WO2001009386A2 - Digital amplification - Google Patents

Digital amplification Download PDF

Info

Publication number
WO2001009386A2
WO2001009386A2 PCT/US2000/020740 US0020740W WO0109386A2 WO 2001009386 A2 WO2001009386 A2 WO 2001009386A2 US 0020740 W US0020740 W US 0020740W WO 0109386 A2 WO0109386 A2 WO 0109386A2
Authority
WO
WIPO (PCT)
Prior art keywords
genetic sequence
assay samples
sequence
selected genetic
molecular beacon
Prior art date
Application number
PCT/US2000/020740
Other languages
French (fr)
Other versions
WO2001009386A3 (en
Inventor
Kenneth W. Kinzler
Bert Vogelstein
Original Assignee
The Johns Hopkins University
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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=26844308&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO2001009386(A2) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by The Johns Hopkins University filed Critical The Johns Hopkins University
Priority to JP2001513641A priority Critical patent/JP4653366B2/en
Priority to CA2376929A priority patent/CA2376929C/en
Priority to AU65028/00A priority patent/AU781440B2/en
Priority to EP00952304A priority patent/EP1255856B1/en
Priority to DE60018616T priority patent/DE60018616T2/en
Priority to AT00952304T priority patent/ATE290608T1/en
Publication of WO2001009386A2 publication Critical patent/WO2001009386A2/en
Publication of WO2001009386A3 publication Critical patent/WO2001009386A3/en

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
    • 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
    • C12Q1/6813Hybridisation assays
    • C12Q1/6816Hybridisation assays characterised by the detection means
    • C12Q1/6818Hybridisation assays characterised by the detection means involving interaction of two or more labels, e.g. resonant energy transfer
    • 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
    • C12Q1/6851Quantitative 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
    • C12Q1/6844Nucleic acid amplification reactions
    • C12Q1/686Polymerase chain reaction [PCR]
    • 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
    • C12Q1/6869Methods for sequencing
    • C12Q1/6874Methods for sequencing involving nucleic acid arrays, e.g. sequencing by hybridisation
    • 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
    • C12Q2600/158Expression markers

Definitions

  • This invention is related to diagnostic genetic analyses. In particular it relates to detection of genetic changes and gene expression. BACKGROUND OF THE INVENTION
  • Mutant-specific oligonucleotides can sometimes be used to detect mutations present in a minor proportion of the cells analyzed, but the signal to noise ratio distinguishing mutant and wild-type ( WT) templates is variable (20-22)
  • the use of mutant-specific primers or the digestion of polymerase chain reaction (PCR) products with specific restriction endonucleases are extremely sensitive methods for detecting such mutations, but it is difficult to quantitate the fraction of mutant molecules in the starting population with these techniques (23-28).
  • Other innovative approaches for the detection of somatic mutations have been reviewed (29-32). A general problem with these methods is that it is difficult or impossible to independently confirm the existence of any mutations that are identified. Thus there is a need in the art for methods for accurately and quantitatively detecting genetic sequences in mixed populations of sequences.
  • a biological sample comprising nucleic acid template molecules is diluted to form a set of assay samples.
  • the template molecules within the assay samples are amplified to form a population of amplified molecules in the assay samples of the set.
  • the amplified molecules in the assay samples of the set are then analyzed to determine a first number of assay samples which contain the selected genetic sequence and a second number of assay samples which contain a reference genetic sequence. The first number is then compared to the second number to ascertain a ratio which reflects the composition of the biological sample.
  • Another embodiment of the invention is a method for determining the ratio of a selected genetic sequence in a population of genetic sequences.
  • Template molecules within a set comprising a plurality of assay samples are amplified to form a population of amplified molecules in each of the assay samples of the set.
  • the amplified molecules in the assay samples of the set are analyzed to determine a first number of assay samples which contain the selected genetic sequence and a second number of assay samples which contain a reference genetic sequence. The first number is compared to the second number to ascertain a ratio which reflects the composition of the biological sample.
  • a molecular beacon probe comprises an oligonucleotide with a stem-loop structure having a photoluminescent dye at one of the 5 1 or 3' ends and a quenching agent at the opposite 5' or 3 1 end.
  • the loop consists of 16 base pairs and has a T m of 50-51 °C.
  • the stem consists of 4 base pairs having a sequence 5'-CACG-3'.
  • a second type of molecular beacon probe comprises an oligonucleotide with a stem-loop structure having a photoluminescent dye at one of the 5' or 3' ends and a quenching agent at the opposite 5' or 3' end.
  • the loop consists of 19-20 base pairs and has a T m of 54-56°C.
  • the stem consists of 4 base pairs having a sequence 5'- CACG-3'.
  • FIG. 1 Schematic of experimental design. (A) The basic two steps involved:
  • PCR on diluted DNA samples is followed by addition of fluorescent probes which discriminate between WT and mutant alleies and subsequent fluorometry.
  • B Principle of molecular beacon analysis. In the stem-loop configuration, fluorescence from a dye at the 5' end of the oligonucleotide probe is quenched by a Dabcyl group at the 3' end. Upon hybridization to a template, the dye is separated from the quencher, resulting in increased fluorescence. Modified from Marras et al. .
  • C Oligonucleotide design. Primers FI and Rl are used to amplify the genomic region of interest. Primer INT is used to produce single stranded DNA from the original PCR products during a subsequent asymmetric PCR step (see Materials and Methods).
  • MB-RED is a Molecular Beacon which detects any appropriate PCR product, whether it is WT or mutant at the queried codons.
  • MB-GREEN is a Molecular Beacon which preferentially detects the WT PCR product.
  • RED/GREEN ratios were determined from the fluorescence of MB-RED and MB-GREEN as described in Materials and Methods.
  • the wells shown are the same as those illustrated in Fig. 3.
  • the sequences of PCR products from the indicated wells were determined as described in Materials and Methods.
  • the method devised by the present inventors involves separately amplifying small numbers of template molecules so that the resultant products have a proportion of the analyte sequence which is detectable by the detection means chosen.
  • single template molecules can be amplified so that the products are completely mutant or completely wild-type (WT).
  • WT completely wild-type
  • the method requires analyzing a large number of amplified products simply and reliably. Techniques for such assessments were developed, with the output providing a digital readout of the fraction of mutant alleies in the analyzed population.
  • the biological sample is diluted to a point at which a practically usable number of the diluted samples contain a proportion of the selected genetic sequence (analyte) relative to total template molecules such that the analyzing technique being used can detect the analyte.
  • a practically usable number of diluted samples will depend on cost of the analysis method. Typically it would be desirable that at least 1/50 of the diluted samples have a detectable proportion of analyte. At least 1/10, 1/5, 3/10, 2/5, 1/2, 3/5, 7/10, 4/5, or 9/10 of the diluted samples may have a detectable proportion of analyte. The higher the fraction of samples which will provide useful information, the more economical will be the overall assay.
  • a particularly preferred degree of dilution is to a point where each of the assay samples has on average one-half of a template.
  • the dilution can be performed from more concentrated samples. Alternatively, dilute sources of template nucleic acids can be used. All of the samples may contain amplifable template molecules. Desirably each assay sample prior to amplification will contain less than a hundred or less than ten template molecules.
  • Digital amplification can be used to detect mutations present at relatively low levels in the samples to be analyzed. The limit of detection is defined by the number of wells that can be analyzed and the intrinsic mutation rate of the polymerase used for amplification.
  • 384 well PCR plates are commercially available and 1536 well plates are on the horizon, theoretically allowing sensitivities for mutation detection at the -0.1% level. It is also possible that Digital Amplification can be performed in microarray format, potentially increasing the sensitivity by another order of magnitude. This sensitivity may ultimately be limited by polymerase errors.
  • the effective error rate in PCR as performed under our conditions was ⁇ 0.3%, i.e., in control experiments with DNA from normal cells, none of 340 wells containing PCR products exhibited RED/GREEN ratios >3.0.
  • Any individual mutation (such as a G- to C- transversion at the second position of codon 12 o ⁇ c-Ki-ras) is expected to occur in ⁇ 1 in 50 polymerase-generated mutants (there are at least 50 base substitutions within or surrounding codons 12 and 13 that should yield high RED/GREEN ratios).
  • Determining the sequence of the putative mutants in the positive wells by direct sequencing as performed here or by any of the other techniques, provides unequivocal validation of a prospective mutation: a significant fraction of the mutations found in individual wells should be identical if the mutation occurred in vivo. Significance can be established through rigorous statistical analysis, as positive signals should be distributed according to Poisson probabilities. Moreover, the error rate in particular Digital Amplification experiments can be precisely determined through performance of Digital Amplification on DNA templates from normal cells.
  • Digital Amplification is as easily applied to RT-PCR products generated from RNA templates as it is to genomic DNA.
  • the fraction of alternatively spliced or mutant transcripts from a gene can be easily determined using photoluminescent probes specific for each of the PCR products generated.
  • Digital Amplification can be used to quantitate relative levels of gene expression within an RNA population.
  • each well would contain primers which are used to amplify a reference transcript expressed constitutively as well as primers specific for the experimental transcript.
  • One photoluminescent probe would then be used to detect PCR products from the reference transcript and a second photoluminescent probe used for the test transcript.
  • the number of wells in which the test transcript is amplified divided by the number of wells in which the reference transcript is amplified provides a quantitative measure of gene expression. Another group of examples involves the investigations of allelic status when two mutations are observed upon sequence analysis of a standard
  • each diluted sample has on average one half a template molecule. This is the same as one half of the diluted samples having one template molecule. This can be empirically determined by amplification. Either the analyte (selected genetic sequence) or the reference genetic sequence can be used for this determination. If the analysis method being used can detect analyte when present at a level of 20%, then one must dilute such that a significant number of diluted assay samples contain more than 20% of analyte. If the analysis method being used requires 100% analyte to detect, then dilution down to the single template molecule level will be required.
  • the digital amplification method requires analysis of a large number of samples to get meaningful results.
  • the accuracy of the determination will improve as the number of samples increases, up to a point.
  • the steps of amplifying and analyzing are performed in the same receptacle. This makes the method an in situ, or "one-pot" method.
  • the method can be used to find a tumor mutation in a population of cells which is not purely tumor cells.
  • a probe for a particular mutation need not be used, but diminution in binding to a wild-type probe can be used as an indicator of the presence of one or more mutations.
  • Chromosomal translocations which are characteristic of leukemias or lymphomas can be detected as a measure of the efficacy of therapy.
  • Gene amplifications are characteristic of certain disease states. These can be measured using digital amplification.
  • spliced forms of a transcript can be detected and quantitated relative to other forms of the transcript using digital amplification on cDNA made from mRNA.
  • cDNA made from mRNA one can determine relative levels of transcription of two different genes.
  • Biological samples which can be used as the starting material for the analyses may be from any tissue or body sample from which DNA or mRNA can be isolated. Preferred sources include stool, blood, and lymph nodes.
  • the biological sample is a cell-free lysate.
  • Molecular beacon probes can utilize any photoluminescent moiety as a detectable moiety. Typically these are dyes. Often these are fluorescent dyes. Photoluminescence is any process in which a material is excited by radiation such as light, is raised to an excited electronic or vibronic state, and subsequently re-emits that excitation energy as a photon of light. Such processes include fluorescence, which denotes emission accompanying descent from an excited state with paired electrons (a "singlet” state) or unpaired electrons (a "triplet” state) to a lower state with the same multiplicity, i.e., a quantum-mechanically “allowed” transition.
  • Photoluminescence also includes phosphorescence which denotes emission accompanying descent from an excited triplet or singlet state to a lower state of different multiplicity, i.e., a quantum mechanically "forbidden” transition. Compared to “allowed” transitions, "forbidden” transitions are associated with relatively longer excited state lifetimes.
  • the quenching of photoluminescence may be analyzed by a variety of methods which vary primarily in terms of signal transduction. Quenching may be transduced as changes in the intensity of photoluminescence or as changes in the ratio of photoluminescence intensities at two different wavelengths, or as changes in photoluminescence lifetimes, or even as changes in the polarization (anisotropy) of photoluminescence. Skilled practitioners will recognize that instrumentation for the measurement of these varied photoluminescent responses are known.
  • the particular ratiometric methods for the analysis of quenching in the instant examples should not be construed as limiting the invention to any particular form of signal transduction. Ratiometric measurements of photoluminescence intensity can include the measurement of changes in intensity, photoluminescence lifetimes, or even polarization (anisotropy).
  • molecular beacon probes as the means of analysis of the amplified dilution samples
  • other techniques can be used as well. These include sequencing, gel electrophoresis, hybridization with other types of probes, including TaqManTM (dual-labeled fluorogenic) probes (Perkin Elmer CorpVApplied Biosystems, Foster City, Calif), pyrene-labeled probes, and other biochemical assays.
  • Step 1 PCR amplifications.
  • the optimal conditions for PCR described in this section were determined by varying the parameters described in the
  • PCR was performed in 7 ul volumes in 96 well polypropylene PCR plates (Marsh Biomedical Products, Rochester, NY). The composition of the reactions was: 67 mM Tris, pH 8.8, 16.6 mM NH 4 SO 4 6.7 mM MgCl 2 , 10 mM ⁇ -mercaptoethanol, 1 mM dATP, 1 mM dCTP, 1 mM dGTP, 1 mM TTP, 6% DMSO, 1 uM primer F 1 , 1 uM primer Rl , 0.05 units/ul Platinum Taq polymerase (Life Technologies, Inc.), and "one-half genome equivalent" of DNA. To determine the amount of DNA corresponding to one-half genome equivalent, DNA samples were serially diluted and tested via PCR. The amount that yielded amplification products in half the wells, usually ⁇ 1.5 pg of total DNA, was defined as
  • Step 2 Fluorescence analysis. 3.5 ul of a solution with the following composition was added to each well: 67 mM Tris, pH 8.8, 16.6 mM NH 4 SO 4 6.7 mM MgCl 2 , 10 mM ⁇ -mercaptoethanol, 1 mM dATP, 1 mM dCTP, 1 mM dGTP, 1 mM TTP, 6% DMSO, 5 uM primer INT, 1 uM
  • MB-GREEN 1 uM MB-RED, 0.1 units/ul Platinum Taq polymerase.
  • the plates were centrifuged for 20 seconds at 6000 g and fluorescence read at excitation emission wavelengths of 485 nm/530 nm for MB-GREEN and 530 nm/590 nm for MB-RED.
  • the fluorescence in wells without template was typically 10,000 to 20,000 fluorescence "units", with about 75% emanating from the fluorometer background and the remainder from the MB probes.
  • the plates were then placed in a thermal cycler for asymmetric amplification at the following temperatures: 94° for one minute; 10 - 15 cycles of 94° for 15 sec, 55° for 15 sec, 70° for 15 seconds; 94° for one minute; and 60° for five minutes.
  • the plates were then incubated at room temperature for ten to sixty minutes and fluorescence measured as described above. Specific fluorescence was defined as the difference in fluorescence before and after the asymmetric amplification.
  • RED/GREEN ratios were defined as the specific fluorescence of MB-RED divided by that of MB-GREEN. RED/GREEN ratios were normalized to the ratio exhibited by the positive controls (25 genome equivalents of DNA from normal cells, as defined above in Example 1).
  • the experiment is outlined in Fig. 1A.
  • PCR products resulting from the amplification of single template molecules should be homogeneous in sequence, a variety of standard techniques could be used to assess their presence. Fluorescent probe-based technologies, which can be performed on the PCR products
  • MB probes are oligonucleotides with stem-loop structures that contain a fluorescent dye at the 5' end and a quenching agent (Dabcyl) at the 3' end (Fig. IB).
  • the degree of quenching via fluorescence energy resonance transfer is inversely proportional to the 6 th power of the distance between the Dabcyl group and the fluorescent dye.
  • MB probes reform a stem-loop structure which quenches the fluorescent signal from the dye (41). If a PCR product whose sequence is complementary to the loop sequence is present during the heating/cooling cycle, hybridization of the MB to one strand of the PCR product will increase the distance between the Dabcyl and the dye, resulting in increased fluorescence.
  • oligonucleotides used for Digital Amplifications shown in Fig. lC.
  • Two unmodified oligonucleotides are used as primers for the PCR reaction.
  • Two MB probes, each labeled with a different fluorophore, are used to detect the PCR products.
  • MB-GREEN has a loop region that is complementary to the portion of the WT PCR product that is queried for mutations. Mutations within the corresponding sequence of the
  • PCR product should significantly impede its hybridization to the MB probe (33,34).
  • MB-RED has a loop region that is complementary to a different portion of the PCR product, one not expected to be mutant. It thus should produce a signal whenever a well contains a PCR product, whether that product is WT or mutant in the region queried by MB-GREEN. Both MB probes are used together to simultaneously detect the presence of a PCR product and its mutational status.
  • the first step involves amplification from single template molecules.
  • Most protocols for amplification from small numbers of template molecules use a nesting procedure, wherein a product resulting from one set of primers is used as template in a second reaction employing internal primers.
  • nesting would be inconvenient and could lead to contamination problems.
  • conditions were sought that would achieve robust amplification without nesting. The most important of these conditions involved the use of a polymerase that was activated only after heating (44,45) and optimized concentrations of dNTP's, primers, buffer components, and temperature.
  • Examples 1-3 The conditions specified in Examples 1-3 were defined after individually optimizing each of these components and proved suitable for amplification of several different human genomic DNA sequences.
  • the time required for PCR was not particularly long (-2.5 hr)
  • the number of cycles used was high and excessive compared to the number of cycles required to amplify the "average" single template molecule.
  • the large cycle number was necessary because the template in some wells might not begin to be amplified until several PCR cycles had been completed.
  • the large number of cycles ensured that every well (not simply the average well) would generate a substantial and roughly equal amount of PCR product if a template molecule were present within it.
  • the second step in Fig 1A involves the detection of these PCR products. It was necessary to considerably modify the standard MB probe approach in order for it to function efficiently in Digital Amplification applications. Theoretically, one separate MB probe could be used to detect each specific mutation that might occur within the queried sequence. By inclusion of one MB corresponding to WT sequence and another corresponding to mutant sequence, the nature of the PCR product would be revealed. Though this strategy could obviously be used effectively in some situations, it becomes complex when several different mutations are expected to occur within the same queried sequence. For example, in the c-Ki-Ras gene example explored here, twelve different base substitutions resulting in missense mutations could theoretically occur within codons 12 and 13, and at least seven of these are observed in naturally-occurring human cancers.
  • MB-GREEN probe For discrimination between WT and mutant sequences (MB-GREEN probe), we found that a 16 base pair loop, of melting temperature (Tm) 50-51 °, and a 4 bp stem, of sequence
  • Fig. 2 Examples of the ratios obtained in replicate wells containing DNA templates from colorectal tumor cells with mutations of c-Ki-Ras are shown in Fig. 2.
  • Fig. 2 Five genome equivalents of DNA were added to each well prior to amplification.
  • Each of six tested mutants yielded ratios of RED/GREEN fluorescence that were significantly in excess of the ratio obtained with DNA from normal cells (1.5 to 3.4 in the mutants compared to 1.0 in normal DNA; p ⁇ 0.0001 in each case, Student's t-Test). The reproducibility of the ratios can be observed in this figure.
  • Direct DNA sequencing of the PCR products used for fluorescence analysis showed that the RED/GREEN ratios were dependent on the relative fraction of mutant genes within the template population (Fig. 2).
  • the DNA from cells containing one mutant c-Ki-Ras allele per every two WT c-Ki-Ras allele yielded a RED/GREEN ratio of 1.5 (Glyl2Arg mutation) while the cells containing three mutant c-Ki-Ras alleies per WT allele exhibited a ratio of 3.4 (Gly 12 Asp).
  • Fig. 4. Again, a biphasic distribution was observed.
  • 64% of the positive wells exhibited RED/GREEN ratios in excess of 3.0 while the other 36% of the positive wells exhibited ratios ranging from 0.8 to 1.1.
  • 54% of the positive wells exhibited RED/GREEN ratios >3.0 while the other positive wells yielded ratios ranging from 0.9 to 1.1.
  • the PCR products from 16 positive wells were used as sequencing templates (Fig. 4).
  • MB-RED fluorescence indicated that 102 of these 288 experimental wells contained PCR products (mean +/- s.d. of 47,000 +/- 18,000 SFU) while the other 186 wells did not (2600 +/- 1500 SFU).
  • the RED/GREEN ratios of the 102 positive wells suggested that five contained mutant c-Ki-Ras genes, with ratios ranging from 2.1 to 5.1.
  • the other 97 wells exhibited ratios ranging from 0.7 to 1.2, identical to those observed in the positive control wells.
  • the PCR products were directly sequenced.
  • the four wells exhibiting RED/GREEN ratios in excess of 3.0 were completely composed of mutant c-Ki-Ras sequence (Fig. 5).
  • the sequence of three of these PCR products revealed Gly 12 Ala mutations (GGT to GCT at codon 12), while the sequence of the fourth indicated a silent C to T transition at the third position of codon 13. This transition presumably resulted from a PCR error during the first productive cycle of amplification from a WT template.
  • the well with a ratio of 2.1 contained a -1:1 mix of WT and Glyl2Ala mutant sequences.

Abstract

The identification of pre-defined mutations expected to be present in a minor fraction of a cell population is important for a variety of basic research and clinical applications. The exponential, analog nature of the polymerase chain reaction is transformed into a linear, digital signal suitable for this purpose. Single molecules can be isolated by dilution and individual amplified; each product is then separately analyzed for the presence of mutations. The process provides a reliable and quantitative measure of the proportion of variant sequences within a DNA example.

Description

DIGITAL AMPLIFICATION
This application claims the benefit of U.S. Serial No. 60/146,792, filed August 2, 1999.
The U.S. government retains certain rights in this invention by virtue of its support of the underlying research, supported by grants CA 43460, CA
57345, and CA 62924 from the National Institutes of Health. TECHNICAL FIELD OF THE INVENTION
This invention is related to diagnostic genetic analyses. In particular it relates to detection of genetic changes and gene expression. BACKGROUND OF THE INVENTION
In classical genetics, only mutations of the germ-line were considered important for understanding disease. With the realization that somatic mutations are the primary cause of cancer (1), and may also play a role in aging (2,3), new genetic principles have arisen. These discoveries have provided a wealth of new opportunities for patient management as well as for basic research into the pathogenesis of neoplasia. However, many of these opportunities hinge upon detection of a small number of mutant-containing cells among a large excess of normal cells. Examples include the detection of neoplastic cells in urine (4), stool (5,6), and sputum (7,8) of patients with cancers of the bladder, colorectum, and lung, respectively. Such detection has been shown in some cases to be possible at a stage when the primary tumors are still curable and the patients asymptomatic. Mutant sequences from the DNA of neoplastic cells have also been found in the blood of cancer patients (9-11). The detection of residual disease in lymph nodes or surgical margins may be useful in predicting which patients might benefit most from further therapy (12-14). From a basic research standpoint, analysis of the early effects of carcinogens is often dependent on the ability to detect small populations of mutant cells (15-17). Because of the importance of this issue in so many settings, many useful techniques have been developed for the detection of mutations. DNA sequencing is the gold standard for the detection of germ line mutations, but is useful only when the fraction of mutated alleles is greater than ~20% (18,19). Mutant-specific oligonucleotides can sometimes be used to detect mutations present in a minor proportion of the cells analyzed, but the signal to noise ratio distinguishing mutant and wild-type ( WT) templates is variable (20-22) The use of mutant-specific primers or the digestion of polymerase chain reaction (PCR) products with specific restriction endonucleases are extremely sensitive methods for detecting such mutations, but it is difficult to quantitate the fraction of mutant molecules in the starting population with these techniques (23-28). Other innovative approaches for the detection of somatic mutations have been reviewed (29-32). A general problem with these methods is that it is difficult or impossible to independently confirm the existence of any mutations that are identified. Thus there is a need in the art for methods for accurately and quantitatively detecting genetic sequences in mixed populations of sequences. SUMMARY OF THE INVENTION
It is an object of the present invention to provide methods for determining the presence of a selected genetic sequence in a population of genetic sequences.
It is another object of the present invention to provide molecular beacon probes useful in the method of the invention.
These and other objects of the invention are achieved by providing a method for determining the presence of a selected genetic sequence in a population of genetic sequences. A biological sample comprising nucleic acid template molecules is diluted to form a set of assay samples. The template molecules within the assay samples are amplified to form a population of amplified molecules in the assay samples of the set. The amplified molecules in the assay samples of the set are then analyzed to determine a first number of assay samples which contain the selected genetic sequence and a second number of assay samples which contain a reference genetic sequence. The first number is then compared to the second number to ascertain a ratio which reflects the composition of the biological sample.
Another embodiment of the invention is a method for determining the ratio of a selected genetic sequence in a population of genetic sequences. Template molecules within a set comprising a plurality of assay samples are amplified to form a population of amplified molecules in each of the assay samples of the set. The amplified molecules in the assay samples of the set are analyzed to determine a first number of assay samples which contain the selected genetic sequence and a second number of assay samples which contain a reference genetic sequence. The first number is compared to the second number to ascertain a ratio which reflects the composition of the biological sample.
According to another embodiment of the invention, a molecular beacon probe is provided. It comprises an oligonucleotide with a stem-loop structure having a photoluminescent dye at one of the 51 or 3' ends and a quenching agent at the opposite 5' or 31 end. The loop consists of 16 base pairs and has a Tm of 50-51 °C. The stem consists of 4 base pairs having a sequence 5'-CACG-3'.
A second type of molecular beacon probe is provided in another embodiment. It comprises an oligonucleotide with a stem-loop structure having a photoluminescent dye at one of the 5' or 3' ends and a quenching agent at the opposite 5' or 3' end. The loop consists of 19-20 base pairs and has a Tm of 54-56°C. The stem consists of 4 base pairs having a sequence 5'- CACG-3'.
Another embodiment provides the two types of molecular beacon probes, either mixed together or provided in a divided container as a kit. The invention thus provides the art with the means to obtain quantitative assessments of particular DNA or RNA sequences in mixed populations of sequences using digital (binary) signals. BRTEF DESCRIPTION OF THE DRAWINGS FIG. 1. Schematic of experimental design. (A) The basic two steps involved:
PCR on diluted DNA samples is followed by addition of fluorescent probes which discriminate between WT and mutant alleies and subsequent fluorometry. (B) Principle of molecular beacon analysis. In the stem-loop configuration, fluorescence from a dye at the 5' end of the oligonucleotide probe is quenched by a Dabcyl group at the 3' end. Upon hybridization to a template, the dye is separated from the quencher, resulting in increased fluorescence. Modified from Marras et al. . (C) Oligonucleotide design. Primers FI and Rl are used to amplify the genomic region of interest. Primer INT is used to produce single stranded DNA from the original PCR products during a subsequent asymmetric PCR step (see Materials and Methods).
MB-RED is a Molecular Beacon which detects any appropriate PCR product, whether it is WT or mutant at the queried codons. MB-GREEN is a Molecular Beacon which preferentially detects the WT PCR product.
FlG.2. Discrimination between WT and mutant PCR products by Molecular Beacons. Ten separate PCR products, each generated from ~50 genome equivalents of DNA of cells containing the indicated mutations of c-Ki-Ras, were analyzed with the Molecular Beacon probes described in the text. Representative examples of the PCR products used for Molecular Beacon analysis were purified and directly sequenced. In the cases with Glyl2Cys and Gly 12 Arg mutations, contaminating non-neoplastic cells within the tumor presumably accounted for the relatively low ratios. In the cases with Glyl2Ser and Gly 12 Asp, there were apparently two or more alleies of mutant c-Ki-Ras for every WT allele; both these tumors were aneuploid.
FlG. 3. Detecting Dig-PCR products with MB-RED. Specific Fluorescence Units of representative wells from an experiment employing colorectal cancer cells with Glyl2Asp or Gly 13 Asp mutations of the c-Ki-Ras gene. Wells with values > 10,000 are shaded yellow. Polyacrylamide gel electrophoretic analyses of the PCR products from selected wells are shown. Wells with fluorescence values <3500 had no PCR product of the correct size while wells with fluorescence values > 10,000 SFU always contained PCR products of 129 bp. Non-specific products generated during the large number of cycles required for Dig-PCR did not affect the fluorescence analysis. Ml and M2 are molecular weight markers used to determine the size of fragments indicated on the left (in base pairs).
FlG.4. Discriminating WT from mutant PCR products obtained in Dig-PCR.
RED/GREEN ratios were determined from the fluorescence of MB-RED and MB-GREEN as described in Materials and Methods. The wells shown are the same as those illustrated in Fig. 3. The sequences of PCR products from the indicated wells were determined as described in Materials and Methods. The wells with RED/GREEN ratios >3.0 each contained mutant sequences while those with RED/GREEN ratios of -1.0 contained WT sequences.
FlG. 5. Dig-PCR of DNA from a stool sample. The 384 wells used in the experiment are displayed. Those colored blue contained 25 genome equivalents of DNA from normal cells. Each of these registered positive with MB-RED and the RED/GREEN ratios were 1.0 +/- 0.1 (mean +/- 1 standard deviation). The wells colored yellow contained no template DNA and each was negative with MB-RED (i.e., fluorescence <3500 fluorescence units.). The other 288 wells contained diluted DNA from the stool sample prepared by alkaline extraction. (Rubeck et al., 1998, BioTechniques 25:588-592.) Those registering as positive with MB-RED were colored either red or green, depending on their RED/GREEN ratios. Those registering negative with MB-RED were colored white. PCR products from the indicated wells were used for automated sequence analysis.
DETAILED DESCRIPTION OF THE INVENTION The method devised by the present inventors involves separately amplifying small numbers of template molecules so that the resultant products have a proportion of the analyte sequence which is detectable by the detection means chosen. At its limit, single template molecules can be amplified so that the products are completely mutant or completely wild-type (WT). The homogeneity of these amplification products makes them trivial to distinguish through existing techniques.
The method requires analyzing a large number of amplified products simply and reliably. Techniques for such assessments were developed, with the output providing a digital readout of the fraction of mutant alleies in the analyzed population.
The biological sample is diluted to a point at which a practically usable number of the diluted samples contain a proportion of the selected genetic sequence (analyte) relative to total template molecules such that the analyzing technique being used can detect the analyte. A practically usable number of diluted samples will depend on cost of the analysis method. Typically it would be desirable that at least 1/50 of the diluted samples have a detectable proportion of analyte. At least 1/10, 1/5, 3/10, 2/5, 1/2, 3/5, 7/10, 4/5, or 9/10 of the diluted samples may have a detectable proportion of analyte. The higher the fraction of samples which will provide useful information, the more economical will be the overall assay. Over-dilution will also lead to a loss of economy, as many samples will be analyzed and provide no signal. A particularly preferred degree of dilution is to a point where each of the assay samples has on average one-half of a template. The dilution can be performed from more concentrated samples. Alternatively, dilute sources of template nucleic acids can be used. All of the samples may contain amplifable template molecules. Desirably each assay sample prior to amplification will contain less than a hundred or less than ten template molecules. Digital amplification can be used to detect mutations present at relatively low levels in the samples to be analyzed. The limit of detection is defined by the number of wells that can be analyzed and the intrinsic mutation rate of the polymerase used for amplification. 384 well PCR plates are commercially available and 1536 well plates are on the horizon, theoretically allowing sensitivities for mutation detection at the -0.1% level. It is also possible that Digital Amplification can be performed in microarray format, potentially increasing the sensitivity by another order of magnitude. This sensitivity may ultimately be limited by polymerase errors. The effective error rate in PCR as performed under our conditions was <0.3%, i.e., in control experiments with DNA from normal cells, none of 340 wells containing PCR products exhibited RED/GREEN ratios >3.0. Any individual mutation (such as a G- to C- transversion at the second position of codon 12 oϊc-Ki-ras) is expected to occur in <1 in 50 polymerase-generated mutants (there are at least 50 base substitutions within or surrounding codons 12 and 13 that should yield high RED/GREEN ratios). Determining the sequence of the putative mutants in the positive wells, by direct sequencing as performed here or by any of the other techniques, provides unequivocal validation of a prospective mutation: a significant fraction of the mutations found in individual wells should be identical if the mutation occurred in vivo. Significance can be established through rigorous statistical analysis, as positive signals should be distributed according to Poisson probabilities. Moreover, the error rate in particular Digital Amplification experiments can be precisely determined through performance of Digital Amplification on DNA templates from normal cells.
Digital Amplification is as easily applied to RT-PCR products generated from RNA templates as it is to genomic DNA. For example, the fraction of alternatively spliced or mutant transcripts from a gene can be easily determined using photoluminescent probes specific for each of the PCR products generated. Similarly, Digital Amplification can be used to quantitate relative levels of gene expression within an RNA population. For this amplification, each well would contain primers which are used to amplify a reference transcript expressed constitutively as well as primers specific for the experimental transcript. One photoluminescent probe would then be used to detect PCR products from the reference transcript and a second photoluminescent probe used for the test transcript. The number of wells in which the test transcript is amplified divided by the number of wells in which the reference transcript is amplified provides a quantitative measure of gene expression. Another group of examples involves the investigations of allelic status when two mutations are observed upon sequence analysis of a standard
DNA sample. To distinguish whether one variant is present in each allele (vs. both occurring in one allele), cloning of PCR products is generally performed. The approach described here would simplify the analysis by eliminating the need for cloning. Other potential applications of Digital Amplification are listed in Table 1. When the goal is the quantitation of the proportion of two relatively common alleies or transcripts rather than the detection of rare alleies, techniques such as those employing TaqMan and real time PCR provide an excellent alternative to use of molecular beacons. Advantages of real time PCR methods include their simplicity and the ability to analyze multiple samples simultaneously. However, Digital Amplification may prove useful for these applications when the expected differences are small, (e.g., only ~2-fold, such as occurs with allelic imbalances (55))
The ultimate utility of Digital Amplification lies in its ability to convert the intrinsically exponential nature of PCR to a linear one. It should thereby prove useful for experiments requiring the investigation of individual alleies, rare variants/mutations, or quantitative analysis of PCR products.
In one preferred embodiment each diluted sample has on average one half a template molecule. This is the same as one half of the diluted samples having one template molecule. This can be empirically determined by amplification. Either the analyte (selected genetic sequence) or the reference genetic sequence can be used for this determination. If the analysis method being used can detect analyte when present at a level of 20%, then one must dilute such that a significant number of diluted assay samples contain more than 20% of analyte. If the analysis method being used requires 100% analyte to detect, then dilution down to the single template molecule level will be required. To achieve a dilution to approximately a single template molecule level, one can dilute such that between 0.1 and 0.9 of the assay samples yield an amplification product. More preferably the dilution will be to between 0.1 and 0.6, more preferably to between 0.3 and 0.5 of the assay samples yielding an amplification product.
The digital amplification method requires analysis of a large number of samples to get meaningful results. Preferably at least ten diluted assay samples are amplified and analyzed. More preferably at least 15, 20, 25, 30, 40, 50, 75, 100, 500, or 1000 diluted assay samples are amplified and analyzed. As in any method, the accuracy of the determination will improve as the number of samples increases, up to a point. Because a large number of samples must be analyzed, it is desirable to reduce the manipulative steps, especially sample transfer steps. Thus it is preferred that the steps of amplifying and analyzing are performed in the same receptacle. This makes the method an in situ, or "one-pot" method.
The number of different situations in which the digital amplification method will find application is large. Some of these are listed in Table 1. As shown in the examples, the method can be used to find a tumor mutation in a population of cells which is not purely tumor cells. As described in the examples, a probe for a particular mutation need not be used, but diminution in binding to a wild-type probe can be used as an indicator of the presence of one or more mutations. Chromosomal translocations which are characteristic of leukemias or lymphomas can be detected as a measure of the efficacy of therapy. Gene amplifications are characteristic of certain disease states. These can be measured using digital amplification. Alternatively spliced forms of a transcript can be detected and quantitated relative to other forms of the transcript using digital amplification on cDNA made from mRNA. Similarly, using cDNA made from mRNA one can determine relative levels of transcription of two different genes. One can use digital amplification to distinguish between a situation where one allele carries two mutations and one mutation is carried on each of two alleies in an individual. Allelic imbalances often result from a disease state. These can be detected using digital amplification. Biological samples which can be used as the starting material for the analyses may be from any tissue or body sample from which DNA or mRNA can be isolated. Preferred sources include stool, blood, and lymph nodes. Preferably the biological sample is a cell-free lysate.
Figure imgf000012_0001
w w w
H
r w
Figure imgf000012_0002
Molecular beacon probes according to the present invention can utilize any photoluminescent moiety as a detectable moiety. Typically these are dyes. Often these are fluorescent dyes. Photoluminescence is any process in which a material is excited by radiation such as light, is raised to an excited electronic or vibronic state, and subsequently re-emits that excitation energy as a photon of light. Such processes include fluorescence, which denotes emission accompanying descent from an excited state with paired electrons (a "singlet" state) or unpaired electrons (a "triplet" state) to a lower state with the same multiplicity, i.e., a quantum-mechanically "allowed" transition. Photoluminescence also includes phosphorescence which denotes emission accompanying descent from an excited triplet or singlet state to a lower state of different multiplicity, i.e., a quantum mechanically "forbidden" transition. Compared to "allowed" transitions, "forbidden" transitions are associated with relatively longer excited state lifetimes.
The quenching of photoluminescence may be analyzed by a variety of methods which vary primarily in terms of signal transduction. Quenching may be transduced as changes in the intensity of photoluminescence or as changes in the ratio of photoluminescence intensities at two different wavelengths, or as changes in photoluminescence lifetimes, or even as changes in the polarization (anisotropy) of photoluminescence. Skilled practitioners will recognize that instrumentation for the measurement of these varied photoluminescent responses are known. The particular ratiometric methods for the analysis of quenching in the instant examples should not be construed as limiting the invention to any particular form of signal transduction. Ratiometric measurements of photoluminescence intensity can include the measurement of changes in intensity, photoluminescence lifetimes, or even polarization (anisotropy).
Although the working examples demonstrate the use of molecular beacon probes as the means of analysis of the amplified dilution samples, other techniques can be used as well. These include sequencing, gel electrophoresis, hybridization with other types of probes, including TaqMan™ (dual-labeled fluorogenic) probes (Perkin Elmer CorpVApplied Biosystems, Foster City, Calif), pyrene-labeled probes, and other biochemical assays.
The above disclosure generally describes the present invention. A more complete understanding can be obtained by reference to the following specific examples which are provided herein for purposes of illustration only, and are not intended to limit the scope of the invention.
EXAMPLE 1
Step 1: PCR amplifications. The optimal conditions for PCR described in this section were determined by varying the parameters described in the
Results. PCR was performed in 7 ul volumes in 96 well polypropylene PCR plates (Marsh Biomedical Products, Rochester, NY). The composition of the reactions was: 67 mM Tris, pH 8.8, 16.6 mM NH4SO4 6.7 mM MgCl2, 10 mM β-mercaptoethanol, 1 mM dATP, 1 mM dCTP, 1 mM dGTP, 1 mM TTP, 6% DMSO, 1 uM primer F 1 , 1 uM primer Rl , 0.05 units/ul Platinum Taq polymerase (Life Technologies, Inc.), and "one-half genome equivalent" of DNA. To determine the amount of DNA corresponding to one-half genome equivalent, DNA samples were serially diluted and tested via PCR. The amount that yielded amplification products in half the wells, usually ~1.5 pg of total DNA, was defined as
"one-half genome equivalent" and used in each well of subsequent Digital Amplification experiments. Fifty ul light mineral oil (Sigma M-3516) was added to each well and reactions performed in a HybAid Thermal cycler at the following temperatures: denaturation at 94° for one min; 60 cycles of 94 ° for 15 sec, 55 ° for 15 sec, 70° for 15 seconds; 70 ° for five minutes.
Reactions were read immediately or stored at room temperature for up to
36 hours before fluorescence analysis. EXAMPLE 2
Step 2: Fluorescence analysis. 3.5 ul of a solution with the following composition was added to each well: 67 mM Tris, pH 8.8, 16.6 mM NH4SO4 6.7 mM MgCl2, 10 mM β-mercaptoethanol, 1 mM dATP, 1 mM dCTP, 1 mM dGTP, 1 mM TTP, 6% DMSO, 5 uM primer INT, 1 uM
MB-GREEN, 1 uM MB-RED, 0.1 units/ul Platinum Taq polymerase. The plates were centrifuged for 20 seconds at 6000 g and fluorescence read at excitation emission wavelengths of 485 nm/530 nm for MB-GREEN and 530 nm/590 nm for MB-RED. The fluorescence in wells without template was typically 10,000 to 20,000 fluorescence "units", with about 75% emanating from the fluorometer background and the remainder from the MB probes. The plates were then placed in a thermal cycler for asymmetric amplification at the following temperatures: 94° for one minute; 10 - 15 cycles of 94° for 15 sec, 55° for 15 sec, 70° for 15 seconds; 94° for one minute; and 60° for five minutes. The plates were then incubated at room temperature for ten to sixty minutes and fluorescence measured as described above. Specific fluorescence was defined as the difference in fluorescence before and after the asymmetric amplification. RED/GREEN ratios were defined as the specific fluorescence of MB-RED divided by that of MB-GREEN. RED/GREEN ratios were normalized to the ratio exhibited by the positive controls (25 genome equivalents of DNA from normal cells, as defined above in Example 1). We found that the ability of MB probes to discriminate between WT and mutant sequences under our conditions could not be reliably determined from experiments in which they were tested by hybridization to relatively short complementary single stranded oligonucleotides, and that actual PCR products had to be used for validation.
EXAMPLE 3
Oligonucleotides and DNA sequencing. Primer FI 5*-CATGTTCTAATATAGTCACATTTTCA-3'; Primer Rl
5'-TCTGAATTAGCTGTATCGTCAAGG-3'; Primer INT 5'-TAGCTGTATCGTCAAGGCAC-3'; MB-RED:
5'-Cy3-CACGGGCCTGCTGAAAATGACTGCGTG-Dabcyl-3'; M B - G R E E N : 5'-Fluorescein-CACGGGAGCTGGTGGCGTAGCGTG-Dabcyl-3'. Molecular Beacons (33,34) were synthesized by Midland Scientific and other oligonucleotides were synthesized by Gene Link (Thornwood, NY). All were dissolved at 50 uM in TE (10 mM Tris, pH 8.0/ 1 mM EDTA) and kept frozen and in the dark until use. PCR products were purified using QIAquick PCR purification kits (Qiagen). In the relevant experiments described in the text, 20% of the product from single wells was used for gel electrophoresis and 40% was used for each sequencing reaction. The primer used for sequencing was
5'-CATTATTTTTATTATAAGGCCTGC-3\ Sequencing was performed using fluorescently-labeled ABI Big Dye terminators and an ABI 377 automated sequencer.
EXAMPLE 4
Principles underlying experiment. The experiment is outlined in Fig. 1A. First, the DNA is diluted into multiwell plates so that there is, on average, one template molecule per two wells, and PCR is performed. Second, the individual wells are analyzed for the presence of PCR products of mutant and WT sequence using fluorescent probes.
As the PCR products resulting from the amplification of single template molecules should be homogeneous in sequence, a variety of standard techniques could be used to assess their presence. Fluorescent probe-based technologies, which can be performed on the PCR products
"in situ" (i.e., in the same wells) are particularly well-suited for this application (31, 33-40). We chose to explore the utility of one such technology, involving Molecular Beacons (MB), for this purpose (33,34). MB probes are oligonucleotides with stem-loop structures that contain a fluorescent dye at the 5' end and a quenching agent (Dabcyl) at the 3' end (Fig. IB). The degree of quenching via fluorescence energy resonance transfer is inversely proportional to the 6th power of the distance between the Dabcyl group and the fluorescent dye. After heating and cooling, MB probes reform a stem-loop structure which quenches the fluorescent signal from the dye (41). If a PCR product whose sequence is complementary to the loop sequence is present during the heating/cooling cycle, hybridization of the MB to one strand of the PCR product will increase the distance between the Dabcyl and the dye, resulting in increased fluorescence.
A schematic of the oligonucleotides used for Digital Amplifications shown in Fig. lC. Two unmodified oligonucleotides are used as primers for the PCR reaction. Two MB probes, each labeled with a different fluorophore, are used to detect the PCR products. MB-GREEN has a loop region that is complementary to the portion of the WT PCR product that is queried for mutations. Mutations within the corresponding sequence of the
PCR product should significantly impede its hybridization to the MB probe (33,34). MB-RED has a loop region that is complementary to a different portion of the PCR product, one not expected to be mutant. It thus should produce a signal whenever a well contains a PCR product, whether that product is WT or mutant in the region queried by MB-GREEN. Both MB probes are used together to simultaneously detect the presence of a PCR product and its mutational status.
Practical Considerations. Numerous conditions were optimized to define conditions that could be reproducibly and generally applied. As outlined in Fig. 1A, the first step involves amplification from single template molecules. Most protocols for amplification from small numbers of template molecules use a nesting procedure, wherein a product resulting from one set of primers is used as template in a second reaction employing internal primers. As many applications of digital amplification are expected to require hundreds or thousands of separate amplifications, such nesting would be inconvenient and could lead to contamination problems. Hence, conditions were sought that would achieve robust amplification without nesting. The most important of these conditions involved the use of a polymerase that was activated only after heating (44,45) and optimized concentrations of dNTP's, primers, buffer components, and temperature.
The conditions specified in Examples 1-3 were defined after individually optimizing each of these components and proved suitable for amplification of several different human genomic DNA sequences. Though the time required for PCR was not particularly long (-2.5 hr), the number of cycles used was high and excessive compared to the number of cycles required to amplify the "average" single template molecule. The large cycle number was necessary because the template in some wells might not begin to be amplified until several PCR cycles had been completed. The large number of cycles ensured that every well (not simply the average well) would generate a substantial and roughly equal amount of PCR product if a template molecule were present within it.
The second step in Fig 1A involves the detection of these PCR products. It was necessary to considerably modify the standard MB probe approach in order for it to function efficiently in Digital Amplification applications. Theoretically, one separate MB probe could be used to detect each specific mutation that might occur within the queried sequence. By inclusion of one MB corresponding to WT sequence and another corresponding to mutant sequence, the nature of the PCR product would be revealed. Though this strategy could obviously be used effectively in some situations, it becomes complex when several different mutations are expected to occur within the same queried sequence. For example, in the c-Ki-Ras gene example explored here, twelve different base substitutions resulting in missense mutations could theoretically occur within codons 12 and 13, and at least seven of these are observed in naturally-occurring human cancers. To detect all twelve mutations as well as the WT sequence with individual Molecular Beacons would require 13 different probes. Inclusion of such a large number of MB probes would raise the background fluorescence and cost of the assay. We therefore attempted to develop a single probe that would react with WT sequences better than any mutant sequence within the queried sequence. We found that the length of the loop sequence, its melting temperature, and the length and sequence of the stem were each important in determining the efficacy of such probes.
Loops ranging from 14 to 26 bases and stems ranging from 4 to 6 bases, as well as numerous sequence variations of both stems and loops, were tested during the optimization procedure. For discrimination between WT and mutant sequences (MB-GREEN probe), we found that a 16 base pair loop, of melting temperature (Tm) 50-51 °, and a 4 bp stem, of sequence
5'-CACG-3', were optimal. For MB-RED probes, the same stem, with a 19-20 bp loop of Tm 54-56°, proved optimal. The differences in the loop sizes and melting temperatures between MB-GREEN and MB-RED probes reflected the fact that only the GREEN probe is designed to discriminate between closely related sequences, with a shorter region of homology facilitating such discrimination.
Examples of the ratios obtained in replicate wells containing DNA templates from colorectal tumor cells with mutations of c-Ki-Ras are shown in Fig. 2. In this experiment, fifty genome equivalents of DNA were added to each well prior to amplification. Each of six tested mutants yielded ratios of RED/GREEN fluorescence that were significantly in excess of the ratio obtained with DNA from normal cells (1.5 to 3.4 in the mutants compared to 1.0 in normal DNA; p < 0.0001 in each case, Student's t-Test). The reproducibility of the ratios can be observed in this figure. Direct DNA sequencing of the PCR products used for fluorescence analysis showed that the RED/GREEN ratios were dependent on the relative fraction of mutant genes within the template population (Fig. 2). Thus, the DNA from cells containing one mutant c-Ki-Ras allele per every two WT c-Ki-Ras allele yielded a RED/GREEN ratio of 1.5 (Glyl2Arg mutation) while the cells containing three mutant c-Ki-Ras alleies per WT allele exhibited a ratio of 3.4 (Gly 12 Asp). These data suggested that wells containing only mutant alleies (no WT) would yield ratios in excess of 3.0, with the exact value dependent on the specific mutation.
Though this mode is the most convenient for many applications, we found it useful to add the MB probes after the PCR-amplification was complete (Fig. 1). This allowed us to use a standard multiwell plate fiuorometer to sequentially analyze a large number of multiwell plates containing pre-formed PCR products and bypassed the requirement for multiple real time PCR instruments. Additionally, we found that the fluorescent signals obtained could be considerably enhanced if several cycles of asymmetric, linear amplification were performed in the presence of the MB probes. Asymmetric amplification was achieved by including an excess of a single internal primer (primer INT in Fig. IC) at the time of addition of the MB probes.
EXAMPLE 5
Analysis of DNA from tumor cells. The principles and practical considerations described above were illlustrated with DNA from two colorectal cancer cell lines, one with a mutation in c-Ki-Ras codon 12 and the other in codon 13. Representative examples of the MB-RED fluorescence values obtained are shown in Fig. 3. There was a clear biphasic distribution, with "positive" wells yielding values in excess of
10,000 specific fluorescence units (SFU, as defined in Materials and Methods) and "negative" wells yielding values less than 3500 SFU. Gel electrophoreses of 127 such wells demonstrated that all positive wells, but no negative wells, contained PCR products of the expected size (Fig. 3). The RED/GREEN fluorescence ratios of the positive wells are shown in
Fig. 4. Again, a biphasic distribution was observed. In the experiment with the tumor containing a Gly 12 Asp mutation, 64% of the positive wells exhibited RED/GREEN ratios in excess of 3.0 while the other 36% of the positive wells exhibited ratios ranging from 0.8 to 1.1. In the case of the tumor with the Glyl3Asp mutation, 54% of the positive wells exhibited RED/GREEN ratios >3.0 while the other positive wells yielded ratios ranging from 0.9 to 1.1. The PCR products from 16 positive wells were used as sequencing templates (Fig. 4). All the wells yielding a ratio in excess of 3.0 were found to contain mutant c-Ki-Ras fragments of the expected sequence, while WT sequence was found in the other PCR products. The presence of homogeneous WT or mutant sequence confirmed that the amplification products were usually derived from single template molecules. The ratios of WT to mutant PCR products determined from the Digital Amplificationassay was also consistent with the fraction of mutant alleies inferred from direct sequence analysis of genomic DNA from the two tumor lines (Fig. 2).
Digital Analysis of DNA from stool. As a more practical example, we analyzed the DNA from stool specimens of colorectal cancer patients. A representative result of such an experiment is illustrated in Fig. 5. From previous analyses of stool specimens from patients whose tumors contained c-Ki-Ras gene mutations, we expected that 1% to 10% of the c-Ki-Ras genes purified from stool would be mutant. We therefore set up a 384 well Digital Amplificationexperiment. As positive controls, 48 of the wells contained 25 genome equivalents of DNA (defined in Materials and Methods) from normal cells. Another 48 wells served as negative controls (no DNA template added). The other 288 wells contained an appropriate dilution of stool DNA. MB-RED fluorescence indicated that 102 of these 288 experimental wells contained PCR products (mean +/- s.d. of 47,000 +/- 18,000 SFU) while the other 186 wells did not (2600 +/- 1500 SFU). The RED/GREEN ratios of the 102 positive wells suggested that five contained mutant c-Ki-Ras genes, with ratios ranging from 2.1 to 5.1. The other 97 wells exhibited ratios ranging from 0.7 to 1.2, identical to those observed in the positive control wells. To determine the nature of the mutant c-Ki-Ras genes in the five positive wells from stool, the PCR products were directly sequenced. The four wells exhibiting RED/GREEN ratios in excess of 3.0 were completely composed of mutant c-Ki-Ras sequence (Fig. 5). The sequence of three of these PCR products revealed Gly 12 Ala mutations (GGT to GCT at codon 12), while the sequence of the fourth indicated a silent C to T transition at the third position of codon 13. This transition presumably resulted from a PCR error during the first productive cycle of amplification from a WT template. The well with a ratio of 2.1 contained a -1:1 mix of WT and Glyl2Ala mutant sequences.
Thus 3.9% (4/102) of the c-Ki-Ras alleies present in this stool sample contained a Gly 12 Ala mutation. The mutant alleies in the stool presumably arose from the colorectal cancer of the patient, as direct sequencing of PCR products generated from DNA of the cancer revealed the identical Gly 12 Ala mutation (not shown).
References
1. Vogelstein & Kinzler, (1998) The Genetic Basis of Human Cancer (MCGraw-Hill, Toronto).
2. Lee et al., (1997) Free Radical Biol Med 22, 1259-69.
3. Ozawa, T. ( 1997) Physiol Rev 77, 425-64.
4. Sidransky et al., ( 1991 ) Science 252, 706-09.
5. Sidransky et al., (1992) Science 256, 102-05.
6. Smith-Ravin et al., (1995) Gut 36, 81-6.
7. Mills et al., (1995) J. Natl Cancer Inst 87, 1056-60.
8. Mao et al. (1994) Cancer Res 54, 1634-37.
9. Brossart et al., (1995) Caner Res 55, 4065-68.
10. Tada et al. (1993) Cancer Res 53, 2472-74.
11. Nawroz et al. (1996) Nature Med 2, 1035-37.
12. Hayashi et al., (1994) Cancer Res 54, 3853-56.
13. Sidransky, (1997) Science 278, 1054-59.
14. Koch et al. (1994) Arch Otolaryngol Head Neck Surg 120, 943-47.
15. Kumar et al, (1990) Science 248, 1101-04. 16. Jonason et al., (1996) Proc Natl Acad Sci USA 93, 14025-29.
17. Ananthaswamy et al. (1999) J Invest Dermatol 112, 763-68.
18. Bar-Eli et al. (1989) Blood 73, 281-83.
19. Collins et al. (1989) Blood 73, 1028-32. 20. Saiki et al. (1986) Nature 324, 163-66.
21. Bos et al. (1987) Nature 327, 293-97.
22. Dicker et al. (1990) Genes Chromosomes Cancer 1, 257-69.
23. Haque et al. (1998) Diagn Mol Pathol 7, 248-52.
24. Haliassos et al. (1989) Nucleic Acids Res 17, 8093-99. 25. Chen et al. ( 1997) Anal Biochem 244, 191 -94.
26. Cha et al. (1992) PCR Methods Appl 2, 14-20.
27. Jiang et al. (1989) Oncogene 4, 923-28.
28. Kahn et al. (1991) Oncogene 6, 1079-83.
29. Kahn et al. (1995) Methods Enzymol 255, 452-64. 30. Laken et al., (1998) Nature Biotechnol 16, 1352-56.
31. Whitcombe et al. (1998) Curr Opin Biotechnol 9, 602-08.
32. Day et al. (1999) Nucleic Acids Res 27, 1820-18.
33. Tyagi et al. (1998) Nature Biotechnol 16, 49-53.
34. Tyagi et al. (1996) Nat Biotechnol 14, 303-08. 35. Lee et al. (1993) Nucleic Acids Res 21, 3761-66.
36. Chiang et al. (1996) Genome Res 6, 1013-26.
37. Heid et al. (1996) Genome Res 6, 986-94.
38. Paris et al. (1998) Nucleic Acids Res 26, 3789-93.
39. Gibson et al. (1997) Clin Chem 43, 1336-41. 40. Chen et al. (1999) Genome Res 9, 492-98.
41. Szollosi et al. (1998) Cytometry 34, 159-79.
42. Cortopassi et al. (1992) Mutat Res 277, 239-49.
43. Monckton et al. (1991) Genomics 11, 465-67. 44. Chou et al. ( 1992) Nucleic Acids Res 20, 1717-23.
45. Kellogg et al. (1994) Biotechniques 16, 1134-37.
46. Li et al. (1988) Nature 335, 414-17.
47. Schmitt et al. (1994) Forensic Sci Int 66, 129-41.
48. Navidi et al. (1991) Hum Reprod 6, 836-49. 49. Zhang et al. (1992) Proc Natl Acad Sci USA 89, 5847-51.
50. Jeffreys et al. (1995) Electrophoresis 16, 1577-85.
51. Ruano et al. (1990) Proc Natl Acad Sci USA, 6296-300.
52. Sidransky et al. (1992) Nature 355, 846-47.
53. Parsons et al. (1995) Science 268, 738-40. 54. Lizardi et al. (1998) Nature Genet 19, 225-32.
55. Vogelstein (1989) Science 244, 207-11.
56. Marras et al. (1999) Genet Anal 14, 151-56.

Claims

1. A method for determining the ratio of a selected genetic sequence in a population of genetic sequences, comprising the steps of:
diluting nucleic acid template molecules in a biological sample to form a set comprising a plurality of assay samples;
amplifying the template molecules within the assay samples to form a population of amplified molecules in the assay samples of the set;
analyzing the amplified molecules in the assay samples of the set to determine a first number of assay samples which contain the selected genetic sequence and a second number of assay samples which contain a reference genetic sequence;
comparing the first number to the second number to ascertain a ratio which reflects the composition of the biological sample.
2. The method of claim 1 wherein the step of diluting is performed until at least one-tenth of the assay samples in the set comprise a number (N) of molecules such that 1/N is larger than the ratio of selected genetic sequences to total genetic sequences required for the step of analyzing to determine the presence of the selected genetic sequence.
3. The method of claim 1 wherein the step of diluting is performed until between 0.1 and 0.9 of the assay samples yield an amplification product when subjected to a polymerase chain reaction.
4. The method of claim 1 wherein the step of diluting is performed until all of the assay samples yield an amplification product when subjected to a polymerase chain reaction and each assay sample contains less than 10 nucleic acid template molecules containing the reference genetic sequence.
5. The method of claim 1 wherein the step of diluting is performed until all of the assay samples yield an amplification product when subjected to a polymerase chain reaction and each assay sample contains less than 100 nucleic acid template molecules containing the reference genetic sequence.
6. The method of claim 1 wherein the biological sample is cell- free.
7. The method of claim 1 wherein the number of assay samples within the set is greater than 10.
8. The method of claim 1 wherein the number of assay samples within the set is greater than 50.
9. The method of claim 1 wherein the number of assay samples within the set is greater than 100.
10. The method of claim 1 wherein the number of assay samples within the set is greater than 500.
11. The method of claim 1 wherein the number of assay samples within the set is greater than 1000.
12. The method of claim 1 wherein the step of amplifying and the step of analyzing are performed on assay samples in the same receptacle.
13. The method of claim 1 wherein a molecular beacon probe is used in the step of analyzing, wherein a molecular beacon probe is an oligonucleotide with a stem-loop structure having a photoluminescent dye at one of the 5' or 3' ends and a quenching agent at the opposite 5' or 3' end.
14. The method of claim 1 wherein the step of analyzing employs gel electrophoresis.
15. The method of claim 1 wherein the step of analyzing employs hybridization to at least one nucleic acid probe.
16. The method of claim 1 wherein the step of analyzing employs hybridization to at least two nucleic acid probe.
17. The method of claim 13 wherein two molecular beacon probes are used, each having a different photoluminescent dye.
18. The method of claim 13 wherein the molecular beacon probe detects a wild-type selected genetic sequence better than a mutant selected genetic sequence.
19. The method of claim 1 wherein the step of amplifying employs a single pair of primers.
20. The method of claim 1 wherein the step of amplifying employs a polymerase which is activated only after heating.
21. The method of claim 1 wherein the step of amplifying employs at least 40 cycles of heating and cooling.
22. The method of claim 1 wherein the step of amplifying employs at least 50 cycles of heating and cooling.
23. The method of claim 1 wherein the step of amplifying employs at least 60 cycles of heating and cooling.
24. The method of claim 1 wherein the biological sample is selected from the group consisting of stool, blood, and lymph nodes.
25. The method of claim 1 wherein the biological sample is blood or bone marrow of a leukemia or lymphoma patient who has received anti- cancer therapy.
26. The method of claim 1 wherein the selected genetic sequence is a translocated allele.
27. The method of claim 1 wherein the selected genetic sequence is a wild-type allele.
28. The method of claim 1 wherein the selected genetic sequence is within an amplicon which is amplified during neoplastic development.
29. The method of claim 1 wherein the selected genetic sequence is a rare exon sequence.
30. The method of claim 1 wherein the nucleic acid template molecules comprise cDNA of RNA transcripts and the selected genetic sequence is present on a cDNA of a first transcript and the reference genetic sequence is present on a cDNA of a second transcript.
31. The method of claim 1 wherein the selected genetic sequence comprises a first mutation and the reference genetic sequence comprises a second mutation.
32. The method of claim 1 wherein the selected genetic sequence and the reference genetic sequence are on distinct chromosomes.
33. A molecular beacon probe comprising:
an oligonucleotide with a stem-loop structure having a photoluminescent dye at one of the 5' or 3' ends and a quenching agent at the opposite 5' or 3' end, wherein the loop consists of 16 base pairs, wherein the loop has a Tm of 50-51 °C and the stem consists of 4 base pairs having a sequence 5'-CACG-3'.
34. The probe of claim 33 wherein the molecular beacon probe detects a wild-type selected genetic sequence better than a mutant selected genetic sequence.
35. The probe of claim 33 wherein the molecular beacon probe detects a mutant genetic sequence better than a wild-type genetic sequence.
36. A molecular beacon probe comprising:
an oligonucleotide with a stem-loop structure having a photoluminescent dye at one of the 5' or 3' ends and a quenching agent at the opposite 5' or 3' end, wherein the loop consists of 19-20 base pairs, wherein the loop has a Tm of 54-56 °C and the stem consists of 4 base pairs having a sequence 5'-CACG-3'.
37. A pair of molecular beacon probes comprising: a first molecular beacon probe which is an oligonucleotide with a stem-loop structure having a first photoluminescent dye at one of the 5' or
3' ends and a quenching agent at the opposite 5' or 3' end, wherein the loop consists of 16 base pairs having a Tm of 50-51 °C and the stem consists of
4 base pairs having a sequence 5'-CACG-3'; and a second molecular beacon probe which is an oligonucleotide with a stem-loop structure having a second photoluminescent dye at one of the 5' or 3' ends and a quenching agent at the opposite 5' or 3' end, wherein the loop consists of 19-20 base pairs having a Tm of 54-56°C and the stem consists of 4 base pairs having a sequence 5'-CACG-3'; wherein the first and the second photoluminescent dyes are distinct.
38. A method for determining the ratio of a selected genetic sequence in a population of genetic sequences, comprising the steps of: amplifying template molecules within a set comprising a plurality of assay samples to form a population of amplified molecules in each of the assay samples of the set; analyzing the amplified molecules in the assay samples of the set to determine a first number of assay samples which contain the selected genetic sequence and a second number of assay samples which contain a reference genetic sequence, wherein at least one-fiftieth of the assay samples in the set comprise a number (N) of molecules such that 1/N is larger than the ratio of selected genetic sequences to total genetic sequences required to determine the presence of the selected genetic sequence; comparing the first number to the second number to ascertain a ratio which reflects the composition of the biological sample.
39. The method of claim 38 wherein the number of assay samples within the set is greater than 10.
40. The method of claim 38 wherein the number of assay samples within the set is greater than 50.
41. The method of claim 38 wherein the number of assay samples within the set is greater than 100.
42. The method of claim 38 wherein the number of assay samples within the set is greater than 500.
43. The method of claim 38 wherein the number of assay samples within the set is greater than 1000.
44. The method of claim 38 wherein the step of amplifying and the step of analyzing are performed on assay samples in the same receptacle.
45. The method of claim 38 wherein a molecular beacon probe is used in the step of analyzing, wherein a molecular beacon probe is an oligonucleotide with a stem-loop structure having a photoluminescent dye at one of the 5' or 3' ends and a quenching agent at the opposite 5' or 3' end.
46. The method of claim 38 wherein the step of analyzing employs gel electrophoresis.
47. The method of claim 38 wherein the step of analyzing employs hybridization to at least one nucleic acid probe.
48. The method of claim 38 wherein the step of analyzing employs hybridization to at least two nucleic acid probe.
49. The method of claim 45 wherein two molecular beacon probes are used, each having a different photoluminescent dye.
50. The method of claim 45 wherein the molecular beacon probe detects a wild-type selected genetic sequence better than a mutant selected genetic sequence.
51. The method of claim 38 wherein the step of amplifying employs a single pair of primers.
52. The method of claim 38 wherein the step of amplifying employs a polymerase which is activated only after heating.
53. The method of claim 38 wherein the step of amplifying employs at least 40 cycles of heating and cooling.
54. The method of claim 38 wherein the step of amplifying employs at least 50 cycles of heating and cooling.
55. The method of claim 38 wherein the step of amplifying employs at least 60 cycles of heating and cooling.
56. The method of claim 38 wherein the template molecules are obtained from a body sample selected from the group consisting of stool, blood, and lymph nodes.
57. The method of claim 38 wherein the template molecules are obtained from a body sample of a leukemia or lymphoma patient who has received anti-cancer therapy, said body sample being selected from the group consisting of blood and bone marrow.
58. The method of claim 38 wherein the selected genetic sequence is a translocated allele.
59. The method of claim 38 wherein the selected genetic sequence is a wild-type allele.
60. The method of claim 38 wherein the selected genetic sequence is within an amplicon which is amplified during neoplastic development.
61. The method of claim 38 wherein the selected genetic sequence is a rare exon sequence.
62. The method of claim 38 wherein the nucleic acid template molecules comprise cDNA of RNA transcripts and the selected genetic sequence is present on a cDNA of a first transcript and the reference genetic sequence is present on a cDNA of a second transcript.
63. The method of claim 38 wherein the selected genetic sequence comprises a first mutation and the reference genetic sequence comprises a second mutation.
64. The method of claim 38 wherein the selected genetic sequence and the reference genetic sequence are on distinct chromosomes.
PCT/US2000/020740 1999-08-02 2000-07-31 Digital amplification WO2001009386A2 (en)

Priority Applications (6)

Application Number Priority Date Filing Date Title
JP2001513641A JP4653366B2 (en) 1999-08-02 2000-07-31 Digital amplification
CA2376929A CA2376929C (en) 1999-08-02 2000-07-31 Digital amplification
AU65028/00A AU781440B2 (en) 1999-08-02 2000-07-31 Digital amplification
EP00952304A EP1255856B1 (en) 1999-08-02 2000-07-31 Digital amplification
DE60018616T DE60018616T2 (en) 1999-08-02 2000-07-31 Digital Amplification
AT00952304T ATE290608T1 (en) 1999-08-02 2000-07-31 DIGITAL AMPLIFICATION

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US14679299P 1999-08-02 1999-08-02
US60/146,792 1999-08-02
US09/613,826 US6440706B1 (en) 1999-08-02 2000-07-11 Digital amplification
US09/613,826 2000-07-11

Publications (2)

Publication Number Publication Date
WO2001009386A2 true WO2001009386A2 (en) 2001-02-08
WO2001009386A3 WO2001009386A3 (en) 2002-09-12

Family

ID=26844308

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2000/020740 WO2001009386A2 (en) 1999-08-02 2000-07-31 Digital amplification

Country Status (8)

Country Link
US (8) US6440706B1 (en)
EP (1) EP1255856B1 (en)
JP (1) JP4653366B2 (en)
AT (1) ATE290608T1 (en)
AU (1) AU781440B2 (en)
CA (3) CA2376929C (en)
DE (1) DE60018616T2 (en)
WO (1) WO2001009386A2 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1456648A2 (en) * 2001-11-20 2004-09-15 Exact Sciences Corporation Automated sample preparation methods and devices
EP1476572A2 (en) * 2002-02-01 2004-11-17 The Johns Hopkins University, School of Medicine, Office of Technology Licencing Digital amplification for detection of mismatch repair deficient tumor cells
EP1523573A2 (en) * 2001-12-06 2005-04-20 The Johns Hopkins University School Of Medicine Disease detection by digital protein truncation assays
WO2007057669A2 (en) * 2005-11-15 2007-05-24 Genoid Kft. Method of detecting pathogens
WO2011085491A1 (en) * 2010-01-15 2011-07-21 The University Of British Columbia Multiplex amplification for the detection of nucleic acid variations
EP1907583B1 (en) 2005-06-15 2016-10-05 Complete Genomics Inc. Single molecule arrays for genetic and chemical analysis

