US20050042639A1 - Single molecule amplification and detection of DNA length - Google Patents

Single molecule amplification and detection of DNA length Download PDF

Info

Publication number
US20050042639A1
US20050042639A1 US10/845,996 US84599604A US2005042639A1 US 20050042639 A1 US20050042639 A1 US 20050042639A1 US 84599604 A US84599604 A US 84599604A US 2005042639 A1 US2005042639 A1 US 2005042639A1
Authority
US
United States
Prior art keywords
nucleic acid
interest
sample
amplification
probes
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US10/845,996
Other languages
English (en)
Inventor
Michael Knapp
Jill Baker
Andrea Chow
Anne Kopf-Sill
Michael Spaid
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Caliper Life Sciences Inc
Original Assignee
Caliper Life Sciences Inc
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
Priority claimed from US10/741,162 external-priority patent/US20040224325A1/en
Priority to US10/845,996 priority Critical patent/US20050042639A1/en
Application filed by Caliper Life Sciences Inc filed Critical Caliper Life Sciences Inc
Assigned to CALIPER LIFE SCIENCES, INC. reassignment CALIPER LIFE SCIENCES, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: SPAID, MICHAEL, KOPF-SILL, ANNE R., BAKER, JILL M., CHOW, ANDREA W., KNAPP, MICHAEL R.
Publication of US20050042639A1 publication Critical patent/US20050042639A1/en
Priority to PCT/US2005/017065 priority patent/WO2005113148A1/en
Priority to JP2007513461A priority patent/JP4740237B2/ja
Priority to AU2005245448A priority patent/AU2005245448A1/en
Priority to EP05749873A priority patent/EP1755785A1/en
Priority to CA002566698A priority patent/CA2566698A1/en
Priority to US11/867,626 priority patent/US7645581B2/en
Priority to US12/684,601 priority patent/US8275554B2/en
Abandoned legal-status Critical Current

Links

Images

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
    • C12Q1/6851Quantitative amplification
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01LCHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
    • B01L3/00Containers or dishes for laboratory use, e.g. laboratory glassware; Droppers
    • B01L3/50Containers for the purpose of retaining a material to be analysed, e.g. test tubes
    • B01L3/502Containers for the purpose of retaining a material to be analysed, e.g. test tubes with fluid transport, e.g. in multi-compartment structures
    • B01L3/5027Containers for the purpose of retaining a material to be analysed, e.g. test tubes with fluid transport, e.g. in multi-compartment structures by integrated microfluidic structures, i.e. dimensions of channels and chambers are such that surface tension forces are important, e.g. lab-on-a-chip
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01LCHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
    • B01L3/00Containers or dishes for laboratory use, e.g. laboratory glassware; Droppers
    • B01L3/50Containers for the purpose of retaining a material to be analysed, e.g. test tubes
    • B01L3/502Containers for the purpose of retaining a material to be analysed, e.g. test tubes with fluid transport, e.g. in multi-compartment structures
    • B01L3/5027Containers for the purpose of retaining a material to be analysed, e.g. test tubes with fluid transport, e.g. in multi-compartment structures by integrated microfluidic structures, i.e. dimensions of channels and chambers are such that surface tension forces are important, e.g. lab-on-a-chip
    • B01L3/502761Containers for the purpose of retaining a material to be analysed, e.g. test tubes with fluid transport, e.g. in multi-compartment structures by integrated microfluidic structures, i.e. dimensions of channels and chambers are such that surface tension forces are important, e.g. lab-on-a-chip specially adapted for handling suspended solids or molecules independently from the bulk fluid flow, e.g. for trapping or sorting beads, for physically stretching molecules
    • 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
    • C12Q1/6813Hybridisation assays
    • C12Q1/6834Enzymatic or biochemical coupling of nucleic acids to a solid phase
    • C12Q1/6837Enzymatic or biochemical coupling of nucleic acids to a solid phase using probe arrays or probe chips
    • 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
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N21/00Investigating or analysing materials by the use of optical means, i.e. using sub-millimetre waves, infrared, visible or ultraviolet light
    • G01N21/62Systems in which the material investigated is excited whereby it emits light or causes a change in wavelength of the incident light
    • G01N21/63Systems in which the material investigated is excited whereby it emits light or causes a change in wavelength of the incident light optically excited
    • G01N21/64Fluorescence; Phosphorescence
    • G01N21/6428Measuring fluorescence of fluorescent products of reactions or of fluorochrome labelled reactive substances, e.g. measuring quenching effects, using measuring "optrodes"
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01LCHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
    • B01L2200/00Solutions for specific problems relating to chemical or physical laboratory apparatus
    • B01L2200/10Integrating sample preparation and analysis in single entity, e.g. lab-on-a-chip concept
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01LCHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
    • B01L2200/00Solutions for specific problems relating to chemical or physical laboratory apparatus
    • B01L2200/16Reagents, handling or storing thereof
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01LCHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
    • B01L2300/00Additional constructional details
    • B01L2300/08Geometry, shape and general structure
    • B01L2300/0809Geometry, shape and general structure rectangular shaped
    • B01L2300/0816Cards, e.g. flat sample carriers usually with flow in two horizontal directions
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01LCHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
    • B01L2300/00Additional constructional details
    • B01L2300/08Geometry, shape and general structure
    • B01L2300/0861Configuration of multiple channels and/or chambers in a single devices
    • B01L2300/0864Configuration of multiple channels and/or chambers in a single devices comprising only one inlet and multiple receiving wells, e.g. for separation, splitting
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01LCHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
    • B01L2300/00Additional constructional details
    • B01L2300/08Geometry, shape and general structure
    • B01L2300/0861Configuration of multiple channels and/or chambers in a single devices
    • B01L2300/0867Multiple inlets and one sample wells, e.g. mixing, dilution
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01LCHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
    • B01L2300/00Additional constructional details
    • B01L2300/18Means for temperature control
    • B01L2300/1805Conductive heating, heat from thermostatted solids is conducted to receptacles, e.g. heating plates, blocks
    • B01L2300/1822Conductive heating, heat from thermostatted solids is conducted to receptacles, e.g. heating plates, blocks using Peltier elements
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01LCHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
    • B01L2300/00Additional constructional details
    • B01L2300/18Means for temperature control
    • B01L2300/1805Conductive heating, heat from thermostatted solids is conducted to receptacles, e.g. heating plates, blocks
    • B01L2300/1827Conductive heating, heat from thermostatted solids is conducted to receptacles, e.g. heating plates, blocks using resistive heater
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01LCHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
    • B01L2300/00Additional constructional details
    • B01L2300/18Means for temperature control
    • B01L2300/1833Means for temperature control using electrical currents in the sample itself
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01LCHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
    • B01L2400/00Moving or stopping fluids
    • B01L2400/04Moving fluids with specific forces or mechanical means
    • B01L2400/0403Moving fluids with specific forces or mechanical means specific forces
    • B01L2400/0406Moving fluids with specific forces or mechanical means specific forces capillary forces
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01LCHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
    • B01L2400/00Moving or stopping fluids
    • B01L2400/04Moving fluids with specific forces or mechanical means
    • B01L2400/0403Moving fluids with specific forces or mechanical means specific forces
    • B01L2400/0415Moving fluids with specific forces or mechanical means specific forces electrical forces, e.g. electrokinetic
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01LCHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
    • B01L2400/00Moving or stopping fluids
    • B01L2400/04Moving fluids with specific forces or mechanical means
    • B01L2400/0475Moving fluids with specific forces or mechanical means specific mechanical means and fluid pressure
    • B01L2400/0487Moving fluids with specific forces or mechanical means specific mechanical means and fluid pressure fluid pressure, pneumatics
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01LCHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
    • B01L7/00Heating or cooling apparatus; Heat insulating devices
    • B01L7/52Heating or cooling apparatus; Heat insulating devices with provision for submitting samples to a predetermined sequence of different temperatures, e.g. for treating nucleic acid samples
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01LCHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
    • B01L7/00Heating or cooling apparatus; Heat insulating devices
    • B01L7/52Heating or cooling apparatus; Heat insulating devices with provision for submitting samples to a predetermined sequence of different temperatures, e.g. for treating nucleic acid samples
    • B01L7/525Heating or cooling apparatus; Heat insulating devices with provision for submitting samples to a predetermined sequence of different temperatures, e.g. for treating nucleic acid samples with physical movement of samples between temperature zones
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N21/00Investigating or analysing materials by the use of optical means, i.e. using sub-millimetre waves, infrared, visible or ultraviolet light
    • G01N21/62Systems in which the material investigated is excited whereby it emits light or causes a change in wavelength of the incident light
    • G01N21/63Systems in which the material investigated is excited whereby it emits light or causes a change in wavelength of the incident light optically excited
    • G01N21/64Fluorescence; Phosphorescence
    • G01N21/6428Measuring fluorescence of fluorescent products of reactions or of fluorochrome labelled reactive substances, e.g. measuring quenching effects, using measuring "optrodes"
    • G01N2021/6439Measuring fluorescence of fluorescent products of reactions or of fluorochrome labelled reactive substances, e.g. measuring quenching effects, using measuring "optrodes" with indicators, stains, dyes, tags, labels, marks
    • G01N2021/6441Measuring fluorescence of fluorescent products of reactions or of fluorochrome labelled reactive substances, e.g. measuring quenching effects, using measuring "optrodes" with indicators, stains, dyes, tags, labels, marks with two or more labels

Definitions

  • the invention is in the field of single molecule detection, e.g., by amplification of single molecules from complex mixtures, e.g., for disease diagnosis, detection of pathogens, environmental contaminants, or the like.
  • Amplifications can be conducted in high throughput systems, e.g., microfluidic systems, to provide an ability to detect rare molecules in complex samples that are aliquotted into low copy number reaction mixtures, whereby a rare copy nucleic acid of interest is detected, e.g., by amplifying large numbers of aliquots of the complex samples.
  • the methods and systems can determine whether an individual nucleic acids of interest has a given length.
  • nucleic acids The detection of nucleic acids is central to medicine, forensic science, industrial processing, crop and animal breeding, and many other fields.
  • the ability to detect disease conditions e.g., cancer
  • infectious organisms e.g., HIV
  • genetic lineage e.g., DNA, RNA, and DNA.
  • Determination of the integrity of a nucleic acid of interest can be relevant to the pathology of an infection or cancer.
  • nucleic acid amplification One of the most powerful and basic technologies for nucleic acid detection is nucleic acid amplification. That is, in many typical formats, such as the polymerase chain reaction (PCR), reverse-transcriptase PCR (RT-PCR), ligase chain reaction (LCR), and Q- ⁇ replicase and other RNA/transcription mediated techniques (e.g., NASBA), amplification of a nucleic acid of interest precedes detection of the nucleic acid of interest, because it is easier to detect many copies of a nucleic acid than it is to detect a single copy.
  • PCR polymerase chain reaction
  • RT-PCR reverse-transcriptase PCR
  • LCR ligase chain reaction
  • Q- ⁇ replicase and other RNA/transcription mediated techniques e.g., NASBA
  • PCR, RT-PCR and LCR are in particularly broad use, in many different fields. Details regarding the use of these and other amplification methods can be found in any of a variety of standard texts, including, e.g.,: Sambrook et al., Molecular Cloning—A Laboratory Manual (3rd Ed.), Vol. 1-3, Cold Spring Harbor Laboratory, Cold Spring Harbor, N.Y., 2000 (“Sambrook”); Current Protocols in Molecular Biology, F. M. Ausubel et al., eds., Current Protocols, a joint venture between Greene Publishing Associates, Inc.
  • nucleic acids particularly rare copy nucleic acids. This is particularly true where the amplification reagents amplify a high copy nucleic acid in a given sample in addition to the rare nucleic acid and the two nucleic acids differ by only one or a few nucleotides in the same sample.
  • Another difficulty with amplification methods that is completely unaddressed in the prior art is that it can be quite difficult to perform quantitative analysis on rare nucleic acids.
  • the problems noted above for detection apply to quantitative analysis as well, with the additional problem that quantification is impacted by the presence of high copy number nucleic acids in the sample, even if the rare nucleic acid can be amplified. This is because, even if the amplification is sufficiently specific for detection of the rare nucleic acid, the high copy number of other amplified nucleic acids still has competitive effects on the amplification reaction for reaction components during the amplification reaction.
  • Sizing of nucleic acids of interest is an area where identification of rare sequences can be of particular interest.
  • the length of a nucleic acid of interest in a complex mixture of other nucleic acids can be diagnostic.
  • U.S. Pat. No. 6,586,177 to Shuber Jul. 1, 2003
  • METHODS FOR DISEASE DETECTION clinical samples are amplified in a multiwell format with two or more primer pairs followed by agarose gel electrophoresis of the amplification reactions to visualize the amplicons.
  • Shuber suggests the assay can be useful to determine proportions of degraded DNA from cells after apoptosis relative to full length DNA from cancer cells in certain clinical samples.
  • this method can be nonspecific, slow and labor intensive, fail to confirm separated amplicons were amplified from the same nucleic acid strands (i.e., ambiguous), fail to distinguish between long nucleic acids with marginally different lengths, fail to determine length directly from a homogenous mixture, and fail to detect nucleic acids of interest against a background of other nucleic acids in many complex clinical samples.
  • the present invention relates to the surprising discovery that single molecule amplification can be used for the detection and statistical characterization of rare nucleic acids of interest in a sample, e.g., for disease diagnosis (e.g., cancer diagnosis), detection of pathogens, detection of rare environmental nucleic acids, and the like.
  • disease diagnosis e.g., cancer diagnosis
  • pathogens e.g., detection of pathogens
  • detection of rare environmental nucleic acids e.g., cancer diagnosis
  • many individual amplification reactions can be performed on reaction mixtures derived from a sample comprising a rare nucleic acid of interest, where each reaction mixture has few (e.g., 1) or no copies of the rare a nucleic acid of interest, e.g., until the nucleic acid of interest is identified in a reaction mixture.
  • Additional nucleic acids in the sample can also be amplified in low copy number reactions and statistical methods can be used to determine the relative ratio of the nucleic acid of interest to the additional nucleic acid, e.g., to determine relative or absolute concentration of the nucleic acid of interest including proportions of full length and fragmented forms.
  • most or all of the steps in the methods herein can be performed in a continuous flow format to greatly speed the rate of the overall method.
  • one or more of the steps can be performed in a stopped flow mode, e.g., where the detector is configured to simultaneously scan multiple amplification regions at once (simultaneous detection provides for increased throughput in these embodiments).
  • High throughput amplification systems such as those embodied in high throughput microfluidic systems are particularly well adapted to performing these methods, which can be used to detect nucleic acids of interest that are present at exceedingly low concentrations in a sample to be analyzed, e.g., by performing many low copy number amplification reactions until the nucleic acid of interest is detected, and/or until enough copies of the nucleic acid of interest are detected that reliable statistical evaluations can be performed.
  • the invention also provides new ways of determining whether and how many copies of an initial nucleic acid are present in a reaction mixture (or whether the initial nucleic acid is present in a reaction mixture) by considering how far signal from the initial nucleic acid disperses during amplification and comparing the dispersion to expected dispersion arising from thermal diffusivity and/or Taylor Aris dispersion, or related phenomena (or even simply by comparison of the observed dispersion to empirically observed control reactions). This can include monitoring the shape (amplitude, signal width, and/or other signal shape features) of a signal generated from an aliquot comprising the nucleic acid of interest to a predicted or empirically observed signal shape.
  • the nucleic acids that are quantified can be known (e.g., controls) or unknown in composition. They can include experimental nucleic acids (the nucleic acids of primary interest in the experiment at issue) or can be other unknown nucleic acids (e.g., uncharacterized genomic and/or cDNA from a biological sample of interest).
  • a sample comprising the nucleic acid of interest and one or more additional nucleic acid is aliquotted into a plurality of reaction mixtures. At least two of the reaction mixtures are single copy reaction mixtures, each comprising a single copy of the nucleic acid of interest.
  • the plurality of reaction mixtures additionally comprise at least one additional reaction mixture comprising at least one copy of the additional nucleic acid.
  • the plurality of reaction mixtures are subjected to one or more amplification reaction (in this context, the amplification reaction may or may not amplify the nucleic acid of interest, i.e., if the reaction has zero copies of the nucleic acid of interest, it will not be amplified; if it has one or more copy it will).
  • the nucleic acid of interest is detected in one or more of the single copy reaction mixtures.
  • Statistical inferences and mathematical relationships can be determined based on the plurality of results from such analyses. For example, the absolute numbers and/or relative proportions of full length and fragmented nucleic acids of interest in a complex sample can be determined.
  • the invention includes methods of detecting a low copy nucleic acid of interest in a sample that has one or more higher copy additional nucleic acid that is different from the low copy nucleic acid.
  • the method includes aliquotting the sample into a plurality of reaction mixtures.
  • the mixtures can include a plurality (e.g., about 5, or more, about 10 or more, about 50 or more, about 100 or more, about 150 or more, or about 500 or more) of zero copy reaction mixtures that include zero copies of the nucleic acid of interest and at least one single copy reaction mixture comprising a single copy of the nucleic acid of interest.
  • the zero and single copy reaction mixtures are subjected to an amplification reaction (whether an amplification actually occurs or not).
  • the nucleic acid of interest is then detected in the single copy reaction mixture (this includes the possibility that the nucleic acid of interest is detected in one or in multiple individual single copy reactions).
  • the proportion of zero copy and single copy reaction mixtures can be incorporated into mathematical formulas, along with, e.g., sample dilution data, to determine the concentration of the nucleic acid of interest in a sample, and/or the proportions of the nucleic acid in full length or fragmented forms.
  • related methods of quantifying a nucleic acid of interest in a sample are provided.
  • the sample can be aliquotted into at least 25 reaction mixtures comprising 2 or fewer copies of the nucleic acid of interest each (and generally 1 or fewer).
  • the reaction mixtures can be subjected to one or more amplification reactions.
  • the nucleic acid of interest is then detected in a plurality of the reaction mixtures.
  • statistical evaluations of the nucleic acid of interest are performed based upon the detection of the nucleic acid of interest in the plurality of reaction mixtures.
  • at least 50 or more, at least 75 or more, or at least 100 or more reaction mixtures, comprising the 2 or fewer copies are subjected to the one or more amplification reactions.
  • methods of detecting a low copy nucleic acid of interest are provided.
  • a sample comprising the low copy nucleic acid of interest is aliquotted into a plurality of reaction mixtures.
  • a plurality of the reaction mixtures contain zero copies of the nucleic acid of interest and at least one of the reaction mixtures comprises at least one copy of the nucleic acid of interest.
  • a plurality of the plurality of zero copy reaction mixtures is subjected to one or more amplification reaction in a microfluidic device comprising at least one microchamber or microchannel.
  • the nucleic acid of interest is determined not to be present in the zero copy reaction mixtures.
  • At least one additional zero copy reaction mixture and the reaction mixture comprising the nucleic acid of interest are subjected to one or more amplification reaction.
  • the nucleic acid of interest is detected in the reaction mixture comprising the nucleic acid of interest.
  • the reaction mixtures are amplified and checked for the presence of the nucleic acid of interest in the microfluidic device, at least until the nucleic acid is detected. For a low copy number nucleic acid, this can require a large number of amplification reactions be performed on the zero copy reaction mixtures until the nucleic acid of interest is found.
  • the invention also provides methods for quantifying a nucleic acid of interest in a sample, e.g., by taking diffusion/dispersion into consideration.
  • a sample comprising a copy of the nucleic acid of interest, or a complement thereof, is aliquotted into at least one reaction mixture.
  • the reaction mixture is subjected to at least one amplification reaction, thereby amplifying the copy of the nucleic acid of interest.
  • a shape, volume, width, length, height, area, or the like, in which the nucleic acid of interest, or a signal corresponding thereto, is present is detected.
  • the shape, volume, width, height, length, or area is interpreted to indicate a number of copies of the nucleic acid of interest in the reaction mixture or sample, thereby quantifying the nucleic acid of interest in the sample. Because these shape features of the signal are extremely reproducible, it is straightforward to distinguish signals of interest from background random signal fluctuations. In a related aspect, knowledge of diffusion/dispersion and the reproducibility of these phenomena can be used to reliably distinguish the signal of a one or more target molecule(s) from random baseline system fluctuations.
  • this interpretation can be performed in any of a variety of ways, e.g., by comparing the shape, volume, width, height, length and/or other signal shape features to predicted values taking thermal diffusivity and/or Taylor-Aris dispersion into account and/or by back calculation from empirically observed values for known reactions performed in the system. It is worth noting that this method is particularly relevant to continuous flow systems, where materials disperse during flow.
  • signals can be read in parallel from homogenous mixtures, e.g., by using probes emitting signals at different wavelengths. Quantitation of the two or more forms (lengths) of the nucleic acid can separately be based on interpretation of the same or different signal parameters.
  • high throughput stopped flow methods of detecting rare nucleic acids are provided.
  • methods of detecting a nucleic acid of interest are provided, in which a sample comprising the nucleic acid of interest is aliquotted into a plurality of reaction mixtures. At least two of the reaction mixtures are single copy reaction mixtures, each comprising a single copy of the nucleic acid of interest.
  • the reaction mixtures are flowed throughout a network of microchannels and subjected to one or more amplification reaction under stopped flow conditions in the network of microchannels.
  • the nucleic acid of interest is detected in the single copy reaction mixtures under the stopped flow conditions.
  • the detection step can include detection of multiple reaction products simultaneously.
  • a CCD array or appropriate image processor can be used to scan an entire chip (or sub-regions thereof) for “clouds” of signal from amplified products. That is, an entire channel or network of channels can be scanned simultaneously (e.g., at two or more frequencies to detect two or more probes) after amplification and any or all regions where signal arising from amplification can be detected simultaneously (or in more than one pass of the scanner/detector, if desired). Where size detection employs two or more probes with different signals, system software can compare signal locations for coincident (typically indicating a nucleic acid of interest has a given length) and non-coincident signals (typically indicating a nucleic acid of interest is fragmented).
  • any or all of the above methods can be practiced in a continuous flow format to improve throughput of the relevant method, and/or can use stopped flow in combination with image analysis of multiple regions of (or an entire) microchannel network.
  • the reaction mixture can comprise the nucleic acid of interest and one or a plurality of additional nucleic acids (typically sample native nucleic acids not of interest or control nucleic acids), with the relevant method including detecting the nucleic acid of interest and/or the plurality of additional nucleic acids in the reaction mixture.
  • the methods optionally include adding up the number of nucleic acids of interest, or the plurality of additional nucleic acids, or both, in the reaction mixture or the sample, or both.
  • a ratio of the nucleic acid of interest or the plurality of additional nucleic acids in the reaction mixture to the sum of the nucleic acid of interest and/or the plurality of additional nucleic acids in the reaction mixture or sample can be determined.
  • a concentration or proportion of the nucleic acid of interest in the reaction mixture or sample can be determined.
  • the sum of the number of nucleic acids of interest and the plurality of additional nucleic acids can provide an indication of the total number of nucleic acids in the reaction mixture.
  • aliquotting the sample or reaction mixture can comprise diluting the sample into a plurality of reaction containers (e.g., wells in a microtiter plate), and/or flowing the sample into a microfluidic dilution channel or chamber.
  • the sample is optionally diluted in the microfluidic dilution channel or chamber (a form of dilution module in the systems of the invention), whereby the sample is aliquotted into multiple diluted aliquots in the microfluidic dilution channel or chamber.
  • the part or all of the aliquotting/dilution process can be multiplexed for high throughput, e.g., by flowing a plurality of samples into the device or reaction containers simultaneously.
  • Samples, aliquots, reaction mixtures, etc. can be flowed under pressure (e.g., into the microfluidic device) or via electroosmosis, or by any other available method.
  • the sample can be diluted from a common reaction component reservoir, e.g., comprising some or all of the reaction and/or buffer components for the amplification reactions (e.g., polymerase, primers, locus specific reagents, labels, salts, magnesium, water and/or the like).
  • one or more component can be located in one or more additional reservoirs and the components can be mixed prior to amplification.
  • any or all of these steps can be practiced in a continuous flow format, or utilizing the stopped flow/simultaneous image analysis methods noted herein.
  • the concentration of the nucleic acids of interest and/or any additional nucleic acid is optionally low in the methods of the invention, e.g., about 1 molecule per aliquot.
  • the sample can be diluted to a concentration of about 1 molecule of interest per nanoliter or less.
  • diluted aliquots are each diluted to the same degree; however, diluted aliquots can also be differentially diluted (e.g., to form a dilution series).
  • the volume of the aliquots can be quite low to keep reagent costs low, e.g., in microfluidic applications.
  • the aliquots can be less than about 100 nl in volume, e.g., less than about 10 nl in volume, or, e.g., about 1 nl in volume or less.
  • At least one of the reaction mixtures is in an aqueous solution (the enzymes used in typical amplification reactions typically function well in an aqueous environment) dispersed as an emulsion.
  • This can take the form of individually resolved reaction mixture droplets in a microfluidic device, fluid in reservoirs of a microtiter plate, or other forms such as where at least one of the reaction mixtures is formulated in an aqueous phase of an emulsion comprising aqueous droplets suspended in an immiscible liquid (in this embodiment, amplification can be performed on the reaction mixture when it is formulated in the emulsion).
  • the nucleic acid of interest is optionally present as a single copy in at least one aqueous droplet of the aqueous phase prior to performing the amplification reaction.
  • the nucleic acid of interest is detected in the emulsion after the amplification reaction is performed.
  • a plurality of additional nucleic acids are also formulated in the aqueous phase of the emulsion and the method comprises detecting the plurality of additional nucleic acids.
  • statistical analysis can be performed on, e.g., the ratio of the nucleic acid sizes nucleic acids in the emulsion, e.g., to determine the concentration and/or proportions of the nucleic acids of interest having a given length in the emulsion.
  • At least 10 of the reaction mixtures are optionally low copy reaction mixtures (e.g., comprising 100 or fewer, usually 50 or fewer, typically 10 or fewer, generally 2 or fewer and often 1 or fewer copies of the nucleic acid of interest and/or of the additional nucleic acid).
  • at least 25, at least 50, at least 100, at least 150, at least 500 or more of the reaction mixtures are low copy reaction mixtures.
  • the low copy reaction mixtures can comprise at least 10, at least 25, at least 50, at least 100, at least 150 at least 500 or more single or zero copy reaction mixtures comprising 1 or fewer copies of the nucleic acid of interest.
  • the reaction mixtures can, and often do, comprise no copies of the nucleic acid of interest.
  • a plurality of the reaction mixtures can comprise a plurality of zero copy reaction mixtures that comprise no copies of the nucleic acid of interest. That is, at least about 10, 25, 50, 100, 150, 500, 1,000 or even 10,000 or more of the reaction mixtures can be zero copy reaction mixtures that have no copies of the nucleic acid of interest.
  • the invention provides the ability to rapidly search through many such zero copy reaction mixtures to individually identify a full length or fragmented nucleic acid of interest.
  • the sample comprises at least one additional nucleic acid that is different than the nucleic acid of interest.
  • the additional nucleic acid can, and often does, exist at a higher copy number in the sample than the nucleic acid of interest.
  • the additional nucleic acid can be a known nucleic acid (e.g., a control or hybridization blocking nucleic acid) or can itself be unknown with respect to part or all of the composition (a common occurrence where the nucleic acid of interest is to be detected in a biological sample, e.g., a cell or tissue sample from a patient).
  • the additional nucleic acid can be present at a concentration at least about 100 ⁇ , at least about 1,000 ⁇ , at least about 10,000 ⁇ , at least about 100,000 ⁇ , at least about 1,000,000 ⁇ or greater as high as the nucleic acid of interest in the sample (that is, can have at least about 100 ⁇ , at least about 1,000 ⁇ , at least about 10,000 ⁇ , at least about 100,000 ⁇ , at least about 1,000,000 ⁇ or greater as many copies as the nucleic acid of interest in the sample).
  • the nucleic acid of interest can be detected regardless of its relative concentration.
  • the additional nucleic acid can be detected independent of the nucleic acid of interest.
  • a ratio of the nucleic acid of interest to the additional nucleic acid can be determined, e.g., for statistical analysis of the nucleic acid of interest and/or the additional nucleic acid.
  • the number of nucleic acids in the reaction mixture (whether the nucleic acid(s) of interest, the additional nucleic acids, or other nucleic acids) can be added up and the concentration of the nucleic acids (or the relative concentrations) can be determined in the sample, or in any of the various aliquots and reaction mixtures herein.
  • the ratio(s) and/or quantities of fragmented nucleic acid of interest, nucleic acid of a given length, and/or an additional nucleic acid can be determined using methods of the invention.
  • the nucleic acid of interest can be essentially any detectable nucleic acid. Examples include SNPs, low copy nucleic acids, cancer associated nucleic acids, infective or pathogen associated nucleic acids, forensic nucleic acids, and the like. Because of the ability of the methods of the invention to identify extremely low copy number nucleic acids, and/or distinguish nucleic acids by size, the invention is suitably applied to early stage disease diagnosis where cancer cells or pathogens are present at low concentrations. For example, colon cancer cells can be present in stool samples, but, at least in the early stages of colon cancer, the concentration of cancer cell DNA is small compared to the overall DNA in such a sample (typically much less than 1% of the cells from which the DNA sample was derived).
  • nucleic acids from the cancer cells is relatively more full length than nucleic acids from other (apoptotic) cells in the sample.
  • the present invention can be used to identify, proportion, and quantify cancer DNA in such a sample, providing a new method for disease diagnosis and prognostication. Similar approaches can be used to identify cancerous DNAs or pathogen nucleic acids from any fluid or tissue from which such samples are normally taken or derived, e.g., blood, urine, serum, plasma, saliva, tears, sputum, stool, ejaculatory fluid, cervical swabs, vaginal secretions, or the like.
  • infective/pathogenic agents such as viruses (e.g., HIV, herpes virus, pox virus, etc.), parasites (e.g., malarial parasites ( Plasmodium ), nematodes, etc.), bacteria (e.g., pathogenic E. coli, salmonella, etc.) can be identified.
  • viruses e.g., HIV, herpes virus, pox virus, etc.
  • parasites e.g., malarial parasites ( Plasmodium ), nematodes, etc.
  • bacteria e.g., pathogenic E. coli, salmonella, etc.
  • pathogenic E. coli e.g., salmonella, etc.
  • Methods can distinguish nucleic acids from living bacteria from those of lysed bacteria, e.g., in a clinical sample.
  • the methods of the invention utilize thermocyclic amplification reactions, although non-thermocyclic reactions (e.g., using denaturants in place of heat, a procedure that is relatively practical in microscale applications) can also be used.
  • the reaction mixtures are subjected to one or more amplification reaction(s) by thermocycling the reaction mixtures in one or more microscale amplification chamber or channel.
  • thermocycling methods can be used in a microscale device (or in reaction containers), e.g., heating by applying electrical current to fluid of the reaction mixture (e.g., in the microscale amplification chambers or channels), resistively heating a heating element that contacts or is in proximity to the reaction mixture (e.g., in the microscale amplification chambers or channels), heating with a Joule-Thompson or Peltier device, or any other available heating or heating and cooling method(s).
  • the components of the system can be treated with one or more reagents between operational runs to reduce cross contamination between operations.
  • the amplification channel can have acid or base flowed into the channel between amplification reactions to reduce unwanted contamination from one or more previous amplification products.
  • detecting can include real time homogenous PCR detection, e.g., via use of TaqManTM probes (operating by detecting a double-labeled probe before, during, or after polymerase-mediated digestion of the double labeled probe), use of molecular beacons, or the like.
  • Real time detection can be omitted, e.g., simply by detecting amplicons via labeled probes, e.g., after separation of the amplicon from unlabeled probe.
  • the detecting step(s) can include quantifying the nucleic acids of interest in the reaction mixtures, or the sample, or both.
  • the nucleic acids can be quantified separate from the detection step.
  • quantifying the nucleic acid of interest optionally comprises detecting the nucleic acid in a plurality of single-copy reaction mixtures and performing statistical or probabilistic analysis to determine a percentage or distribution of reaction mixtures comprising a single copy of the nucleic acid of interest.
  • the statistical or probabilistic analysis can comprise any available technique or combination thereof, e.g., Poisson analysis, Monte Carlo analysis, application of a genetic algorithm, neural network training, Markov modeling, hidden Markov modeling, multidimensional scaling, partial least squares (PLS) analysis, or principle component analysis (PCA).
  • Poisson analysis Monte Carlo analysis
  • PCA principle component analysis
  • the initial starting concentration of a nucleic acid of interest can be determined, e.g., by detecting a reproducible shape, length, width, height, volume or area of associated signal(s) for the nucleic acid of interest in a given reaction mixture.
  • the signal can be detected from a label bound to the nucleic acid of interest.
  • the shape, length, width, height, volume or area is optionally correlated to a number of nucleic acids interest present in one of the reaction mixtures, and/or present in the sample based upon a Taylor-Aris dispersion calculation, or a thermal diffusivity calculation, or both, or by comparison to an empirically observed set of reaction mixtures having a known number of starting nucleic acids for amplification.
  • the invention comprises calculating diffusion, or dispersion, or both, of one or more amplified nucleic acids in the given reaction mixture, and correlating the diffusion, or the dispersion, or both, to a number of copies of the nucleic acid of interest in one of the given reaction mixtures prior to amplification.
  • Systems and/or kits adapted for practicing the methods herein are a feature of the invention.
  • the systems and/or kits can include system instructions (e.g., embodied in a computer or in a computer readable medium, e.g., as system software) for practicing any of the method steps herein.
  • Fluid handling elements for storing, transferring, aliquotting, or diluting samples, e.g., microfluidic handling elements, and detector elements can also be components of the systems and kits herein.
  • packaging materials, integration elements (e.g., instrument cases, power supplies, etc.), instructions for using the systems and kits and the like can be features of the invention.
  • the invention provides a system for detecting low copy nucleic acids of interest in a sample.
  • the system includes a dilution module that dilutes the sample into multiple aliquots and a microfluidic device comprising an amplification channel or chamber configured to thermocycle one or more of the multiple aliquots.
  • a detector integral with or proximal to the microfluidic device is also included, where the detector is configured to detect one or more amplified copies of the nucleic acid of interest in or on the microfluidic device.
  • System instructions that direct the dilution module to aliquot the sample into a plurality of aliquots, including a plurality of zero copy aliquots comprising no copies of the nucleic acids of interest and one or more single copy aliquot comprising a single copy of the nucleic acid of interest are also included.
  • the system also includes system software that correlates a reproducible signal shape, length, width, volume or area occupied by amplified copies of the nucleic acid of interest, as detected by the detector, to the number of copies of the nucleic acid of interest present in one of the aliquots, or to the number of copies of the nucleic acid of interest present in the sample, or both.
  • the system can typically evaluate the absolute or relative number of nucleic acids of interest having different lengths to provide concentrations and/or proportions of the nucleic acids. Any or all of the system components can be selected to operate such that a sample of interest is continuously flowed during operation of the system. Alternately, the stopped flow/simultaneous image analysis methods noted herein can be applied.
  • systems for quantifying one or more low copy nucleic acid of interest in a sample are provided.
  • a dilution module dilutes the sample into multiple aliquots.
  • a microfluidic device comprising an amplification channel or chamber is configured to thermocycle one or more of the multiple aliquots.
  • a detector integral with or proximal to the microfluidic device is configured to detect a reproducible shape, length, width, volume or area occupied by signals from amplified copies of the nucleic acid of interest (often hybridized to a detectable probe or represented by released but previously hybridized probe) present in one of the aliquots following thermocycling of the reaction mixture aliquots.
  • the system can also include system software that correlates the shape, length, width, volume or area occupied by amplified copies of the nucleic acid of interest to the number of copies of the nucleic acid of interest present in one of the aliquots, or to the number of copies of the nucleic acid of interest present in the sample, proportion of nucleic acids with different lengths, etc.
  • the system includes system instructions that direct the dilution module to aliquot the sample into a plurality of aliquots, including a plurality of zero copy aliquots comprising no copies of the nucleic acids of interest and one or more single copy aliquot comprising a single copy of the nucleic acid of interest.
  • the dilution module can optionally be integral with the microfluidic device, e.g., as a dilution channel.
  • the microfluidic device can also include one or more electrodes positioned to flow electrical current into the microchamber or channel. Flow of current into the microchamber or channel can be used to heat fluid in the microchamber or channel.
  • the microfluidic device optionally includes or is coupled to one or more heating element (e.g., a resistive heating element, a Peltier device or a Joule Thompson device) positioned within or proximal to the microchamber or channel, which heats fluid in the microchamber or channel.
  • the detector is typically configured to detect one or more electromagnetic energy signal in or on the microfluidic device, although other in device sensors (e.g., pH, conductivity, etc.) can also be used.
  • the detector can detect fluorescence, luminescence, and/or fluorescence polarization of the sample.
  • the detector can be an off-device instrument, such as, e.g., size selective chromatography instrumentation or a mass spectrometer.
  • the system optionally comprises software with instructions for performing any of the method steps herein.
  • the system can include statistical or probabilistic system software that performs one or more statistical or probabilistic analysis of signals received from one or more of the aliquots subjected to thermocycling.
  • the statistical or probabilistic analysis can include Poisson analysis, Monte Carlo analysis, application of a genetic algorithm, neural network training, Markov modeling, hidden Markov modeling, multidimensional scaling, PLS analysis, and/or PCA analysis.
  • the statistical or probabilistic analysis optionally comprises quantitatively determining a concentration, proportion, or number of the nucleic acids of interest in the sample.
  • the systems above also optionally include fluid handling or storage features such as sample storage modules that store the samples until they are to be diluted by the dilution module, a sample retrieval module that retrieves the sample from the sample storage module and delivers it to the dilution module, or the like.
  • fluid handling or storage features such as sample storage modules that store the samples until they are to be diluted by the dilution module, a sample retrieval module that retrieves the sample from the sample storage module and delivers it to the dilution module, or the like.
  • These features are optionally designed to provide for continuous flow of fluid (e.g., comprising the sample) through the system (thereby providing for higher sample throughput).
  • stopped flow/simultaneous image analysis can be used in the systems herein.
  • Important aspects of the present invention are methods and systems to determine whether a nucleic acid of interest is at least a given length based on the presence, or absence of signals from low or single copy reactions mixtures.
  • the reaction mixtures for such determinations typically contain two or more probes complimentary to sequences at positions spaced along one or more strands of the nucleic acid of interest. Coincident detection of two or more probes in the reaction mixture can indicate that individual nucleic acid molecules are not fragmented between probe hybridization sites.
  • Systems useful in determining length by the two probe single copy reaction mixture techniques can include dilution modules and microfluidic devices to prepare and detect the reaction mixtures, and computers to interpret and correlate signal data acquired from detectors.
  • Methods of determining whether a nucleic acid of interest in a sample comprises at least a given length can include contacting the nucleic acid of interest in a reaction mixture with two or more different probes having detectable markers, and flowing the nucleic acid into a detection region to detect one or more signals from the probes. Coincident detection of two or more signals from different probes can indicate the nucleic acid of interest is not fragmented between the probes. Detection of a single signal can indicate the nucleic acid is fragmented. Such determinations can be considered assays of integrity for a nucleic acid of interest in a sample.
  • Samples for nucleic acid length determinations and differentiations include, e.g., whole blood, serum, plasma, stool, urine, vaginal secretions, ejaculatory fluid, synovial fluid, a biopsy, cerebrospinal fluid, amniotic fluid, sputum, saliva, lymph, tears, sweat, and urine.
  • an amplification reaction can be used to enhance the sensitivity of the assay.
  • the nucleic acid of interest is contacted with a first primer pair and a second primer pair having at least one primer that is outside of the sequence defined by the first primer pair, the nucleic acid of interest is amplified in a reaction mixture in a microchannel or microchamber with polymerase extensions from the primers to produce first amplicons defined by the first primer pair or second amplicons defined by the second primer pair.
  • First and second probes complimentary to the first and second amplicon and having detectable markers are introduced into the reaction mixture to hybridize with available complimentary sequences, and one or more signals are detected from the probes.
  • reaction mixture detected contains only a single copy of the nucleic acid of interest.
  • Reaction mixtures detected in the methods can be homogenous mixtures, e.g., not requiring separation of labeled constituents before detection of signals.
  • the concentration of the nucleic acid in samples is adjusted so that desired numbers of low, single, and zero copy reactions can be independently detected.
  • the adjustment can be a concentration, e.g., by immunoprecipitation, capture to a solid support, or ultrafiltration.
  • the adjustment can be a dilution, e.g., by serial dilution or fluidic mixing.
  • the nucleic acid of interest is diluted or concentrated to provide a concentration of about 1 molecule per nanoliter or less in a reaction mixture.
  • detection results from multiple low copy, single copy, and zero copy reactions are compiled to obtain confirmatory data and to allow statistical inferences with a suitable level of confidence.
  • it is an aspect of the invention that for quantitative results it is preferred to aliquot a sample into at least 25 reaction mixtures comprising 2 or fewer copies of the nucleic acid of interest each for hybridization with probes and counting the number of aliquots resulting in detection of a signal from one probe and/or signals from two or more probes. It is preferred that the aliquotting (concentration, dilution, and/or segregation into a small volume) result in one or more reaction mixtures having single copies or zero copies of the nucleic acid of interest; particularly for quantitation or proportion analyses. From the compiled data, the number of one marker signals and two marker signals can be evaluated to determine a proportion of nucleic acids of interest having different lengths. Thresholds can be established for confident correlation of some such proportions with certain disease states.
  • one primer pair in an amplification acts as a control for amplification and/or hybridization efficiency. It is preferred that these primer pairs define amplicons of about 100 base pairs in length. Primer pairs for probe target amplicons are preferably about the same length for each probe and can range from more than about 1000 base pairs to about 20 base pairs, or from about 200 base pairs to about 50 base pairs, or about 100 base pairs.
  • the larger amplicons generally range in length from about 5000 base pairs to about 200 base pairs, or about 1000 base pairs.
  • at least one of the probes is complimentary to the amplicon sequence defined by one primer pair but not complimentary to the amplicon sequence defined by another primer pair.
  • Amplifications in the methods of determining length are generally provided by constituting amplification reactions containing a polymerizing enzyme to increase the amount of target (e.g., nucleic acid of interest) sequence, increase the amount of hybridized probe, or increase the signal from such probes.
  • Amplifying a nucleic acid in the methods typically involves incorporation of a polymerase into the amplifying reaction, such as a heat stable DNA polymerase for a polymerase chain reaction (PCR), a reverse-transcriptase for RT-PCR, ligase for a ligase chain reaction (LCR), a Q- ⁇ replicase, or enzymes for RNA/transcription mediated techniques.
  • Probes used in the length determinations can have different specificity for sequences along the length of the nucleic acid of interest.
  • the detectable markers on probes with different specificity can have the same signal or, preferably, probes hybridizing to different compliments on the nucleic acid or associated amplicons have detectably different signals.
  • the probes can have any suitable detectable markers, but preferred markers are based on fluorescent dyes. In particular, probes are favored that include a fluorescent resonant energy transfer (FRET) detectable marker or a molecular beacon (MB) marker.
  • FRET fluorescent resonant energy transfer
  • MB molecular beacon
  • Methods of determining given lengths for nucleic acids of interest can quantify the nucleic acid and its fragmentation state. Such quantitation can simply involve counting the number of signals from probes in low or single copy reactions and calculating an amount of the nucleic acid based on known dilution factors, efficiency factors, standard curves, and the like. The quantifying can separately determine the amount of various fragmentation forms of the nucleic acid of interest based on the number of signals from two or more different probes, e.g., having different detectable marker signals.
  • Signal parameters such as shape, volume, width, height, length, area, or ratio, of the one or more signal (e.g., chart peaks) can be interpreted to confirm an actual signal (i.e., that the signal is not an artifact) and/or to indicate a certain quantity of a nucleic acid in a sample.
  • Quantification can be based on comparison of signal peak parameters to an internal standard signal.
  • quantitation can be based on comparison of signals from two or more reaction mixtures comprising different degrees of amplification to standard reaction mixtures with similar degrees of amplification.
  • thermocycler different degrees of amplification can be obtained by flowing reaction mixtures through a thermocycler at different flow rates, flowing reaction mixtures different distances into a thermocycler, retaining reaction mixtures in a thermocycler for different amounts of time, or exposing reaction mixtures to different numbers of amplification cycles.
  • Samples containing unknown amounts of a nucleic acid of interest can be quantified by comparing to their signal peaks to sets of standard signal peaks.
  • a nucleic acid of interest in a sample can be quantified by amplifying a dilution series of standard materials containing known amounts of the nucleic acid of interest through a certain number of amplification cycles, detecting signals associated with standard amplicons produced from the standard materials, amplifying the sample nucleic acid of interest the number of amplification cycles, detecting a signal associated with sample amplicons produced from the sample nucleic acid of interest, and comparing one or more standard amplicon signals to the sample amplicon signal to determine a concentration value for the nucleic acid of interest in the sample.
  • Sample and standard signal parameters for comparison can include, e.g., the shape of their signal peaks, points of inflection on the signal peaks, slopes of the signal peaks, signal peak amplitudes, signal peak areas, signal peak widths at half height, and/or the like.
  • the reliability of results can be enhanced through various schemes of repeated testing.
  • the amplifying, detecting, and comparing steps can be repeated one or more times, with different numbers of amplification cycles, to determine additional concentration values for the sample nucleic acid of interest for statistical evaluation providing more precise or more accurate concentration value results for the nucleic acid of interest in the sample.
  • Improved assay results can be obtained by gathering signal data after amplifications through two or more different numbers of cycles.
  • a major benefit of running the quantitative assay at different amplifications is to broaden the usable range of the assay.
  • statistical evaluation of the additional data provided by analysis at multiple amplification levels can enhance other assay parameters, such as precision, accuracy, and sensitivity.
  • Quantifying a nucleic acid of interest in a sample based on detection of multiple amplifications can include: amplifying the nucleic acid of interest through more than one number of amplification cycles, detecting signals associated with amplicons produced from two or more of the amplification cycle numbers, preparing a sample curve of a signal parameter versus number of amplification cycles, and comparing one or more identifiable points from the sample curve to a standard curve of the identifiable points versus concentration to quantify the nucleic acid of interest.
  • Exemplary identifiable points from signal curves include points of inflection, points having a certain slope, points having a certain signal amplitude, points having a certain fraction of a maximum signal amplitude, and/or the like.
  • Such quantitative assays can be used to quantitate or proportion fragmented and unfragmented nucleic acid of interest in evaluations of integrity.
  • Proportions of fragmented and given length nucleic acid of interest can be determined in a sample by: amplifying the nucleic acid of interest through a plurality of amplification cycles in a reaction mixture defining two or more different amplicons of the nucleic acid of interest; detecting, from homogenous reaction mixtures, different signals associated with each of the different amplicons after at least two different numbers of amplification cycles; preparing sample curves for each of the different signals versus numbers of amplification cycles; and, comparing one or more identifiable points from the sample curves to one or more standard curves describing nucleic acid of interest concentrations versus identifiable points.
  • Each of the amplicons can be relatively or absolutely quantitated to determine the amount of nucleic acid of interest sequences in the sample.
  • the amplification reaction mixtures detected are low copy or single copy reaction mixtures, thus allowing unambiguous determinations of fragmented and given length nucleic acid. That is, coincident detection of two or more or the different signals from low or single copy mixtures can indicate a nucleic acid of a given length, or the detection of a one of the different signals can indicate a fragmented nucleic acid.
  • the number of amplification cycles experienced by samples and standards can be controlled, e.g., by flowing the amplification reactions through a thermocycler at different flow rates, flowing the amplification reactions different distances into a thermocycler, retaining the amplification reactions in a thermocycler for different amounts of time, or exposing the amplification reactions to different numbers of amplification cycles.
  • the systems can basically include a microfluidic device with an amplification microchannel or microchamber containing one or more reaction mixtures under conditions that provide one or more amplicons of the nucleic acid of interest, a detector integral with or proximal to the microfluidic device and configured to detect the amplicons as one or more signals from a homogenous mixture, and a software system that interprets one or more coincidently detected signals to lengths of one or more individual nucleic acid molecules of interest to differentiate lengths of the nucleic acids of interest.
  • High throughput aspects of the system can be advanced by provision of multiple amplification channels in the microfluidic device.
  • the system can include affinity molecules, such as oligonucleotides, on a solid support to capture nucleotides of interest before or during preparation of the reaction mixture, or to capture amplicons for detection.
  • affinity molecules such as oligonucleotides
  • Other system elements e.g., to enhance high throughput aspects of the invention include sample storage modules, sample retrieval modules, and computers.
  • the systems of the invention can incorporate a dilution module to adjust the concentration of reaction mixture constituents.
  • the dilution module can be configured to dilute the sample to a concentration providing one or more single copy reaction mixtures for nucleic acids of interest in the amplification microchannel or microchamber.
  • a dilution module can be equipment to prepare serial multiwell plate dilutions, or a dilution channel in the microfluidic device.
  • the system can include instructions that direct the dilution module to aliquot the sample or reaction mixture into a plurality of aliquots, including a plurality of zero copy aliquots comprising no copies of the nucleic acid of interest and one or more single copy aliquots comprising a single copy of the nucleic acid of interest.
  • Such dilutions (or concentrations) can provide substantial numbers of non-overlapping reaction mixture aliquots for discrete counting of signals.
  • Reaction mixtures of the systems can include constituents associated with amplification of the nucleic acid, hybridization reactions of primers or probes, and/or detection of detectable marker signals.
  • a typical reaction mixture for detection of length can include the nucleic acid of interest, a first primer pair, a second primer pair with at least one primer complimentary to a sequence of the nucleic acid of interest outside a sequence defined by the first primer pair, and a polymerase that can synthesize amplicons defined by the primer pairs.
  • a control primer pair defines amplicons of 100 base pairs or less in length while a test primer pair defines longer amplicons ranging in length from about 100 base pairs to about 3000 base pairs.
  • control amplicons usually overlap sequences of the longer amplicons with probe signals for longer and control probes indicating the proportion of the nucleic acid that is fragmented.
  • the two sets of primer pairs define amplicons of about the same length but the amplicons do not overlap.
  • coincident signals from a low or single copy reaction can indicate the nucleic acid of interest is of a given length (the length defined by the probes and the distance between them).
  • Amplification reaction mixtures and/or hybridization mixtures can include one or more probes to determine the length of a nucleic acid of interest.
  • the probes can have one or more detectable markers and a sequence complimentary to one or more of the amplicons so that the detectable markers provide a signal detectable by the detector.
  • the probes can be complimentary to an amplicon sequence defined by one primer pair but not complimentary to an amplicon sequence defined by another primer pair, while, in some methods described above, the probes can be complimentary to a sequence common to both a first amplicon and a second amplicon.
  • two or more different probes each comprise different signals for easy independent monitoring of coincident signals.
  • detectable markers on different probes can have different fluorescent emission wavelengths.
  • the probes can be, e.g., fluorescent resonant energy transfer (FRET) detectable marker or a molecular beacon (MB) marker.
  • FRET fluorescent resonant energy transfer
  • MB molecular beacon
  • the probe has a detectable marker includes a quencher removable from the FRET probe by nuclease activity, so that one or more positive signals can be detected from a homogenous mixture against low levels of background noise.
  • Systems to determine length can include amplification channels or chambers that provide conditions for amplification of a reaction constituent.
  • the chambers are thermocyclers and the nucleic acid of interest is amplified by a polymerase reaction.
  • the amplification microchannel or microchamber can include, e.g., electrodes to apply a heating current to the microchannel, a resistive heating element, a Joule-Thompson device, a Peltier device, and/or the like.
  • the amplification microchannel or microchamber can be configured to thermocycle the reaction mixture producing amplicons of the nucleic acid of interest in a volume sufficiently small to substantially separate amplification products of a single nucleic acid of interest molecule from other nucleic acid of interest molecules in the sample or from additional nucleic acids in the sample.
  • the amplicons can be detected without resolution of different amplicons or different probes, e.g., in a size selective media or affinity media.
  • Software systems can work in computers to enhance the high throughput aspects of the methods of determining length and automate interpretation of detected signals.
  • the system software can interpret signal volumes, widths, heights, lengths, areas, and/or ratios, from the detector to indicate a number of copies of the nucleic acid of interest in the sample, a number of the nucleic acids of interest having a given length, or a proportion of nucleic acids of interest having different lengths.
  • Detectors in the systems can detect signals from any suitable detectable marker.
  • Detectors can include technologies, such as, e.g., fluorometers, charge coupled device, lasers, enzymes or chromogenic enzyme substrates, photo multiplier tubes, spectrophotometers, scanning detectors, microscopes, galvo-scanners, and/or the like.
  • the detector can independently detect signals from two or more detectable markers with different signals; e.g., a fluorometer detector that can simultaneously detect emissions at two or more frequencies.
  • FIG. 1 schematically illustrates a chip design for an 8-channel PCR sipper chip used in many of the Examples herein.
  • FIG. 2 is a graph of percent amplification versus input copy number for 2 experimental runs, with a comparison to a predicted (Poisson) value.
  • Panels A and B provide peak area and peak width bar graphs.
  • FIG. 4 Panels A-D are graphs illustrating peak width for amplification reactions.
  • FIG. 5 is a graphical analysis of single molecule amplification peak widths.
  • FIG. 6 is a schematic representation of a system of the invention.
  • FIG. 7 is a schematic representation of a system of the invention.
  • FIG. 8 is a schematic representation of a stopped flow system that uses simultaneous image processing of a network of channels to scan for nucleic acids of interest.
  • FIG. 9 is a schematic of a fluidic network after thermocycling. Spots represent the fluorescence “clouds” from single copy amplification reactions. The spots are counted for quantitative PCR analysis.
  • FIG. 10 is a data graph showing single molecule DNA amplification.
  • FIG. 11 is a data graph showing single molecule DNA amplification (6 passes).
  • FIG. 12 is a data graph showing single molecule DNA amplification (3 panels).
  • FIG. 13 provides a graph of detection of 2 mutation sites relevant to cancer detection developed on-chip using TaqMan probes.
  • FIG. 14 shows a schematic diagram of low to single copy detection of nucleic acids of interest amplified using two amplicon sequences that do not overlap.
  • FIG. 15 shows a schematic diagram of low to single copy detection of nucleic acid of interest amplified using two amplicon sequences that overlap at a sequence complimentary to at least one probe.
  • FIGS. 16A to 16 B show schematic diagrams of amplification curves generated by flowing amplification reactions different distances within an actively cycling amplification region.
  • FIG. 17 shows detected signal peaks for a series of nucleic acid standard materials with amplification cycles numbering 25 cycles or 40 cycles.
  • FIGS. 18A to 18 C show schematic charts demonstrating the quantitation of a nucleic acid of interest based on the number of amplification cycles required to reach an identifiable point of maximum slope.
  • FIG. 19 shows a schematic diagram of a system of the invention for differentiating the length of nucleic acids of interest.
  • the present invention derives, in part, from a surprising conceptual shift in considering how rare nucleic acids can be amplified and detected in or from a sample.
  • detection of rare nucleic acids was performed by trying to find ways of improving the specificity and sensitivity of amplification and detection reactions. This is because the better the reaction can specifically amplify and identify a nucleic acid of interest, the better the reliability and throughput of the system.
  • the present invention takes an entirely different approach to identifying nucleic acids of interest. Instead of trying to fish the nucleic acid of interest out of a complex sample directly, the entire sample is simply deconstructed into low copy number aliquots and the low copy number aliquots are subjected to amplification reactions and individual detection until the nucleic acid of interest is found. Continuing with the simple analogy, the entire haystack is broken apart into individual pieces of hay and each is examined to see if it is hay or needle. This low or single copy amplification concept can provide analyses with high sensitivity against a very low background.
  • Such low or single copy amplifications can be especially useful in the present invention for evaluation of a nucleic acid length.
  • hybridization of a dot blotted sample with a pair probes specific to opposite ends of a target nucleic acid can yield ambiguous results on the integrity of the target. Whether or not the target nucleic acid is fragmented, signals from both probes will be detected on the same blot.
  • detection of signals from both probes would indicate target not fragmented between sequences complimentary to the probes.
  • Modem high-throughput systems make this new conceptual approach possible, i.e., the ability to run massively high numbers of amplification reactions at low cost, e.g., using microfluidic amplification technologies, makes it possible to much more exhaustively sample for any particular nucleic acid of interest in a sample.
  • the continuous flow or high throughput stopped flow nature of these systems further facilitates the approach.
  • examination of a sample by such exhaustive sampling methods provides a great deal of quantitative information (and the concomitant possibility of statistical analysis) with respect to the composition of the sample and the proportions of fragmented or unfragmented nucleic acid of interest. This, in turn, provides diagnostic and prognostic information associated with to the abundance (or relative abundance) of the nucleic acids of interest.
  • An “aliquot” is a portion of a component of interest (e.g., a sample or reaction mixture).
  • the aliquot can be diluted, concentrated or undiluted as compared to the component of interest.
  • a “nucleic acid of interest” is any nucleic acid to be amplified, detected and/or quantified in a sample.
  • a nucleic acid of interest can be detected and identified in fragmented form and/or in unfragmented form using methods and systems of the invention.
  • An “amplification reaction” is a reaction that 1) results in amplification of a template, or 2) would result in amplification of a template if the template were present.
  • an “amplification reaction” can be performed on a sample aliquot that comprises a nucleic acid to be amplified, or on a sample aliquot that does not comprise the nucleic acid. Actual amplification of a template is not a requirement for performing an amplification reaction.
  • reaction mixture refers to a mixture of constituents of an amplification reaction and/or a hybridization reaction. An aliquot of a reaction mixture containing a nucleic acid of interest, or not, can still be considered a reaction mixture.
  • a single copy reaction mixture includes constituents of a reaction mixture in a volume where a nucleic acid of interest, and any associated amplicons, do not overlap with another nucleic acid of interest or its associated amplicons.
  • a “zero copy” reaction mixture or aliquot is a reaction mixture or aliquot that has no copies of the relevant nucleic acid (e.g., a nucleic acid of interest, or an additional nucleic acid). It can comprise nucleic acids from a sample other than the relevant nucleic acid(s), or it can be completely devoid of any template nucleic acids from the sample.
  • the relevant nucleic acid e.g., a nucleic acid of interest, or an additional nucleic acid.
  • a “single copy” reaction mixture has 1 copy of the relevant nucleic acid.
  • the reaction mixture can be an amplification reaction mixture or hybridization mixture containing, e.g., a single copy of a nucleic acid of a given length, or fragment thereof.
  • a “low copy” reaction mixture or aliquot is a reaction mixture or aliquot that has only a few copies of the relevant nucleic acid(s). Typically, such a reaction will have 50 or fewer, generally 25 or fewer, usually 10 or fewer and often 5 or fewer, 2 or fewer or 1 or fewer copies of the relevant nucleic acid(s).
  • a “high copy” nucleic acid reaction mixture or aliquot has at least 1 order of magnitude more copies than the low copy number reaction mixture or aliquot, and generally 2, 3, 4, or even 5 or more orders of magnitude more than the low copy number reaction mixture.
  • a nucleic acid is “quantified” or “quantitated” in a sample when an absolute or relative amount of the nucleic acid in a sample is determined. This can be expressed as a number of copies, a concentration of the nucleic acid, a ratio or proportion of the nucleic acid to some other constituent of the sample (e.g., another nucleic acid), or any other appropriate expression.
  • a “given length” of a nucleic acid of interest refers to a distance between two probes hybridized to the nucleic acid plus the sequences complimentary to the probes.
  • the given length can be a known distance, measured, e.g., in units of base pairs, or an unknown distance determined to exist as an unfragmented sequence, e.g., by detection of coincident signals from a low or singly copy reaction mixture.
  • probes refers to probes complimentary to or specifically hybridizing to different target sequences under stringent hybridization conditions.
  • different detectable markers refers to detectable markers that provide signals distinguishable by a detector in the invention.
  • the nucleic acid of interest to be detected in the methods of the invention can be essentially any nucleic acid.
  • the sequences for many nucleic acids and amino acids are available. No attempt is made to identify the hundreds of thousands of known nucleic acids, any of which can be detected in the methods of the invention.
  • Common sequence repositories for known nucleic acids include GenBank EMBL, DDBJ and the NCBI. Other repositories can easily be identified by searching the internet.
  • the nucleic acid can be an RNA (e.g., where amplification includes RT-PCR or LCR) or DNA (e.g., where amplification includes PCR or LCR), or an analogue thereof (e.g., for detection of synthetic nucleic acids or analogues thereof).
  • Any variation in a nucleic acid can be detected, e.g., a mutation, a single nucleotide polymorphism (SNP), an allele, an isotype, a fragment, a full-length nucleic acid, an amplicon, etc.
  • SNP single nucleotide polymorphism
  • the present invention is quantitative, one can detect variations in expression levels, fragmentation, or gene copy numbers by the methods.
  • the methods of the invention are particularly useful in screening samples derived from patients for the nucleic acids of interest, e.g., from bodily fluids and/or waste from the patient. This is because samples derived from relatively large volumes of such materials can be screened in the methods of the invention (removal of such materials is also relatively non-invasive).
  • the nucleic acids of interest e.g., present in cancer cells
  • can easily comprise 1% or less of the related nucleic acid population of the sample e.g., about 1%, 0.1%, 0.001%, 0.0001% or less of the alleles for a gene of interest).
  • whole blood, serum, plasma, stool, urine, vaginal secretions, ejaculatory fluid, synovial fluid, a biopsy, cerebrospinal fluid, and amniotic fluid, sputum, saliva, lymph, tears, sweat, or urine, or the like can easily be screened for rare nucleic acids or fragmentation by the methods of the invention, as can essentially any tissue of interest.
  • samples are typically taken, following informed consent, from a patient by standard medical laboratory methods.
  • nucleic acids Prior to aliquotting and amplification, nucleic acids are optionally purified from the samples by any available method, e.g., those taught in Berger and Kimmel, Guide to Molecular Cloning Techniques, Methods in Enzymology volume 152 Academic Press, Inc., San Diego, Calif. (“Berger”); Sambrook et al., Molecular Cloning—A Laboratory Manual (3rd Ed.), Vol. 1-3, Cold Spring Harbor Laboratory, Cold Spring Harbor, N.Y., 2000 (“Sambrook”); and/or Current Protocols in Molecular Biology, F. M. Ausubel et al., eds., Current Protocols, a joint venture between Greene Publishing Associates, Inc.
  • kits are also commercially available for the purification of nucleic acids from cells or other samples (see, e.g., EasyPrepTM, FlexiPrepTM, both from Pharmacia Biotech; StrataCleanTM, from Stratagene; and, QIAprepTM from Qiagen). Alternately, samples can simply be directly subjected to amplification, e.g., following aliquotting and dilution.
  • One advantage of single molecule detection is that the low concentration of sample components in the reaction can reduce the need for nucleic acid purification. That is, dilution of the sample reduces the abundance of unwanted components at the same time it distributes the nucleic acid of interest into reaction mixtures.
  • nucleic acids of interest to be detected in the methods herein are those involved in cancer.
  • Any nucleic acid that is associated with cancer can be detected in the methods of the invention, e.g., those that encode over expressed or mutated polypeptide growth factors (e.g., sis), over expressed or mutated growth factor receptors (e.g., erb-B1), over expressed or mutated signal transduction proteins such as G-proteins (e.g., Ras), or non-receptor tyrosine kinases (e.g., abl), or over expressed or mutated regulatory proteins (e.g., myc, myb, jun, fos, etc.) and/or the like.
  • polypeptide growth factors e.g., sis
  • erb-1 over expressed or mutated signal transduction proteins
  • G-proteins e.g., Ras
  • non-receptor tyrosine kinases e.g., abl
  • nucleic acids of interest are screened for the amount of fragmentation, with high fragmentation generally associated with apoptosis of normal cells and less fragmentation associated, e.g., with sloughing of cancer cells.
  • cancer can often be linked to signal transduction molecules and corresponding oncogene products, e.g., nucleic acids encoding Mos, Ras, Raf, and Met; and transcriptional activators and suppressors, e.g., p53, Tat, Fos, Myc, Jun, Myb, Rel, and/or nuclear receptors.
  • p53 colloquially referred to as the “molecular policeman” of the cell, is of particular relevance, as about 50% of all known cancers can be traced to one or more genetic lesion in p53.
  • nuclear receptors include those for glucocorticoids (GRs), androgens (ARs), mineralocorticoids (MRs), progestins (PRs), estrogens (ERs), thyroid hormones (TRs), vitamin D (VDRs), retinoids (RARs and RXRs), and the peroxisome proliferator activated receptors (PPARs) that bind eicosanoids.
  • GRs glucocorticoids
  • ARs mineralocorticoids
  • PRs progestins
  • PRs progestins
  • ERs estrogens
  • TRs thyroid hormones
  • VDRs vitamin D
  • RARs and RXRs retinoids
  • PPARs peroxisome proliferator activated receptors
  • nucleic acids of interest are those that are diagnostic of colon cancer, e.g., in samples derived from stool.
  • Colon cancer is a common disease that can be sporadic or inherited.
  • the molecular basis of various patterns of colon cancer is known in some detail.
  • germline mutations are the basis of inherited colon cancer syndromes, while an accumulation of somatic mutations is the basis of sporadic colon cancer.
  • Ashkenazi Jews a mutation that was previously thought to be a polymorphism may cause familial colon cancer. Mutations of at least three different classes of genes have been described in colon cancer etiology: oncogenes, suppressor genes, and mismatch repair genes.
  • nucleic acid encodes DCC (deleted in colon cancer), a cell adhesion molecule with homology to fibronectin.
  • An additional form of colon cancer is an autosomal dominant gene, hMSH2, that comprises a lesion. Familial adenomatous polyposis is another form of colon cancer with a lesion in the MCC locus on chromosome #5.
  • colon cancers and colon cancer markers that can be detected in stool, see, e.g., Boland (2002) “Advances in Colorectal Cancer Screening: Molecular Basis for Stool-Based DNA Tests for Colorectal Cancer: A Primer for Clinicians” Reviews In Gastroenterological Disorders Volume 2, Supp. 1 and the references cited therein.
  • mutations in a variety of other genes that correlate with cancer, such as Ras and p53 are useful diagnostic indicators for cancer.
  • detection of fragmentation levels using methods of the present invention can be particularly useful in detection of colon cancer. For example, as the amount of total patient DNA available in a stool specimen is low, the amplification aspect of the present invention can be beneficial to examination of the DNA.
  • DNA entering the colon lumen from a colon tumor cells can remain generally unfragmented. Detecting the presence of a proportion of unfragmented nucleic acids over a certain threshold in a stool specimen can correlate to presence of a colon cancer.
  • Cervical cancer is another preferred target for detection, e.g., in samples obtained from vaginal secretions. Cervical cancer can be caused by the papova virus and has two oncogenes, E6 and E7. E6 binds to and removes p53 and E7 binds to and removes PRB. The loss of p53 and uncontrolled action of E2F/DP growth factors without the regulation of pRB is one mechanism that leads to cervical cancer. Furthermore, as with colon cancer, detecting the presence of a proportion of unfragmented nucleic acids over a certain threshold in a vaginal swab can correlate to the presence of a cervical cancer.
  • Retinoblastoma is a tumor of the eyes which results from inactivation of the pRB gene. It has been found to transmit heritably when a parent has a mutated pRB gene (and, of course, somatic mutation can cause non-heritable forms of the cancer).
  • Neurofibromatosis Type 1 can be detected in the methods of the invention.
  • the NF1 gene is inactivated, which activates the GTPase activity of the ras oncogene. If NF1 is missing, ras is overactive and causes neural tumors.
  • the methods of the invention can be used to detect Neurofibromatosis Type 1 in CSF or via tissue sampling.
  • cancers that can be detected by detecting appropriate lesions or fragmentation values include cancers of the lymph, blood, stomach, gut, colon, testicles, pancreas, bladder, cervix, uterus, skin, and essentially all others for which an associated genetic lesion or fragmentation threshold exists.
  • nucleic acids from pathogenic or infectious organisms can be detected by the methods of the invention, e.g., for infectious fungi, e.g., Aspergillus, or Candida species; bacteria, particularly E. coli, which serves a model for pathogenic bacteria (and, of course certain strains of which are pathogenic), as well as medically important bacteria such as Staphylococci (e.g., aureus ), or Streptococci (e.g., pneumoniae ); protozoa such as sporozoa (e.g., Plasmodia ), rhizopods (e.g., Entamoeba ) and flagellates ( Trypanosoma, Leishmania, Trichomonas, Giardia, etc.); viruses such as (+) RNA viruses (examples include Poxviruses e.g., vaccinia; Picornaviruses, e.g.
  • RNA viruses e.g., Rhabdoviruses, e.g., VSV; Paramyxovimses, e.g., RSV; Orthomyxovimses, e.g., influenza; Bunyaviruses; and Arenaviruses
  • dsDNA viruses Reoviruses, for example
  • RNA to DNA viruses i.e., Retroviruses, e.g., H[V and HTLV, and certain DNA to RNA viruses such as Hepatitis B.
  • Single and low copy amplification methods of the invention can be useful in many cases, e.g., in exudates from bacterial infections to identify living (having full length nucleic acids) versus dead and lysed pathogens (having fragmented nucleic acids).
  • nucleic acid encoding enzymes can also be detected according to the methods herein, such as amidases, amino acid racemases, acylases, dehalogenases, dioxygenases, diarylpropane peroxidases, epimerases, epoxide hydrolases, esterases, isomerases, kinases, glucose isomerases, glycosidases, glycosyl transferases, haloperoxidases, monooxygenases (e.g., p450s), lipases, lignin peroxidases, nitrile hydratases, nitrilases, proteases, phosphatases, subtilisins, transaminase, and nucleases.
  • amidases e.g., amino acid racemases, acylases, dehalogenases, dioxygenases, diarylpropane peroxidases, epimerases, epoxide hydrolases, esterases, isome
  • agriculturally related proteins such as insect resistance proteins (e.g., the Cry proteins), starch and lipid production enzymes, plant and insect toxins, toxin-resistance proteins, Mycotoxin detoxification proteins, plant growth enzymes (e.g., Ribulose 1,5-Bisphosphate Carboxylase/Oxygenase, “RUBISCO”), lipoxygenase (LOX), and Phosphoenolpyruvate (PEP) carboxylase can also be detected.
  • insect resistance proteins e.g., the Cry proteins
  • starch and lipid production enzymes e.g., plant and insect toxins, toxin-resistance proteins, Mycotoxin detoxification proteins
  • plant growth enzymes e.g., Ribulose 1,5-Bisphosphate Carboxylase/Oxygenase, “RUBISCO”
  • LOX lipoxygenase
  • Phosphoenolpyruvate (PEP) carboxylase can also be detected.
  • the sample can be aliquotted and/or diluted using standard or microfluidic fluid handling approaches (or combinations thereof).
  • Standard fluid handling approaches for dilution/aliquotting include, e.g., pipetting appropriate volumes of the sample into microtiter trays and adding an appropriate diluent. These operations can be performed manually or using available high throughput fluid handlers, such as, e.g., those designed to use serially dilute solutions in microtiter trays.
  • High throughput equipment e.g., incorporating automated pipettors and robotic microtiter tray handling
  • the present invention contemplates making and using high numbers of aliquots of a sample of interest.
  • a conventional high throughput systems can be used in place of, or in conjunction with microfluidic systems (for example, conventional systems can be used to aliquot samples into microtiter trays, from which microfluidic systems can draw materials) in practicing the methods of the invention.
  • emulsions are created, where sample aliquots comprise or consist of droplets within the emulsions.
  • the emulsions can be amplified by standard thermocyclic reactions and amplified nucleic acids detected within droplets of the emulsions using standard equipment (e.g., flow cytometers, microscope stations, or CCD arrays).
  • Microfluidic systems provide a preferred fluid handling and amplification technology that can conveniently be applied to the present invention.
  • samples are drawn into microfluidic devices that comprise networks of microscale cavities (channels, chambers, etc., having at least one dimension less than about 500 ⁇ M in size and often less than about 100 ⁇ M) and the samples are mixed, diluted, aliquotted or otherwise manipulated in the network of cavities (e.g., channels and/or chambers).
  • the microscale device can comprise one or more capillary, in fluid communication with the network, extending outward from a body structure of the microscale device.
  • Negative pressure is applied to the capillary and fluids are drawn into the network from a container (e.g., a well on a microtiter tray).
  • a container e.g., a well on a microtiter tray.
  • This process can be multiplexed by using a device that comprises multiple capillary channels, permitting many samples to be drawn into the network and processed simultaneously. Alternately, multiple samples can be sequentially drawn into the microfluidic device and routed internally to multiple channels for simultaneous processing and analysis.
  • Sample interfaces with dried samples can also be performed using this basic system, e.g., by partly or completely expelling fluid from the capillary to hydrate samples prior to drawing them into the microfluidic device (the fluid is typically contacted to the samples as a hanging drop on the tip of the capillary and then drawn back into the capillary).
  • U.S. Pat. No. 6,482,364 to Parce, et al. Nov. 19, 2002
  • MICROFLUIDIC SYSTEMS INCLUDING PIPETTOR ELEMENTS U.S. Pat. No. 6,042,709 to Parce, et al. (Mar. 28, 2000) MICROFLUIDIC SAMPLING SYSTEM AND METHODS; U.S.
  • Samples and components to be mixed/diluted or aliquotted can be brought into the microscale device through pipettor elements or from reaction component reservoirs on the device itself, or, commonly, both.
  • the sample can be brought into the microfluidic device through a pipettor channel and diluted and supplied with common reagents from an on device dilution and/or reagent reservoir(s).
  • Locus specific reagents e.g., amplification primer pairs
  • the functions the chip performs typically include reaction assembly (assembly of reaction mixtures), thermocycling, and acting as a “cuvette” for an optical system during an imaging (detection) step.
  • reaction assembly the reaction mixture components (particularly magnesium and the enzyme) which get combined at the last second before heating begins are assembled. This is called a “hot start” and provides advantages of specificity.
  • thermocycling the system optionally provides both constant fluid movement and a continuous sequence of temperature changes.
  • a high data rate CCD is useful in providing an adequate dynamic range using the dispersion/diffusion methods of quantification.
  • the aliquotting and/or dilution events can be performed to achieve particular results.
  • a sample can be diluted equally in each aliquot, or, alternately, the aliquots can be differentially diluted (e.g., a dilution series can be made).
  • the aliquots themselves can be of a volume that is appropriate to the fluid handling approach being used by the system, e.g., on the order of a few microliters for microtiter plates to 100 nL, 10 nL or even 1 nL or less for microfluidic approaches.
  • the aliquots can be selected to have high or low copy numbers of any relevant nucleic acid (e.g., for low copy number aliquots, 50 or fewer, generally 25 or fewer, usually 10 or fewer and often 5 or fewer, 2 or fewer or 1 or fewer copies of the relevant nucleic acid(s)).
  • the number of aliquots generated will depend on the size of the sample and the amount of quantitative information desired by the practitioner. For example, where simple detection of a rare nucleic acid is desired, enough low and/or single copy number aliquots are made of the sample to detect the nucleic acid in one of the aliquots. Where more quantitative information is needed, enough copies are made to provide reliable statistical information, e.g., to a given confidence value.
  • this can include anywhere from 1 aliquot to 10 9 or more aliquots, e.g., 10, 100, 1,000, 10,000, 100,000, 1,000,000, 1,000,000,000 or more aliquots.
  • aliquots there is no theoretical limit on the number of aliquots that can be made and assessed for a nucleic acid of interest according to the present invention, though there are practical considerations with respect to the throughput of the system and the size of the sample (the lower the throughput, the fewer aliquots can be analyzed in a given time; the larger the sample size the more aliquots can be made of the sample).
  • reagent usage and concomitant reagent costs
  • the systems of the invention can greatly speed the process of searching many different samples for a nucleic acid of interest.
  • stopped flow approaches simultaneous processing of signals from PCR reactions can be used to speed the process of searching samples for a nucleic acid of interest.
  • about 150 aliquots for each dilution range was sufficient to provide reasonable quantitative information for Poisson statistics for model samples. Obviously, more or fewer aliquots can be used in the methods as well.
  • the flow rate can be used to calculate how many aliquots have passed (undetected) by a detector prior to detection of the nucleic acid of interest.
  • non-continuous flow systems e.g., microwell plate based systems
  • any event anywhere from 1 to 10 6 or more zero copy reactions can be made and assessed by the system, e.g., about 10, 25, 50, 100, 500, 1,000, 10,000, 100,000, or 1,000,000 or more zero copy reactions can be detected in the process of detecting a nucleic acid of interest.
  • additional nucleic acids other than the nucleic acid of interest e.g., controls, or alternate alleles of a nucleic acid of interest that are also amplified by the relevant locus specific reagent
  • the proportion of such alternate nucleic acids in the system to the nucleic acid of interest can range from less than 1 to 10 9 or more, e.g., 1 ⁇ , 10 ⁇ , 100 ⁇ , 1,000 ⁇ , 10,000 ⁇ , 100,000 ⁇ , 1,000,000 ⁇ , 1,000,000,000 ⁇ or more.
  • the continuous flow format is a surprisingly efficient system, meaning that a high proportion of single molecules that get into the system are amplified. This efficiency is useful in ensuring that very rare molecules are detected, if present, for example in a biowarfare or infectious disease detection applications.
  • Evidence for high efficiency is in the examples, tables and figures herein.
  • the systems of the invention can be used to amplify at least 90%, generally 95%, often 99% or more of the rare molecules that are present in sample of interest, or that are present in a collection of aliquots that are subjected to amplification. Efficiency factors can be determined, e.g., empirically, for adjustment of mathematical formulas for more accurate quantitative interpretations of signal data.
  • the methods of the invention include amplifying one or more sequences of a nucleic acid of interest from a sample or aliquot and, optionally, one or more additional nucleic acids. Typically two or more sequences of a nucleic acid of interest are amplified at separated positions to allow interpretation of the nucleic acid length.
  • Any available amplification method can be used, including PCR, RT-PCR, LCR, and/or any of the various RNA mediated amplification methods.
  • PCR, RT-PCR and LCR are preferred amplification methods for amplifying a nucleic acid of interest in the methods of the invention.
  • Real time PCR and/or RT-PCR e.g., mediated via TaqManTM probes or molecular beacon-based probes
  • a molecular beacon is an oligonucleotide or PNA which, under appropriate hybridization conditions, self-hybridizes to form a stem and loop structure.
  • the MB has a label and a quencher at the termini of the oligonucleotide or PNA; thus, under conditions that permit intra-molecular hybridization, the label is typically quenched (or at least altered in its fluorescence) by the quencher. Under conditions where the MB does not display intra-molecular hybridization (e.g., when bound to a target nucleic acid, e.g., to a region of an amplicon during amplification), the MB label is unquenched.
  • MBs are robust reagents for detecting and quantifying nucleic acids, including in real time, e.g., during PCR, LCR or other nucleic acid amplification reactions (e.g., MBs can be used to detect targets as they are formed).
  • a variety of commercial suppliers produce standard and custom molecular beacons, including Cruachem (cruachem.com), Oswel Research Products Ltd. (UK; oswel.com), Research Genetics (a division of Invitrogen, Huntsville Ala. (resgen.com)), the Midland Certified Reagent Company (Midland, Tex. mcrc.com) and Gorilla Genomics, LLC (Alameda, Calif.).
  • kits which utilize molecular beacons are also commercially available, such as the SentinelTM Molecular Beacon Allelic Discrimination Kits from Stratagene (La Jolla, Calif.) and various kits from Eurogentec SA (Belgium, eurogentec.com) and Isogen Bioscience BV (The Netherlands, isogen.com).
  • MB components can be synthesized using conventional methods.
  • oligos or peptide nucleic acids PNAs
  • PNAs peptide nucleic acids
  • Labels can be attached to the oligos or PNAs either during automated synthesis or by post-synthetic reactions which have been described before see, e.g., Tyagi and Kramer (1996) “Molecular beacons: probes that fluoresce upon hybridization” Nature Biotechnology 14:303-308 and U.S. Pat. No. 6,037,130 to Tyagi et al (Mar.
  • Labels/quenchers can be introduced to the oligonucleotides or PNAs, e.g., by using a controlled-pore glass column to introduce, e.g., the quencher (e.g., a 4-dimethylaminoazobenzene-4′-sulfonyl moiety (DABSYL).
  • the quencher e.g., a 4-dimethylaminoazobenzene-4′-sulfonyl moiety (DABSYL).
  • the quencher can be added at the 3′ end of oligonucleotides during automated synthesis; a succinimidyl ester of 4-(4′-dimethylaminophenylazo)benzoic acid (DABCYL) can be used when the site of attachment is a primary amino group; and 4-dimethylaminophenylazophenyl-4′-maleimide (DABMI) can be used when the site of attachment is a sulphydryl group.
  • DBCYL succinimidyl ester of 4-(4′-dimethylaminophenylazo)benzoic acid
  • DABMI 4-dimethylaminophenylazophenyl-4′-maleimide
  • fluorescein can be introduced in the oligos, either using a fluorescein phosphoramadite that replaces a nucleoside with fluorescein, or by using a fluorescein dT phosphoramadite that introduces a fluorescein moiety at a thymidine ring via a spacer.
  • fluorescein dT phosphoramadite that introduces a fluorescein moiety at a thymidine ring via a spacer.
  • iodoacetoamidofluorescein can be coupled to a sulphydryl group.
  • Tetrachlorofluorescein (TET) can be introduced during automated synthesis using a 5′-tetrachloro-fluorescein phosphoramadite.
  • PCR quantification using dual-labeled fluorogenic oligonucleotide probes can be performed according to the present invention.
  • These probes are composed of short (e.g., 20-25 base) oligodeoxynucleotides that are labeled with two different fluorescent dyes. On the 5′ terminus of each probe is a reporter dye, and on the 3′ terminus of each probe a quenching dye is found.
  • the oligonucleotide probe sequence can be complementary to an internal target sequence present in a PCR amplicon.
  • the probe When the probe is intact, energy transfer occurs between the two fluorophores and emission from the reporter is quenched by the quencher (fluorescent resonant energy transfer or FRET).
  • FRET fluorescent resonant energy transfer
  • the probe is cleaved by 5′ nuclease activity of the polymerase used in the reaction, thereby releasing the reporter from the oligonucleotide-quencher and producing an increase in reporter emission intensity.
  • TaqManTM probes are oligonucleotides that have a label and a quencher, where the label is released after hybridization and during amplification by the exonuclease action of the polymerase used in amplification. This provides a real time measure of amplification during synthesis.
  • a variety of TaqManTM reagents are commercially available, e.g., from Applied Biosystems (Division Headquarters in Foster City, Calif.) as well as from a variety of specialty vendors such as Biosearch Technologies (e.g., black hole quencher probes).
  • oligonucleotides In general, synthetic methods for making oligonucleotides, including probes, molecular beacons, PNAs, LNAs (locked nucleic acids), etc., are well known.
  • oligonucleotides can be synthesized chemically according to the solid phase phosphoramidite triester method described by Beaucage and Caruthers (1981), Tetrahedron Letts., 22(20):1859-1862, e.g., using a commercially available automated synthesizer, e.g., as described in Needham-VanDevanter et al. (1984) Nucleic Acids Res., 12:6159-6168.
  • Oligonucleotides can also be ordered from a variety of commercial sources known to persons of skill. There are many commercial providers of oligo synthesis services, and thus this is a broadly accessible technology. Any nucleic acid can be custom ordered from any of a variety of commercial sources, such as The Midland Certified Reagent Company (mcrc@oligos.com), The Great American Gene Company (www.genco.com), ExpressGen Inc. (www.expressgen.com), Operon Technologies Inc. (Alameda, Calif.) and many others. Similarly, PNAs can be custom ordered from any of a variety of sources, such as PeptidoGenic (pkim@ccnet.com), HTI Bio-products, inc. (www.htibio.com), BMA Biomedicals Ltd (U.K.), Bio-Synthesis, Inc., and many others.
  • PeptidoGenic pkim@ccnet.com
  • HTI Bio-products inc.
  • microfluidic system can have one or more capillaries extending outwards from the body structure of the microfluidic system for drawing materials into the body structure.
  • microfluidic cavities within the body structure are microfluidic cavities (channels, chambers, or the like having at least one dimension smaller than about 500 microns, and, typically smaller than about 100 microns) in which the amplification reactions are performed.
  • the capillaries that extend out from the body structure can access standard reagent storage elements (microtiter plates, or the like) by drawing fluid into the capillary, e.g., due to application of a vacuum or electroosmotic force.
  • the capillaries can access dried reagent libraries on substrates (e.g., the LibraryCardTM reagent array made by Caliper Technologies) by partly or completely expelling fluid to rehydrate library members and then by drawing the rehydration fluid back into the capillary.
  • the capillary can partly expel fluid to form a hanging drop on the capillary, which is then contacted to the material to be hydrated. The material in the hanging drop is then drawn back into the capillary.
  • molecular beacons or TaqManTM probes can be incorporated into the relevant amplification reaction and detected in the microfluidic device to provide for real time PCR detection.
  • PCR amplicons can be detected by conventional methods, such as hybridization to a labeled probe, e.g., prior to or following a separation operation that separates unhybridized probe from hybridized probe.
  • an electrophoretic separation can be performed in a channel of the microscale device.
  • standard fluid handling approaches are used in place of, or in conjunction with, microfluidic approaches.
  • PCR can be performed in standard reaction vessels (e.g., microtiter plates), as can dilutions or other operations relevant to the present invention.
  • Various high-throughput systems are available for non-microfluidic approaches to fluid handling (typically involving plates comprising several reaction chambers, e.g., 96 well, 384 well or 1536 well microtiter plates). These approaches can utilize conventional robotics to perform fluid handling operations and can use conventional commercially available thermocyclers to perform amplification reactions. See above, for a discussion of automated fluid handling systems.
  • any available method for detecting amplified nucleic acids can be used in the present invention.
  • Common approaches include real time amplification detection with molecular beacons or TaqManTM probes, detection of intercalating dyes (ethidium bromide or sybergreen), detection of labels incorporated into the amplification probes or the amplified nucleic acids themselves, e.g., following electrophoretic separation of the amplification products from unincorporated label), and/or detection of secondary reagents that bind to the nucleic acids. Details on these general approaches is found in the references cited herein, e.g., Sambrook (2000), Ausubel (2002), and the references in the sections herein related to real time PCR detection.
  • Amplified nucleic acids can be detected in homogenous (substantially unseparated) reaction mixtures or solutions (e.g., using molecular beacons or TaqManTM probes) or during or after separation (e.g., by electrophoresis). Details on these strategies can be found in the preceding references.
  • Amplification and detection are commonly integrated in a system comprising a microfluidic device in the present invention.
  • Available microfluidic systems that include detection features for detecting nucleic acids include the 250 HTS system and AMS 90 SE from Caliper Technologies (Mountain View, Calif.), as well as the Agilent 2100 bioanalyzer (Agilent, Palo Alto, Calif.). Additional details regarding systems that comprise detection (and separation/detection) capabilities are well described in the patent literature, e.g., the references already noted herein and in Parce et al. “High Throughput Screening Assay Systems in Microscale Fluidic Devices” WO 98/00231.
  • the devices herein optionally include signal detectors, e.g., which detect fluorescence, phosphorescence, radioactivity, pH, charge, absorbance, luminescence, temperature, magnetism or the like. Fluorescent detection is especially preferred and generally used for detection of amplified nucleic acids (however, upstream and/or downstream operations can be performed on amplicons, which can involve other detection methods, such as mass spectroscopy or size exclusion).
  • signal detectors e.g., which detect fluorescence, phosphorescence, radioactivity, pH, charge, absorbance, luminescence, temperature, magnetism or the like.
  • Fluorescent detection is especially preferred and generally used for detection of amplified nucleic acids (however, upstream and/or downstream operations can be performed on amplicons, which can involve other detection methods, such as mass spectroscopy or size exclusion).
  • the detector(s) optionally monitor one or a plurality of signals from an amplification reaction and/or hybridization reaction.
  • the detector can monitor optical signals which correspond to “real time” amplification assay results.
  • the detector can monitor a single type of signal, or, e.g., simultaneously monitor multiple different signals.
  • Example detectors include photo multiplier tubes, spectrophotometers, CCD arrays, scanning detectors, microscopes, galvo-scanns and/or the like. Amplicons or other components which emit a detectable signal can be flowed past the detector, or, alternatively, the detector can move relative to the site of the amplification reaction (or, the detector can simultaneously monitor a number of spatial positions corresponding to channel regions, or microtiter wells e.g., as in a CCD array). Detectors in the present invention can detect signals from probes associated with nucleic acids of the invention that flow into one or more detection regions, e.g., of a microfluidic device.
  • the detector can include or be operably linked to a computer (or other logic device), e.g., which has software for converting detector signal information into assay result information (e.g., presence of a nucleic acid of interest, the length of a nucleic acid of interest, proportions of nucleic acid of interest lengths, and/or correlations with disease states), or the like.
  • a computer or other logic device
  • assay result information e.g., presence of a nucleic acid of interest, the length of a nucleic acid of interest, proportions of nucleic acid of interest lengths, and/or correlations with disease states
  • Signals are optionally calibrated, e.g., by calibrating the microfluidic system by monitoring a signal from a known source.
  • signals can be calibrated against a reference light source, internal reference signals, or normalized for detection of positive signals over background.
  • a microfluidic system can also employ multiple different detection systems for monitoring signals in the system.
  • Detection systems of the present invention are used to detect and monitor the materials in a particular channel region (or other reaction detection region). Once detected, the flow rate and velocity of any cells or droplets in the channels can be optionally measured by sensors and controlled as described above.
  • detection systems useful in methods and systems of the invention can include optical sensors, temperature sensors, pressure sensors, pH sensors, conductivity sensors, and the like. Each of these types of sensors is readily incorporated into the microfluidic systems described herein. In these systems, such detectors can be placed either within or adjacent to the microfluidic device or one or more channels, chambers or conduits of the device, such that the detector is within sensory communication with the device, channel, or chamber.
  • within sensory communication generally refers to the placement of the detector in a position such that the detector is capable of detecting the property of the microfluidic device, a portion of the microfluidic device, or the contents of a portion of the microfluidic device, for which that detector was intended.
  • a pH sensor placed in sensory communication with a microscale channel is capable of determining the pH of a fluid disposed in that channel.
  • a temperature sensor placed in sensory communication with the body of a microfluidic device is capable of determining the temperature of the device itself.
  • Particularly preferred detection systems include optical detection systems for detecting an optical property of a material within the channels and/or chambers of the microfluidic devices that are incorporated into the microfluidic systems described herein. Such optical detection systems are typically placed adjacent to a microscale channel of a microfluidic device, and are in sensory communication with the channel via an optical detection window that is disposed across the channel or chamber of the device. Optical detection systems include systems that are capable of measuring the light emitted from material within the channel, the transmissivity or absorbance of the material, as well as the materials spectral characteristics. In preferred aspects, the detector measures an amount of light emitted from the material, such as a fluorescent or chemiluminescent material.
  • the detection system will typically include collection optics for gathering a light based signal transmitted through the detection window, and transmitting that signal to an appropriate light detector.
  • Microscope objectives of varying power, field diameter, and focal length are readily utilized as at least a portion of this optical train.
  • the light detectors are optionally spectrophotometers, photodiodes, avalanche photodiodes, photomultiplier tubes, diode arrays, or in some cases, imaging systems, such as charged coupled devices (CCDs) and the like.
  • the detection system is typically coupled to a computer, via an analog to digital or digital to analog converter, for transmitting detected light data to the computer for analysis, storage and data manipulation.
  • the detector typically includes a light source that produces light at an appropriate wavelength for activating the fluorescent material, as well as optics for directing the light source through the detection window to the material contained in the channel or chamber.
  • the light source can be any number of light sources that provides an appropriate wavelength, including lasers, laser diodes, and LEDs. Other light sources are used in other detection systems. For example, broad band light sources are typically used in light scattering/transmissivity detection schemes, and the like. Typically, light selection parameters are well known to those of skill in the art.
  • the detector can exist as a separate unit, but can also be integrated with the system or microfluidic device, into a single instrument. Integration of these functions into a single unit facilitates connection of these instruments with the computer, by permitting the use of few or a single communication port(s) for transmitting information between the controller, the detector and the computer.
  • One feature of the present invention is that it provides for robust quantitation of rare (and other) nucleic acids in a sample.
  • This robust quantitation provides the ability to perform statistical or probabilistic analysis of the sample. For example, Poisson analysis, Monte Carlo analysis, application of genetic algorithms, neural network training, Markov modeling, hidden Markov modeling, multidimensional scaling, partial least squares (PLS) analysis, or principle component analysis (PCA) can all be applied to data generated by the present invention.
  • PLS partial least squares
  • PCA principle component analysis
  • These statistical evaluations can be used to determine, e.g., the concentration, abundance, or length proportions of a given nucleic acid in a sample and to correlate abundance or proportions to diagnosis or prognosis associated with the diagnosis or prognosis.
  • One feature of the invention is the discovery that the highly reproducible peak parameters, e.g., amplitude, width area, and/or shape features of a signal from an amplification reaction can be correlated to the starting copy number for the reaction and/or used to discriminate signals of interest from background fluctuations.
  • This correlation can be performed at the theoretical level, taking thermal diffusivity and Taylor Aris diffusion into account, or it can be performed by comparison to standards (e.g., comparisons to peak shapes, e.g., heights, widths, or general shape profiles for amplification reactions that have known copy numbers for starting materials).
  • the same or different peak parameters can be evaluated in interpretation of detector signals for two on more probes in determination of nucleic acid length.
  • the Taylor dispersion (D T ) is dependent on the dimensions and shape of the microfluidic cavity through which the label is flowed, the flow velocity (u) and the thermal diffusivity (D).
  • D K(d 2 u 2 )/D, where K is a proportionality factor which is a function of the microfluidic cavity through which the label is flowed and d is a characteristic microfluidic cavity length.
  • K is a proportionality factor which is a function of the microfluidic cavity through which the label is flowed
  • d is a characteristic microfluidic cavity length.
  • the systems of the invention can include microfluidic devices, reaction mixtures, detectors, sample storage elements (microtiter plates, dried arrays of components, etc.), flow controllers, amplification devices or microfluidic modules, computers and/or the like. These systems can be used for aliquoting, amplifying and analyzing the nucleic acids of interest.
  • the microfluidic devices, amplification components, detectors and storage elements of the systems have already been described in some detail above. The following discussion describes appropriate controllers and computers, though many configurations are available and one of skill would be expected to be familiar in their use and would understand how they can be applied to the present invention.
  • controlling instrumentation is optionally utilized in conjunction with the microfluidic devices described herein, for controlling the transport and direction of fluids and/or materials within the devices of the present invention, e.g., by pressure-based or electrokinetic control.
  • fluid transport and direction are controlled in whole or in part, using pressure based flow systems that incorporate external or internal pressure sources to drive fluid flow.
  • Internal sources include microfabricated pumps, e.g., diaphragm pumps, thermal pumps, Lamb wave pumps and the like that have been described in the art. See, e.g., U.S. Pat. Nos. 5,271,724, 5,277,556, and 5,375,979 and Published PCT Application Nos. WO 94/05414 and WO 97/02357.
  • the systems described herein can also utilize electrokinetic material direction and transport systems.
  • external pressure sources are used, and applied to ports at channel termini. These applied pressures, or vacuums, generate pressure differentials across the lengths of channels to drive fluid flow through them.
  • differential flow rates on volumes are optionally accomplished by applying different pressures or vacuums at multiple ports, or preferably, by applying a single vacuum at a common waste port and configuring the various channels with appropriate resistance to yield desired flow rates.
  • Example systems are described in U.S. Ser. No. 09/238,467 filed Jan. 28, 1999.
  • the controller systems are appropriately configured to receive or interface with a microfluidic device or system element as described herein.
  • the controller and/or detector optionally includes a stage upon which a microfluidic device is mounted to facilitate appropriate interfacing between the controller and/or detector and the device.
  • the stage includes an appropriate mounting/alignment structural element, such as a nesting well, alignment pins and/or holes, asymmetric edge structures (to facilitate proper device alignment), and the like. Many such configurations are described in the references cited herein.
  • the controlling instrumentation discussed above is also optionally used to provide for electrokinetic injection or withdrawal of material downstream of the region of interest to control an upstream flow rate.
  • the same instrumentation and techniques described above are also utilized to inject a fluid into a downstream port to function as a flow control element.
  • either or both of the controller system and/or the detection system can be coupled to an appropriately programmed processor or computer (logic device) which functions to instruct the operation of these instruments in accordance with preprogrammed or user input instructions, receive data and information from these instruments, and interpret, manipulate and report this information to the user.
  • the computer is typically appropriately coupled to one or both of these instruments (e.g., including an analog to digital or digital to analog converter as needed).
  • the computer typically includes appropriate software for receiving user instructions, either in the form of user input into a set parameter fields, e.g., in a GUI, or in the form of preprogrammed instructions, e.g., preprogrammed for a variety of different specific operations.
  • the software then converts these instructions to appropriate language for instructing the operation of the fluid direction and transport controller to carry out the desired operation.
  • the computer then receives the data from the one or more sensors/detectors included within the system, and interprets the data, either provides it in a user understood format, or uses that data to initiate further controller instructions, in accordance with the programming, e.g., such as in monitoring and control of flow rates (including for continuous flow), temperatures, applied voltages, and the like.
  • the systems and/or kits can include system instructions (e.g., embodied in a computer or in a computer readable medium, e.g., as system software) for practicing any of the method steps herein.
  • the system optionally includes system software that correlates a shape, length, width, volume and/or area occupied by amplified copies of the nucleic acid of interest, as detected by the detector, to the number of copies of the nucleic acid of interest present in one of the aliquots, or to the number of copies of the nucleic acid of interest present in the sample, or both.
  • system optionally includes system instructions that direct the dilution module to aliquot the sample into a plurality of aliquots, including a plurality of zero copy aliquots comprising no copies of the nucleic acids of interest and one or more single copy aliquot comprising a single copy of the nucleic acid of interest.
  • the computer can include statistical or probabilistic system software that performs one or more statistical or probabilistic analysis of signals received from one or more of the aliquots subjected to amplification (e.g., via thermocycling).
  • the statistical or probabilistic analysis can include Poisson analysis, Monte Carlo analysis, application of a genetic algorithm, neural network training, Markov modeling, hidden Markov modeling, multidimensional scaling, PLS analysis, and/or PCA analysis.
  • the statistical or probabilistic analysis software optionally quantitatively determines a concentration, proportion, or number of the nucleic acids of interest in the sample.
  • Computers and software of the systems receive and evaluate signal data from one or more analyses to provide quantitation and/or proportionality determinations for nucleic acids of interest.
  • the amplitude or integrated area of a signal can be adjusted with a conversion factor for an output in desired units, such as, e.g., copies per nL, ng/ ⁇ L, and the like.
  • one or more standard materials of known concentration can be analyzed to provide data for regression analyses wherein changes in detectable signals with changes in concentration are expressed as an equation (standard curve) from which unknown concentrations can be determined by insertion of one or more signal parameters into the equation.
  • quantitation of a nucleic acid of interest can be based on the number of amplification cycles required to obtain a signal of a certain intensity.
  • the computer typically includes software for the monitoring of materials in the channels. Additionally, the software is optionally used to control electrokinetic or pressure modulated injection or withdrawal of material. The injection or withdrawal is used to modulate the flow rate as described above, to mix components, and the like.
  • FIGS. 6 and 7 provide a schematic illustration of a model system of the invention.
  • system 600 includes microfluidic device 601 .
  • Device 601 includes main channel 604 fabricated therein.
  • Amplification components are flowed, e.g., from reservoir 606 , e.g., by applying a vacuum at vacuum source 608 (and/or at any of the reservoirs or wells noted below) through main channel 604 .
  • Amplification components can also be flowed from wells 610 or 612 and into main channel 604 , for example to form a reaction mixture.
  • Materials can be also flowed from wells 606 or 608 , or materials can be flowed into these wells, e.g., when they are used as waste wells, or when they are coupled to a vacuum source.
  • Flow from wells 614 , 612 , 610 , 606 , or 608 can be performed by modulating fluid pressure, or by electrokinetic approaches.
  • an arrangement such as the device of FIG. 1 can be substituted.
  • a variety of other appropriate microfluidic configurations are set forth in the references noted herein.
  • the integrated system can include pipettor channel 620 (sipper), e.g., protruding from device 601 , for accessing an outside source of reagents.
  • pipettor channel 620 can access microwell plate 622 , which includes samples or sample aliquots, or locus specific reagents, or other reagents useful in the practice of the invention in the wells of the plate. Aliquots or reagents relevant to amplification can be flowed into channel 604 through pipettor channel 620 .
  • Detector 624 is in sensory communication with channel 604 , detecting signals resulting, e.g., from the interaction of a label with an amplicon as described above. Detector 624 is operably linked to Computer 626 , which digitizes, stores and manipulates signal information detected by detector 624 .
  • Voltage/pressure controller 628 controls voltage, pressure, or both, e.g., at the wells of the system, or at vacuum couplings fluidly coupled to channel 604 (or the other channels, wells, or chambers noted above).
  • computer 626 controls voltage/pressure controller 628 .
  • computer 626 uses signal information to select further reaction parameters. For example, upon detecting amplification of a nucleic acid of interest in a well from plate 622 , the computer optionally directs withdrawal of additional aliquots from the well for analysis through pipettor channel 620 , e.g., to deliver different concentrations of the aliquot to the amplification reaction.
  • computer 626 can direct controller 628 to process another aliquot. If statistical information is desired, computer 626 directs controller 628 to perform appropriate fluid manipulations to generate enough data for the statistical analysis.
  • Computer 626 is optionally coupled to or comprises a user viewable display, permitting control of the computer by the user and providing a readout for the user to view results detected by the system.
  • FIG. 7 depicts an alternate embodiment, in which a solid phase array of reagents or samples is accessed by a microfluidic system.
  • system 700 includes microfluidic device 701 .
  • Device 701 includes pipettor channel 720 and a microfluidic network fabricated within the device.
  • Amplification components such as primer pairs, polymerases, buffers, probes, etc., are flowed through device 701 , typically by applying pressure (positive or negative) and/or electrokinetic pressure in the microfluidic network.
  • the integrated system can include pipettor channel 720 , e.g., protruding from device 701 , for accessing an outside source of reagents.
  • pipettor channel 720 can access solid phase array 725 , which includes samples or sample aliquots, or locus specific reagents, or other reagents useful in the practice of the invention. Fluids are partly or completely expelled from channel 720 to rehydrate materials on array 725 .
  • channel 720 can comprise a hanging drop that is used to rehydrate materials, with the drop being withdrawn into channel 720 for distribution into microfluidic device 701 .
  • Detector 724 is in sensory communication with device 701 and computer/controller 726 .
  • Computer/controller 726 can be operated in a manner similar to computer 626 of FIG. 6 . In either case, computer 626 or computer controller 726 optionally control movement of tray 622 or array 725 , and/or microfluidic device 601 or 701 to permit the relevant pipettor channel to process samples or other materials on the array or in the wells of the tray.
  • heating systems can be used in the present invention.
  • winding the channel around fixed heating areas can be performed.
  • Robotics or fluid system elements can be used to heat fluids in multiple different temperature water baths (e.g., 3 baths for a typical amplification reaction at typical annealing, reaction and dissociation conditions).
  • kits for carrying out the methods described herein.
  • these kits typically include system components described herein, as well as additional components to facilitate the performance of the methods by an investigator.
  • the kit also typically includes a receptacle in which the system component is packaged.
  • the elements of the kits of the present invention are typically packaged together in a single package or set of related packages.
  • the package optionally includes reagents used in the assays herein, e.g., buffers, amplification reagents, sizing probe pairs, standard reagents, and the like, as well as written instructions for carrying out the assay in accordance with the methods described herein.
  • kits optionally include pre-measured or pre-dosed reagents that are ready to incorporate into the methods without measurement, e.g., pre-measured fluid aliquots, or pre-weighed or pre-measured solid reagents that may be easily reconstituted by the end-user of the kit.
  • the microfluidic devices described herein are optionally packaged to include reagents for performing the device's preferred function.
  • the kits can include any of microfluidic devices described along with assay components, reagents, sample materials, control materials, or the like.
  • Such kits also typically include appropriate instructions for using the devices and reagents, and in cases where reagents are not predisposed in the devices themselves, with appropriate instructions for introducing the reagents into the channels and/or chambers of the device.
  • kits optionally include special ancillary devices for introducing materials into the microfluidic systems, e.g., appropriately configured syringes/pumps, or the like (in one preferred embodiment, the device itself comprises a pipettor element, such as an electropipettor for introducing material into channels and chambers within the device).
  • the device itself comprises a pipettor element, such as an electropipettor for introducing material into channels and chambers within the device.
  • such kits typically include a microfluidic device with necessary reagents predisposed in the channels/chambers of the device. Generally, such reagents are provided in a stabilized form, so as to prevent degradation or other loss during prolonged storage, e.g., from leakage.
  • a number of stabilizing processes are widely used for reagents that are to be stored, such as the inclusion of chemical stabilizers (i.e., enzymatic inhibitors, microcides/bacteriostats, anticoagulants), the physical stabilization of the material, e.g., through immobilization on a solid support, entrapment in a matrix (i.e., a gel), lyophilization, or the like.
  • chemical stabilizers i.e., enzymatic inhibitors, microcides/bacteriostats, anticoagulants
  • the physical stabilization of the material e.g., through immobilization on a solid support, entrapment in a matrix (i.e., a gel), lyophilization, or the like.
  • PCR polymerase chain reaction
  • SNP single nucleotide polymorphism
  • the amplified product is a pure “clone” of a single parent DNA template, instead of a mixture of many DNA parent templates.
  • Single molecule amplification and detection results in some interesting applications that are not achievable otherwise.
  • One such application is the detection of cancer genes.
  • This example describes (1) a method to perform single molecule PCR using microfluidic technology, (2) analysis and detection of single molecule amplification, and (3) example applications using single molecule PCR detection for cancer detection.
  • a DNA sample (e.g., a genomic DNA) was brought onto chip 100 through a sipper using a pressure gradient into distribution channel 105 .
  • the sample was first mixed with a common reagent from an on-chip reagent reservoir through common reagent channel 106 , then split into 8 equal aliquots into 8 independent analysis channels 110 - 118 .
  • Each aliquot was mixed with locus-specific reagents supplied from a channel-specific chip reservoir to form a reaction mixture, then flowed through heated region 130 comprising metal traces proximal to amplification microchannel 110 - 118 to provide controlled heated regions of chip 100 .
  • Reagent addition for channel specific reagents into channels 110 - 118 provides an elegant microfluidic method of providing for an on-chip “hot start,” in which all of the reagents are added to analysis channels just before amplification.
  • the temperature of the region was cycled appropriately (temperature set points and respective dwell times are controlled) for PCR conditions in the channels in heated region 130 .
  • 8 channel detection region 135 comprises an appropriate detector for detecting PCR amplicons in channels 110 - 118 .
  • PCR sipper chip illustrated in FIG. 1 was used to demonstrate single molecule PCR amplification, experimentally, in a continuous flow format.
  • Table 1 summarizes results of a set of PCR experiments when the average copy number of DNA in each of the 8 channels varied from 0.02 to 48. For each DNA concentration, 8 PCR experiments were done simultaneously. The number of occurrences of a measurable PCR fluorescent signal for each sample was recorded in Table 1, with the maximum occurrence being 8 and the minimum being 0. The percent of occurrence of PCR was calculated and compared with a Poisson statistics prediction. A very good agreement between the measured and predicted percent occurrence of PCR was found.
  • Table 2 summarizes a replication of similar sets of experiments on another day.
  • FIG. 2 is a graphical comparison of predicted (Poisson) and measured statistics (Run 1 and 2) for both sets of experiments. Predicted and actual measurements show close agreement.
  • FIGS. 3A and 3B summarize analysis of peak area and peak (slug) width as a function of starting copy number of DNA in channels. The lower copy number amplifications in fact showed narrow peaks as expected (and vice-versa).
  • Evidence for the system amplifying and typing single molecules also includes the fact that when the sample is a heterozygote, all peaks are positive for one or the other TaqMan probe, but not both.
  • TaqManTM or molecular beacon assays can be developed for sequences that are located close together in the genome. Those assays can be used to determine whether the proximal sequences are present on the same amplified molecule. This is an indirect way of doing a sizing assay: one can ask whether individual molecules have both TaqManTM/beacon sites, providing an indication of how often molecules are of a size that encompasses both sites. One can also type the two sites, providing a haplotyping method.
  • FIG. 5 shows the peak width at 1 ⁇ 2 max vs. calculated input copy number per channel (on-chip).
  • PCR reagents were loaded on-chip.
  • One DNA sample was diluted in assay buffer in a 384-well plate (0.72 ng/ ⁇ L to 11.5 ng/ ⁇ L).
  • the amplification cycle time was 17 seconds (5 seconds at 90° C., 7 seconds at 58° C. and 5 seconds at 72° C.). All injected samples were subjected to a total of 35 amplification cycles. Samples were injected for a total of 200 seconds, with a buffer wash (between samples) of 350 seconds. Width of PCR signal (peak) was measured at 1 ⁇ 2 the peak maximum for each microfluidic channel on-chip (8 total).
  • FIG. 4 illustrates raw fluorescence intensity measurements for SNP analysis at very low starting copy number to below 1 copy per channel on average.
  • FIGS. 10-12 show additional data from additional experimental runs, demonstrating single molecule amplification.
  • a first set of experimental data with 100% of a first SNP allele is shown on the left, while a second set of experimental data with 1% of a second SNP allele (and 99% of the first allele) is shown on the right.
  • the top signal line on the figure is data using a first dye detection system (which provides a longer wavelength “red” signal) for detecting amplification of the first allele, while the bottom line is data from a second dye detection system (a shorter wavelength “blue” signal) for detecting amplification of the second allele.
  • the data represents about 700 total detected DNA molecules in one sample slug.
  • FIG. 11 provides results for 6 separate experimental runs, demonstrating that characteristic peak shapes from molecule to molecule is constant, providing experimental evidence that both PCR and dispersion of the resulting amplicons are very reproducible.
  • a LabChip®-based system allows unlimited sensitivity to rare molecules in that: 1) it is practical, in a microfluidic system, to spread the sample out through the channel such that rare molecules are present amongst smaller numbers of wild-type molecules (reducing the problems created by proportional amplification of starting materials in each aliquot); and 2) reproducible fluidic handling and analysis gives a predictable single molecule peak shape that can be used to discriminate between molecular signals and random signal fluctuations.
  • FIG. 12 provides a titration of a first SNP against a second SNP, showing that the signal from the amplicons corresponding to the first SNP (“FAM DNA,” in the upper trace) and the second SNP (“VIC DNA,” lower trace) show an appropriate signal correlation.
  • the given percentages correspond to the percentage of DNA from a first homozygous sample (both alleles in the first homozygous sample are “FAM”, that is, the material from the homozygous sample is “FF” homozygous) and a second homozygous sample (both alleles in the are VIC DNA sample, “VV”).
  • FAM DNA stands for a DNA sequence that is probed for by a specific oligo sequence with a FAM dye label
  • VIC DNA stands for a DNA sequence that is probed for by a specific oligo sequence with a VIC dye label
  • FF stands for a homozygous DNA sample for the “FAM” (oligo) sequence
  • VV stands for a homozygous DNA sample for the “VIC” (oligo) sequence.
  • FIG. 13 provides an example of detection of 2 mutation sites relevant to cancer detection developed on-chip, using TaqMan probes.
  • cancer e.g., colorectal cancer
  • Two of those assays are shown in FIG. 13 : one for the K-RAS gene and one for the p53 gene, both diagnostic markers for a variety of cancers, such as colon cancer.
  • the data trace shows fluorescence at two wavelengths vs. time for one microfluidic channel.
  • Two TaqMan probes, one specific for the normal allele, and one specific for the mutant allele, were designed and tested in this on-chip assay format.
  • the presence of normal DNA is detected with the wild-type probe (a “red” signal, designated in the black and white reproduction on the top data trace) and mutant DNA molecules are detected with the mutant probe (a “blue” signal, represented in the black and white reproduction as the bottom data trace).
  • Most of the DNA molecules (approximately 500) in the sample slug are normal, shown by the high “red” top fluorescent signal and low “blue” bottom fluorescent signal. This signal is produced by the allele-specific (red, top) and background (blue, bottom) TaqMan probe cleavage surrounding the amplification products of normal genomic DNA molecules.
  • a mutant molecule synthetic DNA template with the appropriate point mutation
  • Continuous flow PCR systems allow for spatial separation of individual low, single copy, and zero copy amplification reaction mixtures in a microfluidic processing environment. Normally, spatial separation is used to separate different reactions, where the starting template concentration is high enough to ensure accurate representation of alleles coming from both parents (e.g., about 50 genome equivalents are often used).
  • the same task is accomplished by diluting the DNA enough such that individual template molecules are separated such that the amplification and detection products for each one are fluidically separated. If the detection product is allele specific, a signal for only one of the two alleles is detected. One can the count the results for each allele, giving the genotype quite accurately.
  • genotyping by this method is that the throughput decreases: one needs many reactions to get a genotype, instead of just one.
  • Genotyping is typically performed with one reaction because the starting concentrations in a two allele system is usually about 50/50 (or at least on the same order of magnitude) and the signal-to-noise ratio of the genotyping biochemistry is good.
  • the genotyping biochemistry is not good enough to give an accurate quantitation of the under-represented allele.
  • the mutant/normal ratio can be quite low (1 in thousands) and therefore undetectable by conventional biochemical methods.
  • PCR in a microfluidic device demonstrated in this application is the ability to quantitate single copies of nucleic acids. This allows one to count the number of infected cells, or virus particles in a sample of interest, at biologically relevant concentrations of cells or virus particles.
  • quantitative single molecule PCR from a starting volume on the order of 10 microliters (an initial pre-concentration step taking the sample from ⁇ 1 ml to about 10 ul can be performed by standard techniques, e.g. immunoprecipitation or hybridization capture into magnetic beads).
  • the ⁇ 10 ul of concentrated solution containing e.g., >50 virus particles can be completely processed (or a substantial fraction of the volume processed) on a microfluidic chip in the following manner.
  • the sample is mixed on-chip with the reagents necessary for PCR (at, e.g., a 1:1 ratio), e.g. primers, probes, dNTPs, etc.
  • the mixture is pressure loaded into a microfluidic network that has a holdup volume on the order of 10 ul (see, FIG. 8 ), and the flow is stopped. As shown, the schematic device of FIG.
  • thermocycling comprises PCR reagent well 801 , sample well 802 vacuum/waste well 803 , imaging area 804 (a detection region) and microfluidic network 805 .
  • the contents of the network are then thermocycled by applying heat externally to the chip, or, optionally, via resistive or Joule heating.
  • the chip is imaged to locate all of the “clouds” of fluorescence (see, FIG. 9 ), each corresponding, typically, to a single copy of DNA from a virus particle.
  • FIG. 9 is a schematic of the fluidic network of FIG. 8 after thermocycling.
  • Spots 806 represent the fluorescence “clouds” from single amplicons (e.g., in one example, virus particle amplicons). Spots 806 can be counted for quantitative PCR.
  • CCD imaging e.g., CCD imaging
  • the detector e.g., a photodiode
  • continuous flow can, alternatively, be used.
  • the active area of the chip for imaging is typically on the order of 20 ⁇ 30 mm (but can, optionally, be smaller or larger). This area is compatible with high resolution imaging ( ⁇ 1-2 um resolution) via techniques commonly used for DNA array applications. These can include CCD imaging, confocal laser scanning, and/or the like.
  • the dynamic range for quantification is typically at least 2-3 orders of magnitude, depending, in part, on the size of the chip. For a typical size of 20 ⁇ 30 mm, the dynamic range is about 2 orders of magnitude.
  • One way to estimate the dynamic range is to examine the average separation between copies, and then compute the expected diffusion distances over the course of an experiment. A rough calculation to demonstrate that these types of volumes can be processed on a microfluidic chip is presented below.
  • the total volume in these channels is 6.2 ⁇ L. If it is further assumed that in the 6.2 ⁇ L, half of the volume comes from PCR reagents and the other half comes from the original 10 ⁇ L sample, then roughly 3 ⁇ L out of 10 ⁇ L are sampled per run, which is a reasonable volume from a statistical sampling or a practical ease of use standpoint. Furthermore, if the 10 ⁇ L concentrated sample contains 100 particles from an initial starting 1 mL volume of plasma, then one can detect about 30 PCR clouds per run, if the PCR efficiency is close to 100%. These clouds would be 62 mm apart, on average, from each other along the channel, or about 1 cloud in every 2 channels.
  • a series of parallel channels e.g. 64
  • chip size and detection practicality If the 64 (2 ⁇ circumflex over ( ) ⁇ n binary split) parallel channels are packed together with 200 um landing area between, they will occupy 21 mm. So an area of 30 mm ⁇ 21 mm can be imaged (or scanned) to find the 30 PCR clouds (in stop flow mode) that should appear in the channels. This is similar to the size of a typical DNA assay chip, meaning that available chip scanners can be used for the detection.
  • One aspect of the invention provides methods to ensure stopped flow conditions on a chip.
  • methods to ensure stopped flow conditions on a chip There are a number of methods that can be employed. For example, one can use temperature sensitive materials (e.g. polymers), to create the stop-flow condition.
  • a simple method to achieve stopped flow is to use standard chip-capillary or chip-tubing connections combined with a valve.
  • Single molecule amplification techniques of the invention can be used to unambiguously determine whether a nucleotide of interest in a reaction mixture is, e.g., fragmented or has a given length between probes. For example, simultaneous signals from two or more different probes hybridized to opposite ends of a nucleic acid of interest in a single copy reaction mixture can provide a high level of confidence that the nucleic acid is not fragmented. This contrasts with conventional methods, such as dot blot hybridizations to multiple target nucleic acid copies, wherein coincidental detection of probes to each end of the nucleic acid of interest can indicate the presence of full length nucleic acid of interest and/or the presence of a one or more pair of unassociated fragmented nucleic acids of interest.
  • the methods and systems for determining length can detect the presence of individual full length or fragmented nucleic acids of interest, as well as provide counts indicating proportions or concentrations of fragmented nucleic acids of interest, even in complex mixtures containing large amounts of additional nucleic acids. Such counts can be subjected to statistical analyses for reporting of validated assay results and correlation to associated disease states.
  • Samples containing nucleic acids of interest can be diluted and/or contained in volumes small enough to provide reaction volumes that include on the average about 1 individual nucleic acid of interest (single copy) in a reaction mixture. If the single copy nucleic acid of interest is hybridized with two probes complimentary to separated sequences on the nucleic acid of interest single copy, detection of the two hybridized probes at the same time in the reaction mixture indicates the nucleic acid is not fragmented between the separate probe target sequences. On the other hand, if the nucleic acid is fragmented between the target sequences, the single copy reaction mixture will contain only one of the fragments and only one of the hybridized probes would be detected.
  • nucleic acids of interest include at least a length (a given length) including the sequences complimentary to particular probes and the nucleic acid between the probes.
  • Methods of determining whether a nucleic acid is of a given length can generally include, e.g., adjusting the concentration of a sample containing the nucleic acid; contacting the nucleic acid with probes, primer pairs, and a polymerase in a reaction mixture; amplifying the nucleic acid to produce specifically detectable amplicons; hybridizing probes to the nucleic acid and/or amplicons in the reaction mixture; flowing reaction mixture constituents into a detection region; detecting signals from hybridized probes; and, interpreting the signals to provide fragment and given length nucleic acid quantities or proportions.
  • Such quantities or proportions can be correlated to disease states that may be associated with the sample source.
  • the concentration of nucleic acids of interest can be adjusted to provide useful numbers of low copy, single copy, and/or zero copy reaction mixtures in the methods of determining whether the nucleic acids are of a given length.
  • Samples can be concentrated, e.g., by ultrafiltration, affinity capture, or immunoprecipitation so that a suitable copy number is obtained in reaction mixtures and detectors of the invention.
  • Samples can be diluted, e.g., by serial dilution in microwell plates or admixing with buffers or reagents in a fluidic system dilution channel, to obtain a desirable concentration of nucleic acids. In many microfluidic embodiments, a concentration of about one nucleic acid of interest molecule per nanoliter is desirable for reaction or detection.
  • the nucleic acid of interest can be adjusted to a range from about 100 molecules per nL to about 0.01 molecules per nL in the reaction mixtures, or from about 10 molecules per nL to about 0.1 molecules per nL, or from about 3 molecules per nL to about 1 molecule per nL.
  • nucleic acids of interest are amplified in a reaction mixture containing a polymerizing enzyme that can make copies (amplicons) of sequences (and/or compliments) from nucleic acids of interest.
  • nucleic acid(s) of interest are amplified by contacting with two or more primer pairs and a polymerase enzyme in a polymerase chain reaction (PCR).
  • the PCR amplification reaction takes place in an amplification microchannel or microchamber of a microfluidic device.
  • This can provide high throughput, low sample use, and isolation of single copy reaction mixtures.
  • reaction mixture constituents including a temperature stable DNA polymerase and one or more primer pairs can flow into a temperature controlled amplification microchannel with the nucleic acid of interest.
  • Amplicons can be extended by polymerization from primer pairs hybridized to the nucleic acid of interest at specific locations, thus the primers define the amplicons.
  • nucleic acid melting, annealing to primers, and primer extension by the polymerase can increase by many orders of magnitude the amount of nucleic acid having the sequence (and second strand compliment) defined by the primer pairs.
  • two or more primer pairs are typically provided in the reaction mixture to amplify two or more regions of the nucleic acid of interest associated with the intended probes. Regions of the amplified nucleic acid can include sequences complimentary to the probes and spaced a given (known or unknown) sequence length apart.
  • the probes themselves can be members of the primer pairs defining the amplicons.
  • the nucleic acid of interest (and/or any amplicons) can be contacted with two or more different probes to determine whether the nucleic acid of interest includes the given length between sequences complimentary to the probes.
  • two probes can hybridize to complimentary sequences spaced by the given length along the strand.
  • the probes can be complimentary to and hybridize to sequences on either strand or both strands separated at a distance of the given sequence length.
  • the amplicons defined by two or more primer pairs do not overlap and are separated by sequences of about the given length, as shown in FIG. 14 . If the nucleic acid of interest 140 is not fragmented between the first amplicon sequences defined by first primer pair 141 and the second amplicon sequences defined by second primer pair 142 , low or single copy amplification reactions will include both first 143 and second 144 amplicons. Hybridization of such a reaction mixture with a first and second probe, specific to the first and second amplicon sequences, respectively, will provide coincident probe signals 145 .
  • single copy amplification reactions will include either first 143 or second 144 amplicons, but not both. Hybridization of these single copy reaction mixtures with the first and second probes, specific to the first and second amplicon sequences, respectively, either first probe signals 146 , or second probe signals 147 will be detected, but not both.
  • Primer pairs in such amplifications typically define amplicon sequences of about 100 base pairs, with the amplicons ranging from more than about 5000 base pairs to about 20 base pairs, or from about 50 base pairs to about 1000 base pairs.
  • first strand primer 150 is shared between second strand primers 151 and 152 for an amplification that overlaps at the sequence of the shorter amplicon. If the nucleic acid of interest 153 is not fragmented between the shorter amplicon 154 sequence and the longer second strand primer 152 sequence, the amplification reaction will provide both shorter 154 and longer 155 amplicons. In such a case, hybridization to a probe specific to the shorter amplicon and a probe specific to the longer amplicon will yield short probe signals 156 coincident with long probe signals 157 .
  • the amplification reaction will provide only shorter 154 amplicon and detection of a single copy reaction mixture will yield only short probe signals 156 .
  • the short amplicon can act as a control sequence confirming the effectiveness of the amplification and hybridization reactions.
  • the short amplicon can act as an internal reference to indicate the total number of total whole and fragmented nucleic acid of interest present in the sample of reaction mixture.
  • the region defined in the shorter amplicon can be far less likely to be fragmented than the region defined in the longer amplicon.
  • determination of a given length can be based on detection a probe hybridizing to a sequence outside of the shorter amplicon region, i.e., the determination does not require coincidence of signals in a single copy reaction.
  • these embodiments can benefit, e.g., from the discrete counting provided by single copy detection methods to enhance the precision of counts and the confidence of statistical inferences from the data.
  • Primer pairs in such amplifications typically define shorter amplicon sequences of about 100 base pairs, as above.
  • Primer pairs defining the longer amplicon are typically separated by sequence lengths ranging from more than about 5000 base pairs to about 200 base pairs, or from about 500 base pairs to about 2000 base pairs, or about 3000 base pairs.
  • Amplification reactions can be carried out using any appropriate technique known in the art and as described in the Amplifying the Aliquots section above.
  • the amplification method can be PCR, RT-PCR, LCR, and/or any of the various RNA mediated amplification methods.
  • additional information about the length and fragmentation status of a nucleic acid of interest can be obtained by using more than two primer pairs or probes.
  • the additional probes and/or primer pairs can enhance the resolution of length determination between and/or outside of the first and second primer pairs and probes. For example, with an additional primer pair amplifying a sequence between the first and second primer pairs, detection of probed amplicons can yield additional information from low or single copy reactions. In such a case, coincident detection of only the first and additional probes can indicate the nucleic acid of interest has the given length between the probes but a break between the additional probe compliment and the second probe compliment.
  • Coincident detection of only the additional and second signals can indicate the nucleic acid of interest has the given length between the additional and second probe complimentary sequences but a break between the first probe compliment and the additional probe compliment.
  • a non-fragmented nucleic of interest in this case would have coincident signals from each of the first, additional, and second probes in detection of a single copy reaction.
  • detection of amplicons interrogated with the three appropriate probes can yield useful information from low or single copy reactions.
  • coincident detection of only the first and additional probes can indicate the nucleic acid of interest has the given length between the probes but a break exists between the first probe compliment and the second probe compliment.
  • Coincident additional and second signals can indicate the nucleic acid of interest has the given length between the additional and second probe compliments but a break exists between the first probe compliment and the second probe compliment.
  • a non-fragmented nucleic of interest in this case would have coincident signals from each of the first, additional, and second probes in a single copy reaction.
  • Such a nucleic acid of interest would have at least the given length between the additional probe and the nearest of the first or second probe, plus the given length between the first and second probe.
  • Further additional probes and/or associated primer pairs can yield additional nucleic acid length information, as can be appreciated by those skilled in the art.
  • Complimentary probes can specifically hybridize to nucleic acids of interest and/or associated amplicons to provide one or more signals from low or single copy hybridizations thereby yielding information useful in determining whether the nucleic acid has at least a given length.
  • the probes can be hybridized under conditions of stringency (e.g., buffer ionic strength and temperature) suitable to provide the required level of specificity.
  • probes are hybridized to nucleic acids under highly stringent conditions.
  • the probes are molecular beacon (MB) probes, fluorescent resonance energy transfer (FRET) probes, or TaqMan® probes, as described in the Amplification of Aliquots section above.
  • detectable markers provide qualitatively different signals unique to each of the different probes.
  • two or more different probes can have the same signal and the coincident presence of the two probes in a single copy reaction mixture can be detected as a signal, e.g., of double amplitude or area.
  • Target nucleic acids of interest and/or associated amplicons hybridized to complimentary probes can be subjected to marker signal detection procedures.
  • Amplification and/or hybridization reaction mixtures can flow into a detection region for detection of any signals present in the mixtures.
  • the detection region can be, e.g., microchambers or microchannels, a region where the amplification reaction mixture was formed or amplified, a region where the hybridization reaction took place, a cuvette region downstream from reaction regions, detection regions integral with or proximal to a microfluidic device, and/or the like.
  • the detector can be any type appropriate to the marker signal and compatible with other system hardware, as described above.
  • the probes, hybridized and/or released from the nucleic acid or amplicon can be detected by flowing into or through the detection region, the detector can be scanned across the probes, or the probes can be detected in a two or three dimensional detection region, e.g., using imaging technologies known in the art.
  • Detected signals can be interpreted to provide detection of a nucleic acid of interest and a determination of whether the nucleic acid has at least a given length.
  • coincident detection of one of more probes in a low or single copy reaction mixture can provide information about the length of a nucleic acid of interest.
  • primers and amplicons are not necessary to determining whether a nucleic acid of interest has a given length or not, but amplification schemes can be useful to enhancing the sensitivity of such determinations.
  • Detection of signals in a low or single copy reaction mixture from two or more probes that have been specifically hybridized to a nucleic acid (or associated amplicons) at sequences spaced a given distance along the nucleic acid can indicate the nucleic acid is not fragmented between the probes. Detection of a signal from only one probe can indicate the presence of a break in the nucleic acid. Accumulated data of detections from multiple low, single, and zero copy reaction mixtures can yield information useful in quantitation, proportioning, and correlating the nucleic acids present in samples.
  • Nucleic acids of interest of differing length can be quantified essentially as described throughout this specification, particularly in the Counting and Statistically Analyzing a Nucleic Acid of Interest section above. Signals detected from low, single, and/or zero copy reaction mixtures can be interpreted and counted to accumulate data useful in calculation of quantities. For example, inferences can be made about the total amount of full length or fragmented nucleic acid of interest in a sample based on counts of coincident signals, solitary signals, and no signal (zero copy) reactions. Appropriate adjustments can be made according to dilution factors, efficiency factors, internal reference values, and the like. Proportions of full length to fragmented nucleic acids of interest can be determined based on the proportions of associated signal counts.
  • the shape, volume, width, height, length, area, or ratio, of the one or more signals can be evaluated to provide quantitative information about nucleic acids of interest.
  • These peak parameters of acquired signals can be subjected to regression analyses to identify standard curve equations that most closely reflect the parameter change with changed concentrations of the nucleic acids of interest in samples.
  • an assay includes detection of different signals from two or more probes with different detectable markers, the same or different peak parameters can be input for regression analysis of the different detected signals.
  • the quantity of a nucleic acid of interest in a sample can be determined by measuring the change in a signal with increasing numbers of amplification cycles. Plotting the change in signal strength with increasing amplification cycles often results in a sigmoid curve. Certain precise points along the curve, such as points of inflection, points with certain slopes, points having a certain absolute signal strength, points having a certain fraction of maximum (plateau) signal strength, and/or the like, can be identified with high precision.
  • Useful standard curves can be prepared based on regression analyses of any of these identifiable points from assay of standard materials of known nucleic acid concentration. For example, a standard curve can be prepared representing known nucleic acid of interest concentrations versus amplification cycles required to attain the chosen identifiable point.
  • Such standard curves can be, e.g., plotted curves of standard data or mathematical representations of such curves. Concentrations of nucleic acids for unknown samples can be determined with reference to the standard curve. Different degrees of amplification can be consistently obtained, e.g., by flowing amplification reactions through an active amplification region at different rates, for different times, and/or different distances to provide a series of reaction mixtures experiencing different numbers of amplification cycles.
  • different degrees of, e.g., PCR amplification are provided by flowing the amplification reaction mixture different distances in an actively cycling heated region of a thermocycler channel.
  • amplification reactions can flow into an actively cycling amplification region for a certain distance before thermocycling is stopped.
  • the front edge of the flowing slug of reaction mixtures experiences more amplification cycles while late entering mixtures nearer the trailing edge experience fewer amplification cycles. If such a slug of reaction mixtures were to continue flowing past a detector, the detector signal output can be a reverse sigmoid curve as shown in FIG. 16A .
  • a slug of amplification reaction mixtures can flow into an inactive amplification region before starting thermocycling so that reaction mixtures near the front edge flow out of the thermocycler sooner to experience fewer amplification cycles than reaction mixtures nearer the trailing edge. Detection of this slug of reaction mixtures flowing past a detector can provide a sigmoid curve detector signal output, as shown in FIG. 16B , indicating weak amplification at the front edge of the slug and higher amplification for the trailing edge.
  • Information about the fragmentation state or integrity of nucleic acids of interest can be correlated to disease states, e.g., of the sample source organism. Correlation analyses known in the art can be carried out, e.g., comparing qualitative, quantitative, and/or proportion data on the length of certain nucleic acids obtained using methods of the invention.
  • disease states can be correlated to a quantity of unfragmented nucleic acid, or a proportion of fragmented nucleic to nucleic acid having a given length.
  • a proportional threshold or quantitative threshold can be established using statistical analyses to provide an acceptable degree of confidence in identification of samples possibly positive for the correlated disease state without unacceptable false positive results.
  • a certain proportion of fragmented to unfragmented nucleic acid in a stool or cervical swab sample can identify the sample as likely to originate from a patient having, e.g., colon or cervical cancer, respectively.
  • Methods and systems of the invention can be used in various formats to quantify nucleic acids of interest.
  • the quantitative assays can be configured to provide a desired quality of output results.
  • assay parameters such as, e.g., sensitivity, accuracy, precision, and rates of false positives or false negatives can be influenced by the design of particular assays. Repetition of assays can increase precision.
  • the quantitative assays can be improved by evaluating signal peak parameters best suited to provide valid results, e.g., with the desired range, sensitivity and/or accuracy.
  • Comparison of a sample signal output peak to a series of standard signal output peaks can indicate the concentration of a nucleic acid of interest in a sample.
  • a series of standard materials containing known mounts of the nucleic acid of interest can each be amplified through the same number of amplification cycles to produce a series of detectable signals, as shown in FIG. 17 .
  • a different detectable signal peak associated with the amplicons can be detected.
  • a sample with an unknown concentration of the nucleic acid of interest is amplified the same number of cycles as the standard materials. The resultant amplicons are detected, with the same probe and detectable marker system as for the standards, to provide a signal with certain distinctive peak parameters.
  • the signal from the sample can be evaluated to identify signal peak parameters (e.g., the shape of signal peaks, points of inflection on signal peaks, slopes of signal peaks, signal peak amplitudes, signal peak areas, signal peak widths at half height, etc.) most suitable for the analytical goal. For example, peak area might provide the most accurate quantitative comparison, while peak height might provide a more precise comparison, and peak shape might provide suitable quantitative comparisons over a broader range of concentrations. Identifying an appropriate signal peak parameter for comparison in a particular instance can be determined, e.g., using methods of assay development and validation procedures well known in the art. The precision and/or accuracy of nucleic acid quantification can be enhanced by interpolating the comparison to intermediate values between standard values, by running replicate standards, by statistical analyses of repeated sample assays, by running comparisons at two or more amplification levels, and/or the like.
  • signal peak parameters e.g., the shape of signal peaks, points of inflection on signal peaks, slopes of signal peaks, signal
  • One method of enhancing reliability of nucleic acid quantification by using multiple assay results depends on comparison of amplification signal response curves between samples and standards. For example, the amount of amplicon associated signal generated over a number of amplification cycles can be compared to the signals for standards of various known concentrations. As shown schematically in FIG. 18A , a signal from an amplified sample can start low 180 , increase logarithmically at some point 181 , and taper off to a maximum signal plateau 182 with increasing numbers of amplification cycles, thus describing a sigmoid curve. Standard materials with different known concentrations of the nucleic acid of interest can be amplified, as shown in FIG. 18B , to provide a series of standard sigmoid curves.
  • Points along the sample and standard curves can be identified, e.g., with various levels consistency. Identifiable points can be, e.g., points of inflection, points having a certain slope, points having a certain signal amplitude, points having a certain fraction of a maximum (e.g., plateau asymptotic) signal amplitude, and the like.
  • a standard curve of concentration versus cycles to an identifiable point can be prepared, as shown in FIG. 18C , so that the concentration of the nucleic acid in a sample can be determined from the number of amplification cycles it takes for the sample to reach the identifiable point. For example, a point of maximum slope 183 (maximum rate of signal increase) can be precisely identified on each of the standard curves.
  • the cycles to maximum slope can be plotted versus concentration to prepare a standard curve.
  • the nucleic acid of interest concentration of an unknown sample can be read as the concentration 184 providing the number of cycles 185 to the maximum slope from the standard curve.
  • preparing curves includes all means of expressing the relationships between relevant factors (e.g., concentration and slope, signal parameter and amplification cycles, identifiable points and concentration, etc.), such as, e.g., data plotting, regression analysis, curve fitting, determination of an equation, and/or the like whether accomplished manually or with the aid of an analog or digital computer and software.
  • nucleic acid of interest quantitation if is preferred standard and sample reaction mixtures be cycled through different numbers of amplification cycles, e.g., as described in the Methods of Determining the Fragmentation Status of Nucleic Acids section above.
  • the quantities and proportions of nucleic acids of a given length and/or fragmented nucleic acids of interest can be determined with high precision, e.g., by reference to standard curves of concentration versus amplification cycles to identifiable points generated for two or more probe signals.
  • detectable signals associated with fragmented or unfragmented nucleic acid of interest can be quantitated separately, e.g., according to the methods described in the paragraph above, to evaluate the integrity of the nucleic acid of interest in a sample.
  • standard curves of concentration versus amplification cycles to an identifiable point can be separately plotted for signals associated with two or more amplicons of the nucleic acid of interest.
  • a signal is associated with an amplicon, e.g., if it originates from a nucleic acid probe that has hybridized to a sequence of the amplicon and has detectable marker providing the signal.) Cycles to the identifiable point for the sample can be compared to the standard curves for each amplicon to separately determine fragmented and given length nucleic acid of interest concentrations.
  • the signals are detected from homogenous reaction mixtures. In another preferred embodiment, the signals are detected from low or single copy reaction mixtures to provide highly resolved data on the integrity of the nucleic acid of interest.
  • Systems of the invention can provide efficient processing and well adapted hardware for high sensitivity differentiation of the lengths of nucleic acids of interest in samples.
  • Systems for differentiating lengths of nucleic acids can be essentially as described herein for single molecule amplifications, but, e.g., incorporating additional elements for amplification, detection, interpretation, and/or correlation of multiple probes.
  • the core system for differentiating nucleic acid length includes, e.g., a microfluidic device capable of containing low and single copy reaction mixtures in microchannels or microchambers, detectors capable of distinguishing one or more signals from a homogenous reaction mixture, and a software system configured to interpret single or coincidently detected signals to lengths of individual nucleic acids from a sample.
  • Additional subsystems can include sample storage modules, retrieval modules, dilution modules, and computers, as discussed above for single molecule amplification systems in general.
  • An exemplary system for differentiating lengths of nucleic acids can function as follows. Samples 190 in wells of microtiter plates 191 can be held in storage module 192 until retrieval by retrieval module 193 and delivery for sampling by microfluidic device 194 . Samples are aspirated up a capillary sipper tube 195 to microchannels 196 or microchambers of the microfluidic device where integral dilution module 197 (e.g., a dilution channel) appropriately dilutes the sample with reagents 198 and buffers 199 to constitute an amplification reaction mixture.
  • integral dilution module 197 e.g., a dilution channel
  • the reaction mixtures can be separately aliquotted into each channel in multiple amplification region 200 where low, single, and/or zero copy reaction mixtures can be exposed to amplification conditions (e.g., thermocycling).
  • Amplified reaction mixture aliquots can flow into detection region 201 where signals from detectable markers on one or more different probes can be detected by detector 202 .
  • the detector can communicate with computer 203 to transmit detector signals for interpretation and/or correlation by system software.
  • Samples for nucleic acid differentiation in the systems can be any that contain natural or unnatural nucleic acids.
  • the samples can include a nucleic acid with single nucleotide polymorphism (SNP), a cancer associated nucleic acid, a nucleic acid from an infective agent, whole blood, serum, plasma, stool, urine, a vaginal secretion, ejaculatory fluid, synovial fluid, a biopsy, cerebrospinal fluid, amniotic fluid, or a forensic nucleic acid.
  • Samples can be stored in a storage module, as described above, under environmental conditions of temperature, light, and humidity conducive to long storage life of the samples. Libraries of samples can be stored, e.g., as matrices of various liquid aliquots in multiwell plates or dry spots on slides. The samples can be located at positions in the storage module trackable with an inventory system and accessible by a sample retrieval system.
  • Retrieval modules can be employed in the systems of differentiating nucleic acid length, e.g., to enhance the high throughput capabilities of the system.
  • the retrieval modules can remove designated samples from a storage module and deliver them to dilution modules or microfluidic devices for analysis.
  • the retrieval modules can have, e.g., robotic arms with 6 degrees of freedom of movement, x-y plotted tray graspers, belt driven conveyors, and/or the like. Bar code readers or radio frequency identification systems can be incorporated into the retrieval modules to identify and track tagged samples.
  • Reaction mixtures for determination of nucleic acid length can be prepared to include a nucleic acid of interest, two or more polymerase primer pairs, nucleotide triphosphates, buffers, and/or two or more detectable probes.
  • the reaction mixture constituents can be combined in any order at any suitable time or place.
  • the reaction mixtures can be constituted outside the microfluidic device, e.g., in microwell plates using manual or automated pipettor systems.
  • the reaction mixture can be diluted outside of the microfluidic device, e.g., by manual serial dilutions or by using an automated dilution module.
  • the dilutions can provide a concentration of the nucleic acid of interest resulting in desired amounts of low, single, and zero copy reaction mixtures in amplification microchannels, hybridization reactions, and/or detection regions of the system.
  • the reaction mixture can include, e.g., two or more primer pairs defining amplicons and bracketing sequences complimentary to probes a given distance apart on the nucleic acid of interest.
  • Preferred primer pairs define amplicons ranging in size from about 50 base pairs to about 3000 base pairs, or about 100 base pairs.
  • the primer pairs have roughly the same melting temperatures and define amplicons of about the same length.
  • the amplicons include a shorter amplicon that overlaps a longer amplicon in a region having sequences complimentary to one or more probes.
  • the systems can include, e.g., solid supports to provide affinity concentrations of sample constituents for analysis, or concentration of hybridization reaction mixture constituents.
  • Solid supports can have affinity elements, such as oligonucleotides complimentary to reaction constituents (e.g., nucleic acids of interest, primers, probes, etc.), capable of specifically capturing the constituents.
  • the solid supports can be useful in adjusting the concentration of nucleic acids of interest for input into the systems.
  • the solid supports can immobilize constituents during amplification or hybridization steps. Probes that have been hybridized to the nucleic acids of interest and/or their associated amplicons can be captured in a detection region for detection. Although certain reaction mixture constituents can be immobilized on the solid supports while certain other constituents can flow past to be removed, the mixture can still be considered homogenous if different probes are present in the same mixture at the time of detection.
  • One or more probes can be present in the amplification reaction mixture and/or in the hybridization mixture to hybridize with the nucleic acid of interest or associated amplicons.
  • signals from a single probe can confirm the presence of unfragmented nucleic acid.
  • the system includes two or more probes with specificity for nucleic acid sequences separated a given distance along the nucleic acid of interest.
  • the signal can be enhanced by providing alternate probes specific for sequences in the second strand of a double stranded nucleic acid at about each end of the given length; typically, the alternate probes hybridize near the first strand probes but are not complimentary to the first strand probes in order to avoid hybridization of primers to each other.
  • Preferred probes have significant signal changes associated with binding to target nucleic acid of interest. This can provide, e.g., a positive signal against a low background from a homogenous mixture (i.e., without the need to separate unhybridized probe from the reaction mixture).
  • probes can have any of the variety of detectable markers described above, preferred probes for differentiating the length of a nucleic acid are molecular beacon (MB), TaqMan®, and fluorescent resonance energy transfer (FRET) probes.
  • MB molecular beacon
  • TaqMan® TaqMan®
  • FRET fluorescent resonance energy transfer
  • a reaction mixture can include independently detectable probe pairs for more than one nucleic acid of interest.
  • a reaction mixture can be constituted to include, e.g., a pair of probes complimentary to sequences at the ends of a first given length of a first nucleic acid of interest, and another pair of probes complimentary to sequences at the ends of a second given length of another nucleic acid of interest.
  • the four probes can each have different detectable markers so they can be individually detected in reaction mixtures.
  • Microfluidic devices in systems for differentiating nucleic acid lengths can include chips with reagent wells, gas, liquid and electrical contact ports, sample sippers, microchannels and microchambers, amplification microchannels, and detection regions, as described above in the Example Systems section.
  • the microfluidic device includes such features as a sipper, multiple amplification channels (such as shown in FIG. 1 ), and detection region configured for laser excitation and fluorescent detection at multiple frequencies.
  • Detectors in the systems can be any configured with a capability to detect two or more signals from a homogenous mixture of reaction mixture constituents.
  • the detector can be appropriate for the type of detectable marker signal provided by the hybridized probes.
  • the detector can include, e.g., a fluorometer, a charge coupled device, a laser, an enzyme, or an enzyme substrate, a photo multiplier tube, a spectrophotometer, scanning detector, microscope, a galvo-scanner, etc.
  • the detectors can monitor a detection region into which amplification reaction mixtures, aliquots, hybridization reaction mixtures, or reaction mixture constituents have flowed.
  • hybridization actuated fluorescent probes are preferred in the methods, preferred detectors in the systems are fluorometers.
  • the detector comprises a laser light excitation source directed to the detection region through optic fibers, and a photodiode array capable of simultaneously detecting two or more emission wavelengths.
  • a software system can be an element of the system to interpret one or more signals and/or to correlate the signals to a disease state.
  • the software system can include, e.g., algorithms to count signals, calculate concentrations, calculate proportions, prepare standard curves, and/or evaluate correlations, e.g., to disease states.
  • the software can interpret, e.g., coincidence of two or more signals to indicate the presence of a nucleic acid of a given length in the detection region.
  • the software can interpret, e.g., a single signal to indicate the presence of a fragmented nucleic acid in the detection region.
  • the software can calculate a quantity of a nucleic acid of interest by factoring in information, such as, e.g., the number of signal counts, reaction volumes, dilution or concentration factors, efficiency factors, known input values and constants, signal peak shape, and/or the like.
  • the software can interpret peak parameters, such as a volume, width, height, length, area, and/or a ratio, of the signals detected by the detector to indicate a number of copies of the nucleic acid of interest in the sample, a number of the nucleic acids of interest having a given length, or a proportion of nucleic acids of interest having different lengths.
  • Software systems can correlate results of one or more signal detection to disease states, e.g., by comparing a validated quantity or proportion threshold to assay results for a sample.
  • Computers can be an important element of systems for differentiating lengths of nucleic acids.
  • Computers can coordinate control activities in the system, such as, e.g., sample identification, sample retrieval, sample sipping, control of microchip pressures and voltages, receipt of detector signals, and software interpretation of signals.
  • the computer is in communication with the signal detector to receive, store, and evaluate signals from sample or standard assays.
  • Computers in the systems can be as described in the Computers section above.
  • systems in the present invention can include, e.g., a digital computer with data sets and instruction sets entered into a software system to practice the methods of determining lengths described herein.
  • the computer can be, e.g., a PC (Intel x86 or Pentium chip—compatible with DOS®, OS2®, WINDOWS® operating systems) a MACINTOSH®, Power PC, or SUN® work station (compatible with a LINUX or UNIX operating system) or other commercially available computer which is known to one of skill.
  • the computer can be, e.g., a simple logic device, such as an integrated circuit or processor with memory, integrated into the system. Software for interpretation of detector signals is available, or can easily be constructed by one of skill using a standard programming language such as Visualbasic, Fortran, Basic, Java, or the like.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Engineering & Computer Science (AREA)
  • Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Immunology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • Genetics & Genomics (AREA)
  • General Engineering & Computer Science (AREA)
  • Biophysics (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biotechnology (AREA)
  • Microbiology (AREA)
  • Dispersion Chemistry (AREA)
  • Hematology (AREA)
  • Clinical Laboratory Science (AREA)
  • Pathology (AREA)
  • General Physics & Mathematics (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Optics & Photonics (AREA)
  • Fluid Mechanics (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Investigating Or Analysing Materials By The Use Of Chemical Reactions (AREA)
US10/845,996 2002-12-20 2004-05-14 Single molecule amplification and detection of DNA length Abandoned US20050042639A1 (en)

Priority Applications (8)

Application Number Priority Date Filing Date Title
US10/845,996 US20050042639A1 (en) 2002-12-20 2004-05-14 Single molecule amplification and detection of DNA length
CA002566698A CA2566698A1 (en) 2004-05-14 2005-05-16 Single molecule amplification and detection of dna length
PCT/US2005/017065 WO2005113148A1 (en) 2004-05-14 2005-05-16 Single molecule amplification and detection of dna length
EP05749873A EP1755785A1 (en) 2004-05-14 2005-05-16 Single molecule amplification and detection of dna length
AU2005245448A AU2005245448A1 (en) 2004-05-14 2005-05-16 Single molecule amplification and detection of DNA length
JP2007513461A JP4740237B2 (ja) 2004-05-14 2005-05-16 単分子増幅及びdna長の検出
US11/867,626 US7645581B2 (en) 2002-12-20 2007-10-04 Determining nucleic acid fragmentation status by coincident detection of two labeled probes
US12/684,601 US8275554B2 (en) 2002-12-20 2010-01-08 System for differentiating the lengths of nucleic acids of interest in a sample

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US43609802P 2002-12-20 2002-12-20
US46238403P 2003-04-11 2003-04-11
US10/741,162 US20040224325A1 (en) 2002-12-20 2003-12-19 Single molecule amplification and detection of DNA
US10/845,996 US20050042639A1 (en) 2002-12-20 2004-05-14 Single molecule amplification and detection of DNA length

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US10/741,162 Continuation-In-Part US20040224325A1 (en) 2002-12-20 2003-12-19 Single molecule amplification and detection of DNA

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US11/867,626 Division US7645581B2 (en) 2002-12-20 2007-10-04 Determining nucleic acid fragmentation status by coincident detection of two labeled probes
US12/684,601 Division US8275554B2 (en) 2002-12-20 2010-01-08 System for differentiating the lengths of nucleic acids of interest in a sample

Publications (1)

Publication Number Publication Date
US20050042639A1 true US20050042639A1 (en) 2005-02-24

Family

ID=34969685

Family Applications (2)

Application Number Title Priority Date Filing Date
US10/845,996 Abandoned US20050042639A1 (en) 2002-12-20 2004-05-14 Single molecule amplification and detection of DNA length
US11/867,626 Expired - Lifetime US7645581B2 (en) 2002-12-20 2007-10-04 Determining nucleic acid fragmentation status by coincident detection of two labeled probes

Family Applications After (1)

Application Number Title Priority Date Filing Date
US11/867,626 Expired - Lifetime US7645581B2 (en) 2002-12-20 2007-10-04 Determining nucleic acid fragmentation status by coincident detection of two labeled probes

Country Status (6)

Country Link
US (2) US20050042639A1 (ja)
EP (1) EP1755785A1 (ja)
JP (1) JP4740237B2 (ja)
AU (1) AU2005245448A1 (ja)
CA (1) CA2566698A1 (ja)
WO (1) WO2005113148A1 (ja)

Cited By (124)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040180346A1 (en) * 2003-03-14 2004-09-16 The Regents Of The University Of California. Chemical amplification based on fluid partitioning
US20070026421A1 (en) * 2000-11-16 2007-02-01 Caliper Life Sciences, Inc. Method and apparatus for generating thermal melting curves in a microfluidic device
US20070154895A1 (en) * 2005-12-30 2007-07-05 Caliper Life Sciences, Inc. Multi-assay microfluidic chips
US20070161868A1 (en) * 2006-01-10 2007-07-12 Biosignia, Inc. Method and system for determining whether additional laboratory tests will yield values beyond a threshold level
US20070184547A1 (en) * 2005-10-11 2007-08-09 Kalyan Handique Polynucleotide sample preparation device
US20070207272A1 (en) * 2006-03-03 2007-09-06 Puri Ishwar K Method and apparatus for magnetic mixing in micron size droplets
US20070292941A1 (en) * 2006-03-24 2007-12-20 Handylab, Inc. Integrated system for processing microfluidic samples, and method of using the same
WO2007147063A2 (en) * 2006-06-16 2007-12-21 Sequenom, Inc. Methods and compositions for the amplification, detection and quantification of nucleic acid from a sample
FR2902441A1 (fr) * 2006-06-14 2007-12-21 Siemens Ag Procede de determination de la concentration d'acides nucleiques
US20080003585A1 (en) * 2006-06-29 2008-01-03 Bio-Rad Laboratories, Inc., A Corporation Of The State Of Delaware Purification and amplification of nucleic acids in a microfluidic device
US20080003594A1 (en) * 2006-06-30 2008-01-03 Hasson Kenton C Systems and methods for monitoring the amplification and dissociation behavior of DNA molecules
US20080130971A1 (en) * 2006-11-30 2008-06-05 Canon U.S. Life Sciences, Inc. Systems and methods for monitoring the amplification and dissociation behavior of dna molecules
US20090053719A1 (en) * 2007-08-03 2009-02-26 The Chinese University Of Hong Kong Analysis of nucleic acids by digital pcr
US20090060795A1 (en) * 2007-08-29 2009-03-05 Canon U.S. Life Sciences, Inc. Microfluidic devices with integrated resistive heater electrodes including systems and methods for controlling and measuring the temperatures of such heater electrodes
US20090112484A1 (en) * 2007-10-25 2009-04-30 Canon U.S. Life Sciences, Inc. High-resolution melting analysis
US20090111149A1 (en) * 2007-10-25 2009-04-30 Canon U.S. Life Sciences, Inc. Method of reducing cross-contamination in continuous amplification reactions in a channel
US20090130746A1 (en) * 2007-10-25 2009-05-21 Canon U.S. Life Sciences, Inc. Microchannel surface coating
US20090134069A1 (en) * 2007-07-13 2009-05-28 Handylab, Inc. Integrated Heater and Magnetic Separator
US20090261086A1 (en) * 2008-03-21 2009-10-22 Neil Reginald Beer Laser Heating of Aqueous Samples on a Micro-Optical-Electro-Mechanical System
US20090317874A1 (en) * 2008-06-23 2009-12-24 Canon U.S. Life Sciences, Inc. Systems and methods for amplifying nucleic acids
US20090318306A1 (en) * 2008-06-23 2009-12-24 Canon U.S. Life Sciences, Inc. System and method for temperature referencing for melt curve data collection
US20100021910A1 (en) * 2008-07-18 2010-01-28 Canon U.S. Life Sciences, Inc. Methods and Systems for Microfluidic DNA Sample Preparation
US20100173393A1 (en) * 2006-11-14 2010-07-08 Handy Lab, Inc. Microfluidic valve and method of making same
US20100184020A1 (en) * 2007-12-27 2010-07-22 Lawrence Livermore National Security, Llc. Chip-Based Sequencing Nucleic Acids
US20100197008A1 (en) * 2003-07-31 2010-08-05 Handylab, Inc. Processing particle-containing samples
US20100233687A1 (en) * 2009-03-10 2010-09-16 Canon U.S. Life Sciences, Inc. Method and system for temperature correction in thermal melt analysis
US20100285571A1 (en) * 2009-05-08 2010-11-11 Canon U.S. Life Sciences, Inc. Systems and methods for auto-calibration of resistive temperature sensors
US20110010103A1 (en) * 2009-04-13 2011-01-13 Canon U.S. Life Sciences, Inc. Rapid method of pattern recognition, machine learning, and automated genotype classification through correlation analysis of dynamic signals
US20110014605A1 (en) * 2009-07-17 2011-01-20 Canon U.S. Life Sciences, Inc. Methods and systems for dna isolation on a microfluidic device
WO2011008217A1 (en) 2009-07-17 2011-01-20 Canon U.S. Life Sciences, Inc. Methods and systems for dna isolation on a microfluidic device
US20110048547A1 (en) * 2009-06-29 2011-03-03 Canon U.S. Life Sciences, Inc. Microfluidic systems and methods for thermal control
US20110053798A1 (en) * 2009-09-02 2011-03-03 Quantalife, Inc. System for mixing fluids by coalescence of multiple emulsions
US20110056926A1 (en) * 2007-08-29 2011-03-10 Canon U.S. Life Sciences, Inc. Microfluidic devices with integrated resistive heater electrodes including systems and methods for controlling and measuring the temperatures of such heater electrodes
US20110064628A1 (en) * 2009-05-14 2011-03-17 Canon U.S. Life Sciences, Inc. Microfluidic chip features for optical and thermal isolation
US20110077897A1 (en) * 2007-08-29 2011-03-31 Canon U.S. Life Sciences, Inc. Microfluidic devices with integrated resistive heater electrodes including systems and methods for controlling and measuring the temperatures of such heater electrodes
US20110086780A1 (en) * 2008-09-23 2011-04-14 Quantalife, Inc. System for forming an array of emulsions
US20110190146A1 (en) * 2008-04-28 2011-08-04 President And Fellows Of Harvard College Microfluidic device for storage and well-defined arrangement of droplets
US20110212516A1 (en) * 2008-09-23 2011-09-01 Ness Kevin D Flow-based thermocycling system with thermoelectric cooler
US20110217712A1 (en) * 2010-03-02 2011-09-08 Quantalife, Inc. Emulsion chemistry for encapsulated droplets
US20110215798A1 (en) * 2007-12-27 2011-09-08 Lawrence Livermore National Security, Llc. Signal Enhancement Using a Switchable Magnetic Trap
US20110264380A1 (en) * 2008-07-21 2011-10-27 Cottet Herve Determination of the hydrodynamic radii and/or content of constituents of a mixture by analysis of the taylor dispersion of the mixture in a capillary tube
US20120028342A1 (en) * 2009-03-24 2012-02-02 Ismagilov Rustem F Slip chip device and methods
WO2012011660A3 (ko) * 2010-07-23 2012-05-03 (주)바이오니아 시료 내장 마이크로 챔버 플레이트 및 분석용 마이크로 챔버 프레이트의 제조 방법, 분석용 마이크로 챔버 플레이트 및 시료 내장 마이크로 챔버 플레이트 제조 장치 셋
US8483972B2 (en) 2009-04-13 2013-07-09 Canon U.S. Life Sciences, Inc. System and method for genotype analysis and enhanced monte carlo simulation method to estimate misclassification rate in automated genotyping
US20130217583A1 (en) * 2006-01-11 2013-08-22 Darren Link Microfluidic devices and methods of use in the formation and control of nanoreactors
US8652780B2 (en) 2007-03-26 2014-02-18 Sequenom, Inc. Restriction endonuclease enhanced polymorphic sequence detection
US8663920B2 (en) 2011-07-29 2014-03-04 Bio-Rad Laboratories, Inc. Library characterization by digital assay
US8685341B2 (en) 2001-09-12 2014-04-01 Handylab, Inc. Microfluidic devices having a reduced number of input and output connections
US8703069B2 (en) 2001-03-28 2014-04-22 Handylab, Inc. Moving microdroplets in a microfluidic device
US8709762B2 (en) 2010-03-02 2014-04-29 Bio-Rad Laboratories, Inc. System for hot-start amplification via a multiple emulsion
US8710211B2 (en) 2007-07-13 2014-04-29 Handylab, Inc. Polynucleotide capture materials, and methods of using same
US8722336B2 (en) 2008-03-26 2014-05-13 Sequenom, Inc. Restriction endonuclease enhanced polymorphic sequence detection
US8730479B2 (en) 2010-03-25 2014-05-20 Bio-Rad Laboratories, Inc. Detection system for droplet-based assays
US8734733B2 (en) 2001-02-14 2014-05-27 Handylab, Inc. Heat-reduction methods and systems related to microfluidic devices
US8768517B2 (en) 2001-03-28 2014-07-01 Handylab, Inc. Methods and systems for control of microfluidic devices
US20140248623A1 (en) * 2011-04-01 2014-09-04 Life Technologies Corporation System and Method for Determining Copies-per-Unit-Volume Using PCR and Flow Control of Droplets
US8852862B2 (en) 2004-05-03 2014-10-07 Handylab, Inc. Method for processing polynucleotide-containing samples
US8883490B2 (en) 2006-03-24 2014-11-11 Handylab, Inc. Fluorescence detector for microfluidic diagnostic system
US8895311B1 (en) 2001-03-28 2014-11-25 Handylab, Inc. Methods and systems for control of general purpose microfluidic devices
US8894947B2 (en) 2001-03-28 2014-11-25 Handylab, Inc. Systems and methods for thermal actuation of microfluidic devices
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
US8962252B2 (en) 2010-08-31 2015-02-24 Canon U.S. Life Sciences, Inc. Optical system for high resolution thermal melt detection
US9061279B2 (en) 2010-08-31 2015-06-23 Canon U.S. Life Sciences, Inc. Composition and method for in-system priming microfluidic devices
US9080207B2 (en) 2006-03-24 2015-07-14 Handylab, Inc. Microfluidic system for amplifying and detecting polynucleotides in parallel
US9089844B2 (en) 2010-11-01 2015-07-28 Bio-Rad Laboratories, Inc. System for forming emulsions
RU2562161C2 (ru) * 2009-09-02 2015-09-10 Эккуджен Пти Лтд Улучшенный способ количественного определения нуклеиновой кислоты
US9132394B2 (en) 2008-09-23 2015-09-15 Bio-Rad Laboratories, Inc. System for detection of spaced droplets
US9168529B2 (en) 2010-08-31 2015-10-27 Canon U.S. Life Sciences, Inc. Air cooling systems and methods for microfluidic devices
USD742027S1 (en) 2011-09-30 2015-10-27 Becton, Dickinson And Company Single piece reagent holder
US9186677B2 (en) 2007-07-13 2015-11-17 Handylab, Inc. Integrated apparatus for performing nucleic acid extraction and diagnostic testing on multiple biological samples
US9222954B2 (en) 2011-09-30 2015-12-29 Becton, Dickinson And Company Unitized reagent strip
US9222128B2 (en) 2011-03-18 2015-12-29 Bio-Rad Laboratories, Inc. Multiplexed digital assays with combinatorial use of signals
US9238223B2 (en) 2007-07-13 2016-01-19 Handylab, Inc. Microfluidic cartridge
US9259734B2 (en) 2007-07-13 2016-02-16 Handylab, Inc. Integrated apparatus for performing nucleic acid extraction and diagnostic testing on multiple biological samples
US9347586B2 (en) 2007-07-13 2016-05-24 Handylab, Inc. Automated pipetting apparatus having a combined liquid pump and pipette head system
US9347059B2 (en) 2011-04-25 2016-05-24 Bio-Rad Laboratories, Inc. Methods and compositions for nucleic acid analysis
US9366632B2 (en) 2010-02-12 2016-06-14 Raindance Technologies, Inc. Digital analyte analysis
US9393560B2 (en) 2010-03-25 2016-07-19 Bio-Rad Laboratories, Inc. Droplet transport system for detection
US9399215B2 (en) 2012-04-13 2016-07-26 Bio-Rad Laboratories, Inc. Sample holder with a well having a wicking promoter
US9404152B2 (en) 2009-01-26 2016-08-02 Canon U.S. Life Sciences, Inc. Microfluidic flow monitoring
US9417190B2 (en) 2008-09-23 2016-08-16 Bio-Rad Laboratories, Inc. Calibrations and controls for droplet-based assays
US9447458B2 (en) 2011-11-16 2016-09-20 Canon U.S. Life Sciences, Inc. Detection of neighboring variants
US9492797B2 (en) 2008-09-23 2016-11-15 Bio-Rad Laboratories, Inc. System for detection of spaced droplets
US9500664B2 (en) 2010-03-25 2016-11-22 Bio-Rad Laboratories, Inc. Droplet generation for droplet-based assays
US9630158B2 (en) 2009-04-10 2017-04-25 Canon U.S. Life Sciences, Inc. Method of delivering PCR solution to microfluidic PCR chamber
USD787087S1 (en) 2008-07-14 2017-05-16 Handylab, Inc. Housing
US9765389B2 (en) 2011-04-15 2017-09-19 Becton, Dickinson And Company Scanning real-time microfluidic thermocycler and methods for synchronized thermocycling and scanning optical detection
US9764322B2 (en) 2008-09-23 2017-09-19 Bio-Rad Laboratories, Inc. System for generating droplets with pressure monitoring
US10066977B2 (en) 2009-01-26 2018-09-04 Canon U.S. Life Sciences, Inc. Microfluidic flow monitoring
US10123380B2 (en) 2008-08-08 2018-11-06 Lawrence Livermore National Security, Llc Instantaneous in-line heating of samples on a monolithic microwave integrated circuit microfluidic device
US10179910B2 (en) 2007-07-13 2019-01-15 Handylab, Inc. Rack for sample tubes and reagent holders
US10196700B2 (en) 2009-03-24 2019-02-05 University Of Chicago Multivolume devices, kits and related methods for quantification and detection of nucleic acids and other analytes
US10351905B2 (en) 2010-02-12 2019-07-16 Bio-Rad Laboratories, Inc. Digital analyte analysis
US10370705B2 (en) 2009-03-24 2019-08-06 University Of Chicago Analysis devices, kits, and related methods for digital quantification of nucleic acids and other analytes
US10512910B2 (en) 2008-09-23 2019-12-24 Bio-Rad Laboratories, Inc. Droplet-based analysis method
US10639597B2 (en) 2006-05-11 2020-05-05 Bio-Rad Laboratories, Inc. Microfluidic devices
US10647981B1 (en) 2015-09-08 2020-05-12 Bio-Rad Laboratories, Inc. Nucleic acid library generation methods and compositions
US10822644B2 (en) 2012-02-03 2020-11-03 Becton, Dickinson And Company External files for distribution of molecular diagnostic tests and determination of compatibility between tests
CN112236512A (zh) * 2018-11-13 2021-01-15 北卡罗来纳-查佩尔山大学 具有微流体装置的分析系统、微流体装置和相关方法
US10900066B2 (en) 2006-03-24 2021-01-26 Handylab, Inc. Microfluidic system for amplifying and detecting polynucleotides in parallel
US10927407B2 (en) 2006-05-11 2021-02-23 Bio-Rad Laboratories, Inc. Systems and methods for handling microfluidic droplets
US10946384B2 (en) 2013-09-11 2021-03-16 Osaka University Thermal convection generating chip, thermal convection generating device, and thermal convection generating method
US10957421B2 (en) 2014-12-03 2021-03-23 Syracuse University System and method for inter-species DNA mixture interpretation
US10960397B2 (en) 2007-04-19 2021-03-30 President And Fellows Of Harvard College Manipulation of fluids, fluid components and reactions in microfluidic systems
US11077415B2 (en) 2011-02-11 2021-08-03 Bio-Rad Laboratories, Inc. Methods for forming mixed droplets
US11130128B2 (en) 2008-09-23 2021-09-28 Bio-Rad Laboratories, Inc. Detection method for a target nucleic acid
US11168353B2 (en) 2011-02-18 2021-11-09 Bio-Rad Laboratories, Inc. Compositions and methods for molecular labeling
US11174509B2 (en) 2013-12-12 2021-11-16 Bio-Rad Laboratories, Inc. Distinguishing rare variations in a nucleic acid sequence from a sample
US11187702B2 (en) 2003-03-14 2021-11-30 Bio-Rad Laboratories, Inc. Enzyme quantification
US11254968B2 (en) 2010-02-12 2022-02-22 Bio-Rad Laboratories, Inc. Digital analyte analysis
EP4001433A1 (en) * 2020-11-20 2022-05-25 Université de Paris A method for determining the level of dna integrity
US11371092B2 (en) * 2007-02-05 2022-06-28 Qiagen Sciences, Llc Detection device and methods of use
CN114717330A (zh) * 2022-04-13 2022-07-08 中国农业科学院北京畜牧兽医研究所 与绵羊单胎产羔数相关的snp分子标记、引物组、试剂盒及检测方法和应用
US11390917B2 (en) 2010-02-12 2022-07-19 Bio-Rad Laboratories, Inc. Digital analyte analysis
US11453906B2 (en) 2011-11-04 2022-09-27 Handylab, Inc. Multiplexed diagnostic detection apparatus and methods
US11511242B2 (en) 2008-07-18 2022-11-29 Bio-Rad Laboratories, Inc. Droplet libraries
CN115551639A (zh) * 2020-04-21 2022-12-30 赫孚孟拉罗股份公司 具有单分子传感器阵列的高通量核酸定序
US11635427B2 (en) 2010-09-30 2023-04-25 Bio-Rad Laboratories, Inc. Sandwich assays in droplets
US11786872B2 (en) 2004-10-08 2023-10-17 United Kingdom Research And Innovation Vitro evolution in microfluidic systems
US11806718B2 (en) 2006-03-24 2023-11-07 Handylab, Inc. Fluorescence detector for microfluidic diagnostic system
US11819849B2 (en) 2007-02-06 2023-11-21 Brandeis University Manipulation of fluids and reactions in microfluidic systems
US11898193B2 (en) 2011-07-20 2024-02-13 Bio-Rad Laboratories, Inc. Manipulating droplet size
US11901041B2 (en) 2013-10-04 2024-02-13 Bio-Rad Laboratories, Inc. Digital analysis of nucleic acid modification
US12030050B2 (en) 2020-11-04 2024-07-09 Handylab, Inc. Microfluidic cartridge and method of making same

Families Citing this family (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6048734A (en) 1995-09-15 2000-04-11 The Regents Of The University Of Michigan Thermal microvalves in a fluid flow method
US6696022B1 (en) * 1999-08-13 2004-02-24 U.S. Genomics, Inc. Methods and apparatuses for stretching polymers
JP5344817B2 (ja) * 2004-05-03 2013-11-20 ハンディーラブ インコーポレイテッド ポリヌクレオチド含有サンプルの処理
US7351538B2 (en) * 2004-08-23 2008-04-01 U.S. Genomics Systems and methods for detecting and analyzing polymers
FR2906032B1 (fr) * 2006-09-15 2008-12-12 Centre Nat Rech Scient Determination des rayons hydrodynamiques des constituants d'un melange par analyse d'une dispersion de taylor effectuee suite a une separation par electrophese capillaire.
ES2674101T3 (es) 2007-04-04 2018-06-27 Ande Corporation Plataformas de separación y detección microfluídicas de plástico
WO2009009127A2 (en) * 2007-07-12 2009-01-15 U.S. Genomics, Inc. Method and system for transferring and/or concentrating a sample
USD621060S1 (en) 2008-07-14 2010-08-03 Handylab, Inc. Microfluidic cartridge
US20090136385A1 (en) 2007-07-13 2009-05-28 Handylab, Inc. Reagent Tube
JP2010004781A (ja) * 2008-06-25 2010-01-14 Canon Inc 核酸増幅装置およびそれを用いた核酸分析装置
USD618820S1 (en) 2008-07-11 2010-06-29 Handylab, Inc. Reagent holder
EP2443254A2 (en) * 2009-06-15 2012-04-25 NetBio, Inc. Improved methods for forensic dna quantitation
ES2549491T3 (es) 2010-03-01 2015-10-28 Fida-Tech Aps Cuantificación de analito usando un análisis de dispersión inducida por flujo
US20110312732A1 (en) * 2010-06-17 2011-12-22 Geneasys Pty Ltd Test module using lanthanide metal-ligand complex, electrochemiluminescent luminophores
US20130046153A1 (en) 2011-08-16 2013-02-21 Elwha LLC, a limited liability company of the State of Delaware Systematic distillation of status data relating to regimen compliance
EP2776554B1 (en) 2011-11-11 2019-03-27 Icubate, Inc. Systems and methods for performing amplicon rescue multiplex polymerase chain reaction (pcr)
US9028776B2 (en) 2012-04-18 2015-05-12 Toxic Report Llc Device for stretching a polymer in a fluid sample
US8956815B2 (en) 2012-04-18 2015-02-17 Toxic Report Llc Intercalation methods and devices
US9422602B2 (en) 2012-08-15 2016-08-23 Bio-Rad Laboratories, Inc. Methods and compositions for determining nucleic acid degradation
US10479128B2 (en) 2017-10-27 2019-11-19 Assa Abloy Ab Security feature
DE102019200929A1 (de) * 2019-01-25 2020-07-30 Robert Bosch Gmbh Verfahren und Vorrichtung zum optischen Erfassen einer Länge eines Makromoleküls
US20230392188A1 (en) * 2022-06-02 2023-12-07 Perkinelmer Health Sciences, Inc. Electrophoresis-mediated characterization of dna content of adeno-associated virus capsids

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6174670B1 (en) * 1996-06-04 2001-01-16 University Of Utah Research Foundation Monitoring amplification of DNA during PCR
US6303305B1 (en) * 1999-03-30 2001-10-16 Roche Diagnostics, Gmbh Method for quantification of an analyte
US6440706B1 (en) * 1999-08-02 2002-08-27 Johns Hopkins University Digital amplification
US6586177B1 (en) * 1999-09-08 2003-07-01 Exact Sciences Corporation Methods for disease detection
US20050069904A1 (en) * 2003-09-29 2005-03-31 Stuart Peirson Analysis of real-time PCR data
US7081339B2 (en) * 2002-04-12 2006-07-25 Primera Biosystems, Inc. Methods for variation detection

Family Cites Families (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5277556A (en) * 1990-07-10 1994-01-11 Westonbridge International Limited Valve and micropump incorporating said valve
ES2075459T3 (es) * 1990-08-31 1995-10-01 Westonbridge Int Ltd Valvula equipada con detector de posicion y microbomba que incorpora dicha valvula.
US5498392A (en) * 1992-05-01 1996-03-12 Trustees Of The University Of Pennsylvania Mesoscale polynucleotide amplification device and method
DE4220077A1 (de) * 1992-06-19 1993-12-23 Bosch Gmbh Robert Mikropumpe
US5639423A (en) 1992-08-31 1997-06-17 The Regents Of The University Of Calfornia Microfabricated reactor
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
US5601982A (en) * 1995-02-07 1997-02-11 Sargent; Jeannine P. Method and apparatus for determining the sequence of polynucleotides
US5856174A (en) 1995-06-29 1999-01-05 Affymetrix, Inc. Integrated nucleic acid diagnostic device
US5939291A (en) * 1996-06-14 1999-08-17 Sarnoff Corporation Microfluidic method for nucleic acid amplification
CN1173776C (zh) 1996-06-28 2004-11-03 卡钳技术有限公司 在微规模流体性设备里的高通过量的筛选分析系统
CN1329729C (zh) * 1996-06-28 2007-08-01 卡钳生命科学股份有限公司 微流体系统
US6235471B1 (en) * 1997-04-04 2001-05-22 Caliper Technologies Corp. Closed-loop biochemical analyzers
US6391622B1 (en) * 1997-04-04 2002-05-21 Caliper Technologies Corp. Closed-loop biochemical analyzers
US5869004A (en) * 1997-06-09 1999-02-09 Caliper Technologies Corp. Methods and apparatus for in situ concentration and/or dilution of materials in microfluidic systems
US5965410A (en) * 1997-09-02 1999-10-12 Caliper Technologies Corp. Electrical current for controlling fluid parameters in microchannels
US6174675B1 (en) * 1997-11-25 2001-01-16 Caliper Technologies Corp. Electrical current for controlling fluid parameters in microchannels
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
US6503718B2 (en) * 1999-01-10 2003-01-07 Exact Sciences Corporation Methods for detecting mutations using primer extension for detecting disease
US6171850B1 (en) * 1999-03-08 2001-01-09 Caliper Technologies Corp. Integrated devices and systems for performing temperature controlled reactions and analyses
US6303343B1 (en) * 1999-04-06 2001-10-16 Caliper Technologies Corp. Inefficient fast PCR
WO2001002850A1 (en) * 1999-07-06 2001-01-11 Caliper Technologies Corp. Microfluidic systems and methods for determining modulator kinetics
US6828098B2 (en) 2000-05-20 2004-12-07 The Regents Of The University Of Michigan Method of producing a DNA library using positional amplification based on the use of adaptors and nick translation
AUPR221400A0 (en) * 2000-12-20 2001-01-25 Murdoch Childrens Research Institute, The Diagnostic assay

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6174670B1 (en) * 1996-06-04 2001-01-16 University Of Utah Research Foundation Monitoring amplification of DNA during PCR
US6303305B1 (en) * 1999-03-30 2001-10-16 Roche Diagnostics, Gmbh Method for quantification of an analyte
US6440706B1 (en) * 1999-08-02 2002-08-27 Johns Hopkins University Digital amplification
US6586177B1 (en) * 1999-09-08 2003-07-01 Exact Sciences Corporation Methods for disease detection
US7081339B2 (en) * 2002-04-12 2006-07-25 Primera Biosystems, Inc. Methods for variation detection
US20050069904A1 (en) * 2003-09-29 2005-03-31 Stuart Peirson Analysis of real-time PCR data

Cited By (327)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9376718B2 (en) 2000-11-16 2016-06-28 Caliper Life Sciences, Inc. Method and apparatus for generating thermal melting curves in a microfluidic device
US11162910B2 (en) 2000-11-16 2021-11-02 Caliper Life Sciences, Inc. Method and apparatus for generating thermal melting curves in a microfluidic device
US20070026421A1 (en) * 2000-11-16 2007-02-01 Caliper Life Sciences, Inc. Method and apparatus for generating thermal melting curves in a microfluidic device
US8900811B2 (en) 2000-11-16 2014-12-02 Caliper Life Sciences, Inc. Method and apparatus for generating thermal melting curves in a microfluidic device
US8734733B2 (en) 2001-02-14 2014-05-27 Handylab, Inc. Heat-reduction methods and systems related to microfluidic devices
US9528142B2 (en) 2001-02-14 2016-12-27 Handylab, Inc. Heat-reduction methods and systems related to microfluidic devices
US9051604B2 (en) 2001-02-14 2015-06-09 Handylab, Inc. Heat-reduction methods and systems related to microfluidic devices
US9259735B2 (en) 2001-03-28 2016-02-16 Handylab, Inc. Methods and systems for control of microfluidic devices
US8895311B1 (en) 2001-03-28 2014-11-25 Handylab, Inc. Methods and systems for control of general purpose microfluidic devices
US10351901B2 (en) 2001-03-28 2019-07-16 Handylab, Inc. Systems and methods for thermal actuation of microfluidic devices
US10571935B2 (en) 2001-03-28 2020-02-25 Handylab, Inc. Methods and systems for control of general purpose microfluidic devices
US10619191B2 (en) 2001-03-28 2020-04-14 Handylab, Inc. Systems and methods for thermal actuation of microfluidic devices
US9677121B2 (en) 2001-03-28 2017-06-13 Handylab, Inc. Systems and methods for thermal actuation of microfluidic devices
US8703069B2 (en) 2001-03-28 2014-04-22 Handylab, Inc. Moving microdroplets in a microfluidic device
US8768517B2 (en) 2001-03-28 2014-07-01 Handylab, Inc. Methods and systems for control of microfluidic devices
US8894947B2 (en) 2001-03-28 2014-11-25 Handylab, Inc. Systems and methods for thermal actuation of microfluidic devices
US8685341B2 (en) 2001-09-12 2014-04-01 Handylab, Inc. Microfluidic devices having a reduced number of input and output connections
US9028773B2 (en) 2001-09-12 2015-05-12 Handylab, Inc. Microfluidic devices having a reduced number of input and output connections
USRE45539E1 (en) * 2003-03-14 2015-06-02 Lawrence Livermore National Security, Llc Method for chemical amplification based on fluid partitioning in an immiscible liquid
USRE43365E1 (en) 2003-03-14 2012-05-08 Lawrence Livermore National Security, Llc Apparatus for chemical amplification based on fluid partitioning in an immiscible liquid
USRE41780E1 (en) 2003-03-14 2010-09-28 Lawrence Livermore National Security, Llc Chemical amplification based on fluid partitioning in an immiscible liquid
USRE46322E1 (en) * 2003-03-14 2017-02-28 Lawrence Livermore National Security, Llc Method for chemical amplification based on fluid partitioning in an immiscible liquid
US11187702B2 (en) 2003-03-14 2021-11-30 Bio-Rad Laboratories, Inc. Enzyme quantification
USRE47080E1 (en) * 2003-03-14 2018-10-09 Lawrence Livermore National Security, Llc Chemical amplification based on fluid partitioning
US7041481B2 (en) * 2003-03-14 2006-05-09 The Regents Of The University Of California Chemical amplification based on fluid partitioning
US20040180346A1 (en) * 2003-03-14 2004-09-16 The Regents Of The University Of California. Chemical amplification based on fluid partitioning
USRE48788E1 (en) * 2003-03-14 2021-10-26 Lawrence Livermore National Security, Llc Chemical amplification based on fluid partitioning
US11078523B2 (en) 2003-07-31 2021-08-03 Handylab, Inc. Processing particle-containing samples
US10731201B2 (en) 2003-07-31 2020-08-04 Handylab, Inc. Processing particle-containing samples
US10865437B2 (en) 2003-07-31 2020-12-15 Handylab, Inc. Processing particle-containing samples
US8679831B2 (en) 2003-07-31 2014-03-25 Handylab, Inc. Processing particle-containing samples
US20100197008A1 (en) * 2003-07-31 2010-08-05 Handylab, Inc. Processing particle-containing samples
US9670528B2 (en) 2003-07-31 2017-06-06 Handylab, Inc. Processing particle-containing samples
US10494663B1 (en) 2004-05-03 2019-12-03 Handylab, Inc. Method for processing polynucleotide-containing samples
US11441171B2 (en) 2004-05-03 2022-09-13 Handylab, Inc. Method for processing polynucleotide-containing samples
US8852862B2 (en) 2004-05-03 2014-10-07 Handylab, Inc. Method for processing polynucleotide-containing samples
US10364456B2 (en) 2004-05-03 2019-07-30 Handylab, Inc. Method for processing polynucleotide-containing samples
US10443088B1 (en) 2004-05-03 2019-10-15 Handylab, Inc. Method for processing polynucleotide-containing samples
US10604788B2 (en) 2004-05-03 2020-03-31 Handylab, Inc. System for processing polynucleotide-containing samples
US11786872B2 (en) 2004-10-08 2023-10-17 United Kingdom Research And Innovation Vitro evolution in microfluidic systems
US20070184547A1 (en) * 2005-10-11 2007-08-09 Kalyan Handique Polynucleotide sample preparation device
US20070154895A1 (en) * 2005-12-30 2007-07-05 Caliper Life Sciences, Inc. Multi-assay microfluidic chips
US20070161868A1 (en) * 2006-01-10 2007-07-12 Biosignia, Inc. Method and system for determining whether additional laboratory tests will yield values beyond a threshold level
US20170067047A1 (en) * 2006-01-11 2017-03-09 Raindance Technologies, Inc. Microfluidic devices and methods of use in the formation and control of nanoreactors
US20130217583A1 (en) * 2006-01-11 2013-08-22 Darren Link Microfluidic devices and methods of use in the formation and control of nanoreactors
US9328344B2 (en) * 2006-01-11 2016-05-03 Raindance Technologies, Inc. Microfluidic devices and methods of use in the formation and control of nanoreactors
US9534216B2 (en) * 2006-01-11 2017-01-03 Raindance Technologies, Inc. Microfluidic devices and methods of use in the formation and control of nanoreactors
US9752141B2 (en) * 2006-01-11 2017-09-05 Raindance Technologies, Inc. Microfluidic devices and methods of use in the formation and control of nanoreactors
JP2014138611A (ja) * 2006-01-11 2014-07-31 Raindance Technologies Inc ナノリアクターの形成および制御において使用するマイクロ流体デバイスおよび方法
JP2016027827A (ja) * 2006-01-11 2016-02-25 レインダンス テクノロジーズ, インコーポレイテッド ナノリアクターの形成および制御において使用するマイクロ流体デバイスおよび方法
US20140323317A1 (en) * 2006-01-11 2014-10-30 Raindance Technologies, Inc. Microfluidic devices and methods of use in the formation and control of nanoreactors
JP2014079252A (ja) * 2006-02-09 2014-05-08 Caliper Life Sciences Inc マイクロ流体素子内の熱融解曲線を生成する方法および装置
JP2016208994A (ja) * 2006-02-09 2016-12-15 カリパー ライフ サイエンシズ,インコーポレイテッド マイクロ流体素子内の熱融解曲線を生成する方法および装置
EP2402460A1 (en) 2006-02-09 2012-01-04 Caliper Life Sciences, Inc. Method and apparatus for generating thermal melting curves in a microfluidic device
US20070207272A1 (en) * 2006-03-03 2007-09-06 Puri Ishwar K Method and apparatus for magnetic mixing in micron size droplets
US20070292941A1 (en) * 2006-03-24 2007-12-20 Handylab, Inc. Integrated system for processing microfluidic samples, and method of using the same
US10695764B2 (en) 2006-03-24 2020-06-30 Handylab, Inc. Fluorescence detector for microfluidic diagnostic system
US11959126B2 (en) 2006-03-24 2024-04-16 Handylab, Inc. Microfluidic system for amplifying and detecting polynucleotides in parallel
US8883490B2 (en) 2006-03-24 2014-11-11 Handylab, Inc. Fluorescence detector for microfluidic diagnostic system
US11142785B2 (en) 2006-03-24 2021-10-12 Handylab, Inc. Microfluidic system for amplifying and detecting polynucleotides in parallel
US10799862B2 (en) 2006-03-24 2020-10-13 Handylab, Inc. Integrated system for processing microfluidic samples, and method of using same
US10821436B2 (en) 2006-03-24 2020-11-03 Handylab, Inc. Integrated system for processing microfluidic samples, and method of using the same
US11141734B2 (en) 2006-03-24 2021-10-12 Handylab, Inc. Fluorescence detector for microfluidic diagnostic system
US10821446B1 (en) 2006-03-24 2020-11-03 Handylab, Inc. Fluorescence detector for microfluidic diagnostic system
US11085069B2 (en) 2006-03-24 2021-08-10 Handylab, Inc. Microfluidic system for amplifying and detecting polynucleotides in parallel
US11666903B2 (en) 2006-03-24 2023-06-06 Handylab, Inc. Integrated system for processing microfluidic samples, and method of using same
US9080207B2 (en) 2006-03-24 2015-07-14 Handylab, Inc. Microfluidic system for amplifying and detecting polynucleotides in parallel
US10843188B2 (en) 2006-03-24 2020-11-24 Handylab, Inc. Integrated system for processing microfluidic samples, and method of using the same
US9040288B2 (en) 2006-03-24 2015-05-26 Handylab, Inc. Integrated system for processing microfluidic samples, and method of using the same
US11806718B2 (en) 2006-03-24 2023-11-07 Handylab, Inc. Fluorescence detector for microfluidic diagnostic system
US9802199B2 (en) 2006-03-24 2017-10-31 Handylab, Inc. Fluorescence detector for microfluidic diagnostic system
US10900066B2 (en) 2006-03-24 2021-01-26 Handylab, Inc. Microfluidic system for amplifying and detecting polynucleotides in parallel
US10913061B2 (en) 2006-03-24 2021-02-09 Handylab, Inc. Integrated system for processing microfluidic samples, and method of using the same
US10857535B2 (en) 2006-03-24 2020-12-08 Handylab, Inc. Integrated system for processing microfluidic samples, and method of using same
US10927407B2 (en) 2006-05-11 2021-02-23 Bio-Rad Laboratories, Inc. Systems and methods for handling microfluidic droplets
US10639597B2 (en) 2006-05-11 2020-05-05 Bio-Rad Laboratories, Inc. Microfluidic devices
US11351510B2 (en) 2006-05-11 2022-06-07 Bio-Rad Laboratories, Inc. Microfluidic devices
US20070298429A1 (en) * 2006-06-14 2007-12-27 Walter Gumbrecht Process for determining the concentration of nucleic acids
US9347097B2 (en) 2006-06-14 2016-05-24 Boehringer Ingelheim Vetmedica Gmbh Arrangement for determining the concentration of nucleic acids
US8715924B2 (en) 2006-06-14 2014-05-06 Siemens Aktiengesellschaft Process for determining the concentration of nucleic acids
FR2902441A1 (fr) * 2006-06-14 2007-12-21 Siemens Ag Procede de determination de la concentration d'acides nucleiques
US20080096766A1 (en) * 2006-06-16 2008-04-24 Sequenom, Inc. Methods and compositions for the amplification, detection and quantification of nucleic acid from a sample
WO2007147063A2 (en) * 2006-06-16 2007-12-21 Sequenom, Inc. Methods and compositions for the amplification, detection and quantification of nucleic acid from a sample
WO2007147063A3 (en) * 2006-06-16 2009-04-02 Sequenom Inc Methods and compositions for the amplification, detection and quantification of nucleic acid from a sample
US20080003585A1 (en) * 2006-06-29 2008-01-03 Bio-Rad Laboratories, Inc., A Corporation Of The State Of Delaware Purification and amplification of nucleic acids in a microfluidic device
US20080003588A1 (en) * 2006-06-30 2008-01-03 Canon U.S. Life Sciences, Inc. Real-time PCR in micro-channels
US10583442B2 (en) 2006-06-30 2020-03-10 Canon U.S.A., Inc. Real-time PCR in micro-channels
US8232094B2 (en) 2006-06-30 2012-07-31 Canon U.S. Life Sciences, Inc. Real-time PCR in micro-channels
US8409848B2 (en) 2006-06-30 2013-04-02 Shulin Zeng System and method for rapid thermal cycling
US9573132B2 (en) 2006-06-30 2017-02-21 Canon U.S. Life Sciences, Inc. System and methods for monitoring the amplification and dissociation behavior of DNA molecules
US20100028980A1 (en) * 2006-06-30 2010-02-04 Canon U.S. Life Sciences, Inc. Real-time pcr in micro-channels
US7629124B2 (en) 2006-06-30 2009-12-08 Canon U.S. Life Sciences, Inc. Real-time PCR in micro-channels
US8058054B2 (en) 2006-06-30 2011-11-15 Canon U.S. Life Sciences, Inc. Systems and methods for real-time PCR
US9592510B2 (en) 2006-06-30 2017-03-14 Canon U.S. Life Sciences, Inc. Real-time PCR in micro-channels
US20080003594A1 (en) * 2006-06-30 2008-01-03 Hasson Kenton C Systems and methods for monitoring the amplification and dissociation behavior of DNA molecules
US20090053726A1 (en) * 2006-06-30 2009-02-26 Canon U.S. Life Sciences, Inc. Systems and methods for real-time pcr
US20080003593A1 (en) * 2006-06-30 2008-01-03 Hasson Kenton C System and methods for monitoring the amplification and dissociation behavior of DNA molecules
US7906319B2 (en) * 2006-06-30 2011-03-15 Canon U.S. Life Sciences, Inc. Systems and methods for monitoring the amplification and dissociation behavior of DNA molecules
US20080176230A1 (en) * 2006-06-30 2008-07-24 Canon U.S. Life Sciences, Inc. Systems and methods for real-time pcr
US20080176289A1 (en) * 2006-06-30 2008-07-24 Canon U.S. Life Sciences, Inc. System and method for rapid thermal cycling
US9283563B2 (en) 2006-06-30 2016-03-15 Canon U.S. Life Sciences, Inc. Systems and methods for real-time PCR
US9815057B2 (en) 2006-11-14 2017-11-14 Handylab, Inc. Microfluidic cartridge and method of making same
US20100173393A1 (en) * 2006-11-14 2010-07-08 Handy Lab, Inc. Microfluidic valve and method of making same
US8765076B2 (en) 2006-11-14 2014-07-01 Handylab, Inc. Microfluidic valve and method of making same
US10710069B2 (en) 2006-11-14 2020-07-14 Handylab, Inc. Microfluidic valve and method of making same
US8709787B2 (en) 2006-11-14 2014-04-29 Handylab, Inc. Microfluidic cartridge and method of using same
US7593560B2 (en) 2006-11-30 2009-09-22 Canon U.S. Life Sciences, Inc. Systems and methods for monitoring the amplification and dissociation behavior of DNA molecules
US20090324037A1 (en) * 2006-11-30 2009-12-31 Canon U.S. Life Sciences, Inc. Systems and methods for monitoring the amplification and dissociation behavior of dna molecules
US20080130971A1 (en) * 2006-11-30 2008-06-05 Canon U.S. Life Sciences, Inc. Systems and methods for monitoring the amplification and dissociation behavior of dna molecules
US9732380B2 (en) 2006-11-30 2017-08-15 Canon U.S. Life Sciences, Inc. Systems and methods for monitoring the amplification and dissociation behavior of DNA molecules
US8989466B2 (en) 2006-11-30 2015-03-24 Canon U.S. Life Sciences, Inc. Systems and methods for monitoring the amplification and dissociation behavior of DNA molecules
US8306294B2 (en) 2006-11-30 2012-11-06 Canon U.S. Life Sciences, Inc. Systems and methods for monitoring the amplification and dissociation behavior of DNA molecules
US11371092B2 (en) * 2007-02-05 2022-06-28 Qiagen Sciences, Llc Detection device and methods of use
US11819849B2 (en) 2007-02-06 2023-11-21 Brandeis University Manipulation of fluids and reactions in microfluidic systems
US8652780B2 (en) 2007-03-26 2014-02-18 Sequenom, Inc. Restriction endonuclease enhanced polymorphic sequence detection
US11618024B2 (en) 2007-04-19 2023-04-04 President And Fellows Of Harvard College Manipulation of fluids, fluid components and reactions in microfluidic systems
US11224876B2 (en) 2007-04-19 2022-01-18 Brandeis University Manipulation of fluids, fluid components and reactions in microfluidic systems
US10960397B2 (en) 2007-04-19 2021-03-30 President And Fellows Of Harvard College Manipulation of fluids, fluid components and reactions in microfluidic systems
US9238223B2 (en) 2007-07-13 2016-01-19 Handylab, Inc. Microfluidic cartridge
US10844368B2 (en) 2007-07-13 2020-11-24 Handylab, Inc. Diagnostic apparatus to extract nucleic acids including a magnetic assembly and a heater assembly
US10632466B1 (en) 2007-07-13 2020-04-28 Handylab, Inc. Integrated apparatus for performing nucleic acid extraction and diagnostic testing on multiple biological samples
US10071376B2 (en) 2007-07-13 2018-09-11 Handylab, Inc. Integrated apparatus for performing nucleic acid extraction and diagnostic testing on multiple biological samples
US10625261B2 (en) 2007-07-13 2020-04-21 Handylab, Inc. Integrated apparatus for performing nucleic acid extraction and diagnostic testing on multiple biological samples
US11549959B2 (en) 2007-07-13 2023-01-10 Handylab, Inc. Automated pipetting apparatus having a combined liquid pump and pipette head system
US10625262B2 (en) 2007-07-13 2020-04-21 Handylab, Inc. Integrated apparatus for performing nucleic acid extraction and diagnostic testing on multiple biological samples
US20090134069A1 (en) * 2007-07-13 2009-05-28 Handylab, Inc. Integrated Heater and Magnetic Separator
US10875022B2 (en) 2007-07-13 2020-12-29 Handylab, Inc. Integrated apparatus for performing nucleic acid extraction and diagnostic testing on multiple biological samples
US10590410B2 (en) 2007-07-13 2020-03-17 Handylab, Inc. Polynucleotide capture materials, and methods of using same
US11466263B2 (en) 2007-07-13 2022-10-11 Handylab, Inc. Diagnostic apparatus to extract nucleic acids including a magnetic assembly and a heater assembly
US9618139B2 (en) 2007-07-13 2017-04-11 Handylab, Inc. Integrated heater and magnetic separator
US11060082B2 (en) 2007-07-13 2021-07-13 Handy Lab, Inc. Polynucleotide capture materials, and systems using same
US9347586B2 (en) 2007-07-13 2016-05-24 Handylab, Inc. Automated pipetting apparatus having a combined liquid pump and pipette head system
US9186677B2 (en) 2007-07-13 2015-11-17 Handylab, Inc. Integrated apparatus for performing nucleic acid extraction and diagnostic testing on multiple biological samples
US11845081B2 (en) 2007-07-13 2023-12-19 Handylab, Inc. Integrated apparatus for performing nucleic acid extraction and diagnostic testing on multiple biological samples
US9217143B2 (en) 2007-07-13 2015-12-22 Handylab, Inc. Polynucleotide capture materials, and methods of using same
US9701957B2 (en) 2007-07-13 2017-07-11 Handylab, Inc. Reagent holder, and kits containing same
US10234474B2 (en) 2007-07-13 2019-03-19 Handylab, Inc. Automated pipetting apparatus having a combined liquid pump and pipette head system
US10179910B2 (en) 2007-07-13 2019-01-15 Handylab, Inc. Rack for sample tubes and reagent holders
US8710211B2 (en) 2007-07-13 2014-04-29 Handylab, Inc. Polynucleotide capture materials, and methods of using same
US10139012B2 (en) 2007-07-13 2018-11-27 Handylab, Inc. Integrated heater and magnetic separator
US10717085B2 (en) 2007-07-13 2020-07-21 Handylab, Inc. Integrated apparatus for performing nucleic acid extraction and diagnostic testing on multiple biological samples
US10100302B2 (en) 2007-07-13 2018-10-16 Handylab, Inc. Polynucleotide capture materials, and methods of using same
US11266987B2 (en) 2007-07-13 2022-03-08 Handylab, Inc. Microfluidic cartridge
US11254927B2 (en) 2007-07-13 2022-02-22 Handylab, Inc. Polynucleotide capture materials, and systems using same
US9259734B2 (en) 2007-07-13 2016-02-16 Handylab, Inc. Integrated apparatus for performing nucleic acid extraction and diagnostic testing on multiple biological samples
US10065185B2 (en) 2007-07-13 2018-09-04 Handylab, Inc. Microfluidic cartridge
US20110183330A1 (en) * 2007-08-03 2011-07-28 The Chinese University Of Hong Kong Analysis for Nucleic Acids by Digital PCR
AU2008285465B2 (en) * 2007-08-03 2014-04-17 The Chinese University Of Hong Kong Analysis of nucleic acids of varying lengths by digital PCR
US8722334B2 (en) * 2007-08-03 2014-05-13 The Chinese University Of Hong Kong Analysis for nucleic acids by digital PCR
US20090053719A1 (en) * 2007-08-03 2009-02-26 The Chinese University Of Hong Kong Analysis of nucleic acids by digital pcr
US8306773B2 (en) 2007-08-29 2012-11-06 Canon U.S. Life Sciences, Inc. Microfluidic devices with integrated resistive heater electrodes including systems and methods for controlling and measuring the temperatures of such heater electrodes
US9221056B2 (en) 2007-08-29 2015-12-29 Canon U.S. Life Sciences, Inc. Microfluidic devices with integrated resistive heater electrodes including systems and methods for controlling and measuring the temperatures of such heater electrodes
US9527083B2 (en) 2007-08-29 2016-12-27 Canon U.S. Life Sciences, Inc. Microfluidic devices with integrated resistive heater electrodes including systems and methods for controlling and measuring the temperatures of such heater electrodes
US20090060795A1 (en) * 2007-08-29 2009-03-05 Canon U.S. Life Sciences, Inc. Microfluidic devices with integrated resistive heater electrodes including systems and methods for controlling and measuring the temperatures of such heater electrodes
US9873122B2 (en) 2007-08-29 2018-01-23 Canon U.S. Life Sciences, Inc. Microfluidic devices with integrated resistive heater electrodes including systems and methods for controlling and measuring the temperatures of such heater electrodes
US9267852B2 (en) 2007-08-29 2016-02-23 Canon U.S. Life Sciences, Inc. Microfluidic devices with integrated resistive heater electrodes including systems and methods for controlling and measuring the temperatures of such heater electrodes
US9829389B2 (en) 2007-08-29 2017-11-28 Canon U.S. Life Sciences, Inc. Microfluidic devices with integrated resistive heater electrodes including systems and methods for controlling and measuring the temperatures of such heater electrodes
US9823135B2 (en) 2007-08-29 2017-11-21 Canon U.S. Life Sciences, Inc. Microfluidic devices with integrated resistive heater electrodes including systems and methods for controlling and measuring the temperatures of such heater electrodes
US20090248349A1 (en) * 2007-08-29 2009-10-01 Canon U.S. Life Sciences, Inc. Microfluidic devices with integrated resistive heater electrodes including systems and methods for controlling and measuring the temperatures of such heater electrodes
US8380457B2 (en) 2007-08-29 2013-02-19 Canon U.S. Life Sciences, Inc. Microfluidic devices with integrated resistive heater electrodes including systems and methods for controlling and measuring the temperatures of such heater electrodes
US20110077897A1 (en) * 2007-08-29 2011-03-31 Canon U.S. Life Sciences, Inc. Microfluidic devices with integrated resistive heater electrodes including systems and methods for controlling and measuring the temperatures of such heater electrodes
US9492826B2 (en) 2007-08-29 2016-11-15 Canon U.S. Life Sciences, Inc. Microfluidic devices with integrated resistive heater electrodes including systems and methods for controlling and measuring the temperatures of such heater electrodes
US20110056926A1 (en) * 2007-08-29 2011-03-10 Canon U.S. Life Sciences, Inc. Microfluidic devices with integrated resistive heater electrodes including systems and methods for controlling and measuring the temperatures of such heater electrodes
US20090112484A1 (en) * 2007-10-25 2009-04-30 Canon U.S. Life Sciences, Inc. High-resolution melting analysis
US9292653B2 (en) 2007-10-25 2016-03-22 Canon U.S. Life Sciences, Inc. High-resolution melting analysis
US20090111149A1 (en) * 2007-10-25 2009-04-30 Canon U.S. Life Sciences, Inc. Method of reducing cross-contamination in continuous amplification reactions in a channel
US8180572B2 (en) 2007-10-25 2012-05-15 Canon U.S. Life Sciences, Inc. High-resolution melting analysis
US9114397B2 (en) 2007-10-25 2015-08-25 Canon U.S. Life Sciences, Inc. Method of reducing cross-contamination in continuous amplification reactions in a channel
US8145433B2 (en) 2007-10-25 2012-03-27 Canon U.S. Life Sciences, Inc. High-resolution melting analysis
US20090130746A1 (en) * 2007-10-25 2009-05-21 Canon U.S. Life Sciences, Inc. Microchannel surface coating
US8189186B2 (en) 2007-12-27 2012-05-29 Lawrence Livermore National Security, Llc. Signal enhancement using a switchable magnetic trap
US20110215798A1 (en) * 2007-12-27 2011-09-08 Lawrence Livermore National Security, Llc. Signal Enhancement Using a Switchable Magnetic Trap
US8815576B2 (en) 2007-12-27 2014-08-26 Lawrence Livermore National Security, Llc. Chip-based sequencing nucleic acids
US20100184020A1 (en) * 2007-12-27 2010-07-22 Lawrence Livermore National Security, Llc. Chip-Based Sequencing Nucleic Acids
US8610032B2 (en) 2008-03-21 2013-12-17 Lawrence Livermore National Security, Llc Laser heating of aqueous samples on a micro-optical-electro-mechanical system
US20090261086A1 (en) * 2008-03-21 2009-10-22 Neil Reginald Beer Laser Heating of Aqueous Samples on a Micro-Optical-Electro-Mechanical System
US8367976B2 (en) 2008-03-21 2013-02-05 Lawrence Livermore National Security, Llc Laser heating of aqueous samples on a micro-optical-electro-mechanical system
US8722336B2 (en) 2008-03-26 2014-05-13 Sequenom, Inc. Restriction endonuclease enhanced polymorphic sequence detection
US10828641B2 (en) 2008-04-28 2020-11-10 President And Fellows Of Harvard College Microfluidic device for storage and well-defined arrangement of droplets
US9664619B2 (en) * 2008-04-28 2017-05-30 President And Fellows Of Harvard College Microfluidic device for storage and well-defined arrangement of droplets
US20110190146A1 (en) * 2008-04-28 2011-08-04 President And Fellows Of Harvard College Microfluidic device for storage and well-defined arrangement of droplets
US11498072B2 (en) 2008-04-28 2022-11-15 President And Fellows Of Harvard College Microfluidic device for storage and well-defined arrangement of droplets
US20090317874A1 (en) * 2008-06-23 2009-12-24 Canon U.S. Life Sciences, Inc. Systems and methods for amplifying nucleic acids
US9393566B2 (en) 2008-06-23 2016-07-19 Canon U.S. Life Sciences, Inc. System and method for temperature referencing for melt curve data collection
US10376891B2 (en) 2008-06-23 2019-08-13 Canon U.S. Life Sciences, Inc. System and method for temperature referencing for melt curve data collection
US20090318306A1 (en) * 2008-06-23 2009-12-24 Canon U.S. Life Sciences, Inc. System and method for temperature referencing for melt curve data collection
US9724695B2 (en) 2008-06-23 2017-08-08 Canon U.S. Life Sciences, Inc. Systems and methods for amplifying nucleic acids
US10632470B2 (en) 2008-06-23 2020-04-28 Canon U.S.A., Inc. Systems and methods for amplifying nucleic acids
USD787087S1 (en) 2008-07-14 2017-05-16 Handylab, Inc. Housing
US9513196B2 (en) 2008-07-18 2016-12-06 Canon U.S. Life Sciences, Inc. Methods and systems for microfluidic DNA sample preparation
US8313906B2 (en) 2008-07-18 2012-11-20 Canon U.S. Life Sciences, Inc. Methods and systems for microfluidic DNA sample preparation
US20100021910A1 (en) * 2008-07-18 2010-01-28 Canon U.S. Life Sciences, Inc. Methods and Systems for Microfluidic DNA Sample Preparation
US11511242B2 (en) 2008-07-18 2022-11-29 Bio-Rad Laboratories, Inc. Droplet libraries
US11534727B2 (en) 2008-07-18 2022-12-27 Bio-Rad Laboratories, Inc. Droplet libraries
US11596908B2 (en) 2008-07-18 2023-03-07 Bio-Rad Laboratories, Inc. Droplet libraries
US20110264380A1 (en) * 2008-07-21 2011-10-27 Cottet Herve Determination of the hydrodynamic radii and/or content of constituents of a mixture by analysis of the taylor dispersion of the mixture in a capillary tube
US10123380B2 (en) 2008-08-08 2018-11-06 Lawrence Livermore National Security, Llc Instantaneous in-line heating of samples on a monolithic microwave integrated circuit microfluidic device
US20110092373A1 (en) * 2008-09-23 2011-04-21 Quantalife, Inc. System for transporting emulsions from an array to a detector
US11130128B2 (en) 2008-09-23 2021-09-28 Bio-Rad Laboratories, Inc. Detection method for a target nucleic acid
US11612892B2 (en) 2008-09-23 2023-03-28 Bio-Rad Laboratories, Inc. Method of performing droplet-based assays
US11633739B2 (en) 2008-09-23 2023-04-25 Bio-Rad Laboratories, Inc. Droplet-based assay system
US9126160B2 (en) 2008-09-23 2015-09-08 Bio-Rad Laboratories, Inc. System for forming an array of emulsions
US9764322B2 (en) 2008-09-23 2017-09-19 Bio-Rad Laboratories, Inc. System for generating droplets with pressure monitoring
US9623384B2 (en) * 2008-09-23 2017-04-18 Bio-Rad Laboratories, Inc. System for transporting emulsions from an array to a detector
US9132394B2 (en) 2008-09-23 2015-09-15 Bio-Rad Laboratories, Inc. System for detection of spaced droplets
US9636682B2 (en) 2008-09-23 2017-05-02 Bio-Rad Laboratories, Inc. System for generating droplets—instruments and cassette
US20110086780A1 (en) * 2008-09-23 2011-04-14 Quantalife, Inc. System for forming an array of emulsions
US9156010B2 (en) 2008-09-23 2015-10-13 Bio-Rad Laboratories, Inc. Droplet-based assay system
US9248417B2 (en) 2008-09-23 2016-02-02 Bio-Rad Laboratories, Inc. System for droplet-based assays using an array of emulsions
US9243288B2 (en) 2008-09-23 2016-01-26 Bio-Rad Laboratories, Inc. Cartridge with lysis chamber and droplet generator
US20110092376A1 (en) * 2008-09-23 2011-04-21 Quantalife, Inc. System for droplet-based assays using an array of emulsions
US8633015B2 (en) 2008-09-23 2014-01-21 Bio-Rad Laboratories, Inc. Flow-based thermocycling system with thermoelectric cooler
US9417190B2 (en) 2008-09-23 2016-08-16 Bio-Rad Laboratories, Inc. Calibrations and controls for droplet-based assays
US10512910B2 (en) 2008-09-23 2019-12-24 Bio-Rad Laboratories, Inc. Droplet-based analysis method
US20110212516A1 (en) * 2008-09-23 2011-09-01 Ness Kevin D Flow-based thermocycling system with thermoelectric cooler
US9649635B2 (en) 2008-09-23 2017-05-16 Bio-Rad Laboratories, Inc. System for generating droplets with push-back to remove oil
US9492797B2 (en) 2008-09-23 2016-11-15 Bio-Rad Laboratories, Inc. System for detection of spaced droplets
US9216392B2 (en) 2008-09-23 2015-12-22 Bio-Rad Laboratories, Inc. System for forming an array of emulsions
US10279350B2 (en) 2008-09-23 2019-05-07 Bio-Rad Laboratories, Inc. Method of generating droplets
US10258988B2 (en) 2008-09-23 2019-04-16 Bio-Rad Laboratories, Inc. Device for generating droplets
US10258989B2 (en) 2008-09-23 2019-04-16 Bio-Rad Laboratories, Inc. Method of making a device for generating droplets
US11130134B2 (en) 2008-09-23 2021-09-28 Bio-Rad Laboratories, Inc. Method of performing droplet-based assays
US9404152B2 (en) 2009-01-26 2016-08-02 Canon U.S. Life Sciences, Inc. Microfluidic flow monitoring
US10066977B2 (en) 2009-01-26 2018-09-04 Canon U.S. Life Sciences, Inc. Microfluidic flow monitoring
US20100233687A1 (en) * 2009-03-10 2010-09-16 Canon U.S. Life Sciences, Inc. Method and system for temperature correction in thermal melt analysis
US9542526B2 (en) 2009-03-10 2017-01-10 Canon U.S. Life Sciences, Inc. Method and system for temperature correction in thermal melt analysis
US9415392B2 (en) * 2009-03-24 2016-08-16 The University Of Chicago Slip chip device and methods
US20120028342A1 (en) * 2009-03-24 2012-02-02 Ismagilov Rustem F Slip chip device and methods
KR20130008439A (ko) * 2009-03-24 2013-01-22 유니버시티 오브 시카고 슬립칩 장치 및 방법
US10370705B2 (en) 2009-03-24 2019-08-06 University Of Chicago Analysis devices, kits, and related methods for digital quantification of nucleic acids and other analytes
US10196700B2 (en) 2009-03-24 2019-02-05 University Of Chicago Multivolume devices, kits and related methods for quantification and detection of nucleic acids and other analytes
KR101702154B1 (ko) * 2009-03-24 2017-02-03 유니버시티 오브 시카고 반응을 수행하기 위한 장치
US10543485B2 (en) 2009-03-24 2020-01-28 University Of Chicago Slip chip device and methods
US9630158B2 (en) 2009-04-10 2017-04-25 Canon U.S. Life Sciences, Inc. Method of delivering PCR solution to microfluidic PCR chamber
US8412466B2 (en) 2009-04-13 2013-04-02 Canon U.S. Life Sciences, Inc. Rapid method of pattern recognition, machine learning, and automated genotype classification through correlation analysis of dynamic signals
US8483972B2 (en) 2009-04-13 2013-07-09 Canon U.S. Life Sciences, Inc. System and method for genotype analysis and enhanced monte carlo simulation method to estimate misclassification rate in automated genotyping
US20110010103A1 (en) * 2009-04-13 2011-01-13 Canon U.S. Life Sciences, Inc. Rapid method of pattern recognition, machine learning, and automated genotype classification through correlation analysis of dynamic signals
US8337082B2 (en) 2009-05-08 2012-12-25 Canon U.S. Life Sciences, Inc. Systems and methods for auto-calibration of resistive temperature sensors
US20100285571A1 (en) * 2009-05-08 2010-11-11 Canon U.S. Life Sciences, Inc. Systems and methods for auto-calibration of resistive temperature sensors
US9939336B2 (en) 2009-05-08 2018-04-10 Canon U.S. Life Sciences, Inc. Systems and methods for auto-calibration of resistive temperature sensors
US8794831B2 (en) 2009-05-08 2014-08-05 Canon U.S. Life Sciences, Inc. Systems and methods for auto-calibration of resistive temperature sensors
US9234236B2 (en) 2009-05-14 2016-01-12 Canon U.S. Life Sciences, Inc. Microfluidic chip features for optical and thermal isolation
US8852527B2 (en) 2009-05-14 2014-10-07 Canon U.S. Life Sciences, Inc. Microfluidic chip features for optical and thermal isolation
US20110064628A1 (en) * 2009-05-14 2011-03-17 Canon U.S. Life Sciences, Inc. Microfluidic chip features for optical and thermal isolation
US8329117B2 (en) 2009-05-14 2012-12-11 Canon U.S. Life Sciences, Inc. Microfluidic chip features for optical and thermal isolation
US9061278B2 (en) 2009-06-29 2015-06-23 Canon U.S. Life Sciences, Inc. Microfluidic systems and methods for thermal control
US20110048547A1 (en) * 2009-06-29 2011-03-03 Canon U.S. Life Sciences, Inc. Microfluidic systems and methods for thermal control
US20110014605A1 (en) * 2009-07-17 2011-01-20 Canon U.S. Life Sciences, Inc. Methods and systems for dna isolation on a microfluidic device
US8304185B2 (en) 2009-07-17 2012-11-06 Canon U.S. Life Sciences, Inc. Methods and systems for DNA isolation on a microfluidic device
WO2011008217A1 (en) 2009-07-17 2011-01-20 Canon U.S. Life Sciences, Inc. Methods and systems for dna isolation on a microfluidic device
US9938519B2 (en) 2009-07-17 2018-04-10 Canon U.S. Life Sciences, Inc. Methods and systems for DNA isolation on a microfluidic device
US9116088B2 (en) 2009-07-17 2015-08-25 Canon U.S. Life Sciences, Inc. Methods and systems for DNA isolation on a microfluidic device
US9194861B2 (en) 2009-09-02 2015-11-24 Bio-Rad Laboratories, Inc. Method of mixing fluids by coalescence of multiple emulsions
US10166522B2 (en) 2009-09-02 2019-01-01 Bio-Rad Laboratories, Inc. System for mixing fluids by coalescence of multiple emulsions
US20110053798A1 (en) * 2009-09-02 2011-03-03 Quantalife, Inc. System for mixing fluids by coalescence of multiple emulsions
RU2562161C2 (ru) * 2009-09-02 2015-09-10 Эккуджен Пти Лтд Улучшенный способ количественного определения нуклеиновой кислоты
US10677693B2 (en) 2009-09-02 2020-06-09 Bio-Rad Laboratories, Inc. System for mixing fluids by coalescence of multiple emulsions
US11254968B2 (en) 2010-02-12 2022-02-22 Bio-Rad Laboratories, Inc. Digital analyte analysis
US10808279B2 (en) 2010-02-12 2020-10-20 Bio-Rad Laboratories, Inc. Digital analyte analysis
US11390917B2 (en) 2010-02-12 2022-07-19 Bio-Rad Laboratories, 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
US8709762B2 (en) 2010-03-02 2014-04-29 Bio-Rad Laboratories, Inc. System for hot-start amplification via a multiple emulsion
US9598725B2 (en) 2010-03-02 2017-03-21 Bio-Rad Laboratories, Inc. Emulsion chemistry for encapsulated droplets
US20110217712A1 (en) * 2010-03-02 2011-09-08 Quantalife, Inc. Emulsion chemistry for encapsulated droplets
US11866771B2 (en) 2010-03-02 2024-01-09 Bio-Rad Laboratories, Inc. Emulsion chemistry for encapsulated droplets
US11060136B2 (en) 2010-03-02 2021-07-13 Bio-Rad Laboratories, Inc. Emulsion chemistry for encapsulated droplets
US10378048B2 (en) 2010-03-02 2019-08-13 Bio-Rad Laboratories, Inc. Emulsion chemistry for encapsulated droplets
US10272432B2 (en) 2010-03-25 2019-04-30 Bio-Rad Laboratories, Inc. Device for generating droplets
US9500664B2 (en) 2010-03-25 2016-11-22 Bio-Rad Laboratories, Inc. Droplet generation for droplet-based assays
US10744506B2 (en) 2010-03-25 2020-08-18 Bio-Rad Laboratories, Inc. Device for generating droplets
US8730479B2 (en) 2010-03-25 2014-05-20 Bio-Rad Laboratories, Inc. Detection system for droplet-based assays
US10099219B2 (en) 2010-03-25 2018-10-16 Bio-Rad Laboratories, Inc. Device for generating droplets
US9393560B2 (en) 2010-03-25 2016-07-19 Bio-Rad Laboratories, Inc. Droplet transport system for detection
WO2012011660A3 (ko) * 2010-07-23 2012-05-03 (주)바이오니아 시료 내장 마이크로 챔버 플레이트 및 분석용 마이크로 챔버 프레이트의 제조 방법, 분석용 마이크로 챔버 플레이트 및 시료 내장 마이크로 챔버 플레이트 제조 장치 셋
US9151714B2 (en) 2010-07-23 2015-10-06 Bioneer Corporation Method of manufacturing micro chamber plate with built-in sample and analytic micro chamber plate, analytic micro chamber plate and apparatus set for manufacturing analytic micro chamber plate with built-in sample
US8962252B2 (en) 2010-08-31 2015-02-24 Canon U.S. Life Sciences, Inc. Optical system for high resolution thermal melt detection
US9701997B2 (en) 2010-08-31 2017-07-11 Canon U.S. Life Sciences, Inc. Composition and method for in-system priming microfluidic devices
US10266873B2 (en) 2010-08-31 2019-04-23 Canon U.S. Life Sciences, Inc. Optical system for high resolution thermal melt detection
US9919314B2 (en) 2010-08-31 2018-03-20 Canon U.S. Life Sciences, Inc. Air cooling systems and methods for microfluidic devices
US9168529B2 (en) 2010-08-31 2015-10-27 Canon U.S. Life Sciences, Inc. Air cooling systems and methods for microfluidic devices
US9061279B2 (en) 2010-08-31 2015-06-23 Canon U.S. Life Sciences, Inc. Composition and method for in-system priming microfluidic devices
US11635427B2 (en) 2010-09-30 2023-04-25 Bio-Rad Laboratories, Inc. Sandwich assays in droplets
US9089844B2 (en) 2010-11-01 2015-07-28 Bio-Rad Laboratories, Inc. System for forming emulsions
US11077415B2 (en) 2011-02-11 2021-08-03 Bio-Rad Laboratories, Inc. Methods for forming mixed droplets
US11168353B2 (en) 2011-02-18 2021-11-09 Bio-Rad Laboratories, Inc. Compositions and methods for molecular labeling
US11965877B2 (en) 2011-02-18 2024-04-23 Bio-Rad Laboratories, Inc. Compositions and methods for molecular labeling
US11768198B2 (en) 2011-02-18 2023-09-26 Bio-Rad Laboratories, Inc. Compositions and methods for molecular labeling
US11747327B2 (en) 2011-02-18 2023-09-05 Bio-Rad Laboratories, Inc. Compositions and methods for molecular labeling
US9222128B2 (en) 2011-03-18 2015-12-29 Bio-Rad Laboratories, Inc. Multiplexed digital assays with combinatorial use of signals
US20140248623A1 (en) * 2011-04-01 2014-09-04 Life Technologies Corporation System and Method for Determining Copies-per-Unit-Volume Using PCR and Flow Control of Droplets
US10557174B2 (en) 2011-04-01 2020-02-11 Life Technologies Corporation System and method for determining copies-per-unit-volume using PCR and flow control of droplets
US9322055B2 (en) * 2011-04-01 2016-04-26 Life Technologies Corporation System and method for determining copies-per-unit-volume using PCR and flow control of droplets
US11788127B2 (en) 2011-04-15 2023-10-17 Becton, Dickinson And Company Scanning real-time microfluidic thermocycler and methods for synchronized thermocycling and scanning optical detection
US10781482B2 (en) 2011-04-15 2020-09-22 Becton, Dickinson And Company Scanning real-time microfluidic thermocycler and methods for synchronized thermocycling and scanning optical detection
US9765389B2 (en) 2011-04-15 2017-09-19 Becton, Dickinson And Company Scanning real-time microfluidic thermocycler and methods for synchronized thermocycling and scanning optical detection
US11939573B2 (en) 2011-04-25 2024-03-26 Bio-Rad Laboratories, Inc. Methods and compositions for nucleic acid analysis
US9347059B2 (en) 2011-04-25 2016-05-24 Bio-Rad Laboratories, Inc. Methods and compositions for nucleic acid analysis
US10760073B2 (en) 2011-04-25 2020-09-01 Bio-Rad Laboratories, Inc. Methods and compositions for nucleic acid analysis
US10190115B2 (en) 2011-04-25 2019-01-29 Bio-Rad Laboratories, Inc. Methods and compositions for nucleic acid analysis
US9885034B2 (en) 2011-04-25 2018-02-06 Bio-Rad Laboratories, Inc. Methods and compositions for nucleic acid analysis
US11754499B2 (en) 2011-06-02 2023-09-12 Bio-Rad Laboratories, Inc. Enzyme quantification
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
US11898193B2 (en) 2011-07-20 2024-02-13 Bio-Rad Laboratories, Inc. Manipulating droplet size
US8663920B2 (en) 2011-07-29 2014-03-04 Bio-Rad Laboratories, Inc. Library characterization by digital assay
US9222954B2 (en) 2011-09-30 2015-12-29 Becton, Dickinson And Company Unitized reagent strip
USD831843S1 (en) 2011-09-30 2018-10-23 Becton, Dickinson And Company Single piece reagent holder
US9480983B2 (en) 2011-09-30 2016-11-01 Becton, Dickinson And Company Unitized reagent strip
USD905269S1 (en) 2011-09-30 2020-12-15 Becton, Dickinson And Company Single piece reagent holder
USD742027S1 (en) 2011-09-30 2015-10-27 Becton, Dickinson And Company Single piece reagent holder
US10076754B2 (en) 2011-09-30 2018-09-18 Becton, Dickinson And Company Unitized reagent strip
USD1029291S1 (en) 2011-09-30 2024-05-28 Becton, Dickinson And Company Single piece reagent holder
US11453906B2 (en) 2011-11-04 2022-09-27 Handylab, Inc. Multiplexed diagnostic detection apparatus and methods
US9447458B2 (en) 2011-11-16 2016-09-20 Canon U.S. Life Sciences, Inc. Detection of neighboring variants
US10822644B2 (en) 2012-02-03 2020-11-03 Becton, Dickinson And Company External files for distribution of molecular diagnostic tests and determination of compatibility between tests
US9399215B2 (en) 2012-04-13 2016-07-26 Bio-Rad Laboratories, Inc. Sample holder with a well having a wicking promoter
US10946384B2 (en) 2013-09-11 2021-03-16 Osaka University Thermal convection generating chip, thermal convection generating device, and thermal convection generating method
US11901041B2 (en) 2013-10-04 2024-02-13 Bio-Rad Laboratories, Inc. Digital analysis of nucleic acid modification
US11174509B2 (en) 2013-12-12 2021-11-16 Bio-Rad Laboratories, Inc. Distinguishing rare variations in a nucleic acid sequence from a sample
US10957421B2 (en) 2014-12-03 2021-03-23 Syracuse University System and method for inter-species DNA mixture interpretation
US10647981B1 (en) 2015-09-08 2020-05-12 Bio-Rad Laboratories, Inc. Nucleic acid library generation methods and compositions
CN112236512A (zh) * 2018-11-13 2021-01-15 北卡罗来纳-查佩尔山大学 具有微流体装置的分析系统、微流体装置和相关方法
CN115551639A (zh) * 2020-04-21 2022-12-30 赫孚孟拉罗股份公司 具有单分子传感器阵列的高通量核酸定序
US12030050B2 (en) 2020-11-04 2024-07-09 Handylab, Inc. Microfluidic cartridge and method of making same
EP4001433A1 (en) * 2020-11-20 2022-05-25 Université de Paris A method for determining the level of dna integrity
CN114717330B (zh) * 2022-04-13 2023-09-22 中国农业科学院北京畜牧兽医研究所 与绵羊单胎产羔数相关的snp分子标记、引物组、试剂盒及检测方法和应用
CN114717330A (zh) * 2022-04-13 2022-07-08 中国农业科学院北京畜牧兽医研究所 与绵羊单胎产羔数相关的snp分子标记、引物组、试剂盒及检测方法和应用

Also Published As

Publication number Publication date
CA2566698A1 (en) 2005-12-01
US7645581B2 (en) 2010-01-12
US20080085521A1 (en) 2008-04-10
WO2005113148A1 (en) 2005-12-01
AU2005245448A1 (en) 2005-12-01
JP4740237B2 (ja) 2011-08-03
JP2008502325A (ja) 2008-01-31
EP1755785A1 (en) 2007-02-28

Similar Documents

Publication Publication Date Title
US7645581B2 (en) Determining nucleic acid fragmentation status by coincident detection of two labeled probes
US10428377B2 (en) Methods of detecting low copy nucleic acids
US8275554B2 (en) System for differentiating the lengths of nucleic acids of interest in a sample
US10583442B2 (en) Real-time PCR in micro-channels
Cao et al. Advances in digital polymerase chain reaction (dPCR) and its emerging biomedical applications
WO2008005248A2 (en) Real-time pcr in micro-channels
US6524830B2 (en) Microfluidic devices and systems for performing inefficient fast PCR
US20140272996A1 (en) Droplet generator with collection tube
US20160310949A1 (en) Digital pcr systems and methods using digital microfluidics
US20080124726A1 (en) Biochemical analysis of partitioned cells
US20050064465A1 (en) Continuous and non-continuous flow bioreactor

Legal Events

Date Code Title Description
AS Assignment

Owner name: CALIPER LIFE SCIENCES, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:KNAPP, MICHAEL R.;CHOW, ANDREA W.;SPAID, MICHAEL;AND OTHERS;REEL/FRAME:015873/0397;SIGNING DATES FROM 20040903 TO 20040930

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION