WO2019071474A1 - Modified nucleoside or nucleotide - Google Patents

Modified nucleoside or nucleotide Download PDF

Info

Publication number
WO2019071474A1
WO2019071474A1 PCT/CN2017/105734 CN2017105734W WO2019071474A1 WO 2019071474 A1 WO2019071474 A1 WO 2019071474A1 CN 2017105734 W CN2017105734 W CN 2017105734W WO 2019071474 A1 WO2019071474 A1 WO 2019071474A1
Authority
WO
WIPO (PCT)
Prior art keywords
nucleic acid
compound
group
polymerase
kod
Prior art date
Application number
PCT/CN2017/105734
Other languages
French (fr)
Chinese (zh)
Inventor
刘二凯
陈奥
章文蔚
Original Assignee
深圳华大智造科技有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 深圳华大智造科技有限公司 filed Critical 深圳华大智造科技有限公司
Priority to PCT/CN2017/105734 priority Critical patent/WO2019071474A1/en
Priority to CN201780090915.8A priority patent/CN110650968B/en
Publication of WO2019071474A1 publication Critical patent/WO2019071474A1/en

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H19/00Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof
    • C07H19/02Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof sharing nitrogen
    • C07H19/04Heterocyclic radicals containing only nitrogen atoms as ring hetero atom
    • C07H19/06Pyrimidine radicals
    • C07H19/10Pyrimidine radicals with the saccharide radical esterified by phosphoric or polyphosphoric acids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids

Definitions

  • the invention relates to the field of nucleic acid sequencing.
  • the invention provides a modified nucleoside or nucleotide, the 3'-OH of the modified nucleoside or nucleotide is reversibly blocked, and the modified nucleoside or nucleotide Carry a detectable mark.
  • the invention also relates to a kit comprising the nucleoside or nucleotide, a method of making the nucleoside or nucleotide, and a sequencing method based on the nucleoside or nucleotide.
  • DNA sequencing technology includes the first generation DNA sequencing technology represented by Sanger sequencing, and the second generation DNA sequencing technology represented by Illumina Hiseq 2500, Roche 454, ABI Solid, BGI SEQ-500, and the like.
  • the Sanger sequencing method has the characteristics of simple experimental operation, intuitive and accurate results, and short experimental period. It has wide application in the fields of clinical gene mutation detection and genotyping, which require high timeliness of detection results.
  • the disadvantages of the Sanger sequencing method are small throughput and high cost, which limits its application in large-scale gene sequencing.
  • the second generation DNA sequencing technology Compared with the first generation of DNA sequencing technology, the second generation DNA sequencing technology has the characteristics of large sequencing throughput, low cost, high degree of automation and single molecule sequencing.
  • the sequencing technology of Hiseq 2500V2 as an example, an experimental procedure can generate 10-200 G base data, and the average cost per base is less than 1/1000 of the sequencing cost of the Sanger sequencing method, and the obtained sequencing result is obtained. It can be processed and analyzed directly by computer. Therefore, second generation DNA sequencing technology is very suitable for large-scale sequencing.
  • the most critical part of the second-generation sequencing technology is sequencing by side synthesis.
  • the usual approach is to reversibly block the deoxyribonucleoside triphosphate (dNTP).
  • dNTP deoxyribonucleoside triphosphate
  • this process involves: catalysis by polymerase, Incorporating a dNTP with a reversible blocking group and a detectable label into a growing nucleic acid strand, wherein the reversible blocking group protects the 3'-OH of the dNTP, preventing the dNTP that has been incorporated into the nucleic acid strand from passing through 3' -OH reacts with free dNTPs; detectable labels are detected to determine the type of dNTP introduced; thereafter, the blocking groups and detectable labels on the introduced dNTPs are removed and the next round of sequencing is initiated.
  • Illumina blocks the 3'-OH by azide methylene, and after polymerization and detection, the azide is removed using an organic phosphine. Methylene, release 3'-OH, so that the next cycle can be sequenced;
  • Columbia University Jingyue Ju research group uses allyl block 3'-OH, the process uses metal palladium and triphenylphosphine triple sulfonate
  • the ligand of the acid trisodium salt removes the blocking group by a palladium-catalyzed deallyl reaction; in addition, researchers have used the carbonate and sulfur-sulfur bond to reversibly block, in the cut After the sulfur-sulfur bond is broken, the resulting mercapto group can cleave the carbonate to which the 3'-OH is attached, thereby completing the deblocking.
  • the blocking method is very strict.
  • Illumina's blocking method using azide methylene is successfully used for sequencing, and other methods. It faces a variety of problems in use and has not been successfully applied. The reasons for this situation include: 1) DNA sequencing reactions must be carried out in a near neutral aqueous system, which makes many chemical reactions and blocking groups unusable; 2) the groups generated after blocking must be very Stable, and the resection reaction needs to be rapid and efficient, which also eliminates many types of reactions, such as the above carbonate and sulfur-sulfur bond methods, which are difficult to apply due to the fast resection rate; 3) the resection reaction does not affect the sequencing system.
  • the allyl group is used as a blocking group, and it needs to be catalyzed by a palladium metal complex during excision. In practical applications, it may be unsuitable for the chip, and eventually no mature product appears on the market. .
  • Illumina optimizes the azidomethylene, which increases the stability, but although the resection efficiency can reach 100%, it still requires a higher temperature, and the excision reagent is not good for DNA. influences.
  • Illumina uses a phospho-blocked dNTP for sequencing while synthesizing, but it is clearly shown in the patent that only the hydroxyl groups on the phosphoric acid are all protected before being polymerized by the polymerase, and the hydroxyl groups are all The protected phosphate group cannot be cleaved by endonuclease IV.
  • the method disclosed in the patent comprises: polymerizing a dNTP in which 3'-OH is blocked by a phosphate group, wherein the hydroxyl group on the phosphate group is completely protected, followed by stepwise excision, that is, first removing the phosphate group A hydroxy protecting group on the cleavage, followed by excision of the entire phosphate group by endonuclease IV, wherein the method used to remove a protecting group includes photocleavage, and Ada (methyltransferase) removes the methyl group.
  • the method disclosed in the patent comprises: polymerizing a dNTP in which 3'-OH is blocked by a phosphate group, wherein the phosphate group, although theoretically feasible, is very cumbersome in practical use.
  • the inventors of the present application have developed a novel 3'-OH reversibly blocked nucleotide which has higher stability, can be efficiently or even completely polymerized by a polymerase, and can be Efficient or even 100% resection without damage to DNA.
  • the inventors of the present application have developed a novel modified nucleoside or nucleotide in which 3'-OH is blocked by a blocking group, and the blocking group can be cleaved without destroying the nucleic acid strand.
  • the reagent is excised to form a free 3'-OH.
  • the modified nucleoside or nucleotide also carries a detectable label, and the detectable label is linked to the base in the nucleotide by a linker comprising a phosphodiester bond, and the excision reagent can be utilized (with excision)
  • the excision reagent of the blocking group is the same or different), and the phosphodiester bond is cleaved to remove the detectable label without destroying the nucleic acid strand.
  • the application provides a compound having the structure of formula (I),
  • R 1 and R 3 are each independently selected from
  • A is selected from the group consisting of phenyl, naphthyl, anthryl and pyridyl;
  • R 2 is selected from the group consisting of a nitro group, a halogenated C 1-4 alkyl group, a halogen, a hydrogen, an aldehyde group, Wherein Q is independently selected from C 1-4 alkyl;
  • R 4 is selected from -H, a monophosphate group (-PO 3 H 2 ), a diphosphate group (-PO 3 H-PO 3 H 2 ), a triphosphate group (-PO 3 H-PO 3 H-PO 3 H 2 ) and a tetraphosphoric acid group (-PO 3 H-PO 3 H-PO 3 H-PO 3 H 2 );
  • R 6 is hydrogen or a hydroxyl group
  • n are each independently selected from 0, 1, 2, 3, 4, 5;
  • L is a linking group or does not exist
  • Base represents a base, such as a purine base or a pyrimidine base, for example, one selected from the group consisting of A, T, U, C, and G;
  • Label indicates a detectable label, such as a fluorophore
  • Blocker indicates a blocking group.
  • the application also provides a method of making a modified nucleoside or nucleotide as described above.
  • sequencing is performed while synthesizing a polynucleotide in which a target single-stranded polynucleotide is complementary to each other.
  • the application provides a method of preparing a growing polynucleotide complementary to a target single-stranded polynucleotide in a sequencing reaction, comprising incorporating a compound as defined above into the complementary of said growth A polynucleotide, wherein the incorporation of the compound prevents any subsequent nucleotides from being introduced into the growing complementary polynucleotide.
  • the application provides a method of determining the sequence of a single-stranded polynucleotide of interest, comprising: monitoring sequential incorporation of complementary nucleotides, wherein at least one complementary nucleotide that is incorporated is as described above A defined compound, and detecting a detectable label carried by the compound.
  • the method of determining the sequence of a single-stranded polynucleotide of interest comprises:
  • duplex comprising a growing nucleic acid strand and a nucleic acid molecule to be sequenced
  • the reaction cycle further comprises the step (iv) of removing the detectable label on the nucleic acid intermediate using a scavenging reagent.
  • the ablation reagents used in steps (iii) and (iv) are the same reagents. In certain embodiments, the ablation reagents used in steps (iii) and (iv) are different reagents.
  • the invention provides a kit comprising first, second, third and fourth compounds, each of said first, second, third and fourth compounds being of the formula A compound of I) which is a derivative of nucleotides A, (T/U), C and G, respectively, and which has a base complementary pairing ability.
  • Example 1 is a process in which a dTTP having a Cy3-tag and a 3'-OH is blocked is prepared in Example 1, and is involved in HPLC chromatogram of intermediate compound IV-1.
  • Figure 2 is an HPLC chromatogram of dTTP with Cy3-labeled and 3'-OH blocked in Example 1.
  • Figure 3 is an HPLC chart of the intermediate compound IV-2 involved in the preparation of dATP with Cy3-labeled and 3'-OH blocked in Example 2.
  • Fig. 4 exemplarily illustrates the process of determining the polymerization efficiency of dTTP in Example 3.
  • Fig. 5 exemplarily illustrates the process of determining the excision efficiency of the blocking group in Example 4.
  • Figure 6 shows the efficiency of the blocking group being cleaved at different excision times in Example 4.
  • Fig. 7 exemplarily illustrates the process of determining the excision efficiency of the fluorophore in Example 5.
  • Figure 8 shows the efficiency of fluorophore excision at different cut-off times in Example 5.
  • Figure 9 is an LC spectrum of dTTP blocked by methyl phosphate in Example 6 after stability testing.
  • Figure 10 is a LC map of the azide methylene-blocked dTTP in Example 6 after stability testing.
  • Sequence 1 (SEQ ID NO: 1): 33 bp
  • Sequence 3 (SEQ ID NO: 3): 32 bp
  • C 1-4 alkyl refers to a straight or branched alkyl group having from 1 to 4 carbon atoms, including but not limited to C 1-2 alkyl, C 1-3 Alkyl, C 2-4 alkyl, for example: methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, isobutyl, tert-butyl and the like.
  • halo refers to the replacement of a hydrogen on a group or compound by one or more (eg, 2, 3, 4, 5, 6, 7, 8, 9) halogen atoms, including Fully halogenated and partially halogenated.
  • halogen includes fluoro, chloro, bromo, iodo.
  • halo C 1-4 alkyl refers to a radical derived from one or more halogen atoms substituted by one or more hydrogen atoms on a C 1-4 alkyl group, said "halogen” And “C 1-4 alkyl” are as defined above.
  • Halo C 1-4 alkyl includes, but is not limited to, halo C 1-2 alkyl, halo C 1-3 alkyl, halo C 2-4 alkyl, such as halomethyl (eg fluoromethyl) , chloromethyl), haloethyl (eg fluoroethyl, chloroethyl), halopropyl, haloisopropyl, halo-n-butyl, halo sec-butyl, halogenated Isobutyl, halogenated tert-butyl.
  • blocking refers to the formation of a 3'-OH formation of a nucleoside or nucleotide using a specific group to polymerize a possible polymerase (eg, a DNA polymerase). termination.
  • the group that is used for blocking is referred to as a "blocking group.”
  • the blocking group can be removed such that the blocked hydroxyl group can be converted to a reactive hydroxyl group, such blockage being referred to as "reversible blocking.”
  • the group used for reversible blocking is referred to as a "reversible blocking group.”
  • the term "support” refers to any material (solid or semi-solid) that allows for stable attachment of nucleic acids, such as latex beads, dextran beads, polystyrene, polypropylene, polyacrylamide gels, Gold thin layers, glass and silicon wafers.
  • the support is optically clear, such as glass.
  • stable attachment means that the linkage between the nucleic acid molecule and the support is sufficiently strong that the nucleic acid molecule is not subjected to various reaction or treatment conditions (eg, polymerization and washing treatment). Detach the support.
  • the term "connected" is intended to cover any form of linkage, such as covalent linkages and non-covalent linkages.
  • the nucleic acid molecule is preferably linked to the support by a covalent means.
  • fragmentation refers to the process of converting a large nucleic acid fragment (eg, a large DNA fragment) into a small nucleic acid fragment (eg, a small DNA fragment).
  • large nucleic acid fragment is intended to encompass nucleic acid molecules (eg, DNA) greater than 5 kb, greater than 10 kb, greater than 25 kb, eg, greater than 500 kb, greater than 1 Mb, greater than 5 Mb or greater nucleic acid molecules (eg, DNA) ).
  • end-filled refers to the process of complementing the ends of a nucleic acid molecule having an overhanging end to form a nucleic acid molecule having a blunt end.
  • linker and “linker sequence” are used interchangeably.
  • linker and linker sequence refer to a stretch of oligonucleotide sequences introduced human at the 5' and/or 3' end of a nucleic acid molecule.
  • a connector can typically contain one or more regions for achieving a particular function.
  • the linker when a linker is introduced artificially at the 5' and/or 3' end of the nucleic acid molecule, the linker will be able to perform the specific function, thereby facilitating subsequent applications.
  • the linker can comprise one or more primer binding regions to facilitate binding of the primers.
  • the linker may comprise one or more primer binding regions, such as a primer binding region capable of hybridizing to a primer for amplification, and/or capable of hybridizing to a primer for use in a sequencing reaction. Primer binding region.
  • the linker comprises a universal linker sequence capable of hybridizing to a universal primer, for example, a universal linker sequence capable of hybridizing to a universal amplification primer and/or a universal sequencing primer.
  • the nucleic acid molecule carrying the linker can be conveniently amplified and/or sequenced by using universal amplification primers and/or universal sequencing primers.
  • the linker can also comprise a tag or tag sequence.
  • the terms “tag” and “tag sequence” are used interchangeably.
  • the terms “tag” and “tag sequence” refer to a segment of an oligonucleotide sequence introduced at the 5' and/or 3' end of a nucleic acid molecule with a particular base sequence. Labels are commonly used to identify/distinguish the source of nucleic acid molecules.
  • different tags can be introduced in different nucleic acid molecules from different sources, whereby when these nucleic acid molecules of different origin are mixed together, each nucleic acid molecule can be accurately determined by the unique tag sequence carried on each nucleic acid molecule. origin of.
  • the tag sequence can have any length, such as 2-50 bp, such as 2, 3, 4, 5, 10, 15, 20, 25, 30, 35, 40, 45 or 50 bp, depending on actual needs.
  • hybridization generally refers to hybridization under stringent conditions.
  • the stringent conditions include, for example, moderate stringency conditions (eg, hybridization at about 45 ° C in 6 x sodium chloride / sodium citrate (SSC), then in 0.2 x SSC / 0.1% SDS One or more washes at about 50-65 ° C; high stringency conditions (eg, hybridization at about 45 ° C in 6 x SSC, then at about 68 ° C in 0.1 x SSC / 0.2% SDS or Multiple washes; and other stringent hybridization conditions known to those skilled in the art (see, for example, Ausubel, FM et al, 1989, Current Protocols in Molecular Biology, Vol. 1, Green Publishing Associates, Inc., and John Wiley & Sons, Inc. New York, pages 6.3.1-6.3.6 and 2.10.3).
  • reaction system containing a solution phase and a solid phase means that the reaction system of the present invention comprises both a support and a substance attached to the support (solid phase), and a solution/solvent dissolved in the solution/solvent.
  • removing the solution phase of the reaction system means removing the solution in the reaction system and the substance (solution phase) contained therein, and retaining only the support in the reaction system and the substance attached to the support. Quality (solid phase).
  • the substance (solid phase) attached to the support may comprise a nucleic acid molecule to be sequenced, a growing nucleic acid strand, and/or a duplex formed by the nucleic acid molecule to be sequenced and the growing nucleic acid strand.
  • primer refers to an oligonucleotide sequence that hybridizes to a complementary sequence and initiates a specific polymerization reaction.
  • sequence of the primer is selected/designed to have maximum hybridization activity for the complementary sequence, while having very low non-specific hybridization activity for other sequences, thereby minimizing non-specific amplification.
  • Methods for designing primers are well known to those skilled in the art and can be performed using commercially available software (e.g., Primer Premier version 6.0, Oligo version 7.36, etc.).
  • polymerase refers to an enzyme capable of performing a nucleotide polymerization reaction. Such an enzyme is capable of introducing a nucleotide paired with a nucleotide at a position corresponding to a template nucleic acid at the 3' end of the growing nucleic acid strand according to the principle of base complementary pairing.
  • the expressions "A, (T/U), C, and G” are intended to cover two instances: “A, T, C, and G” and "A, U, C, and G.”
  • the expression “the four compounds are derivatives of nucleotides A, (T/U), C and G, respectively” is intended to mean that the four compounds are nucleotides A, T, C and G, respectively. Derivatives, or derivatives of nucleotides A, U, C and G, respectively.
  • a compound having a base complementary pairing ability means that the compound is capable of pairing with a corresponding base and forming a hydrogen bond according to the principle of base complementary pairing.
  • base A can be paired with base T or U
  • base G can be paired with base C.
  • a compound having a base complementary pairing ability when a compound having a base complementary pairing ability is a derivative of nucleotide A, it will be able to pair with a base T or U; when a compound having a base complementary pairing ability is a derivative of a nucleotide T or U When it is a substance, it will be able to pair with base A; when a compound having a base complementary pairing ability is a derivative of nucleotide C, it will be able to pair with base G; when a compound having a base complementary pairing ability is In the case of a derivative of nucleotide G, it will be able to pair with base C.
  • the inventors of the present application have developed a novel modified nucleoside or nucleotide in which 3'-OH is blocked by a blocking group, and the blocking group can be cleaved without destroying the nucleic acid strand.
  • the reagent is excised to form a free 3'-OH.
  • the modified nucleoside or nucleotide also carries a detectable label, and the detectable label is linked to the base in the nucleotide by a linker comprising a phosphodiester bond, and the excision reagent can be utilized (with excision)
  • the excision reagent of the blocking group is the same or different), and the phosphodiester bond is cleaved to remove the detectable label without destroying the nucleic acid strand.
  • the application provides a compound having the structure of formula (I),
  • R 1 and R 3 are each independently selected from
  • A is selected from the group consisting of phenyl, naphthyl, anthryl and pyridyl;
  • R 2 is selected from the group consisting of nitro, halo C 1-4 alkyl (eg, fluoro C 1-4 alkyl), halogen (eg, fluorine, chlorine, bromine, iodine), hydrogen, aldehyde, Wherein Q is independently selected from C 1-4 alkyl (eg, C 1-2 alkyl, C 1-3 alkyl, C 2-4 alkyl, such as methyl, ethyl, n-propyl, isopropyl , n-butyl, sec-butyl, isobutyl, tert-butyl);
  • C 1-4 alkyl eg, C 1-2 alkyl, C 1-3 alkyl, C 2-4 alkyl, such as methyl, ethyl, n-propyl, isopropyl , n-butyl, sec-butyl, isobutyl, tert-butyl
  • R 4 is selected from -H, a monophosphate group (-PO 3 H 2 ), a diphosphate group (-PO 3 H-PO 3 H 2 ), a triphosphate group (-PO 3 H-PO 3 H-PO 3 H 2 ) and a tetraphosphoric acid group (-PO 3 H-PO 3 H-PO 3 H-PO 3 H 2 );
  • R 6 is hydrogen or a hydroxyl group
  • n are each independently selected from 0, 1, 2, 3, 4, 5;
  • L is a linking group or does not exist
  • Base represents a base, such as a purine base or a pyrimidine base, for example, one selected from the group consisting of A, T, U, C, and G;
  • Label indicates a detectable label, such as a fluorophore
  • Blocker indicates a blocking group.
  • A is an aromatic group, and after the phosphodiester bond attached thereto is broken, an aromatic ring having a hydroxyl group can be formed, which is advantageous for the stable existence of the cut product;
  • R 1 , R 2 and R 3 are electron withdrawing
  • the base helps the phosphodiester bond between A and R 1 to be cleaved by the excision reagent.
  • R 1 is selected from In certain embodiments, R 1 is
  • R 2 is selected from the group consisting of nitro, trifluoromethyl, fluoro, chloro, hydrogen, and aldehyde. In certain embodiments, R 2 is a nitro group.
  • R 3 is selected from In certain embodiments, R 3 is
  • the compound of the present invention may be a nucleoside or a nucleotide, and thus R 4 may be -H, a monophosphate group (-PO 3 H 2 ), a diphosphate group (-PO 3 H-PO 3 H 2 ), three Phosphoric acid group (-PO 3 H-PO 3 H-PO 3 H 2 ) or tetraphosphoric acid group (-PO 3 H-PO 3 H-PO 3 H-PO 3 H 2 ).
  • R 4 is a monophosphate group (-PO 3 H 2 ), a diphosphate group (-PO 3 H-PO 3 H 2 ), a triphosphate group (-PO 3 H-PO 3 H-PO 3 H 2 ) or tetraphosphate group (-PO 3 H-PO 3 H-PO 3 H-PO 3 H 2 ) nucleotide.
  • the compound is a nucleotide.
  • R 4 is a triphosphate group (-PO 3 H-PO 3 H-PO 3 H 2 ).
  • the compound is a nucleoside triphosphate.
  • R 4 is -H.
  • the compound is a nucleoside.
  • Blocker can have a variety of structures.
  • the structure of the Blocker is:
  • Ra 1 and Ra 2 are each independently selected from the group consisting of H, F, -CF 3 , -CHF 2 , -CH 2 F, -CH 2 W, -COOW, -CONHW; W is selected from C 1 -C 6 alkyl groups. .
  • the structure of the Blocker is:
  • Rb 1 , Rb 2 , Rb 3 , Rb 4 and Rb 5 are each independently selected from H and C 1 -C 6 alkyl.
  • the structure of the Blocker is:
  • Rc 1 and Rc 2 are each independently selected from the group consisting of H, F, Cl and -CF 3 .
  • the structure of the Blocker is:
  • R 5 is selected from C 1-4 alkyl (eg, C 1-2 alkyl, C 1-3 alkyl, C 2-4 alkyl, such as methyl, ethyl, n-propyl, isopropyl, n-Butyl, sec-butyl, isobutyl, tert-butyl).
  • C 1-4 alkyl eg, C 1-2 alkyl, C 1-3 alkyl, C 2-4 alkyl, such as methyl, ethyl, n-propyl, isopropyl, n-Butyl, sec-butyl, isobutyl, tert-butyl.
  • R 5 is selected from C 1-4 alkyl (eg, C 1-2 alkyl, C 1-3 alkyl, C 2-4 alkyl, such as methyl, ethyl, n-propyl, isopropyl, n-Butyl, sec-butyl, isobutyl, tert-butyl).
  • C 1-4 alkyl eg, C 1-2 alkyl, C 1-3 alkyl, C 2-4 alkyl, such as methyl, ethyl, n-propyl, isopropyl, n-Butyl, sec-butyl, isobutyl, tert-butyl.
  • R 5 is methyl or ethyl.
  • a detectable label is linked to a base by a phosphodiester bond; the phosphodiester bond can be cleaved by a scavenging reagent to remove the detectable label from the compound.
  • the detectable label is a fluorophore.
  • fluorophores that can be used in the present invention include, but are not limited to, various known fluorescent labels such as AF532, ALEX-350, FAM, VIC, TET, CAL.
  • the Label in the compounds of the invention is Cy3, Cy3.5, Cy5 or Cy5.5.
  • the compounds of the invention may be deoxyribonucleotides, ribonucleotides, deoxyribonucleosides or ribonucleosides. Therefore, R 6 may be hydrogen or a hydroxyl group. In certain embodiments, R 6 is hydrogen. In this case, the compound is a deoxyribonucleotide or a deoxyribonucleoside.
  • L functions as a linking group in which L may or may not be present.
  • m is one.
  • n is one.
  • L is
  • the compounds of the invention have the structure of formula (II)
  • R 2 in formula (II) is a nitro group.
  • the Label in Formula (II) is Cy3.
  • the compounds of the invention have the structure:
  • An exemplary preparation process includes steps 1 - 3:
  • step 1
  • Step 1 further includes the following steps:
  • Step 1-1 Using Compound I as a starting material, an oxidation reaction is carried out to produce a compound II (intermediate).
  • step 1 comprises: adding methanol and tetrazole to acetonitrile, followed by the addition of a solution in which Compound I is dissolved, and stirring at room temperature to replace the diisopropyl group on compound I with a methoxy group on methanol. Amine; then iodine is added and stirred at room temperature to give compound II.
  • the methanol and/or tetrazole is in excess relative to Compound I.
  • the molar ratio of methanol to Compound I is from 2 to 5:1, for example, 2:1, 3:1, 4:1, or 5:1, such as 3:1.
  • the molar ratio of the tetrazole to Compound I is from 2 to 5:1, for example, 2:1, 3:1, 4:1, or 5:1, such as 3:1.
  • the iodine is added to the reaction system as a solution, for example, a solution comprising water and/or an organic solvent (eg, a solution comprising water, pyridine, and/or tetrahydrofuran).
  • a solution comprising water and/or an organic solvent (eg, a solution comprising water, pyridine, and/or tetrahydrofuran).
  • the iodine is in excess relative to Compound I.
  • the molar ratio of Compound I to iodine is 1: 1.5-5, such as 1:1.5, 1:2, 1:2.5, 1:3, 1:3.5, 1:4, 1:4.5 or 1:5, for example 1:1.5.
  • step 1-1 further comprises: after the oxidation reaction of Compound I is completed, sodium sulfite is added to remove unreacted iodine.
  • step 1-1 further comprises purifying Compound II.
  • Step 1-2 Deprotection of Compound II to give Compound III.
  • step 1-2 comprises mixing compound II with trichloroacetic acid and stirring.
  • step 1-2 comprises: adding compound II to a solution comprising trichloroacetic acid (eg, a solution of trichloroacetic acid in dichloromethane), and stirring at room temperature to provide compound III.
  • a solution comprising trichloroacetic acid eg, a solution of trichloroacetic acid in dichloromethane
  • the trichloroacetic acid is in excess relative to Compound II.
  • steps 1-2 further comprise purifying compound III.
  • Step 1-3 The compound III is subjected to a triphosphate reaction and a deprotection reaction to produce a compound IV.
  • steps 1-3 comprise: adding 2-chloro-4H-1,3,2-benzophosphono-4- to a solution comprising Compound III under argon-protected conditions.
  • the ketone was stirred at room temperature; then tri-n-butylammonium pyrophosphate and n-butylamine were added and stirred at room temperature; then iodine was added and stirred at room temperature.
  • the solution comprising Compound III further comprises 1,4-dioxane and anhydrous pyridine.
  • the 2-chloro-4H-1,3,2-benzodioxan-4-one is added to the solution in the form of a solution (eg, a 1,4-dioxane solution).
  • a solution eg, a 1,4-dioxane solution.
  • the molar ratio of compound III to 2-chloro-4H-1,3,2-benzodioxan-4-one is 1:1-2, such as 1:1.0, 1:1.1. 1:1.2, 1:1.3, 1:1.4, 1:1.5, 1:1.6, 1:1.7, 1:1.8, 1:1.9 or 1:2.0, for example 1:1.1.
  • the tri-n-butylammonium pyrophosphate and/or n-butylamine is added to the reaction system in the form of a solution (eg, N,N-dimethylformamide solution).
  • the tri-n-butylammonium pyrophosphate is in excess relative to Compound III.
  • the molar ratio of Compound III to tri-n-butylammonium pyrophosphate is 1:1.5-5, such as 1:1.5, 1:2, 1:2.5, 1:3, 1:3.5, 1: 4. 1:4.5 or 1:5, for example 1:1.5.
  • the iodine is added to the reaction system as a solution, for example, a solution comprising water and/or an organic solvent (eg, a solution comprising water, pyridine, and/or tetrahydrofuran).
  • a solution comprising water and/or an organic solvent (eg, a solution comprising water, pyridine, and/or tetrahydrofuran).
  • the iodine is in excess relative to Compound III.
  • the molar ratio of Compound I to iodine is 1: 1.5-5, such as 1:1.5, 1:2, 1:2.5, 1:3, 1:3.5, 1:4, 1:4.5 or 1:5, for example 1:1.5.
  • steps 1-3 further comprise: after the triphosphate reaction of the compound III and the deprotection reaction are completed, sodium sulfite is added to remove unreacted iodine.
  • steps 1-3 further comprise purifying compound IV.
  • Step 2 further includes the following steps:
  • Step 2-1 2-Hydroxyacetic acid and N-ethylenediamine trifluoroacetamide are reacted to form Compound V.
  • step 2-1 comprises: adding O-(N-succinimidyl)-N,N,N',N'-tetramethylurea tetrafluoroborate to 2-hydroxyacetic acid and N,N-diisopropylethylamine was stirred at room temperature, then N-ethylenediamine trifluoroacetamide was added and stirred at room temperature to give Compound V.
  • the molar ratio of 2-hydroxyacetic acid to N-ethylenediamine trifluoroacetamide is from 1:1 to 1:2, such as 1:1.0, 1:1.1, 1:1.2, 1:1.3, 1:1.4, 1:1.5, 1:1.6, 1:1.7, 1:1.8, 1:1.9 or 1:2.0, for example 1:1.2.
  • the molar ratio of 2-hydroxyacetic acid to tetrafluoroboric acid O-(N-succinimidyl)-N,N,N',N'-tetramethylurea is 1:1-1. : 2, for example 1:1.0, 1:1.1, 1:1.2, 1:1.3, 1:1.4, 1:1.5, 1:1.6, 1:1.7, 1:1.8, 1:1.9 or 1:2.0, for example 1 :1.2.
  • the molar ratio of 2-hydroxyacetic acid to N,N-diisopropylethylamine is from 1:1 to 1:2, such as 1:1.0, 1:1.1, 1:1.2, 1: 1.3, 1:1.4, 1:1.5, 1:1.6, 1:1.7, 1:1.8, 1:1.9 or 1:2.0, for example 1:1.5.
  • step 2-1 further comprises purifying compound V.
  • Step 2-2 Compound V is reacted in the presence of N,N-diisopropylethylamine and 2-cyanoethyl N,N-diisopropylchlorophosphoramidite to form compound VI.
  • step 2-2 comprises adding 2-cyanoethyl N,N-diiso to a solution comprising compound V and N,N-diisopropylethylamine at 0 °C.
  • the propyl chlorophosphoramidite was stirred at 0 ° C for a while, slowly warmed to room temperature and stirring was continued to give compound VI.
  • the solution comprising Compound V and N,N-diisopropylethylamine further contains dichloromethane.
  • 2-cyanoethyl N,N-diisopropylchlorophosphoramidite is added as a solution (eg, a solution of dichloromethane).
  • the molar ratio of compound V to N,N-diisopropylethylamine is from 1:1 to 1:2, such as 1:1.0, 1:1.1, 1:1.2, 1:1.3, 1:1.4, 1:1.5, 1:1.6, 1:1.7, 1:1.8, 1:1.9 or 1:2.0, for example 1:1.5.
  • the molar ratio of compound V to 2-cyanoethyl N,N-diisopropylchlorophosphoramidite is from 1:1 to 1:2, such as 1:1.0, 1:1.1, 1 : 1.2, 1:1.3, 1:1.4, 1:1.5, 1:1.6, 1:1.7, 1:1.8, 1:1.9 or 1:2.0, for example 1:1.
  • step 2-2 further comprises purifying compound VI.
  • Step 2-3 Compound VI is subjected to an oxidation reaction to give compound VI'.
  • step 2-3 comprises: mixing compound VI with tetrazole and 2-nitro-5-hydroxy-benzoic acid tert-butyl ester, stirring at room temperature for a period of time, adding iodine, and continuing to stir at room temperature , the compound VI' is produced.
  • the tetrazole is in excess relative to Compound VI.
  • the molar ratio of compound VI to tetrazole is 1:5, such as 1:1, 1:2, 1:3, 1:4, or 1:5, such as 1:2.
  • 2-nitro-5-hydroxy-benzoic acid tert-butyl ester is in excess relative to Compound VI.
  • the molar ratio of compound VI to 2-nitro-5-hydroxy-benzoic acid tert-butyl ester is 1:5, such as 1:1, 1:2, 1:3, 1:4 or 1 :5, for example 1:2.
  • Compound VI is dissolved in acetonitrile prior to the reaction.
  • the tetrazole and 2-nitro-5-hydroxy-benzoic acid tert-butyl ester are combined with Compound VI in the form of a solution (eg, an acetonitrile solution).
  • the iodine is added to the reaction system as a solution, for example, a solution comprising water and/or an organic solvent (eg, a solution comprising water, pyridine, and/or tetrahydrofuran).
  • a solution comprising water and/or an organic solvent (eg, a solution comprising water, pyridine, and/or tetrahydrofuran).
  • the iodine is in excess relative to the compound VI.
  • the molar ratio of compound VI to iodine is 1: 1.5-5, such as 1:1.5, 1:2, 1:2.5, 1:3, 1:3.5, 1:4, 1:4.5 or 1:5, for example 1:1.5.
  • steps 2-3 further comprise: after the oxidation reaction of Compound VI is complete, sodium sulfite is added to remove unreacted iodine.
  • Step 2-4 Deprotection of compound VI'.
  • steps 2-4 include: adding aqueous ammonia to compound VI' to remove trifluoroacetyl and cyanoethyl groups; removing aqueous ammonia, and adding potassium hydroxide to remove tert-butyl groups to provide compound VII.
  • steps 2-4 include: adding excess aqueous ammonia to compound VI', stirring at room temperature; removing aqueous ammonia, adding excess potassium hydroxide, and stirring at room temperature.
  • potassium hydroxide is added as a solution (eg, an aqueous solution).
  • steps 2-4 further comprise: purifying compound VII.
  • Step 3-1 Compound VII is reacted with an N-hydroxysuccinimide ester of Cy3 under weakly basic conditions to give compound VIII.
  • step 3-1 comprises: adding N,N-diisopropylethylamine and compound VII to the N-hydroxysuccinimide ester of Cy3, and stirring at room temperature to provide compound VIII.
  • step 3-1 comprises: adding a DMF solution comprising N,N-diisopropylethylamine and Compound VII to a solution of N-hydroxysuccinimide ester of Cy3 in DMF, room temperature Stirring gives compound VIII.
  • step 3-1 further comprises: purifying compound VIII.
  • Step 3-2 Reaction of Compound IV with Compound VIII to form a modified nucleoside or nucleotide of the invention.
  • step 3-2 comprises: compound VIII, O-(N-succinimidyl)-N,N,N',N'-tetramethylurea and N,N tetrafluoroborate - Diisopropylethylamine is mixed, and then compound IV is added and stirred at room temperature to obtain a compound of the present invention.
  • reaction of Step 3-2 is carried out in DMF.
  • Compound IV is added to the reaction system as a solution (eg, a solution comprising sodium bicarbonate).
  • step 3-2 further comprises: purifying the modified nucleoside or nucleotide of the invention.
  • sequencing is performed while synthesizing a polynucleotide in which a target single-stranded polynucleotide is complementary to each other.
  • the application provides a method of preparing a growing polynucleotide complementary to a target single-stranded polynucleotide in a sequencing reaction, comprising incorporating a compound as defined above into the complementary of said growth A polynucleotide, wherein the incorporation of the compound prevents any subsequent nucleotides from being introduced into the growing complementary polynucleotide.
  • the incorporation of the compound is achieved by a terminal transferase, a terminal polymerase, or a reverse transcriptase.
  • the method comprises: incorporating the compound into a growing complementary polynucleotide using a polymerase.
  • the method comprises: polymerizing a polymerase using a polymerase under conditions that allow the polymerase to undergo nucleotide polymerization, thereby incorporating the compound into the growing complementary polynucleotide 3' end.
  • the application provides a method of determining the sequence of a single-stranded polynucleotide of interest, comprising: monitoring sequential incorporation of complementary nucleotides, wherein at least one complementary nucleotide that is incorporated is as described above A defined compound, and detecting a detectable label carried by the compound.
  • the blocking group and the detectable label in the compound are removed prior to introduction of the next complementary nucleotide.
  • the blocking group and the detectable label are removed simultaneously.
  • the blocking group and the detectable label are removed sequentially.
  • the blocking group is removed before or after the detectable label is removed.
  • the method of determining the sequence of a single-stranded polynucleotide of interest comprises:
  • duplex comprising a growing nucleic acid strand and a nucleic acid molecule to be sequenced
  • the reaction cycle further comprises the step (iv) of removing the detectable label on the nucleic acid intermediate using a scavenging reagent.
  • the ablation reagents used in steps (iii) and (iv) are the same reagents. In certain embodiments, the ablation reagents used in steps (iii) and (iv) are different reagents.
  • the method of determining the sequence of a single-stranded polynucleotide of interest comprises:
  • duplex comprising a growing nucleic acid strand and a nucleic acid molecule to be sequenced, the duplex being linked to a support;
  • a scavenging reagent such that the duplex or the growing nucleic acid strand is contacted with a scavenging reagent in a reaction system containing a solution phase and a solid phase; wherein the excision reagent enables the incorporation of the growing nucleic acid strand
  • the phosphodiester bond (1) and/or the phosphodiester bond (2) in the compound at the 3' end are cleaved and do not affect the phosphodiester bond on the duplex backbone;
  • the method further comprises the steps of:
  • the washing operation is performed after any of the steps including the removing operation.
  • a washing operation is performed between step (4) and step (5).
  • a washing operation is performed after step (6).
  • the duplex is obtained by a method comprising the steps of:
  • a primer is provided to anneal the primer to the nucleic acid molecule to be sequenced, which serves as the initial growing nucleic acid strand, together with the nucleic acid molecule to be sequenced, forms a duplex attached to the support.
  • the nucleic acid molecule to be sequenced may be any nucleic acid molecule of interest.
  • the nucleic acid molecule to be sequenced comprises deoxyribonucleotides, ribonucleotides, modified deoxyribonucleotides, modified ribonucleotides, or any combination thereof.
  • the nucleic acid molecule to be sequenced is not limited by its type.
  • the nucleic acid molecule to be sequenced is DNA or RNA.
  • the nucleic acid molecule to be sequenced can be genomic DNA, mitochondrial DNA, chloroplast DNA, mRNA, cDNA, miRNA, or siRNA.
  • the nucleic acid molecule to be sequenced is linear or circular. In certain preferred embodiments, the nucleic acid molecule to be sequenced is double-stranded or single-stranded.
  • the nucleic acid molecule to be sequenced may be single-stranded DNA (ssDNA), double-stranded DNA (dsDNA), single-stranded RNA (ssRNA), double-stranded RNA (dsRNA), or a hybrid of DNA and RNA.
  • the nucleic acid molecule to be sequenced is a single stranded DNA. In certain preferred embodiments, the nucleic acid molecule to be sequenced is a double stranded DNA.
  • the nucleic acid molecule to be sequenced is not limited by its source.
  • the nucleic acid molecule to be sequenced can be obtained from any source, for example, any cell, tissue or organism (eg, viruses, bacteria, fungi, plants, and animals).
  • the nucleic acid molecule to be sequenced is derived from a mammal (eg, a human, a non-human primate, a rodent or a canine), a plant, a bird, a reptile, a fish, Fungus, bacteria or virus.
  • nucleic acid molecules from cells, tissues or organisms are well known to those skilled in the art. Suitable methods include, but are not limited to, ethanol precipitation, chloroform extraction, and the like. A detailed description of such methods can be found, for example, in J. Sambrook et al., Molecular Cloning: A Laboratory Manual, 2nd Edition, Cold Spring Harbor Laboratory Press, 1989, and FMAusubel et al., Guide to Molecular Biology Experiments , 3rd edition, John Wiley & Sons, Inc., 1995. In addition, various commercial kits can be used to extract nucleic acid molecules from a variety of sources, such as cells, tissues or organisms.
  • the nucleic acid molecule to be sequenced is not limited by its length.
  • the nucleic acid molecule to be sequenced can be at least 10 bp, at least 20 bp, at least 30 bp, at least 40 bp, at least 50 bp, at least 100 bp, at least 200 bp, at least 300 bp, at least 400 bp, at least 500 bp, at least 1000 bp in length. , or at least 2000bp.
  • the nucleic acid molecule to be sequenced may be 10-20 bp, 20-30 bp, 30-40 bp, 40-50 bp, 50-100 bp, 100-200 bp, 200-300 bp, 300-400 bp, 400-500 bp, 500-1000 bp, 1000-2000 bp, or more than 2000 bp.
  • the nucleic acid molecule to be sequenced can have a length of 10-1000 bp to facilitate high throughput sequencing.
  • the nucleic acid molecule can be pretreated prior to attaching the nucleic acid molecule to the support.
  • pretreatments include, but are not limited to, fragmentation of nucleic acid molecules, complementation of ends, addition of linkers, addition of tags, repair of nicks, amplification of nucleic acid molecules, isolation and purification of nucleic acid molecules, and any combination thereof.
  • nucleic acid molecules can be subjected to fragmentation in order to obtain nucleic acid molecules of suitable length.
  • fragmentation of a nucleic acid molecule can be performed by any method known to those of ordinary skill in the art.
  • fragmentation can be carried out by enzymatic or mechanical means.
  • the mechanical method can be ultrasonic or physical shear.
  • the enzymatic method can be carried out by digestion with a nuclease (for example, deoxyribonuclease) or restriction endonuclease.
  • the fragmentation results in an end of the sequence that is not known.
  • the fragmentation results in a known end of the sequence.
  • the enzymatic method uses DNase I to fragment a nucleic acid molecule.
  • DNase I is a universal enzyme that non-specifically cleaves double-stranded DNA (dsDNA) to release 5' phosphorylated dinucleotide, trinucleotide and oligonucleotide products.
  • dsDNA double-stranded DNA
  • DNase I has optimal activity in buffers containing Mn 2+ , Mg 2+ and Ca 2+ but no other salts, which are commonly used to fragment a large DNA genome into small DNA fragments, followed by The resulting small DNA fragments can be used to construct a DNA library.
  • DNase I The cleavage properties of DNase I will result in random digestion of DNA molecules (ie, no sequence bias) and, when used in the presence of buffers containing manganese ions, produce predominantly blunt-end dsDNA fragments (Melgar, E. And DAGoldthwait.1968. Deoxyribonucleic acid nucleases. II. The effects of metal on the mechanism of action of deoxyribonuclease IJ Biol. Chem. 243: 4409).
  • DNase I three factors can be considered: (i) amount of enzyme used (unit); (ii) digestion temperature (°C); and (iii) incubation time (minutes).
  • large DNA fragments or whole genomic DNA can be digested with DNase I for 1-2 minutes between 10 ° C and 37 ° C to produce DNA molecules of suitable length.
  • the nucleic acid molecule of interest (the nucleic acid molecule to be sequenced) is fragmented prior to step (1').
  • the nucleic acid molecules to be sequenced are subjected to fragmentation by enzymatic or mechanical means.
  • the nucleic acid molecule to be sequenced by DNase I is fragmented.
  • the nucleic acid molecules to be sequenced are subjected to fragmentation by sonication.
  • the fragmented nucleic acid molecule is 50-2000 bp in length, such as 50-100 bp, 100-200 bp, 200-300 bp, 300-400 bp, 400-500 bp, 500-1000 bp, 1000-2000 bp. , 50-1500 bp, or 50-1000 bp.
  • Fragmentation of double-stranded nucleic acid molecules can produce nucleic acid fragments having blunt ends or overhangs of one or two nucleotides in length.
  • genomic DNA gDNA
  • DNase I the product may comprise a DNA fragment having a blunt end or overhang.
  • the end of the nucleic acid molecule having an overhang can be made up using a polymerase to form a nucleic acid molecule having a blunt end to facilitate subsequent applications (e.g., to facilitate ligation of the fragmented nucleic acid molecule to a linker).
  • the fragmented nucleic acid molecule is treated with a DNA polymerase to produce a DNA fragment having a blunt end.
  • the DNA polymerase can be any known DNA polymerase, such as T4 DNA polymerase, Pfu DNA polymerase, Klenow DNA polymerase.
  • Pfu DNA polymerase may be advantageous because Pfu DNA polymerase not only complements the overhangs to form blunt ends, but also has 3'-5' exonuclease activity, Removal of single nucleotide and dinucleotide overhangs to further increase the number of DNA fragments with blunt ends (Costa, GL and MP Weiner. 1994a. Protocols for cloning and analysis of blunt-ended PCR-generated DNA fragments. PCR Methods Appl 3(5): S95; Costa, GLt A. Grafsky and MP Wemer. 1994b. Cloning and analysis of PCR-generated DNA fragments. PCR Methods Appl 3(6): 338; Costa, GL and MPWeiner. 1994c. Polishing with T4or Pfu polymerase increases the efficiency of cloning of PCR products. Nucleic Acids Res. 22(12): 2423).
  • a linker can be introduced at the 5' and/or 3' end of the nucleic acid molecule to be sequenced.
  • the linker is an oligonucleotide sequence and it can be any sequence, of any length.
  • Linkers of suitable length and sequence can be selected by methods well known in the art.
  • a linker ligated to the end of a nucleic acid molecule to be sequenced is typically 5 to 100 nucleotides in length (eg, 5-10 bp, 10-20 bp, 20-30 bp, 30-40 bp, 40-50 bp, 50-100 bp).
  • the linker can have a primer binding region.
  • the linker has one or more primer binding regions. In certain preferred embodiments, the linker has one or more regions that are capable of hybridizing to a primer for amplification. In certain preferred embodiments, the linker has one or more regions that are capable of hybridizing to primers used in the sequencing reaction. In certain preferred embodiments, a linker is introduced at the 5' end of the nucleic acid molecule to be sequenced. In certain preferred embodiments, a linker is introduced at the 3' end of the nucleic acid molecule to be sequenced.
  • a linker is introduced at the 5' and 3' ends of the nucleic acid molecule to be sequenced.
  • the linker comprises A universal linker sequence sufficient to hybridize to a universal primer.
  • the linker comprises a universal linker sequence that is capable of hybridizing to a universal amplification primer and/or a universal sequencing primer.
  • the tag sequence can be introduced into the nucleic acid molecule to be sequenced, or the tag sequence can be introduced into the linker described above.
  • a tag sequence refers to a segment of an oligonucleotide having a particular base sequence.
  • the tag sequence can have any length, such as 2-50 bp, such as 2, 3, 4, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50 bp, depending on actual needs.
  • each nucleic acid molecule to be sequenced is subjected to a tag sequence containing a particular sequence to facilitate discrimination of the source of each nucleic acid molecule to be sequenced.
  • the tag sequence can be introduced directly at the 5' and/or 3' end of the nucleic acid molecule to be sequenced.
  • a tag sequence can be introduced into the linker and then ligated to the 5' and/or 3' end of the nucleic acid molecule to be sequenced.
  • the tag sequence can be located anywhere in the linker sequence, such as the 5' and/or 3' end of the linker sequence.
  • the linker comprises a primer binding region and a tag sequence.
  • the primer binding region comprises a universal linker sequence that is recognized by universal primers, and preferably, the tag sequence can be located at the 3' end of the primer binding region.
  • different tag sequences are used to label/distinguish nucleic acid molecules from different sources.
  • the same tag sequence is introduced into a nucleic acid molecule of the same source, and for each nucleic acid source, a unique tag sequence is used.
  • nucleic acid molecules of different origins can be combined to form a library, and the source of each nucleic acid molecule in the library can be identified/differentiated by the unique tag sequence carried on each nucleic acid molecule.
  • the nucleic acid molecule to be sequenced can be ligated to a linker or tag sequence by methods well known in the art (eg, PCR or ligation reactions). For example, if a part of the sequence of the nucleic acid molecule to be sequenced is known, the nucleic acid molecule to be sequenced by PCR can be carried out using an appropriate PCR primer containing a linker sequence and a sequence capable of specifically recognizing the nucleic acid molecule to be sequenced. Amplification. The amplified product obtained is the nucleic acid molecule to be tested which introduces a linker at the 5' and/or 3' end.
  • a nucleic acid molecule can be linked to a linker using a non-specific ligase (eg, T4 DNA ligase).
  • a nucleic acid molecule and a linker can be treated with a restriction enzyme such that they have the same cohesive ends, and then the ligase can be used to link nucleic acid molecules and linkers having the same cohesive ends together. Thereby obtaining a nucleic acid molecule linked to the linker.
  • the resulting product may have a nick at the junction.
  • a polymerase can be used to repair this incision.
  • a DNA polymerase that loses 3'-5' exonuclease activity but exhibits 5'-3' exonuclease activity can have an incision and repair The ability to complex incisions (Hamilton, S. C., J. W. Farchaus and M. C. Davis. 2001. DNA polymerases as engines for biotechnology. BioTechniques 31: 370).
  • DNA polymerases which can be used for this purpose include, for example, polI of Thermoanaerobacter thermosulfuricus, DNA polI of E. coli, and phage phi29.
  • polI of Bacillus stearothermophilus is used to repair the nick of dsDNA and form unnotched dsDNA.
  • the nucleic acid molecules to be sequenced can also be amplified to increase the amount or copy number of the nucleic acid molecule.
  • Methods for amplifying nucleic acid molecules are well known to those skilled in the art, a typical example of which is PCR.
  • nucleic acid molecules can be amplified using the following methods: (i) polymerase chain reaction (PCR) requiring temperature cycling (see, eg, Saiki et al, 1995. Science 230: 1350-1354), ligase chain Reaction (see, for example, Barany, 1991. Proc. Natl. Acad. Sci. USA 88: 189-193; Barringer et al, 1990.
  • PCR polymerase chain reaction
  • the nucleic acid molecule to be sequenced is amplified by PCR, and the primers used for PCR amplification comprise a linker sequence and/or a tag sequence.
  • the PCR product thus produced will carry a linker sequence and/or a tag sequence, which can be conveniently used for subsequent applications (e.g., high throughput sequencing).
  • the nucleic acid molecules to be sequenced are also isolated and purified before or after various pretreatment steps. Such separation and purification steps may be advantageous.
  • the isolation and purification steps can be used to obtain nucleic acid molecules of a suitable length (eg, 50-1000 bp) to be sequenced for subsequent applications (eg, high throughput sequencing).
  • agarose gel electrophoresis can be utilized to separate and purify the nucleic acid molecules to be sequenced.
  • the nucleic acid molecules to be sequenced can be isolated and purified by size exclusion chromatography or sucrose settling.
  • the pre-treatment steps described above are merely exemplary and not limiting.
  • Those skilled in the art can perform various desired pretreatments on the nucleic acid molecules to be sequenced according to actual needs, and each pretreatment step is not limited by a specific order.
  • the nucleic acid molecule can be first fragmented and a linker added prior to amplification.
  • the nucleic acid molecule can be amplified first, Then fragment and add the linker.
  • the nucleic acid molecule is fragmented and a linker is added without an amplification step.
  • the nucleic acid molecule of interest e.g., genomic DNA
  • pretreatment e.g., genomic DNA
  • nucleic acid molecule of interest eg, a large nucleic acid fragment, eg, genomic DNA
  • linker sequence comprising, for example, a primer binding region capable of hybridizing to a universal amplification primer, a primer binding region capable of hybridizing to a universal sequencing primer, and/or a tag sequence, and Optionally performing isolation, purification, and denaturation to produce a nucleic acid molecule to be sequenced;
  • support is sometimes also referred to as “solid support” or “solid support.”
  • solid support or “solid support.”
  • the “support” referred to herein is not limited to a solid, it may also be a semi-solid (eg, a gel).
  • the support for ligation of the nucleic acid molecule to be sequenced may be made of various suitable materials.
  • materials include, for example, inorganics, natural polymers, synthetic polymers, and any combination thereof.
  • Specific examples include, but are not limited to, cellulose, cellulose derivatives (such as nitrocellulose), acrylic resins, glass, silica gel, polystyrene, gelatin, polyvinylpyrrolidone, copolymers of vinyl and acrylamide, and Crosslinked polyphenylene bromide such as vinyl benzene (see, for example, Merrifield Biochemistry 1964, 3, 1385-1390), polyacrylamide, latex, dextran, rubber, silicon, plastic, natural sponge, metal plastic, cross-linking dextran (e.g., Sephadex TM), agarose gel (Sepharose TM), and other supports known to the skilled person.
  • Sephadex TM Sephadex TM
  • agarose gel Sepharose TM
  • the support for ligation of the nucleic acid molecule to be sequenced may be a solid support comprising an inert substrate or matrix (eg, slides, polymer beads, etc.), said inert substrate or matrix Functionalization has been functionalized, for example, by the use of intermediate materials containing reactive groups that allow covalent attachment of biomolecules such as polynucleotides.
  • inert substrate or matrix e.g., slides, polymer beads, etc.
  • Functionalization has been functionalized, for example, by the use of intermediate materials containing reactive groups that allow covalent attachment of biomolecules such as polynucleotides.
  • supports include, but are not limited to, polyacrylamide hydrogels supported on an inert substrate such as glass, in particular polyacrylamide hydrogels as described in WO 2005/065814 and US 2008/0280773, wherein The content of the patent application is hereby incorporated by reference in its entirety.
  • a biomolecule eg, a polynucleotide
  • an intermediate material eg, a hydrogel
  • the intermediate material itself can be non-covalently attached to a substrate or matrix (eg, a glass substrate)
  • the support is a slide or wafer having a surface modified with a layer of avidin, amino, acrylamide silane or aldehyde based chemical groups.
  • the support or solid support is not limited by its size, shape and configuration.
  • the support or solid support is a planar structure, such as a slide, chip, microchip, and/or array.
  • the surface of such a support may be in the form of a planar layer.
  • the support or surface thereof is non-planar, such as the inner or outer surface of a tube or container.
  • the support or solid support comprises microspheres or beads.
  • microsphere or “bead” or “particle” or grammatical equivalent refers to a small discrete particle.
  • Suitable bead ingredients include, but are not limited to, plastics, ceramics, glass, polystyrene, methyl styrene, acrylic polymers, paramagnetic materials, cerium oxide sol, carbon graphite, titanium dioxide, latex, cross-linked dextran such as Sepharose , cellulose, nylon, crosslinked micelles and teflon, as well as any other materials outlined herein for the preparation of solid supports.
  • the beads may be spherical or non-spherical. In some embodiments, spherical beads can be used. In some embodiments, irregular particles can be used. In addition, the beads can also be porous.
  • the support for ligation of the nucleic acid molecule to be sequenced is an array of beads or wells (also referred to as a chip).
  • the array can be prepared using any of the materials outlined herein for preparing a solid support, and preferably, the surface of the beads or pores on the array is functionalized to facilitate ligation of the nucleic acid molecules.
  • the number of beads or holes on the array is not limited.
  • each array may comprise 10-10 2 , 10 2 -10 3 , 10 3 -10 4 , 10 4 -10 5 , 10 5 -10 6 , 10 6 -10 7 , 10 7 -10 8 , 10 8 -109 or more beads or holes.
  • each bead or well can be joined to one or more nucleic acid molecules.
  • each array can be connected 10-10 2 , 10 2 -10 3 , 10 3 -10 4 , 10 4 -10 5 , 10 5 -10 6 , 10 6 -10 7 , 10 7 -10 8 , 10 8 - 10 9 or more nucleic acid molecules.
  • arrays can be used particularly advantageously for high throughput sequencing of nucleic acid molecules.
  • Such techniques include, but are not limited to, photolithography, stamping techniques, plastic film technology, and microetching techniques. As will be appreciated by those skilled in the art, the techniques used will depend on the composition, structure and shape of the support.
  • the nucleic acid molecule to be sequenced can be linked (e.g., covalently or non-covalently linked) to the support by any method known to those of ordinary skill in the art.
  • covalent connection or by The irreversible passive adsorption, or the intermolecular affinity (eg, the affinity between biotin and avidin), connects the nucleic acid molecule to be sequenced to the support.
  • the linkage between the nucleic acid molecule to be sequenced and the support is sufficiently strong that the nucleic acid molecule does not detach from the support due to the conditions used in the various reactions (e.g., polymerization) and the washing of the water or buffer solution.
  • the 5' end of the nucleic acid molecule to be sequenced carries a device capable of covalently attaching the nucleic acid molecule to a support, such as a chemically modified functional group.
  • a device capable of covalently attaching the nucleic acid molecule to a support such as a chemically modified functional group.
  • functional groups include, but are not limited to, a phosphate group, a carboxylic acid molecule, an aldehyde molecule, a thiol, a hydroxyl group, a dimethoxytrityl group (DMT), or an amino group.
  • the 5' end of the nucleic acid molecule to be sequenced may be modified with a chemical functional group (eg, a phosphoric acid, a thiol or an amino group), and the support (eg, a porous glass bead) may be an amino-alkane.
  • a chemical functional group eg, a phosphoric acid, a thiol or an amino group
  • the support eg, a porous glass bead
  • Oxysilane for example, aminopropyltrimethoxysilane, aminopropyltriethoxysilane, 4-aminobutyltriethoxysilane, etc.
  • the molecule is covalently attached to the support.
  • the 5' end of the nucleic acid molecule to be sequenced can be modified with a carboxylic acid or an aldehyde group, and the support (eg, latex beads) is derivatized with hydrazine so that the chemistry between the reactive groups can be The reaction covalently attaches the nucleic acid molecule to the support (Kremsky et al., 1987).
  • cross-linking agent can be used to link the nucleic acid molecule of interest to the support.
  • crosslinking agents include, for example, succinic anhydride, phenyl diisothiocyanate (Guo et al., 1994), maleic anhydride (Yang et al., 1998), 1-ethyl-3-(3-di).
  • Methylaminopropyl)-carbodiimide hydrochloride EDC
  • m-maleimidobenzoic acid-N-hydroxysuccinimide ester MBS
  • N-succinimidyl group [4] -iodoacetyl]aminobenzoic acid SIAB
  • 4-(N-maleimidomethyl)cyclohexane-1-carboxylic acid succinimide SCC
  • GMBS N- ⁇ -maleyl Iminobutyryloxy-succinimide ester
  • SMPB 4-(p-maleimidophenyl)butyric acid succinimide
  • corresponding thio compounds water soluble
  • the support can also be derivatized with a bifunctional crosslinker such as a homobifunctional crosslinker and a heterobifunctional crosslinker to provide a modified functionalized surface.
  • a bifunctional crosslinker such as a homobifunctional crosslinker and a heterobifunctional crosslinker to provide a modified functionalized surface.
  • a nucleic acid molecule having a 5'-phosphate, thiol or amino group is capable of interacting with a functionalized surface to form a covalent linkage between the nucleic acid and the support.
  • a large number of bifunctional crosslinkers and methods of use thereof are well known in the art (see, for example, Pierce Catalog and Handbook, pages 155-200).
  • the primer may be of any length and may comprise any sequence or any base as long as it can specifically anneal to a region of the target nucleic acid molecule.
  • Primers are not limited by their length, structure and composition.
  • the primers may be 5-50 bp in length, such as 5-10, 10-15, 15-20, 20-25, 25-30, 30-35, 35-40, 40-45, 45-50 bp.
  • the primers are capable of forming a secondary structure (eg, a hairpin structure).
  • the primer does not form any secondary structure (eg, a hairpin structure).
  • the primers may comprise naturally occurring or non-naturally occurring nucleotides.
  • the primer comprises or consists of a naturally occurring nucleotide.
  • the primer comprises a modified nucleotide, such as a locked nucleic acid (LNA).
  • LNA locked nucleic acid
  • the primer is capable of hybridizing to a nucleic acid of interest under stringent conditions, such as moderately stringent conditions or highly stringent conditions.
  • the primer has a sequence that is fully complementary to a target sequence in a nucleic acid molecule of interest.
  • the primer is partially complementary to a target sequence in a nucleic acid molecule of interest (eg, a mismatch is present).
  • the primer comprises a universal primer sequence.
  • the nucleic acid molecule to be sequenced comprises a linker, and the linker comprises a sequence capable of hybridizing to a universal primer, and the primer used is a universal primer.
  • a suitable polymerase can be used for nucleotide polymerization.
  • the polymerase is capable of synthesizing a new DNA strand (eg, a DNA polymerase) using DNA as a template.
  • the polymerase is capable of synthesizing a new DNA strand (eg, a reverse transcriptase) using RNA as a template.
  • the polymerase is capable of synthesizing a new RNA strand (eg, RNA polymerase) using DNA or RNA as a template.
  • the polymerase is selected from the group consisting of a DNA polymerase, an RNA polymerase, and a reverse transcriptase.
  • a suitable polymerase can be selected for nucleotide polymerization according to actual needs.
  • the polymerization reaction is a polymerase chain reaction (PCR).
  • the polymerization reaction is a reverse transcription reaction.
  • nucleotide polymerization can be carried out using KOD polymerase or a mutant thereof.
  • KOD polymerase or a mutant thereof e.g., KOD POL151, KOD POL157, KOD POL171, KOD POL174, KOD POL376, KOD POL391
  • KOD POL391 and KOD POL171 have acceptable polymerization efficiencies for the modified nucleotides of the invention.
  • KOD POL391 or KOD POL171 has a polymerization efficiency for the modified nucleotides of the invention of greater than 70%, such as from 70% to 80%, from 80% to 90%, or from 90% to 100%.
  • steps (2) - (6) or steps (2) - (4) may be repeated.
  • one or more rounds of nucleotide polymerization can be carried out.
  • the nucleotide polymerization can be carried out in one or more steps.
  • the same or different polymerases can be used for each round of nucleotide polymerization.
  • a first DNA polymerase can be used in the first round of nucleotide polymerization
  • a second DNA polymerase can be used in the second round of nucleotide polymerization.
  • the same polymerase eg, the same DNA polymerase
  • the same polymerase is used in all nucleotide polymerizations.
  • the polymerization of nucleotides is carried out under suitable conditions.
  • suitable polymerization conditions include the composition of the solution phase and the concentration of each component, the pH of the solution phase, the polymerization temperature, and the like. Polymerization under suitable conditions is advantageous in obtaining acceptable, even high, polymerization efficiencies.
  • the solution phase in which the polymerization occurs comprises monovalent salt ions (eg, sodium ions, chloride ions) and/or divalent salt ions (eg, magnesium ions, sulfate ions).
  • concentration of the monovalent salt or divalent salt ion in the solution phase is 1-200 mM, such as 1 mM, 3 mM, 10 mM, 20 mM, 50 mM, 100 mM, 150 mM, or 200 mM.
  • the solution phase in which the polymerization occurs comprises a buffer solution, such as a buffer solution comprising Tris.
  • concentration of Tris in the solution phase is from 10 mM to 200 mM, such as 10 mM, 20 mM, 50 mM, 100 mM, 150 mM, or 200 mM.
  • the solution phase in which the polymerization occurs comprises an organic solvent such as DMSO or glycerol (glycerol).
  • the organic solvent has a mass content in the solution phase of from 0.01% to 10%, such as 0.01%, 0.02%, 0.05%, 1%, 2%, 5%, or 10%.
  • the pH of the solution phase in which the polymerization occurs is from 7.0 to 9.0, such as 7.0, 7.1, 7.2, 7.3, 7.4, 7.5, 7.6, 7.7, 7.8, 7.9, 8.0, 8.1, 8.2, 8.3, 8.4. , 8.5, 8.6, 8.7, 8.8, 8.9 or 9.0.
  • the solution phase in which the polymerization occurs comprises: a monovalent salt ion (eg, sodium ion, chloride ion), a divalent salt ion (eg, magnesium ion, sulfate ion), a buffer solution (eg, a buffer containing Tris) Solution) and organic solvents (such as DMSO or glycerol).
  • a monovalent salt ion eg, sodium ion, chloride ion
  • a divalent salt ion eg, magnesium ion, sulfate ion
  • a buffer solution eg, a buffer containing Tris
  • organic solvents such as DMSO or glycerol
  • the polymerization is at 50-65 ° C (eg, 50 ° C, 51 ° C, 52 ° C, 53 ° C, 54 ° C, 55 ° C, 56 ° C, 57 ° C, 58 ° C, 59 ° C, 60 ° C, 61 It is carried out at ° C, 62 ° C, 63 ° C, 64 ° C or 65 ° C).
  • the time during which the polymerization is carried out can be determined according to actual needs, for example, 1 min to 5 min, 5 min to 10 min, 10 min to 30 min or 30 min to 1 h.
  • the four compounds used in the step (2) are derivatives of nucleotides A, (T/U), C and G, respectively.
  • the four compounds are derivatives of ribose or deoxyribonucleotides A, T, C, and G, respectively.
  • the four compounds are derivatives of ribose or deoxyribonucleotides A, U, C, and G, respectively. It is particularly advantageous that the four compounds do not undergo a chemical reaction with each other during the nucleotide polymerization.
  • the four compounds described have a base complementary pairing ability.
  • the compound when the compound is a derivative of nucleotide A, it will be capable of pairing with the base T or U.
  • the compound When the compound is a derivative of nucleotide T or U, it will be able to pair with base A.
  • the compound When the compound is a derivative of nucleotide C, it will be able to pair with base G.
  • the polymerase eg, DNA polymerase
  • the polymerase will incorporate a compound capable of complementary pairing with a base at a corresponding position in the template nucleic acid into the growing nucleic acid strand according to the principle of base complementary pairing.
  • the base at the corresponding position in the template nucleic acid can be determined by the principle of base complementary pairing. type. For example, if a compound incorporated at the 3' end of the growing nucleic acid strand is identified as a derivative of nucleotide A, then the base at the corresponding position in the template nucleic acid can be determined to be T or U.
  • a compound incorporated at the 3' end of the growing nucleic acid strand is identified as a derivative of nucleotide T or U, then it is determined that the base at the corresponding position in the template nucleic acid is A. If a compound incorporated at the 3' end of the growing nucleic acid strand is identified as a derivative of nucleotide C, then it is determined that the base at the corresponding position in the template nucleic acid is G. If a compound incorporated at the 3' end of the growing nucleic acid strand is identified as a derivative of nucleotide G, then it is determined that the base at the corresponding position in the template nucleic acid is C.
  • the hydroxyl group (-OH) at the 3' position of the ribose or deoxyribose of the four compounds is protected.
  • the hydroxyl groups (-OH) at the 3' position of the ribose or deoxyribose of the four compounds are protected by a protecting group, whereby they are capable of terminating the polymerization of a polymerase such as a DNA polymerase.
  • the protecting group at the 3' position of the ribose or deoxyribose of the four compounds can be removed.
  • the protecting group is removed and converted to a free hydroxyl group (-OH).
  • the polymerase and the four compounds can be used to carry out the next round of polymerization of the grown nucleic acid strand and introduce one more base.
  • the four compounds used in step (2) are reversibly blocked: when they are incorporated into the 3' end of the growing nucleic acid strand (eg, in step (3)), they The polymerase will be terminated to continue the polymerization, terminating the further extension of the growing nucleic acid strand; and, after the blocking group they contain is removed, the polymerase will continue to polymerize the growing nucleic acid strand and continue to extend the nucleic acid chain.
  • a detectable signal in a duplex or a growing nucleic acid strand is detected after each round of polymerization.
  • the type of compound incorporated into the growing nucleic acid strand can be determined to determine the type of base at the corresponding position in the nucleic acid molecule to be sequenced.
  • the detectable label is a fluorophore.
  • the sequencing method of the present invention further comprises, after step (4), based on the principle of base complementary pairing, according to the type of compound incorporated in the 3' end of the growing nucleic acid strand in step (3) Determining the type of base at the corresponding position in the nucleic acid molecule to be sequenced. For example, if a compound incorporated at the 3' end of the growing nucleic acid strand is identified as a derivative of nucleotide A, then it is determined that the base at the corresponding position in the nucleic acid molecule to be sequenced is a derivative capable of binding to nucleotide A Paired bases (eg T or U).
  • Paired bases eg T or U
  • a compound incorporated at the 3' end of the growing nucleic acid strand is identified as a derivative of nucleotide A, then it is determined that the base at the corresponding position in the nucleic acid molecule to be sequenced is T or U. If the compound incorporated at the 3' end of the growing nucleic acid strand is determined to be a derivative of nucleotide T or U, then it is determined that the base at the corresponding position in the nucleic acid molecule to be sequenced is A. If the compound incorporated at the 3' end of the growing nucleic acid strand is determined to be a derivative of nucleotide C, then it is determined that the base at the corresponding position in the nucleic acid molecule to be sequenced is G. If the compound incorporated at the 3' end of the growing nucleic acid strand is determined to be a derivative of nucleotide G, then it is determined that the base at the corresponding position in the nucleic acid molecule to be sequenced is C.
  • each round of polymerization may involve one signal detection, and, in addition to the last round of polymerization, each round of polymerization may involve excision of the duplex or growing nucleic acid strand. deal with. After the last round of polymerization, the duplex or the growing nucleic acid strand may be excised or not excised.
  • the treatment in step (5) can be used to remove blocking groups in the compound incorporated into the 3' end of the growing nucleic acid strand (so that a new round of polymerization can begin) and remove the double A detectable label that may be carried on the chain or growing nucleic acid strand (so as to avoid interference with subsequent detection).
  • the excision reagent for excising the blocking group and the excision reagent for excising the detectable label are the same reagent, or, although different reagents, can be excised under the same conditions, It does not cause mutual interference between the two resection reactions. Therefore, in the step (5), the excision of the blocking group and the excision of the detectable label can be simultaneously performed.
  • the excision reagent for excising the blocking group and the excision reagent for excising the detectable label are different reagents, and the excision needs to be performed under different conditions. In order to avoid mutual interference between the excision reactions, excision of the blocking group and excision of the detectable label can be performed stepwise.
  • step (5) comprises:
  • Step (5-1) adding a first excision reagent, contacting the duplex or the grown nucleic acid strand with the first excision reagent in a reaction system containing a solution phase and a solid phase without affecting the duplex a phosphodiester bond (1) in a compound incorporated at the 3' end of the growing nucleic acid strand under conditions of a phosphodiester bond on the backbone;
  • the phosphodiester bond (2) in the compound incorporated at the 3' end of the growing nucleic acid strand is cleaved under the condition of a phosphodiester bond on the backbone.
  • step (5-1) there is no fixed sequence between step (5-1) and step (5-2), and step (5-1) may be performed first, or step (5-2) may be performed first.
  • step (5-1) may be performed first, or step (5-2) may be performed first.
  • step (5-2) may be performed first.
  • the use of "first” and “second” merely distinguishes between excising agents and does not represent the order of use of the two excising agents.
  • the time during which the excision reaction is performed can be determined according to actual needs, for example, 1 min to 5 min, 5 min to 10 min, 10 min to 30 min, or 30 min to 1 h.
  • a blocking group and/or a detectable label in the compound of the formula (I) is removed using a scavenging agent.
  • the excision reagent of the present invention is capable of cleaving the phosphodiester bond (1) and/or the phosphodiester bond (2) in the compound of the formula (I'), and Does not break the phosphodiester bond in the nucleic acid strand backbone to maintain the integrity of the nucleic acid strand backbone.
  • excision reagents that can be used to remove blocking groups include, but are not limited to, endonuclease IV.
  • the endonuclease IV can selectively cleave the phosphodiester bond (1) in the compound represented by the formula (I') without affecting the phosphodiester bond in the nucleic acid strand skeleton.
  • excision reagents that can be used to remove detectable labels include, but are not limited to, alkaline phosphatase.
  • the alkaline phosphatase can selectively cleave the phosphodiester bond (2) in the compound represented by the formula (I') without affecting the phosphodiester bond in the nucleic acid strand skeleton.
  • the conditions of the excision reaction may depend on the excision reagent.
  • the excision reagent used is a commercially available enzyme, and thus, the conditions of the excision reaction can be determined according to the use conditions recommended by the supplier (for example, a recommended buffer solution, temperature, etc.). .
  • the washing step can be increased as needed.
  • the washing step can be increased at any desired stage, and optionally, the washing step can be performed one or more times.
  • one or more washings may be performed to sufficiently remove the residual solution phase.
  • Such a washing step may be advantageous in that it can be used to substantially remove free (ie, unincorporated growth nucleic acid strands) compounds that carry detectable labels, minimizing non-specific signals.
  • step (6) after removing the solution phase of the reaction system, one or more washings may be performed to sufficiently remove the residual solution phase.
  • a washing step may be advantageous, which can be used to sufficiently remove the ablation reagent applied in step (5), thereby minimizing the adverse effects on subsequent reactions.
  • the washing step can be carried out using a variety of suitable washing solutions.
  • suitable washing solutions include, but are not limited to, phosphate buffer, citrate buffer, Tris-HCl buffer, acetate buffer, carbonate buffer, and the like. It is within the ability of those skilled in the art to select a suitable wash solution (including suitable ingredients, concentrations, ionic strength, pH, etc.) depending on the actual needs.
  • the invention provides a kit comprising first, second, third and fourth compounds, each of said first, second, third and fourth compounds being of the formula A compound of I) which is a derivative of nucleotides A, (T/U), C and G, respectively, and which has a base complementary pairing ability.
  • the Labels in the four compound structures are each different, such as a different fluorophore.
  • kits of the invention further comprise: reagents and/or devices for extracting nucleic acid molecules from a sample; reagents for pretreating nucleic acid molecules; and supports for ligation of nucleic acid molecules to be sequenced a reagent for linking (eg, covalently or non-covalently linking) a nucleic acid molecule to be sequenced to a support; a primer for initial nucleotide polymerization; a polymerase for performing nucleotide polymerization One or more buffer solutions; one or more wash solutions; or any combination thereof.
  • kits of the invention further comprise reagents and/or devices for extracting nucleic acid molecules from a sample.
  • Methods for extracting nucleic acid molecules from a sample are well known in the art. Therefore, various reagents and/or devices for extracting nucleic acid molecules, such as reagents for disrupting cells, reagents for precipitating DNA, reagents for washing DNA, may be disposed in the kit of the present invention as needed.
  • a reagent for dissolving DNA for example, when the nucleic acid molecule of interest is RNA
  • An agent for removing RNA for example, when the nucleic acid molecule of interest is DNA
  • the kit of the invention further comprises an agent for pretreating the nucleic acid molecule.
  • the reagent for pretreating the nucleic acid molecule is not limited, and may be selected according to actual needs.
  • the reagent for pretreating a nucleic acid molecule includes, for example, a reagent for fragmentation of a nucleic acid molecule (for example, DNase I), a reagent for complementing the end of a nucleic acid molecule (for example, a DNA polymerase such as T4 DNA polymerase, Pfu DNA) a polymerase, Klenow DNA polymerase, a linker molecule, a tag molecule, an agent for linking a linker molecule to a nucleic acid molecule of interest (eg, a ligase, such as T4 DNA ligase), an agent for repairing a nucleic acid nick (eg, loss 3'-5' exonuclease activity but a DNA polyme
  • the kit of the invention further comprises a support for ligation of the nucleic acid molecule to be sequenced.
  • the support may have any of the technical features detailed above for the support and any combination thereof.
  • the support may be made of various suitable materials.
  • materials include, for example, inorganics, natural polymers, synthetic polymers, and any combination thereof.
  • Specific examples include, but are not limited to, cellulose, cellulose derivatives (such as nitrocellulose), acrylic resins, glass, silica gel, polystyrene, gelatin, polyvinylpyrrolidone, copolymers of vinyl and acrylamide, and Crosslinked polyphenylene bromide such as vinyl benzene (see, for example, Merrifield Biochemistry 1964, 3, 1385-1390), polyacrylamide, latex, dextran, rubber, silicon, plastic, natural sponge, metal plastic, cross-linking dextran (e.g., Sephadex TM), agarose gel (Sepharose TM), and other supports known to the skilled person.
  • cellulose cellulose derivatives (such as nitrocellulose)
  • acrylic resins glass, silica gel, polystyrene, gelatin, polyvinylpyrrolidone, copolymers of
  • the support for ligation of the nucleic acid molecule to be sequenced may be a solid support comprising an inert substrate or matrix (eg, slides, polymer beads, etc.), said inert substrate or matrix Functionalization has been functionalized, for example, by the use of intermediate materials containing reactive groups that allow covalent attachment of biomolecules such as polynucleotides.
  • inert substrate or matrix e.g., slides, polymer beads, etc.
  • Functionalization has been functionalized, for example, by the use of intermediate materials containing reactive groups that allow covalent attachment of biomolecules such as polynucleotides.
  • supports include, but are not limited to, polyacrylamide hydrogels supported on an inert substrate such as glass, in particular polyacrylamide hydrogels as described in WO 2005/065814 and US 2008/0280773, wherein The content of the patent application is hereby incorporated by reference in its entirety.
  • the biomolecule eg, a polynucleotide
  • an intermediate material eg, a hydrogel
  • the intermediate material itself can be non-covalently attached to the substrate or matrix (e.g, a glass substrate).
  • the support is a slide or wafer having a surface modified with a layer of avidin, amino, acrylamide silane or aldehyde based chemical groups.
  • the support or solid support is not limited by its size, shape and configuration.
  • the support or solid support is a planar structure, such as a slide, chip, microchip, and/or array.
  • the surface of such a support may be in the form of a planar layer.
  • the support or surface thereof is non-planar, such as the inner or outer surface of a tube or container.
  • the support or solid support comprises microspheres or beads.
  • the support for ligation of the nucleic acid molecule to be sequenced is an array of beads or wells.
  • kits of the invention further comprise reagents for attaching (eg, covalently or non-covalently linking) a nucleic acid molecule to be sequenced to a support.
  • agents include, for example, agents that activate or modify a nucleic acid molecule (eg, at its 5' end), such as phosphoric acid, a thiol, an amine, a carboxylic acid, or an aldehyde; an agent that activates or modifies the surface of the support, such as an amino group - Alkoxysilane (for example, aminopropyltrimethoxysilane, aminopropyltriethoxysilane, 4-aminobutyltriethoxysilane, etc.); crosslinking agent such as succinic anhydride, phenyl diisosulfide Cyanate (Guo et al., 1994), maleic anhydride (Yang et al., 1998), 1-ethyl-3-(3-di
  • kits of the invention further comprise primers for initiating nucleotide polymerization.
  • the primer is not subject to any limitation as long as it can specifically anneal to a region of the target nucleic acid molecule.
  • the primers may be 5-50 bp in length, such as 5-10, 10-15, 15-20, 20-25, 25-30, 30-35, 35-40, 40- 45, 45-50bp.
  • the primers may comprise naturally occurring or non-naturally occurring nucleotides.
  • the primer comprises or consists of a naturally occurring nucleotide.
  • the primer comprises a modified nucleotide, such as a locked nucleic acid (LNA).
  • the primer comprises a universal primer sequence.
  • the kit of the invention further comprises a polymerase for performing a nucleotide polymerization reaction.
  • polymerization can be carried out using various suitable polymerases.
  • the polymerase is capable of synthesizing a new DNA strand (eg, a DNA polymerase) using DNA as a template.
  • the polymerase is capable of synthesizing a new DNA strand (eg, a reverse transcriptase) using RNA as a template.
  • the polymerase is capable of synthesizing a new RNA strand (eg, RNA polymerase) using DNA or RNA as a template.
  • the polymerase is selected from the group consisting of a DNA polymerase, an RNA polymerase, and a reverse transcriptase.
  • the kit of the invention comprises KOD polymerase or a mutant thereof.
  • the mutant is selected from the group consisting of KOD POL151, KOD POL157, KOD POL171, KOD POL174, KOD POL376, KOD POL391.
  • the kit of the invention comprises KOD POL391, KOD POL171, or a combination thereof.
  • the kit of the invention further comprises a excision reagent capable of cleaving the phosphodiester bond (1) and/or the phosphodiester bond (2) in formula (I') And, does not affect the phosphodiester bond on the duplex backbone.
  • the excision agent is selected from the group consisting of endonuclease IV and alkaline phosphatase.
  • the kit of the invention further comprises one or more buffer solutions.
  • buffers include, but are not limited to, buffer solutions for DNase I, buffer solutions for DNA polymerases, buffer solutions for ligases, buffer solutions for eluting nucleic acid molecules, and lysing nucleic acid molecules.
  • the kit of the present invention may comprise any one or more of the above buffer solutions.
  • the buffer solution for DNA polymerase comprises a monovalent salt ion (eg, sodium ion, chloride ion) and/or a divalent salt ion (eg, magnesium ion, sulfate ion).
  • a monovalent salt ion eg, sodium ion, chloride ion
  • a divalent salt ion eg, magnesium ion, sulfate ion
  • the monovalent salt The concentration of the ion or divalent salt ion in the buffer solution is 1-200 mM, such as 1 mM, 3 mM, 10 mM, 20 mM, 50 mM, 100 mM, 150 mM or 200 mM.
  • the buffer solution for DNA polymerase comprises Tris.
  • the concentration of Tris in the buffer solution is from 10 mM to 200 mM, such as 10 mM, 20 mM, 50 mM, 100 mM, 150 mM, or 200 mM.
  • the buffer solution for DNA polymerase comprises an organic solvent such as DMSO or glycerol (glycerol).
  • the organic solvent is present in the buffer solution in an amount of from 0.01% to 10%, such as 0.01%, 0.02%, 0.05%, 1%, 2%, 5%, or 10% by mass.
  • the buffer solution for DNA polymerase has a pH of 7.0-9.0, such as 7.0, 7.1, 7.2, 7.3, 7.4, 7.5, 7.6, 7.7, 7.8, 7.9, 8.0, 8.1, 8.2. , 8.3, 8.4, 8.5, 8.6, 8.7, 8.8, 8.9 or 9.0.
  • the buffer solution for DNA polymerase comprises: a monovalent salt ion (eg, sodium ion, chloride ion), a divalent salt ion (eg, magnesium ion, sulfate ion), Tris, and an organic solvent (eg DMSO or glycerol).
  • a monovalent salt ion eg, sodium ion, chloride ion
  • a divalent salt ion eg, magnesium ion, sulfate ion
  • Tris eg, Tris
  • an organic solvent eg DMSO or glycerol
  • the kit of the invention comprises one or more excision reagents capable of allowing a phosphodiester bond (1) and/or a phosphodiester in a compound of formula (I')
  • the bond (2) is cleaved and does not cleave the phosphodiester bond in the nucleic acid strand backbone to maintain the integrity of the nucleic acid strand backbone.
  • the excision agent is selected from the group consisting of an endonuclease and an IV alkaline phosphatase.
  • the kit of the invention further comprises one or more wash solutions.
  • wash solutions include, but are not limited to, phosphate buffer, citrate buffer, Tris-HCl buffer, acetate buffer, carbonate buffer, and the like.
  • the kit of the present invention may comprise any one or more of the above washing solutions.
  • modified nucleosides or nucleotides of the invention can be used to determine the sequence of a single-stranded polynucleotide of interest.
  • the present invention also provides the use of a compound as defined in any one of the above, and a kit as defined in any one of the above, for determining the sequence of a single-stranded polynucleotide of interest.
  • the modified nucleoside or nucleotide provided by the invention has higher stability in sequencing, and the blocking group and the detectable label thereof can be excised under mild conditions without damage to DNA, and Higher resection efficiency can be achieved and even complete resection can be achieved.
  • the modified nucleosides or nucleotides provided by the present invention can be polymerized by a commercially available DNA polymerase and have an acceptable polymerization efficiency, and under suitable conditions, even complete polymerization can be achieved without removing the phosphate group.
  • the ester protecting group can be directly excised, which simplifies the method of excision.
  • Example 1 Using the base T as an example, a commercially available compound I was used as a starting material to prepare a 3'-OH blocked dTTP having a Cy3 fluorescent group.
  • Pyridine was added to 52.4 mg (0.1 mmol) of compound III-1, and after several times of drying under high vacuum, a new anhydrous pyridine was added, and 800 ⁇ L of 1,4-diox was added to the solid after drying. Hexacyclohexane and 300 ⁇ L of anhydrous pyridine were added under argon atmosphere to 1,4-dioxol dissolved in 0.11 mmol (22 mg) of 2-chloro-4H-1,3,2-benzodioxan-4-one. 100 ⁇ L of six rings, reacted at room temperature, and stirred for 10 minutes.
  • Compound IV-1 is linked to compound VIII-1 to form dTTP having a fluorescent group Cy3 and 3'-OH is reversibly blocked.
  • Example 2 taking base A as an example, prepares a 3'-OH blocked nucleotide (dATP) having a Cy3 fluorescent group.
  • dATP 3'-OH blocked nucleotide
  • the modified dTTP of the present invention was polymerized using an exemplary template and primers, and the polymerization efficiency was tested to examine the effect of the polymerase and polymerization conditions on the polymerization efficiency.
  • Theminator (NEB), Taq (NEB), BST 2.0 (NEB), BST 3.0 (NEB), 9°Nm (NEB), KOD (merckmillipore)
  • KOD POL151 KOD POL151, KOD POL157, KOD POL171, KOD POL174, KOD POL376, KOD POL391
  • the above polymerase or a mutant thereof is obtained by purchase.
  • Figure 4 exemplifies the testing process.
  • the chip used for the test consisted of two polymerization reaction zones: zone 1 and zone 2, wherein zone 1 was the reaction of the reference group and zone 2 was the reaction of the group to be tested. It is known that Cy3-modified dideoxy dTTP can be polymerized 100% by Taq DNA polymerase under the test conditions mentioned below, thus serving as a reference group.
  • Cy3 modified dideoxy dTTP (triethylamine salt) was purchased from Jena Bioscience and its chemical structure is as follows:
  • the chip is photographed, and the background signal value is collected. Then, the reaction solution 1 containing Taq DNA polymerase and Cy3 modified dideoxy dTTP is added to the region 1, and the reaction solution 2 is added to the region 2, which contains the test to be tested.
  • the polymerase and the reversibly blocked dTTP with the fluorophore Cy3 prepared in Example 1 were each polymerized at a suitable temperature for 10 minutes, and the concentration of dTTP was 10 ⁇ M.
  • the buffer was polymerase product specification. Recommended buffer system. After the reaction, the chips were repeatedly washed with 1X phosphate buffer to remove unreacted dTTP; 1X phosphate buffer containing 10 mM of vitamin C was pumped, and both sides of the chip were photographed to collect signal values.
  • Each test yielded four values, including the blank background values (a and b) of the reference group and the test group, and the signal values (A and B) of the reference group and the test group after polymerization. It is known that the polymerization efficiency of the control group is 100%, and the polymerization efficiency of the test group can be calculated by the following equation:
  • the polymerization efficiency of the test group (B-b) / (A - a) * 100%.
  • test results showed that the polymerases in Table 1 were able to polymerize the dTTP prepared in Example 1, but the polymerization efficiency of other polymerases was lower than that of KOD.
  • the polymerization efficiency using KOD as a polymerase is acceptable.
  • mutants KOD POL151, KOD POL157, KOD POL171, using KOD polymerase, KOD POL174, KOD POL376, and KOD POL391 were tested as polymerases. These mutants have a poor polymerization effect when polymerizing some reversible blocking dNTPs in the prior art.
  • the dideoxy dTTP modified with Taq DNA polymerase and Cy3 was used as a reference group, and the template shown in SEQ ID NO: 1 and the primer shown in SEQ ID NO: 2, and the same experimental method as above, were used for polymerization for 10 minutes. .
  • the test results are shown in Table 2.
  • the inventors optimized the reaction conditions (including the pH of the reaction solution, the salt concentration, the buffer system concentration, the additive, and the reaction temperature) for polymerization using KOD POL391.
  • the initial reaction conditions were as follows: the pH of the reaction solution was 9.0, and contained 50 mM sodium chloride, 1 mM magnesium sulfate, 50 mM Tris, 0.05% Tween-20, and the reaction temperature was 55 °C.
  • the reaction conditions were optimized, the reference group was not used, but a comparison test was performed on the two reaction areas of the chip, and the polymerization time was 5 minutes. The better conditions that were first optimized were used in subsequent tests. The test results are shown in Table 3-1 to Table 3-6.
  • the reaction solution contained 50 mM sodium chloride, 1 mM magnesium sulfate, 50 mM Tris, 0.05% Tween-20, and the reaction temperature was 55 °C.
  • the reaction solution was pH 7.8, and contained 1 mM magnesium sulfate, 50 mM Tris, 0.05% Tween-20, and the reaction temperature was 55 °C.
  • the reaction solution was pH 7.8, and contained 20 mM sodium chloride, 50 mM Tris, 0.05% Tween-20, and the reaction temperature was 55 °C.
  • the reaction solution was pH 7.8, and contained 20 mM sodium chloride, 3 mM magnesium sulfate, 0.05% Tween-20, and the reaction temperature was 55 °C.
  • the reaction solution was pH 7.8, and contained 20 mM sodium chloride, 3 mM magnesium sulfate, 50 mM Tris, 0.05% Tween-20, and the reaction temperature was 55 °C.
  • reaction solution was pH 7.8 and contained 20 mM sodium chloride, 3 mM magnesium sulfate, 50 mM Tris, 0.05% Tween-20, 5% DMSO.
  • the polymerization efficiency of the polymerase KOD POL391 and KOD POL171 against the dTTP obtained in Example 1 was measured under the conditions of the final optimization, and the polymerization reaction was carried out for 10 minutes.
  • the reaction of the Cy3-modified dideoxy dTTP polymerized by Taq DNA polymerase was used as a reference group. The results are shown in Table 4.
  • the Cy3-modified reversibly blocked dTTP prepared in Example 1 can be efficiently polymerized by KOD POL391 or KOD POL171, and the polymerization efficiency is close to 100%.
  • the dTTP which is reversibly blocked but does not have a fluorescent group is first polymerized.
  • the dTTP which was used in the present embodiment to be reversibly blocked without a fluorescent group was the compound IV-1 in Example 1, and the structure was as follows. Under optimized conditions, it can be 100% polymerized.
  • the blocking group was excised using endonuclease IV (NEB), and the efficiency of excision was tested by the addition of the next base after excision.
  • NEB endonuclease IV
  • Figure 5 exemplifies the testing process.
  • a chip having two reaction regions (region 1 and region 2) is loaded, wherein the reaction of the reference group is performed on the region 1, and the reaction of the group to be tested is performed on the region 2.
  • Two different primers are added to the two regions, wherein the primer of the reference group is as shown in SEQ ID NO: 3, which is one more T at the 3 end than the primer of the test group (as shown in SEQ ID NO: 2).
  • the primer of the reference group is as shown in SEQ ID NO: 3, which is one more T at the 3 end than the primer of the test group (as shown in SEQ ID NO: 2).
  • the reference group and the test composition are systems having the same sequence, and the excision efficiency of the test group can be determined by comparing the signals of the next base.
  • Table 5 and Figure 6 show the efficiency of the ablation at different ablation times.
  • the fluorophore was removed by cleaving the phosphodiester bond (2) in the modified nucleotide of the present invention, and the efficiency of excision was tested.
  • Figure 7 exemplifies the testing process.
  • the CyT-modified and reversibly blocked dTTP prepared in Example 1 was polymerized on a chip, and the polymerase was KOD POL391.
  • the reaction conditions were the final optimized reaction conditions of Example 1, and the concentration of dTTP was 10 ⁇ M.
  • the signal values are collected before and after the polymerization, wherein the signal value before the polymerization is the background A1, and the signal value after the polymerization is a, and then the alkaline phosphatase (NEB, item number M0371S) is used for excision, and the 2, 5, 10 or 20 are respectively cut off. Minutes, after the excision, wash off the reagents, and then collect the signal value, which is A2, and calculate the resection efficiency according to the following equation:
  • the fluorophore on the nucleic acid strand can be completely removed after 20 min of excision.
  • Example 6 tested the stability of the azide methylene blocking group and the stability of the methyl phosphate blocking group.
  • a nucleotide with a protecting group at the 3' end and a fluorescent modification at the base 1482.3 (MH) - .

