WO2011069677A1 - Amplification spécifique à un allèle d'acides nucléiques - Google Patents

Amplification spécifique à un allèle d'acides nucléiques Download PDF

Info

Publication number
WO2011069677A1
WO2011069677A1 PCT/EP2010/007560 EP2010007560W WO2011069677A1 WO 2011069677 A1 WO2011069677 A1 WO 2011069677A1 EP 2010007560 W EP2010007560 W EP 2010007560W WO 2011069677 A1 WO2011069677 A1 WO 2011069677A1
Authority
WO
WIPO (PCT)
Prior art keywords
oligonucleotide
target sequence
variant
target
nucleic acid
Prior art date
Application number
PCT/EP2010/007560
Other languages
English (en)
Inventor
Alison Tsan
Nicolas Newton
Stephen G. Will
Original Assignee
Roche Diagnostics Gmbh
F. Hoffmann-La Roche Ag
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 Roche Diagnostics Gmbh, F. Hoffmann-La Roche Ag filed Critical Roche Diagnostics Gmbh
Priority to CA2781984A priority Critical patent/CA2781984C/fr
Priority to EP10795240.0A priority patent/EP2510117B1/fr
Priority to JP2012542402A priority patent/JP5886755B2/ja
Priority to ES10795240.0T priority patent/ES2530273T3/es
Priority to CN201080055770.6A priority patent/CN102656278A/zh
Publication of WO2011069677A1 publication Critical patent/WO2011069677A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6844Nucleic acid amplification reactions
    • C12Q1/6858Allele-specific amplification
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • 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
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/156Polymorphic or mutational markers
    • 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
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/166Oligonucleotides used as internal standards, controls or normalisation probes

Definitions

  • the invention relates to the field of nucleic acid amplification and specifically to the field of allele-specific amplification.
  • Allele-specific amplification of nucleic acids allows for simultaneous amplification and analysis of the target sequence. Allele-specific amplification is commonly used when the target nucleic acid has one or more variations (polymorphisms) in its sequence. Nucleic acid polymorphisms are used in DNA profile analysis (forensics, paternity testing, tissue typing for organ transplants), genetic mapping, distinguishing between pathogenic strains of microorganisms as well as detection of rare mutations, such as those occurring in cancer cells, existing in the background of cells with normal DNA. In a successful allele-specific amplification, the desired variant of the target nucleic acid is amplified, while the other variants are not, at least not to a detectable level.
  • a typical allele-specific amplification assay involves a polymerase chain reaction (PCR) with at least one allele-specific primer designed such that primer extension occurs only when the primer forms a hybrid with the desired variant of the target sequence. When the primer hybridizes to an undesired variant of the target sequence, primer extension is inhibited.
  • PCR polymerase chain reaction
  • the invention relates to a method of allele-specific amplification of a variant of a target sequence, the target existing in the form of several variant sequences, the method comprising (a) hybridizing a first and a second oligonucleotides to at least one variant of the target sequence; wherein the first oligonucleotide is at least partially complementary to one or more variants of the target sequence, and the second oligonucleotide is at least partially complementary to one or more variants of the target sequence, and has at least one internal selective nucleotide complementary to only one variant of the target sequence;
  • the invention in a second aspect, relates to a method of detecting a variant of a target sequence, the target existing in the form of several variant sequences, the method comprising
  • said second oligonucleotide is at least partially complementary to one or more variants of the target sequence, and has at least one internal selective nucleotide complementary to only one variant of the target sequence;
  • the invention in a third aspect, relates to a kit for allele-specific amplification of a target sequence, said target existing in the form of several variant sequences, the kit comprising (a) a first oligonucleotide, at least partially complementary to one or more variant of the target sequence; and
  • a second oligonucleotide at least partially complementary to one or more variants of the target sequence having at least one internal selective nucleotide complementary to only one variant of the target sequence.
  • the invention relates to an oligonucleotide for performing an allele- specific amplification of a target sequence, said target existing in the form of several variant sequences, the oligonucleotide comprising
  • the invention relates to a reaction mixture for allele-specific amplification of a target sequence, said target existing in the form of several variant sequences, the mixture comprising
  • a second oligonucleotide at least partially complementary to one or more variants of the target sequence but having at least one internal selective nucleotide complementary to only one variant of the target sequence.
  • Figure 1 shows the results of allele-specific amplification using various nucleic acid polymerases and primers with internal selective nucleotide according to the present invention.
  • Figure 2 shows the results of allele-specific amplification using various nucleic acid polymerases and various primers with 3' selective nucleotide as a control.
  • Figure 3 shows the results of allele-specific amplification using various nucleic acid polymerases and various primers with internal selective nucleotide according to the present invention, including primers having a scorpion ARMS format.
  • Figure 4 shows a schematic representation of the structure of a scorpion ARMS format that can be used according to the invention.
  • nucleic acid refers to polymers of nucleotides (e.g., ribonucleotides,
  • deoxyribonucleotides nucleotide analogs etc.
  • deoxyribonucleic acids DNA
  • RNA ribonucleic acids
  • DNA-RNA hybrids DNA-RNA hybrids
  • oligonucleotides polynucleotides, aptamers, peptide nucleic acids (PNAs), PNA-DNA conjugates, PNA-RNA conjugates, etc., that comprise nucleotides covalently linked together, either in a linear or branched fashion.
  • PNAs peptide nucleic acids
  • a nucleic acid is typically single-stranded or double-stranded and will generally contain phosphodiester bonds, although in some cases, nucleic acid analogs are included that may have alternate backbones, including, for example, phosphoramide (Beaucage et al. ( 1993) Tetrahedron 49( 10):1925); phosphorothioate (Mag et al. ( 1991) Nucleic Acids Res. 19:1437; and U.S. Pat. No. 5,644,048), phosphorodithioate (Briu et al. (1989) J. Am. Chem. Soc. 111:2321 ), O-methylphophoroamidite linkages (see Eckstein, Oligonucleotides and
  • nucleotide analogs also may include non-naturally occurring heterocyclic bases, such as those described in, e.g., Seela et al. (1999) Helv. Chim. Acta 82:1640. Certain bases used in nucleotide analogs act as melting temperature (Tm) modifiers.
  • Tm melting temperature
  • some of these include 7-deazapurines (e.g., 7-deazaguanine, 7-deazaadenine, etc.), pyrazolo[3,4-d]pyrimidines, propynyl-dN (e.g., propynyl-dU, propynyl-dC, etc.), and the like. See, e.g., U.S. Patent No. 5,990,303.
  • heterocyclic bases include, e.g., hypoxanthine, inosine, xanthine; 8-aza derivatives of 2-aminopurine, 2,6-diaminopurine, 2-amino-6-chloropurine, hypoxanthine, inosine and xanthine; 7-deaza-8-aza derivatives of adenine, guanine, 2-aminopurine, 2,6- diaminopurine, 2-amino-6-chloropurine, hypoxanthine, inosine and xanthine; 6- azacytidine; 5-fluorocytidine; 5-chlorocytidine; 5-iodocytidine; 5-bromocytidine; 5- methylcytidine; 5-propynylcytidine; 5-bromovinyluracil; 5-fluorouracil; 5-chlorouracil; 5- iodouracil; 5-bromouracil; 5-trifluor
  • template nucleic acid refers to a nucleic acid of interest to which a primer can hybridize and be extended under suitable conditions.
  • target is preferably a region of nucleic acid, consisting of sequences at least partially complementary to at least two primer sequences, and an intervening sequence. (If the target is a single stranded nucleic acid, it consists of a sequence at least partially complementary to one primer and a sequence at least partially identical to the second primer.)
  • Template nucleic acids can exist as isolated nucleic acid fragments or be a part of a larger nucleic acid fragment.
  • Target nucleic acids can be derived or isolated from essentially any source, such as cultured microorganisms, uncultured microorganisms, complex biological mixtures, tissues, sera, ancient or preserved tissues or samples, environmental isolates or the like. Further, template nucleic acids optionally include or are derived from cDNA, RNA, genomic DNA, cloned genomic DNA, genomic DNA libraries, enzymatically fragmented DNA or RNA, chemically fragmented DNA or RNA, physically fragmented DNA or RNA, or the like. Template nucleic acids can also be chemically synthesized using techniques known in the art.
  • oligonucleotide refers to a nucleic acid polymer that includes at least two, but typically 5-50 nucleotides and more typically, between 15 and 35 nucleotides. Oligonucleotides may be prepared by any suitable method known in the art, including, for example, cloning and restriction digestion of appropriate sequences, or direct chemical synthesis by a method such as the phosphotriester method of Narang et al. (1979) Meth. Enzymol. 68:90-99; the phosphodiester method of Brown et al. (1979) Meth. Enzymol. 68:109-151; the
  • a “primer” is an oligonucleotide that can hybridize to a template nucleic acid and permit chain extension or elongation using a nucleotide polymerase. Although other primer lengths are sometimes utilized, primers typically range from 15 to 35 nucleotides. Short primers generally form sufficiently stable hybrids with template nucleic acids at cooler temperatures. A primer need not be perfectly complementary to the template nucleic acids for the extension to occur. A primer that is at least partially complementary to the template nucleic acid is typically capable of hybridizing with the template nucleic acid for extension to occur.
  • a primer nucleic acid can be labeled, if desired, by incorporating a label detectable by radiological, spectroscopic, photochemical, biochemical, immunochemical, or chemical techniques.
  • an "allele-specific primer” is a primer that can hybridize to several variants of the template nucleic acid, but permit elongation by the polymerase when hybridized with only some of the variants of the template nucleic acid. With other variants of the template nucleic acid the primer-template hybrid may not be extended or is extended less efficiently by the polymerase.
  • Nucleic acids are "extended” or “elongated” when additional nucleotides are incorporated into the nucleic acids, for example by a nucleotide incorporating biocatalyst, at the 3' end of a nucleic acid.
  • An amplification assay is "selective” or “allele-selective” if it yields predominance (i.e., a majority but less than 100%) of one product over other possible products.
  • An assay is described as “allele-selective” as long as amplification of the undesired (mismatched) variant of the target sequence is detectable.
  • the term "specific” or “allele-specific” with respect to amplification assay is used if one of the possible products is formed exclusively.
  • allele-specific An assay where amplification of the undesired target is undetectable is called "allele-specific.”
