEK 293T细胞中的稳定性的qPCR结果图。Figure 43 is the qPCR results of the stability of chemically modified mRNA in HEK 293T cells.
图44为化学修饰的mRNA在HeLa细胞中的稳定性测试的qPCR结果图。FIG. 44 is a graph showing the qPCR results of the stability test of chemically modified mRNA in HeLa cells.
图45为化学修饰的mRNA在RAW264.7 cell中的免疫原性测试的蛋白印迹结果。Figure 45 shows the protein blotting results of the immunogenicity test of chemically modified mRNA in RAW264.7 cells.
图46是N6-丙酰基腺苷的1H NMR表征图。FIG. 46 is a 1 H NMR characterization chart of N6-propionyl adenosine.
图47是N6-丙酰基腺苷的13C NMR表征图。FIG. 47 is a 13 C NMR characterization chart of N6-propionyl adenosine.
图48是N6-丙酰基腺苷的HRMS表征图。 FIG. 48 is a HRMS characterization chart of N6-propionyl adenosine.
图49是N6-丙酰基腺苷三磷酸的1H NMR表征图。FIG. 49 is a 1 H NMR characterization chart of N6-propionyl adenosine triphosphate.
图50是N6-丙酰基腺苷三磷酸的13C NMR表征图。FIG. 50 is a 13 C NMR characterization chart of N6-propionyl adenosine triphosphate.
图51是N6-丙酰基腺苷三磷酸的HRMS表征图。FIG. 51 is a HRMS characterization chart of N6-propionyl ATP.
图52是N6-丁酰基腺苷的1H NMR表征图。FIG. 52 is a 1 H NMR characterization chart of N6-butyryladenosine.
图53是N6-丁酰基腺苷的13C NMR表征图。FIG. 53 is a 13 C NMR characterization chart of N6-butyryladenosine.
图54是N6-丁酰基腺苷的HRMS表征。Figure 54 is a HRMS characterization of N6-butyryladenosine.
图55是N6-丁酰基腺苷三磷酸的1H NMR表征图。FIG. 55 is a 1 H NMR characterization chart of N6-butyryl adenosine triphosphate.
图56是N6-丁酰基腺苷三磷酸的13C NMR表征图。FIG. 56 is a 13 C NMR characterization chart of N6-butyryl adenosine triphosphate.
图57是N6-丁酰基腺苷三磷酸的HRMS表征。Figure 57 is a HRMS characterization of N6-butyryl adenosine triphosphate.
图58是N6-戊酰基腺苷的1H NMR表征图。FIG. 58 is a 1 H NMR characterization chart of N6-pentanoyl adenosine.
图59是N6-戊酰基腺苷的13C NMR表征图。FIG. 59 is a 13 C NMR characterization chart of N6-pentanoyl adenosine.
图60是N6-戊酰基腺苷的HRMS表征图。Figure 60 is a HRMS characterization chart of N6-pentanoyl adenosine.
图61是N6-戊酰基腺苷三磷酸的1H NMR表征图。FIG. 61 is a 1 H NMR characterization chart of N6-pentanoyl adenosine triphosphate.
图62是N6-戊酰基腺苷三磷酸的13C NMR表征图。FIG. 62 is a 13 C NMR characterization chart of N6-pentanoyl adenosine triphosphate.
图63是N6-戊酰基腺苷三磷酸的HRMS表征图。FIG. 63 is a HRMS characterization chart of N6-pentanoyl adenosine triphosphate.
图64是N6-己酰基腺苷的1H NMR表征图。FIG. 64 is a 1 H NMR characterization chart of N6-hexanoyl adenosine.
图65是N6-己酰基腺苷的13C NMR表征图。FIG. 65 is a 13 C NMR characterization chart of N6-hexanoyl adenosine.
图66是N6-己酰基腺苷的HRMS表征图。Figure 66 is a HRMS characterization chart of N6-hexanoyl adenosine.
图67是N6-己酰基腺苷三磷酸的1H NMR表征图。FIG. 67 is a 1 H NMR characterization chart of N6-hexanoyl adenosine triphosphate.
图68是N6-己酰基腺苷三磷酸的13C NMR表征图。FIG. 68 is a 13 C NMR characterization chart of N6-hexanoyl adenosine triphosphate.
图69是N6-己酰基腺苷三磷酸的HRMS表征图。FIG. 69 is a HRMS characterization chart of N6-hexanoyl adenosine triphosphate.
图70是N6-苯甲酰基腺苷三磷酸的HRMS表征图。FIG. 70 is a HRMS characterization chart of N6-benzoyl adenosine triphosphate.
图71是N6-甲基腺苷三磷酸的HRMS表征图。Figure 71 is the HRMS characterization chart of N6-methyladenosine triphosphate.
图72是N6-乙酰基腺苷三磷酸的HRMS表征图。Figure 72 is a HRMS characterization chart of N6-acetyl adenosine triphosphate.
图73是2-氨基腺苷三磷酸的HRMS表征图。FIG. 73 is a HRMS characterization chart of 2-aminoadenosine triphosphate.
图74是7-脱氮腺苷三磷酸的HRMS表征图。FIG. 74 is a HRMS characterization chart of 7-deaza-ATP.
图75是以E.Coli polyA聚合酶对eGFP mRNA进行多聚腺苷化修饰的效果图。Figure 75 is a diagram showing the effect of polyadenylation modification of eGFP mRNA by E. coli polyA polymerase.
图76是以Yeast polyA聚合酶对eGFP mRNA进行多聚腺苷化修饰的效果图。Figure 76 shows the effect of polyadenylation modification of eGFP mRNA by Yeast polyA polymerase.
图77是化学修饰的腺苷和假尿苷联用的荧光素酶mRNA在兔网织红细胞裂解物中孵育90min后表达的效果图。Figure 77 is a graph showing the expression of luciferase mRNA in combination with chemically modified adenosine and pseudouridine after incubation in rabbit reticulocyte lysate for 90 minutes.
图78是化学修饰的腺苷全局替换天然腺苷的荧光素酶mRNA在兔网织红细胞裂解物中孵育90min后表达的效果图。FIG78 is a graph showing the expression effect of luciferase mRNA in which chemically modified adenosine globally replaces natural adenosine after incubation in rabbit reticulocyte lysate for 90 minutes.
图79是化学修饰的腺苷全局替换天然腺苷的荧光素酶mRNA在转染进HEK 293T细胞后24h表达的效果图。Figure 79 is a graph showing the expression effect of luciferase mRNA in which chemically modified adenosine globally replaces natural adenosine 24 hours after being transfected into HEK 293T cells.
图80是化学修饰的腺苷和假尿苷联用的荧光素酶mRNA在转染进HeLa细胞后24h表达的效果图。Figure 80 is a graph showing the expression effect of luciferase mRNA combined with chemically modified adenosine and pseudouridine 24 hours after transfection into HeLa cells.
图81是化学修饰的腺苷多聚腺苷化修饰的eGFP mRNA在HEK 293T细胞中表达效果的荧光成像图。Figure 81 is a fluorescence imaging image of the expression effect of chemically modified adenosine polyadenylation-modified eGFP mRNA in HEK 293T cells.
下面通过具体实施方式来进一步说明本申请的技术方案。本领域技术人员应该明了,所述实施例仅仅是帮助理解本申请,不应视为对本申请的具体限制。The technical solution of the present application is further described below through specific implementation methods. Those skilled in the art should understand that the embodiments are only to help understand the present application and should not be regarded as specific limitations of the present application.
实施例中未注明具体技术或条件者,按照本领域内的文献所描述的技术或条件,或者按照产品说明书进行。所用试剂或仪器未注明生产厂商者,均为可通过正规渠道商购获得的常规产品。If no specific techniques or conditions are specified in the examples, the techniques or conditions described in the literature in the field or the product instructions are used. If no manufacturer is specified for the reagents or instruments used, they are all conventional products that can be purchased through regular channels.
实施例1化学修饰胞苷三磷酸的制备方法Example 1 Preparation method of chemically modified cytidine triphosphate
本实施例提供一种化学修饰胞苷三磷酸的制备方法,化学修饰胞苷三磷酸的制备技术路线如图2所示。其中,各步骤的反应条件如下所示,并采用核磁共振和质谱对化合物结构进行表征:This embodiment provides a method for preparing chemically modified cytidine triphosphate, and the preparation technology route of chemically modified cytidine triphosphate is shown in Figure 2. The reaction conditions of each step are as follows, and the structure of the compound is characterized by nuclear magnetic resonance and mass spectrometry:
a.取3mmol(1eq)胞苷以15mL N,N-二甲基甲酰胺溶解,然后依次加入12mmol(4eq)叔丁基二甲基氯硅烷和15mmol(5eq)咪唑,25℃下搅拌过夜;反应结束后以10mL乙酸乙酯萃取反应液,重复3次,水洗有机相,重复3次,收集有机相,无水硫酸钠干燥,过滤,浓缩,硅胶柱色谱纯化(二氯甲烷/甲醇=100/1),得到白色固体化合物1(即式12化合物);化合物1的结构由1H NMR、13C NMR和HRMS确认。a. 3 mmol (1 eq) of cytidine was dissolved in 15 mL of N,N-dimethylformamide, and then 12 mmol (4 eq) of tert-butyldimethylsilyl chloride and 15 mmol (5 eq) of imidazole were added in sequence, and stirred at 25°C overnight; after the reaction, the reaction solution was extracted with 10 mL of ethyl acetate, and the extraction was repeated 3 times; the organic phase was washed with water, and the extraction was repeated 3 times; the organic phase was collected, dried over anhydrous sodium sulfate, filtered, concentrated, and purified by silica gel column chromatography (dichloromethane/methanol = 100/1) to obtain a white solid compound 1 (i.e., the compound of formula 12); the structure of compound 1 was confirmed by 1 H NMR, 13 C NMR and HRMS.
b.以15mL无水吡啶溶解2mmol(1eq)化合物1,然后滴加1.2eq相应酸酐,之后加入10mg 4-二甲氨基吡啶,25℃下搅拌过夜反应;反应结束后以10mL乙酸乙酯萃取反应液,重复3次,水洗有机相,重复3次,收集有机相,无水硫酸钠干燥,过滤,浓缩,硅胶柱色谱纯化(二氯甲烷/甲醇=200/1),得到白色胶状化合物2(即式13化合物);化合物2的结构由1H NMR、13C NMR和HRMS确认。b. 2 mmol (1 eq) of compound 1 was dissolved in 15 mL of anhydrous pyridine, and then 1.2 eq of the corresponding acid anhydride was added dropwise, followed by 10 mg of 4-dimethylaminopyridine. The mixture was stirred at 25°C overnight. After the reaction, the reaction solution was extracted with 10 mL of ethyl acetate, and the extraction was repeated 3 times. The organic phase was washed with water, and the extraction was repeated 3 times. The organic phase was collected, dried over anhydrous sodium sulfate, filtered, concentrated, and purified by silica gel column chromatography (dichloromethane/methanol = 200/1) to obtain a white colloidal compound 2 (i.e., the compound of formula 13). The structure of compound 2 was confirmed by 1 H NMR, 13 C NMR and HRMS.
c.以15mL无水四氢呋喃溶解1.5mmol(1eq)化合物2,然后滴加4.5mL(3eq)四丁基氟化铵溶液(1M四氢呋喃溶液),室温下搅拌反应0.5h,然后减压浓缩,硅胶柱色谱纯化(二氯甲烷/甲醇=20/1至10/1),得到白色固体化合物3(即式14化合物)。化合物3的结构由1H NMR、13C NMR和HRMS确认。c. Dissolve 1.5 mmol (1 eq) of compound 2 in 15 mL of anhydrous tetrahydrofuran, then dropwise add 4.5 mL (3 eq) of tetrabutylammonium fluoride solution (1 M tetrahydrofuran solution), stir at room temperature for 0.5 h, then concentrate under reduced pressure, and purify by silica gel column chromatography (dichloromethane/methanol=20/1 to 10/1) to obtain a white solid compound 3 (i.e., compound of formula 14). The structure of compound 3 was confirmed by 1 H NMR, 13 C NMR and HRMS.
d.以2mL无水吡啶溶解0.1mmol化合物3,并减压共蒸除水3次;在氮气保护下,注入1mL干燥磷酸三甲酯,待固体完全溶解后将试管置于冰浴中冷却2min,并用注射器滴加1.5eq三氯氧磷;冰浴下搅拌反应2.5h。d. Dissolve 0.1 mmol of compound 3 in 2 mL of anhydrous pyridine and remove water under reduced pressure three times; inject 1 mL of dry trimethyl phosphate under nitrogen protection, and after the solid is completely dissolved, place the test tube in an ice bath to cool for 2 min, and add 1.5 eq of phosphorus oxychloride dropwise with a syringe; stir and react in an ice bath for 2.5 h.
e.之后在冰浴条件下依次注入1.5eq干燥三丁胺,1.5mL(8eq)三丁基焦磷酸铵溶液(0.5M N,N-二甲基甲酰胺溶液),移至室温下反应0.5h。e. Then, inject 1.5 eq of dry tributylamine and 1.5 mL (8 eq) of tributylammonium pyrophosphate solution (0.5 M N,N-dimethylformamide solution) in sequence under ice bath conditions, and move to room temperature to react for 0.5 h.
f.反应完成后,冰浴下以5mL 2.0M饱和三乙胺-碳酸水溶液淬灭反应,然后以10mL二氯甲烷萃取反应液,收集水相,重复4次;以快速制备液相色谱仪C18柱分离纯化收集的水相,并以50mM饱和三乙胺-碳酸水溶液和色谱级乙腈为流动相;最后将收集纯化后的溶液冻干,得到白色固体4(式9化合物)。化合物4的结构由1H NMR、31P NMR和HRMS确认。f. After the reaction is completed, the reaction is quenched with 5 mL of 2.0 M saturated triethylamine-carbonic acid aqueous solution under ice bath, and then the reaction solution is extracted with 10 mL of dichloromethane, and the aqueous phase is collected, and repeated 4 times; the collected aqueous phase is separated and purified by a rapid preparative liquid chromatograph C18 column, and 50 mM saturated triethylamine-carbonic acid aqueous solution and chromatographic grade acetonitrile are used as mobile phases; finally, the collected and purified solution is freeze-dried to obtain a white solid 4 (compound of formula 9). The structure of compound 4 is confirmed by 1 H NMR, 31 P NMR and HRMS.
本实施例中化学修饰的胞苷三磷酸的制备方法中,仅进行步骤a、步骤b和步骤c即为化学修饰的胞苷的制备方法。In the method for preparing chemically modified cytidine triphosphate in this embodiment, only step a, step b and step c are performed to obtain the method for preparing chemically modified cytidine.
实施例2 N4-丙酰基胞苷三磷酸Example 2 N4-Propionylcytidine triphosphate
本实施例提供一种化学修饰胞苷三磷酸,所述化学修饰胞苷三磷酸为N4-丙酰基胞苷三磷酸。制备的各步骤的反应条件如下所示,并采用核磁共振和质谱对化合物结构进行表征:This embodiment provides a chemically modified cytidine triphosphate, wherein the chemically modified cytidine triphosphate is N4-propionyl cytidine triphosphate. The reaction conditions of each step of the preparation are as follows, and the structure of the compound is characterized by nuclear magnetic resonance and mass spectrometry:
a.取3mmol(1eq)胞苷以15mL N,N-二甲基甲酰胺溶解,然后依次加入12mmol(4eq)叔丁基二甲基氯硅烷和15mmol(5eq)咪唑,25℃下搅拌过夜;反应结束后以10mL乙酸 乙酯萃取反应液,重复3次,水洗有机相,重复3次,收集有机相,无水硫酸钠干燥,过滤,浓缩,硅胶柱色谱纯化(二氯甲烷/甲醇=100/1),得到白色固体化合物1(即式12化合物),收率95%;化合物1的结构已由1H NMR、13C NMR和HRMS确认。a. Dissolve 3 mmol (1 eq) of cytidine in 15 mL of N,N-dimethylformamide, then add 12 mmol (4 eq) of tert-butyldimethylsilyl chloride and 15 mmol (5 eq) of imidazole, and stir overnight at 25 °C. After the reaction, add 10 mL of acetic acid. The reaction solution was extracted with ethyl ester for 3 times, and the organic phase was washed with water for 3 times. The organic phase was collected, dried over anhydrous sodium sulfate, filtered, concentrated, and purified by silica gel column chromatography (dichloromethane/methanol=100/1) to obtain white solid compound 1 (i.e. compound 12) with a yield of 95%. The structure of compound 1 has been confirmed by 1 H NMR, 13 C NMR and HRMS.
b.以15mL无水吡啶溶解2mmol(1eq)化合物1,然后滴加1.2eq丙酸酐,之后加入10mg 4-二甲氨基吡啶,25℃下搅拌过夜反应;反应结束后以10mL乙酸乙酯萃取反应液,重复3次,水洗有机相,重复3次,收集有机相,无水硫酸钠干燥,过滤,浓缩,硅胶柱色谱纯化(二氯甲烷/甲醇=200/1),得到白色胶状化合物2(即式13化合物),收率92%;化合物2的结构已由1H NMR、13C NMR和HRMS确认。b. 2 mmol (1 eq) of compound 1 was dissolved in 15 mL of anhydrous pyridine, and then 1.2 eq of propionic anhydride was added dropwise, followed by 10 mg of 4-dimethylaminopyridine. The mixture was stirred at 25°C overnight to react. After the reaction, the reaction solution was extracted with 10 mL of ethyl acetate, and the extraction was repeated 3 times. The organic phase was washed with water, and the extraction was repeated 3 times. The organic phase was collected, dried over anhydrous sodium sulfate, filtered, concentrated, and purified by silica gel column chromatography (dichloromethane/methanol=200/1) to obtain a white colloidal compound 2 (i.e., the compound of formula 13) with a yield of 92%. The structure of compound 2 was confirmed by 1 H NMR, 13 C NMR and HRMS.
c.以15mL无水四氢呋喃溶解1.5mmol(1eq)化合物2,然后滴加4.5mL(3eq)四丁基氟化铵溶液(1M四氢呋喃溶液),室温下搅拌反应0.5h,然后减压浓缩,硅胶柱色谱纯化(二氯甲烷/甲醇=20/1至10/1),得到白色固体化合物3(即式14化合物)。化合物3的结构已由1H NMR、13C NMR和HRMS确认。c. Dissolve 1.5 mmol (1 eq) of compound 2 in 15 mL of anhydrous tetrahydrofuran, then dropwise add 4.5 mL (3 eq) of tetrabutylammonium fluoride solution (1 M tetrahydrofuran solution), stir and react at room temperature for 0.5 h, then concentrate under reduced pressure, and purify by silica gel column chromatography (dichloromethane/methanol=20/1 to 10/1) to obtain a white solid compound 3 (i.e., the compound of formula 14). The structure of compound 3 has been confirmed by 1 H NMR, 13 C NMR and HRMS.
d.以2mL无水吡啶溶解0.1mmol化合物3,并减压共蒸除水3次;在氮气保护下,注入1mL干燥磷酸三甲酯,待固体完全溶解后将试管置于冰浴中冷却2min,并用注射器滴加1.5eq三氯氧磷;冰浴下搅拌反应2.5h。d. Dissolve 0.1 mmol of compound 3 in 2 mL of anhydrous pyridine and remove water under reduced pressure three times; inject 1 mL of dry trimethyl phosphate under nitrogen protection, and after the solid is completely dissolved, place the test tube in an ice bath to cool for 2 min, and add 1.5 eq of phosphorus oxychloride dropwise with a syringe; stir and react in an ice bath for 2.5 h.
e.之后在冰浴条件下依次注入1.5eq干燥三丁胺,1.5mL(8eq)三丁基焦磷酸铵溶液(0.5M N,N-二甲基甲酰胺溶液),移至室温下反应0.5h。e. Then, inject 1.5 eq of dry tributylamine and 1.5 mL (8 eq) of tributylammonium pyrophosphate solution (0.5 M N,N-dimethylformamide solution) in sequence under ice bath conditions, and move to room temperature to react for 0.5 h.
f.反应完成后,冰浴下以5mL 2.0M饱和三乙胺-碳酸水溶液淬灭反应,然后以10mL二氯甲烷萃取反应液,收集水相,重复4次;以快速制备液相色谱仪C18柱分离纯化收集的水相,并以50mM饱和三乙胺-碳酸水溶液和色谱级乙腈为流动相;最后将收集纯化后的溶液冻干,得到白色固体4(式9化合物)。化合物4的结构已由1H NMR、31P NMR和HRMS确认。f. After the reaction is completed, the reaction is quenched with 5 mL of 2.0 M saturated triethylamine-carbonic acid aqueous solution under ice bath, and then the reaction solution is extracted with 10 mL of dichloromethane, and the aqueous phase is collected, and repeated 4 times; the collected aqueous phase is separated and purified by a rapid preparative liquid chromatograph C18 column, and 50 mM saturated triethylamine-carbonic acid aqueous solution and chromatographic grade acetonitrile are used as mobile phases; finally, the collected and purified solution is freeze-dried to obtain a white solid 4 (compound of formula 9). The structure of compound 4 has been confirmed by 1 H NMR, 31 P NMR and HRMS.
图3为N4-丙酰基胞苷三磷酸的1H NMR谱图,从图中可知,峰的位置(单位:ppm)依次为:1.07,1.09,1.11;2.44,2.46,2.48,2.49;4.27,4.28,4.30,4.31,4.32,4.38,4.40,4.41;5.92,5.93;7.29,7.31;8.39,8.40。图4为N4-丙酰基胞苷三磷酸的31P NMR谱图,从图中可知,峰的位置(单位:ppm)依次为:-22.70,-22.57,-22.45;-11.48,-11.46,-11.35,-11.34;-6.53,-6.40;19.45,19.51,19.57。图5为N4-丙酰基胞苷三磷酸的质谱图。Figure 3 is the 1 H NMR spectrum of N4-propionylcytidine triphosphate. It can be seen from the figure that the peak positions (unit: ppm) are: 1.07, 1.09, 1.11; 2.44, 2.46, 2.48, 2.49; 4.27, 4.28, 4.30, 4.31, 4.32, 4.38, 4.40, 4.41; 5.92, 5.93; 7.29, 7.31; 8.39, 8.40. Figure 4 is a 31 P NMR spectrum of N4-propionyl cytidine triphosphate, from which it can be seen that the peak positions (unit: ppm) are: -22.70, -22.57, -22.45; -11.48, -11.46, -11.35, -11.34; -6.53, -6.40; 19.45, 19.51, 19.57. Figure 5 is a mass spectrum of N4-propionyl cytidine triphosphate.
实施例3 N4-丁酰基胞苷三磷酸Example 3 N4-butyryl cytidine triphosphate
本实施例提供一种化学修饰胞苷三磷酸,所述化学修饰胞苷三磷酸为N4-丁酰基胞苷三磷酸。制备的各步骤的反应条件如下所示,并采用核磁共振和质谱对化合物结构进行表征:This embodiment provides a chemically modified cytidine triphosphate, wherein the chemically modified cytidine triphosphate is N4-butyryl cytidine triphosphate. The reaction conditions of each step of the preparation are as follows, and the structure of the compound is characterized by nuclear magnetic resonance and mass spectrometry:
a.取3mmol(1eq)胞苷以15mL N,N-二甲基甲酰胺溶解,然后依次加入12mmol(4eq)叔丁基二甲基氯硅烷和15mmol(5eq)咪唑,25℃下搅拌过夜;反应结束后以10mL乙酸乙酯萃取反应液,重复3次,水洗有机相,重复3次,收集有机相,无水硫酸钠干燥,过滤,浓缩,硅胶柱色谱纯化(二氯甲烷/甲醇=100/1),得到白色固体化合物1(即式12化合物),收率93%;化合物1的结构已由1H NMR、13C NMR和HRMS确认。a. 3 mmol (1 eq) of cytidine was dissolved in 15 mL of N,N-dimethylformamide, and then 12 mmol (4 eq) of tert-butyldimethylsilyl chloride and 15 mmol (5 eq) of imidazole were added in sequence, and stirred at 25°C overnight; after the reaction, the reaction solution was extracted with 10 mL of ethyl acetate, and the extraction was repeated 3 times; the organic phase was washed with water, and the extraction was repeated 3 times; the organic phase was collected, dried over anhydrous sodium sulfate, filtered, concentrated, and purified by silica gel column chromatography (dichloromethane/methanol=100/1) to obtain a white solid compound 1 (i.e., the compound of formula 12) with a yield of 93%; the structure of compound 1 has been confirmed by 1 H NMR, 13 C NMR and HRMS.
b.以15mL无水吡啶溶解2mmol(1eq)化合物1,然后滴加1.2eq丁酸酐,之后加入10mg 4-二甲氨基吡啶,25℃下搅拌过夜反应;反应结束后以10mL乙酸乙酯萃取反应液,重复3次,水洗有机相,重复3次,收集有机相,无水硫酸钠干燥,过滤,浓缩,硅胶柱色谱纯化(二氯甲烷/甲醇=200/1),得到白色胶状化合物2(即式13化合物),收率92%;化合物2的结构已由1H NMR、13C NMR和HRMS确认。 b. 2 mmol (1 eq) of compound 1 was dissolved in 15 mL of anhydrous pyridine, and then 1.2 eq of butyric anhydride was added dropwise, followed by 10 mg of 4-dimethylaminopyridine. The mixture was stirred at 25°C overnight to react. After the reaction, the reaction solution was extracted with 10 mL of ethyl acetate, and the extraction was repeated 3 times. The organic phase was washed with water, and the extraction was repeated 3 times. The organic phase was collected, dried over anhydrous sodium sulfate, filtered, concentrated, and purified by silica gel column chromatography (dichloromethane/methanol=200/1) to obtain a white colloidal compound 2 (i.e., the compound of formula 13) with a yield of 92%. The structure of compound 2 was confirmed by 1 H NMR, 13 C NMR and HRMS.
c.以15mL无水四氢呋喃溶解1.5mmol(1eq)化合物2,然后滴加4.5mL(3eq)四丁基氟化铵溶液(1M四氢呋喃溶液),室温下搅拌反应0.5h,然后减压浓缩,硅胶柱色谱纯化(二氯甲烷/甲醇=20/1至10/1),得到白色固体化合物3(即式14化合物)。化合物3的结构已由1H NMR、13C NMR和HRMS确认。c. Dissolve 1.5 mmol (1 eq) of compound 2 in 15 mL of anhydrous tetrahydrofuran, then dropwise add 4.5 mL (3 eq) of tetrabutylammonium fluoride solution (1 M tetrahydrofuran solution), stir and react at room temperature for 0.5 h, then concentrate under reduced pressure, and purify by silica gel column chromatography (dichloromethane/methanol=20/1 to 10/1) to obtain a white solid compound 3 (i.e., the compound of formula 14). The structure of compound 3 has been confirmed by 1 H NMR, 13 C NMR and HRMS.
d.以2mL无水吡啶溶解0.1mmol化合物3,并减压共蒸除水3次;在氮气保护下,注入1mL干燥磷酸三甲酯,待固体完全溶解后将试管置于冰浴中冷却2min,并用注射器滴加1.5eq三氯氧磷;冰浴下搅拌反应2.5h。d. Dissolve 0.1 mmol of compound 3 in 2 mL of anhydrous pyridine and remove water under reduced pressure three times; inject 1 mL of dry trimethyl phosphate under nitrogen protection, and after the solid is completely dissolved, place the test tube in an ice bath to cool for 2 min, and add 1.5 eq of phosphorus oxychloride dropwise with a syringe; stir and react in an ice bath for 2.5 h.
e.之后在冰浴条件下依次注入1.5eq干燥三丁胺,1.5mL(8eq)三丁基焦磷酸铵溶液(0.5M N,N-二甲基甲酰胺溶液),移至室温下反应0.5h。e. Then, inject 1.5 eq of dry tributylamine and 1.5 mL (8 eq) of tributylammonium pyrophosphate solution (0.5 M N,N-dimethylformamide solution) in sequence under ice bath conditions, and move to room temperature to react for 0.5 h.
f.反应完成后,冰浴下以5mL 2.0M饱和三乙胺-碳酸水溶液淬灭反应,然后以10mL二氯甲烷萃取反应液,收集水相,重复4次;以快速制备液相色谱仪C18柱分离纯化收集的水相,并以50mM饱和三乙胺-碳酸水溶液和色谱级乙腈为流动相;最后将收集纯化后的溶液冻干,得到白色固体4(式9化合物)。化合物4的结构已由1H NMR、31P NMR和HRMS确认。f. After the reaction is completed, the reaction is quenched with 5 mL of 2.0 M saturated triethylamine-carbonic acid aqueous solution under ice bath, and then the reaction solution is extracted with 10 mL of dichloromethane, and the aqueous phase is collected, and repeated 4 times; the collected aqueous phase is separated and purified by a rapid preparative liquid chromatograph C18 column, and 50 mM saturated triethylamine-carbonic acid aqueous solution and chromatographic grade acetonitrile are used as mobile phases; finally, the collected and purified solution is freeze-dried to obtain a white solid 4 (compound of formula 9). The structure of compound 4 has been confirmed by 1 H NMR, 31 P NMR and HRMS.
图6为N4-丁酰基胞苷三磷酸的1H NMR谱图,从图中可知,峰的位置(单位:ppm)依次为:0.87,0.89,0.91;1.59,1.61,1.63,1.65;2.41,2.42,2.44;4.24,4.27,4.28,4.30,4.31,4.32,4.39,4.40,4.42;5.92,5.93;7.28,7.30;8.39,8.41。图7为N4-丁酰基胞苷三磷酸的31P NMR谱图,从图中可知,峰的位置(单位:ppm)依次为:-22.68,-22.56,-22.43;-11.47,-11.45,-11.34,-11.33;-6.46,-6.33;19.52,19.58。图30为N4-丁酰基胞苷三磷酸的质谱图。Figure 6 is the 1 H NMR spectrum of N4-butyrylcytidine triphosphate. It can be seen from the figure that the peak positions (unit: ppm) are: 0.87, 0.89, 0.91; 1.59, 1.61, 1.63, 1.65; 2.41, 2.42, 2.44; 4.24, 4.27, 4.28, 4.30, 4.31, 4.32, 4.39, 4.40, 4.42; 5.92, 5.93; 7.28, 7.30; 8.39, 8.41. Figure 7 is a 31 P NMR spectrum of N4-butyryl cytidine triphosphate, from which it can be seen that the peak positions (unit: ppm) are: -22.68, -22.56, -22.43; -11.47, -11.45, -11.34, -11.33; -6.46, -6.33; 19.52, 19.58. Figure 30 is a mass spectrum of N4-butyryl cytidine triphosphate.
实施例4 N4-戊酰基胞苷三磷酸Example 4 N4-Valerylcytidine triphosphate
本实施例提供一种化学修饰胞苷三磷酸,所述化学修饰胞苷三磷酸为N4-戊酰基胞苷三磷酸。制备的各步骤的反应条件参照实施例1。步骤b中所使用的酸酐为戊酸酐。This embodiment provides a chemically modified cytidine triphosphate, wherein the chemically modified cytidine triphosphate is N4-pentanoyl cytidine triphosphate. The reaction conditions of each step of the preparation are as described in Example 1. The anhydride used in step b is valeric anhydride.
图8为N4-戊酰基胞苷三磷酸的1H NMR谱图,从图中可知,峰的位置(单位:ppm)依次为:0.80,0.82,0.84;1.26,1.28;1.52,1.53,1.55,1.57,1.59;2.40,2.42,2.44;4.21,4.24,4.25,4.27,4.28,4.29,4.35,4.37,4.38;5.89,5.90;7.25,7.27;8.36,8.38。图9为N4-戊酰基胞苷三磷酸的31P NMR谱图,从图中可知,峰的位置(单位:ppm)依次为:-22.69,-22.57,-22.44;-11.51,-11.49,-11.39,-11.36,-11.35,-11.32;-6.54,-6.41。图31为N4-戊酰基胞苷三磷酸的质谱图。Figure 8 is the 1 H NMR spectrum of N4-pentanoylcytidine triphosphate. It can be seen from the figure that the peak positions (unit: ppm) are: 0.80, 0.82, 0.84; 1.26, 1.28; 1.52, 1.53, 1.55, 1.57, 1.59; 2.40, 2.42, 2.44; 4.21, 4.24, 4.25, 4.27, 4.28, 4.29, 4.35, 4.37, 4.38; 5.89, 5.90; 7.25, 7.27; 8.36, 8.38. Figure 9 is a 31 P NMR spectrum of N4-pentanoyl cytidine triphosphate, from which it can be seen that the peak positions (unit: ppm) are: -22.69, -22.57, -22.44; -11.51, -11.49, -11.39, -11.36, -11.35, -11.32; -6.54, -6.41. Figure 31 is a mass spectrum of N4-pentanoyl cytidine triphosphate.
实施例5 N4-己酰基胞苷三磷酸Example 5 N4-hexanoyl cytidine triphosphate
本实施例提供一种化学修饰胞苷三磷酸,所述化学修饰胞苷三磷酸为N4-己酰基胞苷三磷酸。制备的各步骤的反应条件参照实施例1。步骤b中所使用的酸酐为己酸酐。This embodiment provides a chemically modified cytidine triphosphate, wherein the chemically modified cytidine triphosphate is N4-hexanoyl cytidine triphosphate. The reaction conditions of each step of the preparation are referred to Example 1. The anhydride used in step b is hexanoic anhydride.
图10为N4-己酰基胞苷三磷酸的1H NMR谱图,从图中可知,峰的位置(单位:ppm)依次为:0.77,0.79,0.80;1.23,1.24;1.56,1.58,1.60;2.39,2.41,2.43;4.24,4.25,4.28,4.35,4.37,4.38;5.90;7.26,7.28;8.36,8.38。图11为N4-己酰基胞苷三磷酸的31PNMR谱图,从图中可知,峰的位置(单位:ppm)依次为:-22.64,-22.51,-22.38;-11.45,-11.33;-6.50,-6.37。图32为N4-己酰基胞苷三磷酸的质谱图。Figure 10 is a 1 H NMR spectrum of N4-hexanoyl cytidine triphosphate. It can be seen from the figure that the peak positions (unit: ppm) are: 0.77, 0.79, 0.80; 1.23, 1.24; 1.56, 1.58, 1.60; 2.39, 2.41, 2.43; 4.24, 4.25, 4.28, 4.35, 4.37, 4.38; 5.90; 7.26, 7.28; 8.36, 8.38. Figure 11 is a 31 P NMR spectrum of N4-hexanoyl cytidine triphosphate. It can be seen from the figure that the peak positions (unit: ppm) are: -22.64, -22.51, -22.38; -11.45, -11.33; -6.50, -6.37. FIG32 is a mass spectrum of N4-hexanoyl cytidine triphosphate.
实施例6 5-甲基-N4-乙酰基胞苷三磷酸Example 6 5-Methyl-N4-acetylcytidine triphosphate
本实施例提供一种化学修饰的胞苷三磷酸,所述化学修饰胞苷三磷酸为5-甲基-N4-乙酰基胞苷三磷酸。制备的各步骤的反应条件参照实施例1,区别在于使用的反应起始物为5-甲 基胞苷,步骤b中所使用的酸酐为乙酸酐。This embodiment provides a chemically modified cytidine triphosphate, wherein the chemically modified cytidine triphosphate is 5-methyl-N4-acetyl cytidine triphosphate. The reaction conditions of each step of the preparation are referred to Example 1, except that the reaction starting material used is 5-methyl-N4-acetyl cytidine triphosphate. Cytidine, the acid anhydride used in step b is acetic anhydride.
图12为5-甲基-N4-乙酰基胞苷三磷酸的1H NMR谱图,从图中可知,峰的位置(单位:ppm)依次为:2.06;2.24,2.25;4.24,4.25,4.27,4.28;4.36,4.37,4.39;5.89,5.90,5.91;8.22,8.30。图13为5-甲基-N4-乙酰基胞苷三磷酸的31P NMR谱图,从图中可知,峰的位置(单位:ppm)依次为:-22.99,-22.86,-22.74;-11.77,-11.75,-11.65,-11.63;-8.19,-8.05;19.42,19.47,19.53。图33为5-甲基-N4-乙酰基胞苷三磷酸的质谱图。Figure 12 is the 1 H NMR spectrum of 5-methyl-N4-acetylcytidine triphosphate. It can be seen from the figure that the peak positions (unit: ppm) are: 2.06; 2.24, 2.25; 4.24, 4.25, 4.27, 4.28; 4.36, 4.37, 4.39; 5.89, 5.90, 5.91; 8.22, 8.30. Figure 13 is the 31 P NMR spectrum of 5-methyl-N4-acetylcytidine triphosphate. It can be seen from the figure that the peak positions (unit: ppm) are: -22.99, -22.86, -22.74; -11.77, -11.75, -11.65, -11.63; -8.19, -8.05; 19.42, 19.47, 19.53. FIG33 is a mass spectrum of 5-methyl-N4-acetylcytidine triphosphate.
实施例7 5-甲基-N4-丁酰基胞苷三磷酸Example 7 5-Methyl-N4-butyryl cytidine triphosphate
本实施例提供一种化学修饰胞苷三磷酸,所述化学修饰胞苷三磷酸为5-甲基-N4-丁酰基胞苷三磷酸。制备的各步骤的反应条件参照实施例1,区别在于使用的反应起始物为5-甲基胞苷,步骤b中所使用的酸酐为丁酸酐。This embodiment provides a chemically modified cytidine triphosphate, wherein the chemically modified cytidine triphosphate is 5-methyl-N4-butyryl cytidine triphosphate. The reaction conditions of each step of the preparation refer to those of Example 1, except that the reaction starting material used is 5-methylcytidine, and the anhydride used in step b is butyric anhydride.
图14为5-甲基-N4-丁酰基胞苷三磷酸的1H NMR谱图,从图中可知,峰的位置(单位:ppm)依次为:0.84,0.86,0.88;1.56,1.57,1.59,1.61;2.03;2.43,2.45,2.47;4.19,4.21,4.24,4.25,4.25,4.36,4.37,4.39;5.87,5.88;8.22。图15为5-甲基-N4-丁酰基胞苷三磷酸的31P NMR谱图,从图中可知,峰的位置(单位:ppm)依次为:-22.74,-22.61,-22.48;-11.77,-11.76,-11.65,-11.63;-6.52,-6.39。图34为5-甲基-N4-丁酰基胞苷三磷酸的质谱图。Figure 14 is the 1 H NMR spectrum of 5-methyl-N4-butyryl cytidine triphosphate. It can be seen from the figure that the peak positions (unit: ppm) are: 0.84, 0.86, 0.88; 1.56, 1.57, 1.59, 1.61; 2.03; 2.43, 2.45, 2.47; 4.19, 4.21, 4.24, 4.25, 4.25, 4.36, 4.37, 4.39; 5.87, 5.88; 8.22. Figure 15 is a 31 P NMR spectrum of 5-methyl-N4-butyryl cytidine triphosphate. From the figure, it can be seen that the peak positions (unit: ppm) are: -22.74, -22.61, -22.48; -11.77, -11.76, -11.65, -11.63; -6.52, -6.39. Figure 34 is a mass spectrum of 5-methyl-N4-butyryl cytidine triphosphate.
实施例8 N4-苯甲酰基胞苷三磷酸Example 8 N4-benzoylcytidine triphosphate
本实施例提供一种化学修饰胞苷三磷酸,所述化学修饰胞苷三磷酸为N4-苯甲酰基胞苷三磷酸。制备的各步骤的反应条件参照实施例1。步骤b中所使用的酸酐为苯甲酸酐。This embodiment provides a chemically modified cytidine triphosphate, wherein the chemically modified cytidine triphosphate is N4-benzoyl cytidine triphosphate. The reaction conditions of each step of the preparation are as in Example 1. The anhydride used in step b is benzoic anhydride.
图16为N4-苯甲酰基胞苷三磷酸的1H NMR谱图;从图中可知,峰的位置(单位:ppm)依次为:4.28,4.28,4.29,4.30,4.31,4.33,4.34,4.34,4.35,4.39,4.41,4.42;5.96,5.97;7.43,7.45,7.51,7.53,7.55,7.63,7.65,7.67;7.87,7.87,7.89;8.45,8.47。图17为N4-苯甲酰基胞苷三磷酸的31P NMR谱图,从图中可知,峰的位置(单位:ppm)依次为:-22.88,-22.75,-22.63;-11.51,-11.48,-11.46,-11.43,-11.38,-11.35,-11.34,-11.31;-7.88,-7.77。Figure 16 is the 1 H NMR spectrum of N4-benzoylcytidine triphosphate; it can be seen from the figure that the peak positions (unit: ppm) are: 4.28, 4.28, 4.29, 4.30, 4.31, 4.33, 4.34, 4.34, 4.35, 4.39, 4.41, 4.42; 5.96, 5.97; 7.43, 7.45, 7.51, 7.53, 7.55, 7.63, 7.65, 7.67; 7.87, 7.87, 7.89; 8.45, 8.47. Figure 17 is the 31 P NMR spectrum of N4-benzoylcytidine triphosphate. It can be seen from the figure that the peak positions (unit: ppm) are: -22.88, -22.75, -22.63; -11.51, -11.48, -11.46, -11.43, -11.38, -11.35, -11.34, -11.31; -7.88, -7.77.
实施例9 N4-乙酰基胞苷三磷酸Example 9 N4-acetylcytidine triphosphate
本实施例提供一种化学修饰胞苷三磷酸,所述化学修饰胞苷三磷酸为N4-乙酰基胞苷三磷酸。制备的各步骤的反应条件参照实施例1。步骤b中所使用的酸酐为乙酸酐。This embodiment provides a chemically modified cytidine triphosphate, wherein the chemically modified cytidine triphosphate is N4-acetyl cytidine triphosphate. The reaction conditions of each step of the preparation are as described in Example 1. The anhydride used in step b is acetic anhydride.
图18为N4-乙酰基胞苷三磷酸的1H NMR谱图,从图中可知,峰的位置(单位:ppm)依次为:2.18;4.24,4.27;4.28;4.31,4.31,4.32,4.39,4.40,4.41;5.92,5.93;7.26,7.27;8.39,8.41。图19为N4-乙酰基胞苷三磷酸的31P NMR谱图,从图中可知,峰的位置(单位:ppm)依次为:-22.68,-22.56,-22.43;-11.46,-11.34;-6.46,-6.33。图35为N4-乙酰基胞苷三磷酸的质谱图。Figure 18 is a 1 H NMR spectrum of N4-acetylcytidine triphosphate. It can be seen from the figure that the peak positions (unit: ppm) are: 2.18; 4.24, 4.27; 4.28; 4.31, 4.31, 4.32, 4.39, 4.40, 4.41; 5.92, 5.93; 7.26, 7.27; 8.39, 8.41. Figure 19 is a 31 P NMR spectrum of N4-acetylcytidine triphosphate. It can be seen from the figure that the peak positions (unit: ppm) are: -22.68, -22.56, -22.43; -11.46, -11.34; -6.46, -6.33. Figure 35 is a mass spectrum of N4-acetylcytidine triphosphate.
实施例10 N4-对甲氧基苯甲酰基胞苷三磷酸Example 10 N4-p-Anisyl cytidine triphosphate
本实施例提供一种化学修饰胞苷三磷酸,所述化学修饰胞苷三磷酸为N4-对甲氧基苯甲酰基胞苷三磷酸。制备的各步骤的反应条件参照实施例1。步骤b中所使用的酰氯为对甲氧基苯甲酰氯(4-甲氧基苯甲酰氯)。This embodiment provides a chemically modified cytidine triphosphate, wherein the chemically modified cytidine triphosphate is N4-p-methoxybenzoyl cytidine triphosphate. The reaction conditions of each step of the preparation are referred to Example 1. The acyl chloride used in step b is p-methoxybenzoyl chloride (4-methoxybenzoyl chloride).
对所得N4-对甲氧基苯甲酰基胞苷三磷酸进行检测,核磁数据为:1H NMR(400MHz,D2O)δ9.93(s,1H),8.94(d,J=0.8Hz,1H),7.95(d,J=2.4Hz,2H),7.03(d,J=2.8Hz,2H),5.85(d,J=2.0Hz,1H),5.53(d,J=4.4Hz,1H),4.04-4.57(m,7H),4.20(m,4H),3.81(s,3H)。 The obtained N4-p-methoxybenzoylcytidine triphosphate was detected, and the nuclear magnetic resonance data were: 1 H NMR (400 MHz, D 2 O) δ9.93 (s, 1H), 8.94 (d, J=0.8 Hz, 1H), 7.95 (d, J=2.4 Hz, 2H), 7.03 (d, J=2.8 Hz, 2H), 5.85 (d, J=2.0 Hz, 1H), 5.53 (d, J=4.4 Hz, 1H), 4.04-4.57 (m, 7H), 4.20 (m, 4H), 3.81 (s, 3H).
实施例11 N4-对硝基苯甲酰基胞苷三磷酸Example 11 N4-p-nitrobenzoylcytidine triphosphate
本实施例提供一种化学修饰胞苷三磷酸,所述化学修饰胞苷三磷酸为N4-对硝基苯甲酰基胞苷三磷酸。制备的各步骤的反应条件参照实施例1。步骤b中所使用的酰氯为对硝基苯甲酰氯(4-硝基苯甲酰氯)。This embodiment provides a chemically modified cytidine triphosphate, wherein the chemically modified cytidine triphosphate is N4-p-nitrobenzoyl cytidine triphosphate. The reaction conditions of each step of the preparation are referred to Example 1. The acyl chloride used in step b is p-nitrobenzoyl chloride (4-nitrobenzoyl chloride).
对所得N4-对硝基苯甲酰基胞苷三磷酸进行检测,核磁数据为:1H NMR(400MHz,D2O)δ9.91(s,1H),8.96(d,J=0.8Hz,1H),8.33(d,J=2.4Hz,2H),8.20(d,J=3.2Hz,2H),5.84(d,J=2.0Hz,1H),5.54(d,J=4.8Hz,1H),4.03-4.57(m,7H),4.21(m,4H),3.81(s,3H)。The obtained N4-p-nitrobenzoylcytidine triphosphate was detected, and the nuclear magnetic resonance data were: 1 H NMR (400 MHz, D 2 O) δ9.91 (s, 1H), 8.96 (d, J=0.8 Hz, 1H), 8.33 (d, J=2.4 Hz, 2H), 8.20 (d, J=3.2 Hz, 2H), 5.84 (d, J=2.0 Hz, 1H), 5.54 (d, J=4.8 Hz, 1H), 4.03-4.57 (m, 7H), 4.21 (m, 4H), 3.81 (s, 3H).
实施例12 5-甲基胞苷三磷酸Example 12 5-Methylcytidine triphosphate
本实施例提供一种化学修饰胞苷三磷酸,所述化学修饰胞苷三磷酸为5-甲基胞苷三磷酸。制备的各步骤的反应条件参照实施例1,区别在于以5-甲基胞苷为起始反应物,直接以步骤d的条件进行三磷酸化反应。This example provides a chemically modified cytidine triphosphate, wherein the chemically modified cytidine triphosphate is 5-methylcytidine triphosphate. The reaction conditions of each step of the preparation are referred to Example 1, except that 5-methylcytidine is used as the starting reactant and the triphosphorylation reaction is directly carried out under the conditions of step d.
图20为5-甲基胞苷三磷酸的1H NMR谱图,从图中可知,峰的位置(单位:ppm)依次为:1.91;4.15,4.16,4.18;4.23,4.25,4.26;4.36,4.37,4.38;5.91,5.92;7.73。图21为5-甲基胞苷三磷酸的31P NMR谱图,从图中可知,峰的位置(单位:ppm)依次为:-23.09,-22.66,-22.54,-22.41;-11.68,-11.56;-7.53,-6.49,-6.36。图36为5-甲基胞苷三磷酸的质谱图。Figure 20 is a 1 H NMR spectrum of 5-methylcytidine triphosphate. It can be seen from the figure that the peak positions (unit: ppm) are: 1.91; 4.15, 4.16, 4.18; 4.23, 4.25, 4.26; 4.36, 4.37, 4.38; 5.91, 5.92; 7.73. Figure 21 is a 31 P NMR spectrum of 5-methylcytidine triphosphate. It can be seen from the figure that the peak positions (unit: ppm) are: -23.09, -22.66, -22.54, -22.41; -11.68, -11.56; -7.53, -6.49, -6.36. Figure 36 is a mass spectrum of 5-methylcytidine triphosphate.
实施例13 5-甲基-N4-己酰基胞苷三磷酸Example 13 5-Methyl-N4-hexanoyl cytidine triphosphate
本实施例提供一种化学修饰胞苷三磷酸,所述化学修饰胞苷三磷酸为5-甲基-N4-己酰基胞苷三磷酸。制备的各步骤的反应条件参照实施例1,区别在于使用的反应起始物为5-甲基胞苷,步骤b中所使用的酸酐为己酸酐。This embodiment provides a chemically modified cytidine triphosphate, wherein the chemically modified cytidine triphosphate is 5-methyl-N4-hexanoyl cytidine triphosphate. The reaction conditions of each step of the preparation are referred to Example 1, except that the reaction starting material used is 5-methylcytidine, and the acid anhydride used in step b is hexanoic anhydride.
图22为5-甲基-N4-己酰基胞苷三磷酸的1H NMR谱图,从图中可知,峰的位置(单位:ppm)依次为:0.79,0.80,0.80,0.82;1.24,1.25,1.26;1.60,1.62;2.06,2.06;2.49,2.51,2.52;4.24,4.25,4.28,4.28,4.34,4.35,4.37;5.90,5.91;8.22。图23为5-甲基-N4-己酰基胞苷三磷酸的31P NMR谱图,从图中可知,峰的位置(单位:ppm)依次为:-22.68,-22.56,-22.43;-11.73,-11.61;-6.49,-6.36。图37为5-甲基-N4-己酰基胞苷三磷酸的质谱图。Figure 22 is the 1 H NMR spectrum of 5-methyl-N4-hexanoyl cytidine triphosphate. It can be seen from the figure that the peak positions (unit: ppm) are: 0.79, 0.80, 0.80, 0.82; 1.24, 1.25, 1.26; 1.60, 1.62; 2.06, 2.06; 2.49, 2.51, 2.52; 4.24, 4.25, 4.28, 4.28, 4.34, 4.35, 4.37; 5.90, 5.91; 8.22. Figure 23 is a 31 P NMR spectrum of 5-methyl-N4-hexanoyl cytidine triphosphate, from which it can be seen that the peak positions (unit: ppm) are: -22.68, -22.56, -22.43; -11.73, -11.61; -6.49, -6.36. Figure 37 is a mass spectrum of 5-methyl-N4-hexanoyl cytidine triphosphate.
实施例14 N4-辛酰基胞苷三磷酸Example 14 N4-octanoyl cytidine triphosphate
本实施例提供一种化学修饰胞苷三磷酸,所述化学修饰胞苷三磷酸为N4-辛酰基胞苷三磷酸,制备的各步骤的反应条件参照实施例1。步骤b中所使用的酸酐为辛酸酐。This example provides a chemically modified cytidine triphosphate, wherein the chemically modified cytidine triphosphate is N4-octanoyl cytidine triphosphate, and the reaction conditions of each step of the preparation are referred to Example 1. The anhydride used in step b is octanoic anhydride.
图24为N4-辛酰基胞苷三磷酸的1H NMR谱图,从图中可知,峰的位置(单位:ppm)依次为:0.74,0.76,0.78;1.23,1.32,1.34,1.56,1.57,1.59;2.39,2.41,2.43;4.24,4.25,4.26,4.28,4.29,4.30;5.89,5.90;7.24,7.26,7.27;8.32,8.34。Figure 24 is the 1 H NMR spectrum of N4-octanoylcytidine triphosphate. It can be seen from the figure that the peak positions (unit: ppm) are: 0.74, 0.76, 0.78; 1.23, 1.32, 1.34, 1.56, 1.57, 1.59; 2.39, 2.41, 2.43; 4.24, 4.25, 4.26, 4.28, 4.29, 4.30; 5.89, 5.90; 7.24, 7.26, 7.27; 8.32, 8.34.
实施例15 N4-癸酰基胞苷三磷酸Example 15 N4-decanoylcytidine triphosphate
本实施例提供一种化学修饰胞苷三磷酸,所述化学修饰胞苷三磷酸为N4-癸酰基胞苷三磷酸,制备的各步骤的反应条件参照实施例1。步骤b中所使用的酸酐为癸酸酐。This example provides a chemically modified cytidine triphosphate, wherein the chemically modified cytidine triphosphate is N4-decanoyl cytidine triphosphate, and the reaction conditions of each step of the preparation are as described in Example 1. The anhydride used in step b is decanoic anhydride.
图25为N4-癸酰基胞苷三磷酸的1H NMR谱图,从图中可知,峰的位置(单位:ppm)依次为:0.73,0.75,0.76;1.20,1.22;1.56,1.57,1.59;2.40,2.42,2.44;4.24,4.26,4.27,4.28,4.30;5.89,5.90;7.19,7.21;8.36,8.38。Figure 25 is the 1 H NMR spectrum of N4-decanoylcytidine triphosphate. It can be seen from the figure that the peak positions (unit: ppm) are: 0.73, 0.75, 0.76; 1.20, 1.22; 1.56, 1.57, 1.59; 2.40, 2.42, 2.44; 4.24, 4.26, 4.27, 4.28, 4.30; 5.89, 5.90; 7.19, 7.21; 8.36, 8.38.
实施例16 5-甲基-N4-苯甲酰基胞苷三磷酸Example 16 5-Methyl-N4-benzoylcytidine triphosphate
本实施例提供一种化学修饰胞苷三磷酸,所述化学修饰胞苷三磷酸为5-甲基-N4-苯甲酰 基胞苷三磷酸,制备的各步骤的反应条件参照实施例1,区别在于使用的反应起始物为5-甲基胞苷,步骤b中所使用的酸酐为苯甲酸酐。This embodiment provides a chemically modified cytidine triphosphate, wherein the chemically modified cytidine triphosphate is 5-methyl-N4-benzoyl The reaction conditions of each step of the preparation are referred to Example 1, except that the reaction starting material used is 5-methylcytidine, and the acid anhydride used in step b is benzoic anhydride.
图26为5-甲基-N4-苯甲酰基胞苷三磷酸的1H NMR谱图,从图中可知,峰的位置(单位:ppm)依次为:2.10;4.24,4.25,4.27,4.28,4.31,4.32,4.33,4.41,4.42,4.43;5.92,5.93;7.47,7.49,7.51,7.59,7.61,7.63;7.86,7.88;8.24。Figure 26 is the 1 H NMR spectrum of 5-methyl-N4-benzoylcytidine triphosphate. It can be seen from the figure that the peak positions (unit: ppm) are: 2.10; 4.24, 4.25, 4.27, 4.28, 4.31, 4.32, 4.33, 4.41, 4.42, 4.43; 5.92, 5.93; 7.47, 7.49, 7.51, 7.59, 7.61, 7.63; 7.86, 7.88; 8.24.
实施例17 2’-氟-N4-乙酰基胞苷三磷酸Example 17 2'-Fluoro-N4-acetylcytidine triphosphate
本实施例提供一种化学修饰胞苷三磷酸,所述化学修饰胞苷三磷酸为2’-氟-N4-乙酰基胞苷三磷酸,制备的各步骤的反应条件参照实施例1,区别在于使用的反应起始物为2’-氟代胞苷,步骤b中所使用的酸酐为乙酸酐。This embodiment provides a chemically modified cytidine triphosphate, wherein the chemically modified cytidine triphosphate is 2'-fluoro-N4-acetyl cytidine triphosphate, and the reaction conditions of each step of the preparation are referred to Example 1, except that the reaction starting material used is 2'-fluorocytidine, and the acid anhydride used in step b is acetic anhydride.
图27为2’-氟-N4-乙酰基胞苷三磷酸的1H NMR谱图,从图中可知,峰的位置(单位:ppm)依次为:2.14;4.22,4.24,4.25,4.28,4.30,4.33,4.36,4.40,4.41,4.42,4.44,4.47,4.48,4.49,4.50;4.98,4.99;5.11,5.12;5.98,6.02;7.23,7.25;8.35,8.37。Figure 27 is the 1 H NMR spectrum of 2'-fluoro-N4-acetylcytidine triphosphate. It can be seen from the figure that the peak positions (unit: ppm) are: 2.14; 4.22, 4.24, 4.25, 4.28, 4.30, 4.33, 4.36, 4.40, 4.41, 4.42, 4.44, 4.47, 4.48, 4.49, 4.50; 4.98, 4.99; 5.11, 5.12; 5.98, 6.02; 7.23, 7.25; 8.35, 8.37.
实施例18 2’-氟-N4-丁酰基胞苷三磷酸Example 18 2'-Fluoro-N4-butyryl cytidine triphosphate
本实施例提供一种化学修饰胞苷三磷酸,所述化学修饰胞苷三磷酸为2’-氟-N4-丁酰基胞苷三磷酸,制备的各步骤的反应条件参照实施例1,区别在于使用的反应起始物为2’-氟代胞苷,步骤b中所使用的酸酐为丁酸酐。This embodiment provides a chemically modified cytidine triphosphate, wherein the chemically modified cytidine triphosphate is 2'-fluoro-N4-butyryl cytidine triphosphate. The reaction conditions of each step of the preparation are referred to Example 1, except that the reaction starting material used is 2'-fluorocytidine, and the anhydride used in step b is butyric anhydride.
图28为2’-氟-N4-丁酰基胞苷三磷酸的1H NMR谱图,从图中可知,峰的位置(单位:ppm)依次为:0.83,0.84,0.86;1.55,1.56,1.58,1.60;2.36,2.38,2.40;4.22,4.24,4.27,4.28,4.30,4.33,4.36,4.40,4.41,4.43,4.43,4.47,4.48,4.49,4.50;4.98,4.99;5.11,5.12;5.98,6.02;7.25,7.27;8.34,8.36。Figure 28 is the 1 H NMR spectrum of 2'-fluoro-N4-butyryl cytidine triphosphate. It can be seen from the figure that the peak positions (unit: ppm) are: 0.83, 0.84, 0.86; 1.55, 1.56, 1.58, 1.60; 2.36, 2.38, 2.40; 4.22, 4.24, 4.27, 4.28, 4.30, 4.33, 4.36, 4.40, 4.41, 4.43, 4.43, 4.47, 4.48, 4.49, 4.50; 4.98, 4.99; 5.11, 5.12; 5.98, 6.02; 7.25, 7.27; 8.34, 8.36.
实施例19 2’-氟-N4-己酰基胞苷三磷酸Example 19 2'-Fluoro-N4-hexanoyl cytidine triphosphate
本实施例提供一种化学修饰胞苷三磷酸,所述化学修饰胞苷三磷酸为2’-氟-N4-己酰基胞苷三磷酸,制备的各步骤的反应条件参照实施例1,区别在于使用的反应起始物为2’-氟代胞苷,步骤b中所使用的酸酐为己酸酐。This embodiment provides a chemically modified cytidine triphosphate, wherein the chemically modified cytidine triphosphate is 2'-fluoro-N4-hexanoyl cytidine triphosphate. The reaction conditions of each step of the preparation are referred to Example 1, except that the reaction starting material used is 2'-fluorocytidine, and the acid anhydride used in step b is hexanoic anhydride.
图29为2’-氟-N4-己酰基胞苷三磷酸的1H NMR谱图,从图中可知,峰的位置(单位:ppm)依次为:0.76,0.77,0.79;1.22,1.22;1.54,1.56,1.58;2.38,2.40,2.41;4.22,4.25,4.27,4.34,4.37,4.39,4.40,4.41,4.42,4.46,4.47,4.48,4.49;4.98,4.99;5.11,5.12;5.98,6.02;7.26,7.27;8.33,8.36。Figure 29 is the 1 H NMR spectrum of 2'-fluoro-N4-hexanoyl cytidine triphosphate. It can be seen from the figure that the peak positions (unit: ppm) are: 0.76, 0.77, 0.79; 1.22, 1.22; 1.54, 1.56, 1.58; 2.38, 2.40, 2.41; 4.22, 4.25, 4.27, 4.34, 4.37, 4.39, 4.40, 4.41, 4.42, 4.46, 4.47, 4.48, 4.49; 4.98, 4.99; 5.11, 5.12; 5.98, 6.02; 7.26, 7.27; 8.33, 8.36.
实施例20 2’-甲氧基胞苷三磷酸Example 20 2'-Methoxycytidine triphosphate
本实施例提供一种化学修饰胞苷三磷酸,所述化学修饰胞苷三磷酸为2’-甲氧基胞苷三磷酸,制备的各步骤的反应条件参照实施例1,区别在于使用的反应起始物为2’-甲氧基胞苷。This example provides a chemically modified cytidine triphosphate, wherein the chemically modified cytidine triphosphate is 2'-methoxycytidine triphosphate. The reaction conditions of each step of the preparation are referred to Example 1, except that the reaction starting material used is 2'-methoxycytidine.
对所得2’-甲氧基胞苷三磷酸进行检测,核磁数据为:1H NMR(400MHz,D2O)δ7.96(d,J=0.8Hz,1H),6.05(d,J=0.8Hz,1H),5.96(d,J=1.2Hz,1H),5.88(d,J=3.6Hz,1H),4.72(t,1H),4.03-4.51(m,5H),3.40(s,3H)。The obtained 2'-methoxycytidine triphosphate was detected, and the nuclear magnetic resonance data were: 1 H NMR (400 MHz, D 2 O) δ7.96 (d, J=0.8 Hz, 1H), 6.05 (d, J=0.8 Hz, 1H), 5.96 (d, J=1.2 Hz, 1H), 5.88 (d, J=3.6 Hz, 1H), 4.72 (t, 1H), 4.03-4.51 (m, 5H), 3.40 (s, 3H).
实施例21 2’-乙氧基胞苷三磷酸Example 21 2'-ethoxycytidine triphosphate
本实施例提供一种化学修饰胞苷三磷酸,所述化学修饰胞苷三磷酸为2’-乙氧基胞苷三磷酸,制备的各步骤的反应条件参照实施例1,区别在于使用的反应起始物为2’-乙氧基胞苷。This example provides a chemically modified cytidine triphosphate, wherein the chemically modified cytidine triphosphate is 2'-ethoxycytidine triphosphate. The reaction conditions of each step of the preparation are referred to Example 1, except that the reaction starting material used is 2'-ethoxycytidine.
对所得2’-乙氧基胞苷三磷酸进行检测,核磁数据为:1H NMR(400MHz,D2O)δ7.97(d,J=0.8Hz,1H),6.67(s,2H),6.03(d,J=0.8Hz,1H),5.53(d,J=3.6Hz,1H),4.70(t,1H),4.02-4.52(m,5H),3.87(q,2H),1.19(t,3H)。 The obtained 2'-ethoxycytidine triphosphate was detected, and the nuclear magnetic resonance data were: 1 H NMR (400 MHz, D 2 O) δ7.97 (d, J=0.8 Hz, 1H), 6.67 (s, 2H), 6.03 (d, J=0.8 Hz, 1H), 5.53 (d, J=3.6 Hz, 1H), 4.70 (t, 1H), 4.02-4.52 (m, 5H), 3.87 (q, 2H), 1.19 (t, 3H).
实施例22 DNA模板制备Example 22 DNA template preparation
以编码荧光素酶的质粒为模板,利用DNA聚合酶在PCR小管中进行。具体步骤如下:The plasmid encoding luciferase is used as a template and DNA polymerase is used in a PCR tube. The specific steps are as follows:
PCR法制备荧光素酶DNA模板,在96孔PCR仪器中进行。The luciferase DNA template was prepared by PCR and carried out in a 96-well PCR instrument.
PCR产物中至少包括:A)一个启动子序列(T7启动子);B)包含至少一个Kozak序列的5’UTR;C)3’UTR;D)荧光素酶编码序列;E)聚A尾(polyA tail)。本实施例涉及的具体序列如表1所示。The PCR product includes at least: A) a promoter sequence (T7 promoter); B) a 5'UTR containing at least one Kozak sequence; C) a 3'UTR; D) a luciferase coding sequence; E) a poly A tail. The specific sequences involved in this embodiment are shown in Table 1.
表1
Table 1
按如下反应体系进行DNA模板的扩增:反应体积,50μL(为单个管的反应体积,一次同时反应多管),具体的反应体系如表2所示。The DNA template was amplified according to the following reaction system: reaction volume, 50 μL (reaction volume of a single tube, multiple tubes were reacted at one time), and the specific reaction system is shown in Table 2.
表2
Table 2
反应程序如下:预变性95℃,1分钟,变性95℃,15s,退火52℃,5s,延伸72℃,2分钟,共32个循环;最后延伸72℃,5分钟。The reaction program was as follows: pre-denaturation at 95°C for 1 min, denaturation at 95°C for 15 s, annealing at 52°C for 5 s, extension at 72°C for 2 min, for a total of 32 cycles; and final extension at 72°C for 5 min.
反应结束后将反应液合并于1.5mL管中。取2μL进行DNA琼脂糖凝胶电泳检测以确定反应成功(琼脂糖凝胶电泳检测条件:2%琼脂糖,5V/分钟,30分钟)。After the reaction is completed, the reaction solutions are combined in a 1.5 mL tube, and 2 μL is taken for DNA agarose gel electrophoresis to confirm the success of the reaction (agarose gel electrophoresis detection conditions: 2% agarose, 5 V/min, 30 min).
反应产物用HiPure Gel Pure DNA Mini Kit纯化。纯化过程如下:The reaction product was purified using HiPure Gel Pure DNA Mini Kit. The purification process is as follows:
(1)短暂离心PCR产物。(1) Briefly centrifuge the PCR product.
(2)加入等倍体积的GDP缓冲液,颠倒混匀。(2) Add an equal volume of GDP buffer and mix by inversion.
(3)将HiPure DNA柱子套在收集管中,把混合液转移至DNA柱子中,12000g离心30-60s。倒弃滤液,把柱子套回2mL离心管中。加入600μL缓冲液DW2(已用无水乙醇稀释)至柱子中,12000g离心30-60s。(3) Place the HiPure DNA column in a collection tube, transfer the mixture to the DNA column, and centrifuge at 12,000 g for 30-60 seconds. Discard the filtrate and place the column back in a 2 mL centrifuge tube. Add 600 μL of buffer DW2 (diluted with anhydrous ethanol) to the column and centrifuge at 12,000 g for 30-60 seconds.
(4)倒弃滤液,把柱子套回2mL离心管中。加入600μL缓冲液DW2(已用无水乙醇稀释)至柱子中,12000g离心30-60s。倒弃滤液,把柱子套回2mL离心管中,12000g离心2分钟。(4) Discard the filtrate and put the column back into a 2 mL centrifuge tube. Add 600 μL of buffer DW2 (diluted with anhydrous ethanol) to the column and centrifuge at 12,000 g for 30-60 seconds. Discard the filtrate and put the column back into a 2 mL centrifuge tube and centrifuge at 12,000 g for 2 minutes.
(5)把柱子套回1.5mL离心管中,加入15-30μL洗脱缓冲液至柱子膜中央,室温放置2分钟,12000g离心1分钟。丢弃柱子,用NanoDrop Spectrophotometer(超微量分光光度计)检测模板DNA的浓度定量,以及260/280、260/230的比值,然后将DNA保存于-20℃。(5) Put the column back into a 1.5 mL centrifuge tube, add 15-30 μL of elution buffer to the center of the column membrane, leave at room temperature for 2 minutes, and centrifuge at 12,000 g for 1 minute. Discard the column, use a NanoDrop Spectrophotometer to detect the concentration of the template DNA, as well as the ratios of 260/280 and 260/230, and then store the DNA at -20°C.
实施例23化学修饰核苷全局替代天然核苷的mRNA的体外转录合成Example 23 In vitro transcription synthesis of mRNA with chemically modified nucleosides replacing natural nucleosides
分别采用所制备的化学修饰的核苷全局替代天然核苷,合成化学修饰的mRNA。化学修饰胞苷三磷酸包含的修饰包括如下所示:N4-乙酰基,N4-丙酰基,N4-丁酰基,N4-戊酰基,N4-己酰基,N4-苯甲酰基,5-甲基-N4-乙酰基,5-甲基-N4-丁酰基,5-甲基-N4-己酰基,5-甲基。The prepared chemically modified nucleosides are used to globally replace natural nucleosides to synthesize chemically modified mRNA. The chemically modified cytidine triphosphate includes modifications as follows: N4-acetyl, N4-propionyl, N4-butyryl, N4-pentanoyl, N4-hexanoyl, N4-benzoyl, 5-methyl-N4-acetyl, 5-methyl-N4-butyryl, 5-methyl-N4-hexanoyl, 5-methyl.
化学修饰的mRNA的合成是利用T7 RNA聚合酶体外转录实现的,以50μL的反应体系为例,在0.2mL的PCR反应管中依次加入如表3所示的成分。The synthesis of chemically modified mRNA was achieved by in vitro transcription using T7 RNA polymerase. Taking a 50 μL reaction system as an example, the components listed in Table 3 were added sequentially into a 0.2 mL PCR reaction tube.
表3
Table 3
CTP*为胞嘧啶核苷三磷酸或化学修饰胞嘧啶核苷三磷酸。Cap Analogue#为市售帽结构类似物;在本实例中,采用的为New England Biolabs提供的3’-O-Me-m7-G(5’)ppp(5’)G RNA Cap Structure Analog,货号为S1411S。CTP* is cytosine triphosphate or chemically modified cytosine triphosphate. Cap Analogue# is a commercially available cap structure analog; in this example, the one used is 3'-O-Me-m 7 -G(5')ppp(5')G RNA Cap Structure Analog provided by New England Biolabs, with the catalog number S1411S.
反应程序如下:将恒温器的热盖启动,设置为60℃。点击恒温器反应系统,37℃,2-4h。孵育完成以后,mRNA纯化方法如下:The reaction procedure is as follows: Turn on the thermostat's hot cover and set it to 60°C. Click the thermostat reaction system, 37°C, 2-4h. After the incubation is completed, the mRNA purification method is as follows:
(1)在每个50μL反应体系中加入1μL DNA酶Ⅰ(RNase-free),37℃下孵育15分钟消化体系中的DNA模板。(1) Add 1 μL DNase I (RNase-free) to each 50 μL reaction system and incubate at 37°C for 15 minutes to digest the DNA template in the system.
(2)在每个50μL反应体系中加入2.5μL乙二胺四乙酸二钠盐溶液(500mM),65℃下孵育10分钟。(2) Add 2.5 μL of ethylenediaminetetraacetic acid disodium salt solution (500 mM) to each 50 μL reaction system and incubate at 65°C for 10 minutes.
(3)将纤维素溶液悬浮在洗脱溶液中,制备成0.2g纤维素/mL的洗脱液(配制方法参考文献:A Facile Method for the Removal of dsRNA Contaminants from In Vitro Transcribed mRNA,Molecular Therapy Nucleic Acids,2019,15,26-35)。将DNA吸附柱套在2mL接收管中,(广州美基生物,DNA/RNA小量吸附柱,货号为M021),然后取630μL悬浊液于DNA柱子中,涡旋震荡2h进行活化。(3) Suspend the cellulose solution in the elution solution to prepare an elution solution with a concentration of 0.2 g cellulose/mL (reference for preparation method: A Facile Method for the Removal of dsRNA Contaminants from In Vitro Transcribed mRNA, Molecular Therapy Nucleic Acids, 2019, 15, 26-35). Put the DNA adsorption column in a 2 mL receiving tube (Guangzhou Meiji Biotechnology, DNA/RNA small-volume adsorption column, item number M021), then take 630 μL of the suspension into the DNA column and vortex for 2 h for activation.
(4)将活化后的纤维素悬浊液在13000g离心1分钟,丢弃滤液,然后重新加入500μL洗脱液进行悬浊。然后将体外转录反应混合物溶液加入到DNA柱子中,涡旋震荡30分钟。(4) The activated cellulose suspension was centrifuged at 13,000 g for 1 minute, the filtrate was discarded, and 500 μL of the elution solution was added to resuspend. The in vitro transcription reaction mixture solution was then added to the DNA column and vortexed for 30 minutes.
(5)将DNA柱子在13000g下离心1分钟,收集滤液,加入到一根新的活化好的DNA柱子中(活化步骤如步骤3所述),涡旋震荡30分钟。(5) Centrifuge the DNA column at 13,000 g for 1 min, collect the filtrate, add it to a new activated DNA column (activation step as described in step 3), and vortex for 30 min.
(6)将DNA柱子在13000g下离心1分钟,收集滤液,然后将滤液转移至一个新的在冰面预冷的无核酸酶的1.5mL离心管中。(6) Centrifuge the DNA column at 13,000 g for 1 minute, collect the filtrate, and then transfer the filtrate to a new 1.5 mL nuclease-free centrifuge tube pre-chilled on ice.
(7)将无水异丙醇和3M的醋酸钠溶液提前在冰面预冷,然后在步骤(6)中的离心管加入500μL异丙醇和50μL醋酸钠,轻柔颠倒混合均匀,然后在4℃下14000g离心10分钟。(7) Pre-cool anhydrous isopropanol and 3 M sodium acetate solution on ice, then add 500 μL of isopropanol and 50 μL of sodium acetate to the centrifuge tube in step (6), gently invert to mix, and then centrifuge at 14,000 g for 10 minutes at 4°C.
(8)去除上清溶液,然后在离心管中加入300μL 4℃预冷的无水乙醇,然后在4℃下14000g离心10分钟。(8) Remove the supernatant solution, then add 300 μL of 4°C precooled anhydrous ethanol into the centrifuge tube, and then centrifuge at 14,000 g at 4°C for 10 minutes.
(9)去除上清溶液,然后将沉淀在室温下风干30分钟。(9) Remove the supernatant solution and air-dry the precipitate at room temperature for 30 minutes.
(10)将所得的mRNA产物溶解在20-50μL无核酶水中,用超微量分光光度计测定纯化后的mRNA浓度,并同时测定260/230、260/280的比值。(10) The obtained mRNA product was dissolved in 20-50 μL of nuclease-free water, and the concentration of the purified mRNA was measured using an ultra-micro spectrophotometer, and the ratios of 260/230 and 260/280 were also measured simultaneously.
(11)取500ng纯化后的mRNA,进行琼脂糖凝胶电泳检测其片段完整性。(11) Take 500 ng of purified mRNA and perform agarose gel electrophoresis to detect the integrity of its fragments.
(12)将mRNA溶液于-80℃下保存。(12) Store the mRNA solution at -80°C.
图38为化学修饰的C系列核苷三磷酸的体外转录结果示意图,图中,泳道1为天然胞嘧啶,泳道2为5-甲基胞嘧啶,泳道3为5-甲基-N4-乙酰基胞嘧啶,泳道4为5-甲基-N4-丁酰基胞嘧啶,泳道5为5-甲基-N4-己酰基,泳道6为5-甲基-N4-苯甲酰基胞嘧啶,泳道7为N4-乙酰基胞嘧啶,泳道8为N4-丙酰基胞嘧啶,泳道9为N4-丁酰基胞嘧啶,泳道10为N4-己酰基胞嘧啶的mRNA;泳道M为RNA分子量标准。Figure 38 is a schematic diagram of the in vitro transcription results of chemically modified C-series nucleoside triphosphates. In the figure, lane 1 is natural cytosine, lane 2 is 5-methylcytosine, lane 3 is 5-methyl-N4-acetylcytosine, lane 4 is 5-methyl-N4-butyrylcytosine, lane 5 is 5-methyl-N4-hexanoyl, lane 6 is 5-methyl-N4-benzoylcytosine, lane 7 is N4-acetylcytosine, lane 8 is N4-propionylcytosine, lane 9 is N4-butyrylcytosine, and lane 10 is the mRNA of N4-hexanoylcytosine; lane M is the RNA molecular weight standard.
若PCR法所得的DNA模板中不含有聚腺苷酸系列(polyA),则此处需有加尾反应,加尾反应的具体步骤如下:室温下在0.2mL PCR Tube管中配制如表7的反应体系;表4为加尾反应体系,总体积为20μL。 If the DNA template obtained by PCR does not contain polyadenylic acid series (polyA), a tailing reaction is required here. The specific steps of the tailing reaction are as follows: prepare the reaction system as shown in Table 7 in a 0.2mL PCR Tube at room temperature; Table 4 is the tailing reaction system, and the total volume is 20μL.
表4
Table 4
反应程序如下:设置恒温反应器,盖温为60℃,37℃下孵育30分钟。孵育完成,以上述描述的纤维素法纯化加尾后的mRNA,然后以琼脂糖凝胶电泳检测mRNA加尾长度及加尾后的mRNA片段完整性。The reaction procedure is as follows: set the thermostatic reactor with a lid temperature of 60°C and incubate at 37°C for 30 minutes. After the incubation is completed, the tailed mRNA is purified by the cellulose method described above, and then the mRNA tail length and the integrity of the tailed mRNA fragment are detected by agarose gel electrophoresis.
实施例24化学修饰核苷与N1-甲基假尿苷联用全局替代天然核苷的mRNA的体外转录合成Example 24 In vitro transcription synthesis of mRNA using chemically modified nucleosides and N1-methylpseudouridine to globally replace natural nucleosides
与实施例23采用相同的方法,区别在于在本实施例中,化学修饰胞嘧啶核苷三磷酸与N1-甲基假尿苷三磷酸按50/50的比例混合,全局替换天然的胞嘧啶核苷三磷酸,进行荧光素酶mRNA的体外转录合成。采用的组合包括为N4-乙酰基胞嘧啶三磷酸/N1-甲基假尿苷三磷酸=50/50,N4-丁酰基胞嘧啶三磷酸/N1-甲基假尿苷三磷酸=50/50,N4-己酰基胞嘧啶三磷酸/N1-甲基假尿苷三磷酸=50/50。The same method as in Example 23 was used, except that in this example, chemically modified cytosine triphosphate and N1-methyl pseudouridine triphosphate were mixed in a 50/50 ratio, and natural cytosine triphosphate was replaced globally to perform in vitro transcription synthesis of luciferase mRNA. The combinations used included N4-acetylcytosine triphosphate/N1-methyl pseudouridine triphosphate=50/50, N4-butyrylcytosine triphosphate/N1-methyl pseudouridine triphosphate=50/50, and N4-hexanoylcytosine triphosphate/N1-methyl pseudouridine triphosphate=50/50.
实施例25化学修饰的mRNA在细胞中的表达效率Example 25 Expression efficiency of chemically modified mRNA in cells
合成编码荧光素酶的含有单一种类胞苷修饰的mRNA或者单一种类修饰胞苷与N1甲基假尿苷联用修饰的mRNA,检测其在细胞中的表达效率;将其转染进细胞之后,翻译成目标蛋白荧光素酶,然后以荧光素为底物,利用翻译的荧光素酶氧化荧光素化学发光,利用酶标仪检测其发光强度,以此确定天然的和化学修饰的mRNA的翻译效率。本实施例分别将所述化学修饰的mRNA转染到HEK 293T细胞和HeLa细胞中,并进行发光强度检测。Synthesize mRNA containing a single type of cytidine modification or mRNA modified by a single type of modified cytidine combined with N1 methyl pseudouridine encoding luciferase, and detect its expression efficiency in cells; after transfecting it into cells, translate it into the target protein luciferase, and then use luciferin as a substrate to use the translated luciferase to oxidize luciferin chemiluminescence, and use a microplate reader to detect its luminescence intensity, so as to determine the translation efficiency of natural and chemically modified mRNA. In this example, the chemically modified mRNA is transfected into HEK 293T cells and HeLa cells, respectively, and the luminescence intensity is detected.
化学修饰的mRNA转染HEK 293T细胞的具体步骤如下:The specific steps for transfecting HEK 293T cells with chemically modified mRNA are as follows:
(1)细胞转染(1) Cell transfection
接种完人胚肾293T细胞(购自中国科学院细胞库)后约24h,观察24孔板内的细胞状态,汇合度在70%。在生物安全柜内,配制90%(体积百分含量)DMEM+10%(体积百分含量)FBS培养基。转染前30分钟弃掉孔板的培养基,每孔加入500μL新鲜培养基,即90%(体积百分含量)DMEM+10%(体积百分含量)FBS培养基。About 24 hours after inoculation of human embryonic kidney 293T cells (purchased from the Chinese Academy of Sciences Cell Bank), observe the cell status in the 24-well plate, and the confluence is 70%. In the biological safety cabinet, prepare 90% (volume percentage) DMEM + 10% (volume percentage) FBS culture medium. Discard the culture medium in the well plate 30 minutes before transfection, and add 500 μL of fresh culture medium, i.e. 90% (volume percentage) DMEM + 10% (volume percentage) FBS culture medium, to each well.
(2)配制转染体系(2) Preparation of transfection system
取50μL Opti-MEM,加入800ng mRNA,混合均匀。另外准备2μL Lipofectamine 2000与50μL Opti-MEM的混合溶液,室温孵育10分钟。然后将Lipofectamine 2000混合液加入到mRNA混合液中混合均匀,孵育20分钟。将混合后的溶液加入到孔板中。转染后6h换液,吸掉旧的培养基,每孔换为500μL新鲜培养基,即90%(体积百分含量)DMEM+10%(体积百分含量)FBS培养基。转染后24h/48h收获。吸掉旧的培养基,用1mL PBS清洗一遍。吸掉PBS,用200μL 1×Cell Lysis Buffer(Abcam,ab152163)将细胞消化5分钟,然后用移液枪将细胞吹打下来,收集于1.5mL离心管中,在4℃下14000g离心10分钟,然后取170μL上清溶液于一个新的1.5mL离心管中,用于荧光素发光检测。 Take 50μL Opti-MEM, add 800ng mRNA, and mix well. Also prepare a mixed solution of 2μL Lipofectamine 2000 and 50μL Opti-MEM, and incubate at room temperature for 10 minutes. Then add the Lipofectamine 2000 mixture to the mRNA mixture, mix well, and incubate for 20 minutes. Add the mixed solution to the well plate. Change the medium 6h after transfection, aspirate the old culture medium, and replace each well with 500μL fresh culture medium, that is, 90% (volume percentage) DMEM + 10% (volume percentage) FBS culture medium. Harvest 24h/48h after transfection. Aspirate the old culture medium and wash once with 1mL PBS. Aspirate the PBS, digest the cells with 200 μL 1× Cell Lysis Buffer (Abcam, ab152163) for 5 minutes, then blow down the cells with a pipette, collect them in a 1.5 mL centrifuge tube, centrifuge at 14000 g for 10 minutes at 4°C, and then take 170 μL of the supernatant solution in a new 1.5 mL centrifuge tube for luciferin luminescence detection.
酶标仪检测发光的实验步骤如下:The experimental steps for detecting luminescence with an ELISA reader are as follows:
在96孔板中加入50μL上述提取的含有荧光素酶的蛋白溶液,然后在室温下孵育10分钟,之后加入50μL Luciferase Assay Reagent(Promega,E1483),用移液枪吹打均匀,并且在5分钟之内在酶标仪上完成发光强度检测。每个样品设置两个复孔。Add 50 μL of the above-extracted protein solution containing luciferase to a 96-well plate, then incubate at room temperature for 10 minutes, then add 50 μL Luciferase Assay Reagent (Promega, E1483), pipette evenly, and complete the luminescence intensity detection on the microplate reader within 5 minutes. Set up two replicate wells for each sample.
图39为转录的mRNA转染HEK 293T细胞24h后的表达效率图,图中横坐标为样本类型,图中:Figure 39 is a graph showing the expression efficiency of transcribed mRNA transfected into HEK 293T cells 24 hours after transfection. The horizontal axis in the figure is the sample type. In the figure:
1表示以天然核苷三磷酸为底物转录的荧光素酶mRNA;1 indicates luciferase mRNA transcribed using natural nucleoside triphosphates as substrates;
2表示以N4-乙酰基胞苷三磷酸替代天然胞苷三磷酸为底物转录的荧光素酶mRNA;2 indicates luciferase mRNA transcribed using N4-acetylcytidine triphosphate instead of natural cytidine triphosphate as substrate;
3表示以N4-丙酰基胞苷三磷酸替代天然胞苷三磷酸为底物转录的荧光素酶mRNA;3 indicates luciferase mRNA transcribed using N4-propionyl cytidine triphosphate instead of natural cytidine triphosphate as substrate;
4表示以N4-丁酰基胞苷三磷酸替代天然胞苷三磷酸为底物转录的荧光素酶mRNA;4 indicates luciferase mRNA transcribed using N4-butyryl cytidine triphosphate instead of natural cytidine triphosphate as substrate;
5表示以N4-戊酰基胞苷三磷酸替代天然胞苷三磷酸为底物转录的荧光素酶mRNA;5 indicates luciferase mRNA transcribed using N4-pentanoyl cytidine triphosphate instead of natural cytidine triphosphate as substrate;
6表示以N4-己酰基胞苷三磷酸替代天然胞苷三磷酸为底物转录的荧光素酶mRNA;6 represents luciferase mRNA transcribed using N4-hexanoyl cytidine triphosphate instead of natural cytidine triphosphate as substrate;
7表示以N4-苯甲酰基胞苷三磷酸替代天然胞苷三磷酸为底物转录的荧光素酶mRNA。7 represents luciferase mRNA transcribed using N4-benzoylcytidine triphosphate instead of natural cytidine triphosphate as substrate.
从图39可知,N4-丙酰基胞嘧啶、N4-丁酰基胞嘧啶、N4-戊酰基胞嘧啶、N4-己酰基胞嘧啶全局替换的荧光素酶mRNA在HEK 293T细胞中的翻译效率均高于天然胞嘧啶转录的荧光素酶mRNA,其中N4-丁酰基胞嘧啶全局替换的荧光素酶mRNA的翻译效率可增强至2.91倍,证明化学修饰的胞嘧啶可以有效增强转录后的mRNA的翻译效率。As can be seen from Figure 39, the translation efficiency of luciferase mRNA with global replacement of N4-propionylcytosine, N4-butyrylcytosine, N4-pentanoylcytosine and N4-hexanoylcytosine in HEK 293T cells is higher than that of luciferase mRNA transcribed from natural cytosine, among which the translation efficiency of luciferase mRNA with global replacement of N4-butyrylcytosine can be enhanced to 2.91 times, proving that chemically modified cytosine can effectively enhance the translation efficiency of transcribed mRNA.
化学修饰的mRNA转染HeLa细胞的步骤参照转染HEK 293T细胞的方法进行。图41为转录的mRNA转染HEK 293T细胞24h后的表达效率图,图中:The steps of chemically modified mRNA transfection of HeLa cells refer to the method of transfecting HEK 293T cells. Figure 41 is the expression efficiency diagram of transcribed mRNA transfected HEK 293T cells 24 hours after transfection. In the figure:
1表示以天然核苷三磷酸为底物转录的荧光素酶mRNA;1 indicates luciferase mRNA transcribed using natural nucleoside triphosphates as substrates;
2表示以N4-乙酰基胞苷三磷酸替代天然胞苷三磷酸为底物转录的荧光素酶mRNA;2 indicates luciferase mRNA transcribed using N4-acetylcytidine triphosphate instead of natural cytidine triphosphate as substrate;
3表示以N4-丙酰基胞苷三磷酸替代天然胞苷三磷酸为底物转录的荧光素酶mRNA;3 indicates luciferase mRNA transcribed using N4-propionyl cytidine triphosphate instead of natural cytidine triphosphate as substrate;
4表示以N4-丁酰基胞苷三磷酸替代天然胞苷三磷酸为底物转录的荧光素酶mRNA;4 indicates luciferase mRNA transcribed using N4-butyryl cytidine triphosphate instead of natural cytidine triphosphate as substrate;
5表示以N4-戊酰基胞苷三磷酸替代天然胞苷三磷酸为底物转录的荧光素酶mRNA;5 indicates luciferase mRNA transcribed using N4-pentanoyl cytidine triphosphate instead of natural cytidine triphosphate as substrate;
6表示以N4-己酰基胞苷三磷酸替代天然胞苷三磷酸为底物转录的荧光素酶mRNA;6 represents luciferase mRNA transcribed using N4-hexanoyl cytidine triphosphate instead of natural cytidine triphosphate as substrate;
7表示以N4-苯甲酰基胞苷三磷酸替代天然胞苷三磷酸为底物转录的荧光素酶mRNA。7 represents luciferase mRNA transcribed using N4-benzoylcytidine triphosphate instead of natural cytidine triphosphate as substrate.
从图41可知,N4-丙酰基胞嘧啶、N4-丁酰基胞嘧啶、N4-戊酰基胞嘧啶、N4-己酰基胞嘧啶全局替换的荧光素酶mRNA在HeLa细胞中的翻译效率均高于天然胞嘧啶转录的荧光素酶mRNA,其中N4-丙酰基胞嘧啶、N4-丁酰基胞嘧啶和N4-戊酰基胞嘧啶全局替换的荧光素酶mRNA的翻译效率均可增强10倍以上,证明化学修饰的胞嘧啶可以有效增强转录后的mRNA的翻译效率。As can be seen from Figure 41, the translation efficiency of luciferase mRNA with global replacement of N4-propionylcytosine, N4-butyrylcytosine, N4-pentanoylcytosine and N4-hexanoylcytosine in HeLa cells is higher than that of luciferase mRNA transcribed from natural cytosine, among which the translation efficiency of luciferase mRNA with global replacement of N4-propionylcytosine, N4-butyrylcytosine and N4-pentanoylcytosine can be enhanced by more than 10 times, proving that chemically modified cytosine can effectively enhance the translation efficiency of transcribed mRNA.
图40为N1-甲基假尿苷和修饰胞苷联用转录的mRNA转染HEK 293T细胞24h后的表达效率图,图中:Figure 40 is a graph showing the expression efficiency of mRNA transcribed by N1-methyl pseudouridine and modified cytidine after transfection into HEK 293T cells for 24 hours. In the figure:
1表示以天然核苷三磷酸为底物转录的荧光素酶mRNA;1 indicates luciferase mRNA transcribed using natural nucleoside triphosphates as substrates;
2表示以N4-乙酰基胞苷三磷酸和N1-甲基假尿苷三磷酸替代天然胞苷三磷酸和尿苷三磷酸为底物转录的荧光素酶mRNA;2 indicates luciferase mRNA transcribed using N4-acetylcytidine triphosphate and N1-methylpseudouridine triphosphate instead of natural cytidine triphosphate and uridine triphosphate as substrates;
3表示以N4-丙酰基胞苷三磷酸和N1-甲基假尿苷三磷酸替代天然胞苷三磷酸和尿苷三磷酸为底物转录的荧光素酶mRNA;3 indicates luciferase mRNA transcribed using N4-propionylcytidine triphosphate and N1-methylpseudouridine triphosphate instead of natural cytidine triphosphate and uridine triphosphate as substrates;
4表示以N4-丁酰基胞苷三磷酸和N1-甲基假尿苷三磷酸替代天然胞苷三磷酸和尿苷三磷酸替代天然胞苷三磷酸为底物转录的荧光素酶mRNA; 4 represents luciferase mRNA transcribed with N4-butyrylcytidine triphosphate and N1-methylpseudouridine triphosphate instead of natural cytidine triphosphate and uridine triphosphate instead of natural cytidine triphosphate as substrates;
5表示以N4-戊酰基胞苷三磷酸和N1-甲基假尿苷三磷酸替代天然胞苷三磷酸和尿苷三磷酸替代天然胞苷三磷酸为底物转录的荧光素酶mRNA;5 indicates luciferase mRNA transcribed with N4-pentanoylcytidine triphosphate and N1-methylpseudouridine triphosphate instead of natural cytidine triphosphate and uridine triphosphate instead of natural cytidine triphosphate as substrates;
6表示以N4-己酰基胞苷三磷酸和N1-甲基假尿苷三磷酸替代天然胞苷三磷酸和尿苷三磷酸替代天然胞苷三磷酸为底物转录的荧光素酶mRNA;6 represents luciferase mRNA transcribed with N4-hexanoylcytidine triphosphate and N1-methylpseudouridine triphosphate instead of natural cytidine triphosphate and uridine triphosphate instead of natural cytidine triphosphate as substrates;
7表示以N1-甲基假尿苷三磷酸替代尿苷三磷酸为底物转录的荧光素酶mRNA。7 represents luciferase mRNA transcribed using N1-methylpseudouridine triphosphate instead of uridine triphosphate as substrate.
从图中可知,与天然胞嘧啶相比,N4-乙酰基胞苷三磷酸和N1-甲基假尿苷三磷酸、N4-丙酰基胞苷三磷酸和N1-甲基假尿苷三磷酸联用后,转录的荧光素酶mRNA在HEK 293T细胞中,有2.63-3.32倍的增强的翻译效率。单独的N1-甲基假尿苷三磷酸全局替换的mRNA能到1.87倍的增强的翻译效率。As can be seen from the figure, compared with natural cytosine, the translation efficiency of luciferase mRNA transcribed by combining N4-acetylcytidine triphosphate with N1-methyl pseudouridine triphosphate, N4-propionylcytidine triphosphate with N1-methyl pseudouridine triphosphate in HEK 293T cells was enhanced by 2.63-3.32 times. The mRNA with global replacement of N1-methyl pseudouridine triphosphate alone can achieve an enhanced translation efficiency of 1.87 times.
图42为N1-甲基假尿苷和修饰胞苷联用转录的mRNA转染HeLa细胞24h后的表达效率图,图中:FIG42 is a graph showing the expression efficiency of mRNA transcribed by N1-methyl pseudouridine and modified cytidine after transfection into HeLa cells for 24 hours, in which:
1表示以天然核苷三磷酸为底物转录的荧光素酶mRNA;1 indicates luciferase mRNA transcribed using natural nucleoside triphosphates as substrates;
2表示以N4-乙酰基胞苷三磷酸和N1-甲基假尿苷三磷酸替代天然胞苷三磷酸和尿苷三磷酸为底物转录的荧光素酶mRNA;2 indicates luciferase mRNA transcribed using N4-acetylcytidine triphosphate and N1-methylpseudouridine triphosphate instead of natural cytidine triphosphate and uridine triphosphate as substrates;
3表示以N4-丙酰基胞苷三磷酸和N1-甲基假尿苷三磷酸替代天然胞苷三磷酸和尿苷三磷酸为底物转录的荧光素酶mRNA;3 indicates luciferase mRNA transcribed using N4-propionylcytidine triphosphate and N1-methylpseudouridine triphosphate instead of natural cytidine triphosphate and uridine triphosphate as substrates;
4表示以N4-丁酰基胞苷三磷酸和N1-甲基假尿苷三磷酸替代天然胞苷三磷酸和尿苷三磷酸替代天然胞苷三磷酸为底物转录的荧光素酶mRNA;4 represents luciferase mRNA transcribed with N4-butyrylcytidine triphosphate and N1-methylpseudouridine triphosphate instead of natural cytidine triphosphate and uridine triphosphate instead of natural cytidine triphosphate as substrates;
5表示以N4-戊酰基胞苷三磷酸和N1-甲基假尿苷三磷酸替代天然胞苷三磷酸和尿苷三磷酸替代天然胞苷三磷酸为底物转录的荧光素酶mRNA;5 indicates luciferase mRNA transcribed with N4-pentanoylcytidine triphosphate and N1-methylpseudouridine triphosphate instead of natural cytidine triphosphate and uridine triphosphate instead of natural cytidine triphosphate as substrates;
6表示以N4-己酰基胞苷三磷酸和N1-甲基假尿苷三磷酸替代天然胞苷三磷酸和尿苷三磷酸替代天然胞苷三磷酸为底物转录的荧光素酶mRNA;6 represents luciferase mRNA transcribed with N4-hexanoylcytidine triphosphate and N1-methylpseudouridine triphosphate instead of natural cytidine triphosphate and uridine triphosphate instead of natural cytidine triphosphate as substrates;
7表示以N1-甲基假尿苷三磷酸替代尿苷三磷酸为底物转录的荧光素酶mRNA。7 represents luciferase mRNA transcribed using N1-methylpseudouridine triphosphate instead of uridine triphosphate as substrate.
从图中可知,与天然胞嘧啶相比,N4-乙酰基胞苷三磷酸、N4-丙酰基胞苷三磷酸、N4-丁酰基胞苷三磷酸、N4-戊酰基胞苷三磷酸与N1-甲基假尿苷联用后,转录的荧光素酶mRNA在HeLa细胞中,有1.66到10.82倍的增强的翻译效率。单独的N1-甲基假尿苷全局替换的mRNA的翻译效率增强9.5倍。As can be seen from the figure, compared with natural cytosine, the translation efficiency of luciferase mRNA transcribed by N4-acetylcytidine triphosphate, N4-propionylcytidine triphosphate, N4-butyrylcytidine triphosphate, and N4-pentanoylcytidine triphosphate in HeLa cells was enhanced by 1.66 to 10.82 times after the combination of N4-acetylcytidine triphosphate, N4-propionylcytidine triphosphate, N4-butyrylcytidine triphosphate and N1-methyl pseudouridine. The translation efficiency of mRNA replaced by N1-methyl pseudouridine alone was enhanced by 9.5 times.
实施例26化学修饰的mRNA在细胞中的稳定性Example 26 Stability of chemically modified mRNA in cells
合成编码荧光素酶的含有单一种类胞苷修饰的mRNA,检测其在细胞中的稳定性;其稳定性是指将mRNA转染进细胞一定时间以后,提取的残余的完整mRNA的相对量。An mRNA encoding luciferase containing a single type of cytidine modification was synthesized and its stability in cells was tested; its stability refers to the relative amount of residual intact mRNA extracted after the mRNA was transfected into the cells for a certain period of time.
本实施例分别将所述化学修饰的mRNA转染到HEK 293T细胞和HeLa细胞中,并进行qPCR检测。化学修饰的mRNA转染HEK 293T细胞并进行qPCR检测的具体步骤如下:In this example, the chemically modified mRNA was transfected into HEK 293T cells and HeLa cells, and qPCR detection was performed. The specific steps of chemically modified mRNA transfection into HEK 293T cells and qPCR detection are as follows:
细胞接种及细胞转染步骤参照实施例25中所述。在转染后3h,倒弃培养液,用1×PBS漂洗细胞一次,然后吸干PBS洗液。加入200μL TransZol Up裂解液(全式金生物,ET1111),水平放置片刻,使裂解液均匀分布于细胞表面并裂解细胞,然后用移液枪吹打细胞使其脱落。之后将裂解液转移至离心管中,用移液枪反复吹打,直至裂解物中无明显沉淀,室温静置5分钟。加入40μL氯仿,剧烈震荡30秒,然后室温孵育3分钟。在4℃下10000g离心15分钟,此时样品分为3层,上层为无色的水相,中间层和下层为粉红色有机相。此时RNA在水相中,用移液枪抽取90μL无色的水相,转移至新的离心管中,加入90μL无水乙醇,轻轻颠倒混合均匀。将得到的沉淀和溶液一起转移至离心柱中,室温下12000g离心30秒,弃流 出液。加入100μL CB9,室温下12000g离心30秒,弃流出液,并重复一次。加入100μL WB9,室温下12000g离心30秒,弃流出液,并重复一次。然后室温下12000g离心2分钟,彻底去除残留的无水乙醇。将离心柱放入无核酸酶管中,加入20μL无核酸酶水于离心柱的中央,室温静置1分钟。然后室温12000g离心1分钟,洗脱RNA。得到的总RNA用NanoDrop定量。之后取1μg总RNA,以Hifair 1st Strand cDNA Synthesis Kit(gDNA digester plus)(翊圣生物,11139ES10)合成cDNA。得到的cDNA以NanoDrop定量,取500ng cDNA,然后以Hieff UNICON Universal Blue qPCR SYBR Green Master Mix(翊圣生物,11184ES08)进行荧光定量扩增。所用荧光定量PCR仪器为qTower。mRNA的稳定性是以得到的Ct值按2-Δ法计算其相对值来表示。Cell inoculation and cell transfection steps are described in Example 25. 3h after transfection, the culture medium was discarded, the cells were rinsed once with 1×PBS, and then the PBS washing solution was aspirated. 200μL TransZol Up lysis solution (Quanshi Jin Bio, ET1111) was added, and it was placed horizontally for a moment to evenly distribute the lysis solution on the cell surface and lyse the cells, and then the cells were blown off with a pipette. After that, the lysis solution was transferred to a centrifuge tube, and it was blown repeatedly with a pipette until there was no obvious precipitation in the lysate, and it was left to stand at room temperature for 5 minutes. 40μL chloroform was added, and it was shaken violently for 30 seconds, and then incubated at room temperature for 3 minutes. Centrifuged at 10000g for 15 minutes at 4°C, at which time the sample was divided into 3 layers, the upper layer was a colorless aqueous phase, and the middle layer and the lower layer were pink organic phases. At this time, RNA was in the aqueous phase, and 90μL of colorless aqueous phase was extracted with a pipette, transferred to a new centrifuge tube, 90μL of anhydrous ethanol was added, and gently inverted to mix evenly. The obtained precipitate and solution were transferred to the centrifuge column and centrifuged at 12000g for 30 seconds at room temperature. Elute. Add 100 μL CB9, centrifuge at 12000g for 30 seconds at room temperature, discard the effluent, and repeat once. Add 100 μL WB9, centrifuge at 12000g for 30 seconds at room temperature, discard the effluent, and repeat once. Then centrifuge at 12000g for 2 minutes at room temperature to completely remove the residual anhydrous ethanol. Place the centrifuge column in a nuclease-free tube, add 20 μL nuclease-free water to the center of the centrifuge column, and let it stand at room temperature for 1 minute. Then centrifuge at 12000g for 1 minute at room temperature to elute the RNA. The total RNA obtained was quantified using NanoDrop. Then take 1 μg of total RNA and synthesize cDNA using Hifair 1st Strand cDNA Synthesis Kit (gDNA digester plus) (Yishen Bio, 11139ES10). The obtained cDNA was quantified by NanoDrop, and 500 ng of cDNA was taken and then fluorescent quantitative amplification was performed using Hieff UNICON Universal Blue qPCR SYBR Green Master Mix (Yisheng Bio, 11184ES08). The fluorescent quantitative PCR instrument used was qTower. The stability of mRNA was expressed by calculating the relative value of the obtained Ct value using the 2 -Δ method.
图43为化学修饰的mRNA在HEK 293T细胞中的稳定性的qPCR结果图,图中:FIG43 is a graph showing the qPCR results of the stability of chemically modified mRNA in HEK 293T cells, in which:
1表示以天然核苷三磷酸为底物转录的荧光素酶mRNA;1 indicates luciferase mRNA transcribed using natural nucleoside triphosphates as substrates;
2表示以N4-乙酰基胞苷三磷酸替代天然胞苷三磷酸为底物转录的荧光素酶mRNA;2 indicates luciferase mRNA transcribed using N4-acetylcytidine triphosphate instead of natural cytidine triphosphate as substrate;
3表示以N4-丙酰基胞苷三磷酸替代天然胞苷三磷酸为底物转录的荧光素酶mRNA;3 indicates luciferase mRNA transcribed using N4-propionyl cytidine triphosphate instead of natural cytidine triphosphate as substrate;
4表示以N4-丁酰基胞苷三磷酸替代天然胞苷三磷酸为底物转录的荧光素酶mRNA;4 indicates luciferase mRNA transcribed using N4-butyryl cytidine triphosphate instead of natural cytidine triphosphate as substrate;
5表示以N4-戊酰基胞苷三磷酸替代天然胞苷三磷酸为底物转录的荧光素酶mRNA;5 indicates luciferase mRNA transcribed using N4-pentanoyl cytidine triphosphate instead of natural cytidine triphosphate as substrate;
6表示以N4-己酰基胞苷三磷酸替代天然胞苷三磷酸为底物转录的荧光素酶mRNA;6 represents luciferase mRNA transcribed using N4-hexanoyl cytidine triphosphate instead of natural cytidine triphosphate as substrate;
7表示以N4-苯甲酰基胞苷三磷酸替代天然胞苷三磷酸为底物转录的荧光素酶mRNA。7 represents luciferase mRNA transcribed using N4-benzoylcytidine triphosphate instead of natural cytidine triphosphate as substrate.
从图43可知,N4-丙酰基胞嘧啶、N4-丁酰基胞嘧啶、N4-戊酰基胞嘧啶、N4-己酰基胞嘧啶全局替换的荧光素酶mRNA在HEK 293T细胞中的稳定性均高于天然胞嘧啶转录的荧光素酶mRNA,证明了化学修饰的胞嘧啶全局替换后可增强转录的mRNA在细胞内的稳定性。As can be seen from Figure 43, the stability of luciferase mRNA with global replacement of N4-propionylcytosine, N4-butyrylcytosine, N4-pentanoylcytosine, and N4-hexanoylcytosine in HEK 293T cells is higher than that of luciferase mRNA transcribed from natural cytosine, proving that global replacement of chemically modified cytosine can enhance the stability of transcribed mRNA in cells.
化学修饰的mRNA转染HeLa细胞并进行qPCR检测的步骤参照转染HEK 293T细胞的方法进行。图44为化学修饰的mRNA在HeLa细胞中的稳定性测试的qPCR结果图,图中:The steps of chemically modified mRNA transfection into HeLa cells and qPCR detection are carried out in accordance with the method of transfecting HEK 293T cells. Figure 44 is a graph showing the qPCR results of the stability test of chemically modified mRNA in HeLa cells, in which:
1表示以天然核苷三磷酸为底物转录的荧光素酶mRNA;1 indicates luciferase mRNA transcribed using natural nucleoside triphosphates as substrates;
2表示以N4-乙酰基胞苷三磷酸替代天然胞苷三磷酸为底物转录的荧光素酶mRNA;2 indicates luciferase mRNA transcribed using N4-acetylcytidine triphosphate instead of natural cytidine triphosphate as substrate;
3表示以N4-丙酰基胞苷三磷酸替代天然胞苷三磷酸为底物转录的荧光素酶mRNA;3 indicates luciferase mRNA transcribed using N4-propionyl cytidine triphosphate instead of natural cytidine triphosphate as substrate;
4表示以N4-丁酰基胞苷三磷酸替代天然胞苷三磷酸为底物转录的荧光素酶mRNA;4 indicates luciferase mRNA transcribed using N4-butyryl cytidine triphosphate instead of natural cytidine triphosphate as substrate;
5表示以N4-戊酰基胞苷三磷酸替代天然胞苷三磷酸为底物转录的荧光素酶mRNA;5 indicates luciferase mRNA transcribed using N4-pentanoyl cytidine triphosphate instead of natural cytidine triphosphate as substrate;
6表示以N4-己酰基胞苷三磷酸替代天然胞苷三磷酸为底物转录的荧光素酶mRNA。6 represents luciferase mRNA transcribed using N4-hexanoyl cytidine triphosphate instead of natural cytidine triphosphate as a substrate.
从图44可知,N4-丙酰基胞嘧啶和N4-戊酰基胞嘧啶、N4-己酰基胞嘧啶全局替换的荧光素酶mRNA在HEK 293T细胞中的稳定性均高于天然胞嘧啶转录的荧光素酶mRNA,证明了化学修饰的胞嘧啶全局替换后可增强转录的mRNA在细胞内的稳定性。As can be seen from Figure 44, the stability of luciferase mRNA with global replacement of N4-propionylcytosine, N4-pentanoylcytosine and N4-hexanoylcytosine in HEK 293T cells is higher than that of luciferase mRNA transcribed from natural cytosine, proving that global replacement of chemically modified cytosine can enhance the stability of transcribed mRNA in cells.
实施例27化学修饰的mRNA在细胞中的免疫原性Example 27 Immunogenicity of chemically modified mRNA in cells
合成编码荧光素酶的含有单一种类胞苷修饰的mRNA,检测其免疫原性。将mRNA转染进巨噬细胞RAW264.7,然后在转染后24h,检测RAW264.7细胞产生的免疫因子包括肿瘤坏死因子α和白细胞介素6。具体步骤如下:Synthesize mRNA encoding luciferase containing a single type of cytidine modification and test its immunogenicity. Transfect the mRNA into macrophages RAW264.7, and then detect the immune factors produced by RAW264.7 cells 24 hours after transfection, including tumor necrosis factor α and interleukin 6. The specific steps are as follows:
细胞接种及细胞转染步骤参照实施例25中所述。在本实施例中所用的细胞为巨噬细胞RAW264.7。在细胞转染后24h,用200μL 1×Cell Lysis Buffer(Abcam,ab152163)将细胞消化5分钟,然后用移液枪将细胞吹打下来,收集于1.5mL离心管中,在4℃下14000g离心10分钟,然后取170μL上清溶液于一个新的1.5mL离心管中。取50μL提取的蛋白溶液,加入100μL BCA protein quantification kit solution(翊圣生物,20201ES76),然后用移液枪吹 打均匀,在30℃下孵育30分钟,然后用Epoch Microplate Reader(酶标仪)检测其在562nm处的吸光度。根据标准曲线计算提取的总蛋白浓度。荧光素酶mRMA的免疫原性以其刺激RAW264.7细胞后分泌的细胞因子,包括肿瘤坏死因子α和白细胞介素6的量来表达。The cell inoculation and cell transfection steps refer to those described in Example 25. The cells used in this example are macrophages RAW264.7. 24 hours after cell transfection, digest the cells with 200 μL 1×Cell Lysis Buffer (Abcam, ab152163) for 5 minutes, then blow down the cells with a pipette and collect them in a 1.5 mL centrifuge tube. Centrifuge at 14000g for 10 minutes at 4°C, and then take 170 μL of the supernatant solution in a new 1.5 mL centrifuge tube. Take 50 μL of the extracted protein solution, add 100 μL of BCA protein quantification kit solution (Yishen Bio, 20201ES76), and then blow down with a pipette. Beat evenly, incubate at 30°C for 30 minutes, and then detect its absorbance at 562nm using Epoch Microplate Reader. Calculate the total protein concentration extracted according to the standard curve. The immunogenicity of luciferase mRMA is expressed by the amount of cytokines secreted by it after stimulating RAW264.7 cells, including tumor necrosis factor α and interleukin 6.
细胞因子的量以蛋白印迹法来定量,其具体步骤如下:取20μg总蛋白样品,加入1/5体积的6×Protein Loading Buffer(全式金生物,DL101),然后在95℃煮10分钟,然后在冰面冷却至4℃。短暂离心,然后将全部样品以12% SDS-PAGE胶进行分离。之后将样品转移至PVDF膜上,然后用5%的脱脂牛奶在室温下将膜封闭2h,然后用1×的TBST Buffer将膜漂洗3次,每次20mL。之后用1:3000稀释的对应一抗(一抗分别是目标蛋白的一抗和内参蛋白的一抗,故为两种,分别包括:重组Anti-TNF alpha抗体,EPR19147,Abcam和Antib-beta Actin抗体,AC-15,Abcam)在4℃孵育过夜。之后用1×的TBST Buffer将膜漂洗3次,每次20mL。之后用1:5000稀释的对应二抗(Goat Anti-Rabbit IgG H&L HRP,ab6721,Abcam和Goat Anti-Mouse IgG H&L HRP,ab6728,Abcam)孵育1h,然后用1×的TBST Buffer将膜漂洗3次,每次10mL。然后配制增强型ECL化学发光曝光试剂(翊圣生物,36222ES60),进行化学曝光。产生的细胞因子的相对含量以提取的条带的强度表达。The amount of cytokines was quantified by western blotting, and the specific steps were as follows: 20 μg of total protein sample was taken, 1/5 volume of 6× Protein Loading Buffer (All-Gold Bio, DL101) was added, and then boiled at 95°C for 10 minutes, and then cooled to 4°C on ice. After a brief centrifugation, all samples were separated by 12% SDS-PAGE gel. The sample was then transferred to a PVDF membrane, and the membrane was blocked with 5% skim milk at room temperature for 2 hours, and then rinsed with 1× TBST Buffer 3 times, 20 mL each time. Then, the corresponding primary antibodies (the primary antibodies are the primary antibodies of the target protein and the primary antibodies of the internal reference protein, so there are two types, including: recombinant Anti-TNF alpha antibody, EPR19147, Abcam and Antib-beta Actin antibody, AC-15, Abcam) diluted 1:3000 were incubated at 4°C overnight. Then, the membrane was rinsed 3 times with 1× TBST Buffer, 20 mL each time. After that, the membrane was incubated with the corresponding secondary antibody (Goat Anti-Rabbit IgG H&L HRP, ab6721, Abcam and Goat Anti-Mouse IgG H&L HRP, ab6728, Abcam) diluted 1:5000 for 1 hour, and then the membrane was rinsed 3 times with 1× TBST Buffer, 10 mL each time. Then, the enhanced ECL chemiluminescence exposure reagent (Yisheng Bio, 36222ES60) was prepared for chemical exposure. The relative content of the produced cytokines was expressed as the intensity of the extracted bands.
图45为化学修饰的mRNA在RAW264.7 cell中的免疫原性测试的蛋白印迹结果图,图中:Figure 45 is a Western blot result of the immunogenicity test of chemically modified mRNA in RAW264.7 cells, in which:
1表示缓冲溶液PBS;1 represents buffer solution PBS;
2表示以天然核苷三磷酸为底物转录的荧光素酶mRNA;2 indicates luciferase mRNA transcribed using natural nucleoside triphosphates as substrates;
3表示以N4-乙酰基胞苷三磷酸替代天然胞苷三磷酸为底物转录的荧光素酶mRNA;3 indicates luciferase mRNA transcribed using N4-acetylcytidine triphosphate instead of natural cytidine triphosphate as substrate;
4表示以N4-丙酰基胞苷三磷酸替代天然胞苷三磷酸为底物转录的荧光素酶mRNA;4 indicates luciferase mRNA transcribed using N4-propionylcytidine triphosphate instead of natural cytidine triphosphate as substrate;
5表示以N4-丁酰基胞苷三磷酸替代天然胞苷三磷酸为底物转录的荧光素酶mRNA;5 indicates luciferase mRNA transcribed using N4-butyryl cytidine triphosphate instead of natural cytidine triphosphate as substrate;
6表示以N4-戊酰基胞苷三磷酸替代天然胞苷三磷酸为底物转录的荧光素酶mRNA;6 indicates luciferase mRNA transcribed using N4-pentanoyl cytidine triphosphate instead of natural cytidine triphosphate as substrate;
7表示以N4-己酰基胞苷三磷酸替代天然胞苷三磷酸为底物转录的荧光素酶mRNA;7 indicates luciferase mRNA transcribed using N4-hexanoyl cytidine triphosphate instead of natural cytidine triphosphate as substrate;
8表示以N4-苯甲酰基胞苷三磷酸替代天然胞苷三磷酸为底物转录的荧光素酶mRNA。8 represents luciferase mRNA transcribed using N4-benzoylcytidine triphosphate instead of natural cytidine triphosphate as substrate.
从图中可知,与天然胞嘧啶转录的荧光素酶mRNA相比,化学修饰的胞嘧啶全局替换的荧光素酶mRNA在RAW264.7细胞中,没有显示出减弱的免疫原性。As can be seen from the figure, compared with the luciferase mRNA transcribed from natural cytosine, the luciferase mRNA with global replacement of chemically modified cytosine did not show weakened immunogenicity in RAW264.7 cells.
实施例28化学修饰腺苷三磷酸的合成Example 28 Synthesis of chemically modified adenosine triphosphate
本实施例提供一种化学修饰腺苷三磷酸的制备方法,制备技术路线如下所示,在本实施例中,所述化学修饰腺苷三磷酸的R5’为氢,Rn为羟基,M为氮。并采用核磁共振和液相色谱联用的质谱对化合物结构进行表征。
This embodiment provides a method for preparing chemically modified adenosine triphosphate, and the preparation technology route is as follows. In this embodiment, R 5' of the chemically modified adenosine triphosphate is hydrogen, R n is hydroxyl, and M is nitrogen. The structure of the compound is characterized by nuclear magnetic resonance and liquid chromatography-mass spectrometry.
a.取3mmol(1eq)腺苷以15mL N,N-二甲基甲酰胺溶解,然后依次加入12mmol(4eq)叔丁基二甲基氯硅烷和15mmol(5eq)咪唑,25℃下搅拌过夜;反应结束后,以10mL乙酸乙酯萃取反应液,重复3次,水洗有机相,重复3次,收集有机相,无水硫酸钠干燥,过滤,浓缩,硅胶柱色谱纯化(二氯甲烷/甲醇=100/1),得到白色固体化合物5(即式18化合物);化合物5的结构由1H NMR、13C NMR和HRMS确认。a. 3 mmol (1 eq) of adenosine was dissolved in 15 mL of N,N-dimethylformamide, and then 12 mmol (4 eq) of tert-butyldimethylsilyl chloride and 15 mmol (5 eq) of imidazole were added in sequence, and stirred at 25°C overnight; after the reaction, the reaction solution was extracted with 10 mL of ethyl acetate, and the extraction was repeated 3 times. The organic phase was washed with water, and the extraction was repeated 3 times. The organic phase was collected, dried over anhydrous sodium sulfate, filtered, concentrated, and purified by silica gel column chromatography (dichloromethane/methanol = 100/1) to obtain a white solid compound 5 (i.e., the compound of formula 18); the structure of compound 5 was confirmed by 1 H NMR, 13 C NMR and HRMS.
b.以15mL无水吡啶溶解12mmol(1eq)化合物5,然后滴加1.2eq相应酰氯,之后加入10mg 4-二甲氨基吡啶,25℃下搅拌过夜反应;反应结束后以10mL乙酸乙酯萃取反应液,重复3次,水洗有机相,重复3次,收集有机相,无水硫酸钠干燥,过滤,浓缩,硅胶柱色谱纯化(二氯甲烷/甲醇=200/1),得到白色泡沫状固体化合物6(即式19化合物);化合物6的结构由1H NMR、13C NMR和HRMS确认。b. 12 mmol (1 eq) of compound 5 was dissolved in 15 mL of anhydrous pyridine, and then 1.2 eq of the corresponding acid chloride was added dropwise, followed by 10 mg of 4-dimethylaminopyridine. The mixture was stirred at 25°C overnight. After the reaction, the reaction solution was extracted with 10 mL of ethyl acetate, and the extraction was repeated 3 times. The organic phase was washed with water, and the extraction was repeated 3 times. The organic phase was collected, dried over anhydrous sodium sulfate, filtered, concentrated, and purified by silica gel column chromatography (dichloromethane/methanol = 200/1) to obtain a white foamy solid compound 6 (i.e., compound 19). The structure of compound 6 was confirmed by 1 H NMR, 13 C NMR and HRMS.
c.以15mL无水四氢呋喃溶解1.5mmol(1eq)化合物6,然后滴加4.5mL(3eq)四丁基氟化铵溶液(1M四氢呋喃溶液),室温下搅拌反应0.5h,然后减压浓缩,硅胶柱色谱纯化(二氯甲烷/甲醇=20/1至10/1),得到白色固体化合物7(即式20化合物)。化合物7的结构由1H NMR、13C NMR和HRMS确认。c. Dissolve 1.5 mmol (1 eq) of compound 6 in 15 mL of anhydrous tetrahydrofuran, then dropwise add 4.5 mL (3 eq) of tetrabutylammonium fluoride solution (1 M tetrahydrofuran solution), stir at room temperature for 0.5 h, then concentrate under reduced pressure, and purify by silica gel column chromatography (dichloromethane/methanol=20/1 to 10/1) to obtain white solid compound 7 (i.e., compound of formula 20). The structure of compound 7 was confirmed by 1 H NMR, 13 C NMR and HRMS.
d.以2mL无水吡啶溶解0.1mmol化合物7,并减压共蒸除水3次;在氮气保护下,注入1mL干燥磷酸三甲酯,待固体完全溶解后将试管置于冰浴中冷却2min,并用注射器滴加1.5eq三氯氧磷;冰浴下搅拌反应2.5h。d. Dissolve 0.1 mmol of compound 7 in 2 mL of anhydrous pyridine and remove water under reduced pressure three times; inject 1 mL of dry trimethyl phosphate under nitrogen protection, and after the solid is completely dissolved, place the test tube in an ice bath for 2 min, and add 1.5 eq of phosphorus oxychloride dropwise with a syringe; stir and react in an ice bath for 2.5 h.
e.之后在冰浴条件下依次注入1.5eq干燥三丁胺,1.5mL(8eq)三丁基焦磷酸铵溶液(0.5M N,N-二甲基甲酰胺溶液),移至室温下反应0.5h。e. Then, inject 1.5 eq of dry tributylamine and 1.5 mL (8 eq) of tributylammonium pyrophosphate solution (0.5 M N,N-dimethylformamide solution) in sequence under ice bath conditions, and move to room temperature to react for 0.5 h.
f.反应完成后,冰浴下以5mL 2.0M饱和三乙胺-碳酸水溶液淬灭反应,然后以10mL二氯甲烷萃取反应液,收集水相,重复4次;以快速制备液相色谱仪C18柱分离纯化收集的水相,并以50mM饱和三乙胺-碳酸水溶液和色谱级乙腈为流动相;最后将收集纯化后的溶 液冻干,得到白色固体化合物8(即式23化合物)。化合物8的结构已由1H NMR、31P NMR、13C NMR和HRMS确认。f. After the reaction was completed, the reaction was quenched with 5 mL of 2.0 M saturated triethylamine-carbonic acid aqueous solution under ice bath, and then the reaction solution was extracted with 10 mL of dichloromethane, and the aqueous phase was collected, and repeated 4 times; the collected aqueous phase was separated and purified by rapid preparative liquid chromatography C18 column, and 50 mM saturated triethylamine-carbonic acid aqueous solution and chromatographic grade acetonitrile were used as mobile phases; finally, the collected purified solution was The solution was lyophilized to obtain a white solid compound 8 (ie, the compound of formula 23). The structure of compound 8 was confirmed by 1 H NMR, 31 P NMR, 13 C NMR and HRMS.