Families Citing this family (270)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994003624A1 (en) * 1992-08-04 1994-02-17 Auerbach Jeffrey I Methods for the isothermal amplification of nucleic acid molecules
US6261808B1 (en) * 1992-08-04 2001-07-17 Replicon, Inc. Amplification of nucleic acid molecules via circular replicons
US6440706B1 (en) 1999-08-02 2002-08-27 Johns Hopkins University Digital amplification
US7654998B1 (en) 1999-08-23 2010-02-02 Aeris Therapeutics, Inc. Tissue volume reduction
US6610043B1 (en) 1999-08-23 2003-08-26 Bistech, Inc. Tissue volume reduction
US6306628B1 (en) 1999-08-25 2001-10-23 Ambergen, Incorporated Methods for the detection, analysis and isolation of Nascent proteins
US6586177B1 (en) 1999-09-08 2003-07-01 Exact Sciences Corporation Methods for disease detection
US7440684B2 (en) * 2001-04-12 2008-10-21 Spaid Michael A Method and apparatus for improved temperature control in microfluidic devices
ATE397095T1 (en) 2001-04-20 2008-06-15 Penn State Res Found METHOD FOR MANIPULATION OF NUCLEIC ACIDS
JP2002372533A (en) * 2001-06-13 2002-12-26 Kiwamu Akagi Examination method of blood, examination chip, and examination device
EP1444365A4 (en) * 2001-10-25 2005-07-20 Gorilla Genomics Inc Asymmetric pcr with nuclease-free polymerase or nuclease-resistant molecular beacons
US7691333B2 (en) 2001-11-30 2010-04-06 Fluidigm Corporation Microfluidic device and methods of using same
JP4355210B2 (en) 2001-11-30 2009-10-28 フルイディグム コーポレイション Microfluidic device and method of using microfluidic device
JP2006523082A (en) * 2002-03-01 2006-10-12 ラブジェン, インコーポレイテッド Rapid analysis of mutations in the genome
US6977162B2 (en) * 2002-03-01 2005-12-20 Ravgen, Inc. Rapid analysis of variations in a genome
US7727720B2 (en) * 2002-05-08 2010-06-01 Ravgen, Inc. Methods for detection of genetic disorders
US20070178478A1 (en) * 2002-05-08 2007-08-02 Dhallan Ravinder S Methods for detection of genetic disorders
US7442506B2 (en) * 2002-05-08 2008-10-28 Ravgen, Inc. Methods for detection of genetic disorders
CA2510166A1 (en) * 2002-12-20 2004-09-30 Caliper Life Sciences, Inc. Single molecule amplification and detection of dna
US20050042639A1 (en) * 2002-12-20 2005-02-24 Caliper Life Sciences, Inc. Single molecule amplification and detection of DNA length
US8275554B2 (en) * 2002-12-20 2012-09-25 Caliper Life Sciences, Inc. System for differentiating the lengths of nucleic acids of interest in a sample
WO2004071948A2 (en) * 2003-02-10 2004-08-26 Reveo, Inc. Micro-nozzle, nano-nozzle, manufacturing methods therefor, applications therefor
US7041481B2 (en) 2003-03-14 2006-05-09 The Regents Of The University Of California Chemical amplification based on fluid partitioning
US20060078893A1 (en) 2004-10-12 2006-04-13 Medical Research Council Compartmentalised combinatorial chemistry by microfluidic control
GB0307403D0 (en) 2003-03-31 2003-05-07 Medical Res Council Selection by compartmentalised screening
GB0307428D0 (en) 2003-03-31 2003-05-07 Medical Res Council Compartmentalised combinatorial chemistry
US20050145496A1 (en) 2003-04-03 2005-07-07 Federico Goodsaid Thermal reaction device and method for using the same
US7476363B2 (en) 2003-04-03 2009-01-13 Fluidigm Corporation Microfluidic devices and methods of using same
US7604965B2 (en) 2003-04-03 2009-10-20 Fluidigm Corporation Thermal reaction device and method for using the same
US8828663B2 (en) * 2005-03-18 2014-09-09 Fluidigm Corporation Thermal reaction device and method for using the same
US20050064452A1 (en) * 2003-04-25 2005-03-24 Schmid Matthew J. System and method for the detection of analytes
JP4571786B2 (en) * 2003-06-10 2010-10-27 則夫 清水 Target nucleic acid detection method
JP5183063B2 (en) * 2003-07-05 2013-04-17 ザ ジョンズ ホプキンス ユニバーシティ Methods and compositions for detection and enumeration of genetic variations
US20060024690A1 (en) * 2003-09-19 2006-02-02 Kao H P Normalization of data using controls
US20050221339A1 (en) 2004-03-31 2005-10-06 Medical Research Council Harvard University Compartmentalised screening by microfluidic control
WO2006041524A2 (en) * 2004-04-07 2006-04-20 Access Bio, Inc. Nucleic acid detection system
US7968287B2 (en) 2004-10-08 2011-06-28 Medical Research Council Harvard University In vitro evolution in microfluidic systems
WO2006094149A2 (en) * 2005-03-01 2006-09-08 Exact Sciences Corporation Methods and compositions for detecting adenoma
ES2313143T3 (en) 2005-04-06 2009-03-01 Maurice Stroun METHOD FOR THE CANCER DIAGNOSIS THROUGH CIRCULATING DNA AND RNA DETECTION.
EP2703499A1 (en) * 2005-06-02 2014-03-05 Fluidigm Corporation Analysis using microfluidic partitioning devices to generate single cell samples
US11111544B2 (en) 2005-07-29 2021-09-07 Natera, Inc. System and method for cleaning noisy genetic data and determining chromosome copy number
US10083273B2 (en) 2005-07-29 2018-09-25 Natera, Inc. System and method for cleaning noisy genetic data and determining chromosome copy number
US10081839B2 (en) 2005-07-29 2018-09-25 Natera, Inc System and method for cleaning noisy genetic data and determining chromosome copy number
US8532930B2 (en) 2005-11-26 2013-09-10 Natera, Inc. Method for determining the number of copies of a chromosome in the genome of a target individual using genetic data from genetically related individuals
US9424392B2 (en) 2005-11-26 2016-08-23 Natera, Inc. System and method for cleaning noisy genetic data from target individuals using genetic data from genetically related individuals
US11111543B2 (en) 2005-07-29 2021-09-07 Natera, Inc. System and method for cleaning noisy genetic data and determining chromosome copy number
US7713288B2 (en) * 2005-08-03 2010-05-11 Applied Spine Technologies, Inc. Spring junction and assembly methods for spinal device
US20070111234A1 (en) * 2005-09-12 2007-05-17 Christian Birkner Detection of biological DNA
WO2007081387A1 (en) 2006-01-11 2007-07-19 Raindance Technologies, Inc. Microfluidic devices, methods of use, and kits for performing diagnostics
WO2007087312A2 (en) * 2006-01-23 2007-08-02 Population Genetics Technologies Ltd. Molecular counting
DK3002338T3 (en) 2006-02-02 2019-08-05 Univ Leland Stanford Junior NON-INVASIVE FEET GENETIC SCREENING BY DIGITAL ANALYSIS
US9562837B2 (en) 2006-05-11 2017-02-07 Raindance Technologies, Inc. Systems for handling microfludic droplets
US20080003142A1 (en) 2006-05-11 2008-01-03 Link Darren R Microfluidic devices
US9074242B2 (en) * 2010-02-12 2015-07-07 Raindance Technologies, Inc. Digital analyte analysis
EP2589668A1 (en) 2006-06-14 2013-05-08 Verinata Health, Inc Rare cell analysis using sample splitting and DNA tags
US20080070792A1 (en) 2006-06-14 2008-03-20 Roland Stoughton Use of highly parallel snp genotyping for fetal diagnosis
US8137912B2 (en) * 2006-06-14 2012-03-20 The General Hospital Corporation Methods for the diagnosis of fetal abnormalities
US20080050739A1 (en) 2006-06-14 2008-02-28 Roland Stoughton Diagnosis of fetal abnormalities using polymorphisms including short tandem repeats
US9012390B2 (en) 2006-08-07 2015-04-21 Raindance Technologies, Inc. Fluorocarbon emulsion stabilizing surfactants
US8772046B2 (en) 2007-02-06 2014-07-08 Brandeis University Manipulation of fluids and reactions in microfluidic systems
WO2008130623A1 (en) 2007-04-19 2008-10-30 Brandeis University Manipulation of fluids, fluid components and reactions in microfluidic systems
CA3176319A1 (en) 2007-07-23 2009-01-29 The Chinese University Of Hong Kong Analyzing tumor dna in a cell-free sample
US20100112590A1 (en) * 2007-07-23 2010-05-06 The Chinese University Of Hong Kong Diagnosing Fetal Chromosomal Aneuploidy Using Genomic Sequencing With Enrichment
ES2380844T3 (en) 2007-09-07 2012-05-18 Fluidigm Corporation Determination of the variation in the number of copies, methods and systems
US8741815B2 (en) 2008-02-19 2014-06-03 Intelligent Bio Systems, Inc. Methods and devices for amplification of nucleic acid
GB2469424B (en) * 2008-02-19 2013-04-17 Intelligent Bio Systems Inc Non-emulsion methods and masked biomolecules
US8709726B2 (en) * 2008-03-11 2014-04-29 Sequenom, Inc. Nucleic acid-based tests for prenatal gender determination
EP4047367A1 (en) 2008-07-18 2022-08-24 Bio-Rad Laboratories, Inc. Method for detecting target analytes with droplet libraries
CN104732118B (en) 2008-08-04 2017-08-22 纳特拉公司 Allele calls the method with ploidy calling
US8962247B2 (en) 2008-09-16 2015-02-24 Sequenom, Inc. Processes and compositions for methylation-based enrichment of fetal nucleic acid from a maternal sample useful for non invasive prenatal diagnoses
US8476013B2 (en) 2008-09-16 2013-07-02 Sequenom, Inc. Processes and compositions for methylation-based acid enrichment of fetal nucleic acid from a maternal sample useful for non-invasive prenatal diagnoses
EP2329021B1 (en) 2008-09-16 2016-08-10 Sequenom, Inc. Processes and compositions for methylation-based enrichment of fetal nucleic acid from a maternal sample useful for non invasive prenatal diagnoses
SI2334812T1 (en) 2008-09-20 2017-05-31 The Board of Trustees of the Leland Stanford Junior University Office of the General Counsel Building 170 Noninvasive diagnosis of fetal aneuploidy by sequencing
US9492797B2 (en) 2008-09-23 2016-11-15 Bio-Rad Laboratories, Inc. System for detection of spaced droplets
US8951939B2 (en) 2011-07-12 2015-02-10 Bio-Rad Laboratories, Inc. Digital assays with multiplexed detection of two or more targets in the same optical channel
US10512910B2 (en) 2008-09-23 2019-12-24 Bio-Rad Laboratories, Inc. Droplet-based analysis method
WO2011120024A1 (en) 2010-03-25 2011-09-29 Quantalife, Inc. Droplet generation for droplet-based assays
US9156010B2 (en) 2008-09-23 2015-10-13 Bio-Rad Laboratories, Inc. Droplet-based assay system
US9417190B2 (en) 2008-09-23 2016-08-16 Bio-Rad Laboratories, Inc. Calibrations and controls for droplet-based assays
US9132394B2 (en) 2008-09-23 2015-09-15 Bio-Rad Laboratories, Inc. System for detection of spaced droplets
US8709762B2 (en) 2010-03-02 2014-04-29 Bio-Rad Laboratories, Inc. System for hot-start amplification via a multiple emulsion
US11130128B2 (en) 2008-09-23 2021-09-28 Bio-Rad Laboratories, Inc. Detection method for a target nucleic acid
US9764322B2 (en) 2008-09-23 2017-09-19 Bio-Rad Laboratories, Inc. System for generating droplets with pressure monitoring
US8633015B2 (en) 2008-09-23 2014-01-21 Bio-Rad Laboratories, Inc. Flow-based thermocycling system with thermoelectric cooler
US8748103B2 (en) 2008-11-07 2014-06-10 Sequenta, Inc. Monitoring health and disease status using clonotype profiles
SG195652A1 (en) 2008-11-07 2013-12-30 Sequenta Inc Methods of monitoring conditions by sequence analysis
US9365901B2 (en) 2008-11-07 2016-06-14 Adaptive Biotechnologies Corp. Monitoring immunoglobulin heavy chain evolution in B-cell acute lymphoblastic leukemia
US9506119B2 (en) 2008-11-07 2016-11-29 Adaptive Biotechnologies Corp. Method of sequence determination using sequence tags
US9528160B2 (en) 2008-11-07 2016-12-27 Adaptive Biotechnolgies Corp. Rare clonotypes and uses thereof
US8628927B2 (en) 2008-11-07 2014-01-14 Sequenta, Inc. Monitoring health and disease status using clonotype profiles
DK3059337T3 (en) 2009-01-15 2019-07-22 Adaptive Biotechnologies Corp Adaptive immunity profiling and methods for producing monoclonal antibodies
US8528589B2 (en) 2009-03-23 2013-09-10 Raindance Technologies, Inc. Manipulation of microfluidic droplets
GB0904957D0 (en) 2009-03-23 2009-05-06 Univ Erasmus Medical Ct Tumour gene profile
US9309557B2 (en) 2010-12-17 2016-04-12 Life Technologies Corporation Nucleic acid amplification
US20120156728A1 (en) 2010-12-17 2012-06-21 Life Technologies Corporation Clonal amplification of nucleic acid on solid surface with template walking
US9309566B2 (en) 2010-12-17 2016-04-12 Life Technologies Corporation Methods, compositions, systems, apparatuses and kits for nucleic acid amplification
US9334531B2 (en) 2010-12-17 2016-05-10 Life Technologies Corporation Nucleic acid amplification
JP2012531202A (en) 2009-06-25 2012-12-10 フレッド ハチンソン キャンサー リサーチ センター How to measure adaptive immunity
JP6155418B2 (en) 2009-09-02 2017-07-05 バイオ−ラッド・ラボラトリーズ・インコーポレーテッド System for mixing fluids by combining multiple emulsions
ES2640776T3 (en) 2009-09-30 2017-11-06 Natera, Inc. Methods for non-invasively calling prenatal ploidy
WO2011042564A1 (en) 2009-10-09 2011-04-14 Universite De Strasbourg Labelled silica-based nanomaterial with enhanced properties and uses thereof
JP2013511991A (en) * 2009-11-25 2013-04-11 クアンタライフ, インコーポレイテッド Methods and compositions for detecting genetic material
US9315857B2 (en) 2009-12-15 2016-04-19 Cellular Research, Inc. Digital counting of individual molecules by stochastic attachment of diverse label-tags
US8835358B2 (en) 2009-12-15 2014-09-16 Cellular Research, Inc. Digital counting of individual molecules by stochastic attachment of diverse labels
EP2516680B1 (en) 2009-12-22 2016-04-06 Sequenom, Inc. Processes and kits for identifying aneuploidy
EP2517025B1 (en) 2009-12-23 2019-11-27 Bio-Rad Laboratories, Inc. Methods for reducing the exchange of molecules between droplets
US20120010085A1 (en) 2010-01-19 2012-01-12 Rava Richard P Methods for determining fraction of fetal nucleic acids in maternal samples
US10388403B2 (en) 2010-01-19 2019-08-20 Verinata Health, Inc. Analyzing copy number variation in the detection of cancer
US9260745B2 (en) 2010-01-19 2016-02-16 Verinata Health, Inc. Detecting and classifying copy number variation
US20120100548A1 (en) 2010-10-26 2012-04-26 Verinata Health, Inc. Method for determining copy number variations
WO2011091063A1 (en) 2010-01-19 2011-07-28 Verinata Health, Inc. Partition defined detection methods
WO2011091046A1 (en) * 2010-01-19 2011-07-28 Verinata Health, Inc. Identification of polymorphic sequences in mixtures of genomic dna by whole genome sequencing
DK3260555T3 (en) 2010-01-19 2019-01-21 Verinata Health Inc Hitherto UNKNOWN PROTOCOL FOR PREPARING SEQUENCE LIBRARIES
US9323888B2 (en) 2010-01-19 2016-04-26 Verinata Health, Inc. Detecting and classifying copy number variation
US20110312503A1 (en) 2010-01-23 2011-12-22 Artemis Health, Inc. Methods of fetal abnormality detection
US9399797B2 (en) 2010-02-12 2016-07-26 Raindance Technologies, Inc. Digital analyte analysis
US10351905B2 (en) 2010-02-12 2019-07-16 Bio-Rad Laboratories, Inc. Digital analyte analysis
US9366632B2 (en) 2010-02-12 2016-06-14 Raindance Technologies, Inc. Digital analyte analysis
US8399198B2 (en) 2010-03-02 2013-03-19 Bio-Rad Laboratories, Inc. Assays with droplets transformed into capsules
EP2556170A4 (en) 2010-03-25 2014-01-01 Quantalife Inc Droplet transport system for detection
CA2767113A1 (en) 2010-03-25 2011-09-29 Bio-Rad Laboratories, Inc. Detection system for droplet-based assays
JP6158080B2 (en) 2010-05-06 2017-07-05 アダプティヴ バイオテクノロジーズ コーポレーション Health and disease status monitoring using chronotype profiles
US11339429B2 (en) 2010-05-18 2022-05-24 Natera, Inc. Methods for non-invasive prenatal ploidy calling
US11332793B2 (en) 2010-05-18 2022-05-17 Natera, Inc. Methods for simultaneous amplification of target loci
US11939634B2 (en) 2010-05-18 2024-03-26 Natera, Inc. Methods for simultaneous amplification of target loci
US11326208B2 (en) 2010-05-18 2022-05-10 Natera, Inc. Methods for nested PCR amplification of cell-free DNA
US11408031B2 (en) 2010-05-18 2022-08-09 Natera, Inc. Methods for non-invasive prenatal paternity testing
US11332785B2 (en) 2010-05-18 2022-05-17 Natera, Inc. Methods for non-invasive prenatal ploidy calling
US20190010543A1 (en) 2010-05-18 2019-01-10 Natera, Inc. Methods for simultaneous amplification of target loci
CA2798758C (en) 2010-05-18 2019-05-07 Natera, Inc. Methods for non-invasive prenatal ploidy calling
US9677118B2 (en) 2014-04-21 2017-06-13 Natera, Inc. Methods for simultaneous amplification of target loci
US11322224B2 (en) 2010-05-18 2022-05-03 Natera, Inc. Methods for non-invasive prenatal ploidy calling
US10316362B2 (en) 2010-05-18 2019-06-11 Natera, Inc. Methods for simultaneous amplification of target loci
SG186787A1 (en) 2010-07-23 2013-02-28 Esoterix Genetic Lab Llc Identification of differentially represented fetal or maternal genomic regions and uses thereof
ES2690753T3 (en) 2010-09-21 2018-11-22 Agilent Technologies, Inc. Increased confidence in allele identifications with molecular count
WO2012045012A2 (en) 2010-09-30 2012-04-05 Raindance Technologies, Inc. Sandwich assays in droplets
EP3574990B1 (en) 2010-11-01 2022-04-06 Bio-Rad Laboratories, Inc. System for forming emulsions
AU2011348100B2 (en) 2010-12-22 2016-08-25 Natera, Inc. Methods for non-invasive prenatal paternity testing
ES2860945T3 (en) * 2010-12-27 2021-10-05 Abbott Molecular Inc Quantification of high titer samples by digital PCR
US10152568B2 (en) 2011-01-05 2018-12-11 The Chinese University Of Hong Kong Noninvasive prenatal genotyping of fetal sex chromosomes
EP3795699B1 (en) 2011-02-02 2023-12-20 Exact Sciences Corporation Digital sequence analysis of dna methylation
CA2824387C (en) 2011-02-09 2019-09-24 Natera, Inc. Methods for non-invasive prenatal ploidy calling
CA2826748C (en) 2011-02-09 2020-08-04 Bio-Rad Laboratories, Inc. Method of detecting variations in copy number of a target nucleic acid
EP3859011A1 (en) 2011-02-11 2021-08-04 Bio-Rad Laboratories, Inc. Methods for forming mixed droplets
WO2012112804A1 (en) 2011-02-18 2012-08-23 Raindance Technoligies, Inc. Compositions and methods for molecular labeling
AU2012231098B2 (en) 2011-03-18 2016-09-29 Bio-Rad Laboratories, Inc. Multiplexed digital assays with combinatorial use of signals
US9260753B2 (en) 2011-03-24 2016-02-16 President And Fellows Of Harvard College Single cell nucleic acid detection and analysis
DK2697392T3 (en) 2011-04-12 2016-03-29 Verinata Health Inc SOLUTION OF GENOME FRACTIONS USING Polymorphism COUNTS
GB2484764B (en) 2011-04-14 2012-09-05 Verinata Health Inc Normalizing chromosomes for the determination and verification of common and rare chromosomal aneuploidies
US9411937B2 (en) 2011-04-15 2016-08-09 Verinata Health, Inc. Detecting and classifying copy number variation
CA2834291A1 (en) 2011-04-25 2012-11-01 Biorad Laboratories, Inc. Methods and compositions for nucleic acid analysis
CA2834218C (en) 2011-04-29 2021-02-16 Sequenom, Inc. Quantification of a minority nucleic acid species using inhibitory oligonucleotides
GB201107466D0 (en) 2011-05-05 2011-06-15 Loktionov Alexandre Device and method for non-invasive collection of colorectal mucocellular layer and disease detection
WO2012167142A2 (en) 2011-06-02 2012-12-06 Raindance Technolgies, Inc. Enzyme quantification
US8841071B2 (en) 2011-06-02 2014-09-23 Raindance Technologies, Inc. Sample multiplexing
US20130178378A1 (en) * 2011-06-09 2013-07-11 Andrew C. Hatch Multiplex digital pcr
US8658430B2 (en) 2011-07-20 2014-02-25 Raindance Technologies, Inc. Manipulating droplet size
WO2013019751A1 (en) 2011-07-29 2013-02-07 Bio-Rad Laboratories, Inc., Library characterization by digital assay
US10385475B2 (en) 2011-09-12 2019-08-20 Adaptive Biotechnologies Corp. Random array sequencing of low-complexity libraries
CA2853088C (en) 2011-10-21 2018-03-13 Adaptive Biotechnologies Corporation Quantification of adaptive immune cell genomes in a complex mixture of cells
GB201120711D0 (en) 2011-12-01 2012-01-11 Univ Erasmus Medical Ct Method for classifying tumour cells
ES2867924T3 (en) 2011-12-09 2021-10-21 Adaptive Biotechnologies Corp Diagnosis of Lymphoid Malignant Tumors and Detection of Minimal Residual Disease
US9499865B2 (en) 2011-12-13 2016-11-22 Adaptive Biotechnologies Corp. Detection and measurement of tissue-infiltrating lymphocytes
US9222115B2 (en) 2011-12-30 2015-12-29 Abbott Molecular, Inc. Channels with cross-sectional thermal gradients
JP2013158290A (en) * 2012-02-03 2013-08-19 Gunma Univ Nucleic acid detection probe bonded with silylated fluorescent, and method for detecting nucleic acid by the same
EP2820174B1 (en) 2012-02-27 2019-12-25 The University of North Carolina at Chapel Hill Methods and uses for molecular tags
WO2013130674A1 (en) 2012-02-27 2013-09-06 Cellular Research, Inc. Compositions and kits for molecular counting
US9670529B2 (en) 2012-02-28 2017-06-06 Population Genetics Technologies Ltd. Method for attaching a counter sequence to a nucleic acid sample
EP4155401A1 (en) 2012-03-02 2023-03-29 Sequenom, Inc. Methods and processes for non-invasive assessment of genetic variations
EP2823060B1 (en) 2012-03-05 2018-02-14 Adaptive Biotechnologies Corporation Determining paired immune receptor chains from frequency matched subunits
US9892230B2 (en) 2012-03-08 2018-02-13 The Chinese University Of Hong Kong Size-based analysis of fetal or tumor DNA fraction in plasma
EP2825675B1 (en) 2012-03-13 2017-12-27 Patel, Abhijit Ajit Measurement of nucleic acid variants using highly-multiplexed error-suppressed deep sequencing
CN107841543B (en) 2012-04-06 2021-12-31 香港中文大学 Non-invasive prenatal diagnosis of fetal trisomy by allele ratio analysis using targeted massively parallel sequencing
WO2013155531A2 (en) 2012-04-13 2013-10-17 Bio-Rad Laboratories, Inc. Sample holder with a well having a wicking promoter
EP2844768B1 (en) 2012-04-30 2019-03-13 Raindance Technologies, Inc. Digital analyte analysis
EP2831276B1 (en) 2012-05-08 2016-04-20 Adaptive Biotechnologies Corporation Compositions and method for measuring and calibrating amplification bias in multiplexed pcr reactions
US9920361B2 (en) 2012-05-21 2018-03-20 Sequenom, Inc. Methods and compositions for analyzing nucleic acid
CN104471077B (en) 2012-05-21 2017-05-24 富鲁达公司 Single-particle analysis of particle populations
CA2878979C (en) 2012-07-13 2021-09-14 Sequenom, Inc. Processes and compositions for methylation-based enrichment of fetal nucleic acid from a maternal sample useful for non-invasive prenatal diagnoses
US20160040229A1 (en) 2013-08-16 2016-02-11 Guardant Health, Inc. Systems and methods to detect rare mutations and copy number variation
CA3190199A1 (en) 2012-09-04 2014-03-13 Guardant Health, Inc. Systems and methods to detect rare mutations and copy number variation