  • allele-specific is meant to encompass both strictly allele-specific, as well as allele-selective amplification.
  • a “genotype” refers to all or part of the genetic constitution of a cell or subject, or group of cells or subjects.
  • a genotype includes the particular mutations and/or alleles (e.g., polymorphisms, such as single nucleotide polymorphisms (SNPs) or the like) present at a given locus or distributed in a genome.
  • SNPs single nucleotide polymorphisms
  • nucleic acid polymerase refers to an enzyme that catalyzes the incorporation of nucleotides into a nucleic acid.
  • exemplary nucleic acid polymerases include DNA polymerases, NA polymerases, terminal transferases, reverse transcriptases, telomerases and the like.
  • thermoostable enzyme refers to an enzyme that is stable (i.e., resists breakdown or denaturation) and retains sufficient catalytic activity when subjected to elevated
  • thermostable polymerase retains sufficient activity to effect subsequent primer extension reactions, when subjected to elevated temperatures for the time necessary to denature double-stranded nucleic acids. Heating conditions necessary for nucleic acid denaturation are well known in the art and are exemplified in U.S. Pat. Nos. 4,683,202 and 4,683,195.
  • a thermostable polymerase is typically suitable for use in a temperature cycling reaction such as the polymerase chain reaction ("PCR").
  • PCR polymerase chain reaction
  • polymerases include Thermus aquaticus Taq DNA polymerase, Thermus sp. Z05 polymerase, Thermus flavus polymerase, Thermotoga maritima polymerases, such as TMA- 25 and TMA-30 polymerases, Tth DNA polymerase, and the like.
  • a “modified” enzyme refers to an enzyme comprising an amino acid polymer in which at least one monomer differs from the reference sequence, such as a native or wild-type form of the enzyme. Exemplary modifications include monomer insertions, deletions, and substitutions. Modified enzymes also include chimeric enzymes that have identifiable component sequences (e.g., structural or functional domains, etc.) derived from two or more parent enzymes. Also included within the definition of modified enzymes are those comprising chemical modifications of the reference sequence.
  • modified polymerases include G46E E678G CS5 DNA polymerase, G46E L329A E678G CS5 DNA polymerase, G46E L329A D640G S671F CS5 DNA polymerase, G46E L329A D640G S671F E678G CS5 DNA polymerase, a G46E E678G CS6 DNA polymerase, ⁇ 05 polymerase, AZ05-Gold polymerase, AZ05R polymerase, E615G Taq DNA polymerase, E678G TMA-25 polymerase, E678G TMA-30 polymerase, and the like.
  • 5' to 3' nuclease activity or "5'-3' nuclease activity” refers to an activity of a nucleic acid polymerase, typically associated with the nucleic acid strand synthesis, whereby nucleotides are removed from the 5' end of A nucleic acid strand, e.g., E. coli DNA polymerase I has this activity, whereas the Klenow fragment does not.
  • 3' to 5' nuclease activity or "3'-5' nuclease activity” or "proof-reading activity” refers to an activity of a nucleic acid polymerase, whereby nucleotides are removed from the 3' end of the nucleic acid strand.
  • E. coli DNA polymerase III has this activity, whereas the Thermus aquaticus (Taq) DNA polymerase does not.
  • label refers to a moiety attached (covalently or non-covalently), to a molecule and capable of providing information about the molecule.
  • exemplary labels include fluorescent labels, colorimetric labels, chemiluminescent labels, bioluminescent labels, radioactive labels, mass-modifying groups, antibodies, antigens, biotin, haptens, and enzymes
  • a “fidelity” or “replication fidelity” is the ability of a nucleic acid polymerase to incorporate a correct nucleotide during template-dependent polymerization.
  • "correct nucleotide” on the nascent nucleotide strand is the nucleotide paired with the template nucleotide via standard Watson-Crick base pairing.
  • Replication fidelity of a particular polymerase results from a combination of incorporating correct nucleotides and removing incorrect nucleotides from the 3'-terminus of the nascent nucleotide strand via the 3'-5' nuclease activity of the polymerase.
  • a “hot start enzyme” is an enzyme, typically a nucleic acid polymerase, capable of acting as the
  • a “selective nucleotide” is a nucleotide in an allele-specific primer that confers allele selectivity to the primer.
  • the selective nucleotide is complementary to a corresponding nucleotide in the desired variant of the target nucleic acids but not complementary to the corresponding nucleotide in the undesired variants of the target nucleic acid.
  • more than one nucleotide may be complementary to a nucleotide in the desired variants of the target nucleic acids but not complementary to the corresponding nucleotide in the undesired variants of the target nucleic acid.
  • the selective nucleotide is located at a position within the primer that affects the specificity of the primer.
  • the selective nucleotide permits efficient or inefficient amplification of the target nucleic acid, depending on whether or not it finds or does not find a complementary partner in the target nucleic acid.
  • a primer may contain more than one selective nucleotide.
  • Watson-Crick base pairing refers to “conventional” hydrogen bonding within a double-stranded nucleic acid molecule.
  • Watson-Crick base pairing is hydrogen bonding between complementary bases, such as bonding between adenine and thymine, between guanine and cytosine, between adenine and uracil, and between analogs of these bases.
  • corpion scorpion-like or “Scorpion ARMS-like” as used herein denote unimolecular primer-probe combination as described in Whitcombe et al., ( 1999).
  • Scorpion or scorpion-like primers within the meaning of the present invention incorporate the typical elements of the scorpion, namely a probe portion, a stem loop portion and a primer portion.
  • An example of "scorpion" or “scorpion-like" unimolecular primer-probe format is illustrated in Fig. 4.
  • oligonucleotide at least partially complementary to one or more variants of the target sequence, but having at least one internal selective nucleotide complementary to only one variant of the target sequence
  • oligonucleotide preferentially when said selective nucleotide forms a base pair with the target, and substantially less when said selective nucleotide does not form a base pair with the target means that extension of the second oligonucleotide by the polymerase is more efficient when the selective nucleotide forms a base pair with the target, than when said selective nucleotide does not form a base pair with the target.
  • This can for example be measured or quantified with the material and methods described in example 1, the results of which are shown on Figure 1.
  • the present invention teaches a new allele-specific amplification primer, a method of designing such primer, a method of using the primer in allele-specific amplification a reaction mixture and a kit including the primer.
  • the method of designing the primer may be used alone or in conjunction with existing methods of designing allele-specific primers.
  • a typical allele-specific primer is designed to hybridize to a polymorphic region of the target sequence and contain at least one selective nucleotide, i.e. nucleotide complementary to the desired variants of the polymorphic nucleotide in the target and non-complementary to the undesired variants of the target.
  • the present inventors have discovered that internal placement of the selective nucleotide is sufficient to ensure allele-specificity of the primer.
  • the terminal mismatch is not required to confer specificity upon the primer.
  • a sole internal mismatch is sufficient to inhibit extension of the mismatched primer by a nucleotide polymerase.
  • the selective nucleotide is placed internally of the 3'-end of the primer, between 1 and 5 nucleotides internally of the 3'-end.
  • the invention is an oligonucleotide (primer) for use in allele-specific PCR.
  • the primer of the invention comprises 10-50, more preferably 15-35 nucleotides, the majority of them complementary to a sequence in more then one variant of the target sequence.
  • the primer also contains at least one internal selective nucleotide
  • the allele-specific primer further contains one or more nucleotides with chemical modifications that further increase its specificity.
  • modifications at the exocyclic amine of a nucleobase have been described in U.S. Patent No. 6,001,611.
  • the allele specific primer according to the present invention may have a modification at the exocyclic amine of one or more nucleobases.
  • the modified-base nucleotide occurs between 1 and 5, but preferably 3 nucleotides upstream of the 3'-terminal nucleotide. In other embodiments, the modified-base nucleotide is the 3'-terminal nucleotide.
  • the modified-base nucleotide occurs both at the 3'- terminus as well as elsewhere within the oligonucleotide primer. In yet other embodiments, the modification may be placed on the selective nucleotide within the allele-specific primer.
  • a suitable modification of the exocyclic amino group may be selected based on the presence of the following properties: ( 1 ) the modification interferes with, but does not prevent, Watson-Crick base pairing of the modified base with the complementary base in the double-stranded nucleic acid; (2) the modification interferes with but does not prevent the extension of the primer containing the modified base by the nucleic acid polymerase; (3) the modification allows synthesis of the strand complementary to the strand incorporating the modified base; and (4) the modification increases selectivity of a primer incorporating the modification.
  • exocyclic amino groups include the amino groups in the 6-position of adenosine, 2-position of guanosine and 4-position of cytidine.
  • Exocyclic amino groups that take part in base pairing with the complementary nucleic acid strand may also occur in various unconventional nitrogenous bases in nucleotides.
  • nucleosides with unconventional bases include, without limitation, 3-methyladenosine, 7-methylguanosine, 3-methylguanosine, 5-methylcytidine, and 5-hydroxymethylcytidine. Suitable
  • modifications of exocyclic amino groups of such unconventional bases may also be selected according to the empirical method of the present invention.
  • the structures of the modified nucleotides containing a modified adenine, guanine, and cytosine base, respectively, are shown below,
  • modifier groups have the structure:
  • Ri and R 2 are independently selected from the group consisting of hydrogen, alkyl, alkoxy, unsubstituted or substituted aryl and phenoxy.
  • Alkyl groups may be branched or unbranched Alkyl groups can be C]-C 2 o alkyls, in particular Q-Cio alkyls.
  • Alkoxy groups can be Q-C 20 alkoxy, in particular Q-Qo alkoxy.
  • Aryl can be unsubstituted or substituted phenyl or naphtyl.
  • R is a benzyl group or a substituted benzyl group.
  • substituted benzyl groups can have the following structure: wherein R 3 represents a Q-C6 branched or unbranched alkyl group, more preferably a Q- C4 branched or unbranched alkyl group, an alkoxy group, or a nitro group. Preferably, R 3 is attached in the para-position.
  • the modifier groups are represented by structures shown below:
  • suitability of a particular group is determined empirically by using the primers with modified nucleotides in an allele- specific amplification reaction.
  • the suitability of the modification is indicated by the increased selectivity of the reaction utilizing a primer with the base modification, when compared to an identical reaction with an unmodified primer. Additional mismatches between the primer and the template further destabilize the hybrid between the primer and the undesired variant of the target sequence.
  • the method for optimizing the design of the allele-specific primers can be found in Newton et al. ( 1989) supra.
  • the allele-specific primer of the present invention may incorporate various aspects of primer design known in the art.
  • the primer may take the form of a unimolecular primer-probe combination termed "scorpion" and described in Whitcombe et al., (1999) Detection of PCR products using self-probing amplicons and fluorescence, Nature Biotech. 17:804-807.
  • the scorpion primer designed according to the present invention incorporates the typical elements of the scorpion, namely a probe portion, a stem loop portion and a primer portion. Further, in a scorpion designed according to the present invention, the primer portion contains the internally placed selective nucleotide.
  • the 5'-portion and the 3'-portion may contain additional selective nucleotides that are complementary only to the desired version of the target sequence, as long as the 3'-terminal nucleotide of the primer is complementary to both desired and undesired version of the target sequence.
  • Empirical selection of a suitable 5'-portion and 3'-portion of the allele-specific primer can be carried out routinely by one of skill in the art. Specifically, the length, degree of complementarity of the 5'-portion and the 3'-portion and chemical modifications of nucleotides in the 5'-portion and the 3'-portion of the primer can be varied, as long as the primer possesses the four characteristics set forth above.
  • suitability of a particular allele-specific primer is determined empirically by using the primer in an allele- specific amplification. The suitability of the primer is indicated by the selectivity of the amplification utilizing the primer.
  • the present invention is a method of allele-specific amplification of a variant of a target sequence, which exists in the form of several variant sequences, the method comprising: providing a sample, possibly containing at least one variant of a target sequence; providing a first oligonucleotide, at least partially complementary to one or more variants of the target sequence; providing a second oligonucleotide, at least partially complementary to one or more variants of the target sequence, but having at least one internal selective nucleotide complementary to only one variant of the target sequence; providing conditions suitable for the hybridization of said first and second oligonucleotides to at least one variant of the target sequence; providing conditions suitable for the oligonucleotide extension by a nucleic acid polymerase; wherein said polymerase is capable of extending said second oligonucleotide when it is hybridized to the variant of the target sequence for which it has said complementary internal selective nucleotide, and
  • the amplification involves the polymerase chain reaction, i.e. repeated cycles of template denaturation, annealing (hybridization) of the oligonucleotide primer to the template, and extension of the primer by the nucleic acid polymerase. In some embodiments, annealing and extension occur at the same
  • the allele-specific amplification assay is real-time PCR assay.
  • the measure of amplification is the "threshold cycle" or Ct value.
  • the difference in Ct values between the matched and the mismatched templates is a measure of discrimination between the alleles or the selectivity of the assay. A greater difference indicates a greater delay in amplification of the mismatched template and thus a greater discrimination between alleles.
  • the mismatched template is present in much greater amounts than the matched template. For example, in tissue samples, only a small fraction of cells may be malignant and carry the mutation targeted by the allele-specific amplification assay
  • the mismatched template present in normal cells may be amplified less efficiently, but the overwhelming numbers of normal cells will overcome any delay in amplification and erase any advantage of the mutant template.
  • the allele-specific amplification assay of the present invention may employ any suitable nucleic acid polymerase known in the art.
  • any thermostable nucleic acid polymerase may be used.
  • a modified, engineered or chimeric polymerase may also be used.
  • an enzyme without the proofreading (3'-5'-exonuclease) activity such as for example, Taq DNA polymerase. It may also be desirable to use enzymes, substantially or entirely lacking the 5'- 3' nuclease activity, such as described in U.S. Patent No. 5,795,762.
  • One example of such an enzyme is ⁇ 05 polymerase.
  • an enzyme with a "hot start” capability such as the reversibly modified enzymes described in U.S. Patent Nos. 5,677,152 and 5,773,528.
  • a hot-start enzyme is AZ05-Gold polymerase. It is generally known that the specificity of an allele-specific primer may vary somewhat among different enzymes.
  • a special advantage of the allele-specific PCR of the present invention is the ability to use polymerases with proofreading 3'-5'-nuclease activity. Examples of such enzymes can be found in US 7,148,049. Such enzymes comprise for example Thermatoga Maritima. These enzymes typically have higher fidelity (i.e.
  • error rate for Taq DNA polymerase (which does not have a proofreading function) is about 10 "4 .
  • the allele- specific primer has a selective nucleotide not at the 3'-end but internally.
  • the internal mismatch is an inefficient substrate for the exonuclease activity of a proofreading enzyme. It has been observed that the ability of the exonuclease to remove mismatched nucleotides drops dramatically went the mismatch is located away from the 3'-end.
  • the primer with an internal selective nucleotide of the present invention may be used with a proofreading nucleotide polymerase.
  • a person of skill in the art would recognize how to optimize reaction conditions, for example by changing the composition of the reaction buffer and concentration of nucleic acid precursors in order to minimize exonuclease activity of the enzyme without compromising allele-specific amplification. See e.g. Goodman et al. ( 1993) Biochemical basis of DNA replication fidelity, Crit. Rev. Biochem. ol. Biol. 28:83-126 for conditions favoring polymerization and conditions favoring nuclease digestion activities of various nucleic acid polymerases.
  • the amplification products may be detected by any technique known in the art, including but not limited to the use of labeled primers and probes as well as various nucleic acid-binding dyes.
  • the means of detection may be specific to one variant of the target sequence, or may be generic to all variants of the target sequence or even to all double-stranded DNA.
  • the amplification products may be detected after the amplification has been completed, for example, by gel electrophoresis of unlabeled products and staining of the gel with a nucleic acid-binding dye.
  • the amplification products may carry a radioactive or a chemical label, either by virtue of incorporation during synthesis or by virtue of being the extension products of a labeled primer.
  • the labeled amplification products may be detected with suitable radiological or chemical tools known in the art.
  • the products may also be detected with a target-specific probe labeled by any one of the methods known in the art.
  • the labeled probe may also be applied to the target without electrophoresis, i.e. in a "dot blot" assay or the like.
  • the presence of the amplification product may be detected in a homogeneous assay, i.e. an assay where the nascent product is detected during the cycles of amplification, or at least in the same unopened tube, and no post-amplification handling is required.
  • a homogeneous assay has been described for example, in U.S.
  • Homogeneous amplification assay using nucleic acid-intercalating dyes has been described for example, in U.S. Patent Nos. 5,871,908 and 6,569,627.
  • the homogeneous assay may also employ fluorescent probes labeled with two interacting fluorophores, such as "molecular beacon" probes (Tyagi et al., ( 1996) Nat. Biotechnol., 14:303-308) or fluorescently labeled nuclease probes (Livak et al, (1995) PCR Meth. Appl., 4:357-362).
  • the amplification products may also be detected using a unimolecular primer- probe combination termed "scorpion.” Whitcombe et al., (1999) Detection of PCR products using self-probing amplicons and fluorescence, Nature Biotech. 17:804-807.
  • the primer portion of the scorpion oligonucleotide may be an allele-specific primer designed according to the present invention.
  • the amplification product may also be identified by virtue of its distinctive melting temperature, see U.S. Patent Nos.
  • the invention provides a reaction mixture for selectively amplifying the desired variant of the target sequence, the target sequence existing in the form of several variant sequences, the mixture comprising a first oligonucleotide, at least partially complementary to one or more variants of the target sequence; and a second oligonucleotide, at least partially complementary to one or more variants of the target sequence, but having at least one internal selective nucleotide complementary to only one variant of the target sequence.
  • the reaction mixture may also contain a nucleic acid polymerase which is capable of extending said second oligonucleotide when it is hybridized to the variant of the target sequence for which it has said complementary internal selective nucleotide, and substantially less when said second oligonucleotide is hybridized to the variant of the target sequence for which it has a non-complementary internal selective nucleotide.
  • the reaction mixture further comprises the reagents and solutions generally necessary for the amplification of nucleic acids, including nucleic acid precursors, i.e. nucleoside triphosphates, and organic and inorganic ions, suitable for the support of the activity of the nucleotide polymerase.
  • the invention provides kits for conducting allele-specific amplification according to the invention.
  • the kit generally includes assay-specific components as well as components generally required for performing nucleic acid amplification.
  • the allele-specific amplification kit of the present invention contains a first oligonucleotide, at least partially complementary to one or more variants of the target sequence; a second oligonucleotide, at least partially
  • control nucleic acid sequence comprising an amount of at least one variant of the target sequence, at least partially complementary to the oligonucleotides enclosed in the kit. In some embodiments, more than one variant of the control nucleic acid sequence may be enclosed.
  • the kit of the present invention may include one or more of a nucleic acid polymerase, nucleic acid precursors, such as nucleoside triphosphates deoxy-ribonucleoside triphosphates or ribonucleoside triphosphates, a pyrophosphatase, for minimizing pyrophosphorolysis of nucleic acids, a uracil N-glycosylase (UNG) for protection against carry-over contamination of amplification reactions, pre-made reagents and buffers necessary for the amplification reaction and detection, and a set of instructions for conducting allele-specific amplification according to the present invention.
  • nucleic acid polymerase such as nucleoside triphosphates deoxy-ribonucleoside triphosphates or ribonucleoside triphosphates
  • a pyrophosphatase for minimizing pyrophosphorolysis of nucleic acids
  • UNG uracil N-glycosylase
  • two variants of the template sequence were used: a matched variant, with a sequence complementary to the selective nucleotide in the allele-specific primer and a mismatched variant, with a sequence non-complementary to the selective nucleotide in the allele-specific primer.
  • the examples utilized the V600E mutation of the human BRAF gene (GeneBank reference).
  • the matched variant was a plasmid DNA with the insert incorporating the BRAF V600E mutant sequence (SEQ ID NO: 19), while the mismatched variant was the same plasmid with the BRAF wild-type sequence (SEQ ID NO: 20).
  • SEQ ID NO: 19 (BRAF V600E mutant sequence fragment)
  • SEQ ID NO: 20 (BRAF wild-type sequence fragment) 5'-AGTAAAAATAGGTGATTTTGGTCTAGCTACAGTGAAATCTCGATGGAGTGGG TCCCATCAGTTTGAACAGTTGTCTGGATCCATTTTGTGGATGGTAAGAATTGA GGCTA-3'
  • T thymine
  • A adenine
  • the mutation is found in many cancers and is thought to contribute to cancer progression, as it results in constitutive activation of the MAPK pathway. Detection of this single nucleotide change in a population of tumor cells has utility in the diagnosis and treatment of human cancers.
  • the mutant target is "matched", i.e. forms an A-T Watson-Crick pair with the selective nucleotide of each of the allele-specific primers (Table A).
  • the mismatched target is the wild-type BRAF sequence.
  • the mismatched target forms a mismatch with the selective nucleotide of each of the allele-specific primers.
  • two variants of the template sequence were used: a matched variant, with a sequence complementary to the selective nucleotide in the allele-specific primer and a mismatched variant, with a sequence non-complementary to the selective nucleotide in the allele-specific primer.
  • the matched variant was a plasmid DNA with the insert representing the BRAF sequence with a V600E mutation.
  • the mismatched variant was the same plasmid with the BRAF wild-type sequence.
  • the forward primers (SEQ ID NO: 1-4) and reverse primer (SEQ ID NO: 11) are shown in Table A.
  • the forward allele-specific primers were designed with the selective nucleotide internal of the 3' terminus, at the N-2 position. Some primers contained chemical modifications where indicated.
  • Each 50 ⁇ reaction contained 10 5 copies of either target, 8% glycerol, 50 mM tricine (pH 7.7), 45 mM potassium acetate (pH 7.5), 200 ⁇ each dATP, dCTP and dGTP, 400 ⁇ dUTP, 0.1 ⁇ forward primer, 0.7 ⁇ reverse primer, 2 ⁇ Syto-13 intercalating dye, 1% DMSO, 2 units of uracil-N-glycosylase (UNG), 50 units of AZ05-Gold DNA polymerase, and 3 mM mangnesium acetate. Amplification and analysis were done using the Roche LightCycler 480 instrument.
  • the reactions were subjected to the following temperature profile: 50°C for 5 minutes (UNG step), 95°C for 10 minutes, followed by 60-70 cycles of 95°C for 15 seconds and 59°C for 40 seconds. Fluorescence data was collected at 495- 525nm at the end of each 59°C step.
  • Example 2 Allele-specific amplification using primers with an internal selective nucleotide and different nucleic acid polymerases
  • Example 2 the same matched (mutant) and mismatched (wild-type) targets as in Example 1 were amplified using primers shown in Table A. Amplification was carried out in the presence of Z05, ⁇ 05, or AZ05-Gold polymerase. All reactions were done in triplicate, in 15 ⁇ , volumes containing 10 5 copies of either target, 200 ⁇ each dATP, dCTP and dGTP, 400 ⁇ dUTP, 0.1 ⁇ forward primer, 0.7 ⁇
  • Z05 reactions contained 3U of polymerase, 130 mM potassium acetate (pH 7.5), 5% glycerol, and 50 mM Tricine (pH 8.3).
  • ⁇ 05 reactions contained 3U of polymerase, 25 mM potassium acetate (pH 7.5), 5% glycerol, and 50 mM Tricine (pH 8.3).
  • AZ05-Gold reactions contained 15U of polymerase, 45 mM potassium acetate (pH 7.5), 8% glycerol, and 50 mM Tricine (pH 7.7).
  • Amplification and analysis were done using the Roche LightCycler 480 instrument. The reactions were subjected to the following temperature profile: 50°C for 5 minutes (UNG step), 95°C for 10 minutes, followed by 99 cycles of 95°C for 15 seconds and 59°C for 40 seconds. Fluorescence data was collected at 465-5 lOnm at the end of each 59°C step.
  • position N denotes the nucleotide position relative to the 3' end.
  • two variants of the template sequence were present in equal amounts, a matched variant, complementary to the primer sequence and a mismatched variant.
  • the matched variant was a plasmid DNA with the insert representing the BRAF V600E mutant sequence (SEQ ID NO: 1), while the mismatched variant was the same plasmid with the BRAF wild-type sequence (SEQ ID NO: 2).
  • the forward primers (SEQ ID NO: 6, 9, 12- 17) and reverse primer (SEQ ID NO: 11) are as described in Table 3.
  • the forward, ASPCR primers were designed with the SNP at, or near the 3' terminal position, either with or without N6-tert-butyl-benzyl-dA modification(s).
  • Amplification and analysis were done using the Roche LightCycler 480 instrument. The reactions were subjected to the following temperature profile: 50°C for 5 minutes (UNG step) followed by 95 cycles of 95°C for 15 seconds and 59°C for 40 seconds. Fluorescence data was collected at the 495-525nm range at the end of each 59°C anneal/ extend step.
  • the results are shown on Figure 2 and Table 4.
  • the selectivity of the amplification is measured by the difference in the Ct value (ACt) between the matched and the mismatched targets. ACt for each experiment is indicated on each diagram and summarized in Table 4.
  • the data shows that the matched (mutant) variant of the target was amplified selectively over the mismatched (wild-type) variant using either the individual primer and probe or the primer and probe linked in a closed Scorpion ARMS-like format. Discrimination was achieved whether the selective nucleotide was at the 3' terminus or internal. Additionally, the selectivity of amplification was enhanced by addition of one or more alkyl modifications.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Engineering & Computer Science (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Immunology (AREA)
  • Genetics & Genomics (AREA)
  • Analytical Chemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biotechnology (AREA)
  • Microbiology (AREA)
  • Molecular Biology (AREA)
  • General Health & Medical Sciences (AREA)
  • Physics & Mathematics (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Pathology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Oncology (AREA)
  • Hospice & Palliative Care (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Apparatus Associated With Microorganisms And Enzymes (AREA)