Abstract

The present invention relates to the field of nucleotide sequencing. Specifically, the invention relates to a modified nucleoside or nucleotide, wherein the 3'-OH of said modified nucleoside or said nucleotide is reversibly blocked, and the modified nucleoside or said nucleotid carries a detectable marker. The invention also relates to a kit containing said nucleoside or nucleotide, a method for preparing said nucleoside or nucleotide, and a sequencing method based on said nucleoside or nucleotide.

Description

修饰的核苷或核苷酸Modified nucleoside or nucleotide 技术领域Technical field
本发明涉及核酸测序领域。特别地,本发明提供了一种修饰的核苷或核苷酸,所述修饰的核苷或核苷酸的3’-OH被可逆阻断,并且,所述修饰的核苷或核苷酸携带有可检测标记。本发明还涉及包含所述核苷或核苷酸的试剂盒,制备所述核苷或核苷酸的方法,以及基于所述核苷或核苷酸的测序方法。The invention relates to the field of nucleic acid sequencing. In particular, the invention provides a modified nucleoside or nucleotide, the 3'-OH of the modified nucleoside or nucleotide is reversibly blocked, and the modified nucleoside or nucleotide Carry a detectable mark. The invention also relates to a kit comprising the nucleoside or nucleotide, a method of making the nucleoside or nucleotide, and a sequencing method based on the nucleoside or nucleotide.
背景技术Background technique
DNA测序技术包括以桑格(Sanger)测序法为代表的第一代DNA测序技术,以及以Illumina Hiseq2500,Roche 454,ABI Solid,BGISEQ-500等为代表的第二代DNA测序技术。桑格测序法具有实验操作简单、结果直观准确和实验周期短等特点,在对检测结果时效性要求很高的临床基因突变检测以及基因分型等领域有着广泛的应用。然而,桑格测序法的缺点是通量小、成本高,这限制了其在大规模基因测序中的应用。DNA sequencing technology includes the first generation DNA sequencing technology represented by Sanger sequencing, and the second generation DNA sequencing technology represented by Illumina Hiseq 2500, Roche 454, ABI Solid, BGI SEQ-500, and the like. The Sanger sequencing method has the characteristics of simple experimental operation, intuitive and accurate results, and short experimental period. It has wide application in the fields of clinical gene mutation detection and genotyping, which require high timeliness of detection results. However, the disadvantages of the Sanger sequencing method are small throughput and high cost, which limits its application in large-scale gene sequencing.
第二代DNA测序技术与第一代DNA测序技术相比,具有测序通量大、成本低、自动化程度高和单分子测序的特点。以Hiseq2500V2的测序技术为例,其一个实验流程可以产生10-200G碱基的数据,平均每个碱基的测序成本不到桑格测序法的测序成本的1/1000,并且所获得的测序结果可通过计算机直接进行处理和分析。因此,第二代DNA测序技术非常适合于大规模测序。Compared with the first generation of DNA sequencing technology, the second generation DNA sequencing technology has the characteristics of large sequencing throughput, low cost, high degree of automation and single molecule sequencing. Taking the sequencing technology of Hiseq 2500V2 as an example, an experimental procedure can generate 10-200 G base data, and the average cost per base is less than 1/1000 of the sequencing cost of the Sanger sequencing method, and the obtained sequencing result is obtained. It can be processed and analyzed directly by computer. Therefore, second generation DNA sequencing technology is very suitable for large-scale sequencing.
第二代测序技术中最关键的部分为边合成边测序,通常采取的手段是对脱氧核糖核苷三磷酸(dNTP)进行可逆阻断,简言之,这一过程包括:通过聚合酶催化,使带有可逆阻断基团和可检测标记的dNTP并入生长的核酸链,其中,可逆阻断基团对dNTP的3’-OH形成保护,可以防止已经并入核酸链的dNTP通过3’-OH与游离dNTP发生反应;对可检测标记进行检测,以判断引入的dNTP的种类;之后,将引入的dNTP上的阻断基团和可检测标记除去,并开始下一轮测序。The most critical part of the second-generation sequencing technology is sequencing by side synthesis. The usual approach is to reversibly block the deoxyribonucleoside triphosphate (dNTP). In short, this process involves: catalysis by polymerase, Incorporating a dNTP with a reversible blocking group and a detectable label into a growing nucleic acid strand, wherein the reversible blocking group protects the 3'-OH of the dNTP, preventing the dNTP that has been incorporated into the nucleic acid strand from passing through 3' -OH reacts with free dNTPs; detectable labels are detected to determine the type of dNTP introduced; thereafter, the blocking groups and detectable labels on the introduced dNTPs are removed and the next round of sequencing is initiated.
目前,用于对3’-OH进行可逆阻断的基团有多种,例如,Illumina采用叠氮亚甲基阻断3’位-OH,在聚合和检测之后,使用有机膦化物切除叠氮亚甲基,释放3’位-OH,从而可以继续下一个循环的测序;哥伦比亚大学Jingyue Ju研究组采用烯丙基阻断3’-OH,切除过程采用金属钯和三苯基膦三间磺酸三钠盐的配体,通过钯催化的去烯丙基反应除去阻断基团;此外,还有研究者利用碳酸酯和硫硫键进行可逆阻断,在切 断硫硫键之后,生成的巯基可以切断连接3’-OH的碳酸酯,从而完成去阻断。At present, there are various groups for reversible blocking of 3'-OH. For example, Illumina blocks the 3'-OH by azide methylene, and after polymerization and detection, the azide is removed using an organic phosphine. Methylene, release 3'-OH, so that the next cycle can be sequenced; Columbia University Jingyue Ju research group uses allyl block 3'-OH, the process uses metal palladium and triphenylphosphine triple sulfonate The ligand of the acid trisodium salt removes the blocking group by a palladium-catalyzed deallyl reaction; in addition, researchers have used the carbonate and sulfur-sulfur bond to reversibly block, in the cut After the sulfur-sulfur bond is broken, the resulting mercapto group can cleave the carbonate to which the 3'-OH is attached, thereby completing the deblocking.
在边合成边测序的方法中,对阻断方法的要求非常严格,以上所提到的阻断方法中,仅Illumina的利用叠氮亚甲基的阻断方法被成功用于测序上,其他方法使用起来面临各种各样的问题,因而目前还未被成功应用。造成这种局面的原因包括:1)DNA测序反应必须在接近中性的水溶液体系中进行,这使得许多化学上的反应和阻断基团无法应用;2)阻断后生成的基团必需非常稳定,而且切除反应需要迅速和高效,这样也排除了很多反应类型,例如上面的碳酸酯和硫硫键的方法,由于切除速率不够快而难以得到应用;3)切除反应不能对测序体系有影响,例如,将烯丙基作为阻断基团,在切除时需要用钯金属配合物进行催化,在实际应用中可能遇到不适于芯片的情况,最终导致并未有成熟的产品出现在市场上。In the method of sequencing while synthesizing, the blocking method is very strict. Among the above blocking methods, only Illumina's blocking method using azide methylene is successfully used for sequencing, and other methods. It faces a variety of problems in use and has not been successfully applied. The reasons for this situation include: 1) DNA sequencing reactions must be carried out in a near neutral aqueous system, which makes many chemical reactions and blocking groups unusable; 2) the groups generated after blocking must be very Stable, and the resection reaction needs to be rapid and efficient, which also eliminates many types of reactions, such as the above carbonate and sulfur-sulfur bond methods, which are difficult to apply due to the fast resection rate; 3) the resection reaction does not affect the sequencing system. For example, the allyl group is used as a blocking group, and it needs to be catalyzed by a palladium metal complex during excision. In practical applications, it may be unsuitable for the chip, and eventually no mature product appears on the market. .
此外,虽然Illumina采用的叠氮亚甲基阻断技术已经比较成熟,但也存在一些问题,包括叠氮的稳定性不够,会导致测序质量降低。为了解决这些问题,Illumina在专利申请WO2014139596A1中,优化了叠氮亚甲基,增加了稳定性,但尽管切除效率能达到100%,但是仍然需要较高温度,而且切除试剂对DNA有不好的影响。此外,Illumina在其专利申请US20130085073A1中,使用磷酸阻断的dNTP进行边合成边测序,但是,该专利中明确显示了,只有磷酸上的羟基全部被保护之后才能被聚合酶聚合,而羟基全部被保护的磷酸基团无法被内切酶IV切除。因此,该专利公开的方法包括:对3’-OH被磷酸基团所阻断的dNTP进行聚合,其中,磷酸基团上的羟基被全部保护,之后进行分步切除,即,首先去掉磷酸基团上的一个羟基保护基,之后通过内切酶IV将整个磷酸基团切除,其中,去掉一个保护基团所用的方法包括光切断法,以及Ada(甲基转移酶)去除甲基。该专利公开的方法包括:对3’-OH被磷酸基团所阻断的dNTP进行聚合,其中,磷酸基团上的虽然理论上可行,但在实际应用中非常麻烦繁琐。In addition, although the azide methylene blocking technology used by Illumina is relatively mature, there are also some problems, including insufficient stability of the azide, which leads to a decrease in sequencing quality. In order to solve these problems, in the patent application WO2014139596A1, Illumina optimizes the azidomethylene, which increases the stability, but although the resection efficiency can reach 100%, it still requires a higher temperature, and the excision reagent is not good for DNA. influences. In addition, in its patent application US20130085073A1, Illumina uses a phospho-blocked dNTP for sequencing while synthesizing, but it is clearly shown in the patent that only the hydroxyl groups on the phosphoric acid are all protected before being polymerized by the polymerase, and the hydroxyl groups are all The protected phosphate group cannot be cleaved by endonuclease IV. Thus, the method disclosed in the patent comprises: polymerizing a dNTP in which 3'-OH is blocked by a phosphate group, wherein the hydroxyl group on the phosphate group is completely protected, followed by stepwise excision, that is, first removing the phosphate group A hydroxy protecting group on the cleavage, followed by excision of the entire phosphate group by endonuclease IV, wherein the method used to remove a protecting group includes photocleavage, and Ada (methyltransferase) removes the methyl group. The method disclosed in the patent comprises: polymerizing a dNTP in which 3'-OH is blocked by a phosphate group, wherein the phosphate group, although theoretically feasible, is very cumbersome in practical use.
发明内容Summary of the invention
为了解决上述问题,本申请的发明人开发了一种新的3’-OH被可逆阻断的核苷酸,其具有更高的稳定性,可以被聚合酶高效甚至完全聚合,并且,可以在对DNA无损伤的条件下高效甚至100%切除。In order to solve the above problems, the inventors of the present application have developed a novel 3'-OH reversibly blocked nucleotide which has higher stability, can be efficiently or even completely polymerized by a polymerase, and can be Efficient or even 100% resection without damage to DNA.
发明概述 Summary of invention
本申请的发明人开发了一种新的修饰的核苷或核苷酸,其中,3’-OH被阻断基团阻断,阻断基团可以在不破坏核酸链的条件下,被切除试剂切除,生成游离的3’-OH。所述修饰的核苷或核苷酸还带有可检测标记,所述可检测标记通过包含磷酸二酯键的连接体与所述核苷酸中的碱基相连,可以利用切除试剂(与切除阻断基团的切除试剂相同或不同),在不破坏核酸链的条件下,使所述磷酸二酯键断裂,以去除可检测标记。The inventors of the present application have developed a novel modified nucleoside or nucleotide in which 3'-OH is blocked by a blocking group, and the blocking group can be cleaved without destroying the nucleic acid strand. The reagent is excised to form a free 3'-OH. The modified nucleoside or nucleotide also carries a detectable label, and the detectable label is linked to the base in the nucleotide by a linker comprising a phosphodiester bond, and the excision reagent can be utilized (with excision) The excision reagent of the blocking group is the same or different), and the phosphodiester bond is cleaved to remove the detectable label without destroying the nucleic acid strand.
因此,在一个方面,本申请提供了具有通式(I)所示结构的化合物,Thus, in one aspect, the application provides a compound having the structure of formula (I),
Figure PCTCN2017105734-appb-000001
Figure PCTCN2017105734-appb-000001
其中,R1和R3各自独立地选自
Figure PCTCN2017105734-appb-000002
Figure PCTCN2017105734-appb-000003
Wherein R 1 and R 3 are each independently selected from
Figure PCTCN2017105734-appb-000002
Figure PCTCN2017105734-appb-000003
A选自苯基、萘基、吲哚基和吡啶基;A is selected from the group consisting of phenyl, naphthyl, anthryl and pyridyl;
R2选自硝基、卤代C1-4烷基、卤素、氢、醛基、
Figure PCTCN2017105734-appb-000004
其中,Q独立地选自C1-4烷基;
R 2 is selected from the group consisting of a nitro group, a halogenated C 1-4 alkyl group, a halogen, a hydrogen, an aldehyde group,
Figure PCTCN2017105734-appb-000004
Wherein Q is independently selected from C 1-4 alkyl;
R4选自-H,单磷酸基团(-PO3H2),二磷酸基团(-PO3H-PO3H2),三磷酸基团(-PO3H-PO3H-PO3H2)和四磷酸基团(-PO3H-PO3H-PO3H-PO3H2);R 4 is selected from -H, a monophosphate group (-PO 3 H 2 ), a diphosphate group (-PO 3 H-PO 3 H 2 ), a triphosphate group (-PO 3 H-PO 3 H-PO 3 H 2 ) and a tetraphosphoric acid group (-PO 3 H-PO 3 H-PO 3 H-PO 3 H 2 );
R6为氢或羟基;R 6 is hydrogen or a hydroxyl group;
m和n各自独立地选自0、1、2、3、4、5;m and n are each independently selected from 0, 1, 2, 3, 4, 5;
L为连接基团或不存在;L is a linking group or does not exist;
Base代表碱基,例如为嘌呤碱基或嘧啶碱基,例如选自A、T、U、C和G中的一种;Base represents a base, such as a purine base or a pyrimidine base, for example, one selected from the group consisting of A, T, U, C, and G;
Label表示可检测标记,例如为荧光基团;Label indicates a detectable label, such as a fluorophore;
Blocker表示阻断基团。Blocker indicates a blocking group.
在一个方面,本申请还提供了制备如上文所述的修饰的核苷或核苷酸的方法。In one aspect, the application also provides a method of making a modified nucleoside or nucleotide as described above.
基于本发明的修饰的核苷或核苷酸,本申请的发明人还开发了一种多核苷酸的测序方法。本发明的测序方法中,一边合成目标单链多核苷酸互补的生长的多核苷酸,一边进行测序。 Based on the modified nucleosides or nucleotides of the present invention, the inventors of the present application have also developed a sequencing method for polynucleotides. In the sequencing method of the present invention, sequencing is performed while synthesizing a polynucleotide in which a target single-stranded polynucleotide is complementary to each other.
因此,在一个方面,本申请提供了一种制备在测序反应中与目标单链多核苷酸互补的生长的多核苷酸的方法,其包括将如上文所定义的化合物并入所述生长的互补多核苷酸,其中,所述化合物的并入防止了任何后续的核苷酸引入所述生长的互补多核苷酸中。Thus, in one aspect, the application provides a method of preparing a growing polynucleotide complementary to a target single-stranded polynucleotide in a sequencing reaction, comprising incorporating a compound as defined above into the complementary of said growth A polynucleotide, wherein the incorporation of the compound prevents any subsequent nucleotides from being introduced into the growing complementary polynucleotide.
在另一个方面,本申请提供了一种测定目标单链多核苷酸的序列的方法,其包括:监测互补核苷酸的顺序并入,其中并入的至少一个互补核苷酸是如上文所定义的化合物,以及,检测所述化合物携带的可检测标记。In another aspect, the application provides a method of determining the sequence of a single-stranded polynucleotide of interest, comprising: monitoring sequential incorporation of complementary nucleotides, wherein at least one complementary nucleotide that is incorporated is as described above A defined compound, and detecting a detectable label carried by the compound.
在某些实施方案中,所述测定目标单链多核苷酸的序列的方法包括:In certain embodiments, the method of determining the sequence of a single-stranded polynucleotide of interest comprises:
(a)提供包含双链体、通式(I)的化合物、聚合酶和切除试剂的混合物;所述双链体包含生长的核酸链以及待测序的核酸分子;(a) providing a mixture comprising a duplex, a compound of formula (I), a polymerase, and a excision reagent; said duplex comprising a growing nucleic acid strand and a nucleic acid molecule to be sequenced;
(b)进行包含以下步骤(i)、(ii)和(iii)的反应循环:(b) Carry out a reaction cycle comprising the following steps (i), (ii) and (iii):
步骤(i):使用聚合酶,使所述化合物并入生长的核酸链,形成包含阻断基团和可检测标记的核酸中间体;Step (i): using a polymerase to incorporate the compound into a growing nucleic acid strand to form a nucleic acid intermediate comprising a blocking group and a detectable label;
步骤(ii):对所述核酸中间体上的可检测标记进行检测;Step (ii): detecting a detectable label on the nucleic acid intermediate;
步骤(iii):使用切除试剂将核酸中间体上的阻断基团除去。Step (iii): The blocking group on the nucleic acid intermediate is removed using a resection reagent.
在某些实施方案中,所述反应循环还包括步骤(iv):使用切除试剂将核酸中间体上的可检测标记除去。In certain embodiments, the reaction cycle further comprises the step (iv) of removing the detectable label on the nucleic acid intermediate using a scavenging reagent.
在某些实施方案中,所述步骤(iii)和步骤(iv)中使用的切除试剂是同样的试剂。在某些实施方案中,所述步骤(iii)和步骤(iv)中使用的切除试剂是不同的试剂。In certain embodiments, the ablation reagents used in steps (iii) and (iv) are the same reagents. In certain embodiments, the ablation reagents used in steps (iii) and (iv) are different reagents.
在一个方面,本发明提供了一种试剂盒,其包含第一、第二、第三和第四化合物,所述第一、第二、第三和第四化合物各自为如上定义的通式(I)的化合物,所述四种化合物分别为核苷酸A、(T/U)、C和G的衍生物,且具有碱基互补配对能力。In one aspect, the invention provides a kit comprising first, second, third and fourth compounds, each of said first, second, third and fourth compounds being of the formula A compound of I) which is a derivative of nucleotides A, (T/U), C and G, respectively, and which has a base complementary pairing ability.
下面将结合附图和发明详述来对本发明的实施方案进行详细阐释。但是,本领域技术人员将理解,下列附图和发明详述仅用于说明本发明,而不是对本发明的范围的限定。根据附图和发明详述的详细公开内容,本发明的各种目的和有利方面对于本领域技术人员来说将变得显然。The embodiments of the present invention are explained in detail below in conjunction with the drawings and the detailed description of the invention. However, those skilled in the art will understand that the following drawings and the detailed description of the invention are only intended to illustrate the invention and not to limit the scope of the invention. The various objects and advantageous aspects of the invention will be apparent to those skilled in the <
附图简述BRIEF DESCRIPTION OF THE DRAWINGS
图1为实施例1中,制备带有Cy3-标记且3’-OH被阻断的dTTP的过程中,所涉及 的中间化合物IV-1的HPLC图谱。1 is a process in which a dTTP having a Cy3-tag and a 3'-OH is blocked is prepared in Example 1, and is involved in HPLC chromatogram of intermediate compound IV-1.
图2为实施例1制得的带有Cy3-标记且3’-OH被阻断的dTTP的HPLC图谱。Figure 2 is an HPLC chromatogram of dTTP with Cy3-labeled and 3'-OH blocked in Example 1.
图3为实施例2中,制备带有Cy3-标记且3’-OH被阻断的dATP的过程中,所涉及的中间化合物IV-2的HPLC图谱。Figure 3 is an HPLC chart of the intermediate compound IV-2 involved in the preparation of dATP with Cy3-labeled and 3'-OH blocked in Example 2.
图4示例性地说明了实施例3中,测定dTTP聚合效率的过程。Fig. 4 exemplarily illustrates the process of determining the polymerization efficiency of dTTP in Example 3.
图5示例性地说明了实施例4中,测定阻断基团的切除效率的过程。Fig. 5 exemplarily illustrates the process of determining the excision efficiency of the blocking group in Example 4.
图6显示了实施例4中,不同切除时间下阻断基团被切除的效率。Figure 6 shows the efficiency of the blocking group being cleaved at different excision times in Example 4.
图7示例性地说明了实施例5中,测定荧光基团的切除效率的过程。Fig. 7 exemplarily illustrates the process of determining the excision efficiency of the fluorophore in Example 5.
图8显示了实施例5中,不同切除时间下荧光基团被切除的效率。Figure 8 shows the efficiency of fluorophore excision at different cut-off times in Example 5.
图9为实施例6中被磷酸甲酯阻断的dTTP在稳定性测试后的LC图谱。Figure 9 is an LC spectrum of dTTP blocked by methyl phosphate in Example 6 after stability testing.
图10为实施例6中被叠氮亚甲基阻断的dTTP在稳定性测试后的LC图谱。Figure 10 is a LC map of the azide methylene-blocked dTTP in Example 6 after stability testing.
本发明涉及的序列的信息提供于下表中The information of the sequences involved in the present invention is provided in the following table.
序列号(SEQ ID NO:)Serial number (SEQ ID NO:) 描述 description
11 模板template
22 引物Primer
33 引物Primer
序列信息Sequence information
序列1(SEQ ID NO:1):33bpSequence 1 (SEQ ID NO: 1): 33 bp
Figure PCTCN2017105734-appb-000005
Figure PCTCN2017105734-appb-000005
序列2(SEQ ID NO:2):31bpSequence 2 (SEQ ID NO: 2): 31 bp
Figure PCTCN2017105734-appb-000006
Figure PCTCN2017105734-appb-000006
序列3(SEQ ID NO:3):32bpSequence 3 (SEQ ID NO: 3): 32 bp
Figure PCTCN2017105734-appb-000007
Figure PCTCN2017105734-appb-000007
发明详述Detailed description of the invention
在本发明中,除非另外定义,否则本文所用的全部技术和科学术语都具有与本发明所属领域普通技术人员通常所理解的相同意思。在本发明的实施方案中,可以使用与本文所述的那些方法和材料类似或等同的方法和材料,下文仅仅是描述了例示性的适合的方法和材料。将所有公开出版物、专利申请、专利和其它参考文献并入本文作为参考。此外,所述材料、方法和实施例仅仅是示范性的而非眼制性的。同时,为了更好地理解本发明,下面提供相关术语的定义和解释。 In the present invention, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which the invention belongs, unless otherwise defined. In the embodiments of the present invention, methods and materials similar or equivalent to those described herein can be used, and only the exemplary suitable methods and materials are described below. All publications, patent applications, patents, and other references are incorporated herein by reference. Moreover, the materials, methods, and examples are illustrative only and not imaginative. Also, for a better understanding of the present invention, definitions and explanations of related terms are provided below.
如本文中所使用的,术语“C1-4烷基”是指直链或支链的含有1-4个碳原子的烷基,包括但不限于C1-2烷基、C1-3烷基、C2-4烷基,例如:甲基、乙基、正丙基、异丙基、正丁基、仲丁基、异丁基、叔丁基等。As used herein, the term "C 1-4 alkyl" refers to a straight or branched alkyl group having from 1 to 4 carbon atoms, including but not limited to C 1-2 alkyl, C 1-3 Alkyl, C 2-4 alkyl, for example: methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, isobutyl, tert-butyl and the like.
如本文中所使用的,术语“卤代”是指基团或化合物上的氢被一个或多个(例如2、3、4、5、6、7、8、9个)卤素原子取代,包括全卤代和部分卤代。As used herein, the term "halo" refers to the replacement of a hydrogen on a group or compound by one or more (eg, 2, 3, 4, 5, 6, 7, 8, 9) halogen atoms, including Fully halogenated and partially halogenated.
如本文中所使用的,术语“卤素”包括氟、氯、溴、碘。As used herein, the term "halogen" includes fluoro, chloro, bromo, iodo.
如本文中所使用的,术语“卤代C1-4烷基”指一至多个卤素原子取代C1-4烷基上的一个或多个氢原子所衍生的基团,所述“卤素”和“C1-4烷基”如前文所定义。卤代C1-4烷基包括但不限于卤代C1-2烷基、卤代C1-3烷基、卤代C2-4烷基,例如卤代甲基(例如氟代甲基、氯代甲基)、卤代乙基(例如氟代乙基、氯代乙基)、卤代正丙基、卤代异丙基、卤代正丁基、卤代仲丁基、卤代异丁基、卤代叔丁基。As used herein, the term "halo C 1-4 alkyl" refers to a radical derived from one or more halogen atoms substituted by one or more hydrogen atoms on a C 1-4 alkyl group, said "halogen" And "C 1-4 alkyl" are as defined above. Halo C 1-4 alkyl includes, but is not limited to, halo C 1-2 alkyl, halo C 1-3 alkyl, halo C 2-4 alkyl, such as halomethyl (eg fluoromethyl) , chloromethyl), haloethyl (eg fluoroethyl, chloroethyl), halopropyl, haloisopropyl, halo-n-butyl, halo sec-butyl, halogenated Isobutyl, halogenated tert-butyl.
如本文中所使用的,术语“阻断”是指,使用特定的基团对核苷或核苷酸的3’-OH形成保护,以使可能的聚合酶(例如DNA聚合酶)的聚合作用终止。被用于阻断的基团被称为“阻断基团”。在某些优选的实施方案中,阻断基团能够被去除,从而,受阻断的羟基能转化为具有反应性的羟基,这样的阻断被称为“可逆阻断”。被用于可逆阻断的基团被称为“可逆阻断基团”。As used herein, the term "blocking" refers to the formation of a 3'-OH formation of a nucleoside or nucleotide using a specific group to polymerize a possible polymerase (eg, a DNA polymerase). termination. The group that is used for blocking is referred to as a "blocking group." In certain preferred embodiments, the blocking group can be removed such that the blocked hydroxyl group can be converted to a reactive hydroxyl group, such blockage being referred to as "reversible blocking." The group used for reversible blocking is referred to as a "reversible blocking group."
如本文中所使用的,术语“支持物”是指允许核酸稳定附着的任何材料(固体或半固体),例如乳胶珠、葡聚糖珠、聚苯乙烯、聚丙烯、聚丙烯酰胺凝胶、金薄层、玻璃和硅片等。在一些示例性实施方案中,所述支持物是光学透明的,例如玻璃。如本文中所使用的,“稳定附着”是指,核酸分子与支持物之间的连接足够强,从而核酸分子不会因各种反应或处理(例如聚合反应和洗涤处理)所使用的条件而脱离支持物。As used herein, the term "support" refers to any material (solid or semi-solid) that allows for stable attachment of nucleic acids, such as latex beads, dextran beads, polystyrene, polypropylene, polyacrylamide gels, Gold thin layers, glass and silicon wafers. In some exemplary embodiments, the support is optically clear, such as glass. As used herein, "stable attachment" means that the linkage between the nucleic acid molecule and the support is sufficiently strong that the nucleic acid molecule is not subjected to various reaction or treatment conditions (eg, polymerization and washing treatment). Detach the support.
如本文中所使用的,术语“连接”意欲涵盖任何形式的连接,例如共价连接和非共价连接。在某些示例性实施方案中,核酸分子优选地通过共价方式与支持物相连接。As used herein, the term "connected" is intended to cover any form of linkage, such as covalent linkages and non-covalent linkages. In certain exemplary embodiments, the nucleic acid molecule is preferably linked to the support by a covalent means.
如本文中所使用的,术语“片段化”是指,将大的核酸片段(例如大的DNA片段)转变成小的核酸片段(例如小的DNA片段)的过程。在某些实施方案中,术语“大的核酸片段”意欲涵盖大于5kb,大于10kb,大于25kb的核酸分子(例如DNA),例如大于500kb,大于1Mb,大于5Mb或更大的核酸分子(例如DNA)。As used herein, the term "fragmentation" refers to the process of converting a large nucleic acid fragment (eg, a large DNA fragment) into a small nucleic acid fragment (eg, a small DNA fragment). In certain embodiments, the term "large nucleic acid fragment" is intended to encompass nucleic acid molecules (eg, DNA) greater than 5 kb, greater than 10 kb, greater than 25 kb, eg, greater than 500 kb, greater than 1 Mb, greater than 5 Mb or greater nucleic acid molecules (eg, DNA) ).
如本文中所使用的,术语“末端补齐”是指,将具有悬突末端的核酸分子的末端补齐,形成具有钝末端的核酸分子的过程。 As used herein, the term "end-filled" refers to the process of complementing the ends of a nucleic acid molecule having an overhanging end to form a nucleic acid molecule having a blunt end.
在本文中,术语“接头”和“接头序列”可互换使用。如本文中所使用的,术语“接头”和“接头序列”是指,在核酸分子的5'和/或3'端人为引入的一段寡核苷酸序列。接头通常可包含一个或多个用于实现特定功能的区域。由此,当在核酸分子的5'和/或3'端人为引入接头时,接头将能够实现所述特定功能,从而利于后续应用。例如,接头可包含一个或多个引物结合区,以便于引物的结合。在一些示例性实施方案中,所述接头可包含一个或多个引物结合区,例如能够与用于进行扩增的引物杂交的引物结合区,和/或能够与用于测序反应的引物杂交的引物结合区。在某些优选实施方案中,所述接头包含,能够与通用引物杂交的通用接头序列,例如,能够与通用扩增引物和/或通用测序引物杂交的通用接头序列。由此,可方便地通过使用通用扩增引物和/或通用测序引物,对携带有接头的核酸分子进行扩增和/或测序。在一些示例性实施方案中,所述接头还可包含标签或标签序列。As used herein, the terms "linker" and "linker sequence" are used interchangeably. As used herein, the terms "linker" and "linker sequence" refer to a stretch of oligonucleotide sequences introduced human at the 5' and/or 3' end of a nucleic acid molecule. A connector can typically contain one or more regions for achieving a particular function. Thus, when a linker is introduced artificially at the 5' and/or 3' end of the nucleic acid molecule, the linker will be able to perform the specific function, thereby facilitating subsequent applications. For example, the linker can comprise one or more primer binding regions to facilitate binding of the primers. In some exemplary embodiments, the linker may comprise one or more primer binding regions, such as a primer binding region capable of hybridizing to a primer for amplification, and/or capable of hybridizing to a primer for use in a sequencing reaction. Primer binding region. In certain preferred embodiments, the linker comprises a universal linker sequence capable of hybridizing to a universal primer, for example, a universal linker sequence capable of hybridizing to a universal amplification primer and/or a universal sequencing primer. Thus, the nucleic acid molecule carrying the linker can be conveniently amplified and/or sequenced by using universal amplification primers and/or universal sequencing primers. In some exemplary embodiments, the linker can also comprise a tag or tag sequence.
在本文中,术语“标签”和“标签序列”可互换使用。如本文中所使用的,术语“标签”和“标签序列”是指,在核酸分子的5'和/或3'端人为引入的、具有特定碱基序列的一段寡核苷酸序列。标签通常用于鉴别/区分核酸分子的来源。例如,可在不同来源的核酸分子中分别引入不同的标签,由此,当这些不同来源的核酸分子混合在一起时,可通过各个核酸分子上携带的独特标签序列,准确地确定每一个核酸分子的来源。根据实际需要,标签序列可以具有任意长度,例如2-50bp,例如2、3、4、5、10、15、20、25、30、35、40、45或50bp。As used herein, the terms "tag" and "tag sequence" are used interchangeably. As used herein, the terms "tag" and "tag sequence" refer to a segment of an oligonucleotide sequence introduced at the 5' and/or 3' end of a nucleic acid molecule with a particular base sequence. Labels are commonly used to identify/distinguish the source of nucleic acid molecules. For example, different tags can be introduced in different nucleic acid molecules from different sources, whereby when these nucleic acid molecules of different origin are mixed together, each nucleic acid molecule can be accurately determined by the unique tag sequence carried on each nucleic acid molecule. origin of. The tag sequence can have any length, such as 2-50 bp, such as 2, 3, 4, 5, 10, 15, 20, 25, 30, 35, 40, 45 or 50 bp, depending on actual needs.
如本文中所使用的,术语“杂交”通常是指,严紧条件下的杂交。在分子生物学领域中杂交技术是熟知的。为了举例说明的目的,所述严紧条件包括例如,中度严紧条件(例如,在6×氯化钠/柠檬酸钠(SSC)中约45℃下杂交,然后在0.2×SSC/0.1%SDS中于约50-65℃下进行一次或多次洗涤);高度严紧条件(例如,在6×SSC中约45℃下杂交,然后在0.1×SSC/0.2%SDS中于约68℃下进行一次或多次洗涤);以及本领域技术人员已知的其它严紧杂交条件(参见例如Ausubel,F.M.等编,1989,Current Protocols in Molecular Biology,第1卷,Green Publishing Associates,Inc.,和John Wiley&Sons,Inc.,纽约,第6.3.1-6.3.6和2.10.3页)。As used herein, the term "hybridization" generally refers to hybridization under stringent conditions. Hybridization techniques are well known in the field of molecular biology. For illustrative purposes, the stringent conditions include, for example, moderate stringency conditions (eg, hybridization at about 45 ° C in 6 x sodium chloride / sodium citrate (SSC), then in 0.2 x SSC / 0.1% SDS One or more washes at about 50-65 ° C; high stringency conditions (eg, hybridization at about 45 ° C in 6 x SSC, then at about 68 ° C in 0.1 x SSC / 0.2% SDS or Multiple washes; and other stringent hybridization conditions known to those skilled in the art (see, for example, Ausubel, FM et al, 1989, Current Protocols in Molecular Biology, Vol. 1, Green Publishing Associates, Inc., and John Wiley & Sons, Inc. New York, pages 6.3.1-6.3.6 and 2.10.3).
如本文中所使用的,表述“含有溶液相和固相的反应体系”是指,本发明的反应体系既包含支持物以及连接于支持物的物质(固相),又包含溶解于溶液/溶剂中的物质(溶液相)。相应地,表述“移除反应体系的溶液相”是指,将反应体系中的溶液以及其包含的物质(溶液相)移除,而仅保留反应体系中的支持物以及连接于支持物的物 质(固相)。在本发明的背景中,连接于支持物的物质(固相)可包括,待测序的核酸分子,生长的核酸链,和/或由待测序的核酸分子和生长的核酸链形成的双链体。As used herein, the expression "reaction system containing a solution phase and a solid phase" means that the reaction system of the present invention comprises both a support and a substance attached to the support (solid phase), and a solution/solvent dissolved in the solution/solvent. The substance in the solution (solution phase). Accordingly, the expression "removing the solution phase of the reaction system" means removing the solution in the reaction system and the substance (solution phase) contained therein, and retaining only the support in the reaction system and the substance attached to the support. Quality (solid phase). In the context of the present invention, the substance (solid phase) attached to the support may comprise a nucleic acid molecule to be sequenced, a growing nucleic acid strand, and/or a duplex formed by the nucleic acid molecule to be sequenced and the growing nucleic acid strand. .
如本文中所使用的,术语“引物”是指,含有能与互补序列杂交并引发特异性聚合反应的寡核苷酸序列。在通常情况下,选择/设计引物的序列,以使其对互补序列具有最大杂交活性,而对其他序列具有非常低的非特异性杂交活性,从而尽可能减少非特异性扩增。设计引物的方法是本领域技术人员公知的,并且可用商业化的软件(例如Primer Premier version 6.0,Oligo version 7.36等)来执行。As used herein, the term "primer" refers to an oligonucleotide sequence that hybridizes to a complementary sequence and initiates a specific polymerization reaction. In general, the sequence of the primer is selected/designed to have maximum hybridization activity for the complementary sequence, while having very low non-specific hybridization activity for other sequences, thereby minimizing non-specific amplification. Methods for designing primers are well known to those skilled in the art and can be performed using commercially available software (e.g., Primer Premier version 6.0, Oligo version 7.36, etc.).
如本文中所使用的,术语“聚合酶”是指,能够进行核苷酸聚合反应的酶。此类酶能够按照碱基互补配对原则,在生长的核酸链的3'端引入与模板核酸相应位置的核苷酸配对的核苷酸。As used herein, the term "polymerase" refers to an enzyme capable of performing a nucleotide polymerization reaction. Such an enzyme is capable of introducing a nucleotide paired with a nucleotide at a position corresponding to a template nucleic acid at the 3' end of the growing nucleic acid strand according to the principle of base complementary pairing.
如本文中所使用的,表述“A、(T/U)、C和G”意欲涵盖两种情况:“A、T、C和G”和“A、U、C和G”。因此,表述“所述四种化合物分别为核苷酸A、(T/U)、C和G的衍生物”意欲表示,所述四种化合物分别为核苷酸A、T、C和G的衍生物,或者分别为核苷酸A、U、C和G的衍生物。As used herein, the expressions "A, (T/U), C, and G" are intended to cover two instances: "A, T, C, and G" and "A, U, C, and G." Thus, the expression "the four compounds are derivatives of nucleotides A, (T/U), C and G, respectively" is intended to mean that the four compounds are nucleotides A, T, C and G, respectively. Derivatives, or derivatives of nucleotides A, U, C and G, respectively.
如本文中所使用的,表述“化合物具有碱基互补配对能力”是指,化合物能够按照碱基互补配对原则,与对应的碱基配对并形成氢键。按照碱基互补配对原则,碱基A能够与碱基T或U配对,碱基G能够与碱基C配对。因此,当具有碱基互补配对能力的化合物为核苷酸A的衍生物时,其将能够与碱基T或U配对;当具有碱基互补配对能力的化合物为核苷酸T或U的衍生物时,其将能够与碱基A配对;当具有碱基互补配对能力的化合物为核苷酸C的衍生物时,其将能够与碱基G配对;当具有碱基互补配对能力的化合物为核苷酸G的衍生物时,其将能够与碱基C配对。As used herein, the expression "a compound having a base complementary pairing ability" means that the compound is capable of pairing with a corresponding base and forming a hydrogen bond according to the principle of base complementary pairing. According to the principle of base complementary pairing, base A can be paired with base T or U, and base G can be paired with base C. Therefore, when a compound having a base complementary pairing ability is a derivative of nucleotide A, it will be able to pair with a base T or U; when a compound having a base complementary pairing ability is a derivative of a nucleotide T or U When it is a substance, it will be able to pair with base A; when a compound having a base complementary pairing ability is a derivative of nucleotide C, it will be able to pair with base G; when a compound having a base complementary pairing ability is In the case of a derivative of nucleotide G, it will be able to pair with base C.
(一)修饰的核苷或核苷酸及其制备方法(1) Modified nucleoside or nucleotide and preparation method thereof
修饰的核苷或核苷酸Modified nucleoside or nucleotide
本申请的发明人开发了一种新的修饰的核苷或核苷酸,其中,3’-OH被阻断基团阻断,阻断基团可以在不破坏核酸链的条件下,被切除试剂切除,生成游离的3’-OH。所述修饰的核苷或核苷酸还带有可检测标记,所述可检测标记通过包含磷酸二酯键的连接体与所述核苷酸中的碱基相连,可以利用切除试剂(与切除阻断基团的切除试剂相同或不同),在不破坏核酸链的条件下,使所述磷酸二酯键断裂,以去除可检测标记。 The inventors of the present application have developed a novel modified nucleoside or nucleotide in which 3'-OH is blocked by a blocking group, and the blocking group can be cleaved without destroying the nucleic acid strand. The reagent is excised to form a free 3'-OH. The modified nucleoside or nucleotide also carries a detectable label, and the detectable label is linked to the base in the nucleotide by a linker comprising a phosphodiester bond, and the excision reagent can be utilized (with excision) The excision reagent of the blocking group is the same or different), and the phosphodiester bond is cleaved to remove the detectable label without destroying the nucleic acid strand.
因此,在一个方面,本申请提供了具有通式(I)所示结构的化合物,Thus, in one aspect, the application provides a compound having the structure of formula (I),
Figure PCTCN2017105734-appb-000008
Figure PCTCN2017105734-appb-000008
其中,R1和R3各自独立地选自
Figure PCTCN2017105734-appb-000009
Figure PCTCN2017105734-appb-000010
Wherein R 1 and R 3 are each independently selected from
Figure PCTCN2017105734-appb-000009
Figure PCTCN2017105734-appb-000010
A选自苯基、萘基、吲哚基和吡啶基;A is selected from the group consisting of phenyl, naphthyl, anthryl and pyridyl;
R2选自硝基、卤代C1-4烷基(例如氟代C1-4烷基)、卤素(例如氟、氯、溴、碘)、氢、醛基、
Figure PCTCN2017105734-appb-000011
其中,Q独立地选自C1-4烷基(例如C1-2烷基、C1-3烷基、C2-4烷基,例如甲基、乙基、正丙基、异丙基、正丁基、仲丁基、异丁基、叔丁基);
R 2 is selected from the group consisting of nitro, halo C 1-4 alkyl (eg, fluoro C 1-4 alkyl), halogen (eg, fluorine, chlorine, bromine, iodine), hydrogen, aldehyde,
Figure PCTCN2017105734-appb-000011
Wherein Q is independently selected from C 1-4 alkyl (eg, C 1-2 alkyl, C 1-3 alkyl, C 2-4 alkyl, such as methyl, ethyl, n-propyl, isopropyl , n-butyl, sec-butyl, isobutyl, tert-butyl);
R4选自-H,单磷酸基团(-PO3H2),二磷酸基团(-PO3H-PO3H2),三磷酸基团(-PO3H-PO3H-PO3H2)和四磷酸基团(-PO3H-PO3H-PO3H-PO3H2);R 4 is selected from -H, a monophosphate group (-PO 3 H 2 ), a diphosphate group (-PO 3 H-PO 3 H 2 ), a triphosphate group (-PO 3 H-PO 3 H-PO 3 H 2 ) and a tetraphosphoric acid group (-PO 3 H-PO 3 H-PO 3 H-PO 3 H 2 );
R6为氢或羟基;R 6 is hydrogen or a hydroxyl group;
m和n各自独立地选自0、1、2、3、4、5;m and n are each independently selected from 0, 1, 2, 3, 4, 5;
L为连接基团或不存在;L is a linking group or does not exist;
Base代表碱基,例如为嘌呤碱基或嘧啶碱基,例如选自A、T、U、C和G中的一种;Base represents a base, such as a purine base or a pyrimidine base, for example, one selected from the group consisting of A, T, U, C, and G;
Label表示可检测标记,例如为荧光基团;Label indicates a detectable label, such as a fluorophore;
Blocker表示阻断基团。Blocker indicates a blocking group.
本发明的化合物中,A为芳香基团,在与其相连的磷酸二酯键断裂后,可形成带羟基的芳香环,有利于切断产物的稳定存在;R1、R2和R3为吸电子基,有助于A与R1之间的磷酸二酯键被切除试剂切断。 In the compound of the present invention, A is an aromatic group, and after the phosphodiester bond attached thereto is broken, an aromatic ring having a hydroxyl group can be formed, which is advantageous for the stable existence of the cut product; R 1 , R 2 and R 3 are electron withdrawing The base helps the phosphodiester bond between A and R 1 to be cleaved by the excision reagent.
在某些实施方案中,R1选自
Figure PCTCN2017105734-appb-000012
在某些实施方案中,R1
Figure PCTCN2017105734-appb-000013
In certain embodiments, R 1 is selected from
Figure PCTCN2017105734-appb-000012
In certain embodiments, R 1 is
Figure PCTCN2017105734-appb-000013
在某些实施方案中,R2选自硝基、三氟甲基、氟、氯、氢和醛基。在某些实施方案中,R2为硝基。In certain embodiments, R 2 is selected from the group consisting of nitro, trifluoromethyl, fluoro, chloro, hydrogen, and aldehyde. In certain embodiments, R 2 is a nitro group.
在某些实施方案中,R3选自
Figure PCTCN2017105734-appb-000014
在某些实施方案中,R3
Figure PCTCN2017105734-appb-000015
In certain embodiments, R 3 is selected from
Figure PCTCN2017105734-appb-000014
In certain embodiments, R 3 is
Figure PCTCN2017105734-appb-000015
在某些实施方案中,
Figure PCTCN2017105734-appb-000016
选自:
In certain embodiments,
Figure PCTCN2017105734-appb-000016
From:
Figure PCTCN2017105734-appb-000017
Figure PCTCN2017105734-appb-000017
在某些实施方案中,
Figure PCTCN2017105734-appb-000018
Figure PCTCN2017105734-appb-000019
In certain embodiments,
Figure PCTCN2017105734-appb-000018
for
Figure PCTCN2017105734-appb-000019
本发明的化合物可以是核苷或核苷酸,因此R4可以是-H、单磷酸基团(-PO3H2)、二磷酸基团(-PO3H-PO3H2)、三磷酸基团(-PO3H-PO3H-PO3H2)或四磷酸基团(-PO3H-PO3H-PO3H-PO3H2)。The compound of the present invention may be a nucleoside or a nucleotide, and thus R 4 may be -H, a monophosphate group (-PO 3 H 2 ), a diphosphate group (-PO 3 H-PO 3 H 2 ), three Phosphoric acid group (-PO 3 H-PO 3 H-PO 3 H 2 ) or tetraphosphoric acid group (-PO 3 H-PO 3 H-PO 3 H-PO 3 H 2 ).
在某些实施方案中,R4为单磷酸基团(-PO3H2)、二磷酸基团(-PO3H-PO3H2)、三磷酸基团(-PO3H-PO3H-PO3H2)或四磷酸基团(-PO3H-PO3H-PO3H-PO3H2)核苷酸。在此情况下,所述化合物为核苷酸。In certain embodiments, R 4 is a monophosphate group (-PO 3 H 2 ), a diphosphate group (-PO 3 H-PO 3 H 2 ), a triphosphate group (-PO 3 H-PO 3 H-PO 3 H 2 ) or tetraphosphate group (-PO 3 H-PO 3 H-PO 3 H-PO 3 H 2 ) nucleotide. In this case, the compound is a nucleotide.
在某些实施方案中,R4为三磷酸基团(-PO3H-PO3H-PO3H2)。在此情况下,所述化合物为核苷三磷酸。In certain embodiments, R 4 is a triphosphate group (-PO 3 H-PO 3 H-PO 3 H 2 ). In this case, the compound is a nucleoside triphosphate.
在某些实施方案中,R4为-H。在此情况下,所述化合物为核苷。In certain embodiments, R 4 is -H. In this case, the compound is a nucleoside.
本发明的化合物中,Blocker可以有多种结构。在某些实施方案中,Blocker的结构为:Among the compounds of the present invention, Blocker can have a variety of structures. In certain embodiments, the structure of the Blocker is:
Figure PCTCN2017105734-appb-000020
其中,Ra1和Ra2各自独立地选自H、F、-CF3、-CHF2、-CH2F、-CH2W,-COOW、-CONHW;W选自C1-C6烷基。
Figure PCTCN2017105734-appb-000020
Wherein, Ra 1 and Ra 2 are each independently selected from the group consisting of H, F, -CF 3 , -CHF 2 , -CH 2 F, -CH 2 W, -COOW, -CONHW; W is selected from C 1 -C 6 alkyl groups. .
在某些实施方案中,Blocker的结构为: In certain embodiments, the structure of the Blocker is:
Figure PCTCN2017105734-appb-000021
其中,Rb1、Rb2、Rb3、Rb4、Rb5各自独立地选自H和C1-C6烷基。
Figure PCTCN2017105734-appb-000021
Wherein Rb 1 , Rb 2 , Rb 3 , Rb 4 and Rb 5 are each independently selected from H and C 1 -C 6 alkyl.
在某些实施方案中,Blocker的结构为:In certain embodiments, the structure of the Blocker is:
Figure PCTCN2017105734-appb-000022
其中,Rc1、Rc2各自独立地选自H、F、Cl和-CF3
Figure PCTCN2017105734-appb-000022
Wherein Rc 1 and Rc 2 are each independently selected from the group consisting of H, F, Cl and -CF 3 .
在某些实施方案中,Blocker的结构为:In certain embodiments, the structure of the Blocker is:
Figure PCTCN2017105734-appb-000023
其中,R5选自C1-4烷基(例如C1-2烷基、C1-3烷基、C2-4烷基,例如甲基、乙基、正丙基、异丙基、正丁基、仲丁基、异丁基、叔丁基)。因此,在某些实施方案中,本发明的化合物具有通式(I’)所示的结构:
Figure PCTCN2017105734-appb-000023
Wherein R 5 is selected from C 1-4 alkyl (eg, C 1-2 alkyl, C 1-3 alkyl, C 2-4 alkyl, such as methyl, ethyl, n-propyl, isopropyl, n-Butyl, sec-butyl, isobutyl, tert-butyl). Thus, in certain embodiments, the compounds of the invention have the structure of formula (I'):
Figure PCTCN2017105734-appb-000024
Figure PCTCN2017105734-appb-000024
其中,R5选自C1-4烷基(例如C1-2烷基、C1-3烷基、C2-4烷基,例如甲基、乙基、正丙基、异丙基、正丁基、仲丁基、异丁基、叔丁基)。Wherein R 5 is selected from C 1-4 alkyl (eg, C 1-2 alkyl, C 1-3 alkyl, C 2-4 alkyl, such as methyl, ethyl, n-propyl, isopropyl, n-Butyl, sec-butyl, isobutyl, tert-butyl).
在某些实施方案中,R5为甲基或乙基。In certain embodiments, R 5 is methyl or ethyl.
本发明的化合物中,可检测标记(Label)通过磷酸二酯键与碱基(Base)相连;磷酸二酯键可以被切除试剂切断,从而使可检测标记从化合物中去除。在某些实施方案中,所述可检测标记为荧光基团。可用于本发明的荧光基团的实例包括但不限于各种已知的荧光标记物,例如AF532,ALEX-350,FAM,VIC,TET,CAL
Figure PCTCN2017105734-appb-000025
Gold 540,JOE,HEX,CAL Fluor Orange 560,TAMRA,CAL Fluor Red 590,ROX,CAL Fluor Red 610,TEXAS RED,CAL Fluor Red 635,Quasar 670,Cy3,Cy3.5,Cy5,Cy5.5,Quasar 705等。此类荧光基团及其检测方法是本领域公知的,并且可根据实际需要进行选择。在某些实施方案中,本发明化合物中的Label为Cy3、Cy3.5、Cy5或Cy5.5。
In the compounds of the present invention, a detectable label (Label) is linked to a base by a phosphodiester bond; the phosphodiester bond can be cleaved by a scavenging reagent to remove the detectable label from the compound. In certain embodiments, the detectable label is a fluorophore. Examples of fluorophores that can be used in the present invention include, but are not limited to, various known fluorescent labels such as AF532, ALEX-350, FAM, VIC, TET, CAL.
Figure PCTCN2017105734-appb-000025
Gold 540, JOE, HEX, CAL Fluor Orange 560, TAMRA, CAL Fluor Red 590, ROX, CAL Fluor Red 610, TEXAS RED, CAL Fluor Red 635, Quasar 670, Cy3, Cy3.5, Cy5, Cy5.5, Quasar 705 and so on. Such fluorophores and methods for their detection are well known in the art and can be selected according to actual needs. In certain embodiments, the Label in the compounds of the invention is Cy3, Cy3.5, Cy5 or Cy5.5.
本发明的化合物可以是脱氧核糖核苷酸、核糖核苷酸、脱氧核糖核苷或核糖核苷。因此,R6可以为氢或羟基。在某些实施方案中,R6为氢。在此情况下,所述化合物为脱氧核糖核苷酸或脱氧核糖核苷。 The compounds of the invention may be deoxyribonucleotides, ribonucleotides, deoxyribonucleosides or ribonucleosides. Therefore, R 6 may be hydrogen or a hydroxyl group. In certain embodiments, R 6 is hydrogen. In this case, the compound is a deoxyribonucleotide or a deoxyribonucleoside.
本发明的化合物中,
Figure PCTCN2017105734-appb-000026
L起到连接基团的作用,其中,L可以存在或不存在。在某些实施方案中,m为1。在某些实施方案中,n为1。在某些实施方案中,L为
Figure PCTCN2017105734-appb-000027
Among the compounds of the present invention,
Figure PCTCN2017105734-appb-000026
L functions as a linking group in which L may or may not be present. In certain embodiments, m is one. In certain embodiments, n is one. In certain embodiments, L is
Figure PCTCN2017105734-appb-000027
在某些实施方案中,本发明的化合物具有通式(II)所示的结构In certain embodiments, the compounds of the invention have the structure of formula (II)
Figure PCTCN2017105734-appb-000028
Figure PCTCN2017105734-appb-000028
在某些实施方案中,通式(II)中的R2为硝基。In certain embodiments, R 2 in formula (II) is a nitro group.
在某些实施方案中,通式(II)中的Label为Cy3。In certain embodiments, the Label in Formula (II) is Cy3.
在某些实施方案中,本发明的化合物具有以下结构:In certain embodiments, the compounds of the invention have the structure:
Figure PCTCN2017105734-appb-000029
Figure PCTCN2017105734-appb-000029
Figure PCTCN2017105734-appb-000030
Figure PCTCN2017105734-appb-000030
制备修饰的核苷或核苷酸的方法Method for preparing modified nucleosides or nucleotides
本申请还提供了制备如上文所述的修饰的核苷或核苷酸的方法。示例性的制备过程包括步骤1-步骤3:The present application also provides methods of making modified nucleosides or nucleotides as described above. An exemplary preparation process includes steps 1 - 3:
步骤1: step 1:
Figure PCTCN2017105734-appb-000031
Figure PCTCN2017105734-appb-000031
步骤1进一步包括以下步骤: Step 1 further includes the following steps:
步骤1-1:以化合物I为起始原料,进行氧化反应,生成化合物II(中间体)。Step 1-1: Using Compound I as a starting material, an oxidation reaction is carried out to produce a compound II (intermediate).
在某些实施方案中,步骤1包括:在乙腈中加入甲醇和四唑,之后加入溶解有化合物I的溶液,在室温下搅拌,使甲醇上的甲氧基取代化合物I上的二异丙基胺基;之后加入碘,在室温下搅拌,得到化合物II。In certain embodiments, step 1 comprises: adding methanol and tetrazole to acetonitrile, followed by the addition of a solution in which Compound I is dissolved, and stirring at room temperature to replace the diisopropyl group on compound I with a methoxy group on methanol. Amine; then iodine is added and stirred at room temperature to give compound II.
在某些实施方案中,所述甲醇和/或四唑相对于化合物I是过量的。在某些实施方案中,所述甲醇与化合物I的摩尔比为2-5:1,例如,2:1、3:1、4:1或5:1,例如3:1。在某些实施方案中,所述四唑与化合物I的摩尔比为2-5:1,例如,2:1、3:1、4:1或5:1,例如3:1。In certain embodiments, the methanol and/or tetrazole is in excess relative to Compound I. In certain embodiments, the molar ratio of methanol to Compound I is from 2 to 5:1, for example, 2:1, 3:1, 4:1, or 5:1, such as 3:1. In certain embodiments, the molar ratio of the tetrazole to Compound I is from 2 to 5:1, for example, 2:1, 3:1, 4:1, or 5:1, such as 3:1.
在某些实施方案中,所述碘以溶液的形式被加入到反应体系中,例如,包含水和/或有机溶剂的溶液(例如,包含水、吡啶和/或四氢呋喃的溶液)。In certain embodiments, the iodine is added to the reaction system as a solution, for example, a solution comprising water and/or an organic solvent (eg, a solution comprising water, pyridine, and/or tetrahydrofuran).
在某些实施方案中,所述碘相对于化合物I是过量的。在某些实施方案中,化合物I与碘的摩尔比为1:1.5-5,例如1:1.5、1:2、1:2.5、1:3、1:3.5、1:4、1:4.5或1:5,例如1:1.5。In certain embodiments, the iodine is in excess relative to Compound I. In certain embodiments, the molar ratio of Compound I to iodine is 1: 1.5-5, such as 1:1.5, 1:2, 1:2.5, 1:3, 1:3.5, 1:4, 1:4.5 or 1:5, for example 1:1.5.
在某些实施方案中,步骤1-1还包括:在化合物I的氧化反应结束后,加入亚硫酸钠,以除去未反应的碘。In certain embodiments, step 1-1 further comprises: after the oxidation reaction of Compound I is completed, sodium sulfite is added to remove unreacted iodine.
在某些实施方案中,步骤1-1还包括对化合物II进行纯化。In certain embodiments, step 1-1 further comprises purifying Compound II.
步骤1-2:对化合物II进行脱保护反应,生成化合物III。 Step 1-2: Deprotection of Compound II to give Compound III.
在某些实施方案中,步骤1-2包括:使化合物II与三氯乙酸混合,并进行搅拌。In certain embodiments, step 1-2 comprises mixing compound II with trichloroacetic acid and stirring.
在某些实施方案中,步骤1-2包括:在包含三氯乙酸的溶液(例如三氯乙酸的二氯甲烷溶液)中加入化合物II,在室温下搅拌,得到化合物III。In certain embodiments, step 1-2 comprises: adding compound II to a solution comprising trichloroacetic acid (eg, a solution of trichloroacetic acid in dichloromethane), and stirring at room temperature to provide compound III.
在某些实施方案中,所述三氯乙酸相对于化合物II是过量的。In certain embodiments, the trichloroacetic acid is in excess relative to Compound II.
在某些实施方案中,步骤1-2还包括对化合物III进行纯化。In certain embodiments, steps 1-2 further comprise purifying compound III.
步骤1-3:对化合物III进行三磷酸反应和脱保护反应,生成化合物IV。Step 1-3: The compound III is subjected to a triphosphate reaction and a deprotection reaction to produce a compound IV.
在某些实施方案中,步骤1-3包括:在氩气保护的条件下,向包含化合物III的溶液中,加入2-氯-4H-1,3,2-苯并二氧磷-4-酮,室温搅拌;再加入三正丁基焦磷酸铵和正丁基胺,室温搅拌;之后加入碘,室温搅拌。In certain embodiments, steps 1-3 comprise: adding 2-chloro-4H-1,3,2-benzophosphono-4- to a solution comprising Compound III under argon-protected conditions. The ketone was stirred at room temperature; then tri-n-butylammonium pyrophosphate and n-butylamine were added and stirred at room temperature; then iodine was added and stirred at room temperature.
在某些实施方案中,所述包含化合物III的溶液还含有1,4-二氧六环和无水吡啶。In certain embodiments, the solution comprising Compound III further comprises 1,4-dioxane and anhydrous pyridine.
在某些实施方案中,所述2-氯-4H-1,3,2-苯并二氧磷-4-酮以溶液的形式(例如1,4-二氧六环溶液)被加入到包含化合物III的溶液中。In certain embodiments, the 2-chloro-4H-1,3,2-benzodioxan-4-one is added to the solution in the form of a solution (eg, a 1,4-dioxane solution). In solution of compound III.
在某些实施方案中,化合物III与2-氯-4H-1,3,2-苯并二氧磷-4-酮的摩尔比为1:1-2,例如1:1.0、1:1.1、1:1.2、1:1.3、1:1.4、1:1.5、1:1.6、1:1.7、1:1.8、1:1.9或1:2.0,例如1:1.1。In certain embodiments, the molar ratio of compound III to 2-chloro-4H-1,3,2-benzodioxan-4-one is 1:1-2, such as 1:1.0, 1:1.1. 1:1.2, 1:1.3, 1:1.4, 1:1.5, 1:1.6, 1:1.7, 1:1.8, 1:1.9 or 1:2.0, for example 1:1.1.
在某些实施方案中,所述三正丁基焦磷酸铵和/或正丁基胺以溶液(例如N,N-二甲基甲酰胺溶液)的形式被加入到反应体系中。In certain embodiments, the tri-n-butylammonium pyrophosphate and/or n-butylamine is added to the reaction system in the form of a solution (eg, N,N-dimethylformamide solution).
在某些实施方案中,所述三正丁基焦磷酸铵相对于化合物III是过量的。在某些实施方案中,化合物III与三正丁基焦磷酸铵的摩尔比为1:1.5-5,例如1:1.5、1:2、1:2.5、1:3、1:3.5、1:4、1:4.5或1:5,例如1:1.5。In certain embodiments, the tri-n-butylammonium pyrophosphate is in excess relative to Compound III. In certain embodiments, the molar ratio of Compound III to tri-n-butylammonium pyrophosphate is 1:1.5-5, such as 1:1.5, 1:2, 1:2.5, 1:3, 1:3.5, 1: 4. 1:4.5 or 1:5, for example 1:1.5.
在某些实施方案中,所述碘以溶液的形式被加入到反应体系中,例如,包含水和/或有机溶剂的溶液(例如,包含水、吡啶和/或四氢呋喃的溶液)中。In certain embodiments, the iodine is added to the reaction system as a solution, for example, a solution comprising water and/or an organic solvent (eg, a solution comprising water, pyridine, and/or tetrahydrofuran).
在某些实施方案中,所述碘相对于化合物III是过量的。在某些实施方案中,化合物I与碘的摩尔比为1:1.5-5,例如1:1.5、1:2、1:2.5、1:3、1:3.5、1:4、1:4.5或1:5,例如1:1.5。In certain embodiments, the iodine is in excess relative to Compound III. In certain embodiments, the molar ratio of Compound I to iodine is 1: 1.5-5, such as 1:1.5, 1:2, 1:2.5, 1:3, 1:3.5, 1:4, 1:4.5 or 1:5, for example 1:1.5.
在某些实施方案中,步骤1-3还包括:在化合物III的三磷酸反应和脱保护反应结束后,加入亚硫酸钠,以除去未反应的碘。In certain embodiments, steps 1-3 further comprise: after the triphosphate reaction of the compound III and the deprotection reaction are completed, sodium sulfite is added to remove unreacted iodine.
在某些实施方案中,步骤1-3还包括对化合物IV进行纯化。In certain embodiments, steps 1-3 further comprise purifying compound IV.
步骤2 Step 2
Figure PCTCN2017105734-appb-000032
Figure PCTCN2017105734-appb-000032
步骤2进一步包括以下步骤:Step 2 further includes the following steps:
步骤2-1:使2-羟基乙酸和N-乙二胺三氟乙酰胺发生反应,生成化合物V。Step 2-1: 2-Hydroxyacetic acid and N-ethylenediamine trifluoroacetamide are reacted to form Compound V.
在某些实施方案中,步骤2-1包括:向2-羟基乙酸中加入四氟硼酸O-(N-琥珀酰亚胺基)-N,N,N′,N′-四甲基脲和N,N-二异丙基乙基胺,室温搅拌,之后加入N-乙二胺三氟乙酰胺,室温搅拌,得到化合物V。In certain embodiments, step 2-1 comprises: adding O-(N-succinimidyl)-N,N,N',N'-tetramethylurea tetrafluoroborate to 2-hydroxyacetic acid and N,N-diisopropylethylamine was stirred at room temperature, then N-ethylenediamine trifluoroacetamide was added and stirred at room temperature to give Compound V.
在某些实施方案中,2-羟基乙酸和N-乙二胺三氟乙酰胺的摩尔比为1:1-1:2,例如1:1.0、1:1.1、1:1.2、1:1.3、1:1.4、1:1.5、1:1.6、1:1.7、1:1.8、1:1.9或1:2.0,例如1:1.2。In certain embodiments, the molar ratio of 2-hydroxyacetic acid to N-ethylenediamine trifluoroacetamide is from 1:1 to 1:2, such as 1:1.0, 1:1.1, 1:1.2, 1:1.3, 1:1.4, 1:1.5, 1:1.6, 1:1.7, 1:1.8, 1:1.9 or 1:2.0, for example 1:1.2.
在某些实施方案中,2-羟基乙酸和四氟硼酸O-(N-琥珀酰亚胺基)-N,N,N′,N′-四甲基脲的摩尔比为1:1-1:2,例如1:1.0、1:1.1、1:1.2、1:1.3、1:1.4、1:1.5、1:1.6、1:1.7、1:1.8、1:1.9或1:2.0,例如1:1.2。In certain embodiments, the molar ratio of 2-hydroxyacetic acid to tetrafluoroboric acid O-(N-succinimidyl)-N,N,N',N'-tetramethylurea is 1:1-1. : 2, for example 1:1.0, 1:1.1, 1:1.2, 1:1.3, 1:1.4, 1:1.5, 1:1.6, 1:1.7, 1:1.8, 1:1.9 or 1:2.0, for example 1 :1.2.
在某些实施方案中,2-羟基乙酸和N,N-二异丙基乙基胺的摩尔比为1:1-1:2,例如1:1.0、1:1.1、1:1.2、1:1.3、1:1.4、1:1.5、1:1.6、1:1.7、1:1.8、1:1.9或1:2.0,例如1:1.5。In certain embodiments, the molar ratio of 2-hydroxyacetic acid to N,N-diisopropylethylamine is from 1:1 to 1:2, such as 1:1.0, 1:1.1, 1:1.2, 1: 1.3, 1:1.4, 1:1.5, 1:1.6, 1:1.7, 1:1.8, 1:1.9 or 1:2.0, for example 1:1.5.
在某些实施方案中,步骤2-1还包括对化合物V进行纯化。In certain embodiments, step 2-1 further comprises purifying compound V.
步骤2-2:使化合物V在N,N-二异丙基乙基胺和2-氰乙基N,N-二异丙基氯代亚磷酰胺的存在下,反应生成化合物VI。 Step 2-2: Compound V is reacted in the presence of N,N-diisopropylethylamine and 2-cyanoethyl N,N-diisopropylchlorophosphoramidite to form compound VI.
在某些实施方案中,步骤2-2包括:在0℃下,向包含化合物V和N,N-二异丙基乙基胺的溶液中,加入2-氰乙基N,N-二异丙基氯代亚磷酰胺,在0℃下搅拌一段时间,缓慢升温到室温,继续搅拌,得到化合物VI。In certain embodiments, step 2-2 comprises adding 2-cyanoethyl N,N-diiso to a solution comprising compound V and N,N-diisopropylethylamine at 0 °C. The propyl chlorophosphoramidite was stirred at 0 ° C for a while, slowly warmed to room temperature and stirring was continued to give compound VI.
在某些实施方案中,包含化合物V和N,N-二异丙基乙基胺的溶液还含有二氯甲烷。In certain embodiments, the solution comprising Compound V and N,N-diisopropylethylamine further contains dichloromethane.
在某些实施方案中,2-氰乙基N,N-二异丙基氯代亚磷酰胺以溶液(例如二氯甲烷溶液)的形式被加入。In certain embodiments, 2-cyanoethyl N,N-diisopropylchlorophosphoramidite is added as a solution (eg, a solution of dichloromethane).
在某些实施方案中,化合物V和N,N-二异丙基乙基胺的摩尔比为1:1-1:2,例如1:1.0、1:1.1、1:1.2、1:1.3、1:1.4、1:1.5、1:1.6、1:1.7、1:1.8、1:1.9或1:2.0,例如1:1.5。In certain embodiments, the molar ratio of compound V to N,N-diisopropylethylamine is from 1:1 to 1:2, such as 1:1.0, 1:1.1, 1:1.2, 1:1.3, 1:1.4, 1:1.5, 1:1.6, 1:1.7, 1:1.8, 1:1.9 or 1:2.0, for example 1:1.5.
在某些实施方案中,化合物V和2-氰乙基N,N-二异丙基氯代亚磷酰胺的摩尔比为1:1-1:2,例如1:1.0、1:1.1、1:1.2、1:1.3、1:1.4、1:1.5、1:1.6、1:1.7、1:1.8、1:1.9或1:2.0,例如1:1。In certain embodiments, the molar ratio of compound V to 2-cyanoethyl N,N-diisopropylchlorophosphoramidite is from 1:1 to 1:2, such as 1:1.0, 1:1.1, 1 : 1.2, 1:1.3, 1:1.4, 1:1.5, 1:1.6, 1:1.7, 1:1.8, 1:1.9 or 1:2.0, for example 1:1.
在某些实施方案中,步骤2-2还包括对化合物VI进行纯化。In certain embodiments, step 2-2 further comprises purifying compound VI.
步骤2-3:使化合物VI进行氧化反应,生成化合物VI’。Step 2-3: Compound VI is subjected to an oxidation reaction to give compound VI'.
在某些实施方案中,步骤2-3包括:将化合物VI与四唑和2-硝基-5-羟基-苯甲酸叔丁酯混合,在室温下搅拌一段时间,加入碘,室温下继续搅拌,生成化合物VI’。In certain embodiments, step 2-3 comprises: mixing compound VI with tetrazole and 2-nitro-5-hydroxy-benzoic acid tert-butyl ester, stirring at room temperature for a period of time, adding iodine, and continuing to stir at room temperature , the compound VI' is produced.
在某些实施方案中,四唑相对于化合物VI是过量的。在某些实施方案中,化合物VI与四唑的摩尔比为1:5,例如1:1、1:2、1:3、1:4或1:5,例如1:2。In certain embodiments, the tetrazole is in excess relative to Compound VI. In certain embodiments, the molar ratio of compound VI to tetrazole is 1:5, such as 1:1, 1:2, 1:3, 1:4, or 1:5, such as 1:2.
在某些实施方案中,2-硝基-5-羟基-苯甲酸叔丁酯相对于化合物VI是过量的。在某些实施方案中,化合物VI与2-硝基-5-羟基-苯甲酸叔丁酯的摩尔比为1:5,例如1:1、1:2、1:3、1:4或1:5,例如1:2。In certain embodiments, 2-nitro-5-hydroxy-benzoic acid tert-butyl ester is in excess relative to Compound VI. In certain embodiments, the molar ratio of compound VI to 2-nitro-5-hydroxy-benzoic acid tert-butyl ester is 1:5, such as 1:1, 1:2, 1:3, 1:4 or 1 :5, for example 1:2.
在某些实施方案中,化合物VI在反应前被溶解于乙腈中。在某些实施方案中,四唑和2-硝基-5-羟基-苯甲酸叔丁酯以溶液(例如乙腈溶液)的形式与化合物VI混合。In certain embodiments, Compound VI is dissolved in acetonitrile prior to the reaction. In certain embodiments, the tetrazole and 2-nitro-5-hydroxy-benzoic acid tert-butyl ester are combined with Compound VI in the form of a solution (eg, an acetonitrile solution).
在某些实施方案中,所述碘以溶液的形式被加入到反应体系中,例如,包含水和/或有机溶剂的溶液(例如,包含水、吡啶和/或四氢呋喃的溶液)中。In certain embodiments, the iodine is added to the reaction system as a solution, for example, a solution comprising water and/or an organic solvent (eg, a solution comprising water, pyridine, and/or tetrahydrofuran).
在某些实施方案中,碘相对于化合物VI是过量的。在某些实施方案中,化合物VI与碘的摩尔比为1:1.5-5,例如1:1.5、1:2、1:2.5、1:3、1:3.5、1:4、1:4.5或1:5,例如1:1.5。In certain embodiments, the iodine is in excess relative to the compound VI. In certain embodiments, the molar ratio of compound VI to iodine is 1: 1.5-5, such as 1:1.5, 1:2, 1:2.5, 1:3, 1:3.5, 1:4, 1:4.5 or 1:5, for example 1:1.5.
在某些实施方案中,步骤2-3还包括:在化合物VI的氧化反应结束后,加入亚硫酸钠,以除去未反应的碘。 In certain embodiments, steps 2-3 further comprise: after the oxidation reaction of Compound VI is complete, sodium sulfite is added to remove unreacted iodine.
步骤2-4:对化合物VI’进行脱保护反应。Step 2-4: Deprotection of compound VI'.
在某些实施方案中,步骤2-4包括:向化合物VI’中加入氨水,以除去三氟乙酰基和氰乙基;除去氨水,加入氢氧化钾,以除去叔丁基,得到化合物VII。In certain embodiments, steps 2-4 include: adding aqueous ammonia to compound VI' to remove trifluoroacetyl and cyanoethyl groups; removing aqueous ammonia, and adding potassium hydroxide to remove tert-butyl groups to provide compound VII.
在某些实施方案中,步骤2-4包括:向化合物VI’中加入过量氨水,室温搅拌;除去氨水,加入过量氢氧化钾,室温搅拌。In certain embodiments, steps 2-4 include: adding excess aqueous ammonia to compound VI', stirring at room temperature; removing aqueous ammonia, adding excess potassium hydroxide, and stirring at room temperature.
在某些实施方案中,氢氧化钾以溶液(例如水溶液)的形式被加入。In certain embodiments, potassium hydroxide is added as a solution (eg, an aqueous solution).
在某些实施方案中,步骤2-4还包括:对化合物VII进行纯化。In certain embodiments, steps 2-4 further comprise: purifying compound VII.
步骤3:Step 3:
Figure PCTCN2017105734-appb-000033
Figure PCTCN2017105734-appb-000033
Figure PCTCN2017105734-appb-000034
Figure PCTCN2017105734-appb-000034
Figure PCTCN2017105734-appb-000035
Figure PCTCN2017105734-appb-000035
步骤3-1:使化合物VII与Cy3的N-羟基琥珀酰亚胺酯在弱碱性条件下反应,生成化合物VIII。Step 3-1: Compound VII is reacted with an N-hydroxysuccinimide ester of Cy3 under weakly basic conditions to give compound VIII.
在某些实施方案中,步骤3-1包括:向Cy3的N-羟基琥珀酰亚胺酯中加入N,N-二异丙基乙基胺和化合物VII,室温搅拌,得到化合物VIII。In certain embodiments, step 3-1 comprises: adding N,N-diisopropylethylamine and compound VII to the N-hydroxysuccinimide ester of Cy3, and stirring at room temperature to provide compound VIII.
在某些实施方案中,步骤3-1包括:向Cy3的N-羟基琥珀酰亚胺酯的DMF溶液中,加入包含N,N-二异丙基乙基胺和化合物VII的DMF溶液,室温搅拌,得到化合物VIII。In certain embodiments, step 3-1 comprises: adding a DMF solution comprising N,N-diisopropylethylamine and Compound VII to a solution of N-hydroxysuccinimide ester of Cy3 in DMF, room temperature Stirring gives compound VIII.
在某些实施方案中,步骤3-1还包括:对化合物VIII进行纯化。In certain embodiments, step 3-1 further comprises: purifying compound VIII.
步骤3-2:使化合物IV和化合物VIII反应,生成本发明的修饰的核苷或核苷酸。Step 3-2: Reaction of Compound IV with Compound VIII to form a modified nucleoside or nucleotide of the invention.
在某些实施方案中,步骤3-2包括:使化合物VIII、四氟硼酸O-(N-琥珀酰亚胺基)-N,N,N′,N′-四甲基脲和N,N-二异丙基乙基胺混合,之后加入化合物IV,室温搅拌,得到本发明的化合物。In certain embodiments, step 3-2 comprises: compound VIII, O-(N-succinimidyl)-N,N,N',N'-tetramethylurea and N,N tetrafluoroborate - Diisopropylethylamine is mixed, and then compound IV is added and stirred at room temperature to obtain a compound of the present invention.
在某些实施方案中,步骤3-2的反应在DMF中进行。In certain embodiments, the reaction of Step 3-2 is carried out in DMF.
在某些实施方案中,化合物IV以溶液(例如包含碳酸氢钠的溶液)的形式被加入到反应体系中。In certain embodiments, Compound IV is added to the reaction system as a solution (eg, a solution comprising sodium bicarbonate).
在某些实施方案中,步骤3-2还包括:对本发明的修饰的核苷或核苷酸进行纯化。In certain embodiments, step 3-2 further comprises: purifying the modified nucleoside or nucleotide of the invention.
(二)测序方法(two) sequencing method
基于本发明的修饰的核苷或核苷酸,本申请的发明人还开发了一种多核苷酸的测序方法。本发明的测序方法中,一边合成目标单链多核苷酸互补的生长的多核苷酸,一边进行测序。Based on the modified nucleosides or nucleotides of the present invention, the inventors of the present application have also developed a sequencing method for polynucleotides. In the sequencing method of the present invention, sequencing is performed while synthesizing a polynucleotide in which a target single-stranded polynucleotide is complementary to each other.
因此,在一个方面,本申请提供了一种制备在测序反应中与目标单链多核苷酸互补的生长的多核苷酸的方法,其包括将如上文所定义的化合物并入所述生长的互补多核苷酸,其中,所述化合物的并入防止了任何后续的核苷酸引入所述生长的互补多核苷酸中。Thus, in one aspect, the application provides a method of preparing a growing polynucleotide complementary to a target single-stranded polynucleotide in a sequencing reaction, comprising incorporating a compound as defined above into the complementary of said growth A polynucleotide, wherein the incorporation of the compound prevents any subsequent nucleotides from being introduced into the growing complementary polynucleotide.
在某些实施方案中,所述化合物的并入通过末端转移酶、末端聚合酶或逆转录酶来实现。In certain embodiments, the incorporation of the compound is achieved by a terminal transferase, a terminal polymerase, or a reverse transcriptase.
在某些实施方案中,所述方法包括:使用聚合酶,使所述化合物并入生长的互补多核苷酸。In certain embodiments, the method comprises: incorporating the compound into a growing complementary polynucleotide using a polymerase.
在某些实施方案中,所述方法包括:在允许聚合酶进行核苷酸聚合反应的条件下,使用聚合酶进行核苷酸聚合反应,从而将所述化合物并入生长的互补多核苷酸的3'端。In certain embodiments, the method comprises: polymerizing a polymerase using a polymerase under conditions that allow the polymerase to undergo nucleotide polymerization, thereby incorporating the compound into the growing complementary polynucleotide 3' end.
在另一个方面,本申请提供了一种测定目标单链多核苷酸的序列的方法,其包括:监测互补核苷酸的顺序并入,其中并入的至少一个互补核苷酸是如上文所定义的化合物,以及,检测所述化合物携带的可检测标记。In another aspect, the application provides a method of determining the sequence of a single-stranded polynucleotide of interest, comprising: monitoring sequential incorporation of complementary nucleotides, wherein at least one complementary nucleotide that is incorporated is as described above A defined compound, and detecting a detectable label carried by the compound.
在某些实施方案中,在引入所述下一个互补核苷酸之前,将所述化合物中的阻断基团和所述可检测标记除去。In certain embodiments, the blocking group and the detectable label in the compound are removed prior to introduction of the next complementary nucleotide.
在某些实施方案中,所述阻断基团和所述可检测标记被同时除去。In certain embodiments, the blocking group and the detectable label are removed simultaneously.
在某些实施方案中,所述阻断基团和所述可检测标记被先后除去。例如,在所述可检测标记被除去之前或之后,所述阻断基团被除去。In certain embodiments, the blocking group and the detectable label are removed sequentially. For example, the blocking group is removed before or after the detectable label is removed.
在某些实施方案中,所述测定目标单链多核苷酸的序列的方法包括:In certain embodiments, the method of determining the sequence of a single-stranded polynucleotide of interest comprises:
(a)提供包含双链体、通式(I)的化合物、聚合酶和切除试剂的混合物;所述双链体包含生长的核酸链以及待测序的核酸分子;(a) providing a mixture comprising a duplex, a compound of formula (I), a polymerase, and a excision reagent; said duplex comprising a growing nucleic acid strand and a nucleic acid molecule to be sequenced;
(b)进行包含以下步骤(i)、(ii)和(iii)的反应循环:(b) Carry out a reaction cycle comprising the following steps (i), (ii) and (iii):
步骤(i):使用聚合酶,使所述化合物并入生长的核酸链,形成包含阻断基团和可检测标记的核酸中间体;Step (i): using a polymerase to incorporate the compound into a growing nucleic acid strand to form a nucleic acid intermediate comprising a blocking group and a detectable label;
步骤(ii):对所述核酸中间体上的可检测标记进行检测;Step (ii): detecting a detectable label on the nucleic acid intermediate;
步骤(iii):使用切除试剂将核酸中间体上的阻断基团除去。 Step (iii): The blocking group on the nucleic acid intermediate is removed using a resection reagent.
在某些实施方案中,所述反应循环还包括步骤(iv):使用切除试剂将核酸中间体上的可检测标记除去。In certain embodiments, the reaction cycle further comprises the step (iv) of removing the detectable label on the nucleic acid intermediate using a scavenging reagent.
在某些实施方案中,所述步骤(iii)和步骤(iv)中使用的切除试剂是同样的试剂。在某些实施方案中,所述步骤(iii)和步骤(iv)中使用的切除试剂是不同的试剂。In certain embodiments, the ablation reagents used in steps (iii) and (iv) are the same reagents. In certain embodiments, the ablation reagents used in steps (iii) and (iv) are different reagents.
在某些实施方案中,例如,并入的至少一个互补核苷酸是如通式(I’)所示的化合物的实施方案中,所述测定目标单链多核苷酸的序列的方法包括:In certain embodiments, for example, wherein the at least one complementary nucleotide incorporated is an embodiment of a compound of formula (I&apos;), the method of determining the sequence of a single-stranded polynucleotide of interest comprises:
(1)提供双链体,其包含生长的核酸链以及待测序的核酸分子,所述双链体连接于支持物上;(1) providing a duplex comprising a growing nucleic acid strand and a nucleic acid molecule to be sequenced, the duplex being linked to a support;
(2)添加用于进行核苷酸聚合反应的聚合酶,以及第一、第二、第三和第四化合物,从而形成含有溶液相和固相的反应体系;其中,所述四种化合物分别为核苷酸A、(T/U)、C和G的衍生物,均具有通式(I’)所示的结构并且具有碱基互补配对能力;(2) adding a polymerase for performing nucleotide polymerization, and first, second, third, and fourth compounds, thereby forming a reaction system containing a solution phase and a solid phase; wherein the four compounds are respectively Derivatives of nucleotides A, (T/U), C and G, each having the structure represented by the general formula (I') and having a base complementary pairing ability;
(3)在允许聚合酶进行核苷酸聚合反应的条件下,使用聚合酶进行核苷酸聚合反应,从而将所述四种化合物中的一种并入生长的核酸链的3'端;(3) performing nucleotide polymerization using a polymerase under conditions allowing the polymerase to undergo nucleotide polymerization, thereby incorporating one of the four compounds into the 3' end of the growing nucleic acid strand;
(4)移除前一步骤的反应体系的溶液相,保留连接于支持物上的双链体,并检测所述双链体或所述生长的核酸链上的可检测标记所发出的信号;(4) removing the solution phase of the reaction system of the previous step, leaving the duplex attached to the support, and detecting the signal emitted by the detectable label on the duplex or the growing nucleic acid strand;
(5)添加切除试剂,使所述双链体或所述生长的核酸链在含有溶液相和固相的反应体系中与切除试剂接触;其中,所述切除试剂能够使并入生长的核酸链3'端的化合物中的磷酸二酯键(1)和/或磷酸二酯键(2)断裂,并且不会影响双链体骨架上的磷酸二酯键;(5) adding a scavenging reagent such that the duplex or the growing nucleic acid strand is contacted with a scavenging reagent in a reaction system containing a solution phase and a solid phase; wherein the excision reagent enables the incorporation of the growing nucleic acid strand The phosphodiester bond (1) and/or the phosphodiester bond (2) in the compound at the 3' end are cleaved and do not affect the phosphodiester bond on the duplex backbone;
(6)移除前一步骤的反应体系的溶液相。(6) The solution phase of the reaction system of the previous step is removed.
在某些优选的实施方案中,所述方法还包括下述步骤:In certain preferred embodiments, the method further comprises the steps of:
(7)重复进行步骤(2)-(6)或步骤(2)-(4)一次或多次。(7) Repeat steps (2)-(6) or steps (2)-(4) one or more times.
任选地,在任意一个包含移除操作的步骤之后,进行洗涤操作。在某些优选的实施方案中,在步骤(4)和步骤(5)之间,进行洗涤操作。在某些优选的实施方案中,在步骤(6)之后,进行洗涤操作。Optionally, the washing operation is performed after any of the steps including the removing operation. In certain preferred embodiments, a washing operation is performed between step (4) and step (5). In certain preferred embodiments, after step (6), a washing operation is performed.
在某些实施方案中,所述双链体通过包含以下步骤的方法得到:In certain embodiments, the duplex is obtained by a method comprising the steps of:
提供引物,使引物退火至待测序的核酸分子上,所述引物作为起始的生长的核酸链,与所述待测序的核酸分子一起形成连接于支持物上的双链体。 A primer is provided to anneal the primer to the nucleic acid molecule to be sequenced, which serves as the initial growing nucleic acid strand, together with the nucleic acid molecule to be sequenced, forms a duplex attached to the support.
核酸分子Nucleic acid molecule
在本发明的方法中,待测序的核酸分子可以是任何目的核酸分子。在某些优选的实施方案中,所述待测序的核酸分子包含脱氧核糖核苷酸、核糖核苷酸、经修饰的脱氧核糖核苷酸、经修饰的核糖核苷酸、或其任何组合。在本发明的方法中,待测序的核酸分子不受其类型的限制。在某些优选的实施方案中,所述待测序的核酸分子为DNA或RNA。在某些优选的实施方案中,所述待测序的核酸分子可以为基因组DNA,线粒体DNA,叶绿体DNA,mRNA,cDNA,miRNA,或siRNA。在某些优选的实施方案中,所述待测序的核酸分子为线性的或者环状的。在某些优选的实施方案中,所述待测序的核酸分子为双链的或者单链的。例如,所述待测序的核酸分子可以为单链DNA(ssDNA),双链DNA(dsDNA),单链RNA(ssRNA),双链RNA(dsRNA),或者DNA和RNA的杂合体。在某些优选的实施方案中,所述待测序的核酸分子为单链DNA。在某些优选的实施方案中,所述待测序的核酸分子为双链DNA。In the method of the invention, the nucleic acid molecule to be sequenced may be any nucleic acid molecule of interest. In certain preferred embodiments, the nucleic acid molecule to be sequenced comprises deoxyribonucleotides, ribonucleotides, modified deoxyribonucleotides, modified ribonucleotides, or any combination thereof. In the methods of the invention, the nucleic acid molecule to be sequenced is not limited by its type. In certain preferred embodiments, the nucleic acid molecule to be sequenced is DNA or RNA. In certain preferred embodiments, the nucleic acid molecule to be sequenced can be genomic DNA, mitochondrial DNA, chloroplast DNA, mRNA, cDNA, miRNA, or siRNA. In certain preferred embodiments, the nucleic acid molecule to be sequenced is linear or circular. In certain preferred embodiments, the nucleic acid molecule to be sequenced is double-stranded or single-stranded. For example, the nucleic acid molecule to be sequenced may be single-stranded DNA (ssDNA), double-stranded DNA (dsDNA), single-stranded RNA (ssRNA), double-stranded RNA (dsRNA), or a hybrid of DNA and RNA. In certain preferred embodiments, the nucleic acid molecule to be sequenced is a single stranded DNA. In certain preferred embodiments, the nucleic acid molecule to be sequenced is a double stranded DNA.
在本发明的方法中,待测序的核酸分子不受其来源的限制。在某些优选的实施方案中,待测序的核酸分子可以获自任何来源,例如,任何细胞、组织或生物体(例如,病毒,细菌,真菌,植物和动物)。在某些优选的实施方案中,待测序的核酸分子源自哺乳动物(例如,人、非人灵长类动物、啮齿类动物或犬科动物)、植物、鸟类、爬行类、鱼类、真菌、细菌或病毒。In the methods of the invention, the nucleic acid molecule to be sequenced is not limited by its source. In certain preferred embodiments, the nucleic acid molecule to be sequenced can be obtained from any source, for example, any cell, tissue or organism (eg, viruses, bacteria, fungi, plants, and animals). In certain preferred embodiments, the nucleic acid molecule to be sequenced is derived from a mammal (eg, a human, a non-human primate, a rodent or a canine), a plant, a bird, a reptile, a fish, Fungus, bacteria or virus.
从细胞、组织或生物体中提取或获得核酸分子的方法是本领域技术人员公知的。合适的方法包括但不限于乙醇沉淀法,氯仿抽提法等。关于此类方法的详细描述可参见例如,J.Sambrook等人,分子克隆:实验室手册,第2版,冷泉港实验室出版社,1989,以及F.M.Ausubel等人,精编分子生物学实验指南,第3版,John Wiley&Sons,Inc.,1995。另外,还可使用各种商业化的试剂盒来从各种来源(例如细胞、组织或生物体)提取核酸分子。Methods for extracting or obtaining nucleic acid molecules from cells, tissues or organisms are well known to those skilled in the art. Suitable methods include, but are not limited to, ethanol precipitation, chloroform extraction, and the like. A detailed description of such methods can be found, for example, in J. Sambrook et al., Molecular Cloning: A Laboratory Manual, 2nd Edition, Cold Spring Harbor Laboratory Press, 1989, and FMAusubel et al., Guide to Molecular Biology Experiments , 3rd edition, John Wiley & Sons, Inc., 1995. In addition, various commercial kits can be used to extract nucleic acid molecules from a variety of sources, such as cells, tissues or organisms.
在本发明的方法中,待测序的核酸分子不受其长度的限制。在某些优选的实施方案中,待测序的核酸分子的长度可以为至少10bp,至少20bp,至少30bp,至少40bp,至少50bp,至少100bp,至少200bp,至少300bp,至少400bp,至少500bp,至少1000bp,或者至少2000bp。在某些优选的实施方案中,待测序的核酸分子的长度可以为10-20bp,20-30bp,30-40bp,40-50bp,50-100bp,100-200bp,200-300bp,300-400bp,400-500bp,500-1000bp,1000-2000bp,或者超过2000bp。在某些优选的实施方案中,待测序的核酸分子可具有10-1000bp的长度,以利于进行高通量测序。 In the method of the invention, the nucleic acid molecule to be sequenced is not limited by its length. In certain preferred embodiments, the nucleic acid molecule to be sequenced can be at least 10 bp, at least 20 bp, at least 30 bp, at least 40 bp, at least 50 bp, at least 100 bp, at least 200 bp, at least 300 bp, at least 400 bp, at least 500 bp, at least 1000 bp in length. , or at least 2000bp. In certain preferred embodiments, the nucleic acid molecule to be sequenced may be 10-20 bp, 20-30 bp, 30-40 bp, 40-50 bp, 50-100 bp, 100-200 bp, 200-300 bp, 300-400 bp, 400-500 bp, 500-1000 bp, 1000-2000 bp, or more than 2000 bp. In certain preferred embodiments, the nucleic acid molecule to be sequenced can have a length of 10-1000 bp to facilitate high throughput sequencing.
在某些优选的实施方案中,在将核酸分子连接于支持物之前,可以对核酸分子进行预处理。此类预处理包括但不限于,核酸分子的片段化,末端的补齐,接头的添加,标签的添加,切口的修复,核酸分子的扩增,核酸分子的分离和纯化,以及其任何组合。In certain preferred embodiments, the nucleic acid molecule can be pretreated prior to attaching the nucleic acid molecule to the support. Such pretreatments include, but are not limited to, fragmentation of nucleic acid molecules, complementation of ends, addition of linkers, addition of tags, repair of nicks, amplification of nucleic acid molecules, isolation and purification of nucleic acid molecules, and any combination thereof.
例如,在某些优选的实施方案中,为了获得具有合适长度的核酸分子,可以对核酸分子进行片段化处理。在本发明的方法中,可以通过本领域普通技术人员已知的任何方法进行核酸分子(例如DNA)的片段化。例如,可通过酶学方法或机械方法进行片段化。所述机械方法可以是超声波或物理剪切。所述酶学方法可以通过用核酸酶(例如,脱氧核糖核酸酶)或限制性核酸内切酶消化来进行。在某些优选的实施方案中,所述片段化导致序列未知的末端。在某些优选的实施方案中,所述片段化导致序列已知的末端。For example, in certain preferred embodiments, nucleic acid molecules can be subjected to fragmentation in order to obtain nucleic acid molecules of suitable length. In the methods of the invention, fragmentation of a nucleic acid molecule (e.g., DNA) can be performed by any method known to those of ordinary skill in the art. For example, fragmentation can be carried out by enzymatic or mechanical means. The mechanical method can be ultrasonic or physical shear. The enzymatic method can be carried out by digestion with a nuclease (for example, deoxyribonuclease) or restriction endonuclease. In certain preferred embodiments, the fragmentation results in an end of the sequence that is not known. In certain preferred embodiments, the fragmentation results in a known end of the sequence.
在某些优选的实施方案中,所述酶学方法使用DNA酶I来对核酸分子进行片段化。DNA酶I是一种非特异性剪切双链DNA(dsDNA)以释放5'磷酸化的二核苷酸、三核苷酸和寡核苷酸产物的通用酶。DNA酶I在含有Mn2+、Mg2+和Ca2+但不含其它盐的缓冲液中具有最佳活性,其通常用于将一个大的DNA基因组片段化成为小的DNA片段,随后所产生的小的DNA片段可用于构建DNA文库。In certain preferred embodiments, the enzymatic method uses DNase I to fragment a nucleic acid molecule. DNase I is a universal enzyme that non-specifically cleaves double-stranded DNA (dsDNA) to release 5' phosphorylated dinucleotide, trinucleotide and oligonucleotide products. DNase I has optimal activity in buffers containing Mn 2+ , Mg 2+ and Ca 2+ but no other salts, which are commonly used to fragment a large DNA genome into small DNA fragments, followed by The resulting small DNA fragments can be used to construct a DNA library.
DNA酶I的剪切特性将导致DNA分子的随机消化(即,没有序列偏向性),并且当在含有锰离子的缓冲液存在的情况下使用时,主要产生钝末端dsDNA片段(Melgar,E.和D.A.Goldthwait.1968.Deoxyribonucleic acid nucleases.II.The effects of metal on the mechanism of action of deoxyribonuclease I.J.Biol.Chem.243:4409)。当使用DNA酶I处理基因组DNA时,可考虑以下三种因素:(i)所用酶的量(单位);(ii)消化温度(℃);和(iii)温育时间(分钟)。通常,可以在10℃-37℃之间,用DNA酶I消化大的DNA片段或全基因组DNA 1-2分钟,以产生具有合适长度的DNA分子。The cleavage properties of DNase I will result in random digestion of DNA molecules (ie, no sequence bias) and, when used in the presence of buffers containing manganese ions, produce predominantly blunt-end dsDNA fragments (Melgar, E. And DAGoldthwait.1968. Deoxyribonucleic acid nucleases. II. The effects of metal on the mechanism of action of deoxyribonuclease IJ Biol. Chem. 243: 4409). When genomic DNA is treated with DNase I, three factors can be considered: (i) amount of enzyme used (unit); (ii) digestion temperature (°C); and (iii) incubation time (minutes). Generally, large DNA fragments or whole genomic DNA can be digested with DNase I for 1-2 minutes between 10 ° C and 37 ° C to produce DNA molecules of suitable length.
因此,在某些优选的实施方案中,在步骤(1’)之前,对目的核酸分子(待测序的核酸分子)进行片段化。在某些优选的实施方案中,通过酶学方法或机械方法对待测序的核酸分子进行片段化处理。在某些优选的实施方案中,使用DNA酶I对待测序的核酸分子进行片段化。在某些优选的实施方案中,通过超声处理来待测序的核酸分子进行片段化。在某些优选的实施方案中,经片段化的核酸分子的长度为50-2000bp,例如50-100bp,100-200bp,200-300bp,300-400bp,400-500bp,500-1000bp,1000-2000bp,50-1500bp,或50-1000bp。 Thus, in certain preferred embodiments, the nucleic acid molecule of interest (the nucleic acid molecule to be sequenced) is fragmented prior to step (1'). In certain preferred embodiments, the nucleic acid molecules to be sequenced are subjected to fragmentation by enzymatic or mechanical means. In certain preferred embodiments, the nucleic acid molecule to be sequenced by DNase I is fragmented. In certain preferred embodiments, the nucleic acid molecules to be sequenced are subjected to fragmentation by sonication. In certain preferred embodiments, the fragmented nucleic acid molecule is 50-2000 bp in length, such as 50-100 bp, 100-200 bp, 200-300 bp, 300-400 bp, 400-500 bp, 500-1000 bp, 1000-2000 bp. , 50-1500 bp, or 50-1000 bp.
双链核酸分子(例如dsDNA,基因组DNA)的片段化可产生具有钝末端的或长度为一个或两个核苷酸的悬突的核酸片段。例如,当用超声法或者DNA酶I处理基因组DNA(gDNA)时,其产物可包含具有钝末端或悬突的DNA片段。在这种情况下,可使用聚合酶将具有悬突的核酸分子的末端补齐,形成具有钝末端的核酸分子,以利于后续的应用(例如便于将经片段化的核酸分子与接头连接)。Fragmentation of double-stranded nucleic acid molecules (eg, dsDNA, genomic DNA) can produce nucleic acid fragments having blunt ends or overhangs of one or two nucleotides in length. For example, when genomic DNA (gDNA) is treated by sonication or DNase I, the product may comprise a DNA fragment having a blunt end or overhang. In this case, the end of the nucleic acid molecule having an overhang can be made up using a polymerase to form a nucleic acid molecule having a blunt end to facilitate subsequent applications (e.g., to facilitate ligation of the fragmented nucleic acid molecule to a linker).
因此,在某些优选的实施方案中,在对待测序的核酸分子(例如dsDNA)进行片段化后,用DNA聚合酶来处理经片段化的核酸分子,以产生具有钝末端的DNA片段。在某些优选的实施方案中,所述DNA聚合酶可以是任何已知的DNA聚合酶,例如T4DNA聚合酶,Pfu DNA聚合酶,Klenow DNA聚合酶。在某些情况下,Pfu DNA聚合酶的使用可能是有利的,这是因为Pfu DNA聚合酶不仅能够补齐悬突部分形成钝末端,而且其具有3'-5'核酸外切酶活性,能够去除单核苷酸和双核苷酸突起,从而进一步增加具有钝末端的DNA片段的数量(Costa,G.L.和M.P.Weiner.1994a.Protocols for cloning and analysis of blunt-ended PCR-generated DNA fragments.PCR Methods Appl 3(5):S95;Costa,G.L.t A.Grafsky和M.P.Wemer.1994b.Cloning and analysis of PCR-generated DNA fragments.PCR Methods Appl 3(6):338;Costa,G.L.和M.P.Weiner.1994c.Polishing with T4or Pfu polymerase increases the efficiency of cloning of PCR products.Nucleic Acids Res.22(12):2423)。Thus, in certain preferred embodiments, after fragmentation of a nucleic acid molecule to be sequenced (e.g., dsDNA), the fragmented nucleic acid molecule is treated with a DNA polymerase to produce a DNA fragment having a blunt end. In certain preferred embodiments, the DNA polymerase can be any known DNA polymerase, such as T4 DNA polymerase, Pfu DNA polymerase, Klenow DNA polymerase. In some cases, the use of Pfu DNA polymerase may be advantageous because Pfu DNA polymerase not only complements the overhangs to form blunt ends, but also has 3'-5' exonuclease activity, Removal of single nucleotide and dinucleotide overhangs to further increase the number of DNA fragments with blunt ends (Costa, GL and MP Weiner. 1994a. Protocols for cloning and analysis of blunt-ended PCR-generated DNA fragments. PCR Methods Appl 3(5): S95; Costa, GLt A. Grafsky and MP Wemer. 1994b. Cloning and analysis of PCR-generated DNA fragments. PCR Methods Appl 3(6): 338; Costa, GL and MPWeiner. 1994c. Polishing with T4or Pfu polymerase increases the efficiency of cloning of PCR products. Nucleic Acids Res. 22(12): 2423).
在某些优选的实施方案中,可以在待测序的核酸分子的5'和/或3'端引入接头。在一般情况下,接头为寡核苷酸序列,并且其可以是任何序列,任意长度。可用本领域熟知的方法选择具有合适长度和序列的接头。例如,连接于待测序的核酸分子末端的接头通常是长度为5-100个核苷酸(例如5-10bp,10-20bp,20-30bp,30-40bp,40-50bp,50-100bp)之间的相对较短的核苷酸序列。在某些优选的实施方案中,所述接头可具有引物结合区。此类引物结合区可与引物发生退火或杂交,从而可用于引发特异性聚合酶反应。在某些优选的实施方案中,所述接头具有一个或多个引物结合区。在某些优选的实施方案中,所述接头具有能够与用于进行扩增的引物杂交的一个或多个区域。在某些优选的实施方案中,所述接头具有能够与用于测序反应的引物杂交的一个或多个区域。在某些优选的实施方案中,在待测序的核酸分子的5'端引入接头。在某些优选的实施方案中,在待测序的核酸分子的3'端引入接头。在某些优选的实施方案中,在待测序的核酸分子的5'端和3'端引入接头。在一些实施方案中,所述接头包含,能 够与通用引物杂交的通用接头序列。在一些实施方案中,所述接头包含,能够与通用扩增引物和/或通用测序引物杂交的通用接头序列。In certain preferred embodiments, a linker can be introduced at the 5' and/or 3' end of the nucleic acid molecule to be sequenced. In general, the linker is an oligonucleotide sequence and it can be any sequence, of any length. Linkers of suitable length and sequence can be selected by methods well known in the art. For example, a linker ligated to the end of a nucleic acid molecule to be sequenced is typically 5 to 100 nucleotides in length (eg, 5-10 bp, 10-20 bp, 20-30 bp, 30-40 bp, 40-50 bp, 50-100 bp). A relatively short nucleotide sequence between. In certain preferred embodiments, the linker can have a primer binding region. Such primer binding regions can anneal or hybridize to primers and can be used to elicit specific polymerase reactions. In certain preferred embodiments, the linker has one or more primer binding regions. In certain preferred embodiments, the linker has one or more regions that are capable of hybridizing to a primer for amplification. In certain preferred embodiments, the linker has one or more regions that are capable of hybridizing to primers used in the sequencing reaction. In certain preferred embodiments, a linker is introduced at the 5' end of the nucleic acid molecule to be sequenced. In certain preferred embodiments, a linker is introduced at the 3' end of the nucleic acid molecule to be sequenced. In certain preferred embodiments, a linker is introduced at the 5' and 3' ends of the nucleic acid molecule to be sequenced. In some embodiments, the linker comprises A universal linker sequence sufficient to hybridize to a universal primer. In some embodiments, the linker comprises a universal linker sequence that is capable of hybridizing to a universal amplification primer and/or a universal sequencing primer.
在某些优选的实施方案中,可以在待测序的核酸分子中引入标签序列,或者可以在上文所述的接头中引入标签序列。标签序列是指具有特定碱基序列的一段寡核苷酸。根据实际需要,标签序列可以具有任意长度,例如2-50bp,例如2、3、4、5、10、15、20、25、30、35、40、45、50bp。在某些优选的实施方案中,使每个待测序的核酸分子均带上含特定序列的标签序列,以利于区分每个待测序的核酸分子的来源。在某些优选的实施方案中,可直接在待测序的核酸分子的5'和/或3'端引入标签序列。在某些优选的实施方案中,可在接头中引入标签序列,然后再将接头连接至待测序的核酸分子的5'和/或3'端。标签序列可以位于接头序列的任何位置,例如接头序列的5'和/或3'端。在某些优选的实施方案中,接头包含引物结合区和标签序列。在某些进一步优选的实施方案中,所述引物结合区包含可被通用引物识别的通用接头序列,并且优选地,标签序列可位于引物结合区的3'端。In certain preferred embodiments, the tag sequence can be introduced into the nucleic acid molecule to be sequenced, or the tag sequence can be introduced into the linker described above. A tag sequence refers to a segment of an oligonucleotide having a particular base sequence. The tag sequence can have any length, such as 2-50 bp, such as 2, 3, 4, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50 bp, depending on actual needs. In certain preferred embodiments, each nucleic acid molecule to be sequenced is subjected to a tag sequence containing a particular sequence to facilitate discrimination of the source of each nucleic acid molecule to be sequenced. In certain preferred embodiments, the tag sequence can be introduced directly at the 5' and/or 3' end of the nucleic acid molecule to be sequenced. In certain preferred embodiments, a tag sequence can be introduced into the linker and then ligated to the 5' and/or 3' end of the nucleic acid molecule to be sequenced. The tag sequence can be located anywhere in the linker sequence, such as the 5' and/or 3' end of the linker sequence. In certain preferred embodiments, the linker comprises a primer binding region and a tag sequence. In certain further preferred embodiments, the primer binding region comprises a universal linker sequence that is recognized by universal primers, and preferably, the tag sequence can be located at the 3' end of the primer binding region.
在某些优选的实施方案中,使用不同的标签序列来标记/区分来自不同来源的核酸分子。在此类实施方案中,优选地,将相同的标签序列引入相同来源的核酸分子,并且针对每一种核酸来源,使用一种独特的标签序列。随后,可将不同来源的核酸分子组合在一起,构成文库,并通过各个核酸分子上所携带的独特标签序列来鉴别/区分文库中的各个核酸分子的来源。In certain preferred embodiments, different tag sequences are used to label/distinguish nucleic acid molecules from different sources. In such embodiments, preferably, the same tag sequence is introduced into a nucleic acid molecule of the same source, and for each nucleic acid source, a unique tag sequence is used. Subsequently, nucleic acid molecules of different origins can be combined to form a library, and the source of each nucleic acid molecule in the library can be identified/differentiated by the unique tag sequence carried on each nucleic acid molecule.
可通过本领域熟知的方法(例如,PCR或连接反应),将待测序的核酸分子与接头或标签序列相连接。例如,如果待测序的核酸分子的一部分序列是已知的,那么,可使用适当的PCR引物(其含有接头序列以及能够特异性识别待测序的核酸分子的序列)通过PCR对待测序的核酸分子进行扩增。所获得的扩增产物即是在5'和/或3'端引入了接头的待测核酸分子。在某些实施方案中,可使用非特异性的连接酶(例如T4DNA连接酶),将核酸分子与接头相连接。在某些实施方案中,可使用限制性内切酶对核酸分子和接头进行处理,从而使它们具有相同的粘性末端,随后可使用连接酶将具有相同的粘性末端的核酸分子和接头连接在一起,从而获得与接头相连接的核酸分子。The nucleic acid molecule to be sequenced can be ligated to a linker or tag sequence by methods well known in the art (eg, PCR or ligation reactions). For example, if a part of the sequence of the nucleic acid molecule to be sequenced is known, the nucleic acid molecule to be sequenced by PCR can be carried out using an appropriate PCR primer containing a linker sequence and a sequence capable of specifically recognizing the nucleic acid molecule to be sequenced. Amplification. The amplified product obtained is the nucleic acid molecule to be tested which introduces a linker at the 5' and/or 3' end. In certain embodiments, a nucleic acid molecule can be linked to a linker using a non-specific ligase (eg, T4 DNA ligase). In certain embodiments, a nucleic acid molecule and a linker can be treated with a restriction enzyme such that they have the same cohesive ends, and then the ligase can be used to link nucleic acid molecules and linkers having the same cohesive ends together. Thereby obtaining a nucleic acid molecule linked to the linker.
在某些实施方案中,在使用连接酶将核酸分子和接头连接在一起后,所获得的产物在接合处可存在切口。在这种情况下,可使用聚合酶来修复这种切口。例如,丧失3'-5'核酸外切酶活性但显示5'-3'核酸外切酶活性的DNA聚合酶可具有识别切口并修 复切口的能力(Hamilton,S.C.,J.W.Farchaus and M.C.Davis.2001.DNA polymerases as engines for biotechnology.BioTechniques 31:370)。可用于这种用途的DNA聚合酶包括例如硫化氢热厌氧杆菌(Thermoanaerobacter thermosulfuricus)的polI、大肠肝菌(E.coli)的DNA polI、和噬菌体phi29。在一个优选的实施方案中,将嗜热脂肪芽孢杆菌(Bacillus stearothermophilus)的polI用于修复dsDNA的切口并且形成无缺口的dsDNA。In certain embodiments, after the nucleic acid molecule and linker are joined together using a ligase, the resulting product may have a nick at the junction. In this case, a polymerase can be used to repair this incision. For example, a DNA polymerase that loses 3'-5' exonuclease activity but exhibits 5'-3' exonuclease activity can have an incision and repair The ability to complex incisions (Hamilton, S. C., J. W. Farchaus and M. C. Davis. 2001. DNA polymerases as engines for biotechnology. BioTechniques 31: 370). DNA polymerases which can be used for this purpose include, for example, polI of Thermoanaerobacter thermosulfuricus, DNA polI of E. coli, and phage phi29. In a preferred embodiment, polI of Bacillus stearothermophilus is used to repair the nick of dsDNA and form unnotched dsDNA.
在某些优选的实施方案中,还可以对待测序的核酸分子进行扩增,以增加核酸分子的量或者拷贝数。用于扩增核酸分子的方法是本领域技术人员所熟知的,其典型实例为PCR。例如,可使用下述方法来扩增核酸分子:(i)需要温度循环的聚合酶链式反应(PCR)(参见例如,Saiki等人,1995.Science 230:1350-1354),连接酶链式反应(参见例如,Barany,1991.Proc.Natl.Acad.Sci.USA 88:189-193;Barringer等人,1990.Gene 89:117-122),和基于转录的扩增(参见例如,Kwoh等人,1989.Proc.Natl.Acad.Sci.USA 86:1173-1177);(ii)等温扩增系统(参见例如,Guatelli等人,1990.Proc.Natl.Acad.Sci.USA 87:1874-1878);QP复制酶系统(参见例如,Lizardi等人,1988.BioTechnology 6:1197-1202);和链置换扩增(Nucleic Acids Res.1992Apr 11;20(7):1691-6)。在某些优选的实施方案中,通过PCR来扩增待测序的核酸分子,并且,用于进行PCR扩增的引物包含接头序列和/或标签序列。由此所产生的PCR产物将带有接头序列和/或标签序列,从而可方便地用于后续应用(例如高通量测序)。In certain preferred embodiments, the nucleic acid molecules to be sequenced can also be amplified to increase the amount or copy number of the nucleic acid molecule. Methods for amplifying nucleic acid molecules are well known to those skilled in the art, a typical example of which is PCR. For example, nucleic acid molecules can be amplified using the following methods: (i) polymerase chain reaction (PCR) requiring temperature cycling (see, eg, Saiki et al, 1995. Science 230: 1350-1354), ligase chain Reaction (see, for example, Barany, 1991. Proc. Natl. Acad. Sci. USA 88: 189-193; Barringer et al, 1990. Gene 89: 117-122), and transcription-based amplification (see, for example, Kwoh et al. Human, 1989. Proc. Natl. Acad. Sci. USA 86: 1173-1177); (ii) Isothermal amplification system (see, for example, Guatelli et al., 1990. Proc. Natl. Acad. Sci. USA 87:1874- 1878); QP replicase system (see, eg, Lizardi et al, 1988. BioTechnology 6: 1197-1202); and strand displacement amplification (Nucleic Acids Res. 1992 Apr 11; 20(7): 1691-6). In certain preferred embodiments, the nucleic acid molecule to be sequenced is amplified by PCR, and the primers used for PCR amplification comprise a linker sequence and/or a tag sequence. The PCR product thus produced will carry a linker sequence and/or a tag sequence, which can be conveniently used for subsequent applications (e.g., high throughput sequencing).
在某些优选的实施方案中,在进行各种预处理步骤之前或之后,还可以对待测序的核酸分子进行分离和纯化。此类分离和纯化步骤可能是有利的。例如,在某些优选的实施方案中,所述分离和纯化步骤可用于获得具有适宜长度(例如50-1000bp)的待测序的核酸分子,以利于后续应用(例如高通量测序)。在某些优选的实施方案中,可利用琼脂糖凝胶电泳来分离和纯化待测序的核酸分子。在某些优选的实施方案中,可以通过大小排阻层析法或蔗糖沉降来分离和纯化待测序的核酸分子。In certain preferred embodiments, the nucleic acid molecules to be sequenced are also isolated and purified before or after various pretreatment steps. Such separation and purification steps may be advantageous. For example, in certain preferred embodiments, the isolation and purification steps can be used to obtain nucleic acid molecules of a suitable length (eg, 50-1000 bp) to be sequenced for subsequent applications (eg, high throughput sequencing). In certain preferred embodiments, agarose gel electrophoresis can be utilized to separate and purify the nucleic acid molecules to be sequenced. In certain preferred embodiments, the nucleic acid molecules to be sequenced can be isolated and purified by size exclusion chromatography or sucrose settling.
应当理解的是,上述的预处理步骤(例如,片段化,末端补齐,添加接头,添加标签,切口修复,扩增,分离和纯化)仅仅是示例性的,而非限制性的。本领域技术人员可以根据实际需要,对待测序的核酸分子进行各种期望的预处理,并且各个预处理步骤不受特定的顺序限制。例如,在某些实施方案中,可以先对核酸分子进行片段化并添加接头,然后再进行扩增。在另一些实施方案中,可以先对核酸分子进行扩增, 然后再进行片段化和添加接头。在某些实施方案中,对核酸分子进行片段化并添加接头,而不进行扩增步骤。It should be understood that the pre-treatment steps described above (eg, fragmentation, end-filling, addition of linkers, tag addition, nick repair, amplification, separation, and purification) are merely exemplary and not limiting. Those skilled in the art can perform various desired pretreatments on the nucleic acid molecules to be sequenced according to actual needs, and each pretreatment step is not limited by a specific order. For example, in certain embodiments, the nucleic acid molecule can be first fragmented and a linker added prior to amplification. In other embodiments, the nucleic acid molecule can be amplified first, Then fragment and add the linker. In certain embodiments, the nucleic acid molecule is fragmented and a linker is added without an amplification step.
在某些示例性实施方案中,在步骤(1’)之前,对目的核酸分子(例如,基因组DNA)进行下述预处理:In certain exemplary embodiments, prior to step (1'), the nucleic acid molecule of interest (e.g., genomic DNA) is subjected to the following pretreatment:
(i)对所述目的核酸分子(例如大的核酸片段,例如基因组DNA)进行片段化,从而产生经片段化的核酸分子;(i) fragmenting the nucleic acid molecule of interest (eg, a large nucleic acid fragment, eg, genomic DNA) to produce a fragmented nucleic acid molecule;