本实施例中化学修饰的腺苷三磷酸的制备方法中,仅进行步骤a、步骤b和步骤c即为化学修饰腺苷的制备方法,在此不一一赘述。In the method for preparing chemically modified adenosine triphosphate in this embodiment, only step a, step b and step c are performed to form the method for preparing chemically modified adenosine, which will not be described in detail here.
实施例29 N6-丙酰基腺苷及N6-丙酰基腺苷三磷酸的合成Example 29 Synthesis of N6-propionyl adenosine and N6-propionyl adenosine triphosphate
本实施例提供一种化学修饰腺苷和化学修饰腺苷三磷酸的制备方法,所述化学修饰腺苷为N6-丙酰基腺苷,所述化学修饰腺苷三磷酸为N6-丙酰基腺苷三磷酸,结构如下所示。
This embodiment provides a method for preparing chemically modified adenosine and chemically modified adenosine triphosphate, wherein the chemically modified adenosine is N6-propionyl adenosine, and the chemically modified adenosine triphosphate is N6-propionyl adenosine triphosphate, and the structures are shown below.
制备的各步骤的反应条件如下所示,并采用核磁共振和质谱对化合物结构进行表征:The reaction conditions of each step of the preparation are shown below, and the structure of the compound was characterized by nuclear magnetic resonance and mass spectrometry:
a.取3mmol(1eq)腺苷以15mL N,N-二甲基甲酰胺溶解,然后依次加入12mmol(4eq)叔丁基二甲基氯硅烷和15mmol(5eq)咪唑,25℃下搅拌过夜;反应结束后,以10mL乙酸乙酯萃取反应液,重复3次,水洗有机相,重复3次,收集有机相,无水硫酸钠干燥,过滤,浓缩,硅胶柱色谱纯化(二氯甲烷/甲醇=100/1),得到白色固体化合物5,收率95%;化合物1的结构已由1H NMR、13C NMR和HRMS确认。a. 3 mmol (1 eq) of adenosine was dissolved in 15 mL of N,N-dimethylformamide, and then 12 mmol (4 eq) of tert-butyldimethylsilyl chloride and 15 mmol (5 eq) of imidazole were added in sequence, and stirred at 25°C overnight; after the reaction, the reaction solution was extracted with 10 mL of ethyl acetate, and the extraction was repeated 3 times; the organic phase was washed with water, and the extraction was repeated 3 times; the organic phase was collected, dried over anhydrous sodium sulfate, filtered, concentrated, and purified by silica gel column chromatography (dichloromethane/methanol=100/1) to obtain a white solid compound 5 with a yield of 95%; the structure of compound 1 has been confirmed by 1 H NMR, 13 C NMR and HRMS.
b.以15mL无水吡啶溶解12mmol(1eq)化合物5,然后滴加1.2eq丙酰氯,之后加入10mg 4-二甲氨基吡啶,25℃下搅拌过夜反应;反应结束后以10mL乙酸乙酯萃取反应液,重复3次,水洗有机相,重复3次,收集有机相,无水硫酸钠干燥,过滤,浓缩,硅胶柱色谱纯化(二氯甲烷/甲醇=200/1),得到白色泡固体化合物6,收率92%;化合物2的结构已由1H NMR、13C NMR和HRMS确认。b. 12 mmol (1 eq) of compound 5 was dissolved in 15 mL of anhydrous pyridine, and then 1.2 eq of propionyl chloride was added dropwise, followed by 10 mg of 4-dimethylaminopyridine. The mixture was stirred at 25°C overnight to react. After the reaction, the reaction solution was extracted with 10 mL of ethyl acetate, repeated 3 times, and the organic phase was washed with water, repeated 3 times. The organic phase was collected, dried over anhydrous sodium sulfate, filtered, concentrated, and purified by silica gel column chromatography (dichloromethane/methanol=200/1) to obtain compound 6 as a white foamy solid with a yield of 92%. The structure of compound 2 was confirmed by 1 H NMR, 13 C NMR and HRMS.
c.以15mL无水四氢呋喃溶解1.5mmol(1eq)化合物6,然后滴加4.5mL(3eq)四丁基氟化铵溶液(1M四氢呋喃溶液),室温下搅拌反应0.5h,然后减压浓缩,硅胶柱色谱纯化(二氯甲烷/甲醇=20/1至10/1),得到白色固体化合物7(N6-丙酰基腺苷)。化合物7的结构已由1H NMR、13C NMR和HRMS确认。N6-丙酰基腺苷的1H NMR表征图如图46所示,N6-丙酰基腺苷的13C NMR表征图如图47所示,N6-丙酰基腺苷的HRMS表征图如图48所示。c. Dissolve 1.5 mmol (1 eq) of compound 6 in 15 mL of anhydrous tetrahydrofuran, then dropwise add 4.5 mL (3 eq) of tetrabutylammonium fluoride solution (1 M tetrahydrofuran solution), stir the reaction at room temperature for 0.5 h, then concentrate under reduced pressure, and purify by silica gel column chromatography (dichloromethane/methanol=20/1 to 10/1) to obtain a white solid compound 7 (N6-propionyl adenosine). The structure of compound 7 has been confirmed by 1 H NMR, 13 C NMR and HRMS. The 1 H NMR characterization of N6-propionyl adenosine is shown in FIG46 , the 13 C NMR characterization of N6-propionyl adenosine is shown in FIG47 , and the HRMS characterization of N6-propionyl adenosine is shown in FIG48 .
d.以2mL无水吡啶溶解0.1mmol化合物7,并减压共蒸除水3次;在氮气保护下,注入1mL干燥磷酸三甲酯,待固体完全溶解后将试管置于冰浴中冷却2min,并用注射器滴加1.5eq三氯氧磷;冰浴下搅拌反应2.5h。d. Dissolve 0.1 mmol of compound 7 in 2 mL of anhydrous pyridine and remove water under reduced pressure three times; inject 1 mL of dry trimethyl phosphate under nitrogen protection, and after the solid is completely dissolved, place the test tube in an ice bath for 2 min, and add 1.5 eq of phosphorus oxychloride dropwise with a syringe; stir and react in an ice bath for 2.5 h.
e.之后在冰浴条件下依次注入1.5eq干燥三丁胺,1.5mL(8eq)三丁基焦磷酸铵溶液(0.5M N,N-二甲基甲酰胺溶液),移至室温下反应0.5h。e. Then, inject 1.5 eq of dry tributylamine and 1.5 mL (8 eq) of tributylammonium pyrophosphate solution (0.5 M N,N-dimethylformamide solution) in sequence under ice bath conditions, and move to room temperature to react for 0.5 h.
f.反应完成后,冰浴下以5mL 2.0M饱和三乙胺-碳酸水溶液淬灭反应,然后以10mL二氯甲烷萃取反应液,收集水相,重复4次;以快速制备液相色谱仪C18柱分离纯化收集的水相,并以50mM饱和三乙胺-碳酸水溶液和色谱级乙腈为流动相;最后将收集纯化后的溶液冻干,得到白色固体化合物8(N6-丙酰基腺苷三磷酸)。化合物8的结构已由1H NMR,31PNMR和HRMS确认。N6-丙酰基腺苷三磷酸的1H NMR表征图如图49所示,N6-丙酰基腺苷三磷酸的13C NMR表征图如图50所示,N6-丙酰基腺苷三磷酸的HRMS表征图如图51所示。 f. After the reaction is completed, quench the reaction with 5 mL of 2.0 M saturated triethylamine-carbonic acid aqueous solution under ice bath, then extract the reaction solution with 10 mL of dichloromethane, collect the aqueous phase, and repeat 4 times; separate and purify the collected aqueous phase with a rapid preparative liquid chromatograph C18 column, and use 50 mM saturated triethylamine-carbonic acid aqueous solution and chromatographic grade acetonitrile as mobile phases; finally, freeze-dry the collected and purified solution to obtain white solid compound 8 (N6-propionyl adenosine triphosphate). The structure of compound 8 has been confirmed by 1 H NMR, 31 PNMR and HRMS. The 1 H NMR characterization of N6-propionyl adenosine triphosphate is shown in Figure 49, the 13 C NMR characterization of N6-propionyl adenosine triphosphate is shown in Figure 50, and the HRMS characterization of N6-propionyl adenosine triphosphate is shown in Figure 51.
实施例30 N6-丁酰基腺苷及N6-丁酰基腺苷三磷酸的合成Example 30 Synthesis of N6-butyryladenosine and N6-butyryladenosine triphosphate
本实施例提供一种化学修饰腺苷和化学修饰腺苷三磷酸的制备方法,所述化学修饰腺苷为N6-丁酰基腺苷,所述化学修饰腺苷三磷酸为N6-丁酰基腺苷三磷酸,结构如下所示。
This embodiment provides a method for preparing chemically modified adenosine and chemically modified adenosine triphosphate, wherein the chemically modified adenosine is N6-butyryladenosine, and the chemically modified adenosine triphosphate is N6-butyryladenosine triphosphate, and the structures are shown below.
制备方法与实施例29的区别在于,步骤b中采用丁酰氯进行反应,制备的各步骤的反应条件参照实施例29,并采用核磁共振和质谱对化合物结构进行表征。The difference between the preparation method and Example 29 is that butyryl chloride is used for the reaction in step b, the reaction conditions of each preparation step refer to Example 29, and the structure of the compound is characterized by nuclear magnetic resonance and mass spectrometry.
N6-丁酰基腺苷的1H NMR表征图如图52所示,N6-丁酰基腺苷的13C NMR表征图如图53所示,N6-丁酰基腺苷的HRMS表征如图54所示。N6-丁酰基腺苷三磷酸的1H NMR表征图如图55所示,N6-丁酰基腺苷三磷酸的13C NMR表征图如图56所示,N6-丁酰基腺苷三磷酸的HRMS表征如图57所示。The 1 H NMR characterization chart of N6-butyryladenosine is shown in Figure 52, the 13 C NMR characterization chart of N6-butyryladenosine is shown in Figure 53, and the HRMS characterization chart of N6-butyryladenosine is shown in Figure 54. The 1 H NMR characterization chart of N6-butyryladenosine triphosphate is shown in Figure 55, the 13 C NMR characterization chart of N6-butyryladenosine triphosphate is shown in Figure 56, and the HRMS characterization chart of N6-butyryladenosine triphosphate is shown in Figure 57.
实施例31 N6-戊酰基腺苷及N6-戊酰基腺苷三磷酸的合成Example 31 Synthesis of N6-pentanoyl adenosine and N6-pentanoyl adenosine triphosphate
本实施例提供一种化学修饰腺苷和化学修饰腺苷三磷酸的制备方法,所述化学修饰腺苷为N6-戊酰基腺苷,所述化学修饰腺苷三磷酸为N6-戊酰基腺苷三磷酸,结构如下所示。
This embodiment provides a method for preparing chemically modified adenosine and chemically modified adenosine triphosphate, wherein the chemically modified adenosine is N6-pentanoyl adenosine, and the chemically modified adenosine triphosphate is N6-pentanoyl adenosine triphosphate, and the structures are shown below.
制备方法与实施例29的区别在于,步骤b中采用戊酰氯进行反应,制备的各步骤的反应条件参照实施例29,并采用核磁共振和质谱对化合物结构进行表征。The difference between the preparation method and Example 29 is that valeryl chloride is used for the reaction in step b, the reaction conditions of each step of the preparation refer to Example 29, and the structure of the compound is characterized by nuclear magnetic resonance and mass spectrometry.
N6-戊酰基腺苷的1H NMR表征图如图58所示,N6-戊酰基腺苷的13C NMR表征图如图59所示,N6-戊酰基腺苷的HRMS表征图如图60所示。N6-戊酰基腺苷三磷酸的1H NMR表征图如图61所示,N6-戊酰基腺苷三磷酸的13C NMR表征图如图62所示,N6-戊酰基腺苷三磷酸的HRMS表征图如图63所示。The 1 H NMR characterization chart of N6-pentanoyl adenosine is shown in Figure 58, the 13 C NMR characterization chart of N6-pentanoyl adenosine is shown in Figure 59, and the HRMS characterization chart of N6-pentanoyl adenosine is shown in Figure 60. The 1 H NMR characterization chart of N6-pentanoyl adenosine triphosphate is shown in Figure 61, the 13 C NMR characterization chart of N6-pentanoyl adenosine triphosphate is shown in Figure 62, and the HRMS characterization chart of N6-pentanoyl adenosine triphosphate is shown in Figure 63.
实施例32 N6-己酰基腺苷及N6-己酰基腺苷三磷酸的合成Example 32 Synthesis of N6-hexanoyl adenosine and N6-hexanoyl adenosine triphosphate
本实施例提供一种化学修饰腺苷和化学修饰腺苷三磷酸的制备方法,所述化学修饰腺苷为N6-己酰基腺苷,所述化学修饰腺苷三磷酸为N6-己酰基腺苷三磷酸,结构如下所示。
This embodiment provides a method for preparing chemically modified adenosine and chemically modified adenosine triphosphate, wherein the chemically modified adenosine is N6-hexanoyl adenosine, and the chemically modified adenosine triphosphate is N6-hexanoyl adenosine triphosphate, and the structures are shown below.
制备方法与实施例29的区别在于,步骤b中采用己酰氯进行反应,制备的各步骤的反应条件参照实施例29,并采用核磁共振和质谱对化合物结构进行表征。 The difference between the preparation method and Example 29 is that hexanoyl chloride is used for the reaction in step b, the reaction conditions of each preparation step refer to Example 29, and the structure of the compound is characterized by nuclear magnetic resonance and mass spectrometry.
N6-己酰基腺苷的1H NMR表征图如图64所示,N6-己酰基腺苷的13C NMR表征图如图65所示,N6-己酰基腺苷的HRMS表征图如图66所示。N6-己酰基腺苷三磷酸的1H NMR表征图如图67所示,N6-己酰基腺苷三磷酸的13C NMR表征图如图68所示,N6-己酰基腺苷三磷酸的HRMS表征图如图69所示。The 1 H NMR characterization chart of N6-hexanoyl adenosine is shown in Figure 64, the 13 C NMR characterization chart of N6-hexanoyl adenosine is shown in Figure 65, and the HRMS characterization chart of N6-hexanoyl adenosine is shown in Figure 66. The 1 H NMR characterization chart of N6-hexanoyl adenosine triphosphate is shown in Figure 67, the 13 C NMR characterization chart of N6-hexanoyl adenosine triphosphate is shown in Figure 68, and the HRMS characterization chart of N6-hexanoyl adenosine triphosphate is shown in Figure 69.
实施例33 N6-苯甲酰基腺苷三磷酸Example 33 N6-Benzoyl adenosine triphosphate
本实施例提供一种化学修饰腺苷三磷酸的制备方法,所述化学修饰腺苷三磷酸为N6-苯甲酰基腺苷三磷酸,结构如下所示。
This embodiment provides a method for preparing chemically modified adenosine triphosphate, wherein the chemically modified adenosine triphosphate is N6-benzoyl adenosine triphosphate, and the structure is shown below.
制备方法与实施例29的区别在于,步骤b中采用苯甲酸酐进行反应,制备的各步骤的反应条件参照实施例29,并采用核磁共振和质谱对化合物结构进行表征。N6-苯甲酰基腺苷三磷酸的HRMS表征图如图70所示。The difference between the preparation method and Example 29 is that benzoic anhydride is used for the reaction in step b, the reaction conditions of each step of the preparation are referred to Example 29, and the structure of the compound is characterized by nuclear magnetic resonance and mass spectrometry. The HRMS characterization chart of N6-benzoyl adenosine triphosphate is shown in Figure 70.
实施例34 N6-甲基腺苷三磷酸Example 34 N6-methyladenosine triphosphate
本实施例提供一种化学修饰腺苷三磷酸的制备方法,所述化学修饰腺苷三磷酸为N6-甲基腺苷三磷酸,结构如下所示。
This embodiment provides a method for preparing chemically modified adenosine triphosphate, wherein the chemically modified adenosine triphosphate is N6-methyladenosine triphosphate, and the structure is shown below.
制备方法与实施例29的区别在于,直接以商品化的N6-甲基腺苷为原料(CAS号:1867-73-8)进行三磷酸化反应,制备的各步骤的反应条件参照实施例29,并采用核磁共振和质谱对化合物结构进行表征。N6-甲基腺苷三磷酸的HRMS表征图如图71所示。The difference between the preparation method and Example 29 is that commercial N6-methyladenosine (CAS No.: 1867-73-8) is used as a raw material for triphosphorylation reaction, the reaction conditions of each step of the preparation are referred to Example 29, and the structure of the compound is characterized by nuclear magnetic resonance and mass spectrometry. The HRMS characterization chart of N6-methyladenosine triphosphate is shown in Figure 71.
实施例35 N6-乙酰基腺苷三磷酸Example 35 N6-acetyl adenosine triphosphate
本实施例提供一种化学修饰腺苷三磷酸的制备方法,所述化学修饰腺苷三磷酸为N6-乙酰基腺苷三磷酸,结构如下所示。
This embodiment provides a method for preparing chemically modified adenosine triphosphate, wherein the chemically modified adenosine triphosphate is N6-acetyl adenosine triphosphate, and the structure is shown below.
制备方法与实施例29的区别在于,步骤b中采用乙酰氯进行反应,制备的各步骤的反应 条件参照实施例29,并采用核磁共振和质谱对化合物结构进行表征。N6-乙酰基腺苷三磷酸的HRMS表征图如图72所示。The difference between the preparation method and Example 29 is that acetyl chloride is used for reaction in step b, and the reaction of each step of the preparation is The conditions were similar to those in Example 29, and the structure of the compound was characterized by nuclear magnetic resonance and mass spectrometry. The HRMS characterization graph of N6-acetyl adenosine triphosphate is shown in FIG72 .
实施例36 2-氨基腺苷三磷酸Example 36 2-aminoadenosine triphosphate
本实施例提供一种化学修饰腺苷三磷酸的制备方法,所述化学修饰腺苷三磷酸为2-氨基腺苷三磷酸,结构如下所示。
This embodiment provides a method for preparing chemically modified adenosine triphosphate, wherein the chemically modified adenosine triphosphate is 2-aminoadenosine triphosphate, and the structure is shown below.
制备方法与实施例29的区别在于,直接以商品化的2-氨基腺苷为原料(CAS号:2096-10-8)进行反应,制备的各步骤的反应条件参照实施例29,并采用核磁共振和质谱对化合物结构进行表征。2-氨基腺苷三磷酸的HRMS表征图如图73所示。The difference between the preparation method and Example 29 is that commercial 2-aminoadenosine (CAS No.: 2096-10-8) is directly used as a raw material for the reaction, the reaction conditions of each step of the preparation are referred to Example 29, and the structure of the compound is characterized by nuclear magnetic resonance and mass spectrometry. The HRMS characterization chart of 2-aminoadenosine triphosphate is shown in Figure 73.
实施例37 7-脱氮腺苷三磷酸Example 37 7-Deazaadenosine triphosphate
本实施例提供一种化学修饰腺苷三磷酸的制备方法,所述化学修饰腺苷三磷酸为7-脱氮腺苷三磷酸,结构如下所示。
This embodiment provides a method for preparing chemically modified adenosine triphosphate, wherein the chemically modified adenosine triphosphate is 7-deazaadenosine triphosphate, and the structure is shown below.
制备方法与实施例29的区别在于,直接以商品化的7-脱氮腺苷为原料(CAS号:69-33-0)进行反应,制备的各步骤的反应条件参照实施例29,并采用核磁共振和质谱对化合物结构进行表征。7-脱氮腺苷三磷酸的HRMS表征图如图74所示。The difference between the preparation method and Example 29 is that commercial 7-deazaadenosine (CAS No.: 69-33-0) is directly used as a raw material for the reaction, the reaction conditions of each step of the preparation are referred to Example 29, and the structure of the compound is characterized by nuclear magnetic resonance and mass spectrometry. The HRMS characterization chart of 7-deazaadenosine triphosphate is shown in Figure 74.
实施例38编译增强绿色荧光蛋白的DNA模板的制备Example 38 Preparation of DNA Template for Compiling Enhanced Green Fluorescent Protein
编译增强绿色荧光蛋白的DNA模板是将含有目的基因的质粒以内切酶进行线性化处理制备得到的。The DNA template for compiling enhanced green fluorescent protein is prepared by linearizing the plasmid containing the target gene with an endonuclease.
1)酶切法制备增强绿色荧光蛋白线性DNA模板,在96孔PCR仪器中进行。线性化后的产物中至少包括:A)一个启动子序列(T7启动子);B)包含至少一个Kozak序列的5’UTR;C)3’UTR;D)eGFP编码序列。本实施例涉及的具体序列如表5所示。1) Enzyme digestion was used to prepare an enhanced green fluorescent protein linear DNA template in a 96-well PCR instrument. The linearized product at least includes: A) a promoter sequence (T7 promoter); B) a 5'UTR containing at least one Kozak sequence; C) a 3'UTR; D) an eGFP coding sequence. The specific sequences involved in this embodiment are shown in Table 5.
表5
Table 5
配制如下反应体系,进行DNA模板的线性化:反应体积,20μL(为单个管的反应体积,一次同时反应多管),具体的反应体系如表6所示。The following reaction system was prepared to linearize the DNA template: reaction volume, 20 μL (reaction volume of a single tube, multiple tubes were reacted at one time), and the specific reaction system is shown in Table 6.
表6
Table 6
将恒温器的热盖启动,设置为60℃。点击恒温器反应系统,37℃,15min。酶切产物用HiPure Gel Pure DNA Mini Kit(广州美基生物科技有限公司,货号:D2111)纯化。纯化过程如下:Turn on the thermostat's hot cover and set it to 60°C. Click the thermostat reaction system, 37°C, 15 min. The enzyme digestion product was purified using HiPure Gel Pure DNA Mini Kit (Guangzhou Meiji Biotechnology Co., Ltd., catalog number: D2111). The purification process is as follows:
(1)短暂离心酶切反应产物。(1) Briefly centrifuge the digestion reaction products.
(2)加入等倍体积的缓冲液GDP,颠倒混匀。(2) Add an equal volume of GDP buffer and mix by inversion.
(3)将HiPure DNA柱子套在收集管中,把混合液转移至DNA柱子中,12000g离心30-60s。倒弃滤液,把柱子套回2mL离心管中。加入600μL缓冲液DW2(已用无水乙醇稀释)至柱子中,12000g离心30-60s。(3) Place the HiPure DNA column in a collection tube, transfer the mixture to the DNA column, and centrifuge at 12,000 g for 30-60 seconds. Discard the filtrate and place the column back in a 2 mL centrifuge tube. Add 600 μL of buffer DW2 (diluted with anhydrous ethanol) to the column and centrifuge at 12,000 g for 30-60 seconds.
(4)倒弃滤液,把柱子套回2mL离心管中。加入600μL缓冲液DW2(已用无水乙醇稀释)至柱子中,12000g离心30-60s。倒弃滤液,把柱子套回2mL离心管中,12000g离心2分钟。(4) Discard the filtrate and put the column back into a 2 mL centrifuge tube. Add 600 μL of buffer DW2 (diluted with anhydrous ethanol) to the column and centrifuge at 12,000 g for 30-60 seconds. Discard the filtrate and put the column back into a 2 mL centrifuge tube and centrifuge at 12,000 g for 2 minutes.
(5)把柱子套回1.5mL离心管中,加入15-30μL洗脱缓冲液至柱子膜中央,室温放置2分钟,12000g离心1分钟。丢弃柱子,用NanoDrop分光光度计检测模板DNA的浓度,以及260/280、260/230的比值,然后将DNA保存于-20℃。(5) Put the column back into a 1.5 mL centrifuge tube, add 15-30 μL of elution buffer to the center of the column membrane, leave at room temperature for 2 minutes, and centrifuge at 12,000 g for 1 minute. Discard the column, measure the concentration of the template DNA, and the ratios of 260/280 and 260/230 using a NanoDrop spectrophotometer, and then store the DNA at -20°C.
实施例39编译荧光素酶的DNA模板的制备Example 39 Preparation of DNA template for encoding luciferase
以编译荧光素酶的质粒为模板,利用DNA聚合酶在PCR小管中进行DNA扩增。具体步骤如下:Using the plasmid encoding luciferase as a template, DNA amplification was performed in a PCR tube using DNA polymerase. The specific steps are as follows:
PCR法制备荧光素酶DNA模板,在96孔PCR仪器中进行。The luciferase DNA template was prepared by PCR and carried out in a 96-well PCR instrument.
PCR产物中至少包括:A)一个启动子序列(T7启动子);B)包含至少一个Kozak序列 的5’UTR;C)3’UTR;D)荧光素酶编码序列;E)聚A尾(polyA尾)。本实施例涉及的具体序列如表7所示The PCR product contains at least: A) a promoter sequence (T7 promoter); B) at least one Kozak sequence 5'UTR; C) 3'UTR; D) luciferase coding sequence; E) poly A tail (polyA tail). The specific sequences involved in this example are shown in Table 7
表7
Table 7
配制如下反应体系进行DNA模板的扩增:反应体积,50μL(为单个管的反应体积,一次同时反应多管),具体的反应体系如表8所示。The following reaction system was prepared for amplification of the DNA template: reaction volume, 50 μL (reaction volume of a single tube, multiple tubes were reacted at one time), and the specific reaction system is shown in Table 8.
表8
Table 8
反应程序如下:预变性95℃,1分钟,变性95℃,15秒,退火52℃,5秒,延伸72℃,2分钟,共32个循环;最后延伸72℃,5分钟。The reaction program was as follows: pre-denaturation at 95°C for 1 minute, denaturation at 95°C for 15 seconds, annealing at 52°C for 5 seconds, extension at 72°C for 2 minutes, for a total of 32 cycles; and final extension at 72°C for 5 minutes.
反应结束后将反应液合并于1.5mL管中。取2μL进行DNA琼脂糖凝胶电泳检测以确定反应成功(琼脂糖凝胶电泳检测条件:2%琼脂糖,5V/分钟,电泳30分钟)。After the reaction is completed, the reaction solution is combined in a 1.5 mL tube. 2 μL is taken for DNA agarose gel electrophoresis to confirm the success of the reaction (agarose gel electrophoresis detection conditions: 2% agarose, 5 V/min, electrophoresis for 30 minutes).
反应产物用HiPure Gel Pure DNA Mini Kit纯化。纯化过程如下:The reaction product was purified using HiPure Gel Pure DNA Mini Kit. The purification process is as follows:
(1)短暂离心PCR产物。(1) Briefly centrifuge the PCR product.
(2)加入等倍体积的缓冲液GDP,颠倒混匀。(2) Add an equal volume of GDP buffer and mix by inversion.
(3)将HiPure DNA柱子套在收集管中,把混合液转移至DNA柱子中,12000g离心30-60s。倒弃滤液,把柱子套回2mL离心管中。加入600μL缓冲液DW2(已用无水乙醇稀释)至柱子中,12000g离心30-60s。(3) Place the HiPure DNA column in a collection tube, transfer the mixture to the DNA column, and centrifuge at 12,000 g for 30-60 seconds. Discard the filtrate and place the column back in a 2 mL centrifuge tube. Add 600 μL of buffer DW2 (diluted with anhydrous ethanol) to the column and centrifuge at 12,000 g for 30-60 seconds.
(4)倒弃滤液,把柱子套回2mL离心管中。加入600μL缓冲液DW2(已用无水乙醇稀释)至柱子中,12000g离心30-60s。倒弃滤液,把柱子套回2mL离心管中,12000g离心2分钟。(4) Discard the filtrate and put the column back into a 2 mL centrifuge tube. Add 600 μL of buffer DW2 (diluted with anhydrous ethanol) to the column and centrifuge at 12,000 g for 30-60 seconds. Discard the filtrate and put the column back into a 2 mL centrifuge tube and centrifuge at 12,000 g for 2 minutes.
(5)把柱子套回1.5mL离心管中,加入15-30μL洗脱缓冲液至柱子膜中央,室温放置2分钟,12000g离心1分钟。丢弃柱子,用NanoDrop分光光度计检测模板DNA的浓度,以及260/280、260/230的比值,然后将DNA保存于-20℃。 (5) Put the column back into a 1.5 mL centrifuge tube, add 15-30 μL of elution buffer to the center of the column membrane, leave at room temperature for 2 minutes, and centrifuge at 12,000 g for 1 minute. Discard the column, measure the concentration of the template DNA, and the ratios of 260/280 and 260/230 using a NanoDrop spectrophotometer, and then store the DNA at -20°C.
实施例40化学修饰的腺苷全局替代天然腺苷的mRNA的体外转录合成Example 40 In vitro transcription synthesis of mRNA in which chemically modified adenosine globally replaces natural adenosine
化学修饰的腺苷全局替换天然腺苷的mRNA的合成是利用T7 RNA聚合酶以体外转录实现的,以50μL的反应体系为例,在0.2mL的PCR反应管中依次加入表9所示成分。The synthesis of mRNA in which chemically modified adenosine globally replaces natural adenosine is achieved by in vitro transcription using T7 RNA polymerase. Taking a 50 μL reaction system as an example, the components listed in Table 9 are added sequentially into a 0.2 mL PCR reaction tube.
表9
Table 9
*当体外转录制备eGFP mRNA时,模板则为增强绿色荧光蛋白DNA模板。**为腺嘌呤核苷三磷酸或化学修饰胞腺嘌呤核苷三磷酸。Cap Analogue#为市售帽结构类似物;在本实例中,采用的为New England Biolabs提供的3’-O-Me-m7-G(5’)ppp(5’)G RNA Cap Structure Analog,货号为S1411S。*When preparing eGFP mRNA by in vitro transcription, the template is an enhanced green fluorescent protein DNA template. **Adenosine triphosphate or chemically modified cytosine triphosphate. Cap Analogue# is a commercially available cap structure analog; in this example, the one used is 3’-O-Me-m7-G(5’)ppp(5’)G RNA Cap Structure Analog provided by New England Biolabs, with the catalog number S1411S.
将恒温器的热盖启动,设置为60℃。点击恒温器反应系统,37℃,2-4h。孵育完成以后,mRNA纯化方法如下:Turn on the thermostat's hot cover and set it to 60°C. Click on the thermostat reaction system, 37°C, 2-4h. After the incubation is complete, the mRNA purification method is as follows:
1.在每个50μL反应体系中加入1μL DNA酶Ⅰ(RNase-free),37℃下孵育15分钟消化体系中的DNA模板。1. Add 1 μL DNase I (RNase-free) to each 50 μL reaction system and incubate at 37°C for 15 minutes to digest the DNA template in the system.
2.在每个50μL反应体系中加入2.5μL乙二胺四乙酸二钠盐溶液(500mM),65℃下孵育10分钟。2. Add 2.5 μL of ethylenediaminetetraacetic acid disodium salt solution (500 mM) to each 50 μL reaction system and incubate at 65°C for 10 minutes.
3.将纤维素溶液悬浮在洗脱溶液中,制备成0.2g纤维素/mL的悬浊液(配制方法参考文献:A Facile Method for the Removal of dsRNA Contaminants from In Vitro Transcribed mRNA,MolecμLar Therapy Nucleic Acids,2019,15,26-35)。将DNA吸附柱套在2mL接收管中,(广州美基生物,DNA/RNA小量吸附柱,货号为M021),然后取630μL悬浊液于DNA柱子中,涡旋震荡2h进行活化。3. Suspend the cellulose solution in the elution solution to prepare a suspension of 0.2 g cellulose/mL (reference for preparation method: A Facile Method for the Removal of dsRNA Contaminants from In Vitro Transcribed mRNA, Molec μLar Therapy Nucleic Acids, 2019, 15, 26-35). Put the DNA adsorption column in a 2 mL receiving tube (Guangzhou Meiji Biotechnology, DNA/RNA small amount adsorption column, product number is M021), then take 630 μL of the suspension into the DNA column and vortex for 2 hours for activation.
4.将活化后的纤维素悬浊液在13000g离心1分钟,丢弃滤液,然后重新加入500μL洗脱液进行悬浊。然后将体外转录反应混合物溶液加入到DNA柱子中,涡旋震荡30分钟。4. Centrifuge the activated cellulose suspension at 13000g for 1 minute, discard the filtrate, and then resuspend it with 500μL of elution solution. Then add the in vitro transcription reaction mixture solution to the DNA column and vortex for 30 minutes.
5.将DNA柱子在13000g下离心1分钟,收集滤液,加入到一根新的活化好的DNA柱子中(活化步骤如步骤3所述),涡旋震荡30分钟。5. Centrifuge the DNA column at 13000g for 1 minute, collect the filtrate, add it to a new activated DNA column (activation step as described in step 3), and vortex for 30 minutes.
6.将DNA柱子在13000g下离心1分钟,收集滤液,然后将滤液转移至一个新的在冰面预冷的无核酸酶的1.5mL离心管中。6. Centrifuge the DNA column at 13,000 g for 1 minute, collect the filtrate, and transfer the filtrate to a new 1.5 mL nuclease-free centrifuge tube pre-chilled on ice.
7.将无水异丙醇和3M的醋酸钠溶液提前在冰面预冷,然后在步骤6中的离心管加入500μL无水异丙醇和50μL醋酸钠溶液,轻柔颠倒混合均匀,然后在4℃下14000g离心10分钟。7. Pre-cool anhydrous isopropanol and 3M sodium acetate solution on ice, then add 500 μL of anhydrous isopropanol and 50 μL of sodium acetate solution to the centrifuge tube in step 6, gently invert to mix, and then centrifuge at 14000g at 4°C for 10 minutes.
8.去除上清溶液,然后在离心管中加入300μL 4℃预冷的无水乙醇,然后在4℃下14000 g离心10分钟。8. Remove the supernatant solution, then add 300 μL of 4°C pre-cooled anhydrous ethanol to the centrifuge tube, and then incubate at 4°C at 14000 Centrifuge at 4 °C for 10 min.
9.去除上清溶液,然后将沉淀在室温下风干30分钟。9. Remove the supernatant solution and air-dry the pellet at room temperature for 30 minutes.
10.将所得的mRNA产物溶解在20-50μL无核酸酶水中,用NanoDrop分光光度计测定纯化后的mRNA浓度,并同时测定260/230、260/280的比值。10. Dissolve the obtained mRNA product in 20-50 μL of nuclease-free water, and measure the concentration of the purified mRNA using a NanoDrop spectrophotometer, and simultaneously measure the ratios of 260/230 and 260/280.
11.取500ng纯化后的mRNA,进行琼脂糖凝胶电泳检测其片段完整性。11. Take 500 ng of purified mRNA and perform agarose gel electrophoresis to detect the integrity of its fragments.
12.将mRNA溶液于-80℃下保存。12. Store the mRNA solution at -80°C.
其中化学修饰的腺嘌呤核苷包括以下情况:N6-甲基腺苷、N6-乙酰基腺苷、N6-丙酰基腺苷、N6-丁酰基腺苷、N6-戊酰基腺苷、N6-己酰基腺苷、N6-苯甲酰基腺苷、2-氨基腺苷或7-脱氮腺苷等。The chemically modified adenine nucleosides include the following: N6-methyladenosine, N6-acetyladenosine, N6-propionyladenosine, N6-butyryladenosine, N6-pentanoyladenosine, N6-hexanoyladenosine, N6-benzoyladenosine, 2-aminoadenosine or 7-deazaadenosine, etc.
实施例41化学修饰的腺苷与假尿苷联用全局替代天然腺苷和天然尿苷的mRNA的体外转录合成Example 41 In vitro transcription and synthesis of mRNA using chemically modified adenosine and pseudouridine to globally replace natural adenosine and natural uridine
本实施例与实施例40中的条件和操作方法完全相同,其不同点在于,做体外转录的底物中,天然尿苷被假尿苷完全替换。其余纯化和表征方法则完全一样。The conditions and operation methods of this example are exactly the same as those of Example 40, except that natural uridine is completely replaced by pseudouridine in the substrate for in vitro transcription. The rest of the purification and characterization methods are exactly the same.
表10
Table 10
**为腺嘌呤核苷三磷酸或化学修饰胞腺嘌呤核苷三磷酸。Cap Analogue#为市售帽结构类似物;在本实例中,采用的为New England Biolabs提供的3’-O-Me-m7-G(5’)ppp(5’)G RNA Cap Structure Analog,货号为S1411S。** is adenosine triphosphate or chemically modified adenosine triphosphate. Cap Analogue# is a commercially available cap structure analog; in this example, 3’-O-Me-m7-G(5’)ppp(5’)G RNA Cap Structure Analog provided by New England Biolabs was used, with the catalog number S1411S.
实施例42体外转录的eGFP mRNA的多聚腺苷化修饰Example 42 Polyadenylation of in vitro transcribed eGFP mRNA
化学修饰腺苷对体外转录的eGFP mRNA进行多聚腺苷化修饰是以E.Coli polyA聚合酶或者Yeast polyA聚合酶实现的,以20μL的反应体系为例,在0.2mL的PCR管中,依次加入如表11和表12所示组分。Chemically modified adenosine is used to perform polyadenylation modification on in vitro transcribed eGFP mRNA using E. coli polyA polymerase or Yeast polyA polymerase. Taking a 20 μL reaction system as an example, add the components shown in Tables 11 and 12 in a 0.2 mL PCR tube.
表11
Table 11
*为腺嘌呤核苷三磷酸或化学修饰腺嘌呤核苷三磷酸。缓冲液A(5×)组分如下:500mM乙酸钾,100mM Tris-HCl(pH 8.0),10mM乙酸镁,0.25% NP-40。* is adenosine triphosphate or chemically modified adenosine triphosphate. The components of buffer A (5×) are as follows: 500 mM potassium acetate, 100 mM Tris-HCl (pH 8.0), 10 mM magnesium acetate, 0.25% NP-40.
表12
Table 12
*为腺嘌呤核苷三磷酸或化学修饰腺嘌呤核苷三磷酸。缓冲液B(5×)组分如下:50%甘油,125mM Tris-HCl(pH 7.0),0.5mg/mL BSA,3.5mM氯化锰,250mM氯化钾,0.05mM EDTA,2.5mM二硫苏糖醇。* is adenosine triphosphate or chemically modified adenosine triphosphate. The components of buffer B (5×) are as follows: 50% glycerol, 125 mM Tris-HCl (pH 7.0), 0.5 mg/mL BSA, 3.5 mM manganese chloride, 250 mM potassium chloride, 0.05 mM EDTA, 2.5 mM dithiothreitol.
将恒温器的热盖启动,设置为60℃。点击恒温器反应系统,37℃,30min。孵育完成以后,加尾的mRNA纯化方法如实施例40中所描述。Turn on the thermostat's hot cover and set it to 60° C. Click on the thermostat reaction system, 37° C., 30 min. After the incubation is complete, the tailed mRNA is purified as described in Example 40.
取500ng纯化后的mRNA,进行琼脂糖凝胶电泳检测以确定反应成功(琼脂糖凝胶电泳检测条件:2%琼脂糖,5V/厘米,电泳30分钟)。500 ng of purified mRNA was taken and subjected to agarose gel electrophoresis to confirm the success of the reaction (agarose gel electrophoresis detection conditions: 2% agarose, 5 V/cm, electrophoresis for 30 minutes).
图75为以E.Coli polyA聚合酶对eGFP mRNA进行多聚腺苷化修饰的效果图,图中,泳道1-13分别为:泳道1:eGFP mRNA模板;泳道2:eGFP mRNA的天然腺苷加尾产物;泳道3:eGFP mRNA的2’-甲氧基腺苷加尾产物;泳道4:eGFP mRNA的2’-氟代腺苷加尾产物;泳道5:eGFP mRNA的N6-甲基腺苷加尾产物;泳道6:eGFP mRNA的N6-乙酰基腺苷加尾产物;泳道7:eGFP mRNA的N6-丙酰基腺苷加尾产物;泳道8:eGFP mRNA的N6-丁酰基腺苷加尾产物;泳道9:eGFP mRNA的N6-戊酰基腺苷加尾产物;泳道10:eGFP mRNA的N6-己酰基腺苷加尾产物;泳道11:eGFP mRNA的N6-苯甲酰基腺苷加尾产物;泳道12:eGFP mRNA的2-氨基腺苷加尾产物;泳道13:eGFP mRNA的7-脱氮腺苷加尾产物。Figure 75 is a diagram showing the effect of polyadenylation modification of eGFP mRNA by E. coli polyA polymerase. In the figure, lanes 1-13 are: lane 1: eGFP mRNA template; lane 2: natural adenosine tailing product of eGFP mRNA; lane 3: 2'-methoxyadenosine tailing product of eGFP mRNA; lane 4: 2'-fluoroadenosine tailing product of eGFP mRNA; lane 5: N6-methyladenosine tailing product of eGFP mRNA; lane 6: N6-acetyladenosine tailing product of eGFP mRNA. Lane 7: N6-propionyladenosine tailing products of eGFP mRNA; Lane 8: N6-butyryladenosine tailing products of eGFP mRNA; Lane 9: N6-pentanoyladenosine tailing products of eGFP mRNA; Lane 10: N6-hexanoyladenosine tailing products of eGFP mRNA; Lane 11: N6-benzoyladenosine tailing products of eGFP mRNA; Lane 12: 2-aminoadenosine tailing products of eGFP mRNA; Lane 13: 7-deazaadenosine tailing products of eGFP mRNA.
图76为以Yeast polyA聚合酶对eGFP mRNA进行多聚腺苷化修饰的效果图,图中,泳道1-13分别为:泳道1:eGFP mRNA模板;泳道2:eGFP mRNA的天然腺苷加尾产物;泳道3:eGFP mRNA的2’-甲氧基腺苷加尾产物;泳道4:eGFP mRNA的2’-氟代腺苷加尾产物;泳道5:eGFP mRNA的N6-甲基腺苷加尾产物;泳道6:eGFP mRNA的N6-乙酰基腺苷加尾产物;泳道7:eGFP mRNA的N6-丙酰基腺苷加尾产物;泳道8:eGFP mRNA的N6-丁酰基腺苷加尾产物;泳道9:eGFP mRNA的N6-戊酰基腺苷加尾产物;泳道10:eGFP mRNA的N6-己酰基腺苷加尾产物;泳道11:eGFP mRNA的N6-苯甲酰基腺苷加尾产物;泳道12:eGFP mRNA的2-氨基腺苷加尾产物;泳道13:eGFP mRNA的7-脱氮腺苷加尾产物。Figure 76 is a diagram showing the effect of polyadenylation modification of eGFP mRNA by Yeast polyA polymerase. In the figure, lanes 1-13 are respectively: lane 1: eGFP mRNA template; lane 2: natural adenosine tailing product of eGFP mRNA; lane 3: 2'-methoxyadenosine tailing product of eGFP mRNA; lane 4: 2'-fluoroadenosine tailing product of eGFP mRNA; lane 5: N6-methyladenosine tailing product of eGFP mRNA; lane 6: N6-acetyladenosine tailing product of eGFP mRNA. Lane 10: N6-hexanoyladenosine tailing product of eGFP mRNA; Lane 11: N6-benzoyladenosine tailing product of eGFP mRNA; Lane 12: 2-aminoadenosine tailing product of eGFP mRNA; Lane 13: 7-deazaadenosine tailing product of eGFP mRNA.
实施例43化学修饰的腺苷全局替换天然腺苷的荧光素酶mRNA或化学修饰的腺苷和假尿苷联用的荧光素酶mRNA在兔网织红细胞裂解液体系中的翻译Example 43 Translation of luciferase mRNA in which chemically modified adenosine globally replaces natural adenosine or luciferase mRNA in which chemically modified adenosine and pseudouridine are combined in rabbit reticulocyte lysate system
兔网织红细胞裂解液体系如表13所示。 The rabbit reticulocyte lysate system is shown in Table 13.
表13
Table 13
将恒温器的热盖启动,设置为60℃。点击恒温器反应系统,30℃,90min。孵育完成的产物直接用于荧光素的发光实验。Turn on the thermostat cover and set it to 60°C. Click the thermostat reaction system, 30°C, 90min. The incubated product is directly used for the luminescence experiment of luciferin.
实施例44体外翻译制备的荧光素酶氧化荧光素的发光实验Example 44 Luminescence experiment of luciferase oxidized luciferin prepared by in vitro translation
酶标仪检测发光的实验步骤如下:在96孔板中加入2.5μL实施例43中的孵育产物,然后在室温下孵育10分钟,之后加入50μL荧光素酶检测试剂(Promega,E1483),用移液枪吹打均匀,去除体系中的气泡,且于5分钟之内在酶标仪上完成发光强度检测。每个样品设置两个复孔。The experimental steps for detecting luminescence by microplate reader are as follows: 2.5 μL of the incubation product in Example 43 was added to a 96-well plate, and then incubated at room temperature for 10 minutes, followed by adding 50 μL of luciferase detection reagent (Promega, E1483), pipetting evenly to remove bubbles in the system, and completing the luminescence intensity detection on the microplate reader within 5 minutes. Two replicate wells were set for each sample.
图77是化学修饰的腺苷和假尿苷联用的荧光素酶mRNA在兔网织红细胞裂解物中孵育90min后表达的效果图。图中1-6分别表示,1:无修饰的荧光素酶mRNA的表达效果图;2:假尿苷替换的荧光素酶mRNA的表达效果图;3:N6-乙酰基腺苷和假尿苷联用替换的荧光素酶mRNA的表达效果图;4:N6-丙酰基腺苷和假尿苷联用替换的荧光素酶mRNA的表达效果图;5:N6-丁酰基腺苷和假尿苷联用替换的荧光素酶mRNA的表达效果图;6:2-氨基腺苷和假尿苷联用替换的荧光素酶mRNA的表达效果图。结果表明,当将化学修饰的腺苷和假尿苷联用时,修饰的mRNA在兔网织红细胞裂解液中的表达效率更高,超过假尿苷单独修饰的mRNA的表达效率,因此化学修饰的腺苷可以与假尿苷联用以进一步增强修饰的mRNA的表达效率,作为假尿苷修饰的补充选择。Figure 77 is a graph showing the expression of luciferase mRNA in combination with chemically modified adenosine and pseudouridine after incubation in rabbit reticulocyte lysate for 90 minutes. Figures 1-6 respectively represent: 1: expression effect graph of unmodified luciferase mRNA; 2: expression effect graph of luciferase mRNA replaced with pseudouridine; 3: expression effect graph of luciferase mRNA replaced with N6-acetyladenosine and pseudouridine; 4: expression effect graph of luciferase mRNA replaced with N6-propionyladenosine and pseudouridine; 5: expression effect graph of luciferase mRNA replaced with N6-butyryladenosine and pseudouridine; 6: expression effect graph of luciferase mRNA replaced with 2-aminoadenosine and pseudouridine. The results showed that when chemically modified adenosine and pseudouridine were used together, the expression efficiency of the modified mRNA in rabbit reticulocyte lysate was higher, exceeding the expression efficiency of mRNA modified with pseudouridine alone. Therefore, chemically modified adenosine can be used in combination with pseudouridine to further enhance the expression efficiency of the modified mRNA as a supplementary option to pseudouridine modification.
图78是化学修饰的腺苷全局替换天然腺苷的荧光素酶mRNA在兔网织红细胞裂解物中孵育90min后表达的效果图。图中1-10分别表示,1:无修饰的荧光素酶mRNA的表达效果图;2:N6-甲基腺苷替换的荧光素酶mRNA的表达效果图;3:N6-乙酰基腺苷替换的荧光素酶mRNA的表达效果图;4:N6-丙酰基腺苷替换的荧光素酶mRNA的表达效果图;5:N6-丁酰基腺苷替换的荧光素酶mRNA的表达效果图;6:N6-戊酰基腺苷替换的荧光素酶mRNA的表达效果图;7:N6-己酰基腺苷替换的荧光素酶mRNA的表达效果图;8:N6-苯甲酰基腺苷替换的荧光素酶mRNA的表达效果图;9:2-氨基腺苷替换的荧光素酶mRNA的表达效果图;10:7-脱氮腺苷替换的荧光素酶mRNA的表达效果图。结果表明,化学修饰的腺苷全局替换天然腺苷的荧光素酶mRNA,相对于无修饰的mRNA,在兔网织红细胞裂解液中的表达效率更高,其中修饰类型包括N6-乙酰基修饰,N6-丙酰基修饰,N6-丁酰基修饰和N6-苯甲酰基修饰。FIG78 is a graph showing the expression effect of luciferase mRNA in which chemically modified adenosine globally replaces natural adenosine after incubation in rabbit reticulocyte lysate for 90 minutes. 1-10 in the figure respectively represent, 1: the expression effect diagram of unmodified luciferase mRNA; 2: the expression effect diagram of luciferase mRNA replaced by N6-methyladenosine; 3: the expression effect diagram of luciferase mRNA replaced by N6-acetyladenosine; 4: the expression effect diagram of luciferase mRNA replaced by N6-propionyladenosine; 5: the expression effect diagram of luciferase mRNA replaced by N6-butyryladenosine; 6: the expression effect diagram of luciferase mRNA replaced by N6-pentanoyladenosine; 7: the expression effect diagram of luciferase mRNA replaced by N6-hexanoyladenosine; 8: the expression effect diagram of luciferase mRNA replaced by N6-benzoyladenosine; 9: the expression effect diagram of luciferase mRNA replaced by 2-aminoadenosine; 10: the expression effect diagram of luciferase mRNA replaced by 7-deazaadenosine. The results showed that luciferase mRNA in which chemically modified adenosine globally replaced natural adenosine had higher expression efficiency in rabbit reticulocyte lysate than unmodified mRNA, where the modification types included N6-acetyl modification, N6-propionyl modification, N6-butyryl modification and N6-benzoyl modification.
实施例45细胞内翻译制备的荧光素酶氧化荧光素的发光实验Example 45 Luminescence experiment of luciferase oxidized luciferin prepared by intracellular translation
从细胞里提取目标蛋白荧光素酶的实验步骤如下:以24孔板进行细胞培养和mRNA转染,在荧光素酶mRNA转染细胞24h以后,将24孔板置于冰面上,去除培养液,用冷PBS溶液洗两遍,然后加入200μL RIPA裂解液(翊圣,货号20115ES60,使用前加入1mM PMSF), 用移液枪吹打5min使细胞充分裂解,收集细胞裂解液,转移至1.5mL EP管中,4℃下15000转/分进行离心10min,取150μL上层清液,用于后续检测。The experimental steps for extracting the target protein luciferase from cells are as follows: cell culture and mRNA transfection are performed in a 24-well plate. 24 hours after the luciferase mRNA transfection cells, the 24-well plate is placed on ice, the culture medium is removed, and the plate is washed twice with cold PBS solution, and then 200 μL RIPA lysis buffer (Yisheng, catalog number 20115ES60, 1 mM PMSF is added before use). The cells were fully lysed by blowing with a pipette for 5 minutes, and the cell lysate was collected and transferred to a 1.5 mL EP tube. The tube was centrifuged at 15,000 rpm for 10 minutes at 4°C, and 150 μL of the supernatant was taken for subsequent detection.
酶标仪检测发光的实验步骤如下:酶标仪检测发光的实验步骤如实施例44,区别在于在96孔板中加入的是50μL本实施例中提取的蛋白溶液,然后加入50μL荧光素酶检测试剂(Promega,E1483)进行检测。每个样品设置两个复孔。The experimental steps of detecting luminescence by an ELISA instrument are as follows: The experimental steps of detecting luminescence by an ELISA instrument are as in Example 44, except that 50 μL of the protein solution extracted in this example is added to the 96-well plate, and then 50 μL of luciferase detection reagent (Promega, E1483) is added for detection. Two replicate wells are set for each sample.
图79为化学修饰的腺苷全局替换天然腺苷的荧光素酶mRNA在转染进HEK 293T细胞后24h表达的效果图,图中,1-9分别表示,1:无修饰的荧光素酶mRNA的表达效果图;2:N6-甲基腺苷替换的荧光素酶mRNA的表达效果图;3:N6-乙酰基腺苷替换的荧光素酶mRNA的表达效果图;4:N6-丙酰基腺苷替换的荧光素酶mRNA的表达效果图;5:N6-丁酰基腺苷替换的荧光素酶mRNA的表达效果图;6:N6-戊酰基腺苷替换的荧光素酶mRNA的表达效果图;7:N6-己酰基腺苷替换的荧光素酶mRNA的表达效果图;8:2-氨基腺苷替换的荧光素酶mRNA的表达效果图;9:7-脱氮腺苷替换的荧光素酶mRNA的表达效果图。从图34可知,N6-乙酰基腺苷替换天然腺苷修饰的荧光素酶mRNA,在HEK 293T细胞中的翻译效率相对于无修饰的mRNA有接近3倍的增强。Figure 79 shows the expression effect of luciferase mRNA in which chemically modified adenosine globally replaces natural adenosine 24 hours after transfection into HEK 293T cells. In the figure, 1-9 respectively represent, 1: expression effect of unmodified luciferase mRNA; 2: expression effect of luciferase mRNA replaced by N6-methyladenosine; 3: expression effect of luciferase mRNA replaced by N6-acetyladenosine; 4: expression effect of luciferase mRNA replaced by N6-propionyladenosine; 5: expression effect of luciferase mRNA replaced by N6-butyryladenosine; 6: expression effect of luciferase mRNA replaced by N6-pentanoyladenosine; 7: expression effect of luciferase mRNA replaced by N6-hexanoyladenosine; 8: expression effect of luciferase mRNA replaced by 2-aminoadenosine; 9: expression effect of luciferase mRNA replaced by 7-deazaadenosine. As can be seen from Figure 34, the translation efficiency of luciferase mRNA modified by replacing natural adenosine with N6-acetyladenosine in HEK 293T cells is nearly 3 times higher than that of unmodified mRNA.
图80是化学修饰的腺苷和假尿苷联用的荧光素酶mRNA在转染进HeLa细胞后24h表达的效果图。图中,1-6分别表示,1:无修饰的荧光素酶mRNA的表达效果图;2:假尿苷替换的荧光素酶mRNA的表达效果图;3:N6-乙酰基腺苷和假尿苷联用替换的荧光素酶mRNA的表达效果图;4:N6-丙酰基腺苷和假尿苷联用替换的荧光素酶mRNA的表达效果图;5:N6-丁酰基腺苷和假尿苷联用替换的荧光素酶mRNA的表达效果图;6:2-氨基腺苷和假尿苷联用替换的荧光素酶mRNA的表达效果图。结果表明,将化学修饰的腺苷和假尿苷联用时,修饰的mRNA在细胞体系中的表达效率更高,超过假尿苷单独修饰的mRNA的表达效率,因此化学修饰的腺苷可以与假尿苷联用以进一步增强修饰的mRNA的表达效率,作为假尿苷修饰的补充选择。Figure 80 is a graph showing the expression of chemically modified adenosine and pseudouridine combined luciferase mRNA 24h after transfection into HeLa cells. In the figure, 1-6 respectively represent, 1: expression effect graph of unmodified luciferase mRNA; 2: expression effect graph of luciferase mRNA replaced by pseudouridine; 3: expression effect graph of luciferase mRNA replaced by N6-acetyladenosine and pseudouridine; 4: expression effect graph of luciferase mRNA replaced by N6-propionyladenosine and pseudouridine; 5: expression effect graph of luciferase mRNA replaced by N6-butyryladenosine and pseudouridine; 6: expression effect graph of luciferase mRNA replaced by 2-aminoadenosine and pseudouridine. The results showed that when chemically modified adenosine and pseudouridine were used together, the expression efficiency of the modified mRNA in the cell system was higher, exceeding the expression efficiency of mRNA modified by pseudouridine alone. Therefore, chemically modified adenosine can be used in combination with pseudouridine to further enhance the expression efficiency of the modified mRNA as a supplementary option for pseudouridine modification.
实施例46含有化学修饰的多聚腺苷尾巴的eGFP mRNA在细胞中的表达效率Example 46 Expression efficiency of eGFP mRNA containing a chemically modified polyadenosine tail in cells
合成含有化学修饰的多聚腺苷尾巴的eGFP mRNA,检测其在细胞中的表达效率;将其转染进细胞之后,翻译成目标蛋白增强绿色荧光蛋白,利用激光共聚焦荧光显微镜检测其荧光强度,以此确定含有天然的和化学修饰的多聚腺苷尾巴的eGFP mRNA的翻译效率。eGFP mRNA containing a chemically modified polyadenosine tail was synthesized and its expression efficiency in cells was tested. After being transfected into cells, it was translated into the target protein enhanced green fluorescent protein, and its fluorescence intensity was detected using a laser confocal fluorescence microscope to determine the translation efficiency of eGFP mRNA containing natural and chemically modified polyadenosine tails.
化学修饰的mRNA转染HEK 293T细胞的具体步骤如下:The specific steps for transfecting HEK 293T cells with chemically modified mRNA are as follows:
(1)细胞转染(1) Cell transfection
接种完人胚肾293T细胞(购自中国科学院细胞库)后约24h,观察35mm孔板内的细胞状态,汇合度在70%。在生物安全柜内,配制90%(体积百分含量)DMEM+10%(体积百分含量)FBS培养基。转染前30分钟弃掉孔板的培养基,每孔加入3mL新鲜培养基,即90%(体积百分含量)DMEM+10%(体积百分含量)FBS培养基。About 24 hours after inoculation of human embryonic kidney 293T cells (purchased from the Chinese Academy of Sciences Cell Bank), the cell state in the 35mm well plate was observed, and the confluence was 70%. In a biological safety cabinet, 90% (volume percentage) DMEM + 10% (volume percentage) FBS culture medium was prepared. 30 minutes before transfection, the culture medium in the well plate was discarded, and 3 mL of fresh culture medium, i.e. 90% (volume percentage) DMEM + 10% (volume percentage) FBS culture medium, was added to each well.
(2)配制转染体系(2) Preparation of transfection system
取50μL Opti-MEM,加入800ng mRNA,混合均匀。另外准备2μL Lipofectamine 2000(脂质体2000)与50μL Opti-MEM的混合溶液,室温孵育10分钟。然后将脂质体2000混合液加入到mRNA混合液中混合均匀,孵育20分钟。将混合后的溶液加入到孔板中。转染后6h换液,吸掉旧的培养基,每孔换为3mL新鲜培养基,即90%(体积百分含量)DMEM+10%(体积百分含量)FBS培养基。转染后24h/48h更换新鲜培养基,以激光共聚焦荧光显微镜对细胞荧光强度进行检测。 Take 50μL Opti-MEM, add 800ng mRNA, and mix well. Also prepare a mixed solution of 2μL Lipofectamine 2000 (liposome 2000) and 50μL Opti-MEM, and incubate at room temperature for 10 minutes. Then add the liposome 2000 mixture to the mRNA mixture, mix well, and incubate for 20 minutes. Add the mixed solution to the well plate. Change the medium 6h after transfection, aspirate the old culture medium, and replace each well with 3mL of fresh culture medium, that is, 90% (volume percentage) DMEM + 10% (volume percentage) FBS culture medium. Replace the fresh culture medium 24h/48h after transfection, and detect the fluorescence intensity of the cells with a laser confocal fluorescence microscope.
图81为化学修饰的腺苷多聚腺苷化修饰的eGFP mRNA在HEK 293T细胞中表达效果的荧光成像图,图中,1表示无poly(A)尾的eGFP mRNA表达效果的荧光成像图;2表示具有多聚天然腺苷尾的eGFP mRNA表达效果的荧光成像图;3表示具有多聚N6-乙酰基腺苷尾的eGFP mRNA表达效果的荧光成像图;4表示具有多聚N6-丙酰基腺苷尾的eGFP mRNA表达效果的荧光成像图,图中比例尺为50μm。Figure 81 is a fluorescence imaging diagram of the expression effect of chemically modified adenosine polyadenylation-modified eGFP mRNA in HEK 293T cells. In the figure, 1 represents the fluorescence imaging diagram of the expression effect of eGFP mRNA without poly (A) tail; 2 represents the fluorescence imaging diagram of the expression effect of eGFP mRNA with poly natural adenosine tail; 3 represents the fluorescence imaging diagram of the expression effect of eGFP mRNA with poly N6-acetyl adenosine tail; 4 represents the fluorescence imaging diagram of the expression effect of eGFP mRNA with poly N6-propionyl adenosine tail. The scale bar in the figure is 50 μm.
从表81可知,具有多聚N6-乙酰基腺苷进行尾的eGFP mRNA在转染进HEK 293T细胞后荧光强度更高,蛋白表达效率超过具有多聚天然腺苷尾的mRNA。As can be seen from Table 81, the eGFP mRNA with a poly-N6-acetyladenosine tail has a higher fluorescence intensity after being transfected into HEK 293T cells, and the protein expression efficiency exceeds that of the mRNA with a poly-natural adenosine tail.
综上,本申请提供了一种化学修饰核苷和化学修饰核苷三磷酸,为mRNA修饰技术提供补充和新选择。以所述化学修饰胞嘧啶核苷三磷酸全局替换天然胞嘧啶核苷三磷酸,得到化学修饰的mRNA,所述化学修饰的mRNA在无细胞体系和细胞内均具有更高的翻译效率,且更稳定。以所述化学修饰腺嘌呤核苷三磷酸全局替换mRNA中的天然腺嘌呤核苷,或者对mRNA进行多聚腺苷化加尾修饰,能够增强目标mRNA的翻译效率和稳定性,降低免疫原性,为mRNA修饰技术提供补充和新选择。In summary, the present application provides a chemically modified nucleoside and a chemically modified nucleoside triphosphate, which provide supplements and new options for mRNA modification technology. The chemically modified cytosine triphosphate is used to globally replace the natural cytosine triphosphate to obtain a chemically modified mRNA, which has a higher translation efficiency in both the cell-free system and the cell, and is more stable. The natural adenine nucleosides in the mRNA are globally replaced with the chemically modified adenine triphosphate, or the mRNA is polyadenylated and tailed, which can enhance the translation efficiency and stability of the target mRNA, reduce immunogenicity, and provide supplements and new options for mRNA modification technology.
申请人声明,以上所述仅为本申请的具体实施方式,但本申请的保护范围并不局限于此,所属技术领域的技术人员应该明了,任何属于本技术领域的技术人员在本申请揭露的技术范围内,可轻易想到的变化或替换,均落在本申请的保护范围和公开范围之内。 The applicant declares that the above is only a specific implementation method of the present application, but the protection scope of the present application is not limited thereto. Technical personnel in the relevant technical field should understand that any changes or substitutions that can be easily thought of by technical personnel in the relevant technical field within the technical scope disclosed in the present application are within the protection scope and disclosure scope of the present application.
Claims (13)
A chemically modified nucleoside selected from a compound having a structure as shown in Formula 1, a salt thereof or an isomer thereof:
Y is selected from a cytosine ring containing a substituent or an adenine ring containing a substituent, wherein the cytosine ring containing a substituent is as shown in Formula 2, and the adenine ring containing a substituent is as shown in Formula 3. Indicates the connection location;
The chemically modified nucleoside according to claim 1, wherein the chemically modified nucleoside is a chemically modified cytosine nucleoside, and the chemically modified cytosine nucleoside is selected from a compound having a structure as shown in Formula 4, a salt thereof, or an isomer thereof:
The chemically modified nucleoside according to claim 1, wherein the chemically modified nucleoside is a chemically modified adenine nucleoside. The chemically modified adenine nucleoside is selected from a compound having a structure as shown in Formula 5, a salt thereof or an isomer thereof:
A chemically modified nucleoside triphosphate selected from a compound having a structure as shown in Formula 6, a salt thereof or an isomer thereof:
W is selected from a cytosine ring containing a substituent or an adenine ring containing a substituent, wherein the cytosine ring containing a substituent is as shown in Formula 7, and the adenine ring containing a substituent is as shown in Formula 8. Indicates the connection location;
The chemically modified nucleoside triphosphate according to claim 4, wherein the chemically modified nucleoside triphosphate is a chemically modified cytosine triphosphate, and the chemically modified cytosine triphosphate is selected from a compound having a structure as shown in Formula 9, a salt thereof, or an isomer thereof:
The chemically modified nucleoside triphosphate according to claim 4, wherein the chemically modified nucleoside triphosphate is a chemically modified adenine nucleoside triphosphate, and the chemically modified adenine nucleoside triphosphate is selected from a compound having a structure as shown in Formula 10, a salt thereof, or an isomer thereof:
The chemically modified nucleoside triphosphate according to claim 5, wherein the chemically modified nucleoside triphosphate is prepared by the following preparation method:
The chemically modified nucleoside triphosphate according to claim 6, wherein the chemically modified nucleoside triphosphate is prepared by the following preparation method:
Applications Claiming Priority (4)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| CN202310655937.1A CN119080855A (en) | 2023-06-05 | 2023-06-05 | A chemically modified cytosine triphosphate and its preparation method and application |
| CN202310655937.1 | 2023-06-05 | ||
| CN202311722227.2 | 2023-12-14 | ||
| CN202311722227.2A CN120157727A (en) | 2023-12-14 | 2023-12-14 | A chemically modified adenine nucleoside triphosphate and its preparation method and application |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2024250923A1 true WO2024250923A1 (en) | 2024-12-12 |
Family
ID=93795002
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/CN2024/093272 Pending WO2024250923A1 (en) | 2023-06-05 | 2024-05-15 | Chemically modified nucleoside, chemically modified nucleoside triphosphate and use thereof |
Country Status (1)
| Country | Link |
|---|---|
| WO (1) | WO2024250923A1 (en) |
Citations (8)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| CN103974724A (en) * | 2011-10-03 | 2014-08-06 | 现代治疗公司 | Modified nucleosides, nucleotides and nucleic acids and uses thereof |
| CN104968354A (en) * | 2011-12-21 | 2015-10-07 | 现代治疗公司 | Methods of increasing the viability or longevity of an organ or organ explant |
| US20150307542A1 (en) * | 2012-10-03 | 2015-10-29 | Moderna Therapeutics, Inc. | Modified nucleic acid molecules and uses thereof |
| WO2020229831A1 (en) * | 2019-05-14 | 2020-11-19 | Nuclera Nucleics Ltd | Nucleic acid polymer with amine-masked bases |
| CN112921014A (en) * | 2019-12-05 | 2021-06-08 | 左炽健 | T7RNA polymerase mutant, mRNA, gene, expression vector and cell |
| CN114656511A (en) * | 2022-03-29 | 2022-06-24 | 上海吉量医药工程有限公司 | Preparation method of acetylated cytosine triphosphate and intermediate thereof |
| CN114807154A (en) * | 2020-12-10 | 2022-07-29 | 深圳市瑞吉生物科技有限公司 | Modified nucleic acid and application thereof |
| WO2022235838A1 (en) * | 2021-05-07 | 2022-11-10 | Helix Nanotechnologies, Inc. | Modified ribonucleic acids and uses thereof |
-
2024
- 2024-05-15 WO PCT/CN2024/093272 patent/WO2024250923A1/en active Pending
Patent Citations (8)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| CN103974724A (en) * | 2011-10-03 | 2014-08-06 | 现代治疗公司 | Modified nucleosides, nucleotides and nucleic acids and uses thereof |
| CN104968354A (en) * | 2011-12-21 | 2015-10-07 | 现代治疗公司 | Methods of increasing the viability or longevity of an organ or organ explant |
| US20150307542A1 (en) * | 2012-10-03 | 2015-10-29 | Moderna Therapeutics, Inc. | Modified nucleic acid molecules and uses thereof |
| WO2020229831A1 (en) * | 2019-05-14 | 2020-11-19 | Nuclera Nucleics Ltd | Nucleic acid polymer with amine-masked bases |
| CN112921014A (en) * | 2019-12-05 | 2021-06-08 | 左炽健 | T7RNA polymerase mutant, mRNA, gene, expression vector and cell |
| CN114807154A (en) * | 2020-12-10 | 2022-07-29 | 深圳市瑞吉生物科技有限公司 | Modified nucleic acid and application thereof |
| WO2022235838A1 (en) * | 2021-05-07 | 2022-11-10 | Helix Nanotechnologies, Inc. | Modified ribonucleic acids and uses thereof |
| CN114656511A (en) * | 2022-03-29 | 2022-06-24 | 上海吉量医药工程有限公司 | Preparation method of acetylated cytosine triphosphate and intermediate thereof |
Non-Patent Citations (4)
| Title |
|---|
| A. GABBAI, T. POSTERNAK: "Nucleotide Derivatives of Biological Interest. IX. Enzymic Assay with ATP Analogs", HELVETICA CHIMICA ACTA, VERLAG HELVETICA CHIMICA ACTA., HOBOKEN, USA, vol. 54, no. 7, 30 August 1971 (1971-08-30), Hoboken, USA, pages 2141 - 2142, XP009559339, ISSN: 0018-019X * |
| DATABASE Registry 16 November 1984 (1984-11-16), ANONYMOUS: "Heptanamide, N-(1-β-D-arabinofuranosyl-1,2-dihydro-2-oxo-4- pyrimidinyl)- (CA INDEX NAME)", XP093247990, Database accession no. 55726-40-4 * |
| HAMPTON ALEXANDER, HAMPTON LEWIS A SLOTIN: "Inactivation of Rabbit, Pig, and Carp Adenylate Kinases by N6-oand /7-Fluorobenzoyladenosine S'-Triphosphates* 1"", BIOCHEMISTRY, vol. 14, no. 25, 1 January 1975 (1975-01-01), pages 5438 - 5444, XP093247984 * |
| JEVGENIJA JAKUBOVSKA, DAIVA TAURAITė, LUKAS BIRšTONAS, ROLANDAS MEšKYS: "N 4-acyl-2′-deoxycytidine-5′-triphosphates for the enzymatic synthesis of modified DNA", NUCLEIC ACIDS RESEARCH, OXFORD UNIVERSITY PRESS, GB, vol. 46, no. 12, 6 July 2018 (2018-07-06), GB , pages 5911 - 5923, XP055746184, ISSN: 0305-1048, DOI: 10.1093/nar/gky435 * |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| JP7019639B2 (en) | Modified nucleosides, nucleotides and nucleic acids, and how to use them | |
| CN111778254B (en) | Small interfering nucleic acid for inhibiting novel coronavirus, composition and application | |
| EP2931319B1 (en) | Modified nucleic acid molecules and uses thereof | |
| JP2017500034A (en) | Modified nucleic acid molecules and uses thereof | |
| JP5603933B2 (en) | Nucleic acid aptamer capable of specifically binding to pancreatic cancer cells or tissues and use thereof | |
| CN114853836B (en) | Initial capping oligonucleotide primer containing GNA structure and preparation method and application thereof | |
| WO2024188370A1 (en) | Capped polynucleotide, capped mrna and composition thereof, pharmaceutical protein, and preparation method therefor and use thereof, and pharmaceutical preparation | |
| Hu et al. | TLR8 activation and inhibition by guanosine analogs in RNA: Importance of functional groups and chain length | |
| WO2024250923A1 (en) | Chemically modified nucleoside, chemically modified nucleoside triphosphate and use thereof | |
| WO2024250607A1 (en) | Chemically modified cytidine triphosphate and preparation method therefor and use thereof | |
| CN108948109B (en) | Raffinolactone glycoside compound and application thereof | |
| CN119060138B (en) | Targeting Nectin-4 cyclic peptide and preparation method and application thereof | |
| WO2024153245A1 (en) | Compound for rna capping and use thereof | |
| CN117813383A (en) | SiRNA for inhibiting CIDEB gene expression, medicine and application thereof | |
| WO2023246860A1 (en) | Initially capped oligonucleotide primer, method for preparing same, and use thereof | |
| CN119177266A (en) | In vitro transcription buffer system, in vitro transcription solution containing same and in vitro transcription method using same | |
| Beasley | Synthetic Strategies towards the Development of New RNA Structure and RNA Labeling Tools | |
| CN117820388A (en) | A deuterated capping compound for mRNA synthesis and its preparation method and application | |
| CN118834250A (en) | Novel delivery vehicle | |
| CN120665128A (en) | Compounds, pharmaceutical compositions, kits for capping RNA transcripts, and methods of in vitro transcription | |
| HK40032374B (en) | Modified nucleosides, nucleotides, and nucleic acids, and uses thereof | |
| HK40032374A (en) | Modified nucleosides, nucleotides, and nucleic acids, and uses thereof | |
| HK40009706A (en) | Modified nucleosides, nucleotides, and nucleic acids, and uses thereof | |
| HK40009706B (en) | Modified nucleosides, nucleotides, and nucleic acids, and uses thereof | |
| HK1200730B (en) | Modified nucleosides, nucleotides, and nucleic acids, and uses thereof |
Legal Events
| Date | Code | Title | Description |
|---|---|---|---|
| 121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 24818456 Country of ref document: EP Kind code of ref document: A1 |