Abstract

La présente invention concerne un procédé d'amplification spécifique à un allèle, utilisant un oligonucléotide spécifique à un allèle, au moins partiellement complémentaire de plus d'une variante de la séquence cible, contenant un nucléotide sélectif placé en interne complémentaire d'une seule variante de la séquence cible, l'oligonucléotide spécifique à l'allèle étant étendu par une polymérase d'acide nucléique principalement ou exclusivement lorsqu'il est hybridé avec la variante de la séquence cible pour laquelle il contient ledit nucléotide sélectif complémentaire.
PCT/EP2010/007560 2009-12-11 2010-12-10 Amplification spécifique à un allèle d'acides nucléiques WO2011069677A1 (fr)

Priority Applications (5)

Application Number Priority Date Filing Date Title
CA2781984A CA2781984C (fr) 2009-12-11 2010-12-10 Amplification specifique a un allele d'acides nucleiques
EP10795240.0A EP2510117B1 (fr) 2009-12-11 2010-12-10 Amplification spécifique à un allèle d'acides nucléiques
JP2012542402A JP5886755B2 (ja) 2009-12-11 2010-12-10 核酸の対立遺伝子特異的増幅
ES10795240.0T ES2530273T3 (es) 2009-12-11 2010-12-10 Amplificación de ácidos nucleicos específica de alelo
CN201080055770.6A CN102656278A (zh) 2009-12-11 2010-12-10 核酸的等位基因特异性扩增

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US28569009P 2009-12-11 2009-12-11
US61/285,690 2009-12-11

Publications (1)

Publication Number Publication Date
WO2011069677A1 true WO2011069677A1 (fr) 2011-06-16

Family

ID=43531218

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2010/007560 WO2011069677A1 (fr) 2009-12-11 2010-12-10 Amplification spécifique à un allèle d'acides nucléiques

Country Status (7)

Country Link
US (1) US9238832B2 (fr)
EP (1) EP2510117B1 (fr)
JP (2) JP5886755B2 (fr)
CN (2) CN102656278A (fr)
CA (1) CA2781984C (fr)
ES (1) ES2530273T3 (fr)
WO (1) WO2011069677A1 (fr)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013022807A1 (fr) * 2011-08-05 2013-02-14 Unitaq Bio Procédés et compositions pour détection d'acides nucléiques à l'aide de clivage par 5'-nucléase et d'amplification
WO2014090837A1 (fr) * 2012-12-13 2014-06-19 Roche Diagnostics Gmbh Amorces à phosphate modifié et base pour pcr spécifique d'allèles
JP2014532434A (ja) * 2011-11-10 2014-12-08 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffma 上皮増殖因子受容体キナーゼ・ドメイン内の新規な複合突然変異
JP2016510982A (ja) * 2013-03-13 2016-04-14 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft ヒトpi3kca(pik3ca)遺伝子変異検出のための方法及び組成物

Families Citing this family (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012142213A2 (fr) 2011-04-15 2012-10-18 The Johns Hopkins University Système de séquençage sûr
BR112014006419A2 (pt) 2011-09-19 2018-08-07 Genentech Inc métodos para tratar um paciente com câncer, kit e artigo
EP3447495B2 (fr) 2012-10-29 2024-03-13 The Johns Hopkins University Test de papanicolaou pour cancers de l'endomètre et de l'ovaire
CN103911445B (zh) * 2014-03-20 2015-08-12 上海科亦生物科技有限公司 一种as-pcr引物设计方法、基因多态性检测方法及试剂盒
CN104845967B (zh) * 2015-04-15 2020-12-11 苏州新海生物科技股份有限公司 寡聚核苷酸片段及使用其的选择性扩增目标核酸序列变异体的方法及应用
US11286531B2 (en) 2015-08-11 2022-03-29 The Johns Hopkins University Assaying ovarian cyst fluid
CN116949140A (zh) * 2016-02-17 2023-10-27 哈佛学院院长及董事 分子编程工具
JP2018164421A (ja) * 2017-03-28 2018-10-25 東洋紡株式会社 核酸増幅方法
MX2020001575A (es) 2017-08-07 2020-11-18 Univ Johns Hopkins Materiales y métodos para evaluar y tratar el cáncer.
WO2019099420A1 (fr) * 2017-11-15 2019-05-23 Yan Wang Procédé de détection de multiples mutations d'adn et de variations du nombre de copies

Citations (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4458066A (en) 1980-02-29 1984-07-03 University Patents, Inc. Process for preparing polynucleotides
US4683202A (en) 1985-03-28 1987-07-28 Cetus Corporation Process for amplifying nucleic acid sequences
US4683195A (en) 1986-01-30 1987-07-28 Cetus Corporation Process for amplifying, detecting, and/or-cloning nucleic acid sequences
US5210015A (en) 1990-08-06 1993-05-11 Hoffman-La Roche Inc. Homogeneous assay system using the nuclease activity of a nucleic acid polymerase
US5338671A (en) 1992-10-07 1994-08-16 Eastman Kodak Company DNA amplification with thermostable DNA polymerase and polymerase inhibiting antibody
US5411876A (en) 1990-02-16 1995-05-02 Hoffmann-La Roche Inc. Use of grease or wax in the polymerase chain reaction
US5639611A (en) 1988-12-12 1997-06-17 City Of Hope Allele specific polymerase chain reaction
US5644048A (en) 1992-01-10 1997-07-01 Isis Pharmaceuticals, Inc. Process for preparing phosphorothioate oligonucleotides
US5677152A (en) 1995-08-25 1997-10-14 Roche Molecular Systems, Inc. Nucleic acid amplification using a reersibly inactivated thermostable enzyme
US5773528A (en) 1990-10-31 1998-06-30 International Business Machines Corporation Dual cure epoxy backseal formulation
US5795762A (en) 1986-08-22 1998-08-18 Roche Molecular Systems, Inc. 5' to 3' exonuclease mutations of thermostable DNA polymerases
US5840867A (en) 1991-02-21 1998-11-24 Gilead Sciences, Inc. Aptamer analogs specific for biomolecules
US5871908A (en) 1992-02-05 1999-02-16 Evotec Biosystems Gmbh Process for the determination of in vitro amplified nucleic acids
US5990303A (en) 1985-08-16 1999-11-23 Roche Diagnostics Gmbh Synthesis of 7-deaza-2'deoxyguanosine nucleotides
US6001611A (en) 1997-03-20 1999-12-14 Roche Molecular Systems, Inc. Modified nucleic acid amplification primers
US6569627B2 (en) 1996-06-04 2003-05-27 University Of Utah Research Foundation Monitoring hybridization during PCR using SYBR™ Green I
WO2003072814A2 (fr) * 2002-02-26 2003-09-04 Roche Diagnostics Gmbh Amelioration de la methode de pcr specifique d'allele
US20060246476A1 (en) * 2005-01-24 2006-11-02 David Polsky Methods for detecting circulating mutant BRAF DNA
US7148049B2 (en) 2002-04-02 2006-12-12 Roche Molecular Systems, Inc. Thermostable or thermoactive DNA polymerase molecules with attenuated 3′-5′ exonuclease activity
US20070117118A1 (en) * 2005-11-21 2007-05-24 Hidenobu Yaku Discrimination method of target base in DNA, and allele specific primer used in the method of the same
WO2010046067A1 (fr) * 2008-10-20 2010-04-29 Roche Diagnostics Gmbh Amplification allèle-spécifique améliorée

Family Cites Families (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IE61148B1 (en) 1988-03-10 1994-10-05 Ici Plc Method of detecting nucleotide sequences
US5521301A (en) 1988-12-12 1996-05-28 City Of Hope Genotyping of multiple allele systems
US5137806A (en) 1989-12-11 1992-08-11 Board Of Regents, The University Of Texas System Methods and compositions for the detection of sequences in selected DNA molecules
DE69813203T2 (de) 1998-07-21 2004-02-12 Keygene N.V. Verbesserte Primer zur AFLP Amplifizierung
US6200757B1 (en) 1999-01-19 2001-03-13 Dade Behring Inc. Method for controlling the extension of an oligonucleotide
EP1088891B1 (fr) * 1999-09-28 2005-01-12 Roche Diagnostics GmbH Enzyme thermostable pour augmenter la fidélité de polymèrase d'ADN thermostable - pour l'amélioration de la synthèse des acides nucléiques et d'amplification in vitro
EP1241266A4 (fr) 1999-12-10 2004-11-17 Toyo Boseki Procede de detection de polymorphisme nucleotidique
US7582420B2 (en) * 2001-07-12 2009-09-01 Illumina, Inc. Multiplex nucleic acid reactions
AU2002232177B2 (en) 2001-02-15 2006-11-09 Takara Bio Inc. Method of detecting nucleotide polymorphism
US20050019918A1 (en) * 2003-06-03 2005-01-27 Hidetoshi Sumimoto Treatment of cancer by inhibiting BRAF expression
US7408051B2 (en) 2004-04-14 2008-08-05 Applera Corporation Modified oligonucleotides and applications thereof
WO2006047484A2 (fr) * 2004-10-22 2006-05-04 Redpath Integrated Pathology, Inc. Analyse moleculaire de fluides cellulaires et d'echantillons cytologiques liquides pour diagnostic clinique, caracterisation, et integration a l'evaluation des pathologiques microscopiques
US8349556B2 (en) 2006-04-28 2013-01-08 Igor Kutyavin Use of base-modified deoxynucleoside triphosphates to improve nucleic acid detection
JP2009077712A (ja) * 2007-09-11 2009-04-16 F Hoffmann La Roche Ag B−Rafキナーゼ阻害剤に対する感受性についての診断試験
EP2418515A1 (fr) 2010-07-21 2012-02-15 Astrium GmbH Procédé d'intégrité pour corrections différentielles

Patent Citations (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4458066A (en) 1980-02-29 1984-07-03 University Patents, Inc. Process for preparing polynucleotides
US4683202A (en) 1985-03-28 1987-07-28 Cetus Corporation Process for amplifying nucleic acid sequences
US4683202B1 (fr) 1985-03-28 1990-11-27 Cetus Corp
US5990303A (en) 1985-08-16 1999-11-23 Roche Diagnostics Gmbh Synthesis of 7-deaza-2'deoxyguanosine nucleotides
US4683195A (en) 1986-01-30 1987-07-28 Cetus Corporation Process for amplifying, detecting, and/or-cloning nucleic acid sequences
US4683195B1 (fr) 1986-01-30 1990-11-27 Cetus Corp
US5795762A (en) 1986-08-22 1998-08-18 Roche Molecular Systems, Inc. 5' to 3' exonuclease mutations of thermostable DNA polymerases
US5639611A (en) 1988-12-12 1997-06-17 City Of Hope Allele specific polymerase chain reaction
US5411876A (en) 1990-02-16 1995-05-02 Hoffmann-La Roche Inc. Use of grease or wax in the polymerase chain reaction
US5210015A (en) 1990-08-06 1993-05-11 Hoffman-La Roche Inc. Homogeneous assay system using the nuclease activity of a nucleic acid polymerase
US5773528A (en) 1990-10-31 1998-06-30 International Business Machines Corporation Dual cure epoxy backseal formulation
US5840867A (en) 1991-02-21 1998-11-24 Gilead Sciences, Inc. Aptamer analogs specific for biomolecules
US5644048A (en) 1992-01-10 1997-07-01 Isis Pharmaceuticals, Inc. Process for preparing phosphorothioate oligonucleotides
US5871908A (en) 1992-02-05 1999-02-16 Evotec Biosystems Gmbh Process for the determination of in vitro amplified nucleic acids
US5338671A (en) 1992-10-07 1994-08-16 Eastman Kodak Company DNA amplification with thermostable DNA polymerase and polymerase inhibiting antibody
US5677152A (en) 1995-08-25 1997-10-14 Roche Molecular Systems, Inc. Nucleic acid amplification using a reersibly inactivated thermostable enzyme
US6569627B2 (en) 1996-06-04 2003-05-27 University Of Utah Research Foundation Monitoring hybridization during PCR using SYBR™ Green I
US6001611A (en) 1997-03-20 1999-12-14 Roche Molecular Systems, Inc. Modified nucleic acid amplification primers
WO2003072814A2 (fr) * 2002-02-26 2003-09-04 Roche Diagnostics Gmbh Amelioration de la methode de pcr specifique d'allele
US7148049B2 (en) 2002-04-02 2006-12-12 Roche Molecular Systems, Inc. Thermostable or thermoactive DNA polymerase molecules with attenuated 3′-5′ exonuclease activity
US20060246476A1 (en) * 2005-01-24 2006-11-02 David Polsky Methods for detecting circulating mutant BRAF DNA
US20070117118A1 (en) * 2005-11-21 2007-05-24 Hidenobu Yaku Discrimination method of target base in DNA, and allele specific primer used in the method of the same
WO2010046067A1 (fr) * 2008-10-20 2010-04-29 Roche Diagnostics Gmbh Amplification allèle-spécifique améliorée

Non-Patent Citations (20)

* Cited by examiner, † Cited by third party
Title
ANDRE ET AL.: "Fidelity and mutational spectrum of Pfu DNA polymerase on a human mitochondrial DNA sequence", GENOME RES., vol. 7, 1997, pages 843 - 852
BEAUCAGE ET AL., TETRAHEDRON LETT., vol. 22, 1981, pages 1859 - 1862
BEAUCAGE ET AL., TETRAHEDRON, vol. 49, no. 10, 1993, pages 1925
BRIU ET AL., J. AM. CHEM. SOC., vol. 111, 1989, pages 2321
BROWN ET AL., METH. ENZYMOL., vol. 68, 1979, pages 109 - 151
ECKSTEIN: "Oligonucleotides and Analogues: a Practical Approach", 1992, OXFORD UNIVERSITY PRESS
EGHOLM, J. AM. CHEM. SOC., vol. 114, 1992, pages 1895
FIDALGO-DA SILVA ET AL.: "DNA polymerase proofreading: active site switching catalyzed by the bacteriophage T4 DNA polymerase", NUCL. ACIDS RES., vol. 35, 2007, pages 5452 - 5463
GOODMAN ET AL.: "Biochemical basis of DNA replication fidelity", CRIT. REV. BIOCHEM. MOL. BIOL., vol. 28, 1993, pages 83 - 126
LIVAK ET AL., PCR METH. APPL., vol. 4, 1995, pages 357 - 362
MAG ET AL., NUCLEIC ACIDS RES., vol. 19, 1991, pages 1437
MATTEUCCI ET AL., J. AM. CHEM. SOC., vol. 103, 1981, pages 3185 - 3191
NARANG ET AL., METH. ENZYMOL., vol. 68, 1979, pages 90 - 99
NEWTON ET AL.: "Analysis of any point mutation in DNA. The amplification refractory mutation system (ARMS)", NUCL. ACIDS RES., vol. 17, 1989, pages 2503 - 2516
REDDY ET AL.: "Processive proofreading is intrinsic to T4 DNA polymerase", J. BIOL. CHEM., vol. 267, 1992, pages 14157 - 14166
SEELA ET AL., HELV. CHIM. ACTA, vol. 82, 1999, pages 1640
TINDALL ET AL.: "Fidelity of DNA synthesis by the Thermus aquaticus DNA polymerase", BIOCHEMISTRY, vol. 27, 1988, pages 6008 - 6013
TYAGI ET AL., NAT. BIOTECHNOL., vol. 14, 1996, pages 303 - 308
WHITCOMBE D ET AL: "Detection of PCR products using self-probing amplicons and fluorescence", NATURE BIOTECHNOLOGY, NATURE PUBLISHING GROUP, NEW YORK, NY, US, vol. 17, no. 8, 1 August 1999 (1999-08-01), pages 804 - 807, XP002226672, ISSN: 1087-0156, DOI: DOI:10.1038/11751 *
WHITCOMBE ET AL.: "Detection of PCR products using self-probing amplicons and fluorescence", NATURE BIOTECH., vol. 17, 1999, pages 804 - 807

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013022807A1 (fr) * 2011-08-05 2013-02-14 Unitaq Bio Procédés et compositions pour détection d'acides nucléiques à l'aide de clivage par 5'-nucléase et d'amplification
JP2014532434A (ja) * 2011-11-10 2014-12-08 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffma 上皮増殖因子受容体キナーゼ・ドメイン内の新規な複合突然変異
US9738935B2 (en) 2011-11-10 2017-08-22 Roche Molecular Systems, Inc. Complex mutations in the epidermal growth factor receptor kinase domain
WO2014090837A1 (fr) * 2012-12-13 2014-06-19 Roche Diagnostics Gmbh Amorces à phosphate modifié et base pour pcr spécifique d'allèles
US9382581B2 (en) 2012-12-13 2016-07-05 Roche Molecular Systems, Inc. Primers with modified phosphate and base in allele-specific PCR
JP2016510982A (ja) * 2013-03-13 2016-04-14 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft ヒトpi3kca(pik3ca)遺伝子変異検出のための方法及び組成物

Also Published As

Publication number Publication date
EP2510117B1 (fr) 2014-12-10
US20110311968A1 (en) 2011-12-22
CN102656278A (zh) 2012-09-05
CA2781984C (fr) 2015-02-17
JP2016013137A (ja) 2016-01-28
CN106987624A (zh) 2017-07-28
JP5886755B2 (ja) 2016-03-16
CA2781984A1 (fr) 2011-06-16
US9238832B2 (en) 2016-01-19
JP2013513360A (ja) 2013-04-22
ES2530273T3 (es) 2015-02-27
EP2510117A1 (fr) 2012-10-17

Similar Documents

Publication Publication Date Title
CA2781984C (fr) Amplification specifique a un allele d'acides nucleiques
US9506111B2 (en) Allele-specific amplification
EP2931917B1 (fr) Amorces à phosphate modifié et base pour pcr spécifique d'allèles
US20160369330A1 (en) Compositions for improved allele-specific pcr
EP2794916A1 (fr) Procédés et réactifs pour la réduction d'une amplification non spécifique
US8614071B2 (en) Preferential amplification of mRNA over DNA using chemically modified primers
US9909169B2 (en) Allele-specific amplification of nucleic acids using blocking oligonucleotides for wild type suppression

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 201080055770.6

Country of ref document: CN

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

Ref document number: 10795240

Country of ref document: EP

Kind code of ref document: A1

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

Ref document number: 2010795240

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2781984

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2012542402

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE