US20050145496A1 - Thermal reaction device and method for using the same - Google Patents

Thermal reaction device and method for using the same Download PDF

Info

Publication number
US20050145496A1
US20050145496A1 US10/876,046 US87604604A US2005145496A1 US 20050145496 A1 US20050145496 A1 US 20050145496A1 US 87604604 A US87604604 A US 87604604A US 2005145496 A1 US2005145496 A1 US 2005145496A1
Authority
US
United States
Prior art keywords
channel
reagent
sample
reaction
devices
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
US10/876,046
Inventor
Federico Goodsaid
Marc Unger
Jiang Huang
Emerson Quan
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.)
Standard Biotools Corp
Original Assignee
Individual
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 US10/818,642 external-priority patent/US7666361B2/en
Priority claimed from US10/837,885 external-priority patent/US7476363B2/en
Priority to US10/876,046 priority Critical patent/US20050145496A1/en
Application filed by Individual filed Critical Individual
Assigned to FLUIDIGM CORPORATION reassignment FLUIDIGM CORPORATION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: HUANG, JIANG, GOODSAID, FEDERICO, QUAN, EMERSON, UNGER, MARC
Priority to US11/084,357 priority patent/US7604965B2/en
Priority to EP05766055.7A priority patent/EP1796824A4/en
Priority to CN2005800225837A priority patent/CN1997454B/en
Priority to SG2013006333A priority patent/SG2013006333A/en
Priority to SG200902863-0A priority patent/SG166024A1/en
Priority to CN2010101546166A priority patent/CN101811074B/en
Priority to JP2007511507A priority patent/JP5502275B2/en
Priority to PCT/US2005/015352 priority patent/WO2005107938A2/en
Priority to CN201210270352XA priority patent/CN102778432A/en
Publication of US20050145496A1 publication Critical patent/US20050145496A1/en
Assigned to FLUIDIGM CORPORATION - A DELAWARE CORPORATION reassignment FLUIDIGM CORPORATION - A DELAWARE CORPORATION REINCORPORATION ASSIGNMENT Assignors: FLUIDIGM CORPORATION - A CALIFORNIA CORPORATION
Priority to US11/929,436 priority patent/US7749737B2/en
Priority to US11/929,370 priority patent/US7867454B2/en
Priority to US12/579,347 priority patent/US8247178B2/en
Priority to JP2012020787A priority patent/JP2012085659A/en
Priority to US13/548,068 priority patent/US9150913B2/en
Priority to US14/477,602 priority patent/US20150174575A1/en
Priority to US14/873,958 priority patent/US20160129441A1/en
Priority to US15/805,720 priority patent/US10131934B2/en
Priority to US15/829,665 priority patent/US20180243741A1/en
Priority to US16/150,096 priority patent/US20190169676A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • 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
    • B01L3/5025Containers for the purpose of retaining a material to be analysed, e.g. test tubes with fluid transport, e.g. in multi-compartment structures for parallel transport of multiple samples
    • 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
    • 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
    • B01L3/502707Containers 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 characterised by the manufacture of the container or its components
    • 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
    • B01L3/50273Containers 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 characterised by the means or forces applied to move the fluids
    • 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
    • B01L3/502738Containers 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 characterised by integrated valves
    • 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
    • B01L2200/06Fluid handling related problems
    • B01L2200/0642Filling fluids into wells by specific techniques
    • 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
    • B01L2200/14Process control and prevention of errors
    • B01L2200/142Preventing evaporation
    • 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
    • B01L2200/14Process control and prevention of errors
    • B01L2200/143Quality control, feedback systems
    • B01L2200/147Employing temperature sensors
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01LCHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
    • B01L2300/00Additional constructional details
    • B01L2300/06Auxiliary integrated devices, integrated components
    • B01L2300/0627Sensor or part of a sensor is integrated
    • B01L2300/0636Integrated biosensor, microarrays
    • 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
    • B01L2300/0809Geometry, shape and general structure rectangular shaped
    • B01L2300/0819Microarrays; Biochips
    • 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
    • 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
    • B01L2300/12Specific details about materials
    • B01L2300/123Flexible; Elastomeric
    • 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
    • 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
    • B01L2400/0481Moving fluids with specific forces or mechanical means specific mechanical means and fluid pressure squeezing of channels or chambers
    • 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
    • B01L2400/00Moving or stopping fluids
    • B01L2400/06Valves, specific forms thereof
    • B01L2400/0633Valves, specific forms thereof with moving parts
    • B01L2400/0638Valves, specific forms thereof with moving parts membrane valves, flap valves
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01LCHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
    • B01L2400/00Moving or stopping fluids
    • B01L2400/06Valves, specific forms thereof
    • B01L2400/0633Valves, specific forms thereof with moving parts
    • B01L2400/0655Valves, specific forms thereof with moving parts pinch valves
    • 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
    • B01L3/02Burettes; Pipettes
    • B01L3/0241Drop counters; Drop formers
    • B01L3/0244Drop counters; Drop formers using pins
    • B01L3/0248Prongs, quill pen type dispenser
    • 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
    • B01L3/502715Containers 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 characterised by interfacing components, e.g. fluidic, electrical, optical or mechanical interfaces
    • 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
    • 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]
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N27/00Investigating or analysing materials by the use of electric, electrochemical, or magnetic means
    • G01N27/26Investigating or analysing materials by the use of electric, electrochemical, or magnetic means by investigating electrochemical variables; by using electrolysis or electrophoresis
    • G01N27/416Systems
    • G01N27/447Systems using electrophoresis
    • G01N27/453Cells therefor