(ii)将经片段化的核酸分子与接头序列(其例如包含,能够与通用扩增引物杂交的引物结合区,能够与通用测序引物杂交的引物结合区,和/或标签序列)连接,并任选地进行分离、纯化和变性,从而产生待测序的核酸分子;(ii) linking the fragmented nucleic acid molecule to a linker sequence comprising, for example, a primer binding region capable of hybridizing to a universal amplification primer, a primer binding region capable of hybridizing to a universal sequencing primer, and/or a tag sequence, and Optionally performing isolation, purification, and denaturation to produce a nucleic acid molecule to be sequenced;
(iii)将待测序的核酸分子与支持物相连接,从而获得连接于支持物上的待测序的核酸分子。(iii) linking the nucleic acid molecule to be sequenced to a support to obtain a nucleic acid molecule to be sequenced attached to the support.
支持物Support
在通常情况下,用于连接待测序的核酸分子的支持物呈固相,以便于操作。因此,在本公开内容中,“支持物”有时也被称为“固体支持物”或“固相支持物”。然而,应当理解的是,本文所提及的“支持物”并不限于固体,其还可以是半固体(例如凝胶)。In the usual case, the support for ligation of the nucleic acid molecule to be sequenced is in a solid phase for ease of handling. Thus, in the present disclosure, "support" is sometimes also referred to as "solid support" or "solid support." However, it should be understood that the "support" referred to herein is not limited to a solid, it may also be a semi-solid (eg, a gel).
在本发明的方法中,用于连接待测序的核酸分子的支持物可以由各种合适的材料制成。此类材料包括例如:无机物、天然聚合物、合成聚合物,以及其任何组合。具体的例子包括但不限于:纤维素、纤维素衍生物(例如硝化纤维素)、丙烯酸树脂、玻璃、硅胶、聚苯乙烯、明胶、聚乙烯吡咯烷酮、乙烯基和丙烯酰胺的共聚物、与二乙烯基苯等交联的聚苯乙缔(参见例如,Merrifield Biochemistry 1964,3,1385-1390)、聚丙烯酰胺、乳胶、葡聚糖、橡胶、硅、塑料、天然海绵、金属塑料、交联的葡聚糖(例如,SephadexTM)、琼脂糖凝胶(SepharoseTM),以及本领域技术人员已知的其他支持物。In the method of the present invention, the support for ligation of the nucleic acid molecule to be sequenced may be made of various suitable materials. Such materials include, for example, inorganics, natural polymers, synthetic polymers, and any combination thereof. Specific examples include, but are not limited to, cellulose, cellulose derivatives (such as nitrocellulose), acrylic resins, glass, silica gel, polystyrene, gelatin, polyvinylpyrrolidone, copolymers of vinyl and acrylamide, and Crosslinked polyphenylene bromide such as vinyl benzene (see, for example, Merrifield Biochemistry 1964, 3, 1385-1390), polyacrylamide, latex, dextran, rubber, silicon, plastic, natural sponge, metal plastic, cross-linking dextran (e.g., Sephadex TM), agarose gel (Sepharose TM), and other supports known to the skilled person.
在某些优选的实施方案中,用于连接待测序的核酸分子的支持物可以是包括惰性基底或基质(例如,载玻片、聚合物珠等)的固体支持物,所述惰性基底或基质已例如通过应用含有活性基团的中间材料而被功能化,所述活性基团允许共价连接诸如多核苷酸的生物分子。此类支持物的实例包括但不限于,负载于诸如玻璃的惰性基底上的聚丙酰胺水凝胶,特别是WO 2005/065814和US 2008/0280773中描述的聚丙烯酰胺水凝胶,其中,所述专利申请的内容通过引用以其全文并入本文。在此类实施方案 中,生物分子(例如多核苷酸)可被直接地共价地连接至中间材料(例如水凝胶),而中间材料其自身可被非共价地连接至基底或基质(例如,玻璃基底)。在某些优选的实施方案中,所述支持物为表面修饰了一层亲和素、氨基、丙烯酰胺硅烷或醛基化学基团的玻片或硅片。In certain preferred embodiments, the support for ligation of the nucleic acid molecule to be sequenced may be a solid support comprising an inert substrate or matrix (eg, slides, polymer beads, etc.), said inert substrate or matrix Functionalization has been functionalized, for example, by the use of intermediate materials containing reactive groups that allow covalent attachment of biomolecules such as polynucleotides. Examples of such supports include, but are not limited to, polyacrylamide hydrogels supported on an inert substrate such as glass, in particular polyacrylamide hydrogels as described in WO 2005/065814 and US 2008/0280773, wherein The content of the patent application is hereby incorporated by reference in its entirety. In such an implementation A biomolecule (eg, a polynucleotide) can be directly covalently attached to an intermediate material (eg, a hydrogel), while the intermediate material itself can be non-covalently attached to a substrate or matrix (eg, a glass substrate) . In certain preferred embodiments, the support is a slide or wafer having a surface modified with a layer of avidin, amino, acrylamide silane or aldehyde based chemical groups.
在本发明中,支持物或固体支持物不受限于其大小、形状和构造。在一些实施方案中,支持物或固体支持物是平面结构,例如载片、芯片、微芯片和/或阵列。此类支持物的表面可以是平面层的形式。In the present invention, the support or solid support is not limited by its size, shape and configuration. In some embodiments, the support or solid support is a planar structure, such as a slide, chip, microchip, and/or array. The surface of such a support may be in the form of a planar layer.
在一些实施方案中,支持物或其表面是非平面的,例如管或容器的内表面或外表面。在一些实施方案中,支持物或固体支持物包括微球或珠。本文中“微球”或“珠”或“颗粒”或语法上的等同物是指小的离散颗粒。合适的珠成分包括但不限于塑料、陶瓷、玻璃、聚苯乙烯、甲基苯乙烯、丙烯酸聚合物、顺磁材料、二氧化钍溶胶、碳石墨、二氧化钛、乳胶、交联葡聚糖例如Sepharose、纤维素、尼龙、交联胶束和teflon,以及本文概述的用于制备固体支持物的任何其他材料。此外,珠可以是球形的,也可以是非球形的。在一些实施方案中,可以使用球形的珠。在一些实施方案中,可以使用不规则的颗粒。此外,珠还可以是多孔的。In some embodiments, the support or surface thereof is non-planar, such as the inner or outer surface of a tube or container. In some embodiments, the support or solid support comprises microspheres or beads. As used herein, "microsphere" or "bead" or "particle" or grammatical equivalent refers to a small discrete particle. Suitable bead ingredients include, but are not limited to, plastics, ceramics, glass, polystyrene, methyl styrene, acrylic polymers, paramagnetic materials, cerium oxide sol, carbon graphite, titanium dioxide, latex, cross-linked dextran such as Sepharose , cellulose, nylon, crosslinked micelles and teflon, as well as any other materials outlined herein for the preparation of solid supports. In addition, the beads may be spherical or non-spherical. In some embodiments, spherical beads can be used. In some embodiments, irregular particles can be used. In addition, the beads can also be porous.
在某些优选的实施方案中,用于连接待测序的核酸分子的支持物为珠或孔的阵列(其也被称为芯片)。所述阵列可以使用本文概述的用于制备固体支持物的任何材料来制备,并且优选地,阵列上的珠或孔的表面进行了官能化,以利于核酸分子的连接。阵列上的珠或孔的数目不受限制。例如,每一个阵列可包含10-102、102-103、103-104、104-105、105-106、106-107、107-108、108-109或更多个珠或孔。在某些示例性实施方案中,每个珠或孔的表面可连接一个或多个核酸分子。相应地,每一个阵列可连接10-102、102-103、103-104、104-105、105-106、106-107、107-108、108-109或更多个核酸分子。因此,此类阵列可特别有利地用于核酸分子的高通量测序。In certain preferred embodiments, the support for ligation of the nucleic acid molecule to be sequenced is an array of beads or wells (also referred to as a chip). The array can be prepared using any of the materials outlined herein for preparing a solid support, and preferably, the surface of the beads or pores on the array is functionalized to facilitate ligation of the nucleic acid molecules. The number of beads or holes on the array is not limited. For example, each array may comprise 10-10 2 , 10 2 -10 3 , 10 3 -10 4 , 10 4 -10 5 , 10 5 -10 6 , 10 6 -10 7 , 10 7 -10 8 , 10 8 -109 or more beads or holes. In certain exemplary embodiments, the surface of each bead or well can be joined to one or more nucleic acid molecules. Correspondingly, each array can be connected 10-10 2 , 10 2 -10 3 , 10 3 -10 4 , 10 4 -10 5 , 10 5 -10 6 , 10 6 -10 7 , 10 7 -10 8 , 10 8 - 10 9 or more nucleic acid molecules. Thus, such arrays can be used particularly advantageously for high throughput sequencing of nucleic acid molecules.
如本领域普遍所知的,可利用多种技术来制造支持物。此类技术包括但不限于照相平板印刷术、冲压技术、塑膜技术和显微蚀刻技术。如将被本领域人员所领会的,所使用的技术将取决于支持物的组成、结构和形状。As is generally known in the art, a variety of techniques can be utilized to make the support. Such techniques include, but are not limited to, photolithography, stamping techniques, plastic film technology, and microetching techniques. As will be appreciated by those skilled in the art, the techniques used will depend on the composition, structure and shape of the support.
待测序的核酸分子与支持物的连接The connection of the nucleic acid molecule to be sequenced to the support
在本发明的方法中,可以通过本领域普通技术人员已知的任何方法将待测序的核酸分子与支持物连接(例如,共价或非共价连接)。例如,可通过共价连接,或通过 不可逆的被动吸附,或通过分子间的亲和力(例如,生物素与亲和素之间的亲和力),将待测序的核酸分子与支持物相连接。然而优选的是,待测序的核酸分子与支持物之间的连接足够强,从而核酸分子不会因各种反应(例如聚合反应)所使用的条件以及水或缓冲溶液的洗涤而脱离支持物。In the methods of the invention, the nucleic acid molecule to be sequenced can be linked (e.g., covalently or non-covalently linked) to the support by any method known to those of ordinary skill in the art. For example, by covalent connection, or by The irreversible passive adsorption, or the intermolecular affinity (eg, the affinity between biotin and avidin), connects the nucleic acid molecule to be sequenced to the support. Preferably, however, the linkage between the nucleic acid molecule to be sequenced and the support is sufficiently strong that the nucleic acid molecule does not detach from the support due to the conditions used in the various reactions (e.g., polymerization) and the washing of the water or buffer solution.
例如,在某些优选的实施方案中,待测序的核酸分子的5'端携带有能够将该核酸分子共价连接于支持物的装置,例如化学修饰的官能团。此类官能团的实例包括但不限于磷酸基团、羧酸分子、醛分子、硫醇、羟基、二甲氧基三苯甲基(DMT)或氨基。For example, in certain preferred embodiments, the 5' end of the nucleic acid molecule to be sequenced carries a device capable of covalently attaching the nucleic acid molecule to a support, such as a chemically modified functional group. Examples of such functional groups include, but are not limited to, a phosphate group, a carboxylic acid molecule, an aldehyde molecule, a thiol, a hydroxyl group, a dimethoxytrityl group (DMT), or an amino group.
例如,在某些优选的实施方案中,待测序的核酸分子的5'端可用化学官能团(例如磷酸、硫醇或氨基基团)进行修饰,并且支持物(例如多孔玻璃珠)用氨基-烷氧基硅烷(例如氨基丙基三甲氧基硅烷、氨基丙基三乙氧基硅烷、4-氨基丁基三乙氧基硅烷等)进行衍生,从而可通过活性基团之间的化学反应将核酸分子共价连接在支持物上。在某些优选的实施方案中,待测序的核酸分子的5'端可用羧酸或醛基进行修饰,并且支持物(例如乳胶珠)用肼进行衍生,从而可通过活性基团之间的化学反应将核酸分子共价连接在支持物上(Kremsky等人,1987)。For example, in certain preferred embodiments, the 5' end of the nucleic acid molecule to be sequenced may be modified with a chemical functional group (eg, a phosphoric acid, a thiol or an amino group), and the support (eg, a porous glass bead) may be an amino-alkane. Oxysilane (for example, aminopropyltrimethoxysilane, aminopropyltriethoxysilane, 4-aminobutyltriethoxysilane, etc.) is derivatized so that the nucleic acid can be exchanged by a chemical reaction between the reactive groups The molecule is covalently attached to the support. In certain preferred embodiments, the 5' end of the nucleic acid molecule to be sequenced can be modified with a carboxylic acid or an aldehyde group, and the support (eg, latex beads) is derivatized with hydrazine so that the chemistry between the reactive groups can be The reaction covalently attaches the nucleic acid molecule to the support (Kremsky et al., 1987).
另外,还可以采用交联剂将目的核酸分子与支持物相连接。此类交联剂包括例如,琥珀酰酐、苯基二异硫氰酸盐(Guo等人,1994)、马来酸酐(Yang等人,1998)、1-乙基-3-(3-二甲基氨基丙基)-碳二亚胺盐酸盐(EDC)、间-马来酰亚胺基苯甲酸-N-羟基琥珀酰亚胺酯(MBS)、N-琥珀酰亚胺基[4-碘代乙酰基]氨基苯甲酸(SIAB)、4-(N-马来酰亚胺基甲基)环己烷-1-羧酸琥珀酰亚胺(SMCC)、N-γ-马来酰亚胺基丁酰氧基-琥珀酰亚胺酯(GMBS)、4-(p-马来酰亚胺基苯基)丁酸琥珀酰亚胺(SMPB),以及相应的硫代化合物(水溶性的)。Alternatively, a cross-linking agent can be used to link the nucleic acid molecule of interest to the support. Such crosslinking agents include, for example, succinic anhydride, phenyl diisothiocyanate (Guo et al., 1994), maleic anhydride (Yang et al., 1998), 1-ethyl-3-(3-di). Methylaminopropyl)-carbodiimide hydrochloride (EDC), m-maleimidobenzoic acid-N-hydroxysuccinimide ester (MBS), N-succinimidyl group [4] -iodoacetyl]aminobenzoic acid (SIAB), 4-(N-maleimidomethyl)cyclohexane-1-carboxylic acid succinimide (SMCC), N-γ-maleyl Iminobutyryloxy-succinimide ester (GMBS), 4-(p-maleimidophenyl)butyric acid succinimide (SMPB), and corresponding thio compounds (water soluble) of).
此外,还可用双功能交联剂(例如同源双功能交联剂和异源双功能交联剂)对支持物进行衍生,从而提供经修饰的功能化表面。随后,具有5'-磷酸、硫醇或氨基基团的核酸分子即能够与功能化表面相互作用,形成核酸和支持物之间的共价连接。大量的双功能交联剂及其使用方法是本领域公知的(参见例如,Pierce Catalog and Handbook,第155-200页)。In addition, the support can also be derivatized with a bifunctional crosslinker such as a homobifunctional crosslinker and a heterobifunctional crosslinker to provide a modified functionalized surface. Subsequently, a nucleic acid molecule having a 5'-phosphate, thiol or amino group is capable of interacting with a functionalized surface to form a covalent linkage between the nucleic acid and the support. A large number of bifunctional crosslinkers and methods of use thereof are well known in the art (see, for example, Pierce Catalog and Handbook, pages 155-200).
引物Primer
在本发明的方法中,引物可以是任何长度,并且可以包含任何序列或任何碱基,只要它能够特异性地退火到目标核酸分子的一个区域上。换言之,在本发明的方法中, 引物不受限于其长度、结构和组成。例如,在一些示例性实施方案中,所述引物的长度可以为5-50bp,例如5-10、10-15、15-20、20-25、25-30、30-35、35-40、40-45、45-50bp。在一些示例性实施方案中,所述引物能够形成二级结构(例如发夹结构)。在一些示例性实施方案中,所述引物不形成任何二级结构(例如发夹结构)。在一些示例性实施方案中,所述引物可包含天然存在或非天然存在的核苷酸。在一些示例性实施方案中,所述引物包含天然存在的核苷酸或者由天然存在的核苷酸组成。在一些示例性实施方案中,所述引物包含经修饰的核苷酸,例如锁核酸(LNA)。在一些示例性实施方案中,所述引物能够在严紧条件(例如中度严紧条件或高度严紧条件)下与目的核酸分子杂交。在一些示例性实施方案中,所述引物具有与目的核酸分子中的靶序列完全互补的序列。在一些示例性实施方案中,所述引物与目的核酸分子中的靶序列是部分互补的(例如,存在错配)。在一些示例性实施方案中,所述引物包含通用引物序列。在一些示例性实施方案中,所述待测序的核酸分子包含接头,并且所述接头包含能够与通用引物杂交的序列,并且所使用的引物为通用引物。In the method of the present invention, the primer may be of any length and may comprise any sequence or any base as long as it can specifically anneal to a region of the target nucleic acid molecule. In other words, in the method of the present invention, Primers are not limited by their length, structure and composition. For example, in some exemplary embodiments, the primers may be 5-50 bp in length, such as 5-10, 10-15, 15-20, 20-25, 25-30, 30-35, 35-40, 40-45, 45-50 bp. In some exemplary embodiments, the primers are capable of forming a secondary structure (eg, a hairpin structure). In some exemplary embodiments, the primer does not form any secondary structure (eg, a hairpin structure). In some exemplary embodiments, the primers may comprise naturally occurring or non-naturally occurring nucleotides. In some exemplary embodiments, the primer comprises or consists of a naturally occurring nucleotide. In some exemplary embodiments, the primer comprises a modified nucleotide, such as a locked nucleic acid (LNA). In some exemplary embodiments, the primer is capable of hybridizing to a nucleic acid of interest under stringent conditions, such as moderately stringent conditions or highly stringent conditions. In some exemplary embodiments, the primer has a sequence that is fully complementary to a target sequence in a nucleic acid molecule of interest. In some exemplary embodiments, the primer is partially complementary to a target sequence in a nucleic acid molecule of interest (eg, a mismatch is present). In some exemplary embodiments, the primer comprises a universal primer sequence. In some exemplary embodiments, the nucleic acid molecule to be sequenced comprises a linker, and the linker comprises a sequence capable of hybridizing to a universal primer, and the primer used is a universal primer.
聚合酶Polymerase
在本发明的制备多核苷酸的方法或测序方法中,可使用合适的聚合酶来进行核苷酸聚合反应。在一些示例性实施方案中,所述聚合酶能够以DNA为模板合成新的DNA链(例如DNA聚合酶)。在一些示例性实施方案中,所述聚合酶能够以RNA为模板合成新的DNA链(例如反转录酶)。在一些示例性实施方案中,所述聚合酶能够以DNA或RNA为模板合成新的RNA链(例如RNA聚合酶)。因此,在某些优选的实施方案中,所述聚合酶选自DNA聚合酶,RNA聚合酶,和反转录酶。可根据实际需要,选择合适的聚合酶来进行核苷酸聚合反应。在某些优选的实施方案中,所述聚合反应为聚合酶链式反应(PCR)。在某些优选的实施方案中,所述聚合反应为反转录反应。In the method of preparing a polynucleotide of the present invention or the sequencing method, a suitable polymerase can be used for nucleotide polymerization. In some exemplary embodiments, the polymerase is capable of synthesizing a new DNA strand (eg, a DNA polymerase) using DNA as a template. In some exemplary embodiments, the polymerase is capable of synthesizing a new DNA strand (eg, a reverse transcriptase) using RNA as a template. In some exemplary embodiments, the polymerase is capable of synthesizing a new RNA strand (eg, RNA polymerase) using DNA or RNA as a template. Accordingly, in certain preferred embodiments, the polymerase is selected from the group consisting of a DNA polymerase, an RNA polymerase, and a reverse transcriptase. A suitable polymerase can be selected for nucleotide polymerization according to actual needs. In certain preferred embodiments, the polymerization reaction is a polymerase chain reaction (PCR). In certain preferred embodiments, the polymerization reaction is a reverse transcription reaction.
在本发明的方法中,可以使用KOD聚合酶或其突变体进行核苷酸聚合反应。KOD聚合酶或其突变体(例如KOD POL151、KOD POL157、KOD POL171、KOD POL174、KOD POL376、KOD POL391)对本发明的修饰的核苷或核苷酸具有可接受的聚合效率。KOD POL391和KOD POL171对本发明的修饰的核苷酸的具有可接受的聚合效率。在某些实施方案中,KOD POL391或KOD POL171对本发明的修饰的核苷酸的聚合效率在70%以上,例如70%-80%、80%-90%或90%-100%。 In the method of the present invention, nucleotide polymerization can be carried out using KOD polymerase or a mutant thereof. KOD polymerase or a mutant thereof (e.g., KOD POL151, KOD POL157, KOD POL171, KOD POL174, KOD POL376, KOD POL391) has acceptable polymerization efficiency for the modified nucleoside or nucleotide of the present invention. KOD POL391 and KOD POL171 have acceptable polymerization efficiencies for the modified nucleotides of the invention. In certain embodiments, KOD POL391 or KOD POL171 has a polymerization efficiency for the modified nucleotides of the invention of greater than 70%, such as from 70% to 80%, from 80% to 90%, or from 90% to 100%.
此外,如上文所描述的,在本发明的方法中,可重复进行步骤(2)-(6)或步骤(2)-(4)。因此,在本发明的某些优选实施方案中,可进行一轮或多轮的核苷酸聚合反应。换言之,在本发明的某些优选实施方案中,可在一个或多个步骤中进行核苷酸聚合反应。在这种情况下,每一轮核苷酸聚合反应可以使用相同或不同的聚合酶。例如,可在第一轮核苷酸聚合反应中使用第一种DNA聚合酶,并且可在第二轮核苷酸聚合反应中使用第二种DNA聚合酶。然而,在某些示例性实施方案中,在所有的核苷酸聚合反应中,使用相同的聚合酶(例如相同的DNA聚合酶)。Further, as described above, in the method of the present invention, steps (2) - (6) or steps (2) - (4) may be repeated. Thus, in certain preferred embodiments of the invention, one or more rounds of nucleotide polymerization can be carried out. In other words, in certain preferred embodiments of the invention, the nucleotide polymerization can be carried out in one or more steps. In this case, the same or different polymerases can be used for each round of nucleotide polymerization. For example, a first DNA polymerase can be used in the first round of nucleotide polymerization, and a second DNA polymerase can be used in the second round of nucleotide polymerization. However, in certain exemplary embodiments, the same polymerase (eg, the same DNA polymerase) is used in all nucleotide polymerizations.
聚合条件Polymerization condition
在本发明的制备多核苷酸的方法或测序方法中,核苷酸的聚合反应在适宜的条件下进行。适宜的聚合条件包括溶液相的组成以及各成分的浓度、溶液相的pH、聚合温度等。在适宜的条件下进行聚合,有利于获得可接受的、甚至高的聚合效率。In the method of preparing a polynucleotide of the present invention or the sequencing method, the polymerization of nucleotides is carried out under suitable conditions. Suitable polymerization conditions include the composition of the solution phase and the concentration of each component, the pH of the solution phase, the polymerization temperature, and the like. Polymerization under suitable conditions is advantageous in obtaining acceptable, even high, polymerization efficiencies.
在某些实施方案中,发生聚合反应的溶液相中包含一价盐离子(例如钠离子、氯离子)和/或二价盐离子(例如镁离子、硫酸根离子)。在某些实施方案中,所述一价盐离子或二价盐离子在所述溶液相中的浓度为1-200mM,例如1mM、3mM、10mM、20mM、50mM、100mM、150mM或200mM。In certain embodiments, the solution phase in which the polymerization occurs comprises monovalent salt ions (eg, sodium ions, chloride ions) and/or divalent salt ions (eg, magnesium ions, sulfate ions). In certain embodiments, the concentration of the monovalent salt or divalent salt ion in the solution phase is 1-200 mM, such as 1 mM, 3 mM, 10 mM, 20 mM, 50 mM, 100 mM, 150 mM, or 200 mM.
在某些实施方案中,发生聚合反应的溶液相包含缓冲溶液,例如包含三羟甲基氨基甲烷(Tris)的缓冲溶液。在某些实施方案中,Tris在所述溶液相中的浓度为10mM-200mM,例如10mM、20mM、50mM、100mM、150mM或200mM。In certain embodiments, the solution phase in which the polymerization occurs comprises a buffer solution, such as a buffer solution comprising Tris. In certain embodiments, the concentration of Tris in the solution phase is from 10 mM to 200 mM, such as 10 mM, 20 mM, 50 mM, 100 mM, 150 mM, or 200 mM.
在某些实施方案中,发生聚合反应的溶液相包含有机溶剂,例如DMSO或丙三醇(甘油)。在某些实施方案中,所述有机溶剂在溶液相中的质量含量为0.01%-10%,例如0.01%、0.02%、0.05%、1%、2%、5%或10%。In certain embodiments, the solution phase in which the polymerization occurs comprises an organic solvent such as DMSO or glycerol (glycerol). In certain embodiments, the organic solvent has a mass content in the solution phase of from 0.01% to 10%, such as 0.01%, 0.02%, 0.05%, 1%, 2%, 5%, or 10%.
在某些实施方案中,发生聚合反应的溶液相的pH为7.0-9.0,例如7.0、7.1、7.2、7.3、7.4、7.5、7.6、7.7、7.8、7.9、8.0、8.1、8.2、8.3、8.4、8.5、8.6、8.7、8.8、8.9或9.0。In certain embodiments, the pH of the solution phase in which the polymerization occurs is from 7.0 to 9.0, such as 7.0, 7.1, 7.2, 7.3, 7.4, 7.5, 7.6, 7.7, 7.8, 7.9, 8.0, 8.1, 8.2, 8.3, 8.4. , 8.5, 8.6, 8.7, 8.8, 8.9 or 9.0.
在某些实施方案中,发生聚合反应的溶液相包含:一价盐离子(例如钠离子、氯离子)、二价盐离子(例如镁离子、硫酸根离子)、缓冲溶液(例如包含Tris的缓冲溶液)和有机溶剂(例如DMSO或甘油)。在某些实施方案中,所述溶液相的pH为7.8。 In certain embodiments, the solution phase in which the polymerization occurs comprises: a monovalent salt ion (eg, sodium ion, chloride ion), a divalent salt ion (eg, magnesium ion, sulfate ion), a buffer solution (eg, a buffer containing Tris) Solution) and organic solvents (such as DMSO or glycerol). In certain embodiments, the pH of the solution phase is 7.8.
在某些实施方案中,聚合反应在50-65℃(例如50℃、51℃、52℃、53℃、54℃、55℃、56℃、57℃、58℃、59℃、60℃、61℃、62℃、63℃、64℃或65℃)下进行。In certain embodiments, the polymerization is at 50-65 ° C (eg, 50 ° C, 51 ° C, 52 ° C, 53 ° C, 54 ° C, 55 ° C, 56 ° C, 57 ° C, 58 ° C, 59 ° C, 60 ° C, 61 It is carried out at ° C, 62 ° C, 63 ° C, 64 ° C or 65 ° C).
聚合反应进行的时间可以根据实际需要确定,例如可以进行1min-5min、5min-10min、10min-30min或30min-1h。The time during which the polymerization is carried out can be determined according to actual needs, for example, 1 min to 5 min, 5 min to 10 min, 10 min to 30 min or 30 min to 1 h.
碱基衍生物Base derivative
在本发明的方法中,步骤(2)中所使用的四种化合物分别为核苷酸A、(T/U)、C和G的衍生物。在某些示例性实施方案中,所述的四种化合物分别为核糖或脱氧核糖核苷酸A、T、C和G的衍生物。在某些示例性实施方案中,所述的四种化合物分别为核糖或脱氧核糖核苷酸A、U、C和G的衍生物。特别有利地,所述的四种化合物在核苷酸聚合反应过程中,彼此之间不会发生化学反应。In the method of the present invention, the four compounds used in the step (2) are derivatives of nucleotides A, (T/U), C and G, respectively. In certain exemplary embodiments, the four compounds are derivatives of ribose or deoxyribonucleotides A, T, C, and G, respectively. In certain exemplary embodiments, the four compounds are derivatives of ribose or deoxyribonucleotides A, U, C, and G, respectively. It is particularly advantageous that the four compounds do not undergo a chemical reaction with each other during the nucleotide polymerization.
此外,所述的四种化合物具有碱基互补配对能力。例如,当所述化合物为核苷酸A的衍生物时,其将能够与碱基T或U配对。当所述化合物为核苷酸T或U的衍生物时,其将能够与碱基A配对。当所述化合物为核苷酸C的衍生物时,其将能够与碱基G配对。当所述化合物为核苷酸G的衍生物时,其将能够与碱基C配对。由此,在步骤(3)中,聚合酶(例如DNA聚合酶)将根据碱基互补配对原则,将能够与模板核酸中相应位置处的碱基互补配对的化合物并入生长的核酸链的3'端。相应地,在通过可检测标记(例如荧光基团)发出的信号确定并入生长的核酸链3'端的化合物的类型后,可通过碱基互补配对原则,确定模板核酸中相应位置处的碱基的类型。例如,如果并入生长的核酸链3'端的化合物被确定为核苷酸A的衍生物,那么即可确定模板核酸中相应位置处的碱基为T或U。如果并入生长的核酸链3'端的化合物被确定为核苷酸T或U的衍生物,那么即可确定模板核酸中相应位置处的碱基为A。如果并入生长的核酸链3'端的化合物被确定为核苷酸C的衍生物,那么即可确定模板核酸中相应位置处的碱基为G。如果并入生长的核酸链3'端的化合物被确定为核苷酸G的衍生物,那么即可确定模板核酸中相应位置处的碱基为C。In addition, the four compounds described have a base complementary pairing ability. For example, when the compound is a derivative of nucleotide A, it will be capable of pairing with the base T or U. When the compound is a derivative of nucleotide T or U, it will be able to pair with base A. When the compound is a derivative of nucleotide C, it will be able to pair with base G. When the compound is a derivative of nucleotide G, it will be able to pair with base C. Thus, in step (3), the polymerase (eg, DNA polymerase) will incorporate a compound capable of complementary pairing with a base at a corresponding position in the template nucleic acid into the growing nucleic acid strand according to the principle of base complementary pairing. 'end. Correspondingly, after determining the type of compound incorporated into the 3' end of the growing nucleic acid strand by a signal emitted by a detectable label (eg, a fluorophore), the base at the corresponding position in the template nucleic acid can be determined by the principle of base complementary pairing. type. For example, if a compound incorporated at the 3' end of the growing nucleic acid strand is identified as a derivative of nucleotide A, then the base at the corresponding position in the template nucleic acid can be determined to be T or U. If a compound incorporated at the 3' end of the growing nucleic acid strand is identified as a derivative of nucleotide T or U, then it is determined that the base at the corresponding position in the template nucleic acid is A. If a compound incorporated at the 3' end of the growing nucleic acid strand is identified as a derivative of nucleotide C, then it is determined that the base at the corresponding position in the template nucleic acid is G. If a compound incorporated at the 3' end of the growing nucleic acid strand is identified as a derivative of nucleotide G, then it is determined that the base at the corresponding position in the template nucleic acid is C.
在本发明中,所述四种化合物的核糖或脱氧核糖的3'位置处的羟基(-OH)是受保护的。换言之,在所述四种化合物的核糖或脱氧核糖的3'位置处的羟基(-OH)被保护基团保护,从而,它们能够终止聚合酶(例如DNA聚合酶)的聚合作用。例如,当所述四种化合物中的任一种被引入生长的核酸链的3'端时,由于该化合物的核糖或 脱氧核糖的3'位置处不存在游离的羟基(-OH),聚合酶将无法继续进行下一轮的聚合反应,从而聚合反应将被终止。在这种情况下,在每一轮的聚合反应中,将有且只有一个碱基被并入生长的核酸链。In the present invention, the hydroxyl group (-OH) at the 3' position of the ribose or deoxyribose of the four compounds is protected. In other words, the hydroxyl groups (-OH) at the 3' position of the ribose or deoxyribose of the four compounds are protected by a protecting group, whereby they are capable of terminating the polymerization of a polymerase such as a DNA polymerase. For example, when any of the four compounds is introduced into the 3' end of the growing nucleic acid strand, due to the ribose or There is no free hydroxyl group (-OH) at the 3' position of the deoxyribose, and the polymerase will not be able to proceed to the next round of polymerization, so that the polymerization will be terminated. In this case, in each round of polymerization, one and only one base will be incorporated into the growing nucleic acid strand.
此外,在某些实施方案中,所述四种化合物的核糖或脱氧核糖的3'位置处的保护基团能够被去除。在步骤(5)中,将所述保护基团去除,并转变为游离的羟基(-OH)。随后,可使用聚合酶和所述四种化合物对生长的核酸链进行下一轮的聚合反应,并再次引入一个碱基。Furthermore, in certain embodiments, the protecting group at the 3' position of the ribose or deoxyribose of the four compounds can be removed. In step (5), the protecting group is removed and converted to a free hydroxyl group (-OH). Subsequently, the polymerase and the four compounds can be used to carry out the next round of polymerization of the grown nucleic acid strand and introduce one more base.
因此,在某些实施方案中,步骤(2)中所使用的四种化合物被可逆阻断:当它们被并入生长的核酸链的3'端(例如在步骤(3)中)时,它们将终止聚合酶继续进行聚合作用,终止生长的核酸链的进一步延伸;并且,在它们所包含的阻断基团被去除后,聚合酶将能够继续对生长的核酸链进行聚合作用,继续延伸核酸链。Thus, in certain embodiments, the four compounds used in step (2) are reversibly blocked: when they are incorporated into the 3' end of the growing nucleic acid strand (eg, in step (3)), they The polymerase will be terminated to continue the polymerization, terminating the further extension of the growing nucleic acid strand; and, after the blocking group they contain is removed, the polymerase will continue to polymerize the growing nucleic acid strand and continue to extend the nucleic acid chain.
并入生长的核酸链的化合物的确定Determination of a compound incorporated into a growing nucleic acid strand
在本发明的测序方法中,在每一轮聚合反应之后,对双链体或生长的核酸链中的可检测信号进行检测。通过检测,可以确定并入生长的核酸链的化合物的类型,从而确定待测序的核酸分子中相应位置处的碱基类型。In the sequencing method of the present invention, a detectable signal in a duplex or a growing nucleic acid strand is detected after each round of polymerization. By detection, the type of compound incorporated into the growing nucleic acid strand can be determined to determine the type of base at the corresponding position in the nucleic acid molecule to be sequenced.
在某些优选的实施方案中,所述可检测标记为荧光基团。In certain preferred embodiments, the detectable label is a fluorophore.
在某些优选的实施方案中,本发明的测序方法还包括,在步骤(4)之后,基于碱基互补配对原则,根据步骤(3)中并入生长的核酸链的3'端的化合物的类型,确定待测序的核酸分子中相应位置处的碱基类型。例如,如果并入生长的核酸链3'端的化合物被确定为核苷酸A的衍生物,那么即可确定待测序的核酸分子中相应位置处的碱基为能够与核苷酸A的衍生物配对的碱基(例如T或U)。In certain preferred embodiments, the sequencing method of the present invention further comprises, after step (4), based on the principle of base complementary pairing, according to the type of compound incorporated in the 3' end of the growing nucleic acid strand in step (3) Determining the type of base at the corresponding position in the nucleic acid molecule to be sequenced. For example, if a compound incorporated at the 3' end of the growing nucleic acid strand is identified as a derivative of nucleotide A, then it is determined that the base at the corresponding position in the nucleic acid molecule to be sequenced is a derivative capable of binding to nucleotide A Paired bases (eg T or U).
更具体而言,如果并入生长的核酸链3'端的化合物被确定为核苷酸A的衍生物,那么即可确定待测序的核酸分子中相应位置处的碱基为T或U。如果并入生长的核酸链3'端的化合物被确定为核苷酸T或U的衍生物,那么即可确定待测序的核酸分子中相应位置处的碱基为A。如果并入生长的核酸链3'端的化合物被确定为核苷酸C的衍生物,那么即可确定待测序的核酸分子中相应位置处的碱基为G。如果并入生长的核酸链3'端的化合物被确定为核苷酸G的衍生物,那么即可确定待测序的核酸分子中相应位置处的碱基为C。 More specifically, if a compound incorporated at the 3' end of the growing nucleic acid strand is identified as a derivative of nucleotide A, then it is determined that the base at the corresponding position in the nucleic acid molecule to be sequenced is T or U. If the compound incorporated at the 3' end of the growing nucleic acid strand is determined to be a derivative of nucleotide T or U, then it is determined that the base at the corresponding position in the nucleic acid molecule to be sequenced is A. If the compound incorporated at the 3' end of the growing nucleic acid strand is determined to be a derivative of nucleotide C, then it is determined that the base at the corresponding position in the nucleic acid molecule to be sequenced is G. If the compound incorporated at the 3' end of the growing nucleic acid strand is determined to be a derivative of nucleotide G, then it is determined that the base at the corresponding position in the nucleic acid molecule to be sequenced is C.
双链体或生长的核酸链的处理Processing of duplexes or growing nucleic acid strands
在本发明的测序方法中,每一轮的聚合反应可涉及一次信号检测,并且,除了最后一轮聚合反应之外,每一轮的聚合反应可涉及对双链体或生长的核酸链进行切除处理。最后一轮聚合反应之后,可对双链体或生长的核酸链进行切除处理,也可不进行切除处理。In the sequencing method of the present invention, each round of polymerization may involve one signal detection, and, in addition to the last round of polymerization, each round of polymerization may involve excision of the duplex or growing nucleic acid strand. deal with. After the last round of polymerization, the duplex or the growing nucleic acid strand may be excised or not excised.
在某些实施方案中,步骤(5)中的处理可用于去除并入生长的核酸链3'端的化合物中的阻断基团(从而可开始新一轮的聚合反应),并去除所述双链体或生长的核酸链上可能携带的可检测标记(从而可避免对后续的检测造成干扰)。In certain embodiments, the treatment in step (5) can be used to remove blocking groups in the compound incorporated into the 3' end of the growing nucleic acid strand (so that a new round of polymerization can begin) and remove the double A detectable label that may be carried on the chain or growing nucleic acid strand (so as to avoid interference with subsequent detection).
在某些实施方案中,用于切除阻断基团的切除试剂和用于切除可检测标记的切除试剂是同一种试剂,或者,虽然是不同的试剂,但可以在同样的条件下进行切除,而不会造成两个切除反应之间互相干扰。因此,步骤(5)中,对阻断基团的切除和对可检测标记的切除可以同时进行。In certain embodiments, the excision reagent for excising the blocking group and the excision reagent for excising the detectable label are the same reagent, or, although different reagents, can be excised under the same conditions, It does not cause mutual interference between the two resection reactions. Therefore, in the step (5), the excision of the blocking group and the excision of the detectable label can be simultaneously performed.
在某些实施方案中,用于切除阻断基团的切除试剂和用于切除可检测标记的切除试剂是不同的试剂,并且,需要在不同的条件下进行切除。为了避免切除反应之间互相干扰,对阻断基团的切除和对可检测标记的切除可以分步进行。In certain embodiments, the excision reagent for excising the blocking group and the excision reagent for excising the detectable label are different reagents, and the excision needs to be performed under different conditions. In order to avoid mutual interference between the excision reactions, excision of the blocking group and excision of the detectable label can be performed stepwise.
在某些实施方案中,例如,并入的至少一个互补核苷酸是如通式(I’)所示的化合物的实施方案中,步骤(5)包括:In certain embodiments, for example, wherein the at least one complementary nucleotide incorporated is an embodiment of a compound of formula (I'), step (5) comprises:
步骤(5-1):添加第一切除试剂,使所述双链体或所述生长的核酸链在含有溶液相和固相的反应体系中与第一切除试剂接触,在不影响双链体骨架上的磷酸二酯键的条件下,使并入生长的核酸链3'端的化合物中的磷酸二酯键(1)断裂;Step (5-1): adding a first excision reagent, contacting the duplex or the grown nucleic acid strand with the first excision reagent in a reaction system containing a solution phase and a solid phase without affecting the duplex a phosphodiester bond (1) in a compound incorporated at the 3' end of the growing nucleic acid strand under conditions of a phosphodiester bond on the backbone;
步骤(5-2):添加第二切除试剂,使所述双链体或所述生长的核酸链在含有溶液相和固相的反应体系中与第而切除试剂接触,在不影响双链体骨架上的磷酸二酯键的条件下,使并入生长的核酸链3'端的化合物中的磷酸二酯键(2)断裂。Step (5-2): adding a second excision reagent, so that the duplex or the grown nucleic acid strand is contacted with the first excision reagent in a reaction system containing a solution phase and a solid phase without affecting the duplex The phosphodiester bond (2) in the compound incorporated at the 3' end of the growing nucleic acid strand is cleaved under the condition of a phosphodiester bond on the backbone.
需要指出的是,步骤(5-1)和步骤(5-2)之间没有固定的先后顺序,可以先进行步骤(5-1),也可先进行步骤(5-2)。用“第一”和“第二”只是对切除试剂进行区分,并不代表两种切除试剂的使用顺序。It should be noted that there is no fixed sequence between step (5-1) and step (5-2), and step (5-1) may be performed first, or step (5-2) may be performed first. The use of "first" and "second" merely distinguishes between excising agents and does not represent the order of use of the two excising agents.
切除反应进行的时间可以根据实际需要确定,例如可以进行1min-5min、5min-10min、10min-30min或30min-1h。The time during which the excision reaction is performed can be determined according to actual needs, for example, 1 min to 5 min, 5 min to 10 min, 10 min to 30 min, or 30 min to 1 h.
切除试剂Resection reagent
本发明中,利用切除试剂除去通式(I)化合物中的阻断基团和/或可检测标记。当使用通式(I’)所示的化合物时,本发明的切除试剂能够使通式(I’)化合物中的磷酸二酯键(1)和/或磷酸二酯键(2)断裂,并且,不会使核酸链骨架中的磷酸二酯键断裂,以保持核酸链骨架的完整性。In the present invention, a blocking group and/or a detectable label in the compound of the formula (I) is removed using a scavenging agent. When the compound of the formula (I') is used, the excision reagent of the present invention is capable of cleaving the phosphodiester bond (1) and/or the phosphodiester bond (2) in the compound of the formula (I'), and Does not break the phosphodiester bond in the nucleic acid strand backbone to maintain the integrity of the nucleic acid strand backbone.
本发明中,可用于除去阻断基团的切除试剂包括但不限于内切酶IV。内切酶IV可以选择性地使通式(I’)所示的化合物中的磷酸二酯键(1)断裂,而不影响核酸链骨架中的磷酸二酯键。In the present invention, excision reagents that can be used to remove blocking groups include, but are not limited to, endonuclease IV. The endonuclease IV can selectively cleave the phosphodiester bond (1) in the compound represented by the formula (I') without affecting the phosphodiester bond in the nucleic acid strand skeleton.
本发明中,可用于除去可检测标记的切除试剂包括但不限于碱性磷酸酶。碱性磷酸酶可以选择性地使通式(I’)所示的化合物中的磷酸二酯键(2)断裂,而不影响核酸链骨架中的磷酸二酯键。In the present invention, excision reagents that can be used to remove detectable labels include, but are not limited to, alkaline phosphatase. The alkaline phosphatase can selectively cleave the phosphodiester bond (2) in the compound represented by the formula (I') without affecting the phosphodiester bond in the nucleic acid strand skeleton.
切除反应的条件可以视切除试剂而定,例如,所使用的切除试剂是商购的酶,从而,可以依据供应商推荐的使用条件(例如推荐的缓冲溶液、温度等),确定切除反应的条件。The conditions of the excision reaction may depend on the excision reagent. For example, the excision reagent used is a commercially available enzyme, and thus, the conditions of the excision reaction can be determined according to the use conditions recommended by the supplier (for example, a recommended buffer solution, temperature, etc.). .
洗涤步骤Washing step
在本发明的测序方法中,可根据需要,增加洗涤步骤。可在任何期望的阶段,增加所述洗涤步骤,并且任选地,所述洗涤步骤可进行一次或多次。In the sequencing method of the present invention, the washing step can be increased as needed. The washing step can be increased at any desired stage, and optionally, the washing step can be performed one or more times.
例如,在步骤(4)中,在移除反应体系的溶液相之后,可进行一次或多次洗涤,以充分除去残留的溶液相。此类洗涤步骤可能是有利的,其可用于充分去除游离的(即,未并入生长的核酸链的)携带可检测标记的化合物,尽可能减少非特异性的信号。For example, in the step (4), after removing the solution phase of the reaction system, one or more washings may be performed to sufficiently remove the residual solution phase. Such a washing step may be advantageous in that it can be used to substantially remove free (ie, unincorporated growth nucleic acid strands) compounds that carry detectable labels, minimizing non-specific signals.
类似地,在步骤(6)中,在移除反应体系的溶液相之后,可进行一次或多次洗涤,以充分除去残留的溶液相。此类洗涤步骤可能是有利的,其可用于充分去除步骤(5)中应用的切除试剂,从而尽可能减少对后续反应的不利影响。Similarly, in the step (6), after removing the solution phase of the reaction system, one or more washings may be performed to sufficiently remove the residual solution phase. Such a washing step may be advantageous, which can be used to sufficiently remove the ablation reagent applied in step (5), thereby minimizing the adverse effects on subsequent reactions.
可使用各种合适的洗涤溶液来进行洗涤步骤。此类洗涤溶液的实例包括但不限于,磷酸盐缓冲液,柠檬酸盐缓冲液,Tris-HCl缓冲液,醋酸盐缓冲液,碳酸盐缓冲液等等。可根据实际需要来选择合适的洗涤溶液(包括合适的成分,浓度,离子强度,pH值等),这在本领域技术人员的能力范围之内。The washing step can be carried out using a variety of suitable washing solutions. Examples of such wash solutions include, but are not limited to, phosphate buffer, citrate buffer, Tris-HCl buffer, acetate buffer, carbonate buffer, and the like. It is within the ability of those skilled in the art to select a suitable wash solution (including suitable ingredients, concentrations, ionic strength, pH, etc.) depending on the actual needs.
(三)试剂盒(three) kit
在一个方面,本发明提供了一种试剂盒,其包含第一、第二、第三和第四化合物,所述第一、第二、第三和第四化合物各自为如上定义的通式(I)的化合物,所述四种化合物分别为核苷酸A、(T/U)、C和G的衍生物,且具有碱基互补配对能力。In one aspect, the invention provides a kit comprising first, second, third and fourth compounds, each of said first, second, third and fourth compounds being of the formula A compound of I) which is a derivative of nucleotides A, (T/U), C and G, respectively, and which has a base complementary pairing ability.
在某些实施方案中,所述四种化合物结构中的Label各不相同,例如为不同的荧光基团。In certain embodiments, the Labels in the four compound structures are each different, such as a different fluorophore.
在某些实施方案中,本发明的试剂盒还包含:用于从样品中提取核酸分子的试剂和/或装置;用于预处理核酸分子的试剂;用于连接待测序的核酸分子的支持物;用于将待测序的核酸分子与支持物连接(例如,共价或非共价连接)的试剂;用于起始核苷酸聚合反应的引物;用于进行核苷酸聚合反应的聚合酶;一种或多种缓冲溶液;一种或多种洗涤溶液;或其任何组合。In certain embodiments, kits of the invention further comprise: reagents and/or devices for extracting nucleic acid molecules from a sample; reagents for pretreating nucleic acid molecules; and supports for ligation of nucleic acid molecules to be sequenced a reagent for linking (eg, covalently or non-covalently linking) a nucleic acid molecule to be sequenced to a support; a primer for initial nucleotide polymerization; a polymerase for performing nucleotide polymerization One or more buffer solutions; one or more wash solutions; or any combination thereof.
在某些实施方案中,本发明的试剂盒还包含,用于从样品中提取核酸分子的试剂和/或装置。用于从样品中提取核酸分子的方法是本领域熟知的。因此,可根据需要,在本发明的试剂盒中配置各种用于提取核酸分子的试剂和/或装置,例如用于破碎细胞的试剂,用于沉淀DNA的试剂,用于洗涤DNA的试剂,用于溶解DNA的试剂,用于沉淀RNA的试剂,用于洗涤RNA的试剂,用于溶解RNA的试剂,用于去除蛋白的试剂,用于去除DNA的试剂(例如当目的核酸分子为RNA时),用于去除RNA的试剂(例如当目的核酸分子为DNA时),及其任何组合。In certain embodiments, the kits of the invention further comprise reagents and/or devices for extracting nucleic acid molecules from a sample. Methods for extracting nucleic acid molecules from a sample are well known in the art. Therefore, various reagents and/or devices for extracting nucleic acid molecules, such as reagents for disrupting cells, reagents for precipitating DNA, reagents for washing DNA, may be disposed in the kit of the present invention as needed. a reagent for dissolving DNA, a reagent for precipitating RNA, a reagent for washing RNA, a reagent for dissolving RNA, a reagent for removing protein, and a reagent for removing DNA (for example, when the nucleic acid molecule of interest is RNA) An agent for removing RNA (for example, when the nucleic acid molecule of interest is DNA), and any combination thereof.
在某些实施方案中,本发明的试剂盒还包含,用于预处理核酸分子的试剂。在本发明的试剂盒中,用于预处理核酸分子的试剂不受额外限制,并且可根据实际需要选择。所述用于预处理核酸分子的试剂包括例如,用于核酸分子片段化的试剂(例如DNA酶I),用于补齐核酸分子末端的试剂(例如DNA聚合酶,例如T4DNA聚合酶,Pfu DNA聚合酶,Klenow DNA聚合酶),接头分子,标签分子,用于将接头分子与目的核酸分子相连接的试剂(例如连接酶,例如T4DNA连接酶),用于修复核酸切口的试剂(例如,丧失3'-5'核酸外切酶活性但显示5'-3'核酸外切酶活性的DNA聚合酶),用于扩增核酸分子的试剂(例如,DNA聚合酶,引物,dNTP),用于分离和纯化核酸分子的试剂(例如层析柱),以及其任何组合。In certain embodiments, the kit of the invention further comprises an agent for pretreating the nucleic acid molecule. In the kit of the present invention, the reagent for pretreating the nucleic acid molecule is not limited, and may be selected according to actual needs. The reagent for pretreating a nucleic acid molecule includes, for example, a reagent for fragmentation of a nucleic acid molecule (for example, DNase I), a reagent for complementing the end of a nucleic acid molecule (for example, a DNA polymerase such as T4 DNA polymerase, Pfu DNA) a polymerase, Klenow DNA polymerase, a linker molecule, a tag molecule, an agent for linking a linker molecule to a nucleic acid molecule of interest (eg, a ligase, such as T4 DNA ligase), an agent for repairing a nucleic acid nick (eg, loss 3'-5' exonuclease activity but a DNA polymerase showing 5'-3' exonuclease activity), reagents for amplifying nucleic acid molecules (eg, DNA polymerase, primers, dNTPs), for An agent that separates and purifies the nucleic acid molecule (eg, a chromatography column), and any combination thereof.
在某些实施方案中,本发明的试剂盒还包含用于连接待测序的核酸分子的支持物。所述支持物可以具有上文中针对支持物所详细描述的任何技术特征以及其任何组合。In certain embodiments, the kit of the invention further comprises a support for ligation of the nucleic acid molecule to be sequenced. The support may have any of the technical features detailed above for the support and any combination thereof.
例如,在本发明中,所述支持物可以由各种合适的材料制成。此类材料包括例如:无机物、天然聚合物、合成聚合物,以及其任何组合。具体的例子包括但不限于:纤维素、 纤维素衍生物(例如硝化纤维素)、丙烯酸树脂、玻璃、硅胶、聚苯乙烯、明胶、聚乙烯吡咯烷酮、乙烯基和丙烯酰胺的共聚物、与二乙烯基苯等交联的聚苯乙缔(参见例如,Merrifield Biochemistry 1964,3,1385-1390)、聚丙烯酰胺、乳胶、葡聚糖、橡胶、硅、塑料、天然海绵、金属塑料、交联的葡聚糖(例如,SephadexTM)、琼脂糖凝胶(SepharoseTM),以及本领域技术人员已知的其他支持物。For example, in the present invention, the support may be made of various suitable materials. Such materials include, for example, inorganics, natural polymers, synthetic polymers, and any combination thereof. Specific examples include, but are not limited to, cellulose, cellulose derivatives (such as nitrocellulose), acrylic resins, glass, silica gel, polystyrene, gelatin, polyvinylpyrrolidone, copolymers of vinyl and acrylamide, and Crosslinked polyphenylene bromide such as vinyl benzene (see, for example, Merrifield Biochemistry 1964, 3, 1385-1390), polyacrylamide, latex, dextran, rubber, silicon, plastic, natural sponge, metal plastic, cross-linking dextran (e.g., Sephadex TM), agarose gel (Sepharose TM), and other supports known to the skilled person.
在某些优选的实施方案中,用于连接待测序的核酸分子的支持物可以是包括惰性基底或基质(例如,载玻片、聚合物珠等)的固体支持物,所述惰性基底或基质已例如通过应用含有活性基团的中间材料而被功能化,所述活性基团允许共价连接诸如多核苷酸的生物分子。此类支持物的实例包括但不限于,负载于诸如玻璃的惰性基底上的聚丙酰胺水凝胶,特别是WO 2005/065814和US 2008/0280773中描述的聚丙烯酰胺水凝胶,其中,所述专利申请的内容通过引用以其全文并入本文。在此类实施方案中,生物分子(例如多核苷酸)可被直接地共价地连接至中间材料(例如水凝胶),而中间材料其自身可被非共价地连接至基底或基质(例如,玻璃基底)。在某些优选的实施方案中,所述支持物为表面修饰了一层亲和素、氨基、丙烯酰胺硅烷或醛基化学基团的玻片或硅片。In certain preferred embodiments, the support for ligation of the nucleic acid molecule to be sequenced may be a solid support comprising an inert substrate or matrix (eg, slides, polymer beads, etc.), said inert substrate or matrix Functionalization has been functionalized, for example, by the use of intermediate materials containing reactive groups that allow covalent attachment of biomolecules such as polynucleotides. Examples of such supports include, but are not limited to, polyacrylamide hydrogels supported on an inert substrate such as glass, in particular polyacrylamide hydrogels as described in WO 2005/065814 and US 2008/0280773, wherein The content of the patent application is hereby incorporated by reference in its entirety. In such embodiments, the biomolecule (eg, a polynucleotide) can be directly covalently attached to an intermediate material (eg, a hydrogel), while the intermediate material itself can be non-covalently attached to the substrate or matrix ( For example, a glass substrate). In certain preferred embodiments, the support is a slide or wafer having a surface modified with a layer of avidin, amino, acrylamide silane or aldehyde based chemical groups.
在本发明中,支持物或固体支持物不受限于其大小、形状和构造。在一些实施方案中,支持物或固体支持物是平面结构,例如载片、芯片、微芯片和/或阵列。此类支持物的表面可以是平面层的形式。在一些实施方案中,支持物或其表面是非平面的,例如管或容器的内表面或外表面。在一些实施方案中,支持物或固体支持物包括微球或珠。在某些优选的实施方案中,用于连接待测序的核酸分子的支持物为珠或孔的阵列。In the present invention, the support or solid support is not limited by its size, shape and configuration. In some embodiments, the support or solid support is a planar structure, such as a slide, chip, microchip, and/or array. The surface of such a support may be in the form of a planar layer. In some embodiments, the support or surface thereof is non-planar, such as the inner or outer surface of a tube or container. In some embodiments, the support or solid support comprises microspheres or beads. In certain preferred embodiments, the support for ligation of the nucleic acid molecule to be sequenced is an array of beads or wells.
在某些优选的实施方案中,本发明的试剂盒还包含用于将待测序的核酸分子与支持物连接(例如,共价或非共价连接)的试剂。此类试剂包括例如对核酸分子(例如其5'端)进行活化或修饰的试剂,例如磷酸、硫醇、胺、羧酸或醛;对支持物的表面进行活化或修饰的试剂,例如氨基-烷氧基硅烷(例如氨基丙基三甲氧基硅烷、氨基丙基三乙氧基硅烷、4-氨基丁基三乙氧基硅烷等);交联剂,例如琥珀酰酐、苯基二异硫氰酸盐(Guo等人,1994)、马来酸酐(Yang等人,1998)、1-乙基-3-(3-二甲基氨基丙基)-碳二亚胺盐酸盐(EDC)、间-马来酰亚胺基苯甲酸-N-羟基琥珀酰亚胺酯(MBS)、N-琥珀酰亚胺基[4-碘代乙酰基]氨基苯甲酸(SIAB)、4-(N-马来酰亚胺基甲基)环己烷-1-羧酸琥珀酰亚胺(SMCC)、N-γ-马来酰亚胺基丁酰氧基-琥珀酰亚胺酯(GMBS)、4-(p-马来酰亚胺基苯基)丁酸琥珀酰亚胺(SMPB);以及其任何组合。 In certain preferred embodiments, the kits of the invention further comprise reagents for attaching (eg, covalently or non-covalently linking) a nucleic acid molecule to be sequenced to a support. Such agents include, for example, agents that activate or modify a nucleic acid molecule (eg, at its 5' end), such as phosphoric acid, a thiol, an amine, a carboxylic acid, or an aldehyde; an agent that activates or modifies the surface of the support, such as an amino group - Alkoxysilane (for example, aminopropyltrimethoxysilane, aminopropyltriethoxysilane, 4-aminobutyltriethoxysilane, etc.); crosslinking agent such as succinic anhydride, phenyl diisosulfide Cyanate (Guo et al., 1994), maleic anhydride (Yang et al., 1998), 1-ethyl-3-(3-dimethylaminopropyl)-carbodiimide hydrochloride (EDC) , m-maleimidobenzoic acid-N-hydroxysuccinimide ester (MBS), N-succinimidyl [4-iodoacetyl]aminobenzoic acid (SIAB), 4-(N -maleimidomethyl)cyclohexane-1-carboxylic acid succinimide (SMCC), N-γ-maleimidobutyryloxy-succinimide ester (GMBS), 4-(p-maleimidophenyl)butyric acid succinimide (SMPB); and any combination thereof.
在某些优选的实施方案中,本发明的试剂盒还包含用于起始核苷酸聚合反应的引物。在本发明中,引物不受额外的限制,只要它能够特异性地退火到目标核酸分子的一个区域上。在一些示例性实施方案中,所述引物的长度可以为5-50bp,例如5-10、10-15、15-20、20-25、25-30、30-35、35-40、40-45、45-50bp。在一些示例性实施方案中,所述引物可包含天然存在或非天然存在的核苷酸。在一些示例性实施方案中,所述引物包含天然存在的核苷酸或者由天然存在的核苷酸组成。在一些示例性实施方案中,所述引物包含经修饰的核苷酸,例如锁核酸(LNA)。在某些优选的实施方案中,所述引物包含通用引物序列。In certain preferred embodiments, the kits of the invention further comprise primers for initiating nucleotide polymerization. In the present invention, the primer is not subject to any limitation as long as it can specifically anneal to a region of the target nucleic acid molecule. In some exemplary embodiments, the primers may be 5-50 bp in length, such as 5-10, 10-15, 15-20, 20-25, 25-30, 30-35, 35-40, 40- 45, 45-50bp. In some exemplary embodiments, the primers may comprise naturally occurring or non-naturally occurring nucleotides. In some exemplary embodiments, the primer comprises or consists of a naturally occurring nucleotide. In some exemplary embodiments, the primer comprises a modified nucleotide, such as a locked nucleic acid (LNA). In certain preferred embodiments, the primer comprises a universal primer sequence.
在某些优选的实施方案中,本发明的试剂盒还包含用于进行核苷酸聚合反应的聚合酶。在本发明中,可使用各种合适的聚合酶进行聚合反应。在一些示例性实施方案中,所述聚合酶能够以DNA为模板合成新的DNA链(例如DNA聚合酶)。在一些示例性实施方案中,所述聚合酶能够以RNA为模板合成新的DNA链(例如反转录酶)。在一些示例性实施方案中,所述聚合酶能够以DNA或RNA为模板合成新的RNA链(例如RNA聚合酶)。因此,在某些优选的实施方案中,所述聚合酶选自DNA聚合酶,RNA聚合酶,和反转录酶。In certain preferred embodiments, the kit of the invention further comprises a polymerase for performing a nucleotide polymerization reaction. In the present invention, polymerization can be carried out using various suitable polymerases. In some exemplary embodiments, the polymerase is capable of synthesizing a new DNA strand (eg, a DNA polymerase) using DNA as a template. In some exemplary embodiments, the polymerase is capable of synthesizing a new DNA strand (eg, a reverse transcriptase) using RNA as a template. In some exemplary embodiments, the polymerase is capable of synthesizing a new RNA strand (eg, RNA polymerase) using DNA or RNA as a template. Accordingly, in certain preferred embodiments, the polymerase is selected from the group consisting of a DNA polymerase, an RNA polymerase, and a reverse transcriptase.
在某些优选的实施方案中,本发明的试剂盒包含KOD聚合酶或其突变体。在某些优选的实施方案中,所述突变体选自KOD POL151、KOD POL157、KOD POL171、KOD POL174、KOD POL376、KOD POL391。在某些优选的实施方案中,本发明的试剂盒包含KOD POL391、KOD POL171或其组合。In certain preferred embodiments, the kit of the invention comprises KOD polymerase or a mutant thereof. In certain preferred embodiments, the mutant is selected from the group consisting of KOD POL151, KOD POL157, KOD POL171, KOD POL174, KOD POL376, KOD POL391. In certain preferred embodiments, the kit of the invention comprises KOD POL391, KOD POL171, or a combination thereof.
在某些优选的实施方案中,本发明的试剂盒还包含切除试剂,所述切除试剂能够使式(I’)中的磷酸二酯键(1)和/或磷酸二酯键(2)断裂,并且,不会影响双链体骨架上的磷酸二酯键。In certain preferred embodiments, the kit of the invention further comprises a excision reagent capable of cleaving the phosphodiester bond (1) and/or the phosphodiester bond (2) in formula (I') And, does not affect the phosphodiester bond on the duplex backbone.
在某些优选的实施方案中,所述切除试剂选自内切酶IV和碱性磷酸酶。In certain preferred embodiments, the excision agent is selected from the group consisting of endonuclease IV and alkaline phosphatase.
在某些优选的实施方案中,本发明的试剂盒还包含一种或多种缓冲溶液。此类缓冲液包括但不限于,用于DNA酶I的缓冲溶液,用于DNA聚合酶的缓冲溶液,用于连接酶的缓冲溶液,用于洗脱核酸分子的缓冲溶液,用于溶解核酸分子的缓冲溶液,用于进行核苷酸聚合反应(例如PCR)的缓冲溶液,和用于进行连接反应的缓冲溶液。本发明的试剂盒可包含上述缓冲溶液的任一种或多种。In certain preferred embodiments, the kit of the invention further comprises one or more buffer solutions. Such buffers include, but are not limited to, buffer solutions for DNase I, buffer solutions for DNA polymerases, buffer solutions for ligases, buffer solutions for eluting nucleic acid molecules, and lysing nucleic acid molecules. A buffer solution, a buffer solution for performing a nucleotide polymerization reaction (for example, PCR), and a buffer solution for performing a ligation reaction. The kit of the present invention may comprise any one or more of the above buffer solutions.
在某些实施方案中,所述用于DNA聚合酶的缓冲溶液包含一价盐离子(例如钠离子、氯离子)和/或二价盐离子(例如镁离子、硫酸根离子)。在某些实施方案中,所述一价盐 离子或二价盐离子在所述缓冲溶液中的浓度为1-200mM,例如1mM、3mM、10mM、20mM、50mM、100mM、150mM或200mM。In certain embodiments, the buffer solution for DNA polymerase comprises a monovalent salt ion (eg, sodium ion, chloride ion) and/or a divalent salt ion (eg, magnesium ion, sulfate ion). In certain embodiments, the monovalent salt The concentration of the ion or divalent salt ion in the buffer solution is 1-200 mM, such as 1 mM, 3 mM, 10 mM, 20 mM, 50 mM, 100 mM, 150 mM or 200 mM.
在某些实施方案中,所述用于DNA聚合酶的缓冲溶液包含三羟甲基氨基甲烷(Tris)。在某些实施方案中,Tris在所述缓冲溶液中的浓度为10mM-200mM,例如10mM、20mM、50mM、100mM、150mM或200mM。In certain embodiments, the buffer solution for DNA polymerase comprises Tris. In certain embodiments, the concentration of Tris in the buffer solution is from 10 mM to 200 mM, such as 10 mM, 20 mM, 50 mM, 100 mM, 150 mM, or 200 mM.
在某些实施方案中,所述用于DNA聚合酶的缓冲溶液包含有机溶剂,例如DMSO或丙三醇(甘油)。在某些实施方案中,所述有机溶剂在所述缓冲溶液中的质量含量为0.01%-10%,例如0.01%、0.02%、0.05%、1%、2%、5%或10%。In certain embodiments, the buffer solution for DNA polymerase comprises an organic solvent such as DMSO or glycerol (glycerol). In certain embodiments, the organic solvent is present in the buffer solution in an amount of from 0.01% to 10%, such as 0.01%, 0.02%, 0.05%, 1%, 2%, 5%, or 10% by mass.
在某些实施方案中,所述用于DNA聚合酶的缓冲溶液的pH为7.0-9.0,例如7.0、7.1、7.2、7.3、7.4、7.5、7.6、7.7、7.8、7.9、8.0、8.1、8.2、8.3、8.4、8.5、8.6、8.7、8.8、8.9或9.0。In certain embodiments, the buffer solution for DNA polymerase has a pH of 7.0-9.0, such as 7.0, 7.1, 7.2, 7.3, 7.4, 7.5, 7.6, 7.7, 7.8, 7.9, 8.0, 8.1, 8.2. , 8.3, 8.4, 8.5, 8.6, 8.7, 8.8, 8.9 or 9.0.
在某些实施方案中,所述用于DNA聚合酶的缓冲溶液包含:一价盐离子(例如钠离子、氯离子)、二价盐离子(例如镁离子、硫酸根离子)、Tris和有机溶剂(例如DMSO或甘油)。在某些实施方案中,所述缓冲溶液相的pH为7.8。In certain embodiments, the buffer solution for DNA polymerase comprises: a monovalent salt ion (eg, sodium ion, chloride ion), a divalent salt ion (eg, magnesium ion, sulfate ion), Tris, and an organic solvent (eg DMSO or glycerol). In certain embodiments, the buffer solution phase has a pH of 7.8.
在某些优选的实施方案中,本发明的试剂盒包含一种或多种切除试剂,所述切除试剂能够使式(I’)化合物中的磷酸二酯键(1)和/或磷酸二酯键(2)断裂,并且,不会使核酸链骨架中的磷酸二酯键断裂,以保持核酸链骨架的完整性。In certain preferred embodiments, the kit of the invention comprises one or more excision reagents capable of allowing a phosphodiester bond (1) and/or a phosphodiester in a compound of formula (I') The bond (2) is cleaved and does not cleave the phosphodiester bond in the nucleic acid strand backbone to maintain the integrity of the nucleic acid strand backbone.
在某些实施方案中,所述切除试剂选自内切酶和IV碱性磷酸酶。In certain embodiments, the excision agent is selected from the group consisting of an endonuclease and an IV alkaline phosphatase.
在某些优选的实施方案中,本发明的试剂盒还包含一种或多种洗涤溶液。此类洗涤溶液的实例包括但不限于,磷酸盐缓冲液,柠檬酸盐缓冲液,Tris-HCl缓冲液,醋酸盐缓冲液,碳酸盐缓冲液等。本发明的试剂盒可包含上述洗涤溶液的任一种或多种。In certain preferred embodiments, the kit of the invention further comprises one or more wash solutions. Examples of such wash solutions include, but are not limited to, phosphate buffer, citrate buffer, Tris-HCl buffer, acetate buffer, carbonate buffer, and the like. The kit of the present invention may comprise any one or more of the above washing solutions.
修饰的核苷或核苷酸,以及试剂盒的用途Modified nucleoside or nucleotide, and the use of the kit
本发明的修饰的核苷或核苷酸可以用于测定目标单链多核苷酸的序列。The modified nucleosides or nucleotides of the invention can be used to determine the sequence of a single-stranded polynucleotide of interest.
因此,本发明还提供了如上任一项定义的化合物,以及如上任一项定义的试剂盒用于测定目标单链多核苷酸的序列的用途。Accordingly, the present invention also provides the use of a compound as defined in any one of the above, and a kit as defined in any one of the above, for determining the sequence of a single-stranded polynucleotide of interest.
发明的有益效果Advantageous effects of the invention
与现有技术相比,本发明的技术方案具有以下有益效果: Compared with the prior art, the technical solution of the present invention has the following beneficial effects:
本发明提供的修饰的核苷或核苷酸,在测序中拥有更高的稳定性,其带有的阻断基团和可检测标记可以在温和的条件下被切除,对DNA无损伤,并且可以达到较高的切除效率,甚至可以完全切除。本发明提供的修饰的核苷或核苷酸可以被市售的DNA聚合酶聚合,并且具有可接受的聚合效率,在适宜的条件下,甚至可以达到完全聚合,并且不需要去掉磷酸基团上的酯保护基,可直接进行切除,简化了切除的方法。The modified nucleoside or nucleotide provided by the invention has higher stability in sequencing, and the blocking group and the detectable label thereof can be excised under mild conditions without damage to DNA, and Higher resection efficiency can be achieved and even complete resection can be achieved. The modified nucleosides or nucleotides provided by the present invention can be polymerized by a commercially available DNA polymerase and have an acceptable polymerization efficiency, and under suitable conditions, even complete polymerization can be achieved without removing the phosphate group. The ester protecting group can be directly excised, which simplifies the method of excision.
下面将结合附图和实施例对本发明的实施方案进行详细描述,但是本领域技术人员将理解,下列附图和实施例仅用于说明本发明,而不是对本发明的范围的限定。根据附图和优选实施方案的下列详细描述,本发明的各种目的和有利方面对于本领域技术人员来说将变得显然。The embodiments of the present invention will be described in detail below with reference to the accompanying drawings and embodiments. The various objects and advantageous aspects of the invention will be apparent to those skilled in the <
具体实施方式Detailed ways
现参照下列意在举例说明本发明(而非限定本发明)的实施例来描述本发明。本领域技术人员知晓,实施例以举例方式描述本发明,且不意欲限制本发明所要求保护的范围。The invention is described with reference to the following examples which are intended to illustrate, but not limit the invention. The invention is described by way of example, and is not intended to limit the scope of the invention.
实施例1以碱基T为例,以市售的化合物I作为起始原料,制备带有Cy3荧光基团的、3’-OH被阻断的dTTP。Example 1 Using the base T as an example, a commercially available compound I was used as a starting material to prepare a 3'-OH blocked dTTP having a Cy3 fluorescent group.
(1)将化合物I-1转化为化合物II-1(1) Conversion of Compound I-1 to Compound II-1
Figure PCTCN2017105734-appb-000036
Figure PCTCN2017105734-appb-000036
首先在10mL无水乙腈溶剂中加入3mmol的甲醇和3mmol四唑,之后缓慢加入溶解有0.88g(1mmol)的化合物I-1的5mL无水乙腈溶液,室温搅拌反应1小时。之后,加入碘溶液10mL,碘溶液的溶剂为水/吡啶/四氢呋喃(体积比2/20/78),碘的浓度为0.15M,含1.5mmol碘单质,室温搅拌30分钟。之后,缓慢加入5%的亚硫酸钠溶液,直至溶液中碘单质的颜色消失。加入100mL二氯甲烷和100mL饱和氯化钠溶液,萃取有机相,在水相中加入另外100mL二氯甲烷再次萃取,合并有机相,加入硫酸镁进行干燥,旋转蒸发去除溶剂。可以再用硅胶柱色谱、以体积比1:1的乙酸乙酯/正己烷溶剂对产物分离纯化, 得到化合物II-1,产率:95%(0.78g)。First, 3 mmol of methanol and 3 mmol of tetrazole were added to 10 mL of anhydrous acetonitrile solvent, and then a solution of 0.88 g (1 mmol) of Compound I-1 dissolved in 5 mL of anhydrous acetonitrile was slowly added, and the reaction was stirred at room temperature for 1 hour. Thereafter, 10 mL of an iodine solution was added, and the solvent of the iodine solution was water/pyridine/tetrahydrofuran (volume ratio 2/20/78), the concentration of iodine was 0.15 M, and 1.5 mmol of iodine was contained, and the mixture was stirred at room temperature for 30 minutes. Thereafter, a 5% sodium sulfite solution was slowly added until the color of the iodine element in the solution disappeared. 100 mL of dichloromethane and 100 mL of a saturated sodium chloride solution were added, the organic phase was extracted, and another 100 mL of dichloromethane was added to the aqueous phase for re-extraction. The organic phases were combined, dried over magnesium sulfate and evaporated to remove solvent. The product can be separated and purified by silica gel column chromatography with a 1:1 ratio of ethyl acetate/n-hexane solvent. The compound II-1 was obtained in a yield: 95% (0.78 g).
化合物II-1的核磁和质谱数据:1H NMR(300MHz,CDCl3):δ(ppm)7.84(s,1H),7.3-7.18(m,9H),6.77(m,4H),6.40(dd,1H),5.05(s,1H,),4.21(s,1H),4.09(m,3H),3.75(s,6H),3.47(m,1H),3.32(d,1H),2.67(m,1H),2.60(m,2H),2.36(m,1H),31P NMR(163MHz,CDCl3):δ(ppm)-2.48,-2.66.LC-MS(ESI):m/z 827.2(M+H+).Nuclear magnetic and mass spectral data of compound II-1: 1 H NMR (300 MHz, CDCl 3 ): δ (ppm) 7.84 (s, 1H), 7.3-7.18 (m, 9H), 6.77 (m, 4H), 6.40 (dd , 1H), 5.05 (s, 1H,), 4.21 (s, 1H), 4.09 (m, 3H), 3.75 (s, 6H), 3.47 (m, 1H), 3.32 (d, 1H), 2.67 (m) , 1H), 2.60 (m, 2H), 2.36 (m, 1H), 31 P NMR (163 MHz, CDCl 3 ): δ (ppm) - 2.48, - 2.66. LC-MS (ESI): m/z 827.2 ( M+H + ).
(2)将化合物II-1转化为化合物III-1(2) Conversion of compound II-1 to compound III-1
Figure PCTCN2017105734-appb-000037
Figure PCTCN2017105734-appb-000037
在3%三氯乙酸的二氯甲烷溶液(5mL)中加入0.82g(1mmol)化合物II-1,室温搅拌3小时,旋转蒸发浓缩,使溶液体积为约1mL,在硅胶柱上用4:1的乙酸乙酯/正己烷溶剂分离纯化,得到化合物III-1,产率:92%(0.48g)。0.82 g (1 mmol) of compound II-1 was added to a solution of 3% trichloroacetic acid in dichloromethane (5 mL), stirred at room temperature for 3 hours, concentrated by rotary evaporation to give a solution volume of about 1 mL, and 4:1 on a silica gel column. The ethyl acetate/n-hexane solvent was separated and purified to give Compound III-1, yield: 92% (0.48 g).
化合物III-1的核磁和质谱数据:1H NMR(300MHz,CDCl3):δ(ppm)8.24(s,1H),6.26(dd,1H),5.39–5.34(m,1H),4.23–4.13(m,3H),4.09(m,3H),3.96(dd,1H),3.89(dd,1H),3.80-3.70(s,6H)2.54–2.47(m,1H),2.33–2.42(m,1H′),LC-MS(ESI):m/z 525.4(M+H+).Nuclear magnetic and mass spectral data of compound III-1: 1 H NMR (300 MHz, CDCl 3 ): δ (ppm) 8.24 (s, 1H), 6.26 (dd, 1H), 5.39 - 5.34 (m, 1H), 4.23 - 4.13 ( m, 3H), 4.09 (m, 3H), 3.96 (dd, 1H), 3.89 (dd, 1H), 3.80-3.70 (s, 6H) 2.54 - 2.47 (m, 1H), 2.33 - 2.42 (m, 1H) '), LC-MS (ESI): m/z 525.4 (M+H+).
(3)将化合物III-1转化为化合物IV-1(3) Conversion of compound III-1 to compound IV-1
Figure PCTCN2017105734-appb-000038
Figure PCTCN2017105734-appb-000038
在52.4mg(0.1mmol)化合物III-1中加入吡啶,多次在高真空条件下抽干后,再加入新的无水吡啶,在抽干之后的固体中加入800μL的1,4-二氧六环和300μL无水吡啶,在氩气保护条件下加入溶解有0.11mmol(22mg)2-氯-4H-1,3,2-苯并二氧磷-4-酮的1,4-二氧六环100μL,室温反应,搅拌10分钟。加入溶解有1.5倍当量(1.5mmol,71.6mg) 的三正丁基焦磷酸铵和100μL正丁基胺的N,N-二甲基甲酰胺300μL,室温搅拌,反应15分钟。之后,加入碘溶液1mL,溶剂为水/吡啶/四氢呋喃(2/20/78),碘的浓度为0.15M,含0.15mmol碘单质,室温搅拌反应30分钟。之后缓慢加入5%的亚硫酸钠溶液,直至溶液中碘单质的颜色消失。旋转蒸发去除所有溶剂,剩余固体加入5mL水溶解,之后加入5mL 30%氨水,室温搅拌,反应3个小时,旋转蒸发去除所有溶剂,用微量水溶解剩余固体,用6:4:1的1,4二氧六环/水/氨水作为溶剂,在制备型薄层色谱上分离产物,得到化合物IV-1,产率:30%(18mg)。Pyridine was added to 52.4 mg (0.1 mmol) of compound III-1, and after several times of drying under high vacuum, a new anhydrous pyridine was added, and 800 μL of 1,4-diox was added to the solid after drying. Hexacyclohexane and 300 μL of anhydrous pyridine were added under argon atmosphere to 1,4-dioxol dissolved in 0.11 mmol (22 mg) of 2-chloro-4H-1,3,2-benzodioxan-4-one. 100 μL of six rings, reacted at room temperature, and stirred for 10 minutes. Add 1.5 times equivalent (1.5mmol, 71.6mg) 300 μL of N-N-dimethylformamide of 100 μL of n-butylammonium pyrophosphate and 100 μL of n-butylamine were stirred at room temperature for 15 minutes. Thereafter, 1 mL of an iodine solution was added, and the solvent was water/pyridine/tetrahydrofuran (2/20/78), the concentration of iodine was 0.15 M, and 0.15 mmol of iodine was contained, and the reaction was stirred at room temperature for 30 minutes. Thereafter, a 5% sodium sulfite solution was slowly added until the color of the iodine element in the solution disappeared. Rotate to remove all solvents, add the remaining solids to 5 mL of water to dissolve, then add 5 mL of 30% ammonia water, stir at room temperature, react for 3 hours, remove all solvents by rotary evaporation, and dissolve the remaining solids with a trace of water, with a ratio of 6:4:1. 4 Dioxane/water/ammonia water was used as a solvent to isolate the product on preparative thin-layer chromatography to give Compound IV-1, yield: 30% (18 mg).
化合物IV-1的质谱数据:MS(MALDI–):m/z=614.6(M–1).Mass spectral data of compound IV-1: MS (MALDI-): m/z = 614.6 (M - 1).
在高效液相色谱(HPLC)上,用1:1乙腈/水作为流动相进行测试,流出时间如图1所示。On high performance liquid chromatography (HPLC), 1:1 acetonitrile/water was used as the mobile phase for the flow phase as shown in Figure 1.
(4)合成化合物V-1(4) Synthesis of compound V-1
Figure PCTCN2017105734-appb-000039
Figure PCTCN2017105734-appb-000039
在380mg(5mmol)2-羟基乙酸中加入50mL DMF,加入1.8g(6mmol)四氟硼酸O-(N-琥珀酰亚胺基)-N,N,N′,N′-四甲基脲和1.04mL N,N-二异丙基乙基胺,室温搅拌,反应30分钟。之后加入780mg N-乙二胺三氟乙酰胺,室温搅拌,反应2个小时。旋转蒸发去除所有溶剂,加入100mL乙酸乙酯溶解剩余固体。用0.1M盐酸溶液萃取(2×100mL),之后用饱和碳酸钠溶液萃取(2×150mL),之后用硫酸镁干燥,过滤后旋转蒸发去除溶剂,即得到化合物V-1,产率:75%(0.8g)。Add 50 mL of DMF to 380 mg (5 mmol) of 2-hydroxyacetic acid, and add 1.8 g (6 mmol) of O-(N-succinimidyl)-N,N,N',N'-tetramethylurea tetrafluoroborate and 1.04 mL of N,N-diisopropylethylamine was stirred at room temperature for 30 minutes. Thereafter, 780 mg of N-ethylenediamine trifluoroacetamide was added, and the mixture was stirred at room temperature for 2 hours. All solvents were removed by rotary evaporation and 100 mL of ethyl acetate was added to dissolve the residual solid. It was extracted with a 0.1 M hydrochloric acid solution (2×100 mL), and then extracted with a saturated sodium carbonate solution (2×150 mL), then dried over magnesium sulfate, filtered and evaporated to remove solvent to yield compound V-1, yield: 75% (0.8g).
化合物V-1的核磁和质谱数据:1H NMR(300MHz,CDCl3):δ(ppm)4.05(s,2H),3.40-3.30(m,4H),LC-MS(ESI):m/z 215.2(M+H+).Nuclear magnetic and mass spectral data of the compound V-1: 1 H NMR (300 MHz, CDCl 3 ): δ (ppm) 4.05 (s, 2H), 3.40 - 3.30 (m, 4H), LC-MS (ESI): m/z 215.2 (M+H+).
(5)将化合物V-1转化为合成化合物VI-1(5) Conversion of compound V-1 to synthetic compound VI-1
Figure PCTCN2017105734-appb-000040
Figure PCTCN2017105734-appb-000040
在10mL无水二氯甲烷中加入214mg(1mmol)化合物V-1,加入205μL N,N-二异丙基乙基胺,之后在0℃下加入溶解有283mg(1.2mmol)的2-氰乙基N,N-二异丙基氯代亚磷酰胺的无水二氯甲烷2mL,继续在0℃下搅拌10分钟。缓慢升温到室温,反应2小时。 之后加入20mL二氯甲烷和50mL饱和碳酸氢钠溶液,萃取之后,加入硫酸镁干燥,去除溶剂。利用1:1的乙酸乙酯/正己烷为洗脱剂,在硅胶柱上分离得到化合物VI-1,产率:85%(352mg)。214 mg (1 mmol) of compound V-1 was added to 10 mL of anhydrous dichloromethane, and 205 μL of N,N-diisopropylethylamine was added, followed by the addition of 283 mg (1.2 mmol) of 2-cyanoethyl at 0 °C. 2 mL of anhydrous dichloromethane of N,N-diisopropylchlorophosphoramidite was further stirred at 0 °C for 10 minutes. The temperature was slowly raised to room temperature and reacted for 2 hours. Thereafter, 20 mL of dichloromethane and 50 mL of a saturated sodium hydrogencarbonate solution were added, and after extraction, magnesium sulfate was added to dryness to remove the solvent. Compound VI-1 was isolated on a silica gel column using 1:1 ethyl acetate / n-hexane as eluent. Yield: 85% (352 mg).
化合物VI-1的核磁和质谱数据:1H NMR(300MHz,CDCl3):δ(ppm)4.16(s,2H),3.8(m,2H),3.40-3.30(m,6H),3.10(t,2H),1.16(d,12H)LC-MS(ESI):m/z 415.3(M+H+).Nuclear magnetic and mass spectral data of compound VI-1: 1 H NMR (300 MHz, CDCl 3 ): δ (ppm) 4.16 (s, 2H), 3.8 (m, 2H), 3.40 - 3.30 (m, 6H), 3.10 (t, 2H), 1.16 (d, 12H) LC-MS (ESI): m/z 415.3 (M+H+).
(6)将化合物VI-1转化为化合物VII-1(6) Conversion of compound VI-1 to compound VII-1
Figure PCTCN2017105734-appb-000041
Figure PCTCN2017105734-appb-000041
在3mL无水乙腈中溶解207mg(0.5mmol)化合物VI-1,将该溶液缓慢加入到溶有70mg(1mmol)的四唑和239mg(1mmol)的2-硝基-5-羟基-苯甲酸叔丁酯的无水乙腈(7mL)中,室温搅拌,反应30分钟。之后加入碘溶液5mL,溶剂为水/吡啶/四氢呋喃(2/20/78),碘的浓度为0.15M,含0.75mmol碘单质,室温搅拌,反应30分钟。之后缓慢加入5%的亚硫酸钠溶液,直至溶液中碘单质的颜色消失。旋转蒸发去除所有溶剂,加入5mL水溶解剩余固体,加入5mL 2M氢氧化钾,室温搅拌2个小时。调节溶液的pH至偏酸性,使用大量甲醇溶解固体,之后旋转蒸发去除溶剂。利用水为溶剂对剩余固体重结晶,得到化合物VII-1,产率:36%(131mg)。207 mg (0.5 mmol) of compound VI-1 was dissolved in 3 mL of anhydrous acetonitrile, and the solution was slowly added to a solution of 70 mg (1 mmol) of tetrazole and 239 mg (1 mmol) of 2-nitro-5-hydroxy-benzoic acid. The butyl ester in anhydrous acetonitrile (7 mL) was stirred at room temperature for 30 minutes. Then, 5 mL of an iodine solution was added, and the solvent was water/pyridine/tetrahydrofuran (2/20/78), the concentration of iodine was 0.15 M, and 0.75 mmol of iodine was contained, and the mixture was stirred at room temperature for 30 minutes. Thereafter, a 5% sodium sulfite solution was slowly added until the color of the iodine element in the solution disappeared. All solvents were removed by rotary evaporation, 5 mL of water was added to dissolve the remaining solid, and 5 mL of 2M potassium hydroxide was added and stirred at room temperature for 2 hours. The pH of the solution was adjusted to be acidic, and a large amount of methanol was used to dissolve the solid, followed by rotary evaporation to remove the solvent. Recrystallization of the remaining solid using water as a solvent gave Compound VII-1, yield: 36% (131 mg).
化合物VII-1的核磁和质谱数据:1H NMR(300MHz,CDCl3):δ(ppm)8.23-8.11(m,2H),7.64(m,1H),4.45(s,2H),3.40-3.30(t,2H),2.90(t,2H),LC-MS(ESI):m/z362.1(M-H-).Nuclear magnetic and mass spectral data of compound VII-1: 1 H NMR (300 MHz, CDCl 3 ): δ (ppm) 8.23 - 8.11 (m, 2H), 7.64 (m, 1H), 4.45 (s, 2H), 3.40 - 3.30 ( t, 2H), 2.90 (t, 2H), LC-MS (ESI): m/z 362.1 (MH-).
(7)对化合物VII-1进行荧光标记,得到化合物VIII-1(7) Fluorescent labeling of compound VII-1 to obtain compound VIII-1
Figure PCTCN2017105734-appb-000042
Figure PCTCN2017105734-appb-000042
将12.8mg(20umol)Cy3的N-羟基琥珀酰亚胺酯溶解于1mL DMF中,向其中加入溶解有4.2μL(24μmol)N,N-二异丙基乙基胺和8.7mg(24μmol)化合物VII-1的DMF (1mL),室温搅拌,反应2个小时,之后旋转蒸发去除溶剂。加入尽可能少的DMF溶解剩余固体,之后利用100%乙腈作为流动相,在HPLC上纯化,得到化合物VIII-1,产率:95%(18.6mg)。12.8 mg (20 umol) of Cy3 N-hydroxysuccinimide ester was dissolved in 1 mL of DMF, and 4.2 μL (24 μmol) of N,N-diisopropylethylamine and 8.7 mg (24 μmol) of compound dissolved therein were added thereto. DMF of VII-1 (1 mL), stirred at room temperature, reacted for 2 hours, then evaporated to remove solvent. The remaining solid was dissolved by adding as little DMF as possible, and then purified by HPLC using 100% acetonitrile as a mobile phase to give Compound VIII-1, yield: 95% (18.6 mg).
化合物VIII-1的质谱数据:LC-MS(ESI):m/z 975.2(M-H-).Mass spectral data for compound VIII-1: LC-MS (ESI): m/z 975.2 (M-H-).
(8)化合物IV-1与化合物VIII-1连接,生成带有荧光基团Cy3且3’-OH被可逆阻断的dTTP(8) Compound IV-1 is linked to compound VIII-1 to form dTTP having a fluorescent group Cy3 and 3'-OH is reversibly blocked.
Figure PCTCN2017105734-appb-000043
Figure PCTCN2017105734-appb-000043
在溶有9.8mg(10μmol)化合物VIII-1的1mL DMF中,加入2μL(12μmol)N,N-二异丙基乙基胺和3.6mg(12μmol)四氟硼酸O-(N-琥珀酰亚胺基)-N,N,N′,N′-四甲基脲,反应30分钟之后,加入溶解有6.2mg(10μmol)化合物IV-1的1mL 0.1M的碳酸氢钠溶液,室温搅拌,反应2小时。高真空去除所有的溶剂,加入尽可能少的水,之后用1:1的水/乙腈和0.05M三乙胺溶液(pH7.0)作为流动相,在HPLC上纯化,得到标记有Cy3且3’-OH被阻断的dTTP。产率:90%(14.1mg)。2 μL (12 μmol) of N,N-diisopropylethylamine and 3.6 mg (12 μmol) of tetrafluoroboric acid O-(N-succinyl) were added to 1 mL of DMF in which 9.8 mg (10 μmol) of Compound VIII-1 was dissolved. Amino)-N,N,N',N'-tetramethylurea, after reacting for 30 minutes, add 1 mL of 0.1 M sodium hydrogen carbonate solution dissolved in 6.2 mg (10 μmol) of compound IV-1, and stir at room temperature. 2 hours. Remove all solvents in high vacuum, add as little water as possible, then use 1:1 water/acetonitrile and 0.05 M triethylamine solution (pH 7.0) as the mobile phase, and purify on HPLC to obtain the label labeled Cy3 and 3. '-OH blocked dTTP. Yield: 90% (14.1 mg).
终产物的质谱数据:LC-MS(ESI):m/z 1573.2(M-H-);HPLC图谱见图2。Mass spectral data of the final product: LC-MS (ESI): m/z 1573.2 (M-H-);
实施例2以碱基A为例,制备带有Cy3荧光基团的、3’-OH被阻断的核苷酸(dATP)。Example 2, taking base A as an example, prepares a 3'-OH blocked nucleotide (dATP) having a Cy3 fluorescent group.
参照实施例1(1)-(3)的过程,制备用于合成dATP的中间体(化合物IV-2)。化合物IV-2 的结构如下:An intermediate (Compound IV-2) for the synthesis of dATP was prepared by referring to the procedures of Examples 1 (1) to (3). Compound IV-2 The structure is as follows:
Figure PCTCN2017105734-appb-000044
Figure PCTCN2017105734-appb-000044
化合物IV-2的质谱数据:MS(MALDI–):m/z=636.1(M–1)。Mass spectral data for compound IV-2: MS (MALDI -): m/z = 636.1 (M -1).
在高效液相色谱上,用1:1的乙腈/水作为流动相,流出时间如图3所示。On high performance liquid chromatography, 1:1 acetonitrile/water was used as the mobile phase, and the effluent time was as shown in Fig. 3.
参照实施例1的过程,制备带有Cy3荧光基团的、3’-OH被阻断的dATP。Referring to the procedure of Example 1, 3'-OH blocked dATP having a Cy3 fluorescent group was prepared.
实施例3修饰的dTTP的聚合Example 3 Polymerization of Modified dTTP
本实施例中,使用示例性的模板和引物对本发明的修饰的dTTP进行聚合,对聚合效率进行测试,考察聚合酶和聚合条件对聚合效率的影响。In this example, the modified dTTP of the present invention was polymerized using an exemplary template and primers, and the polymerization efficiency was tested to examine the effect of the polymerase and polymerization conditions on the polymerization efficiency.
(1)模板和引物序列(1) Template and primer sequences
模板:5’-CAACAGAAGGATTCTGGCGAACCGGAGGCTGAA--3’(SEQ ID NO:1)Template: 5'-CAACAGAAGGATTCTGGCGAACCGGAGGCTGAA--3' (SEQ ID NO: 1)
引物:3’-TGTCTTCCTAAGACCGCTTGGCCTCCGACTT-5’(SEQ ID NO:2)Primer: 3'-TGTCTTCCTAAGACCGCTTGGCCTCCGACTT-5' (SEQ ID NO: 2)
(2)聚合酶(2) polymerase
theminator(NEB)、Taq(NEB)、BST 2.0(NEB)、BST 3.0(NEB)、9°Nm(NEB)、KOD(merckmillipore)Theminator (NEB), Taq (NEB), BST 2.0 (NEB), BST 3.0 (NEB), 9°Nm (NEB), KOD (merckmillipore)
KOD聚合酶的突变体:KOD POL151、KOD POL157、KOD POL171、KOD POL174、KOD POL376、KOD POL391Mutants of KOD polymerase: KOD POL151, KOD POL157, KOD POL171, KOD POL174, KOD POL376, KOD POL391
上述聚合酶或其突变体均通过购买得到。The above polymerase or a mutant thereof is obtained by purchase.
(3)测序系统(3) Sequencing system
BGISEQ-500测序系统(操作方法参考BGISEQ-500用户手册)BGISEQ-500 Sequencing System (refer to the BGISEQ-500 User Manual for the method of operation)
(4)测试过程(4) Test process
图4示例性地说明了测试过程。Figure 4 exemplifies the testing process.
测试使用的芯片包括两个聚合反应区域:区域1和区域2,其中,区域1上进行的是参照组的反应,区域2上进行的是待测组的反应。已知在下文中提到的测试条件下,Cy3修饰的双脱氧的dTTP可以被Taq DNA聚合酶100%的聚合,因此作为参照组。The chip used for the test consisted of two polymerization reaction zones: zone 1 and zone 2, wherein zone 1 was the reaction of the reference group and zone 2 was the reaction of the group to be tested. It is known that Cy3-modified dideoxy dTTP can be polymerized 100% by Taq DNA polymerase under the test conditions mentioned below, thus serving as a reference group.
Cy3修饰的双脱氧的dTTP(三乙胺盐)购自Jena Bioscience,化学结构如下: Cy3 modified dideoxy dTTP (triethylamine salt) was purchased from Jena Bioscience and its chemical structure is as follows:
Figure PCTCN2017105734-appb-000045
Figure PCTCN2017105734-appb-000045
将芯片装载好。首先对芯片进行拍照,采集背景信号值,之后,在区域1上加入反应液1,其中含有Taq DNA聚合酶以及Cy3修饰的双脱氧的dTTP,在区域2上加入反应液2,其中含有待测的聚合酶以及实施例1制备的被可逆阻断且带有荧光基团Cy3的dTTP,分别在各自合适的温度下进行聚合反应10分钟,dTTP的浓度均为10μM,缓冲液使用聚合酶产品说明书推荐的缓冲体系。反应结束后,用1X磷酸盐缓冲液重复清洗芯片两边,去除未反应的dTTP;泵入含有10mM的维生素C的1X磷酸盐缓冲液,对芯片两边进行拍照,采集信号值。Load the chip. First, the chip is photographed, and the background signal value is collected. Then, the reaction solution 1 containing Taq DNA polymerase and Cy3 modified dideoxy dTTP is added to the region 1, and the reaction solution 2 is added to the region 2, which contains the test to be tested. The polymerase and the reversibly blocked dTTP with the fluorophore Cy3 prepared in Example 1 were each polymerized at a suitable temperature for 10 minutes, and the concentration of dTTP was 10 μM. The buffer was polymerase product specification. Recommended buffer system. After the reaction, the chips were repeatedly washed with 1X phosphate buffer to remove unreacted dTTP; 1X phosphate buffer containing 10 mM of vitamin C was pumped, and both sides of the chip were photographed to collect signal values.
每次测试得到4个数值,包括参照组和待测组的空白背景值(a和b),以及参照组和待测组在聚合之后的信号值(A和B)。已知对照组的聚合效率为100%,待测组的聚合效率可以用下面的方程式计算:Each test yielded four values, including the blank background values (a and b) of the reference group and the test group, and the signal values (A and B) of the reference group and the test group after polymerization. It is known that the polymerization efficiency of the control group is 100%, and the polymerization efficiency of the test group can be calculated by the following equation:
待测组的聚合效率=(B-b)/(A-a)*100%。The polymerization efficiency of the test group = (B-b) / (A - a) * 100%.
使用不同聚合酶进行聚合,测试结果如表1所示(不同批次测试的背景值之间、以及参照组的信号之间略有差异)。Polymerization was carried out using different polymerases, and the test results are shown in Table 1 (the difference between the background values of the different batch tests and the signals of the reference group).
表1Table 1
Figure PCTCN2017105734-appb-000046
Figure PCTCN2017105734-appb-000046
测试结果显示,表1中的聚合酶对实施例1制得的dTTP能够聚合,但是,除了KOD外,其他聚合酶的聚合效率比较低。使用KOD作为聚合酶的聚合效率是可接受的。The test results showed that the polymerases in Table 1 were able to polymerize the dTTP prepared in Example 1, but the polymerization efficiency of other polymerases was lower than that of KOD. The polymerization efficiency using KOD as a polymerase is acceptable.
进一步地,使用KOD聚合酶的突变体KOD POL151、KOD POL157、KOD POL171、 KOD POL174、KOD POL376、KOD POL391作为聚合酶,进行测试。这些突变体在对现有技术中的某些可逆阻断dNTP进行聚合时,聚合效果较差。Further, mutants KOD POL151, KOD POL157, KOD POL171, using KOD polymerase, KOD POL174, KOD POL376, and KOD POL391 were tested as polymerases. These mutants have a poor polymerization effect when polymerizing some reversible blocking dNTPs in the prior art.
采用Taq DNA聚合酶和Cy3修饰的双脱氧的dTTP为参照组,采用SEQ ID NO:1所示的模板和SEQ ID NO:2所示的引物,以及与上文相同的实验方法,聚合10分钟。测试结果如表2所示。The dideoxy dTTP modified with Taq DNA polymerase and Cy3 was used as a reference group, and the template shown in SEQ ID NO: 1 and the primer shown in SEQ ID NO: 2, and the same experimental method as above, were used for polymerization for 10 minutes. . The test results are shown in Table 2.
表2Table 2
Figure PCTCN2017105734-appb-000047
Figure PCTCN2017105734-appb-000047
从表2可以看出,聚合酶KOD POL391和KOD POL171对实施例1制得的dTTP有较高的聚合效率。As can be seen from Table 2, the polymerases KOD POL391 and KOD POL171 had higher polymerization efficiencies for the dTTP prepared in Example 1.
进一步地,发明人对使用KOD POL391进行聚合的反应条件(包括反应溶液的pH、盐浓度、缓冲体系浓度、添加剂,以及反应温度)进行优化。Further, the inventors optimized the reaction conditions (including the pH of the reaction solution, the salt concentration, the buffer system concentration, the additive, and the reaction temperature) for polymerization using KOD POL391.
最初的反应条件为:反应溶液的pH为9.0,包含50mM氯化钠、1mM硫酸镁、50mM Tris、0.05%吐温-20,反应温度为55℃。在优化反应条件时,不使用参照组,而是在芯片的两个反应区域上进行对比测试,聚合时间为5分钟。先被优化得到的更好的条件被用于后续的测试中。测试结果如表3-1~表3-6所示。The initial reaction conditions were as follows: the pH of the reaction solution was 9.0, and contained 50 mM sodium chloride, 1 mM magnesium sulfate, 50 mM Tris, 0.05% Tween-20, and the reaction temperature was 55 °C. When the reaction conditions were optimized, the reference group was not used, but a comparison test was performed on the two reaction areas of the chip, and the polymerization time was 5 minutes. The better conditions that were first optimized were used in subsequent tests. The test results are shown in Table 3-1 to Table 3-6.
表3-1Table 3-1
pHpH pH 9.0pH 9.0 pH 8.5pH 8.5 pH 8.0pH 8.0 pH7.8pH 7.8 pH 7.5pH 7.5 pH 7.0pH 7.0
聚合效率Polymerization efficiency 55.1255.12 76.6576.65 89.9089.90 93.7693.76 80.2180.21 37.3837.38
其他条件:反应溶液包含50mM氯化钠、1mM硫酸镁、50mM Tris、0.05%吐温-20,反应温度为55℃。Other conditions: The reaction solution contained 50 mM sodium chloride, 1 mM magnesium sulfate, 50 mM Tris, 0.05% Tween-20, and the reaction temperature was 55 °C.
表3-2Table 3-2
氯化钠浓度Sodium chloride concentration 20mM20mM 100mM100mM
聚合效率Polymerization efficiency 96.3396.33 92.5092.50
其他条件:反应溶液pH7.8,包含1mM硫酸镁、50mM Tris、0.05%吐温-20,反应温度为55℃。Other conditions: The reaction solution was pH 7.8, and contained 1 mM magnesium sulfate, 50 mM Tris, 0.05% Tween-20, and the reaction temperature was 55 °C.
表3-3Table 3-3
硫酸镁浓度Magnesium sulfate concentration 10mM10mM 3mM3mM
聚合效率Polymerization efficiency 93.9493.94 97.4097.40
其他条件:反应溶液pH7.8,包含20mM氯化钠、50mM Tris、0.05%吐温-20,反应温度为55℃。Other conditions: The reaction solution was pH 7.8, and contained 20 mM sodium chloride, 50 mM Tris, 0.05% Tween-20, and the reaction temperature was 55 °C.
表3-4Table 3-4
Tris浓度Tris concentration 100mM100mM 20mM20mM
聚合效率Polymerization efficiency 94.6794.67 93.2893.28
其他条件:反应溶液pH7.8,包含20mM氯化钠、3mM硫酸镁、0.05%吐温-20,反应温度为55℃。Other conditions: The reaction solution was pH 7.8, and contained 20 mM sodium chloride, 3 mM magnesium sulfate, 0.05% Tween-20, and the reaction temperature was 55 °C.
表3-5Table 3-5
添加剂additive 2%DMSO2% DMSO 5%DMSO5% DMSO 5%甘油5% glycerin
聚合效率Polymerization efficiency 98.4598.45 99.1099.10 88.9988.99
其他条件:反应溶液pH7.8,包含20mM氯化钠、3mM硫酸镁、50mM Tris、0.05%吐温-20,反应温度为55℃。Other conditions: The reaction solution was pH 7.8, and contained 20 mM sodium chloride, 3 mM magnesium sulfate, 50 mM Tris, 0.05% Tween-20, and the reaction temperature was 55 °C.
表3-6Table 3-6
反应温度temperature reflex 57℃57°C 60℃60 ° C 63℃63°C
聚合效率Polymerization efficiency 99.0299.02 98.4398.43 93.7793.77
其他条件:反应溶液pH7.8,包含20mM氯化钠、3mM硫酸镁、50mM Tris、0.05%吐温-20、5%DMSO。Other conditions: The reaction solution was pH 7.8 and contained 20 mM sodium chloride, 3 mM magnesium sulfate, 50 mM Tris, 0.05% Tween-20, 5% DMSO.
从表3-1~表3-6可以看出,改变pH到7.8和加入5%DMSO对聚合效率影响最大,其他条件对pH几乎无正面提升。最终优化得到的反应条件是:反应溶液pH7.8,包含20mM氯化钠、3mM硫酸镁、50mM Tris、5%DMSO、0.05%吐温-20,反应温度为57℃。It can be seen from Table 3-1 to Table 3-6 that changing the pH to 7.8 and adding 5% DMSO have the greatest influence on the polymerization efficiency, and other conditions have almost no positive effect on the pH. The reaction conditions finally optimized were: pH 7.8 of the reaction solution, containing 20 mM sodium chloride, 3 mM magnesium sulfate, 50 mM Tris, 5% DMSO, 0.05% Tween-20, and the reaction temperature was 57 °C.
在最终优化的条件下,测定聚合酶KOD POL391和KOD POL171对实施例1得到的dTTP的聚合效率,聚合反应进行10分钟。以Taq DNA聚合酶聚合Cy3修饰的双脱氧的dTTP的反应作为参照组。结果如表4所示。The polymerization efficiency of the polymerase KOD POL391 and KOD POL171 against the dTTP obtained in Example 1 was measured under the conditions of the final optimization, and the polymerization reaction was carried out for 10 minutes. The reaction of the Cy3-modified dideoxy dTTP polymerized by Taq DNA polymerase was used as a reference group. The results are shown in Table 4.
表4Table 4
Figure PCTCN2017105734-appb-000048
Figure PCTCN2017105734-appb-000048
Figure PCTCN2017105734-appb-000049
Figure PCTCN2017105734-appb-000049
如表4所示,实施例1制得的Cy3修饰的可逆阻断的dTTP可以被KOD POL391或KOD POL171高效聚合,聚合效率接近100%。As shown in Table 4, the Cy3-modified reversibly blocked dTTP prepared in Example 1 can be efficiently polymerized by KOD POL391 or KOD POL171, and the polymerization efficiency is close to 100%.
实施例4阻断基团的去除Example 4 Removal of Blocking Groups
本实施例中,首先对被可逆阻断但不带荧光基团的dTTP进行聚合。用于本实施例的被可逆阻断但不带荧光基团的dTTP为实施例1中的化合物IV-1,结构如下。在优化的条件下,其可以被100%聚合。In this example, the dTTP which is reversibly blocked but does not have a fluorescent group is first polymerized. The dTTP which was used in the present embodiment to be reversibly blocked without a fluorescent group was the compound IV-1 in Example 1, and the structure was as follows. Under optimized conditions, it can be 100% polymerized.
Figure PCTCN2017105734-appb-000050
Figure PCTCN2017105734-appb-000050
聚合之后,使用内切酶IV(NEB)对阻断基团进行切除,并通过切除之后加入下一位碱基的方式测试切除的效率。After the polymerization, the blocking group was excised using endonuclease IV (NEB), and the efficiency of excision was tested by the addition of the next base after excision.
图5示例性地说明了测试过程。Figure 5 exemplifies the testing process.
首先,将有两个反应区域(区域1和区域2)的芯片装载好,其中,区域1上进行的是参照组的反应,区域2上进行的是待测组的反应。两个区域分别加入两个不同的引物,其中,参照组的引物如SEQ ID NO:3所示,其比待测组的引物(如SEQ ID NO:2所示)在3端多了一个T碱基,因此,在待测组加入一个T碱基并进行切除之后,参照组和待测组成为拥有相同序列的体系,可以通过对比下一个碱基的信号来确定待测组的切除效率。First, a chip having two reaction regions (region 1 and region 2) is loaded, wherein the reaction of the reference group is performed on the region 1, and the reaction of the group to be tested is performed on the region 2. Two different primers are added to the two regions, wherein the primer of the reference group is as shown in SEQ ID NO: 3, which is one more T at the 3 end than the primer of the test group (as shown in SEQ ID NO: 2). Base, therefore, after adding a T base to the test group and excising, the reference group and the test composition are systems having the same sequence, and the excision efficiency of the test group can be determined by comparing the signals of the next base.
在区域1和区域2中,均使用聚合酶KOD POL391,在实施例3得到的最终优化的反应条件下,对化合物IV-1进行聚合,化合物IV的浓度为10μM,聚合进行10分钟。洗掉未反应的试剂,以内切酶4(100U/mL)作为切除试剂,在NEB缓冲液3中进行切除反应,反应温度为37℃,切除时间为10min、20min、40min或60min。对芯片进行洗涤,之后对芯片采集信号,获得参照组和待测组的背景值a和b。加入Taq DNA和Cy3修饰的ddGTP,进行聚合反应10分钟,之后洗掉未反应的ddGTP,拍照采集信号,获得参照组和待测组的信号值A和B。利用下面的方程可算出切除效率,In Region 1 and Region 2, polymerase KOD POL391 was used, and under the finally optimized reaction conditions obtained in Example 3, Compound IV-1 was polymerized, the concentration of Compound IV was 10 μM, and polymerization was carried out for 10 minutes. The unreacted reagent was washed away, and the excision reagent 4 (100 U/mL) was used as the excision reagent, and the excision reaction was carried out in NEB buffer 3 at a reaction temperature of 37 ° C, and the excision time was 10 min, 20 min, 40 min or 60 min. The chip is washed, and then a signal is acquired on the chip to obtain background values a and b of the reference group and the group to be tested. Taq DNA and Cy3 modified ddGTP were added, and polymerization was carried out for 10 minutes, after which unreacted ddGTP was washed away, and a signal was collected to obtain signal values A and B of the reference group and the test group. The cutting efficiency can be calculated using the equation below.
切除效率==(B-b)/(A-a)*100%。Excision efficiency == (B-b) / (A-a) * 100%.
表5和图6显示了不同切除时间下的切除效率。 Table 5 and Figure 6 show the efficiency of the ablation at different ablation times.
表5table 5
Figure PCTCN2017105734-appb-000051
Figure PCTCN2017105734-appb-000051
从表5和图6可以看出,切除效率在20分钟之后进入平台期,最终的切除效率达到100%。As can be seen from Table 5 and Figure 6, the cutting efficiency entered the plateau after 20 minutes, and the final cutting efficiency reached 100%.
实施例5荧光基团的去除Example 5 Removal of Fluorescent Groups
本实施例中,通过切断本发明修饰核苷酸中的磷酸二酯键(2),去除荧光基团,并测试切除的效率。In this example, the fluorophore was removed by cleaving the phosphodiester bond (2) in the modified nucleotide of the present invention, and the efficiency of excision was tested.
图7示例性地说明了测试过程。Figure 7 exemplifies the testing process.
在芯片上对实施例1制得的被Cy-3修饰且被可逆阻断的dTTP进行聚合,聚合酶为KOD POL391,反应条件为实施例1最终优化的反应条件,dTTP的浓度为10μM。聚合前后分别采集信号值,其中,聚合之前的信号值为背景A1,聚合之后的信号值为a,之后使用碱性磷酸酶(NEB,货号M0371S)进行切除,分别切除2、5、10或20分钟,切除之后洗掉反应试剂,再采集信号值,为A2,按照下边的方程计算切除效率:The CyT-modified and reversibly blocked dTTP prepared in Example 1 was polymerized on a chip, and the polymerase was KOD POL391. The reaction conditions were the final optimized reaction conditions of Example 1, and the concentration of dTTP was 10 μM. The signal values are collected before and after the polymerization, wherein the signal value before the polymerization is the background A1, and the signal value after the polymerization is a, and then the alkaline phosphatase (NEB, item number M0371S) is used for excision, and the 2, 5, 10 or 20 are respectively cut off. Minutes, after the excision, wash off the reagents, and then collect the signal value, which is A2, and calculate the resection efficiency according to the following equation:
切除效率=(a-A2)/(a-A1)*100%。Excision efficiency = (a-A2) / (a-A1) * 100%.
测试结果如表6和图8所示。The test results are shown in Table 6 and Figure 8.
表6Table 6
切除时间(min)Cutting time (min) A1A1 aa A2A2 切除效率Resection efficiency
22 434434 60126012 13821382 83%83%
55 333333 45674567 672672 92%92%
1010 282282 47184718 370370 98%98%
2020 426426 58405840 412412 100%100%
从表6和图8可以看出,经过20min的切除,核酸链上的荧光基团可以被完全除去。As can be seen from Table 6 and Figure 8, the fluorophore on the nucleic acid strand can be completely removed after 20 min of excision.
实施例6测试叠氮亚甲基阻断基团的稳定性和磷酸甲酯阻断基团的稳定性。Example 6 tested the stability of the azide methylene blocking group and the stability of the methyl phosphate blocking group.
待测化合物:Test compound:
(1)实施例1制得的带有荧光基团Cy3且3’-OH被磷酸甲酯阻断的dTTP;(1) dTTP having a fluorescent group Cy3 and 3'-OH blocked by methyl phosphate prepared in Example 1;
(2)带有荧光基团Cy3且3’-OH被叠氮亚甲基阻断的dTTP(结构如下,由mychemlab购买)。 (2) dTTP having a fluorescent group Cy3 and 3'-OH blocked by an azide methylene group (structure is as follows, purchased by mychemlab).
Figure PCTCN2017105734-appb-000052
Figure PCTCN2017105734-appb-000052
测试过程:Testing process:
分别在1mg的两个化合物样品中加入1M tris-buffer(pH 8.0)(0.3mL)和人血清蛋白,65℃下混合搅拌36小时;之后,使用液相色谱-质谱联用(LC)对两个样品进行分析(LC-MS),用1:1的乙腈:乙酸三乙胺盐的水溶液(0.05M,pH为7)为流动相,检测两个样品是否有3’端保护基团脱掉的碱基或者荧光修饰基团脱掉的分子质谱峰出现。1 M tris-buffer (pH 8.0) (0.3 mL) and human serum albumin were added to 1 mg of the two compound samples, respectively, and stirred at 65 ° C for 36 hours; then, using liquid chromatography-mass spectrometry (LC) for two One sample was analyzed (LC-MS), and a 1:1 acetonitrile:acetic acid triethylamine salt aqueous solution (0.05 M, pH 7) was used as a mobile phase to detect whether two samples had a 3' end protecting group removed. Molecular mass peaks of molecular bases or fluorescent modified groups are removed.
被磷酸甲酯阻断的dTTP的LC结果如图9所示,LC图谱上产生了一个非常小的新峰,经质谱鉴定,分子量为780.1(M-H)-,结构如下。The LC results of dTTP blocked by methyl phosphate are shown in Figure 9. A very small new peak was generated on the LC map and identified by mass spectrometry with a molecular weight of 780.1 (MH) - and the structure is as follows.
Figure PCTCN2017105734-appb-000053
Figure PCTCN2017105734-appb-000053
被叠氮亚甲基阻断的dTTP的LC结果如图10所示,LC图谱上有较大的两个新峰和较小的1个新峰。经质谱鉴定,结构和分子量为(按图中从左到右的顺序):The LC results of dTTP blocked by azide methylene group are shown in Figure 10. There are two larger new peaks and one smaller new peak on the LC map. Identification by mass spectrometry, structure and molecular weight (from left to right in the figure):
Figure PCTCN2017105734-appb-000054
Figure PCTCN2017105734-appb-000054
3’端不带保护基团而碱基带荧光修饰的核苷酸,1482.3(M-H)-A nucleotide with a protecting group at the 3' end and a fluorescent modification at the base, 1482.3 (MH) - .
尽管本发明的具体实施方式已经得到详细的描述,但本领域技术人员将理解:根据已经公开的所有教导,可以对细节进行各种修改和变动,并且这些改变均在本发明的保护范围之内。本发明的全部范围由所附权利要求及其任何等同物给出。 While the invention has been described in detail, the embodiments of the invention . The full scope of the invention is given by the appended claims and any equivalents thereof.

Claims (32)

  1. 具有通式(I)所示结构的化合物,a compound having the structure represented by the formula (I),
    Figure PCTCN2017105734-appb-100001
    Figure PCTCN2017105734-appb-100001
    其中,R1和R3各自独立地选自
    Figure PCTCN2017105734-appb-100002
    Figure PCTCN2017105734-appb-100003
    Wherein R 1 and R 3 are each independently selected from
    Figure PCTCN2017105734-appb-100002
    Figure PCTCN2017105734-appb-100003
    A选自苯基、萘基、吲哚基和吡啶基;A is selected from the group consisting of phenyl, naphthyl, anthryl and pyridyl;
    R2选自硝基、卤代C1-4烷基、卤素、氢、醛基、
    Figure PCTCN2017105734-appb-100004
    其中,Q独立地选自C1-4烷基;
    R 2 is selected from the group consisting of a nitro group, a halogenated C 1-4 alkyl group, a halogen, a hydrogen, an aldehyde group,
    Figure PCTCN2017105734-appb-100004
    Wherein Q is independently selected from C 1-4 alkyl;
    R4选自-H,单磷酸基团(-PO3H2),二磷酸基团(-PO3H-PO3H2),三磷酸基团(-PO3H-PO3H-PO3H2)和四磷酸基团(-PO3H-PO3H-PO3H-PO3H2);R 4 is selected from -H, a monophosphate group (-PO 3 H 2 ), a diphosphate group (-PO 3 H-PO 3 H 2 ), a triphosphate group (-PO 3 H-PO 3 H-PO 3 H 2 ) and a tetraphosphoric acid group (-PO 3 H-PO 3 H-PO 3 H-PO 3 H 2 );
    R6为氢或羟基;R 6 is hydrogen or a hydroxyl group;
    m和n各自独立地选自0、1、2、3、4、5;m and n are each independently selected from 0, 1, 2, 3, 4, 5;
    L为连接基团或不存在;L is a linking group or does not exist;
    Base代表碱基,例如为嘌呤碱基或嘧啶碱基,例如选自A、T、U、C和G中的一种;Base represents a base, such as a purine base or a pyrimidine base, for example, one selected from the group consisting of A, T, U, C, and G;
    Label表示可检测标记,例如为荧光基团;Label indicates a detectable label, such as a fluorophore;
    Blocker表示阻断基团。Blocker indicates a blocking group.
  2. 权利要求1的化合物,其中,R1选自
    Figure PCTCN2017105734-appb-100005
    优选地,R1
    Figure PCTCN2017105734-appb-100006
    The compound of claim 1 wherein R 1 is selected from
    Figure PCTCN2017105734-appb-100005
    Preferably, R 1 is
    Figure PCTCN2017105734-appb-100006
  3. 权利要求1或2的化合物,其中,R2选自硝基、三氟甲基、氟、氯、氢和醛基;优选地,R2为硝基。 The compound of claim 1 or 2, wherein R 2 is selected from the group consisting of nitro, trifluoromethyl, fluoro, chloro, hydrogen and aldehyde; preferably, R 2 is nitro.
  4. 权利要求1-3任一项的化合物,其中,R3选自
    Figure PCTCN2017105734-appb-100007
    优选地,R3
    Figure PCTCN2017105734-appb-100008
    The compound according to any one of claims 1 to 3, wherein R 3 is selected from
    Figure PCTCN2017105734-appb-100007
    Preferably, R 3 is
    Figure PCTCN2017105734-appb-100008
  5. 权利要求1-4任一项的化合物,其中,
    Figure PCTCN2017105734-appb-100009
    选自:
    A compound according to any one of claims 1 to 4, wherein
    Figure PCTCN2017105734-appb-100009
    From:
    Figure PCTCN2017105734-appb-100010
    Figure PCTCN2017105734-appb-100010
    优选地,
    Figure PCTCN2017105734-appb-100011
    Figure PCTCN2017105734-appb-100012
    Preferably,
    Figure PCTCN2017105734-appb-100011
    for
    Figure PCTCN2017105734-appb-100012
  6. 权利要求1-5任一项的化合物,其中,R4为三磷酸基团(-PO3H-PO3H-PO3H2)。A compound according to any one of claims 1 to 5, wherein R 4 is a triphosphate group (-PO 3 H-PO 3 H-PO 3 H 2 ).
  7. 权利要求1-6任一项的化合物,其中,R6为氢。A compound according to any one of claims 1 to 6, wherein R 6 is hydrogen.
  8. 权利要求1-7任一项的化合物,其中,Label选自Cy3、Cy3.5、Cy5和Cy5.5。The compound according to any one of claims 1 to 7, wherein the Label is selected from the group consisting of Cy3, Cy3.5, Cy5 and Cy5.5.
  9. 权利要求1-8任一项的化合物,其中,m为1。A compound according to any one of claims 1-8, wherein m is 1.
  10. 权利要求1-9任一项的化合物,其中,n为1。The compound of any one of claims 1-9, wherein n is 1.
  11. 权利要求1-10任一项的化合物,其中,L为
    Figure PCTCN2017105734-appb-100013
    A compound according to any one of claims 1 to 10, wherein L is
    Figure PCTCN2017105734-appb-100013
  12. 权利要求1-11任一项的化合物,其中,Blocker的结构为:The compound of any one of claims 1-11, wherein the structure of the Blocker is:
    Figure PCTCN2017105734-appb-100014
    Figure PCTCN2017105734-appb-100014
    其中,Ra1和Ra2各自独立地选自H、F、-CF3、-CHF2、-CH2F、-CH2W,-COOW、-CONHW;Wherein, Ra 1 and Ra 2 are each independently selected from the group consisting of H, F, -CF 3 , -CHF 2 , -CH 2 F, -CH 2 W, -COOW, -CONHW;
    W选自C1-C6烷基。 W is selected from a C 1 -C 6 alkyl group.
  13. 权利要求1-11任一项的化合物,其中,Blocker的结构为:The compound of any one of claims 1-11, wherein the structure of the Blocker is:
    Figure PCTCN2017105734-appb-100015
    Figure PCTCN2017105734-appb-100015
    其中,Rb1、Rb2、Rb3、Rb4、Rb5各自独立地选自H和C1-C6烷基。Wherein Rb 1 , Rb 2 , Rb 3 , Rb 4 and Rb 5 are each independently selected from H and C 1 -C 6 alkyl.
  14. 权利要求1-11任一项的化合物,其中,Blocker的结构为:The compound of any one of claims 1-11, wherein the structure of the Blocker is:
    Figure PCTCN2017105734-appb-100016
    Figure PCTCN2017105734-appb-100016
    其中,Rc1、Rc2各自独立地选自H、F、Cl和-CF3Wherein Rc 1 and Rc 2 are each independently selected from the group consisting of H, F, Cl and -CF 3 .
  15. 权利要求1-11任一项的化合物,其具有通式(I’)所示的结构:A compound according to any one of claims 1 to 11, which has the structure represented by the formula (I'):
    Figure PCTCN2017105734-appb-100017
    Figure PCTCN2017105734-appb-100017
    其中,R5选自C1-4烷基;Wherein R 5 is selected from C 1-4 alkyl;
    优选地,R5为甲基或乙基。Preferably, R 5 is methyl or ethyl.
  16. 权利要求1-15任一项的化合物,其具有通式(II)所示的结构A compound according to any one of claims 1 to 15, which has the structure represented by the formula (II)
    Figure PCTCN2017105734-appb-100018
    Figure PCTCN2017105734-appb-100018
    优选地,R2为硝基;Preferably, R 2 is a nitro group;
    优选地,Label为Cy3。Preferably, the Label is Cy3.
  17. 权利要求1-16任一项的化合物,所述化合物选自: A compound according to any one of claims 1 to 16 which is selected from the group consisting of:
    Figure PCTCN2017105734-appb-100019
    Figure PCTCN2017105734-appb-100019
    Figure PCTCN2017105734-appb-100020
    Figure PCTCN2017105734-appb-100020
  18. 制备在测序反应中与目标单链多核苷酸互补的生长的多核苷酸的方法,其包括将权利要求1-17中任一项的化合物并入所述生长的互补多核苷酸,其中,所述化合物的并入防止了任何后续的核苷酸引入所述生长的互补多核苷酸中;A method of preparing a growing polynucleotide that is complementary to a target single-stranded polynucleotide in a sequencing reaction, comprising incorporating the compound of any one of claims 1-17 into the growing complementary polynucleotide, wherein Incorporation of the compounds prevents any subsequent nucleotides from being introduced into the growing complementary polynucleotide;
    优选地,所述化合物的并入通过末端转移酶、末端聚合酶或逆转录酶来实现;Preferably, the incorporation of the compound is achieved by a terminal transferase, a terminal polymerase or a reverse transcriptase;
    优选地,所述方法包括:使用聚合酶,使所述化合物并入生长的互补多核苷酸;Preferably, the method comprises: incorporating the compound into a growing complementary polynucleotide using a polymerase;
    优选地,所述方法包括:在允许聚合酶进行核苷酸聚合反应的条件下,使用聚合酶进行核苷酸聚合反应,从而将所述化合物并入生长的互补多核苷酸的3'端。Preferably, the method comprises performing a nucleotide polymerization using a polymerase under conditions allowing the polymerase to undergo nucleotide polymerization, thereby incorporating the compound into the 3' end of the growing complementary polynucleotide.
  19. 测定目标单链多核苷酸的序列的方法,其包括:监测互补核苷酸的顺序并入,其中并入的至少一个互补核苷酸是权利要求1-17中任一项的化合物,以及,检测所述化合物携带的可检测标记;A method of determining the sequence of a single-stranded polynucleotide of interest, comprising: monitoring the sequential incorporation of a complementary nucleotide, wherein the at least one complementary nucleotide incorporated is a compound of any of claims 1-17, and Detecting a detectable label carried by the compound;
    优选地,在引入所述下一个互补核苷酸之前,将所述化合物中的阻断基团和所述可检测标记除去;Preferably, the blocking group and the detectable label in the compound are removed prior to introduction of the next complementary nucleotide;
    优选地,所述阻断基团和所述可检测标记被同时除去;Preferably, the blocking group and the detectable label are simultaneously removed;
    优选地,所述阻断基团和所述可检测标记被先后除去;例如,在所述可检测标记被除去之前或之后,所述阻断基团被除去。Preferably, the blocking group and the detectable label are removed in succession; for example, the blocking group is removed before or after the detectable label is removed.
  20. 权利要求19的方法,包括以下步骤:The method of claim 19 comprising the steps of:
    (a)提供包含双链体、权利要求1-17中任一项的化合物、聚合酶和切除试剂的混合物;所述双链体包含生长的核酸链以及待测序的核酸分子;(a) providing a mixture comprising a duplex, a compound according to any one of claims 1-17, a polymerase and a excision reagent; the duplex comprising a growing nucleic acid strand and a nucleic acid molecule to be sequenced;
    (b)进行包含以下步骤(i)、(ii)和(iii)的反应循环: (b) Carry out a reaction cycle comprising the following steps (i), (ii) and (iii):
    步骤(i):使用聚合酶,使所述化合物并入生长的核酸链,形成包含阻断基团和可检测标记的核酸中间体;Step (i): using a polymerase to incorporate the compound into a growing nucleic acid strand to form a nucleic acid intermediate comprising a blocking group and a detectable label;
    步骤(ii):对所述核酸中间体上的可检测标记进行检测;Step (ii): detecting a detectable label on the nucleic acid intermediate;
    步骤(iii):使用切除试剂将核酸中间体上的阻断基团除去;Step (iii): removing the blocking group on the nucleic acid intermediate using a resection reagent;
    优选地,所述反应循环还包括步骤(iv):使用切除试剂将核酸中间体上的可检测标记除去;Preferably, the reaction cycle further comprises the step (iv): removing the detectable label on the nucleic acid intermediate using a scavenging reagent;
    优选地,所述步骤(iii)和步骤(iv)中使用的切除试剂是同样的试剂;Preferably, the ablation reagent used in the step (iii) and the step (iv) is the same reagent;
    优选地,所述步骤(iii)和步骤(iv)中使用的切除试剂是不同的试剂。Preferably, the ablation reagents used in step (iii) and step (iv) are different reagents.
  21. 权利要求19的方法,其中,并入的至少一个互补核苷酸是如通式(I’)所示的化合物,所述方法包括以下步骤:The method of claim 19, wherein the at least one complementary nucleotide incorporated is a compound of the formula (I'), the method comprising the steps of:
    (1)提供双链体,其包含生长的核酸链以及待测序的核酸分子,所述双链体连接于支持物上;(1) providing a duplex comprising a growing nucleic acid strand and a nucleic acid molecule to be sequenced, the duplex being linked to a support;
    (2)添加用于进行核苷酸聚合反应的聚合酶,以及第一、第二、第三和第四化合物,从而形成含有溶液相和固相的反应体系;其中,所述四种化合物分别为核苷酸A、(T/U)、C和G的衍生物,均具有通式(I)所示的结构并且具有碱基互补配对能力;(2) adding a polymerase for performing nucleotide polymerization, and first, second, third, and fourth compounds, thereby forming a reaction system containing a solution phase and a solid phase; wherein the four compounds are respectively Derivatives of nucleotides A, (T/U), C and G, each having the structure represented by the general formula (I) and having a base complementary pairing ability;
    (3)在允许聚合酶进行核苷酸聚合反应的条件下,使用聚合酶进行核苷酸聚合反应,从而将所述四种化合物中的一种并入生长的核酸链的3'端;(3) performing nucleotide polymerization using a polymerase under conditions allowing the polymerase to undergo nucleotide polymerization, thereby incorporating one of the four compounds into the 3' end of the growing nucleic acid strand;
    (4)移除前一步骤的反应体系的溶液相,保留连接于支持物上的双链体,并检测所述双链体或所述生长的核酸链上的可检测标记所发出的信号;(4) removing the solution phase of the reaction system of the previous step, leaving the duplex attached to the support, and detecting the signal emitted by the detectable label on the duplex or the growing nucleic acid strand;
    (5)添加切除试剂,使所述双链体或所述生长的核酸链在含有溶液相和固相的反应体系中与切除试剂接触;其中,所述切除试剂能够使并入生长的核酸链3'端的化合物中的磷酸二酯键(1)和/或磷酸二酯键(2)断裂,并且不会影响双链体骨架上的磷酸二酯键;(5) adding a scavenging reagent such that the duplex or the growing nucleic acid strand is contacted with a scavenging reagent in a reaction system containing a solution phase and a solid phase; wherein the excision reagent enables the incorporation of the growing nucleic acid strand The phosphodiester bond (1) and/or the phosphodiester bond (2) in the compound at the 3' end are cleaved and do not affect the phosphodiester bond on the duplex backbone;
    (6)移除前一步骤的反应体系的溶液相;(6) removing the solution phase of the reaction system of the previous step;
    优选地,所述方法还包括下述步骤:Preferably, the method further comprises the steps of:
    (7)重复进行步骤(2)-(6)或步骤(2)-(4)一次或多次;(7) repeating steps (2)-(6) or steps (2)-(4) one or more times;
    任选地,在任意一个包含移除操作的步骤之后,进行洗涤操作。Optionally, the washing operation is performed after any of the steps including the removing operation.
  22. 权利要求21的方法,其中,步骤(1)中的双链体通过包含以下步骤的方法得到:The method of claim 21, wherein the duplex in step (1) is obtained by a method comprising the steps of:
    提供引物,使引物退火至待测序的核酸分子上,所述引物作为起始的生长的核酸链, 与所述待测序的核酸分子一起形成连接于支持物上的双链体。Primers are provided to anneal the primer to the nucleic acid molecule to be sequenced, the primer being the starting nucleic acid strand for growth, A duplex attached to the support is formed with the nucleic acid molecule to be sequenced.
  23. 权利要求21或22的方法,所述步骤(3)中的聚合酶选自KOD聚合酶或其突变体(例如KOD POL151、KOD POL157、KOD POL171、KOD POL174、KOD POL376、KOD POL391);The method of claim 21 or 22, wherein the polymerase in the step (3) is selected from the group consisting of KOD polymerase or a mutant thereof (for example, KOD POL151, KOD POL157, KOD POL171, KOD POL174, KOD POL376, KOD POL391);
    优选地,所述聚合酶选自KOD POL391和KOD POL171。Preferably, the polymerase is selected from the group consisting of KOD POL391 and KOD POL171.
  24. 权利要求21-23任一项的方法,所述步骤(2)中的溶液相包含一价盐离子(例如钠离子、氯离子)和/或二价盐离子(例如镁离子、硫酸根离子);The method of any one of claims 21 to 23, wherein the solution phase in the step (2) comprises a monovalent salt ion (e.g., sodium ion, chloride ion) and/or a divalent salt ion (e.g., magnesium ion, sulfate ion) ;
    优选地,所述一价盐离子或二价盐离子在所述溶液相中的浓度为1-200mM;Preferably, the concentration of the monovalent salt ion or divalent salt ion in the solution phase is 1-200 mM;
    优选地,所述步骤(2)中的溶液相包含缓冲溶液,例如Tris缓冲溶液;Preferably, the solution phase in the step (2) comprises a buffer solution, such as a Tris buffer solution;
    优选地,Tris在所述溶液相中的浓度为10mM-200mM;Preferably, the concentration of Tris in the solution phase is from 10 mM to 200 mM;
    优选地,所述步骤(2)中的溶液相包含有机溶剂,例如DMSO或丙三醇;Preferably, the solution phase in the step (2) comprises an organic solvent such as DMSO or glycerol;
    优选地,所述有机溶剂在溶液相中的质量含量为0.01%-10%;Preferably, the organic solvent has a mass content in the solution phase of 0.01% to 10%;
    优选地,所述步骤(2)中的溶液相的pH为7.0-9.0;Preferably, the pH of the solution phase in the step (2) is 7.0-9.0;
    优选地,所述步骤(3)中的聚合反应在50-65℃下进行。Preferably, the polymerization in the step (3) is carried out at 50 to 65 °C.
  25. 权利要求21-24任一项的方法,所述方法还包括,在步骤(4)之后,基于碱基互补配对原则,根据步骤(3)中并入生长的核酸链的3'端的化合物的类型,确定待测序的核酸分子中相应位置处的碱基类型。The method according to any one of claims 21 to 24, further comprising, after step (4), based on the principle of base complementary pairing, according to the type of compound incorporated in the 3' end of the growing nucleic acid strand in step (3) Determining the type of base at the corresponding position in the nucleic acid molecule to be sequenced.
  26. 权利要求21-25任一项的方法,所述步骤(5)中,对阻断基团的切除和对可检测标记的切除同时进行,或者,对阻断基团的切除和对可检测标记的切除分步进行;The method according to any one of claims 21 to 25, wherein in the step (5), the excision of the blocking group and the excision of the detectable label are performed simultaneously, or the excision of the blocking group and the detection of the detectable label Excision step by step;
    优选地,所述步骤(5)包括:Preferably, the step (5) comprises:
    步骤(5-1):添加第一切除试剂,使所述双链体或所述生长的核酸链在含有溶液相和固相的反应体系中与第一切除试剂接触,在不影响双链体骨架上的磷酸二酯键的条件下,使并入生长的核酸链3'端的化合物中的磷酸二酯键(1)断裂;Step (5-1): adding a first excision reagent, contacting the duplex or the grown nucleic acid strand with the first excision reagent in a reaction system containing a solution phase and a solid phase without affecting the duplex a phosphodiester bond (1) in a compound incorporated at the 3' end of the growing nucleic acid strand under conditions of a phosphodiester bond on the backbone;
    步骤(5-2):添加第二切除试剂,使所述双链体或所述生长的核酸链在含有溶液相和固相的反应体系中与第而切除试剂接触,在不影响双链体骨架上的磷酸二酯键的条件下,使并入生长的核酸链3'端的化合物中的磷酸二酯键(2)断裂; Step (5-2): adding a second excision reagent, so that the duplex or the grown nucleic acid strand is contacted with the first excision reagent in a reaction system containing a solution phase and a solid phase without affecting the duplex a phosphodiester bond (2) in a compound incorporated at the 3' end of the growing nucleic acid strand under conditions of a phosphodiester bond on the backbone;
    优选地,所述第一切除试剂为内切酶IV;Preferably, the first excision reagent is endonuclease IV;
    优选地,所述第二切除试剂为碱性磷酸酶。Preferably, the second excision reagent is alkaline phosphatase.
  27. 一种试剂盒,其包含第一、第二、第三和第四化合物,所述第一、第二、第三和第四化合物各自为权利要求1-17任一项的化合物,所述四种化合物分别为核苷酸A、(T/U)、C和G的衍生物,且具有碱基互补配对能力;A kit comprising first, second, third and fourth compounds, each of said first, second, third and fourth compounds being a compound according to any one of claims 1-17, said four The compounds are derivatives of nucleotides A, (T/U), C and G, respectively, and have a base complementary pairing ability;
    优选地,所述四种化合物结构中的Label各不相同,例如为不同的荧光基团。Preferably, the Labels in the four compound structures are different, for example, different fluorophores.
  28. 权利要求27的试剂盒,所述试剂盒包含用于进行核苷酸聚合反应的聚合酶;The kit of claim 27, the kit comprising a polymerase for performing a nucleotide polymerization reaction;
    优选地,所述试剂盒包含KOD聚合酶或其突变体;Preferably, the kit comprises KOD polymerase or a mutant thereof;
    优选地,所述突变体选自KOD POL151、KOD POL157、KOD POL171、KOD POL174、KOD POL376、KOD POL391中的一种或多种;Preferably, the mutant is selected from one or more of KOD POL151, KOD POL157, KOD POL171, KOD POL174, KOD POL376, KOD POL391;
    优选地,所述试剂盒包含KOD POL391、KOD POL171或其组合。Preferably, the kit comprises KOD POL391, KOD POL171 or a combination thereof.
  29. 权利要求27或29的试剂盒,所述试剂盒还包含切除试剂,所述切除试剂能够使式(I’)中的磷酸二酯键(1)和/或磷酸二酯键(2)断裂,并且,不会影响双链体骨架上的磷酸二酯键;The kit according to claim 27 or 29, further comprising a excision reagent capable of cleavage of a phosphodiester bond (1) and/or a phosphodiester bond (2) in the formula (I'), Moreover, it does not affect the phosphodiester bond on the duplex backbone;
    优选地,所述切除试剂选自内切酶IV和碱性磷酸酶。Preferably, the excision agent is selected from the group consisting of endonuclease IV and alkaline phosphatase.
  30. 权利要求27-29任一项的试剂盒,所述试剂盒还包含:用于从样品中提取核酸分子的试剂和/或装置;用于预处理核酸分子的试剂;用于连接待测序的核酸分子的支持物;用于将待测序的核酸分子与支持物连接(例如,共价或非共价连接)的试剂;用于起始核苷酸聚合反应的引物;用于进行核苷酸聚合反应的聚合酶;一种或多种缓冲溶液;一种或多种洗涤溶液;或其任何组合。The kit according to any one of claims 27 to 29, further comprising: reagents and/or means for extracting nucleic acid molecules from the sample; reagents for pretreating the nucleic acid molecules; and for connecting nucleic acids to be sequenced a support for a molecule; an agent for linking (eg, covalently or non-covalently linking) a nucleic acid molecule to be sequenced to a support; a primer for initial nucleotide polymerization; for performing nucleotide polymerization The reacted polymerase; one or more buffer solutions; one or more wash solutions; or any combination thereof.
  31. 权利要求27-30任一项的试剂盒,所述试剂盒包含缓冲溶液;A kit according to any one of claims 27 to 30, the kit comprising a buffer solution;
    优选地,所述缓冲溶液包含一价盐离子(例如钠离子、氯离子)和/或二价盐离子(例如镁离子、硫酸根离子);Preferably, the buffer solution comprises monovalent salt ions (such as sodium ions, chloride ions) and/or divalent salt ions (such as magnesium ions, sulfate ions);
    优选地,所述一价盐离子或二价盐离子在所述缓冲溶液中的浓度为1-200mM;Preferably, the concentration of the monovalent salt ion or divalent salt ion in the buffer solution is 1-200 mM;
    优选地,所述缓冲溶液包含Tris; Preferably, the buffer solution comprises Tris;
    优选地,Tris在所述缓冲溶液中的浓度为10mM-200mM;Preferably, the concentration of Tris in the buffer solution is 10 mM to 200 mM;
    优选地,所述缓冲溶液的pH为7.0-9.0;Preferably, the pH of the buffer solution is 7.0-9.0;
    优选地,所述缓冲溶液包含有机溶剂,例如DMSO或丙三醇。Preferably, the buffer solution comprises an organic solvent such as DMSO or glycerol.
  32. 权利要求1-17任一项的化合物,或权利要求27-31任一项的试剂盒用于测定目标单链多核苷酸的序列的用途。 Use of a compound according to any one of claims 1-17, or a kit according to any one of claims 27-31, for determining the sequence of a single-stranded polynucleotide of interest.
PCT/CN2017/105734 2017-10-11 2017-10-11 Modified nucleoside or nucleotide WO2019071474A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
PCT/CN2017/105734 WO2019071474A1 (en) 2017-10-11 2017-10-11 Modified nucleoside or nucleotide
CN201780090915.8A CN110650968B (en) 2017-10-11 2017-10-11 Modified nucleosides or nucleotides

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/CN2017/105734 WO2019071474A1 (en) 2017-10-11 2017-10-11 Modified nucleoside or nucleotide

Publications (1)

Publication Number Publication Date
WO2019071474A1 true WO2019071474A1 (en) 2019-04-18

Family

ID=66100193

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2017/105734 WO2019071474A1 (en) 2017-10-11 2017-10-11 Modified nucleoside or nucleotide

Country Status (2)

Country Link
CN (1) CN110650968B (en)
WO (1) WO2019071474A1 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020259678A1 (en) * 2019-06-26 2020-12-30 南京金斯瑞生物科技有限公司 Oligonucleotide containing blocker
CN113748216A (en) * 2019-05-15 2021-12-03 深圳华大智造极创科技有限公司 Single-channel sequencing method based on self-luminescence
WO2022083686A1 (en) * 2020-10-21 2022-04-28 深圳华大生命科学研究院 Modified nucleoside or nucleotide
US20220298201A1 (en) * 2018-02-21 2022-09-22 Singular Genomics Systems, Inc. Fluorinated nucleotides and uses thereof
WO2022206922A1 (en) * 2021-04-01 2022-10-06 深圳华大生命科学研究院 Nucleotide analogue for sequencing
WO2023280156A1 (en) * 2021-07-08 2023-01-12 深圳华大生命科学研究院 Modified nucleoside or nucleotide

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115820380B (en) * 2023-01-04 2024-01-30 深圳赛陆医疗科技有限公司 Microfluidic chip and preparation method and application thereof

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102030792A (en) * 2009-09-29 2011-04-27 韩国科学技术研究院 3'-o-fluorescently modified nucleotides and uses thereof
WO2012083249A2 (en) * 2010-12-17 2012-06-21 The Trustees Of Columbia University In The City Of New York Dna sequencing by synthesis using modified nucleotides and nanopore detection
US20130085073A1 (en) * 2011-09-29 2013-04-04 Illumina Cambridge Limited Continuous extension and deblocking in reactions for nuclleic acids synthesis and sequencing
WO2016020691A1 (en) * 2014-08-08 2016-02-11 Illumina Cambridge Limited Modified nucleotide linkers

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102030792A (en) * 2009-09-29 2011-04-27 韩国科学技术研究院 3'-o-fluorescently modified nucleotides and uses thereof
WO2012083249A2 (en) * 2010-12-17 2012-06-21 The Trustees Of Columbia University In The City Of New York Dna sequencing by synthesis using modified nucleotides and nanopore detection
US20130085073A1 (en) * 2011-09-29 2013-04-04 Illumina Cambridge Limited Continuous extension and deblocking in reactions for nuclleic acids synthesis and sequencing
WO2016020691A1 (en) * 2014-08-08 2016-02-11 Illumina Cambridge Limited Modified nucleotide linkers

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20220298201A1 (en) * 2018-02-21 2022-09-22 Singular Genomics Systems, Inc. Fluorinated nucleotides and uses thereof
CN113748216A (en) * 2019-05-15 2021-12-03 深圳华大智造极创科技有限公司 Single-channel sequencing method based on self-luminescence
CN113748216B (en) * 2019-05-15 2024-04-23 青岛华大智造科技有限责任公司 Single-channel sequencing method based on self-luminescence
WO2020259678A1 (en) * 2019-06-26 2020-12-30 南京金斯瑞生物科技有限公司 Oligonucleotide containing blocker
WO2022083686A1 (en) * 2020-10-21 2022-04-28 深圳华大生命科学研究院 Modified nucleoside or nucleotide
WO2022206922A1 (en) * 2021-04-01 2022-10-06 深圳华大生命科学研究院 Nucleotide analogue for sequencing
CN115197290A (en) * 2021-04-01 2022-10-18 深圳华大生命科学研究院 Nucleotide analogs for sequencing
CN115197289A (en) * 2021-04-01 2022-10-18 深圳华大生命科学研究院 Nucleotide analogs for sequencing
CN115197291A (en) * 2021-04-01 2022-10-18 深圳华大生命科学研究院 Nucleotide analogs for sequencing
WO2023280156A1 (en) * 2021-07-08 2023-01-12 深圳华大生命科学研究院 Modified nucleoside or nucleotide

Also Published As

Publication number Publication date
CN110650968B (en) 2022-07-05
CN110650968A (en) 2020-01-03

Similar Documents

Publication Publication Date Title
CN110650968B (en) Modified nucleosides or nucleotides
JP7164276B2 (en) Tagmentation using immobilized transposomes with linkers
EP3615671B1 (en) Compositions and methods for improving sample identification in indexed nucleic acid libraries
WO2018121587A1 (en) Single fluorescent dye based sequencing method
EP3570972B1 (en) Methods and reagents for synthesising polynucleotide molecules
KR20160138579A (en) Systems and methods for clonal replication and amplification of nucleic acid molecules for genomic and therapeutic applications
WO2020227953A1 (en) Single-channel sequencing method based on self-luminescence
WO2023168443A1 (en) Double-stranded splint adaptors and methods of use
CN115348896A (en) Nucleic acid molecules comprising cleavable or excisable moieties
WO2022271954A1 (en) Methods and compositions for combinatorial indexing of bead-based nucleic acids
US20230105642A1 (en) Method and compositions for preparing nucleic acid libraries
US20070049745A1 (en) Oligonucleotide synthesis using periodate salts
KR20230124636A (en) Compositions and methods for highly sensitive detection of target sequences in multiplex reactions
KR20230035237A (en) Generation of Nucleic Acids with Modified Bases Using Recombinant Terminal Deoxynucleotidyl Transferases
EP3802876A1 (en) Method of attaching adaptors to single-stranded regions of double-stranded polynucleotides
CN117813390A (en) Method for metal directed cleavage of surface-bound polynucleotides
WO2023235865A1 (en) Compositions and methods for reducing base call errors by removing deaminated nucleotides from a nucleic acid library
WO2023122491A1 (en) Periodate compositions and methods for chemical cleavage of surface-bound polynucleotides
CA3211172A1 (en) Methods of preparing directional tagmentation sequencing libraries using transposon-based technology with unique molecular identifiers for error correction
WO2023122499A1 (en) Periodate compositions and methods for chemical cleavage of surface-bound polynucleotides
WO2024011145A1 (en) Pcr-free library preparation using double-stranded splint adaptors and methods of use
CN117940577A (en) Periodate compositions and methods for chemically cleaving surface-bound polynucleotides
WO2004018691A1 (en) A method for amplification micro-excised chromosomal dna and the use 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: 17928574

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 17928574

Country of ref document: EP

Kind code of ref document: A1