US10876152B2 (en) 2012-09-04 2020-12-29 Guardant Health, Inc. Systems and methods to detect rare mutations and copy number variation
US11913065B2 (en) 2012-09-04 2024-02-27 Guardent Health, Inc. Systems and methods to detect rare mutations and copy number variation
US20160002731A1 (en) 2012-10-01 2016-01-07 Adaptive Biotechnologies Corporation Immunocompetence assessment by adaptive immune receptor diversity and clonality characterization
US10150996B2 (en) 2012-10-19 2018-12-11 Adaptive Biotechnologies Corp. Quantification of adaptive immune cell genomes in a complex mixture of cells
US10975423B2 (en) 2013-03-11 2021-04-13 Elitechgroup, Inc. Methods for true isothermal strand displacement amplification
EP3597774A1 (en) 2013-03-13 2020-01-22 Sequenom, Inc. Primers for dna methylation analysis
EP2986762B1 (en) 2013-04-19 2019-11-06 Bio-Rad Laboratories, Inc. Digital analyte analysis
ES2654631T3 (en) 2013-05-13 2018-02-14 Elitechgroup B.V. Digital PCR in drops with short minor groove probes
PL3004388T5 (en) 2013-05-29 2023-10-09 Chronix Biomedical Detection and quantification of donor cell-free dna in the circulation of organ transplant recipients
US9708657B2 (en) 2013-07-01 2017-07-18 Adaptive Biotechnologies Corp. Method for generating clonotype profiles using sequence tags
WO2015013681A1 (en) 2013-07-25 2015-01-29 Bio-Rad Laboratories, Inc. Genetic assays
WO2015031691A1 (en) 2013-08-28 2015-03-05 Cellular Research, Inc. Massively parallel single cell analysis
US10262755B2 (en) 2014-04-21 2019-04-16 Natera, Inc. Detecting cancer mutations and aneuploidy in chromosomal segments
US10577655B2 (en) 2013-09-27 2020-03-03 Natera, Inc. Cell free DNA diagnostic testing standards
US9499870B2 (en) 2013-09-27 2016-11-22 Natera, Inc. Cell free DNA diagnostic testing standards
US11901041B2 (en) 2013-10-04 2024-02-13 Bio-Rad Laboratories, Inc. Digital analysis of nucleic acid modification
CN105745528A (en) 2013-10-07 2016-07-06 赛卢拉研究公司 Methods and systems for digitally counting features on arrays
US9944977B2 (en) 2013-12-12 2018-04-17 Raindance Technologies, Inc. Distinguishing rare variations in a nucleic acid sequence from a sample
ES2784450T3 (en) 2013-12-28 2020-09-25 Guardant Health Inc Methods and systems to detect genetic variants
WO2015103367A1 (en) 2013-12-31 2015-07-09 Raindance Technologies, Inc. System and method for detection of rna species
CA2941612A1 (en) 2014-03-05 2015-09-11 Adaptive Biotechnologies Corporation Methods using randomer-containing synthetic molecules
WO2015138774A1 (en) 2014-03-13 2015-09-17 Sequenom, Inc. Methods and processes for non-invasive assessment of genetic variations
US10066265B2 (en) 2014-04-01 2018-09-04 Adaptive Biotechnologies Corp. Determining antigen-specific t-cells
EP3134541B1 (en) 2014-04-21 2020-08-19 Natera, Inc. Detecting copy number variations (cnv) of chromosomal segments in cancer
WO2015173651A1 (en) 2014-05-14 2015-11-19 Mark Davies Microfluidic device with channel plates
PT3201361T (en) 2014-10-01 2020-05-13 Chronix Biomedical Methods of quantifying cell-free dna
US10392663B2 (en) 2014-10-29 2019-08-27 Adaptive Biotechnologies Corp. Highly-multiplexed simultaneous detection of nucleic acids encoding paired adaptive immune receptor heterodimers from a large number of samples
EP3216852A4 (en) 2014-11-04 2018-09-26 Toppan Printing Co., Ltd. Method for introducing nucleic acid, method for detecting nucleic acid, method for analyzing biological component, array device for biological component assay, and kit for analyzing biological component
US10246701B2 (en) 2014-11-14 2019-04-02 Adaptive Biotechnologies Corp. Multiplexed digital quantitation of rearranged lymphoid receptors in a complex mixture
US11066705B2 (en) 2014-11-25 2021-07-20 Adaptive Biotechnologies Corporation Characterization of adaptive immune response to vaccination or infection using immune repertoire sequencing
EP3242938B1 (en) * 2015-01-09 2020-01-08 Bio-Rad Laboratories, Inc. Detection of genome editing
US10364467B2 (en) 2015-01-13 2019-07-30 The Chinese University Of Hong Kong Using size and number aberrations in plasma DNA for detecting cancer
EP3259371B1 (en) 2015-02-19 2020-09-02 Becton, Dickinson and Company High-throughput single-cell analysis combining proteomic and genomic information
CA2976580A1 (en) 2015-02-24 2016-09-01 Adaptive Biotechnologies Corp. Methods for diagnosing infectious disease and determining hla status using immune repertoire sequencing
US9727810B2 (en) 2015-02-27 2017-08-08 Cellular Research, Inc. Spatially addressable molecular barcoding
CN107406888A (en) 2015-03-30 2017-11-28 赛卢拉研究公司 For combining the method and composition of bar coding
US11041202B2 (en) 2015-04-01 2021-06-22 Adaptive Biotechnologies Corporation Method of identifying human compatible T cell receptors specific for an antigenic target
EP3286326A1 (en) 2015-04-23 2018-02-28 Cellular Research, Inc. Methods and compositions for whole transcriptome amplification
US11479812B2 (en) 2015-05-11 2022-10-25 Natera, Inc. Methods and compositions for determining ploidy
CN107636167B (en) * 2015-05-18 2022-09-02 传奇诊断股份公司 Detection of target nucleic acids and variants
CN104846103A (en) * 2015-05-26 2015-08-19 南京杰蒙生物技术有限公司 Application of MDPCR (multiple digital PCR (polymerase chain reaction)) technology to chromosome aneuploidy screening
WO2016196229A1 (en) 2015-06-01 2016-12-08 Cellular Research, Inc. Methods for rna quantification
US11302416B2 (en) 2015-09-02 2022-04-12 Guardant Health Machine learning for somatic single nucleotide variant detection in cell-free tumor nucleic acid sequencing applications
JP7009993B2 (en) 2015-09-08 2022-01-26 凸版印刷株式会社 Biomaterial detection method and biomaterial introduction method
US10647981B1 (en) 2015-09-08 2020-05-12 Bio-Rad Laboratories, Inc. Nucleic acid library generation methods and compositions
JP6940484B2 (en) 2015-09-11 2021-09-29 セルラー リサーチ, インコーポレイテッド Methods and compositions for library normalization
WO2017106768A1 (en) 2015-12-17 2017-06-22 Guardant Health, Inc. Methods to determine tumor gene copy number by analysis of cell-free dna
EP3452614B1 (en) 2016-05-02 2023-06-28 Becton, Dickinson and Company Accurate molecular barcoding
US10301677B2 (en) 2016-05-25 2019-05-28 Cellular Research, Inc. Normalization of nucleic acid libraries
CN109074430B (en) 2016-05-26 2022-03-29 贝克顿迪金森公司 Molecular marker counting adjustment method
US10202641B2 (en) 2016-05-31 2019-02-12 Cellular Research, Inc. Error correction in amplification of samples
US10640763B2 (en) 2016-05-31 2020-05-05 Cellular Research, Inc. Molecular indexing of internal sequences
WO2018031625A2 (en) 2016-08-09 2018-02-15 Integrated Dna Technologies, Inc. Rnase h mutants in an emulsion
US10428325B1 (en) 2016-09-21 2019-10-01 Adaptive Biotechnologies Corporation Identification of antigen-specific B cell receptors
AU2017331459B2 (en) 2016-09-26 2023-04-13 Becton, Dickinson And Company Measurement of protein expression using reagents with barcoded oligonucleotide sequences
US9850523B1 (en) 2016-09-30 2017-12-26 Guardant Health, Inc. Methods for multi-resolution analysis of cell-free nucleic acids
CN109642250A (en) 2016-09-30 2019-04-16 夸登特健康公司 The method of multiresolution analysis for cell-free nucleic acid
WO2018067517A1 (en) 2016-10-04 2018-04-12 Natera, Inc. Methods for characterizing copy number variation using proximity-litigation sequencing
AU2017359047A1 (en) 2016-11-08 2019-05-02 Becton, Dickinson And Company Methods for cell label classification
CN109952612B (en) 2016-11-08 2023-12-01 贝克顿迪金森公司 Method for classifying expression profiles
US10011870B2 (en) 2016-12-07 2018-07-03 Natera, Inc. Compositions and methods for identifying nucleic acid molecules
US10870891B2 (en) 2017-01-05 2020-12-22 Biodesix, Inc. Diagnostic test system for specific, sensitive and reproducible detection of circulating nucleic acids in whole blood
US10722880B2 (en) 2017-01-13 2020-07-28 Cellular Research, Inc. Hydrophilic coating of fluidic channels
CN110382708A (en) 2017-02-01 2019-10-25 赛卢拉研究公司 Selective amplification is carried out using blocking property oligonucleotides
EP3585889A1 (en) 2017-02-21 2020-01-01 Natera, Inc. Compositions, methods, and kits for isolating nucleic acids
CN110719959B (en) 2017-06-05 2021-08-06 贝克顿迪金森公司 Sample indexing for single cells
JP6897655B2 (en) * 2017-11-13 2021-07-07 株式会社リコー Device and inspection method
US11254980B1 (en) 2017-11-29 2022-02-22 Adaptive Biotechnologies Corporation Methods of profiling targeted polynucleotides while mitigating sequencing depth requirements
WO2019126209A1 (en) 2017-12-19 2019-06-27 Cellular Research, Inc. Particles associated with oligonucleotides
JP7348603B2 (en) 2018-04-02 2023-09-21 エニュメラ・モレキュラー・インコーポレイテッド Methods, systems, and compositions for counting nucleic acid molecules
AU2019262048A1 (en) 2018-05-03 2020-11-12 Becton, Dickinson And Company High throughput multiomics sample analysis
JP7358388B2 (en) 2018-05-03 2023-10-10 ベクトン・ディキンソン・アンド・カンパニー Molecular barcoding at opposite transcript ends
US11525159B2 (en) 2018-07-03 2022-12-13 Natera, Inc. Methods for detection of donor-derived cell-free DNA
WO2020072380A1 (en) 2018-10-01 2020-04-09 Cellular Research, Inc. Determining 5' transcript sequences
JP2022506546A (en) 2018-11-08 2022-01-17 ベクトン・ディキンソン・アンド・カンパニー Single-cell whole transcriptome analysis using random priming
US11492660B2 (en) 2018-12-13 2022-11-08 Becton, Dickinson And Company Selective extension in single cell whole transcriptome analysis
JP2022517963A (en) 2019-01-10 2022-03-11 アイオバンス バイオセラピューティクス,インコーポレイテッド Systems and methods for monitoring the clonality and persistence of adoptive cell therapy
US11371076B2 (en) 2019-01-16 2022-06-28 Becton, Dickinson And Company Polymerase chain reaction normalization through primer titration
EP4242322A3 (en) 2019-01-23 2023-09-20 Becton, Dickinson and Company Oligonucleotides associated with antibodies
CN113661249A (en) 2019-01-31 2021-11-16 夸登特健康公司 Compositions and methods for isolating cell-free DNA
EP3947718A4 (en) 2019-04-02 2022-12-21 Enumera Molecular, Inc. Methods, systems, and compositions for counting nucleic acid molecules
US11939622B2 (en) 2019-07-22 2024-03-26 Becton, Dickinson And Company Single cell chromatin immunoprecipitation sequencing assay
WO2021080629A1 (en) 2019-10-23 2021-04-29 Elitechgroup, Inc. Methods for true isothermal strand displacement amplification
WO2021092386A1 (en) 2019-11-08 2021-05-14 Becton Dickinson And Company Using random priming to obtain full-length v(d)j information for immune repertoire sequencing
EP4090763A1 (en) 2020-01-13 2022-11-23 Becton Dickinson and Company Methods and compositions for quantitation of proteins and rna
CN115605614A (en) 2020-05-14 2023-01-13 贝克顿迪金森公司(Us) Primers for immune repertoire profiling
US11932901B2 (en) 2020-07-13 2024-03-19 Becton, Dickinson And Company Target enrichment using nucleic acid probes for scRNAseq
EP4214243A1 (en) 2020-09-21 2023-07-26 Progenity, Inc. Compositions and methods for isolation of cell-free dna
EP4247967A1 (en) 2020-11-20 2023-09-27 Becton, Dickinson and Company Profiling of highly expressed and lowly expressed proteins

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0643140A1 (en) * 1993-09-13 1995-03-15 Canon Kabushiki Kaisha Determination of nucleic acid by PCR, measurement of number of microbial cells, genes, or gene-copies by PCR, and measuring-kit employed for the same
WO1995013399A1 (en) * 1993-11-12 1995-05-18 The Public Health Research Institute Of The City Of New York, Inc. Hybridization probes for nucleic acid detection, universal stems, methods and kits
US5804383A (en) * 1992-08-21 1998-09-08 The Regents Of The University Of California Method and assay for detection of the expression of allele-specific mutations by allele-specific in situ reverse transcriptase polymerase chain reaction
US5858663A (en) * 1992-10-01 1999-01-12 Life Technologies, Inc. Method for the rapid and ultra-sensitive detection of leukemic cells
WO1999013113A1 (en) * 1997-09-12 1999-03-18 The Public Health Research Institute Of The City Of New York, Inc. Non-competitive co-amplification methods

Family Cites Families (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5213961A (en) * 1989-08-31 1993-05-25 Brigham And Women's Hospital Accurate quantitation of RNA and DNA by competetitive polymerase chain reaction
US5712125A (en) 1990-07-24 1998-01-27 Cemv Bioteknik Ab Competitive PCR for quantitation of DNA
WO1992021694A1 (en) * 1991-06-06 1992-12-10 Baylor College Of Medicine Molecular diagnosis of autosomal dominant charcot-marie-tooth disease
DE69233285T2 (en) 1991-08-02 2004-11-25 bioMérieux B.V. Quantification of nucleic acids
ES2161715T3 (en) 1992-03-04 2001-12-16 Univ California COMPARATIVE GENOMIC HYBRIDIZATION (HGC).
US5518901A (en) * 1993-04-19 1996-05-21 Murtagh; James J. Methods for adapting nucleic acid for detection, sequencing, and cloning using exonuclease
WO1995006750A1 (en) 1993-09-03 1995-03-09 Cellpro, Incorporated Methods for quantifying the number of cells containing a selected nucleic acid sequence in a heterogenous population of cells
US5925517A (en) * 1993-11-12 1999-07-20 The Public Health Research Institute Of The City Of New York, Inc. Detectably labeled dual conformation oligonucleotide probes, assays and kits
AU694502B2 (en) 1994-03-18 1998-07-23 Myriad Genetics, Inc. Mts gene, mutations therein, and methods for diagnosing cancer using mts gene sequence
RU2161309C2 (en) 1994-03-18 2000-12-27 Исследовательский Фонд Университета Юты, Mutations of mts gene in embryonic line and method of detection of predisposition to malignant tumors
DE69519783T2 (en) 1994-04-29 2001-06-07 Perkin Elmer Corp METHOD AND DEVICE FOR REAL-TIME DETECTION OF PRODUCTS OF NUCLEIC ACID AMPLIFICATION
CA2223241C (en) 1995-06-07 2009-11-17 Gen-Probe Incorporated Methods and kits for determining pre-amplification levels of a nucleic acid target sequence from post-amplification levels of product
US6291166B1 (en) * 1997-04-16 2001-09-18 Xtrana, Inc. Nucleic acid archiving
US5773258A (en) 1995-08-25 1998-06-30 Roche Molecular Systems, Inc. Nucleic acid amplification using a reversibly inactivated thermostable enzyme
US5736330A (en) 1995-10-11 1998-04-07 Luminex Corporation Method and compositions for flow cytometric determination of DNA sequences
US5670325A (en) 1996-08-14 1997-09-23 Exact Laboratories, Inc. Method for the detection of clonal populations of transformed cells in a genomically heterogeneous cellular sample
US5736333A (en) * 1996-06-04 1998-04-07 The Perkin-Elmer Corporation Passive internal references for the detection of nucleic acid amplification products
US5928870A (en) 1997-06-16 1999-07-27 Exact Laboratories, Inc. Methods for the detection of loss of heterozygosity
US6020137A (en) 1996-08-14 2000-02-01 Exact Laboratories, Inc. Methods for the detection of loss of heterozygosity
CA2410120A1 (en) 1996-08-28 1998-03-05 The Johns Hopkins University School Of Medicine Method for detecting cell proliferative disorders
US6143496A (en) 1997-04-17 2000-11-07 Cytonix Corporation Method of sampling, amplifying and quantifying segment of nucleic acid, polymerase chain reaction assembly having nanoliter-sized sample chambers, and method of filling assembly
GB9712512D0 (en) 1997-06-16 1997-08-20 Bioinvent Int Ab A method for in vitro molecular evolution of protein function
CA2325343A1 (en) 1998-04-23 1999-10-28 Genentech, Inc. Quantitative analysis of gene expression
US6066458A (en) 1998-05-18 2000-05-23 Becton, Dickinson And Company Methods, apparatus and computer program products for determining quantities of nucleic acid sequences in samples using standard curves and amplification ratio estimates
US6037130A (en) * 1998-07-28 2000-03-14 The Public Health Institute Of The City Of New York, Inc. Wavelength-shifting probes and primers and their use in assays and kits
JP2001017179A (en) 1999-07-05 2001-01-23 Nippon Flour Mills Co Ltd Amplification of nucleic acid molecule
US6440706B1 (en) 1999-08-02 2002-08-27 Johns Hopkins University Digital amplification

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5804383A (en) * 1992-08-21 1998-09-08 The Regents Of The University Of California Method and assay for detection of the expression of allele-specific mutations by allele-specific in situ reverse transcriptase polymerase chain reaction
US5858663A (en) * 1992-10-01 1999-01-12 Life Technologies, Inc. Method for the rapid and ultra-sensitive detection of leukemic cells
EP0643140A1 (en) * 1993-09-13 1995-03-15 Canon Kabushiki Kaisha Determination of nucleic acid by PCR, measurement of number of microbial cells, genes, or gene-copies by PCR, and measuring-kit employed for the same
WO1995013399A1 (en) * 1993-11-12 1995-05-18 The Public Health Research Institute Of The City Of New York, Inc. Hybridization probes for nucleic acid detection, universal stems, methods and kits
WO1999013113A1 (en) * 1997-09-12 1999-03-18 The Public Health Research Institute Of The City Of New York, Inc. Non-competitive co-amplification methods

Non-Patent Citations (7)

* Cited by examiner, † Cited by third party
Title
DATABASE BIOSIS [Online] BIOSCIENCES INFORMATION SERVICE, PHILADELPHIA, PA, US; April 1999 (1999-04) EVERETT KARIN D E ET AL: "Identification of nine species of the Chlamydiaceae using PCR-RFLP." Database accession no. PREV199900271658 XP002185145 & INTERNATIONAL JOURNAL OF SYSTEMATIC BACTERIOLOGY, vol. 49, no. 2, April 1999 (1999-04), pages 803-813, ISSN: 0020-7713 *
HALFORD WILLIAM P ET AL: "The inherent quantitative capacity of the reverse transcription-polymerase chain reaction." ANALYTICAL BIOCHEMISTRY, vol. 266, no. 2, 15 January 1999 (1999-01-15), pages 181-191, XP000791527 ISSN: 0003-2697 *
PIATEK AMY S ET AL: "Molecular beacon sequence analysis for detecting drug resistance in Mycobacterium tuberculosis." NATURE BIOTECHNOLOGY, vol. 16, no. 4, April 1998 (1998-04), pages 359-363, XP000891876 ISSN: 1087-0156 *
TYAGI SANJAY ET AL: "Molecular beacons: Probes that fluoresce upon hybridization." NATURE BIOTECHNOLOGY, vol. 14, no. 3, 1996, pages 303-308, XP002914999 ISSN: 1087-0156 *
TYAGI SANJAY ET AL: "Multicolor molecular beacons for allele discrimination." NATURE BIOTECHNOLOGY, vol. 16, no. 1, January 1998 (1998-01), pages 49-53, XP002143901 ISSN: 1087-0156 *
VET JACQUELINE A M ET AL: "Multiplex detection of four pathogenic retroviruses using molecular beacons." PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES, vol. 96, no. 11, 25 May 1999 (1999-05-25), pages 6394-6399, XP002145609 May 25, 1999 ISSN: 0027-8424 *
VOGELSTEIN BERT ET AL: "Digital PCR." PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES, vol. 96, no. 16, 3 August 1999 (1999-08-03), pages 9236-9241, XP002185144 Aug. 3, 1999 ISSN: 0027-8424 *

Cited By (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1456648A4 (en) * 2001-11-20 2005-06-29 Exact Sciences Corp Automated sample preparation methods and devices
EP1456648A2 (en) * 2001-11-20 2004-09-15 Exact Sciences Corporation Automated sample preparation methods and devices
US7910300B2 (en) 2001-12-06 2011-03-22 Johns Hopkins University Disease detection by protein truncation assays
EP1523573A4 (en) * 2001-12-06 2006-03-08 Univ Johns Hopkins Med Disease detection by digital protein truncation assays
EP1523573A2 (en) * 2001-12-06 2005-04-20 The Johns Hopkins University School Of Medicine Disease detection by digital protein truncation assays
EP2192194A3 (en) * 2001-12-06 2010-10-20 The Johns Hopkins University Disease detection by digital protein truncation assays
US8669062B2 (en) 2001-12-06 2014-03-11 The Johns Hopkins University Disease detection by digital protein truncation assays
EP2960343A1 (en) * 2001-12-06 2015-12-30 The Johns Hopkins University Detection of mutations by digital protein amplification assays
EP1476572A4 (en) * 2002-02-01 2005-08-17 Univ Johns Hopkins Digital amplification for detection of mismatch repair deficient tumor cells
EP1476572A2 (en) * 2002-02-01 2004-11-17 The Johns Hopkins University, School of Medicine, Office of Technology Licencing Digital amplification for detection of mismatch repair deficient tumor cells
EP1907583B1 (en) 2005-06-15 2016-10-05 Complete Genomics Inc. Single molecule arrays for genetic and chemical analysis
EP1907583B2 (en) 2005-06-15 2019-10-23 Complete Genomics Inc. Single molecule arrays for genetic and chemical analysis
US10351909B2 (en) 2005-06-15 2019-07-16 Complete Genomics, Inc. DNA sequencing from high density DNA arrays using asynchronous reactions
US9944984B2 (en) 2005-06-15 2018-04-17 Complete Genomics, Inc. High density DNA array
EP2620510B1 (en) 2005-06-15 2016-10-12 Complete Genomics Inc. Single molecule arrays for genetic and chemical analysis
WO2007057669A3 (en) * 2005-11-15 2007-08-02 Genoid Kft Method of detecting pathogens
US7993881B2 (en) 2005-11-15 2011-08-09 Genoid Kft Method for detecting pathogens using molecular beacons
EA014648B1 (en) * 2005-11-15 2010-12-30 Генойд Кфт. Method of detecting pathogens
WO2007057669A2 (en) * 2005-11-15 2007-05-24 Genoid Kft. Method of detecting pathogens
WO2011085491A1 (en) * 2010-01-15 2011-07-21 The University Of British Columbia Multiplex amplification for the detection of nucleic acid variations

Also Published As

Publication number Publication date
CA2756673C (en) 2015-03-10
CA2376929C (en) 2015-05-19
JP4653366B2 (en) 2011-03-16
JP2003511009A (en) 2003-03-25
DE60018616T2 (en) 2006-02-23
CA2376929A1 (en) 2001-02-08
CA2756675A1 (en) 2001-02-08
WO2001009386A3 (en) 2002-09-12
AU781440B2 (en) 2005-05-26
US6753147B2 (en) 2004-06-22
US7915015B2 (en) 2011-03-29
CA2756673A1 (en) 2001-02-08
US6440706B1 (en) 2002-08-27
DE60018616D1 (en) 2005-04-14
US20110201004A1 (en) 2011-08-18
US8859206B2 (en) 2014-10-14
US20020090629A1 (en) 2002-07-11
US7824889B2 (en) 2010-11-02
AU6502800A (en) 2001-02-19
ATE290608T1 (en) 2005-03-15
EP1255856B1 (en) 2005-03-09
US20150038341A1 (en) 2015-02-05
US20080241830A1 (en) 2008-10-02
US9970058B2 (en) 2018-05-15
US20050130176A1 (en) 2005-06-16
CA2756675C (en) 2015-05-05
US20100209921A1 (en) 2010-08-19
US20180327858A1 (en) 2018-11-15
EP1255856A2 (en) 2002-11-13

Similar Documents

Publication Publication Date Title
EP1255856B1 (en) Digital amplification
Vogelstein et al. Digital pcr
ES2273367T3 (en) MULTIPLEX LOCI AMPLIFICATION OF SHORT REPETITIONS IN TANDEM.
JP3589638B2 (en) Real-time quantification method with nucleic acid efficiency correction
EP1288314B1 (en) Real-time gene quantification with internal standards
CN102344960B (en) Quantification of gene expression
JP5745774B2 (en) Multiplex quantitative nucleic acid amplification and melting assays
JP4919568B2 (en) Assays to detect short sequence variants
KR19990063053A (en) Detection of target primers and labeling primers for use in detection of target nucleic acids
Vet et al. Molecular beacons: colorful analysis of nucleic acids
JP2001515734A (en) Non-competitive co-amplification method
WO2014152185A1 (en) Multiplex allele detection
EP2798086A1 (en) Reagents and methods for autoligation chain reaction
JP2007530033A (en) Nucleic acid amplification measurement method using probe labeled with intercalating dye
JP5813263B1 (en) Method for detecting gene mutation and fluorescently labeled oligonucleotide used therefor
CN112301096A (en) Novel nucleic acid probe labeling method

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CR CU CZ DE DK DM DZ EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG US UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
WWE Wipo information: entry into national phase

Ref document number: 65028/00

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2376929

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2000952304

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

AK Designated states

Kind code of ref document: A3

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CR CU CZ DE DK DM DZ EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG US UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A3

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

WWP Wipo information: published in national office

Ref document number: 2000952304

Country of ref document: EP

WWG Wipo information: grant in national office

Ref document number: 2000952304

Country of ref document: EP