Definitions

  • microfluidic devices have the potential to be adapted for use with automated systems, thereby providing the additional benefits of further cost reductions and decreased operator errors because of the reduction in human involvement.
  • Microfluidic devices have been proposed for use in a variety of applications, including, for instance, capillary electrophoresis, gas chromatography and cell separations.
  • microfluidic devices designed for use in conducting a variety of chemical and biochemical analyses. Because of its importance in modern biochemistry, there is a particular need for devices that can be utilized to conduct a variety of nucleic acid amplification reactions, while having sufficient versatility for use in other types of analyses as well.
  • Devices with the ability to conduct nucleic acid amplifications would have diverse utilities. For example, such devices could be used as an analytical tool to determine whether a particular target nucleic acid of interest is present or absent in a sample. Thus, the devices could be utilized to test for the presence of particular pathogens (e.g., viruses, bacteria or fungi), and for identification purposes (e.g., paternity and forensic applications). Such devices could also be utilized to detect or characterize specific nucleic acids previously correlated with particular diseases or genetic disorders. When used as analytical tools, the devices could also be utilized to conduct genotyping analyses and gene expression analyses (e.g., differential gene expression studies).
  • pathogens e.g., viruses, bacteria or fungi
  • identification purposes e.g., paternity and forensic applications
  • Such devices could also be utilized to detect or characterize specific nucleic acids previously correlated with particular diseases or genetic disorders.
  • the devices When used as analytical tools, the devices could also be utilized to conduct genotyping analyses and gene expression analyses (e.g.
  • the devices can be used in a preparative fashion to amplify sufficient nucleic acid for further analysis such as sequencing of amplified product, cell-typing, DNA fingerprinting and the like.
  • Amplified products can also be used in various genetic engineering applications, such as insertion into a vector that can then be used to transform cells for the production of a desired protein product.
  • a variety of devices and methods for conducting microfluidic analyses are provided herein, including devices that can be utilized to conduct thermal cycling reactions such as nucleic acid amplification reactions.
  • the devices differ from conventional microfluidic devices in that they include elastomeric components; in some instances, much or all of the device is composed of elastomeric material.
  • Certain devices are designed to conduct thermal cycling reactions (e.g., PCR) with devices that include one or more elastomeric valves to regulate solution flow through the device.
  • thermal cycling reactions e.g., PCR
  • devices that include one or more elastomeric valves to regulate solution flow through the device.
  • Some of the devices include blind flow channels which include a region that functions as a reaction site. Certain such devices include a flow channel formed within an elastomeric material, and a plurality of blind flow channels in fluid communication with the flow channel, with a region of each blind flow channel defining a reaction site.
  • the devices can also include one or more control channels overlaying and intersecting each of the blind flow channels, wherein an elastomeric membrane separates the one or more control channels from the blind flow channels at each intersection.
  • the elastomeric membrane in such devices is disposed to be deflected into or withdrawn from the blind flow channel in response to an actuation force.
  • the devices can optionally further include a plurality of guard channels formed within the elastomeric material and overlaying the flow channel and/or one or more of the reaction sites.
  • the guard channels are designed to have fluid flow therethrough to reduce evaporation from the flow channels and reaction sites of the device.
  • the devices can optionally include one or more reagents deposited within each of the reaction sites.
  • the flow channel is one of a plurality of flow channels, each of the flow channels in fluid communication with multiple blind flow channels which branch therefrom.
  • the plurality of flow channels are interconnected with one another such that fluid can be introduced into each of the reaction sites via a single inlet.
  • the plurality of flow channels are isolated from each other such that fluid introduced into one flow channel cannot flow to another flow channel, and each flow channel comprises an inlet at one or both ends into which fluid can be introduced.
  • Other devices include an array of reaction sites having a density of at least 50 sites/cm 2 , with the reaction sites typically formed within an elastomeric material. Other devices have even higher densities such as at least 250, 500 or 1000 sites/cm 2 , for example.
  • Still other device include a reaction site formed within an elastomeric substrate, at which a reagent for conducting a reaction is non-covalently immobilized.
  • the reagent can be one or more reagents for conducting essentially any type of reaction.
  • the reagent in some devices includes one reagents for conducting a nucleic acid amplification reaction.
  • the reagent comprises a primer, polymerase and one or more nucleotides.
  • the reagent is a nucleic acid template.
  • a variety of matrix or array-based devices are also provided. Certain of these devices include: (i) a first plurality of flow channels formed in an elastomeric substrate, (ii) a second plurality of flow channels formed in the elastomeric substrate that intersect the first plurality of flow channels to define an array of reaction sites, (iii) a plurality of isolation valves disposed within the first and second plurality of flow channels that can be actuated to isolate solution within each of the reaction sites from solution at other reaction sites, and (iv) a plurality of guard channels overlaying one or more of the flow channels and/or one or more of the reaction sites to prevent evaporation of solution therefrom.
  • the foregoing devices can be utilized to conduct a number of different types of reactions, including those involving temperature regulation (e.g., thermocycling of nucleic acid analyses).
  • Methods conducted with certain blind channel type devices involve providing a microfluidic device that comprises a flow channel formed within an elastomeric material; and a plurality of blind flow channels in fluid communication with the flow channel, with an end region of each blind flow channel defining a reaction site. At least one reagent is introduced into each of the reaction sites, and then a reaction is detected at one or more of the reaction sites.
  • the method can optionally include heating the at least one reagent within the reaction site.
  • a method can involve introducing the components for a nucleic acid amplification reaction and then thermocycling the components to form amplified product.
  • Other methods involve providing a microfluidic device comprising one or more reaction sites, each reaction site comprising a first reagent for conducting an analysis that is non-covalently deposited on an elastomeric substrate.
  • a second reagent is then introduced into the one or more reaction sites, whereby the first and second reagents mix to form a reaction mixture.
  • a reaction between the first and second reagents at one or more of the reaction sites is subsequently detected.
  • Still other methods involve providing a microfluidic device comprising an array of reaction sites formed within a substrate and having a density of at least 50 sites/cm 2 . At least one reagent is introduced into each of the reaction sites. A reaction at one or more of the reaction sites is then detected.
  • Yet other methods involve providing a microfluidic device comprising at least one reaction site which is formed within an elastomeric substrate and a plurality of guard channels also formed within the elastomeric substrate. At least one reagent is introduced into each of the reaction sites and then heated within the reaction sites. A fluid is flowed through the guard channels before or during heating to reduce evaporation from the at least one reaction site. A reaction within the at least one reaction site is subsequently detected.
  • Additional devices designed to reduce evaporation of fluid from the device comprise a cavity that is part of a microfluidic network formed in an elastomeric substrate; and a plurality of guard channels overlaying the cavity and separated from the cavity by an elastomeric membrane.
  • the guard channel in such devices is sized (i) to allow solution flow therethrough, and (ii) such that there is not a substantial reduction in solution flow in, out or through the cavity due to deflection of the membrane(s) upon application of an actuation force to the guard channels.
  • Other such devices include (i) one or more flow channels and/or one or more reaction sites; and (ii) a plurality of guard channels overlaying the microfluidic system and separated therefrom by elastomer, wherein the spacing between guard channels is between 1 ⁇ m to 1 mm. In other devices the spacing is between 5 ⁇ m and 500 ⁇ m, in other devices between 10 ⁇ m and 100 ⁇ m, and in still other devices between 40 ⁇ m and 75 ⁇ m.
  • compositions for conducting nucleic acid analyses in reaction sites of certain microfluidic devices include one or more of the following: an agent that blocks protein binding sites on an elastomeric material and a detergent.
  • the blocking agent is typically selected from the group consisting of a protein (e.g., gelatin or albumin, such as bovine serum albumin (BSA)).
  • the detergent can be SDS or Triton, for example.
  • FIG. 1A is a schematic representation of an exemplary device with a matrix design of intersecting vertical and horizontal flow channels.
  • FIGS. 1 B-E show enlarged views of a portion of the device shown in FIG. 1A and illustrates its operation.
  • FIG. 1F is a schematic representation of another exemplary matrix design device that utilizes guard channels to reduce sample evaporation.
  • FIG. 2 is a plan view of an exemplary blind channel device.
  • FIG. 3A is a plan view of another exemplary blind channel device.
  • FIG. 3B is a schematic representation of a more complex blind channel device based upon the unit of the general design depicted in FIG. 3A .
  • FIG. 3C is an enlarged view of a region of the device shown in FIG. 3B , and illustrates the orientation of the guard flow channels in this particular design.
  • FIG. 4 is a plan view of a device utilizing the hybrid design.
  • FIG. 5 is a chart showing ramp up and down times to conduct a thermocycling reaction.
  • FIG. 6 shows the location of spotted reagents within reaction sites in a blind channel type device illustrating proper alignment of the reagents within reaction sites at the corners of the device.
  • FIGS. 7A and 7B respectively are a cross-sectional view and a schematic diagram of another hybrid type microfluidic device and represents the type of device used to conduct the experiments described in Examples 1-4.
  • FIG. 8 is a bar graph in which the average FAM/PR1/Control ratios are plotted for six different ⁇ -actin TaqMan reactions.
  • the reactions were thermocycled in the micro fluidic device (chip) shown in FIG. 7B (solid bars) and Macro TaqMan reactions (striped bars).
  • the controls are the first and fourth bar sets that have no DNA.
  • the error bars are the standard deviation of the ratios.
  • FIG. 9 is a diagram depicting an exemplary pin spotting process.
  • Reagents are picked up from a source (e.g., a microtiter plate) and then printed by bringing the loaded pin into contact with the substrate.
  • the wash step consists of agitation in deionized water followed by vacuum drying.
  • FIG. 10 is a bar graph depicting FAM signal strength for the microfluidic device (chip) described in Example 1 (see FIG. 7B ) based on the experiments described in Example 2.
  • the data are in the form of (FAM signal/PR1 signal) scaled by the FAM/PR1 ratio for the reference lanes. Error bars are the standard deviation along a lane.
  • the “1.3X” and “1X” designations refer to the concentration of the spotted primers and probes, in relation to their nominal values.
  • FIG. 11 is a bar graph showing average VIC/PF1/Control ratios for 9-10 wells for Macro TaqMan (striped bars), and TaqMan reactions in the microfluidic device (solid bars).
  • Two negative controls (Control) and two samples with 100 pg/nl genomic DNA were thermocycled with reaction components as described above with 4 ⁇ the standard amount of primer/probe.
  • the error bars represent the standard deviation of the average ratios.
  • FIG. 12 is a bar graph that shows FAM/PR1/Control ratios for each of 10-1 nl wells branching from a single flow channel of a microfluidic device (see FIG. 7B ).
  • the amount of genomic DNA was 0.25 pg/nl, which results in an average of one target copy per well.
  • FIG. 13 is a bar graph depicting the average VIC/PR1/Control ratios for CYP2D6 SNP reactions using the microfluidic device shown in FIG. 7B .
  • Allele 1 (Al-1) is the positive control for the VIC probe against the reference or wild type allele CYP2D6*1.
  • Allele 2 (Al-2) is the positive control for the FAM probe against the variant or mutant allele, CYP2D6*3.
  • the control has no DNA template. Genomic DNA was used at either 100 pg/nl or 20 pg/nl.
  • the error bars are the standard deviation of the ratios.
  • FIG. 14 is a bar graph showing the average FAM/PR1/Control ratios for CYP2D6 SNP reactions in the microfluidic device shown in FIG. 7B .
  • the samples are the same as described with respect to FIG. 13 and in Example 3.
  • FIG. 15 is a schematic diagram of the microfluidic device used for the experiments in Example 4.
  • FIG. 16 is a polyacrylamide gel containing PCR product from Macro PCR and PCR reactions in the microfluidic device shown in FIG. 7B .
  • the results on the left show the approximate migration of different DNA base pair lengths.
  • the lanes containing interspersed bands are molecular weight markers.
  • the lanes labeled “Macro” are the PCR products from the Macro reactions at different dilutions.
  • the lanes labeled “In chip” are PCR products generated in the chip.
  • the lanes containing many bands throughout the gel are nonspecific background signals.
  • FIGS. 17 a - 17 d depict two preferred designs of a partitioning microfluidic device in a valve off and valve actuated state.
  • FIGS. 18 a and 18 b depict images of a partitioning microfluidic devices after a thermocycling reaction was performed.
  • FIG. 18 a depicts a two color image
  • FIG. 18 b depicts the remaining signal after subtraction of the control red signal.
  • FIG. 19 depicts a graph of comparing the average number of copies per well to the number of positive wells.
  • FIG. 20 depicts an isothermic amplification scheme—SCORPION
  • FIG. 21 depicts an exemplary matrix microfluidic device plan view.
  • a “flow channel” refers generally to a flow path through which a solution can flow.
  • valve refers to a configuration in which a flow channel and a control channel intersect and are separated by an elastomeric membrane that can be deflected into or retracted from the flow channel in response to an actuation force.
  • blind channel or a “dead-end channel” refers to a flow channel which has an entrance but not a separate exit. Accordingly, solution flow in and out of the blind channel occurs at the same location. The process of filling one or more blind channels is sometimes simply referred to as “blind fill.”
  • an “isolated reaction site” generally refers to a reaction site that is not in fluid communication with other reactions sites present on the device. When used with respect to a blind channel, the isolated reaction site is the region at the end of the blind channel that can be blocked off by a valve associated with the blind channel.
  • a “via” refers to a channel formed in an elastomeric device to provide fluid access between an external port of the device and one or more flow channels.
  • a via can serve as a sample input or output, for example.
  • elastomer and “elastomeric” has its general meaning as used in the art.
  • Allcock et al. Contemporary Polymer Chemistry, 2nd Ed.
  • elastomeric materials exhibit elastic properties because the polymer chains readily undergo torsional motion to permit uncoiling of the backbone chains in response to a force, with the backbone chains recoiling to assume the prior shape in the absence of the force.
  • elastomers deform when force is applied, but then return to their original shape when the force is removed.
  • the elasticity exhibited by elastomeric materials can be characterized by a Young's modulus.
  • the elastomeric materials utilized in the microfluidic devices disclosed herein typically have a Young's modulus of between about 1 Pa-1 TPa, in other instances between about 10 Pa-100 GPa, in still other instances between about 20 Pa-1 GPa, in yet other instances between about 50 Pa-10 MPa, and in certain instances between about 100 Pa-1 MPa. Elastomeric materials having a Young's modulus outside of these ranges can also be utilized depending upon the needs of a particular application.
  • microfluidic devices described herein are fabricated from an elastomeric polymer such as GE RTV 615 (formulation), a vinyl-silane crosslinked (type) silicone elastomer (family).
  • elastomeric polymer such as GE RTV 615 (formulation), a vinyl-silane crosslinked (type) silicone elastomer (family).
  • the present microfluidic systems are not limited to this one formulation, type or even this family of polymer; rather, nearly any elastomeric polymer is suitable. Given the tremendous diversity of polymer chemistries, precursors, synthetic methods, reaction conditions, and potential additives, there are a large number of possible elastomer systems that can be used to make monolithic elastomeric microvalves and pumps.
  • nucleic acid refers to include a polymeric form of nucleotides of any length, including, but not limited to, ribonucleotides or deoxyribonucleotides. There is no intended distinction in length between these terms. Further, these terms refer only to the primary structure of the molecule. Thus, in certain embodiments these terms can include triple-, double- and single-stranded DNA, as well as triple-, double- and single-stranded RNA. They also include modifications, such as by methylation and/or by capping, and unmodified forms of the polynucleotide.
  • nucleic acid examples include polydeoxyribonucleotides (containing 2-deoxy-D-ribose), polyribonucleotides (containing D-ribose), any other type of polynucleotide which is an N— or C-glycoside of a purine or pyrimidine base, and other polymers containing nonnucleotidic backbones, for example, polyamide (e.g., peptide nucleic acids (PNAs)) and polymorpholino (commercially available from the Anti-Virals, Inc., Corvallis, Oreg., as Neugene) polymers, and other synthetic sequence-specific nucleic acid polymers providing that the polymers contain nucleobases in a configuration which allows for base pairing and base stacking, such as is found in DNA and RNA.
  • PNAs peptide nucleic acids
  • a “probe” is an nucleic acid capable of binding to a target nucleic acid of complementary sequence through one or more types of chemical bonds, usually through complementary base pairing, usually through hydrogen bond formation, thus forming a duplex structure.
  • the probe binds or hybridizes to a “probe binding site.”
  • the probe can be labeled with a detectable label to permit facile detection of the probe, particularly once the probe has hybridized to its complementary target.
  • the label attached to the probe can include any of a variety of different labels known in the art that can be detected by chemical or physical means, for example.
  • Suitable labels that can be attached to probes include, but are not limited to, radioisotopes, fluorophores, chromophores, mass labels, electron dense particles, magnetic particles, spin labels, molecules that emit chemiluminescence, electrochemically active molecules, enzymes, cofactors, and enzyme substrates.
  • Probes can vary significantly in size. Some probes are relatively short. Generally, probes are at least 7 to 15 nucleotides in length. Other probes are at least 20, 30 or 40 nucleotides long. Still other probes are somewhat longer, being at least 50, 60, 70, 80, 90 nucleotides long. Yet other probes are longer still, and are at least 100, 150, 200 or more nucleotides long. Probes can be of any specific length that falls within the foregoing ranges as well.
  • a “primer” is a single-stranded polynucleotide capable of acting as a point of initiation of template-directed DNA synthesis under appropriate conditions (i.e., in the presence of four different nucleoside triphosphates and an agent for polymerization, such as, DNA or RNA polymerase or reverse transcriptase) in an appropriate buffer and at a suitable temperature.
  • the appropriate length of a primer depends on the intended use of the primer but typically is at least 7 nucleotides long and, more typically range from 10 to 30 nucleotides in length. Other primers can be somewhat longer such as 30 to 50 nucleotides long. Short primer molecules generally require cooler temperatures to form sufficiently stable hybrid complexes with the template.
  • a primer need not reflect the exact sequence of the template but must be sufficiently complementary to hybridize with a template.
  • primer site or “primer binding site” refers to the segment of the target DNA to which a primer hybridizes.
  • primer pair means a set of primers including a 5′ “upstream primer” that hybridizes with the complement of the 5′ end of the DNA sequence to be amplified and a 3′ “downstream primer” that hybridizes with the 3′ end of the sequence to be amplified.
  • a primer that is “perfectly complementary” has a sequence fully complementary across the entire length of the primer and has no mismatches.
  • the primer is typically perfectly complementary to a portion (subsequence) of a target sequence.
  • a “mismatch” refers to a site at which the nucleotide in the primer and the nucleotide in the target nucleic acid with which it is aligned are not complementary.
  • substantially complementary when used in reference to a primer means that a primer is not perfectly complementary to its target sequence; instead, the primer is only sufficiently complementary to hybridize selectively to its respective strand at the desired primer-binding site.
  • nucleic acid is identical to, or hybridizes selectively to, another nucleic acid molecule.
  • Selectivity of hybridization exists when hybridization occurs that is more selective than total lack of specificity.
  • selective hybridization will occur when there is at least about 55% identity over a stretch of at least 14-25 nucleotides, preferably at least 65%, more preferably at least 75%, and most preferably at least 90%.
  • one nucleic acid hybridizes specifically to the other nucleic acid. See M. Kanehisa, Nucleic Acids Res. 12: 203 (1984).
  • label refers to a molecule or an aspect of a molecule that can be detected by physical, chemical, electromagnetic and other related analytical techniques.
  • detectable labels include, but are not limited to, radioisotopes, fluorophores, chromophores, mass labels, electron dense particles, magnetic particles, spin labels, molecules that emit chemiluminescence, electrochemically active molecules, enzymes, cofactors, enzymes linked to nucleic acid probes and enzyme substrates.
  • detectably labeled means that an agent has been conjugated with a label or that an agent has some inherent characteristic (e.g., size, shape or color) that allows it to be detected without having to be conjugated to a separate label.
  • a “polymorphic marker” or “polymorphic site” is the locus at which divergence occurs. Preferred markers have at least two alleles, each occurring at frequency of greater than 1%, and more preferably greater than 10% or 20% of a selected population. A polymorphic locus may be as small as one base pair. Polymorphic markers include restriction fragment length polymorphisms, variable number of tandem repeats (VNTR's), hypervariable regions, minisatellites, dinucleotide repeats, trinucleotide repeats, tetranucleotide repeats, simple sequence repeats, and insertion elements such as Alu. The first identified allelic form is arbitrarily designated as the reference form and other allelic forms are designated as alternative or variant alleles.
  • allelic form occurring most frequently in a selected population is sometimes referred to as the wildtype form. Diploid organisms may be homozygous or heterozygous for allelic forms.
  • a diallelic polymorphism has two forms.
  • a triallelic polymorphism has three forms.
  • a “single nucleotide polymorphism” occurs at a polymorphic site occupied by a single nucleotide, which is the site of variation between allelic sequences. The site is usually preceded by and followed by highly conserved sequences of the allele (e.g., sequences that vary in less than 1/100 or 1/11000 members of the populations).
  • a single nucleotide polymorphism usually arises due to substitution of one nucleotide for another at the polymorphic site.
  • a transition is the replacement of one purine by another purine or one pyrimidine by another pyrimidine.
  • a transversion is the replacement of a purine by a pyrimidine or vice versa.
  • Single nucleotide polymorphisms can also arise from a deletion of a nucleotide or an insertion of a nucleotide relative to a reference allele.
  • a “reagent” refers broadly to any agent used in a reaction.
  • a reagent can include a single agent which itself can be monitored (e.g., a substance that is monitored as it is heated) or a mixture of two or more agents.
  • a reagent may be living (e.g., a cell) or non-living.
  • Exemplary reagents for a nucleic acid amplification reaction include, but are not limited to, buffer, metal ions, polymerase, primers, template nucleic acid, nucleotides, labels, dyes, nucleases and the like.
  • Reagents for enzyme reactions include, for example, substrates, cofactors, coupling enzymes, buffer, metal ions, inhibitors and activators.
  • Reagents for cell-based reactions include, but are not limited to, cells, cell specific dyes and ligands (e.g., agonists and antagonists) that bind to cellular receptors.
  • a “ligand” is any molecule for which there exists another molecule (i.e., an “antiligand”) that specifically or non-specifically binds to the ligand, owing to recognition of some portion of the ligand by the antiligand.
  • microfluidic devices also sometimes referred to as chips
  • the devices are designed for use in analyses requiring temperature control, especially analyses involving thermocycling (e.g., nucleic acid amplification reactions).
  • the microfluidic devices incorporate certain design features that: give the devices a significantly smaller footprint than many conventional microfluidic devices, enable the devices to be readily integrated with other instrumentation and provide for automated analysis.
  • blind channel or “blind fill” are characterized in part by having a plurality of blind channels, which, as indicated in the definition section, are flow channels having a dead end or isolated end such that solution can only enter and exit the blind channel at one end (i.e., there is not a separate inlet and outlet for the blind channel).
  • blind channels which, as indicated in the definition section, are flow channels having a dead end or isolated end such that solution can only enter and exit the blind channel at one end (i.e., there is not a separate inlet and outlet for the blind channel).
  • These devices require only a single valve for each blind channel to isolate a region of the blind channel to form an isolated reaction site.
  • one or more reagents for conducting an analysis are deposited at the reaction sites, thereby resulting in a significant reduction in the number of input and outputs.
  • the blind channels are connected to an interconnected network of channels such that all the reaction sites can be filled from a single, or limited number, of sample inputs. Because of the reduction in complexity in inputs and outputs and the use of only a single valve to isolate each reaction site, the space available for reaction sites is increased. Thus, the features of these devices means that each device can include a large number of reaction sites (e.g., up to tens of thousands) and can achieve high reaction site densities (e.g., over 1,000-4,000 reaction sites/cm 2 ). Individually and collectively, these features also directly translate into a significant reduction in the size of these devices compared to traditional microfluidic devices.
  • microfluidic devices that are disclosed herein utilize a matrix design.
  • microfluidic devices of this type utilize a plurality of intersecting horizontal and vertical flow channels to define an array of reaction sites at the points of intersection.
  • devices of this design also have an array or reaction sites; however, there is a larger number of sample inputs and corresponding outputs to accommodate the larger number of samples with this design.
  • a valve system referred to as a switchable flow array architecture enables solution be flowed selectively through just the horizontal or flow channels, thus allowing for switchable isolation of various flow channels in the matrix.
  • the matrix devices are constructed to analyze a large number of sample under a limited number of conditions. Still other devices are hybrids of these two general design types.
  • microfluidic devices that are described are further characterized in part by utilizing various components such as flow channels, control channels, valves and/or pumps from elastomeric materials.
  • various components such as flow channels, control channels, valves and/or pumps from elastomeric materials.
  • essentially the entire device is made of elastomeric material. Consequently, such devices differ significantly in form and function from the majority of conventional microfluidic devices that are formed from silicon-based material.
  • the design of the devices enables them to be utilized in combination with a number of different heating systems.
  • the devices are useful in conducting diverse analyses that require temperature control.
  • those microfluidic devices for use in heating applications can incorporate a further design feature to minimize evaporation of sample from the reaction sites.
  • Devices of this type in general include a number of guard channels formed within the elastomeric device through which water can be flowed to increase the water vapor pressure within the elastomeric material from which the device is formed, thereby reducing evaporation of sample from the reaction sites.
  • a temperature cycling device may be used to control the temperature of the microfluidic devices.
  • the microfluidic device would be adapted to make thermal contact with the microfluidic device.
  • a window may be formed in a region of the carrier or slide such that the microfluidic device, preferably a device having an elastomeric block, may directly contact the heating/cooling block of the temperature cycling device.
  • the heating/cooling block has grooves therein in communication with a vacuum source for applying a suction force to the microfluidic device, preferably the portion wherein the reactions are taking place.
  • a rigid thermally conductive plate may be bonded to the microfluidic device that then mates with the heating and cooling block for efficient thermal conduction resulting.
  • the array format of certain of the devices means the devices can achieve high throughput.
  • the high throughput and temperature control capabilities make the devices useful for performing large numbers of nucleic acid amplifications (e.g., polymerase chain reaction—PCR).
  • PCR polymerase chain reaction
  • Such reactions will be discussed at length herein as illustrative of the utility of the devices, especially of their use in any reaction requiring temperature control.
  • the devices can be utilized in a wide variety of other types of analyses or reactions. Examples include analyses of protein-ligand interactions and interactions between cells and various compounds. Further examples are provided infra.
  • microfluidic devices disclosed herein are typically constructed at least in part from elastomeric materials and constructed by single and multilayer soft lithography (MSL) techniques and/or sacrificial-layer encapsulation methods (see, e.g., Unger et al. (2000) Science 288: 113-116, and PCT Publication WO 01/01025, both of which are incorporated by reference herein in their entirety for all purposes).
  • MSL soft lithography
  • sacrificial-layer encapsulation methods see, e.g., Unger et al. (2000) Science 288: 113-116, and PCT Publication WO 01/01025, both of which are incorporated by reference herein in their entirety for all purposes.
  • microfluidic devices can be designed in which solution flow through flow channels of the device is controlled, at least in part, with one or more control channels that are separated from the flow channel by an elastomeric membrane or segment.
  • This membrane or segment can be deflected into or retracted from the flow channel with which a control channel is associated by applying an actuation force to the control channels.
  • solution flow can be slowed or entirely blocked through the flow channel.
  • the devices provided herein incorporate such pumps and valves to isolate selectively a reaction site at which reagents are allowed to react.
  • the reaction sites can be located at any of a number of different locations within the device. For example, in some matrix-type devices, the reaction site is located at the intersection of a set of flow channels. In blind channel devices, the reaction site is located at the end of the blind channel.
  • the elastomeric device is typically fixed to a support (e.g., a glass slide).
  • a support e.g., a glass slide.
  • the resulting structure can then be placed on a temperature control plate, for example, to control the temperature at the various reaction sites.
  • the device can be placed on any of a number of thermocycling plates.
  • the devices are made of elastomeric materials that are relatively optically transparent, reactions can be readily monitored using a variety of different detection systems at essentially any location on the microfluidic device. Most typically, however, detection occurs at the reaction site itself (e.g., within a region that includes an intersection of flow channels or at the blind end of a flow channel).
  • detection occurs at the reaction site itself (e.g., within a region that includes an intersection of flow channels or at the blind end of a flow channel).
  • the fact that the device is manufactured from substantially transparent materials also means that certain detection systems can be utilized with the current devices that are not usable with traditional silicon-based microfluidic devices. Detection can be achieved using detectors that are incorporated into the device or that are separate from the device but aligned with the region of the device to be detected.
  • a plurality of guard channels can be formed in the devices.
  • the guard channels are similar to the control channels in that typically they are formed in a layer of elastomer that overlays the flow channels and/or reaction site.
  • the guard channels are separated from the underlying flow channels and/or reaction sites by a membrane or segment of elastomeric material.
  • the guard channels are considerably smaller in cross-sectional area. In general, a membrane with smaller area will deflect less than a membrane with larger area under the same applied pressure.
  • the guard channels are designed to be pressurized to allow solution (typically water) to be flowed into the guard channel.
  • solution typically water
  • Water vapor originating from the guard channel can diffuse into the pores of the elastomer adjacent a flow channel or reaction site, thus increasing the water vapor concentration adjacent the flow channel or reaction site and reducing evaporation of solution therefrom.
  • the guard channels are sufficiently small such that when pressurized the membrane that separates the guard channel from the underlying flow channel or reaction site does not substantially restrict solution flow in, out, or through the flow channel or reaction site which the guard channel overlays.
  • substantially restrict or other similar terms means that solution flow is not reduced in, out or through the flow channel or reaction site by more than 40%, typically less than 30%, usually less than 20%, and in some instances less than 10%, as compared to solution flow in, to or through the flow channel or reaction site under the same conditions, when the guard channel is not pressurized to achieve solution flow therethrough.
  • the guard channels have a cross-sectional area of between 100 ⁇ m 2 and 50,000 ⁇ m 2 , or any integral or non-integral cross-sectional area therebetween.
  • the cross-sectional area is less than 50,000 ⁇ m 2 , in other instances less than 10,000 ⁇ m 2 , in still other instances less than 10,00 ⁇ m 2 , and in yet other instances less than 100 ⁇ m 2 .
  • the guard channels can have any of a variety of shapes including, but not limited to, circular, elliptical, square, rectangular, hexagonal and octahedral shapes.
  • the guard channels are designed to reduce the evaporation of sample and reagents from the device during the time and under the conditions that it takes to conduct a thermocycling reaction to less than 50%, in other instance less than 45%, 40%, 35%, 30%, 25%, 20%, 15%, 10%, 5% or 1%. Thus, for example, a typical PCR reaction involving 40 cycles can be conducted within 120 minutes.
  • the guard channel system is designed to reduce evaporation during approximately this time frame to the foregoing set of limits. To achieve this level of evaporation reduction, the guard channels are typically present at a density of at least 10 lines/cm 2 to 1000 lines/cm 2 , or any integral density level therebetween.
  • the guard channels are generally at least 25 lines/cm 2 , in other instances at least 50 lines/cm 2 , in still other instances at least 100 lines/cm 2 , and in yet other instances at least 500 lines/cm 2 .
  • the guard channels are typically present at a spacing between 1 mm to 1 ⁇ m as measured from the outer edge of one line to the nearest outer edge of adjacent line, or any integral density level therebetween. More specifically, the guard channels are generally spaced between 500 ⁇ m to 5 ⁇ m, in other instances between 100 ⁇ m to 10 ⁇ m, in still other instances between 75 ⁇ m to 40 ⁇ m.
  • the spacing is typically at least 1 ⁇ m, but is less than 1 mm, in other instances less than 500 ⁇ m, in still other instances less than 400 ⁇ m, in yet other instances less than 300 ⁇ m, in other instances less than 200 ⁇ m, and in still other instances less than 100 ⁇ m, 50 ⁇ m or 25 ⁇ m.
  • the guard channels can be formed as a separate network of channels or can be smaller channels that branch off of the control channels.
  • the guard channels can extend across the device or only a particular region or regions of the device.
  • the guard channels are placed adjacent and over flow channels and reaction sites as these are the primary locations at which evaporation is the primary concern. Exemplary locations of guard channels on certain matrix devices are illustrated in FIG. 1C , and on certain blind channel devices in FIGS. 3B and 3C , and discussed in greater detail infra.
  • the solution flowed through the guard channel includes any substance that can reduce evaporation of water.
  • the substance can be one that increases the water vapor concentration adjacent a flow line and/or reaction site, or one that while not increasing the water vapor concentration nonetheless blocks evaporation of water from the flow line and/or reaction site (blocking agent).
  • blocking agent include, but are not limited to, water and buffered solution (e.g., TaqMan buffer solution, and phosphate buffered saline).
  • suitable blocking agents include, for example, mineral oil.
  • Guard channels are typically formed in the elastomer utilizing the MSL techniques and/or sacrificial-layer encapsulation methods cited above.
  • Devices utilizing the matrix design generally have a plurality of vertical and horizontal flow channel that intersect to form an array of junctions. Because a different sample and reagent (or set of reagents) can be introduced into each of the flow channels, a large number of samples can be tested against a relatively large number of reaction conditions in a high throughput format. Thus, for example, if a different sample is introduced into each of M different vertical flow channels and a different reagent (or set of reagents) is introduced into each of N horizontal flow channels, then M ⁇ N different reactions can be conducted at the same time.
  • matrix devices include valves that allow for switchable isolation of the vertical and horizontal flow channels.
  • valves are positioned to allow selective flow just through the vertical flow channels or just through the horizontal flow channels. Because devices of this type allow flexibility with respect to the selection of the type and number of samples, as well as the number and type of reagents, these devices are well-suited for conducting analyses in which one wants to screen a large number of samples against a relatively large number of reaction conditions.
  • the matrix devices can optionally incorporate guard channels to help prevent evaporation of sample and reactants.
  • FIG. 21 depicts an exemplary matrix design wherein a first elastomeric layer (1st layer) and a second elastomeric layer (2d layer) each having fluid channels formed therein.
  • a reagent fluid channel in the first layer is connected to a reagent fluid channel in the second layer through a via, while the second layer also has sample channels therein, the sample channels and the reagent channels terminating in sample and reagent chambers, respectively.
  • the sample and reagent chambers are in fluid communication with each other through an interface channel that has an interface valve associated therewith to control fluid communication between each of the chambers of a reaction cell.
  • the interface is first closed, then reagent is introduced into the reagent channel from the reagent inlet and sample is introduced into the sample channel through the sample inlet, containment valves are then closed to isolate each reaction cell from other reaction cells. Once the reaction cells are isolated, the interface valve is opened to cause the sample chamber and the reagent chamber to be in fluid communication with each other so that a desired reaction may take place.
  • a preferred aspect of the invention provides for a microfluidic device for reacting M number of different samples with N number of different reagents comprising: a plurality of reaction cells, each reaction cell comprising a sample chamber and a reagent chamber, the sample chamber and the reagent chamber being in fluid communication through an interface channel having an interface valve associated therewith for controlling fluid communication between the sample chamber and the reagent chamber; a plurality of sample inlets each in fluid communication with the sample chambers; a plurality of reagent inlets each in fluid communication with the reagent chambers; wherein one of the sample inlets or reagent inlets is in fluid communication with one of the sample chambers or one of the reagent chambers, respectively, through a via.
  • Certain embodiments include having the reaction cells be formed within an elastomeric block formed from a plurality of layers bonded together and the interface valve is deflectable membrane; having the sample inlets be in communication with the sample chamber through a sample channel and the reagent inlet is in fluid communication with the reagent chamber through a reagent channel, a portion of the sample channel and a portion of the reagent channel being oriented about parallel to each other and each having a containment valve associated therewith for controlling fluid communication therethrough; having the valve associated with the sample channel and the valve associated with the reagent channel are in operable communication with each other through a common containment control channel; having the containment common control channel located along a line about normal to one of the sample channel or the reagent channel
  • Another aspect of the invention provides for a method for making a feature in an elastomeric block comprising the steps of: providing a first elastomeric layer; applying a photoresist layer upon a surface of the first elastomeric layer; applying a light pattern to the photoresist layer to form a pattern of reacted photoresist material; removing unreacted photoresist material leaving the pattern of reacted photoresist upon the surface of the first elastomeric layer; applying an etching reagent to the first elastomeric surface to etch the surface of the first elastomeric layer not covered by the pattern of reacted photoresist material thereby removing regions of the first elastomeric layer not covered by the pattern of reacted photoresist and leaving a pattern of the elastomeric layer corresponding to the pattern of reacted photoresist material.
  • the method include having a step of removing the pattern of reacted photoresist material; having the removing is caused by applying an adhesive tape to the surface of the elastomeric layer and the pattern of reacted photoresist material, then separating the adhesive tape from the elastomeric layer while some or all of the pattern of reacted photoresist material is removed from the surface of the elastomeric layer; having the photo resist be SU8; having the etching reagent comprises tetrabutylammoniumfluoride-trihydrate; having the feature be a via; having the elastomeric block comprise a plurality of elastomeric layers bonded together, wherein two or more elastomeric layers have recesses formed therein and one recess of one elastomeric layer is in communication with a recess of another elastomeric layer through the via.
  • microfluidic devices of the present invention may be further integrated into the carrier devices described in copending and co-owned U.S. patent application Ser. No. 60/557,715 by Unger filed on Mar. 29, 2004, which is herein incorporated for all purposes.
  • the carrier of Unger provides on-board continuous fluid pressure to maintain valve closure away from a source of fluid pressure, e.g., house air pressure.
  • Unger further provides for an automated system for charging and actuating the valves of the present invention as described therein.
  • FIG. 1A provides an illustration of one exemplary matrix device.
  • This device 100 comprises seven vertical flow channels 102 and seven horizontal flow channels 104 that intersect to form an array of 49 different intersections or reaction sites 106 .
  • This particular device thus enables seven samples to be reacted with seven different reagents or sets of reagents.
  • Column valves 110 that regulate solution flow in the vertical direction can be controlled by control channels 118 that can all be actuated at a single inlet 114 .
  • row valves 108 regulate solution flow in the horizontal direction; these are controlled by control channels 116 that are actuated by a single control inlet 112 .
  • the control channels 116 that regulate the row valves 108 vary in width depending upon location. When a control channel 116 crosses a vertical flow channel 102 , the control channel 116 is sufficiently narrow that when it is actuated it does not deflect into the vertical flow channel 102 to reduce substantially solution flow therethrough. However, the width of the control channel 116 is increased when it overlays one of the horizontal flow channels 104 ; this makes the membrane of the control channel sufficiently large to block solution flow through the horizontal flow channel 104 .
  • reagents R 1 -R 7 are introduced into their respective horizontal flow channels 104 and samples S 1 -S 7 are injected into their respective vertical flow channels 102 .
  • the reagents in each horizontal flow channel 104 thus mix with the samples in each of the vertical flow channels 102 at the intersections 106 , which in this particular device are in the shape of a well or chamber.
  • the reagents necessary for the amplification reaction are introduced into each of the horizontal flow channels 104 .
  • Different nucleic acid templates are introduced into the vertical flow channels 102 .
  • the primer introduced as part of the reagent mixture that is introduced into each of the horizontal flow channels 104 might differ between flow channels. This allows each nucleic acid template to be reacted with a number of different primers.
  • FIGS. 1 B-E show enlarged plan views of adjacent reaction sites in the device depicted in FIG. 1A to illustrate more specifically how the device operates during an analysis.
  • the intersections 106 are not shown in the form of reaction wells and control channels 116 , 118 have been omitted, with just the column and row valves 110 , 108 being shown (rectangular boxes).
  • FIG. 1B an analysis is commenced by closing column valves 110 and opening row valves 108 to allow solution flow through horizontal flow channel 104 while blocking flow through vertical flow channels 102 .
  • Reagent R 1 is then introduced into horizontal flow channel 104 and flowed completely through the length of the horizontal flow channel 104 such that all the reaction sites 106 are filled.
  • Solution flow through horizontal channel 104 can be achieved by an external pump, but more typically is achieved by incorporating a peristaltic pump into the elastomeric device itself as described in detail in Unger et al. (2000) Science 288: 113-116, and PCT Publication WO 01/01025, for example.
  • row valves 108 are closed and column valves 102 opened (see FIG. 1C ). This allows samples S 1 and S 2 to be introduced into vertical flow channels 102 and to flow through their respective flow channels. As the samples flow through the vertical flow channels 102 , they expel R 1 from the reaction sites 106 , thus leaving sample at reaction sites 106 . Then, as shown in FIG. 1D , row valves 108 are opened to allow S 1 and S 2 to diffuse and mix with R 1 . Thus, a mixture of sample and reactant (R 1 S 1 and R 1 S 2 ) is obtained in the region of each intersection or reaction site 106 .
  • all row and column valves 108 , 110 are closed to isolate S 1 and S 2 within the region of their respective reaction sites 106 and to prevent intermixing of S 1 and S 2 (see FIG. 1E ).
  • the mixtures are then allowed to react and the reactions detected by monitoring the intersection 106 or the cross-shaped region that includes the intersection 106 .
  • the device is placed on a heater and heated while the samples remain isolated.
  • FIG. 1F A modified version of the device shown in FIG. 1A is shown in FIG. 1F .
  • the general structure bears many similarities with that depicted in FIG. 1A , and common elements in both figures share the same reference numbers.
  • the device 150 illustrated in FIG. 1F differs in that pairs of horizontal flow channels 104 are joined to a common inlet 124 . This essentially enables duplicate sets of reagents to be introduced into two adjacent flow channels with just a single injection into inlet 124 .
  • the use of a common inlet is further extended with respect to the vertical flow channels 102 . In this particular example, each sample is introduced into five vertical flow channels 102 with a single injection into sample inlet 120 .
  • the device shown in FIG. 1F also includes a separate control channel inlet 128 that regulates control channel 130 that can be used to govern solution flow toward outlets 132 and another control channel inlet 132 that regulates control channel 134 that regulates solution flow to outlets 136 .
  • device 150 incorporates guard channels 138 .
  • the guard channels 138 are formed as part of control channels 116 .
  • the guard channels 138 are smaller than the row valves 108 ; consequently, the membranes of the guard channels 138 are not deflected into the underlying horizontal flow channels 104 such that solution flow is disrupted.
  • the devices include one or more flow channels from which one or more blind channels branch. As indicated above, the end region of such channels can serve as a reaction site. A valve formed by an overlaying flow channel can be actuated to isolate the reaction site at the end of the blind channel. The valves provide a mechanism for switchably isolating the reaction sites.
  • the flow channel network in communication with the blind channels is configured such that all or most of the reaction sites can be filled with a single or a limited number of inlets (e.g., less than 5 or less than 10).
  • a limited number of inlets e.g., less than 5 or less than 10.
  • the ability to fill a blind flow channel is made possible because the devices are made from elastomeric material.
  • the elastomeric material is sufficiently porous such that air within the flow channels and blind channels can escape through these pores as solution is introduced into the channels.
  • the lack of porosity of materials utilized in other microfluidic devices precludes use of the blind channel design because air in a blind channel has no way to escape as solution is injected.
  • a third characteristic is that one or more reagents are non-covalently deposited on a base layer of elastomer during manufacture (see infra for further details on the fabrication process) within the reaction sites.
  • the reagent(s) are non-covalently attached because the reagents are designed to become dissolved when sample is introduced into the reaction site.
  • a different reactant or set of reactants is deposited at each of the different reaction sites.
  • Certain blind channel devices are designed such that the reaction sites are arranged in the form of an array.
  • one or more of the reagents required for conducting the extension reaction are deposited at each of the reaction sites during manufacture of the device.
  • reagents include, for example, all or some of the following: primers, polymerase, nucleotides, cofactors, metal ions, buffers, intercalating dyes and the like.
  • primers selected to amplify different regions of DNA are deposited at each reaction site. Consequently, when a nucleic acid template is introduced into the reaction sites via inlet, a large number of extension reactions can be performed at different segments of the template.
  • Thermocycling necessary for an amplification reaction can be accomplished by placing the device on a thermocycling plate and cycling the device between the various required temperatures.
  • the reagents can be immobilized in a variety of ways. For example, in some instances one or more of the reagents are non-covalently deposited at the reaction site, whereas in other instances one or more of the reagents is covalently attached to the substrate at the reaction site. If covalently attached, the reagents can be linked to the substrate via a linker.
  • linker types can be utilized such as photochemical/photolabile linkers, themolabile linkers, and linkers that can be cleaved enzymatically.
  • linkers are bifunctional (i.e., the linker contains a functional group at each end that is reactive with groups located on the element to which the linker is to be attached); the functional groups at each end can be the same or different.
  • suitable linkers that can be used in some assays include straight or branched-chain carbon linkers, heterocyclic linkers and peptide linkers.
  • a variety of types of linkers are available from Pierce Chemical Company in Rockford, Ill. and are described in EPA 188,256; U.S. Pat. Nos. 4,671,958; 4,659,839; 4,414,148; 4,669,784; 4,680,338, 4,569, 789 and 4,589,071, and by Eggenweiler, H.
  • NVOC (6 nitroveratryloxycarbonyl) linkers and other NVOC-related linkers are examples of suitable photochemical linkers (see, e.g., WO 90/15070 and WO 92/10092). Peptides that have protease cleavage sites are discussed, for example, in U.S. Pat. No. 5,382,513.
  • FIG. 2 is a simplified plan view of one exemplary device utilizing the blind channel design.
  • the device 200 includes a flow channel 204 and a set of branch flow channels 206 branching therefrom that are formed in an elastomeric substrate 202 .
  • Each branch flow channel 206 terminates in a reaction site 208 , thereby forming an array of reaction sites.
  • Overlaying the branch flow channels 206 is a control channel 210 that is separated from the branch flow channels 206 by membranes 212 . Actuation of control channel 210 causes membranes 212 to deflect into the branch flow channels 206 (i.e., to function as a valve), thus enabling each of the reaction sites 208 to be isolated from the other reaction sites.
  • Operation of such a device involves injecting a test sample into flow channel 204 with solution subsequently flowing into each of branch channels 206 .
  • control channel 210 is actuated to cause activation of valves/membranes 212 to deflect into branch channels 206 , thereby sealing off each of reaction sites 208 .
  • the sample flows into and remains in reaction sites 208 , it dissolves reagents previously spotted at each of the reaction sites 208 . Once dissolved, the reagents can react with the sample. Valves 212 prevent the dissolved reagents at each reaction site 208 from intermixing by diffusion. Reaction between sample and reagents are then detected, typically within reaction site 208 . Reactions can optionally be heated as described in the temperature control section infra.
  • FIG. 3A illustrates an example of a somewhat more complex blind flow channel design.
  • each of a set of horizontal flow channels 304 are connected at their ends to two vertical flow channels 302 .
  • a plurality of branch flow channels 306 extend from each of the horizontal flow channels 304 .
  • the branch flow channels 304 in this particular design are interleaved such that the branch channel 306 attached to any given horizontal flow channel 304 is positioned between two branch channels 306 joined to an immediately adjacent horizontal flow channel 304 , or positioned between a branch flow channel 306 joined to an immediately adjacent flow channel 304 and one of the vertical flow channels 302 .
  • each branch flow channel 306 terminates in a reaction site 308 .
  • a control channel 310 overlays each of the branch channels and is separated from the underlying branch channel by membrane 312 .
  • the control channel is actuated at port 316 .
  • the vertical and horizontal flow channels 302 , 304 are interconnected such that injection of sample into inlet 314 allows solution to flow throughout the horizontal and vertical flow channel network and ultimately into each of the reaction sites 308 via the branch flow channels 306 .
  • sample is injected into inlet to introduce solution into each of the reaction sites.
  • valves/membranes are actuated to trap solution within the reaction sites by pressurizing the control channels at port.
  • Reagents previously deposited in the reaction sites become resuspended within the reaction sites, thereby allowing reaction between the deposited reagents and sample within each reaction site.
  • Reactions within the reaction sites are monitored by a detector. Again, reactions can optionally be controllably heated according to the methods set forth in the temperature control section below.
  • FIG. 3B An even more complicated version of the general design illustrated in FIG. 3A is shown in FIG. 3B .
  • the device shown in FIG. 3B is one in which the unit organization of the horizontal and branch flow channels 302 shown in FIG. 3A is repeated multiple times.
  • the device shown in FIG. 3B further illustrates the inclusion of guard channels 320 in those devices to be utilized in applications that involve heating (e.g., thermocycling).
  • An exemplary orientation of the guard channels 320 with respect to the flow channels 304 and branch channels 306 is shown in the enlarged view depicted in FIG. 3C .
  • the guard channels 320 overlay the branch flow channels 306 and reaction sites 308 .
  • water is flowed through the guard channels 320 during heating of the device 300 to increase the local concentration of water in the device, thereby reducing evaporation of water from solution in the flow channels 306 and reaction sites 308 .
  • blind channel devices minimizes the footprint of the device and enable a large number of reaction sites to be formed on the device and for high densities to be obtained.
  • devices of this type having 2500 reaction sites can readily be manufactured to fit on a standard microscope slides (25 mm ⁇ 75 mm).
  • the foregoing features also enable very high densities of reaction sites to be obtained with devices utilizing the blind channel design. For example, densities of at least 50, 60, 70, 80, 90 or 100 reaction sites/cm 2 or any integral density value therebetween can be readily obtained. However, certain devices have even higher densities ranging, for example, between 100 to 4000 reaction sites/cm 2 , or any integral density value therebetween.
  • some devices have densities of at least 100, 150, 200, 250, 300, 400, 500, 600, 700, 800, 900 or 1000 sites/cm 2 .
  • Devices with very high densities of at least, 2000, 3000, or 4000 sites/cm 2 are also obtainable.
  • Such high densities directly translate into a very large number of reaction sites on the device.
  • Devices utilizing the blind channel architecture typically have at least 10-100 reaction sites, or any integral number of sites therebetween. More typically, the devices have at least 100-1,000 reaction sites, or any integral number of sites therebetween. Higher density devices can have even more reaction sites, such as at least 1,000-100,000 reaction sites, or any integral number of sites therebetween.
  • certain devices have at least 100; 500; 1,000; 2,000; 3,000; 4,000; 5,000; 6,000; 7,000; 8,000; 9,000; 10,000; 20,000; 30,000; 40,000; 50,000; or 100,000 reaction sites depending upon the overall size of the device.
  • the large number of reaction sites and densities that can be obtained is also a consequence of the ability to fabricate very small wells or cavities.
  • the cavities or wells typically have a volume of less than 50 nL; in other instances less than 40 nL, 30 nL, 20 nL or 10 nL; and in still other instances less than 5 nL or 1 nL.
  • certain devices have wells that are 300 microns long, 300 microns wide and 10 microns deep.
  • the blind channel devices provided herein can utilize certain design features and methodologies discussed in PCT Applications PCT/US01/44549 (published as WO 02/43615) and PCT/US02/10875 (published as WO 02/082047), including, for example, strategies for filling dead-ended channels, liquid priming, pressurized outgas priming, as well as various strategies for displacing gas during the filling of microfluidic channels. Both of these PCT publications are incorporated herein by reference in their entirety for all purposes.
  • Still other devices are hybrids of the matrix and blind fill designs.
  • the design of devices of this type is similar to the blind channel device shown in FIG. 3A , except that each horizontal flow channel is connected to its own sample inlet port(s) and the horizontal flow channels are not interconnected via vertical flow channels. Consequently, sample introduced into any given horizontal flow channel fills only that horizontal flow channel and reaction sites attached thereto.
  • sample can flow between the horizontal flow channels 304 via vertical flow channels 302 .
  • Device 400 comprises a plurality of horizontal flow channels 404 , each of which has a plurality of branch flow channels 406 extending from it and its own sample inlet 414 .
  • a control channel 410 overlays each of the branch flow channels 406 and membrane (valve) 412 separates the control channel 410 from the underlying branch flow channel 406 .
  • actuation of the control channel at inlet 416 causes deflection of membranes 412 into the branch flow channels 406 and isolation of reaction sites 408 .
  • each horizontal flow channel 404 can include an inlet 414 at each end, thus allowing sample to be introduced from both ends.
  • reaction mixtures can be prepared prior to injection on the chip. Once the mixtures are injected, they can be analyzed or further treated (e.g., heated).
  • each sample By injecting different samples into each of the horizontal flow channels, a large number of samples can be rapidly analyzed. Assuming reagents have been previously deposited at the reaction sites, the presence of the same reagent at each reaction site associated with any given horizontal flow channel provides a facile way to conduct a number of replicate reactions with each sample. If instead, the reagent at the reaction sites differ for any given flow channel, then each sample is essentially simultaneously exposed to a variety of different reaction conditions.
  • the devices provided herein are tailored for a variety of different types of investigations. If an investigation involves screening of a relatively large number of different samples under user controlled conditions (e.g., 100 samples against 100 user selected reagents), then the matrix devices provide a useful solution. If, however, the investigation involves analyzing one or a limited number of samples under a wide variety of reaction conditions (e.g., one sample against 10,000 reaction conditions), then the blind channel design is useful. Finally, if one wants to examine a relatively large number of samples against defined reaction conditions without having to inject reagents (e.g., 100 samples against 100 previously defined reagents), then the hybrid devices are useful.
  • reagents e.g., 100 samples against 100 previously defined reagents
  • temperature controller is meant broadly to refer to a device or element that can regulate temperature of the entire microfluidic device or within a portion of the microfluidic device (e.g., within a particular temperature region or at one or more junctions in a matrix of blind channel-type microfluidic device).
  • the devices are placed on a thermal cycling plate to thermal cycle the device.
  • a thermal cycling plate to thermal cycle the device.
  • a variety of such plates are readily available from commercial sources, including for example the ThermoHybaid Px2 (Franklin, Mass.), MJ Research PTC-200 (South San Francisco, Calif.), Eppendorf Part# E5331 (Westbury, N.Y.), Techne Part# 205330 (Princeton, N.J.).
  • thermocouple One structure for detecting temperature is a thermocouple.
  • a thermocouple could be created as thin film wires patterned on the underlying substrate material, or as wires incorporated directly into the microfabricated elastomer material itself.
  • Temperature can also be sensed through a change in electrical resistance.
  • change in resistance of a thermistor fabricated on an underlying semiconductor substrate utilizing conventional techniques can be calibrated to a given temperature change.
  • a thermistor could be inserted directly into the microfabricated elastomer material.
  • Still another approach to detection of temperature by resistance is described in Wu et al. in “MEMS Flow Sensors for Nano-fluidic Applications”, Sensors and Actuators A 89 152-158 (2001), which is hereby incorporated by reference in its entirety.
  • This paper describes the use of doped polysilicon structures to both control and sense temperature.
  • the temperature coefficient of resistance can be precisely controlled by the identity and amount of dopant, thereby optimizing performance of the sensor for a given application.
  • Thermo-chromatic materials are another type of structure available to detect temperature on regions of an amplification device. Specifically, certain materials dramatically and reproducibly change color as they pass through different temperatures. Such a material could be added to the solution as they pass through different temperatures. Thermo-chromatic materials could be formed on the underlying substrate or incorporated within the elastomer material. Alternatively, thermo-chromatic materials could be added to the sample solution in the form of particles.
  • Another approach to detecting temperature is through the use of an infrared camera.
  • An infrared camera in conjunction with a microscope could be utilized to determine the temperature profile of the entire amplification structure. Permeability of the elastomer material to radiation would facilitate this analysis.
  • pyroelectric sensors Specifically, some crystalline materials, particularly those materials also exhibiting piezoelectric behavior, exhibit the pyroelectric effect. This effect describes the phenomena by which the polarization of the material's crystal lattice, and hence the voltage across the material, is highly dependent upon temperature. Such materials could be incorporated onto the substrate or elastomer and utilized to detect temperature.
  • blind channel devices have a base layer onto which reagents are placed.
  • the structure comprising the two layers containing the flow channels and control channels is overlayed on the base layer such that the flow channels are aligned with the deposited reagents.
  • the other side of the base layer is then placed upon a substrate (e.g., glass).
  • the reaction site at which reaction occurs is about 100-150 microns above the substrate/glass interface.
  • FIG. 5 illustrates the rapidity at which the desired temperature is achieved using a blind channel device.
  • temperature may be measured by using double stranded oligonucleotide polymers having known tms wherein an intercollating dye whose intercollation indicates the whether the oligonucleotide is hybridized or denatured, such as SYBR GreenTM or ethidium bromide for example, wherein by introducing a solution containing the oligonucleotide with the dye into the chambers of the microfluidic device having an array of reaction chambers can be used to determine the extent to which the temperature of each chamber is consistent across the array.
  • an intercollating dye whose intercollation indicates the whether the oligonucleotide is hybridized or denatured
  • the intercalating dye changes its relation to the oligonucleotide upon it sdenataturation into a single stranded oligonucleotide.
  • the temperature is above the tm and is lowered, an the intercollation of the dye into the now annealed oligonucleotide may be monitored.
  • the use of the dye in essence provides for an “oligonucleotide thermometer” which changes a property, such as fluorescence, in response to a temperature change relative to the tm of the oligonucleotide.
  • a number of different detection strategies can be utilized with the microfluidic devices that are provided herein. Selection of the appropriate system is informed in part on the type of event and/or agent being detected.
  • the detectors can be designed to detect a number of different signal types including, but not limited to, signals from radioisotopes, fluorophores, chromophores, electron dense particles, magnetic particles, spin labels, molecules that emit chemiluminescence, electrochemically active molecules, enzymes, cofactors, enzymes linked to nucleic acid probes and enzyme substrates.
  • Illustrative detection methodologies suitable for use with the present microfluidic devices include, but are not limited to, light scattering, multichannel fluorescence detection, UV and visible wavelength absorption, luminescence, differential reflectivity, and confocal laser scanning. Additional detection methods that can be used in certain application include scintillation proximity assay techniques, radiochemical detection, fluorescence polarization, fluorescence correlation spectroscopy (FCS), time-resolved energy transfer (TRET), fluorescence resonance energy transfer (FRET) and variations such as bioluminescence resonance energy transfer (BRET). Additional detection options include electrical resistance, resistivity, impedance, and voltage sensing.
  • Detection occurs at a “detection section,” or “detection region.” These terms and other related terms refer to the portion of the microfluidic device at which detection occurs. As indicated above, with devices utilizing the blind channel design, the detection section is generally the reaction site as isolated by the valve associated with each reaction site. The detection section for matrix-based devices is usually within regions of flow channels that are adjacent an intersection, the intersection itself, or a region that encompasses the intersection and a surrounding region.
  • the detection section can be in communication with one or more microscopes, diodes, light stimulating devices (e.g., lasers), photomultiplier tubes, processors and combinations of the foregoing, which cooperate to detect a signal associated with a particular event and/or agent.
  • the signal being detected is an optical signal that is detected in the detection section by an optical detector.
  • the optical detector can include one or more photodiodes (e.g., avalanche photodiodes), a fiber-optic light guide leading, for example, to a photomultiplier tube, a microscope, and/or a video camera (e.g., a CCD camera).
  • Detectors can be microfabricated within the microfluidic device, or can be a separate element. If the detector exists as a separate element and the microfluidic device includes a plurality of detection sections, detection can occur within a single detection section at any given moment.
  • scanning systems can be used. For instance, certain automated systems scan the light source relative to the microfluidic device; other systems scan the emitted light over a detector, or include a multichannel detector.
  • the microfluidic device can be attached to a translatable stage and scanned under a microscope objective. A signal so acquired is then routed to a processor for signal interpretation and processing. Arrays of photomultiplier tubes can also be utilized. Additionally, optical systems that have the capability of collecting signals from all the different detection sections simultaneously while determining the signal from each section can be utilized.
  • External detectors are usable because the devices that are provided are completely or largely manufactured of materials that are optically transparent at the wavelength being monitored. This feature enables the devices described herein to utilize a number of optical detection systems that are not possible with conventional silicon-based microfluidic devices.
  • a particularly preferred detector uses a CCD camera and an optical path that provides for a large field of view and a high numerical aperture to maximize the amount of light collected from each reaction chamber.
  • the CCD is used as an array of photodetectors wherein each pixel or group of pixels corresponds to a reaction chamber rather than being used to produce an image of the array.
  • the optics may be altered such that image quality is reduced or defocused to increase the depth of field of the optical system to collect more light from each reaction chamber.
  • a detector can include a light source for stimulating a reporter that generates a detectable signal.
  • the type of light source utilized depends in part on the nature of the reporter being activated. Suitable light sources include, but are not limited to, lasers, laser diodes and high intensity lamps. If a laser is utilized, the laser can be utilized to scan across a set of detection sections or a single detection section. Laser diodes can be microfabricated into the microfluidic device itself. Alternatively, laser diodes can be fabricated into another device that is placed adjacent to the microfluidic device being utilized to conduct a thermal cycling reaction such that the laser light from the diode is directed into the detection section.
  • the detector can also include, for example, a temperature sensor, a conductivity sensor, a potentiometric sensor (e.g., pH electrode) and/or an amperometric sensor (e.g., to monitor oxidation and reduction reactions).
  • a temperature sensor e.g., a thermocouple
  • a conductivity sensor e.g., a thermocouple
  • a potentiometric sensor e.g., pH electrode
  • an amperometric sensor e.g., to monitor oxidation and reduction reactions.
  • a number of commercially-available external detectors can be utilized. Many of these are fluorescent detectors because of the ease in preparing fluorescently labeled reagents. Specific examples of detectors that are available include, but are not limited to, Applied Precision ArrayWoRx (Applied Precision, Issaquah, Wash.)).
  • intercalation dyes that only fluoresce upon binding to double-stranded DNA can be used to detect double-stranded amplified DNA.
  • suitable dyes include, but are not limited to, SYBRTM and Pico Green (from Molecular Probes, Inc. of Eugene, Oreg.), ethidium bromide, propidium iodide, chromomycin, acridine orange, Hoechst 33258, Toto-1, Yoyo-1, and DAPI (4′,6-diamidino-2-phenylindole hydrochloride). Additional discussion regarding the use of intercalation dyes is provided by Zhu et al., Anal. Chem. 66: 1941-1948 (1994), which is incorporated by reference in its entirety.
  • Detection methods of this type involve detecting a change in fluorescence from a donor (reporter) and/or acceptor (quencher) fluorophore in a donor/acceptor fluorophore pair.
  • the donor and acceptor fluorophore pair are selected such that the emission spectrum of the donor overlaps the excitation spectrum of the acceptor.
  • energy transfer from the donor to the acceptor can occur. This energy transfer can be detected.
  • FRET and template extension reactions FRET and template extension reactions. These methods generally utilize a primer labeled with one member of a donor/acceptor pair and a nucleotide labeled with the other member of the donor/acceptor pair. Prior to incorporation of the labeled nucleotide into the primer during an template-dependent extension reaction, the donor and acceptor are spaced far enough apart that energy transfer cannot occur. However, if the labeled nucleotide is incorporated into the primer and the spacing is sufficiently close, then energy transfer occurs and can be detected. These methods are particularly useful in conducting single base pair extension reactions in the detection of single nucleotide polymorphisms (see infra) and are described in U.S. Pat. No. 5,945,283 and PCT Publication WO 97/22719.
  • Quantitative RT-PCR A variety of so-called “real time amplification” methods or “real time quantitative PCR” methods can also be utilized to determine the quantity of a target nucleic acid present in a sample by measuring the amount of amplification product formed during or after the amplification process itself.
  • Fluorogenic nuclease assays are one specific example of a real time quantitation method which can be used successfully with the devices described herein. This method of monitoring the formation of amplification product involves the continuous measurement of PCR product accumulation using a dual-labeled fluorogenic oligonucleotide probe—an approach frequently referred to in the literature as the “TaqMan” method.
  • the probe used in such assays is typically a short (ca. 20-25 bases) polynucleotide that is labeled with two different fluorescent dyes.
  • the 5′ terminus of the probe is typically attached to a reporter dye and the 3′ terminus is attached to a quenching dye, although the dyes can be attached at other locations on the probe as well.
  • the probe is designed to have at least substantial sequence complementarity with the probe binding site on the target nucleic acid. Upstream and downstream PCR primers that bind to regions that flank the probe binding site are also included in the reaction mixture.
  • the probe When the probe is intact, energy transfer between the two fluorophors occurs and the quencher quenches emission from the reporter.
  • the probe is cleaved by the 5′ nuclease activity of a nucleic acid polymerase such as Taq polymerase, thereby releasing the reporter from the polynucleotide-quencher and resulting in an increase of reporter emission intensity which can be measured by an appropriate detector.
  • a nucleic acid polymerase such as Taq polymerase
  • One detector which is specifically adapted for measuring fluorescence emissions such as those created during a fluorogenic assay is the ABI 7700 manufactured by Applied Biosystems, Inc. in Foster City, Calif.
  • Computer software provided with the instrument is capable of recording the fluorescence intensity of reporter and quencher over the course of the amplification. These recorded values can then be used to calculate the increase in normalized reporter emission intensity on a continuous basis and ultimately quantify the amount of the mRNA being amplified.
  • Intercalation dyes such as described above can also be utilized in a different approach to quantitative PCR methods. As noted above, these dyes preferentially bind to double stranded DNA (e.g., SYBR GREEN) and only generate signal once bound. Thus, as an amplification reaction progresses, an increasing amount of dye becomes bound and is accompanied by a concomitant increase in signal that can be detected.
  • SYBR GREEN double stranded DNA
  • Molecular Beacons With molecular beacons, a change in conformation of the probe as it hybridizes to a complementary region of the amplified product results in the formation of a detectable signal.
  • the probe itself includes two sections: one section at the 5′ end and the other section at the 3′ end. These sections flank the section of the probe that anneals to the probe binding site and are complementary to one another. One end section is typically attached to a reporter dye and the other end section is usually attached to a quencher dye.
  • the two end sections can hybridize with each other to form a hairpin loop.
  • the reporter and quencher dye are in sufficiently close proximity that fluorescence from the reporter dye is effectively quenched by the quencher dye.
  • Hybridized probe results in a linearized conformation in which the extent of quenching is decreased.
  • Probes of this type and methods of their use is described further, for example, by Piatek, A. S., et al., Nat. Biotechnol. 16: 359-63 (1998); Tyagi, S. and Kramer, F. R., Nature Biotechnology 14: 303-308 (1996); and Tyagi, S. et al., Nat. Biotechnol. 16: 49-53 (1998), each of which is incorporated by reference herein in their entirety for all purposes.
  • Invader assays are used for SNP genotyping and utilize an oligonucleotide, designated the signal probe, that is complementary to the target nucleic acid (DNA or RNA) or polymorphism site.
  • a second oligonucleotide, designated the Invader Oligo contains the same 5′ nucleotide sequence, but the 3′ nucleotide sequence contains a nucleotide polymorphism.
  • the Invader Oligo interferes with the binding of the signal probe to the target nucleic acid such that the 5′ end of the signal probe forms a “flap” at the nucleotide containing the polymorphism.
  • Cleavase enzyme cleaves the 5′ flap of the nucleotides. The released flap binds with a third probe bearing FRET labels, thereby forming another duplex structure recognized by the Cleavase enzyme. This time the Cleavase enzyme cleaves a fluorophore away from a quencher and produces a fluorescent signal.
  • the signal probe will be designed to hybridize with either the reference (wild type) allele or the variant (mutant) allele.
  • NASBA Nucleic Acid Sequence Based Amplification
  • a primer complementary to the RNA contains the sequence for the T7 promoter site. This primer is allowed to bind with the template RNA and Reverse Transcriptase (RT) added to generate the complementary strand from 3′ to 5′. RNase H is subsequently added to digest away the RNA, leaving single stranded cDNA behind. A second copy of the primer can then bind the single stranded cDNA and make double stranded cDNA.
  • T7 RNA polymerase is added to generate many copies of the RNA from the T7 promoter site that was incorporated into the cDNA sequence by the first primer. All the enzymes mentioned are capable of functioning at 41° C. (See, e.g., Compton, J. Nucleic Acid Sequence-based Amplification, Nature 350: 91-91, 1991.)
  • FIG. 20 depicts the scheme thereof, wherein Scorpion probing mechanism is as follows.
  • Step 1 initial denaturation of target and Scorpion stem sequence.
  • Step 2 annealing of Scorpion primer to target.
  • Step 3 extension of Scorpion primer produces double-stranded DNA.
  • Step 4 denaturation of double-stranded DNA produced in step 3. This gives a single-stranded target molecule with the Scorpion primer attached.
  • Step 5 on cooling, the Scorpion probe sequence binds to its target in an intramolecular manner.
  • a Scorpion (as shown in FIG. 24 ) consists of a specific probe sequence that is held in a hairpin loop configuration by complementary stem sequences on the 5′ and 3′ sides of the probe. The fluorophore attached to the 5′-end is quenched by a moiety (normally methyl red) joined to the 3′-end of the loop. The hairpin loop is linked to the 5′-end of a primer via a PCR stopping sequence (stopper). After extension of the primer during PCR amplification, the specific probe sequence is able to bind to its complement within the same strand of DNA.
  • This hybridization event opens the hairpin loop so that fluorescence is no longer quenched and an increase in signal is observed.
  • the PCR stoping sequence prevents read-through, that could lead to opening of the hairpin loop in the absence of the specific target sequence. Such read-through would lead to the detection of non-specific PCR products, e.g. primer dimers or mispriming events.
  • nucleic acids within the flow channel e.g., the substantially circular flow channel of FIG. 1 or the reaction chambers of FIG. 2
  • solution containing amplified product is withdrawn and then subjected to the electric field.
  • Reactions conducted with the microfluidic devices disclosed herein are typically conducted with certain additives to enhance the reactions. So, for example, in the case of devices in which reagents are deposited, these additives can be spotted with one or more reactants at a reaction site, for instance.
  • One set of additives are blocking reagents that block protein binding sites on the elastomeric substrate.
  • a wide variety of such compounds can be utilized including a number of different proteins (e.g., gelatin and various albumin proteins, such as bovine serum albumin) and glycerol.
  • a detergent additive can also be useful. Any of a number of different detergents can be utilized. Examples include, but are not limited to SDS and the various Triton detergents.
  • additives that promote the amplification reaction.
  • enhancers include, but are not limited to, reagents that reduce secondary structure in the nucleic acid (e.g., betaine), and agents that reduce mispriming events (e.g., tetramethylammonium chloride).
  • exemplary polymerases that can be utilized include, but are not limited to, rTH polymerase XL, which is a combination of Thermus thermophilus (Tth) and Thermococcus litoralis (Tli), hyperthermo-philic archaebacterium Pyrosoccus woesei (Pwo), and Tgo DNA Polymerase.
  • rTH polymerase XL is a combination of Thermus thermophilus (Tth) and Thermococcus litoralis (Tli), hyperthermo-philic archaebacterium Pyrosoccus woesei (Pwo), and Tgo DNA Polymerase.
  • Example 1 infra Further details regarding additives useful in conducting reactions with certain of the devices disclosed herein, including nucleic acid amplification reactions, are provided in Example 1 infra.
  • the microfluidic devices provided herein can be manufactured to include a large number of reaction sites, the devices are useful in a wide variety of screening and analytical methods.
  • the devices can be utilized to detect reactions between species that react to form a detectable signal, or a product that upon interaction with another species generates a detectable signal.
  • the devices can also be utilized in a number of different types of analyses or reactions requiring temperature control.
  • amplification reactions can be linear amplifications, (amplifications with a single primer), as well as exponential amplifications (i.e., amplifications conducted with a forward and reverse primer set).
  • the reagents that are typically deposited within the reaction sites are those reagents necessary to perform the desired type of amplification reaction. Usually this means that some or all of the following are deposited, primers, polymerase, nucleotides, metal ions, buffer, and cofactors, for example.
  • the sample introduced into the reaction site in such cases is the nucleic acid template.
  • the template can be deposited and the amplification reagents flowed into the reaction sites.
  • samples containing nucleic acid template are flowed through the vertical flow channels and the amplification reagents through the horizontal flow channels or vice versa.
  • PCR is perhaps the best known amplification technique.
  • the devices are not limited to conducting PCR amplifications.
  • Other types of amplification reactions that can be conducted include, but are not limited to, (i) ligase chain reaction (LCR) (see Wu and Wallace, Genomics 4: 560 (1989) and Landegren et al., Science 241: 1077 (1988)); (ii) transcription amplification (see Kwoh et al., Proc. Natl. Acad. Sci. USA 86: 1173 (1989)); (iii) self-sustained sequence replication (see Guatelli et al., Proc. Nat. Acad. Sci.
  • LCR ligase chain reaction
  • transcription amplification see Kwoh et al., Proc. Natl. Acad. Sci. USA 86: 1173 (1989)
  • self-sustained sequence replication see Guatelli et al., Proc. Nat. Acad. Sci.
  • NASBA nucleic acid based sequence amplification
  • Detection of the resulting amplified product can be accomplished using any of the detection methods described supra for detecting amplified DNA.
  • Genotyping involves the determination of whether a diploid organism (i.e., an organism with two copies of each gene) contains two copies of a reference allele (a reference-type homozygote), one copy each of the reference and a variant allele (i.e., a heterozygote), or contains two copies of the variant allele (i.e., a variant-type homozygote).
  • a diploid organism i.e., an organism with two copies of each gene
  • a variant allele i.e., a heterozygote
  • the methods of the invention can be utilized to interrogate a single variant site.
  • the methods can also be used to determine the genotype of an individual in many different DNA loci, either on the same gene, different genes or combinations thereof.
  • Devices to be utilized for conducting genotyping analyses are designed to utilize reaction sites of appropriate size to ensure from a statistical standpoint that a copy of each of the two alleles for a diploid subject are present in the reaction site at a workable DNA concentrations. Otherwise, an analysis could yield results suggesting that a heterozygote is a homozygote simply because a copy of the second allele is not present at the reaction site.
  • Table 2 below indicates the number of copies of the genome present in a 1 nl reaction volume at various exemplary DNA concentrations that can be utilized with the devices described herein. TABLE 2 Number of genome copies present in a 1 nL volume at the indicated DNA concentration. Volume (nL) [DNA] (ug/uL) N 1 0.33 100 1 0.10 32 1 0.05 16 1 0.01 3 1 0.003 1
  • Genotyping analyses using certain devices are typically conducted with samples having a DNA concentration of approximately 0.10 ug/uL, although the current inventors have run successful TaqMan reactions at concentrations in which there is a single genome per reaction site.
  • Genotyping analyses can be conducted using a variety of different approaches. In these methods, it is generally sufficient to obtain a “yes” or “no” result, i.e., detection need only be able to answer the question whether a given allele is present. Thus, analyses can be conducted only with the primers or nucleotides necessary to detect the presence of one allele potentially at a polymorphic site. However, more typically, primers and nucleotides to detect the presence of each allele potentially at the polymorphic site are included. Examples of suitable approaches follow.
  • SBPE Single Base Pair Extension Reactions.
  • SBPE reactions are one technique specifically developed for conducting genotyping analyses.
  • SPBE assays a number of SPBE assays have been developed, the general approach is quite similar. Typically, these assays involve hybridizing a primer that is complementary to a target nucleic acid such that the 3′ end of the primer is immediately 5′ of the variant site or is adjacent thereto. Extension is conducted in the presence of one or more labeled non-extendible nucleotides that are complementary to the nucleotide(s) that occupy the variant site and a polymerase.
  • the non-extendible nucleotide is a nucleotide analog that prevents further extension by the polymerase once incorporated into the primer.
  • a labeled non-extendible nucleotide is incorporated onto the 3′ end of the primer to generate a labeled extension product.
  • extended primers provide an indication of which nucleotide is present at the variant site of a target nucleic acid.
  • Detection of the extended products can be detected utilizing the FRET detection approach described for extension reactions in the detection section supra.
  • a reagent mixture containing a primer labeled with one member of a donor/acceptor fluorophore, one to four labeled non-extendible nucleotides (differentially labeled if more than one non-extendible nucleotide is included), and polymerase are introduced (or previously spotted) at a reaction site.
  • a sample containing template DNA is then introduced into the reaction site to allow template extension to occur.
  • Any extension product formed is detected by the formation of a FRET signal (see, e.g., U.S. Pat. No. 5,945,283 and PCT Publication WO 97/22719.).
  • the reactions can optionally be thermocycled to increase signal using the temperature control methods and apparatus described above.
  • the allele to which a probe is complementary is present in the target DNA, then amplification occurs, thereby resulting in a detectable signal as described in the detection above. Based upon which of the differential signal is obtained, the identity of the nucleotide at the polymorphic site can be determined. If both signals are detected, then both alleles are present. Thermocycling during the reaction is performed as described in the temperature control section supra.
  • Gene expression analysis involves determining the level at which one or more genes is expressed in a particular cell. The determination can be qualitative, but generally is quantitative.
  • the levels of the gene(s) in one cell e.g., a test cell
  • the expression levels of the same genes in another cell control cell.
  • a wide variety of such comparisons can be made. Examples include, but are not limited to, a comparison between healthy and diseased cells, between cells from an individual treated with one drug and cells from another untreated individual, between cells exposed to a particular toxicant and cells not exposed, and so on. Genes whose expression levels vary between the test and control cells can serve as markers and/or targets for therapy.
  • genes can serve as markers of the disease and can potentially be utilized as the basis for diagnostic tests. These genes could also be targets.
  • a strategy for treating the disease might include procedures that result in a reduction of expression of the up-regulated genes.
  • the design of the devices disclosed herein is helpful in facilitating a variety of gene expression analyses. Because the devices contain a large number of reaction sites, a large number of genes and/or samples can be tested at the same time. Using the blind flow channel devices, for instance, the expression levels of hundreds or thousands of genes can be determined at the same time.
  • the devices also facilitate differential gene expression analyses. With the matrix design, for example, a sample obtained from a healthy cell can be tested in one flow channel, with a sample from a diseased cell run in an immediately adjacent channel. This feature enhances the ease of detection and the accuracy of the results because the two samples are run on the same device at the same time and under the same conditions.
  • nucleic acid sample comprising mRNA transcript(s) of the gene(s) or gene fragments, or nucleic acids derived from the mRNA transcript(s) is obtained.
  • a nucleic acid derived from an mRNA transcript refers to a nucleic acid for whose synthesis the mRNA transcript or a subsequence thereof has ultimately served as a template.
  • a cDNA reverse transcribed from an mRNA, an RNA transcribed from that cDNA, a DNA amplified from the cDNA, an RNA transcribed from the amplified DNA are all derived from the mRNA transcript and detection of such derived products is indicative of the presence and/or abundance of the original transcript in a sample.
  • suitable samples include, but are not limited to, mRNA transcripts of the gene or genes, cDNA reverse transcribed from the mRNA, cRNA transcribed from the cDNA, DNA amplified from the genes, RNA transcribed from amplified DNA.
  • a nucleic acid sample is the total mRNA isolated from a biological sample; in other instances, the nucleic acid sample is the total RNA from a biological sample.
  • biological sample refers to a sample obtained from an organism or from components of an organism, such as cells, biological tissues and fluids. In some methods, the sample is from a human patient. Such samples include sputum, blood, blood cells (e.g., white cells), tissue or fine needle biopsy samples, urine, peritoneal fluid, and fleural fluid, or cells therefrom. Biological samples can also include sections of tissues such as frozen sections taken for histological purposes. Often two samples are provided for purposes of comparison. The samples can be, for example, from different cell or tissue types, from different individuals or from the same original sample subjected to two different treatments (e.g., drug-treated and control).
  • RNA isolation technique that does not select against the isolation of mRNA can be utilized for the purification of such RNA samples.
  • methods of isolation and purification of nucleic acids are described in detail in WO 97/10365, WO 97/27317, Chapter 3 of Laboratory Techniques in Biochemistry and Molecular Biology: Hybridization With Nucleic Acid Probes, Part 1. Theory and Nucleic Acid Preparation, (P. Tijssen, ed.) Elsevier, N.Y. (1993); Chapter 3 of Laboratory Techniques in Biochemistry and Molecular Biology: Hybridization With Nucleic Acid Probes, Part 1. Theory and Nucleic Acid Preparation, (P. Tijssen, ed.) Elsevier, N.Y.
  • a further calculation demonstrates that the certain of the devices provided herein which utilize a 1 nanoliter reaction site contain sufficient DNA to achieve accurate expression results.
  • a typical mRNA preparation procedure yields approximately 10 ug of mRNA. It has been demonstrated that typically there are 1 to 10,000 copies of each mRNA per cell. Of the mRNAs that are expressed within any given cell, approximately the four most common messages comprise about 13% of the total mRNA levels. Thus, such highly expressed messages comprise 1.3 ug of mRNA (each is 4 ⁇ 10 ⁇ 12 mole or approximately 2.4 ⁇ 10 12 copies).
  • rare messages are expected to be present at a level of about 2 ⁇ 10 ⁇ 8 copies. If in a standard analysis the mRNA sample is dissolved in 10 ul, the concentration of a rare message is approximately 2 ⁇ 10 7 copies/ul; this concentration corresponds to 20,000 copies per 1 nl well (or 4 ⁇ 10 11 M).
  • the reagents that are introduced (or previously spotted) in the reaction sites can include one or all of the following: primer, labeled probe, nucleotides and polymerase. If an intercalation dye is utilized, the reagent mixture typically includes one or all of the following: primer, nucleotides, polymerase, and intercalation dye.
  • the array-based devices described herein are inherently designed to conduct a large number of amplification reactions at the same time. This feature, however, can readily be further expanded upon by conducting multiple analyses (e.g., genotyping and expression analyses) within each reaction site.
  • Multiplex amplifications can even be performed within a single reaction site by, for example, utilizing a plurality of primers, each specific for a particular target nucleic acid of interest, during the thermal cycling process.
  • the presence of the different amplified products can be detected using differentially labeled probes to conduct a quantitative RT-PCR reaction or by using differentially labeled molecular beacons (see supra).
  • each differentially labeled probes is designed to hybridize only to a particular amplified target.
  • Fluorescence Spectroscopy (Pesce et al., Eds.) Marcel Dekker, New York, (1971); White et al., Fluorescence Analysis: A Practical Approach , Marcel Dekker, New York, (1970); Berlman, Handbook of Fluorescence Spectra of Aromatic Molecules, 2 nd ed., Academic Press, New York, (1971); Griffiths, Colour and Constitution of Organic Molecules , Academic Press, New York, (1976); Indicators (Bishop, Ed.). Pergamon Press, Oxford, 19723; and Haugland, Handbook of Fluorescent Probes and Research Chemicals , Molecular Probes, Eugene (1992).
  • genotyping and expression analyses can optionally be conducted at each reaction site. If quantitative PCR methods such as TaqMan is utilized, then primers for amplifying different regions of a target DNA of interest are included within a single reaction site. Differentially labeled probes for each region are utilized to distinguish product that is formed.
  • quantitative PCR methods such as TaqMan
  • the devices can also be utilized in a number of other applications as well. As indicated earlier, the devices can be utilized to analyze essentially any interaction between two or more species that generates a detectable signal or a reaction product that can reacted with a detection reagent that generates a signal upon interaction with the reaction product.
  • the devices can be utilized in a number of screening applications to identify test agents that have a particular desired activity.
  • the devices can be utilized to screen compounds for activity as a substrate or inhibitor of one or more enzymes.
  • test compound and other necessary enzymatic assay reagents e.g., buffer, metal ions, cofactors and substrates
  • test compound and other necessary enzymatic assay reagents are introduced (if not previously deposited) in the reaction site.
  • the enzyme sample is then introduced and reaction (if the test compound is a substrate) or inhibition of the reaction (if the test compound is an inhibitor) is detected.
  • Such reactions or inhibition can be accomplished by standard techniques, such as directly or indirectly monitoring the loss of substrate and/or appearance of product.
  • Devices with sufficiently large flow channels and reaction sites can also be utilized to conduct cellular assays to detect interaction between a cell and one or more reagents. For instance, certain analyses involve determination of whether a particular cell type is present in a sample. One example for accomplishing this is to utilize cell-specific dyes that preferentially reaction with certain cell types. Thus, such dyes can be introduced into the reaction sites and then cells added. Staining of cells can be detected using standard microscopic techniques. As another illustration, test compounds can be screened for ability to trigger or inhibit a cellular response, such as a signal transduction pathway. In such an analysis, test compound is introduced into a site and then the cell added. The reaction site is then checked to detect formation of the cellular response.
  • the microfluidic devices that are provided are generally constructed utilizing single and multilayer soft lithography (MSL) techniques and/or sacrificial-layer encapsulation methods.
  • MSL soft lithography
  • the basic MSL approach involves casting a series of elastomeric layers on a micro-machined mold, removing the layers from the mold and then fusing the layers together.
  • sacrificial-layer encapsulation approach patterns of photoresist are deposited wherever a channel is desired.
  • the foregoing fabrication methods initially involve fabricating mother molds for top layers (e.g., the elastomeric layer with the control channels) and bottom layers (e.g., the elastomeric layer with the flow channels) on silicon wafers by photolithography with photoresist (Shipley SJR 5740).
  • Channel heights can be controlled precisely by the spin coating rate.
  • Photoresist channels are formed by exposing the photoresist to UV light followed by development. Heat reflow process and protection treatment is typically achieved as described by M. A. Unger, H.-P. Chou, T. Throsen, A. Scherer and S. R. Quake, Science (2000) 288: 113, which is incorporated herein by reference in its entirety.
  • a mixed two-part-silicone elastomer (GE RTV 615) is then spun into the bottom mold and poured onto the top mold, respectively.
  • spin coating can be utilized to control the thickness of bottom polymeric fluid layer.
  • the partially cured top layer is peeled off from its mold after baking in the oven at 80° C. for 25 minutes, aligned and assembled with the bottom layer.
  • a 1.5-hour final bake at 80° C. is used to bind these two layers irreversibly.
  • this RTV device is typically treated with HCL (0.1N, 30 min at 80° C.). This treatment acts to cleave some of the Si—O—Si bonds, thereby exposing hydroxy groups that make the channels more hydrophilic.
  • the device can then optionally be hermetically sealed to a support.
  • the support can be manufactured of essentially any material, although the surface should be flat to ensure a good seal, as the seal formed is primarily due to adhesive forces. Examples of suitable supports include glass, plastics and the like.
  • the devices formed according to the foregoing method result in the substrate (e.g., glass slide) forming one wall of the flow channel.
  • the substrate e.g., glass slide
  • the device once removed from the mother mold is sealed to a thin elastomeric membrane such that the flow channel is totally enclosed in elastomeric material.
  • the resulting elastomeric device can then optionally be joined to a substrate support.
  • Microfluidic devices based on the blind channel design in which reagents are deposited at the reaction sites during manufacture are typically formed of three layers.
  • the bottom layer is the layer upon which reagents are deposited.
  • the bottom layer can be formed from various elastomeric materials as described in the references cited above on MLS methods. Typically, the material is polydimethylsiloxane (PMDS) elastomer.
  • PMDS polydimethylsiloxane
  • the deposited reagents are deposited such that a covalent bond is not formed between the reagent and the surface of the elastomer because, as described earlier, the reagents are intended to dissolve in the sample solution once it is introduced into the reaction site.
  • the other two layers of the device are the layer in which the flow channels are formed and the layer in which the control and optionally guard channels are formed. These two layers are prepared according to the general methods set forth earlier in this section. The resulting two layer structure is then placed on top of the first layer onto which the reagents have been deposited.
  • a specific example of the composition of the three layers is as follows (ration of component A to component B): first layer (sample layer) 30:1 (by weight); second layer (flow channel layer) 30:1; and third layer (control layer) 4:1. It is anticipated, however, that other compositions and ratios of the elastomeric components can be utilized as well.
  • FIG. 6 is a set of photographs taken from the four corners of a device; these photographs demonstrate that the deposited reagents can be accurately aligned within the reaction sites utilizing the foregoing approach. These photographs show guard channels and reaction site located at the end of branch flow channel. The white circle indicates the location of the deposited reagent relative to the reaction site. As indicated, each reagent spot is well within the confines of the reaction site.
  • the reagents can be deposited utilizing any of a number of commercially available reagent spotters and using a variety of established spotting techniques.
  • suitable spotter that can be utilized in the preparation of the devices include pin spotters, acoustic spotters, automated micropipettors, electrophoretic pumps, ink jet printer devices, ink drop printers, and certain osmotic pumps.
  • commercially available spotters include: Cartesian Technologies MicroSys 5100 (Irvine, Calif.), Hitach SPBIO (Alameda, Calif.), Genetix Q-Array (United Kingdom), Affymetrix 417 (Santa Clara, Calif.) and Packard Bioscience SpotArray (Meriden, Conn.).
  • spots of reagents are deposited; usually spots of less than 10 nl are deposited, in other instances less than 5 nl, 2 nl or 1 nl, and in still other instances, less than 0.5 nl, 0.25 nl, or 0.1 nl.
  • Arrays of materials may also be formed by the methods described in Foder, et al., U.S. Pat. No. 5,445,934: titled “Array of oligonucleotides on a solid substrate”, which is herein incorporated by reference, wherein oligonucleotide probes, such as SNP probes, are synthesized in situ using spatial light directed photolithography.
  • Such arrays would be used as the substrate or base of the microfluidic devices of the present invention such that the regions of the substrate corresponding to the reaction sites, for example, blind fill chambers, would be contain one, or preferably more than one, oligonucleotide probes arrayed in known locations on the substrate.
  • the reaction sites depicted as square boxes along the serpentine, fluid channel, would contain a plurality of different SNP probes, preferably a collection of SNP probes suitable for identifying an individual from a population of individuals, and preferably wherein a plurality of reaction sites along the serpentine fluid channel, such that if a fluid sample containing nucleic acid sequences from a plurality of individuals where introduced into the serpentine flow channel, and a plurality of valve in communication with the serpentine flow channel such that when actuated causes the serpentine flow channel to be partitioned thereby isolating each reaction site from one another to contain a fraction of the fluid sample in each reaction site.
  • Amplification of the components of the sample may be performed to increase the number of molecules, for example nucleic acid molecules, for binding to the array of SNP probes located within each reaction site.
  • each of the reaction sites along the serpentine fluid channel would be the same array, that is, have the same SNP probes arrayed, and in other embodiments, two or more of the reaction sites along the serpentine t fluid channel would have a different set of SNP probes.
  • Other partitioning fluid channel architectures could also be used, for example, branched and/or branched branch systems, and so forth.
  • Other arraying techniques, such as spotting described herein, may likewise be used to form the arrays located within the partitionable reactions sites along a serpentine or common, such as in branched, fluid channel(s).
  • FIG. 7A shows a cross-sectional view of the device.
  • the device 700 includes a layer 722 into which is formed the flow channels.
  • This fluid layer 722 is sandwiched between an overlaying layer 720 that includes the control and guard layers and an underlying sealing layer 724 .
  • the sealing layer 724 forms one side of the flow channels.
  • the resulting three-layer structure is affixed to a substrate 726 (in this example, a slide or coverslip), which provides structural stiffness, increases thermal conductivity, and helps to prevent evaporation from the bottom of microfluidic device 700 .
  • a substrate 726 in this example, a slide or coverslip
  • FIG. 7B shows a schematic view of the design of the flow channels in flow layer 722 and of the control channels and guard channel in control/guard layer 720 .
  • Device 700 consists of ten independent flow channels 702 , each with its own inlet 708 , and branching blind channels 704 , each blind channel 704 having a 1 nl reaction site 706 .
  • Device 700 contains a network of control lines 712 , which isolate the reaction sites 706 when sufficient pressure is applied.
  • a series of guard channels 716 are also included to prevent liquid from evaporating out of the reaction sites 706 ; fluid is introduced via inlet 718 .
  • a PCR reaction using ⁇ -actin primers and TaqMan probe to amplify exon 3 of the ⁇ -actin gene from human male genomic DNA was conducted in device 700 .
  • the TaqMan reaction consists of the following components: 1 ⁇ TaqMan Buffer A (50 mM KCl, 10 mM Tris-HCl, 0.01M EDTA, 60 nM Passive Reference 1 (PR1), pH 8.3); 3.5-4.0 mM MgCl; 200 nM dATP, dCTP, dGTP, 400 nM dUTP; 300 nM ⁇ -actin forward primer and reverse primer; 200 nM FAM-labeled ⁇ -actin probe; 0.01 U/ul AmpEraseUNG (Applied Biosystems, Foster City, Calif.); 0.1-0.2 U/ul DyNAzyme (Finnzyme, Espoo, Finland); 0.5% Triton-x-100 (Sigma, St.
  • the TaqMan reaction samples and Control were injected into microfluidic device 700 by using a gel loading pipet tip attached to a 1 ml syringe.
  • the pipet tip was filled with the reaction samples and then inserted into the fluid via 708 .
  • the flow channels 702 were filled by manually applying backpressure to the syringe until all the entire blind channels 704 and reaction sites 706 were filled.
  • Control lines 712 were filled with deionized water and pressurized to 15-20 psi after all of the samples were loaded into the flow lines 702 , 704 .
  • the pressurized control lines 712 were actuated to close the valves and isolate the samples in the 1 nl wells 706 .
  • the guard channels 716 were then filled with deionized water and pressurized to 5-7 psi.
  • Mineral oil (15 ul) (Sigma) was placed on the flatplate of a thermocycler and then the microfluidic device/coverglass 700 was placed on the thermocycler.
  • Micro fluidic device 700 was then thermocycled using an initial ramp and either a three-step or two-step thermocycling profile:
  • MicroAmp tubes (Applied Biosystems, Foster City, Calif.) with the remaining reaction mixture, designated Macro TaqMan reactions to distinguish them from reactions performed in the microfluidic device, were placed in the GeneAmp PCR System 9700 (Applied Biosystems, Foster City, Calif.) and thermocycled in the 9600 mode.
  • the Macro TaqMan reactions served as macroscopic controls for the reactions performed in the micro fluidic device.
  • the thermocycling protocol was set to match that of the microfluidic device, except that the initial ramp rate was not controlled for the Macro TaqMan reactions.
  • thermocycling was completed, the control and guard lines were depressurized and the chip was transferred onto a glass slide (VWR, West Chester, Pa.). The chip was then placed into an Array WoRx Scanner (Applied Precision, Issaquah, Wash.) with a modified carrier. The fluorescence intensity was measured for three different excitation/emission wavelengths: 475/510 nm (FAM), 510/560 nm (VIC), and 580/640 nm (Passive Reference 1 (PR1)). The Array Works Software was used to image the fluorescence in the micro fluidic device and to measure the signal and background intensities of each 1 nl well.
  • FAM 510 nm
  • VIC 510/560 nm
  • PR1 Passive Reference 1
  • results were then analyzed using a Microsoft Excel file to calculate the FAM/PR1 ratio for ⁇ -actin TaqMan reactions.
  • positive samples for target DNA were determined using calculations described in the protocol provided by the manufacturer (TaqMan PCR Reagent Kit Protocol).
  • the signal strength was calculated by dividing the FAM/PR1 ratio of the samples by the FAM/PR1 ratio of the controls.
  • a successful reaction was defined as a sample ratio above the 99% confidence threshold level.
  • AmpliTaq Gold (Applied Biosystems, Foster City, Calif.) was used in TaqMan reactions and FAM/PR1/Control ratios of 1.5-2.0 were produced, compared to Macro TaqMan reaction ratios of 5.0-14.0. Although results were positive, increased signal strength was desired. Therefore, the AmpliTaq Gold polymerase was substituted with DyNAzyme polymerase due to its increased thermostability, proofreading, and resistance to impurities.
  • the standard Macro TaqMan DyNAzyme concentration of 0.025 U/ul was used in the microfluidic experiments. This polymerase change to DyNAzyme produced FAM/ROX/Control ratios of 3.5-5.8. The signal strength was improved, but it was difficult to achieve consistent results.
  • the concentration of the polymerase was increased and surface modifying additives were included.
  • Two increased concentrations of DyNAzyme were tested, 8 ⁇ (0.2 U/ul) and 4 ⁇ (0.1 U/ul) the standard concentration for Macro TaqMan, with 100 pg or 10 pg of genomic DNA per nl in the micro fluidic device.
  • Gelatin, Glycerol, and 0.5% Triton-x-100 were added to prevent the polymerase from attaching to the PDMS.
  • the results of the reactions in the micro fluidic device (chip) and the Macro TaqMan controls are shown in FIG. 8 .
  • the microfluidic TaqMan reaction ratios range from 4.9-8.3, while the Macro TaqMan reactions range from 7.7-9.7. Therefore, the signal strength of the TaqMan reactions in chip is up to 87% of the Macro TaqMan reactions. There was no significant difference between 4 ⁇ or 8 ⁇ DyNAzyme.
  • the results demonstrate that PCR reactions can be done with greater than 50% signal strength, when compared to the Macro TaqMan reactions, in the microfluidic devices. The results have been consistent through at least four attempts.
  • spotted in this context, refers to the placement of small droplets of reagents (spots) on a substrate that is then assembled to become part of a microfluidic device.
  • the spotted reagents are generally a subset of the reagent mixture required for performing PCR.
  • Routine spotting of reagents was performed via a contact printing process. Reagents were picked up from a set of source wells on metal pins, and deposited by contacting the pins to a target substrate. This printing process is further outlined in FIG. 9 . As shown, reagents were picked up from a source (e.g., microtiter plates), and then printed by bringing the loaded pin into contact with the substrate. The wash step consists of agitation in deionized water followed by vacuum drying. The system used to print the reagent spots is a Cartesian Technologies MicroSys 5100 (Irvine, Calif.), employing TeleChem “ChipMaker” brand pins, although other systems can be used as described supra.
  • a source e.g., microtiter plates
  • the wash step consists of agitation in deionized water followed by vacuum drying.
  • the system used to print the reagent spots is a Cartesian Technologies MicroSys 5100 (Irvine, Calif.
  • Pins employed are Telechem ChipMaker 4 pins, which incorporate an electro-milled slot (see FIG. 9 ) to increase the uptake volume (and hence the number of printable spots). Under the operating conditions employed (typically, 75% relative humidity and temperature approximately 25° C.), in excess of one hundred spots were printed per pin, per loading cycle. Under the conditions above, the volume of reagents spotted onto the PDMS substrate is on the order of 0.1 nL.
  • the dimensions of the pin tip are 125 ⁇ 125 ⁇ m.
  • the final spot of dried reagent is substantially smaller than this (as small as 7 ⁇ m in diameter), yet the pin size defines a lower limit to the readily achievable spot spacing.
  • the achievable spacing determines the smallest well-to-well pitch in the final device. Using such a device and the foregoing methods, arrays with spacings of 180 ⁇ m have been achieved. Arrays built into working chips tend to have spacings from 600 to 1300 microns.
  • the flow and control layers of the PCR devices are assembled according to the normal MSL process described above.
  • the microfluidic device design is the same as the one described in Example 1.
  • a substrate layer composed of 150 ⁇ m-thick PDMS with component ratio A:B of 30:1 is formed via spin-coating a blank silicon wafer, and then cured for 90 minutes at 80° C.
  • the cured blank substrate layer of PDMS serves as the target for reagent spotting. Patterns of spots are printed onto the substrate, which is still on the blank wafer.
  • the reagents spotted for PCR reactions were primers and probes, specific to the particular gene to be amplified.
  • the spotted reagent included a 1:1:1 volume ratio of 300 nM ⁇ -actin forward primer (FP), 300 nM ⁇ -actin reverse primer (RP), and 200 nM ⁇ -actin probe (Prb). In some cases, it is useful to further tune the chemistry via concentrating the spotted mixture.
  • the spotted reagent is concentrated to be 3 times and 4 times the concentration of the macro reaction. Concentration of the reagents is performed in a Centrivap heated and evacuated centrifuge and does not alter relative FP:RP:Prb ratios. The increased spot concentration results in the correct final concentration when the reagents are resuspended in a 1 nL reaction volume. Spotted reagents need not be limited to primers and probes; nor must all three (FP, RP and Prb) be spotted. Applications where only the probe, or even one of the primers, is spotted can be performed. Experiments have been conducted in which the sample primer/probe sets spotted were TaqMan ⁇ -actin and TaqMan RNAse-P.
  • the combined flow and control layers i.e., layers 720 and 722 of FIG. 7A
  • the remaining components of the PCR reaction (described in Example 1) are injected into the flow channels of the chip and the chip is thermocycled as described in Example 1.
  • PCR reactions have been successfully and repeatably performed using devices where primer (forward and reverse primers) and probe molecules are spotted.
  • An example of data from a chip in which a reaction has been successfully performed is shown in FIG. 10 .
  • the spotted reagents have resulted in successful PCR reactions as defined in Example 1.
  • Successful reactions have been performed using 2-stage and 3-stage thermocycling protocols.
  • genotyping experiments can be conducted utilizing a microfluidic device or chip such as described herein. Specifically, these experiments were designed to determine if reactions conducted in the device have sufficient sensitivity and to ensure that other primer/probe sets, besides ⁇ -actin, can be performed in the microfluidic device.
  • RNase P TaqMan reactions (Applied Biosystems; Foster City, Calif.) were performed in a microfluidic device as described in Example 1 to demonstrate that other primer/probe sets produce detectable results.
  • RNaseP reactions also require a higher level of sensitivity because the RNaseP primer/probe set detects a single copy gene (2 copies/genome) in contrast to the ⁇ -actin primer/probe set.
  • the ⁇ -actin set detects a single copy ⁇ -actin gene and several pseudogenes, which collectively total approximately 17 copies per genome.
  • the RNase P reactions were run with the same components as described in Example 1, with the exception that the ⁇ -actin primer/probe set was replaced with the RNase P primer/probe set.
  • the RnaseP primer/probe set was used at 4 ⁇ the manufacturer's recommended value to enhance the fluorescence signal.
  • the VIC dye was conjugated to the probe for RNase P and the analysis focused on VIC/PR1 ratios. The results of one of four experiments are shown in FIG. 11 .
  • the VIC/PR1/Control ratios for the Macro TaqMan reactions are 1.23.
  • the corresponding ratios for the TaqMan reactions in the microfluidic device are 1.11 and 1.21.
  • the ratios of the genomic DNA samples in the microfluidic device are above the 99% confidence threshold level.
  • the signal strength of the TaqMan reactions in the microfluidic device is 50% and 93.7% of the Macro TaqMan reactions.
  • the control TaqMan reactions in the microfluidic device have standard deviations of 0.006 and 0.012, demonstrating consistency in the reactions across the micro fluidic device. Therefore, it is determined that the TaqMan reactions in the chip are sensitive enough to detect 2 copies per genome.
  • dilutions of genomic DNA were tested using the ⁇ -actin primer/probe set.
  • Reaction compositions were generally composed as described in Example 1 using 4 ⁇ DyNAzyme and dilutions of genomic DNA.
  • the genomic DNA was diluted down to 0.25 pg/nl, which corresponds to approximately 1 copy per nl.
  • the result of one dilution series is shown in FIG. 12 .
  • the Al-1 (Allele 1, CYP2D6*1 wild type allele) and genomic DNA (100 pg/nl) produced an average VIC/PR1/Control ratio of 3.5 and 2.2, respectively, indicating that the genomic DNA was positive for the CYP2D6*1, wild type allele. These values are above the threshold limit for the reactions.
  • the signal strength of the TaqMan reactions in the microfluidic device is 59% and 40% of the Macro TaqMan controls, respectively.
  • Al-2 (Allele 2, CYP2D6*3 mutant or variant allele) which should be negative in the VIC channel, showed some signal over control (1.5), possibly due to FAM fluorescence leaking into the VIC channel of the detector. The leakage can be minimized with an improved detection process.
  • the Al-2 positive control gave an average FAM/PR1/Control ratio of 3.0, which was 37% of the Macro TaqMan signal and above the calculated threshold limit (see FIG. 14 ).
  • the genomic samples were negative for the CYP2D6*3 mutant allele, an expected result since the frequency of the CYP2D6*3 allele is low. Again, it appears that there is some leakage of the Al-1, VIC probe into the FAM channel of the detector. Overall, the SNP detection reactions were successful in the microfluidic device.
  • PCR reactions compositions were as described in Example 1, except the TaqMan probe was omitted and the ⁇ -actin forward primer was conjugated to FAM.
  • FIG. 15 shows a schematic view of the design.
  • the device 1500 generally consists of a sample region 1502 and a control region 1504 .
  • Sample region 1502 contains three hundred and forty-one 1 nl reaction sites 1508 represented by the rectangles arrayed along flow channel 1506 , which includes inlet via 1510 and outlet via 1512 .
  • Control region 1504 contains three control flow channels 1514 each containing ten 1 nl reaction sites 1518 , also represented by the rectangles and an inlet via 1516 .
  • a network of control lines 1522 isolate each reaction site 1508 , 1518 when sufficient pressure is applied to inlet via 1524 .
  • a series of guard channels 1520 are included to prevent liquid from evaporating out of the reaction sites 1508 , 1518 .
  • the device is a three-layer device as described in Example 1 (see FIG. 7A ). The entire chip is placed onto a coverslip.
  • Microfluidic device 1500 was loaded and thermocycled using the 3 temperature profile described in Example 1. The remaining reaction sample was thermocycled in the GeneAmp 9700 with the same thermocycling profile as for microfluidic device 1500 . The reaction products were recovered after thermocycling was completed. To recover the amplified DNA, 3 ⁇ l of water was injected into sample input via 1506 and 3-4 ⁇ l of product were removed from outlet via 1512 . The reaction products from device 1500 and the Macro reaction were treated with 2 ⁇ l of ExoSAP-IT (USB, Cleveland, Ohio), which is composed of DNA Exonuclease 1 and Shrimp Alkaline Phosphatase, to remove excess nucleotides and primers. The Macro product was diluted from 1:10 to 1:106. The product from device 1500 was dehydrated and resuspended in 4 ⁇ l of formamide.
  • ExoSAP-IT USB, Cleveland, Ohio
  • FIG. 15 shows the gel electrophoresis results.
  • the appropriate size DNA band of 294 base pairs in length is observed in FIG. 16 .
  • the products from the Macro reactions are shown on the left hand side of the gel and correspond to about 294 base pairs, the expected size of the ⁇ -actin PCR product.
  • the negative controls lack the PCR product.
  • the product derived from the device gave the expected ⁇ -actin PCR product. Therefore, target DNA was amplified in the micro fluidic device.
  • PCR polymerase chain reaction
  • FIGS. 17 a - 17 d depict a single bank and dual bank partitioning microfluidic device.
  • MSL multilayer soft lithography
  • thermocycling protocols e.g. long extension times, many cycles
  • precautions against mispriming and non-specific amplification e.g. “hot start” PCR (thermal activation of the polymerase), “booster” PCR, additives to reduce nonspecific hybridization, etc)
  • thermocycling protocols e.g. long extension times, many cycles
  • precautions against mispriming and non-specific amplification e.g. “hot start” PCR (thermal activation of the polymerase), “booster” PCR, additives to reduce nonspecific hybridization, etc
  • this system achieves reliable amplification from single copies using standard conditions—off-the-shelf primers and probes and a single-round, standard thermocycling protocol.
  • This principle of massive partitioning with a digital PCR readout may be used for absolute quantification of the concentration of target in a sample. It can be used, for example, to genotype a pooled sample of genomic DNA simply by counting the numbers of wells that give a positive for a particular allele, or plurality of alleles as described above. Due to the enhanced resistance to side reactions, it should also be useful in quantifying mutants in a background of wild-type DNA—a problem relevant in cancer detection.
  • concentration by partitioning may also be useful in other reactions where detection of single molecules, bacteria, viruses or cells is of interest (e.g. ELISA reactions for protein detection). Digital PCR is described by Brown, et al., U.S. Pat. No.
  • the invention provides for methods and devices for conducting digital PCR in a microfluidic environment comprising the steps of: providing a microfluidic device having a fluid channel therein, said fluid channel having two or more valves associated therewith, the valves, when actuated, being capable of partitioning the fluid channel into two or more reaction sites or chambers; introducing a sample containing at least one target nucleic acid polymer, actuating the valves to partition the fluid sample into two or more portions, wherein at least one portion contains a target nucleic acid polymer and another portion does not contain a target nucleic acid polymer, amplifying the target nucleic acid polymer, and, determining the number of portions of the fluid channel that contained the target molecule.
  • the microfluidic device comprises an elastomeric material, and more preferably, comprises at least one layer comprising an elastomeric material.
  • the microfluidic device further comprises a deflectable membrane wherein the deflectable membrane is deflectable into and out of the fluid channel to control fluid flow within the fluid channel and/or to partition one portion of the fluid channel from another, preferably wherein the deflectable membrane is integral to a layer of the microfluidic device having a channel or recess formed therein, and preferably wherein the deflectable membrane is formed where a first channel in a first layer is overlapped by a second channel in a second layer of the microfluidic device.
  • the sample fluid contains all of the components needed for conducting an amplification reaction
  • the microfluidic device contains at least one component of an amplification reaction prior to the introduction of the sample fluid.
  • the microfluidic device further comprises a detection reagent, preferably one or more nucleic acid polymers complimentary to a least a portion of the target nucleic acid polymer, preferably a plurality of different nucleic acid polymers spatially arrayed within a reaction site or chamber of the microfluidic device.
  • thermocycling reactions such as PCR
  • isothermal reactions such as described by Van Ness et al., in U.S. patent application Ser. No. 10/196,740 which has publishes as U.S. 2003/0138800 A1, which is herein incorporated by reference in its entirety for the purpose of teaching an isothermic amplification scheme.
  • the invention further provides for a protein microcalorimetry assay using a fluorescent dye, for example SYBER green (TM), to measure the conformational changes of a protein, such as denaturation, especially if a protein's denaturation temperature changes when the protein interacts with another moiety such as a ligand or compound or other protein.
  • a fluorescent dye for example SYBER green (TM)
  • TM SYBR Green
  • An additional benefit of using SYBR Green (TM) is that it us used at lower wavelengths than other UV range dyes thus reducing background problems typically associated with many plastic materials.

Abstract

An M×N matrix microfluidic device for performing a matrix of reactions, the device having a plurality of reaction cells in communication with one of either a sample inlet or a reagent inlet through a via formed within an elastomeric block of the device. Methods provided include a method for forming vias in parallel in an elastomeric layer of an elastomeric block of a microfluidic device, the method comprising using patterned photoresist masks and etching reagents to etch away regions or portions of an elastomeric layer of the elastomeric block.

Description

    PRIORITY CLAIM
  • This non-provisional patent application claims is a continuation-in-part application claiming priority to U.S. non-provisional patent application Ser. No. 10/837,835, filed on May 2, 2004, which claims priority to U.S. non-provisional application Ser. No. 10/818,642, filed on Apr. 5, 2004, which claims priority under 35 U.S.C. §119(e) to U.S. provisional patent application Ser. No. 60/460,634, filed on Apr. 3, 2003, by Unger et al., all of which are incorporated by reference in their entirety for all purposes and the specific purposes describe therein and herein.
  • CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application relates to U.S. Non-Provisional application Ser. No. 10/306,798, filed on Nov. 27, 2002, which claims priority under 35 U.S.C. §119(e) to U.S. Provisional Application No. 60/391,529, filed Jun. 24, 2002, and of U.S. Provisional Application No. 60/335,292, filed Nov. 30, 2001, each of which is incorporated by reference in their entirety for all purposes.
  • BACKGROUND
  • Recently, there have been concerted efforts to develop and manufacture microfluidic systems to perform various chemical and biochemical analyses and syntheses, both for preparative and analytical applications. The goal to make such devices arises because of the significant benefits that can be realized from miniaturization with respect to analyses and syntheses conducted on a macro scale. Such benefits include a substantial reduction in time, cost and the space requirements for the devices utilized to conduct the analysis or synthesis. Additionally, microfluidic devices have the potential to be adapted for use with automated systems, thereby providing the additional benefits of further cost reductions and decreased operator errors because of the reduction in human involvement. Microfluidic devices have been proposed for use in a variety of applications, including, for instance, capillary electrophoresis, gas chromatography and cell separations.
  • However, realization of these benefits has often been thwarted because of various complications associated with the microfluidic devices that have thus far been manufactured. For instance, many of the current microfluidic devices are manufactured from silica-based substrates; these materials are difficult and complicated to machine and devices made from such materials are fragile. Furthermore, transport of fluid through many existing microfluidic devices requires regulation of complicated electrical fields to transport fluids in a controlled fashion through the device.
  • Thus, in view of the foregoing benefits that can be achieved with microfluidic devices but the current limitations of existing devices, there remains a need for microfluidic devices designed for use in conducting a variety of chemical and biochemical analyses. Because of its importance in modern biochemistry, there is a particular need for devices that can be utilized to conduct a variety of nucleic acid amplification reactions, while having sufficient versatility for use in other types of analyses as well.
  • Devices with the ability to conduct nucleic acid amplifications would have diverse utilities. For example, such devices could be used as an analytical tool to determine whether a particular target nucleic acid of interest is present or absent in a sample. Thus, the devices could be utilized to test for the presence of particular pathogens (e.g., viruses, bacteria or fungi), and for identification purposes (e.g., paternity and forensic applications). Such devices could also be utilized to detect or characterize specific nucleic acids previously correlated with particular diseases or genetic disorders. When used as analytical tools, the devices could also be utilized to conduct genotyping analyses and gene expression analyses (e.g., differential gene expression studies). Alternatively, the devices can be used in a preparative fashion to amplify sufficient nucleic acid for further analysis such as sequencing of amplified product, cell-typing, DNA fingerprinting and the like. Amplified products can also be used in various genetic engineering applications, such as insertion into a vector that can then be used to transform cells for the production of a desired protein product.
  • SUMMARY
  • A variety of devices and methods for conducting microfluidic analyses are provided herein, including devices that can be utilized to conduct thermal cycling reactions such as nucleic acid amplification reactions. The devices differ from conventional microfluidic devices in that they include elastomeric components; in some instances, much or all of the device is composed of elastomeric material.
  • Certain devices are designed to conduct thermal cycling reactions (e.g., PCR) with devices that include one or more elastomeric valves to regulate solution flow through the device. Thus, methods for conducting amplification reactions with devices of this design are also provided.
  • Some of the devices include blind flow channels which include a region that functions as a reaction site. Certain such devices include a flow channel formed within an elastomeric material, and a plurality of blind flow channels in fluid communication with the flow channel, with a region of each blind flow channel defining a reaction site. The devices can also include one or more control channels overlaying and intersecting each of the blind flow channels, wherein an elastomeric membrane separates the one or more control channels from the blind flow channels at each intersection. The elastomeric membrane in such devices is disposed to be deflected into or withdrawn from the blind flow channel in response to an actuation force. The devices can optionally further include a plurality of guard channels formed within the elastomeric material and overlaying the flow channel and/or one or more of the reaction sites. The guard channels are designed to have fluid flow therethrough to reduce evaporation from the flow channels and reaction sites of the device. Additionally, the devices can optionally include one or more reagents deposited within each of the reaction sites.
  • In certain devices, the flow channel is one of a plurality of flow channels, each of the flow channels in fluid communication with multiple blind flow channels which branch therefrom. Of devices of this design, in some instances the plurality of flow channels are interconnected with one another such that fluid can be introduced into each of the reaction sites via a single inlet. In other devices, however, the plurality of flow channels are isolated from each other such that fluid introduced into one flow channel cannot flow to another flow channel, and each flow channel comprises an inlet at one or both ends into which fluid can be introduced.
  • Other devices include an array of reaction sites having a density of at least 50 sites/cm2, with the reaction sites typically formed within an elastomeric material. Other devices have even higher densities such as at least 250, 500 or 1000 sites/cm2, for example.
  • Still other device include a reaction site formed within an elastomeric substrate, at which a reagent for conducting a reaction is non-covalently immobilized. The reagent can be one or more reagents for conducting essentially any type of reaction. The reagent in some devices includes one reagents for conducting a nucleic acid amplification reaction. Thus, in some devices the reagent comprises a primer, polymerase and one or more nucleotides. In other devices, the reagent is a nucleic acid template.
  • A variety of matrix or array-based devices are also provided. Certain of these devices include: (i) a first plurality of flow channels formed in an elastomeric substrate, (ii) a second plurality of flow channels formed in the elastomeric substrate that intersect the first plurality of flow channels to define an array of reaction sites, (iii) a plurality of isolation valves disposed within the first and second plurality of flow channels that can be actuated to isolate solution within each of the reaction sites from solution at other reaction sites, and (iv) a plurality of guard channels overlaying one or more of the flow channels and/or one or more of the reaction sites to prevent evaporation of solution therefrom.
  • The foregoing devices can be utilized to conduct a number of different types of reactions, including those involving temperature regulation (e.g., thermocycling of nucleic acid analyses). Methods conducted with certain blind channel type devices involve providing a microfluidic device that comprises a flow channel formed within an elastomeric material; and a plurality of blind flow channels in fluid communication with the flow channel, with an end region of each blind flow channel defining a reaction site. At least one reagent is introduced into each of the reaction sites, and then a reaction is detected at one or more of the reaction sites. The method can optionally include heating the at least one reagent within the reaction site. Thus, for example, a method can involve introducing the components for a nucleic acid amplification reaction and then thermocycling the components to form amplified product.
  • Other methods involve providing a microfluidic device comprising one or more reaction sites, each reaction site comprising a first reagent for conducting an analysis that is non-covalently deposited on an elastomeric substrate. A second reagent is then introduced into the one or more reaction sites, whereby the first and second reagents mix to form a reaction mixture. A reaction between the first and second reagents at one or more of the reaction sites is subsequently detected.
  • Still other methods involve providing a microfluidic device comprising an array of reaction sites formed within a substrate and having a density of at least 50 sites/cm2. At least one reagent is introduced into each of the reaction sites. A reaction at one or more of the reaction sites is then detected.
  • Yet other methods involve providing a microfluidic device comprising at least one reaction site which is formed within an elastomeric substrate and a plurality of guard channels also formed within the elastomeric substrate. At least one reagent is introduced into each of the reaction sites and then heated within the reaction sites. A fluid is flowed through the guard channels before or during heating to reduce evaporation from the at least one reaction site. A reaction within the at least one reaction site is subsequently detected.
  • Additional devices designed to reduce evaporation of fluid from the device are also provided. In general, such devices comprise a cavity that is part of a microfluidic network formed in an elastomeric substrate; and a plurality of guard channels overlaying the cavity and separated from the cavity by an elastomeric membrane. The guard channel in such devices is sized (i) to allow solution flow therethrough, and (ii) such that there is not a substantial reduction in solution flow in, out or through the cavity due to deflection of the membrane(s) upon application of an actuation force to the guard channels. Other such devices include (i) one or more flow channels and/or one or more reaction sites; and (ii) a plurality of guard channels overlaying the microfluidic system and separated therefrom by elastomer, wherein the spacing between guard channels is between 1 μm to 1 mm. In other devices the spacing is between 5 μm and 500 μm, in other devices between 10 μm and 100 μm, and in still other devices between 40 μm and 75 μm.
  • Compositions for conducting nucleic acid analyses in reaction sites of certain microfluidic devices are also provided. Certain such compositions include one or more of the following: an agent that blocks protein binding sites on an elastomeric material and a detergent. The blocking agent is typically selected from the group consisting of a protein (e.g., gelatin or albumin, such as bovine serum albumin (BSA)). The detergent can be SDS or Triton, for example.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1A is a schematic representation of an exemplary device with a matrix design of intersecting vertical and horizontal flow channels.
  • FIGS. 1B-E show enlarged views of a portion of the device shown in FIG. 1A and illustrates its operation.
  • FIG. 1F is a schematic representation of another exemplary matrix design device that utilizes guard channels to reduce sample evaporation.
  • FIG. 2 is a plan view of an exemplary blind channel device.
  • FIG. 3A is a plan view of another exemplary blind channel device.
  • FIG. 3B is a schematic representation of a more complex blind channel device based upon the unit of the general design depicted in FIG. 3A.
  • FIG. 3C is an enlarged view of a region of the device shown in FIG. 3B, and illustrates the orientation of the guard flow channels in this particular design.
  • FIG. 4 is a plan view of a device utilizing the hybrid design.
  • FIG. 5 is a chart showing ramp up and down times to conduct a thermocycling reaction.
  • FIG. 6 shows the location of spotted reagents within reaction sites in a blind channel type device illustrating proper alignment of the reagents within reaction sites at the corners of the device.
  • FIGS. 7A and 7B respectively are a cross-sectional view and a schematic diagram of another hybrid type microfluidic device and represents the type of device used to conduct the experiments described in Examples 1-4.
  • FIG. 8 is a bar graph in which the average FAM/PR1/Control ratios are plotted for six different β-actin TaqMan reactions. The reactions were thermocycled in the micro fluidic device (chip) shown in FIG. 7B (solid bars) and Macro TaqMan reactions (striped bars). The controls are the first and fourth bar sets that have no DNA. The error bars are the standard deviation of the ratios.
  • FIG. 9 is a diagram depicting an exemplary pin spotting process. Reagents are picked up from a source (e.g., a microtiter plate) and then printed by bringing the loaded pin into contact with the substrate. The wash step consists of agitation in deionized water followed by vacuum drying.
  • FIG. 10 is a bar graph depicting FAM signal strength for the microfluidic device (chip) described in Example 1 (see FIG. 7B) based on the experiments described in Example 2. The data are in the form of (FAM signal/PR1 signal) scaled by the FAM/PR1 ratio for the reference lanes. Error bars are the standard deviation along a lane. The “1.3X” and “1X” designations refer to the concentration of the spotted primers and probes, in relation to their nominal values.
  • FIG. 11 is a bar graph showing average VIC/PF1/Control ratios for 9-10 wells for Macro TaqMan (striped bars), and TaqMan reactions in the microfluidic device (solid bars). Two negative controls (Control) and two samples with 100 pg/nl genomic DNA were thermocycled with reaction components as described above with 4× the standard amount of primer/probe. The error bars represent the standard deviation of the average ratios.
  • FIG. 12 is a bar graph that shows FAM/PR1/Control ratios for each of 10-1 nl wells branching from a single flow channel of a microfluidic device (see FIG. 7B). The amount of genomic DNA was 0.25 pg/nl, which results in an average of one target copy per well.
  • FIG. 13 is a bar graph depicting the average VIC/PR1/Control ratios for CYP2D6 SNP reactions using the microfluidic device shown in FIG. 7B. Allele 1 (Al-1) is the positive control for the VIC probe against the reference or wild type allele CYP2D6*1. Allele 2 (Al-2) is the positive control for the FAM probe against the variant or mutant allele, CYP2D6*3. The control has no DNA template. Genomic DNA was used at either 100 pg/nl or 20 pg/nl. The error bars are the standard deviation of the ratios.
  • FIG. 14 is a bar graph showing the average FAM/PR1/Control ratios for CYP2D6 SNP reactions in the microfluidic device shown in FIG. 7B. The samples are the same as described with respect to FIG. 13 and in Example 3.
  • FIG. 15 is a schematic diagram of the microfluidic device used for the experiments in Example 4.
  • FIG. 16 is a polyacrylamide gel containing PCR product from Macro PCR and PCR reactions in the microfluidic device shown in FIG. 7B. The results on the left show the approximate migration of different DNA base pair lengths. The lanes containing interspersed bands are molecular weight markers. The lanes labeled “Macro” are the PCR products from the Macro reactions at different dilutions. The lanes labeled “In chip” are PCR products generated in the chip. The lanes containing many bands throughout the gel are nonspecific background signals.
  • FIGS. 17 a-17 d depict two preferred designs of a partitioning microfluidic device in a valve off and valve actuated state.
  • FIGS. 18 a and 18 b depict images of a partitioning microfluidic devices after a thermocycling reaction was performed. FIG. 18 a depicts a two color image, and FIG. 18 b depicts the remaining signal after subtraction of the control red signal.
  • FIG. 19 depicts a graph of comparing the average number of copies per well to the number of positive wells.
  • FIG. 20 depicts an isothermic amplification scheme—SCORPION
  • FIG. 21 depicts an exemplary matrix microfluidic device plan view.
  • DETAILED DESCRIPTION
  • I. Definitions
  • Unless defined otherwise, all technical and scientific terms used herein have the meaning commonly understood by a person skilled in the art to which this invention belongs. The following references provide one of skill with a general definition of many of the terms used in this invention: Singleton et al., DICTIONARY OF MICROBIOLOGY AND MOLECULAR BIOLOGY (2d ed. 1994); THE CAMBRIDGE DICTIONARY OF SCIENCE AND TECHNOLOGY (Walker ed., 1988); THE GLOSSARY OF GENETICS, 5TH ED., R. Rieger et al. (eds.), Springer Verlag (1991); and Hale & Marham, THE HARPER COLLINS DICTIONARY OF BIOLOGY (1991). As used herein, the following terms have the meanings ascribed to them unless specified otherwise.
  • A “flow channel” refers generally to a flow path through which a solution can flow.
  • The term “valve” unless otherwise indicted refers to a configuration in which a flow channel and a control channel intersect and are separated by an elastomeric membrane that can be deflected into or retracted from the flow channel in response to an actuation force.
  • A “blind channel” or a “dead-end channel” refers to a flow channel which has an entrance but not a separate exit. Accordingly, solution flow in and out of the blind channel occurs at the same location. The process of filling one or more blind channels is sometimes simply referred to as “blind fill.”
  • An “isolated reaction site” generally refers to a reaction site that is not in fluid communication with other reactions sites present on the device. When used with respect to a blind channel, the isolated reaction site is the region at the end of the blind channel that can be blocked off by a valve associated with the blind channel.
  • A “via” refers to a channel formed in an elastomeric device to provide fluid access between an external port of the device and one or more flow channels. Thus, a via can serve as a sample input or output, for example.
  • The term “elastomer” and “elastomeric” has its general meaning as used in the art. Thus, for example, Allcock et al. (Contemporary Polymer Chemistry, 2nd Ed.) describes elastomers in general as polymers existing at a temperature between their glass transition temperature and liquefaction temperature. Elastomeric materials exhibit elastic properties because the polymer chains readily undergo torsional motion to permit uncoiling of the backbone chains in response to a force, with the backbone chains recoiling to assume the prior shape in the absence of the force. In general, elastomers deform when force is applied, but then return to their original shape when the force is removed. The elasticity exhibited by elastomeric materials can be characterized by a Young's modulus. The elastomeric materials utilized in the microfluidic devices disclosed herein typically have a Young's modulus of between about 1 Pa-1 TPa, in other instances between about 10 Pa-100 GPa, in still other instances between about 20 Pa-1 GPa, in yet other instances between about 50 Pa-10 MPa, and in certain instances between about 100 Pa-1 MPa. Elastomeric materials having a Young's modulus outside of these ranges can also be utilized depending upon the needs of a particular application.
  • Some of the microfluidic devices described herein are fabricated from an elastomeric polymer such as GE RTV 615 (formulation), a vinyl-silane crosslinked (type) silicone elastomer (family). However, the present microfluidic systems are not limited to this one formulation, type or even this family of polymer; rather, nearly any elastomeric polymer is suitable. Given the tremendous diversity of polymer chemistries, precursors, synthetic methods, reaction conditions, and potential additives, there are a large number of possible elastomer systems that can be used to make monolithic elastomeric microvalves and pumps. The choice of materials typically depends upon the particular material properties (e.g., solvent resistance, stiffness, gas permeability, and/or temperature stability) required for the application being conducted. Additional details regarding the type of elastomeric materials that can be used in the manufacture of the components of the microfluidic devices disclosed herein are set forth in Unger et al. (2000) Science 288: 113-116, and PCT Publications WO 02/43615, and WO 01/01025, which are incorporated herein by reference in their entirety for all purposes.
  • The terms “nucleic acid,” “polynucleotide,” and “oligonucleotide” are used herein to include a polymeric form of nucleotides of any length, including, but not limited to, ribonucleotides or deoxyribonucleotides. There is no intended distinction in length between these terms. Further, these terms refer only to the primary structure of the molecule. Thus, in certain embodiments these terms can include triple-, double- and single-stranded DNA, as well as triple-, double- and single-stranded RNA. They also include modifications, such as by methylation and/or by capping, and unmodified forms of the polynucleotide. More particularly, the terms “nucleic acid,” “polynucleotide,” and “oligonucleotide,” include polydeoxyribonucleotides (containing 2-deoxy-D-ribose), polyribonucleotides (containing D-ribose), any other type of polynucleotide which is an N— or C-glycoside of a purine or pyrimidine base, and other polymers containing nonnucleotidic backbones, for example, polyamide (e.g., peptide nucleic acids (PNAs)) and polymorpholino (commercially available from the Anti-Virals, Inc., Corvallis, Oreg., as Neugene) polymers, and other synthetic sequence-specific nucleic acid polymers providing that the polymers contain nucleobases in a configuration which allows for base pairing and base stacking, such as is found in DNA and RNA.
  • A “probe” is an nucleic acid capable of binding to a target nucleic acid of complementary sequence through one or more types of chemical bonds, usually through complementary base pairing, usually through hydrogen bond formation, thus forming a duplex structure. The probe binds or hybridizes to a “probe binding site.” The probe can be labeled with a detectable label to permit facile detection of the probe, particularly once the probe has hybridized to its complementary target. The label attached to the probe can include any of a variety of different labels known in the art that can be detected by chemical or physical means, for example. Suitable labels that can be attached to probes include, but are not limited to, radioisotopes, fluorophores, chromophores, mass labels, electron dense particles, magnetic particles, spin labels, molecules that emit chemiluminescence, electrochemically active molecules, enzymes, cofactors, and enzyme substrates. Probes can vary significantly in size. Some probes are relatively short. Generally, probes are at least 7 to 15 nucleotides in length. Other probes are at least 20, 30 or 40 nucleotides long. Still other probes are somewhat longer, being at least 50, 60, 70, 80, 90 nucleotides long. Yet other probes are longer still, and are at least 100, 150, 200 or more nucleotides long. Probes can be of any specific length that falls within the foregoing ranges as well.
  • A “primer” is a single-stranded polynucleotide capable of acting as a point of initiation of template-directed DNA synthesis under appropriate conditions (i.e., in the presence of four different nucleoside triphosphates and an agent for polymerization, such as, DNA or RNA polymerase or reverse transcriptase) in an appropriate buffer and at a suitable temperature. The appropriate length of a primer depends on the intended use of the primer but typically is at least 7 nucleotides long and, more typically range from 10 to 30 nucleotides in length. Other primers can be somewhat longer such as 30 to 50 nucleotides long. Short primer molecules generally require cooler temperatures to form sufficiently stable hybrid complexes with the template. A primer need not reflect the exact sequence of the template but must be sufficiently complementary to hybridize with a template. The term “primer site” or “primer binding site” refers to the segment of the target DNA to which a primer hybridizes. The term “primer pair” means a set of primers including a 5′ “upstream primer” that hybridizes with the complement of the 5′ end of the DNA sequence to be amplified and a 3′ “downstream primer” that hybridizes with the 3′ end of the sequence to be amplified.
  • A primer that is “perfectly complementary” has a sequence fully complementary across the entire length of the primer and has no mismatches. The primer is typically perfectly complementary to a portion (subsequence) of a target sequence. A “mismatch” refers to a site at which the nucleotide in the primer and the nucleotide in the target nucleic acid with which it is aligned are not complementary. The term “substantially complementary” when used in reference to a primer means that a primer is not perfectly complementary to its target sequence; instead, the primer is only sufficiently complementary to hybridize selectively to its respective strand at the desired primer-binding site.
  • The term “complementary” means that one nucleic acid is identical to, or hybridizes selectively to, another nucleic acid molecule. Selectivity of hybridization exists when hybridization occurs that is more selective than total lack of specificity. Typically, selective hybridization will occur when there is at least about 55% identity over a stretch of at least 14-25 nucleotides, preferably at least 65%, more preferably at least 75%, and most preferably at least 90%. Preferably, one nucleic acid hybridizes specifically to the other nucleic acid. See M. Kanehisa, Nucleic Acids Res. 12: 203 (1984).
  • The term “label” refers to a molecule or an aspect of a molecule that can be detected by physical, chemical, electromagnetic and other related analytical techniques. Examples of detectable labels that can be utilized include, but are not limited to, radioisotopes, fluorophores, chromophores, mass labels, electron dense particles, magnetic particles, spin labels, molecules that emit chemiluminescence, electrochemically active molecules, enzymes, cofactors, enzymes linked to nucleic acid probes and enzyme substrates. The term “detectably labeled” means that an agent has been conjugated with a label or that an agent has some inherent characteristic (e.g., size, shape or color) that allows it to be detected without having to be conjugated to a separate label.
  • A “polymorphic marker” or “polymorphic site” is the locus at which divergence occurs. Preferred markers have at least two alleles, each occurring at frequency of greater than 1%, and more preferably greater than 10% or 20% of a selected population. A polymorphic locus may be as small as one base pair. Polymorphic markers include restriction fragment length polymorphisms, variable number of tandem repeats (VNTR's), hypervariable regions, minisatellites, dinucleotide repeats, trinucleotide repeats, tetranucleotide repeats, simple sequence repeats, and insertion elements such as Alu. The first identified allelic form is arbitrarily designated as the reference form and other allelic forms are designated as alternative or variant alleles. The allelic form occurring most frequently in a selected population is sometimes referred to as the wildtype form. Diploid organisms may be homozygous or heterozygous for allelic forms. A diallelic polymorphism has two forms. A triallelic polymorphism has three forms.
  • A “single nucleotide polymorphism” (SNP) occurs at a polymorphic site occupied by a single nucleotide, which is the site of variation between allelic sequences. The site is usually preceded by and followed by highly conserved sequences of the allele (e.g., sequences that vary in less than 1/100 or 1/11000 members of the populations). A single nucleotide polymorphism usually arises due to substitution of one nucleotide for another at the polymorphic site. A transition is the replacement of one purine by another purine or one pyrimidine by another pyrimidine. A transversion is the replacement of a purine by a pyrimidine or vice versa. Single nucleotide polymorphisms can also arise from a deletion of a nucleotide or an insertion of a nucleotide relative to a reference allele.
  • A “reagent” refers broadly to any agent used in a reaction. A reagent can include a single agent which itself can be monitored (e.g., a substance that is monitored as it is heated) or a mixture of two or more agents. A reagent may be living (e.g., a cell) or non-living. Exemplary reagents for a nucleic acid amplification reaction include, but are not limited to, buffer, metal ions, polymerase, primers, template nucleic acid, nucleotides, labels, dyes, nucleases and the like. Reagents for enzyme reactions include, for example, substrates, cofactors, coupling enzymes, buffer, metal ions, inhibitors and activators. Reagents for cell-based reactions include, but are not limited to, cells, cell specific dyes and ligands (e.g., agonists and antagonists) that bind to cellular receptors.
  • A “ligand” is any molecule for which there exists another molecule (i.e., an “antiligand”) that specifically or non-specifically binds to the ligand, owing to recognition of some portion of the ligand by the antiligand.
  • II. Overview
  • A number of different microfluidic devices (also sometimes referred to as chips) having unique flow channel architectures are provided herein, as well as methods for using such devices to conduct a variety of high throughput analyses. The devices are designed for use in analyses requiring temperature control, especially analyses involving thermocycling (e.g., nucleic acid amplification reactions). The microfluidic devices incorporate certain design features that: give the devices a significantly smaller footprint than many conventional microfluidic devices, enable the devices to be readily integrated with other instrumentation and provide for automated analysis.
  • Some of the microfluidic devices utilize a design typically referred to herein as “blind channel” or “blind fill” are characterized in part by having a plurality of blind channels, which, as indicated in the definition section, are flow channels having a dead end or isolated end such that solution can only enter and exit the blind channel at one end (i.e., there is not a separate inlet and outlet for the blind channel). These devices require only a single valve for each blind channel to isolate a region of the blind channel to form an isolated reaction site. During manufacture of this type of device, one or more reagents for conducting an analysis are deposited at the reaction sites, thereby resulting in a significant reduction in the number of input and outputs. Additionally, the blind channels are connected to an interconnected network of channels such that all the reaction sites can be filled from a single, or limited number, of sample inputs. Because of the reduction in complexity in inputs and outputs and the use of only a single valve to isolate each reaction site, the space available for reaction sites is increased. Thus, the features of these devices means that each device can include a large number of reaction sites (e.g., up to tens of thousands) and can achieve high reaction site densities (e.g., over 1,000-4,000 reaction sites/cm2). Individually and collectively, these features also directly translate into a significant reduction in the size of these devices compared to traditional microfluidic devices.
  • Other microfluidic devices that are disclosed herein utilize a matrix design. In general, microfluidic devices of this type utilize a plurality of intersecting horizontal and vertical flow channels to define an array of reaction sites at the points of intersection. Thus, devices of this design also have an array or reaction sites; however, there is a larger number of sample inputs and corresponding outputs to accommodate the larger number of samples with this design. A valve system referred to as a switchable flow array architecture enables solution be flowed selectively through just the horizontal or flow channels, thus allowing for switchable isolation of various flow channels in the matrix. Hence, whereas the blind channel devices are designed to conduct a large number of analyses under different conditions with a limited number of samples, the matrix devices are constructed to analyze a large number of sample under a limited number of conditions. Still other devices are hybrids of these two general design types.
  • The microfluidic devices that are described are further characterized in part by utilizing various components such as flow channels, control channels, valves and/or pumps from elastomeric materials. In some instances, essentially the entire device is made of elastomeric material. Consequently, such devices differ significantly in form and function from the majority of conventional microfluidic devices that are formed from silicon-based material.
  • The design of the devices enables them to be utilized in combination with a number of different heating systems. Thus, the devices are useful in conducting diverse analyses that require temperature control. Additionally, those microfluidic devices for use in heating applications can incorporate a further design feature to minimize evaporation of sample from the reaction sites. Devices of this type in general include a number of guard channels formed within the elastomeric device through which water can be flowed to increase the water vapor pressure within the elastomeric material from which the device is formed, thereby reducing evaporation of sample from the reaction sites.
  • In another embodiment, a temperature cycling device may be used to control the temperature of the microfluidic devices. Preferably, the microfluidic device would be adapted to make thermal contact with the microfluidic device. Where the microfluidic device is supported by a substrate material, such as a glass slide or the bottom of a carrier plate, such as a plastic carrier, a window may be formed in a region of the carrier or slide such that the microfluidic device, preferably a device having an elastomeric block, may directly contact the heating/cooling block of the temperature cycling device. In a preferred embodiment, the heating/cooling block has grooves therein in communication with a vacuum source for applying a suction force to the microfluidic device, preferably the portion wherein the reactions are taking place. Alternatively, a rigid thermally conductive plate may be bonded to the microfluidic device that then mates with the heating and cooling block for efficient thermal conduction resulting.
  • The array format of certain of the devices means the devices can achieve high throughput. Collectively, the high throughput and temperature control capabilities make the devices useful for performing large numbers of nucleic acid amplifications (e.g., polymerase chain reaction—PCR). Such reactions will be discussed at length herein as illustrative of the utility of the devices, especially of their use in any reaction requiring temperature control. However, it should be understood that the devices are not limited to these particular applications. The devices can be utilized in a wide variety of other types of analyses or reactions. Examples include analyses of protein-ligand interactions and interactions between cells and various compounds. Further examples are provided infra.
  • III. General Structure of Microfluidic Devices
  • A. Pumps and Valves
  • The microfluidic devices disclosed herein are typically constructed at least in part from elastomeric materials and constructed by single and multilayer soft lithography (MSL) techniques and/or sacrificial-layer encapsulation methods (see, e.g., Unger et al. (2000) Science 288: 113-116, and PCT Publication WO 01/01025, both of which are incorporated by reference herein in their entirety for all purposes). Utilizing such methods, microfluidic devices can be designed in which solution flow through flow channels of the device is controlled, at least in part, with one or more control channels that are separated from the flow channel by an elastomeric membrane or segment. This membrane or segment can be deflected into or retracted from the flow channel with which a control channel is associated by applying an actuation force to the control channels. By controlling the degree to which the membrane is deflected into or retracted out from the flow channel, solution flow can be slowed or entirely blocked through the flow channel. Using combinations of control and flow channels of this type, one can prepare a variety of different types of valves and pumps for regulating solution flow as described in extensive detail in Unger et al. (2000) Science 288: 113-116, and PCT Publication WO/02/43615 and WO 01/01025.
  • The devices provided herein incorporate such pumps and valves to isolate selectively a reaction site at which reagents are allowed to react. The reaction sites can be located at any of a number of different locations within the device. For example, in some matrix-type devices, the reaction site is located at the intersection of a set of flow channels. In blind channel devices, the reaction site is located at the end of the blind channel.
  • If the device is to be utilized in temperature control reactions (e.g., thermocycling reactions), then, as described in greater detail infra, the elastomeric device is typically fixed to a support (e.g., a glass slide). The resulting structure can then be placed on a temperature control plate, for example, to control the temperature at the various reaction sites. In the case of thermocycling reactions, the device can be placed on any of a number of thermocycling plates.
  • Because the devices are made of elastomeric materials that are relatively optically transparent, reactions can be readily monitored using a variety of different detection systems at essentially any location on the microfluidic device. Most typically, however, detection occurs at the reaction site itself (e.g., within a region that includes an intersection of flow channels or at the blind end of a flow channel). The fact that the device is manufactured from substantially transparent materials also means that certain detection systems can be utilized with the current devices that are not usable with traditional silicon-based microfluidic devices. Detection can be achieved using detectors that are incorporated into the device or that are separate from the device but aligned with the region of the device to be detected.
  • B. Guard Channels
  • To reduce evaporation of sample and reagents from the elastomeric microfluidic devices that are provided herein, a plurality of guard channels can be formed in the devices. The guard channels are similar to the control channels in that typically they are formed in a layer of elastomer that overlays the flow channels and/or reaction site. Hence, like control channels, the guard channels are separated from the underlying flow channels and/or reaction sites by a membrane or segment of elastomeric material. Unlike control channels, however, the guard channels are considerably smaller in cross-sectional area. In general, a membrane with smaller area will deflect less than a membrane with larger area under the same applied pressure. The guard channels are designed to be pressurized to allow solution (typically water) to be flowed into the guard channel. Water vapor originating from the guard channel can diffuse into the pores of the elastomer adjacent a flow channel or reaction site, thus increasing the water vapor concentration adjacent the flow channel or reaction site and reducing evaporation of solution therefrom.
  • In general, the guard channels are sufficiently small such that when pressurized the membrane that separates the guard channel from the underlying flow channel or reaction site does not substantially restrict solution flow in, out, or through the flow channel or reaction site which the guard channel overlays. When used in this context, the term “substantially restrict” or other similar terms means that solution flow is not reduced in, out or through the flow channel or reaction site by more than 40%, typically less than 30%, usually less than 20%, and in some instances less than 10%, as compared to solution flow in, to or through the flow channel or reaction site under the same conditions, when the guard channel is not pressurized to achieve solution flow therethrough. Usually this means that the guard channels have a cross-sectional area of between 100 μm2 and 50,000 μm2, or any integral or non-integral cross-sectional area therebetween. Thus, for example, in some instances, the cross-sectional area is less than 50,000 μm2, in other instances less than 10,000 μm2, in still other instances less than 10,00 μm2, and in yet other instances less than 100 μm2. The guard channels can have any of a variety of shapes including, but not limited to, circular, elliptical, square, rectangular, hexagonal and octahedral shapes.
  • The guard channels are designed to reduce the evaporation of sample and reagents from the device during the time and under the conditions that it takes to conduct a thermocycling reaction to less than 50%, in other instance less than 45%, 40%, 35%, 30%, 25%, 20%, 15%, 10%, 5% or 1%. Thus, for example, a typical PCR reaction involving 40 cycles can be conducted within 120 minutes. The guard channel system is designed to reduce evaporation during approximately this time frame to the foregoing set of limits. To achieve this level of evaporation reduction, the guard channels are typically present at a density of at least 10 lines/cm2 to 1000 lines/cm2, or any integral density level therebetween. More specifically, the guard channels are generally at least 25 lines/cm2, in other instances at least 50 lines/cm2, in still other instances at least 100 lines/cm2, and in yet other instances at least 500 lines/cm2. To achieve this level of evaporation reduction, the guard channels are typically present at a spacing between 1 mm to 1 μm as measured from the outer edge of one line to the nearest outer edge of adjacent line, or any integral density level therebetween. More specifically, the guard channels are generally spaced between 500 μm to 5 μm, in other instances between 100 μm to 10 μm, in still other instances between 75 μm to 40 μm. Thus, the spacing is typically at least 1 μm, but is less than 1 mm, in other instances less than 500 μm, in still other instances less than 400 μm, in yet other instances less than 300 μm, in other instances less than 200 μm, and in still other instances less than 100 μm, 50 μm or 25 μm.
  • The guard channels can be formed as a separate network of channels or can be smaller channels that branch off of the control channels. The guard channels can extend across the device or only a particular region or regions of the device. Typically, the guard channels are placed adjacent and over flow channels and reaction sites as these are the primary locations at which evaporation is the primary concern. Exemplary locations of guard channels on certain matrix devices are illustrated in FIG. 1C, and on certain blind channel devices in FIGS. 3B and 3C, and discussed in greater detail infra.
  • The solution flowed through the guard channel includes any substance that can reduce evaporation of water. The substance can be one that increases the water vapor concentration adjacent a flow line and/or reaction site, or one that while not increasing the water vapor concentration nonetheless blocks evaporation of water from the flow line and/or reaction site (blocking agent). Thus, one option is to utilize essentially any aqueous solution in which case suitable solutions include, but are not limited to, water and buffered solution (e.g., TaqMan buffer solution, and phosphate buffered saline). Suitable blocking agents include, for example, mineral oil.
  • Guard channels are typically formed in the elastomer utilizing the MSL techniques and/or sacrificial-layer encapsulation methods cited above.
  • The following sections describe in greater detail a number of specific configurations that can be utilized to conduct a variety of analyses, including analyses requiring temperature control (e.g., nucleic acid amplification reactions). It should be understood, however, that these configurations are exemplary and that modifications of these systems will be apparent to those skilled in the art.
  • IV. Matrix Design
  • A. General
  • Devices utilizing the matrix design generally have a plurality of vertical and horizontal flow channel that intersect to form an array of junctions. Because a different sample and reagent (or set of reagents) can be introduced into each of the flow channels, a large number of samples can be tested against a relatively large number of reaction conditions in a high throughput format. Thus, for example, if a different sample is introduced into each of M different vertical flow channels and a different reagent (or set of reagents) is introduced into each of N horizontal flow channels, then M×N different reactions can be conducted at the same time. Typically, matrix devices include valves that allow for switchable isolation of the vertical and horizontal flow channels. Said differently, the valves are positioned to allow selective flow just through the vertical flow channels or just through the horizontal flow channels. Because devices of this type allow flexibility with respect to the selection of the type and number of samples, as well as the number and type of reagents, these devices are well-suited for conducting analyses in which one wants to screen a large number of samples against a relatively large number of reaction conditions. The matrix devices can optionally incorporate guard channels to help prevent evaporation of sample and reactants.
  • The invention provides for high-density matrix designs that utilize fluid communication vias between layers of the microfluidic device to weave control lines and fluid lines through the device. For example, by having a fluid line in each layer of a two layer elastomeric block, higher density reaction cell arrangements are possible. FIG. 21 depicts an exemplary matrix design wherein a first elastomeric layer (1st layer) and a second elastomeric layer (2d layer) each having fluid channels formed therein. For example, a reagent fluid channel in the first layer is connected to a reagent fluid channel in the second layer through a via, while the second layer also has sample channels therein, the sample channels and the reagent channels terminating in sample and reagent chambers, respectively. The sample and reagent chambers are in fluid communication with each other through an interface channel that has an interface valve associated therewith to control fluid communication between each of the chambers of a reaction cell. In use, the interface is first closed, then reagent is introduced into the reagent channel from the reagent inlet and sample is introduced into the sample channel through the sample inlet, containment valves are then closed to isolate each reaction cell from other reaction cells. Once the reaction cells are isolated, the interface valve is opened to cause the sample chamber and the reagent chamber to be in fluid communication with each other so that a desired reaction may take place.
  • Accordingly, a preferred aspect of the invention provides for a microfluidic device for reacting M number of different samples with N number of different reagents comprising: a plurality of reaction cells, each reaction cell comprising a sample chamber and a reagent chamber, the sample chamber and the reagent chamber being in fluid communication through an interface channel having an interface valve associated therewith for controlling fluid communication between the sample chamber and the reagent chamber; a plurality of sample inlets each in fluid communication with the sample chambers; a plurality of reagent inlets each in fluid communication with the reagent chambers; wherein one of the sample inlets or reagent inlets is in fluid communication with one of the sample chambers or one of the reagent chambers, respectively, through a via. Certain embodiments include having the reaction cells be formed within an elastomeric block formed from a plurality of layers bonded together and the interface valve is deflectable membrane; having the sample inlets be in communication with the sample chamber through a sample channel and the reagent inlet is in fluid communication with the reagent chamber through a reagent channel, a portion of the sample channel and a portion of the reagent channel being oriented about parallel to each other and each having a containment valve associated therewith for controlling fluid communication therethrough; having the valve associated with the sample channel and the valve associated with the reagent channel are in operable communication with each other through a common containment control channel; having the containment common control channel located along a line about normal to one of the sample channel or the reagent channel
  • Another aspect of the invention provides for a method for making a feature in an elastomeric block comprising the steps of: providing a first elastomeric layer; applying a photoresist layer upon a surface of the first elastomeric layer; applying a light pattern to the photoresist layer to form a pattern of reacted photoresist material; removing unreacted photoresist material leaving the pattern of reacted photoresist upon the surface of the first elastomeric layer; applying an etching reagent to the first elastomeric surface to etch the surface of the first elastomeric layer not covered by the pattern of reacted photoresist material thereby removing regions of the first elastomeric layer not covered by the pattern of reacted photoresist and leaving a pattern of the elastomeric layer corresponding to the pattern of reacted photoresist material. In certain preferred embodiments of the method include having a step of removing the pattern of reacted photoresist material; having the removing is caused by applying an adhesive tape to the surface of the elastomeric layer and the pattern of reacted photoresist material, then separating the adhesive tape from the elastomeric layer while some or all of the pattern of reacted photoresist material is removed from the surface of the elastomeric layer; having the photo resist be SU8; having the etching reagent comprises tetrabutylammoniumfluoride-trihydrate; having the feature be a via; having the elastomeric block comprise a plurality of elastomeric layers bonded together, wherein two or more elastomeric layers have recesses formed therein and one recess of one elastomeric layer is in communication with a recess of another elastomeric layer through the via.
  • The microfluidic devices of the present invention may be further integrated into the carrier devices described in copending and co-owned U.S. patent application Ser. No. 60/557,715 by Unger filed on Mar. 29, 2004, which is herein incorporated for all purposes. The carrier of Unger provides on-board continuous fluid pressure to maintain valve closure away from a source of fluid pressure, e.g., house air pressure. Unger further provides for an automated system for charging and actuating the valves of the present invention as described therein.
  • B. Exemplary Designs and Uses
  • FIG. 1A provides an illustration of one exemplary matrix device. This device 100 comprises seven vertical flow channels 102 and seven horizontal flow channels 104 that intersect to form an array of 49 different intersections or reaction sites 106. This particular device thus enables seven samples to be reacted with seven different reagents or sets of reagents. Column valves 110 that regulate solution flow in the vertical direction can be controlled by control channels 118 that can all be actuated at a single inlet 114.
  • Similarly, row valves 108 regulate solution flow in the horizontal direction; these are controlled by control channels 116 that are actuated by a single control inlet 112. As shown in FIG. 1A, the control channels 116 that regulate the row valves 108 vary in width depending upon location. When a control channel 116 crosses a vertical flow channel 102, the control channel 116 is sufficiently narrow that when it is actuated it does not deflect into the vertical flow channel 102 to reduce substantially solution flow therethrough. However, the width of the control channel 116 is increased when it overlays one of the horizontal flow channels 104; this makes the membrane of the control channel sufficiently large to block solution flow through the horizontal flow channel 104.
  • In operation, reagents R1-R7 are introduced into their respective horizontal flow channels 104 and samples S1-S7 are injected into their respective vertical flow channels 102. The reagents in each horizontal flow channel 104 thus mix with the samples in each of the vertical flow channels 102 at the intersections 106, which in this particular device are in the shape of a well or chamber. Thus, in the specific case of a nucleic acid amplification reaction, for example, the reagents necessary for the amplification reaction are introduced into each of the horizontal flow channels 104. Different nucleic acid templates are introduced into the vertical flow channels 102. In certain analyses, the primer introduced as part of the reagent mixture that is introduced into each of the horizontal flow channels 104 might differ between flow channels. This allows each nucleic acid template to be reacted with a number of different primers.
  • FIGS. 1B-E show enlarged plan views of adjacent reaction sites in the device depicted in FIG. 1A to illustrate more specifically how the device operates during an analysis. For the purposes of clarity, the intersections 106 are not shown in the form of reaction wells and control channels 116, 118 have been omitted, with just the column and row valves 110, 108 being shown (rectangular boxes). As shown in FIG. 1B, an analysis is commenced by closing column valves 110 and opening row valves 108 to allow solution flow through horizontal flow channel 104 while blocking flow through vertical flow channels 102. Reagent R1 is then introduced into horizontal flow channel 104 and flowed completely through the length of the horizontal flow channel 104 such that all the reaction sites 106 are filled. Solution flow through horizontal channel 104 can be achieved by an external pump, but more typically is achieved by incorporating a peristaltic pump into the elastomeric device itself as described in detail in Unger et al. (2000) Science 288: 113-116, and PCT Publication WO 01/01025, for example.
  • Once R1 has been introduced, row valves 108 are closed and column valves 102 opened (see FIG. 1C). This allows samples S1 and S2 to be introduced into vertical flow channels 102 and to flow through their respective flow channels. As the samples flow through the vertical flow channels 102, they expel R1 from the reaction sites 106, thus leaving sample at reaction sites 106. Then, as shown in FIG. 1D, row valves 108 are opened to allow S1 and S2 to diffuse and mix with R1. Thus, a mixture of sample and reactant (R1 S1 and R1 S2) is obtained in the region of each intersection or reaction site 106. After allowing a sufficient time for S1 and S2 to diffuse with R1, all row and column valves 108, 110 are closed to isolate S1 and S2 within the region of their respective reaction sites 106 and to prevent intermixing of S1 and S2 (see FIG. 1E). The mixtures are then allowed to react and the reactions detected by monitoring the intersection 106 or the cross-shaped region that includes the intersection 106. For analyses requiring heating (e.g., thermocycling during amplification reactions), the device is placed on a heater and heated while the samples remain isolated.
  • A modified version of the device shown in FIG. 1A is shown in FIG. 1F. The general structure bears many similarities with that depicted in FIG. 1A, and common elements in both figures share the same reference numbers. The device 150 illustrated in FIG. 1F differs in that pairs of horizontal flow channels 104 are joined to a common inlet 124. This essentially enables duplicate sets of reagents to be introduced into two adjacent flow channels with just a single injection into inlet 124. The use of a common inlet is further extended with respect to the vertical flow channels 102. In this particular example, each sample is introduced into five vertical flow channels 102 with a single injection into sample inlet 120. Thus, with this particular device, there are essentially ten replicate reactions for each particular combination of sample and reagent. Of course, the number of replicate reactions can be varied as desired by altering the number of vertical and/or horizontal flow channels 102, 104 that are joined to a common inlet 120, 124.
  • The device shown in FIG. 1F also includes a separate control channel inlet 128 that regulates control channel 130 that can be used to govern solution flow toward outlets 132 and another control channel inlet 132 that regulates control channel 134 that regulates solution flow to outlets 136. Additionally, device 150 incorporates guard channels 138. In this particular design, the guard channels 138 are formed as part of control channels 116. As indicated supra, the guard channels 138 are smaller than the row valves 108; consequently, the membranes of the guard channels 138 are not deflected into the underlying horizontal flow channels 104 such that solution flow is disrupted.
  • Finally, the design shown in FIG. 1F differs in that reaction does not occur in wells at the intersection of the horizontal and vertical flow lines, but in the intersection itself.
  • V. Blind Channel Designs
  • A. General
  • Devices utilizing a blind channel design have certain features. First, the devices include one or more flow channels from which one or more blind channels branch. As indicated above, the end region of such channels can serve as a reaction site. A valve formed by an overlaying flow channel can be actuated to isolate the reaction site at the end of the blind channel. The valves provide a mechanism for switchably isolating the reaction sites.
  • Second, the flow channel network in communication with the blind channels is configured such that all or most of the reaction sites can be filled with a single or a limited number of inlets (e.g., less than 5 or less than 10). The ability to fill a blind flow channel is made possible because the devices are made from elastomeric material. The elastomeric material is sufficiently porous such that air within the flow channels and blind channels can escape through these pores as solution is introduced into the channels. The lack of porosity of materials utilized in other microfluidic devices precludes use of the blind channel design because air in a blind channel has no way to escape as solution is injected.
  • A third characteristic is that one or more reagents are non-covalently deposited on a base layer of elastomer during manufacture (see infra for further details on the fabrication process) within the reaction sites. The reagent(s) are non-covalently attached because the reagents are designed to become dissolved when sample is introduced into the reaction site. To maximize the number of analyses, a different reactant or set of reactants is deposited at each of the different reaction sites.
  • Certain blind channel devices are designed such that the reaction sites are arranged in the form of an array.
  • Thus, in those blind channel devices designed for conducting nucleic acid amplification reactions, for example, one or more of the reagents required for conducting the extension reaction are deposited at each of the reaction sites during manufacture of the device. Such reagents include, for example, all or some of the following: primers, polymerase, nucleotides, cofactors, metal ions, buffers, intercalating dyes and the like. To maximize high throughput analysis, different primers selected to amplify different regions of DNA are deposited at each reaction site. Consequently, when a nucleic acid template is introduced into the reaction sites via inlet, a large number of extension reactions can be performed at different segments of the template. Thermocycling necessary for an amplification reaction can be accomplished by placing the device on a thermocycling plate and cycling the device between the various required temperatures.
  • The reagents can be immobilized in a variety of ways. For example, in some instances one or more of the reagents are non-covalently deposited at the reaction site, whereas in other instances one or more of the reagents is covalently attached to the substrate at the reaction site. If covalently attached, the reagents can be linked to the substrate via a linker. A variety of linker types can be utilized such as photochemical/photolabile linkers, themolabile linkers, and linkers that can be cleaved enzymatically. Some linkers are bifunctional (i.e., the linker contains a functional group at each end that is reactive with groups located on the element to which the linker is to be attached); the functional groups at each end can be the same or different. Examples of suitable linkers that can be used in some assays include straight or branched-chain carbon linkers, heterocyclic linkers and peptide linkers. A variety of types of linkers are available from Pierce Chemical Company in Rockford, Ill. and are described in EPA 188,256; U.S. Pat. Nos. 4,671,958; 4,659,839; 4,414,148; 4,669,784; 4,680,338, 4,569, 789 and 4,589,071, and by Eggenweiler, H. M, Pharmaceutical Agent Discovery Today 1998, 3, 552. NVOC (6 nitroveratryloxycarbonyl) linkers and other NVOC-related linkers are examples of suitable photochemical linkers (see, e.g., WO 90/15070 and WO 92/10092). Peptides that have protease cleavage sites are discussed, for example, in U.S. Pat. No. 5,382,513.
  • B. Exemplary Designs and Uses
  • FIG. 2 is a simplified plan view of one exemplary device utilizing the blind channel design. The device 200 includes a flow channel 204 and a set of branch flow channels 206 branching therefrom that are formed in an elastomeric substrate 202. Each branch flow channel 206 terminates in a reaction site 208, thereby forming an array of reaction sites. Overlaying the branch flow channels 206 is a control channel 210 that is separated from the branch flow channels 206 by membranes 212. Actuation of control channel 210 causes membranes 212 to deflect into the branch flow channels 206 (i.e., to function as a valve), thus enabling each of the reaction sites 208 to be isolated from the other reaction sites.
  • Operation of such a device involves injecting a test sample into flow channel 204 with solution subsequently flowing into each of branch channels 206. Once the sample has filled each branch channel 206, control channel 210 is actuated to cause activation of valves/membranes 212 to deflect into branch channels 206, thereby sealing off each of reaction sites 208. As the sample flows into and remains in reaction sites 208, it dissolves reagents previously spotted at each of the reaction sites 208. Once dissolved, the reagents can react with the sample. Valves 212 prevent the dissolved reagents at each reaction site 208 from intermixing by diffusion. Reaction between sample and reagents are then detected, typically within reaction site 208. Reactions can optionally be heated as described in the temperature control section infra.
  • FIG. 3A illustrates an example of a somewhat more complex blind flow channel design. In this particular design 300, each of a set of horizontal flow channels 304 are connected at their ends to two vertical flow channels 302. A plurality of branch flow channels 306 extend from each of the horizontal flow channels 304. The branch flow channels 304 in this particular design are interleaved such that the branch channel 306 attached to any given horizontal flow channel 304 is positioned between two branch channels 306 joined to an immediately adjacent horizontal flow channel 304, or positioned between a branch flow channel 306 joined to an immediately adjacent flow channel 304 and one of the vertical flow channels 302. As with the design depicted in FIG. 3A, each branch flow channel 306 terminates in a reaction site 308. Also consistent with the design shown in FIG. 3A, a control channel 310 overlays each of the branch channels and is separated from the underlying branch channel by membrane 312. The control channel is actuated at port 316. The vertical and horizontal flow channels 302, 304 are interconnected such that injection of sample into inlet 314 allows solution to flow throughout the horizontal and vertical flow channel network and ultimately into each of the reaction sites 308 via the branch flow channels 306.
  • Hence, in operation, sample is injected into inlet to introduce solution into each of the reaction sites. Once the reaction sites are filled, valves/membranes are actuated to trap solution within the reaction sites by pressurizing the control channels at port. Reagents previously deposited in the reaction sites become resuspended within the reaction sites, thereby allowing reaction between the deposited reagents and sample within each reaction site. Reactions within the reaction sites are monitored by a detector. Again, reactions can optionally be controllably heated according to the methods set forth in the temperature control section below.
  • An even more complicated version of the general design illustrated in FIG. 3A is shown in FIG. 3B. The device shown in FIG. 3B is one in which the unit organization of the horizontal and branch flow channels 302 shown in FIG. 3A is repeated multiple times. The device shown in FIG. 3B further illustrates the inclusion of guard channels 320 in those devices to be utilized in applications that involve heating (e.g., thermocycling). An exemplary orientation of the guard channels 320 with respect to the flow channels 304 and branch channels 306 is shown in the enlarged view depicted in FIG. 3C. The guard channels 320 overlay the branch flow channels 306 and reaction sites 308. As discussed above, water is flowed through the guard channels 320 during heating of the device 300 to increase the local concentration of water in the device, thereby reducing evaporation of water from solution in the flow channels 306 and reaction sites 308.
  • The features of blind channel devices discussed at the outset of this section minimizes the footprint of the device and enable a large number of reaction sites to be formed on the device and for high densities to be obtained. For example, devices of this type having 2500 reaction sites can readily be manufactured to fit on a standard microscope slides (25 mm×75 mm). The foregoing features also enable very high densities of reaction sites to be obtained with devices utilizing the blind channel design. For example, densities of at least 50, 60, 70, 80, 90 or 100 reaction sites/cm2 or any integral density value therebetween can be readily obtained. However, certain devices have even higher densities ranging, for example, between 100 to 4000 reaction sites/cm2, or any integral density value therebetween. For instance, some devices have densities of at least 100, 150, 200, 250, 300, 400, 500, 600, 700, 800, 900 or 1000 sites/cm2. Devices with very high densities of at least, 2000, 3000, or 4000 sites/cm2 are also obtainable. Such high densities directly translate into a very large number of reaction sites on the device. Devices utilizing the blind channel architecture typically have at least 10-100 reaction sites, or any integral number of sites therebetween. More typically, the devices have at least 100-1,000 reaction sites, or any integral number of sites therebetween. Higher density devices can have even more reaction sites, such as at least 1,000-100,000 reaction sites, or any integral number of sites therebetween. Thus, certain devices have at least 100; 500; 1,000; 2,000; 3,000; 4,000; 5,000; 6,000; 7,000; 8,000; 9,000; 10,000; 20,000; 30,000; 40,000; 50,000; or 100,000 reaction sites depending upon the overall size of the device.
  • The large number of reaction sites and densities that can be obtained is also a consequence of the ability to fabricate very small wells or cavities. For example, the cavities or wells typically have a volume of less than 50 nL; in other instances less than 40 nL, 30 nL, 20 nL or 10 nL; and in still other instances less than 5 nL or 1 nL. As a specific example, certain devices have wells that are 300 microns long, 300 microns wide and 10 microns deep.
  • The blind channel devices provided herein can utilize certain design features and methodologies discussed in PCT Applications PCT/US01/44549 (published as WO 02/43615) and PCT/US02/10875 (published as WO 02/082047), including, for example, strategies for filling dead-ended channels, liquid priming, pressurized outgas priming, as well as various strategies for displacing gas during the filling of microfluidic channels. Both of these PCT publications are incorporated herein by reference in their entirety for all purposes.
  • VI. Hybrid Designs
  • Still other devices are hybrids of the matrix and blind fill designs. The design of devices of this type is similar to the blind channel device shown in FIG. 3A, except that each horizontal flow channel is connected to its own sample inlet port(s) and the horizontal flow channels are not interconnected via vertical flow channels. Consequently, sample introduced into any given horizontal flow channel fills only that horizontal flow channel and reaction sites attached thereto. Whereas, in the blind flow channel device shown in FIG. 3A, sample can flow between the horizontal flow channels 304 via vertical flow channels 302.
  • An example of devices of this general device is shown in FIG. 4. Device 400 comprises a plurality of horizontal flow channels 404, each of which has a plurality of branch flow channels 406 extending from it and its own sample inlet 414. A control channel 410 overlays each of the branch flow channels 406 and membrane (valve) 412 separates the control channel 410 from the underlying branch flow channel 406. As with the blind flow channel design, actuation of the control channel at inlet 416 causes deflection of membranes 412 into the branch flow channels 406 and isolation of reaction sites 408. In a variation of this design, each horizontal flow channel 404 can include an inlet 414 at each end, thus allowing sample to be introduced from both ends.
  • In some instances, reagents are deposited at the reaction sites during manufacture of the device. This enables a large number of samples to be tested under a relatively large number of reaction conditions in a short period of time without requiring time-consuming additions of reagents as required with the matrix devices. Alternatively, reaction mixtures can be prepared prior to injection on the chip. Once the mixtures are injected, they can be analyzed or further treated (e.g., heated).
  • By injecting different samples into each of the horizontal flow channels, a large number of samples can be rapidly analyzed. Assuming reagents have been previously deposited at the reaction sites, the presence of the same reagent at each reaction site associated with any given horizontal flow channel provides a facile way to conduct a number of replicate reactions with each sample. If instead, the reagent at the reaction sites differ for any given flow channel, then each sample is essentially simultaneously exposed to a variety of different reaction conditions.
  • Thus, the devices provided herein are tailored for a variety of different types of investigations. If an investigation involves screening of a relatively large number of different samples under user controlled conditions (e.g., 100 samples against 100 user selected reagents), then the matrix devices provide a useful solution. If, however, the investigation involves analyzing one or a limited number of samples under a wide variety of reaction conditions (e.g., one sample against 10,000 reaction conditions), then the blind channel design is useful. Finally, if one wants to examine a relatively large number of samples against defined reaction conditions without having to inject reagents (e.g., 100 samples against 100 previously defined reagents), then the hybrid devices are useful.
  • VII. Temperature Control
  • A. Devices and Components
  • A number of different options of varying sophistication are available for controlling temperature within selected regions of the microfluidic device or the entire device. Thus, as used herein, the term temperature controller is meant broadly to refer to a device or element that can regulate temperature of the entire microfluidic device or within a portion of the microfluidic device (e.g., within a particular temperature region or at one or more junctions in a matrix of blind channel-type microfluidic device).
  • Generally, the devices are placed on a thermal cycling plate to thermal cycle the device. A variety of such plates are readily available from commercial sources, including for example the ThermoHybaid Px2 (Franklin, Mass.), MJ Research PTC-200 (South San Francisco, Calif.), Eppendorf Part# E5331 (Westbury, N.Y.), Techne Part# 205330 (Princeton, N.J.).
  • To ensure the accuracy of thermal cycling steps, in certain devices it is useful to incorporate sensors detecting temperature at various regions of the device. One structure for detecting temperature is a thermocouple. Such a thermocouple could be created as thin film wires patterned on the underlying substrate material, or as wires incorporated directly into the microfabricated elastomer material itself.
  • Temperature can also be sensed through a change in electrical resistance. For example, change in resistance of a thermistor fabricated on an underlying semiconductor substrate utilizing conventional techniques can be calibrated to a given temperature change. Alternatively, a thermistor could be inserted directly into the microfabricated elastomer material. Still another approach to detection of temperature by resistance is described in Wu et al. in “MEMS Flow Sensors for Nano-fluidic Applications”, Sensors and Actuators A 89 152-158 (2001), which is hereby incorporated by reference in its entirety. This paper describes the use of doped polysilicon structures to both control and sense temperature. For polysilicon and other semiconductor materials, the temperature coefficient of resistance can be precisely controlled by the identity and amount of dopant, thereby optimizing performance of the sensor for a given application.
  • Thermo-chromatic materials are another type of structure available to detect temperature on regions of an amplification device. Specifically, certain materials dramatically and reproducibly change color as they pass through different temperatures. Such a material could be added to the solution as they pass through different temperatures. Thermo-chromatic materials could be formed on the underlying substrate or incorporated within the elastomer material. Alternatively, thermo-chromatic materials could be added to the sample solution in the form of particles.
  • Another approach to detecting temperature is through the use of an infrared camera. An infrared camera in conjunction with a microscope could be utilized to determine the temperature profile of the entire amplification structure. Permeability of the elastomer material to radiation would facilitate this analysis.
  • Yet another approach to temperature detection is through the use of pyroelectric sensors. Specifically, some crystalline materials, particularly those materials also exhibiting piezoelectric behavior, exhibit the pyroelectric effect. This effect describes the phenomena by which the polarization of the material's crystal lattice, and hence the voltage across the material, is highly dependent upon temperature. Such materials could be incorporated onto the substrate or elastomer and utilized to detect temperature.
  • Other electrical phenomena, such as capacitance and inductance, can be exploited to detect temperature in accordance with embodiments of the present invention.
  • B. Verification of Accurate Thermocycling
  • As described in greater detail in the fabrication section infra, blind channel devices have a base layer onto which reagents are placed. The structure comprising the two layers containing the flow channels and control channels is overlayed on the base layer such that the flow channels are aligned with the deposited reagents. The other side of the base layer is then placed upon a substrate (e.g., glass). Usually, the reaction site at which reaction occurs is about 100-150 microns above the substrate/glass interface. Using known equations for thermal diffusivity and appropriate values for the elastomers and glass utilized in the device, one can calculate the time required for the temperature within the reaction site to reach the temperature the controller seeks to maintain. The calculated values shown in Table I demonstrate that temperature can rapidly be reached, even using elastomer and glass layers considerably thicker than utilized in devices in which the reaction site is approximately 100-150 microns (i.e., the typical distance for the devices described herein).
    TABLE 1
    Calculated heat diffusion lengths through PDMS and glass layers at the
    indicated time periods.
    1 second 10 seconds 100 seconds
    PDMS
    400 um 1.26 mm 4.0 mm
    Glass 640 um  2.0 mm 6.4 mm
  • FIG. 5 illustrates the rapidity at which the desired temperature is achieved using a blind channel device.
  • In another embodiment, temperature may be measured by using double stranded oligonucleotide polymers having known tms wherein an intercollating dye whose intercollation indicates the whether the oligonucleotide is hybridized or denatured, such as SYBR Green™ or ethidium bromide for example, wherein by introducing a solution containing the oligonucleotide with the dye into the chambers of the microfluidic device having an array of reaction chambers can be used to determine the extent to which the temperature of each chamber is consistent across the array. In this embodiment, as the temperature is raised above the tm, the intercalating dye changes its relation to the oligonucleotide upon it sdenataturation into a single stranded oligonucleotide. Alternatively, the if the temperature is above the tm and is lowered, an the intercollation of the dye into the now annealed oligonucleotide may be monitored. The use of the dye in essence provides for an “oligonucleotide thermometer” which changes a property, such as fluorescence, in response to a temperature change relative to the tm of the oligonucleotide. By designing or using oligonucleotides of a selected tm, the extent to which an array of reaction chambers change temperature in a similar manner can be determined.
  • VIII. Detection
  • A. General
  • A number of different detection strategies can be utilized with the microfluidic devices that are provided herein. Selection of the appropriate system is informed in part on the type of event and/or agent being detected. The detectors can be designed to detect a number of different signal types including, but not limited to, signals from radioisotopes, fluorophores, chromophores, electron dense particles, magnetic particles, spin labels, molecules that emit chemiluminescence, electrochemically active molecules, enzymes, cofactors, enzymes linked to nucleic acid probes and enzyme substrates.
  • Illustrative detection methodologies suitable for use with the present microfluidic devices include, but are not limited to, light scattering, multichannel fluorescence detection, UV and visible wavelength absorption, luminescence, differential reflectivity, and confocal laser scanning. Additional detection methods that can be used in certain application include scintillation proximity assay techniques, radiochemical detection, fluorescence polarization, fluorescence correlation spectroscopy (FCS), time-resolved energy transfer (TRET), fluorescence resonance energy transfer (FRET) and variations such as bioluminescence resonance energy transfer (BRET). Additional detection options include electrical resistance, resistivity, impedance, and voltage sensing.
  • Detection occurs at a “detection section,” or “detection region.” These terms and other related terms refer to the portion of the microfluidic device at which detection occurs. As indicated above, with devices utilizing the blind channel design, the detection section is generally the reaction site as isolated by the valve associated with each reaction site. The detection section for matrix-based devices is usually within regions of flow channels that are adjacent an intersection, the intersection itself, or a region that encompasses the intersection and a surrounding region.
  • The detection section can be in communication with one or more microscopes, diodes, light stimulating devices (e.g., lasers), photomultiplier tubes, processors and combinations of the foregoing, which cooperate to detect a signal associated with a particular event and/or agent. Often the signal being detected is an optical signal that is detected in the detection section by an optical detector. The optical detector can include one or more photodiodes (e.g., avalanche photodiodes), a fiber-optic light guide leading, for example, to a photomultiplier tube, a microscope, and/or a video camera (e.g., a CCD camera).
  • Detectors can be microfabricated within the microfluidic device, or can be a separate element. If the detector exists as a separate element and the microfluidic device includes a plurality of detection sections, detection can occur within a single detection section at any given moment. Alternatively, scanning systems can be used. For instance, certain automated systems scan the light source relative to the microfluidic device; other systems scan the emitted light over a detector, or include a multichannel detector. As a specific illustrative example, the microfluidic device can be attached to a translatable stage and scanned under a microscope objective. A signal so acquired is then routed to a processor for signal interpretation and processing. Arrays of photomultiplier tubes can also be utilized. Additionally, optical systems that have the capability of collecting signals from all the different detection sections simultaneously while determining the signal from each section can be utilized.
  • External detectors are usable because the devices that are provided are completely or largely manufactured of materials that are optically transparent at the wavelength being monitored. This feature enables the devices described herein to utilize a number of optical detection systems that are not possible with conventional silicon-based microfluidic devices.
  • A particularly preferred detector uses a CCD camera and an optical path that provides for a large field of view and a high numerical aperture to maximize the amount of light collected from each reaction chamber. In this regard, the CCD is used as an array of photodetectors wherein each pixel or group of pixels corresponds to a reaction chamber rather than being used to produce an image of the array. Thus, the optics may be altered such that image quality is reduced or defocused to increase the depth of field of the optical system to collect more light from each reaction chamber.
  • A detector can include a light source for stimulating a reporter that generates a detectable signal. The type of light source utilized depends in part on the nature of the reporter being activated. Suitable light sources include, but are not limited to, lasers, laser diodes and high intensity lamps. If a laser is utilized, the laser can be utilized to scan across a set of detection sections or a single detection section. Laser diodes can be microfabricated into the microfluidic device itself. Alternatively, laser diodes can be fabricated into another device that is placed adjacent to the microfluidic device being utilized to conduct a thermal cycling reaction such that the laser light from the diode is directed into the detection section.
  • Detection can involve a number of non-optical approaches as well. For example, the detector can also include, for example, a temperature sensor, a conductivity sensor, a potentiometric sensor (e.g., pH electrode) and/or an amperometric sensor (e.g., to monitor oxidation and reduction reactions).
  • A number of commercially-available external detectors can be utilized. Many of these are fluorescent detectors because of the ease in preparing fluorescently labeled reagents. Specific examples of detectors that are available include, but are not limited to, Applied Precision ArrayWoRx (Applied Precision, Issaquah, Wash.)).
  • B. Detection of Amplified Nucleic Acids
  • 1. Intercalation Dyes
  • Certain intercalation dyes that only fluoresce upon binding to double-stranded DNA can be used to detect double-stranded amplified DNA. Examples of suitable dyes include, but are not limited to, SYBR™ and Pico Green (from Molecular Probes, Inc. of Eugene, Oreg.), ethidium bromide, propidium iodide, chromomycin, acridine orange, Hoechst 33258, Toto-1, Yoyo-1, and DAPI (4′,6-diamidino-2-phenylindole hydrochloride). Additional discussion regarding the use of intercalation dyes is provided by Zhu et al., Anal. Chem. 66: 1941-1948 (1994), which is incorporated by reference in its entirety.
  • 2. FRET Based Detection Methods
  • Detection methods of this type involve detecting a change in fluorescence from a donor (reporter) and/or acceptor (quencher) fluorophore in a donor/acceptor fluorophore pair. The donor and acceptor fluorophore pair are selected such that the emission spectrum of the donor overlaps the excitation spectrum of the acceptor. Thus, when the pair of fluorophores are brought within sufficiently close proximity to one another, energy transfer from the donor to the acceptor can occur. This energy transfer can be detected.
  • FRET and template extension reactions. These methods generally utilize a primer labeled with one member of a donor/acceptor pair and a nucleotide labeled with the other member of the donor/acceptor pair. Prior to incorporation of the labeled nucleotide into the primer during an template-dependent extension reaction, the donor and acceptor are spaced far enough apart that energy transfer cannot occur. However, if the labeled nucleotide is incorporated into the primer and the spacing is sufficiently close, then energy transfer occurs and can be detected. These methods are particularly useful in conducting single base pair extension reactions in the detection of single nucleotide polymorphisms (see infra) and are described in U.S. Pat. No. 5,945,283 and PCT Publication WO 97/22719.
  • Quantitative RT-PCR. A variety of so-called “real time amplification” methods or “real time quantitative PCR” methods can also be utilized to determine the quantity of a target nucleic acid present in a sample by measuring the amount of amplification product formed during or after the amplification process itself. Fluorogenic nuclease assays are one specific example of a real time quantitation method which can be used successfully with the devices described herein. This method of monitoring the formation of amplification product involves the continuous measurement of PCR product accumulation using a dual-labeled fluorogenic oligonucleotide probe—an approach frequently referred to in the literature as the “TaqMan” method.
  • The probe used in such assays is typically a short (ca. 20-25 bases) polynucleotide that is labeled with two different fluorescent dyes. The 5′ terminus of the probe is typically attached to a reporter dye and the 3′ terminus is attached to a quenching dye, although the dyes can be attached at other locations on the probe as well. The probe is designed to have at least substantial sequence complementarity with the probe binding site on the target nucleic acid. Upstream and downstream PCR primers that bind to regions that flank the probe binding site are also included in the reaction mixture.
  • When the probe is intact, energy transfer between the two fluorophors occurs and the quencher quenches emission from the reporter. During the extension phase of PCR, the probe is cleaved by the 5′ nuclease activity of a nucleic acid polymerase such as Taq polymerase, thereby releasing the reporter from the polynucleotide-quencher and resulting in an increase of reporter emission intensity which can be measured by an appropriate detector.
  • One detector which is specifically adapted for measuring fluorescence emissions such as those created during a fluorogenic assay is the ABI 7700 manufactured by Applied Biosystems, Inc. in Foster City, Calif. Computer software provided with the instrument is capable of recording the fluorescence intensity of reporter and quencher over the course of the amplification. These recorded values can then be used to calculate the increase in normalized reporter emission intensity on a continuous basis and ultimately quantify the amount of the mRNA being amplified.
  • Additional details regarding the theory and operation of fluorogenic methods for making real time determinations of the concentration of amplification products are described, for example, in U.S. Pat. No. 5,210,015 to Gelfand, U.S. Pat. No. 5,538,848 to Livak, et al., and U.S. Pat. No. 5,863,736 to Haaland, as well as Heid, C. A., et al., Genome Research, 6: 986-994 (1996); Gibson, U. E. M, et al., Genome Research 6: 995-1001 (1996); Holland, P. M., et al., Proc. Natl. Acad. Sci. USA 88: 7276-7280, (1991); and Livak, K. J., et al., PCR Methods and Applications 357-362 (1995), each of which is incorporated by reference in its entirety.
  • Thus, as the amplification reaction progresses, an increasing amount of dye becomes bound and is accompanied by a concomitant increase in signal.
  • Intercalation dyes such as described above can also be utilized in a different approach to quantitative PCR methods. As noted above, these dyes preferentially bind to double stranded DNA (e.g., SYBR GREEN) and only generate signal once bound. Thus, as an amplification reaction progresses, an increasing amount of dye becomes bound and is accompanied by a concomitant increase in signal that can be detected.
  • Molecular Beacons: With molecular beacons, a change in conformation of the probe as it hybridizes to a complementary region of the amplified product results in the formation of a detectable signal. The probe itself includes two sections: one section at the 5′ end and the other section at the 3′ end. These sections flank the section of the probe that anneals to the probe binding site and are complementary to one another. One end section is typically attached to a reporter dye and the other end section is usually attached to a quencher dye.
  • In solution, the two end sections can hybridize with each other to form a hairpin loop. In this conformation, the reporter and quencher dye are in sufficiently close proximity that fluorescence from the reporter dye is effectively quenched by the quencher dye. Hybridized probe, in contrast, results in a linearized conformation in which the extent of quenching is decreased. Thus, by monitoring emission changes for the two dyes, it is possible to indirectly monitor the formation of amplification product. Probes of this type and methods of their use is described further, for example, by Piatek, A. S., et al., Nat. Biotechnol. 16: 359-63 (1998); Tyagi, S. and Kramer, F. R., Nature Biotechnology 14: 303-308 (1996); and Tyagi, S. et al., Nat. Biotechnol. 16: 49-53 (1998), each of which is incorporated by reference herein in their entirety for all purposes.
  • Invader: Invader assays (Third Wave Technologies, (Madison, Wis.)) are used for SNP genotyping and utilize an oligonucleotide, designated the signal probe, that is complementary to the target nucleic acid (DNA or RNA) or polymorphism site. A second oligonucleotide, designated the Invader Oligo, contains the same 5′ nucleotide sequence, but the 3′ nucleotide sequence contains a nucleotide polymorphism. The Invader Oligo interferes with the binding of the signal probe to the target nucleic acid such that the 5′ end of the signal probe forms a “flap” at the nucleotide containing the polymorphism. This complex is recognized by a structure specific endonuclease, called the Cleavase enzyme. Cleavase cleaves the 5′ flap of the nucleotides. The released flap binds with a third probe bearing FRET labels, thereby forming another duplex structure recognized by the Cleavase enzyme. This time the Cleavase enzyme cleaves a fluorophore away from a quencher and produces a fluorescent signal. For SNP genotyping, the signal probe will be designed to hybridize with either the reference (wild type) allele or the variant (mutant) allele. Unlike PCR, there is a linear amplification of signal with no amplification of the nucleic acid. Further details sufficient to guide one of ordinary skill in the art is provided by, for example, Neri, B. P., et al., Advances in Nucleic Acid and Protein Analysis 3826: 117-125, 2000).
  • Nasba: Nucleic Acid Sequence Based Amplification (NASBA) is a detection method using RNA as the template. A primer complementary to the RNA contains the sequence for the T7 promoter site. This primer is allowed to bind with the template RNA and Reverse Transcriptase (RT) added to generate the complementary strand from 3′ to 5′. RNase H is subsequently added to digest away the RNA, leaving single stranded cDNA behind. A second copy of the primer can then bind the single stranded cDNA and make double stranded cDNA. T7 RNA polymerase is added to generate many copies of the RNA from the T7 promoter site that was incorporated into the cDNA sequence by the first primer. All the enzymes mentioned are capable of functioning at 41° C. (See, e.g., Compton, J. Nucleic Acid Sequence-based Amplification, Nature 350: 91-91, 1991.)
  • Scorpion. This method is described, for example, by Thelwell N., et al. Nucleic Acids Research, 28: 3752-3761, 2000, which is hereby incorporated by reference in its entirety for all purposes, and which FIG. 20 depicts the scheme thereof, wherein Scorpion probing mechanism is as follows. Step 1: initial denaturation of target and Scorpion stem sequence. Step 2: annealing of Scorpion primer to target. Step 3: extension of Scorpion primer produces double-stranded DNA. Step 4: denaturation of double-stranded DNA produced in step 3. This gives a single-stranded target molecule with the Scorpion primer attached. Step 5: on cooling, the Scorpion probe sequence binds to its target in an intramolecular manner. This is favoured over the intermolecular binding of the complementary target strand. A Scorpion (as shown in FIG. 24) consists of a specific probe sequence that is held in a hairpin loop configuration by complementary stem sequences on the 5′ and 3′ sides of the probe. The fluorophore attached to the 5′-end is quenched by a moiety (normally methyl red) joined to the 3′-end of the loop. The hairpin loop is linked to the 5′-end of a primer via a PCR stopping sequence (stopper). After extension of the primer during PCR amplification, the specific probe sequence is able to bind to its complement within the same strand of DNA. This hybridization event opens the hairpin loop so that fluorescence is no longer quenched and an increase in signal is observed. The PCR stoping sequence prevents read-through, that could lead to opening of the hairpin loop in the absence of the specific target sequence. Such read-through would lead to the detection of non-specific PCR products, e.g. primer dimers or mispriming events.
  • 3. Capacitive DNA Detection
  • There is a linear relationship between DNA concentration and the change in capacitance that is evoked by the passage of nucleic acids across a 1-kHz electric field. This relationship has been found to be species independent. (See, e.g., Sohn, et al. (2000) Proc. Natl. Acad. Sci. U.S.A. 97: 10687-10690). Thus, in certain devices, nucleic acids within the flow channel (e.g., the substantially circular flow channel of FIG. 1 or the reaction chambers of FIG. 2) are subjected to such a field to determine concentration of amplified product. Alternatively, solution containing amplified product is withdrawn and then subjected to the electric field.
  • IX. Composition of Mixtures for Conducting Reactions
  • Reactions conducted with the microfluidic devices disclosed herein are typically conducted with certain additives to enhance the reactions. So, for example, in the case of devices in which reagents are deposited, these additives can be spotted with one or more reactants at a reaction site, for instance. One set of additives are blocking reagents that block protein binding sites on the elastomeric substrate. A wide variety of such compounds can be utilized including a number of different proteins (e.g., gelatin and various albumin proteins, such as bovine serum albumin) and glycerol.
  • A detergent additive can also be useful. Any of a number of different detergents can be utilized. Examples include, but are not limited to SDS and the various Triton detergents.
  • In the specific case of nucleic acid amplification reactions, a number of different types of additives can be included. One category are enhancers that promote the amplification reaction. Such additives include, but are not limited to, reagents that reduce secondary structure in the nucleic acid (e.g., betaine), and agents that reduce mispriming events (e.g., tetramethylammonium chloride).
  • It has also been found in conducting certain amplification reactions that some polymerases give enhanced results. For example, while good results were obtained with AmpliTaq Gold polymerase (Applied Biosystems, Foster City, Calif.) from Thermus aquaticus, improved reactions were in some instances obtained using DyNAzyme polymerase from Finnzyme, Espoo, Finland. This polymerase is from the thermophilic bacterium, Thermus brockianus. Other exemplary polymerases that can be utilized include, but are not limited to, rTH polymerase XL, which is a combination of Thermus thermophilus (Tth) and Thermococcus litoralis (Tli), hyperthermo-philic archaebacterium Pyrosoccus woesei (Pwo), and Tgo DNA Polymerase.
  • Further details regarding additives useful in conducting reactions with certain of the devices disclosed herein, including nucleic acid amplification reactions, are provided in Example 1 infra.
  • X. Exemplary Applications
  • Because the microfluidic devices provided herein can be manufactured to include a large number of reaction sites, the devices are useful in a wide variety of screening and analytical methods. In general, the devices can be utilized to detect reactions between species that react to form a detectable signal, or a product that upon interaction with another species generates a detectable signal. In view of their use with various types of temperature control systems, the devices can also be utilized in a number of different types of analyses or reactions requiring temperature control.
  • A. Nucleic Acid Amplification Reactions
  • The devices disclosed herein can be utilized to conduct essentially any type of nucleic acid amplification reaction. Thus, for example, amplification reactions can be linear amplifications, (amplifications with a single primer), as well as exponential amplifications (i.e., amplifications conducted with a forward and reverse primer set).
  • When the blind channel type devices are utilized to perform nucleic acid amplification reactions, the reagents that are typically deposited within the reaction sites are those reagents necessary to perform the desired type of amplification reaction. Usually this means that some or all of the following are deposited, primers, polymerase, nucleotides, metal ions, buffer, and cofactors, for example. The sample introduced into the reaction site in such cases is the nucleic acid template. Alternatively, however, the template can be deposited and the amplification reagents flowed into the reaction sites. As discussed supra, when the matrix device is utilized to conduct an amplification reaction, samples containing nucleic acid template are flowed through the vertical flow channels and the amplification reagents through the horizontal flow channels or vice versa.
  • While PCR is perhaps the best known amplification technique. The devices are not limited to conducting PCR amplifications. Other types of amplification reactions that can be conducted include, but are not limited to, (i) ligase chain reaction (LCR) (see Wu and Wallace, Genomics 4: 560 (1989) and Landegren et al., Science 241: 1077 (1988)); (ii) transcription amplification (see Kwoh et al., Proc. Natl. Acad. Sci. USA 86: 1173 (1989)); (iii) self-sustained sequence replication (see Guatelli et al., Proc. Nat. Acad. Sci. USA, 87: 1874 (1990)); and (iv) nucleic acid based sequence amplification (NASBA) (see, Sooknanan, R. and Malek, L., BioTechnology 13: 563-65 (1995)). Each of the foregoing references are incorporated herein by reference in their entirety for all purposes.
  • Detection of the resulting amplified product can be accomplished using any of the detection methods described supra for detecting amplified DNA.
  • B. SNP Analysis and Genotyping
  • 1. General
  • Many diseases linked to genome modifications, either of the host organism or of infectious organisms, are the consequence of a change in a small number of nucleotides, frequently involving a change in a single nucleotide. Such single nucleotide changes are referred to as single nucleotide polymorphisms or simply SNPs, and the site at which the SNP occurs is typically referred to as a polymorphic site. The devices described herein can be utilized to determine the identify of a nucleotide present at such polymorphic sites. As an extension of this capability, the devices can be utilized in genotyping analyses. Genotyping involves the determination of whether a diploid organism (i.e., an organism with two copies of each gene) contains two copies of a reference allele (a reference-type homozygote), one copy each of the reference and a variant allele (i.e., a heterozygote), or contains two copies of the variant allele (i.e., a variant-type homozygote). When conducting a genotyping analysis, the methods of the invention can be utilized to interrogate a single variant site. However, as described further below in the section on multiplexing, the methods can also be used to determine the genotype of an individual in many different DNA loci, either on the same gene, different genes or combinations thereof.
  • Devices to be utilized for conducting genotyping analyses are designed to utilize reaction sites of appropriate size to ensure from a statistical standpoint that a copy of each of the two alleles for a diploid subject are present in the reaction site at a workable DNA concentrations. Otherwise, an analysis could yield results suggesting that a heterozygote is a homozygote simply because a copy of the second allele is not present at the reaction site. Table 2 below indicates the number of copies of the genome present in a 1 nl reaction volume at various exemplary DNA concentrations that can be utilized with the devices described herein.
    TABLE 2
    Number of genome copies present in a 1 nL volume
    at the indicated DNA concentration.
    Volume (nL) [DNA] (ug/uL) N
    1 0.33 100
    1 0.10 32
    1 0.05 16
    1 0.01 3
    1 0.003 1
  • As a general matter, due to stochastic proportioning of the sample, the copy number present before an amplification reaction is commenced determines the likely error in the measurement. Genotyping analyses using certain devices are typically conducted with samples having a DNA concentration of approximately 0.10 ug/uL, although the current inventors have run successful TaqMan reactions at concentrations in which there is a single genome per reaction site.
  • 2. Methods
  • Genotyping analyses can be conducted using a variety of different approaches. In these methods, it is generally sufficient to obtain a “yes” or “no” result, i.e., detection need only be able to answer the question whether a given allele is present. Thus, analyses can be conducted only with the primers or nucleotides necessary to detect the presence of one allele potentially at a polymorphic site. However, more typically, primers and nucleotides to detect the presence of each allele potentially at the polymorphic site are included. Examples of suitable approaches follow.
  • Single Base Pair Extension (SBPE) Reactions. SBPE reactions are one technique specifically developed for conducting genotyping analyses. Although a number of SPBE assays have been developed, the general approach is quite similar. Typically, these assays involve hybridizing a primer that is complementary to a target nucleic acid such that the 3′ end of the primer is immediately 5′ of the variant site or is adjacent thereto. Extension is conducted in the presence of one or more labeled non-extendible nucleotides that are complementary to the nucleotide(s) that occupy the variant site and a polymerase. The non-extendible nucleotide is a nucleotide analog that prevents further extension by the polymerase once incorporated into the primer. If the added non-extendible nucleotide(s) is(are) complementary to the nucleotide at the variant site, then a labeled non-extendible nucleotide is incorporated onto the 3′ end of the primer to generate a labeled extension product. Hence, extended primers provide an indication of which nucleotide is present at the variant site of a target nucleic acid. Such methods and related methods are discussed, for example, in U.S. Pat. Nos. 5,846,710; 6,004,744; 5,888,819; 5,856,092; and 5,710,028; and in WO 92/16657.
  • Detection of the extended products can be detected utilizing the FRET detection approach described for extension reactions in the detection section supra. Thus, for example, using the devices described herein, a reagent mixture containing a primer labeled with one member of a donor/acceptor fluorophore, one to four labeled non-extendible nucleotides (differentially labeled if more than one non-extendible nucleotide is included), and polymerase are introduced (or previously spotted) at a reaction site. A sample containing template DNA is then introduced into the reaction site to allow template extension to occur. Any extension product formed is detected by the formation of a FRET signal (see, e.g., U.S. Pat. No. 5,945,283 and PCT Publication WO 97/22719.). The reactions can optionally be thermocycled to increase signal using the temperature control methods and apparatus described above.
  • Ouantitative PCR. Genotyping analyses can also be conducted using the quantitative PCR methods described earlier. In this case, differentially labeled probes complementary to each of the allelic forms are included as reagents, together with primers, nucleotides and polymerase. However, reactions can be conducted with only a single probe, although this can create ambiguity as to whether lack of signal is due to absence of a particular allele or simply a failed reaction. For the typical biallelic case in which two alleles are possible for a polymorphic site, two differentially labeled probes, each perfectly complementary to one of the alleles are usually included in the reagent mixture, together with amplification primers, nucleotides and polymerase. Sample containing the target DNA is introduced into the reaction site. If the allele to which a probe is complementary is present in the target DNA, then amplification occurs, thereby resulting in a detectable signal as described in the detection above. Based upon which of the differential signal is obtained, the identity of the nucleotide at the polymorphic site can be determined. If both signals are detected, then both alleles are present. Thermocycling during the reaction is performed as described in the temperature control section supra.
  • B. Gene Expression Analysis
  • 1. General
  • Gene expression analysis involves determining the level at which one or more genes is expressed in a particular cell. The determination can be qualitative, but generally is quantitative. In a differential gene expression analysis, the levels of the gene(s) in one cell (e.g., a test cell) are compared to the expression levels of the same genes in another cell (control cell). A wide variety of such comparisons can be made. Examples include, but are not limited to, a comparison between healthy and diseased cells, between cells from an individual treated with one drug and cells from another untreated individual, between cells exposed to a particular toxicant and cells not exposed, and so on. Genes whose expression levels vary between the test and control cells can serve as markers and/or targets for therapy. For example, if a certain group of genes is found to be up-regulated in diseased cells rather than healthy cells, such genes can serve as markers of the disease and can potentially be utilized as the basis for diagnostic tests. These genes could also be targets. A strategy for treating the disease might include procedures that result in a reduction of expression of the up-regulated genes.
  • The design of the devices disclosed herein is helpful in facilitating a variety of gene expression analyses. Because the devices contain a large number of reaction sites, a large number of genes and/or samples can be tested at the same time. Using the blind flow channel devices, for instance, the expression levels of hundreds or thousands of genes can be determined at the same time. The devices also facilitate differential gene expression analyses. With the matrix design, for example, a sample obtained from a healthy cell can be tested in one flow channel, with a sample from a diseased cell run in an immediately adjacent channel. This feature enhances the ease of detection and the accuracy of the results because the two samples are run on the same device at the same time and under the same conditions.
  • 2. Sample Preparation and Concentration
  • To measure the transcription level (and thereby the expression level) of a gene or genes, a nucleic acid sample comprising mRNA transcript(s) of the gene(s) or gene fragments, or nucleic acids derived from the mRNA transcript(s) is obtained. A nucleic acid derived from an mRNA transcript refers to a nucleic acid for whose synthesis the mRNA transcript or a subsequence thereof has ultimately served as a template. Thus, a cDNA reverse transcribed from an mRNA, an RNA transcribed from that cDNA, a DNA amplified from the cDNA, an RNA transcribed from the amplified DNA, are all derived from the mRNA transcript and detection of such derived products is indicative of the presence and/or abundance of the original transcript in a sample. Thus, suitable samples include, but are not limited to, mRNA transcripts of the gene or genes, cDNA reverse transcribed from the mRNA, cRNA transcribed from the cDNA, DNA amplified from the genes, RNA transcribed from amplified DNA.
  • In some methods, a nucleic acid sample is the total mRNA isolated from a biological sample; in other instances, the nucleic acid sample is the total RNA from a biological sample. The term “biological sample”, as used herein, refers to a sample obtained from an organism or from components of an organism, such as cells, biological tissues and fluids. In some methods, the sample is from a human patient. Such samples include sputum, blood, blood cells (e.g., white cells), tissue or fine needle biopsy samples, urine, peritoneal fluid, and fleural fluid, or cells therefrom. Biological samples can also include sections of tissues such as frozen sections taken for histological purposes. Often two samples are provided for purposes of comparison. The samples can be, for example, from different cell or tissue types, from different individuals or from the same original sample subjected to two different treatments (e.g., drug-treated and control).
  • Any RNA isolation technique that does not select against the isolation of mRNA can be utilized for the purification of such RNA samples. For example, methods of isolation and purification of nucleic acids are described in detail in WO 97/10365, WO 97/27317, Chapter 3 of Laboratory Techniques in Biochemistry and Molecular Biology: Hybridization With Nucleic Acid Probes, Part 1. Theory and Nucleic Acid Preparation, (P. Tijssen, ed.) Elsevier, N.Y. (1993); Chapter 3 of Laboratory Techniques in Biochemistry and Molecular Biology: Hybridization With Nucleic Acid Probes, Part 1. Theory and Nucleic Acid Preparation, (P. Tijssen, ed.) Elsevier, N.Y. (1993); and Sambrook et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Press, N.Y., (1989); Current Protocols in Molecular Biology, (Ausubel, F. M. et al., eds.) John Wiley & Sons, Inc., New York (1987-1993). Large numbers of tissue samples can be readily processed using techniques known in the art, including, for example, the single-step RNA isolation process of Chomczynski, P. described in U.S. Pat. No. 4,843,155.
  • In gene expression analyses utilizing the devices that are described, a significant factor affecting the results is the concentration of the nucleic acid in the sample. At low copy number, noise is related to the square root of copy number. Thus, the level of error that is deemed acceptable governs the copy number required. The required copy number in the particular sample volume gives the required DNA concentration. Although not necessarily optimal, quantitation reactions can be conducted with an error level of up to 50%, but preferably is less. Assuming a 1 nanoliter volume, the DNA concentrations required to achieve a particular error level are shown in Table 3. As can be seen, 1 nanoliter volumes such as used with certain of the devices have sufficient copies of gene expression products at concentrations that are workable with microfluidic devices.
    TABLE 3
    Gene Expression-DNA Quantity
    Error (%) N (Copy No.) Volume (nL) [DNA] (10−12 M)
    2 2500 1 4.2
    10 100 1 0.17
    25 16 1 0.027
    50 4 1 0.0066
  • A further calculation demonstrates that the certain of the devices provided herein which utilize a 1 nanoliter reaction site contain sufficient DNA to achieve accurate expression results. Specifically, a typical mRNA preparation procedure yields approximately 10 ug of mRNA. It has been demonstrated that typically there are 1 to 10,000 copies of each mRNA per cell. Of the mRNAs that are expressed within any given cell, approximately the four most common messages comprise about 13% of the total mRNA levels. Thus, such highly expressed messages comprise 1.3 ug of mRNA (each is 4×10−12 mole or approximately 2.4×1012 copies). In view of the foregoing expression ranges, rare messages are expected to be present at a level of about 2×10−8 copies. If in a standard analysis the mRNA sample is dissolved in 10 ul, the concentration of a rare message is approximately 2×107 copies/ul; this concentration corresponds to 20,000 copies per 1 nl well (or 4×1011 M).
  • 3. Methods
  • Because expression analysis typically involves a quantitative analysis, detection is typically achieved using one of the quantitative real time PCR methods described above. Thus, if a TaqMan approach is utilized, the reagents that are introduced (or previously spotted) in the reaction sites can include one or all of the following: primer, labeled probe, nucleotides and polymerase. If an intercalation dye is utilized, the reagent mixture typically includes one or all of the following: primer, nucleotides, polymerase, and intercalation dye.
  • D. Multiplexing
  • The array-based devices described herein (see, e.g., FIGS. 1A, 1F, 2, 3A and 3B and accompanying text) are inherently designed to conduct a large number of amplification reactions at the same time. This feature, however, can readily be further expanded upon by conducting multiple analyses (e.g., genotyping and expression analyses) within each reaction site.
  • Multiplex amplifications can even be performed within a single reaction site by, for example, utilizing a plurality of primers, each specific for a particular target nucleic acid of interest, during the thermal cycling process. The presence of the different amplified products can be detected using differentially labeled probes to conduct a quantitative RT-PCR reaction or by using differentially labeled molecular beacons (see supra). In such approaches, each differentially labeled probes is designed to hybridize only to a particular amplified target. By judicious choice of the different labels that are utilized, analyses can be conducted in which the different labels are excited and/or detected at different wavelengths in a single reaction. Further guidance regarding the selection of appropriate fluorescent labels that are suitable in such approaches include: Fluorescence Spectroscopy (Pesce et al., Eds.) Marcel Dekker, New York, (1971); White et al., Fluorescence Analysis: A Practical Approach, Marcel Dekker, New York, (1970); Berlman, Handbook of Fluorescence Spectra of Aromatic Molecules, 2nd ed., Academic Press, New York, (1971); Griffiths, Colour and Constitution of Organic Molecules, Academic Press, New York, (1976); Indicators (Bishop, Ed.). Pergamon Press, Oxford, 19723; and Haugland, Handbook of Fluorescent Probes and Research Chemicals, Molecular Probes, Eugene (1992).
  • Multiple genotyping and expression analyses can optionally be conducted at each reaction site. If quantitative PCR methods such as TaqMan is utilized, then primers for amplifying different regions of a target DNA of interest are included within a single reaction site. Differentially labeled probes for each region are utilized to distinguish product that is formed.
  • E. Non-Nucleic Acid Analyses
  • While useful for conducting a wide variety of nucleic acid analyses, the devices can also be utilized in a number of other applications as well. As indicated earlier, the devices can be utilized to analyze essentially any interaction between two or more species that generates a detectable signal or a reaction product that can reacted with a detection reagent that generates a signal upon interaction with the reaction product.
  • Thus, for example, the devices can be utilized in a number of screening applications to identify test agents that have a particular desired activity. As a specific example, the devices can be utilized to screen compounds for activity as a substrate or inhibitor of one or more enzymes. In such analyses, test compound and other necessary enzymatic assay reagents (e.g., buffer, metal ions, cofactors and substrates) are introduced (if not previously deposited) in the reaction site. The enzyme sample is then introduced and reaction (if the test compound is a substrate) or inhibition of the reaction (if the test compound is an inhibitor) is detected. Such reactions or inhibition can be accomplished by standard techniques, such as directly or indirectly monitoring the loss of substrate and/or appearance of product.
  • Devices with sufficiently large flow channels and reaction sites can also be utilized to conduct cellular assays to detect interaction between a cell and one or more reagents. For instance, certain analyses involve determination of whether a particular cell type is present in a sample. One example for accomplishing this is to utilize cell-specific dyes that preferentially reaction with certain cell types. Thus, such dyes can be introduced into the reaction sites and then cells added. Staining of cells can be detected using standard microscopic techniques. As another illustration, test compounds can be screened for ability to trigger or inhibit a cellular response, such as a signal transduction pathway. In such an analysis, test compound is introduced into a site and then the cell added. The reaction site is then checked to detect formation of the cellular response.
  • Further discussion of related devices and applications of such devices is set forth in copending and commonly owned U.S. Provisional application No. 60/335,292, filed Nov. 30, 2001, which is incorporated herein by reference in its entirety for all purposes.
  • XI. Fabrication
  • A. General Aspects
  • As alluded to earlier, the microfluidic devices that are provided are generally constructed utilizing single and multilayer soft lithography (MSL) techniques and/or sacrificial-layer encapsulation methods. The basic MSL approach involves casting a series of elastomeric layers on a micro-machined mold, removing the layers from the mold and then fusing the layers together. In the sacrificial-layer encapsulation approach, patterns of photoresist are deposited wherever a channel is desired. These techniques and their use in producing microfluidic devices is discussed in detail, for example, by Unger et al. (2000) Science 288: 113-116, by Chou, et al. (2000) “Integrated Elastomer Fluidic Lab-on-a-chip-Surface Patterning and DNA Diagnostics, in Proceedings of the Solid State Actuator and Sensor Workshop, Hilton Head, S.C.; and in PCT Publication WO 01/01025, each of which is incorporated herein by reference in their entirety for all purposes.
  • In brief, the foregoing fabrication methods initially involve fabricating mother molds for top layers (e.g., the elastomeric layer with the control channels) and bottom layers (e.g., the elastomeric layer with the flow channels) on silicon wafers by photolithography with photoresist (Shipley SJR 5740). Channel heights can be controlled precisely by the spin coating rate. Photoresist channels are formed by exposing the photoresist to UV light followed by development. Heat reflow process and protection treatment is typically achieved as described by M. A. Unger, H.-P. Chou, T. Throsen, A. Scherer and S. R. Quake, Science (2000) 288: 113, which is incorporated herein by reference in its entirety. A mixed two-part-silicone elastomer (GE RTV 615) is then spun into the bottom mold and poured onto the top mold, respectively. Here, too, spin coating can be utilized to control the thickness of bottom polymeric fluid layer. The partially cured top layer is peeled off from its mold after baking in the oven at 80° C. for 25 minutes, aligned and assembled with the bottom layer. A 1.5-hour final bake at 80° C. is used to bind these two layers irreversibly. Once peeled off from the bottom silicon mother mold, this RTV device is typically treated with HCL (0.1N, 30 min at 80° C.). This treatment acts to cleave some of the Si—O—Si bonds, thereby exposing hydroxy groups that make the channels more hydrophilic.
  • The device can then optionally be hermetically sealed to a support. The support can be manufactured of essentially any material, although the surface should be flat to ensure a good seal, as the seal formed is primarily due to adhesive forces. Examples of suitable supports include glass, plastics and the like.
  • The devices formed according to the foregoing method result in the substrate (e.g., glass slide) forming one wall of the flow channel. Alternatively, the device once removed from the mother mold is sealed to a thin elastomeric membrane such that the flow channel is totally enclosed in elastomeric material. The resulting elastomeric device can then optionally be joined to a substrate support.
  • B. Devices Utilizing Blind Channel Design
  • 1. Layer Formation
  • Microfluidic devices based on the blind channel design in which reagents are deposited at the reaction sites during manufacture are typically formed of three layers. The bottom layer is the layer upon which reagents are deposited. The bottom layer can be formed from various elastomeric materials as described in the references cited above on MLS methods. Typically, the material is polydimethylsiloxane (PMDS) elastomer. Based upon the arrangement and location of the reaction sites that is desired for the particular device, one can determine the locations on the bottom layer at which the appropriate reagents should be spotted. Because PMDS is hydrophobic, the deposited aqueous spot shrinks to form a very small spot. The deposited reagents are deposited such that a covalent bond is not formed between the reagent and the surface of the elastomer because, as described earlier, the reagents are intended to dissolve in the sample solution once it is introduced into the reaction site.
  • The other two layers of the device are the layer in which the flow channels are formed and the layer in which the control and optionally guard channels are formed. These two layers are prepared according to the general methods set forth earlier in this section. The resulting two layer structure is then placed on top of the first layer onto which the reagents have been deposited. A specific example of the composition of the three layers is as follows (ration of component A to component B): first layer (sample layer) 30:1 (by weight); second layer (flow channel layer) 30:1; and third layer (control layer) 4:1. It is anticipated, however, that other compositions and ratios of the elastomeric components can be utilized as well.
  • During this process, the reaction sites are aligned with the deposited reagents such that the reagents are positioned within the appropriate reaction site. FIG. 6 is a set of photographs taken from the four corners of a device; these photographs demonstrate that the deposited reagents can be accurately aligned within the reaction sites utilizing the foregoing approach. These photographs show guard channels and reaction site located at the end of branch flow channel. The white circle indicates the location of the deposited reagent relative to the reaction site. As indicated, each reagent spot is well within the confines of the reaction site.
  • 2. Spotting
  • The reagents can be deposited utilizing any of a number of commercially available reagent spotters and using a variety of established spotting techniques. Examples of suitable spotter that can be utilized in the preparation of the devices include pin spotters, acoustic spotters, automated micropipettors, electrophoretic pumps, ink jet printer devices, ink drop printers, and certain osmotic pumps. Examples of commercially available spotters include: Cartesian Technologies MicroSys 5100 (Irvine, Calif.), Hitach SPBIO (Alameda, Calif.), Genetix Q-Array (United Kingdom), Affymetrix 417 (Santa Clara, Calif.) and Packard Bioscience SpotArray (Meriden, Conn.). In general, very small spots of reagents are deposited; usually spots of less than 10 nl are deposited, in other instances less than 5 nl, 2 nl or 1 nl, and in still other instances, less than 0.5 nl, 0.25 nl, or 0.1 nl.
  • Arrays of materials may also be formed by the methods described in Foder, et al., U.S. Pat. No. 5,445,934: titled “Array of oligonucleotides on a solid substrate”, which is herein incorporated by reference, wherein oligonucleotide probes, such as SNP probes, are synthesized in situ using spatial light directed photolithography. Such arrays would be used as the substrate or base of the microfluidic devices of the present invention such that the regions of the substrate corresponding to the reaction sites, for example, blind fill chambers, would be contain one, or preferably more than one, oligonucleotide probes arrayed in known locations on the substrate. In the case of a partitioning microfluidic structure, such as the one depicted in FIG. 15 herein, the reaction sites, depicted as square boxes along the serpentine, fluid channel, would contain a plurality of different SNP probes, preferably a collection of SNP probes suitable for identifying an individual from a population of individuals, and preferably wherein a plurality of reaction sites along the serpentine fluid channel, such that if a fluid sample containing nucleic acid sequences from a plurality of individuals where introduced into the serpentine flow channel, and a plurality of valve in communication with the serpentine flow channel such that when actuated causes the serpentine flow channel to be partitioned thereby isolating each reaction site from one another to contain a fraction of the fluid sample in each reaction site. Amplification of the components of the sample may be performed to increase the number of molecules, for example nucleic acid molecules, for binding to the array of SNP probes located within each reaction site. In some embodiments, each of the reaction sites along the serpentine fluid channel would be the same array, that is, have the same SNP probes arrayed, and in other embodiments, two or more of the reaction sites along the serpentine t fluid channel would have a different set of SNP probes. Other partitioning fluid channel architectures could also be used, for example, branched and/or branched branch systems, and so forth. Other arraying techniques, such as spotting described herein, may likewise be used to form the arrays located within the partitionable reactions sites along a serpentine or common, such as in branched, fluid channel(s).
  • The following examples are presented to further illustrate certain aspects of the devices and methods that are disclosed herein. The examples are not to be considered as limiting the invention.
  • EXAMPLE 1 Signal Strength Evaluations
  • I. Introduction
  • . The purpose of this set of experiments was to demonstrate that successful PCR reactions can be conducted with a microfluidic device of the design set forth herein with signal strength greater than 50% of the Macro TaqMan reaction.
  • II. Microfluidic Device
  • A three layer microfluidic device, fabricated using the MSL process, was designed and fabricated for conducting the experiments described in the following example. FIG. 7A shows a cross-sectional view of the device. As shown, the device 700 includes a layer 722 into which is formed the flow channels. This fluid layer 722 is sandwiched between an overlaying layer 720 that includes the control and guard layers and an underlying sealing layer 724. The sealing layer 724 forms one side of the flow channels. The resulting three-layer structure is affixed to a substrate 726 (in this example, a slide or coverslip), which provides structural stiffness, increases thermal conductivity, and helps to prevent evaporation from the bottom of microfluidic device 700.
  • FIG. 7B shows a schematic view of the design of the flow channels in flow layer 722 and of the control channels and guard channel in control/guard layer 720. Device 700 consists of ten independent flow channels 702, each with its own inlet 708, and branching blind channels 704, each blind channel 704 having a 1 nl reaction site 706. Device 700 contains a network of control lines 712, which isolate the reaction sites 706 when sufficient pressure is applied. A series of guard channels 716 are also included to prevent liquid from evaporating out of the reaction sites 706; fluid is introduced via inlet 718.
  • II. Experimental Setup
  • A PCR reaction using β-actin primers and TaqMan probe to amplify exon 3 of the β-actin gene from human male genomic DNA (Promega, Madison Wis.) was conducted in device 700. The TaqMan reaction consists of the following components: 1×TaqMan Buffer A (50 mM KCl, 10 mM Tris-HCl, 0.01M EDTA, 60 nM Passive Reference 1 (PR1), pH 8.3); 3.5-4.0 mM MgCl; 200 nM dATP, dCTP, dGTP, 400 nM dUTP; 300 nM β-actin forward primer and reverse primer; 200 nM FAM-labeled β-actin probe; 0.01 U/ul AmpEraseUNG (Applied Biosystems, Foster City, Calif.); 0.1-0.2 U/ul DyNAzyme (Finnzyme, Espoo, Finland); 0.5% Triton-x-100 (Sigma, St. Louis, Mo.); 0.8 ug/ul Gelatin (Calbiochem, San Diego, Calif.); 5.0% Glycerol (Sigma, St. Louis, Mo.); deionized H2O and male genomic DNA. The components of the reaction were added to produce a total reaction volume of 25 μl. Negative controls (Control) composed of all the TaqMan reaction components, except target DNA were included in each set of PCR reactions.
  • Once the TaqMan reaction samples and Control were prepared, they were injected into microfluidic device 700 by using a gel loading pipet tip attached to a 1 ml syringe. The pipet tip was filled with the reaction samples and then inserted into the fluid via 708. The flow channels 702 were filled by manually applying backpressure to the syringe until all the entire blind channels 704 and reaction sites 706 were filled. Control lines 712 were filled with deionized water and pressurized to 15-20 psi after all of the samples were loaded into the flow lines 702, 704. The pressurized control lines 712 were actuated to close the valves and isolate the samples in the 1 nl wells 706. The guard channels 716 were then filled with deionized water and pressurized to 5-7 psi. Mineral oil (15 ul) (Sigma) was placed on the flatplate of a thermocycler and then the microfluidic device/coverglass 700 was placed on the thermocycler. Micro fluidic device 700 was then thermocycled using an initial ramp and either a three-step or two-step thermocycling profile:
      • 1. Initial ramp to 95° C. and maintain for 1 minute (1.0° C./s to 75° C., 0.1° C./sec to 95° C.).
      • 2. Three step thermocycling for 40 cycles (92° C. for 30 sec., 54° C. for 30 sec., and 72° C. for 1 min) or;
      • 3. Two step thermocycling for 40 cycles (92° C. for 30 seconds and 60° C. for 60 sec.)
  • MicroAmp tubes (Applied Biosystems, Foster City, Calif.) with the remaining reaction mixture, designated Macro TaqMan reactions to distinguish them from reactions performed in the microfluidic device, were placed in the GeneAmp PCR System 9700 (Applied Biosystems, Foster City, Calif.) and thermocycled in the 9600 mode. The Macro TaqMan reactions served as macroscopic controls for the reactions performed in the micro fluidic device. The thermocycling protocol was set to match that of the microfluidic device, except that the initial ramp rate was not controlled for the Macro TaqMan reactions.
  • Once thermocycling was completed, the control and guard lines were depressurized and the chip was transferred onto a glass slide (VWR, West Chester, Pa.). The chip was then placed into an Array WoRx Scanner (Applied Precision, Issaquah, Wash.) with a modified carrier. The fluorescence intensity was measured for three different excitation/emission wavelengths: 475/510 nm (FAM), 510/560 nm (VIC), and 580/640 nm (Passive Reference 1 (PR1)). The Array Works Software was used to image the fluorescence in the micro fluidic device and to measure the signal and background intensities of each 1 nl well. The results were then analyzed using a Microsoft Excel file to calculate the FAM/PR1 ratio for β-actin TaqMan reactions. For conventional Macro TaqMan, positive samples for target DNA were determined using calculations described in the protocol provided by the manufacturer (TaqMan PCR Reagent Kit Protocol). The signal strength was calculated by dividing the FAM/PR1 ratio of the samples by the FAM/PR1 ratio of the controls. A successful reaction was defined as a sample ratio above the 99% confidence threshold level.
  • III. Results
  • Initially, AmpliTaq Gold (Applied Biosystems, Foster City, Calif.) was used in TaqMan reactions and FAM/PR1/Control ratios of 1.5-2.0 were produced, compared to Macro TaqMan reaction ratios of 5.0-14.0. Although results were positive, increased signal strength was desired. Therefore, the AmpliTaq Gold polymerase was substituted with DyNAzyme polymerase due to its increased thermostability, proofreading, and resistance to impurities. The standard Macro TaqMan DyNAzyme concentration of 0.025 U/ul was used in the microfluidic experiments. This polymerase change to DyNAzyme produced FAM/ROX/Control ratios of 3.5-5.8. The signal strength was improved, but it was difficult to achieve consistent results. Because it is know that some proteins stick to PDMS, the concentration of the polymerase was increased and surface modifying additives were included. Two increased concentrations of DyNAzyme were tested, 8×(0.2 U/ul) and 4×(0.1 U/ul) the standard concentration for Macro TaqMan, with 100 pg or 10 pg of genomic DNA per nl in the micro fluidic device. Gelatin, Glycerol, and 0.5% Triton-x-100 were added to prevent the polymerase from attaching to the PDMS. The results of the reactions in the micro fluidic device (chip) and the Macro TaqMan controls are shown in FIG. 8.
  • The microfluidic TaqMan reaction ratios range from 4.9-8.3, while the Macro TaqMan reactions range from 7.7-9.7. Therefore, the signal strength of the TaqMan reactions in chip is up to 87% of the Macro TaqMan reactions. There was no significant difference between 4× or 8×DyNAzyme. The results demonstrate that PCR reactions can be done with greater than 50% signal strength, when compared to the Macro TaqMan reactions, in the microfluidic devices. The results have been consistent through at least four attempts.
  • EXAMPLE 2 Spotting Reagents
  • I. Introduction
  • The purpose of the experiment was to demonstrate successful spotted PCR reactions in a microfluidic device. The term “spotted” in this context, refers to the placement of small droplets of reagents (spots) on a substrate that is then assembled to become part of a microfluidic device. The spotted reagents are generally a subset of the reagent mixture required for performing PCR.
  • II. Procedure
  • A. Spotting of Reagents
  • Routine spotting of reagents was performed via a contact printing process. Reagents were picked up from a set of source wells on metal pins, and deposited by contacting the pins to a target substrate. This printing process is further outlined in FIG. 9. As shown, reagents were picked up from a source (e.g., microtiter plates), and then printed by bringing the loaded pin into contact with the substrate. The wash step consists of agitation in deionized water followed by vacuum drying. The system used to print the reagent spots is a Cartesian Technologies MicroSys 5100 (Irvine, Calif.), employing TeleChem “ChipMaker” brand pins, although other systems can be used as described supra.
  • Pins employed are Telechem ChipMaker 4 pins, which incorporate an electro-milled slot (see FIG. 9) to increase the uptake volume (and hence the number of printable spots). Under the operating conditions employed (typically, 75% relative humidity and temperature approximately 25° C.), in excess of one hundred spots were printed per pin, per loading cycle. Under the conditions above, the volume of reagents spotted onto the PDMS substrate is on the order of 0.1 nL.
  • The dimensions of the pin tip are 125×125 μm. The final spot of dried reagent is substantially smaller than this (as small as 7 μm in diameter), yet the pin size defines a lower limit to the readily achievable spot spacing. The achievable spacing determines the smallest well-to-well pitch in the final device. Using such a device and the foregoing methods, arrays with spacings of 180 μm have been achieved. Arrays built into working chips tend to have spacings from 600 to 1300 microns.
  • Spotting was done using only one pin at a time. The system in use, however, has a pin head which can accommodate up to 32 pins. Printing a standard-size chip (array dimensions of order 20×25 mm) takes under 5 minutes.
  • B. Assembly of Spotted Chips
  • The flow and control layers of the PCR devices are assembled according to the normal MSL process described above. The microfluidic device design is the same as the one described in Example 1. In parallel, a substrate layer composed of 150 μm-thick PDMS with component ratio A:B of 30:1 is formed via spin-coating a blank silicon wafer, and then cured for 90 minutes at 80° C.
  • The cured blank substrate layer of PDMS (sealing/substrate layer 724 of FIG. 7A) serves as the target for reagent spotting. Patterns of spots are printed onto the substrate, which is still on the blank wafer. The reagents spotted for PCR reactions were primers and probes, specific to the particular gene to be amplified. The spotted reagent included a 1:1:1 volume ratio of 300 nM β-actin forward primer (FP), 300 nM β-actin reverse primer (RP), and 200 nM β-actin probe (Prb). In some cases, it is useful to further tune the chemistry via concentrating the spotted mixture. It has been found that adjusting the concentrations such that primer and probe concentrations are equal to, or slightly higher than, the normal macroscopic recipe value yields consistently good results. Therefore, the spotted reagent is concentrated to be 3 times and 4 times the concentration of the macro reaction. Concentration of the reagents is performed in a Centrivap heated and evacuated centrifuge and does not alter relative FP:RP:Prb ratios. The increased spot concentration results in the correct final concentration when the reagents are resuspended in a 1 nL reaction volume. Spotted reagents need not be limited to primers and probes; nor must all three (FP, RP and Prb) be spotted. Applications where only the probe, or even one of the primers, is spotted can be performed. Experiments have been conducted in which the sample primer/probe sets spotted were TaqMan β-actin and TaqMan RNAse-P.
  • Following the spotting process onto the substrate layer, the combined flow and control layers (i.e., layers 720 and 722 of FIG. 7A) were aligned with the spot pattern and brought into contact. A further bake at 80° C., for 60-90 minutes, was used to bond the substrate to the rest of the chip. After the chip has been assembled, the remaining components of the PCR reaction (described in Example 1) are injected into the flow channels of the chip and the chip is thermocycled as described in Example 1.
  • III. Results
  • PCR reactions have been successfully and repeatably performed using devices where primer (forward and reverse primers) and probe molecules are spotted. An example of data from a chip in which a reaction has been successfully performed is shown in FIG. 10. The spotted reagents have resulted in successful PCR reactions as defined in Example 1. Successful reactions have been performed using 2-stage and 3-stage thermocycling protocols.
  • EXAMPLE 3 Genotyping
  • I. Introduction
  • The purpose of the following experiments was to demonstrate that genotyping experiments can be conducted utilizing a microfluidic device or chip such as described herein. Specifically, these experiments were designed to determine if reactions conducted in the device have sufficient sensitivity and to ensure that other primer/probe sets, besides β-actin, can be performed in the microfluidic device.
  • II. Methods/Results
  • A. RNase P Experiment
  • RNase P TaqMan reactions (Applied Biosystems; Foster City, Calif.) were performed in a microfluidic device as described in Example 1 to demonstrate that other primer/probe sets produce detectable results. RNaseP reactions also require a higher level of sensitivity because the RNaseP primer/probe set detects a single copy gene (2 copies/genome) in contrast to the β-actin primer/probe set. The β-actin set detects a single copy β-actin gene and several pseudogenes, which collectively total approximately 17 copies per genome. The RNase P reactions were run with the same components as described in Example 1, with the exception that the β-actin primer/probe set was replaced with the RNase P primer/probe set. Further, the RnaseP primer/probe set was used at 4× the manufacturer's recommended value to enhance the fluorescence signal. The VIC dye was conjugated to the probe for RNase P and the analysis focused on VIC/PR1 ratios. The results of one of four experiments are shown in FIG. 11.
  • The VIC/PR1/Control ratios for the Macro TaqMan reactions are 1.23. The corresponding ratios for the TaqMan reactions in the microfluidic device are 1.11 and 1.21. The ratios of the genomic DNA samples in the microfluidic device are above the 99% confidence threshold level. Further, the signal strength of the TaqMan reactions in the microfluidic device is 50% and 93.7% of the Macro TaqMan reactions. The control TaqMan reactions in the microfluidic device have standard deviations of 0.006 and 0.012, demonstrating consistency in the reactions across the micro fluidic device. Therefore, it is determined that the TaqMan reactions in the chip are sensitive enough to detect 2 copies per genome.
  • B. DNA Dilution Experiment
  • To further determine the sensitivity of TaqMan reactions in the microfluidic device, dilutions of genomic DNA were tested using the β-actin primer/probe set. Reaction compositions were generally composed as described in Example 1 using 4×DyNAzyme and dilutions of genomic DNA. The genomic DNA was diluted down to 0.25 pg/nl, which corresponds to approximately 1 copy per nl. The result of one dilution series is shown in FIG. 12.
  • According to a Poisson distribution, 37% of the total number of wells should be negative if the average target number is one. Well numbers 5, 6 and 7 are below the calculated threshold and, therefore, negative. This suggests that the β-actin TaqMan reactions in micro fluidic chip can detect an average of one copy per nl. Therefore, the sensitivity of the reactions in the microfluidic device is sufficient to perform genotyping experiments.
  • C. Genotyping Experiment
  • Because TaqMan in the microfluidic device is capable of detecting low target numbers, preliminary testing of SNP (Single Nucleotide Polymorphism) genotyping was performed using the Predetermined Allelic Discrimination kit (Applied Biosystems; Foster City, Calif.) against the CYP2D6 P450 cytochrome gene. The kit contains one primer set and two probes; FAM labeled for the wildtype or reference allele, CYP2D6*1, and VIC labeled for the CYP2D6*3 mutant or variant allele. Positive controls, PCR products, for each allele along with genomic DNA were tested in the device using the same conditions as described in Example 1. The results from one experiment are shown in FIGS. 13 and 14. The experiment has been repeated at least three times to validate the results and to demonstrate reliability.
  • As shown in FIG. 13, the Al-1 (Allele 1, CYP2D6*1 wild type allele) and genomic DNA (100 pg/nl) produced an average VIC/PR1/Control ratio of 3.5 and 2.2, respectively, indicating that the genomic DNA was positive for the CYP2D6*1, wild type allele. These values are above the threshold limit for the reactions. The signal strength of the TaqMan reactions in the microfluidic device is 59% and 40% of the Macro TaqMan controls, respectively. Al-2 (Allele 2, CYP2D6*3 mutant or variant allele), which should be negative in the VIC channel, showed some signal over control (1.5), possibly due to FAM fluorescence leaking into the VIC channel of the detector. The leakage can be minimized with an improved detection process.
  • The Al-2 positive control gave an average FAM/PR1/Control ratio of 3.0, which was 37% of the Macro TaqMan signal and above the calculated threshold limit (see FIG. 14). The genomic samples were negative for the CYP2D6*3 mutant allele, an expected result since the frequency of the CYP2D6*3 allele is low. Again, it appears that there is some leakage of the Al-1, VIC probe into the FAM channel of the detector. Overall, the SNP detection reactions were successful in the microfluidic device.
  • EXAMPLE 4 Verification of PCR by Gel Electrophoresis
  • I. Introduction
  • As an alternative method to prove amplification of DNA was occurring in the microfluidic device, an experiment to detect PCR product by gel electrophoresis was performed. PCR reactions compositions were as described in Example 1, except the TaqMan probe was omitted and the β-actin forward primer was conjugated to FAM.
  • II. Procedure
  • A. Microfluidic Device
  • A three layer microfluidic device, fabricated using the MSL process, was designed and fabricated for conducting the experiments described in this example; FIG. 15 shows a schematic view of the design. The device 1500 generally consists of a sample region 1502 and a control region 1504. Sample region 1502 contains three hundred and forty-one 1 nl reaction sites 1508 represented by the rectangles arrayed along flow channel 1506, which includes inlet via 1510 and outlet via 1512. Control region 1504 contains three control flow channels 1514 each containing ten 1 nl reaction sites 1518, also represented by the rectangles and an inlet via 1516. A network of control lines 1522 isolate each reaction site 1508, 1518 when sufficient pressure is applied to inlet via 1524. A series of guard channels 1520 are included to prevent liquid from evaporating out of the reaction sites 1508, 1518. The device is a three-layer device as described in Example 1 (see FIG. 7A). The entire chip is placed onto a coverslip.
  • B. Experimental Setup
  • Microfluidic device 1500 was loaded and thermocycled using the 3 temperature profile described in Example 1. The remaining reaction sample was thermocycled in the GeneAmp 9700 with the same thermocycling profile as for microfluidic device 1500. The reaction products were recovered after thermocycling was completed. To recover the amplified DNA, 3 μl of water was injected into sample input via 1506 and 3-4 μl of product were removed from outlet via 1512. The reaction products from device 1500 and the Macro reaction were treated with 2 μl of ExoSAP-IT (USB, Cleveland, Ohio), which is composed of DNA Exonuclease 1 and Shrimp Alkaline Phosphatase, to remove excess nucleotides and primers. The Macro product was diluted from 1:10 to 1:106. The product from device 1500 was dehydrated and resuspended in 4 μl of formamide.
  • III. Results
  • Both products, along with negative controls were analyzed, on a polyacrylamide gel. FIG. 15 shows the gel electrophoresis results. The appropriate size DNA band of 294 base pairs in length is observed in FIG. 16.
  • The products from the Macro reactions are shown on the left hand side of the gel and correspond to about 294 base pairs, the expected size of the β-actin PCR product. The negative controls lack the PCR product. Similarly, the product derived from the device gave the expected β-actin PCR product. Therefore, target DNA was amplified in the micro fluidic device.
  • EXAMPLE 5 Massive Partitioning
  • The polymerase chain reaction (PCR) has become an essential tool in molecular biology. Its combination of sensitivity (amplification of single molecules of DNA), specificity (distinguishing single base mismatches) and dynamic range (105 with realtime instrumentation) make it one of the most powerful analytical tools in existence. We demonstrate here that PCR performance improves as the reaction volume is reduced: we have performed 21,000 simultaneous PCR reactions in a single microfluidic chip, in a volume of 90 pL per reaction and with single template molecule sensitivity.
  • FIGS. 17 a-17 d depict a single bank and dual bank partitioning microfluidic device. where multilayer soft lithography (MSL) (1), was used to create elastomeric microfluidic chips which use active valves to massively partition each of several liquid samples into a multitude of isolated reaction volumes. After injection of the samples into inlet 1703 which is in communication with branched partitioning channel system 1705 of microfluidic device 1701 (FIG. 17 b), 2400 90 pL volumes 1709 of each sample are isolated by closing valves 1707 spaced along (FIG. 17 d) simple microfluidic channels. The chip device is then thermocycled on a flat plate thermocycler and imaged in a commercially available fluorescence reader.
  • We assessed the performance of PCR in the chips by varying the concentration of template DNA and measuring the number of wells that gave a positive Taqman™ signal. We found that a digital amplification is observed when the average number of copies per well is low (FIGS. 18 a and 18 b). A mixture of robust positive and clearly negative signals is observed even when the average number of copies per well is below 1; this implies that even a single copy of target can give good amplification. The number of positive wells was consistent with the number of wells calculated to have ≧1 copy of target by the Poisson distribution (FIG. 19.). This result validates that this system gives amplification consistently even from a single copy of target. Fluorescent signal strengths from microfluidic Taqman™ PCR were comparable to macroscopic PCR reactions with the same DNA concentration—even though the macroscopic reactions contained >104 more template copies per reaction.
  • We believe that the primary source of this remarkable fidelity is the effective concentration of the target: a single molecule in a 90 pL volume is 55,000 times more concentrated than a single molecule in a 5 uL volume. Since the number of molecules of target, nt, does not change (i.e. nt=1) and the number of molecules that can produce side reactions, ns, (i.e. primer-dimers and non-complementary DNA sequences in the sample) is linearly proportional to volume (i.e. ns∝V), the ratio of target to side reactions is inversely proportional to volume: nt/ns∝1/V. Since side reactions are a primary cause of PCR failure (4), the advantage to reducing the volume of the reaction is clear.
  • PCR amplification from single copies of template has been previously reported (5). However, current methods that achieve reliable amplification from single copies in a macroscopic volumes often require altered thermocycling protocols (e.g. long extension times, many cycles), precautions against mispriming and non-specific amplification (e.g. “hot start” PCR (thermal activation of the polymerase), “booster” PCR, additives to reduce nonspecific hybridization, etc), and are almost always done with two rounds of PCR, where an aliquot of the first PCR is used as template in the second reaction. In contrast, this system achieves reliable amplification from single copies using standard conditions—off-the-shelf primers and probes and a single-round, standard thermocycling protocol. Being completely enclosed, it is also nearly invulnerable to environmental contamination. The ability to do massive numbers of PCR reactions simultaneously provides definite logistical, cost and time advantages compared to macroscopic volumes (1 chip with 21,000 reactions vs. 219 separate 96 well plates, and the associated time, equipment, and tracking infrastructure).
  • This principle of massive partitioning with a digital PCR readout may be used for absolute quantification of the concentration of target in a sample. It can be used, for example, to genotype a pooled sample of genomic DNA simply by counting the numbers of wells that give a positive for a particular allele, or plurality of alleles as described above. Due to the enhanced resistance to side reactions, it should also be useful in quantifying mutants in a background of wild-type DNA—a problem relevant in cancer detection. The general principle of concentration by partitioning may also be useful in other reactions where detection of single molecules, bacteria, viruses or cells is of interest (e.g. ELISA reactions for protein detection). Digital PCR is described by Brown, et al., U.S. Pat. No. 6,143,496, titled “Method of sampling, amplifying and quantifying segement of nucleic acid, polymerase chain reaction assembly having nanoliter-sized chambers and methods of filling chambers”, and by Vogelstein, et al, U.S. Pat. No. 6,446,706, titled “Digital PCR”, both of which are hereby incorporated by reference in their entirety. The small volumes achievable using microfluidics allow both a massive degree of parallelization and very high target-to-background concentration ratios. High target-to-background ratios allow single-molecule amplification fidelity. These factors suggest that for PCR, smaller really is better.
  • The invention provides for methods and devices for conducting digital PCR in a microfluidic environment comprising the steps of: providing a microfluidic device having a fluid channel therein, said fluid channel having two or more valves associated therewith, the valves, when actuated, being capable of partitioning the fluid channel into two or more reaction sites or chambers; introducing a sample containing at least one target nucleic acid polymer, actuating the valves to partition the fluid sample into two or more portions, wherein at least one portion contains a target nucleic acid polymer and another portion does not contain a target nucleic acid polymer, amplifying the target nucleic acid polymer, and, determining the number of portions of the fluid channel that contained the target molecule. In preferred embodiments, the microfluidic device comprises an elastomeric material, and more preferably, comprises at least one layer comprising an elastomeric material. In certain preferred embodiments, the microfluidic device further comprises a deflectable membrane wherein the deflectable membrane is deflectable into and out of the fluid channel to control fluid flow within the fluid channel and/or to partition one portion of the fluid channel from another, preferably wherein the deflectable membrane is integral to a layer of the microfluidic device having a channel or recess formed therein, and preferably wherein the deflectable membrane is formed where a first channel in a first layer is overlapped by a second channel in a second layer of the microfluidic device. In some embodiments, the sample fluid contains all of the components needed for conducting an amplification reaction, while in other embodiments, the microfluidic device contains at least one component of an amplification reaction prior to the introduction of the sample fluid. In some embodiments, the microfluidic device further comprises a detection reagent, preferably one or more nucleic acid polymers complimentary to a least a portion of the target nucleic acid polymer, preferably a plurality of different nucleic acid polymers spatially arrayed within a reaction site or chamber of the microfluidic device.
  • Amplification may be achieved by thermocycling reactions such as PCR, or by isothermal reactions, such as described by Van Ness et al., in U.S. patent application Ser. No. 10/196,740 which has publishes as U.S. 2003/0138800 A1, which is herein incorporated by reference in its entirety for the purpose of teaching an isothermic amplification scheme.
  • The invention further provides for a protein microcalorimetry assay using a fluorescent dye, for example SYBER green (TM), to measure the conformational changes of a protein, such as denaturation, especially if a protein's denaturation temperature changes when the protein interacts with another moiety such as a ligand or compound or other protein. An additional benefit of using SYBR Green (TM) is that it us used at lower wavelengths than other UV range dyes thus reducing background problems typically associated with many plastic materials.
  • REFERENCES
    • 1. Unger et al, Science 288, 113-116 (2000).
    • 2. The sample channels and control lines are loaded by “blind filling”—PDMS is sufficiently gas permeable that liquid pressurized at a few psi drives the gas out of the channels, leaving them completely filled with liquid. See Hansen et al, PNAS 99, 16531-16536 (2002)
    • 3. A 294 bp segment of the human β-actin gene was amplified using a 5′-exonuclease assay (Taqman). Forward and reverse primer sequences were 5′-TCACCCACACTGTGCCCATCTACGA3′ and 5′-CAGCGGAACCGCTCATTGCCAATGG3′, respectively. FIG. 1 b was taken with a TAMRA-based FRET probe, sequence 5′-(FAM)ATGCCC-X(TAMRA)-CCCCCATGCCATCCTGCGTp-3′. The data in FIG. 1 c was taken with a dark-quencher based probe, as large numbers of these primer-probe sets are becoming commercially available. Reactions contained 1×Taqman buffer A (50 mM KCl, 10 mM Tris-HCl, 0.01 M EDTA, 60 nM Passive Reference 1, pH 8.3), 4 mM MgCl2, 200 nM dATP, dCTP, dTTP, 400 nM dUTP, 300 nM forward primer, 300 nM reverse primer, 200 nM probe, 0.01 U/uL Amperase UNG (all from Applied Biosystems, Foster City, Calif.), 0.2 U/uL DyNAzyme (Finnzyme, Espoo, Finland), 0.5% Triton-x-100, 0.8 ug/ul Gelatin (Calbiochem, San Diego, Calif.), 5.0% Glycerol, deionized H2O and human male genomic DNA (Promega).
    • 4. Quantitative PCR Technology, Chapter on “Gene Quantification”, L J McBride, K Livak, M Lucero, et al, Editor, Francois Ferre, Birkauser, Boston, Mass. p 97-110, 1998.
    • See E. T. Lagally, I. Medintz, R. A. Mathies, Anal Chem 73(3), 565-570 (2001), as well as B. Vogelstein, K. W. Kinzler, PNAS 96, 9236-9241 (1999)
  • It is understood that the examples and embodiments described herein are for illustrative purposes only and that various modifications or changes in light thereof will be suggested to persons skilled in the art and are to be included within the spirit and purview of this application and scope of the appended claims. All publications, patents, and patent applications cited herein are incorporated by reference in their entirety for all purposes to the same extent as if each individual publication, patent or patent application were specifically and individually indicated to be so incorporated by reference.

Claims (19)

1. A microfluidic device for reacting M number of different samples with N number of different reagents comprising:
a plurality of reaction cells, each reaction cell comprising a sample chamber and a reagent chamber, said sample chamber and said reagent chamber being in fluid communication through an interface channel having an interface valve associated therewith for controlling fluid communication between said sample chamber and said reagent chamber;
a plurality of sample inlets each in fluid communication with said sample chambers;
a plurality of reagent inlets each in fluid communication with said reagent chambers;
wherein at least one of said chambers contains an oligonucleotide or protein polymer and an intercollating dye.
2. The microfluidic device of claim 1 wherein said reaction cells are formed within an elastomeric block formed from a plurality of layers bonded together and said interface value is deflectable membrane.
3. The microfluidic device of claim 1 wherein said sample inlets are in communication with said sample chamber through a sample channel and said reagent inlet is in fluid communication with said reagent chamber through a reagent channel, a portion of said sample channel and a portion of said reagent channel being oriented about parallel to each other and each having a containment valve associated therewith for controlling fluid communication therethrough.
4. The microfluidic device of claim 3 wherein said valve associated with said sample channel and said valve associated with said reagent channel are in operable communication with each other through a common containment control channel.
5. The microfluidic device of claim 4 wherein said containment common control channel located along a line about normal to one of said sample channel or said reagent channel.
6. The device of claim 1 wherein the intercollating dye is SYBR Green (TM).
7. A method for determining the denaturation temperature of protein or oligonucleotide comprising the steps of providing the device of claim 1, altering the temperature of the device, and detecting a change in the dye.
8. A microfluidic device for reacting M number of different samples with N number of different reagents comprising:
a plurality of reaction cells, each reaction cell comprising a sample chamber and a reagent chamber, said sample chamber and said reagent chamber being in fluid communication through an interface channel having an interface valve associated therewith for controlling fluid communication between said sample chamber and said reagent chamber;
a plurality of sample inlets each in fluid communication with said sample chambers;
a plurality of reagent inlets each in fluid communication with said reagent chambers;
wherein one of said sample inlets or reagent inlets is in fluid communication with one of said sample chambers or one of said reagent chambers, respectively, through a via.
9. The microfluidic device of claim 8 wherein said reaction cells are formed within an elastomeric block formed from a plurality of layers bonded together and said interface valve is deflectable membrane.
10. The microfluidic device of claim 8 wherein said sample inlets are in communication with said sample chamber through a sample channel and said reagent inlet is in fluid communication with said reagent chamber through a reagent channel, a portion of said sample channel and a portion of said reagent channel being oriented about parallel to each other and each having a containment valve associated therewith for controlling fluid communication therethrough.
11. The microfluidic device of claim 10 wherein said valve associated with said sample channel and said valve associated with said reagent channel are in operable communication with each other through a common containment control channel.
12. The microfluidic device of claim 11 wherein said containment common control channel located along a line about normal to one of said sample channel or said reagent channel.
13. A method for making a feature in an elastomeric block comprising the steps of:
providing a first elastomeric layer;
applying a photoresist layer upon a surface of said first elastomeric layer;
applying a light pattern to said photoresist layer to form a pattern of reacted photoresist material;
removing unreacted photoresist material leaving said pattern of reacted photoresist upon said surface of said first elastomeric layer;
applying an etching reagent to said first elastomeric surface to etch said surface of said first elastomeric layer not covered by said pattern of reacted photoresist material thereby removing regions of said first elastomeric layer not covered by said pattern of reacted photoresist and leaving a pattern of said elastomeric layer corresponding to said pattern of reacted photoresist material.
14. The method of claim 13 further comprising the step of removing said pattern of reacted photoresist material.
15. The method of claim 14 wherein said removing is caused by applying an adhesive tape to said surface of said elastomeric layer and said pattern of reacted photoresist material, then separating said adhesive tape from said elastomeric layer while some or all of said pattern of reacted photoresist material is removed from said surface of said elastomeric layer.
16. The method of claim 13 wherein said photoresist is SU8.
17. The method of claim 13 wherein said etching reagent comprises tetrabutylammoniumfluoride-trihydrate.
18. The method of claim 13 wherein said feature is a via.
19. The method of claim 18 wherein said elastomeric block comprises a plurality of elastomeric layers bonded together, wherein two or more elastomeric layers have recesses formed therein and one recess of one elastomeric layer is in communication with a recess of another elastomeric layer through said via.
US10/876,046 2003-04-03 2004-06-23 Thermal reaction device and method for using the same Abandoned US20050145496A1 (en)

Priority Applications (20)

Application Number Priority Date Filing Date Title
US10/876,046 US20050145496A1 (en) 2003-04-03 2004-06-23 Thermal reaction device and method for using the same
US11/084,357 US7604965B2 (en) 2003-04-03 2005-03-18 Thermal reaction device and method for using the same
CN201210270352XA CN102778432A (en) 2004-05-02 2005-05-02 Thermal reaction device and using method thereof
EP05766055.7A EP1796824A4 (en) 2004-05-02 2005-05-02 Thermal reaction device and method for using the same
PCT/US2005/015352 WO2005107938A2 (en) 2004-05-02 2005-05-02 Thermal reaction device and method for using the same
JP2007511507A JP5502275B2 (en) 2004-05-02 2005-05-02 Thermal reaction device and method of using the thermal reaction device
CN2005800225837A CN1997454B (en) 2004-05-02 2005-05-02 Thermal reaction device and method for using the same
SG2013006333A SG2013006333A (en) 2004-05-02 2005-05-02 Thermal reaction device and method for using the same
SG200902863-0A SG166024A1 (en) 2004-05-02 2005-05-02 Thermal reaction device and method for using the same
CN2010101546166A CN101811074B (en) 2004-05-02 2005-05-02 Thermal reaction equipment and using method thereof
US11/929,370 US7867454B2 (en) 2003-04-03 2007-10-30 Thermal reaction device and method for using the same
US11/929,436 US7749737B2 (en) 2003-04-03 2007-10-30 Thermal reaction device and method for using the same
US12/579,347 US8247178B2 (en) 2003-04-03 2009-10-14 Thermal reaction device and method for using the same
JP2012020787A JP2012085659A (en) 2004-05-02 2012-02-02 Thermal reaction device and method for using the same
US13/548,068 US9150913B2 (en) 2003-04-03 2012-07-12 Thermal reaction device and method for using the same
US14/477,602 US20150174575A1 (en) 2003-04-03 2015-03-02 Thermal Reaction Device and Method for Using the Same
US14/873,958 US20160129441A1 (en) 2003-04-03 2015-10-02 Thermal reaction device and method for using the same
US15/805,720 US10131934B2 (en) 2003-04-03 2017-11-07 Thermal reaction device and method for using the same
US15/829,665 US20180243741A1 (en) 2004-05-02 2017-12-01 Thermal reaction device and method for using the same
US16/150,096 US20190169676A1 (en) 2003-04-03 2018-10-02 Thermal reaction device and method for using the same

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US46063403P 2003-04-03 2003-04-03
US10/818,642 US7666361B2 (en) 2003-04-03 2004-04-05 Microfluidic devices and methods of using same
US10/837,885 US7476363B2 (en) 2003-04-03 2004-05-02 Microfluidic devices and methods of using same
US10/876,046 US20050145496A1 (en) 2003-04-03 2004-06-23 Thermal reaction device and method for using the same

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US10/837,885 Continuation-In-Part US7476363B2 (en) 2003-04-03 2004-05-02 Microfluidic devices and methods of using same

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US11/084,357 Continuation-In-Part US7604965B2 (en) 2003-04-03 2005-03-18 Thermal reaction device and method for using the same
US11/929,370 Continuation US7867454B2 (en) 2003-04-03 2007-10-30 Thermal reaction device and method for using the same

Publications (1)

Publication Number Publication Date
US20050145496A1 true US20050145496A1 (en) 2005-07-07

Family

ID=46302222

Family Applications (2)

Application Number Title Priority Date Filing Date
US10/876,046 Abandoned US20050145496A1 (en) 2003-04-03 2004-06-23 Thermal reaction device and method for using the same
US11/929,370 Active 2024-09-24 US7867454B2 (en) 2003-04-03 2007-10-30 Thermal reaction device and method for using the same

Family Applications After (1)

Application Number Title Priority Date Filing Date
US11/929,370 Active 2024-09-24 US7867454B2 (en) 2003-04-03 2007-10-30 Thermal reaction device and method for using the same

Country Status (1)

Country Link
US (2) US20050145496A1 (en)

Cited By (47)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060006067A1 (en) * 2004-06-07 2006-01-12 Fluidigm Corporation Optical lens system and method for microfluidic devices
US20070202525A1 (en) * 2006-02-02 2007-08-30 The Board Of Trustees Of The Leland Stanford Junior University Non-invasive fetal genetic screening by digital analysis
US20080137584A1 (en) * 2006-12-08 2008-06-12 Samsung Electronics Co., Ltd. Apparatus and method for selecting frame structure in multihop relay broadband wireless access communication system
US20080138809A1 (en) * 2006-06-14 2008-06-12 Ravi Kapur Methods for the Diagnosis of Fetal Abnormalities
US20080220422A1 (en) * 2006-06-14 2008-09-11 Daniel Shoemaker Rare cell analysis using sample splitting and dna tags
US20090029377A1 (en) * 2007-07-23 2009-01-29 The Chinese University Of Hong Kong Diagnosing fetal chromosomal aneuploidy using massively parallel genomic sequencing
WO2009017627A1 (en) * 2007-07-30 2009-02-05 Gn Biosystems Incorporated Apparatus and method for conducting high-throughput micro-volume experiments
EP2089543A2 (en) * 2006-11-15 2009-08-19 Idaho Technology, Inc. High density self-contained biological analysis
US20090298191A1 (en) * 2006-10-18 2009-12-03 President And Fellows Of Harvard College Lateral Flow and Flow-through Bioassay Devices Based On Patterned Porous Media, Methods of Making Same, and Methods of Using Same
WO2009121041A3 (en) * 2008-03-27 2009-12-17 President And Fellows Of Harvard College Paper-based microfluidic systems
US20100064780A1 (en) * 2005-07-27 2010-03-18 Howard A Stone Pressure Determination In Microfludic Systems
US20100112590A1 (en) * 2007-07-23 2010-05-06 The Chinese University Of Hong Kong Diagnosing Fetal Chromosomal Aneuploidy Using Genomic Sequencing With Enrichment
US20100138165A1 (en) * 2008-09-20 2010-06-03 The Board Of Trustees Of The Leland Stanford Junior University Noninvasive Diagnosis of Fetal Aneuploidy by Sequencing
US7749737B2 (en) 2003-04-03 2010-07-06 Fluidigm Corporation Thermal reaction device and method for using the same
US20110105360A1 (en) * 2008-03-27 2011-05-05 President And Fellows Of Harvard College Paper-based cellular arrays
US20110123398A1 (en) * 2008-03-27 2011-05-26 President And Fellows Of Harvard College Three-dimensional microfluidic devices
US20110129841A1 (en) * 2005-06-02 2011-06-02 Fluidigm Corporation Analysis using microfluidic partitioning devices
US20110189786A1 (en) * 2008-03-27 2011-08-04 President And Fellows Of Harvard College Cotton thread as a low-cost multi-assay diagnostic platform
US20110206576A1 (en) * 2008-08-07 2011-08-25 Fluidigm Corporation Microfluidic mixing and reaction systems for high efficiency screening
US20130224879A1 (en) * 2011-08-26 2013-08-29 The Trustees Of Columbia University In The City Of New York Systems and methods for a dna-based thermometer
US8658418B2 (en) 2002-04-01 2014-02-25 Fluidigm Corporation Microfluidic particle-analysis systems
WO2014068408A2 (en) 2012-10-23 2014-05-08 Caris Life Sciences Switzerland Holdings, S.A.R.L. Aptamers and uses thereof
WO2014100434A1 (en) 2012-12-19 2014-06-26 Caris Science, Inc. Compositions and methods for aptamer screening
US8821810B2 (en) 2010-02-03 2014-09-02 President And Fellows Of Harvard College Devices and methods for multiplexed assays
US8828663B2 (en) 2005-03-18 2014-09-09 Fluidigm Corporation Thermal reaction device and method for using the same
WO2015031694A2 (en) 2013-08-28 2015-03-05 Caris Science, Inc. Oligonucleotide probes and uses thereof
US9128101B2 (en) 2010-03-01 2015-09-08 Caris Life Sciences Switzerland Holdings Gmbh Biomarkers for theranostics
US9192933B2 (en) 2009-03-06 2015-11-24 President And Fellows Of Harvard College Microfluidic, electrochemical devices
US9205468B2 (en) 2009-11-30 2015-12-08 Fluidigm Corporation Microfluidic device regeneration
US9316331B2 (en) * 2005-01-25 2016-04-19 Fluidigm Corporation Multilevel microfluidic systems and methods
WO2016145128A1 (en) 2015-03-09 2016-09-15 Caris Science, Inc. Oligonucleotide probes and uses thereof
US9469876B2 (en) 2010-04-06 2016-10-18 Caris Life Sciences Switzerland Holdings Gmbh Circulating biomarkers for metastatic prostate cancer
WO2017004243A1 (en) 2015-06-29 2017-01-05 Caris Science, Inc. Therapeutic oligonucleotides
WO2017019918A1 (en) 2015-07-28 2017-02-02 Caris Science, Inc. Targeted oligonucleotides
US9625404B2 (en) 2011-01-11 2017-04-18 The Trustees Of Columbia University In The City Of New York Systems and methods for single-molecule detection using nanotubes
WO2017205686A1 (en) 2016-05-25 2017-11-30 Caris Science, Inc. Oligonucleotide probes and uses thereof
US9841416B2 (en) 2012-04-20 2017-12-12 The Trustees Of Columbia University In The City Of New York Systems and methods for single-molecule nucleic-acid assay platforms
US9840732B2 (en) 2012-05-21 2017-12-12 Fluidigm Corporation Single-particle analysis of particle populations
US10053728B2 (en) 2006-11-15 2018-08-21 Biofire Diagnostics, Llc High density self-contained biological analysis
US10364467B2 (en) 2015-01-13 2019-07-30 The Chinese University Of Hong Kong Using size and number aberrations in plasma DNA for detecting cancer
CN110850083A (en) * 2019-10-31 2020-02-28 浙江卫未生物医药科技有限公司 Batch exosome concentration detection kit and operation method
US10591391B2 (en) 2006-06-14 2020-03-17 Verinata Health, Inc. Diagnosis of fetal abnormalities using polymorphisms including short tandem repeats
US10627364B2 (en) 2012-04-10 2020-04-21 The Trustees Of Columbia University In The City Of New York Systems and methods for biological ion channel interfaces
US10704090B2 (en) 2006-06-14 2020-07-07 Verinata Health, Inc. Fetal aneuploidy detection by sequencing
US10731166B2 (en) 2016-03-18 2020-08-04 Caris Science, Inc. Oligonucleotide probes and uses thereof
US10942184B2 (en) 2012-10-23 2021-03-09 Caris Science, Inc. Aptamers and uses thereof
US11624727B2 (en) 2011-02-23 2023-04-11 The Trustees Of Columbia University In The City Of New York Systems and methods for single-molecule detection using nanopores

Families Citing this family (45)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6221654B1 (en) 1996-09-25 2001-04-24 California Institute Of Technology Method and apparatus for analysis and sorting of polynucleotides based on size
US7459022B2 (en) 2001-04-06 2008-12-02 California Institute Of Technology Microfluidic protein crystallography
US7306672B2 (en) 2001-04-06 2007-12-11 California Institute Of Technology Microfluidic free interface diffusion techniques
WO2001001025A2 (en) 1999-06-28 2001-01-04 California Institute Of Technology Microfabricated elastomeric valve and pump systems
US8709153B2 (en) 1999-06-28 2014-04-29 California Institute Of Technology Microfludic protein crystallography techniques
US7144616B1 (en) 1999-06-28 2006-12-05 California Institute Of Technology Microfabricated elastomeric valve and pump systems
US8052792B2 (en) 2001-04-06 2011-11-08 California Institute Of Technology Microfluidic protein crystallography techniques
US7867763B2 (en) 2004-01-25 2011-01-11 Fluidigm Corporation Integrated chip carriers with thermocycler interfaces and methods of using the same
US20050118073A1 (en) 2003-11-26 2005-06-02 Fluidigm Corporation Devices and methods for holding microfluidic devices
US7351376B1 (en) 2000-06-05 2008-04-01 California Institute Of Technology Integrated active flux microfluidic devices and methods
EP1334347A1 (en) 2000-09-15 2003-08-13 California Institute Of Technology Microfabricated crossflow devices and methods
EP1343973B2 (en) 2000-11-16 2020-09-16 California Institute Of Technology Apparatus and methods for conducting assays and high throughput screening
JP4355210B2 (en) 2001-11-30 2009-10-28 フルイディグム コーポレイション Microfluidic device and method of using microfluidic device
US7691333B2 (en) 2001-11-30 2010-04-06 Fluidigm Corporation Microfluidic device and methods of using same
EP2213615A3 (en) 2002-09-25 2012-02-29 California Institute of Technology Microfluidic Large Scale Integration
US8220494B2 (en) 2002-09-25 2012-07-17 California Institute Of Technology Microfluidic large scale integration
JP5695287B2 (en) 2002-10-02 2015-04-01 カリフォルニア インスティテュート オブ テクノロジー Nucleic acid analysis of microfluids
US7695683B2 (en) 2003-05-20 2010-04-13 Fluidigm Corporation Method and system for microfluidic device and imaging thereof
US7407799B2 (en) 2004-01-16 2008-08-05 California Institute Of Technology Microfluidic chemostat
JP2008522795A (en) * 2004-12-03 2008-07-03 カリフォルニア インスティチュート オブ テクノロジー Microfluidic device with chemical reaction circuit
US20080264863A1 (en) 2004-12-03 2008-10-30 California Institute Of Technology Microfluidic Sieve Valves
US7883669B2 (en) 2005-04-20 2011-02-08 Fluidigm Corporation Analysis engine and database for manipulating parameters for fluidic systems on a chip
US20070054293A1 (en) * 2005-08-30 2007-03-08 California Institute Of Technology Microfluidic chaotic mixing systems and methods
EP1938101A2 (en) * 2005-09-13 2008-07-02 Fluidigm Corporation Microfluidic assay devices and methods
US7815868B1 (en) 2006-02-28 2010-10-19 Fluidigm Corporation Microfluidic reaction apparatus for high throughput screening
US8828661B2 (en) * 2006-04-24 2014-09-09 Fluidigm Corporation Methods for detection and quantification of nucleic acid or protein targets in a sample
US8055034B2 (en) 2006-09-13 2011-11-08 Fluidigm Corporation Methods and systems for image processing of microfluidic devices
WO2008067552A2 (en) * 2006-11-30 2008-06-05 Fluidigm Corporation Method and apparatus for biological sample analysis
EP2125219B1 (en) 2007-01-19 2016-08-10 Fluidigm Corporation High precision microfluidic devices and methods
BRPI0816393A2 (en) * 2007-09-07 2015-03-03 Fluidigm Corp METHOD FOR DETERMINING THE NUMBER OF COPIES REGARDING A TARGET POLINUCLEOTIDE SEQUENCE IN A GENOME OF AN INDIVIDUAL
WO2009100449A1 (en) 2008-02-08 2009-08-13 Fluidigm Corporation Dynamic array assay methods
US9579830B2 (en) 2008-07-25 2017-02-28 Fluidigm Corporation Method and system for manufacturing integrated fluidic chips
US8058630B2 (en) * 2009-01-16 2011-11-15 Fluidigm Corporation Microfluidic devices and methods
US8790916B2 (en) 2009-05-14 2014-07-29 Genestream, Inc. Microfluidic method and system for isolating particles from biological fluid
US8551787B2 (en) * 2009-07-23 2013-10-08 Fluidigm Corporation Microfluidic devices and methods for binary mixing
SG169918A1 (en) 2009-10-02 2011-04-29 Fluidigm Corp Microfluidic devices with removable cover and methods of fabrication and application
WO2012054933A2 (en) 2010-10-22 2012-04-26 Fluidigm Corporation Universal probe assay methods
US9168531B2 (en) 2011-03-24 2015-10-27 Fluidigm Corporation Method for thermal cycling of microfluidic samples
CN103635594B (en) 2011-05-09 2018-02-13 富鲁达公司 Detection of nucleic acids based on probe
US9644231B2 (en) 2011-05-09 2017-05-09 Fluidigm Corporation Nucleic acid detection using probes
US9039993B2 (en) 2013-03-14 2015-05-26 Formulatrix, Inc. Microfluidic device
US9163277B2 (en) 2013-03-14 2015-10-20 Formulatrix, Inc. Microfluidic device
DE102014018535A1 (en) * 2014-12-12 2016-06-16 Nanotemper Technologies Gmbh System and method for a seal-free tempering of capillaries
US9890835B2 (en) 2015-04-24 2018-02-13 Allison Transmission, Inc. Multi-speed transmission
US11154864B2 (en) 2018-01-17 2021-10-26 Qiagen Sciences, Llc Microfluidic device with vented microchambers

Citations (96)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2656508A (en) * 1949-08-27 1953-10-20 Wallace H Coulter Means for counting particles suspended in a fluid
US3560754A (en) * 1965-11-17 1971-02-02 Ibm Photoelectric particle separator using time delay
US3570515A (en) * 1969-06-19 1971-03-16 Foxboro Co Aminar stream cross-flow fluid diffusion logic gate
US3747628A (en) * 1971-02-17 1973-07-24 Philips Corp Fluidic function module for use in a system for constructing fluidic circuits
US4046159A (en) * 1974-10-08 1977-09-06 Pegourie Jean Pierre Pneumatic logic circuits and their integrated circuits
US4153855A (en) * 1977-12-16 1979-05-08 The United States Of America As Represented By The Secretary Of The Army Method of making a plate having a pattern of microchannels
US4245673A (en) * 1978-03-01 1981-01-20 La Telemechanique Electrique Pneumatic logic circuit
US4434704A (en) * 1980-04-14 1984-03-06 Halliburton Company Hydraulic digital stepper actuator
US4581624A (en) * 1984-03-01 1986-04-08 Allied Corporation Microminiature semiconductor valve
US4585209A (en) * 1983-10-27 1986-04-29 Harry E. Aine Miniature valve and method of making same
US4675300A (en) * 1985-09-18 1987-06-23 The Board Of Trustees Of The Leland Stanford Junior University Laser-excitation fluorescence detection electrokinetic separation
US4898582A (en) * 1988-08-09 1990-02-06 Pharmetrix Corporation Portable infusion device assembly
US4908112A (en) * 1988-06-16 1990-03-13 E. I. Du Pont De Nemours & Co. Silicon semiconductor wafer for analyzing micronic biological samples
US4992312A (en) * 1989-03-13 1991-02-12 Dow Corning Wright Corporation Methods of forming permeation-resistant, silicone elastomer-containing composite laminates and devices produced thereby
US5032381A (en) * 1988-12-20 1991-07-16 Tropix, Inc. Chemiluminescence-based static and flow cytometry
US5085562A (en) * 1989-04-11 1992-02-04 Westonbridge International Limited Micropump having a constant output
US5088515A (en) * 1989-05-01 1992-02-18 Kamen Dean L Valve system with removable fluid interface
US5096388A (en) * 1990-03-22 1992-03-17 The Charles Stark Draper Laboratory, Inc. Microfabricated pump
US5126115A (en) * 1987-10-27 1992-06-30 Fujitsu Limited Process and apparatus for preparation of single crystal of biopolymer
US5126022A (en) * 1990-02-28 1992-06-30 Soane Tecnologies, Inc. Method and device for moving molecules by the application of a plurality of electrical fields
US5132012A (en) * 1988-06-24 1992-07-21 Hitachi, Ltd. Liquid chromatograph
US5140161A (en) * 1985-08-05 1992-08-18 Biotrack Capillary flow device
US5224843A (en) * 1989-06-14 1993-07-06 Westonbridge International Ltd. Two valve micropump with improved outlet
US5290240A (en) * 1993-02-03 1994-03-01 Pharmetrix Corporation Electrochemical controlled dispensing assembly and method for selective and controlled delivery of a dispensing fluid
US5336062A (en) * 1990-02-27 1994-08-09 Fraunhofer-Gesellschaft Zur Forderung Der Angewandten Forschung E.V. Microminiaturized pump
US5346372A (en) * 1991-07-18 1994-09-13 Aisin Seiki Kabushiki Kaisha Fluid flow regulating device
US5400741A (en) * 1993-05-21 1995-03-28 Medical Foundation Of Buffalo, Inc. Device for growing crystals
US5423287A (en) * 1992-11-25 1995-06-13 Nissan Motor Company, Ltd. Crystal growing cell
US5434049A (en) * 1992-02-28 1995-07-18 Hitachi, Ltd. Separation of polynucleotides using supports having a plurality of electrode-containing cells
US5452878A (en) * 1991-06-18 1995-09-26 Danfoss A/S Miniature actuating device
US5486335A (en) * 1992-05-01 1996-01-23 Trustees Of The University Of Pennsylvania Analysis based on flow restriction
US5498392A (en) * 1992-05-01 1996-03-12 Trustees Of The University Of Pennsylvania Mesoscale polynucleotide amplification device and method
US5500071A (en) * 1994-10-19 1996-03-19 Hewlett-Packard Company Miniaturized planar columns in novel support media for liquid phase analysis
US5512131A (en) * 1993-10-04 1996-04-30 President And Fellows Of Harvard College Formation of microstamped patterns on surfaces and derivative articles
US5529465A (en) * 1991-09-11 1996-06-25 Fraunhofer-Gesellschaft Zur Forderung Der Angewandten Forschung E.V. Micro-miniaturized, electrostatically driven diaphragm micropump
US5558998A (en) * 1992-02-25 1996-09-24 The Regents Of The Univ. Of California DNA fragment sizing and sorting by laser-induced fluorescence
US5593130A (en) * 1993-06-09 1997-01-14 Pharmacia Biosensor Ab Valve, especially for fluid handling bodies with microflowchannels
US5632876A (en) * 1995-06-06 1997-05-27 David Sarnoff Research Center, Inc. Apparatus and methods for controlling fluid flow in microchannels
US5642015A (en) * 1993-07-14 1997-06-24 The University Of British Columbia Elastomeric micro electro mechanical systems
US5641400A (en) * 1994-10-19 1997-06-24 Hewlett-Packard Company Use of temperature control devices in miniaturized planar column devices and miniaturized total analysis systems
US5659171A (en) * 1993-09-22 1997-08-19 Northrop Grumman Corporation Micro-miniature diaphragm pump for the low pressure pumping of gases
US5661222A (en) * 1995-04-13 1997-08-26 Dentsply Research & Development Corp. Polyvinylsiloxane impression material
US5660370A (en) * 1996-03-07 1997-08-26 Integrated Fludics, Inc. Valve with flexible sheet member and two port non-flexing backer member
US5665070A (en) * 1995-01-19 1997-09-09 I-Flow Corporation Infusion pump with magnetic bag compression
US5705018A (en) * 1995-12-13 1998-01-06 Hartley; Frank T. Micromachined peristaltic pump
US5716852A (en) * 1996-03-29 1998-02-10 University Of Washington Microfabricated diffusion-based chemical sensor
US5726751A (en) * 1995-09-27 1998-03-10 University Of Washington Silicon microchannel optical flow cytometer
US5750015A (en) * 1990-02-28 1998-05-12 Soane Biosciences Method and device for moving molecules by the application of a plurality of electrical fields
US5757482A (en) * 1995-04-20 1998-05-26 Perseptive Biosystems, Inc. Module for optical detection in microscale fluidic analyses
US5759014A (en) * 1994-01-14 1998-06-02 Westonbridge International Limited Micropump
US5775371A (en) * 1995-03-08 1998-07-07 Abbott Laboratories Valve control
US5788468A (en) * 1994-11-03 1998-08-04 Memstek Products, Llc Microfabricated fluidic devices
US5800690A (en) * 1996-07-03 1998-09-01 Caliper Technologies Corporation Variable control of electroosmotic and/or electrophoretic forces within a fluid-containing structure via electrical forces
US5856092A (en) * 1989-02-13 1999-01-05 Geneco Pty Ltd Detection of a nucleic acid sequence or a change therein
US5856174A (en) * 1995-06-29 1999-01-05 Affymetrix, Inc. Integrated nucleic acid diagnostic device
US5863801A (en) * 1996-06-14 1999-01-26 Sarnoff Corporation Automated nucleic acid isolation
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
US5875817A (en) * 1995-08-17 1999-03-02 Ortech Corporation Digital gas metering system using tri-stable and bi-stable solenoids
US5876187A (en) * 1995-03-09 1999-03-02 University Of Washington Micropumps with fixed valves
US5880071A (en) * 1996-06-28 1999-03-09 Caliper Technologies Corporation Electropipettor and compensation means for electrophoretic bias
US5904824A (en) * 1997-03-07 1999-05-18 Beckman Instruments, Inc. Microfluidic electrophoresis device
US5932799A (en) * 1997-07-21 1999-08-03 Ysi Incorporated Microfluidic analyzer module
US5942443A (en) * 1996-06-28 1999-08-24 Caliper Technologies Corporation High throughput screening assay systems in microscale fluidic devices
US6107044A (en) * 1997-10-16 2000-08-22 Caliper Technologies Corp. Apparatus and methods for sequencing nucleic acids in microfluidic systems
US6117634A (en) * 1997-03-05 2000-09-12 The Reagents Of The University Of Michigan Nucleic acid sequencing and mapping
US6123769A (en) * 1995-03-01 2000-09-26 Sumitomo Metal Industries, Ltd. Crystallization control method for organic compound and crystallization control solid-state component employed therefor
US6174365B1 (en) * 1996-07-15 2001-01-16 Sumitomo Metal Industries, Ltd. Apparatus for crystal growth and crystal growth method employing the same
US6221654B1 (en) * 1996-09-25 2001-04-24 California Institute Of Technology Method and apparatus for analysis and sorting of polynucleotides based on size
US20020014673A1 (en) * 1992-04-08 2002-02-07 Elm Technology Corporation Method of making membrane integrated circuits
US6345502B1 (en) * 1997-11-12 2002-02-12 California Institute Of Technology Micromachined parylene membrane valve and pump
US6358387B1 (en) * 2000-03-27 2002-03-19 Caliper Technologies Corporation Ultra high throughput microfluidic analytical systems and methods
US20020037499A1 (en) * 2000-06-05 2002-03-28 California Institute Of Technology Integrated active flux microfluidic devices and methods
US6376971B1 (en) * 1997-02-07 2002-04-23 Sri International Electroactive polymer electrodes
US6375871B1 (en) * 1998-06-18 2002-04-23 3M Innovative Properties Company Methods of manufacturing microfluidic articles
US20020054885A1 (en) * 1999-12-22 2002-05-09 Sylvie Laquerre Herpes simplex virus-1 Glycoprotein C mutants for treating unwanted hyperproliferative cell growth
US6409832B2 (en) * 2000-03-31 2002-06-25 Micronics, Inc. Protein crystallization in microfluidic structures
US6408878B2 (en) * 1999-06-28 2002-06-25 California Institute Of Technology Microfabricated elastomeric valve and pump systems
US20020108096A1 (en) * 2000-06-27 2002-08-08 Michael Lee Microfluidic design automation method and system
US20030008308A1 (en) * 2001-04-06 2003-01-09 California Institute Of Technology Nucleic acid amplification utilizing microfluidic devices
US6520936B1 (en) * 1999-06-08 2003-02-18 Medtronic Minimed, Inc. Method and apparatus for infusing liquids using a chemical reaction in an implanted infusion device
US6540895B1 (en) * 1997-09-23 2003-04-01 California Institute Of Technology Microfabricated cell sorter for chemical and biological materials
US20030080442A1 (en) * 2001-08-30 2003-05-01 Fluidigm Corp. Electrostatic/electrostrictive actuation of elastomer structures using compliant electrodes
US6585939B1 (en) * 1999-02-26 2003-07-01 Orchid Biosciences, Inc. Microstructures for use in biological assays and reactions
US20030138829A1 (en) * 2001-11-30 2003-07-24 Fluidigm Corp. Microfluidic device and methods of using same
US20030143120A1 (en) * 2002-01-25 2003-07-31 Waldemar Ruediger Parallel chemistry reactor with interchangeable vessel carrying inserts
US20030175947A1 (en) * 2001-11-05 2003-09-18 Liu Robin Hui Enhanced mixing in microfluidic devices
US6690437B2 (en) * 2000-04-18 2004-02-10 Semiconductor Energy Laboratory Co., Ltd. Electro-optical device
US20040072278A1 (en) * 2002-04-01 2004-04-15 Fluidigm Corporation Microfluidic particle-analysis systems
US20040115838A1 (en) * 2000-11-16 2004-06-17 Quake Stephen R. Apparatus and methods for conducting assays and high throughput screening
US20040141887A1 (en) * 2002-11-08 2004-07-22 Irm, Llc Apparatus and methods to process substrate surface features
US20050053952A1 (en) * 2002-10-02 2005-03-10 California Institute Of Technology Microfluidic nucleic acid analysis
US20050123947A1 (en) * 1997-09-23 2005-06-09 California Institute Of Technology Methods and systems for molecular fingerprinting
US20050129581A1 (en) * 2003-04-03 2005-06-16 Fluidigm Corporation Microfluidic devices and methods of using same
US6929030B2 (en) * 1999-06-28 2005-08-16 California Institute Of Technology Microfabricated elastomeric valve and pump systems
US20050197652A1 (en) * 2002-05-13 2005-09-08 Fluidigm Corporation Drug delivery system
US20050201901A1 (en) * 2004-01-25 2005-09-15 Fluidigm Corp. Crystal forming devices and systems and methods for using the same

Family Cites Families (152)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3839176A (en) 1971-03-08 1974-10-01 North American Rockwell Method and apparatus for removing contaminants from liquids
US3984307A (en) 1973-03-05 1976-10-05 Bio/Physics Systems, Inc. Combined particle sorter and segregation indicator
JPS5941169B2 (en) 1975-12-25 1984-10-05 シチズン時計株式会社 Elastomer
US4373527B1 (en) 1979-04-27 1995-06-27 Univ Johns Hopkins Implantable programmable medication infusion system
GB2097692B (en) 1981-01-10 1985-05-22 Shaw Stewart P D Combining chemical reagents
US4399219A (en) 1981-01-29 1983-08-16 Massachusetts Institute Of Technology Process for isolating microbiologically active material
US4575681A (en) 1982-11-12 1986-03-11 Teleco Oilfield Services Inc. Insulating and electrode structure for a drill string
US4662710A (en) 1982-12-03 1987-05-05 Amp Incorporated Method and apparatus for breaking an optical fiber
GB8408529D0 (en) 1984-04-03 1984-05-16 Health Lab Service Board Concentration of biological particles
JPH07104855B2 (en) 1985-03-28 1995-11-13 インターナショナル・ビジネス・マシーンズ・コーポレーション Numerical simulation device
US4963498A (en) 1985-08-05 1990-10-16 Biotrack Capillary flow device
US5164598A (en) 1985-08-05 1992-11-17 Biotrack Capillary flow device
US4786165A (en) 1986-07-10 1988-11-22 Toa Medical Electronics Co., Ltd. Flow cytometry and apparatus therefor
US5525464A (en) 1987-04-01 1996-06-11 Hyseq, Inc. Method of sequencing by hybridization of oligonucleotide probes
ES2063038T3 (en) 1987-07-31 1995-01-01 Gen Probe Inc ANALYSIS OF POLYNUCLEOTIDES USING OLIGONUCLEOTIDES TO ELIMINATE UNDESIRABLE CROSS REACTIONS.
US4936465A (en) 1987-12-07 1990-06-26 Zoeld Tibor Method and apparatus for fast, reliable, and environmentally safe dispensing of fluids, gases and individual particles of a suspension through pressure control at well defined parts of a closed flow-through system
US4965743A (en) 1988-07-14 1990-10-23 The United States Of America As Represented By The Administrator Of The National Aeronautics And Space Administration Discrete event simulation tool for analysis of qualitative models of continuous processing system
US5100627A (en) 1989-11-30 1992-03-31 The Regents Of The University Of California Chamber for the optical manipulation of microscopic particles
KR910012538A (en) 1989-12-27 1991-08-08 야마무라 가쯔미 Micro pump and its manufacturing method
WO1991013338A2 (en) 1990-02-24 1991-09-05 Hatfield Polytechnic Higher Education Corporation Biorheological measurement
US5858188A (en) 1990-02-28 1999-01-12 Aclara Biosciences, Inc. Acrylic microchannels and their use in electrophoretic applications
GB9008044D0 (en) 1990-04-09 1990-06-06 Hatfield Polytechnic Higher Ed Microfabricated device for biological cell sorting
SE470347B (en) 1990-05-10 1994-01-31 Pharmacia Lkb Biotech Microstructure for fluid flow systems and process for manufacturing such a system
DE69106240T2 (en) 1990-07-02 1995-05-11 Seiko Epson Corp Micropump and method of making a micropump.
US6074818A (en) 1990-08-24 2000-06-13 The University Of Tennessee Research Corporation Fingerprinting of nucleic acids, products and methods
WO1992004569A1 (en) 1990-08-31 1992-03-19 Westonbridge International Limited A valve equipped with a position detector and a micropump incorporating said valve
WO1992019960A1 (en) 1991-05-09 1992-11-12 Nanophore, Inc. Methods for the electrophoretic separation of nucleic acids and other linear macromolecules in gel media with restrictive pore diameters
US5164558A (en) 1991-07-05 1992-11-17 Massachusetts Institute Of Technology Micromachined threshold pressure switch and method of manufacture
US5307186A (en) 1991-08-09 1994-04-26 Sharp Kabushiki Kaisha Liquid crystal light valve having capability of providing high-contrast image
US5265327A (en) 1991-09-13 1993-11-30 Faris Sadeg M Microchannel plate technology
US6569382B1 (en) 1991-11-07 2003-05-27 Nanogen, Inc. Methods apparatus for the electronic, homogeneous assembly and fabrication of devices
GB2264296B (en) 1992-02-07 1995-06-28 Zortech Int Microporous thermal insulation material
GB2264496B (en) 1992-02-25 1995-10-25 Us Energy Sizing of fragments from a nucleic acid sequence
JP2740705B2 (en) 1992-04-08 1998-04-15 本田技研工業株式会社 Fluid torque converter characteristic simulation method and apparatus
US5587128A (en) 1992-05-01 1996-12-24 The Trustees Of The University Of Pennsylvania Mesoscale polynucleotide amplification devices
DE4220077A1 (en) 1992-06-19 1993-12-23 Bosch Gmbh Robert Micro-pump for delivery of gases - uses working chamber warmed by heating element and controlled by silicon wafer valves.
US5629147A (en) 1992-07-17 1997-05-13 Aprogenex, Inc. Enriching and identifying fetal cells in maternal blood for in situ hybridization
US5639423A (en) 1992-08-31 1997-06-17 The Regents Of The University Of Calfornia Microfabricated reactor
US5364742A (en) 1992-09-21 1994-11-15 International Business Machines Corporation Micro-miniature structures and method of fabrication thereof
US5477474A (en) 1992-10-29 1995-12-19 Altera Corporation Computer logic simulation with dynamic modeling
EP0600608B1 (en) 1992-10-29 1999-12-22 Altera Corporation Design verification method for programmable logic design
EP0700485B1 (en) 1993-05-27 1997-08-13 Fraunhofer-Gesellschaft Zur Förderung Der Angewandten Forschung E.V. Microvalve
DE69431994T2 (en) 1993-10-04 2003-10-30 Res Int Inc MICRO-MACHINED FLUID TREATMENT DEVICE WITH FILTER AND CONTROL VALVE
EP0670489A3 (en) 1994-03-03 1996-03-06 Ciba Geigy Ag Apparatus and process for separating fluid substances.
US5580523A (en) 1994-04-01 1996-12-03 Bard; Allen J. Integrated chemical synthesizers
US5587081A (en) 1994-04-26 1996-12-24 Jet-Tech, Inc. Thermophilic aerobic waste treatment process
DE69527585T2 (en) 1994-06-08 2003-04-03 Affymetrix Inc Method and device for packaging chips
US5807522A (en) 1994-06-17 1998-09-15 The Board Of Trustees Of The Leland Stanford Junior University Methods for fabricating microarrays of biological samples
DE4433894A1 (en) 1994-09-22 1996-03-28 Fraunhofer Ges Forschung Method and device for controlling a micropump
DE69531430T2 (en) 1994-10-07 2004-07-01 Bayer Corp. relief valve
US5496009A (en) 1994-10-07 1996-03-05 Bayer Corporation Valve
DE4437274C2 (en) 1994-10-18 1998-11-05 Inst Chemo Biosensorik Analyte selective sensor
US5571410A (en) 1994-10-19 1996-11-05 Hewlett Packard Company Fully integrated miniaturized planar liquid sample handling and analysis device
DE4438785C2 (en) 1994-10-24 1996-11-07 Wita Gmbh Wittmann Inst Of Tec Microchemical reaction and analysis unit
US5608519A (en) 1995-03-20 1997-03-04 Gourley; Paul L. Laser apparatus and method for microscopic and spectroscopic analysis and processing of biological cells
DE19520298A1 (en) 1995-06-02 1996-12-05 Bayer Ag Sorting device for biological cells or viruses
US5589136A (en) 1995-06-20 1996-12-31 Regents Of The University Of California Silicon-based sleeve devices for chemical reactions
US6130098A (en) 1995-09-15 2000-10-10 The Regents Of The University Of Michigan Moving microdroplets
JPH09149799A (en) 1995-11-30 1997-06-10 Hitachi Ltd Analysis or detection of nucleic acid and analyser or inspection device of nucleic acid
KR100207410B1 (en) 1995-12-19 1999-07-15 전주범 Fabrication method for lightpath modulation device
US5863502A (en) 1996-01-24 1999-01-26 Sarnoff Corporation Parallel reaction cassette and associated devices
US6126899A (en) 1996-04-03 2000-10-03 The Perkins-Elmer Corporation Device for multiple analyte detection
BR9710054A (en) 1996-06-28 2000-01-11 Caliper Techn Corp Apparatus for separating test compounds for an effect on a biochemical system and for detecting a effect of a test compound on a biochemical system, procedures for determining whether a sample contains a compound capable of affecting a biochemical system, for separating a plurality of test compounds for an effect on a biochemical system and uses of a microfluidic system and a test substrate.
CN1329729C (en) 1996-06-28 2007-08-01 卡钳生命科学股份有限公司 Electropipettor and compensation means for electrophoretic bias
US5699157A (en) 1996-07-16 1997-12-16 Caliper Technologies Corp. Fourier detection of species migrating in a microchannel
US6136212A (en) 1996-08-12 2000-10-24 The Regents Of The University Of Michigan Polymer-based micromachining for microfluidic devices
EP0929658A4 (en) 1996-08-26 2005-11-02 Univ Princeton Reversibly sealable microstructure sorting devices
US5738799A (en) 1996-09-12 1998-04-14 Xerox Corporation Method and materials for fabricating an ink-jet printhead
US5854684A (en) 1996-09-26 1998-12-29 Sarnoff Corporation Massively parallel detection
US5858187A (en) 1996-09-26 1999-01-12 Lockheed Martin Energy Systems, Inc. Apparatus and method for performing electrodynamic focusing on a microchip
US6056428A (en) 1996-11-12 2000-05-02 Invention Machine Corporation Computer based system for imaging and analyzing an engineering object system and indicating values of specific design changes
US5971355A (en) 1996-11-27 1999-10-26 Xerox Corporation Microdevice valve structures to fluid control
EP0958593A4 (en) 1997-01-27 2006-08-30 California Inst Of Techn Mems electrospray nozzle for mass spectroscopy
JP2001521622A (en) 1997-04-04 2001-11-06 カリパー テクノロジーズ コーポレイション Closed-loop biochemical analyzer
US6143496A (en) 1997-04-17 2000-11-07 Cytonix Corporation Method of sampling, amplifying and quantifying segment of nucleic acid, polymerase chain reaction assembly having nanoliter-sized sample chambers, and method of filling assembly
EP0988529B1 (en) 1997-04-25 2013-06-12 Caliper Life Sciences, Inc. Microfluidic devices incorporating improved channel geometries
WO1999000655A2 (en) 1997-06-27 1999-01-07 Immunetics, Inc. Rapid flow-through binding assay apparatus and method
US6529612B1 (en) 1997-07-16 2003-03-04 Diversified Scientific, Inc. Method for acquiring, storing and analyzing crystal images
US6073482A (en) 1997-07-21 2000-06-13 Ysi Incorporated Fluid flow module
US5965410A (en) 1997-09-02 1999-10-12 Caliper Technologies Corp. Electrical current for controlling fluid parameters in microchannels
US7214298B2 (en) 1997-09-23 2007-05-08 California Institute Of Technology Microfabricated cell sorter
US5842787A (en) 1997-10-09 1998-12-01 Caliper Technologies Corporation Microfluidic systems incorporating varied channel dimensions
US5836750A (en) 1997-10-09 1998-11-17 Honeywell Inc. Electrostatically actuated mesopump having a plurality of elementary cells
US6089534A (en) 1998-01-08 2000-07-18 Xerox Corporation Fast variable flow microelectromechanical valves
US6167910B1 (en) 1998-01-20 2001-01-02 Caliper Technologies Corp. Multi-layer microfluidic devices
US6155282A (en) 1998-01-20 2000-12-05 Triconex, Incorporated Two out of three voting solenoid arrangement
US5997961A (en) 1998-03-06 1999-12-07 Battelle Memorial Institute Method of bonding functional surface materials to substrates and applications in microtechnology and antifouling
WO1999052633A1 (en) 1998-04-14 1999-10-21 Ivd Systems Test cartridge with a single inlet port
US6246330B1 (en) 1998-05-29 2001-06-12 Wyn Y. Nielsen Elimination-absorber monitoring system
AU4719399A (en) 1998-06-26 2000-01-17 University Of Washington Crystallization media
US6406921B1 (en) 1998-07-14 2002-06-18 Zyomyx, Incorporated Protein arrays for high-throughput screening
US6576478B1 (en) 1998-07-14 2003-06-10 Zyomyx, Inc. Microdevices for high-throughput screening of biomolecules
US6132685A (en) 1998-08-10 2000-10-17 Caliper Technologies Corporation High throughput microfluidic systems and methods
EP1876442A3 (en) 1998-09-17 2008-03-05 Advion BioSciences, Inc. Integrated monolithic microfabricated liquid chromatography system and method
US6605472B1 (en) 1998-10-09 2003-08-12 The Governors Of The University Of Alberta Microfluidic devices connected to glass capillaries with minimal dead volume
US6149787A (en) 1998-10-14 2000-11-21 Caliper Technologies Corp. External material accession systems and methods
RU2143343C1 (en) 1998-11-03 1999-12-27 Самсунг Электроникс Ко., Лтд. Microinjector and microinjector manufacture method
US6541539B1 (en) 1998-11-04 2003-04-01 President And Fellows Of Harvard College Hierarchically ordered porous oxides
WO2000050642A1 (en) 1999-02-23 2000-08-31 Caliper Technologies Corp. Sequencing by incorporation
US6749814B1 (en) 1999-03-03 2004-06-15 Symyx Technologies, Inc. Chemical processing microsystems comprising parallel flow microreactors and methods for using same
US6500323B1 (en) 1999-03-26 2002-12-31 Caliper Technologies Corp. Methods and software for designing microfluidic devices
AU779792B2 (en) 1999-04-06 2005-02-10 Uab Research Foundation, The Method for screening crystallization conditions in solution crystal growth
US6296673B1 (en) 1999-06-18 2001-10-02 The Regents Of The University Of California Methods and apparatus for performing array microcrystallizations
US6664104B2 (en) 1999-06-25 2003-12-16 Cepheid Device incorporating a microfluidic chip for separating analyte from a sample
US6818395B1 (en) 1999-06-28 2004-11-16 California Institute Of Technology Methods and apparatus for analyzing polynucleotide sequences
US8550119B2 (en) 1999-06-28 2013-10-08 California Institute Of Technology Microfabricated elastomeric valve and pump systems
US6899137B2 (en) 1999-06-28 2005-05-31 California Institute Of Technology Microfabricated elastomeric valve and pump systems
US6533914B1 (en) 1999-07-08 2003-03-18 Shaorong Liu Microfabricated injector and capillary array assembly for high-resolution and high throughput separation
DE19933614C1 (en) 1999-07-17 2000-11-30 Moeller Gmbh Contact system for current-limiting load switch has 2-armed contact arm carrying contact pieces cooperating with contact pieces of fixed contact rails fitted to pivot axis via elongate slot
DE60037433T2 (en) 1999-07-20 2008-12-04 Sri International, Menlo Park Electroactive polymer generators
CA2380075C (en) 1999-07-23 2007-05-29 The Board Of Trustees Of The University Of Illinois Microfabricated devices and method of manufacturing the same using polymer gel
US6977145B2 (en) 1999-07-28 2005-12-20 Serono Genetics Institute S.A. Method for carrying out a biochemical protocol in continuous flow in a microreactor
US6440706B1 (en) 1999-08-02 2002-08-27 Johns Hopkins University Digital amplification
EP1206697A2 (en) 1999-08-02 2002-05-22 Emerald Biostructures Inc. Method and system for creating a crystallization results database
WO2001032930A1 (en) 1999-11-04 2001-05-10 California Institute Of Technology Methods and apparatuses for analyzing polynucleotide sequences
US6866785B2 (en) 2001-08-13 2005-03-15 The Board Of Trustees Of The Leland Stanford Junior University Photopolymerized sol-gel column and associated methods
WO2001067369A2 (en) 2000-03-03 2001-09-13 California Institute Of Technology Combinatorial array for nucleic acid analysis
US6541071B1 (en) 2000-03-23 2003-04-01 Corning Incorporated Method for fabricating supported bilayer-lipid membranes
US6885982B2 (en) 2000-06-27 2005-04-26 Fluidigm Corporation Object oriented microfluidic design method and system
US6627159B1 (en) 2000-06-28 2003-09-30 3M Innovative Properties Company Centrifugal filling of sample processing devices
JP3542550B2 (en) 2000-07-19 2004-07-14 本田技研工業株式会社 Method of forming fuel cell seal
EP1334347A1 (en) 2000-09-15 2003-08-13 California Institute Of Technology Microfabricated crossflow devices and methods
US6787339B1 (en) 2000-10-02 2004-09-07 Motorola, Inc. Microfluidic devices having embedded metal conductors and methods of fabricating said devices
US7097809B2 (en) 2000-10-03 2006-08-29 California Institute Of Technology Combinatorial synthesis system
US6508988B1 (en) 2000-10-03 2003-01-21 California Institute Of Technology Combinatorial synthesis system
EP1322936A2 (en) 2000-10-03 2003-07-02 California Institute Of Technology Microfluidic devices and methods of use
WO2002030486A2 (en) 2000-10-13 2002-04-18 Fluidigm Corporation Microfluidic device based sample injection system for analytical devices
US7232109B2 (en) 2000-11-06 2007-06-19 California Institute Of Technology Electrostatic valves for microfluidic devices
AU2002248149A1 (en) 2000-11-16 2002-08-12 Fluidigm Corporation Microfluidic devices for introducing and dispensing fluids from microfluidic systems
US6736978B1 (en) 2000-12-13 2004-05-18 Iowa State University Research Foundation, Inc. Method and apparatus for magnetoresistive monitoring of analytes in flow streams
US20050196785A1 (en) 2001-03-05 2005-09-08 California Institute Of Technology Combinational array for nucleic acid analysis
EP1377821A2 (en) 2001-04-03 2004-01-07 Micronics, Inc. Pneumatic valve interface for use in microfluidic structures
US20020164816A1 (en) 2001-04-06 2002-11-07 California Institute Of Technology Microfluidic sample separation device
US6752922B2 (en) 2001-04-06 2004-06-22 Fluidigm Corporation Microfluidic chromatography
WO2002082047A2 (en) 2001-04-06 2002-10-17 California Institute Of Technology High throughput screening of crystallization of materials
US6677131B2 (en) 2001-05-14 2004-01-13 Corning Incorporated Well frame including connectors for biological fluids
US20030049659A1 (en) 2001-05-29 2003-03-13 Lapidus Stanley N. Devices and methods for isolating samples into subsamples for analysis
US6847153B1 (en) 2001-06-13 2005-01-25 The United States Of America As Represented By The Secretary Of The Navy Polyurethane electrostriction
US6444318B1 (en) 2001-07-17 2002-09-03 Surmodics, Inc. Self assembling monolayer compositions
US7192629B2 (en) 2001-10-11 2007-03-20 California Institute Of Technology Devices utilizing self-assembled gel and method of manufacture
US6627076B2 (en) 2001-10-19 2003-09-30 Sandia National Laboratories Compact microchannel system
US7691333B2 (en) 2001-11-30 2010-04-06 Fluidigm Corporation Microfluidic device and methods of using same
US6581441B1 (en) 2002-02-01 2003-06-24 Perseptive Biosystems, Inc. Capillary column chromatography process and system
US6662818B2 (en) 2002-02-01 2003-12-16 Perseptive Biosystems, Inc. Programmable tracking pressure regulator for control of higher pressures in microfluidic circuits
US20060086309A1 (en) 2002-06-24 2006-04-27 Fluiding Corporation Recirculating fluidic network and methods for using the same
EP2213615A3 (en) 2002-09-25 2012-02-29 California Institute of Technology Microfluidic Large Scale Integration
US7186383B2 (en) 2002-09-27 2007-03-06 Ast Management Inc. Miniaturized fluid delivery and analysis system
EP1594694A4 (en) 2002-12-30 2010-01-20 Univ California Methods and apparatus for pathogen detection and analysis
US7041481B2 (en) 2003-03-14 2006-05-09 The Regents Of The University Of California Chemical amplification based on fluid partitioning
US7476363B2 (en) 2003-04-03 2009-01-13 Fluidigm Corporation Microfluidic devices and methods of using same
US7413712B2 (en) 2003-08-11 2008-08-19 California Institute Of Technology Microfluidic rotary flow reactor matrix
US7407799B2 (en) 2004-01-16 2008-08-05 California Institute Of Technology Microfluidic chemostat

Patent Citations (99)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2656508A (en) * 1949-08-27 1953-10-20 Wallace H Coulter Means for counting particles suspended in a fluid
US3560754A (en) * 1965-11-17 1971-02-02 Ibm Photoelectric particle separator using time delay
US3570515A (en) * 1969-06-19 1971-03-16 Foxboro Co Aminar stream cross-flow fluid diffusion logic gate
US3747628A (en) * 1971-02-17 1973-07-24 Philips Corp Fluidic function module for use in a system for constructing fluidic circuits
US4046159A (en) * 1974-10-08 1977-09-06 Pegourie Jean Pierre Pneumatic logic circuits and their integrated circuits
US4153855A (en) * 1977-12-16 1979-05-08 The United States Of America As Represented By The Secretary Of The Army Method of making a plate having a pattern of microchannels
US4245673A (en) * 1978-03-01 1981-01-20 La Telemechanique Electrique Pneumatic logic circuit
US4434704A (en) * 1980-04-14 1984-03-06 Halliburton Company Hydraulic digital stepper actuator
US4585209A (en) * 1983-10-27 1986-04-29 Harry E. Aine Miniature valve and method of making same
US4581624A (en) * 1984-03-01 1986-04-08 Allied Corporation Microminiature semiconductor valve
US5140161A (en) * 1985-08-05 1992-08-18 Biotrack Capillary flow device
US4675300A (en) * 1985-09-18 1987-06-23 The Board Of Trustees Of The Leland Stanford Junior University Laser-excitation fluorescence detection electrokinetic separation
US5126115A (en) * 1987-10-27 1992-06-30 Fujitsu Limited Process and apparatus for preparation of single crystal of biopolymer
US4908112A (en) * 1988-06-16 1990-03-13 E. I. Du Pont De Nemours & Co. Silicon semiconductor wafer for analyzing micronic biological samples
US5132012A (en) * 1988-06-24 1992-07-21 Hitachi, Ltd. Liquid chromatograph
US4898582A (en) * 1988-08-09 1990-02-06 Pharmetrix Corporation Portable infusion device assembly
US5032381A (en) * 1988-12-20 1991-07-16 Tropix, Inc. Chemiluminescence-based static and flow cytometry
US5856092A (en) * 1989-02-13 1999-01-05 Geneco Pty Ltd Detection of a nucleic acid sequence or a change therein
US4992312A (en) * 1989-03-13 1991-02-12 Dow Corning Wright Corporation Methods of forming permeation-resistant, silicone elastomer-containing composite laminates and devices produced thereby
US5085562A (en) * 1989-04-11 1992-02-04 Westonbridge International Limited Micropump having a constant output
US5088515A (en) * 1989-05-01 1992-02-18 Kamen Dean L Valve system with removable fluid interface
US5224843A (en) * 1989-06-14 1993-07-06 Westonbridge International Ltd. Two valve micropump with improved outlet
US5336062A (en) * 1990-02-27 1994-08-09 Fraunhofer-Gesellschaft Zur Forderung Der Angewandten Forschung E.V. Microminiaturized pump
US5126022A (en) * 1990-02-28 1992-06-30 Soane Tecnologies, Inc. Method and device for moving molecules by the application of a plurality of electrical fields
US5750015A (en) * 1990-02-28 1998-05-12 Soane Biosciences Method and device for moving molecules by the application of a plurality of electrical fields
US5096388A (en) * 1990-03-22 1992-03-17 The Charles Stark Draper Laboratory, Inc. Microfabricated pump
US5452878A (en) * 1991-06-18 1995-09-26 Danfoss A/S Miniature actuating device
US5346372A (en) * 1991-07-18 1994-09-13 Aisin Seiki Kabushiki Kaisha Fluid flow regulating device
US5529465A (en) * 1991-09-11 1996-06-25 Fraunhofer-Gesellschaft Zur Forderung Der Angewandten Forschung E.V. Micro-miniaturized, electrostatically driven diaphragm micropump
US5558998A (en) * 1992-02-25 1996-09-24 The Regents Of The Univ. Of California DNA fragment sizing and sorting by laser-induced fluorescence
US5434049A (en) * 1992-02-28 1995-07-18 Hitachi, Ltd. Separation of polynucleotides using supports having a plurality of electrode-containing cells
US20020045297A1 (en) * 1992-04-08 2002-04-18 Elm Technology Corporation. Membrane 3D IC fabrication
US20020014673A1 (en) * 1992-04-08 2002-02-07 Elm Technology Corporation Method of making membrane integrated circuits
US5486335A (en) * 1992-05-01 1996-01-23 Trustees Of The University Of Pennsylvania Analysis based on flow restriction
US5498392A (en) * 1992-05-01 1996-03-12 Trustees Of The University Of Pennsylvania Mesoscale polynucleotide amplification device and method
US5423287A (en) * 1992-11-25 1995-06-13 Nissan Motor Company, Ltd. Crystal growing cell
US5290240A (en) * 1993-02-03 1994-03-01 Pharmetrix Corporation Electrochemical controlled dispensing assembly and method for selective and controlled delivery of a dispensing fluid
US5400741A (en) * 1993-05-21 1995-03-28 Medical Foundation Of Buffalo, Inc. Device for growing crystals
US5593130A (en) * 1993-06-09 1997-01-14 Pharmacia Biosensor Ab Valve, especially for fluid handling bodies with microflowchannels
US5642015A (en) * 1993-07-14 1997-06-24 The University Of British Columbia Elastomeric micro electro mechanical systems
US5659171A (en) * 1993-09-22 1997-08-19 Northrop Grumman Corporation Micro-miniature diaphragm pump for the low pressure pumping of gases
US5512131A (en) * 1993-10-04 1996-04-30 President And Fellows Of Harvard College Formation of microstamped patterns on surfaces and derivative articles
US5759014A (en) * 1994-01-14 1998-06-02 Westonbridge International Limited Micropump
US5500071A (en) * 1994-10-19 1996-03-19 Hewlett-Packard Company Miniaturized planar columns in novel support media for liquid phase analysis
US5641400A (en) * 1994-10-19 1997-06-24 Hewlett-Packard Company Use of temperature control devices in miniaturized planar column devices and miniaturized total analysis systems
US5788468A (en) * 1994-11-03 1998-08-04 Memstek Products, Llc Microfabricated fluidic devices
US5665070A (en) * 1995-01-19 1997-09-09 I-Flow Corporation Infusion pump with magnetic bag compression
US6123769A (en) * 1995-03-01 2000-09-26 Sumitomo Metal Industries, Ltd. Crystallization control method for organic compound and crystallization control solid-state component employed therefor
US5775371A (en) * 1995-03-08 1998-07-07 Abbott Laboratories Valve control
US5876187A (en) * 1995-03-09 1999-03-02 University Of Washington Micropumps with fixed valves
US5661222A (en) * 1995-04-13 1997-08-26 Dentsply Research & Development Corp. Polyvinylsiloxane impression material
US5757482A (en) * 1995-04-20 1998-05-26 Perseptive Biosystems, Inc. Module for optical detection in microscale fluidic analyses
US5632876A (en) * 1995-06-06 1997-05-27 David Sarnoff Research Center, Inc. Apparatus and methods for controlling fluid flow in microchannels
US5856174A (en) * 1995-06-29 1999-01-05 Affymetrix, Inc. Integrated nucleic acid diagnostic device
US6043080A (en) * 1995-06-29 2000-03-28 Affymetrix, Inc. Integrated nucleic acid diagnostic device
US5875817A (en) * 1995-08-17 1999-03-02 Ortech Corporation Digital gas metering system using tri-stable and bi-stable solenoids
US5726751A (en) * 1995-09-27 1998-03-10 University Of Washington Silicon microchannel optical flow cytometer
US5705018A (en) * 1995-12-13 1998-01-06 Hartley; Frank T. Micromachined peristaltic pump
US5660370A (en) * 1996-03-07 1997-08-26 Integrated Fludics, Inc. Valve with flexible sheet member and two port non-flexing backer member
US5716852A (en) * 1996-03-29 1998-02-10 University Of Washington Microfabricated diffusion-based chemical sensor
US5863801A (en) * 1996-06-14 1999-01-26 Sarnoff Corporation Automated nucleic acid isolation
US5942443A (en) * 1996-06-28 1999-08-24 Caliper Technologies Corporation High throughput screening assay systems in microscale fluidic devices
US5880071A (en) * 1996-06-28 1999-03-09 Caliper Technologies Corporation Electropipettor and compensation means for electrophoretic bias
US5800690A (en) * 1996-07-03 1998-09-01 Caliper Technologies Corporation Variable control of electroosmotic and/or electrophoretic forces within a fluid-containing structure via electrical forces
US6174365B1 (en) * 1996-07-15 2001-01-16 Sumitomo Metal Industries, Ltd. Apparatus for crystal growth and crystal growth method employing the same
US6221654B1 (en) * 1996-09-25 2001-04-24 California Institute Of Technology Method and apparatus for analysis and sorting of polynucleotides based on size
US6376971B1 (en) * 1997-02-07 2002-04-23 Sri International Electroactive polymer electrodes
US6117634A (en) * 1997-03-05 2000-09-12 The Reagents Of The University Of Michigan Nucleic acid sequencing and mapping
US5904824A (en) * 1997-03-07 1999-05-18 Beckman Instruments, Inc. Microfluidic electrophoresis device
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
US5932799A (en) * 1997-07-21 1999-08-03 Ysi Incorporated Microfluidic analyzer module
US20050123947A1 (en) * 1997-09-23 2005-06-09 California Institute Of Technology Methods and systems for molecular fingerprinting
US6540895B1 (en) * 1997-09-23 2003-04-01 California Institute Of Technology Microfabricated cell sorter for chemical and biological materials
US6107044A (en) * 1997-10-16 2000-08-22 Caliper Technologies Corp. Apparatus and methods for sequencing nucleic acids in microfluidic systems
US6345502B1 (en) * 1997-11-12 2002-02-12 California Institute Of Technology Micromachined parylene membrane valve and pump
US6375871B1 (en) * 1998-06-18 2002-04-23 3M Innovative Properties Company Methods of manufacturing microfluidic articles
US6585939B1 (en) * 1999-02-26 2003-07-01 Orchid Biosciences, Inc. Microstructures for use in biological assays and reactions
US6520936B1 (en) * 1999-06-08 2003-02-18 Medtronic Minimed, Inc. Method and apparatus for infusing liquids using a chemical reaction in an implanted infusion device
US6929030B2 (en) * 1999-06-28 2005-08-16 California Institute Of Technology Microfabricated elastomeric valve and pump systems
US6408878B2 (en) * 1999-06-28 2002-06-25 California Institute Of Technology Microfabricated elastomeric valve and pump systems
US20020054885A1 (en) * 1999-12-22 2002-05-09 Sylvie Laquerre Herpes simplex virus-1 Glycoprotein C mutants for treating unwanted hyperproliferative cell growth
US6358387B1 (en) * 2000-03-27 2002-03-19 Caliper Technologies Corporation Ultra high throughput microfluidic analytical systems and methods
US6409832B2 (en) * 2000-03-31 2002-06-25 Micronics, Inc. Protein crystallization in microfluidic structures
US6690437B2 (en) * 2000-04-18 2004-02-10 Semiconductor Energy Laboratory Co., Ltd. Electro-optical device
US20020037499A1 (en) * 2000-06-05 2002-03-28 California Institute Of Technology Integrated active flux microfluidic devices and methods
US6767706B2 (en) * 2000-06-05 2004-07-27 California Institute Of Technology Integrated active flux microfluidic devices and methods
US20020108096A1 (en) * 2000-06-27 2002-08-08 Michael Lee Microfluidic design automation method and system
US20040115838A1 (en) * 2000-11-16 2004-06-17 Quake Stephen R. Apparatus and methods for conducting assays and high throughput screening
US20030008308A1 (en) * 2001-04-06 2003-01-09 California Institute Of Technology Nucleic acid amplification utilizing microfluidic devices
US20030080442A1 (en) * 2001-08-30 2003-05-01 Fluidigm Corp. Electrostatic/electrostrictive actuation of elastomer structures using compliant electrodes
US20030175947A1 (en) * 2001-11-05 2003-09-18 Liu Robin Hui Enhanced mixing in microfluidic devices
US20030138829A1 (en) * 2001-11-30 2003-07-24 Fluidigm Corp. Microfluidic device and methods of using same
US20030143120A1 (en) * 2002-01-25 2003-07-31 Waldemar Ruediger Parallel chemistry reactor with interchangeable vessel carrying inserts
US20040072278A1 (en) * 2002-04-01 2004-04-15 Fluidigm Corporation Microfluidic particle-analysis systems
US20050197652A1 (en) * 2002-05-13 2005-09-08 Fluidigm Corporation Drug delivery system
US20050053952A1 (en) * 2002-10-02 2005-03-10 California Institute Of Technology Microfluidic nucleic acid analysis
US20040141887A1 (en) * 2002-11-08 2004-07-22 Irm, Llc Apparatus and methods to process substrate surface features
US20050129581A1 (en) * 2003-04-03 2005-06-16 Fluidigm Corporation Microfluidic devices and methods of using same
US20050201901A1 (en) * 2004-01-25 2005-09-15 Fluidigm Corp. Crystal forming devices and systems and methods for using the same

Cited By (133)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9926521B2 (en) 2000-06-27 2018-03-27 Fluidigm Corporation Microfluidic particle-analysis systems
US8658418B2 (en) 2002-04-01 2014-02-25 Fluidigm Corporation Microfluidic particle-analysis systems
US10131934B2 (en) 2003-04-03 2018-11-20 Fluidigm Corporation Thermal reaction device and method for using the same
US8247178B2 (en) 2003-04-03 2012-08-21 Fluidigm Corporation Thermal reaction device and method for using the same
US7749737B2 (en) 2003-04-03 2010-07-06 Fluidigm Corporation Thermal reaction device and method for using the same
US9150913B2 (en) 2003-04-03 2015-10-06 Fluidigm Corporation Thermal reaction device and method for using the same
US7588672B2 (en) 2004-06-07 2009-09-15 Fluidigm Corporation Optical lens system and method for microfluidic devices
US9234237B2 (en) 2004-06-07 2016-01-12 Fluidigm Corporation Optical lens system and method for microfluidic devices
US8926905B2 (en) 2004-06-07 2015-01-06 Fluidigm Corporation Optical lens system and method for microfluidic devices
US20060006067A1 (en) * 2004-06-07 2006-01-12 Fluidigm Corporation Optical lens system and method for microfluidic devices
US10106846B2 (en) 2004-06-07 2018-10-23 Fluidigm Corporation Optical lens system and method for microfluidic devices
US8721968B2 (en) 2004-06-07 2014-05-13 Fluidigm Corporation Optical lens system and method for microfluidic devices
US7906072B2 (en) 2004-06-07 2011-03-15 Fluidigm Corporation Optical lens system and method for microfluidic devices
US20100320364A1 (en) * 2004-06-07 2010-12-23 Fluidigm Corporation Optical lens system and method for microfluidic devices
US20090294703A1 (en) * 2004-06-07 2009-12-03 Fluidigm Corporation Optical lens system and method for microfluidic devices
US8512640B2 (en) 2004-06-07 2013-08-20 Fluidigm Corporation Optical lens system and method for microfluidic devices
US10745748B2 (en) 2004-06-07 2020-08-18 Fluidigm Corporation Optical lens system and method for microfluidic devices
US7307802B2 (en) 2004-06-07 2007-12-11 Fluidigm Corporation Optical lens system and method for microfluidic devices
US8048378B2 (en) 2004-06-07 2011-11-01 Fluidigm Corporation Optical lens system and method for microfluidic devices
US20080088952A1 (en) * 2004-06-07 2008-04-17 Fluidigm Corporation Optical lens system and method for microfluidic devices
US9663821B2 (en) 2004-06-07 2017-05-30 Fluidigm Corporation Optical lens system and method for microfluidic devices
US9316331B2 (en) * 2005-01-25 2016-04-19 Fluidigm Corporation Multilevel microfluidic systems and methods
US8828663B2 (en) 2005-03-18 2014-09-09 Fluidigm Corporation Thermal reaction device and method for using the same
US20110143949A1 (en) * 2005-06-02 2011-06-16 Fluidigm Corporation Analysis using microfluidic partitioning devices
US20110129841A1 (en) * 2005-06-02 2011-06-02 Fluidigm Corporation Analysis using microfluidic partitioning devices
US9364829B2 (en) 2005-06-02 2016-06-14 Fluidigm Corporation Analysis using microfluidic partitioning devices
US20100064780A1 (en) * 2005-07-27 2010-03-18 Howard A Stone Pressure Determination In Microfludic Systems
US20090170113A1 (en) * 2006-02-02 2009-07-02 The Board Of Trustees Of The Leland Stanford Junior University Non-Invasive Fetal Genetic Screening by Digital Analysis
US7888017B2 (en) 2006-02-02 2011-02-15 The Board Of Trustees Of The Leland Stanford Junior University Non-invasive fetal genetic screening by digital analysis
US20100255492A1 (en) * 2006-02-02 2010-10-07 The Board Of Trustees Of The Leland Stanford Junior University Non-Invasive Fetal Genetic Screening by Digital Analysis
US9777328B2 (en) 2006-02-02 2017-10-03 The Board Of Trustees Of The Leland Stanford Junior University Non-invasive fetal genetic screening by digital analysis
US20100255493A1 (en) * 2006-02-02 2010-10-07 The Board Of Trustees Of The Leland Stanford Junior University Non-Invasive Fetal Genetic Screening by Digital Analysis
US20100256013A1 (en) * 2006-02-02 2010-10-07 The Board Of Trustees Of The Leland Stanford Junior University Non-Invasive Fetal Genetic Screening by Digital Analysis
US9777329B2 (en) 2006-02-02 2017-10-03 The Board Of Trustees Of The Leland Stanford Junior University Non-invasive fetal genetic screening by digital analysis
US20100124751A1 (en) * 2006-02-02 2010-05-20 The Board Of Trustees Of The Leland Stanford Junior University Non-Invasive Fetal Genetic Screening by Digital Analysis
US8008018B2 (en) 2006-02-02 2011-08-30 The Board Of Trustees Of The Leland Stanford Junior University Determination of fetal aneuploidies by massively parallel DNA sequencing
US20100124752A1 (en) * 2006-02-02 2010-05-20 The Board Of Trustees Of The Leland Stanford Junior University Non-Invasive Fetal Genetic Screening by Digital Analysis
US9441273B2 (en) 2006-02-02 2016-09-13 The Board Of Trustees Of The Leland Stanford Junior University Non-invasive fetal genetic screening by digital analysis
US11692225B2 (en) 2006-02-02 2023-07-04 The Board Of Trustees Of The Leland Stanford Junior University Non-invasive fetal genetic screening by digital analysis
US10072295B2 (en) 2006-02-02 2018-09-11 The Board Of Trustees Of The Leland Stanford Junior University Non-invasive fetal genetic screening by digtal analysis
US20090170114A1 (en) * 2006-02-02 2009-07-02 The Board Of Trustees Of The Leland Stanford Junior University Non-Invasive Fetal Genetic Screening by Digital Analysis
US20070202525A1 (en) * 2006-02-02 2007-08-30 The Board Of Trustees Of The Leland Stanford Junior University Non-invasive fetal genetic screening by digital analysis
US8293470B2 (en) 2006-02-02 2012-10-23 The Board Of Trustees Of The Leland Stanford Junior University Non-invasive fetal genetic screening by digital analysis
US10591391B2 (en) 2006-06-14 2020-03-17 Verinata Health, Inc. Diagnosis of fetal abnormalities using polymorphisms including short tandem repeats
US8372584B2 (en) 2006-06-14 2013-02-12 The General Hospital Corporation Rare cell analysis using sample splitting and DNA tags
US20080138809A1 (en) * 2006-06-14 2008-06-12 Ravi Kapur Methods for the Diagnosis of Fetal Abnormalities
US20080220422A1 (en) * 2006-06-14 2008-09-11 Daniel Shoemaker Rare cell analysis using sample splitting and dna tags
US10155984B2 (en) 2006-06-14 2018-12-18 The General Hospital Corporation Rare cell analysis using sample splitting and DNA tags
US8168389B2 (en) 2006-06-14 2012-05-01 The General Hospital Corporation Fetal cell analysis using sample splitting
US9273355B2 (en) 2006-06-14 2016-03-01 The General Hospital Corporation Rare cell analysis using sample splitting and DNA tags
US11781187B2 (en) 2006-06-14 2023-10-10 The General Hospital Corporation Rare cell analysis using sample splitting and DNA tags
US10041119B2 (en) 2006-06-14 2018-08-07 Verinata Health, Inc. Methods for the diagnosis of fetal abnormalities
US8137912B2 (en) 2006-06-14 2012-03-20 The General Hospital Corporation Methods for the diagnosis of fetal abnormalities
US10435751B2 (en) 2006-06-14 2019-10-08 Verinata Health, Inc. Methods for the diagnosis of fetal abnormalities
US9017942B2 (en) 2006-06-14 2015-04-28 The General Hospital Corporation Rare cell analysis using sample splitting and DNA tags
US10704090B2 (en) 2006-06-14 2020-07-07 Verinata Health, Inc. Fetal aneuploidy detection by sequencing
US9347100B2 (en) 2006-06-14 2016-05-24 Gpb Scientific, Llc Rare cell analysis using sample splitting and DNA tags
US11261492B2 (en) 2006-06-14 2022-03-01 The General Hospital Corporation Methods for the diagnosis of fetal abnormalities
US11674176B2 (en) 2006-06-14 2023-06-13 Verinata Health, Inc Fetal aneuploidy detection by sequencing
US8603832B2 (en) 2006-10-18 2013-12-10 President And Fellows Of Harvard College Lateral flow and flow-through bioassay devices based on patterned porous media, methods of making same, and methods of using same
US20090298191A1 (en) * 2006-10-18 2009-12-03 President And Fellows Of Harvard College Lateral Flow and Flow-through Bioassay Devices Based On Patterned Porous Media, Methods of Making Same, and Methods of Using Same
US9193988B2 (en) 2006-10-18 2015-11-24 President And Fellows Of Harvard College Lateral flow and flow-through bioassay devices based on patterned porous media, methods of making same, and methods of using same
US8377710B2 (en) 2006-10-18 2013-02-19 President And Fellows Of Harvard College Lateral flow and flow-through bioassay devices based on patterned porous media, methods of making same, and methods of using same
EP2089543A2 (en) * 2006-11-15 2009-08-19 Idaho Technology, Inc. High density self-contained biological analysis
US10889856B2 (en) 2006-11-15 2021-01-12 Biofire Diagnostics, Llc High density self-contained biological analysis
US11866774B2 (en) 2006-11-15 2024-01-09 Biofire Diagnostics, Llc High density self-contained biological analysis
US10053728B2 (en) 2006-11-15 2018-08-21 Biofire Diagnostics, Llc High density self-contained biological analysis
EP2089543A4 (en) * 2006-11-15 2013-04-17 Biofire Diagnostics Inc High density self-contained biological analysis
US20080137584A1 (en) * 2006-12-08 2008-06-12 Samsung Electronics Co., Ltd. Apparatus and method for selecting frame structure in multihop relay broadband wireless access communication system
US9121069B2 (en) 2007-07-23 2015-09-01 The Chinese University Of Hong Kong Diagnosing cancer using genomic sequencing
US10619214B2 (en) 2007-07-23 2020-04-14 The Chinese University Of Hong Kong Detecting genetic aberrations associated with cancer using genomic sequencing
US20090029377A1 (en) * 2007-07-23 2009-01-29 The Chinese University Of Hong Kong Diagnosing fetal chromosomal aneuploidy using massively parallel genomic sequencing
US8442774B2 (en) 2007-07-23 2013-05-14 The Chinese University Of Hong Kong Diagnosing fetal chromosomal aneuploidy using paired end
US20100112590A1 (en) * 2007-07-23 2010-05-06 The Chinese University Of Hong Kong Diagnosing Fetal Chromosomal Aneuploidy Using Genomic Sequencing With Enrichment
US9051616B2 (en) 2007-07-23 2015-06-09 The Chinese University Of Hong Kong Diagnosing fetal chromosomal aneuploidy using massively parallel genomic sequencing
US11142799B2 (en) 2007-07-23 2021-10-12 The Chinese University Of Hong Kong Detecting chromosomal aberrations associated with cancer using genomic sequencing
US8972202B2 (en) 2007-07-23 2015-03-03 The Chinese University Of Hong Kong Diagnosing fetal chromosomal aneuploidy using massively parallel genomic sequencing
WO2009017627A1 (en) * 2007-07-30 2009-02-05 Gn Biosystems Incorporated Apparatus and method for conducting high-throughput micro-volume experiments
US20110111517A1 (en) * 2008-03-27 2011-05-12 President And Fellows Of Harvard College Paper-based microfluidic systems
US8921118B2 (en) 2008-03-27 2014-12-30 President And Fellows Of Harvard College Paper-based microfluidic systems
US9829488B2 (en) 2008-03-27 2017-11-28 President And Fellows Of Havard College Paper-based cellular arrays
US20110123398A1 (en) * 2008-03-27 2011-05-26 President And Fellows Of Harvard College Three-dimensional microfluidic devices
US20110105360A1 (en) * 2008-03-27 2011-05-05 President And Fellows Of Harvard College Paper-based cellular arrays
US20110189786A1 (en) * 2008-03-27 2011-08-04 President And Fellows Of Harvard College Cotton thread as a low-cost multi-assay diagnostic platform
US8206992B2 (en) 2008-03-27 2012-06-26 President And Fellows Of Harvard College Cotton thread as a low-cost multi-assay diagnostic platform
WO2009121041A3 (en) * 2008-03-27 2009-12-17 President And Fellows Of Harvard College Paper-based microfluidic systems
US8628729B2 (en) 2008-03-27 2014-01-14 President And Fellows Of Harvard College Three-dimensional microfluidic devices
US20110206576A1 (en) * 2008-08-07 2011-08-25 Fluidigm Corporation Microfluidic mixing and reaction systems for high efficiency screening
US9182322B2 (en) 2008-08-07 2015-11-10 Fluidigm Corporation Microfluidic mixing and reaction systems for high efficiency screening
US8617488B2 (en) * 2008-08-07 2013-12-31 Fluidigm Corporation Microfluidic mixing and reaction systems for high efficiency screening
US8296076B2 (en) 2008-09-20 2012-10-23 The Board Of Trustees Of The Leland Stanford Junior University Noninvasive diagnosis of fetal aneuoploidy by sequencing
US8195415B2 (en) 2008-09-20 2012-06-05 The Board Of Trustees Of The Leland Stanford Junior University Noninvasive diagnosis of fetal aneuploidy by sequencing
US10669585B2 (en) 2008-09-20 2020-06-02 The Board Of Trustees Of The Leland Stanford Junior University Noninvasive diagnosis of fetal aneuploidy by sequencing
US9404157B2 (en) 2008-09-20 2016-08-02 The Board Of Trustees Of The Leland Stanford Junior University Noninvasive diagnosis of fetal aneuploidy by sequencing
US9353414B2 (en) 2008-09-20 2016-05-31 The Board Of Trustees Of The Leland Stanford Junior University Noninvasive diagnosis of fetal aneuploidy by sequencing
US8682594B2 (en) 2008-09-20 2014-03-25 The Board Of Trustees Of The Leland Stanford Junior University Noninvasive diagnosis of fetal aneuploidy by sequencing
US20100138165A1 (en) * 2008-09-20 2010-06-03 The Board Of Trustees Of The Leland Stanford Junior University Noninvasive Diagnosis of Fetal Aneuploidy by Sequencing
US9192933B2 (en) 2009-03-06 2015-11-24 President And Fellows Of Harvard College Microfluidic, electrochemical devices
US9205468B2 (en) 2009-11-30 2015-12-08 Fluidigm Corporation Microfluidic device regeneration
US8821810B2 (en) 2010-02-03 2014-09-02 President And Fellows Of Harvard College Devices and methods for multiplexed assays
US9128101B2 (en) 2010-03-01 2015-09-08 Caris Life Sciences Switzerland Holdings Gmbh Biomarkers for theranostics
US9469876B2 (en) 2010-04-06 2016-10-18 Caris Life Sciences Switzerland Holdings Gmbh Circulating biomarkers for metastatic prostate cancer
US9625404B2 (en) 2011-01-11 2017-04-18 The Trustees Of Columbia University In The City Of New York Systems and methods for single-molecule detection using nanotubes
US9891182B2 (en) 2011-01-11 2018-02-13 The Trustees Of Columbia University In The City Of New York Systems and methods for single-molecule detection using nanotubes
US10684240B2 (en) 2011-01-11 2020-06-16 The Trustees Of Columbia University In The City Of New York Systems and methods for single-molecule detection using nanotubes
US11624727B2 (en) 2011-02-23 2023-04-11 The Trustees Of Columbia University In The City Of New York Systems and methods for single-molecule detection using nanopores
US20130224879A1 (en) * 2011-08-26 2013-08-29 The Trustees Of Columbia University In The City Of New York Systems and methods for a dna-based thermometer
US9194801B2 (en) * 2011-08-26 2015-11-24 The Trustees Of Columbia University In The City Of New York Systems and methods for a DNA-based thermometer
US10627364B2 (en) 2012-04-10 2020-04-21 The Trustees Of Columbia University In The City Of New York Systems and methods for biological ion channel interfaces
US9841416B2 (en) 2012-04-20 2017-12-12 The Trustees Of Columbia University In The City Of New York Systems and methods for single-molecule nucleic-acid assay platforms
US10401353B2 (en) 2012-04-20 2019-09-03 The Trustees Of Columbia University In The City Of New York Systems and methods for single-molecule nucleic-acid assay platforms
US9840732B2 (en) 2012-05-21 2017-12-12 Fluidigm Corporation Single-particle analysis of particle populations
US9958448B2 (en) 2012-10-23 2018-05-01 Caris Life Sciences Switzerland Holdings Gmbh Aptamers and uses thereof
EP4170031A1 (en) 2012-10-23 2023-04-26 Caris Science, Inc. Aptamers and uses thereof
WO2014068408A2 (en) 2012-10-23 2014-05-08 Caris Life Sciences Switzerland Holdings, S.A.R.L. Aptamers and uses thereof
US10942184B2 (en) 2012-10-23 2021-03-09 Caris Science, Inc. Aptamers and uses thereof
US9939443B2 (en) 2012-12-19 2018-04-10 Caris Life Sciences Switzerland Holdings Gmbh Compositions and methods for aptamer screening
WO2014100434A1 (en) 2012-12-19 2014-06-26 Caris Science, Inc. Compositions and methods for aptamer screening
WO2015031694A2 (en) 2013-08-28 2015-03-05 Caris Science, Inc. Oligonucleotide probes and uses thereof
US10364467B2 (en) 2015-01-13 2019-07-30 The Chinese University Of Hong Kong Using size and number aberrations in plasma DNA for detecting cancer
WO2016145128A1 (en) 2015-03-09 2016-09-15 Caris Science, Inc. Oligonucleotide probes and uses thereof
US11091765B2 (en) 2015-06-29 2021-08-17 Caris Science, Inc. Therapeutic oligonucleotides
US10590425B2 (en) 2015-06-29 2020-03-17 Caris Science, Inc. Therapeutic oligonucleotides
WO2017004243A1 (en) 2015-06-29 2017-01-05 Caris Science, Inc. Therapeutic oligonucleotides
US10941176B2 (en) 2015-07-28 2021-03-09 Caris Science, Inc. Therapeutic oligonucleotides
WO2017019918A1 (en) 2015-07-28 2017-02-02 Caris Science, Inc. Targeted oligonucleotides
US11725023B2 (en) 2015-07-28 2023-08-15 Caris Science, Inc. Therapeutic oligonucleotides
EP3828272A1 (en) 2016-03-18 2021-06-02 Caris Science, Inc. Oligonucleotide probes and uses thereof
US11332748B2 (en) 2016-03-18 2022-05-17 Caris Science, Inc. Oligonucleotide probes and uses thereof
US10731166B2 (en) 2016-03-18 2020-08-04 Caris Science, Inc. Oligonucleotide probes and uses thereof
US11293017B2 (en) 2016-05-25 2022-04-05 Caris Science, Inc. Oligonucleotide probes and uses thereof
WO2017205686A1 (en) 2016-05-25 2017-11-30 Caris Science, Inc. Oligonucleotide probes and uses thereof
CN110850083A (en) * 2019-10-31 2020-02-28 浙江卫未生物医药科技有限公司 Batch exosome concentration detection kit and operation method

Also Published As

Publication number Publication date
US20080292504A1 (en) 2008-11-27
US7867454B2 (en) 2011-01-11

Similar Documents

Publication Publication Date Title
US10131934B2 (en) Thermal reaction device and method for using the same
US7867454B2 (en) Thermal reaction device and method for using the same
US7476363B2 (en) Microfluidic devices and methods of using same
US7666361B2 (en) Microfluidic devices and methods of using same
US8343442B2 (en) Microfluidic device and methods of using same
US8828663B2 (en) Thermal reaction device and method for using the same
US8163492B2 (en) Microfluidic device and methods of using same
US20180243741A1 (en) Thermal reaction device and method for using the same
CA2807591C (en) Microfluidic devices and methods of using same

Legal Events

Date Code Title Description
AS Assignment

Owner name: FLUIDIGM CORPORATION, COLORADO

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:GOODSAID, FEDERICO;UNGER, MARC;HUANG, JIANG;AND OTHERS;REEL/FRAME:016192/0304;SIGNING DATES FROM 20050104 TO 20050111

AS Assignment

Owner name: FLUIDIGM CORPORATION - A DELAWARE CORPORATION, CAL

Free format text: REINCORPORATION ASSIGNMENT;ASSIGNOR:FLUIDIGM CORPORATION - A CALIFORNIA CORPORATION;REEL/FRAME:019899/0313

Effective date: 20070928

Owner name: FLUIDIGM CORPORATION - A DELAWARE CORPORATION,CALI

Free format text: REINCORPORATION ASSIGNMENT;ASSIGNOR:FLUIDIGM CORPORATION - A CALIFORNIA CORPORATION;REEL/FRAME:019899/0313

Effective date: 20070928

STCB Information on status: application discontinuation

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