US20080014589A1 - Microfluidic devices and methods of use thereof - Google Patents

Microfluidic devices and methods of use thereof Download PDF

Info

Publication number
US20080014589A1
US20080014589A1 US11/803,101 US80310107A US2008014589A1 US 20080014589 A1 US20080014589 A1 US 20080014589A1 US 80310107 A US80310107 A US 80310107A US 2008014589 A1 US2008014589 A1 US 2008014589A1
Authority
US
United States
Prior art keywords
droplets
fluid
sample fluid
channel
continuous phase
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
US11/803,101
Inventor
Darren Link
Michael Weiner
David Marran
Jonathan Rothberg
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.)
Bio Rad Laboratories Inc
Original Assignee
Raindance Technologies Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Raindance Technologies Inc filed Critical Raindance Technologies Inc
Priority to US11/803,101 priority Critical patent/US20080014589A1/en
Assigned to RAINDANCE TECHNOLOGIES, INC. reassignment RAINDANCE TECHNOLOGIES, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: MARRAN, DAVID, LINK, DARREN R., ROTHBERG, JONATHAN M., WEINER, MICHAEL
Publication of US20080014589A1 publication Critical patent/US20080014589A1/en
Assigned to CAPITAL ROYALTY PARTNERS II - PARALLEL FUND "A" L.P., CAPITAL ROYALTY PARTNERS II L.P., PARALLEL INVESTMENT OPPORTUNITIES PARTNERS II L.P. reassignment CAPITAL ROYALTY PARTNERS II - PARALLEL FUND "A" L.P. SHORT-FORM PATENT SECURITY AGREEMENT Assignors: RAINDANCE TECHNOLOGIES, INC.
Assigned to RAINDANCE TECHNOLOGIES, INC. reassignment RAINDANCE TECHNOLOGIES, INC. RELEASE BY SECURED PARTY (SEE DOCUMENT FOR DETAILS). Assignors: CAPITAL ROYALTY PARTNERS II (CAYMAN) L.P., CAPITAL ROYALTY PARTNERS II L.P., CRG ISSUER 2015-1, PARALLEL INVESTMENT OPPORTUNITIES PARTNERS II L.P.
Assigned to BIO-RAD LABORATORIES, INC. reassignment BIO-RAD LABORATORIES, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: RAINDANCE TECHNOLOGIES, INC.
Abandoned legal-status Critical Current

Links

Images

Classifications

    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01FMIXING, e.g. DISSOLVING, EMULSIFYING OR DISPERSING
    • B01F23/00Mixing according to the phases to be mixed, e.g. dispersing or emulsifying
    • B01F23/40Mixing liquids with liquids; Emulsifying
    • B01F23/41Emulsifying
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01FMIXING, e.g. DISSOLVING, EMULSIFYING OR DISPERSING
    • B01F33/00Other mixers; Mixing plants; Combinations of mixers
    • B01F33/30Micromixers
    • B01F33/301Micromixers using specific means for arranging the streams to be mixed, e.g. channel geometries or dispositions
    • B01F33/3011Micromixers using specific means for arranging the streams to be mixed, e.g. channel geometries or dispositions using a sheathing stream of a fluid surrounding a central stream of a different fluid, e.g. for reducing the cross-section of the central stream or to produce droplets from the central stream
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J19/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J19/0093Microreactors, e.g. miniaturised or microfabricated reactors
    • 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
    • B01L3/502769Containers 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 multiphase flow arrangements
    • B01L3/502784Containers 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 multiphase flow arrangements specially adapted for droplet or plug flow, e.g. digital microfluidics
    • 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
    • B01L3/56Labware specially adapted for transferring fluids
    • B01L3/565Seals
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B03SEPARATION OF SOLID MATERIALS USING LIQUIDS OR USING PNEUMATIC TABLES OR JIGS; MAGNETIC OR ELECTROSTATIC SEPARATION OF SOLID MATERIALS FROM SOLID MATERIALS OR FLUIDS; SEPARATION BY HIGH-VOLTAGE ELECTRIC FIELDS
    • B03CMAGNETIC OR ELECTROSTATIC SEPARATION OF SOLID MATERIALS FROM SOLID MATERIALS OR FLUIDS; SEPARATION BY HIGH-VOLTAGE ELECTRIC FIELDS
    • B03C5/00Separating dispersed particles from liquids by electrostatic effect
    • B03C5/005Dielectrophoresis, i.e. dielectric particles migrating towards the region of highest field strength
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B03SEPARATION OF SOLID MATERIALS USING LIQUIDS OR USING PNEUMATIC TABLES OR JIGS; MAGNETIC OR ELECTROSTATIC SEPARATION OF SOLID MATERIALS FROM SOLID MATERIALS OR FLUIDS; SEPARATION BY HIGH-VOLTAGE ELECTRIC FIELDS
    • B03CMAGNETIC OR ELECTROSTATIC SEPARATION OF SOLID MATERIALS FROM SOLID MATERIALS OR FLUIDS; SEPARATION BY HIGH-VOLTAGE ELECTRIC FIELDS
    • B03C5/00Separating dispersed particles from liquids by electrostatic effect
    • B03C5/02Separators
    • B03C5/022Non-uniform field separators
    • B03C5/026Non-uniform field separators using open-gradient differential dielectric separation, i.e. using electrodes of special shapes for non-uniform field creation, e.g. Fluid Integrated Circuit [FIC]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/10Processes for the isolation, preparation or purification of DNA or RNA
    • C12N15/1034Isolating an individual clone by screening libraries
    • C12N15/1068Template (nucleic acid) mediated chemical library synthesis, e.g. chemical and enzymatical DNA-templated organic molecule synthesis, libraries prepared by non ribosomal polypeptide synthesis [NRPS], DNA/RNA-polymerase mediated polypeptide synthesis
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/10Processes for the isolation, preparation or purification of DNA or RNA
    • C12N15/1034Isolating an individual clone by screening libraries
    • C12N15/1086Preparation or screening of expression libraries, e.g. reporter assays
    • 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
    • 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
    • C12Q1/6806Preparing nucleic acids for analysis, e.g. for polymerase chain reaction [PCR] assay
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6813Hybridisation assays
    • C12Q1/6834Enzymatic or biochemical coupling of nucleic acids to a solid phase
    • C12Q1/6837Enzymatic or biochemical coupling of nucleic acids to a solid phase using probe arrays or probe chips
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6844Nucleic acid amplification reactions
    • 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
    • C12Q1/6846Common amplification features
    • 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
    • C12Q1/6853Nucleic acid amplification reactions using modified primers or templates
    • C12Q1/6855Ligating adaptors
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6844Nucleic acid amplification reactions
    • C12Q1/686Polymerase chain reaction [PCR]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6869Methods for sequencing
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6869Methods for sequencing
    • C12Q1/6874Methods for sequencing involving nucleic acid arrays, e.g. sequencing by hybridisation
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N15/00Investigating characteristics of particles; Investigating permeability, pore-volume, or surface-area of porous materials
    • G01N15/10Investigating individual particles
    • G01N15/14Electro-optical investigation, e.g. flow cytometers
    • G01N15/1468Electro-optical investigation, e.g. flow cytometers with spatial resolution of the texture or inner structure of the particle
    • G01N15/147Electro-optical investigation, e.g. flow cytometers with spatial resolution of the texture or inner structure of the particle the analysis being performed on a sample stream
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N21/00Investigating or analysing materials by the use of optical means, i.e. using sub-millimetre waves, infrared, visible or ultraviolet light
    • G01N21/01Arrangements or apparatus for facilitating the optical investigation
    • G01N21/03Cuvette constructions
    • G01N21/05Flow-through cuvettes
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N21/00Investigating or analysing materials by the use of optical means, i.e. using sub-millimetre waves, infrared, visible or ultraviolet light
    • G01N21/62Systems in which the material investigated is excited whereby it emits light or causes a change in wavelength of the incident light
    • G01N21/63Systems in which the material investigated is excited whereby it emits light or causes a change in wavelength of the incident light optically excited
    • G01N21/64Fluorescence; Phosphorescence
    • 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
    • G01N27/28Electrolytic cell components
    • G01N27/30Electrodes, e.g. test electrodes; Half-cells
    • G01N27/327Biochemical electrodes, e.g. electrical or mechanical details for in vitro measurements
    • G01N27/3275Sensing specific biomolecules, e.g. nucleic acid strands, based on an electrode surface reaction
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N35/00Automatic analysis not limited to methods or materials provided for in any single one of groups G01N1/00 - G01N33/00; Handling materials therefor
    • G01N35/08Automatic analysis not limited to methods or materials provided for in any single one of groups G01N1/00 - G01N33/00; Handling materials therefor using a stream of discrete samples flowing along a tube system, e.g. flow injection analysis
    • 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
    • B01L2200/02Adapting objects or devices to another
    • B01L2200/026Fluid interfacing between devices or objects, e.g. connectors, inlet details
    • B01L2200/027Fluid interfacing between devices or objects, e.g. connectors, inlet details for microfluidic devices
    • 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
    • B01L2200/0636Focussing flows, e.g. to laminate flows
    • 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
    • B01L2200/0647Handling flowable solids, e.g. microscopic beads, cells, particles
    • 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
    • B01L2200/0673Handling of plugs of fluid surrounded by immiscible fluid
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01LCHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
    • B01L2200/00Solutions for specific problems relating to chemical or physical laboratory apparatus
    • B01L2200/10Integrating sample preparation and analysis in single entity, e.g. lab-on-a-chip concept
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01LCHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
    • 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
    • B01L2300/0861Configuration 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
    • B01L2300/16Surface properties and coatings
    • B01L2300/161Control and use of surface tension forces, e.g. hydrophobic, hydrophilic
    • B01L2300/165Specific details about hydrophobic, oleophobic surfaces
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01LCHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
    • B01L2400/00Moving or stopping fluids
    • B01L2400/04Moving fluids with specific forces or mechanical means
    • B01L2400/0403Moving fluids with specific forces or mechanical means specific forces
    • B01L2400/0415Moving fluids with specific forces or mechanical means specific forces electrical forces, e.g. electrokinetic
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01LCHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
    • B01L2400/00Moving or stopping fluids
    • B01L2400/04Moving fluids with specific forces or mechanical means
    • B01L2400/0403Moving fluids with specific forces or mechanical means specific forces
    • B01L2400/0415Moving fluids with specific forces or mechanical means specific forces electrical forces, e.g. electrokinetic
    • B01L2400/0424Dielectrophoretic forces
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01LCHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
    • B01L2400/00Moving or stopping fluids
    • B01L2400/04Moving fluids with specific forces or mechanical means
    • B01L2400/0475Moving 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
    • B01L2400/08Regulating or influencing the flow resistance
    • B01L2400/084Passive control of flow resistance
    • B01L2400/086Passive control of flow resistance using baffles or other fixed flow obstructions
    • 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
    • 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
    • B01L3/502746Containers 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 for controlling flow resistance, e.g. flow controllers, baffles
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01LCHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
    • B01L7/00Heating or cooling apparatus; Heat insulating devices
    • B01L7/52Heating or cooling apparatus; Heat insulating devices with provision for submitting samples to a predetermined sequence of different temperatures, e.g. for treating nucleic acid samples
    • B01L7/525Heating or cooling apparatus; Heat insulating devices with provision for submitting samples to a predetermined sequence of different temperatures, e.g. for treating nucleic acid samples with physical movement of samples between temperature zones
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01LCHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
    • B01L9/00Supporting devices; Holding devices
    • B01L9/52Supports specially adapted for flat sample carriers, e.g. for plates, slides, chips
    • B01L9/527Supports specially adapted for flat sample carriers, e.g. for plates, slides, chips for microfluidic devices, e.g. used for lab-on-a-chip
    • 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
    • C12Q2565/00Nucleic acid analysis characterised by mode or means of detection
    • C12Q2565/60Detection means characterised by use of a special device
    • C12Q2565/628Detection means characterised by use of a special device being a surface plasmon resonance spectrometer
    • 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
    • C12Q2565/00Nucleic acid analysis characterised by mode or means of detection
    • C12Q2565/60Detection means characterised by use of a special device
    • C12Q2565/629Detection means characterised by use of a special device being a microfluidic device
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N21/00Investigating or analysing materials by the use of optical means, i.e. using sub-millimetre waves, infrared, visible or ultraviolet light
    • G01N21/01Arrangements or apparatus for facilitating the optical investigation
    • G01N21/03Cuvette constructions
    • G01N2021/0346Capillary cells; Microcells
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N35/00Automatic analysis not limited to methods or materials provided for in any single one of groups G01N1/00 - G01N33/00; Handling materials therefor
    • G01N2035/00178Special arrangements of analysers
    • G01N2035/00237Handling microquantities of analyte, e.g. microvalves, capillary networks
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N35/00Automatic analysis not limited to methods or materials provided for in any single one of groups G01N1/00 - G01N33/00; Handling materials therefor
    • G01N2035/00178Special arrangements of analysers
    • G01N2035/00326Analysers with modular structure
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2201/00Features of devices classified in G01N21/00
    • G01N2201/02Mechanical
    • G01N2201/024Modular construction
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10TTECHNICAL SUBJECTS COVERED BY FORMER US CLASSIFICATION
    • Y10T137/00Fluid handling
    • Y10T137/8593Systems
    • Y10T137/87571Multiple inlet with single outlet
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10TTECHNICAL SUBJECTS COVERED BY FORMER US CLASSIFICATION
    • Y10T137/00Fluid handling
    • Y10T137/8593Systems
    • Y10T137/87571Multiple inlet with single outlet
    • Y10T137/87587Combining by aspiration
    • Y10T137/87619With selectively operated flow control means in inlet
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10TTECHNICAL SUBJECTS COVERED BY FORMER US CLASSIFICATION
    • Y10T137/00Fluid handling
    • Y10T137/8593Systems
    • Y10T137/87571Multiple inlet with single outlet
    • Y10T137/87652With means to promote mixing or combining of plural fluids

Definitions

  • the present invention generally relates to systems and methods for the formation and/or control of fluidic species, and articles produced by such systems and methods. More particularly, the present invention relates to the development of high throughput microfluidic devices for precision fluid handling and use of such systems in various biological, chemical, or diagnostic assays.
  • Microfluidic systems have been described in a variety of contexts, typically in the context of miniaturized laboratory (e.g., clinical) analysis. Other uses have been described as well.
  • International Patent Application Publication No. WO 01/89788 describes multi-level microfluidic systems that can be used to provide patterns of materials, such as biological materials and cells, on surfaces.
  • Other publications describe microfluidic systems including valves, switches, and other components.
  • microfluidic devices Precision manipulation of streams of fluids with microfluidic devices is revolutionizing many fluid-based technologies. Networks of small channels are a flexible platform for the precision manipulation of small amounts of fluids.
  • the utility of such microfluidic devices depends critically on enabling technologies such as the microfluidic peristaltic pump, electrokinetic pumping, dielectrophoretic pump or electrowetting driven flow.
  • the assembly of such modules into complete systems provides a convenient and robust way to construct microfluidic devices.
  • virtually all microfluidic devices are based on flows of streams of fluids; this sets a limit on the smallest volume of reagent that can effectively be used because of the contaminating effects of diffusion and surface adsorption.
  • an electrically addressable emulsification system that combines compartmentalization and electrical manipulation, which allows for multi-step chemical processing, including analysis and sorting, to be initiated in confinement with vibrant timing and metering precision, for use in a variety of chemical, biological, and screening assays, in which the cost and time to perform such assays would be drastically reduced. It would also be desirable to develop a device using dielectrophoretic force (which does not rely on charge density) to manipulate droplets so that more than one electrical pondermotive function can be carried out following a significantly long delay from droplet formation.
  • the present invention provides substrates having individual fluid handling modules that can be combined into fluid processing systems so as to perform multi-step processing of isolated components, which is essential to perform biological, chemical and diagnostic applications, quickly, effectively and inexpensively.
  • the microfluidic substrates of the present invention can encapsulate reagents into droplets, which can be combined, analyzed, and sorted.
  • the present invention provides a method of pairing sample fluids to form a droplet or nanoreactor including, for example, (a) providing a microfluidic substrate including at least two inlet channels adapted to carry at least two dispersed phase sample fluids and at least one main channel adapted to carry at least one continuous phase fluid; (b) flowing a first sample fluid through a first inlet channel which is in fluid communication with the main channel at a junction, wherein the junction includes a first fluidic nozzle designed for flow focusing such that the first sample fluid forms a plurality of highly uniform, monodisperse droplets of a first size in the continuous phase; (c) flowing a second sample fluid through a second inlet channel which is in fluid communication with the main channel at a junction, wherein the junction includes a second fluidic nozzle designed for flow focusing such that the second sample fluid forms a plurality of highly uniform, monodisperse droplets of a second size in the continuous phase, wherein the size of the droplets of the second sample fluid are smaller than the size of the droplets
  • the present invention also provides various methods of forming a droplet emulsion library of a sample fluid.
  • One method includes, for example, (a) providing at least one first channel adapted to carry at least one dispersed phase sample fluid and at least one second channel adapted to carry at least one continuous phase fluid; (b) flowing the sample fluid through the first channel which is in fluid communication with the second channel at a junction, wherein the junction includes a fluidic nozzle such that the sample fluid forms a plurality of highly uniform, monodisperse droplets of a predetermined size in the continuous phase, wherein the fluidic nozzle is isolated from the dispersed phase fluid, has a three dimensional design to permit flow focusing and eliminates surface wetting.
  • the nozzle can be formed from small bore tubing or from the tip of a molded ferrule.
  • Another method of forming a droplet emulsion library of a sample fluid includes, for example, (a) providing a microfluidic substrate including at least one first channel adapted to carry at least one dispersed phase sample fluid and at least one second channel adapted to carry at least one continuous phase fluid; (b) flowing the sample fluid through the first channel which is in fluid communication with the second channel at a junction, wherein the junction includes a fluidic nozzle such that the sample fluid forms a plurality of highly uniform, monodisperse droplets of a predetermined size in the continuous phase.
  • Another method of forming a droplet emulsion library of a sample fluid includes, for example, (a) providing a microfluidic substrate including at least one first channel adapted to carry at least one dispersed phase sample fluid and at least one channel adapted to carry at least one continuous phase fluid; (b) providing a means for storing the sample fluid wherein the storage means is in fluid communication with the first channel and provides means for introducing the sample fluid to the inlet channel, wherein the storage means contains an immiscible phase fluid with a density less than that of the sample fluid; (c) introducing the sample fluid into the storage means wherein the sample fluid flows through the less dense immiscible fluid such that the sample fluid settles at the bottom of the storage means and is subsequently introduced into the first channel; (d) flowing the sample fluid through the first channel which is in fluid communication with the second channel at a junction, wherein the junction includes a fluidic nozzle such that the sample fluid forms a plurality of highly uniform, monodisperse droplets of a predetermined size in the continuous
  • Another method of forming a droplet emulsion library of a sample fluid includes, for example, (a) providing a microfluidic substrate including at least one first channel adapted to carry at least one dispersed phase sample fluid and at least one channel adapted to carry at least one continuous phase fluid; (b) providing a means for storing the sample fluid wherein the storage means is in fluid communication with the first channel and provides means for introducing the sample fluid to the inlet channel, wherein the storage means contains an immiscible phase fluid with a density greater than that of the sample fluid; (c) inserting a sample fluid introduction apparatus into the storage means wherein the sample fluid is forced through the more dense immiscible fluid by the introduction apparatus such that the sample fluid is subsequently introduced into the first channel; (d) flowing the sample fluid through the first channel which is in fluid communication with the second channel at a junction, wherein the junction includes a fluidic nozzle such that the sample fluid forms a plurality of highly uniform, monodisperse droplets of a predetermined size in the continuous phase.
  • the storage means can be a well or reservoir.
  • the fluid introduction apparatus can be a sample tip loading pump.
  • the present invention also provides a method of forming a uniformed sized droplet emulsion library including, for example, (a) providing a means for separating droplets of similar sizes, wherein the means includes a periodic array of geometric parameters defining an obstacle matrix; (b) introducing at least one sample fluid containing various sized droplets to the separating means; (c) subjecting the sample fluid to laminar flow through the microscale obstacles within the separating means, wherein the sample fluids do not mix; and (d) separating and isolating uniformed sized droplets from within the sample fluid by deterministic lateral displacement.
  • the means can be a microfluidic channel, microfluidic lateral diffusion device, tube, syringe, column or a capillary.
  • the present invention also provides various methods for solidifying a droplet or nanoreactor.
  • One method includes, for example, (a) providing a microfluidic substrate including at least one inlet channel adapted to carry at least one dispersed phase sample fluid and at least one main channel adapted to carry at least one continuous phase fluid; (b) incorporating at least one solidifying agent within a sample fluid; (c) flowing the sample fluid through a first inlet channel which is in fluid communication with the main channel at a junction, such that the first sample fluid forms a plurality of highly uniform, monodisperse droplets in the continuous phase; (d) providing a means which activates the solidifying agent such that the droplet or nanoreactor forms a matrix.
  • Another method for solidifying a droplet or nanoreactor includes, for example, (a) providing a microfluidic substrate including at least two inlet channels adapted to carry at least two dispersed phase sample fluids and at least one main channel adapted to carry at least one continuous phase fluid; (b) incorporating at least one solidifying agent within at least one sample fluid; (c) flowing a first sample fluid through a first inlet channel which is in fluid communication with the main channel at a junction, such that the first sample fluid forms a plurality of highly uniform, monodisperse droplets in the continuous phase; (d) flowing a second sample fluid through a second inlet channel which is in fluid communication with the main channel at a junction, such that the second sample fluid forms a plurality of highly uniform, monodisperse droplets in the continuous phase; (e) coalescing at least one droplet formed in step (c) with at least one droplet formed in step (d) as the droplets pass through an electric field, thereby producing a droplet or nanoreactor; and (f) providing
  • the solidifying agent can be a low temperature agarose or a polymerizing solution.
  • the means for activating the solidifying agent can be a physical or chemical means.
  • the present invention also provides various methods for introducing sample fluid to a microfluidic substrate.
  • One method includes, for example, (a) providing a microfluidic substrate including at least one inlet channel adapted to carry at least one dispersed phase sample fluid and at least one main channel adapted to carry at least one continuous phase fluid; (b) providing a means for storing the sample fluid, wherein the storage means is in fluid communication with the inlet channel and provides a means for introducing the sample fluid into the inlet channel; (c) combining the sample fluid with at least one immiscible phase fluid within the storage means, wherein the immiscible phase fluid has a density different from that of the sample fluid such that the fluids separate into distinct layers; (d) providing a force such that the immiscible phase fluid forces the sample fluid completely into the inlet channel of the microfluidic substrate.
  • Another method for introducing sample fluid to a microfluidic substrate includes, for example, (a) providing a microfluidic substrate including at least one inlet channel adapted to carry at least one dispersed phase sample fluid and at least one main channel adapted to carry at least one continuous phase fluid; (b) providing a means for storing the sample fluid, wherein the storage means is in fluid communication with the inlet channel and provides a means for introducing the sample fluid into the inlet channel; (c) combining the sample fluid with at least two immiscible phase fluids within the storage means, wherein a first immiscible phase fluid has a density greater than that of the sample fluid and a second immiscible phase fluid has a density less than that of the sample fluid such that the fluids separate into distinct layers with the sample fluid layer residing between the two immiscible phase layers; (d) providing a force such that the more dense immiscible phase fluid forces the less dense immiscible phase fluid and the sample fluid completely into the inlet channel of the microflu
  • the storage means can be a well or reservoir.
  • the immiscible fluid can be biologically and/or chemically inert.
  • the present invention also provides a method of extracting biological or chemical material from within a droplet or nanoreactor including, for example, (a) providing one or more droplets or nanoreactors formed within a microfluidic substrate; (b) combining the droplets or nanoreactors with a first immiscible fluid with a density greater than that of the droplets or nanoreactors such that the droplets or nanoreactors and immiscible fluid form separate layers; (c) removing the droplet or nanoreactor layer formed in step (b); (d) combining and mixing the droplet or nanoreactor layer with a second immiscible fluid including a destabilizing surfactant such that the droplets or nanoreactors and immiscible fluid form separate layers; and (e) removing the resulting aqueous layer formed step (d), thereby extracting the biological or chemical material from within the droplet or nanoreactor.
  • the immiscible fluid can be biologically and/or chemically inert.
  • the destabilizing surfactant can be a perfluorinated alcohol.
  • the droplets or nanoreactors can reside on top of the immiscible fluids within the separate layers.
  • the present invention also provides various methods for amplifying DNA.
  • One method includes, for example, (a) providing a microfluidic substrate including at least one inlet channel adapted to carry at least one dispersed phase sample fluid and at least one main channel adapted to carry at least one continuous phase fluid, wherein the main channel includes a serpentine line with heating and cooling regions; (b) flowing a sample fluid including one more target DNA molecules to be amplified, PCR primer pair sets, dNTPs, enzymes and buffer components effective to permit PCR amplification through an inlet channel which is in fluid communication with the main channel at a junction, such that the sample fluid forms a plurality of highly uniform, monodisperse droplets in the continuous phase, wherein the junction is heated to 95° C.
  • Another method of amplifying DNA includes, for example, (a) providing a microfluidic substrate including at least two inlet channels adapted to carry at least two dispersed phase sample fluids and at least one main channel adapted to carry at least one continuous phase fluid, wherein the main channel includes a serpentine line with heating and cooling regions; (b) flowing a first sample fluid including one more target DNA molecules to be amplified through a first inlet channel which is in fluid communication with the main channel at a junction, such that the first sample fluid forms a plurality of highly uniform, monodisperse droplets in the continuous phase; (c) flowing a second sample fluid including PCR primer pair sets, dNTPs, enzymes and buffer components effective to permit PCR amplification through a second inlet channel which is in fluid communication with the main channel at a junction, such that the second sample fluid forms a plurality of highly uniform, monodisperse droplets in the continuous phase, wherein the junction is heated to 95° C.
  • step (d) coalescing the droplets including the DNA molecules from step (b) with the droplets including the PCR primer pair sets, dNTPs, enzymes and buffer components from step (c) within an electric field; (e) reacting the contents of the droplets for at least twenty heating and cooling cycles to permit the PCR amplification of the target DNA molecules, such that the amplified target DNA molecules are attached to a semi-solid substrate.
  • Another method of amplifying DNA includes, for example, (a) providing a microfluidic substrate including at least one inlet channel adapted to carry at least one dispersed phase sample fluid and at least one main channel adapted to carry at least one continuous phase fluid, wherein the main channel includes a serpentine line with heating and cooling regions; (b) flowing a sample fluid including one more target DNA molecules to be sequenced, primer pair sets, dNTPs, enzymes and buffer components effective to permit isothermal amplification through an inlet channel which is in fluid communication with the main channel at a junction, such that the sample fluid forms a plurality of highly uniform, monodisperse droplets in the continuous phase, wherein a portion of at least one of the primer pair sets is attached to a semi-solid substrate; (c) reacting the contents of the droplets to permit the isothermal amplification of the target DNA molecules, such that the amplified target DNA molecules are attached to a semi-solid substrate.
  • Another method of amplifying DNA includes, for example, (a) providing a microfluidic substrate including at least two inlet channels adapted to carry at least two dispersed phase sample fluids and at least one main channel adapted to carry at least one continuous phase fluid, wherein the main channel includes a serpentine line with heating and cooling regions; (b) flowing a first sample fluid including one more target DNA molecules to be amplified through a first inlet channel which is in fluid communication with the main channel at a junction, such that the first sample fluid forms a plurality of highly uniform, monodisperse droplets in the continuous phase; (c) flowing a second sample fluid including primer pair sets, dNTPs, enzymes and buffer components effective to permit isothermal amplification through a second inlet channel which is in fluid communication with the main channel at a junction, such that the second sample fluid forms a plurality of highly uniform, monodisperse droplets in the continuous phase, wherein a portion of at least one of the primer pair sets is attached to a semi-solid substrate; (d) coal
  • the present invention also provides various methods for sequencing DNA.
  • One method includes, for example, (a) providing a microfluidic substrate including at least two inlet channels adapted to carry at least two dispersed phase sample fluids and at least one main channel adapted to carry at least one continuous phase fluid, wherein the main channel includes a serpentine line with heating and cooling regions; (b) flowing a first sample fluid including one more target DNA molecules to be sequenced, PCR primer pair sets, dNTPs, enzymes and buffer components effective to permit PCR amplification through a first inlet channel which is in fluid communication with the main channel at a junction, such that the first sample fluid forms a plurality of highly uniform, monodisperse droplets in the continuous phase, wherein the junction is heated to 95° C.
  • step (c) reacting the contents of the droplets for at least twenty heating and cooling cycles to permit the PCR amplification of the target DNA molecules; (d) flowing a second sample fluid including shrimp alkaline phosphatase and exonuclease I through a second inlet channel which is in fluid communication with the main channel at a junction, such that the second sample fluid forms a plurality of highly uniform, monodisperse droplets in the continuous phase; (e) coalescing the droplets including the amplified DNA molecules from step (c) with the droplets including the shrimp alkaline phosphatase and exonuclease I from step (d) within an electric field and reacting the contents of the combined droplets at 37° C.; (f) inactivating the enzymes within the droplets to terminate the reaction from step (e) by heating the reacted droplets to 95° C.; (g) flowing a third sample fluid including universal sequencing primers, labeled ddNTPs and buffer effective to permit nucleotide
  • Another method of sequencing DNA includes, for example, (a) providing a microfluidic substrate including at least two inlet channels adapted to carry at least two dispersed phase sample fluids and at least one main channel adapted to carry at least one continuous phase fluid, wherein the main channel includes a serpentine line with heating and cooling regions; (b) flowing a first sample fluid including one more target DNA molecules to be sequenced, primer pair sets, dNTPs, enzymes and buffer components effective to permit isothermal amplification through a first inlet channel which is in fluid communication with the main channel at a junction, such that the first sample fluid forms a plurality of highly uniform, monodisperse droplets in the continuous phase; (c) reacting the contents of the droplets to permit the isothermal amplification of the target DNA molecules; (d) flowing a second sample fluid including shrimp alkaline phosphatase and exonuclease I through a second inlet channel which is in fluid communication with the main channel at a junction, such that the second sample fluid forms a plurality of
  • Each primer or ddNTP can be labeled with a unique label.
  • a portion of at least one of the primer pair sets can be attached to a semi-solid substrate.
  • the semi-solid substrate can be a bead.
  • the junction between the inlet channel and main channel can include a fluidic nozzle for flow focusing.
  • the present invention also provides a method of detecting a single nucleotide polymorphism (SNP) including, for example, (a) providing a microfluidic substrate including at least one inlet channel adapted to carry at least one dispersed phase sample fluid and at least one main channel adapted to carry at least one continuous phase fluid, wherein the main channel includes a serpentine line with heating and cooling regions; (b) flowing a sample fluid including one more target DNA molecules to be analyzed, labeled PCR primer pair sets, dNTPs, enzymes and buffer components effective to permit PCR amplification through an inlet channel which is in fluid communication with the main channel at a junction, such that the sample fluid forms a plurality of highly uniform, monodisperse droplets in the continuous phase, wherein the junction is heated to 95° C. to provide a hot start; (c) reacting the contents of the droplets for at least twenty heating and cooling cycles to permit the PCR amplification of the target DNA molecules; and (d) analyzing the reaction to determine the presence of absence of
  • the present invention also provides a method of forming enzyme emulsions including, for example, (a) providing a microfluidic substrate including at least one inlet channel adapted to carry at least one dispersed phase fluid and at least one main channel adapted to carry at least one continuous phase fluid; (b) flowing a sample fluid including at least one cell through an inlet channel which is in fluid communication with the main channel at a junction, such that the sample fluid forms a plurality of highly uniform, monodisperse droplets in the continuous phase, wherein the cells includes at least one enzyme which can be secreted by the cells; (c) incubating the cells within the plurality of droplets such that the cells secrete at least one enzyme into the droplet, wherein the cells within the droplets can be incubated within or outside of the microfluidic substrate.
  • the present invention also provides various methods for detecting enzyme activity.
  • One method includes, for example, (a) providing a microfluidic substrate including at least one inlet channel adapted to carry at least one dispersed phase sample fluid and at least one main channel adapted to carry at least one continuous phase fluid; (b) flowing a sample fluid including at least one cell and at least one labeled enzyme substrate through a inlet channel which is in fluid communication with the main channel at a junction, such that the sample fluid forms a plurality of highly uniform, monodisperse droplets in the continuous phase wherein the cells includes at least one enzyme which can be secreted by the cells; (c) collecting the droplets and incubating them on the microfluidic substrate or off the microfluidic substrate at a temperature and duration appropriate to permit the enzyme/substrate reaction to occur; (d) reintroducing the droplets onto the microfluidic substrate if the droplets removed from the microfluidic substrate in step (c); (e) analyzing the contents of the coalesced droplets using the detection
  • Another method of detecting enzyme activity includes, for example, (a) providing a microfluidic substrate including at least two inlet channels adapted to carry at least two dispersed phase sample fluids and at least one main channel adapted to carry at least one continuous phase fluid, wherein the main channel includes a serpentine line with heating and cooling regions; (b) flowing a first sample fluid including at least one cell through a first inlet channel which is in fluid communication with the main channel at a junction, such that the first sample fluid forms a plurality of highly uniform, monodisperse droplets in the continuous phase, wherein the cells includes at least one enzyme which can be secreted by the cells; (c) flowing a second sample fluid including at least one labeled enzyme substrate through a second inlet channel which is in fluid communication with the main channel at a junction, such that the second sample fluid forms a plurality of highly uniform, monodisperse droplets in the continuous phase; (d) coalescing the droplets including the secreted enzymes from step (b) with the droplets including the labeled substrate
  • the method can further include incubating the cells within the plurality of droplets from step (b) such that the cells secrete at least one enzyme into the droplet, prior to flowing the second sample fluid in step (c).
  • the cells can be incubated within the plurality of droplets within the microfluidic substrate or outside the microfluidic substrate.
  • the method can further include lysing the cells within the plurality of droplets from step (b) such that the cells release at least one enzyme into the plurality droplet, prior to flowing the second sample fluid in step (c).
  • the method can further include selecting the coalesced droplets which contain the enzyme with the highest level of activity for the labeled substrate.
  • the channels can be coated with an anti-wetting or blocking agent for the dispersed phase.
  • the channels can be coated with a silica primer layer followed by a perfluoroalkylalkylsilane compound, an amorphous soluble perfluoropolymer, BSA, PEG-silane or fluorosilane.
  • the channels are coated with a silica primer layer followed by a perfluoroalkylalkylsilane compound.
  • the plurality of droplets or nanoreactors can include a biological/chemical material.
  • the biological/chemical material can be tissues, cells, particles, proteins, antibodies, amino acids, nucleotides, small molecules, or pharmaceuticals.
  • the biological/chemical material can include a label.
  • Each biological/chemical material can be labeled with a unique label, such as a protein, a DNA tag, a dye, a quantum dot or a radio frequency identification tag.
  • Each plurality of droplets can be labeled, such as a change in viscosity, a change in opacity, a change in volume, a change in density, a change in pH, a change in temperature, a change in dielectric constant, a change in conductivity, a change in the amount of beads present in the solution, a change in the amount of flocculent in the solution, a change in the amount of a selected solvent within the solution or the change in the amount of any measurable entity within the solution, or combinations thereof.
  • Labels can be detected by any means know in the art, including for example, fluorescence polarization, fluorescence intensity, fluorescence lifetime, fluorescence energy transfer, pH, ionic content, temperature or combinations thereof.
  • the continuous phase can be a non-polar solvent.
  • the continuous phase can be a fluorocarbon oil.
  • the continuous phase can further include one or more additives.
  • the additive is a fluorosurfactant. More preferably, the fluorosurfactant is a perfluorinated polyether.
  • the flow of the dispersed phase fluid and the continuous phase fluid can be pressure driven.
  • the methods of the present invention can include one or more coalescence steps Coalescing droplets or nanoreactors can occur within a coalescence module in fluid communication with the main channel on the microfluidic substrate.
  • the coalescence module can include one or more electrodes that generate an electric field.
  • the channel dimensions in the coalescence module can include an expanded portion of the main channel between the electrodes to bring successive droplets into proximity.
  • the channel dimensions in the coalescence module can include a narrowed portion of the main channel to center droplets within the main channel prior to a expanded portion of the main channel.
  • the droplets or nanoreactors to be coalesced during coalescing steps within the coalescence module have no charge and are coalesced within an electric field.
  • the methods of the present invention can include one or more detection or analysis steps. Interrogating droplet or nanoreactor for at least one predetermined characteristic of the present invention can occur within a detection module in fluid communication with the main channel on the microfluidic substrate.
  • the detection module including a detection apparatus for evaluating the contents or characteristics of the droplet or nanoreactor.
  • the detection apparatus includes an optical or electrical detector.
  • the methods of the present invention can include one or more sorting steps. Sorting a droplet or nanoreactor into or away from a collection module can occur within a sorting module in fluid communication with the main channel on the microfluidic substrate in response to the contents or characterization of the droplet or nanoreactor evaluated in the detection module.
  • the sorting module includes a sorting apparatus adapted to direct the droplet or nanoreactor into or away from the collection module.
  • the sorting apparatus can include one or more electrodes that generate an electric field.
  • the droplets or nanoreactors to be sorted during sorting steps within the sorting module have no charge and are sorted within an electric field.
  • the present invention also provides a methods of detecting or tracking an aqueous solution.
  • a method for detecting an aqueous solution includes, for example, introducing a label into the aqueous solution such that the label alters a physical property of the aqueous solution thereby permitting the detection of aqueous solution including the label.
  • a method of tracking an aqueous solution includes, for example, introducing a label into the aqueous solution such that the label alters a physical property of the aqueous solution thereby permitting the tracking of aqueous solution including the label.
  • the label can be a change in viscosity, a change in opacity, a change in volume, a change in density, a change in pH, a change in temperature, a change in dielectric constant, a change in conductivity, a change in the amount of beads present in the solution, a change in the amount of flocculent in the solution, a change in the amount of a selected solvent within the solution or the change in the amount of any measurable entity within the solution, or combinations thereof.
  • the label can be inducible.
  • the inducible label is detectable when combined with at least one additional solution wherein the solution contains at least one entity to which can alter a property of the label such that the aqueous solution including the label is detectable.
  • the label can alter the light scattering properties of the aqueous solution
  • the labeled aqueous solution can be a labeled aqueous droplet.
  • the labeled aqueous droplet can be detected within a microfluidic substrate.
  • the aqueous solution can further include a biological/chemical material.
  • the biological/chemical material can be tissues, cells, particles, proteins, antibodies, amino acids, nucleotides, small molecules, or pharmaceuticals.
  • the biological/chemical material can include a label. Preferable the label is unique for each biological/chemical material and different from the aqueous solution label.
  • the label can be a protein, a DNA tag, a dye, a quantum dot or a radio frequency identification tag.
  • the label can be detected by any means known in the art including, for example, fluorescence polarization, fluorescence intensity, fluorescence lifetime, fluorescence energy transfer, pH, ionic content, temperature or combinations thereof.
  • FIG. 1 is an schematic illustrating the interacting modules of a microfluidic device of the present invention.
  • FIG. 2 Panels A and B, show dual and single oil versions of the nozzle concept using a small ferrule for the nozzle section. Panels C and D, show the same nozzles made directly out of small bore tubing (the “nozzle” runs the entire length of the tubing).
  • FIG. 3 shows the expansion of the nozzle ferrule concept shown in FIGS. 2A and 2B .
  • FIG. 4 shows the expansion of the nozzle section contained in the ferrule.
  • FIG. 5, 5A shows the operation of the nozzle in Aspiration Mode and 5 B shows the operation of the nozzle in Injection Mode.
  • FIG. 6 shows a reservoir based sample emulsification where the well is initially filled with a fluid with lower density than the sample to be introduced.
  • FIG. 7 illustrates a sample introduction when the sample is less dense than the fluid in the sample port, which is an alternative scheme used to introduce samples that are less dense than the oil used to emulsify the sample.
  • FIG. 8 illustrates a nozzle that formed directly into the fitting used to connect the collection syringe to a syringe tip (e.g. capillary tubing) in order to create a monodisperse emulsion directly from a library well.
  • Step 1 shows the aspiration of the sample can be accomplished by running the collection syringe in withdrawal mode at a flow rate (Q3) above the flow rate of the two oil syringes.
  • Step 2 shows the appropriate volume of sample loaded into the capillary tubing, and the capillary tubing would be removed from the sample well, an air bubble, and possibly a cleaning solution would be aspirated.
  • Step 3 shows when almost all of the sample has been emulsified, the collection syringe withdrawal rate would either be reduced below the oil flow rates, stopped, or set to infuse at some nominal rate.
  • FIG. 9 illustrates a two phase system where the reagent is injected on top of the 2 nd , immiscible phase.
  • A During injection, prior to transition from 1 st phase to 2 nd phase.
  • B 2 nd phase just entering the transfer lines.
  • C 2 nd phase has completely filled the transfer line and pushed the entire volume of reagent through the system.
  • FIG. 10 illustrates sandwiching an ultra-small volume of fluid (i.e., sub-nanoliter) between two solutions having different densities.
  • FIG. 11 illustrates possible interconnect designs for use with PDMS devices.
  • FIG. 12 illustrates self-alignment of fluidic interconnect
  • FIG. 13 illustrates the interconnects needed for each tube molded into a single monolithic self-aligned part.
  • FIG. 14 shows a schematic of a molding tool based on this concept.
  • the pins (orange) are captured within an elestomeric molded sleeve and a compression plate made from a rigid backer plate and foam rubber is used to apply gentle even pressure to the pins and generate the force needed to make the pins uniformly contact the master.
  • FIG. 15 is a schematic diagram of an improved coalescence module that shows an optional small constriction (neckdown) just before this expansion can be used to better align the droplets on their way into the coalescence point.
  • FIG. 16 illustrates that fluorescence polarization (FP) measures the tumbling rate of a compound in solution and is a function of it's volume (in most cases, volume is correlated with MW)
  • FIG. 17 shows the fluorescence polarization of three different compounds. Results of reading polarization in 18,000 drops containing 3 distinct species (FC, BTFC, and BTFC bound to SA). Ideal for reading results of drug screening assays, protein interactions, or DNA hybridization.
  • FIG. 18 A) illustrates encoding a liquid solution using both overall fluorescence polarization and overall dye intensity within droplets; B) shows that multiple colors of fluorescence polarization and FI increases the number of possible labels. Ten intensity levels with ten fluorescence polarization levels on two colors yields 10,000 labels
  • FIG. 19 illustrates FP coding using dyes having different fluorescence lifetimes. These were made one element at a time, stored in a single syringe overnight and then loaded back on chip. The codes were made by using a ratio of two different dyes, one with a short lifetime and hence high FP and one with a long lifetime and correspondingly low FP. The mixtures have intermediate FP signals. The intensity is tuned by controlling the overall concentration of the two dyes.
  • FIG. 20A-20D illustrate the sorting and/or splitting of droplets in accordance with another embodiment of the invention
  • FIG. 21A -F shows the possible flow geometries used in an asymmetric sorting application.
  • FIG. 22 shows the possible electrode geometries used in an asymmetric sorting application.
  • Panel A shows the design using sharp tipped electrodes.
  • Panel B shows broad tipped electrodes to increase the interaction time between the droplets and the electric field (the tips could be many drop diameters long).
  • Panel C shows electrodes straddling the collection line.
  • Panel D shows electrodes on opposite sides of the main channel.
  • Panel E shows an Asymmetric Electrode Pair (the asymmetry may be present on any of the other electrode pair layouts as well).
  • FIG. 23 shows a schematic of a device that split droplets, performs different experiments on the two daughter droplets and then reorders so that they pass sequential through the detector
  • FIG. 24 shows geometric parameters defining the obstacle matrix.
  • Panel B shows three fluid streams.
  • Panel C shows a particle with a radius that is larger than lane 1 follows a streamline passing through the particle's center (black dot).
  • FIG. 25 shows high-resolution separation of fluorescent microspheres with diameters of 0.80 um (green), 0.90 um (red), and 1.03 um (yellow), with a matrix of varying gap size.
  • FIG. 26 is a schematic illustrating the separation by deterministic lateral displacement in an array of microposts, with an example row shift fraction of one-third.
  • FIG. 27 shows a dideoxynucleotide sequencing on a microfabricated chip. Shown is one embodiment for a DNA sequencing chip design. Template DNA and primers are combined at step ‘add 1’ and the reaction is incubated at 95° C. for a hot start (position 1). The reaction then cycles 20-30 times (position 2) before the addition of SAP and ExoI at ‘add 2.’ The reaction is incubated at 37° C. for a predefined time-period and then the SAP and ExoI enzymes are inactivated at 95° C. (position ‘4’). The SAP/ExoI procedure degrades nucleotides and single-stranded DNA (primers) remaining after PCR. The universal sequencing primers, ddNTPs and buffers are added at ‘add 3,’ and the PCR sequencing reaction is allowed to proceed at position ‘5.’ The final reaction product is collected and can be stored off-chip.
  • FIG. 28 Panel A shows a schematic of the TempliPhi amplification process using rolling circle amplification.
  • Panel B illustrates a transcription mediated reaction.
  • Panel C illustrates strand-displacement amplification.
  • Panel D shows a schematic diagram of helicase-dependent amplification.
  • FIG. 29 illustrates emulsion-based sample preparation, sample preparation and DNA sequencing. Random libraries of DNA fragments are generated by shearing an entire genome and isolating single DNA molecules by limiting dilution.
  • FIG. 30 shows one method for isolating antibodies on a microfluidic device.
  • FIG. 31 shows an alternate method for isolating antibodies on a microfluidic device.
  • FIG. 32 shows the method of the present invention for isolating antibodies on the microfluidic device.
  • the right panel is a diagram of individual steps proposed to amplify signal of interacting antibody and antigen.
  • the left panel is a schematic as would be designed for a chip to be used on microfluidic device.
  • FIG. 33 shows the genetic selection for full length antibody clones.
  • a genetic selection can be used to enrich for full-length antibody clones by transforming E. coli and selecting for clones able to grow on medium in which a suitable sugar is the only carbon source.
  • FIG. 34 is a schematic representation of a multi-step chip according to the invention.
  • 1 st stage droplets are sent into C2, the collection port;
  • 2 nd stage 1 st stage emulsion collected into C2 is reinjected back into the chip and merged with the droplets formed in the second stage nozzle.
  • FIG. 35 shows karyotyping using spectral probes that allow all 23 pairs of human chromosomes to be seen at one time, with each pair of chromosomes painted in a difference fluorescent color.
  • FIG. 36 shows the future BioBased Economy with six building blocks based on renewable biomass.
  • FIG. 37 shows the 8 non-renewable building blocks based on petroleum.
  • the microfluidic devices and methods of use described herein are based on the creation and electrical manipulation of aqueous phase droplets completely encapsulated by an inert immiscible oil stream. This combination enables precise droplet generation, highly efficient, electrically addressable, droplet coalescence, and controllable, electrically addressable single droplet sorting.
  • the microfluidic devices include one or more channels and modules. A schematic illustrating one example of interacting modules of a microfluidic substrate is shown in FIG. 1 . The integration of these modules is an essential enabling technology for a droplet based, high-throughput microfluidic reactor system.
  • microfluidic devices of the present invention can be utilized for numerous biological, chemical, or diagnostic applications, as described in further detail herein.
  • the microfluidic device of the present invention includes one or more analysis units.
  • An “analysis unit” is a microsubstrate, e.g., a microchip.
  • the terms microsubstrate, substrate, microchip, and chip are used interchangeably herein.
  • the analysis unit includes at least one inlet channel, at least one main channel, at least one inlet module, at least one coalescence module, and at least one detection module.
  • the analysis unit can further includes one or more sorting modules.
  • the sorting module can be in fluid communication with branch channels which are in fluid communication with one or more outlet modules (collection module or waste module). For sorting applications, at least one detection module cooperates with at least one sorting module to divert flow via a detector-originated signal.
  • modules and channels are in fluid communication with each other and therefore may overlap; i.e., there may be no clear boundary where a module or channel begins or ends.
  • a plurality of analysis units of the invention may be combined in one device. The analysis unit and specific modules are described in further detail herein.
  • the dimensions of the substrate are those of typical microchips, ranging between about 0.5 cm to about 15 cm per side and about I micron to about I cm in thickness.
  • a substrate can be transparent and can be covered with a material having transparent properties, such as a glass coverslip, to permit detection of a reporter, for example, by an optical device such as an optical microscope.
  • the material can be perforated for functional interconnects, such as fluidic, electrical, and/or optical interconnects, and sealed to the back interface of the device so that the junction of the interconnects to the device is leak-proof.
  • Such a device can allow for application of high pressure to fluid channels without leaking.
  • various components of the invention can be formed from solid materials, in which the channels can be formed via molding, micromachining, film deposition processes such as spin coating and chemical vapor deposition, laser fabrication, photolithographic techniques, etching methods including wet chemical or plasma processes, and the like. See, for example, Scientific American, 248:44-55, 1983 (Angell, et al). At least a portion of the fluidic system can be formed of silicone by molding a silicone chip. Technologies for precise and efficient formation of various fluidic systems and devices of the invention from silicone are known.
  • Various components of the systems and devices of the invention can also be formed of a polymer, for example, an elastomeric polymer such as polydimethylsiloxane (“PDMS”), polytetrafluoroethylene (“PTFE”) or Teflon®, or the like.
  • PDMS polydimethylsiloxane
  • PTFE polytetrafluoroethylene
  • Teflon® Teflon®
  • the channels of the invention can be formed, for example by etching a silicon chip using conventional photolithography techniques, or using a micromachining technology called “soft lithography” as described by Whitesides and Xia, Angewandte Chemie International Edition 37, 550 (1998). These and other methods may be used to provide inexpensive miniaturized devices, and in the case of soft lithography, can provide robust devices having beneficial properties such as improved flexibility, stability, and mechanical strength. When optical detection is employed, the invention also provides minimal light scatter from molecule, cell, small molecule or particle suspension and chamber material.
  • a base portion including a bottom wall and side walls can be formed from an opaque material such as silicone or PDMS, and a top portion can be formed from a transparent or at least partially transparent material, such as glass or a transparent polymer, for observation and/or control of the fluidic process.
  • Components can be coated so as to expose a desired chemical functionality to fluids that contact interior channel walls, where the base supporting material does not have a precise, desired functionality.
  • components can be formed as illustrated, with interior channel walls coated with another material.
  • Material used to form various components of the systems and devices of the invention may desirably be selected from among those materials that will not adversely affect or be affected by fluid flowing through the fluidic system, e.g., material(s) that is chemically inert in the presence of fluids to be used within the device.
  • Various components of the invention when formed from polymeric and/or flexible and/or elastomeric materials, and can be conveniently formed of a hardenable fluid, facilitating formation via molding (e.g. replica molding, injection molding, cast molding, etc.).
  • the hardenable fluid can be essentially any fluid that can be induced to solidify, or that spontaneously solidifies, into a solid capable of containing and/or transporting fluids contemplated for use in and with the fluidic network.
  • the hardenable fluid comprises a polymeric liquid or a liquid polymeric precursor (i.e. a “prepolymer”).
  • Suitable polymeric liquids can include, for example, thermoplastic polymers, thermoset polymers, or mixture of such polymers heated above their melting point.
  • a suitable polymeric liquid may include a solution of one or more polymers in a suitable solvent, which solution forms a solid polymeric material upon removal of the solvent, for example, by evaporation.
  • a suitable solvent such polymeric materials, which can be solidified from, for example, a melt state or by solvent evaporation, are well known to those of ordinary skill in the art.
  • a variety of polymeric materials, many of which are elastomeric, are suitable, and are also suitable for forming molds or mold masters, for embodiments where one or both of the mold masters is composed of an elastomeric material.
  • a non-limiting list of examples of such polymers includes polymers of the general classes of silicone polymers, epoxy polymers, and acrylate polymers.
  • Epoxy polymers are characterized by the presence of a three-membered cyclic ether group commonly referred to as an epoxy group, 1,2-epoxide, or oxirane.
  • diglycidyl ethers of bisphenol A can be used, in addition to compounds based on aromatic amine, triazine, and cycloaliphatic backbones.
  • Another example includes the well-known Novolac polymers.
  • Non-limiting examples of silicone elastomers suitable for use according to the invention include those formed from precursors including the chlorosilanes such as methylchlorosilanes, ethylchlorosilanes, phenylchlorosilanes, etc.
  • Silicone polymers are preferred, for example, the silicone elastomer polydimethylsiloxane.
  • Non-limiting examples of PDMS polymers include those sold under the trademark Sylgard by Dow Chemical Co., Midland, Mich., and particularly Sylgard 182, Sylgard 184, and Sylgard 186.
  • Silicone polymers including PDMS have several beneficial properties simplifying formation of the microfluidic structures of the invention. For instance, such materials are inexpensive, readily available, and can be solidified from a prepolymeric liquid via curing with heat.
  • PDMSs are typically curable by exposure of the prepolymeric liquid to temperatures of about, for example, about 65° C. to about 75° C. for exposure times of, for example, about an hour.
  • silicone polymers such as PDMS
  • PDMS polymethyl methacrylate copolymer
  • flexible (e.g., elastomeric) molds or masters can be advantageous in this regard.
  • the present invention provides improved methods of bonding PDMS to incompatible media.
  • Normal methods of bonding various materials (plastic, metals, etc) directly to materials such as PDMS, silicone, Teflon, and PEEK using traditional bonding practices (adhesives, epoxies, etc) do not work well due to the poor adhesion of the bonding agent to materials such as PDMS.
  • Normal surface preparation by commercially available surface activators has not worked well in microfluidic device manufacturing. This problem is eliminated by treating the PDMS surface to be bonded with high intensity oxygen or air plasma. The process converts the top layer of PDMS to glass which bonds extremely well with normal adhesives.
  • Tests using this method to bond external fluid lines to PDMS using a UV-cure adhesive resulted in a bond that is stronger than the PDMS substrate, resulting in fracture of the PDMS prior to failure of the bond.
  • the present method combines high radiant flux, wavelength selection, and cure exposure time to significantly enhance the bond strength of the adhesive.
  • One advantage of forming structures such as microfluidic structures of the invention from silicone polymers, such as PDMS, is the ability of such polymers to be oxidized, for example by exposure to an oxygen-containing plasma such as an air plasma, so that the oxidized structures contain, at their surface, chemical groups capable of cross-linking to other oxidized silicone polymer surfaces or to the oxidized surfaces of a variety of other polymeric and non-polymeric materials.
  • an oxygen-containing plasma such as an air plasma
  • oxidized silicone such as oxidized PDMS can also be sealed irreversibly to a range of oxidized materials other than itself including, for example, glass, silicon, silicon oxide, quartz, silicon nitride, polyethylene, polystyrene, glassy carbon, and epoxy polymers, which have been oxidized in a similar fashion to the PDMS surface (for example, via exposure to an oxygen-containing plasma).
  • Oxidation and sealing methods useful in the context of the present invention, as well as overall molding techniques, are described in the art, for example, in an article entitled “Rapid Prototyping of Microfluidic Systems and Polydimethylsiloxane,” Anal. Chem., 70:474-480, 1998 (Duffy et al.), incorporated herein by reference.
  • microfluidic structures of the invention or interior, fluid-contacting surfaces
  • these surfaces can be much more hydrophilic than the surfaces of typical elastomeric polymers (where a hydrophilic interior surface is desired).
  • Such hydrophilic channel surfaces can thus be more easily filled and wetted with aqueous solutions than can structures comprised of typical, unoxidized elastomeric polymers or other hydrophobic materials.
  • a bottom wall is formed of a material different from one or more side walls or a top wall, or other components.
  • the interior surface of a bottom wall can comprise the surface of a silicon wafer or microchip, or other substrate.
  • Other components can, as described above, be sealed to such alternative substrates.
  • a component comprising a silicone polymer e.g. PDMS
  • the substrate may be selected from the group of materials to which oxidized silicone polymer is able to irreversibly seal (e.g., glass, silicon, silicon oxide, quartz, silicon nitride, polyethylene, polystyrene, epoxy polymers, and glassy carbon surfaces which have been oxidized).
  • other sealing techniques can be used, as would be apparent to those of ordinary skill in the art, including, but not limited to, the use of separate adhesives, thermal bonding, solvent bonding, ultrasonic welding, etc.
  • the microfluidic substrates of the present invention include channels that form the boundary for a fluid.
  • a “channel,” as used herein, means a feature on or in a substrate that at least partially directs the flow of a fluid.
  • the channel may be formed, at least in part, by a single component, e.g., an etched substrate or molded unit.
  • the channel can have any cross-sectional shape, for example, circular, oval, triangular, irregular, square or rectangular (having any aspect ratio), or the like, and can be covered or uncovered (i.e., open to the external environment surrounding the channel).
  • at least one portion of the channel can have a cross-section that is completely enclosed, and/or the entire channel may be completely enclosed along its entire length with the exception of its inlet and outlet.
  • An open channel generally will include characteristics that facilitate control over fluid transport, e.g., structural characteristics (an elongated indentation) and/or physical or chemical characteristics (hydrophobicity vs. hydrophilicity) and/or other characteristics that can exert a force (e.g., a containing force) on a fluid.
  • the fluid within the channel may partially or completely fill the channel.
  • the fluid may be held or confined within the channel or a portion of the channel in some fashion, for example, using surface tension (e.g., such that the fluid is held within the channel within a meniscus, such as a concave or convex meniscus).
  • some (or all) of the channels may be of a particular size or less, for example, having a largest dimension perpendicular to fluid flow of less than about 5 mm, less than about 2 mm, less than about 1 mm, less than about 500 microns, less than about 200 microns, less than about 100 microns, less than about 60 microns, less than about 50 microns, less than about 40 microns, less than about 30 microns, less than about 25 microns, less than about 10 microns, less than about 3 microns, less than about 1 micron, less than about 300 nm, less than about 100 nm, less than about 30 nm, or less than about 10 nm or less in some cases.
  • larger channels, tubes, etc. can be used to store fluids in bulk and/or deliver a fluid to the channel.
  • the channel is a capillary.
  • the dimensions of the channel may be chosen such that fluid is able to freely flow through the channel, for example, if the fluid contains cells.
  • the dimensions of the channel may also be chosen, for example, to allow a certain volumetric or linear flow rate of fluid in the channel.
  • the number of channels and the shape of the channels can be varied by any method known to those of ordinary skill in the art. In some cases, more than one channel or capillary may be used. For example, two or more channels may be used, where they are positioned inside each other, positioned adjacent to each other, etc.
  • the channels of the device are preferably square, with a diameter between about 2 microns and 1 mm. This geometry facilitates an orderly flow of droplets in the channels.
  • the volume of the detection module in an analysis device is typically in the range of between about 0.1 picoliters and 500 nanoliters.
  • a “main channel” is a channel of the device of the invention which permits the flow of molecules, cells, small molecules or particles past a coalescence module for coalescing one or more droplets, a detection module for detection (identification) or measurement of a droplet and a sorting module, if present, for sorting a droplet based on the detection in the detection module.
  • the main channel is typically in fluid communication with the coalescence, detection and/or sorting modules, as well as, an inlet channel of the inlet module.
  • the main channel is also typically in fluid communication with an outlet module and optionally with branch channels, each of which may have a collection module or waste module. These channels permit the flow of molecules, cells, small molecules or particles out of the main channel.
  • An “inlet channel” permits the flow of molecules, cells, small molecules or particles into the main channel.
  • One or more inlet channels communicate with one or more means for introducing a sample into the device of the present invention.
  • the inlet channel communicates with the main channel at an inlet module.
  • the microfluidic substrate can also comprise one or more fluid channels to inject or remove fluid in between droplets in a droplet stream for the purpose of changing the spacing between droplets.
  • the channels of the device of the present invention can be of any geometry as described.
  • the channels of the device can comprise a specific geometry such that the contents of the channel are manipulated, e.g., sorted, mixed, prevent clogging, etc.
  • a microfluidic substrate can also include a specific geometry designed in such a manner as to prevent the aggregation of biological/chemical material and keep the biological/chemical material separated from each other prior to encapsulation in droplets.
  • the geometry of channel dimension can be changed to disturb the aggregates and break them apart by various methods, that can include, but is not limited to, geometric pinching (to force cells through a (or a series of) narrow region(s), whose dimension is smaller or comparable to the dimension of a single cell) or a barricade (place a series of barricades on the way of the moving cells to disturb the movement and break up the aggregates of cells).
  • the channels may have a coating which minimizes adhesion.
  • a coating may be intrinsic to the material from which the device is manufactured, or it may be applied after the structural aspects of the channels have been microfabricated.
  • “TEFLON” is an example of a coating that has suitable surface properties.
  • the surface of the channels of the microfluidic device can be coated with any anti-wetting or blocking agent for the dispersed phase.
  • the channel can be coated with any protein to prevent adhesion of the biological/chemical sample.
  • the channels are coated with BSA, PEG-silane and/or fluorosilane.
  • the channels can be coated with a cyclized transparent optical polymer obtained by copolymerization of perfluoro (alkenyl vinyl ethers), such as the type sold by Asahi Glass Co. under the trademark Cytop.
  • the coating is applied from a 0.1-0.5 wt % solution of Cytop CTL-809M in CT-Solv 180. This solution can be injected into the channels of a microfluidic device via a plastic syringe. The device can then be heated to about 90° C. for 2 hours, followed by heating at 200° C. for an additional 2 hours.
  • the channels can be coated with a hydrophobic coating of the type sold by PPG Industries, Inc. under the trademark Aquapel (e.g., perfluoroalkylalkylsilane surface treatment of plastic and coated plastic substrate surfaces in conjunction with the use of a silica primer layer) and disclosed in U.S. Pat. No. 5,523,162, which patent is hereby incorporated by reference.
  • Aquapel e.g., perfluoroalkylalkylsilane surface treatment of plastic and coated plastic substrate surfaces in conjunction with the use of a silica primer layer
  • U.S. Pat. No. 5,523,162 which patent is hereby incorporated by reference.
  • the surface of the channels in the microfluidic device can be also fluorinated to prevent undesired wetting behaviors.
  • a microfluidic device can be placed in a polycarbonate dessicator with an open bottle of (tridecafluoro-1,1,2,2-tetrahydrooctyl)trichlorosilane. The dessicator is evacuated for 5 minutes, and then sealed for 20-40 minutes. The dessicator is then backfilled with air and removed.
  • This approach uses a simple diffusion mechanism to enable facile infiltration of channels of the microfluidic device with the fluorosilane and can be readily scaled up for simultaneous device fluorination.
  • the microfluidic device of the present invention is capable of controlling the direction and flow of fluids and entities within the device.
  • flow means any movement of liquid or solid through a device or in a method of the invention, and encompasses without limitation any fluid stream, and any material moving with, within or against the stream, whether or not the material is carried by the stream.
  • the movement of molecules, beads, cells or virions through a device or in a method of the invention, e.g. through channels of a microfluidic chip of the invention comprises a flow.
  • any force may be used to provide a flow, including without limitation, pressure, capillary action, electro-osmosis, electrophoresis, dielectrophoresis, optical tweezers, and combinations thereof, without regard for any particular theory or mechanism of action, so long as molecules, cells or virions are directed for detection, measurement or sorting according to the invention. Specific flow forces are described in further detail herein.
  • the flow stream in the main channel is typically, but not necessarily, continuous and may be stopped and started, reversed or changed in speed.
  • a liquid that does not contain sample molecules, cells or particles can be introduced into a sample inlet well or channel and directed through the inlet module, e.g., by capillary action, to hydrate and prepare the device for use.
  • buffer or oil can also be introduced into a main inlet region that communicates directly with the main channel to purge the device (e.g., or “dead” air) and prepare it for use.
  • the pressure can be adjusted or equalized, for example, by adding buffer or oil to an outlet module.
  • fluid stream or “fluidic stream” refers to the flow of a fluid, typically generally in a specific direction.
  • the fluidic stream may be continuous and/or discontinuous.
  • a “continuous” fluidic stream is a fluidic stream that is produced as a single entity, e. g., if a continuous fluidic stream is produced from a channel, the fluidic stream, after production, appears to be contiguous with the channel outlet.
  • the continuous fluidic stream is also referred to as a continuous phase fluid or carrier fluid.
  • the continuous fluidic stream may be laminar, or turbulent in some cases.
  • a “discontinuous” fluidic stream is a fluidic stream that is not produced as a single entity.
  • the discontinuous fluidic stream is also referred to as the dispersed phase fluid or sample fluid.
  • a discontinuous fluidic stream may have the appearance of individual droplets, optionally surrounded by a second fluid.
  • a “droplet,” as used herein, is an isolated portion of a first fluid that completely surrounded by a second fluid.
  • the droplets may be spherical or substantially spherical; however, in other cases, the droplets may be non-spherical, for example, the droplets may have the appearance of “blobs” or other irregular shapes, for instance, depending on the external environment.
  • the dispersed phase fluid can include a biological/chemical material.
  • the biological/chemical material can be tissues, cells, particles, proteins, antibodies, amino acids, nucleotides, small molecules, and pharmaceuticals.
  • the biological/chemical material can include one or more labels known in the art.
  • the label can be a DNA tag, dyes or quantum dot, or combinations thereof.
  • emulsion refers to a preparation of one liquid distributed in small globules (also referred to herein as drops, droplets or NanoReactors) in the body of a second liquid.
  • the first and second fluids are immiscible with each other.
  • the discontinuous phase can be an aqueous solution and the continuous phase can a hydrophobic fluid such as an oil. This is termed a water in oil emulsion.
  • the emulsion may be a oil in water emulsion.
  • the first liquid, which is dispersed in globules is referred to as the discontinuous phase
  • the second liquid is referred to as the continuous phase or the dispersion medium.
  • the continuous phase can be an aqueous solution and the discontinuous phase is a hydrophobic fluid, such as an oil (e.g., decane, tetradecane, or hexadecane).
  • a hydrophobic fluid such as an oil (e.g., decane, tetradecane, or hexadecane).
  • the droplets or globules of oil in an oil in water emulsion are also referred to herein as “micelles”, whereas globules of water in a water in oil emulsion may be referred to as “reverse micelles”.
  • the fluidic droplets may each be substantially the same shape and/or size.
  • the shape and/or size can be determined, for example, by measuring the average diameter or other characteristic dimension of the droplets.
  • the “average diameter” of a plurality or series of droplets is the arithmetic average of the average diameters of each of the droplets. Those of ordinary skill in the art will be able to determine the average diameter (or other characteristic dimension) of a plurality or series of droplets, for example, using laser light scattering, microscopic examination, or other known techniques.
  • the diameter of a droplet, in a non-spherical droplet is the mathematically-defined average diameter of the droplet, integrated across the entire surface.
  • the average diameter of a droplet may be, for example, less than about 1 mm, less than about 500 micrometers, less than about 200 micrometers, less than about 100 micrometers, less than about 75 micrometers, less than about 50 micrometers, less than about 25 micrometers, less than about 10 micrometers, or less than about 5 micrometers in some cases.
  • the average diameter may also be at least about 1 micrometer, at least about 2 micrometers, at least about 3 micrometers, at least about 5 micrometers, at least about 10 micrometers, at least about 15 micrometers, or at least about 20 micrometers in certain cases.
  • NanoReactor and its plural encompass the terms “droplet”, “nanodrop”, “nanodroplet”, “microdrop” or “microdroplet” as defined herein, as well as an integrated system for the manipulation and probing of droplets, as described in detail herein.
  • Nanoreactors as described herein can be 0.1-1000 ⁇ m (e.g., 0.1, 0.2 . . . 5, 10, 15, 20, 25, 30, 35, 40, 45, 50 . . . 1000), or any size within in this range. Droplets at these dimensions tend to conform to the size and shape of the channels, while maintaining their respective volumes. Thus, as droplets move from a wider channel to a narrower channel they become longer and thinner, and vice versa.
  • the microfluidic substrate of this invention most preferably generate round, monodisperse droplets.
  • the droplets can have a diameter that is smaller than the diameter of the microchannel; i.e., preferably 15 to 100 ⁇ m when cells are used; or 10 to 75 ⁇ m when reagents or other chemical or biological agents are used; or 100 to 1000 ⁇ m when droplets are used for sequencing reactions such that droplets will be removed and dispensed into other collection apparatuses, such as microtiter plates or utilized in sequencing devices.
  • Monodisperse droplets are particularly preferably, e.g., in high throughput devices and other embodiments where it is desirable to generate droplets at high frequency and of high uniformity.
  • the droplet forming liquid is typically an aqueous buffer solution, such as ultrapure water (e.g., 18 mega-ohm resistivity, obtained, for example by column chromatography), 10 mM Tris HCl and 1 mM EDTA (TE) buffer, phosphate buffer saline (PBS) or acetate buffer. Any liquid or buffer that is physiologically compatible, with the population of molecules, cells or particles to be analyzed and/or sorted can be used.
  • the fluid passing through the main channel and in which the droplets are formed is one that is immiscible with the droplet forming fluid.
  • the fluid passing through the main channel can be a non-polar solvent, decane (e g., tetradecane or hexadecane), fluorocarbon oil, silicone oil or another oil (for example, mineral oil).
  • the dispersed phase fluid may also contain biological/chemical material (e.g., molecules, cells, or other particles) for combination, analysis and/or sorting in the device.
  • the droplets of the dispersed phase fluid can contain more than one particle or can contain no more than one particle.
  • each droplet preferably contains, on average, no more than one cell.
  • each droplet may contain, on average, at least 1000 cells.
  • the droplets can be detected and/or sorted according to their contents.
  • the concentration (i.e., number) of molecules, cells or particles in a droplet can influence sorting efficiently and therefore is preferably optimized.
  • the sample concentration should be dilute enough that most of the droplets contain no more than a single molecule, cell or particle, with only a small statistical chance that a droplet will contain two or more molecules, cells or particles. This is to ensure that for the large majority of measurements, the level of reporter measured in each droplet as it passes through the detection module corresponds to a single molecule, cell or particle and not to two or more molecules, cells or particles.
  • the parameters which govern this relationship are the volume of the droplets and the concentration of molecules, cells or particles in the sample solution.
  • [cell]” is the concentration of molecules, cells or particles in units of number of molecules, cells or particles per cubic micron ( ⁇ m 3 ), and V is the volume of the droplet in units of ⁇ m 3 .
  • P ⁇ 2 can be minimized by decreasing the concentration of molecules, cells or particles in the sample solution.
  • decreasing the concentration of molecules, cells or particles in the sample solution also results in an increased volume of solution processed through the device and can result in longer run times. Accordingly, it is desirable to minimize to presence of multiple molecules, cells or particles in the droplets (thereby increasing the accuracy of the sorting) and to reduce the volume of sample, thereby permitting a sorted sample in a reasonable time in a reasonable volume containing an acceptable concentration of molecules, cells or particles.
  • the maximum tolerable P ⁇ 2 depends on the desired “purity” of the sorted sample.
  • the “purity” in this case refers to the fraction of sorted molecules, cells or particles that posses a desired characteristic (e.g., display a particular antigen, are in a specified size range or are a particular type of molecule, cell or particle).
  • the purity of the sorted sample is inversely proportional to P ⁇ 2 .
  • P ⁇ 2 a relatively high
  • the fluids used to generate droplets in microfluidic devices are typically immiscible liquids such as oil and water. These two materials generally have very different dielectric constants associated with them. These differences can be exploited to determine droplet rate and size for every drop passing through a small section of a microfluidic device.
  • ⁇ 0 is the permittivity of free space
  • k is the effective dielectric constant (this changes every time a droplet passes through)
  • A is the area of the capacitor and d is the electrode separation. The current measured in the device is then plotted as a function of time.
  • the fluidic droplets may contain additional entities, for example, other chemical, biochemical, or biological entities (e.g., dissolved or suspended in the fluid), cells, particles, gases, molecules, or the like.
  • the droplets may each be substantially the same shape or size, as discussed above.
  • the invention provides for the production of droplets consisting essentially of a substantially uniform number of entities of a species therein (i.e., molecules, cells, particles, etc.). For example, about 90%, about 93%, about 95%, about 97%, about 98%, or about 99%, or more of a plurality or series of droplets may each contain the same number of entities of a particular species.
  • a substantial number of fluidic droplets produced may each contain 1 entity, 2 entities, 3 entities, 4 entities, 5 entities, 7 entities, 10 entities, 15 entities, 20 entities, 25 entities, 30 entities, 40 entities, 50 entities, 60 entities, 70 entities, 80 entities, 90 entities, 100 entities, etc., where the entities are molecules or macromolecules, cells, particles, etc.
  • the droplets may each independently contain a range of entities, for example, less than 20 entities, less than 15 entities, less than 10 entities, less than 7 entities, less than 5 entities, or less than 3 entities in some cases.
  • a droplet may contain 100,000,000 entities. In other embodiments, a droplet may contain 1,000,000 entities.
  • the droplets of fluid may be screened or sorted for those droplets of fluid containing the species as further described below (e.g., using fluorescence or other techniques such as those described above), and in some cases, the droplets may be screened or sorted for those droplets of fluid containing a particular number or range of entities of the species of interest, e.g., as previously described.
  • a plurality or series of fluidic droplets may be enriched (or depleted) in the ratio of droplets that do contain the species, for example, by a factor of at least about 2, at least about 3, at least about 5, at least about 10, at least about 15, at least about 20, at least about 50, at least about 100, at least about 125, at least about 150, at least about 200, at least about 250, at least about 500, at least about 750, at least about 1000, at least about 2000, or at least about 5000 or more in some cases.
  • the enrichment may be in a ratio of at least about 10 4 , at least about 10 5 , at least about 10 6 , at least about 10 7 , at least about 10 8 , at least about 10 9 , at least about 10 10 , at least about 10 11 , at least about 10 12 , at least about 10 13 , at least about 10 14 , at least about 10 15 , or more.
  • a fluidic droplet containing a particular species may be selected from a library of fluidic droplets containing various species, where the library may have about 100, about 10 3 , about 10 4 , about 10 5 , about 10 6 , about 10 7 , about 10 8 , about 10 9 , about 10 10 , about 10 11 , about 10 12 , about 10 13 , about 10 14 , about 10 15 , or more items, for example, a DNA library, an RNA library, a protein library, a combinatorial chemistry library, etc.
  • the droplets carrying the species may then be fused, reacted, or otherwise used or processed, etc., as further described below, for example, to initiate or determine a reaction.
  • Droplets of a sample fluid can be formed within the inlet module on the microfluidic device or droplets (or droplet libraries) can be formed before the sample fluid is introduced to the microfluidic device (“off chip” droplet formation).
  • the droplets comprising each sample to be analyzed must be monodisperse.
  • Droplet size must be highly controlled to ensure that droplets containing the correct contents for analysis and coalesced properly.
  • the present invention provides devices and methods for forming droplets and droplet libraries.
  • the present invention provides compositions and methods for forming sample droplet emulsions on a microfluidic substrate.
  • the present invention also provides embedded microfluidic nozzles.
  • this invention would form a nozzle directly into the fitting used to connect the storage well/reservoir (e.g. syringe) to a syringe tip (e.g. capillary tubing), as shown in FIGS. 2-6 .
  • FIG. 2 Panels A and B, show dual and single oil versions of the nozzle concept using a small ferrule for the nozzle section.
  • FIG. 3 shows the expansion of the nozzle ferrule concept shown in FIGS. 2A and 2B .
  • the tube based nozzles ( FIG. 2C, 2D ) function identically to this, except the “nozzle” runs the entire length of the tube instead of having a short transition.
  • the ability to form droplets is identical in both cases.
  • FIG. 4 shows the expansion of the nozzle section contained in the ferrule. The tee design in FIG.
  • FIG. 5A shows the operation of the nozzle in Aspiration Mode
  • FIG. 5B shows the operation of the nozzle in Injection Mode.
  • the droplets formed are approximately 45 um in diameter, and were formed from PCR mix (210 ul/hr) and SpectraSyn-10 (600 ul/hr). Other tests have been demonstrated with Spectrasyn-2 and PCR mix.
  • the droplets are traveling in 300 urn wide ⁇ 260 urn deep channels.
  • the Nozzle tube used was 100 um in diameter, and the fluids used were PCR Mix and Spectrasyn-10 with surfactant.
  • the nozzle can be made from one or two oil lines providing constant flow of oil into the nozzle, a connection to the capillary tubing, and a connection to the storage well/reservoir (e.g. syringe).
  • the high resolution part of the nozzle can be made out of a small bore tubing or a small, simple part molded or stamped from an appropriate material (Teflon®, plastic, metal, etc). If necessary, the nozzle itself could be formed into the tip of the ferrule using post mold processing such as laser ablation or drilling.
  • This nozzle design eliminates the surface wetting issues surrounding the quasi-2D flow associated with typical microfluidic nozzles made using soft lithography or other standard microfluidic chip manufacturing techniques. This is because the nozzle design is fully 3-dimensional, resulting is a complete isolation of the nozzle section from the continuous aqueous phase. This same design can also be used for generation of emulsions required for immediate use, where the aqueous line would be attached directly to a syringe and the outlet of the nozzle would be used to transport the emulsion to the point of use (e.g. into a microfluidic PCR chip, delay line, etc).
  • the present invention provides compositions and methods to directly emulsify library elements from standard library storage geometries (e.g. 96 well plates, etc).
  • this invention would include microfluidic based nozzles manufactured simultaneously with an appropriately designed fluidic interconnect or well.
  • FIGS. 6 and 7 present two possible approaches to interface with the nozzle.
  • FIG. 6 shows a reservoir based sample emulsification.
  • the well is initially filled with a fluid with lower density than the sample to be introduced.
  • the operation of this device would be very similar to the device described above, with the exception that the sample would be introduced into a port instead of being directly aspirated from a sample well. This could either be emulsification oil obtained directly from the nozzle, or a different material that is loaded or flowing into the well automatically.
  • the oil lines would begin flowing at their prescribed rates ( FIG. 6 e ), while the collection or waste port would begin withdrawing at a rate corresponding to the total oil flow plus the desired sample flow.
  • the sample would be introduced into the port either manually (e.g.
  • the sample volume permitted would be dependent on the port volume. Since the sample is more dense than the fluid in the well, it would settle into the bottom of the well and be transported to the nozzle ( FIGS. 6 a - 6 d ). During this time, either the waste (used during startup only if transients cause problems) or the collection port would be withdrawing emulsified sample and stored. When the sample is completely emulsified the next sample would be introduced and the process repeated. If washing steps are required between runs, the washing fluids would be withdrawn into the waste line. If the pressure drop across the nozzle would cause cavitation on collection then an optional pressurization of the input well can be utilized.
  • FIG. 7 shows sample introduction when the sample is less dense than the fluid in the sample port.
  • FIG. 7 depicts an alternative scheme that could be used to introduce samples that are less dense than the oil used to emulsify the sample.
  • the process of introducing the sample into the port could be run either manually or with a robotic sampling system.
  • the sample port could be filled with the emulsification oil through backflow from the nozzle prior to introduction of the sample ( FIG. 7 a ). If this oil is not appropriate, the port can be filled from the top with a different immiscible fluid that might have more desirable properties than the emulsification oil (e.g. better wetting, less surfactant, etc).
  • This second immiscible fluid could be introduced during startup and flow continuously into the port when the sample tip is not inserted. Keeping the sample port filled with fluid will prevent air entrainment during startup and should improve transient performance.
  • the sample tip is connected to a pump capable of driving the sample into the device, it could be started up as the tip is inserted into the device ( 7 b - c ).
  • the device could be run identically to the “normal” operation of our devices, including having the “transport to waste” line ( 7 e ) not connected to a pump.
  • the sample tip loading pump is not capable of accurately forcing the flow (i.e. not connected to a suitable pump)
  • the back end of the tip could be connected to a valve that would open to either atmospheric pressure (or possibly a pressurized gas supply) when the tip is fully inserted into the port.
  • FIG. 7 also shows another possible configuration of the aspiration probe assembly used for the device in FIG. 6 .
  • the present invention also provides compositions and methods for creating emulsion of the sample fluid (e.g. droplets) prior to the introduction of the sample fluid into the microfluidic devices of the present invention. More specifically, the methods are directed to the creating sample droplet emulsions “off chip”, for example in a syringe.
  • a nozzle is formed directly into the fitting used to connect the collection syringe to a syringe tip (e.g. capillary tubing), as shown in FIG. 8 .
  • the nozzle can be made from one or two oil lines providing constant flow of oil into the nozzle, a connection to the capillary tubing, and a connection to the collection syringe.
  • Aspiration of the sample can be accomplished by running the collection syringe in withdrawal mode at a flow rate (Q3) above the flow rate of the two oil syringes (Step 1 in FIG. 8 ). The difference in flow would correspond to the flow rate aspirated from the sample well.
  • the capillary tubing When the appropriate volume of sample has been loaded into the capillary tubing, the capillary tubing would be removed from the sample well, an air bubble, and possibly a cleaning solution would be aspirated (Step 2 in FIG. 8 ).
  • the collection syringe withdrawal rate would either be reduced below the oil flow rates, stopped, or set to infuse at some nominal rate (Step 3 in FIG. 8 ).
  • the remaining sample, air, cleaning solution, etc, left in the capillary would be flushed back out into a cleaning well and the outside of the capillary would be cleaned at the “wash station.”
  • the process would repeat for the next library element.
  • the nozzle can be formed through using small bore tubing (glass, Teflon®, PEEK tubing or capillaries) or micro-fabrication or molding processes such as PDMS soft lithography, glass etching, hot embossing, or similar high resolution fabrication technology.
  • small bore tubing glass, Teflon®, PEEK tubing or capillaries
  • micro-fabrication or molding processes such as PDMS soft lithography, glass etching, hot embossing, or similar high resolution fabrication technology.
  • the present apparatus can be readily adapted for clinical applications or work where cross contamination must be eliminated, since the region from the nozzle to the syringe are isolated from the sample stream (e.g., the oil wets these surfaces and keeps the sample from directly contacting aqueous sample).
  • the aspiration tip can be designed as a disposable item (like a robotic sampler aspiration tips) and automatically replaced between samples.
  • Multiple nozzle/syringe pairs can be operated in parallel, thus increasing throughput. This allows simultaneous sampling of multiple wells/samples during a single process step. Each sample can be collected into a separate syringe.
  • sample droplet emulsions and emulsion libraries “off chip” are described in Example 1.
  • the present invention provides compositions and methods which eliminates the problems associated with dead volume and reagent waste when working with extremely small volumes of reagents.
  • the primary reagents is combined with a second, immiscible phase in the storage container (e.g. a syringe or other reservoir).
  • a second, immiscible phase in the storage container (e.g. a syringe or other reservoir).
  • This second phase is used to push the entire amount of the first phase into the system with no significant losses. More specifically, when two immiscible fluids are combined in a reservoir, the two fluids will tend to separate into layers as long as the densities of the materials are different. If the fluid of interest (e.g., sample fluid) is closest to the exit of the reservoir, it will be the first to leave when the reservoir is emptied (the exit can be on either the top or bottom, depending on the density difference). Once the reagent has been pumped out of the reservoir, the second phase will follow.
  • the fluid of interest e.g., sample fluid
  • This second phase will then push the first phase completely through the system without any sample fluid loss.
  • oil and water the reagent
  • the syringe would be combined in a syringe. If the oil is denser than the water, the syringe would be oriented with its exit face up, if the oil were less dense, then the syringe would be face down. The oil would be chosen such that the materials of interest in the reagent are not soluble in the oil phase.
  • FIG. 9 is one example of this approach when a syringe is used as the reservoir and the second phase is denser than the reagent phase. If the reagent were more dense, then the syringe orientation would be reversed (i.e. the exit would be facing downward in the figure).
  • FIG. 9 shows a two phase system where the reagent is injected on top of the second, immiscible phase: (A) During injection, prior to transition from first phase to second phase, (B) second phase just entering the transfer line,. (C) second phase has completely filled the transfer line and pushed the entire volume of reagent through the system.
  • a sample solution is sandwiched between two immiscible liquids, wherein one liquid has a density greater than the sample density, and the second liquid has a density less than the sample density.
  • the sample density 1.0
  • the sample can be layered between perflourocarbon oil (density 1.8) and mineral oil (density 0.914).
  • perflourocarbon oil density 1.8
  • mineral oil density 0.914
  • the sample then rises to the injection point after the mineral oil. It is further contemplated that the methods disclosed herein would also work for gases.
  • the gases and/or liquids can be miscible, but of different densities such that they are layered on top each other in a manner that prevents their mixing.
  • the present invention also provides solid phase particles and methods for the forming solid phase particles on a microfluidic device for downstream analysis.
  • the solid phase particles can be used for various biological or chemical analysis (e.g., DNA or protein analyses).
  • amplification reaction within droplets using one of several amplification type methods (described in further detail herein), including, but not limited to; PCR, Rolling Circle Amplification (RCA), Nucleic Acid Sequence Based Amplification (NASBA), ligase chain reaction, etc. followed by encapsulation/solidification of the amplified reaction within the droplets by either polymerizing the droplets using chemical or physical means.
  • amplification type methods including, but not limited to; PCR, Rolling Circle Amplification (RCA), Nucleic Acid Sequence Based Amplification (NASBA), ligase chain reaction, etc.
  • a physical means might be termed ‘gelling’ whereby one incorporates low temperature agarose within the droplet during formulation and keeping the droplet above the solidification temperature until one desires the droplet to solidify.
  • a chemical means might be termed ‘polymerization’ whereby one combines (if needed) the droplet with a polymerizing solution and then polymerizing the droplet using either a polymerization initiator (for example free radicals) or a means such as UV light.
  • a polymerization initiator for example free radicals
  • a means such as UV light.
  • Some other means of gelling or polymerization include matragel, polyacrylamide, mixed polysaccharides, etc.
  • Some example initiators can be temperature, UV irradiation, etc.
  • one of the DNA primers used for amplification can be attached to one of the molecules that will form the polymerized matrix. Attachment can be through a direct chemical attachment, or through a secondary attachment such as biotin-streptavidin attachment. In this example, the DNA will become physically attached to the formed solid-phase that occurs after solidification of the droplet. Using one of several gelling or polymerizing methods it should be possible to further manipulate these droplets.
  • proteins can be either trapped within, or attached to the gel or polymer matrix. If attached, it can be through a covalent linkage or through an affinity tag, for example his6 or avi-tag.
  • the proteins can be added to droplets containing gel or polymer reagent, or they can be formulated along with the gel or polymerization reagent. Variations that include both are also possible.
  • the protein can be added to the droplets. Additionally, it is possible to add DNA to the droplet and allow in vitro transcription/translation to synthesize the protein.
  • the droplets can be kept in liquid form on the microfluidic device and either gelled or polymerized upon removal, or can be gelled or polymerized within the droplets anywhere on the device after the droplets have been formed.
  • multiple plasmids are formulated into a droplet along with an in vitro transcription/translation reaction.
  • Genes, encoded by the plasmids are translated and transcribed to protein molecules.
  • the protein molecules attach to the polymer via an avi-tag, the droplets are allowed to gel and the plasmid molecules become ‘fixed’ or ‘trapped’ within the gel.
  • the gelled droplets are collected, the emulsion is broken and the solidified droplets collected and washed.
  • DNA is amplified within a droplet wherein one primer is physically attached to a polymer monomer.
  • the droplet is then combined with a droplet containing the enzymes DNA polymerase, luciferase and sulfurylase.
  • the merged droplets are allowed to gel or polymerize, they are collected, and if needed, washed. These washed gelled droplets can then used for a DNA sequencing reaction.
  • the fluids used in the invention may contain one or more additives, such as agents which reduce surface tensions (surfactants).
  • Surfactants can include Tween, Span, fluorosurfactants, and other agents that are soluble in oil relative to water.
  • performance is improved by adding a second surfactant to the aqueous phase.
  • Surfactants can aid in controlling or optimizing droplet size, flow and uniformity, for example by reducing the shear force needed to extrude or inject droplets into an intersecting channel. This can affect droplet volume and periodicity, or the rate or frequency at which droplets break off into an intersecting channel.
  • the surfactant can serve to stabilize aqueous emulsions in fluorinated oils from coalescing.
  • the droplets may be coated with a surfactant.
  • Preferred surfactants that may be added to the continuous phase fluid include, but are not limited to, surfactants such as sorbitan-based carboxylic acid esters (e.g., the “Span” surfactants, Fluka Chemika), including sorbitan monolaurate (Span 20), sorbitan monopalmitate (Span 40), sorbitan monostearate (Span 60) and sorbitan monooleate (Span 80), and perfluorinated polyethers (e.g., DuPont Krytox 157 FSL, FSM, and/or FSH).
  • surfactants such as sorbitan-based carboxylic acid esters (e.g., the “Span” surfactants, Fluka Chemika), including sorbitan monolaurate (Span 20), sorbitan monopalmitate (Span 40), sorbitan monostearate (Span 60) and sorbitan monooleate (Span 80), and perfluorin
  • non-ionic surfactants which may be used include polyoxyethylenated alkylphenols (for example, nonyl-, p-dodecyl-, and dinonylphenols), polyoxyethylenated straight chain alcohols, polyoxyethylenated polyoxypropylene glycols, polyoxyethylenated mercaptans, long chain carboxylic acid esters (for example, glyceryl and polyglycerl esters of natural fatty acids, propylene glycol, sorbitol, polyoxyethylenated sorbitol esters, polyoxyethylene glycol esters, etc.) and alkanolamines (e.g., diethanolamine-fatty acid condensates and isopropanolamine-fatty acid condensates).
  • alkylphenols for example, nonyl-, p-dodecyl-, and dinonylphenols
  • polyoxyethylenated straight chain alcohols poly
  • ionic surfactants such as sodium dodecyl sulfate (SDS) may also be used.
  • SDS sodium dodecyl sulfate
  • surfactants are generally less preferably for many embodiments of the invention.
  • a water soluble surfactant such as SDS may denature or inactivate the contents of the droplet.
  • the carrier fluid can be an oil (e.g., decane, tetradecane or hexadecane) or fluorocarbon oil that contains a surfactant (e.g., a non-ionic surfactant such as a Span surfactant) as an additive (preferably between about 0.2 and 5% by volume, more preferably about 2%).
  • a surfactant e.g., a non-ionic surfactant such as a Span surfactant
  • a user can preferably cause the carrier fluid to flow through channels of the microfluidic device so that the surfactant in the carrier fluid coats the channel walls.
  • the fluorosurfactant can be prepared by reacting the perflourinated polyether DuPont Krytox 157 FSL, FSM, or FSH with aqueous ammonium hydroxide in a volatile fluorinated solvent.
  • the solvent and residual water and ammonia can be removed with a rotary evaporator.
  • the surfactant can then be dissolved (e.g., 2.5 wt %) in a fluorinated oil (e.g., Flourinert (3M)), which then serves as the continuous phase of the emulsion.
  • a fluorinated oil e.g., Flourinert (3M)
  • the invention can use pressure drive flow control, e.g., utilizing valves and pumps, to manipulate the flow of cells, particles, molecules, enzymes or reagents in one or more directions and/or into one or more channels of a microfluidic device.
  • pressure drive flow control e.g., utilizing valves and pumps
  • other methods may also be used, alone or in combination with pumps and valves, such as electro-osmotic flow control, electrophoresis and dielectrophoresis (Fulwyer, Science 156, 910 (1974); Li and Harrison, Analytical Chemistry 69, 1564 (1997); Fiedler, et al. Analytical Chemistry 70, 1909-1915 (1998); U.S. Pat. No. 5,656,155).
  • Positive displacement pressure driven flow is a preferred way of controlling fluid flow and dielectrophoresis is a preferred way of manipulating droplets within that flow.
  • the pressure at the inlet module can also be regulated by adjusting the pressure on the main and sample inlet channels, for example, with pressurized syringes feeding into those inlet channels.
  • the size and periodicity of the droplets generated may be regulated.
  • a valve may be placed at or coincident to either the inlet module or the sample inlet channel connected thereto to control the flow of solution into the inlet module, thereby controlling the size and periodicity of the droplets.
  • Periodicity and droplet volume may also depend on channel diameter, the viscosity of the fluids, and shear pressure.
  • electro-osmosis is believed to produce motion in a stream containing ions e.g. a liquid such as a buffer, by application of a voltage differential or charge gradient between two or more electrodes. Neutral (uncharged) molecules or cells can be carried by the stream. Electro-osmosis is particularly suitable for rapidly changing the course, direction or speed of flow. Electrophoresis is believed to produce movement of charged objects in a fluid toward one or more electrodes of opposite charge, and away from one on or more electrodes of like charge. Where an aqueous phase is combined with an oil phase, aqueous droplets are encapsulated or separated from each other by oil.
  • the oil phase is not an electrical conductor and may insulate the droplets from the electro-osmotic field.
  • electro-osmosis may be used to drive the flow of droplets if the oil is modified to carry or react to an electrical field, or if the oil is substituted for another phase that is immiscible in water but which does not insulate the water phase from electrical fields.
  • Dielectrophoresis is believed to produce movement of dielectric objects, which have no net charge, but have regions that are positively or negatively charged in relation to each other.
  • dielectric polarizability of the particles and the suspending medium dielectric particles will move either toward the regions of high field strength or low field strength.
  • the polarizability of living cells depends on their composition, morphology, and phenotype and is highly dependent on the frequency of the applied electrical field.
  • cells of different types and in different physiological states generally possess distinctly different dielectric properties, which may provide a basis for cell separation, e.g., by differential dielectrophoretic forces.
  • the polarizability of droplets also depends upon their size, shape and composition. For example, droplets that contain salts can be polarized. According to formulas provided in Fiedler, et al. Analytical Chemistry 70, 1909-1915 (1998), individual manipulation of single droplets requires field differences (inhomogeneities) with dimensions close to the droplets.
  • dielectrophoretic force gradient means a dielectrophoretic force is exerted on an object in an electric field provided that the object has a different dielectric constant than the surrounding media. This force can either pull the object into the region of larger field or push it out of the region of larger field. The force is attractive or repulsive depending respectively on whether the object or the surrounding media has the larger dielectric constant.
  • Manipulation is also dependent on permittivity (a dielectric property) of the droplets and/or particles with the suspending medium.
  • permittivity a dielectric property
  • polymer particles, living cells show negative dielectrophoresis at high-field frequencies in water.
  • dielectrophoretic forces experienced by a latex sphere in a 0.5 MV/m field (10 V for a 20 micron electrode gap) in water are predicted to be about 0.2 piconewtons (pN) for a 3.4 micron latex sphere to 15 pN for a 15 micron latex sphere (Fiedler, et al. Analytical Chemistry 70, 1909-1915 (1998)).
  • Radiation pressure can also be used in the invention to deflect and move objects, e.g. droplets and particles (molecules, cells, particles, etc.) contained therein, with focused beams of light such as lasers.
  • Flow can also be obtained and controlled by providing a pressure differential or gradient between one or more channels of a device or in a method of the invention.
  • Molecules, cells or particles can be moved by direct mechanical switching, e.g., with on-off valves or by squeezing the channels. Pressure control may also be used, for example, by raising or lowering an output well to change the pressure inside the channels on the chip. See, e.g., the devices and methods described U.S. Pat. No. 6,540,895. These methods and devices can further be used in combination with the methods and devices described in pending U.S. Patent Application Publication No.20010029983 and 20050226742. Different switching and flow control mechanisms can be combined on one chip or in one device and can work independently or together as desired.
  • the microfluidic device of the present invention includes one or more inlet modules.
  • An “inlet module” is an area of a microfluidic substrate device that receives molecules, cells, small molecules or particles for additional coalescence, detection and/or sorting.
  • the inlet module can contain one or more inlet channels, wells or reservoirs, openings, and other features which facilitate the entry of molecules, cells, small molecules or particles into the substrate.
  • a substrate may contain more than one inlet module if desired. Different sample inlet channels can communicate with the main channel at different inlet modules. Alternately, different sample inlet channels can communication with the main channel at the same inlet module.
  • the inlet module is in fluid communication with the main channel.
  • the inlet module generally comprises a junction between the sample inlet channel and the main channel such that a solution of a sample (i.e., a fluid containing a sample such as molecules, cells, small molecules (organic or inorganic) or particles) is introduced to the main channel and forms a plurality of droplets.
  • a sample i.e., a fluid containing a sample such as molecules, cells, small molecules (organic or inorganic) or particles
  • the sample solution can be pressurized.
  • the sample inlet channel can intersect the main channel such that the sample solution is introduced into the main channel at an angle perpendicular to a stream of fluid passing through the main channel.
  • the sample inlet channel and main channel intercept at a T-shaped junction; i.e., such that the sample inlet channel is perpendicular (90 degrees) to the main channel.
  • the sample inlet channel can intercept the main channel at any angle, and need not introduce the sample fluid to the main channel at an angle that is perpendicular to that flow.
  • the angle between intersecting channels is in the range of from about 60 to about 120 degrees. Particular exemplary angles are 45, 60, 90, and 120 degrees.
  • Embodiments of the invention are also provided in which there are two or more inlet modules introducing droplets of samples into the main channel.
  • a first inlet module may introduce droplets of a first sample into a flow of fluid in the main channel and a second inlet module may introduce droplets of a second sample into the flow of fluid in main channel, and so forth.
  • the second inlet module is preferably downstream from the first inlet module (e.g., about 30 ⁇ m).
  • the fluids introduced into the two or more different inlet modules can comprise the same fluid or the same type of fluid (e.g., different aqueous solutions).
  • droplets of an aqueous solution containing an enzyme are introduced into the main channel at the first inlet module and droplets of aqueous solution containing a substrate for the enzyme are introduced into the main channel at the second inlet module.
  • the droplets introduced at the different inlet modules may be droplets of different fluids which may be compatible or incompatible.
  • the different droplets may be different aqueous solutions, or droplets introduced at a first inlet module may be droplets of one fluid (e.g., an aqueous solution) whereas droplets introduced at a second inlet module may be another fluid (e.g., alcohol or oil).
  • the droplet pairs can contain liquids of different compositions and/or volumes, which would then combine to allow for a specific reaction to be investigated.
  • the pair of droplets can come from any of the following: (i) two continuous aqueous streams and an oil stream; (ii) a continuous aqueous stream, an emulsion stream, and an oil stream, or (iii) two emulsion streams and an oil stream.
  • interdigitation means pairing of droplets from separate aqueous streams, or from two separate inlet nozzles, for eventual coalescence.
  • the nozzle designs described herein enhance the interdigitation of droplets and further improves coalescence of droplets due to the better control of the interdigitation and smaller distance between pairs of droplets.
  • the greater control over interdigitation allows for a perfect control over the frequency of either of the droplets.
  • the spacing between droplets and coupling of the droplets can be adjusted by adjusting flow of any of the streams, viscosity of the streams, nozzle design (including orifice diameter, the channel angle, and post-orifice neck of the nozzle).
  • a device of the invention can include a sample solution reservoir or well or other apparatus for introducing a sample to the device, at the inlet module, which is typically in fluid communication with an inlet channel.
  • Reservoirs and wells used for loading one or more samples onto the microfluidic device of the present invention include but are not limited to, syringes, cartridges, vials, eppendorf tubes and cell culture materials (e.g., 96 well plates).
  • a reservoir may facilitate introduction of molecules or cells into the device and into the sample inlet channel of each analysis unit.
  • the microfluidic device can include a syringe (or other glass container) that is treated with a vapor or solution of an appropriate PEG-silane to effect the surface PEG functionalization.
  • a syringe or other glass container
  • the purpose for treating the walls of glass containers (e.g., syringes) with a PEG functionality is to prevent biological adhesion to the inner walls of the container, which frustrates the proper transfer of biological/chemical materials into the microfluidic device of the present invention.
  • the inlet channel is further connected to a means for introducing a sample to said device.
  • the means can be a well or reservoir.
  • the means can be temperature controlled.
  • the inlet module may also contain a connector adapted to receive a suitable piece of tubing, such as liquid chromatography or HPLC tubing, through which a sample may be supplied.
  • a suitable piece of tubing such as liquid chromatography or HPLC tubing
  • Such an arrangement facilitates introducing the sample solution under positive pressure in order to achieve a desired infusion rate at the inlet module.
  • the interconnections including tubes, must be extremely clean and make excellent bonding with the PDMS surface in order to allow proper operation of the device.
  • the difficulty in making a fluidic connection to a microfluidic device is primarily due to the difficulty in transitioning from a macroscopic fluid line into the device while minimizing dead volume.
  • tubes and interconnects for the PDMS slab can be cured in place.
  • the tubes and interconnects can be placed in position by applying a UV-cured adhesive to allow for holding the tubes in place on the silicone wafer.
  • PDMS can be poured over the wafer and cured.
  • the cured PDMS, along with the tubes in place, can be peeled off of the silicone wafer easily. This process can be applied to fluidics channels as well as other connection channels. Once the adhesive is applied onto the wafer, the process will allow for quick templating of PDMS slabs with exact reproducibility of channel locations and cleanliness. Tubes of any size can be implemented for this process.
  • the tubing side of the interconnect can be mounted into a retaining block that provides precise registration of the tubing, while the microfluidic device can be positioned accurately in a carrier that the retaining block would align and clamp to.
  • the total dead volume associated with these designs would be critically dependent on how accurately the two mating surfaces could be positioned relative to each other.
  • the maximum force required to maintain the seal would be limited by the exact shape and composition of the sealing materials as well as the rigidity and strength of the device itself.
  • the shapes of the mating surfaces can be tailored to the minimal leakage potential, sealing force required, and potential for misalignment.
  • the single ring indicated in can be replaced with a series of rings of appropriate cross-sectional shape.
  • Reservoirs and wells used for loading one or more samples onto the microfluidic device of the present invention include but are not limited to, syringes, cartridges, vials, eppendorf tubes and cell culture materials (e.g., 96 well plates) as described above.
  • One of the issues to be resolved in loading samples into the inlet channel at the inlet module of the substrate is the size difference between the loading means or injection means, e.g., capillary or HPLC tubing and the inlet channel. It is necessary to create an interconnect and loading method which limits leaks and minimizes dead volume and compliance problems.
  • the present invention includes one or more inlet modules comprising self-aligning fluidic interconnects proximate to one or more inlet channels to improve the efficiency of sample loading and/or injection.
  • the present invention proposes the use of small interconnects based on creating a radial seal instead of a face seal between the microfluidic device and interconnect.
  • the inserted interconnect would have a larger diameter than the mating feature on the device.
  • the stretching of the chip would provide the sealing force needed to make a leak-free seal between the external fluid lines and the microfluidic device.
  • FIG. 11 details design possibilities for making this seal.
  • the external interconnect In order to handle instrument and chip manufacturing tolerances, the external interconnect must be self aligning and the “capture radius” of the molded hole must be large enough to reliably steer the interconnect to the sealing surfaces.
  • FIG. 12 shows that the entrance to the molded hole is large enough to guarantee capture but tapers down to the sealing surfaces.
  • the external interconnect could be made directly out of the tubing leading up to the microfluidic substrate, thus eliminating potential leak points and unswept volumes. As seen in FIG. 12 , the interconnect is surrounded by the substrate interconnects or “chip dock” for most of its length to make certain it is held within the tolerance stack-up of the system.
  • the external interconnect is made from a hard but flexible material such as 1/32′′ PEEK tubing.
  • the features in the microfluidic device can be molded directly into it during the manufacturing process, while the inserted seals can be molded/machined directly onto the tubing ends or molded as individual pieces and mechanically fastened to the tubing.
  • the retaining ferrule shown in FIG. 12 would be attached during manufacturing and provide good absolute referencing of the tube length.
  • the ferrule could be an off-the-shelf component or a custom manufactured part and be made from, for example, a polymer, an elastomer, or a metal.
  • the tubing end could be tapered on the end (top most diagram) or squared off (the figure above). The specific shape of the end will be controlled by how easily the microfluidic device will gall during insertion.
  • the present invention also provides methods of direct molding of fluidic interconnects into a microfluidic device.
  • Development of a commercial microfluidic platform requires a simple, reliable fluidic interconnect in order to reduce the chance of operator error and leaks. Molding these interconnects directly into the microfluidic device requires precise alignment of the molding pins to the patterned shim (the “master” manufactured from Silicon/photoresist or made from some metal) used to form the microfluidic and electrical channels.
  • the extreme tolerances required when molding with a low viscosity elastomer such as PDMS requires near perfect sealing of the pin face to the master, while still accommodating imperfections in the master and assembly of the molding tool.
  • the present invention provides a precise and repeatable method of molding of interconnects while accommodating the imperfections in the molding process by introducing movable pins captured in an elastomeric sleeve molded directly into the tool.
  • the tool In order to effectively produce at relatively low volume and be able to inexpensively prototype devices, the tool must be able to use masters generated using standard photolithographic processes (e.g. silicon wafers patterned with SU-8).
  • FIG. 14 shows a schematic of a molding tool based on this concept.
  • the pins (orange) are captured within an elestomeric molded sleeve.
  • a compression plate made from a rigid backer plate and foam rubber is used to apply gentle even pressure to the pins and generate the force needed to make the pins uniformly contact the master.
  • the molded sleeve was found to be necessary to consistently prevent the uncured elastomer from penetrating the region between the pin and the top plate.
  • Early designs used pins captured in tight clearance holes, and the pins would frequently bind in place (even with lubricant), preventing smooth motion of the pins and improper contact with the master.
  • the well or reservoir of the inlet module further include an acoustic actuator.
  • an acoustic actuator To obtain one droplet comprising a single element of a specific biological/chemical material (e.g., a cell), separation of biological/chemical material, and uniformity of the number density of biological/chemical materials in a microfluidic channel is desirable.
  • the microfluidic device can include an acoustic actuator.
  • the loaded sample (biological/chemical material) can be well mixed and separated in a small chamber by acoustic wave before sending out to the nozzle region for encapsulation.
  • the frequency of the acoustic wave should be fine tuned so as not to cause any damage to the cells.
  • the biological effects of acoustic mixing have been well studied (e.g., in the ink-jet industry) and many published literatures also showed that piezoelectric microfluidic device can deliver intact biological payloads such as live microorganisms and DNA.
  • the design of the acoustic resonant can use a Piezoelectric bimorph flat plate located on the side of the carved resonant in the PDMS slab.
  • the resonant inlet can connect to the cell flow input channel and the outlet can connect to the cell flow pinching channel.
  • the piezoelectric driving waveform can be carefully optimized to select the critical frequencies that can separate cells in fluids. There are five parameters to optimize beyond the frequency parameter and Lab electronics can be used to optimize the piezoelectric driving waveform. Afterwards, a low cost circuit can be designed to generate only the optimized waveform in a preferred microfluidic device.
  • the microfluidic device of the present invention also includes one or more coalescence modules.
  • a “coalescence module” is within or coincident with at least a portion of the main channel at or downstream of the inlet module where molecules, cells, small molecules or particles comprised within droplets are brought within proximity of other droplets comprising molecules, cells, small molecules or particles and where the droplets in proximity fuse, coalesce or combine their contents.
  • the coalescence module can also include an apparatus, for generating an electric force.
  • the electric force exerted on the fluidic droplet may be large enough to cause the droplet to move within the liquid.
  • the electric force exerted on the fluidic droplet may be used to direct a desired motion of the droplet within the liquid, for example, to or within a channel or a microfluidic channel (e.g., as further described herein), etc.
  • the electric field can be generated from an electric field generator, i.e., a device or system able to create an electric field that can be applied to the fluid.
  • the electric field generator may produce an AC field (i.e., one that varies periodically with respect to time, for example, sinusoidally, sawtooth, square, etc.), a DC field (i.e., one that is constant with respect to time), a pulsed field, etc.
  • the electric field generator may be constructed and arranged to create an electric field within a fluid contained within a channel or a microfluidic channel.
  • the electric field generator may be integral to or separate from the fluidic system containing the channel or microfluidic channel, according to some embodiments.
  • integrated means that portions of the components integral to each other are joined in such a way that the components cannot be in manually separated from each other without cutting or breaking at least one of the components.
  • an electric field is produced by applying voltage across a pair of electrodes, which may be positioned on or embedded within the fluidic system (for example, within a substrate defining the channel or microfluidic channel), and/or positioned proximate the fluid such that at least a portion of the electric field interacts with the fluid.
  • the electrodes can be fashioned from any suitable electrode material or materials known to those of ordinary skill in the art, including, but not limited to, silver, gold, copper, carbon, platinum, copper, tungsten, tin, cadmium, nickel, indium tin oxide (“ITO”), etc., as well as combinations thereof.
  • the device can include channels for use in fluid control and other channels filled with a metal alloy for casting integrated metal alloy components (i.e., electrodes).
  • the electrodes can be manufactured using other technologies (e.g., lithographically patterned electrodes made from indium tin oxide or a metal such as platinum).
  • the microfluidic device can include metal alloy components useful for performing electrical functions on fluids, including but not limited to, coalescing droplets, charging droplets, sorting droplets, detecting droplets and shaking droplets to mix the contents of coalesced droplets.
  • the device can contain more than one of the above mentioned components for more than one of the above mentioned functions.
  • the electrodes comprising metal alloy components may either terminate at fluid channels or be isolated from fluid channels.
  • the electrodes can be constructed by filling the appropriate channels with metal alloy. One way this can be accomplished is to use positive pressure injection of the metal alloy in a melted state, such as with a syringe, into the channels, and then cool the metal alloy to a solid form. Another example is to use negative pressure to draw the metal alloy in a melted state into the channels, and then cool the metal alloy to a solid form. This can be accomplished for example by use of capillary forces. Another method of construction can use any of the above mentioned embodiments, and then flush out the metal alloy in a melted state with another liquid to define the geometry of the metal alloy components.
  • Another example is to use any of the above mentioned embodiments, and then use a localized cold probe to define a solid termination point for the metal alloy, and then cool the remaining metal alloy to a solid form.
  • a further example is to use another material, such as microscopic solder spheres or UV curable conductive ink, to form a barrier between fluid and metal alloy channels, to define the geometry of the metal alloy components.
  • the device can include a combination of both integrated metal alloy components and a patterned electrically conductive layer.
  • the patterned electrically conductive layer can have features patterned such that their boundaries are within a leak-proof seal.
  • the device can have a patterned electrically conductive feature as one of two charging electrodes and one integrated metal alloy component as the other of two charging electrodes.
  • the device can include a plurality of electrodes that are insulated from the fluid present in the device, and the method of operation including appropriate application of dielectrical signals and appropriate fluids.
  • the electrodes are typically in contact with the fluids in order to allow discharge of species that would otherwise screen the applied dielectric field.
  • this screening effect typically arises so quickly that the device is not useful for any significantly extended period of time.
  • the drawbacks of electrodes in contact with the fluids vs.
  • insulated electrodes are (a) degraded reliability against leaking (since the interface between the electrodes and the other components of the device may be more difficult to effect a leak-proof seal), and (b) degraded reliability against electrode corrosion (whose failure mode effects include failure of application of dielectric fields, and fluid channel contamination).
  • the device of the present invention comprising a plurality of electrodes that are insulated from the fluid present in the device counteracts this screening effect by extending the screening rise time and including a polarity switch for all of the different dielectric fields applied in the device.
  • the screening rise time is extended by using fluids with dielectrical properties.
  • a polarity switch for all of the different dielectric fields applied in the device is achieved by using an algorithm for dielectrical control, which switches the polarity of the dielectrical fields at a frequency sufficiently high to maintain proper dielectrical function of the device.
  • This dielectrical control algorithm may also switch the polarity for the dielectric fields in a cascading, time controlled manner starting at the fluid origin point and progressing downstream, so that given fluid components experience one polarity at every point along their course.
  • the device of the present invention can be used with metal alloy electrodes or using a combination of metal alloy electrodes and patterned conductive film electrodes.
  • the invention can provide a microfluidic device using injected electrodes.
  • the interface between the microscopic electrode (typically 25 ⁇ m thick) and the macroscopic interconnect can easily fail if the joint between the two is flexed.
  • the flexing of the joint can be eliminated by securing a firm material that serves to fasten, support, and re-enforce the joint (i.e., a grommet) into the interface.
  • the mating surface of the device can be manufactured from a hard material such as glass or plastic.
  • the electrical connection with the external system can be made by securing the device such that it connects to a spring loaded contact, which is either offset from the grommet (thereby minimizing the force applied to the solder region), or centered on the grommet (as long as the contact does not touch the solder).
  • the metal alloy components are also useful for performing optical functions on fluids, including but not limited to, optical detection of droplets in a geometry which may include a mirror.
  • the microfluidic device can include a layer patterned with channels for fluid control, and another layer with patterned electrically conductive features, where the features are patterned such that their boundaries are within a leak-proof seal.
  • the leak-proof seal can be achieved at the interface between the unpatterned areas of the fluid control layer and the unpatterned areas of the electrically conductive layer.
  • the leak-proof seal can also be achieved by a third interfacial layer between the fluid control layer and the unpatterned areas of the electrically conductive layer.
  • the third interfacial layer can or can not be perforated at specific locations to allow contact between the fluid and the electrically conductive layer.
  • Electrical access ports can also be patterned in the fluid control layer.
  • the electrodes and patterned electrically conductive layers as described can be associated with any module of the device (inlet module, coalescence module, mixing module, delay module, detection module and sorting module) to generate dielectric or electric forces to manipulate and control the droplets and their contents.
  • the microfluidic device according to the present invention can include placing a fluidic channel between two parallel electrodes, which can result in a steep electric field gradient at the entrance to the electrodes due to edge effects at the ends of the electrode pair. Placing these pairs of electrodes at a symmetric channel split can allow precise bi-directional control of droplet within a device. Using the same principle, only with asymmetric splits, can allow single ended control of the droplet direction in the same manner. Alternatively, a variation on this geometry will allow precise control of the droplet phase by shifting.
  • the electric field generator can be constructed and arranged (e.g., positioned) to create an electric field applicable to the fluid of at least about 0.01 V/micrometer, and, in some cases, at least about 0.03 V/micrometer, at least about 0.05 V/micrometer, at least about 0.08 V/micrometer, at least about 0.1 V/micrometer, at least about 0.3 V/micrometer, at least about 0.5 V/micrometer, at least about 0.7 V/micrometer, at least about 1 V/micrometer, at least about 1.2 V/micrometer, at least about 1.4 V/micrometer, at least about 1.6 V/micrometer, or at least about 2 V/micrometer.
  • even higher electric field intensities may be used, for example, at least about 2 V/micrometer, at least about 3 V/micrometer, at least about 5 V/micrometer, at least about 7 V/micrometer, or at least about 10 V/micrometer or more.
  • an electric field may be applied to fluidic droplets to cause the droplets to experience an electric force.
  • the electric force exerted on the fluidic droplets may be, in some cases, at least about 10 ⁇ 16 N/micrometer 3 .
  • the electric force exerted on the fluidic droplets may be greater, e.g., at least about 10 ⁇ 15 N/micrometer 3 , at least about 10 ⁇ 14 N/micrometer 3 , at least about 10 ⁇ 13 N/micrometer 3 , at least about 10 ⁇ 12 N/micrometer 3 , at least about 10 ⁇ 11 N/micrometer 3 , at least about 10 ⁇ 10 N/micrometer 3 , at least about 10 ⁇ 9 N/micrometer 3 , at least about 10 ⁇ 8 N/micrometer 3 , or at least about 10 ⁇ 7 N/micrometer 3 or more.
  • the electric force exerted on the fluidic droplets, relative to the surface area of the fluid may be at least about 10 ⁇ 15 N/micrometer 2 , and in some cases, at least about 10 ⁇ 14 N/micrometer 2 , at least about 10 ⁇ 13 N/micrometer 2 , at least about 10 ⁇ 12 N/micrometer 2 , at least about 10 ⁇ 11 N/micrometer 2 , at least about 10 ⁇ 10 N/micrometer 2 , at least about 10 ⁇ 9 N/micrometer 2 , at least about 10 ⁇ 8 N/micrometer 2 , at least about 10 ⁇ 7 N/micrometer 2 , or at least about 10 ⁇ 6 N/micrometer 2 or more.
  • the electric force exerted on the fluidic droplets may be at least about 10 ⁇ 9 N, at least about 10 ⁇ 8 N, at least about 10 ⁇ 7 N, at least about 10 ⁇ 6 N, at least about 10 ⁇ 5 N, or at least about 10 ⁇ 4 N or more in some cases.
  • droplet coalescence is presently carried out by having two droplet forming nozzles emitting droplets into the same main channel.
  • the size of the nozzles allow for one nozzle to form a large drop that fills the exhaust line while the other nozzle forms a drop that is smaller than the first.
  • the smaller droplet is formed at a rate that is less than the larger droplet rate, which insures that at most one small droplet is between big droplets.
  • the small droplet will catch up to the larger one over a relatively short distance, but sometimes the recirculation zone behind the large drop causes the small drop to separate from the large drop cyclically.
  • the small drop occasionally does not catch up with the large one over the distance between the nozzles and the coalescing electrodes. Thus, in some situations is a need for a more robust coalescence scheme.
  • FIG. 15 is a schematic diagram of the improved coalescence module.
  • a small constriction (neckdown) just before this expansion can be used to better align the droplets on their way into the coalescence point (also shown in the FIG. 15 ).
  • This optional neckdown can help center the small droplet in the channel stream lines, reducing the chance that it will flow around the larger droplet prior to coalescing in the expansion.
  • the electrode pair may be placed on either one side of the channel or on both sides.
  • the expansion in the coalescing region allows for a dramatic catching up of the small drop to the large drop, as shown through micrographs taken on an operating device.
  • the volume of the expansion is big enough to slow the large droplet down so that the small drop always catches up to the large drop, but doesn't allow the next large drop to catch up and make contact with the pair to be coalesced.
  • the electrodes allow for coalescence to take place when the drops are in contact with each other and passing through the field gradient.
  • the microfluidic device of the present invention can also include one or more detection modules.
  • a “detection module” is a location within the device, typically within the main channel where molecules, cells, small molecules or particles are to be detected, identified, measured or interrogated on the basis of at least one predetermined characteristic.
  • the molecules, cells, small molecules or particles can be examined one at a time, and the characteristic is detected or measured optically, for example, by testing for the presence or amount of a reporter.
  • the detection module is in communication with one or more detection apparatuses.
  • the detection apparatuses can be optical or electrical detectors or combinations thereof.
  • detection apparatuses include optical waveguides, microscopes, diodes, light stimulating devices, (e.g., lasers), photo multiplier tubes, and processors (e.g., computers and software), and combinations thereof, which cooperate to detect a signal representative of a characteristic, marker, or reporter, and to determine and direct the measurement or the sorting action at the sorting module.
  • light stimulating devices e.g., lasers
  • photo multiplier tubes e.g., computers and software
  • processors e.g., computers and software
  • determining generally refers to the analysis or measurement of a species, for example, quantitatively or qualitatively, and/or the detection of the presence or absence of the species. “Determining” may also refer to the analysis or measurement of an interaction between two or more species, for example, quantitatively or qualitatively, or by detecting the presence or absence of the interaction.
  • spectroscopy such as infrared, absorption, fluorescence, UV/visible, FTIR (“Fourier Transform Infrared Spectroscopy”), or Raman
  • gravimetric techniques such as infrared, absorption, fluorescence, UV/visible, FTIR (“Fourier Transform Infrared Spectroscopy”), or Raman
  • gravimetric techniques such as ellipsometry; piezoelectric measurements; immunoassays; electrochemical measurements; optical measurements such as optical density measurements; circular dichroism; light scattering measurements such as quasielectric light scattering; polarimetry; refractometry; or turbidity measurements as described further herein.
  • a detection module is within, communicating or coincident with a portion of the main channel at or downstream of the inlet module and, in sorting embodiments, at, proximate to, or upstream of, the sorting module or branch point.
  • the sorting module may be located immediately downstream of the detection module or it may be separated by a suitable distance consistent with the size of the molecules, the channel dimensions and the detection system. Precise boundaries for the detection module are not required, but are preferred.
  • Detection modules used for detecting molecules and cells have a cross-sectional area large enough to allow a desired molecule, cells, bead, or particles to pass through without being substantially slowed down relative to the flow carrying it.
  • the dimensions of the detection module are influenced by the nature of the sample under study and, in particular, by the size of the droplets, beads, particles, molecules or cells (including virions) under study.
  • mammalian cells can have a diameter of about 1 to 50 microns, more typically 10 to 30 microns, although some mammalian cells (e.g., fat cells) can be larger than 120 microns.
  • Plant cells are generally 10 to 100 microns. However, other molecules or particles can be smaller with a diameter from about 20 nm to about 500 nm.
  • the present invention provides self-aligning optical waveguides and optical elements (lenses, prisms, mirrors, interconnects, etc.) for detection and control of droplets.
  • Such waveguides can be used to provide well defmed optical access to the fluidic channels to permit optical scattering, absorption, fluorescence, or any other optical measurement technique.
  • a separate series of channels and useful shapes can be created either simultaneously within the other channels in the substrate (i.e. in the same processing step) or in successive steps.
  • the reusable master created in this way can then used to form the waveguide components and fluid channels without the need for special fixturing or careful alignment in subsequent steps.
  • the extra channels or shapes can then filled with a high index of refraction liquid (for waveguides) or reflective material (for mirrors) through injection into the channel or void.
  • the liquid can either remain as a fluid or be allowed to solidify.
  • UV cure epoxies used by the telecommunications industry are excellent choices for the waveguide materials.
  • Possible waveguide geometry can include a focusing lens and a back-reflecting mirror.
  • One or more detections sensors and/or processors may be positioned to be in sensing communication with the fluidic droplet.
  • Sensing communication means that the sensor may be positioned anywhere such that the fluidic droplet within the fluidic system (e.g., within a channel), and/or a portion of the fluidic system containing the fluidic droplet may be sensed and/or determined in some fashion.
  • the sensor may be in sensing communication with the fluidic droplet and/or the portion of the fluidic system containing the fluidic droplet fluidly, optically or visually, thermally, pneumatically, electronically, or the like.
  • the sensor can be positioned proximate the fluidic system, for example, embedded within or integrally connected to a wall of a channel, or positioned separately from the fluidic system but with physical, electrical, and/or optical communication with the fluidic system so as to be able to sense and/or determine the fluidic droplet and/or a portion of the fluidic system containing the fluidic droplet (e.g., a channel or a microchannel, a liquid containing the fluidic droplet, etc.).
  • a sensor may be free of any physical connection with a channel containing a droplet, but may be positioned so as to detect electromagnetic radiation arising from the droplet or the fluidic system, such as infrared, ultraviolet, or visible light.
  • the electromagnetic radiation may be produced by the droplet, and/or may arise from other portions of the fluidic system (or externally of the fluidic system) and interact with the fluidic droplet and/or the portion of the fluidic system containing the fluidic droplet in such as a manner as to indicate one or more characteristics of the fluidic droplet, for example, through absorption, reflection, diffraction, refraction, fluorescence, phosphorescence, changes in polarity, phase changes, changes with respect to time, etc.
  • a laser may be directed towards the fluidic droplet and/or the liquid surrounding the fluidic droplet, and the fluorescence of the fluidic droplet and/or the surrounding liquid may be determined.
  • “Sensing communication,” as used herein may also be direct or indirect.
  • light from the fluidic droplet may be directed to a sensor, or directed first through a fiber optic system, a waveguide, etc., before being directed to a sensor.
  • Non-limiting examples of detection sensors useful in the invention include optical or electromagnetically-based systems.
  • the sensor may be a fluorescence sensor (e.g., stimulated by a laser), a microscopy system (which may include a camera or other recording device), or the like.
  • the sensor may be an electronic sensor, e.g., a sensor able to determine an electric field or other electrical characteristic.
  • the sensor may detect capacitance, inductance, etc., of a fluidic droplet and/or the portion of the fluidic system containing the fluidic droplet.
  • the sensor may be connected to a processor, which in turn, cause an operation to be performed on the fluidic droplet, for example, by sorting the droplet.
  • Characteristics determinable with respect to the droplet and usable in the invention can be identified by those of ordinary skill in the art. Non-limiting examples of such characteristics include fluorescence, spectroscopy (e.g., optical, infrared, ultraviolet, etc.), radioactivity, mass, volume, density, temperature, viscosity, pH, concentration of a substance, such as a biological substance (e.g., a protein, a nucleic acid, etc.), or the like.
  • a corresponding signal is then produced, for example indicating that “yes” the characteristic is present, or “no” it is not.
  • the signal may correspond to a characteristic qualitatively or quantitatively. That is, the amount of the signal can be measured and can correspond to the degree to which a characteristic is present.
  • the strength of the signal may indicate the size of a molecule, or the potency or amount of an enzyme expressed by a cell, or a positive or negative reaction such as binding or hybridization of one molecule to another, or a chemical reaction of a substrate catalyzed by an enzyme.
  • data can be collected and/or a control system in the sorting module, if present, can be activated to divert a droplet into one branch channel or another for delivery to the collection module or waste module.
  • a control system in the sorting module if present, can be activated to divert a droplet into one branch channel or another for delivery to the collection module or waste module.
  • molecules or cells within a droplet at a sorting module can be sorted into an appropriate branch channel according to a signal produced by the corresponding examination at a detection module.
  • the means of changing the flow path can be accomplished through mechanical, electrical, optical, or some other technique as described herein.
  • a preferred detector is an optical detector, such as a microscope, which may be coupled with a computer and/or other image processing or enhancement devices to process images or information produced by the microscope using known techniques.
  • molecules can be analyzed and/or sorted by size or molecular weight.
  • Enzymes can be analyzed and/or sorted by the extent to which they catalyze chemical reaction of a substrate (conversely, substrate can be analyzed and/or sorted by the level of chemical reactivity catalyzed by an enzyme).
  • Cells can be sorted according to whether they contain or produce a particular protein, by using an optical detector to examine each cell for an optical indication of the presence or amount of that protein.
  • the protein may itself be detectable, for example by a characteristic fluorescence, or it may be labeled or associated with a reporter that produces a detectable signal when the desired protein is present, or is present in at least a threshold amount.
  • a characteristic fluorescence or it may be labeled or associated with a reporter that produces a detectable signal when the desired protein is present, or is present in at least a threshold amount.
  • characteristics that can be identified or measured using the techniques of the invention, which include without limitation surface characteristics of the cell and intracellular characteristics, provided only that the characteristic or characteristics of interest for sorting can be sufficiently identified and detected or measured to distinguish cells having the desired characteristic(s) from those which do not.
  • any label or reporter as described herein can be used as the basis for analyzing and/or sorting molecules or cells, i.e. detecting molecules or cells to be collected.
  • the biological/chemical entity to be analyzed may itself be detectable, for example by a characteristic fluorescence, or it may be labeled or associated with a reporter that produces a detectable signal when the desired protein is present, or is present in at least a threshold amount.
  • Luminescent colloidal semiconductor nanocrystals called quantum dots or q-dots (QD) are inorganic fluorophores that have the potential to circumvent some of the functional limitations encountered by organic dyes.
  • CdSe—ZnS core-shell QDs exhibit size-dependent tunable photoluminescence (PL) with narrow emission bandwidths (FWHM ⁇ 30 to 45 nm) that span the visible spectrum and broad absorption bands. These allow simultaneous excitation of several particle sizes (colors) at a common wavelength. This, in turn, allows simultaneous resolution of several colors using standard instrumentation.
  • CdSe—ZnS QDs also have high quantum yields, are resistant to photodegradation, and can be detected optically at concentrations comparable to organic dyes.
  • Quantum dots are nano-scale semiconductors typically consisting of materials such as crystalline cadmium selenide.
  • the term ‘q-dot’ emphasizes the quantum confinement effect of these materials, and typically refers to fluorescent nanocrystals in the quantum confined size range.
  • Quantum confinement refers to the light emission from bulk (macroscopic) semiconductors such as LEDs which results from exciting the semiconductor either electrically or by shining light on it, creating electron-hole pairs which, when they recombine, emit light. The energy, and therefore the wavelength, of the emitted light is governed by the composition of the semiconductor material.
  • the physical size of the semiconductor is considerably reduced to be much smaller than the natural radius of the electron-hole pair (Bohr radius)
  • additional energy is required to “confine” this excitation within the nanoscopic semiconductor structure leading to a shift in the emission to shorter wavelengths.
  • Three different q-dots in several concentrations each can be placed in a microdroplet, and can then be used with a microfluidic device to decode what is in the drop.
  • the Q-dot readout extension to the fluorescence station can be incorporated into the design of the microfluidic device.
  • a series of dichroic beamsplitters, emission filters, and detectors can be stacked onto the system, allowing measurement of the required five emission channels (two fluorescence polarization signals and three q-dot bands).
  • Fluorescence Polarization (FP) detection technology enables homogeneous assays suitable for high throughput screening assays in the Drug Discovery field.
  • the most common label in the assays is fluorescein.
  • FP-assay the fluorophore is excited with polarized light. Only fluorophores parallel to the light absorb and are excited. The excited state has a lifetime before the light emission occurs. During this time the labeled fluorophore molecule rotates and the polarization of the light emitted differs from the excitation plane.
  • S-plane a polarized emission filter parallel to the excitation filter
  • P-plane a polarized emission filter perpendicular to the excitation filter
  • the rotational speed of a molecule is dependent on the size of the molecule, temperature and viscosity of the solution.
  • Fluorescein has a fluorescence lifetime suitable for the rotation speeds of molecules in bio-affinity assays like receptor-ligand binding assays or immunoassays of haptens.
  • the basic principle is that the labeled compound is small and rotates rapidly (low polarization). When the labeled compound binds to the larger molecule, its rotation slows down considerably (polarization changes from low to high polarization).
  • FP provides a direct readout of the extent of tracer binding to protein, nucleic acids, and other biopolymers.
  • Fluorescence polarization technology has been used in basic research and commercial diagnostic assays for many decades, but has begun to be widely used in drug discovery only in the past six years.
  • FP assays for drug discovery were developed for single-tube analytical instruments, but the technology was rapidly converted to high-throughput screening assays when commercial plate readers with equivalent sensitivity became available.
  • These assays include such well-known pharmaceutical targets such as kinases, phosphatases, proteases, G-protein coupled receptors, and nuclear receptors.
  • Other homogeneous technologies based on fluorescence intensity have been developed. These include energy transfer, quenching, and enhancement assays.
  • FP offers several advantages over these. The assays are usually easier to construct, since the tracers do not have to respond to binding by intensity changes.
  • FP is independent of intensity, it is relatively immune to colored solutions and cloudy suspensions. FP offers several advantages in the area of instrumentation. Because FP is a fundamental property of the molecule, and the reagents are stable, little or no standardization is required. FP is relatively insensitive to drift in detector gain settings and laser power.
  • the dyes chosen for FP are commonly used in most cell- and enzyme-based assays and are designed not to overlap significantly with the q-dots.
  • the dyes are evaluated both independently and together with the q-dots (at first off-instrument) to assess the cross-talk.
  • the liquid q-dot labels are read outside a spectral wavelength band currently used in FACS analysis and sorting (i.e., the dyes flourescein, Cy3, Cy5, etc). This permits the use of currently-available assays (dependent on these dyes). Using specific q-dots, crosstalk is minimized.
  • the present invention provides methods to label droplets and/or nanoreactors formed on a microfluidic device by using only a single dye code to avoid cross-talk with other dyes during FP. Additionally, the present invention provides methods to create FP dye codes to label compounds contained within liquids (including droplets and/or nanoreactors) where the compound is designed to be differentiated by FP on a microfluidic device. In this manner, dye codes having the same color, absorption, and emission could be used to label compounds within liquids.
  • the present invention is directed to the use of fluorescence polarization to label liquids.
  • Droplets can be labeled using several means. These labeling means include, but are not limited to, the use of different dyes, quantum dots, capacitance, opacity, light scattering, fluorecence intensity (FI), fluorescence lifetime (FL), fluorescence polarization (FP), circular dichroism (CD), fluorescenece correlation and combinations of all of these previous labeling means.
  • the following disclosure describes the use of FP and FI as a means to label droplets on a microfluidic device.
  • FL as a means to adjust the overall FP of a solution, and by varying the concentration of the total FI, to create a 2-dimensional encoding scheme is demonstrated.
  • FP is independent of the concentration of the dye; liquids can have vastly different concentrations of FITC in them yet still have identical FP measurements.
  • a FP dye is an organic dye that does not interfere with the assay dye is used. Furthermore, since the total intensity of the FP dye can be quantified, a second dimension in which to label the droplet is provided. Thus, one can exploit the differences in FP to create an encoding scheme of dye within a liquid solution, including droplets.
  • An example is shown in FIG. 17 whereby the droplets are labeled with 3 differently-sized FITC molecules (i.e., three different droplets contain FITC molecules and FITC coupled to either biotin or streptavidin, respectively). Therefore, in a single dimension, FP can be used to create an encoding scheme.
  • the present invention can also use Fluorescence Intensity (FI) of the overall solution to create even more labels in a second dimension.
  • FIG. 18 An example of labeling droplets in 2 dimensions is shown in FIG. 18 .
  • FIG. 17 discusses the use of multiple compounds to which a dye molecule is attached to span a range of FP, it is also possible to span the range using a high and low molecular weight compound set.
  • a dye can be attached to a large compound (for example streptavidin) and kept at a fixed concentration, to which a smaller compound (for example, a free dye molecule) would be titrated into the same solution.
  • the FP of the solution can be adjusted to be in discernable increments from the value of the large molecule to somewhere slightly greater than the FP of the smaller molecule.
  • the [total] dye intensity can be varied by varying the concentration of the mixture of the two dye-attached compounds. By varying total dye concentration and the FP, two dimensions can be used to generate the FP dye codes (FPcodes). Accordingly, many FPcodes can be generated using only two compounds.
  • the present invention is directed labeling solids using properties other than dye emission and dye concentration.
  • the solid can include, for example, a bead or location on a solid support or chip.
  • FI and FL can be two of many dimensions of characteristics used as labels.
  • a linker can be used to couple the dye to the bead.
  • the linker can be varied so as to allow the dye to have differing degrees of freedom in which to rotate (i.e., tumble). Varying the linker in this manner can change the FP of the attached dye, which in unique combinations can be used as a label.
  • the beads can be swollen in organic solvent and the dyes held in place by hydrophobic forces.
  • the FP, FI, FL methods described above for liquid labeling can also be used as a means for labeling the beads.
  • a quenching molecule can also be used to change the characteristics of a dye.
  • Such quenching can be continuous or brought about through the interaction of a molecule, such as a peptide or nucleic acid linker, with differing means of bringing molecules together depending on the strength of linker-internal interaction (e.g., a nucleotide stem loop structure of varying lengths).
  • a molecule such as a peptide or nucleic acid linker
  • the reactions analyzed on the virtual, random and non-random arrays can be also increased beyond the two (cy3 and cy5 intensities) commonly used for multiplexing.
  • different FP, FI, etc can be used as a read-out.
  • Random array decoding Beads of the prior art use one or more pre-attached oligonucleotide-coupled beads that are held in place in a fiber-optic faceplate (for example, those used by Illiumina). The oligos on the beads are decoded using sequential hybridization of a labeled complementary oligo.
  • the assay of the prior art uses a separate oligonucleotide complementary zipcode (‘Illumacode’) attached to each type of bead.
  • the invention described herein is superior to the methods of the prior art in that the FP, FI, FL-labeled bead or mobile solid support can be placed into a random array (e.g., a chip as manufactured by Illumina) and the FP, FI, FL used to decode the bead.
  • the FP, FI, FL of the bead can be decoded before using the chip and the different beads ‘mapped’ as to their specific locations.
  • the bead can be decoded during attachment of the assay read-out.
  • the methods described by the present invention can be used to pre-determine the location of each bead-type either before, or during analysis.
  • Virtual array decoding Methods of the prior art use 2 lasers and 3 detectors to differentiate a set of 100 bead-types.
  • the beads-types are differentiated by the FI of two different dyes present in 1 of 10 concentrations (per dye) contained within the bead, and the assay detector is used to measure fluorescein concentration on the bead.
  • the dyes which are added to organic-solvent swollen beads, are not directly attached to the beads, but remain held within the bead by hydrophobic forces.
  • a second detector to the machines of the prior art used to measure FP can be added, thereby adding a third dimension and extending the encoding scheme beyond the 100 available in the prior art.
  • Non-random array decoding In chips of the prior art (such as those used by Affymetrix) oligonucleotides are synthesized directly on the chip. Decoding is simply a matter of knowing the location of the assay on the chip.
  • the methods as described herein can be advantageously used in conjunction with such chips to increase the number of things that can be simultaneously analyzed (i.e., multiplexed) on the chip.
  • Cy3, Cy5, FL and FP can be used as analysis markers for hybridization reactions.
  • the present invention also provides methods for labeling micro or nano-sized droplets using Radio Frequency Identification (RFID).
  • RFID tags can improve the identification of the contents within the droplets.
  • the droplets are utilized within a microfluidic device.
  • RFID is an automatic identification method, relying on storing and remotely retrieving data using devices called RFID tags or transponders.
  • An RFID tag is an object that can be attached to or incorporated into a product, animal, or person for the purpose of identification using radio waves.
  • Chip-based RFID tags contain silicon chips and antennae. Passive tags require no internal power source, whereas active tags require a power source.
  • Hitachi has “powder” 0.05mm ⁇ 0.05mm RFID chips. The new chips are 64 times smaller than the previous record holder, the 0.4mm ⁇ 0.4mm mu-chips, and nine times smaller than Hitachi's last year prototype, and have room for a 128-bit ROM that can store a unique 38-digit ID number.
  • a solution containing RFID tags are emulsified into droplets and are used as a label for the identification of the material within the droplet solution.
  • Applications include, but are not limited to; genetics, genomics, proteomics, chemical synthesis, biofuels, and others.
  • the detection module may include an apparatus for stimulating a reporter for that characteristic to emit measurable light energy, e.g., a light source such as a laser, laser diode, light emitting diode (LED), high-intensity lamp, (e.g., mercury lamp), and the like.
  • a lamp e.g., the channels are preferably shielded from light in all regions except the detection module.
  • the laser can be set to scan across a set of detection modules from different analysis units.
  • laser diodes or LED's may be microfabricated into the same chip that contains the analysis units.
  • laser diodes or LED's may be incorporated into a second chip (i.e., a laser diode chip) that is placed adjacent to the analysis or microchip such that the laser light from the diodes shines on the detection module(s).
  • An integrated semiconductor laser and/or an integrated photodiode detector can be included on the substrate in the vicinity of the detection module. This design provides the advantages of compactness and a shorter optical path for exciting and/or emitted radiation, thus minimizing distortion and losses.
  • Fluorescence produced by a reporter is excited using a laser beam focused on molecules (e.g., DNA, protein, enzyme or substrate) or cells passing through a detection region.
  • Fluorescent reporters can include, but are not limited to, rhodamine, fluorescein, Texas red, Cy 3, Cy 5, phycobiliprotein (e.g., phycoerythrin), green fluorescent protein (GFP), YOYO-1 and PicoGreen.
  • the reporter labels can be fluorescently labeled single nucleotides, such as fluorescein-dNTP, rhodamine-dNTP, Cy3-dNTP, etc.; where dNTP represents dATP, dTTP, dUTP or dCTP.
  • the reporter can also be chemically-modified single nucleotides, such as biotin-dNTP.
  • the reporter can be fluorescently or chemically labeled amino acids or antibodies (which bind to a particular antigen, or fragment thereof, when expressed or displayed by a cell or virus).
  • the device can analyze and/or sort cells based on the level of expression of selected cell markers, such as cell surface markers, which have a detectable reporter bound thereto, in a manner similar to that currently employed using fluorescence-activated cell sorting (FACS) machines. Proteins or other characteristics within a cell, and which do not necessarily appear on the cell surface, can also be identified and used as a basis for sorting.
  • the device can also determine the size or molecular weight of molecules such as polynucleotides or polypeptides (including enzymes and other proteins) or fragments thereof passing through the detection module. Alternatively, the device can determine the presence or degree of some other characteristic indicated by a reporter. If desired, the cells, particles or molecules can be sorted based on this analysis.
  • the sorted cells, particles or molecules can be collected from the outlet channels in collection modules (or discarded in wasted modules) and used as needed.
  • the collected cells, particles or molecules can be removed from the device or reintroduced to the device for additional coalescence, analysis and sorting.
  • a “processor” or a “microprocessor” is any component or device able to receive a signal from one or more sensors, store the signal, and/or direct one or more responses (e.g., as described above), for example, by using a mathematical formula or an electronic or computational circuit.
  • the signal may be any suitable signal indicative of the environmental factor determined by the sensor, for example a pneumatic signal, an electronic signal, an optical signal, a mechanical signal, etc.
  • the device of the present invention can comprise features, such as integrated metal alloy components and/or features patterned in an electrically conductive layer, for detecting droplets by broadcasting a signal around a droplet and picking up an electrical signal in proximity to the droplet.
  • the droplet content detection can also be achieved by simultaneous detection of contents of multiple droplets in parallel using spectroscopic fluorescence imaging with sensitivity as high as single-molecule limit.
  • One can spatially distribute droplets containing fluorescent entities such as fluorophore biological markers and/or quantum dots in a two-dimensional sheet in a microscopic field-of-view.
  • the filed-of-view of those droplets can then be illuminated by a fluorescence excitation source and the resulting fluorescence can be spectroscopically imaged.
  • the throughput of fluorescence detection compared to a single-drop fluorescence detection method can be increased by a factor of a/b for a given sensitivity, where a is the number of droplets that can be imaged within a given field-of-view, and b is the ratio of the fluorescence sensitivity of a single-drop fluorescence detector compared to that of the multiple drop fluorescence detector.
  • a is the number of droplets that can be imaged within a given field-of-view
  • b is the ratio of the fluorescence sensitivity of a single-drop fluorescence detector compared to that of the multiple drop fluorescence detector.
  • the residence time of the droplet in the field-of-view can be unlimited, thereby allowing sensitivity as high as the single-molecule limit.
  • the device of the present invention also comprises the use of beads and methods for analyzing and sorting beads (i.e, bead reader device).
  • the device can read and either sort or not sort droplets containing one or more of a set of two or more beads. Each bead can be differentiated from each other bead within a set. Beads can be separated by several tags including, but not limited to, quantum dyes, fluorescent dyes, ratios of fluorescent dyes, radioactivity, radio-tags, etc.
  • a set of beads containing a ratio of two dyes in discrete amounts with an apparatus for detecting and differentiating beads containing one discrete ratio from the other beads in this set having a different ratio of the two dyes.
  • the microfluidic device can include paramagnetic beads. The paramagnetic beads can introduce and remove chemical components from droplets using droplet coalescence and breakup events. The paramagnetic beads can also be used for sorting droplets.
  • the present invention provides methods of screening molecular libraries on beads through limited-dilusion-loading and then chemical or optical release inside of droplets.
  • a releasing means chemical, UV light, heat, etc
  • tea-bag synthesis of chemicals on a bead simultaneously with a means for identifying said bead (using, for example, a mass spec tag).
  • a means for identifying said bead using, for example, a mass spec tag.
  • the device of the present invention can comprise column separation prior to bead sorting.
  • Such separating means could include size, charge, hydrophobicity, atomic mass, etc.
  • the separating can be done isocratic or by use of a means for generating a gradient chemically, (for example using salt or hydrophobicity), electrically, by pressure, or etc.
  • a channel is preloaded with Sepharose size exclusion media.
  • a sample is loaded at one end, and the droplets are formed at an opposing end. The sample separates by size prior to becoming incorporated within a droplet.
  • the microfluidic device of the present invention can further include one or more sorting modules.
  • a “sorting module ” is a junction of a channel where the flow of molecules, cells, small molecules or particles can change direction to enter one or more other channels, e.g., a branch channel for delivery to an outlet module (i.e., collection or waste module), depending on a signal received in connection with an examination in the detection module.
  • a sorting module is monitored and/or under the control of a detection module, and therefore a sorting module may “correspond” to such detection module.
  • the sorting region is in communication with and is influenced by one or more sorting apparatuses.
  • a sorting apparatus comprises techniques or control systems, e.g., dielectric, electric, electro-osmotic, (micro-) valve, etc.
  • a control system can employ a variety of sorting techniques to change or direct the flow of molecules, cells, small molecules or particles into a predetermined branch channel.
  • a “branch channel” is a channel which is in communication with a sorting region and a main channel. The main channel can communicate with two or more branch channels at the sorting module or “branch point”, forming, for example, a T-shape or a Y-shape. Other shapes and channel geometries may be used as desired.
  • a branch channel receives molecules, cells, small molecules or particles depending on the molecule, cells, small molecules or particles characteristic of interest as detected by the detection module and sorted at the sorting module.
  • a branch channel can have an outlet module and/or terminate with a well or reservoir to allow collection or disposal (collection module or waste module, respectively) of the molecules, cells, small molecules or particles.
  • a branch channel may be in communication with other channels to permit additional sorting.
  • the device of the present invention can further include one or more outlet modules.
  • An “outlet module” is an area of the device that collects or dispenses molecules, cells, small molecules or particles after coalescence, detection and/or sorting.
  • the outlet module can include a collection module and/or a waste module.
  • the collection module can be connected to a means for storing a sample.
  • the collection module can be a well or reservoir for collecting and containing droplets detected to have a specific predetermined characteristic in the detection module.
  • the collection module can be temperature controlled.
  • the waste module can be connected to a means for discarding a sample.
  • the waste module can be a well or reservoir for collecting and containing droplets detected to not have a specific predetermined characteristic in the detection module.
  • the outlet module is downstream from a sorting module, if present, or downstream from the detection module if a sorting module is not present.
  • the outlet module may contain branch channels or outlet channels for connection to a collection module or waste module.
  • a device can contain more than one outlet module.
  • a characteristic of a fluidic droplet may be sensed and/or determined in some fashion, for example, as described herein (e.g., fluorescence of the fluidic droplet may be determined), and, in response, an electric field may be applied or removed from the fluidic droplet to direct the fluidic droplet to a particular region (e.g. a channel).
  • a fluidic droplet is preferably sorted or steered by inducing a dipole in the uncharged fluidic droplet (which may be initially charged or uncharged), and sorting or steering the droplet using an applied electric field.
  • the electric field may be an AC field, a DC field, etc.
  • a channel 540 containing fluidic droplet 530 and liquid 535 , divides into channel 542 and 544 .
  • Fluidic droplet 530 is uncharged.
  • Electrode 526 is positioned near channel 542
  • electrode 527 is positioned near channel 544 .
  • Electrode 528 is positioned near the junction of channels 540 , 542 , and 544 .
  • a dipole is induced in the fluidic droplet using electrodes 526 , 527 , and/or 528 .
  • a dipole is induced in droplet 530 by applying an electric field 525 to the droplet using electrodes 527 and 528 .
  • droplet 530 Due to the strength of the electric field, the droplet is strongly attracted to the right, into channel 544 . Similarly, in FIG. 20D , a dipole is induced in droplet 530 by applying an electric field 525 to the droplet using electrodes 526 and 528 , causing the droplet to be attracted into channel 542 . Thus, by applying the proper electric field, droplet 530 can be directed to either channel 542 or 544 as desired.
  • the present invention also provides improvements in the efficiency, accuracy, and reliability of the preferred dielectric droplet sorting technique described above.
  • the single sided dielectric sorting relies on a combination of flow imbalance between the two exhaust legs and a switchable electric field to selectively sort out droplets of interest from the main sample stream. Sorting decisions are made based on some form of real time measurement of the droplet and its contents.
  • FIGS. 21 and 22 depict many of the various possible fluid and electrode geometries possible for single sided dielectric sorting.
  • FIG. 21 Panels A-D show possible flow channel geometries that can be used in an asymmetric sorting application.
  • Panel F illustrates the use of a barrier, for example, a barrier where no fluid flow passes on its left side. Note these designs are only conceptual representation of the fluid channels, and actual designs may differ in absolute and relative dimensions as determined by one of ordinary skill in the art.
  • FIG. 22 shows the possible electrode geometries used in an asymmetric sorting application.
  • Panel A shows the use of sharp tipped electrodes.
  • Panel B shows broad tipped electrodes to increase the interaction time between the droplets and the electric field (the tips could be many drop diameters long).
  • Panel C shows electrodes straddling the collection line.
  • Panel D shows electrodes on opposite sides of the main channel.
  • Panel E shows an Asymmetric Electrode Pair (the asymmetry may be present on any of the other electrode pair layouts as well). Note these designs are only conceptual representation of the electrodes, and actual designs may differ in absolute dimensions and electrode shape as determined by one of ordinary skill in the art.
  • the fluid channel geometry is drawn as a “Y” junction, any of the channel geometries shown in FIG. 21 could be substituted in these drawings.
  • the flow rate of the collection leg is set to a value just below the level required to begin pulling droplets into the collection line (indicated as 40% in the figures, although the actual value may be differ from this and is dependent on the actual fluidic and electrode geometry, total flow, as well as droplet size and composition).
  • the collection leg can be operated at a flow rate at which the droplets would normally flow down the Sort collect line (i.e. change the flow splits shown in the diagrams from 40% collect/60% waste to 60% collect/40% waste), and keep the electric field energized until a droplet of interest is detected. At that time, the field would be briefly turned off, and the droplet would be pulled down the collection leg based on fluidic forces instead of electrical forces.
  • a fluidic droplet may be directed by creating an electric charge (e.g., as previously described) on the droplet, and steering the droplet using an applied electric field, which may be an AC field, a DC field, etc.
  • an electric field maybe selectively applied and removed (or a different electric field may be applied) as needed to direct the fluidic droplet to a particular region.
  • the electric field may be selectively applied and removed as needed, in some embodiments, without substantially altering the flow of the liquid containing the fluidic droplet.
  • a liquid may flow on a substantially steady-state basis (i.e., the average flowrate of the liquid containing the fluidic droplet deviates by less than 20% or less than 15% of the steady-state flow or the expected value of the flow of liquid with respect to time, and in some cases, the average flowrate may deviate less than 10% or less than 5%) or other predetermined basis through a fluidic system of the invention (e.g., through a channel or a microchannel), and fluidic droplets contained within the liquid may be directed to various regions, e.g., using an electric field, without substantially altering the flow of the liquid through the fluidic system.
  • a substantially steady-state basis i.e., the average flowrate of the liquid containing the fluidic droplet deviates by less than 20% or less than 15% of the steady-state flow or the expected value of the flow of liquid with respect to time, and in some cases, the average flowrate may deviate less than 10% or less than 5%
  • a fluidic system of the invention e.g.
  • the fluidic droplets may be screened or sorted within a fluidic system of the invention by altering the flow of the liquid containing the droplets. For instance, in one set of embodiments, a fluidic droplet may be steered or sorted by directing the liquid surrounding the fluidic droplet into a first channel, a second channel, etc.
  • pressure within a fluidic system can be controlled to direct the flow of fluidic droplets.
  • a droplet can be directed toward a channel junction including multiple options for further direction of flow (e.g., directed toward a branch, or fork, in a channel defining optional downstream flow channels).
  • Pressure within one or more of the optional downstream flow channels can be controlled to direct the droplet selectively into one of the channels, and changes in pressure can be effected on the order of the time required for successive droplets to reach the junction, such that the downstream flow path of each successive droplet can be independently controlled.
  • the expansion and/or contraction of liquid reservoirs may be used to steer or sort a fluidic droplet into a channel, e.g., by causing directed movement of the liquid containing the fluidic droplet.
  • the liquid reservoirs may be positioned such that, when activated, the movement of liquid caused by the activated reservoirs causes the liquid to flow in a preferred direction, carrying the fluidic droplet in that preferred direction.
  • the expansion of a liquid reservoir may cause a flow of liquid towards the reservoir, while the contraction of a liquid reservoir may cause a flow of liquid away from the reservoir.
  • the expansion and/or contraction of the liquid reservoir may be combined with other flow-controlling devices and methods, e.g., as described herein.
  • Non-limiting examples of devices able to cause the expansion and/or contraction of a liquid reservoir include pistons and piezoelectric components.
  • piezoelectric components may be particularly useful due to their relatively rapid response times, e.g., in response to an electrical signal.
  • the fluidic droplets may be sorted into more than two channels. Alternately, a fluidic droplet may be sorted and/or split into two or more separate droplets, for example, depending on the particular application. Any of the above-described techniques may be used to spilt and/or sort droplets.
  • a fluidic droplet may be directed to a first region or channel; by applying (or removing) a second electric field to the device (or a portion thereof), the droplet may be directed to a second region or channel; by applying a third electric field to the device (or a portion thereof), the droplet may be directed to a third region or channel; etc., where the electric fields may differ in some way, for example, in intensity, direction, frequency, duration, etc.
  • each droplet may be independently sorted and/or split; for example, some droplets may be directed to one location or another, while other droplets may be split into multiple droplets directed to two or more locations.
  • At least about 1 droplet per second may be determined and/or sorted in some cases, and in other cases, at least about 10 droplets per second, at least about 20 droplets per second, at least about 30 droplets per second, at least about 100 droplets per second, at least about 200 droplets per second, at least about 300 droplets per second, at least about 500 droplets per second, at least about 750 droplets per second, at least about 1000 droplets per second, at least about 1500 droplets per second, at least about 2000 droplets per second, at least about 3000 droplets per second, at least about 5000 droplets per second, at least about 7500 droplets per second, at least about 10,000 droplets per second, at least about 15,000 droplets per second, at least about 20,000 droplets per second, at least about 30,000 droplets per second, at least about 50,000 droplets per second, at least about 75,000 droplets per second, at least about 100,000 droplets per second, at least about 150,000 droplets per second,
  • the present invention provides a device comprising multiple channels with the appropriate geometry to split droplets, perform different experiments on the two daughter droplets and then reorder so that they pass sequential through the detector. The sums, ratios or differences in the two signals can then be calculated before the droplets enter the sorting bifurcation.
  • An indicator dye or equivalent material may be added to one or both droplets to indicate when each droplet enters and leaves the laser.
  • a representative sketch is shown in FIG. 23 .
  • the present invention proposes methods for recovering aqueous phase components from aqueous emulsions that have been collected on a microfluidic device in a minimum number of steps and in a gentle manner so as to minimize potential damage to cell viability.
  • a stable aqueous sample droplet emulsion containing aqueous phase components in a continuous phase carrier fluid is allowed to cream to the top of the continuous phase carrier oil.
  • the continuous phase carrier fluid can include a perfluorocarbon oil that can have one or more stabilizing surfactants.
  • the aqueous emulsion rises to the top or separates from the continuous phase carrier fluid by virtue of the density of the continuous phase fluid being greater than that of the aqueous phase emulsion.
  • the perfluorocarbon oil used in one embodiment of the device is 1.8, compared to the density of the aqueous emulsion, which is 1.0.
  • the creamed emulsion is then placed onto a second continuous phase carrier fluid which contains a de-stabilizing surfactant, such as a perfluorinated alchohol (e.g. 1H,1H,2H,2H-Perfluoro-1-octanol).
  • a de-stabilizing surfactant such as a perfluorinated alchohol (e.g. 1H,1H,2H,2H-Perfluoro-1-octanol).
  • the second continuous phase carrier fluid can also be a perfluorocarbon oil.
  • Additional destabilizing surfactants and/or oil combinations can be identified or synthesized to be useful with this invention.
  • the microfluidic device of the present invention can further include one or more mixing modules.
  • coalescence of one or more droplets in one or more coalescence modules can be sufficient to mix the contents of the coalesced droplets (e.g., through rotating vortexes existing within the droplet), it should be noted that when two droplets fuse or coalesce, perfect mixing within the droplet does not instantaneously occur.
  • the coalesced droplet may initially be formed of a first fluid region (from the first droplet) and a second fluid region (from the second droplet).
  • the fluid regions may remain as separate regions, for example, due to internal “counter-revolutionary” flow within the fluidic droplet, thus resulting in a non-uniform fluidic droplet.
  • a “mixing module” can comprise features for shaking or otherwise manipulate droplets so as to mix their contents.
  • the mixing module is preferably downstream from the coalescing module and upstream from the detection module.
  • the mixing module can include, but is not limited to, the use of channel geometries, acoustic actuators, metal alloy component electrodes or electrically conductive patterned electrodes to mix the contents of droplets and to reduce mixing times for fluids combined into a single droplet in the microfluidic device.
  • the fluidic droplet may be passed through one or more channels or other systems which cause the droplet to change its velocity and/or direction of movement. The change of direction may alter convection patterns within the droplet, causing the fluids to be at least partially mixed. Combinations are also possible.
  • the frequency of the acoustic wave should be fine tuned so as not to cause any damage to the cells.
  • the biological effects of acoustic mixing have been well studied (e.g., in the ink-jet industry) and many published literatures also showed that piezoelectric microfluidic device can deliver intact biological payloads such as live microorganisms and DNA.
  • the design of the acoustic resonant uses a Piezoelectric bimorph flat plate located on the side of the carved resonant in the PDMS slab.
  • the piezoelectric driving waveform is carefully optimized to select the critical frequencies that can separate cells in fluids. There are five parameters to optimize beyond the frequency parameter. Lab electronics is used to optimize the piezoelectric driving waveform. Afterwards, a low cost circuit can be designed to generate only the optimized waveform in a preferred microfluidic device.
  • the microfluidic device of the present invention can further include one or more delay modules.
  • the “delay module” can be a delay line.
  • the operation of a microfluidics device where a reaction within a droplet is allowed to occur for a non-trivial length of time requires a delay line to increase the residence time within the device. For reactions demanding extensive residence time, longer or larger delay lines are required. Accordingly, the invention provides methods to increase residence times within microfluidic devices.
  • the delay module is in fluid communication with the main channel or it can be an elongated portion of the main channel itself.
  • the delay module can be located downstream of the coalescence module and upstream of the detection module.
  • the delay module can be a serpentine channel or a buoyant hourglass.
  • the delay module can further comprise heating and cooling regions. The heating and cooling regions can be used for performing on-chip, flow-through PCR as further described herein.
  • the channel dimensions and configurations can be designed to accommodate the required residence time with minimum pressure drops across the device.
  • the device can comprise a multilayered PDMS slab which is composed of several patterned PDMS slabs.
  • the channel dimensions can also be designed so as to allow for required flow, residence time and pressure drop. Some channels may be required to be very large in width and height.
  • the device includes support posts within the channel design. In order to reduce dead volume behind posts and further improve droplet stability, the support posts are designed to optimize a streamlined flow within the channel. These designs can include curved features as opposed to sharp edges.
  • delay lines can also be extended to the outside of the chip.
  • the off-chip delay lines can be tubes within micron-sized internal diameter.
  • the delay lines can be in the form of a tower (i.e., a structure which is vertical with respect to the ambient gravitational field) as to allow buoyant forces to assist controlled droplet transport.
  • Known delay lines involve transporting droplets by emulsifying them in a carrier fluid flowing in a channel and/or tube. Because the velocity profile of the carrier fluid through the cross-section of the channel and/or tube is not uniform, the velocity distribution of the droplets will not be narrow, which causes the delay time distribution of the droplets to not be narrow (i.e., some droplets will be delayed more or less than others).
  • the devices of the present invention can also include buoyancy-assisted microfluidic delay lines.
  • buoyancy-assisted microfluidic delay lines buoyant forces act on droplets emulsified in a fluid in one or more towers. This can include allowing the tower to fill for the desired delay time, and then releasing the droplets.
  • the tower can or can not continue to fill and release droplets as needed.
  • Droplets that have a density less than their carrier fluid are fed into the base of the tower, buoyantly rise to the top of the tower with a substantially uniform velocity distribution, and are funneled into a functional component of the microfluidic device (such as a y-branch).
  • Carrier fluid is exhausted at the base of the tower at the same rate as it is introduced at the apex so that the net flow of carrier fluid through the delay line is zero.
  • the tower and funnel sections can have any cross-sectional shape, such as circular, elliptical, or polygonal.
  • the microfluidic device can include a tower with adjustable length.
  • the capacity of each tower is 0.05*T, where T is the delay time.
  • the concept includes, for example: (a) upon device start-up, filling the first tower for 0.05*T, but stop-cock its exhaust, and also have the other nineteen towers closed; (b) after 0.05*T, closing the first tower and filling the second between 0.05*T and 0.1 0*T; (c) repeating step (b) for the remaining eighteen towers; (d) at time T, allowing the first tower to exhaust; (e) at time 1.05*T, stop-cocking the exhaust of the first tower, allowing the second tower to exhaust, and allowing the first tower to fill; (f) at time 1.10*T, stop-cocking the exhaust of the second tower, allowing the third tower to exhaust, closing the first tower, and allowing the second tower to fill, and (g) repeating step (f)
  • the delay module can also include channels (e.g. the main channel) which has an altered geometry which permits the “parking” (e.g., slowing or stopping) of droplets within the microfluidic device.
  • droplets are able to be parked in wells or channels at predefined locations. This can be done by creating discrete well-like indentions in the channel whereby a droplet ‘falls’ into the well and remains there as the fluid flows over it, or by using a technique entitled ‘by-pass pots’ whereby a droplet is used to block a small outlet in a well, thereby causing the flow to by-pass that droplet-containing well.
  • the instant invention is to use either of these techniques or any related technique, for example just stopping the drops in a channel, to position droplets at either random or predefined places within a microfluidics device. These random or predefined locations can then be queried at a later time-point for a reaction to have occurred, or for removal of the droplets using another means such as resuspension followed by aspiration.
  • a rolling circle amplification reaction is initiated in droplets, the droplets are then parked within the chip, and the amplification reaction allowed to proceed for a set period of time prior to stopping the reaction through the use of heat.
  • the parked droplets are then dried in situ and the covering of the chip disassembled from the chip.
  • One or a set of needle-like devices that are able to be lined up with the droplet parking space are then placed adjacent to or on top of the dried droplets and a liquid solution used to resuspend the material in the dried droplet that has been deposited into the chip, for further downstream processing.
  • the first reactions are created in 10 ⁇ m droplets, the droplets are dried within a channel parking space or by-pass pot which is able to hold a droplet of size larger than 10 ⁇ m, and the droplets are dried in situ.
  • a second set of droplets that are larger than 10 ⁇ m are then allowed to proceed down said channel and when caught in said parking space or by-pass pot are able to resuspend the material from the first droplets that are dried along the walls of the first parking space or by-pass pot.
  • the second droplet is slightly larger than the first and that ensures that the material along the walls is ‘captured’ by the second droplet, and not allowed to diffuse away from the first droplet wall by diffusion.
  • use of surfactants becomes optional in either the first or second droplet formulations.
  • the instant invention also provides the following devices and methods for use in practicing the methods described herein.
  • the PDMS substrate which comprises a portion of the microfluidic device can be covered or coated with an adhesive tape or strip that can removed by peeling.
  • the PDMS substrate can also be bonded by an ultra thin silica that can be pierced by a set of needles.
  • the silica may be spin coated or electro-plated onto a thin backing.
  • Droplets can be dried onto a piece of paper such that can be detected by a second device to determine the Ncode within the droplet and to determine whether an amplification reaction has occurred within the droplet.
  • a plate read comprising dried and undried spots using either an optical array device, such as found in high-end cameras or fiber, optic device is also contemplated.
  • Dry Nitrogen can be utilized to dry the spots by either flowing it through the channel or placing the device into a dry-N2 chamber.
  • Channels can be filled with dried nitrogen or salt run underneath or adjacent to the parking space channels to allow chemical or physical-type gradients to be set up in the chip.
  • the channel walls can be coated with Steptavidin and the produced reactants, for example, DNA biotinylated so that it adheres in situ.
  • Porous beads deposited into the wells can be used in combination with solutions without oils to wash the beads by flow, followed by re-depositing droplets with surfactants to recoat the beads.
  • the wells within the substrate can be filled with many small beads by loading small beads into droplets, storing the droplets into individual wells containing apertures that are slightly smaller than the beads, breaking the droplets by drying or flow of aqueous solutions with or without surfactants into the channels and past the beads, and then re-encapsulating the beads in situ.
  • a set of electrodes within or adjacent to the microfluidic substrate can be used to fuse two droplets in a storage/holding space. The electrodes may be perpendicular to the plane of the channels and either the electrodes or channels moved so as to allow droplet fusions to occur.
  • the microfluidic device of the present invention can further include one or more UV-release modules.
  • the “UV-release module” is in fluid communication with the main channel.
  • the UV-release module is located downstream of the inlet module and upstream of the coalescence module.
  • the UV-module can be a used in bead assays. Compounds from encapsulated beads can be cleaved in a UV-releasing module using UV light. Photolabile linkers can be broken down on demand after a single bead has been encapsulated thus releasing multiple copies of a single compound into solution. In the cell based assay disclosed herein the chemical compound assayed is desired to be in solution in order to penetrate the cell membrane.
  • Photocleavable linkers can be utilized to cleave the compounds of the bead after drop formation by passing the drop through a UV-release module (i.e., laser of the appropriate wavelength).
  • the present invention also provides methods for chemical synthesis on a bead and releasing said chemical attached to the bead using a releasing means (chemical, UV light, heat, etc) within a droplet, and then combining a second droplet to the first droplet for further manipulation.
  • a releasing means chemical, UV light, heat, etc
  • the releasing means is a UV-module.
  • tea-bag synthesis of chemicals on a bead simultaneously with a means for identifying said bead (using, for example, a mass spec tag).
  • a means for identifying said bead using, for example, a mass spec tag.
  • kits also typically include instructions for carrying out the subject assay, and may optionally include the fluid receptacle, e.g., the cuvette, multiwell plate, microfluidic device, etc. in which the reaction is to be carried out.
  • the fluid receptacle e.g., the cuvette, multiwell plate, microfluidic device, etc. in which the reaction is to be carried out.
  • reagents included within the kit are uniquely labeled emulsions containing tissues, cells, particles, proteins, antibodies, amino acids, nucleotides, small molecules, substrates, and/or pharmaceuticals.
  • These reagents may be provided in pre-measured container (e.g., vials or ampoules) which are co-packaged in a single box, pouch or the like that is ready for use.
  • the container holding the reagents can be configured so as to readily attach to the fluid receptacle of the device in which the reaction is to be carried out (e.g., the inlet module of the microfluidic device as described herein).
  • the kit can include an RNAi kit.
  • the kit can include a chemical synthesis kit. It will be appreciated by persons of ordinary skill in the art that these embodiments are merely illustrative and that other kits are also within the scope of the present invention.
  • the microfluidic device of the present invention can be utilized to conduct numerous chemical and biological assays, including but not limited to, creating emulsion libraries, flow cytometry, gene amplification, isothermal gene amplification, DNA sequencing, SNP analysis, drug screening, RNAi analysis, karyotyping, creating microbial strains with improved biomass conversion, moving cells using optical tweezer/cell trapping, transformation of cells by electroporation, ⁇ TAS, and DNA hybridization.
  • chemical and biological assays including but not limited to, creating emulsion libraries, flow cytometry, gene amplification, isothermal gene amplification, DNA sequencing, SNP analysis, drug screening, RNAi analysis, karyotyping, creating microbial strains with improved biomass conversion, moving cells using optical tweezer/cell trapping, transformation of cells by electroporation, ⁇ TAS, and DNA hybridization.
  • “about” or “approximately” shall generally mean within 20 percent, preferably within 10 percent, and more preferably within 5 percent of a given value or range.
  • molecule means any distinct or distinguishable structural unit of matter comprising one or more atoms, and includes for example polypeptides and polynucleotides.
  • polymer means any substance or compound that is composed of two or more building blocks (‘mers’) that are repetitively linked to each other.
  • a “dimer” is a compound in which two building blocks have been joined together.
  • polynucleotide refers to a polymeric molecule having a backbone that supports bases capable of hydrogen bonding to typical polynucleotides, where the polymer backbone presents the bases in a manner to permit such hydrogen bonding in a sequence specific fashion between the polymeric molecule and a typical polynucleotide (e.g., single-stranded DNA).
  • bases are typically inosine, adenosine, guanosine, cytosine, uracil and thymidine.
  • Polymeric molecules include double and single stranded RNA and DNA, and backbone modifications thereof, for example, methylphosphonate linkages.
  • nucleotide sequence is a series of nucleotide bases (also called “nucleotides”) generally in DNA and RNA, and means any chain of two or more nucleotides.
  • a nucleotide sequence typically carries genetic information, including the information used by cellular machinery to make proteins and enzymes. These terms include double or single stranded genomic and cDNA, RNA, any synthetic and genetically manipulated polynucleotide, and both sense and anti-sense polynucleotide (although only sense stands are being represented herein).
  • PNA protein nucleic acids
  • the polynucleotides herein may be flanked by natural regulatory sequences, or may be associated with heterologous sequences, including promoters, enhancers, response elements, signal sequences, polyadenylation sequences, introns, 5′- and 3′-non-coding regions, and the like.
  • the nucleic acids may also be modified by many means known in the art.
  • Non-limiting examples of such modifications include methylation, “caps”, substitution of one or more of the naturally occurring nucleotides with an analog, and internucleotide modifications such as, for example, those with uncharged linkages (e.g., methyl phosphonates, phosphotriesters, phosphoroamidates, carbamates, etc.) and with charged linkages (e.g., phosphorothioates, phosphorodithioates, etc.).
  • uncharged linkages e.g., methyl phosphonates, phosphotriesters, phosphoroamidates, carbamates, etc.
  • charged linkages e.g., phosphorothioates, phosphorodithioates, etc.
  • Polynucleotides may contain one or more additional covalently linked moieties, such as, for example, proteins (e.g., nucleases, toxins, antibodies, signal peptides, poly-L-lysine, etc.), intercalators (e.g., acridine, psoralen, etc.), chelators (e.g., metals, radioactive metals, iron, oxidative metals, etc.), and alkylators.
  • the polynucleotides may be derivatized by formation of a methyl or ethyl phosphotriester or an alkyl phosphoramidate linkage.
  • the polynucleotides herein may also be modified with a label capable of providing a detectable signal, either directly or indirectly. Exemplary labels include radioisotopes, fluorescent molecules, biotin, and the like.
  • dielectrophoretic force gradient means a dielectrophoretic force is exerted on an object in an electric field provided that the object has a different dielectric constant than the surrounding media. This force can either pull the object into the region of larger field or push it out of the region of larger field. The force is attractive or repulsive depending respectively on whether the object or the surrounding media has the larger dielectric constant.
  • DNA deoxyribonucleic acid
  • A adenine
  • G guanine
  • C cytosine
  • T thymine
  • DNA can have one strand of nucleotide bases, or two complimentary strands which may form a double helix structure.
  • RNA ribonucleic acid
  • A adenine
  • G guanine
  • C cytosine
  • U uracil
  • RNA typically has one strand of nucleotide bases.
  • a “polypeptide” (one or more peptides) is a chain of chemical building blocks called amino acids that are linked together by chemical bonds called peptide bonds.
  • a “protein” is a polypeptide produced by a living organism.
  • a protein or polypeptide may be “native” or “wild-type”, meaning that it occurs in nature; or it may be a “mutant”, “variant” or “modified”, meaning that it has been made, altered, derived, or is in some way different or changed from a native protein, or from another mutant.
  • an “enzyme” is a polypeptide molecule, usually a protein produced by a living organism, that catalyzes chemical reactions of other substances. The enzyme is not itself altered or destroyed upon completion of the reaction, and can therefore be used repeatedly to catalyze reactions.
  • a “substrate” refers to any substance upon which an enzyme acts.
  • particles means any substance that may be encapsulated within a droplet for analysis, reaction, sorting, or any operation according to the invention.
  • Particles are not only objects such as microscopic beads (e.g., chromatographic and fluorescent beads), latex, glass, silica or paramagnetic beads, but also includes other encapsulating porous and/or biomaterials such as liposomes, vesicles and other emulsions. Beads ranging in size from 0.1 micron to 1 mm can be used in the devices and methods of the invention and are therefore encompassed with the term “particle” as used herein.
  • the term particle also encompasses biological cells, as well as beads and other microscopic objects of similar size (e.g., from about 0.1 to 120 microns, and typically from about 1 to 50 microns) or smaller (e.g., from about 0.1 to 150 nm).
  • the devices and methods of the invention are also directed to sorting and/or analyzing molecules of any kind, including polynucleotides, polypeptides and proteins (including enzymes) and their substrates and small molecules (organic or inorganic).
  • the term particle further encompasses these materials.
  • the particles are sorted and/or analyzed by encapsulating the particles into individual droplets (e.g., droplets of aqueous solution in oil), and these droplets are then sorted, combined and/or analyzed in a microfabricated device.
  • droplets generally includes anything that is or can be contained within a droplet.
  • a “small molecule” as used herein, is meant to refer to a composition that has a molecular weight of less than about 5 kD and most preferably less than about 4 kD.
  • Small molecules can be, e.g., nucleic acids, peptides, polypeptides, peptidomimetics, carbohydrates, lipids or other organic or inorganic molecules. Libraries of chemical and/or biological mixtures, such as fungal, bacterial, or algal extracts, are known in the art.
  • cell means any cell or cells, as well as viruses or any other particles having a microscopic size, e.g. a size that is similar to or smaller than that of a biological cell, and includes any prokaryotic or eukaryotic cell, e.g., bacteria, fungi, plant and animal cells.
  • Cells are typically spherical, but can also be elongated, flattened, deformable and asymmetrical, i.e., non-spherical.
  • the size or diameter of a cell typically ranges from about 0.1 to 120 microns, and typically is from about 1 to 50 microns.
  • a cell may be living or dead.
  • the microfabricated device of the invention is directed to sorting materials having a size similar to a biological cell (e.g. about 0.1 to 120 microns) or smaller (e.g., about 0.1 to 150 nm) any material having a size similar to or smaller than a biological cell can be characterized and sorted using the microfabricated device of the invention.
  • the term cell shall further include microscopic beads (such as chromatographic and fluorescent beads), liposomes, emulsions, or any other encapsulating biomaterials and porous materials.
  • Non-limiting examples include latex, glass, orparamagnetic beads; and vesicles such as emulsions and liposomes, and other porous materials such as silica beads.
  • Beads ranging in size from 0.1 micron to 1 mm can also be used, for example in sorting a library of compounds produced by combinatorial chemistry.
  • a cell may be charged or uncharged.
  • charged beads may be used to facilitate flow or detection, or as a reporter.
  • Biological cells, living or dead may be charged for example by using a surfactant, such as SDS (sodium dodecyl sulfate).
  • SDS sodium dodecyl sulfate
  • the term cell further encompasses “virions”, whether or not virions are expressly mentioned.
  • a “virion”, “virus particle” is the complete particle of a virus.
  • Viruses typically comprise a nucleic acid core (comprising DNA or RNA) and, in certain viruses, a protein coat or “capsid”. Certain viruses may have an outer protein covering called an “envelope”.
  • a virion may be either living (i.e., “viable”) or dead (i.e., “non-viable”).
  • a living or “viable” virus is one capable of infecting a living cell.
  • Viruses are generally smaller than biological cells and typically range in size from about 20-25 nm diameter or less (parvoviridae, picomoviridae) to approximately 200-450 nm (poxviridae).
  • filamentous viruses may reach lengths of 2000 nm (closterviruses) and are therefore larger than some bacterial cells.
  • the microfabricated device of the invention is particularly suited for sorting materials having a size similar to a virus (i.e., about 0.1 to 150 nm)
  • any material having a size similar to a virion can be characterized and sorted using the microfabricated device of the invention.
  • Non-limiting examples include latex, glass or paramagnetic beads; vesicles such as emulsions and liposomes; and other porous materials such as silica beads. Beads ranging in size from 0.1 to 150 nm can also be used, for example, in sorting a library of compounds produced by combinatorial chemistry.
  • a virion may be charged or uncharged.
  • charged beads may be used to facilitate flow or detection, or as a reporter.
  • Biological viruses whether viable or non-viable, may be charged, for example, by using a surfactant, such as SDS.
  • a “reporter” is any molecule, or a portion thereof, that is detectable, or measurable, for example, by optical detection.
  • the reporter associates with a molecule, cell or virion or with a particular marker or characteristic of the molecule, cell or virion, or is itself detectable to permit identification of the molecule, cell or virion's, or the presence or absence of a characteristic of the molecule, cell or virion.
  • characteristics include size, molecular weight, the presence or absence of particular constituents or moieties (such as particular nucleotide sequences or restrictions sites).
  • reporter In the case of cells, characteristics which may be marked by a reporter includes antibodies, proteins and sugar moieties, receptors, polynucleotides, and fragments thereof.
  • label can be used interchangeably with “reporter”.
  • the reporter is typically a dye, fluorescent, ultraviolet, or chemiluminescent agent, chromophore, or radio-label, any of which may be detected with or without some kind of stimulatory event, e.g., fluoresce with or without a reagent.
  • the reporter is a protein that is optically detectable without a device, e.g. a laser, to stimulate the reporter, such as horseradish peroxidase (HRP).
  • HRP horseradish peroxidase
  • a protein reporter can be expressed in the cell that is to be detected, and such expression may be indicative of the presence of the protein or it can indicate the presence of another protein that may or may not be coexpressed with the reporter.
  • a reporter may also include any substance on or in a cell that causes a detectable reaction, for example by acting as a starting material, reactant or a catalyst for a reaction which produces a detectable product. Cells may be sorted, for example, based on the presence of the substance, or on the ability of the cell to produce the detectable product when the reporter substance is provided.
  • a “marker” is a characteristic of a molecule, cell or virion that is detectable or is made detectable by a reporter, or which may be coexpressed with a reporter.
  • a marker can be particular constituents or moieties, such as restrictions sites or particular nucleic acid sequences in the case of polynucleotides.
  • characteristics may include a protein, including enzyme, receptor and ligand proteins, saccharrides, polynucleotides, and combinations thereof, or any biological material associated with a cell or virion.
  • the product of an enzymatic reaction may also be used as a marker.
  • the marker may be directly or indirectly associated with the reporter or can itself be a reporter.
  • a marker is generally a distinguishing feature of a molecule, cell or virion
  • a reporter is generally an agent which directly or indirectly identifies or permits measurement of a marker.
  • the present invention provides methods for preparing a library of droplet emulsions, where each of the droplets is of the same, predetermined size (monodisperse). Further, present invention provides methods for deterministic lateral displacement for continuous particle separation, which can occur within droplets on a microfluidic device.
  • Particles in solution are usually separated according to size by exclusion or hydrodynamic chromatography.
  • a sample mixture is injected at one end of a tube packed with porous beads and then washed through the tube. Particles smaller than the pore sizes enter the beads, which lengthen their migration path, and so they are on average eluted later than larger particles. Zones of particles of a given size broaden, however, because particles in each zone take many different paths, leading to different retention times. This multipath effect reduces the resolution of size-exclusion chromatography.
  • hydrodynamic chromatography a sample mixture is driven through a capillary by hydrodynamic flow, which has a parabolic flow profile. Large particles cannot intercept the low-velocity fluid near the capillary wall, and thus on average move faster and become separated from small particles. Multipath effects also limit the resolution of hydrodynamic chromatography, because each migration path samples different velocities in the parabolic flow.
  • Lateral displacement means for sizing and separating droplets in solution can be utilized.
  • the present invention relates to the generation of a microfluidic device consisting of raised pillars in both columns and rows that are designed for lateral diffusion.
  • the pillars can be adjusted so as to be a means for separating droplets of similar sizes from a fluid containing various sized droplets.
  • a fluid containing oil, water and a surfactant is mixed so as to create a bulk emulsion.
  • the bulk emulsion is injected into beginning of a microfluidic lateral diffusion device and various fractions are collected at the ending of the device at positions corresponding to specific sizes.
  • Advantages to this lateral diffusion separation means would be the isolation of similarly-sized droplets off-line in a fast and facile manner.
  • Bulk emulsions could be size-selected and then the resulting emulsions, if desired, combined to create sized libraries for re-introduction into a microfluidic device.
  • the lateral diffusion microfluidic devices could be rolled-up into a syringe or designed for parallel processing.
  • Microfabricated devices have also been designed that inherently rely on diffusion for separation. Particle mixtures are either repeatedly subject to spatially asymmetric potentials created by microelectrodes or driven through arrays of micrometer-scale asymmetric obstacles to exploit differences in diffusion lengths. In all of the devices discussed so far, particles in a given zone have many different migration paths, and diffusion is required for separation.
  • the present invention describes a separation process that creates equivalent migration paths for each particle in a mixture, thereby eliminating multipath zone broadening ( FIG. 24 ).
  • FIG. 24 (Panel A) Geometric parameters defining the obstacle matrix. A fluid flow is applied in the vertical direction (orange arrow).
  • Panel C A particle with a radius that is larger than lane 1 follows a streamline passing through the particle's center (black dot), moving toward lane 1. The particle is physically displaced as it enters the next gap. Black dotted lines mark the lanes.
  • the separation process uses laminar flow through a periodic array of micrometer-scale obstacles. Each row of obstacles is shifted horizontally with respect to the previous row by ⁇ , where ⁇ is the center-to-center distance between the obstacles ( FIG. 24 ) . For convenience, let ⁇ / ⁇ be 1 ⁇ 3. Fluid emerging from a gap between two obstacles will encounter an obstacle in the next row and will bifurcate as it moves around the obstacle. Let the flow diverted to the left of the obstacle be ⁇ , where ⁇ is the total fluid flux going through the gap. If the fluid is confined to move straight down through the array, ⁇ must equal ⁇ / ⁇ . Let us then consider the flow through a gap to be made up of three lanes, each of which by definition has a flux of ⁇ /3.
  • Particles that are smaller than the lane width will follow the streamlines.
  • a particle starting in lane 1 will go through lane 3 (right lane with respect to the gap) in the second row, lane 2 (middle lane) in the third row, and back to lane I (left lane) in the fourth row ( FIG. 24B ).
  • particles starting from any of the three lanes will go back to the original lane assignment after three rows, so that net migration is in the average flow direction. This motion is called the “zigzag mode.”
  • particles can diffuse into an adjacent lane.
  • the microscopic path for all lanes is equivalent, unlike the multiple paths particles take when moving through a column of porous beads.
  • a particle with a radius larger than the width of lane I at a gap will behave differently in the array. This is because the center of the particle cannot “fit” into lane I in a gap. As such a particle from lane 2 in one gap moves into the subsequent gap, expecting to move through the gap in lane 1, the particle will be “bumped” and its center will thus be displaced into lane 2 ( FIG. 24C ). The particle will then flow with the fluid in lane 2. This process is repeated every time a large particle approaches a row of obstacles, so that the particle remains in lane 2 as it moves down through the array. This transport pattern is called the “displacement mode.” This is also applicable to electrophoresis by considering ion flows instead of fluid flows.
  • FIG. 25 shows High-resolution separation of fluorescent microspheres with diameters of 0.80 um (green), 0.90 um (red), and 1.03 um (yellow), with a matrix of varying gap size. Whereas the shift in registry and the lattice constants of the matrix remain the same, the obstacle diameters are changed to create gaps, d, of different sizes, which are labeled on the left side of the fluorescent image.
  • the red bars on the fluorescence profile represent the width of the peaks (SD), and the black bars label the 1% inhomogeneity in the bead population. a.u., arbitrary units.
  • FIG. 26 is a schematic illustrating the separation by deterministic lateral displacement in an array of microposts, with an example row shift fraction of one-third. This shift creates three equal flux streamlines.
  • the dashed lines are the boundaries between the streamlines, which are assigned an index in the gaps between the posts.
  • Paths of particles both smaller and larger than the critical threshold are depicted with green and red dotted lines respectively. Small particles stay within a flow stream and large particles are displaced at each obstacle.
  • G is the clear spacing between the gap, is the center-to-center post separation, and d is the relative shift of the post centers in adjacent rows.
  • the present invention provides methods for performing polymerase chain reaction (PCR).
  • PCR can be performed on a drop-by-drop basis in a microfluidic device according to the present invention.
  • a monolithic chip can be provided wherein the heating and cooling lines are built into the chip and a sorting means is provided. Advantages of performing PCR in droplets on such a chip are that the chip is disposable and the reaction can be repeated without cleaning the device between reactions. Furthermore, the chip provides a convenient way of getting all the components to perform PCR in the droplets in the right concentration. Additionally, the PCR is more efficient because the heat transfer is more efficient due to the small volume. This provides for shorter incubation/residence times.
  • Droplets containing the nucleic acids, all PCR primers, and, if present, beads are generated one at a time at rates between 100 and 20,000 droplets per second.
  • the droplets can then be sent through a serpentine path between heating and cooling lines to amplify the genetic material inside the droplets.
  • the droplets may be sent for further on-chip or off-chip processing, directed into another chip, or the emulsion may be broken to release the PCR product.
  • beads may be harvested by passing the emulsion through a filtration device, sedimentation, or centrifugation.
  • the width and depth of the channel can be adjusted to set the residence time at each temperature, which can be controlled to anywhere between less than a second and minutes. At a typical rate of 1000 drops per second, 1 million strands of DNA can be amplified in approximately 20 minutes on one device. A typical flow rate of 250 ⁇ L/hour would correspond to 1000 drops of 50 microns in diameter being generated every second. Flow rates and droplet sizes can be adjusted as needed by controlling the nozzle geometry.
  • the present invention also provides methods for performing dideoxynucleotide sequencing reactions on a microfluidic device.
  • Chain terminator sequencing (Sanger sequencing) is well known to those of ordinary skill in the art.
  • DNA template preparation, cycling sequencing and preparing extension products for electrophoresis are related techniques and also well known to those of skill in the art.
  • Applied Biosystems' “Automated DNA Sequencing: Chemistry Guide” 2000 is an excellent resource covering these techniques and is incorporated herein by reference in its entirety.
  • One method is to sequencing PCR templates which can include single amplification PCR or nested and semi-nested PCR strategies.
  • the target DNA is amplified with a single set of primers and then sequenced using the same primers. For many samples, this works well.
  • optimization of the PCR amplification may be required. Optimizing the PCR minimizes the presence of non-specific product bands and ensures adequate yield.
  • a single PCR amplification is also compatible with the use of a sequencing primer that binds internally (semi-nested or nested) to one or both of the PCR primers. This can be helpful if primer-dimer (primer oligomerization) artifacts are a problem.
  • a nested or semi-nested PCR can be used. These techniques are useful when the target is present in small quantity. They offer more specificity, which provides superior sequencing data with reduced background signal. Both nested and semi-nested PCR require two amplifications. The first amplification is identical for nested and semi-nested, but the second amplification differs as described below. Amplify with one set of PCR primers, which converts a complex sample (such as bacterial genomic DNA) into a non-complex sample consisting of the first PCR product and some side products.
  • a complex sample such as bacterial genomic DNA
  • Nested PCR Amplify 1% or less of the first PCR reaction product using a second set of PCR primers that hybridize at positions internal to the first set.
  • Semi-nested PCR Only one primer of the second set of PCR primers is internal. The other primer is one of the original PCR primers.
  • a PCR primer can be synthesized with a universal sequencing primer binding site added to the 5′ end (e.g., see Appendix E in Applied Biosystems' “Automated DNA Sequencing: Chemistry Guide” for universal primer sequences). This allows any PCR product to be sequenced with universal primers.
  • Universal-tailed PCR primers enable the use of commercially available dye-labeled sequencing primers. This technique is also useful with dye terminator chemistries, because universal sequencing primers have good annealing characteristics. However, the longer PCR primers add to the overall cost of the reactions. Using universal-tailed primers sometimes results in primer oligomerization. As these products have priming sites present, they can result in noisy data for the first 20-100 bases. Redesigning the PCR primer, optimizing the PCR amplification further, and employing Hot Start methods can help overcome this situation.
  • the resulting PCR product is in solution along with PCR primers, dNTPs, enzyme, and buffer components.
  • the method used to prepare the PCR product for sequencing depends on the amounts of these components that are carried over and on the chemistry used for sequencing.
  • Excess PCR primers carried over from the amplification reaction compete with the sequencing primer for binding sites and reagents in the sequencing reaction. This carryover of PCR primers presents more of a problem in dye terminator chemistries because the dye label is incorporated into the extension product after the primer anneals to the template. If more than one primer is present, multiple dye-labeled sequence ladders are generated, resulting in noisy data.
  • Excess dNTPs from the amplification reaction can affect the balance of the sequencing reaction, resulting in decreased termination in shorter extension fragments.
  • Nonspecific PCR products include primer-dimer artifacts and secondary PCR products.
  • the presence of any significant quantity of either in a PCR product can result in poor quality sequencing data.
  • Nonspecific PCR products behave as templates in the sequencing reaction and produce extension products, which results in noisy data. These products often can be visualized on an agarose gel before sequencing. If they are present, the PCR amplification should be optimized and repeated before sequencing. Use of a nested or semi-nested sequencing primer can also allow good sequence data to be obtained.
  • the PCR product of interest can be purified by agarose gel electrophoresis.
  • PCR optimization Innis and Gelfand, 1990: (1) Amount of starting DNA; (2) Careful primer design; (3) Primer concentration, (4) Enzyme concentration, (5) Magnesium ion (Mg2+) concentration, (6) Nucleotide concentration; (7) Buffer composition; (8) Number of cycles; (9) pH; (10) Manual Hot Start method; (11) AmpliTaq Gold® DNA Polymerase as an automatic Hot Start and/or (12) Limiting dNTPs and primers. All of these methods increase the specificity of the PCR amplification and decrease the amount of contaminants that can interfere with a sequencing reaction.
  • the SAP/Exo I procedure degrades nucleotides and single-stranded DNA (primers) remaining after PCR (Werle et al., 1994). This procedure is particularly useful in cases where limiting concentrations of primers and nucleotides cannot be used for direct PCR sequencing.
  • FIG. 27 shows one embodiment for a DNA sequencing chip design.
  • Template DNA and primers are combined at step ‘add 1’ and the reaction is incubated at 95° C. for a hot start (position 1).
  • the reaction then cycles 20-30 times (position 2) before the addition of SAP and ExoI at ‘add 2.’
  • the reaction is incubated at 37° C. for a predefined time-period and then the SAP and ExoI enzymes are inactivated at 95° C. (position ‘4’).
  • the SAP/ExoI procedure degrades nucleotides and single-stranded DNA (primers) remaining after PCR.
  • the universal sequencing primers, ddNTPs and buffers are added at ‘add 3,’ and the PCR sequencing reaction is allowed to proceed at position ‘5.’
  • the final reaction product is collected and can be stored off-chip.
  • PCR protocols that limit amounts of primers and dNTPs allow the product of the reaction to be used for sequencing with no purification. This is usually carried out by setting up the PCR amplification with 5-10 pmol of primers and 20-40 ⁇ M dNTPs, so that most of the primers and dNTPs are exhausted during amplification. If the yield of the desired PCR product is high and the product is specific, i.e., it produces a single band when analyzed by agarose gel electrophoresis, the sample can be diluted before sequencing and will give good results. The dilution ratio depends on the concentration of your PCR product and needs to be determined empirically (start with 1:2 and 1:10 dilutions with deionized water).
  • Direct PCR sequencing is most useful in applications where the same target is being amplified and sequenced repeatedly and PCR conditions have been optimized. Direct PCR sequencing can be done with dye primer chemistries. With dye terminator chemistries, it is much more critical that the PCR primers be consumed. Excess PCR primers will be extended and labeled by the cycle sequencing reaction, resulting in noisy data. Direct PCR sequencing does not work for XL PCR because limiting amounts of primers and dNTPs cannot be used. The PCR product should be purified or the excess primers and nucleotides should be degraded by SAP/Exo I treatment.
  • the present invention provides methods for performing isothermal-type amplification methods on a microfluidic device.
  • Isothermal amplification is an alternative to the standard PCR techniques described herein. Isothermal amplification is used to reduce the relative amount of background DNA in a sample. Primers are generally used in a constant temperature means of amplification. Isothermal amplification is applicable for SNP detection.
  • Once the DNA is amplified by isothermal amplification there are several well-known means for detecting which nucleotide polymorphism is present. These include, but are not limited to; allele specific primer extension, oligonucleotide ligation assay, mini-sequencing, fluorescence polarization, etc.
  • Isothermal amplification is also applicable for DNA sequencing preparation.
  • the isothermally-amplified DNA can be attached to a solid phase within a droplet or placed within a parking space on chip. The beads or parking spaces can be accessed and the amplified DNA used for a DNA sequencing reaction.
  • isothermal amplification is applicable for gene expression analysis. Isothermal amplification can be used to monitor gene expression by the measurement of the amount of cDNA produced in a quantitative fashion. Many methods for isothermal amplification are known in the art, including but not limited to the following examples.
  • Rolling circle amplification A DNA polymerase extends a primer on a circular template, generating tandemly linked copies of the complementary sequence of the template (Fire & Xu, 1995).
  • the TempliPhi amplification process using rolling circle amplification is known in the art. In the process, random hexamer primers anneal to the circular template DNA at multiple sites. Phi29 DNA polymerase extends each of these primers. When the DNA polymerase reaches a downstream-extended primer, strand displacement synthesis occurs. The displaced strand is rendered single-stranded and available to be primed by more hexamer primer. The process continues, resulting in exponential, isothermal amplification.
  • TMA Transcription mediated amplification
  • An RNA polymerase is used to make RNA from a promoter engineered in the primer region, a reverse transcriptase to produce complementary DNA from the RNA templates and RNase H to remove the RNA from cDNA (Guatelli et al, 1990).
  • Strand-displacement amplification SDA
  • a restriction endonuclease is used to nick the unmodified strand of its target DNA and the action of an exonuclease-deficient DNA polymerase to extend the 30 end at the nick and displace the downstream DNA strand (Walker et al, 1992). Strand-displacement amplification is known in the art.
  • HAD Helicase-dependent amplification
  • a DNA helicase is used to generate single-stranded templates for primer hybridization and subsequent primer extension by a DNA polymerase. Schematic diagram of HAD is shown in FIG. 28 . Two complementary DNA strands are shown as two lines: the thick one is the top strand and the thin one is the bottom strand. 1: A helicase (black triangle) separates the two complementary DNA strands, which are bound by SSB (grey circles). 2: Primers (lines with arrow heads) hybridize to the target region on the ssDNA template. 3: A DNA polymerase (squares with mosaic patterns) extends the primers hybridized on the template DNA. 4: Amplified products enter the next round of amplification.
  • Random libraries of DNA fragments are generated by shearing an entire genome and isolating single DNA molecules by limiting dilution. See, FIG. 29 .
  • sequencing reactions such as those performed by Solexa, 454 Life Sciences and others involve randomly fragmenting the entire genome, adding specialized common adapters to the fragments, capturing the individual fragments on their own beads and, within the droplets of an emulsion, clonally amplifing the individual fragment ( FIG. 29 a, 29 b ).
  • their approach does not require subcloning or the handling of individual clones; the templates are handled in bulk within the emulsions. Typically, about 30% of the beads will have DNA, producing 450,000 template-carrying beads per emulsion reaction.
  • FIG. 29 Sample preparation and DNA sequencing is shown in FIG. 29 .
  • Panel A Genomic DNA is isolated, fragmented, ligated to adapters and separated into single strands (top left). Fragments are bound to beads under conditions that favor one fragment per bead, the beads are captured in the droplets of a PCR-reaction-mixture-in-oil emulsion and PCR amplification occurs within each droplet, resulting in beads each carrying ten million copies of a unique DNA template (top, second from the left). The emulsion is broken, the DNA strands are denatured, and beads carrying single-stranded DNA clones are deposited into wells of a fiber-optic slide (bottom left).
  • Panel B Microscope photograph of emulsion showing droplets containing a bead and empty droplets. The thin arrow points to a 28-mm bead; the thick arrow points to an approximately 100-mm droplet.
  • Panel C Scanning electron micrograph of a portion of a fiber-optic slide, showing fiber-optic cladding and wells before bead deposition.
  • Panel D The sequencing instrument consists of the following major subsystems: a fluidic assembly.
  • Panel E a flow chamber that includes the well-containing fiber-optic slide.
  • Panel F a CCD camera-based imaging assembly.
  • Panel G and a computer that provides the necessary user interface and instrument control.
  • Another example is sequencing in fabricated picolitre-sized reaction vessels.
  • One method uses sequencing by synthesis simultaneously in open wells of a fiber-optic slide using a modified pyrosequencing protocol that is designed to take advantage of the small scale of the wells.
  • the fiber optic slides are manufactured by slicing of a fiber-optic block that is obtained by repeated drawing and fusing of optic fibers. At each iteration, the diameters of the individual fibers decrease as they are hexagonally packed into bundles of increasing cross-sectional sizes.
  • Each fiber-optic core is 44 ⁇ m in diameter and surrounded by 2-3 ⁇ m of cladding; etching of each core creates reaction wells approximately 55 ⁇ m in depth with a centre-to-centre distance of 50 ⁇ m ( FIG.
  • the slide containing approximately 1.6 million wells, is loaded with beads and mounted in a flow chamber designed to create a 300- ⁇ m high channel, above the well openings, through which the sequencing reagents flow ( FIG. 29 d ).
  • the unetched base of the slide is in optical contact with a second fiber optic imaging bundle bonded to a charge-coupled device (CCD) sensor, allowing the capture of emitted photons from the bottom of each individual well ( FIG. 29 d ).
  • CCD charge-coupled device
  • reagents flow perpendicularly to the wells.
  • This configuration allows simultaneous extension reactions on template-carrying beads within the open wells and relies on convective and diffusive transport to control the addition or removal of reagents and by-products.
  • the timescale for diffusion into and out of the wells is on the order of 10 s in the current configuration and is dependent on well depth and flow channel height.
  • the timescales for the signal-generating enzymatic reactions are on the order of 0.02-1.5 s.
  • the current reaction is dominated by mass transport effects, and improvements based on faster delivery of reagents are possible.
  • Well depth was selected on the basis of a number of competing requirements: (1) wells need to be deep enough for the DNA-carrying beads to remain in the wells in the presence of convective transport past the wells; (2) they must be sufficiently deep to provide adequate isolation against diffusion of by-products from a well in which incorporation is taking place to a well where no incorporation is occurring; and (3) they must be shallow enough to allow rapid diffusion of nucleotides into the wells and rapid washing out of remaining nucleotides at the end of each flow cycle to enable high sequencing throughput and reduced reagent use. After the flow of each nucleotide, a wash containing a pyrase is used to ensure that nucleotides do not remain in any well before the next nucleotide being introduced.
  • Nucleotide incorporation is detected by the associated release of inorganic pyrophosphate and the generation of photons.
  • Wells containing template-carrying beads are identified by detecting a known four-nucleotide ‘key’ sequence at the beginning of the read.
  • Raw signals are background-subtracted, normalized and corrected.
  • the normalized signal intensity at each nucleotide flow, for a particular well indicates the number of nucleotides, if any, that were incorporated. This linearity in signal is preserved to at least homopolymers of length eight.
  • sequencing by synthesis a very small number of templates on each bead lose synchronism (that is, either get ahead of, or fall behind, all other templates in sequence).
  • NTA Non-template amplification
  • the present invention provides the use of mixed modified and standard hexamer primers in microfluidic reactions to retard NTA while allowing template-based amplification to proceed.
  • the modified primers of the present invention containing nitroindoles and C3 non-replicable elements were studied in an effort to reduce NTA both in bulk and microfluidic reactions. Both nitroindoles and C3 non-replicable elements were found to be effective in reducing NTA, with primers containing two 5′ nitroindoles most effective in NTA suppression. However, increased NTA suppression was tightly linked with reduced yield in template amplification reactions.
  • Amplifications using a ratio of nitroindole to random hexamer primers generated a range of both template and non-template amplification yields, with a 15:85 ratio of nitroindole to random hexamers generating template yields commensurate with random hexamers primers alone, but generating little if any spurious product in the absence of template.
  • a Single Nucleotide Polymorphism is a small genetic change, or variation, that can occur within a person's DNA sequence.
  • the genetic code is specified by the four nucleotide “letters” A (adenine), C (cytosine), T (thymine), and G (guanine).
  • SNP variation occurs when a single nucleotide, such as an A, replaces one of the other three nucleotide letters—C, G, or T.
  • SNP SNP
  • AAGGTTA ATGGTTA
  • T ATGGTTA
  • SNPs SNPs occur in the human population more than 0.1 percent of the time. Because only about 3 to 5 percent of a person's DNA sequence codes for the production of proteins, most SNPs are found outside of “coding sequences”. SNPs found within a coding sequence are of particular interest to researchers because they are more likely to alter the biological function of a protein. Because of the recent advances in technology, coupled with the unique ability of these genetic variations to facilitate gene identification, there has been a recent flurry of SNP discovery and detection.
  • the most appropriate drug for an individual could be determined in advance of treatment by analyzing a patient's SNP profile.
  • personalized medicine The ability to target a drug to those individuals most likely to benefit, referred to as “personalized medicine”, would allow pharmaceutical companies to bring many more drugs to market and allow doctors to prescribe individualized therapies specific to a patient's needs.
  • the microfluidic device of the present invention is capable of performing at least 10,000 SNP analysis per second, such that a full genome scan (i.e., 100K SNPs) with 10 ⁇ overrepresentation can be performed in less than an hour.
  • a full genome scan i.e., 100K SNPs
  • the speed and efficiency permitted using the devices and methods of the present invention will significantly lower the associated costs and reagent consumption for performing SNP analysis.
  • PCR and isothermal amplifications described herein can be very useful in providing necessary sample preparation processes for commercial available DNA sequencers, such as the Solexa's 1G sequencer, which relies upon immobilized DNA for a substrate.
  • single molecules can be amplified using PCR primers treated with moieties used for surface immobliziation, then flow the PCR positive droplets across the surface of the slide, forming a packed emulsion. The emulsion can then be broken, and the primers allowed to bind to the slide due the presence of the appropriate coating to bind the PCR products.
  • the Solexa 1G sequencer currently sequences amplified material that has been amplified in place, on primers bound to the slide, by bridge amplification.
  • template DNA is flowed across the slide surface at very low concentrations, and adapters previously ligated to each template hybridize to complimentary primers attached to the slide in any one of eight lanes.
  • the primers are subjected to bridge amplification, a version of PCR amplification that utilizes immobilized primers—the product of the reaction is a patch of DNA immobilized to the slide.
  • a Solexa slide can be coated with any of the compounds commonly used to permit binding and immobilization (e.g., carboxy-esters, streptavidin, Igg, gold, etc).
  • PCR reactions could be performed as described in the instant microfluidic device using primers modified with the appropriate binding moiety (5′ amines, 5′ biotins, 5′ DNPs or 5′ Thiols, respectively) to efficiently amplify PCR products in solution which could then be efficiently and easily bound to the Solexa slide for subsequent sequencing.
  • the amplification is quite straight forward, conducted with a limiting dilution of template and a set of primer pairs compatible with the adapters ligated to the Solexa templates through their standard sample preparation process.
  • One of the primer pairs would possess a 5′ binding moiety as described earlier, only one of the pair as this will permit removal of the opposing strand and the generation of single stranded immobilized templates on the slide.
  • Proper spacing between the droplets can be obtained by mixing droplets containing only buffer or immiscible oil in with the PCR positive droplets at a ratio sufficient to ensure that the PCR positive droplets are rarely proximal to each other.
  • the droplets can be broken through the application of electrical field and the PCR products allowed to bind to the slide in the same geographic area that the droplet had occupied.
  • the immobilized templates can be rendered single stranded through the application of basic washes, temperature etc. This will permit the rapid amplification of PCR fragments and their subsequent density-controlled deposition onto the Solexa chip for sequencing.
  • the present invention provides methods for detecting the presence and/or sequence of nucleic acids in low copy number in droplets on a microfluidic device.
  • the detection of a specific nucleic acid sequence present in a sample by probing the sample with a complementary sequence of nucleic acids is a well known technique.
  • Nucleic acids are highly specific in binding to complementary nucleic acids and are thus useful to determine whether a specific nucleic acid is present in a sample.
  • One must know the sequence of the specific nucleic acid to be detected and then construct a probe having a complementary nucleic acid sequence to the specific nucleic acid sequence.
  • nucleic acid probes are highly specific, it is preferable in some situations to probe the nucleic acid sequence itself rather than the protein produced by the nucleic acid sequence.
  • a diagnostic method based solely on protein detection would be unreliable for determining the presence of infectious particles of hepatitis B virus, due to the presence of significant levels of non-infectious antigen particles which lack the DNA genome.
  • the various subtypes of human papilloma virus found in either pre-cancerous or benign cervical tumors can be distinguished only by the use of nucleic acid probe hybridization.
  • the specific genetic makeup of an AIDS virus makes it certain that an assay based on the presence of an AIDS virus specific nucleic acid sequence would be superior as a diagnostic.
  • ribosomal RNA up to 100,000 copies per cell
  • GenProbe to facilitate diagnosis of certain bacterial pathogens, such as Legionella and Mycoplasma
  • this strategy cannot be used with non-cellular pathogens, such as viruses, or with probed nucleic acid sequences with low copy numbers.
  • Copy number is a particular problem with the development of a nucleic acid probe method for the detection of AIDS virus, where the integrated provirus may be present in less than one of ten thousand peripheral blood lymphocytes.
  • the particular nucleic acid sequence suspected to be present in a sample could be amplified, the copy number problem could be circumvented and probe assays could be more readily used.
  • One method to amplify is to ‘grow out’ the sample, that is, to arrange conditions so that the living biological material present in the sample can replicate itself. Replication could increase the quantity of nucleic acid sequences to detectable levels.
  • food samples In the food industry, for example, in order to test processed food for the food-poisoning bacteria Salmonella, food samples must be incubated for a number of days to increase the quantity of nucleic acid copy numbers. In clinical samples, pathogens must also be allowed to increase their number by growing out over some considerable time.
  • Current methods utilize a process in which a sample suspected of containing a target DNA sequence is treated with oligonucleotide primers such that a primer extension product is synthesized which in turn serves as a template, resulting in amplification of the target a DNA sequence.
  • the primer extension product is separated from the template using heat denaturation.
  • Current methods also include a process for amplifying a target DNA sequence having two separate complementary strands. The process includes treating the strands with primers to synthesize extension products, separating the primer extension products from the templates, and in turn using the primer extension products as templates.
  • Both of the above methods require either manual or mechanical participation and multi-step operations by the user in the amplification process and are restricted to amplifying DNA only.
  • the steps involved in these methods require the user to heat the sample, cool the sample, add appropriate enzymes and then repeat the steps. The temperature changes cause the enzymes to loose their activity. Hence, the user is required to repeatedly supplement the amplification mixture with aliquots of appropriate enzymes during the amplification process.
  • a nucleotide/peptide nucleic acid (pna) probe can be tethered such that when it binds to a template DNA molecule in low copy number located inside an aqueous emulsion (i.e., droplet) it turns on or activates an enzyme.
  • aqueous emulsion i.e., droplet
  • an alkaline phosphatase conjugate can be placed on one and of the low copy number nucleotide, and an inhibitor to alkaline phosphatase on the other end (like a molecular beacon).
  • the tethers can be a protein complementation assay wherein the binding of the oligo probe to the low copy number template causes the enzyme to be active.
  • Example 1 Taqman chews a beta-gal alpha protein attached at the 3′ end of an oligo probe thereby releasing free alpha subunit to bind to omega fragment in solution.
  • Example 2 Two oligo probes sit down on the low copy number template adjacent to each other, thereby bringing two subunits of a protein complementing assay reagent together.
  • Example 3 A Taqman-like enzyme that releases an active moiety can also be used. The active moiety can include, for example, an enzyme that becomes activated upon release from the oligo probe, or a fluorescent group that is quenched while tethered to the oligo probe.
  • Example 4 The probe has a fluorescent group attached such that the detected hybridization causes the release of the fluorescent group a la Taqman.
  • Example 5 The probe has an inactive enzyme attached such that the detected hybridization causes the release of the active enzyme by a Taqman-type release.
  • Example 6 The probe has an inactive complementing enzyme attached such that the detected hybridization causes the release of the active moiety of the enzyme to be able to complement.
  • Example 7 Two probes have inactive enzyme moieties attached such that the detected hybridization causes the complementation and activation of the enzyme.
  • Example 8 Two probes come together and allow a Fluorescence Resonance Energy Transfer (FRET) reaction to occur. This would require a FRET-oligo library. Almost all SNP or transcriptional profiling method may be amenable to this concept.
  • FRET Fluorescence Resonance Energy Transfer
  • microfluidic devices are including, but not limited to, protein-protein, antibody-antigen, nucleic acid-protein, nucleic acid-nucleic acid, ligand-protein, ligand-nucleic acid, ligand-ligand, eukaryotic or prokaryotic cell surface moiety-second moiety, the measurement of two or more receptors on the surface of an eukaryotic or prokaryotic cell, the development of three-hybrid type systems using tandem fusions, interactor-cofactor, etc. Many other types of interactions are also known that can be adapted to the system. Assays incorporating complementation assays can be used in both proteomics and genomics.
  • the present invention provides methods which reduce the number of interacting proteins by amplifying the signal of molecules that do interact.
  • stable emulsions may not be needed.
  • the amount of each interacting partner may be with-in drop quantifiable by genetically or chemically coupling a reporter molecule (e.g., a dye or quantum dot, GFP protein) to one and or the other.
  • a reporter molecule e.g., a dye or quantum dot, GFP protein
  • the complementation assay described for enzymatic amplification can use one of several different complementing proteins such that the concentration of each partner can be calculated within the droplet by using different enzyme substrates added to the droplets at the same or differing time. Timing of substrate addition is not critical and one of skill in the art would readily recognize that addition can be done at different times. ‘Killing’ by various denaturants, protease, etc is also within the purview of the skilled artisan.
  • complementation assays can be used to add a specific addition to, for example, an IVT synthesized protein.
  • an IVT synthesized protein As an example, the s-peptide of RNaseA and the S protein wherein the S peptide is genetically fused to the IVT-generated Ab and the S protein, upon binding to the S peptide activates the RNaseA activity and thereby stops further IVT synthesis.
  • any two complementing interactrs can be used to generate an activity.
  • the present invention provides methods which can allow the quantification of proteins below that which can be seen by fluorescent spectroscopy in the absence of amplification.
  • amplification may also not be needed if one can genetically add a quantified number of reporters to the end of a molecule. For example, genetically fusing 10 GFP proteins onto the end of a protein would thereby increase the fluorescence intensity 10-fold.
  • a series of small complementing moieties can be fused onto the end of a protein and there obviate the need for long genetic fusions.
  • a series of 10 s peptides spaced apart by a linker would be able to each ‘grab hold’ of an S protein to generate an increased signal over that which can be achieved by a single enzyme.
  • Other examples include either biotin or biotin-binding protein mimetic and streptavidin or avidin, FLAG tags, poly histidines, complementing GFP, etc.
  • Another example includes Qcoding the droplets.
  • FIGS. 30 and 31 show the current method for isolating antibodies on a microfluidic device.
  • DNA beads are made using bulk emulsion PCR; DNA-containing beads preferably isolated before loading onto a microfluidic device.
  • the antigen can be ⁇ 20 kD, (preferably ⁇ 10 kD) and labeled with an appropriate dye, (preferably with several dye-molecules). It may even be preferable to put a poly-lysine tail onto the antigen to increase signal (but be concerned about quenching).
  • the antigen is formulated into the bead-containing droplets at a concentration of 100 nm to allow proper sensitivity in the droplet (use of multiple/different dyes may allow this concentration to drop). In some situations it is possible to add antigen at the same time as the IVT solution.
  • FIG. 32 shows the method of the present invention for isolating antibodies on the microfluidic device.
  • the right panel is a diagram of individual steps proposed to amplify signal of interacting antibody and antigen.
  • the left panel is a schematic as would be designed for a chip to be used on microfluidic device.
  • FIG. 33 shows the genetic selection for full length antibody clones.
  • a genetic selection can be used to enrich for full-length antibody clones by transforming E. coli and selecting for clones able to grow on medium in which a suitable sugar is the only carbon source.
  • the present invention provides a microfluidic device topology and implementation that merges the functionality of a first microfluidic substrate with that of a second microfluidic substrate by using forced withdrawal and reinjection into the same fluidic port.
  • steps must be performed to complete a “two step” experiment: (1) Installation of the first microfluidic substrate; (2) Installation of all first stage reagents; (3) Priming of all fluid lines and stabilization of the device operation; (4) Passive connection of a storage container to the instrument after device has been stabilized; (5) Disconnection of the storage container from the first device when collection complete; (6) Incubation of the collected emulsion; (7) Removal of the first device from the instrument; (8) Cleaning of the fluid lines needed for the second step of the experiment; (9) Installation of a second device to the instrument; (10) Connection of all second stage reagents, including the emulsion collected during the first stage; (11) Priming of the fluidic connections to the second device prior to running the second half of the measurement; and (12) Collection
  • the methods of the present invention replaces current methodology of multi-step assays with the following: (1) Installation of the first microfluidic substrate; (2) Installation of all first and second stage reagents, including the first stage storage container (if it is not already connected); (3) Priming and stabilization of the fluidic lines and device; (4) Controlled collection of the first stage combined droplets by actively withdrawing some fraction of the oil and all of the generated emulsion into the storage container; (5) Incubation of the collected emulsion; (6) Startup and reinjection of the collected droplets back into the second half of the device; and (8) Collection/readout of the second stage.
  • FIG. 34 depicts a schematic representation of this device topology.
  • RNAi short interfering RNA
  • shRNA Short hairpin RNA transcribed from small DNA plasmids within the target cell has also been shown to mediate stable gene silencing and achieve gene knockdown at levels comparable to those obtained by transfection with chemically synthesized siRNA (T. R. Brummelkamp, R. Bemards, R. Agami, Science 296, 550 (2002), P. J. Paddison, A. A. Caudiy, G. J. Hannon, PNAS 99, 1443 (2002)).
  • Possible applications of RNAi for therapeutic purposes are extensive and include silencing and knockdown of disease genes such as oncogenes or viral genes.
  • microfluidic devices are including, but not limited to, protein-protein, antibody-antigen, nucleic acid-protein, nucleic acid-nucleic acid, ligand-protein, ligand-nucleic acid, ligand-ligand, eukaryotic or prokaryotic cell surface moiety-second moiety, the measurement of two or more receptors on the surface of an eukaryotic or prokaryotic cell, the development of three-hybrid type systems using tandem fusions, interactor-cofactor, etc. Many other types of interactions are also known that can be adapted to the system. However, there is a need in the art for improved methods of RNAi screening, quickly and accurately.
  • the present invention provides methods for the screening of lethal and synthetic lethal RNAi-induced phenotype on a microfluidic device.
  • the present invention utilizes a lentiviral library of RNAi where each virus has a unique 60-nt identifying barcode bracketed on either side with nucleotide sequences common to all vectors.
  • the analysis of lethal and synthetic lethal RNAi-induced phenotypes occurs in two steps.
  • the viral library is combined in bulk and infected, also in bulk, into an appropriate host strain.
  • the molar amount of each of the different lentivirus in the library is pre-determined by sequencing on, for example, an appropriate instrument or by gene expression analysis on a microfluidic device.
  • Post infection the treated cells are collected and the 60-nt barcode is amplified from chromosomal DNA using PCR primers based on the bracketing sequence.
  • the PCR amplification product is added to a microfluidic device and analyzed against a labeled droplet library wherein the labeled droplets contain lentiviral-barcode-quantification reagents (e.g., molecular beacons, Taqman probes, etc.) against each of said lentiviral barcodes.
  • lentiviral-barcode-quantification reagents e.g., molecular beacons, Taqman probes, etc.
  • a gene-expression analysis-like analysis is performed to quantify the amount of each lentiviral barcode-type in the treated cells.
  • An absence or significant decrease of any lentiviral barcode in the amplified product can be assumed to be due to the death of that barcode-containing lentivirus in the treated cells.
  • the products within the droplets can also be amplified.
  • step one the viral library is combined in bulk and infected, also in bulk, into an appropriate host strain.
  • the molar amount of each of the different lentivirus in the library is pre-determined by sequencing on, for example, an appropriate instrument or by gene expression analysis on a microfluidic device.
  • Post infection the treated cells are i) collected in bulk, ii) sorted using a phenotype able to be sorted in a microfluidic device (e.g, GFP expression, cell-surface marker, low-copy cell-surface marker, etc) and iii) the 60-nt barcode is amplified from chromosomal DNA using PCR primers based on the bracketing sequence.
  • the PCR amplification product is added to a microfludic device and analyzed against a labeled droplet library wherein the labeled droplets contain lentiviral-barcode-quantification reagents (e.g., molecular beacons, Taqman probes, etc.) against each of said lentiviral barcodes.
  • lentiviral-barcode-quantification reagents e.g., molecular beacons, Taqman probes, etc.
  • a gene-expression analysis-like analysis is performed to quantify the amount of each lentiviral barcode-type in the treated cells. An absence or significant decrease of any lentiviral barcode in the amplified product can be assumed to be due to the death of that barcode-containing lentivirus in the treated cells.
  • chromosomes in cancerous cells frequently exhibit aberrations called translocations, where a piece of one chromosome breaks off and attaches to the end of another chromosome. Identifying such chromosome abnormalities and determining their role in disease is an important step in developing new methods for diagnosing many genetic disorders.
  • Traditional karyotyping using Giemsa staining allows scientists to view the full set of human chromosomes in black and white, a technique that is useful for observing the number and size of the chromosomes.
  • Giemsa staining allows scientists to view the full set of human chromosomes in black and white, a technique that is useful for observing the number and size of the chromosomes.
  • the present invention provides methods for karyotyping.
  • the karyotyping screens occur within droplets on a microfluidic device.
  • Spectral karyotyping SKY is a laboratory technique that allows scientists to visualize all 23 pairs of human chromosomes at one time, with each pair of chromosomes painted in a different fluorescent color. By using SKY, they can easily see instances where a chromosome, painted in one color, has a small piece of a different chromosome, painted in another color, attached to it.
  • chromosomes can be captured within droplets without having to worry about shear-forces.
  • the chromosomes can then be passed through a ‘neck-down’ to stretch them out.
  • Labeling prior to loading with either Giemsa stain or oligonucleotide probes, can be used to karyotype the DNA as it flows.
  • the present invention provides methods of using SKY probes to ‘paint’ individual chromosomes. Also provided is a method used by flow cytometrists for the preparation of chromosomes prior to flow analysis, including flow sorting. The present invention provides methods which allow the adaptation of these methods for use on a microfluidic device.
  • SKY involves the preparation of a large collection of short sequences of single-stranded DNA called probes.
  • Each of the individual probes in this DNA library is complementary to a unique region of one chromosome; together, all of the probes make up a network of DNA that is complementary to all of the chromosomes within the human genome.
  • Each probe is labeled with a fluorescent molecule that corresponds to the chromosome to which it is complementary. For example, probes that are complementary to chromosome 1 are labeled with yellow molecules, while those that are complementary to chromosome 2 are labeled with red molecules, and so on.
  • the probes When these probes are mixed with the chromosomes from a human cell, the probes hybridize, or bind, to the DNA in the chromosomes. As they hybridize, the fluorescent probes essentially paint the set of chromosomes in a rainbow of colors.
  • scientists can then use computers to analyze the painted chromosomes to determine whether any of them exhibit translocations or other structural abnormalities. See, FIG. 35 .
  • chromosomes Prior to analysis chromosomes can be prepared as described in Bee Ling Ng and Nigel P. Carter “Factors Affecting Flow Karyotype Resolution. Cytometry” Part A 69A: 1028-1036 (2006) as follows:
  • the present invention also provides methods of sorting chromosomes for karyotyping wherein individual chromosomes are sorted. This sorting can be performed after chromosome-specific identification (such as hybridization of labeled probes) so as to enrich a population for one or more specific chromosomes. This enriched population can be used in DNA sequencing reactions.
  • the Giemsa-stained and/or labeled-probe-hybridized chromosomes can be sent through a constriction of a channel on a microfluidic device to detect the areas of stain and/or label as a genetic ‘bar-code’ to identify regions of translocation, etc. on individual chromosomes.
  • Identification of chromosomes and karyotyping can be used after enrichment of specific cell-types, for example i) fetal cells from maternal blood, or ii) cancer cells from human blood.
  • Biomass is organic matter such as plant matter, i.e., trees, grasses, agricultural crops, or other biological material such as animal material. It can be used as a solid fuel, or converted into liquid or gaseous forms, for the production of electric power, heat, chemicals, or fuels. Biomass can also be used in formulating other commercial products in other industrial sectors such as textiles, food supply, environmental, communication, housing, etc. For example, biofuel development seeks the development of new microbial strains with improved biomass conversion to ethanol.
  • Lignocellulosic biomass contains five carbon sugars such as xylose (from the hemicellulose) as well as the more “common” six carbon sugars such as glucose found in grains. This makes fermentation and other bioprocessing far more challenging. While some biorefmery scenarios will take advantage of the different sugar streams to produce multiple products, others will be more cost effective if all the sugars can coferment in a single set of equipment. Accordingly, researchers are developing microorganisms that can coferment all the sugars in biomass in order to improve ethanol production economics. With industrial partners, researchers are working to develop designer strains of microorganisms for biomass conversion of specific feedstocks, feedstreams, and processes. Thus, there is a need for devices and methods for the rapid engineering of new microbial strains with improved biomass conversion.
  • the present invention provides a microfluidic device in which to formulate a mutant bacterial, yeast, or fungi strain which can be used for biomass energy conversion.
  • the microorganism strain can be engineered, e.g., by recombinant methods, to include at least one nucleic acid sequence encoding one or more polypeptides of interest, wherein the mutant strain expresses the polypeptides of interest at a higher level than the corresponding non-mutant strain under the same conditions.
  • the nucleic acid sequence can be operably linked to an expression-regulating region selected from the group consisting of a promoter sequence associated with cellulase expression, xylanase expression, or gpdA expression.
  • the nucleic acid sequence can further be optionally linked to a secretion signal sequence.
  • the nucleic acid sequence can be a heterologous nucleic acid sequence selected from heterologous polypeptide-encoding nucleic acid sequences, heterologous signal sequences, or heterologous expression-regulating sequences, or combinations thereof.
  • the nucleic acid sequence can be a heterologous signal sequence, e.g., a secretion signal sequence.
  • the nucleic acid sequence can be a heterologous expression-regulating region, e.g. an inducible promoter or a high expression promoter.
  • the polypeptides of interest can be homologous peptides and are expressed in the mutant strain at a higher level than in the corresponding non-mutant strain under the same conditions.
  • the polypeptides of interest can be selected from one or more of carbohydrate-degrading enzymes, proteases, lipases, esterases, other hydrolases, oxidoreductases, and transferases.
  • the polypeptides of interest can be selected from one or more of fungal enzymes that allow production or overproduction of primary metabolites, organic acids, secondary metabolites, and antibiotics.
  • fungal sequences can include secretion signal sequences, for example, and can be selected from one or more of cellulase, ⁇ -galactosidase, xylanase, pectinase, esterase, protease, amylase, polygalacturonase or hydrophobin.
  • the fungal sequences can include one or more fungal expression-regulating regions.
  • the polypeptides of interest exhibit optimal activity and/or stability at a pH above 6, and/or have more than 70% of its activity and/or stability at a pH above 6.
  • the mutant microorganism can further include a selectable marker.
  • the selectable marker can confer resistance to a drug, for example, or relieve a nutritional defect.
  • the microorganism can be mutated via mutagenesis.
  • Mutagenesis can be achieved, for example, by one or both of UV irradiation or chemical mutagenesis.
  • mutagenesis can include exposing a microorganism to UV irraditation, exposing it to N-methyl-N′-nitro-N-nitrosoguanidine, and exposing it again to UV irradiation.
  • the present invention also provides methods for creating microbial strains with improved biomass conversion.
  • the method includes providing a microfluidic device made of a microfabricated substrate.
  • the microfabricated substrate can have a plurality of electrically addressable, channel bearing modules integrally arranged so as to be in fluid communication with each other, thereby forming at least one main channel adapted to carry at least one continuous phase fluid.
  • the method further includes flowing a buffer, a microbe library, and a media (either in separate solutions or all together in one solution) through a first inlet channel into the main channel of the microfabricated substrate such that one or more droplets is formed in the continuous phase fluid; flowing a substrate through a second inlet channel into the main channel of the microfabricated substrate such that one or more droplets is formed in said continuous phase fluid; coalescing the droplets containing the microbe library with the droplets containing the substrate as the droplets pass through a coalescence module, thereby producing a NanoRefinery; interrogating the NanoRefinery for a predetermined characteristic within a detection module on the microfabricated substrate; and collecting the NanoRefineries containing the microbes of interest in a collection module on the microfabricated substrate.
  • An assay system e.g., a means by which to determine whether a desired product has been produced, or a means by which to determine the absence of a starting substrate material, can also be incorporated into one of the droplets or the NanoRefinery.
  • the assay system can be added either before, after, or simultaneously with the addition of the substrate.
  • the assay system is a dye that can measure the amount of sugar in a solution.
  • the microbe library can include one or more of DNA, bacteria, yeast or fungi.
  • the substrate can include biomass, which can include one or more of fermentation broth, cellulose or other polysaccharide, or plant lignan.
  • the present invention further provides methods for degrading or converting biomass into one or more products.
  • the method includes treating the biomass with an effective amount of a recombinant microorganism, wherein the recombinant microorganism expresses or overexpresses one or more heterologous sequences encoding enzymes that degrade or convert the biomass into one or more products.
  • the biomass can include plant cell wall polysaccharides.
  • the products can include one or more of the commodity chemicals or secondary commodity chemicals used to produce one or more of the Intermediates or finished products and consumer goods listed in FIGS. 36 and 37 .
  • the present invention provides methods of screening for enzymes with improved activity.
  • at least one cell prokaryotic or eukaryotic
  • the cells are allowed to secrete a substance for which a homogeneous assay is available.
  • an individual Bacillus subtilis cell from a mutagen-treated culture is deposited within a 30 micron growth-medium containing droplet, the resulting droplets are collected and allowed to incubate overnight.
  • the bacterium secretes a protease into the droplet.
  • the droplets containing the Bacilli are then individually merged with an assay droplet containing a protease-cleavable dye-labeled peptide.
  • the uncleaved peptide is colorless, while the cleaved peptide becomes red.
  • the droplets are incubated on chip for a sufficient time such as to allow color formation. Droplets that are red are sorted. The collected sorted droplets are then plated onto solid growth medium and the resulting colonies, after overnight incubation, represent individual clonal isolates from the droplets.
  • the assay droplets can be sorted based upon a specific activity, for example enzymes that are more active, indicated by a more intense red droplet after a specific period of time.
  • the conditions within the droplet can be changed during the merging of the two droplets to assay conditions which may itself not be permissive for the bacteria. For example the pH within the droplet is altered in order to find mutant enzymes that work better under either acidic or alkaline conditions. Or the droplets can be heated such that enzymes that are more heat resistant are identified.
  • the merging of the droplets can be right after the individual cell is placed into the droplet or after further incubation.
  • Another method of the instant invention is as described above, except that the cells are lysed before the assay step in order to release the contents of the cell into the droplet.
  • the substrate can be added with- or simultaneously formulated from two or more separate reagent streams with the contained within a droplet.
  • macrophage cells are washed in buffer and incubated with an enzyme-conjugated anti-cell-surface antibody.
  • the cells are then individually loaded into droplets formulated at the same time, with the enzyme substrate.
  • the amount of enzyme substrate turned over within the droplet will be proportional to the number of enzyme molecules within the droplet, which is proportional to the number of antibodies bound to the macrophage surface. By careful calibration it should be possible to estimate the number of cell-surface molecules attached to the cell surface.

Abstract

The present invention provides novel microfluidic substrates and methods that are useful for performing biological, chemical and diagnostic assays. The substrates can include a plurality of electrically addressable, channel bearing fluidic modules integrally arranged such that a continuous channel is provided for flow of immiscible fluids.

Description

    RELATED APPLICATIONS
  • This patent application claims priority to, and the benefit of, U.S. Provisional Application Nos. 60/799,833 filed on May 11, 2006; 60/799,834 filed on May 11, 2006; 60/808,614 filed on May 25, 2006; 60/815,097 filed on Jun. 19, 2006; 60/819,733 filed on Jul. 7, 2006; 60/819,734 filed on Jul. 7, 2006; 60/841,716 filed on Sep. 1, 2006; 60/843,374 filed on Sep. 8, 2006; 60/833,151 filed on Jul. 24, 2006; 60/834,987 filed on Jul. 31, 2006; 60/837,871 filed on Aug. 14, 2006; 60/837,695 filed on Aug. 14, 2006; 60/843,327 filed on Sep. 8, 2006; 60/856,540 filed on Nov. 3, 2006; 60/856,440 filed on Nov. 3, 2006; 60/874,561 filed on Dec. 12, 2006; 60/858,279 filed on Nov. 8, 2006; 60/858,278 filed on Nov. 8, 2006; 60/874,640 filed on Dec. 12, 2006; 60/860,665 filed on Nov. 22, 2006; 60/873,766 filed on Dec. 8, 2006; 60/876,209 filed on Dec. 20, 2006; 60/899,258 filed on Feb. 2, 2007; 60/903,153 filed on Feb. 23, 2007; 60/904,293 filed on Feb. 28, 2007; 60/920,337 filed on Mar. 26, 2007. The contents of each of these applications are incorporated herein by reference in their entirety.
  • FIELD OF INVENTION
  • The present invention generally relates to systems and methods for the formation and/or control of fluidic species, and articles produced by such systems and methods. More particularly, the present invention relates to the development of high throughput microfluidic devices for precision fluid handling and use of such systems in various biological, chemical, or diagnostic assays.
  • BACKGROUND
  • The manipulation of fluids to form fluid streams of desired configuration, discontinuous fluid streams, droplets, particles, dispersions, etc., for purposes of fluid delivery, product manufacture, analysis, and the like, is a relatively well-studied art. For example, highly monodisperse gas bubbles, less than 100 microns in diameter, have been produced using a technique referred to as capillary flow focusing. In this technique, gas is forced out of a capillary tube into a bath of liquid, where the tube is positioned above a small orifice, and the contraction flow of the external liquid through this orifice focuses the gas into a thin jet which subsequently breaks into equal-sized bubbles via a capillary instability. A similar arrangement can be used to produce liquid droplets in air.
  • Microfluidic systems have been described in a variety of contexts, typically in the context of miniaturized laboratory (e.g., clinical) analysis. Other uses have been described as well. For example, International Patent Application Publication No. WO 01/89788 describes multi-level microfluidic systems that can be used to provide patterns of materials, such as biological materials and cells, on surfaces. Other publications describe microfluidic systems including valves, switches, and other components.
  • Precision manipulation of streams of fluids with microfluidic devices is revolutionizing many fluid-based technologies. Networks of small channels are a flexible platform for the precision manipulation of small amounts of fluids. The utility of such microfluidic devices depends critically on enabling technologies such as the microfluidic peristaltic pump, electrokinetic pumping, dielectrophoretic pump or electrowetting driven flow. The assembly of such modules into complete systems provides a convenient and robust way to construct microfluidic devices. However, virtually all microfluidic devices are based on flows of streams of fluids; this sets a limit on the smallest volume of reagent that can effectively be used because of the contaminating effects of diffusion and surface adsorption. As the dimensions of small volumes shrink, diffusion becomes the dominant mechanism for mixing leading to dispersion of reactants; moreover, surface adsorption of reactants, while small, can be highly detrimental when the concentrations are low and volumes are small. As a result current microfluidic technologies cannot be reliably used for applications involving minute quantities of reagent; for example, bioassays on single cells or library searches involving single beads are not easily performed. An alternate approach that overcomes these limitations is the use of aqueous droplets in an immiscible carrier fluid; these provide a well defined, encapsulated microenvironment that eliminates cross contamination or changes in concentration due to diffusion or surface interactions. Droplets provide the ideal microcapsule that can isolate reactive materials, cells, or small particles for further manipulation and study. However, essentially all enabling technology for microfluidic systems developed thus far has focused on single phase fluid flow and there are few equivalent active means to manipulate droplets requiring the development of droplet handling technology. While significant advances have been made in dynamics at the macro-or microfluidic scale, improved techniques and the results of these techniques are still needed. For example, as the scale of these reactors shrinks, contamination effects due to surface adsorption and diffusion limit the smallest quantities that can be used. Confinement of reagents in droplets in an immiscible carrier fluid overcomes these limitations, but demands new fluid-handling technology.
  • Furthermore, the underlying physics of the influence of electric fields on fluids is well known. The attractive and repulsive forces produced by an electric field on positive or negative charges give rise to the forces on charged fluid elements, the polarization of non-polar molecules, and the torque on polar molecules which aligns them with the field. In a non-uniform field, because the force on the positively charged portion of the distribution is different than the force on the negatively charged portion, polar molecules will also experience a net force toward the region of higher field intensity. In the continuum limit, the result is a pondermotive force in the fluid. In the limit of high droplet surface tension, it is useful to describe the net pondermotive force on a droplet as if it were a rigid sphere:
    F=qE+
    Figure US20080014589A1-20080117-P00900
    m)r 3
    Figure US20080014589A1-20080117-P00900
    (K)∇E 2.
    where the first term is the electrophoretic force on the droplet (q is the net droplet charge and E is the electric field), and the second term is the dielectrophoretic force (r is the radius of the sphere,
    Figure US20080014589A1-20080117-P00900
    (K) is the real part of the Clausius-Mossotti factor
    K=(ε*p−ε*m)/(ε*p+2ε*m)
    and ε*p and ε*m are the complex permittivities of the droplet and carrier fluid).
  • Although utility of electrophoretic control of droplets is great, it does have significant limitations. First, the charging of droplets is only effectively accomplished at the nozzle. Second, the discharge path required to eliminate screening effects also discharges the droplets. Third, finite conductivity of the carrier fluid, however small, will eventually discharge the droplets. Therefore, once the droplet is formed, there is essentially only one opportunity to perform any pondermotive function which relies on the droplet's charge density (such as coalescing oppositely charged droplets through their mutual Coulombic attraction, or electrophoretically sorting a droplet), and that function can only be performed as long as sufficient charge has not leaked off of the droplet.
  • Thus, it would be desirable to develop an electrically addressable emulsification system that combines compartmentalization and electrical manipulation, which allows for multi-step chemical processing, including analysis and sorting, to be initiated in confinement with exquisite timing and metering precision, for use in a variety of chemical, biological, and screening assays, in which the cost and time to perform such assays would be drastically reduced. It would also be desirable to develop a device using dielectrophoretic force (which does not rely on charge density) to manipulate droplets so that more than one electrical pondermotive function can be carried out following a significantly long delay from droplet formation.
  • SUMMARY OF THE INVENTION
  • The present invention provides substrates having individual fluid handling modules that can be combined into fluid processing systems so as to perform multi-step processing of isolated components, which is essential to perform biological, chemical and diagnostic applications, quickly, effectively and inexpensively. Using principles based on the electrical manipulation of droplets, the microfluidic substrates of the present invention can encapsulate reagents into droplets, which can be combined, analyzed, and sorted.
  • The present invention provides a method of pairing sample fluids to form a droplet or nanoreactor including, for example, (a) providing a microfluidic substrate including at least two inlet channels adapted to carry at least two dispersed phase sample fluids and at least one main channel adapted to carry at least one continuous phase fluid; (b) flowing a first sample fluid through a first inlet channel which is in fluid communication with the main channel at a junction, wherein the junction includes a first fluidic nozzle designed for flow focusing such that the first sample fluid forms a plurality of highly uniform, monodisperse droplets of a first size in the continuous phase; (c) flowing a second sample fluid through a second inlet channel which is in fluid communication with the main channel at a junction, wherein the junction includes a second fluidic nozzle designed for flow focusing such that the second sample fluid forms a plurality of highly uniform, monodisperse droplets of a second size in the continuous phase, wherein the size of the droplets of the second sample fluid are smaller than the size of the droplets of the first sample fluid; (d) providing a flow and droplet formation rate of the first and second sample fluids wherein the droplets are interdigitized such that a first sample fluid droplet is followed by and paired with a second sample fluid droplet; (e) providing channel dimensions such that the paired first sample fluid and the second sample fluid droplet are brought into proximity; (f) coalescing the paired first and second sample droplets as the paired droplets pass through an electric field, thereby producing a droplet or nanoreactor.
  • The present invention also provides various methods of forming a droplet emulsion library of a sample fluid. One method includes, for example, (a) providing at least one first channel adapted to carry at least one dispersed phase sample fluid and at least one second channel adapted to carry at least one continuous phase fluid; (b) flowing the sample fluid through the first channel which is in fluid communication with the second channel at a junction, wherein the junction includes a fluidic nozzle such that the sample fluid forms a plurality of highly uniform, monodisperse droplets of a predetermined size in the continuous phase, wherein the fluidic nozzle is isolated from the dispersed phase fluid, has a three dimensional design to permit flow focusing and eliminates surface wetting. The nozzle can be formed from small bore tubing or from the tip of a molded ferrule.
  • Another method of forming a droplet emulsion library of a sample fluid includes, for example, (a) providing a microfluidic substrate including at least one first channel adapted to carry at least one dispersed phase sample fluid and at least one second channel adapted to carry at least one continuous phase fluid; (b) flowing the sample fluid through the first channel which is in fluid communication with the second channel at a junction, wherein the junction includes a fluidic nozzle such that the sample fluid forms a plurality of highly uniform, monodisperse droplets of a predetermined size in the continuous phase.
  • Another method of forming a droplet emulsion library of a sample fluid includes, for example, (a) providing a microfluidic substrate including at least one first channel adapted to carry at least one dispersed phase sample fluid and at least one channel adapted to carry at least one continuous phase fluid; (b) providing a means for storing the sample fluid wherein the storage means is in fluid communication with the first channel and provides means for introducing the sample fluid to the inlet channel, wherein the storage means contains an immiscible phase fluid with a density less than that of the sample fluid; (c) introducing the sample fluid into the storage means wherein the sample fluid flows through the less dense immiscible fluid such that the sample fluid settles at the bottom of the storage means and is subsequently introduced into the first channel; (d) flowing the sample fluid through the first channel which is in fluid communication with the second channel at a junction, wherein the junction includes a fluidic nozzle such that the sample fluid forms a plurality of highly uniform, monodisperse droplets of a predetermined size in the continuous phase.
  • Another method of forming a droplet emulsion library of a sample fluid includes, for example, (a) providing a microfluidic substrate including at least one first channel adapted to carry at least one dispersed phase sample fluid and at least one channel adapted to carry at least one continuous phase fluid; (b) providing a means for storing the sample fluid wherein the storage means is in fluid communication with the first channel and provides means for introducing the sample fluid to the inlet channel, wherein the storage means contains an immiscible phase fluid with a density greater than that of the sample fluid; (c) inserting a sample fluid introduction apparatus into the storage means wherein the sample fluid is forced through the more dense immiscible fluid by the introduction apparatus such that the sample fluid is subsequently introduced into the first channel; (d) flowing the sample fluid through the first channel which is in fluid communication with the second channel at a junction, wherein the junction includes a fluidic nozzle such that the sample fluid forms a plurality of highly uniform, monodisperse droplets of a predetermined size in the continuous phase.
  • The storage means can be a well or reservoir. The fluid introduction apparatus can be a sample tip loading pump.
  • The present invention also provides a method of forming a uniformed sized droplet emulsion library including, for example, (a) providing a means for separating droplets of similar sizes, wherein the means includes a periodic array of geometric parameters defining an obstacle matrix; (b) introducing at least one sample fluid containing various sized droplets to the separating means; (c) subjecting the sample fluid to laminar flow through the microscale obstacles within the separating means, wherein the sample fluids do not mix; and (d) separating and isolating uniformed sized droplets from within the sample fluid by deterministic lateral displacement. The means can be a microfluidic channel, microfluidic lateral diffusion device, tube, syringe, column or a capillary.
  • The present invention also provides various methods for solidifying a droplet or nanoreactor. One method includes, for example, (a) providing a microfluidic substrate including at least one inlet channel adapted to carry at least one dispersed phase sample fluid and at least one main channel adapted to carry at least one continuous phase fluid; (b) incorporating at least one solidifying agent within a sample fluid; (c) flowing the sample fluid through a first inlet channel which is in fluid communication with the main channel at a junction, such that the first sample fluid forms a plurality of highly uniform, monodisperse droplets in the continuous phase; (d) providing a means which activates the solidifying agent such that the droplet or nanoreactor forms a matrix.
  • Another method for solidifying a droplet or nanoreactor includes, for example, (a) providing a microfluidic substrate including at least two inlet channels adapted to carry at least two dispersed phase sample fluids and at least one main channel adapted to carry at least one continuous phase fluid; (b) incorporating at least one solidifying agent within at least one sample fluid; (c) flowing a first sample fluid through a first inlet channel which is in fluid communication with the main channel at a junction, such that the first sample fluid forms a plurality of highly uniform, monodisperse droplets in the continuous phase; (d) flowing a second sample fluid through a second inlet channel which is in fluid communication with the main channel at a junction, such that the second sample fluid forms a plurality of highly uniform, monodisperse droplets in the continuous phase; (e) coalescing at least one droplet formed in step (c) with at least one droplet formed in step (d) as the droplets pass through an electric field, thereby producing a droplet or nanoreactor; and (f) providing a means which activates the solidifying agent such that the droplet or nanoreactor forms a matrix.
  • The solidifying agent can be a low temperature agarose or a polymerizing solution. The means for activating the solidifying agent can be a physical or chemical means.
  • The present invention also provides various methods for introducing sample fluid to a microfluidic substrate. One method includes, for example, (a) providing a microfluidic substrate including at least one inlet channel adapted to carry at least one dispersed phase sample fluid and at least one main channel adapted to carry at least one continuous phase fluid; (b) providing a means for storing the sample fluid, wherein the storage means is in fluid communication with the inlet channel and provides a means for introducing the sample fluid into the inlet channel; (c) combining the sample fluid with at least one immiscible phase fluid within the storage means, wherein the immiscible phase fluid has a density different from that of the sample fluid such that the fluids separate into distinct layers; (d) providing a force such that the immiscible phase fluid forces the sample fluid completely into the inlet channel of the microfluidic substrate.
  • Another method for introducing sample fluid to a microfluidic substrate includes, for example, (a) providing a microfluidic substrate including at least one inlet channel adapted to carry at least one dispersed phase sample fluid and at least one main channel adapted to carry at least one continuous phase fluid; (b) providing a means for storing the sample fluid, wherein the storage means is in fluid communication with the inlet channel and provides a means for introducing the sample fluid into the inlet channel; (c) combining the sample fluid with at least two immiscible phase fluids within the storage means, wherein a first immiscible phase fluid has a density greater than that of the sample fluid and a second immiscible phase fluid has a density less than that of the sample fluid such that the fluids separate into distinct layers with the sample fluid layer residing between the two immiscible phase layers; (d) providing a force such that the more dense immiscible phase fluid forces the less dense immiscible phase fluid and the sample fluid completely into the inlet channel of the microfluidic substrate.
  • The storage means can be a well or reservoir. The immiscible fluid can be biologically and/or chemically inert.
  • The present invention also provides a method of extracting biological or chemical material from within a droplet or nanoreactor including, for example, (a) providing one or more droplets or nanoreactors formed within a microfluidic substrate; (b) combining the droplets or nanoreactors with a first immiscible fluid with a density greater than that of the droplets or nanoreactors such that the droplets or nanoreactors and immiscible fluid form separate layers; (c) removing the droplet or nanoreactor layer formed in step (b); (d) combining and mixing the droplet or nanoreactor layer with a second immiscible fluid including a destabilizing surfactant such that the droplets or nanoreactors and immiscible fluid form separate layers; and (e) removing the resulting aqueous layer formed step (d), thereby extracting the biological or chemical material from within the droplet or nanoreactor. The immiscible fluid can be biologically and/or chemically inert. The destabilizing surfactant can be a perfluorinated alcohol. The droplets or nanoreactors can reside on top of the immiscible fluids within the separate layers.
  • The present invention also provides various methods for amplifying DNA. One method includes, for example, (a) providing a microfluidic substrate including at least one inlet channel adapted to carry at least one dispersed phase sample fluid and at least one main channel adapted to carry at least one continuous phase fluid, wherein the main channel includes a serpentine line with heating and cooling regions; (b) flowing a sample fluid including one more target DNA molecules to be amplified, PCR primer pair sets, dNTPs, enzymes and buffer components effective to permit PCR amplification through an inlet channel which is in fluid communication with the main channel at a junction, such that the sample fluid forms a plurality of highly uniform, monodisperse droplets in the continuous phase, wherein the junction is heated to 95° C. to provide a hot start, wherein a portion of at least one of the PCR primer pair sets is attached to a semi-solid substrate; (c) reacting the contents of the droplets for at least twenty heating and cooling cycles to permit the PCR amplification of the target DNA molecules, such that the amplified target DNA molecules are attached to a semi-solid substrate.
  • Another method of amplifying DNA includes, for example, (a) providing a microfluidic substrate including at least two inlet channels adapted to carry at least two dispersed phase sample fluids and at least one main channel adapted to carry at least one continuous phase fluid, wherein the main channel includes a serpentine line with heating and cooling regions; (b) flowing a first sample fluid including one more target DNA molecules to be amplified through a first inlet channel which is in fluid communication with the main channel at a junction, such that the first sample fluid forms a plurality of highly uniform, monodisperse droplets in the continuous phase; (c) flowing a second sample fluid including PCR primer pair sets, dNTPs, enzymes and buffer components effective to permit PCR amplification through a second inlet channel which is in fluid communication with the main channel at a junction, such that the second sample fluid forms a plurality of highly uniform, monodisperse droplets in the continuous phase, wherein the junction is heated to 95° C. to provide a hot start and wherein a portion of at least one of the PCR primer pair sets is attached to a semi-solid substrate; (d) coalescing the droplets including the DNA molecules from step (b) with the droplets including the PCR primer pair sets, dNTPs, enzymes and buffer components from step (c) within an electric field; (e) reacting the contents of the droplets for at least twenty heating and cooling cycles to permit the PCR amplification of the target DNA molecules, such that the amplified target DNA molecules are attached to a semi-solid substrate.
  • Another method of amplifying DNA includes, for example, (a) providing a microfluidic substrate including at least one inlet channel adapted to carry at least one dispersed phase sample fluid and at least one main channel adapted to carry at least one continuous phase fluid, wherein the main channel includes a serpentine line with heating and cooling regions; (b) flowing a sample fluid including one more target DNA molecules to be sequenced, primer pair sets, dNTPs, enzymes and buffer components effective to permit isothermal amplification through an inlet channel which is in fluid communication with the main channel at a junction, such that the sample fluid forms a plurality of highly uniform, monodisperse droplets in the continuous phase, wherein a portion of at least one of the primer pair sets is attached to a semi-solid substrate; (c) reacting the contents of the droplets to permit the isothermal amplification of the target DNA molecules, such that the amplified target DNA molecules are attached to a semi-solid substrate.
  • Another method of amplifying DNA includes, for example, (a) providing a microfluidic substrate including at least two inlet channels adapted to carry at least two dispersed phase sample fluids and at least one main channel adapted to carry at least one continuous phase fluid, wherein the main channel includes a serpentine line with heating and cooling regions; (b) flowing a first sample fluid including one more target DNA molecules to be amplified through a first inlet channel which is in fluid communication with the main channel at a junction, such that the first sample fluid forms a plurality of highly uniform, monodisperse droplets in the continuous phase; (c) flowing a second sample fluid including primer pair sets, dNTPs, enzymes and buffer components effective to permit isothermal amplification through a second inlet channel which is in fluid communication with the main channel at a junction, such that the second sample fluid forms a plurality of highly uniform, monodisperse droplets in the continuous phase, wherein a portion of at least one of the primer pair sets is attached to a semi-solid substrate; (d) coalescing the droplets including the DNA molecules from step (b) with the droplets including the primer pair sets, dNTPs, enzymes and buffer components from step (c) within an electric field; (e) reacting the contents of the droplets for at least twenty heating and cooling cycles to permit the isothermal amplification of the target DNA molecules, such that the amplified target DNA molecules are attached to a semi-solid substrate.
  • The present invention also provides various methods for sequencing DNA. One method includes, for example, (a) providing a microfluidic substrate including at least two inlet channels adapted to carry at least two dispersed phase sample fluids and at least one main channel adapted to carry at least one continuous phase fluid, wherein the main channel includes a serpentine line with heating and cooling regions; (b) flowing a first sample fluid including one more target DNA molecules to be sequenced, PCR primer pair sets, dNTPs, enzymes and buffer components effective to permit PCR amplification through a first inlet channel which is in fluid communication with the main channel at a junction, such that the first sample fluid forms a plurality of highly uniform, monodisperse droplets in the continuous phase, wherein the junction is heated to 95° C. to provide a hot start; (c) reacting the contents of the droplets for at least twenty heating and cooling cycles to permit the PCR amplification of the target DNA molecules; (d) flowing a second sample fluid including shrimp alkaline phosphatase and exonuclease I through a second inlet channel which is in fluid communication with the main channel at a junction, such that the second sample fluid forms a plurality of highly uniform, monodisperse droplets in the continuous phase; (e) coalescing the droplets including the amplified DNA molecules from step (c) with the droplets including the shrimp alkaline phosphatase and exonuclease I from step (d) within an electric field and reacting the contents of the combined droplets at 37° C.; (f) inactivating the enzymes within the droplets to terminate the reaction from step (e) by heating the reacted droplets to 95° C.; (g) flowing a third sample fluid including universal sequencing primers, labeled ddNTPs and buffer effective to permit nucleotide sequencing through a third inlet channel which is in fluid communication with the main channel at a junction, such that the third sample fluid forms a plurality of highly uniform, monodisperse droplets in the continuous phase; (h) coalescing the droplets including the amplified DNA molecules from step (f) with the droplets including the universal sequencing primers, labeled ddNTPs and sequencing buffer from step (g) within an electric field; (i) reacting the contents of the coalesced droplets for at least twenty heating and cooling cycles to permit the sequencing reaction to proceed; and (j) analyzing the sequencing reaction to determine the nucleic acid sequence of the target DNA.
  • Another method of sequencing DNA includes, for example, (a) providing a microfluidic substrate including at least two inlet channels adapted to carry at least two dispersed phase sample fluids and at least one main channel adapted to carry at least one continuous phase fluid, wherein the main channel includes a serpentine line with heating and cooling regions; (b) flowing a first sample fluid including one more target DNA molecules to be sequenced, primer pair sets, dNTPs, enzymes and buffer components effective to permit isothermal amplification through a first inlet channel which is in fluid communication with the main channel at a junction, such that the first sample fluid forms a plurality of highly uniform, monodisperse droplets in the continuous phase; (c) reacting the contents of the droplets to permit the isothermal amplification of the target DNA molecules; (d) flowing a second sample fluid including shrimp alkaline phosphatase and exonuclease I through a second inlet channel which is in fluid communication with the main channel at a junction, such that the second sample fluid forms a plurality of highly uniform, monodisperse droplets in the continuous phase; (e) coalescing the droplets including the amplified DNA molecules from step (c) with the droplets including the shrimp alkaline phosphatase and exonuclease I from step (d) within an electric field and reacting the contents of the combined droplets at 37° C.; (f) inactivating the enzymes within the droplets to terminate the reaction from step (e) by heating the reacted droplets to 95° C.; (g) flowing a third sample fluid including universal sequencing primers, labeled ddNTPs and buffer effective to permit nucleotide sequencing through a third inlet channel which is in fluid communication with the main channel at a junction, such that the third sample fluid forms a plurality of highly uniform, monodisperse droplets in the continuous phase; (h) coalescing the droplets including the amplified DNA molecules from step (f) with the droplets including the universal sequencing primers, labeled ddNTPs and sequencing buffer from step (g) within an electric field; (i) reacting the contents of the coalesced droplets for at least twenty heating and cooling cycles to permit the sequencing reaction to proceed; and (j) analyzing the sequencing reaction to determine the nucleic acid sequence of the target DNA.
  • Each primer or ddNTP can be labeled with a unique label. A portion of at least one of the primer pair sets can be attached to a semi-solid substrate. The semi-solid substrate can be a bead. The junction between the inlet channel and main channel can include a fluidic nozzle for flow focusing.
  • The present invention also provides a method of detecting a single nucleotide polymorphism (SNP) including, for example, (a) providing a microfluidic substrate including at least one inlet channel adapted to carry at least one dispersed phase sample fluid and at least one main channel adapted to carry at least one continuous phase fluid, wherein the main channel includes a serpentine line with heating and cooling regions; (b) flowing a sample fluid including one more target DNA molecules to be analyzed, labeled PCR primer pair sets, dNTPs, enzymes and buffer components effective to permit PCR amplification through an inlet channel which is in fluid communication with the main channel at a junction, such that the sample fluid forms a plurality of highly uniform, monodisperse droplets in the continuous phase, wherein the junction is heated to 95° C. to provide a hot start; (c) reacting the contents of the droplets for at least twenty heating and cooling cycles to permit the PCR amplification of the target DNA molecules; and (d) analyzing the reaction to determine the presence of absence of a SNP.
  • The present invention also provides a method of forming enzyme emulsions including, for example, (a) providing a microfluidic substrate including at least one inlet channel adapted to carry at least one dispersed phase fluid and at least one main channel adapted to carry at least one continuous phase fluid; (b) flowing a sample fluid including at least one cell through an inlet channel which is in fluid communication with the main channel at a junction, such that the sample fluid forms a plurality of highly uniform, monodisperse droplets in the continuous phase, wherein the cells includes at least one enzyme which can be secreted by the cells; (c) incubating the cells within the plurality of droplets such that the cells secrete at least one enzyme into the droplet, wherein the cells within the droplets can be incubated within or outside of the microfluidic substrate.
  • The present invention also provides various methods for detecting enzyme activity. One method includes, for example, (a) providing a microfluidic substrate including at least one inlet channel adapted to carry at least one dispersed phase sample fluid and at least one main channel adapted to carry at least one continuous phase fluid; (b) flowing a sample fluid including at least one cell and at least one labeled enzyme substrate through a inlet channel which is in fluid communication with the main channel at a junction, such that the sample fluid forms a plurality of highly uniform, monodisperse droplets in the continuous phase wherein the cells includes at least one enzyme which can be secreted by the cells; (c) collecting the droplets and incubating them on the microfluidic substrate or off the microfluidic substrate at a temperature and duration appropriate to permit the enzyme/substrate reaction to occur; (d) reintroducing the droplets onto the microfluidic substrate if the droplets removed from the microfluidic substrate in step (c); (e) analyzing the contents of the coalesced droplets using the detection module to detect the presence or absence of an enzyme/substrate reaction; and, (f) selecting the droplets which contain the presence of an enzyme/substrate reaction.
  • Another method of detecting enzyme activity includes, for example, (a) providing a microfluidic substrate including at least two inlet channels adapted to carry at least two dispersed phase sample fluids and at least one main channel adapted to carry at least one continuous phase fluid, wherein the main channel includes a serpentine line with heating and cooling regions; (b) flowing a first sample fluid including at least one cell through a first inlet channel which is in fluid communication with the main channel at a junction, such that the first sample fluid forms a plurality of highly uniform, monodisperse droplets in the continuous phase, wherein the cells includes at least one enzyme which can be secreted by the cells; (c) flowing a second sample fluid including at least one labeled enzyme substrate through a second inlet channel which is in fluid communication with the main channel at a junction, such that the second sample fluid forms a plurality of highly uniform, monodisperse droplets in the continuous phase; (d) coalescing the droplets including the secreted enzymes from step (b) with the droplets including the labeled substrate from step (c) within an electric field; (e) analyzing the contents of the coalesced droplets using a detection module to detect the presence or absence of an enzyme/substrate reaction; and, (f) selecting the coalesced droplets which contain the presence of an enzyme/substrate reaction.
  • The method can further include incubating the cells within the plurality of droplets from step (b) such that the cells secrete at least one enzyme into the droplet, prior to flowing the second sample fluid in step (c). The cells can be incubated within the plurality of droplets within the microfluidic substrate or outside the microfluidic substrate. The method can further include lysing the cells within the plurality of droplets from step (b) such that the cells release at least one enzyme into the plurality droplet, prior to flowing the second sample fluid in step (c). The method can further include selecting the coalesced droplets which contain the enzyme with the highest level of activity for the labeled substrate.
  • The channels (e.g., inlet and main channels) can be coated with an anti-wetting or blocking agent for the dispersed phase. The channels can be coated with a silica primer layer followed by a perfluoroalkylalkylsilane compound, an amorphous soluble perfluoropolymer, BSA, PEG-silane or fluorosilane. Preferably, the channels are coated with a silica primer layer followed by a perfluoroalkylalkylsilane compound.
  • The plurality of droplets or nanoreactors can include a biological/chemical material. The biological/chemical material can be tissues, cells, particles, proteins, antibodies, amino acids, nucleotides, small molecules, or pharmaceuticals. The biological/chemical material can include a label. Each biological/chemical material can be labeled with a unique label, such as a protein, a DNA tag, a dye, a quantum dot or a radio frequency identification tag. Each plurality of droplets can be labeled, such as a change in viscosity, a change in opacity, a change in volume, a change in density, a change in pH, a change in temperature, a change in dielectric constant, a change in conductivity, a change in the amount of beads present in the solution, a change in the amount of flocculent in the solution, a change in the amount of a selected solvent within the solution or the change in the amount of any measurable entity within the solution, or combinations thereof. Labels can be detected by any means know in the art, including for example, fluorescence polarization, fluorescence intensity, fluorescence lifetime, fluorescence energy transfer, pH, ionic content, temperature or combinations thereof.
  • The continuous phase can be a non-polar solvent. The continuous phase can be a fluorocarbon oil. The continuous phase can further include one or more additives. Preferably, the additive is a fluorosurfactant. More preferably, the fluorosurfactant is a perfluorinated polyether. The flow of the dispersed phase fluid and the continuous phase fluid can be pressure driven.
  • The methods of the present invention can include one or more coalescence steps Coalescing droplets or nanoreactors can occur within a coalescence module in fluid communication with the main channel on the microfluidic substrate. The coalescence module can include one or more electrodes that generate an electric field. The channel dimensions in the coalescence module can include an expanded portion of the main channel between the electrodes to bring successive droplets into proximity. The channel dimensions in the coalescence module can include a narrowed portion of the main channel to center droplets within the main channel prior to a expanded portion of the main channel. The droplets or nanoreactors to be coalesced during coalescing steps within the coalescence module have no charge and are coalesced within an electric field.
  • The methods of the present invention can include one or more detection or analysis steps. Interrogating droplet or nanoreactor for at least one predetermined characteristic of the present invention can occur within a detection module in fluid communication with the main channel on the microfluidic substrate. The detection module including a detection apparatus for evaluating the contents or characteristics of the droplet or nanoreactor. The detection apparatus includes an optical or electrical detector.
  • The methods of the present invention can include one or more sorting steps. Sorting a droplet or nanoreactor into or away from a collection module can occur within a sorting module in fluid communication with the main channel on the microfluidic substrate in response to the contents or characterization of the droplet or nanoreactor evaluated in the detection module. The sorting module includes a sorting apparatus adapted to direct the droplet or nanoreactor into or away from the collection module. The sorting apparatus can include one or more electrodes that generate an electric field. The droplets or nanoreactors to be sorted during sorting steps within the sorting module have no charge and are sorted within an electric field.
  • The present invention also provides a methods of detecting or tracking an aqueous solution. A method for detecting an aqueous solution includes, for example, introducing a label into the aqueous solution such that the label alters a physical property of the aqueous solution thereby permitting the detection of aqueous solution including the label. A method of tracking an aqueous solution includes, for example, introducing a label into the aqueous solution such that the label alters a physical property of the aqueous solution thereby permitting the tracking of aqueous solution including the label.
  • The label can be a change in viscosity, a change in opacity, a change in volume, a change in density, a change in pH, a change in temperature, a change in dielectric constant, a change in conductivity, a change in the amount of beads present in the solution, a change in the amount of flocculent in the solution, a change in the amount of a selected solvent within the solution or the change in the amount of any measurable entity within the solution, or combinations thereof. The label can be inducible. The inducible label is detectable when combined with at least one additional solution wherein the solution contains at least one entity to which can alter a property of the label such that the aqueous solution including the label is detectable. The label can alter the light scattering properties of the aqueous solution
  • The labeled aqueous solution can be a labeled aqueous droplet. The labeled aqueous droplet can be detected within a microfluidic substrate.
  • The aqueous solution can further include a biological/chemical material. The biological/chemical material can be tissues, cells, particles, proteins, antibodies, amino acids, nucleotides, small molecules, or pharmaceuticals. The biological/chemical material can include a label. Preferable the label is unique for each biological/chemical material and different from the aqueous solution label. The label can be a protein, a DNA tag, a dye, a quantum dot or a radio frequency identification tag. The label can be detected by any means known in the art including, for example, fluorescence polarization, fluorescence intensity, fluorescence lifetime, fluorescence energy transfer, pH, ionic content, temperature or combinations thereof.
  • Unless otherwise defmed, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, suitable methods and materials are described below. All publications, patent applications, patents, and other references mentioned herein are incorporated by reference in their entirety. In the case of conflict, the present specification, including definitions, will control. In addition, the materials, methods, and examples are illustrative only and not intended to be limiting.
  • Other features and advantages of the invention will be apparent from the following detailed description and claims.
  • BRIEF DESCRIPTION OF DRAWINGS
  • Non-limiting embodiments of the present invention will be described by way of example with reference to the accompanying drawings, which are schematic and are not intended to be drawn to scale. In the drawings, each identical or nearly identical component illustrated is typically represented by a single numeral. For the purposes of clarity, not every component is labeled in every drawing, nor is every component of each embodiment of the invention shown where illustration is not necessary to allow those of ordinary skill in the art to understand the invention. In the drawings:
  • FIG. 1 is an schematic illustrating the interacting modules of a microfluidic device of the present invention.
  • FIG. 2, Panels A and B, show dual and single oil versions of the nozzle concept using a small ferrule for the nozzle section. Panels C and D, show the same nozzles made directly out of small bore tubing (the “nozzle” runs the entire length of the tubing).
  • FIG. 3 shows the expansion of the nozzle ferrule concept shown in FIGS. 2A and 2B.
  • FIG. 4 shows the expansion of the nozzle section contained in the ferrule.
  • FIG. 5, 5A shows the operation of the nozzle in Aspiration Mode and 5B shows the operation of the nozzle in Injection Mode.
  • FIG. 6 shows a reservoir based sample emulsification where the well is initially filled with a fluid with lower density than the sample to be introduced.
  • FIG. 7 illustrates a sample introduction when the sample is less dense than the fluid in the sample port, which is an alternative scheme used to introduce samples that are less dense than the oil used to emulsify the sample.
  • FIG. 8 illustrates a nozzle that formed directly into the fitting used to connect the collection syringe to a syringe tip (e.g. capillary tubing) in order to create a monodisperse emulsion directly from a library well. Step 1 shows the aspiration of the sample can be accomplished by running the collection syringe in withdrawal mode at a flow rate (Q3) above the flow rate of the two oil syringes. Step 2 shows the appropriate volume of sample loaded into the capillary tubing, and the capillary tubing would be removed from the sample well, an air bubble, and possibly a cleaning solution would be aspirated. Step 3 shows when almost all of the sample has been emulsified, the collection syringe withdrawal rate would either be reduced below the oil flow rates, stopped, or set to infuse at some nominal rate.
  • FIG. 9 illustrates a two phase system where the reagent is injected on top of the 2nd, immiscible phase. (A) During injection, prior to transition from 1st phase to 2nd phase. (B) 2nd phase just entering the transfer lines. (C) 2nd phase has completely filled the transfer line and pushed the entire volume of reagent through the system.
  • FIG. 10 illustrates sandwiching an ultra-small volume of fluid (i.e., sub-nanoliter) between two solutions having different densities.
  • FIG. 11 illustrates possible interconnect designs for use with PDMS devices.
  • FIG. 12 illustrates self-alignment of fluidic interconnect
  • FIG. 13 illustrates the interconnects needed for each tube molded into a single monolithic self-aligned part.
  • FIG. 14 shows a schematic of a molding tool based on this concept. The pins (orange) are captured within an elestomeric molded sleeve and a compression plate made from a rigid backer plate and foam rubber is used to apply gentle even pressure to the pins and generate the force needed to make the pins uniformly contact the master.
  • FIG. 15 is a schematic diagram of an improved coalescence module that shows an optional small constriction (neckdown) just before this expansion can be used to better align the droplets on their way into the coalescence point.
  • FIG. 16 illustrates that fluorescence polarization (FP) measures the tumbling rate of a compound in solution and is a function of it's volume (in most cases, volume is correlated with MW)
  • FIG. 17 shows the fluorescence polarization of three different compounds. Results of reading polarization in 18,000 drops containing 3 distinct species (FC, BTFC, and BTFC bound to SA). Ideal for reading results of drug screening assays, protein interactions, or DNA hybridization.
  • FIG. 18, A) illustrates encoding a liquid solution using both overall fluorescence polarization and overall dye intensity within droplets; B) shows that multiple colors of fluorescence polarization and FI increases the number of possible labels. Ten intensity levels with ten fluorescence polarization levels on two colors yields 10,000 labels
  • FIG. 19 illustrates FP coding using dyes having different fluorescence lifetimes. These were made one element at a time, stored in a single syringe overnight and then loaded back on chip. The codes were made by using a ratio of two different dyes, one with a short lifetime and hence high FP and one with a long lifetime and correspondingly low FP. The mixtures have intermediate FP signals. The intensity is tuned by controlling the overall concentration of the two dyes.
  • FIG. 20A-20D illustrate the sorting and/or splitting of droplets in accordance with another embodiment of the invention
  • FIG. 21A-F shows the possible flow geometries used in an asymmetric sorting application.
  • FIG. 22 shows the possible electrode geometries used in an asymmetric sorting application. Panel A shows the design using sharp tipped electrodes. Panel B shows broad tipped electrodes to increase the interaction time between the droplets and the electric field (the tips could be many drop diameters long). Panel C shows electrodes straddling the collection line. Panel D shows electrodes on opposite sides of the main channel. Panel E shows an Asymmetric Electrode Pair (the asymmetry may be present on any of the other electrode pair layouts as well).
  • FIG. 23 shows a schematic of a device that split droplets, performs different experiments on the two daughter droplets and then reorders so that they pass sequential through the detector
  • FIG. 24, (Panel A) shows geometric parameters defining the obstacle matrix. (Panel B) shows three fluid streams. (Panel C) shows a particle with a radius that is larger than lane 1 follows a streamline passing through the particle's center (black dot).
  • FIG. 25 shows high-resolution separation of fluorescent microspheres with diameters of 0.80 um (green), 0.90 um (red), and 1.03 um (yellow), with a matrix of varying gap size.
  • FIG. 26 is a schematic illustrating the separation by deterministic lateral displacement in an array of microposts, with an example row shift fraction of one-third.
  • FIG. 27 shows a dideoxynucleotide sequencing on a microfabricated chip. Shown is one embodiment for a DNA sequencing chip design. Template DNA and primers are combined at step ‘add 1’ and the reaction is incubated at 95° C. for a hot start (position 1). The reaction then cycles 20-30 times (position 2) before the addition of SAP and ExoI at ‘add 2.’ The reaction is incubated at 37° C. for a predefined time-period and then the SAP and ExoI enzymes are inactivated at 95° C. (position ‘4’). The SAP/ExoI procedure degrades nucleotides and single-stranded DNA (primers) remaining after PCR. The universal sequencing primers, ddNTPs and buffers are added at ‘add 3,’ and the PCR sequencing reaction is allowed to proceed at position ‘5.’ The final reaction product is collected and can be stored off-chip.
  • FIG. 28, Panel A shows a schematic of the TempliPhi amplification process using rolling circle amplification. Panel B illustrates a transcription mediated reaction. Panel C illustrates strand-displacement amplification. Panel D shows a schematic diagram of helicase-dependent amplification.
  • FIG. 29 illustrates emulsion-based sample preparation, sample preparation and DNA sequencing. Random libraries of DNA fragments are generated by shearing an entire genome and isolating single DNA molecules by limiting dilution.
  • FIG. 30 shows one method for isolating antibodies on a microfluidic device.
  • FIG. 31 shows an alternate method for isolating antibodies on a microfluidic device.
  • FIG. 32 shows the method of the present invention for isolating antibodies on the microfluidic device. The right panel is a diagram of individual steps proposed to amplify signal of interacting antibody and antigen. The left panel is a schematic as would be designed for a chip to be used on microfluidic device.
  • FIG. 33 shows the genetic selection for full length antibody clones. A genetic selection can be used to enrich for full-length antibody clones by transforming E. coli and selecting for clones able to grow on medium in which a suitable sugar is the only carbon source.
  • FIG. 34 is a schematic representation of a multi-step chip according to the invention. 1st stage: droplets are sent into C2, the collection port; 2nd stage: 1st stage emulsion collected into C2 is reinjected back into the chip and merged with the droplets formed in the second stage nozzle.
  • FIG. 35 shows karyotyping using spectral probes that allow all 23 pairs of human chromosomes to be seen at one time, with each pair of chromosomes painted in a difference fluorescent color.
  • FIG. 36 shows the future BioBased Economy with six building blocks based on renewable biomass.
  • FIG. 37 shows the 8 non-renewable building blocks based on petroleum.
  • DETAILED DESCRIPTION
  • The microfluidic devices and methods of use described herein are based on the creation and electrical manipulation of aqueous phase droplets completely encapsulated by an inert immiscible oil stream. This combination enables precise droplet generation, highly efficient, electrically addressable, droplet coalescence, and controllable, electrically addressable single droplet sorting. The microfluidic devices include one or more channels and modules. A schematic illustrating one example of interacting modules of a microfluidic substrate is shown in FIG. 1. The integration of these modules is an essential enabling technology for a droplet based, high-throughput microfluidic reactor system.
  • The microfluidic devices of the present invention can be utilized for numerous biological, chemical, or diagnostic applications, as described in further detail herein.
  • Substrates
  • The microfluidic device of the present invention includes one or more analysis units. An “analysis unit” is a microsubstrate, e.g., a microchip. The terms microsubstrate, substrate, microchip, and chip are used interchangeably herein. The analysis unit includes at least one inlet channel, at least one main channel, at least one inlet module, at least one coalescence module, and at least one detection module. The analysis unit can further includes one or more sorting modules. The sorting module can be in fluid communication with branch channels which are in fluid communication with one or more outlet modules (collection module or waste module). For sorting applications, at least one detection module cooperates with at least one sorting module to divert flow via a detector-originated signal. It shall be appreciated that the “modules” and “channels” are in fluid communication with each other and therefore may overlap; i.e., there may be no clear boundary where a module or channel begins or ends. A plurality of analysis units of the invention may be combined in one device. The analysis unit and specific modules are described in further detail herein.
  • The dimensions of the substrate are those of typical microchips, ranging between about 0.5 cm to about 15 cm per side and about I micron to about I cm in thickness. A substrate can be transparent and can be covered with a material having transparent properties, such as a glass coverslip, to permit detection of a reporter, for example, by an optical device such as an optical microscope. The material can be perforated for functional interconnects, such as fluidic, electrical, and/or optical interconnects, and sealed to the back interface of the device so that the junction of the interconnects to the device is leak-proof. Such a device can allow for application of high pressure to fluid channels without leaking.
  • A variety of materials and methods, according to certain aspects of the invention, can be used to form any of the described components of the systems and devices of the invention. In some cases, the various materials selected lend themselves to various methods. For example, various components of the invention can be formed from solid materials, in which the channels can be formed via molding, micromachining, film deposition processes such as spin coating and chemical vapor deposition, laser fabrication, photolithographic techniques, etching methods including wet chemical or plasma processes, and the like. See, for example, Scientific American, 248:44-55, 1983 (Angell, et al). At least a portion of the fluidic system can be formed of silicone by molding a silicone chip. Technologies for precise and efficient formation of various fluidic systems and devices of the invention from silicone are known. Various components of the systems and devices of the invention can also be formed of a polymer, for example, an elastomeric polymer such as polydimethylsiloxane (“PDMS”), polytetrafluoroethylene (“PTFE”) or Teflon®, or the like.
  • The channels of the invention can be formed, for example by etching a silicon chip using conventional photolithography techniques, or using a micromachining technology called “soft lithography” as described by Whitesides and Xia, Angewandte Chemie International Edition 37, 550 (1998). These and other methods may be used to provide inexpensive miniaturized devices, and in the case of soft lithography, can provide robust devices having beneficial properties such as improved flexibility, stability, and mechanical strength. When optical detection is employed, the invention also provides minimal light scatter from molecule, cell, small molecule or particle suspension and chamber material.
  • Different components can be formed of different materials. For example, a base portion including a bottom wall and side walls can be formed from an opaque material such as silicone or PDMS, and a top portion can be formed from a transparent or at least partially transparent material, such as glass or a transparent polymer, for observation and/or control of the fluidic process. Components can be coated so as to expose a desired chemical functionality to fluids that contact interior channel walls, where the base supporting material does not have a precise, desired functionality. For example, components can be formed as illustrated, with interior channel walls coated with another material. Material used to form various components of the systems and devices of the invention, e.g., materials used to coat interior walls of fluid channels, may desirably be selected from among those materials that will not adversely affect or be affected by fluid flowing through the fluidic system, e.g., material(s) that is chemically inert in the presence of fluids to be used within the device.
  • Various components of the invention when formed from polymeric and/or flexible and/or elastomeric materials, and can be conveniently formed of a hardenable fluid, facilitating formation via molding (e.g. replica molding, injection molding, cast molding, etc.). The hardenable fluid can be essentially any fluid that can be induced to solidify, or that spontaneously solidifies, into a solid capable of containing and/or transporting fluids contemplated for use in and with the fluidic network. In one embodiment, the hardenable fluid comprises a polymeric liquid or a liquid polymeric precursor (i.e. a “prepolymer”). Suitable polymeric liquids can include, for example, thermoplastic polymers, thermoset polymers, or mixture of such polymers heated above their melting point. As another example, a suitable polymeric liquid may include a solution of one or more polymers in a suitable solvent, which solution forms a solid polymeric material upon removal of the solvent, for example, by evaporation. Such polymeric materials, which can be solidified from, for example, a melt state or by solvent evaporation, are well known to those of ordinary skill in the art. A variety of polymeric materials, many of which are elastomeric, are suitable, and are also suitable for forming molds or mold masters, for embodiments where one or both of the mold masters is composed of an elastomeric material. A non-limiting list of examples of such polymers includes polymers of the general classes of silicone polymers, epoxy polymers, and acrylate polymers. Epoxy polymers are characterized by the presence of a three-membered cyclic ether group commonly referred to as an epoxy group, 1,2-epoxide, or oxirane. For example, diglycidyl ethers of bisphenol A can be used, in addition to compounds based on aromatic amine, triazine, and cycloaliphatic backbones. Another example includes the well-known Novolac polymers. Non-limiting examples of silicone elastomers suitable for use according to the invention include those formed from precursors including the chlorosilanes such as methylchlorosilanes, ethylchlorosilanes, phenylchlorosilanes, etc.
  • Silicone polymers are preferred, for example, the silicone elastomer polydimethylsiloxane. Non-limiting examples of PDMS polymers include those sold under the trademark Sylgard by Dow Chemical Co., Midland, Mich., and particularly Sylgard 182, Sylgard 184, and Sylgard 186. Silicone polymers including PDMS have several beneficial properties simplifying formation of the microfluidic structures of the invention. For instance, such materials are inexpensive, readily available, and can be solidified from a prepolymeric liquid via curing with heat. For example, PDMSs are typically curable by exposure of the prepolymeric liquid to temperatures of about, for example, about 65° C. to about 75° C. for exposure times of, for example, about an hour. Also, silicone polymers, such as PDMS, can be elastomeric and thus may be useful for forming very small features with relatively high aspect ratios, necessary in certain embodiments of the invention. Flexible (e.g., elastomeric) molds or masters can be advantageous in this regard.
  • The present invention provides improved methods of bonding PDMS to incompatible media. Normal methods of bonding various materials (plastic, metals, etc) directly to materials such as PDMS, silicone, Teflon, and PEEK using traditional bonding practices (adhesives, epoxies, etc) do not work well due to the poor adhesion of the bonding agent to materials such as PDMS. Normal surface preparation by commercially available surface activators has not worked well in microfluidic device manufacturing. This problem is eliminated by treating the PDMS surface to be bonded with high intensity oxygen or air plasma. The process converts the top layer of PDMS to glass which bonds extremely well with normal adhesives. Tests using this method to bond external fluid lines to PDMS using a UV-cure adhesive (Loctite 352, 363, and others) resulted in a bond that is stronger than the PDMS substrate, resulting in fracture of the PDMS prior to failure of the bond. The present method combines high radiant flux, wavelength selection, and cure exposure time to significantly enhance the bond strength of the adhesive.
  • One advantage of forming structures such as microfluidic structures of the invention from silicone polymers, such as PDMS, is the ability of such polymers to be oxidized, for example by exposure to an oxygen-containing plasma such as an air plasma, so that the oxidized structures contain, at their surface, chemical groups capable of cross-linking to other oxidized silicone polymer surfaces or to the oxidized surfaces of a variety of other polymeric and non-polymeric materials. Thus, components can be formed and then oxidized and essentially irreversibly sealed to other silicone polymer surfaces, or to the surfaces of other substrates reactive with the oxidized silicone polymer surfaces, without the need for separate adhesives or other sealing means. In most cases, sealing can be completed simply by contacting an oxidized silicone surface to another surface without the need to apply auxiliary pressure to form the seal. That is, the pre-oxidized silicone surface acts as a contact adhesive against suitable mating surfaces. Specifically, in addition to being irreversibly sealable to itself, oxidized silicone such as oxidized PDMS can also be sealed irreversibly to a range of oxidized materials other than itself including, for example, glass, silicon, silicon oxide, quartz, silicon nitride, polyethylene, polystyrene, glassy carbon, and epoxy polymers, which have been oxidized in a similar fashion to the PDMS surface (for example, via exposure to an oxygen-containing plasma). Oxidation and sealing methods useful in the context of the present invention, as well as overall molding techniques, are described in the art, for example, in an article entitled “Rapid Prototyping of Microfluidic Systems and Polydimethylsiloxane,” Anal. Chem., 70:474-480, 1998 (Duffy et al.), incorporated herein by reference.
  • Another advantage to forming microfluidic structures of the invention (or interior, fluid-contacting surfaces) from oxidized silicone polymers is that these surfaces can be much more hydrophilic than the surfaces of typical elastomeric polymers (where a hydrophilic interior surface is desired). Such hydrophilic channel surfaces can thus be more easily filled and wetted with aqueous solutions than can structures comprised of typical, unoxidized elastomeric polymers or other hydrophobic materials.
  • In one embodiment, a bottom wall is formed of a material different from one or more side walls or a top wall, or other components. For example, the interior surface of a bottom wall can comprise the surface of a silicon wafer or microchip, or other substrate. Other components can, as described above, be sealed to such alternative substrates. Where it is desired to seal a component comprising a silicone polymer (e.g. PDMS) to a substrate (bottom wall) of different material, the substrate may be selected from the group of materials to which oxidized silicone polymer is able to irreversibly seal (e.g., glass, silicon, silicon oxide, quartz, silicon nitride, polyethylene, polystyrene, epoxy polymers, and glassy carbon surfaces which have been oxidized). Alternatively, other sealing techniques can be used, as would be apparent to those of ordinary skill in the art, including, but not limited to, the use of separate adhesives, thermal bonding, solvent bonding, ultrasonic welding, etc.
  • Channels
  • The microfluidic substrates of the present invention include channels that form the boundary for a fluid. A “channel,” as used herein, means a feature on or in a substrate that at least partially directs the flow of a fluid. In some cases, the channel may be formed, at least in part, by a single component, e.g., an etched substrate or molded unit. The channel can have any cross-sectional shape, for example, circular, oval, triangular, irregular, square or rectangular (having any aspect ratio), or the like, and can be covered or uncovered (i.e., open to the external environment surrounding the channel). In embodiments where the channel is completely covered, at least one portion of the channel can have a cross-section that is completely enclosed, and/or the entire channel may be completely enclosed along its entire length with the exception of its inlet and outlet.
  • An open channel generally will include characteristics that facilitate control over fluid transport, e.g., structural characteristics (an elongated indentation) and/or physical or chemical characteristics (hydrophobicity vs. hydrophilicity) and/or other characteristics that can exert a force (e.g., a containing force) on a fluid. The fluid within the channel may partially or completely fill the channel. In some cases the fluid may be held or confined within the channel or a portion of the channel in some fashion, for example, using surface tension (e.g., such that the fluid is held within the channel within a meniscus, such as a concave or convex meniscus). In an article or substrate, some (or all) of the channels may be of a particular size or less, for example, having a largest dimension perpendicular to fluid flow of less than about 5 mm, less than about 2 mm, less than about 1 mm, less than about 500 microns, less than about 200 microns, less than about 100 microns, less than about 60 microns, less than about 50 microns, less than about 40 microns, less than about 30 microns, less than about 25 microns, less than about 10 microns, less than about 3 microns, less than about 1 micron, less than about 300 nm, less than about 100 nm, less than about 30 nm, or less than about 10 nm or less in some cases. Of course, in some cases, larger channels, tubes, etc. can be used to store fluids in bulk and/or deliver a fluid to the channel. In one embodiment, the channel is a capillary.
  • The dimensions of the channel may be chosen such that fluid is able to freely flow through the channel, for example, if the fluid contains cells. The dimensions of the channel may also be chosen, for example, to allow a certain volumetric or linear flow rate of fluid in the channel. Of course, the number of channels and the shape of the channels can be varied by any method known to those of ordinary skill in the art. In some cases, more than one channel or capillary may be used. For example, two or more channels may be used, where they are positioned inside each other, positioned adjacent to each other, etc.
  • For particles (e.g., cells) or molecules that are in droplets (i.e., deposited by the inlet module) within the flow of the main channel, the channels of the device are preferably square, with a diameter between about 2 microns and 1 mm. This geometry facilitates an orderly flow of droplets in the channels. Similarly, the volume of the detection module in an analysis device is typically in the range of between about 0.1 picoliters and 500 nanoliters.
  • A “main channel” is a channel of the device of the invention which permits the flow of molecules, cells, small molecules or particles past a coalescence module for coalescing one or more droplets, a detection module for detection (identification) or measurement of a droplet and a sorting module, if present, for sorting a droplet based on the detection in the detection module. The main channel is typically in fluid communication with the coalescence, detection and/or sorting modules, as well as, an inlet channel of the inlet module. The main channel is also typically in fluid communication with an outlet module and optionally with branch channels, each of which may have a collection module or waste module. These channels permit the flow of molecules, cells, small molecules or particles out of the main channel. An “inlet channel” permits the flow of molecules, cells, small molecules or particles into the main channel. One or more inlet channels communicate with one or more means for introducing a sample into the device of the present invention. The inlet channel communicates with the main channel at an inlet module.
  • The microfluidic substrate can also comprise one or more fluid channels to inject or remove fluid in between droplets in a droplet stream for the purpose of changing the spacing between droplets.
  • The channels of the device of the present invention can be of any geometry as described. However, the channels of the device can comprise a specific geometry such that the contents of the channel are manipulated, e.g., sorted, mixed, prevent clogging, etc.
  • A microfluidic substrate can also include a specific geometry designed in such a manner as to prevent the aggregation of biological/chemical material and keep the biological/chemical material separated from each other prior to encapsulation in droplets. The geometry of channel dimension can be changed to disturb the aggregates and break them apart by various methods, that can include, but is not limited to, geometric pinching (to force cells through a (or a series of) narrow region(s), whose dimension is smaller or comparable to the dimension of a single cell) or a barricade (place a series of barricades on the way of the moving cells to disturb the movement and break up the aggregates of cells).
  • To prevent material (e.g., cells and other particles or molecules) from adhering to the sides of the channels, the channels (and coverslip, if used) may have a coating which minimizes adhesion. Such a coating may be intrinsic to the material from which the device is manufactured, or it may be applied after the structural aspects of the channels have been microfabricated. “TEFLON” is an example of a coating that has suitable surface properties. The surface of the channels of the microfluidic device can be coated with any anti-wetting or blocking agent for the dispersed phase. The channel can be coated with any protein to prevent adhesion of the biological/chemical sample. For example, in one embodiment the channels are coated with BSA, PEG-silane and/or fluorosilane. For example, 5 mg/ml BSA is sufficient to prevent attachment and prevent clogging. In another embodiment, the channels can be coated with a cyclized transparent optical polymer obtained by copolymerization of perfluoro (alkenyl vinyl ethers), such as the type sold by Asahi Glass Co. under the trademark Cytop. In such an embodiment, the coating is applied from a 0.1-0.5 wt % solution of Cytop CTL-809M in CT-Solv 180. This solution can be injected into the channels of a microfluidic device via a plastic syringe. The device can then be heated to about 90° C. for 2 hours, followed by heating at 200° C. for an additional 2 hours. In another embodiment, the channels can be coated with a hydrophobic coating of the type sold by PPG Industries, Inc. under the trademark Aquapel (e.g., perfluoroalkylalkylsilane surface treatment of plastic and coated plastic substrate surfaces in conjunction with the use of a silica primer layer) and disclosed in U.S. Pat. No. 5,523,162, which patent is hereby incorporated by reference. By fluorinating the surfaces of the channels, the continuous phase preferentially wets the channels and allows for the stable generation and movement of droplets through the device. The low surface tension of the channel walls thereby minimizes the accumulation of channel clogging particulates.
  • The surface of the channels in the microfluidic device can be also fluorinated to prevent undesired wetting behaviors. For example, a microfluidic device can be placed in a polycarbonate dessicator with an open bottle of (tridecafluoro-1,1,2,2-tetrahydrooctyl)trichlorosilane. The dessicator is evacuated for 5 minutes, and then sealed for 20-40 minutes. The dessicator is then backfilled with air and removed. This approach uses a simple diffusion mechanism to enable facile infiltration of channels of the microfluidic device with the fluorosilane and can be readily scaled up for simultaneous device fluorination.
  • Fluids
  • The microfluidic device of the present invention is capable of controlling the direction and flow of fluids and entities within the device. The term “flow” means any movement of liquid or solid through a device or in a method of the invention, and encompasses without limitation any fluid stream, and any material moving with, within or against the stream, whether or not the material is carried by the stream. For example, the movement of molecules, beads, cells or virions through a device or in a method of the invention, e.g. through channels of a microfluidic chip of the invention, comprises a flow. This is so, according to the invention, whether or not the molecules, beads, cells or virions are carried by a stream of fluid also comprising a flow, or whether the molecules, cells or virions are caused to move by some other direct or indirect force or motivation, and whether or not the nature of any motivating force is known or understood. The application of any force may be used to provide a flow, including without limitation, pressure, capillary action, electro-osmosis, electrophoresis, dielectrophoresis, optical tweezers, and combinations thereof, without regard for any particular theory or mechanism of action, so long as molecules, cells or virions are directed for detection, measurement or sorting according to the invention. Specific flow forces are described in further detail herein.
  • The flow stream in the main channel is typically, but not necessarily, continuous and may be stopped and started, reversed or changed in speed. A liquid that does not contain sample molecules, cells or particles can be introduced into a sample inlet well or channel and directed through the inlet module, e.g., by capillary action, to hydrate and prepare the device for use. Likewise, buffer or oil can also be introduced into a main inlet region that communicates directly with the main channel to purge the device (e.g., or “dead” air) and prepare it for use. If desired, the pressure can be adjusted or equalized, for example, by adding buffer or oil to an outlet module.
  • As used herein, the term “fluid stream” or “fluidic stream” refers to the flow of a fluid, typically generally in a specific direction. The fluidic stream may be continuous and/or discontinuous. A “continuous” fluidic stream is a fluidic stream that is produced as a single entity, e. g., if a continuous fluidic stream is produced from a channel, the fluidic stream, after production, appears to be contiguous with the channel outlet. The continuous fluidic stream is also referred to as a continuous phase fluid or carrier fluid. The continuous fluidic stream may be laminar, or turbulent in some cases.
  • Similarly, a “discontinuous” fluidic stream is a fluidic stream that is not produced as a single entity. The discontinuous fluidic stream is also referred to as the dispersed phase fluid or sample fluid. A discontinuous fluidic stream may have the appearance of individual droplets, optionally surrounded by a second fluid. A “droplet,” as used herein, is an isolated portion of a first fluid that completely surrounded by a second fluid. In some cases, the droplets may be spherical or substantially spherical; however, in other cases, the droplets may be non-spherical, for example, the droplets may have the appearance of “blobs” or other irregular shapes, for instance, depending on the external environment. As used herein, a first entity is “surrounded” by a second entity if a closed loop can be drawn or idealized around the first entity through only the second entity. The dispersed phase fluid can include a biological/chemical material. The biological/chemical material can be tissues, cells, particles, proteins, antibodies, amino acids, nucleotides, small molecules, and pharmaceuticals. The biological/chemical material can include one or more labels known in the art. The label can be a DNA tag, dyes or quantum dot, or combinations thereof.
  • Droplets
  • The term “emulsion” refers to a preparation of one liquid distributed in small globules (also referred to herein as drops, droplets or NanoReactors) in the body of a second liquid. The first and second fluids are immiscible with each other. For example, the discontinuous phase can be an aqueous solution and the continuous phase can a hydrophobic fluid such as an oil. This is termed a water in oil emulsion. Alternatively, the emulsion may be a oil in water emulsion. In that example, the first liquid, which is dispersed in globules, is referred to as the discontinuous phase, whereas the second liquid is referred to as the continuous phase or the dispersion medium. The continuous phase can be an aqueous solution and the discontinuous phase is a hydrophobic fluid, such as an oil (e.g., decane, tetradecane, or hexadecane). The droplets or globules of oil in an oil in water emulsion are also referred to herein as “micelles”, whereas globules of water in a water in oil emulsion may be referred to as “reverse micelles”.
  • The fluidic droplets may each be substantially the same shape and/or size. The shape and/or size can be determined, for example, by measuring the average diameter or other characteristic dimension of the droplets. The “average diameter” of a plurality or series of droplets is the arithmetic average of the average diameters of each of the droplets. Those of ordinary skill in the art will be able to determine the average diameter (or other characteristic dimension) of a plurality or series of droplets, for example, using laser light scattering, microscopic examination, or other known techniques. The diameter of a droplet, in a non-spherical droplet, is the mathematically-defined average diameter of the droplet, integrated across the entire surface. The average diameter of a droplet (and/or of a plurality or series of droplets) may be, for example, less than about 1 mm, less than about 500 micrometers, less than about 200 micrometers, less than about 100 micrometers, less than about 75 micrometers, less than about 50 micrometers, less than about 25 micrometers, less than about 10 micrometers, or less than about 5 micrometers in some cases. The average diameter may also be at least about 1 micrometer, at least about 2 micrometers, at least about 3 micrometers, at least about 5 micrometers, at least about 10 micrometers, at least about 15 micrometers, or at least about 20 micrometers in certain cases.
  • As used herein, the term “NanoReactor” and its plural encompass the terms “droplet”, “nanodrop”, “nanodroplet”, “microdrop” or “microdroplet” as defined herein, as well as an integrated system for the manipulation and probing of droplets, as described in detail herein. Nanoreactors as described herein can be 0.1-1000 μm (e.g., 0.1, 0.2 . . . 5, 10, 15, 20, 25, 30, 35, 40, 45, 50 . . . 1000), or any size within in this range. Droplets at these dimensions tend to conform to the size and shape of the channels, while maintaining their respective volumes. Thus, as droplets move from a wider channel to a narrower channel they become longer and thinner, and vice versa.
  • The microfluidic substrate of this invention most preferably generate round, monodisperse droplets. The droplets can have a diameter that is smaller than the diameter of the microchannel; i.e., preferably 15 to 100 μm when cells are used; or 10 to 75 μm when reagents or other chemical or biological agents are used; or 100 to 1000 μm when droplets are used for sequencing reactions such that droplets will be removed and dispensed into other collection apparatuses, such as microtiter plates or utilized in sequencing devices. Monodisperse droplets are particularly preferably, e.g., in high throughput devices and other embodiments where it is desirable to generate droplets at high frequency and of high uniformity.
  • The droplet forming liquid is typically an aqueous buffer solution, such as ultrapure water (e.g., 18 mega-ohm resistivity, obtained, for example by column chromatography), 10 mM Tris HCl and 1 mM EDTA (TE) buffer, phosphate buffer saline (PBS) or acetate buffer. Any liquid or buffer that is physiologically compatible, with the population of molecules, cells or particles to be analyzed and/or sorted can be used. The fluid passing through the main channel and in which the droplets are formed is one that is immiscible with the droplet forming fluid. The fluid passing through the main channel can be a non-polar solvent, decane (e g., tetradecane or hexadecane), fluorocarbon oil, silicone oil or another oil (for example, mineral oil).
  • The dispersed phase fluid may also contain biological/chemical material (e.g., molecules, cells, or other particles) for combination, analysis and/or sorting in the device. The droplets of the dispersed phase fluid can contain more than one particle or can contain no more than one particle. For example, where the biological material comprises cells, each droplet preferably contains, on average, no more than one cell. However, in some embodiments, each droplet may contain, on average, at least 1000 cells. The droplets can be detected and/or sorted according to their contents.
  • The concentration (i.e., number) of molecules, cells or particles in a droplet can influence sorting efficiently and therefore is preferably optimized. In particular, the sample concentration should be dilute enough that most of the droplets contain no more than a single molecule, cell or particle, with only a small statistical chance that a droplet will contain two or more molecules, cells or particles. This is to ensure that for the large majority of measurements, the level of reporter measured in each droplet as it passes through the detection module corresponds to a single molecule, cell or particle and not to two or more molecules, cells or particles.
  • The parameters which govern this relationship are the volume of the droplets and the concentration of molecules, cells or particles in the sample solution. The probability that a droplet will contain two or more molecules, cells or particles (P≦2) can be expressed as
    P ≦2=1−{1+[cell]×V}×e −[cell]×V
  • where “[cell]” is the concentration of molecules, cells or particles in units of number of molecules, cells or particles per cubic micron (μm3), and V is the volume of the droplet in units of μm3.
  • It will be appreciated that P≦2 can be minimized by decreasing the concentration of molecules, cells or particles in the sample solution. However, decreasing the concentration of molecules, cells or particles in the sample solution also results in an increased volume of solution processed through the device and can result in longer run times. Accordingly, it is desirable to minimize to presence of multiple molecules, cells or particles in the droplets (thereby increasing the accuracy of the sorting) and to reduce the volume of sample, thereby permitting a sorted sample in a reasonable time in a reasonable volume containing an acceptable concentration of molecules, cells or particles.
  • The maximum tolerable P≦2 depends on the desired “purity” of the sorted sample. The “purity” in this case refers to the fraction of sorted molecules, cells or particles that posses a desired characteristic (e.g., display a particular antigen, are in a specified size range or are a particular type of molecule, cell or particle). The purity of the sorted sample is inversely proportional to P≦2. For example, in applications where high purity is not needed or desired a relatively high P≦2 (e.g., P≦2=0.2) may be acceptable. For most applications, maintaining P≦2 at or below about 0. 1, preferably at or below about 0.01, provides satisfactory results.
  • The fluids used to generate droplets in microfluidic devices are typically immiscible liquids such as oil and water. These two materials generally have very different dielectric constants associated with them. These differences can be exploited to determine droplet rate and size for every drop passing through a small section of a microfluidic device. One method to directly monitor this variation in the dielectric constant measures the change in capacitance over time between a pair of closely spaced electrodes. This change in capacitance can be detected by the change in current measured in these electrodes: i = V C t
    Where i is the current, V is the voltage applied across the electrodes, and dC/dt is the change in capacitance with time. Alternatively, the capacitance can be measured directly if a time varying voltage is applied to these same electrodes: i=CdV/dt Where C is the measured capacitance, and dV/dt is the change in voltage with time. As a first approximation, the electrode pair can be determined as a parallel plate capacitor: C = ɛ 0 k A d
  • Where ε0 is the permittivity of free space, k is the effective dielectric constant (this changes every time a droplet passes through), A is the area of the capacitor and d is the electrode separation. The current measured in the device is then plotted as a function of time.
  • The fluidic droplets may contain additional entities, for example, other chemical, biochemical, or biological entities (e.g., dissolved or suspended in the fluid), cells, particles, gases, molecules, or the like. In some cases, the droplets may each be substantially the same shape or size, as discussed above. In certain instances, the invention provides for the production of droplets consisting essentially of a substantially uniform number of entities of a species therein (i.e., molecules, cells, particles, etc.). For example, about 90%, about 93%, about 95%, about 97%, about 98%, or about 99%, or more of a plurality or series of droplets may each contain the same number of entities of a particular species. For instance, a substantial number of fluidic droplets produced, e.g., as described above, may each contain 1 entity, 2 entities, 3 entities, 4 entities, 5 entities, 7 entities, 10 entities, 15 entities, 20 entities, 25 entities, 30 entities, 40 entities, 50 entities, 60 entities, 70 entities, 80 entities, 90 entities, 100 entities, etc., where the entities are molecules or macromolecules, cells, particles, etc. In some cases, the droplets may each independently contain a range of entities, for example, less than 20 entities, less than 15 entities, less than 10 entities, less than 7 entities, less than 5 entities, or less than 3 entities in some cases. In some embodiments, a droplet may contain 100,000,000 entities. In other embodiments, a droplet may contain 1,000,000 entities.
  • In a liquid containing droplets of fluid, some of which contain a species of interest and some of which do not contain the species of interest, the droplets of fluid may be screened or sorted for those droplets of fluid containing the species as further described below (e.g., using fluorescence or other techniques such as those described above), and in some cases, the droplets may be screened or sorted for those droplets of fluid containing a particular number or range of entities of the species of interest, e.g., as previously described. Thus, in some cases, a plurality or series of fluidic droplets, some of which contain the species and some of which do not, may be enriched (or depleted) in the ratio of droplets that do contain the species, for example, by a factor of at least about 2, at least about 3, at least about 5, at least about 10, at least about 15, at least about 20, at least about 50, at least about 100, at least about 125, at least about 150, at least about 200, at least about 250, at least about 500, at least about 750, at least about 1000, at least about 2000, or at least about 5000 or more in some cases. In other cases, the enrichment (or depletion) may be in a ratio of at least about 104, at least about 105, at least about 106, at least about 107, at least about 108, at least about 109, at least about 1010, at least about 1011, at least about 1012, at least about 1013, at least about 1014, at least about 1015, or more. For example, a fluidic droplet containing a particular species may be selected from a library of fluidic droplets containing various species, where the library may have about 100, about 103, about 104, about 105, about 106, about 107, about 108, about 109, about 1010, about 1011, about 1012, about 1013, about 1014, about 1015, or more items, for example, a DNA library, an RNA library, a protein library, a combinatorial chemistry library, etc. In certain embodiments, the droplets carrying the species may then be fused, reacted, or otherwise used or processed, etc., as further described below, for example, to initiate or determine a reaction.
  • Droplets of a sample fluid can be formed within the inlet module on the microfluidic device or droplets (or droplet libraries) can be formed before the sample fluid is introduced to the microfluidic device (“off chip” droplet formation). To permit effective interdigitation, coalescence and detection, the droplets comprising each sample to be analyzed must be monodisperse. As described in more detail herein, in many applications, different samples to be analyzed are contained within droplets of different sizes. Droplet size must be highly controlled to ensure that droplets containing the correct contents for analysis and coalesced properly. As such, the present invention provides devices and methods for forming droplets and droplet libraries.
  • Devices and Methods for Forming Sample Droplets on a Microfluidic Substrate
  • The present invention provides compositions and methods for forming sample droplet emulsions on a microfluidic substrate. The present invention also provides embedded microfluidic nozzles. In order to create a monodisperse emulsion directly from a library well, this invention would form a nozzle directly into the fitting used to connect the storage well/reservoir (e.g. syringe) to a syringe tip (e.g. capillary tubing), as shown in FIGS. 2-6. FIG. 2, Panels A and B, show dual and single oil versions of the nozzle concept using a small ferrule for the nozzle section. FIG. 2, Panels C and D, show the same Nozzles made directly out of small bore tubing (the “nozzle” runs the entire length of the tubing). Both designs can form droplets identically, although the pressure drop will be higher for the tube based nozzle (bottom). FIG. 3 shows the expansion of the nozzle ferrule concept shown in FIGS. 2A and 2B. The tube based nozzles (FIG. 2C, 2D) function identically to this, except the “nozzle” runs the entire length of the tube instead of having a short transition. The ability to form droplets is identical in both cases. FIG. 4 shows the expansion of the nozzle section contained in the ferrule. The tee design in FIG. 2D has been built and tested, with a cross-section cut of this design shown in FIG. 5. FIG. 5A shows the operation of the nozzle in Aspiration Mode and FIG. 5B shows the operation of the nozzle in Injection Mode. The droplets formed are approximately 45 um in diameter, and were formed from PCR mix (210 ul/hr) and SpectraSyn-10 (600 ul/hr). Other tests have been demonstrated with Spectrasyn-2 and PCR mix. The droplets are traveling in 300 urn wide×260 urn deep channels. The Nozzle tube used was 100 um in diameter, and the fluids used were PCR Mix and Spectrasyn-10 with surfactant.
  • Since the flow is three dimensional, under this design surface wetting effects are minimized. The nozzle can be made from one or two oil lines providing constant flow of oil into the nozzle, a connection to the capillary tubing, and a connection to the storage well/reservoir (e.g. syringe). The high resolution part of the nozzle can be made out of a small bore tubing or a small, simple part molded or stamped from an appropriate material (Teflon®, plastic, metal, etc). If necessary, the nozzle itself could be formed into the tip of the ferrule using post mold processing such as laser ablation or drilling.
  • This nozzle design eliminates the surface wetting issues surrounding the quasi-2D flow associated with typical microfluidic nozzles made using soft lithography or other standard microfluidic chip manufacturing techniques. This is because the nozzle design is fully 3-dimensional, resulting is a complete isolation of the nozzle section from the continuous aqueous phase. This same design can also be used for generation of emulsions required for immediate use, where the aqueous line would be attached directly to a syringe and the outlet of the nozzle would be used to transport the emulsion to the point of use (e.g. into a microfluidic PCR chip, delay line, etc).
  • In another embodiment, the present invention provides compositions and methods to directly emulsify library elements from standard library storage geometries (e.g. 96 well plates, etc). In order to create a monodisperse emulsion from fluids contained in a library well plate, this invention would include microfluidic based nozzles manufactured simultaneously with an appropriately designed fluidic interconnect or well. FIGS. 6 and 7 present two possible approaches to interface with the nozzle.
  • FIG. 6 shows a reservoir based sample emulsification. In FIG. 6, the well is initially filled with a fluid with lower density than the sample to be introduced. The operation of this device would be very similar to the device described above, with the exception that the sample would be introduced into a port instead of being directly aspirated from a sample well. This could either be emulsification oil obtained directly from the nozzle, or a different material that is loaded or flowing into the well automatically. The oil lines would begin flowing at their prescribed rates (FIG. 6 e), while the collection or waste port would begin withdrawing at a rate corresponding to the total oil flow plus the desired sample flow. Once the flow has been established, the sample would be introduced into the port either manually (e.g. a pipette) or with a robotic sample handling system. The sample volume permitted would be dependent on the port volume. Since the sample is more dense than the fluid in the well, it would settle into the bottom of the well and be transported to the nozzle (FIGS. 6 a-6 d). During this time, either the waste (used during startup only if transients cause problems) or the collection port would be withdrawing emulsified sample and stored. When the sample is completely emulsified the next sample would be introduced and the process repeated. If washing steps are required between runs, the washing fluids would be withdrawn into the waste line. If the pressure drop across the nozzle would cause cavitation on collection then an optional pressurization of the input well can be utilized.
  • FIG. 7 shows sample introduction when the sample is less dense than the fluid in the sample port. FIG. 7 depicts an alternative scheme that could be used to introduce samples that are less dense than the oil used to emulsify the sample. As with the concept in FIG. 1, the process of introducing the sample into the port could be run either manually or with a robotic sampling system. In this concept, the sample port could be filled with the emulsification oil through backflow from the nozzle prior to introduction of the sample (FIG. 7 a). If this oil is not appropriate, the port can be filled from the top with a different immiscible fluid that might have more desirable properties than the emulsification oil (e.g. better wetting, less surfactant, etc). This second immiscible fluid could be introduced during startup and flow continuously into the port when the sample tip is not inserted. Keeping the sample port filled with fluid will prevent air entrainment during startup and should improve transient performance.
  • If the sample tip is connected to a pump capable of driving the sample into the device, it could be started up as the tip is inserted into the device (7 b-c). When used with this sort of sample introduction, the device could be run identically to the “normal” operation of our devices, including having the “transport to waste” line (7 e) not connected to a pump. If the sample tip loading pump is not capable of accurately forcing the flow (i.e. not connected to a suitable pump), the back end of the tip could be connected to a valve that would open to either atmospheric pressure (or possibly a pressurized gas supply) when the tip is fully inserted into the port. In order to prevent air entrainment into the sample tip and device, this connection could be made through a reservoir filled with the desired immiscible liquid. In either case, the device would run identically to the one described above and shown in FIG. 6. FIG. 7 also shows another possible configuration of the aspiration probe assembly used for the device in FIG. 6.
  • Methods for Forming Sample Droplet Emulsions Prior to Injection on a Microfluidic Substrate
  • The present invention also provides compositions and methods for creating emulsion of the sample fluid (e.g. droplets) prior to the introduction of the sample fluid into the microfluidic devices of the present invention. More specifically, the methods are directed to the creating sample droplet emulsions “off chip”, for example in a syringe. In order to create a monodisperse emulsion directly from a library well, a nozzle is formed directly into the fitting used to connect the collection syringe to a syringe tip (e.g. capillary tubing), as shown in FIG. 8. The nozzle can be made from one or two oil lines providing constant flow of oil into the nozzle, a connection to the capillary tubing, and a connection to the collection syringe. Aspiration of the sample can be accomplished by running the collection syringe in withdrawal mode at a flow rate (Q3) above the flow rate of the two oil syringes (Step 1 in FIG. 8). The difference in flow would correspond to the flow rate aspirated from the sample well. When the appropriate volume of sample has been loaded into the capillary tubing, the capillary tubing would be removed from the sample well, an air bubble, and possibly a cleaning solution would be aspirated (Step 2 in FIG. 8). When almost all of the sample has been emulsified, the collection syringe withdrawal rate would either be reduced below the oil flow rates, stopped, or set to infuse at some nominal rate (Step 3 in FIG. 8). The remaining sample, air, cleaning solution, etc, left in the capillary would be flushed back out into a cleaning well and the outside of the capillary would be cleaned at the “wash station.” When the capillary is completely clean, the process would repeat for the next library element.
  • The nozzle can be formed through using small bore tubing (glass, Teflon®, PEEK tubing or capillaries) or micro-fabrication or molding processes such as PDMS soft lithography, glass etching, hot embossing, or similar high resolution fabrication technology.
  • The present apparatus can be readily adapted for clinical applications or work where cross contamination must be eliminated, since the region from the nozzle to the syringe are isolated from the sample stream (e.g., the oil wets these surfaces and keeps the sample from directly contacting aqueous sample). The aspiration tip can be designed as a disposable item (like a robotic sampler aspiration tips) and automatically replaced between samples.
  • Multiple nozzle/syringe pairs can be operated in parallel, thus increasing throughput. This allows simultaneous sampling of multiple wells/samples during a single process step. Each sample can be collected into a separate syringe.
  • Other methods for forming sample droplet emulsions and emulsion libraries “off chip” are described in Example 1.
  • Methods for Minimizing Sample Volume Loss
  • As described herein, a significant problem when working with very small amounts of reagents comes from the losses associated with dead volumes found in the storage containers and transport lines. As an example, if 50 microliters of a material is injected into a 254 micron internal diameter capillary tube, a 75 cm long tube would consume about 38 microliters of the material (˜75%). To address this problem, the present invention provides compositions and methods which eliminates the problems associated with dead volume and reagent waste when working with extremely small volumes of reagents.
  • In one embodiment, the primary reagents (sample) is combined with a second, immiscible phase in the storage container (e.g. a syringe or other reservoir). This second phase is used to push the entire amount of the first phase into the system with no significant losses. More specifically, when two immiscible fluids are combined in a reservoir, the two fluids will tend to separate into layers as long as the densities of the materials are different. If the fluid of interest (e.g., sample fluid) is closest to the exit of the reservoir, it will be the first to leave when the reservoir is emptied (the exit can be on either the top or bottom, depending on the density difference). Once the reagent has been pumped out of the reservoir, the second phase will follow. This second phase will then push the first phase completely through the system without any sample fluid loss. As an example of this, oil and water (the reagent) would be combined in a syringe. If the oil is denser than the water, the syringe would be oriented with its exit face up, if the oil were less dense, then the syringe would be face down. The oil would be chosen such that the materials of interest in the reagent are not soluble in the oil phase. FIG. 9 is one example of this approach when a syringe is used as the reservoir and the second phase is denser than the reagent phase. If the reagent were more dense, then the syringe orientation would be reversed (i.e. the exit would be facing downward in the figure). Specifically, FIG. 9 shows a two phase system where the reagent is injected on top of the second, immiscible phase: (A) During injection, prior to transition from first phase to second phase, (B) second phase just entering the transfer line,. (C) second phase has completely filled the transfer line and pushed the entire volume of reagent through the system.
  • Alternatively, a sample solution is sandwiched between two immiscible liquids, wherein one liquid has a density greater than the sample density, and the second liquid has a density less than the sample density. Referring to FIG. 10, the sample (density 1.0) can be layered between perflourocarbon oil (density 1.8) and mineral oil (density 0.914). Ideally, when the device is being used to analyze biological reactions the immiscible solutions do not inhibit the reactions of the sample, nor are any test molecules in the sample or the sample itself soluble in either immiscible fluid. The less dense fluid (in FIG. 10, the mineral oil) can be used to ‘prime the pump’ and remove any air or dead-space that occurs during normal injection. The sample then rises to the injection point after the mineral oil. It is further contemplated that the methods disclosed herein would also work for gases. The gases and/or liquids can be miscible, but of different densities such that they are layered on top each other in a manner that prevents their mixing.
  • Solid or Semi-Solid Phase Droplets
  • The present invention also provides solid phase particles and methods for the forming solid phase particles on a microfluidic device for downstream analysis. The solid phase particles can be used for various biological or chemical analysis (e.g., DNA or protein analyses).
  • For DNA analysis, post amplification encapsulation of amplicons occurs within a gel or polymer matrix prior to breaking of the droplet emulsion. Amplification reactions within droplets using one of several amplification type methods (described in further detail herein), including, but not limited to; PCR, Rolling Circle Amplification (RCA), Nucleic Acid Sequence Based Amplification (NASBA), ligase chain reaction, etc. followed by encapsulation/solidification of the amplified reaction within the droplets by either polymerizing the droplets using chemical or physical means.
  • A physical means might be termed ‘gelling’ whereby one incorporates low temperature agarose within the droplet during formulation and keeping the droplet above the solidification temperature until one desires the droplet to solidify.
  • A chemical means might be termed ‘polymerization’ whereby one combines (if needed) the droplet with a polymerizing solution and then polymerizing the droplet using either a polymerization initiator (for example free radicals) or a means such as UV light. Some other means of gelling or polymerization include matragel, polyacrylamide, mixed polysaccharides, etc. Some example initiators can be temperature, UV irradiation, etc.
  • In a further example, one of the DNA primers used for amplification can be attached to one of the molecules that will form the polymerized matrix. Attachment can be through a direct chemical attachment, or through a secondary attachment such as biotin-streptavidin attachment. In this example, the DNA will become physically attached to the formed solid-phase that occurs after solidification of the droplet. Using one of several gelling or polymerizing methods it should be possible to further manipulate these droplets.
  • One could also either exchange or wash away unincorporated nucleotides, primers and exchange buffers so as to remove the initial amplification buffers, polymerases, etc. In an example of further droplet manipulation wherein one of the strands is polymerized from an attached DNA amplification primer, one could treat the polymerized or gelled droplet with a base solution to disassociate the two DNA strands and elute, from the gelled or polymerized droplet, the unattached strand.
  • For protein analysis, proteins can be either trapped within, or attached to the gel or polymer matrix. If attached, it can be through a covalent linkage or through an affinity tag, for example his6 or avi-tag. The proteins can be added to droplets containing gel or polymer reagent, or they can be formulated along with the gel or polymerization reagent. Variations that include both are also possible. The protein can be added to the droplets. Additionally, it is possible to add DNA to the droplet and allow in vitro transcription/translation to synthesize the protein.
  • The droplets can be kept in liquid form on the microfluidic device and either gelled or polymerized upon removal, or can be gelled or polymerized within the droplets anywhere on the device after the droplets have been formed.
  • In an example, multiple plasmids are formulated into a droplet along with an in vitro transcription/translation reaction. Genes, encoded by the plasmids, are translated and transcribed to protein molecules. The protein molecules attach to the polymer via an avi-tag, the droplets are allowed to gel and the plasmid molecules become ‘fixed’ or ‘trapped’ within the gel. The gelled droplets are collected, the emulsion is broken and the solidified droplets collected and washed. As an example application, DNA is amplified within a droplet wherein one primer is physically attached to a polymer monomer. The droplet is then combined with a droplet containing the enzymes DNA polymerase, luciferase and sulfurylase. The merged droplets are allowed to gel or polymerize, they are collected, and if needed, washed. These washed gelled droplets can then used for a DNA sequencing reaction.
  • Surfactants
  • The fluids used in the invention may contain one or more additives, such as agents which reduce surface tensions (surfactants). Surfactants can include Tween, Span, fluorosurfactants, and other agents that are soluble in oil relative to water. In some applications, performance is improved by adding a second surfactant to the aqueous phase. Surfactants can aid in controlling or optimizing droplet size, flow and uniformity, for example by reducing the shear force needed to extrude or inject droplets into an intersecting channel. This can affect droplet volume and periodicity, or the rate or frequency at which droplets break off into an intersecting channel. Furthermore, the surfactant can serve to stabilize aqueous emulsions in fluorinated oils from coalescing.
  • The droplets may be coated with a surfactant. Preferred surfactants that may be added to the continuous phase fluid include, but are not limited to, surfactants such as sorbitan-based carboxylic acid esters (e.g., the “Span” surfactants, Fluka Chemika), including sorbitan monolaurate (Span 20), sorbitan monopalmitate (Span 40), sorbitan monostearate (Span 60) and sorbitan monooleate (Span 80), and perfluorinated polyethers (e.g., DuPont Krytox 157 FSL, FSM, and/or FSH). Other non-limiting examples of non-ionic surfactants which may be used include polyoxyethylenated alkylphenols (for example, nonyl-, p-dodecyl-, and dinonylphenols), polyoxyethylenated straight chain alcohols, polyoxyethylenated polyoxypropylene glycols, polyoxyethylenated mercaptans, long chain carboxylic acid esters (for example, glyceryl and polyglycerl esters of natural fatty acids, propylene glycol, sorbitol, polyoxyethylenated sorbitol esters, polyoxyethylene glycol esters, etc.) and alkanolamines (e.g., diethanolamine-fatty acid condensates and isopropanolamine-fatty acid condensates). In addition, ionic surfactants such as sodium dodecyl sulfate (SDS) may also be used. However, such surfactants are generally less preferably for many embodiments of the invention. For instance, in those embodiments where aqueous droplets are used as nanoreactors for chemical reactions (including biochemical reactions) or are used to analyze and/or sort biomaterials, a water soluble surfactant such as SDS may denature or inactivate the contents of the droplet.
  • The carrier fluid can be an oil (e.g., decane, tetradecane or hexadecane) or fluorocarbon oil that contains a surfactant (e.g., a non-ionic surfactant such as a Span surfactant) as an additive (preferably between about 0.2 and 5% by volume, more preferably about 2%). A user can preferably cause the carrier fluid to flow through channels of the microfluidic device so that the surfactant in the carrier fluid coats the channel walls.
  • In one embodiment, the fluorosurfactant can be prepared by reacting the perflourinated polyether DuPont Krytox 157 FSL, FSM, or FSH with aqueous ammonium hydroxide in a volatile fluorinated solvent. The solvent and residual water and ammonia can be removed with a rotary evaporator. The surfactant can then be dissolved (e.g., 2.5 wt %) in a fluorinated oil (e.g., Flourinert (3M)), which then serves as the continuous phase of the emulsion.
  • Driving Forces
  • The invention can use pressure drive flow control, e.g., utilizing valves and pumps, to manipulate the flow of cells, particles, molecules, enzymes or reagents in one or more directions and/or into one or more channels of a microfluidic device. However, other methods may also be used, alone or in combination with pumps and valves, such as electro-osmotic flow control, electrophoresis and dielectrophoresis (Fulwyer, Science 156, 910 (1974); Li and Harrison, Analytical Chemistry 69, 1564 (1997); Fiedler, et al. Analytical Chemistry 70, 1909-1915 (1998); U.S. Pat. No. 5,656,155). Application of these techniques according to the invention provides more rapid and accurate devices and methods for analysis or sorting, for example, because the sorting occurs at or in a sorting module that can be placed at or immediately after a detection module. This provides a shorter distance for molecules or cells to travel, they can move more rapidly and with less turbulence, and can more readily be moved, examined, and sorted in single file, i.e., one at a time.
  • Positive displacement pressure driven flow is a preferred way of controlling fluid flow and dielectrophoresis is a preferred way of manipulating droplets within that flow.
  • The pressure at the inlet module can also be regulated by adjusting the pressure on the main and sample inlet channels, for example, with pressurized syringes feeding into those inlet channels. By controlling the pressure difference between the oil and water sources at the inlet module, the size and periodicity of the droplets generated may be regulated. Alternatively, a valve may be placed at or coincident to either the inlet module or the sample inlet channel connected thereto to control the flow of solution into the inlet module, thereby controlling the size and periodicity of the droplets. Periodicity and droplet volume may also depend on channel diameter, the viscosity of the fluids, and shear pressure.
  • Without being bound by any theory, electro-osmosis is believed to produce motion in a stream containing ions e.g. a liquid such as a buffer, by application of a voltage differential or charge gradient between two or more electrodes. Neutral (uncharged) molecules or cells can be carried by the stream. Electro-osmosis is particularly suitable for rapidly changing the course, direction or speed of flow. Electrophoresis is believed to produce movement of charged objects in a fluid toward one or more electrodes of opposite charge, and away from one on or more electrodes of like charge. Where an aqueous phase is combined with an oil phase, aqueous droplets are encapsulated or separated from each other by oil. Typically, the oil phase is not an electrical conductor and may insulate the droplets from the electro-osmotic field. In this example, electro-osmosis may be used to drive the flow of droplets if the oil is modified to carry or react to an electrical field, or if the oil is substituted for another phase that is immiscible in water but which does not insulate the water phase from electrical fields.
  • Dielectrophoresis is believed to produce movement of dielectric objects, which have no net charge, but have regions that are positively or negatively charged in relation to each other. Alternating, non-homogeneous electric fields in the presence of droplets and/or particles, such as cells or molecules, cause the droplets and/or particles to become electrically polarized and thus to experience dielectrophoretic forces. Depending on the dielectric polarizability of the particles and the suspending medium, dielectric particles will move either toward the regions of high field strength or low field strength. For example, the polarizability of living cells depends on their composition, morphology, and phenotype and is highly dependent on the frequency of the applied electrical field. Thus, cells of different types and in different physiological states generally possess distinctly different dielectric properties, which may provide a basis for cell separation, e.g., by differential dielectrophoretic forces. Likewise, the polarizability of droplets also depends upon their size, shape and composition. For example, droplets that contain salts can be polarized. According to formulas provided in Fiedler, et al. Analytical Chemistry 70, 1909-1915 (1998), individual manipulation of single droplets requires field differences (inhomogeneities) with dimensions close to the droplets.
  • The term “dielectrophoretic force gradient” means a dielectrophoretic force is exerted on an object in an electric field provided that the object has a different dielectric constant than the surrounding media. This force can either pull the object into the region of larger field or push it out of the region of larger field. The force is attractive or repulsive depending respectively on whether the object or the surrounding media has the larger dielectric constant.
  • Manipulation is also dependent on permittivity (a dielectric property) of the droplets and/or particles with the suspending medium. Thus, polymer particles, living cells show negative dielectrophoresis at high-field frequencies in water. For example, dielectrophoretic forces experienced by a latex sphere in a 0.5 MV/m field (10 V for a 20 micron electrode gap) in water are predicted to be about 0.2 piconewtons (pN) for a 3.4 micron latex sphere to 15 pN for a 15 micron latex sphere (Fiedler, et al. Analytical Chemistry 70, 1909-1915 (1998)). These values are mostly greater than the hydrodynamic forces experienced by the sphere in a stream (about 0.3 pN for a 3.4 micron sphere and 1.5 pN for a 15 micron sphere). Therefore, manipulation of individual cells or particles can be accomplished in a streaming fluid, such as in a cell sorter device, using dielectrophoresis. Using conventional semiconductor technologies, electrodes can be microfabricated onto a substrate to control the force fields in a microfabricated sorting device of the invention. Dielectrophoresis is particularly suitable for moving objects that are electrical conductors. The use of AC current is preferred, to prevent permanent alignment of ions. Megahertz frequencies are suitable to provide a net alignment, attractive force, and motion over relatively long distances. See U.S. Pat. No. 5,454,472.
  • Radiation pressure can also be used in the invention to deflect and move objects, e.g. droplets and particles (molecules, cells, particles, etc.) contained therein, with focused beams of light such as lasers. Flow can also be obtained and controlled by providing a pressure differential or gradient between one or more channels of a device or in a method of the invention.
  • Molecules, cells or particles (or droplets containing molecules, cells or particles) can be moved by direct mechanical switching, e.g., with on-off valves or by squeezing the channels. Pressure control may also be used, for example, by raising or lowering an output well to change the pressure inside the channels on the chip. See, e.g., the devices and methods described U.S. Pat. No. 6,540,895. These methods and devices can further be used in combination with the methods and devices described in pending U.S. Patent Application Publication No.20010029983 and 20050226742. Different switching and flow control mechanisms can be combined on one chip or in one device and can work independently or together as desired.
  • Inlet Module
  • The microfluidic device of the present invention includes one or more inlet modules. An “inlet module” is an area of a microfluidic substrate device that receives molecules, cells, small molecules or particles for additional coalescence, detection and/or sorting. The inlet module can contain one or more inlet channels, wells or reservoirs, openings, and other features which facilitate the entry of molecules, cells, small molecules or particles into the substrate. A substrate may contain more than one inlet module if desired. Different sample inlet channels can communicate with the main channel at different inlet modules. Alternately, different sample inlet channels can communication with the main channel at the same inlet module. The inlet module is in fluid communication with the main channel. The inlet module generally comprises a junction between the sample inlet channel and the main channel such that a solution of a sample (i.e., a fluid containing a sample such as molecules, cells, small molecules (organic or inorganic) or particles) is introduced to the main channel and forms a plurality of droplets. The sample solution can be pressurized. The sample inlet channel can intersect the main channel such that the sample solution is introduced into the main channel at an angle perpendicular to a stream of fluid passing through the main channel. For example, the sample inlet channel and main channel intercept at a T-shaped junction; i.e., such that the sample inlet channel is perpendicular (90 degrees) to the main channel. However, the sample inlet channel can intercept the main channel at any angle, and need not introduce the sample fluid to the main channel at an angle that is perpendicular to that flow. The angle between intersecting channels is in the range of from about 60 to about 120 degrees. Particular exemplary angles are 45, 60, 90, and 120 degrees.
  • Embodiments of the invention are also provided in which there are two or more inlet modules introducing droplets of samples into the main channel. For example, a first inlet module may introduce droplets of a first sample into a flow of fluid in the main channel and a second inlet module may introduce droplets of a second sample into the flow of fluid in main channel, and so forth. The second inlet module is preferably downstream from the first inlet module (e.g., about 30 μm). The fluids introduced into the two or more different inlet modules can comprise the same fluid or the same type of fluid (e.g., different aqueous solutions). For example, droplets of an aqueous solution containing an enzyme are introduced into the main channel at the first inlet module and droplets of aqueous solution containing a substrate for the enzyme are introduced into the main channel at the second inlet module. Alternatively, the droplets introduced at the different inlet modules may be droplets of different fluids which may be compatible or incompatible. For example, the different droplets may be different aqueous solutions, or droplets introduced at a first inlet module may be droplets of one fluid (e.g., an aqueous solution) whereas droplets introduced at a second inlet module may be another fluid (e.g., alcohol or oil).
  • Droplet Interdigitation
  • Particular design embodiments of the microfluidic device described herein allow for a more reproducible and controllable interdigitation of droplets of specific liquids followed by pair-wise coalescence of these droplets, described in further detail herein. The droplet pairs can contain liquids of different compositions and/or volumes, which would then combine to allow for a specific reaction to be investigated. The pair of droplets can come from any of the following: (i) two continuous aqueous streams and an oil stream; (ii) a continuous aqueous stream, an emulsion stream, and an oil stream, or (iii) two emulsion streams and an oil stream. The term “interdigitation” as used herein means pairing of droplets from separate aqueous streams, or from two separate inlet nozzles, for eventual coalescence.
  • The nozzle designs described herein enhance the interdigitation of droplets and further improves coalescence of droplets due to the better control of the interdigitation and smaller distance between pairs of droplets. The greater control over interdigitation allows for a perfect control over the frequency of either of the droplets. To obtain the optimum operation, the spacing between droplets and coupling of the droplets can be adjusted by adjusting flow of any of the streams, viscosity of the streams, nozzle design (including orifice diameter, the channel angle, and post-orifice neck of the nozzle).
  • Reservoir/Well
  • A device of the invention can include a sample solution reservoir or well or other apparatus for introducing a sample to the device, at the inlet module, which is typically in fluid communication with an inlet channel. Reservoirs and wells used for loading one or more samples onto the microfluidic device of the present invention, include but are not limited to, syringes, cartridges, vials, eppendorf tubes and cell culture materials (e.g., 96 well plates). A reservoir may facilitate introduction of molecules or cells into the device and into the sample inlet channel of each analysis unit.
  • Fluidic Interconnects
  • The microfluidic device can include a syringe (or other glass container) that is treated with a vapor or solution of an appropriate PEG-silane to effect the surface PEG functionalization. The purpose for treating the walls of glass containers (e.g., syringes) with a PEG functionality is to prevent biological adhesion to the inner walls of the container, which frustrates the proper transfer of biological/chemical materials into the microfluidic device of the present invention. The inlet channel is further connected to a means for introducing a sample to said device. The means can be a well or reservoir. The means can be temperature controlled. The inlet module may also contain a connector adapted to receive a suitable piece of tubing, such as liquid chromatography or HPLC tubing, through which a sample may be supplied. Such an arrangement facilitates introducing the sample solution under positive pressure in order to achieve a desired infusion rate at the inlet module.
  • The interconnections, including tubes, must be extremely clean and make excellent bonding with the PDMS surface in order to allow proper operation of the device. The difficulty in making a fluidic connection to a microfluidic device is primarily due to the difficulty in transitioning from a macroscopic fluid line into the device while minimizing dead volume.
  • In order to minimize contamination and leakage and allow for greater reproducibility and reliability are improved, tubes and interconnects for the PDMS slab can be cured in place. The tubes and interconnects can be placed in position by applying a UV-cured adhesive to allow for holding the tubes in place on the silicone wafer. Once the tubes are placed in position, PDMS can be poured over the wafer and cured. The cured PDMS, along with the tubes in place, can be peeled off of the silicone wafer easily. This process can be applied to fluidics channels as well as other connection channels. Once the adhesive is applied onto the wafer, the process will allow for quick templating of PDMS slabs with exact reproducibility of channel locations and cleanliness. Tubes of any size can be implemented for this process. This process allows for less stress on the interconnection joints and smaller interconnection footprints in the device (see, for example, PCT/US2006/02186 filed on Jun. 1, 2006; PCT/US2006/021280 filed on Jun. 1, 2006 and PCT/US2006/021380 filed on Jun. 1, 2006, each of which is incorporated by reference in their entirety for all purposes).
  • The tubing side of the interconnect can be mounted into a retaining block that provides precise registration of the tubing, while the microfluidic device can be positioned accurately in a carrier that the retaining block would align and clamp to. The total dead volume associated with these designs would be critically dependent on how accurately the two mating surfaces could be positioned relative to each other. The maximum force required to maintain the seal would be limited by the exact shape and composition of the sealing materials as well as the rigidity and strength of the device itself. The shapes of the mating surfaces can be tailored to the minimal leakage potential, sealing force required, and potential for misalignment. By way of non-limiting example, the single ring indicated in can be replaced with a series of rings of appropriate cross-sectional shape.
  • Reservoirs and wells used for loading one or more samples onto the microfluidic device of the present invention, include but are not limited to, syringes, cartridges, vials, eppendorf tubes and cell culture materials (e.g., 96 well plates) as described above. One of the issues to be resolved in loading samples into the inlet channel at the inlet module of the substrate is the size difference between the loading means or injection means, e.g., capillary or HPLC tubing and the inlet channel. It is necessary to create an interconnect and loading method which limits leaks and minimizes dead volume and compliance problems. Several devices and methods described in further detail herein address and solve these art problems.
  • Self-Aligning Fluidic Interconnects
  • The present invention includes one or more inlet modules comprising self-aligning fluidic interconnects proximate to one or more inlet channels to improve the efficiency of sample loading and/or injection.
  • The present invention proposes the use of small interconnects based on creating a radial seal instead of a face seal between the microfluidic device and interconnect. The inserted interconnect would have a larger diameter than the mating feature on the device. When inserted, the stretching of the chip would provide the sealing force needed to make a leak-free seal between the external fluid lines and the microfluidic device. FIG. 11 details design possibilities for making this seal.
  • Studies were performed with the leftmost design of FIG. 1 using a cast hole in PDMS and 1/32″ PEEK tubing, which showed that the seal was able to withstand more than 90 PSI of pressure without leakage.
  • In order to handle instrument and chip manufacturing tolerances, the external interconnect must be self aligning and the “capture radius” of the molded hole must be large enough to reliably steer the interconnect to the sealing surfaces. FIG. 12 shows that the entrance to the molded hole is large enough to guarantee capture but tapers down to the sealing surfaces. The external interconnect could be made directly out of the tubing leading up to the microfluidic substrate, thus eliminating potential leak points and unswept volumes. As seen in FIG. 12, the interconnect is surrounded by the substrate interconnects or “chip dock” for most of its length to make certain it is held within the tolerance stack-up of the system. The external interconnect is made from a hard but flexible material such as 1/32″ PEEK tubing. The features in the microfluidic device can be molded directly into it during the manufacturing process, while the inserted seals can be molded/machined directly onto the tubing ends or molded as individual pieces and mechanically fastened to the tubing. The retaining ferrule shown in FIG. 12 would be attached during manufacturing and provide good absolute referencing of the tube length. The ferrule could be an off-the-shelf component or a custom manufactured part and be made from, for example, a polymer, an elastomer, or a metal. The tubing end could be tapered on the end (top most diagram) or squared off (the figure above). The specific shape of the end will be controlled by how easily the microfluidic device will gall during insertion.
  • Alternatively, it is also possible to mold all the interconnects needed for each tube into a single monolithic self-aligned part as detailed in FIG. 13. This may help reduce the difficulty in maintaining alignment of many external fluidic lines to the chip.
  • Methods for Molding Fluidic Interconnects Directly on the Substrate
  • The present invention also provides methods of direct molding of fluidic interconnects into a microfluidic device. Development of a commercial microfluidic platform requires a simple, reliable fluidic interconnect in order to reduce the chance of operator error and leaks. Molding these interconnects directly into the microfluidic device requires precise alignment of the molding pins to the patterned shim (the “master” manufactured from Silicon/photoresist or made from some metal) used to form the microfluidic and electrical channels. The extreme tolerances required when molding with a low viscosity elastomer such as PDMS requires near perfect sealing of the pin face to the master, while still accommodating imperfections in the master and assembly of the molding tool. In an embodiment, the present invention provides a precise and repeatable method of molding of interconnects while accommodating the imperfections in the molding process by introducing movable pins captured in an elastomeric sleeve molded directly into the tool. In order to effectively produce at relatively low volume and be able to inexpensively prototype devices, the tool must be able to use masters generated using standard photolithographic processes (e.g. silicon wafers patterned with SU-8).
  • FIG. 14 shows a schematic of a molding tool based on this concept. In FIG. 14, the pins (orange) are captured within an elestomeric molded sleeve. A compression plate made from a rigid backer plate and foam rubber is used to apply gentle even pressure to the pins and generate the force needed to make the pins uniformly contact the master. The molded sleeve was found to be necessary to consistently prevent the uncured elastomer from penetrating the region between the pin and the top plate. Early designs used pins captured in tight clearance holes, and the pins would frequently bind in place (even with lubricant), preventing smooth motion of the pins and improper contact with the master. This would in turn cause a thin film of the elastomer to form between the bottom of the pin and the master (“Flash”). This flash prevents proper operation of the interconnects during chip operation. The addition of the elastomeric sleeves around each pin eliminated this problem, and produce consistent, reliable shutoff between the master and the pins.
  • Acoustic Actuator
  • The well or reservoir of the inlet module further include an acoustic actuator. To obtain one droplet comprising a single element of a specific biological/chemical material (e.g., a cell), separation of biological/chemical material, and uniformity of the number density of biological/chemical materials in a microfluidic channel is desirable. Accordingly, the microfluidic device can include an acoustic actuator. The loaded sample (biological/chemical material) can be well mixed and separated in a small chamber by acoustic wave before sending out to the nozzle region for encapsulation. The frequency of the acoustic wave should be fine tuned so as not to cause any damage to the cells. The biological effects of acoustic mixing have been well studied (e.g., in the ink-jet industry) and many published literatures also showed that piezoelectric microfluidic device can deliver intact biological payloads such as live microorganisms and DNA.
  • The design of the acoustic resonant can use a Piezoelectric bimorph flat plate located on the side of the carved resonant in the PDMS slab. The resonant inlet can connect to the cell flow input channel and the outlet can connect to the cell flow pinching channel. The piezoelectric driving waveform can be carefully optimized to select the critical frequencies that can separate cells in fluids. There are five parameters to optimize beyond the frequency parameter and Lab electronics can be used to optimize the piezoelectric driving waveform. Afterwards, a low cost circuit can be designed to generate only the optimized waveform in a preferred microfluidic device.
  • Coalescence Module
  • The microfluidic device of the present invention also includes one or more coalescence modules. A “coalescence module” is within or coincident with at least a portion of the main channel at or downstream of the inlet module where molecules, cells, small molecules or particles comprised within droplets are brought within proximity of other droplets comprising molecules, cells, small molecules or particles and where the droplets in proximity fuse, coalesce or combine their contents. The coalescence module can also include an apparatus, for generating an electric force.
  • The electric force exerted on the fluidic droplet may be large enough to cause the droplet to move within the liquid. In some cases, the electric force exerted on the fluidic droplet may be used to direct a desired motion of the droplet within the liquid, for example, to or within a channel or a microfluidic channel (e.g., as further described herein), etc.
  • The electric field can be generated from an electric field generator, i.e., a device or system able to create an electric field that can be applied to the fluid. The electric field generator may produce an AC field (i.e., one that varies periodically with respect to time, for example, sinusoidally, sawtooth, square, etc.), a DC field (i.e., one that is constant with respect to time), a pulsed field, etc. The electric field generator may be constructed and arranged to create an electric field within a fluid contained within a channel or a microfluidic channel. The electric field generator may be integral to or separate from the fluidic system containing the channel or microfluidic channel, according to some embodiments. As used herein, “integral” means that portions of the components integral to each other are joined in such a way that the components cannot be in manually separated from each other without cutting or breaking at least one of the components.
  • Techniques for producing a suitable electric field (which may be AC, DC, etc.) are known to those of ordinary skill in the art. For example, in one embodiment, an electric field is produced by applying voltage across a pair of electrodes, which may be positioned on or embedded within the fluidic system (for example, within a substrate defining the channel or microfluidic channel), and/or positioned proximate the fluid such that at least a portion of the electric field interacts with the fluid. The electrodes can be fashioned from any suitable electrode material or materials known to those of ordinary skill in the art, including, but not limited to, silver, gold, copper, carbon, platinum, copper, tungsten, tin, cadmium, nickel, indium tin oxide (“ITO”), etc., as well as combinations thereof.
  • Electrodes
  • The device can include channels for use in fluid control and other channels filled with a metal alloy for casting integrated metal alloy components (i.e., electrodes). Alternatively, the electrodes can be manufactured using other technologies (e.g., lithographically patterned electrodes made from indium tin oxide or a metal such as platinum). The microfluidic device can include metal alloy components useful for performing electrical functions on fluids, including but not limited to, coalescing droplets, charging droplets, sorting droplets, detecting droplets and shaking droplets to mix the contents of coalesced droplets. The device can contain more than one of the above mentioned components for more than one of the above mentioned functions.
  • The electrodes comprising metal alloy components may either terminate at fluid channels or be isolated from fluid channels. The electrodes can be constructed by filling the appropriate channels with metal alloy. One way this can be accomplished is to use positive pressure injection of the metal alloy in a melted state, such as with a syringe, into the channels, and then cool the metal alloy to a solid form. Another example is to use negative pressure to draw the metal alloy in a melted state into the channels, and then cool the metal alloy to a solid form. This can be accomplished for example by use of capillary forces. Another method of construction can use any of the above mentioned embodiments, and then flush out the metal alloy in a melted state with another liquid to define the geometry of the metal alloy components. Another example is to use any of the above mentioned embodiments, and then use a localized cold probe to define a solid termination point for the metal alloy, and then cool the remaining metal alloy to a solid form. A further example is to use another material, such as microscopic solder spheres or UV curable conductive ink, to form a barrier between fluid and metal alloy channels, to define the geometry of the metal alloy components.
  • The device can include a combination of both integrated metal alloy components and a patterned electrically conductive layer. The patterned electrically conductive layer can have features patterned such that their boundaries are within a leak-proof seal. The device can have a patterned electrically conductive feature as one of two charging electrodes and one integrated metal alloy component as the other of two charging electrodes.
  • The device can include a plurality of electrodes that are insulated from the fluid present in the device, and the method of operation including appropriate application of dielectrical signals and appropriate fluids. In known devices, the electrodes are typically in contact with the fluids in order to allow discharge of species that would otherwise screen the applied dielectric field. Whereas, in devices where the electrodes have been insulated from the fluid, this screening effect typically arises so quickly that the device is not useful for any significantly extended period of time. The drawbacks of electrodes in contact with the fluids vs. insulated electrodes are (a) degraded reliability against leaking (since the interface between the electrodes and the other components of the device may be more difficult to effect a leak-proof seal), and (b) degraded reliability against electrode corrosion (whose failure mode effects include failure of application of dielectric fields, and fluid channel contamination).
  • The device of the present invention comprising a plurality of electrodes that are insulated from the fluid present in the device counteracts this screening effect by extending the screening rise time and including a polarity switch for all of the different dielectric fields applied in the device. The screening rise time is extended by using fluids with dielectrical properties. A polarity switch for all of the different dielectric fields applied in the device is achieved by using an algorithm for dielectrical control, which switches the polarity of the dielectrical fields at a frequency sufficiently high to maintain proper dielectrical function of the device. This dielectrical control algorithm may also switch the polarity for the dielectric fields in a cascading, time controlled manner starting at the fluid origin point and progressing downstream, so that given fluid components experience one polarity at every point along their course. The device of the present invention can be used with metal alloy electrodes or using a combination of metal alloy electrodes and patterned conductive film electrodes.
  • The invention can provide a microfluidic device using injected electrodes. The interface between the microscopic electrode (typically 25 μm thick) and the macroscopic interconnect can easily fail if the joint between the two is flexed. The flexing of the joint can be eliminated by securing a firm material that serves to fasten, support, and re-enforce the joint (i.e., a grommet) into the interface. In order to prevent flexing, the mating surface of the device can be manufactured from a hard material such as glass or plastic. The electrical connection with the external system can be made by securing the device such that it connects to a spring loaded contact, which is either offset from the grommet (thereby minimizing the force applied to the solder region), or centered on the grommet (as long as the contact does not touch the solder).
  • The metal alloy components are also useful for performing optical functions on fluids, including but not limited to, optical detection of droplets in a geometry which may include a mirror.
  • To prevent leakage of fluid out of electrodes placed within microfluidic channels, the microfluidic device can include a layer patterned with channels for fluid control, and another layer with patterned electrically conductive features, where the features are patterned such that their boundaries are within a leak-proof seal. The leak-proof seal can be achieved at the interface between the unpatterned areas of the fluid control layer and the unpatterned areas of the electrically conductive layer. The leak-proof seal can also be achieved by a third interfacial layer between the fluid control layer and the unpatterned areas of the electrically conductive layer. The third interfacial layer can or can not be perforated at specific locations to allow contact between the fluid and the electrically conductive layer. Electrical access ports can also be patterned in the fluid control layer.
  • The electrodes and patterned electrically conductive layers as described can be associated with any module of the device (inlet module, coalescence module, mixing module, delay module, detection module and sorting module) to generate dielectric or electric forces to manipulate and control the droplets and their contents.
  • Effective control of uncharged droplets within microfluidic devices can require the generation of extremely strong dielectric field gradients. The fringe fields from the edges of a parallel plate capacitor can provide an excellent topology to form these gradients. The microfluidic device according to the present invention can include placing a fluidic channel between two parallel electrodes, which can result in a steep electric field gradient at the entrance to the electrodes due to edge effects at the ends of the electrode pair. Placing these pairs of electrodes at a symmetric channel split can allow precise bi-directional control of droplet within a device. Using the same principle, only with asymmetric splits, can allow single ended control of the droplet direction in the same manner. Alternatively, a variation on this geometry will allow precise control of the droplet phase by shifting.
  • In some cases, transparent or substantially transparent electrodes can be used. The electric field generator can be constructed and arranged (e.g., positioned) to create an electric field applicable to the fluid of at least about 0.01 V/micrometer, and, in some cases, at least about 0.03 V/micrometer, at least about 0.05 V/micrometer, at least about 0.08 V/micrometer, at least about 0.1 V/micrometer, at least about 0.3 V/micrometer, at least about 0.5 V/micrometer, at least about 0.7 V/micrometer, at least about 1 V/micrometer, at least about 1.2 V/micrometer, at least about 1.4 V/micrometer, at least about 1.6 V/micrometer, or at least about 2 V/micrometer. In some embodiments, even higher electric field intensities may be used, for example, at least about 2 V/micrometer, at least about 3 V/micrometer, at least about 5 V/micrometer, at least about 7 V/micrometer, or at least about 10 V/micrometer or more.
  • As described, an electric field may be applied to fluidic droplets to cause the droplets to experience an electric force. The electric force exerted on the fluidic droplets may be, in some cases, at least about 10−16 N/micrometer3. In certain cases, the electric force exerted on the fluidic droplets may be greater, e.g., at least about 10−15 N/micrometer3, at least about 10−14 N/micrometer3, at least about 10−13 N/micrometer3, at least about 10−12 N/micrometer3, at least about 10−11 N/micrometer3, at least about 10−10 N/micrometer3, at least about 10−9 N/micrometer3, at least about 10−8 N/micrometer3, or at least about 10−7 N/micrometer3 or more. The electric force exerted on the fluidic droplets, relative to the surface area of the fluid, may be at least about 10−15 N/micrometer2, and in some cases, at least about 10−14 N/micrometer2, at least about 10−13 N/micrometer2, at least about 10−12 N/micrometer2, at least about 10−11 N/micrometer2, at least about 10−10 N/micrometer2, at least about 10−9 N/micrometer2, at least about 10−8 N/micrometer2, at least about 10−7 N/micrometer2, or at least about 10−6 N/micrometer2 or more. In yet other embodiments, the electric force exerted on the fluidic droplets may be at least about 10−9 N, at least about 10−8 N, at least about 10−7 N, at least about 10−6 N, at least about 10−5 N, or at least about 10−4 N or more in some cases.
  • Channel Expansion Geometries
  • In preferred embodiments described herein, droplet coalescence is presently carried out by having two droplet forming nozzles emitting droplets into the same main channel. The size of the nozzles allow for one nozzle to form a large drop that fills the exhaust line while the other nozzle forms a drop that is smaller than the first. The smaller droplet is formed at a rate that is less than the larger droplet rate, which insures that at most one small droplet is between big droplets. Normally, the small droplet will catch up to the larger one over a relatively short distance, but sometimes the recirculation zone behind the large drop causes the small drop to separate from the large drop cyclically. In addition, the small drop occasionally does not catch up with the large one over the distance between the nozzles and the coalescing electrodes. Thus, in some situations is a need for a more robust coalescence scheme.
  • Geometric alterations in the coalescence module can create a more robust, reliable coalescence or fusing of droplets over a wider range of sizes and flows. The solution to improve the performance is to place an expansion in the main channel between the electrodes. FIG. 15 is a schematic diagram of the improved coalescence module. Optionally, a small constriction (neckdown) just before this expansion can be used to better align the droplets on their way into the coalescence point (also shown in the FIG. 15). This optional neckdown can help center the small droplet in the channel stream lines, reducing the chance that it will flow around the larger droplet prior to coalescing in the expansion. The electrode pair may be placed on either one side of the channel or on both sides.
  • The expansion in the coalescing region allows for a dramatic catching up of the small drop to the large drop, as shown through micrographs taken on an operating device. The volume of the expansion is big enough to slow the large droplet down so that the small drop always catches up to the large drop, but doesn't allow the next large drop to catch up and make contact with the pair to be coalesced. The electrodes allow for coalescence to take place when the drops are in contact with each other and passing through the field gradient.
  • Detection Module
  • The microfluidic device of the present invention can also include one or more detection modules. A “detection module” is a location within the device, typically within the main channel where molecules, cells, small molecules or particles are to be detected, identified, measured or interrogated on the basis of at least one predetermined characteristic. The molecules, cells, small molecules or particles can be examined one at a time, and the characteristic is detected or measured optically, for example, by testing for the presence or amount of a reporter. For example, the detection module is in communication with one or more detection apparatuses. The detection apparatuses can be optical or electrical detectors or combinations thereof. Examples of suitable detection apparatuses include optical waveguides, microscopes, diodes, light stimulating devices, (e.g., lasers), photo multiplier tubes, and processors (e.g., computers and software), and combinations thereof, which cooperate to detect a signal representative of a characteristic, marker, or reporter, and to determine and direct the measurement or the sorting action at the sorting module. However, other detection techniques can also be employed
  • The term “determining,” as used herein, generally refers to the analysis or measurement of a species, for example, quantitatively or qualitatively, and/or the detection of the presence or absence of the species. “Determining” may also refer to the analysis or measurement of an interaction between two or more species, for example, quantitatively or qualitatively, or by detecting the presence or absence of the interaction. Examples of suitable techniques include, but are not limited to, spectroscopy such as infrared, absorption, fluorescence, UV/visible, FTIR (“Fourier Transform Infrared Spectroscopy”), or Raman; gravimetric techniques; ellipsometry; piezoelectric measurements; immunoassays; electrochemical measurements; optical measurements such as optical density measurements; circular dichroism; light scattering measurements such as quasielectric light scattering; polarimetry; refractometry; or turbidity measurements as described further herein.
  • A detection module is within, communicating or coincident with a portion of the main channel at or downstream of the inlet module and, in sorting embodiments, at, proximate to, or upstream of, the sorting module or branch point. The sorting module may be located immediately downstream of the detection module or it may be separated by a suitable distance consistent with the size of the molecules, the channel dimensions and the detection system. Precise boundaries for the detection module are not required, but are preferred.
  • Detection modules used for detecting molecules and cells have a cross-sectional area large enough to allow a desired molecule, cells, bead, or particles to pass through without being substantially slowed down relative to the flow carrying it. The dimensions of the detection module are influenced by the nature of the sample under study and, in particular, by the size of the droplets, beads, particles, molecules or cells (including virions) under study. For example, mammalian cells can have a diameter of about 1 to 50 microns, more typically 10 to 30 microns, although some mammalian cells (e.g., fat cells) can be larger than 120 microns. Plant cells are generally 10 to 100 microns. However, other molecules or particles can be smaller with a diameter from about 20 nm to about 500 nm.
  • Waveguides
  • The present invention provides self-aligning optical waveguides and optical elements (lenses, prisms, mirrors, interconnects, etc.) for detection and control of droplets. Such waveguides can be used to provide well defmed optical access to the fluidic channels to permit optical scattering, absorption, fluorescence, or any other optical measurement technique.
  • In order to create the waveguides, a separate series of channels and useful shapes (lenses, mirrors, etc) can be created either simultaneously within the other channels in the substrate (i.e. in the same processing step) or in successive steps. The reusable master created in this way can then used to form the waveguide components and fluid channels without the need for special fixturing or careful alignment in subsequent steps. The extra channels or shapes can then filled with a high index of refraction liquid (for waveguides) or reflective material (for mirrors) through injection into the channel or void. The liquid can either remain as a fluid or be allowed to solidify. UV cure epoxies used by the telecommunications industry are excellent choices for the waveguide materials. Possible waveguide geometry can include a focusing lens and a back-reflecting mirror.
  • Sensors
  • One or more detections sensors and/or processors may be positioned to be in sensing communication with the fluidic droplet. “Sensing communication,” as used herein, means that the sensor may be positioned anywhere such that the fluidic droplet within the fluidic system (e.g., within a channel), and/or a portion of the fluidic system containing the fluidic droplet may be sensed and/or determined in some fashion. For example, the sensor may be in sensing communication with the fluidic droplet and/or the portion of the fluidic system containing the fluidic droplet fluidly, optically or visually, thermally, pneumatically, electronically, or the like. The sensor can be positioned proximate the fluidic system, for example, embedded within or integrally connected to a wall of a channel, or positioned separately from the fluidic system but with physical, electrical, and/or optical communication with the fluidic system so as to be able to sense and/or determine the fluidic droplet and/or a portion of the fluidic system containing the fluidic droplet (e.g., a channel or a microchannel, a liquid containing the fluidic droplet, etc.). For example, a sensor may be free of any physical connection with a channel containing a droplet, but may be positioned so as to detect electromagnetic radiation arising from the droplet or the fluidic system, such as infrared, ultraviolet, or visible light. The electromagnetic radiation may be produced by the droplet, and/or may arise from other portions of the fluidic system (or externally of the fluidic system) and interact with the fluidic droplet and/or the portion of the fluidic system containing the fluidic droplet in such as a manner as to indicate one or more characteristics of the fluidic droplet, for example, through absorption, reflection, diffraction, refraction, fluorescence, phosphorescence, changes in polarity, phase changes, changes with respect to time, etc. As an example, a laser may be directed towards the fluidic droplet and/or the liquid surrounding the fluidic droplet, and the fluorescence of the fluidic droplet and/or the surrounding liquid may be determined. “Sensing communication,” as used herein may also be direct or indirect. As an example, light from the fluidic droplet may be directed to a sensor, or directed first through a fiber optic system, a waveguide, etc., before being directed to a sensor.
  • Non-limiting examples of detection sensors useful in the invention include optical or electromagnetically-based systems. For example, the sensor may be a fluorescence sensor (e.g., stimulated by a laser), a microscopy system (which may include a camera or other recording device), or the like. As another example, the sensor may be an electronic sensor, e.g., a sensor able to determine an electric field or other electrical characteristic. For example, the sensor may detect capacitance, inductance, etc., of a fluidic droplet and/or the portion of the fluidic system containing the fluidic droplet. In some cases, the sensor may be connected to a processor, which in turn, cause an operation to be performed on the fluidic droplet, for example, by sorting the droplet.
  • Characteristics
  • Characteristics determinable with respect to the droplet and usable in the invention can be identified by those of ordinary skill in the art. Non-limiting examples of such characteristics include fluorescence, spectroscopy (e.g., optical, infrared, ultraviolet, etc.), radioactivity, mass, volume, density, temperature, viscosity, pH, concentration of a substance, such as a biological substance (e.g., a protein, a nucleic acid, etc.), or the like.
  • A corresponding signal is then produced, for example indicating that “yes” the characteristic is present, or “no” it is not. The signal may correspond to a characteristic qualitatively or quantitatively. That is, the amount of the signal can be measured and can correspond to the degree to which a characteristic is present. For example, the strength of the signal may indicate the size of a molecule, or the potency or amount of an enzyme expressed by a cell, or a positive or negative reaction such as binding or hybridization of one molecule to another, or a chemical reaction of a substrate catalyzed by an enzyme. In response to the signal, data can be collected and/or a control system in the sorting module, if present, can be activated to divert a droplet into one branch channel or another for delivery to the collection module or waste module. Thus, in sorting embodiments, molecules or cells within a droplet at a sorting module can be sorted into an appropriate branch channel according to a signal produced by the corresponding examination at a detection module. The means of changing the flow path can be accomplished through mechanical, electrical, optical, or some other technique as described herein.
  • A preferred detector is an optical detector, such as a microscope, which may be coupled with a computer and/or other image processing or enhancement devices to process images or information produced by the microscope using known techniques. For example, molecules can be analyzed and/or sorted by size or molecular weight. Enzymes can be analyzed and/or sorted by the extent to which they catalyze chemical reaction of a substrate (conversely, substrate can be analyzed and/or sorted by the level of chemical reactivity catalyzed by an enzyme). Cells can be sorted according to whether they contain or produce a particular protein, by using an optical detector to examine each cell for an optical indication of the presence or amount of that protein. The protein may itself be detectable, for example by a characteristic fluorescence, or it may be labeled or associated with a reporter that produces a detectable signal when the desired protein is present, or is present in at least a threshold amount. There is no limit to the kind or number of characteristics that can be identified or measured using the techniques of the invention, which include without limitation surface characteristics of the cell and intracellular characteristics, provided only that the characteristic or characteristics of interest for sorting can be sufficiently identified and detected or measured to distinguish cells having the desired characteristic(s) from those which do not. For example, any label or reporter as described herein can be used as the basis for analyzing and/or sorting molecules or cells, i.e. detecting molecules or cells to be collected.
  • Fluorescence Polarization
  • As described herein, the biological/chemical entity to be analyzed may itself be detectable, for example by a characteristic fluorescence, or it may be labeled or associated with a reporter that produces a detectable signal when the desired protein is present, or is present in at least a threshold amount.
  • Luminescent colloidal semiconductor nanocrystals called quantum dots or q-dots (QD) are inorganic fluorophores that have the potential to circumvent some of the functional limitations encountered by organic dyes. In particular, CdSe—ZnS core-shell QDs exhibit size-dependent tunable photoluminescence (PL) with narrow emission bandwidths (FWHM ˜30 to 45 nm) that span the visible spectrum and broad absorption bands. These allow simultaneous excitation of several particle sizes (colors) at a common wavelength. This, in turn, allows simultaneous resolution of several colors using standard instrumentation. CdSe—ZnS QDs also have high quantum yields, are resistant to photodegradation, and can be detected optically at concentrations comparable to organic dyes.
  • Quantum dots are nano-scale semiconductors typically consisting of materials such as crystalline cadmium selenide. The term ‘q-dot’ emphasizes the quantum confinement effect of these materials, and typically refers to fluorescent nanocrystals in the quantum confined size range. Quantum confinement refers to the light emission from bulk (macroscopic) semiconductors such as LEDs which results from exciting the semiconductor either electrically or by shining light on it, creating electron-hole pairs which, when they recombine, emit light. The energy, and therefore the wavelength, of the emitted light is governed by the composition of the semiconductor material. If, however, the physical size of the semiconductor is considerably reduced to be much smaller than the natural radius of the electron-hole pair (Bohr radius), additional energy is required to “confine” this excitation within the nanoscopic semiconductor structure leading to a shift in the emission to shorter wavelengths. Three different q-dots in several concentrations each can be placed in a microdroplet, and can then be used with a microfluidic device to decode what is in the drop. The Q-dot readout extension to the fluorescence station can be incorporated into the design of the microfluidic device. A series of dichroic beamsplitters, emission filters, and detectors can be stacked onto the system, allowing measurement of the required five emission channels (two fluorescence polarization signals and three q-dot bands).
  • Fluorescence Polarization (FP) detection technology enables homogeneous assays suitable for high throughput screening assays in the Drug Discovery field. The most common label in the assays is fluorescein. In FP-assay the fluorophore is excited with polarized light. Only fluorophores parallel to the light absorb and are excited. The excited state has a lifetime before the light emission occurs. During this time the labeled fluorophore molecule rotates and the polarization of the light emitted differs from the excitation plane. To evaluate the polarization two measurements are needed: the first using a polarized emission filter parallel to the excitation filter (S-plane) and the second with a polarized emission filter perpendicular to the excitation filter (P-plane). The Fluorescence Polarization response is given as mP (milli-Polarization level) and is obtained from the equation:
    Polarization (mP)=1000*(S−G*P)/(S+G*P)
    Where S and P are background subtracted fluorescence count rates and G (grating) is an instrument and assay dependent factor.
  • The rotational speed of a molecule is dependent on the size of the molecule, temperature and viscosity of the solution. Fluorescein has a fluorescence lifetime suitable for the rotation speeds of molecules in bio-affinity assays like receptor-ligand binding assays or immunoassays of haptens. The basic principle is that the labeled compound is small and rotates rapidly (low polarization). When the labeled compound binds to the larger molecule, its rotation slows down considerably (polarization changes from low to high polarization). Thus, FP provides a direct readout of the extent of tracer binding to protein, nucleic acids, and other biopolymers.
  • Fluorescence polarization technology has been used in basic research and commercial diagnostic assays for many decades, but has begun to be widely used in drug discovery only in the past six years. Originally, FP assays for drug discovery were developed for single-tube analytical instruments, but the technology was rapidly converted to high-throughput screening assays when commercial plate readers with equivalent sensitivity became available. These assays include such well-known pharmaceutical targets such as kinases, phosphatases, proteases, G-protein coupled receptors, and nuclear receptors. Other homogeneous technologies based on fluorescence intensity have been developed. These include energy transfer, quenching, and enhancement assays. FP offers several advantages over these. The assays are usually easier to construct, since the tracers do not have to respond to binding by intensity changes. In addition, only one tracer is required and crude receptor preparations may be utilized. Furthermore, since FP is independent of intensity, it is relatively immune to colored solutions and cloudy suspensions. FP offers several advantages in the area of instrumentation. Because FP is a fundamental property of the molecule, and the reagents are stable, little or no standardization is required. FP is relatively insensitive to drift in detector gain settings and laser power.
  • The dyes chosen for FP are commonly used in most cell- and enzyme-based assays and are designed not to overlap significantly with the q-dots. The dyes are evaluated both independently and together with the q-dots (at first off-instrument) to assess the cross-talk. Preferably, the liquid q-dot labels are read outside a spectral wavelength band currently used in FACS analysis and sorting (i.e., the dyes flourescein, Cy3, Cy5, etc). This permits the use of currently-available assays (dependent on these dyes). Using specific q-dots, crosstalk is minimized.
  • Accordingly, the present invention provides methods to label droplets and/or nanoreactors formed on a microfluidic device by using only a single dye code to avoid cross-talk with other dyes during FP. Additionally, the present invention provides methods to create FP dye codes to label compounds contained within liquids (including droplets and/or nanoreactors) where the compound is designed to be differentiated by FP on a microfluidic device. In this manner, dye codes having the same color, absorption, and emission could be used to label compounds within liquids.
  • In one aspect, the present invention is directed to the use of fluorescence polarization to label liquids. Droplets can be labeled using several means. These labeling means include, but are not limited to, the use of different dyes, quantum dots, capacitance, opacity, light scattering, fluorecence intensity (FI), fluorescence lifetime (FL), fluorescence polarization (FP), circular dichroism (CD), fluorescenece correlation and combinations of all of these previous labeling means. The following disclosure describes the use of FP and FI as a means to label droplets on a microfluidic device. In addition, the use of FL as a means to adjust the overall FP of a solution, and by varying the concentration of the total FI, to create a 2-dimensional encoding scheme is demonstrated.
  • In general, molecules that take up more volume will tumble slower than a smaller molecule coupled to the same fluorophore (see FIG. 16). FP is independent of the concentration of the dye; liquids can have vastly different concentrations of FITC in them yet still have identical FP measurements.
  • In a preferred embodiment, a FP dye is an organic dye that does not interfere with the assay dye is used. Furthermore, since the total intensity of the FP dye can be quantified, a second dimension in which to label the droplet is provided. Thus, one can exploit the differences in FP to create an encoding scheme of dye within a liquid solution, including droplets. An example is shown in FIG. 17 whereby the droplets are labeled with 3 differently-sized FITC molecules (i.e., three different droplets contain FITC molecules and FITC coupled to either biotin or streptavidin, respectively). Therefore, in a single dimension, FP can be used to create an encoding scheme. However, the present invention can also use Fluorescence Intensity (FI) of the overall solution to create even more labels in a second dimension. An example of labeling droplets in 2 dimensions is shown in FIG. 18.
  • Interestingly, the differences of the fluorescence lifetime (FL) of two dyes with spectral overlap in the detected emission wavelength to change the overall FP of the combined solution can also be exploited (see FIGS. 18 and 19).
  • Although FIG. 17 discusses the use of multiple compounds to which a dye molecule is attached to span a range of FP, it is also possible to span the range using a high and low molecular weight compound set. As exemplified by FIG. 19, a dye can be attached to a large compound (for example streptavidin) and kept at a fixed concentration, to which a smaller compound (for example, a free dye molecule) would be titrated into the same solution. The FP of the solution can be adjusted to be in discernable increments from the value of the large molecule to somewhere slightly greater than the FP of the smaller molecule. The [total] dye intensity can be varied by varying the concentration of the mixture of the two dye-attached compounds. By varying total dye concentration and the FP, two dimensions can be used to generate the FP dye codes (FPcodes). Accordingly, many FPcodes can be generated using only two compounds.
  • This could also include use of large fluorescent proteins such as GFP and the phycobiliproteins combined with a smaller molecule.
  • Examples of dyes commonly used in biological dyes are listed in the table below.
    Excitation Emission
    Wavelength Wavelength Examples of Compatible Dyes
    450 500 Cyan 500
    483 533 SYBR Green, FAM
    523 568 HEX, VIC
    558 610 RED 610
    615 640 RED 640
    650 670 CY5
  • In another aspect, the present invention is directed labeling solids using properties other than dye emission and dye concentration. In one embodiment the solid can include, for example, a bead or location on a solid support or chip. As demonstrated above for liquids, FI and FL can be two of many dimensions of characteristics used as labels. By way of non-limiting example, it is possible to use two dyes with different FL to change the overall FP for a solid such as a bead or other mobile solid support.
  • In another embodiment, a linker can be used to couple the dye to the bead. The linker can be varied so as to allow the dye to have differing degrees of freedom in which to rotate (i.e., tumble). Varying the linker in this manner can change the FP of the attached dye, which in unique combinations can be used as a label. In some embodiments, the beads can be swollen in organic solvent and the dyes held in place by hydrophobic forces. In this case, the FP, FI, FL methods described above for liquid labeling can also be used as a means for labeling the beads. A quenching molecule can also be used to change the characteristics of a dye. Such quenching can be continuous or brought about through the interaction of a molecule, such as a peptide or nucleic acid linker, with differing means of bringing molecules together depending on the strength of linker-internal interaction (e.g., a nucleotide stem loop structure of varying lengths).
  • The reactions analyzed on the virtual, random and non-random arrays (discussed briefly below) can be also increased beyond the two (cy3 and cy5 intensities) commonly used for multiplexing. For example, different FP, FI, etc can be used as a read-out.
  • Random array decoding: Beads of the prior art use one or more pre-attached oligonucleotide-coupled beads that are held in place in a fiber-optic faceplate (for example, those used by Illiumina). The oligos on the beads are decoded using sequential hybridization of a labeled complementary oligo. The assay of the prior art uses a separate oligonucleotide complementary zipcode (‘Illumacode’) attached to each type of bead.
  • The invention described herein is superior to the methods of the prior art in that the FP, FI, FL-labeled bead or mobile solid support can be placed into a random array (e.g., a chip as manufactured by Illumina) and the FP, FI, FL used to decode the bead. The FP, FI, FL of the bead can be decoded before using the chip and the different beads ‘mapped’ as to their specific locations. Alternatively, the bead can be decoded during attachment of the assay read-out. Significantly, the methods described by the present invention can be used to pre-determine the location of each bead-type either before, or during analysis.
  • Virtual array decoding: Methods of the prior art use 2 lasers and 3 detectors to differentiate a set of 100 bead-types. The beads-types are differentiated by the FI of two different dyes present in 1 of 10 concentrations (per dye) contained within the bead, and the assay detector is used to measure fluorescein concentration on the bead. The dyes, which are added to organic-solvent swollen beads, are not directly attached to the beads, but remain held within the bead by hydrophobic forces.
  • Using the methods of the present invention as described herein, a second detector to the machines of the prior art used to measure FP can be added, thereby adding a third dimension and extending the encoding scheme beyond the 100 available in the prior art.
  • Non-random array decoding: In chips of the prior art (such as those used by Affymetrix) oligonucleotides are synthesized directly on the chip. Decoding is simply a matter of knowing the location of the assay on the chip.
  • The methods as described herein can be advantageously used in conjunction with such chips to increase the number of things that can be simultaneously analyzed (i.e., multiplexed) on the chip. By way of non-limiting example, Cy3, Cy5, FL and FP can be used as analysis markers for hybridization reactions.
  • The present invention also provides methods for labeling micro or nano-sized droplets using Radio Frequency Identification (RFID). RFID tags can improve the identification of the contents within the droplets. Preferably, the droplets are utilized within a microfluidic device.
  • RFID is an automatic identification method, relying on storing and remotely retrieving data using devices called RFID tags or transponders. An RFID tag is an object that can be attached to or incorporated into a product, animal, or person for the purpose of identification using radio waves. Chip-based RFID tags contain silicon chips and antennae. Passive tags require no internal power source, whereas active tags require a power source. Hitachi has “powder” 0.05mm×0.05mm RFID chips. The new chips are 64 times smaller than the previous record holder, the 0.4mm×0.4mm mu-chips, and nine times smaller than Hitachi's last year prototype, and have room for a 128-bit ROM that can store a unique 38-digit ID number.
  • In one embodiment, a solution containing RFID tags are emulsified into droplets and are used as a label for the identification of the material within the droplet solution. Applications include, but are not limited to; genetics, genomics, proteomics, chemical synthesis, biofuels, and others.
  • Lasers
  • To detect a reporter or determine whether a molecule, cell or particle has a desired characteristic, the detection module may include an apparatus for stimulating a reporter for that characteristic to emit measurable light energy, e.g., a light source such as a laser, laser diode, light emitting diode (LED), high-intensity lamp, (e.g., mercury lamp), and the like. Where a lamp is used, the channels are preferably shielded from light in all regions except the detection module. Where a laser is used, the laser can be set to scan across a set of detection modules from different analysis units. In addition, laser diodes or LED's may be microfabricated into the same chip that contains the analysis units. Alternatively, laser diodes or LED's may be incorporated into a second chip (i.e., a laser diode chip) that is placed adjacent to the analysis or microchip such that the laser light from the diodes shines on the detection module(s).
  • An integrated semiconductor laser and/or an integrated photodiode detector can be included on the substrate in the vicinity of the detection module. This design provides the advantages of compactness and a shorter optical path for exciting and/or emitted radiation, thus minimizing distortion and losses.
  • Fluorescence produced by a reporter is excited using a laser beam focused on molecules (e.g., DNA, protein, enzyme or substrate) or cells passing through a detection region. Fluorescent reporters can include, but are not limited to, rhodamine, fluorescein, Texas red, Cy 3, Cy 5, phycobiliprotein (e.g., phycoerythrin), green fluorescent protein (GFP), YOYO-1 and PicoGreen. In molecular fingerprinting applications, the reporter labels can be fluorescently labeled single nucleotides, such as fluorescein-dNTP, rhodamine-dNTP, Cy3-dNTP, etc.; where dNTP represents dATP, dTTP, dUTP or dCTP. The reporter can also be chemically-modified single nucleotides, such as biotin-dNTP. The reporter can be fluorescently or chemically labeled amino acids or antibodies (which bind to a particular antigen, or fragment thereof, when expressed or displayed by a cell or virus).
  • The device can analyze and/or sort cells based on the level of expression of selected cell markers, such as cell surface markers, which have a detectable reporter bound thereto, in a manner similar to that currently employed using fluorescence-activated cell sorting (FACS) machines. Proteins or other characteristics within a cell, and which do not necessarily appear on the cell surface, can also be identified and used as a basis for sorting. The device can also determine the size or molecular weight of molecules such as polynucleotides or polypeptides (including enzymes and other proteins) or fragments thereof passing through the detection module. Alternatively, the device can determine the presence or degree of some other characteristic indicated by a reporter. If desired, the cells, particles or molecules can be sorted based on this analysis. The sorted cells, particles or molecules can be collected from the outlet channels in collection modules (or discarded in wasted modules) and used as needed. The collected cells, particles or molecules can be removed from the device or reintroduced to the device for additional coalescence, analysis and sorting.
  • Processors
  • As used herein, a “processor” or a “microprocessor” is any component or device able to receive a signal from one or more sensors, store the signal, and/or direct one or more responses (e.g., as described above), for example, by using a mathematical formula or an electronic or computational circuit. The signal may be any suitable signal indicative of the environmental factor determined by the sensor, for example a pneumatic signal, an electronic signal, an optical signal, a mechanical signal, etc.
  • The device of the present invention can comprise features, such as integrated metal alloy components and/or features patterned in an electrically conductive layer, for detecting droplets by broadcasting a signal around a droplet and picking up an electrical signal in proximity to the droplet.
  • Parallel Analysis
  • The droplet content detection can also be achieved by simultaneous detection of contents of multiple droplets in parallel using spectroscopic fluorescence imaging with sensitivity as high as single-molecule limit. One can spatially distribute droplets containing fluorescent entities such as fluorophore biological markers and/or quantum dots in a two-dimensional sheet in a microscopic field-of-view. The filed-of-view of those droplets can then be illuminated by a fluorescence excitation source and the resulting fluorescence can be spectroscopically imaged. Therefore, for a given fluorescence detection sensitivity, the throughput of fluorescence detection compared to a single-drop fluorescence detection method can be increased by a factor of a/b for a given sensitivity, where a is the number of droplets that can be imaged within a given field-of-view, and b is the ratio of the fluorescence sensitivity of a single-drop fluorescence detector compared to that of the multiple drop fluorescence detector. Furthermore, unlike single-drop fluorescent detection method where the drops are flowed through a detection volume so that their residence time in the detection volume, and hence the signal integration time and sensitivity, is limited, the residence time of the droplet in the field-of-view can be unlimited, thereby allowing sensitivity as high as the single-molecule limit.
  • Beads
  • The device of the present invention also comprises the use of beads and methods for analyzing and sorting beads (i.e, bead reader device). The device can read and either sort or not sort droplets containing one or more of a set of two or more beads. Each bead can be differentiated from each other bead within a set. Beads can be separated by several tags including, but not limited to, quantum dyes, fluorescent dyes, ratios of fluorescent dyes, radioactivity, radio-tags, etc. For example, a set of beads containing a ratio of two dyes in discrete amounts with an apparatus for detecting and differentiating beads containing one discrete ratio from the other beads in this set having a different ratio of the two dyes. The microfluidic device can include paramagnetic beads. The paramagnetic beads can introduce and remove chemical components from droplets using droplet coalescence and breakup events. The paramagnetic beads can also be used for sorting droplets.
  • The present invention provides methods of screening molecular libraries on beads through limited-dilusion-loading and then chemical or optical release inside of droplets. Provided are methods for chemical synthesis on a bead and releasing said chemical attached to the bead using a releasing means (chemical, UV light, heat, etc) within a droplet, and then combining a second droplet to the first droplet for further manipulation. For example, tea-bag synthesis of chemicals on a bead simultaneously with a means for identifying said bead (using, for example, a mass spec tag). Using the resulting mixed-chemistry beads in a droplet within a fluid flow, and exposing the beads to UV light to release the chemical synthesized from the bead into the droplet environment. Combining the droplet containing the released chemical with a droplet containing a cell, and performing a cell-based assay. Sorting droplets having the desired characteristics (for example, turn on of a reporter gene), and then analyzing the sorted beads using mass spectroscopy.
  • The device of the present invention can comprise column separation prior to bead sorting. A device containing a channel loaded with a separating means for chromatographically sorting the sample prior to droplet formation. Such separating means could include size, charge, hydrophobicity, atomic mass, etc. The separating can be done isocratic or by use of a means for generating a gradient chemically, (for example using salt or hydrophobicity), electrically, by pressure, or etc. For example, a channel is preloaded with Sepharose size exclusion media. A sample is loaded at one end, and the droplets are formed at an opposing end. The sample separates by size prior to becoming incorporated within a droplet.
  • Sorting Module
  • The microfluidic device of the present invention can further include one or more sorting modules. A “sorting module ” is a junction of a channel where the flow of molecules, cells, small molecules or particles can change direction to enter one or more other channels, e.g., a branch channel for delivery to an outlet module (i.e., collection or waste module), depending on a signal received in connection with an examination in the detection module. Typically, a sorting module is monitored and/or under the control of a detection module, and therefore a sorting module may “correspond” to such detection module. The sorting region is in communication with and is influenced by one or more sorting apparatuses. A sorting apparatus comprises techniques or control systems, e.g., dielectric, electric, electro-osmotic, (micro-) valve, etc. A control system can employ a variety of sorting techniques to change or direct the flow of molecules, cells, small molecules or particles into a predetermined branch channel. A “branch channel” is a channel which is in communication with a sorting region and a main channel. The main channel can communicate with two or more branch channels at the sorting module or “branch point”, forming, for example, a T-shape or a Y-shape. Other shapes and channel geometries may be used as desired. Typically, a branch channel receives molecules, cells, small molecules or particles depending on the molecule, cells, small molecules or particles characteristic of interest as detected by the detection module and sorted at the sorting module. A branch channel can have an outlet module and/or terminate with a well or reservoir to allow collection or disposal (collection module or waste module, respectively) of the molecules, cells, small molecules or particles. Alternatively, a branch channel may be in communication with other channels to permit additional sorting.
  • The device of the present invention can further include one or more outlet modules. An “outlet module” is an area of the device that collects or dispenses molecules, cells, small molecules or particles after coalescence, detection and/or sorting. The outlet module can include a collection module and/or a waste module. The collection module can be connected to a means for storing a sample. The collection module can be a well or reservoir for collecting and containing droplets detected to have a specific predetermined characteristic in the detection module. The collection module can be temperature controlled. The waste module can be connected to a means for discarding a sample. The waste module can be a well or reservoir for collecting and containing droplets detected to not have a specific predetermined characteristic in the detection module. The outlet module is downstream from a sorting module, if present, or downstream from the detection module if a sorting module is not present. The outlet module may contain branch channels or outlet channels for connection to a collection module or waste module. A device can contain more than one outlet module.
  • A characteristic of a fluidic droplet may be sensed and/or determined in some fashion, for example, as described herein (e.g., fluorescence of the fluidic droplet may be determined), and, in response, an electric field may be applied or removed from the fluidic droplet to direct the fluidic droplet to a particular region (e.g. a channel). A fluidic droplet is preferably sorted or steered by inducing a dipole in the uncharged fluidic droplet (which may be initially charged or uncharged), and sorting or steering the droplet using an applied electric field. The electric field may be an AC field, a DC field, etc. For example, with reference to FIG. 20A, a channel 540, containing fluidic droplet 530 and liquid 535, divides into channel 542 and 544. Fluidic droplet 530 is uncharged. Electrode 526 is positioned near channel 542, while electrode 527 is positioned near channel 544. Electrode 528 is positioned near the junction of channels 540, 542, and 544. In FIGS. 20C and 20D, a dipole is induced in the fluidic droplet using electrodes 526, 527, and/or 528. In FIG. 20C, a dipole is induced in droplet 530 by applying an electric field 525 to the droplet using electrodes 527 and 528. Due to the strength of the electric field, the droplet is strongly attracted to the right, into channel 544. Similarly, in FIG. 20D, a dipole is induced in droplet 530 by applying an electric field 525 to the droplet using electrodes 526 and 528, causing the droplet to be attracted into channel 542. Thus, by applying the proper electric field, droplet 530 can be directed to either channel 542 or 544 as desired.
  • The present invention also provides improvements in the efficiency, accuracy, and reliability of the preferred dielectric droplet sorting technique described above. The single sided dielectric sorting relies on a combination of flow imbalance between the two exhaust legs and a switchable electric field to selectively sort out droplets of interest from the main sample stream. Sorting decisions are made based on some form of real time measurement of the droplet and its contents. FIGS. 21 and 22 depict many of the various possible fluid and electrode geometries possible for single sided dielectric sorting. FIG. 21, Panels A-D show possible flow channel geometries that can be used in an asymmetric sorting application. Panel F, illustrates the use of a barrier, for example, a barrier where no fluid flow passes on its left side. Note these designs are only conceptual representation of the fluid channels, and actual designs may differ in absolute and relative dimensions as determined by one of ordinary skill in the art.
  • FIG. 22 shows the possible electrode geometries used in an asymmetric sorting application. Panel A shows the use of sharp tipped electrodes. Panel B shows broad tipped electrodes to increase the interaction time between the droplets and the electric field (the tips could be many drop diameters long). Panel C shows electrodes straddling the collection line. Panel D shows electrodes on opposite sides of the main channel. Panel E shows an Asymmetric Electrode Pair (the asymmetry may be present on any of the other electrode pair layouts as well). Note these designs are only conceptual representation of the electrodes, and actual designs may differ in absolute dimensions and electrode shape as determined by one of ordinary skill in the art. Although the fluid channel geometry is drawn as a “Y” junction, any of the channel geometries shown in FIG. 21 could be substituted in these drawings.
  • Typically, the flow rate of the collection leg is set to a value just below the level required to begin pulling droplets into the collection line (indicated as 40% in the figures, although the actual value may be differ from this and is dependent on the actual fluidic and electrode geometry, total flow, as well as droplet size and composition).
  • As an alternative design strategy, the collection leg can be operated at a flow rate at which the droplets would normally flow down the Sort collect line (i.e. change the flow splits shown in the diagrams from 40% collect/60% waste to 60% collect/40% waste), and keep the electric field energized until a droplet of interest is detected. At that time, the field would be briefly turned off, and the droplet would be pulled down the collection leg based on fluidic forces instead of electrical forces.
  • Alternately, a fluidic droplet may be directed by creating an electric charge (e.g., as previously described) on the droplet, and steering the droplet using an applied electric field, which may be an AC field, a DC field, etc. As an example, an electric field maybe selectively applied and removed (or a different electric field may be applied) as needed to direct the fluidic droplet to a particular region. The electric field may be selectively applied and removed as needed, in some embodiments, without substantially altering the flow of the liquid containing the fluidic droplet. For example, a liquid may flow on a substantially steady-state basis (i.e., the average flowrate of the liquid containing the fluidic droplet deviates by less than 20% or less than 15% of the steady-state flow or the expected value of the flow of liquid with respect to time, and in some cases, the average flowrate may deviate less than 10% or less than 5%) or other predetermined basis through a fluidic system of the invention (e.g., through a channel or a microchannel), and fluidic droplets contained within the liquid may be directed to various regions, e.g., using an electric field, without substantially altering the flow of the liquid through the fluidic system.
  • In other embodiments, however, the fluidic droplets may be screened or sorted within a fluidic system of the invention by altering the flow of the liquid containing the droplets. For instance, in one set of embodiments, a fluidic droplet may be steered or sorted by directing the liquid surrounding the fluidic droplet into a first channel, a second channel, etc.
  • In another set of embodiments, pressure within a fluidic system, for example, within different channels or within different portions of a channel, can be controlled to direct the flow of fluidic droplets. For example, a droplet can be directed toward a channel junction including multiple options for further direction of flow (e.g., directed toward a branch, or fork, in a channel defining optional downstream flow channels). Pressure within one or more of the optional downstream flow channels can be controlled to direct the droplet selectively into one of the channels, and changes in pressure can be effected on the order of the time required for successive droplets to reach the junction, such that the downstream flow path of each successive droplet can be independently controlled. In one arrangement, the expansion and/or contraction of liquid reservoirs may be used to steer or sort a fluidic droplet into a channel, e.g., by causing directed movement of the liquid containing the fluidic droplet. The liquid reservoirs may be positioned such that, when activated, the movement of liquid caused by the activated reservoirs causes the liquid to flow in a preferred direction, carrying the fluidic droplet in that preferred direction. For instance, the expansion of a liquid reservoir may cause a flow of liquid towards the reservoir, while the contraction of a liquid reservoir may cause a flow of liquid away from the reservoir. In some cases, the expansion and/or contraction of the liquid reservoir may be combined with other flow-controlling devices and methods, e.g., as described herein. Non-limiting examples of devices able to cause the expansion and/or contraction of a liquid reservoir include pistons and piezoelectric components. In some cases, piezoelectric components may be particularly useful due to their relatively rapid response times, e.g., in response to an electrical signal.
  • In some embodiments, the fluidic droplets may be sorted into more than two channels. Alternately, a fluidic droplet may be sorted and/or split into two or more separate droplets, for example, depending on the particular application. Any of the above-described techniques may be used to spilt and/or sort droplets. As a non-limiting example, by applying (or removing) a first electric field to a device (or a portion thereof), a fluidic droplet may be directed to a first region or channel; by applying (or removing) a second electric field to the device (or a portion thereof), the droplet may be directed to a second region or channel; by applying a third electric field to the device (or a portion thereof), the droplet may be directed to a third region or channel; etc., where the electric fields may differ in some way, for example, in intensity, direction, frequency, duration, etc. In a series of droplets, each droplet may be independently sorted and/or split; for example, some droplets may be directed to one location or another, while other droplets may be split into multiple droplets directed to two or more locations.
  • In some cases, high sorting speeds may be achievable using certain systems and methods of the invention. For instance, at least about 1 droplet per second may be determined and/or sorted in some cases, and in other cases, at least about 10 droplets per second, at least about 20 droplets per second, at least about 30 droplets per second, at least about 100 droplets per second, at least about 200 droplets per second, at least about 300 droplets per second, at least about 500 droplets per second, at least about 750 droplets per second, at least about 1000 droplets per second, at least about 1500 droplets per second, at least about 2000 droplets per second, at least about 3000 droplets per second, at least about 5000 droplets per second, at least about 7500 droplets per second, at least about 10,000 droplets per second, at least about 15,000 droplets per second, at least about 20,000 droplets per second, at least about 30,000 droplets per second, at least about 50,000 droplets per second, at least about 75,000 droplets per second, at least about 100,000 droplets per second, at least about 150,000 droplets per second, at least about 200,000 droplets per second, at least about 300,000 droplets per second, at least about 500,000 droplets per second, at least about 750,000 droplets per second, at least about 1,000,000 droplets per second may be determined and/or sorted in such a fashion.
  • Multiple Measurement Sorting
  • In some embodiments, it may be useful to sort droplets based on two different measurements. For example, one might want to sort based on the ratio of two signals, sum of two signals, or difference between two signals. Specifically, this would be useful for cases when one would like to optimize an enzyme so that it work one substrate, but not another, or so that it works on two substrates. This is not easy to do using multiple rounds of selection on populations of droplets. To overcome this shortcoming of current sorting technology, the present invention provides a device comprising multiple channels with the appropriate geometry to split droplets, perform different experiments on the two daughter droplets and then reorder so that they pass sequential through the detector. The sums, ratios or differences in the two signals can then be calculated before the droplets enter the sorting bifurcation. An indicator dye or equivalent material may be added to one or both droplets to indicate when each droplet enters and leaves the laser. A representative sketch is shown in FIG. 23.
  • Sample Recovery
  • The present invention proposes methods for recovering aqueous phase components from aqueous emulsions that have been collected on a microfluidic device in a minimum number of steps and in a gentle manner so as to minimize potential damage to cell viability.
  • In one aspect, a stable aqueous sample droplet emulsion containing aqueous phase components in a continuous phase carrier fluid is allowed to cream to the top of the continuous phase carrier oil. By way of nonlimiting example, the continuous phase carrier fluid can include a perfluorocarbon oil that can have one or more stabilizing surfactants. The aqueous emulsion rises to the top or separates from the continuous phase carrier fluid by virtue of the density of the continuous phase fluid being greater than that of the aqueous phase emulsion. For example, the perfluorocarbon oil used in one embodiment of the device is 1.8, compared to the density of the aqueous emulsion, which is 1.0.
  • The creamed emulsion is then placed onto a second continuous phase carrier fluid which contains a de-stabilizing surfactant, such as a perfluorinated alchohol (e.g. 1H,1H,2H,2H-Perfluoro-1-octanol). The second continuous phase carrier fluid can also be a perfluorocarbon oil. Upon mixing, the aqueous emulsion begins to coalesce, and coalescence is completed by brief centrifugation at low speed (e.g., 1 minute at 2000 rpm in a microcentrifuge). The coalesced aqueous phase can now be removed (cells can be placed in an appropriate environment for further analysis).
  • Additional destabilizing surfactants and/or oil combinations can be identified or synthesized to be useful with this invention.
  • Mixing Module
  • The microfluidic device of the present invention can further include one or more mixing modules. Although coalescence of one or more droplets in one or more coalescence modules can be sufficient to mix the contents of the coalesced droplets (e.g., through rotating vortexes existing within the droplet), it should be noted that when two droplets fuse or coalesce, perfect mixing within the droplet does not instantaneously occur. Instead, for example, the coalesced droplet may initially be formed of a first fluid region (from the first droplet) and a second fluid region (from the second droplet). Thus, in some cases, the fluid regions may remain as separate regions, for example, due to internal “counter-revolutionary” flow within the fluidic droplet, thus resulting in a non-uniform fluidic droplet. A “mixing module” can comprise features for shaking or otherwise manipulate droplets so as to mix their contents. The mixing module is preferably downstream from the coalescing module and upstream from the detection module. The mixing module can include, but is not limited to, the use of channel geometries, acoustic actuators, metal alloy component electrodes or electrically conductive patterned electrodes to mix the contents of droplets and to reduce mixing times for fluids combined into a single droplet in the microfluidic device. For example, the fluidic droplet may be passed through one or more channels or other systems which cause the droplet to change its velocity and/or direction of movement. The change of direction may alter convection patterns within the droplet, causing the fluids to be at least partially mixed. Combinations are also possible.
  • For acoustic manipulation, the frequency of the acoustic wave should be fine tuned so as not to cause any damage to the cells. The biological effects of acoustic mixing have been well studied (e.g., in the ink-jet industry) and many published literatures also showed that piezoelectric microfluidic device can deliver intact biological payloads such as live microorganisms and DNA. In an example, the design of the acoustic resonant uses a Piezoelectric bimorph flat plate located on the side of the carved resonant in the PDMS slab. The piezoelectric driving waveform is carefully optimized to select the critical frequencies that can separate cells in fluids. There are five parameters to optimize beyond the frequency parameter. Lab electronics is used to optimize the piezoelectric driving waveform. Afterwards, a low cost circuit can be designed to generate only the optimized waveform in a preferred microfluidic device.
  • Other examples of fluidic mixing in droplets are described WO 2004/091763, incorporated herein by reference.
  • Delay Module
  • The microfluidic device of the present invention can further include one or more delay modules. The “delay module” can be a delay line. The operation of a microfluidics device where a reaction within a droplet is allowed to occur for a non-trivial length of time requires a delay line to increase the residence time within the device. For reactions demanding extensive residence time, longer or larger delay lines are required. Accordingly, the invention provides methods to increase residence times within microfluidic devices.
  • The delay module is in fluid communication with the main channel or it can be an elongated portion of the main channel itself. The delay module can be located downstream of the coalescence module and upstream of the detection module. The delay module can be a serpentine channel or a buoyant hourglass. The delay module can further comprise heating and cooling regions. The heating and cooling regions can be used for performing on-chip, flow-through PCR as further described herein.
  • The channel dimensions and configurations can be designed to accommodate the required residence time with minimum pressure drops across the device. For example, to accommodate very long delay lines within the microfluidic device, the device can comprise a multilayered PDMS slab which is composed of several patterned PDMS slabs.
  • The channel dimensions can also be designed so as to allow for required flow, residence time and pressure drop. Some channels may be required to be very large in width and height. In order to avoid collapse of the channels, the device includes support posts within the channel design. In order to reduce dead volume behind posts and further improve droplet stability, the support posts are designed to optimize a streamlined flow within the channel. These designs can include curved features as opposed to sharp edges.
  • To allow for longer period of device operation, delay lines can also be extended to the outside of the chip. The off-chip delay lines can be tubes within micron-sized internal diameter.
  • In order to allow more efficient use of available space and faster operation, in methods where droplets are charged, after charging, asymmetric splitting of oil and drops can be accommodated by siphoning off oil from channels after droplets are charged.
  • The delay lines can be in the form of a tower (i.e., a structure which is vertical with respect to the ambient gravitational field) as to allow buoyant forces to assist controlled droplet transport. Known delay lines involve transporting droplets by emulsifying them in a carrier fluid flowing in a channel and/or tube. Because the velocity profile of the carrier fluid through the cross-section of the channel and/or tube is not uniform, the velocity distribution of the droplets will not be narrow, which causes the delay time distribution of the droplets to not be narrow (i.e., some droplets will be delayed more or less than others).
  • The devices of the present invention can also include buoyancy-assisted microfluidic delay lines. In buoyancy-assisted microfluidic delay lines, buoyant forces act on droplets emulsified in a fluid in one or more towers. This can include allowing the tower to fill for the desired delay time, and then releasing the droplets. The tower can or can not continue to fill and release droplets as needed. In this example, one may desire to have a cylindrical tower section that is capped by a pyramidal funnel section. The tower can effectively functions as an hourglass. Droplets that have a density less than their carrier fluid are fed into the base of the tower, buoyantly rise to the top of the tower with a substantially uniform velocity distribution, and are funneled into a functional component of the microfluidic device (such as a y-branch). Carrier fluid is exhausted at the base of the tower at the same rate as it is introduced at the apex so that the net flow of carrier fluid through the delay line is zero. The tower and funnel sections can have any cross-sectional shape, such as circular, elliptical, or polygonal. The microfluidic device can include a tower with adjustable length.
  • The device can also include a switching network of twenty towers to guarantee a delay time dispersion of 5% (because 1/20=0.05). The capacity of each tower is 0.05*T, where T is the delay time. The concept includes, for example: (a) upon device start-up, filling the first tower for 0.05*T, but stop-cock its exhaust, and also have the other nineteen towers closed; (b) after 0.05*T, closing the first tower and filling the second between 0.05*T and 0.1 0*T; (c) repeating step (b) for the remaining eighteen towers; (d) at time T, allowing the first tower to exhaust; (e) at time 1.05*T, stop-cocking the exhaust of the first tower, allowing the second tower to exhaust, and allowing the first tower to fill; (f) at time 1.10*T, stop-cocking the exhaust of the second tower, allowing the third tower to exhaust, closing the first tower, and allowing the second tower to fill, and (g) repeating step (f) ad infinitum. More than twenty towers may provide an even tighter control over the width of the delay time dispersion. This scheme may require a valve network. This network of towers can be outside the microfluidic device.
  • The delay module can also include channels (e.g. the main channel) which has an altered geometry which permits the “parking” (e.g., slowing or stopping) of droplets within the microfluidic device.
  • In the methods provided herein, droplets are able to be parked in wells or channels at predefined locations. This can be done by creating discrete well-like indentions in the channel whereby a droplet ‘falls’ into the well and remains there as the fluid flows over it, or by using a technique entitled ‘by-pass pots’ whereby a droplet is used to block a small outlet in a well, thereby causing the flow to by-pass that droplet-containing well.
  • The instant invention is to use either of these techniques or any related technique, for example just stopping the drops in a channel, to position droplets at either random or predefined places within a microfluidics device. These random or predefined locations can then be queried at a later time-point for a reaction to have occurred, or for removal of the droplets using another means such as resuspension followed by aspiration.
  • In one example, a rolling circle amplification reaction is initiated in droplets, the droplets are then parked within the chip, and the amplification reaction allowed to proceed for a set period of time prior to stopping the reaction through the use of heat. The parked droplets are then dried in situ and the covering of the chip disassembled from the chip. One or a set of needle-like devices that are able to be lined up with the droplet parking space are then placed adjacent to or on top of the dried droplets and a liquid solution used to resuspend the material in the dried droplet that has been deposited into the chip, for further downstream processing.
  • In another example, to avoid possible diffusion of reactant contents from a first and a second set of reactions in droplets, the first reactions are created in 10 μm droplets, the droplets are dried within a channel parking space or by-pass pot which is able to hold a droplet of size larger than 10 μm, and the droplets are dried in situ. A second set of droplets that are larger than 10 μm are then allowed to proceed down said channel and when caught in said parking space or by-pass pot are able to resuspend the material from the first droplets that are dried along the walls of the first parking space or by-pass pot. In doing so, the second droplet is slightly larger than the first and that ensures that the material along the walls is ‘captured’ by the second droplet, and not allowed to diffuse away from the first droplet wall by diffusion. By doing so, use of surfactants becomes optional in either the first or second droplet formulations.
  • The instant invention also provides the following devices and methods for use in practicing the methods described herein. The PDMS substrate which comprises a portion of the microfluidic device can be covered or coated with an adhesive tape or strip that can removed by peeling. The PDMS substrate can also be bonded by an ultra thin silica that can be pierced by a set of needles. The silica may be spin coated or electro-plated onto a thin backing. Droplets can be dried onto a piece of paper such that can be detected by a second device to determine the Ncode within the droplet and to determine whether an amplification reaction has occurred within the droplet. A plate read comprising dried and undried spots using either an optical array device, such as found in high-end cameras or fiber, optic device is also contemplated. Dry Nitrogen can be utilized to dry the spots by either flowing it through the channel or placing the device into a dry-N2 chamber. Channels can be filled with dried nitrogen or salt run underneath or adjacent to the parking space channels to allow chemical or physical-type gradients to be set up in the chip. The channel walls can be coated with Steptavidin and the produced reactants, for example, DNA biotinylated so that it adheres in situ. Porous beads deposited into the wells can be used in combination with solutions without oils to wash the beads by flow, followed by re-depositing droplets with surfactants to recoat the beads. The wells within the substrate can be filled with many small beads by loading small beads into droplets, storing the droplets into individual wells containing apertures that are slightly smaller than the beads, breaking the droplets by drying or flow of aqueous solutions with or without surfactants into the channels and past the beads, and then re-encapsulating the beads in situ. A set of electrodes within or adjacent to the microfluidic substrate can be used to fuse two droplets in a storage/holding space. The electrodes may be perpendicular to the plane of the channels and either the electrodes or channels moved so as to allow droplet fusions to occur.
  • UV-Release Module
  • The microfluidic device of the present invention can further include one or more UV-release modules. The “UV-release module” is in fluid communication with the main channel. The UV-release module is located downstream of the inlet module and upstream of the coalescence module. The UV-module can be a used in bead assays. Compounds from encapsulated beads can be cleaved in a UV-releasing module using UV light. Photolabile linkers can be broken down on demand after a single bead has been encapsulated thus releasing multiple copies of a single compound into solution. In the cell based assay disclosed herein the chemical compound assayed is desired to be in solution in order to penetrate the cell membrane. Furthermore, to ensure compartmentalization of a single compound with a cell the cleavage of the compound from the solid support can only be done after the bead has been encapsulated. Photocleavable linkers can be utilized to cleave the compounds of the bead after drop formation by passing the drop through a UV-release module (i.e., laser of the appropriate wavelength).
  • The present invention also provides methods for chemical synthesis on a bead and releasing said chemical attached to the bead using a releasing means (chemical, UV light, heat, etc) within a droplet, and then combining a second droplet to the first droplet for further manipulation. Preferably, the releasing means is a UV-module. For example, tea-bag synthesis of chemicals on a bead simultaneously with a means for identifying said bead (using, for example, a mass spec tag). Using the resulting mixed-chemistry beads in a droplet within a fluid flow, and exposing the beads to UV light to release the chemical synthesized from the bead into the droplet environment. Combining the droplet containing the released chemical with a droplet containing a cell, and performing a cell-based assay. Sorting droplets having the desired characteristics (for example, turn on of a reporter gene), and then analyzing the sorted beads using mass spectroscopy.
  • Kits
  • As a matter of convenience, predetermined amounts of the reagents, compound libraries, and/or emulsions described herein and employed in the present invention can be optionally provided in a kit in packaged combination to facilitate the application of the various assays and methods described herein. Such kits also typically include instructions for carrying out the subject assay, and may optionally include the fluid receptacle, e.g., the cuvette, multiwell plate, microfluidic device, etc. in which the reaction is to be carried out.
  • Typically, reagents included within the kit are uniquely labeled emulsions containing tissues, cells, particles, proteins, antibodies, amino acids, nucleotides, small molecules, substrates, and/or pharmaceuticals. These reagents may be provided in pre-measured container (e.g., vials or ampoules) which are co-packaged in a single box, pouch or the like that is ready for use. The container holding the reagents can be configured so as to readily attach to the fluid receptacle of the device in which the reaction is to be carried out (e.g., the inlet module of the microfluidic device as described herein). In one embodiment, the kit can include an RNAi kit. In another embodiment, the kit can include a chemical synthesis kit. It will be appreciated by persons of ordinary skill in the art that these embodiments are merely illustrative and that other kits are also within the scope of the present invention.
  • Methods
  • The microfluidic device of the present invention can be utilized to conduct numerous chemical and biological assays, including but not limited to, creating emulsion libraries, flow cytometry, gene amplification, isothermal gene amplification, DNA sequencing, SNP analysis, drug screening, RNAi analysis, karyotyping, creating microbial strains with improved biomass conversion, moving cells using optical tweezer/cell trapping, transformation of cells by electroporation, μTAS, and DNA hybridization.
  • Definitions
  • The terms used in this specification generally have their ordinary meanings in the art, within the context of this invention and in the specific context where each term is used. Certain terms are discussed below, or elsewhere in the specification, to provide additional guidance to the practitioner in describing the devices and methods of the invention and how to make and use them. It will be appreciated that the same thing can typically be described in more than one way. Consequently, alternative language and synonyms may be used for any one or more of the terms discussed herein. Synonyms for certain terms are provided. However, a recital of one or more synonyms does not exclude the use of other synonyms, nor is any special significance to be placed upon whether or not a term is elaborated or discussed herein. All publications, patent applications, patents, and other references mentioned herein are incorporated by reference. In the case of conflict, the present specification, including definitions, will control. In addition, the materials, methods, and examples are illustrative only and are not intended to be limiting.
  • The invention is also described by means of particular examples. However, the use of such examples anywhere in the specification, including examples of any terms discussed herein, is illustrative only and in no way limits the scope and meaning of the invention or of any exemplified term. Likewise, the invention is not limited to any particular preferred embodiments described herein. Indeed, many modifications and variations of the invention will be apparent to those skilled in the art upon reading this specification and can be made without departing from its spirit and scope. The invention is therefore to be limited only by the terms of the appended claims along with the full scope of equivalents to which the claims are entitled.
  • As used herein, “about” or “approximately” shall generally mean within 20 percent, preferably within 10 percent, and more preferably within 5 percent of a given value or range.
  • The term “molecule” means any distinct or distinguishable structural unit of matter comprising one or more atoms, and includes for example polypeptides and polynucleotides.
  • The term “polymer” means any substance or compound that is composed of two or more building blocks (‘mers’) that are repetitively linked to each other. For example, a “dimer” is a compound in which two building blocks have been joined together.
  • The term “polynucleotide” as used herein refers to a polymeric molecule having a backbone that supports bases capable of hydrogen bonding to typical polynucleotides, where the polymer backbone presents the bases in a manner to permit such hydrogen bonding in a sequence specific fashion between the polymeric molecule and a typical polynucleotide (e.g., single-stranded DNA). Such bases are typically inosine, adenosine, guanosine, cytosine, uracil and thymidine. Polymeric molecules include double and single stranded RNA and DNA, and backbone modifications thereof, for example, methylphosphonate linkages.
  • Thus, a “polynucleotide” or “nucleotide sequence” is a series of nucleotide bases (also called “nucleotides”) generally in DNA and RNA, and means any chain of two or more nucleotides. A nucleotide sequence typically carries genetic information, including the information used by cellular machinery to make proteins and enzymes. These terms include double or single stranded genomic and cDNA, RNA, any synthetic and genetically manipulated polynucleotide, and both sense and anti-sense polynucleotide (although only sense stands are being represented herein). This includes single- and double-stranded molecules, i.e., DNA-DNA, DNA-RNA and RNA-RNA hybrids, as well as “protein nucleic acids” (PNA) formed by conjugating bases to an amino acid backbone. This also includes nucleic acids containing modified bases, for example thio-uracil, thio-guanine and fluoro-uracil.
  • The polynucleotides herein may be flanked by natural regulatory sequences, or may be associated with heterologous sequences, including promoters, enhancers, response elements, signal sequences, polyadenylation sequences, introns, 5′- and 3′-non-coding regions, and the like. The nucleic acids may also be modified by many means known in the art. Non-limiting examples of such modifications include methylation, “caps”, substitution of one or more of the naturally occurring nucleotides with an analog, and internucleotide modifications such as, for example, those with uncharged linkages (e.g., methyl phosphonates, phosphotriesters, phosphoroamidates, carbamates, etc.) and with charged linkages (e.g., phosphorothioates, phosphorodithioates, etc.). Polynucleotides may contain one or more additional covalently linked moieties, such as, for example, proteins (e.g., nucleases, toxins, antibodies, signal peptides, poly-L-lysine, etc.), intercalators (e.g., acridine, psoralen, etc.), chelators (e.g., metals, radioactive metals, iron, oxidative metals, etc.), and alkylators. The polynucleotides may be derivatized by formation of a methyl or ethyl phosphotriester or an alkyl phosphoramidate linkage. Furthermore, the polynucleotides herein may also be modified with a label capable of providing a detectable signal, either directly or indirectly. Exemplary labels include radioisotopes, fluorescent molecules, biotin, and the like.
  • The term “dielectrophoretic force gradient” means a dielectrophoretic force is exerted on an object in an electric field provided that the object has a different dielectric constant than the surrounding media. This force can either pull the object into the region of larger field or push it out of the region of larger field. The force is attractive or repulsive depending respectively on whether the object or the surrounding media has the larger dielectric constant. “DNA” (deoxyribonucleic acid) means any chain or sequence of the chemical building blocks adenine (A), guanine (G), cytosine (C) and thymine (T), called nucleotide bases, that are linked together on a deoxyribose sugar backbone. DNA can have one strand of nucleotide bases, or two complimentary strands which may form a double helix structure. “RNA” (ribonucleic acid) means any chain or sequence of the chemical building blocks adenine (A), guanine (G), cytosine (C) and uracil (U), called nucleotide bases, that are linked together on a ribose sugar backbone. RNA typically has one strand of nucleotide bases.
  • A “polypeptide” (one or more peptides) is a chain of chemical building blocks called amino acids that are linked together by chemical bonds called peptide bonds. A “protein” is a polypeptide produced by a living organism. A protein or polypeptide may be “native” or “wild-type”, meaning that it occurs in nature; or it may be a “mutant”, “variant” or “modified”, meaning that it has been made, altered, derived, or is in some way different or changed from a native protein, or from another mutant.
  • An “enzyme” is a polypeptide molecule, usually a protein produced by a living organism, that catalyzes chemical reactions of other substances. The enzyme is not itself altered or destroyed upon completion of the reaction, and can therefore be used repeatedly to catalyze reactions. A “substrate” refers to any substance upon which an enzyme acts.
  • As used herein, “particles” means any substance that may be encapsulated within a droplet for analysis, reaction, sorting, or any operation according to the invention. Particles are not only objects such as microscopic beads (e.g., chromatographic and fluorescent beads), latex, glass, silica or paramagnetic beads, but also includes other encapsulating porous and/or biomaterials such as liposomes, vesicles and other emulsions. Beads ranging in size from 0.1 micron to 1 mm can be used in the devices and methods of the invention and are therefore encompassed with the term “particle” as used herein. The term particle also encompasses biological cells, as well as beads and other microscopic objects of similar size (e.g., from about 0.1 to 120 microns, and typically from about 1 to 50 microns) or smaller (e.g., from about 0.1 to 150 nm). The devices and methods of the invention are also directed to sorting and/or analyzing molecules of any kind, including polynucleotides, polypeptides and proteins (including enzymes) and their substrates and small molecules (organic or inorganic). Thus, the term particle further encompasses these materials.
  • The particles (including, e.g., cells and molecules) are sorted and/or analyzed by encapsulating the particles into individual droplets (e.g., droplets of aqueous solution in oil), and these droplets are then sorted, combined and/or analyzed in a microfabricated device. Accordingly, the term “droplet” generally includes anything that is or can be contained within a droplet.
  • A “small molecule” as used herein, is meant to refer to a composition that has a molecular weight of less than about 5 kD and most preferably less than about 4 kD. Small molecules can be, e.g., nucleic acids, peptides, polypeptides, peptidomimetics, carbohydrates, lipids or other organic or inorganic molecules. Libraries of chemical and/or biological mixtures, such as fungal, bacterial, or algal extracts, are known in the art.
  • As used herein, “cell” means any cell or cells, as well as viruses or any other particles having a microscopic size, e.g. a size that is similar to or smaller than that of a biological cell, and includes any prokaryotic or eukaryotic cell, e.g., bacteria, fungi, plant and animal cells. Cells are typically spherical, but can also be elongated, flattened, deformable and asymmetrical, i.e., non-spherical. The size or diameter of a cell typically ranges from about 0.1 to 120 microns, and typically is from about 1 to 50 microns. A cell may be living or dead. Since the microfabricated device of the invention is directed to sorting materials having a size similar to a biological cell (e.g. about 0.1 to 120 microns) or smaller (e.g., about 0.1 to 150 nm) any material having a size similar to or smaller than a biological cell can be characterized and sorted using the microfabricated device of the invention. Thus, the term cell shall further include microscopic beads (such as chromatographic and fluorescent beads), liposomes, emulsions, or any other encapsulating biomaterials and porous materials. Non-limiting examples include latex, glass, orparamagnetic beads; and vesicles such as emulsions and liposomes, and other porous materials such as silica beads. Beads ranging in size from 0.1 micron to 1 mm can also be used, for example in sorting a library of compounds produced by combinatorial chemistry. As used herein, a cell may be charged or uncharged. For example, charged beads may be used to facilitate flow or detection, or as a reporter. Biological cells, living or dead, may be charged for example by using a surfactant, such as SDS (sodium dodecyl sulfate). The term cell further encompasses “virions”, whether or not virions are expressly mentioned.
  • A “virion”, “virus particle” is the complete particle of a virus. Viruses typically comprise a nucleic acid core (comprising DNA or RNA) and, in certain viruses, a protein coat or “capsid”. Certain viruses may have an outer protein covering called an “envelope”. A virion may be either living (i.e., “viable”) or dead (i.e., “non-viable”). A living or “viable” virus is one capable of infecting a living cell. Viruses are generally smaller than biological cells and typically range in size from about 20-25 nm diameter or less (parvoviridae, picomoviridae) to approximately 200-450 nm (poxviridae). However, some filamentous viruses may reach lengths of 2000 nm (closterviruses) and are therefore larger than some bacterial cells. Since the microfabricated device of the invention is particularly suited for sorting materials having a size similar to a virus (i.e., about 0.1 to 150 nm), any material having a size similar to a virion can be characterized and sorted using the microfabricated device of the invention. Non-limiting examples include latex, glass or paramagnetic beads; vesicles such as emulsions and liposomes; and other porous materials such as silica beads. Beads ranging in size from 0.1 to 150 nm can also be used, for example, in sorting a library of compounds produced by combinatorial chemistry. As used herein, a virion may be charged or uncharged. For example, charged beads may be used to facilitate flow or detection, or as a reporter. Biological viruses, whether viable or non-viable, may be charged, for example, by using a surfactant, such as SDS.
  • A “reporter” is any molecule, or a portion thereof, that is detectable, or measurable, for example, by optical detection. In addition, the reporter associates with a molecule, cell or virion or with a particular marker or characteristic of the molecule, cell or virion, or is itself detectable to permit identification of the molecule, cell or virion's, or the presence or absence of a characteristic of the molecule, cell or virion. In the case of molecules such as polynucleotides such characteristics include size, molecular weight, the presence or absence of particular constituents or moieties (such as particular nucleotide sequences or restrictions sites). In the case of cells, characteristics which may be marked by a reporter includes antibodies, proteins and sugar moieties, receptors, polynucleotides, and fragments thereof. The term “label” can be used interchangeably with “reporter”. The reporter is typically a dye, fluorescent, ultraviolet, or chemiluminescent agent, chromophore, or radio-label, any of which may be detected with or without some kind of stimulatory event, e.g., fluoresce with or without a reagent. In one embodiment, the reporter is a protein that is optically detectable without a device, e.g. a laser, to stimulate the reporter, such as horseradish peroxidase (HRP). A protein reporter can be expressed in the cell that is to be detected, and such expression may be indicative of the presence of the protein or it can indicate the presence of another protein that may or may not be coexpressed with the reporter. A reporter may also include any substance on or in a cell that causes a detectable reaction, for example by acting as a starting material, reactant or a catalyst for a reaction which produces a detectable product. Cells may be sorted, for example, based on the presence of the substance, or on the ability of the cell to produce the detectable product when the reporter substance is provided.
  • A “marker” is a characteristic of a molecule, cell or virion that is detectable or is made detectable by a reporter, or which may be coexpressed with a reporter. For molecules. a marker can be particular constituents or moieties, such as restrictions sites or particular nucleic acid sequences in the case of polynucleotides. For cells and virions, characteristics may include a protein, including enzyme, receptor and ligand proteins, saccharrides, polynucleotides, and combinations thereof, or any biological material associated with a cell or virion. The product of an enzymatic reaction may also be used as a marker. The marker may be directly or indirectly associated with the reporter or can itself be a reporter. Thus, a marker is generally a distinguishing feature of a molecule, cell or virion, and a reporter is generally an agent which directly or indirectly identifies or permits measurement of a marker. These terms may, however, be used interchangeably.
  • The invention is further described below, by way of the following examples. The examples also illustrate useful methodology for practicing the invention. These examples do not limit the claimed invention.
  • EXAMPLES Example 1
  • The present invention provides methods for preparing a library of droplet emulsions, where each of the droplets is of the same, predetermined size (monodisperse). Further, present invention provides methods for deterministic lateral displacement for continuous particle separation, which can occur within droplets on a microfluidic device.
  • Particles in solution are usually separated according to size by exclusion or hydrodynamic chromatography. In the former, a sample mixture is injected at one end of a tube packed with porous beads and then washed through the tube. Particles smaller than the pore sizes enter the beads, which lengthen their migration path, and so they are on average eluted later than larger particles. Zones of particles of a given size broaden, however, because particles in each zone take many different paths, leading to different retention times. This multipath effect reduces the resolution of size-exclusion chromatography. In hydrodynamic chromatography, a sample mixture is driven through a capillary by hydrodynamic flow, which has a parabolic flow profile. Large particles cannot intercept the low-velocity fluid near the capillary wall, and thus on average move faster and become separated from small particles. Multipath effects also limit the resolution of hydrodynamic chromatography, because each migration path samples different velocities in the parabolic flow.
  • Recently, Huang et al. Science 304(5673):987-90, 2004 and Davis et al. Proc Natl Acad Sci U S A. 103(40): 14779-84, 2006 demonstrate a separation process that creates equivalent migration paths for each particle in a mixture, thereby eliminating multipath zone broadening. They describe a ‘lateral displacement’ means for separation of particles in solution based on particle size.
  • Lateral displacement means for sizing and separating droplets in solution (based on droplet size) can be utilized. The present invention relates to the generation of a microfluidic device consisting of raised pillars in both columns and rows that are designed for lateral diffusion. The pillars can be adjusted so as to be a means for separating droplets of similar sizes from a fluid containing various sized droplets.
  • In an example, a fluid containing oil, water and a surfactant is mixed so as to create a bulk emulsion. The bulk emulsion is injected into beginning of a microfluidic lateral diffusion device and various fractions are collected at the ending of the device at positions corresponding to specific sizes. Advantages to this lateral diffusion separation means would be the isolation of similarly-sized droplets off-line in a fast and facile manner. Bulk emulsions could be size-selected and then the resulting emulsions, if desired, combined to create sized libraries for re-introduction into a microfluidic device. In a further example, the lateral diffusion microfluidic devices could be rolled-up into a syringe or designed for parallel processing.
  • Recently, devices that exploit both techniques have been miniaturized with the use of microfabrication technology. Microfabricated devices have also been designed that inherently rely on diffusion for separation. Particle mixtures are either repeatedly subject to spatially asymmetric potentials created by microelectrodes or driven through arrays of micrometer-scale asymmetric obstacles to exploit differences in diffusion lengths. In all of the devices discussed so far, particles in a given zone have many different migration paths, and diffusion is required for separation.
  • The present invention describes a separation process that creates equivalent migration paths for each particle in a mixture, thereby eliminating multipath zone broadening (FIG. 24). FIG. 24. (Panel A) Geometric parameters defining the obstacle matrix. A fluid flow is applied in the vertical direction (orange arrow). (Panel B) Three fluid streams (red, yellow, and blue) in a gap do not mix as they flow through the matrix. Lane 1 at the first obstacle row becomes lane 3 at the second row, lane 3 becomes lane 2 at the third row, and so on. Small particles following streamlines will thus stay in the same lane. (Panel C) A particle with a radius that is larger than lane 1 follows a streamline passing through the particle's center (black dot), moving toward lane 1. The particle is physically displaced as it enters the next gap. Black dotted lines mark the lanes.
  • The separation process uses laminar flow through a periodic array of micrometer-scale obstacles. Each row of obstacles is shifted horizontally with respect to the previous row by δλ, where λ is the center-to-center distance between the obstacles (FIG. 24) . For convenience, let δλ/λ be ⅓. Fluid emerging from a gap between two obstacles will encounter an obstacle in the next row and will bifurcate as it moves around the obstacle. Let the flow diverted to the left of the obstacle be δφ, where φ is the total fluid flux going through the gap. If the fluid is confined to move straight down through the array, δ must equal δλ/λ. Let us then consider the flow through a gap to be made up of three lanes, each of which by definition has a flux of φ/3. Because the Reynolds number is low (<10−3 in micrometer-scale environments) and flows are laminar, the streams in each lane do not cross or mix (FIG. 24B). Notably, as the lanes go through gaps, their positions relative to the gaps change. The lanes are represented in each gap by 1, 2, and 3, from left to right, respectively. Lane 1 becomes lane 3 in the next gap, lane 2 becomes lane 1, and lane 3 becomes lane 2 (FIG. 24). After three rows, the three lanes rejoin in their original configuration.
  • Particles that are smaller than the lane width will follow the streamlines. A particle starting in lane 1 will go through lane 3 (right lane with respect to the gap) in the second row, lane 2 (middle lane) in the third row, and back to lane I (left lane) in the fourth row (FIG. 24B). In fact, particles starting from any of the three lanes will go back to the original lane assignment after three rows, so that net migration is in the average flow direction. This motion is called the “zigzag mode.” In practice, particles can diffuse into an adjacent lane. However, the microscopic path for all lanes is equivalent, unlike the multiple paths particles take when moving through a column of porous beads. In contrast to the smaller particles, a particle with a radius larger than the width of lane I at a gap will behave differently in the array. This is because the center of the particle cannot “fit” into lane I in a gap. As such a particle from lane 2 in one gap moves into the subsequent gap, expecting to move through the gap in lane 1, the particle will be “bumped” and its center will thus be displaced into lane 2 (FIG. 24C). The particle will then flow with the fluid in lane 2. This process is repeated every time a large particle approaches a row of obstacles, so that the particle remains in lane 2 as it moves down through the array. This transport pattern is called the “displacement mode.” This is also applicable to electrophoresis by considering ion flows instead of fluid flows.
  • FIG. 25 shows High-resolution separation of fluorescent microspheres with diameters of 0.80 um (green), 0.90 um (red), and 1.03 um (yellow), with a matrix of varying gap size. Whereas the shift in registry and the lattice constants of the matrix remain the same, the obstacle diameters are changed to create gaps, d, of different sizes, which are labeled on the left side of the fluorescent image. The red bars on the fluorescence profile represent the width of the peaks (SD), and the black bars label the 1% inhomogeneity in the bead population. a.u., arbitrary units.
  • FIG. 26 is a schematic illustrating the separation by deterministic lateral displacement in an array of microposts, with an example row shift fraction of one-third. This shift creates three equal flux streamlines. The dashed lines are the boundaries between the streamlines, which are assigned an index in the gaps between the posts. Paths of particles both smaller and larger than the critical threshold are depicted with green and red dotted lines respectively. Small particles stay within a flow stream and large particles are displaced at each obstacle. G is the clear spacing between the gap, is the center-to-center post separation, and d is the relative shift of the post centers in adjacent rows.
  • These described methods allow for the quick and efficient formation of uniformed sized droplet emulsion libraries for further use on a microfluidic device of the present invention.
  • Example 2
  • The present invention provides methods for performing polymerase chain reaction (PCR). PCR can be performed on a drop-by-drop basis in a microfluidic device according to the present invention. A monolithic chip can be provided wherein the heating and cooling lines are built into the chip and a sorting means is provided. Advantages of performing PCR in droplets on such a chip are that the chip is disposable and the reaction can be repeated without cleaning the device between reactions. Furthermore, the chip provides a convenient way of getting all the components to perform PCR in the droplets in the right concentration. Additionally, the PCR is more efficient because the heat transfer is more efficient due to the small volume. This provides for shorter incubation/residence times. Droplets containing the nucleic acids, all PCR primers, and, if present, beads are generated one at a time at rates between 100 and 20,000 droplets per second. The droplets can then be sent through a serpentine path between heating and cooling lines to amplify the genetic material inside the droplets. Upon exiting the device the droplets may be sent for further on-chip or off-chip processing, directed into another chip, or the emulsion may be broken to release the PCR product. If present, beads may be harvested by passing the emulsion through a filtration device, sedimentation, or centrifugation.
  • The width and depth of the channel can be adjusted to set the residence time at each temperature, which can be controlled to anywhere between less than a second and minutes. At a typical rate of 1000 drops per second, 1 million strands of DNA can be amplified in approximately 20 minutes on one device. A typical flow rate of 250 μL/hour would correspond to 1000 drops of 50 microns in diameter being generated every second. Flow rates and droplet sizes can be adjusted as needed by controlling the nozzle geometry.
  • The present invention also provides methods for performing dideoxynucleotide sequencing reactions on a microfluidic device. Chain terminator sequencing (Sanger sequencing) is well known to those of ordinary skill in the art. DNA template preparation, cycling sequencing and preparing extension products for electrophoresis are related techniques and also well known to those of skill in the art. Applied Biosystems' “Automated DNA Sequencing: Chemistry Guide” 2000 is an excellent resource covering these techniques and is incorporated herein by reference in its entirety.
  • One method is to sequencing PCR templates which can include single amplification PCR or nested and semi-nested PCR strategies. In the simplest PCR sequencing case, the target DNA is amplified with a single set of primers and then sequenced using the same primers. For many samples, this works well. For the samples that do not work well with this method, optimization of the PCR amplification may be required. Optimizing the PCR minimizes the presence of non-specific product bands and ensures adequate yield. A single PCR amplification is also compatible with the use of a sequencing primer that binds internally (semi-nested or nested) to one or both of the PCR primers. This can be helpful if primer-dimer (primer oligomerization) artifacts are a problem.
  • If difficulty with more complex samples, such as bacterial genomic DNA, is encountered a nested or semi-nested PCR can be used. These techniques are useful when the target is present in small quantity. They offer more specificity, which provides superior sequencing data with reduced background signal. Both nested and semi-nested PCR require two amplifications. The first amplification is identical for nested and semi-nested, but the second amplification differs as described below. Amplify with one set of PCR primers, which converts a complex sample (such as bacterial genomic DNA) into a non-complex sample consisting of the first PCR product and some side products. Nested PCR: Amplify 1% or less of the first PCR reaction product using a second set of PCR primers that hybridize at positions internal to the first set. Semi-nested PCR: Only one primer of the second set of PCR primers is internal. The other primer is one of the original PCR primers.
  • A PCR primer can be synthesized with a universal sequencing primer binding site added to the 5′ end (e.g., see Appendix E in Applied Biosystems' “Automated DNA Sequencing: Chemistry Guide” for universal primer sequences). This allows any PCR product to be sequenced with universal primers. Universal-tailed PCR primers enable the use of commercially available dye-labeled sequencing primers. This technique is also useful with dye terminator chemistries, because universal sequencing primers have good annealing characteristics. However, the longer PCR primers add to the overall cost of the reactions. Using universal-tailed primers sometimes results in primer oligomerization. As these products have priming sites present, they can result in noisy data for the first 20-100 bases. Redesigning the PCR primer, optimizing the PCR amplification further, and employing Hot Start methods can help overcome this situation.
  • After PCR amplification, the resulting PCR product is in solution along with PCR primers, dNTPs, enzyme, and buffer components. The method used to prepare the PCR product for sequencing depends on the amounts of these components that are carried over and on the chemistry used for sequencing. Excess PCR primers carried over from the amplification reaction compete with the sequencing primer for binding sites and reagents in the sequencing reaction. This carryover of PCR primers presents more of a problem in dye terminator chemistries because the dye label is incorporated into the extension product after the primer anneals to the template. If more than one primer is present, multiple dye-labeled sequence ladders are generated, resulting in noisy data. Excess dNTPs from the amplification reaction can affect the balance of the sequencing reaction, resulting in decreased termination in shorter extension fragments.
  • Nonspecific PCR products include primer-dimer artifacts and secondary PCR products. The presence of any significant quantity of either in a PCR product can result in poor quality sequencing data. Nonspecific PCR products behave as templates in the sequencing reaction and produce extension products, which results in noisy data. These products often can be visualized on an agarose gel before sequencing. If they are present, the PCR amplification should be optimized and repeated before sequencing. Use of a nested or semi-nested sequencing primer can also allow good sequence data to be obtained. Alternatively, the PCR product of interest can be purified by agarose gel electrophoresis.
  • There are several ways to minimize contaminants in a PCR amplification: PCR optimization (Innis and Gelfand, 1990): (1) Amount of starting DNA; (2) Careful primer design; (3) Primer concentration, (4) Enzyme concentration, (5) Magnesium ion (Mg2+) concentration, (6) Nucleotide concentration; (7) Buffer composition; (8) Number of cycles; (9) pH; (10) Manual Hot Start method; (11) AmpliTaq Gold® DNA Polymerase as an automatic Hot Start and/or (12) Limiting dNTPs and primers. All of these methods increase the specificity of the PCR amplification and decrease the amount of contaminants that can interfere with a sequencing reaction.
  • There are several methods for purifying PCR products: (1) Column purification; (2) Ethanol precipitation; and/or (3) Gel purification.
  • An alternative to one of the more stringent purification methods listed above is treatment of PCR products with shrimp alkaline phosphatase (SAP) and exonuclease I (Exo I) before sequencing.
  • The SAP/Exo I procedure degrades nucleotides and single-stranded DNA (primers) remaining after PCR (Werle et al., 1994). This procedure is particularly useful in cases where limiting concentrations of primers and nucleotides cannot be used for direct PCR sequencing.
  • FIG. 27 shows one embodiment for a DNA sequencing chip design. Template DNA and primers are combined at step ‘add 1’ and the reaction is incubated at 95° C. for a hot start (position 1). The reaction then cycles 20-30 times (position 2) before the addition of SAP and ExoI at ‘add 2.’ The reaction is incubated at 37° C. for a predefined time-period and then the SAP and ExoI enzymes are inactivated at 95° C. (position ‘4’). The SAP/ExoI procedure degrades nucleotides and single-stranded DNA (primers) remaining after PCR. The universal sequencing primers, ddNTPs and buffers are added at ‘add 3,’ and the PCR sequencing reaction is allowed to proceed at position ‘5.’ The final reaction product is collected and can be stored off-chip.
      • Step Action
      • 1. For each sample, combine the following:
        • SAP(1 Unit/μL), 2 μL
        • Exo I (10 Units/μL), 0.2 μL
        • Deionized water, 6.0 μL
        • Note In general this procedure works well using 0.5 units of each enzyme per microliter of PCR products used. The procedure seems to work equally well with or without the use of SAP buffer, so this has been excluded in this protocol.
      • 2. Add 4.0 μL of PCR product to the above mix.
      • 3. Incubate at 37° C. for 1 hour.
      • 4. Incubate at 72° C. for 15 minutes to inactivate the enzymes.
  • The recommended DNA quantities for sequencing reactions are shown in Table 3-1 below.
    TABLE 3-1
    Recommended Ranges of DNA Template Quantity for Each Chemistry
    Cycle Sequencing Chemistry
    Fluorescein/
    Rhodamine dRhodamine BigDye Rhodamine
    Template Dye Terminator Terminator Terminator Dye Primer BigDye Primer
    PCR product:
    100-200 bp 1-3 ng 1-3 ng 1-3 ng 2-5 ng 2-5 ng
    200-500 bp 3-10 ng 3-10 ng 3-10 ng 5-10 ng 5-10 ng
    500-1000 bp 5-20 ng 5-20 ng 5-20 ng 10-20 ng 10-20 ng
    1000-2000 bp 10-40 ng 10-40 ng 10-40 ng 20-50 ng 20-50 ng
    >2000 bp 40-100 ng 40-100 ng 40-100 ng 50-150 ng 50-150 ng
    single-stranded 100-250 ng 50-100 ng 50-100 ng 150-300 ng 150-400 ng
    double-stranded 200-600 ng 200-500 ng 200-500 ng 300-600 ng 200-800 ng
    cosmid, BAC 0.5-2.0 μg not 0.5-1.0 μg 0.5-2.0 μg 0.5-1.0 μg
    recommended
    bacterial genomic DNA not recommended 2-3 μg not recommended
  • PCR protocols that limit amounts of primers and dNTPs allow the product of the reaction to be used for sequencing with no purification. This is usually carried out by setting up the PCR amplification with 5-10 pmol of primers and 20-40 μM dNTPs, so that most of the primers and dNTPs are exhausted during amplification. If the yield of the desired PCR product is high and the product is specific, i.e., it produces a single band when analyzed by agarose gel electrophoresis, the sample can be diluted before sequencing and will give good results. The dilution ratio depends on the concentration of your PCR product and needs to be determined empirically (start with 1:2 and 1:10 dilutions with deionized water). When you limit concentrations of primers and dNTPs and dilute the PCR products, the PCR parameters have to be robust. Direct PCR sequencing is most useful in applications where the same target is being amplified and sequenced repeatedly and PCR conditions have been optimized. Direct PCR sequencing can be done with dye primer chemistries. With dye terminator chemistries, it is much more critical that the PCR primers be consumed. Excess PCR primers will be extended and labeled by the cycle sequencing reaction, resulting in noisy data. Direct PCR sequencing does not work for XL PCR because limiting amounts of primers and dNTPs cannot be used. The PCR product should be purified or the excess primers and nucleotides should be degraded by SAP/Exo I treatment.
  • Example 3
  • The present invention provides methods for performing isothermal-type amplification methods on a microfluidic device. Isothermal amplification is an alternative to the standard PCR techniques described herein. Isothermal amplification is used to reduce the relative amount of background DNA in a sample. Primers are generally used in a constant temperature means of amplification. Isothermal amplification is applicable for SNP detection. Once the DNA is amplified by isothermal amplification there are several well-known means for detecting which nucleotide polymorphism is present. These include, but are not limited to; allele specific primer extension, oligonucleotide ligation assay, mini-sequencing, fluorescence polarization, etc. Isothermal amplification is also applicable for DNA sequencing preparation. The isothermally-amplified DNA can be attached to a solid phase within a droplet or placed within a parking space on chip. The beads or parking spaces can be accessed and the amplified DNA used for a DNA sequencing reaction. Further, isothermal amplification is applicable for gene expression analysis. Isothermal amplification can be used to monitor gene expression by the measurement of the amount of cDNA produced in a quantitative fashion. Many methods for isothermal amplification are known in the art, including but not limited to the following examples.
  • Rolling circle amplification (RCA). A DNA polymerase extends a primer on a circular template, generating tandemly linked copies of the complementary sequence of the template (Fire & Xu, 1995). The TempliPhi amplification process using rolling circle amplification is known in the art. In the process, random hexamer primers anneal to the circular template DNA at multiple sites. Phi29 DNA polymerase extends each of these primers. When the DNA polymerase reaches a downstream-extended primer, strand displacement synthesis occurs. The displaced strand is rendered single-stranded and available to be primed by more hexamer primer. The process continues, resulting in exponential, isothermal amplification.
  • Transcription mediated amplification (TMA). An RNA polymerase is used to make RNA from a promoter engineered in the primer region, a reverse transcriptase to produce complementary DNA from the RNA templates and RNase H to remove the RNA from cDNA (Guatelli et al, 1990).
  • Strand-displacement amplification (SDA). A restriction endonuclease is used to nick the unmodified strand of its target DNA and the action of an exonuclease-deficient DNA polymerase to extend the 30 end at the nick and displace the downstream DNA strand (Walker et al, 1992). Strand-displacement amplification is known in the art.
  • Helicase-dependent amplification (HDA). A DNA helicase is used to generate single-stranded templates for primer hybridization and subsequent primer extension by a DNA polymerase. Schematic diagram of HAD is shown in FIG. 28. Two complementary DNA strands are shown as two lines: the thick one is the top strand and the thin one is the bottom strand. 1: A helicase (black triangle) separates the two complementary DNA strands, which are bound by SSB (grey circles). 2: Primers (lines with arrow heads) hybridize to the target region on the ssDNA template. 3: A DNA polymerase (squares with mosaic patterns) extends the primers hybridized on the template DNA. 4: Amplified products enter the next round of amplification.
  • One example is emulsion-based sample preparation. Random libraries of DNA fragments are generated by shearing an entire genome and isolating single DNA molecules by limiting dilution. See, FIG. 29. Specifically, sequencing reactions such as those performed by Solexa, 454 Life Sciences and others involve randomly fragmenting the entire genome, adding specialized common adapters to the fragments, capturing the individual fragments on their own beads and, within the droplets of an emulsion, clonally amplifing the individual fragment (FIG. 29 a, 29 b). Unlike in current sequencing technology, their approach does not require subcloning or the handling of individual clones; the templates are handled in bulk within the emulsions. Typically, about 30% of the beads will have DNA, producing 450,000 template-carrying beads per emulsion reaction.
  • Sample preparation and DNA sequencing is shown in FIG. 29. Panel A, Genomic DNA is isolated, fragmented, ligated to adapters and separated into single strands (top left). Fragments are bound to beads under conditions that favor one fragment per bead, the beads are captured in the droplets of a PCR-reaction-mixture-in-oil emulsion and PCR amplification occurs within each droplet, resulting in beads each carrying ten million copies of a unique DNA template (top, second from the left). The emulsion is broken, the DNA strands are denatured, and beads carrying single-stranded DNA clones are deposited into wells of a fiber-optic slide (bottom left). Smaller beads carrying immobilized enzymes required for pyrophosphate sequencing are deposited into each well (bottom, second from the left). Panel B, Microscope photograph of emulsion showing droplets containing a bead and empty droplets. The thin arrow points to a 28-mm bead; the thick arrow points to an approximately 100-mm droplet. Panel C, Scanning electron micrograph of a portion of a fiber-optic slide, showing fiber-optic cladding and wells before bead deposition. Panel D, The sequencing instrument consists of the following major subsystems: a fluidic assembly. Panel E, a flow chamber that includes the well-containing fiber-optic slide. Panel F, a CCD camera-based imaging assembly. Panel G, and a computer that provides the necessary user interface and instrument control.
  • Another example is sequencing in fabricated picolitre-sized reaction vessels. One method uses sequencing by synthesis simultaneously in open wells of a fiber-optic slide using a modified pyrosequencing protocol that is designed to take advantage of the small scale of the wells. The fiber optic slides are manufactured by slicing of a fiber-optic block that is obtained by repeated drawing and fusing of optic fibers. At each iteration, the diameters of the individual fibers decrease as they are hexagonally packed into bundles of increasing cross-sectional sizes. Each fiber-optic core is 44 μm in diameter and surrounded by 2-3 μm of cladding; etching of each core creates reaction wells approximately 55 μm in depth with a centre-to-centre distance of 50 μm (FIG. 29 c), resulting in a calculated well size of 75 pl and a well density of 480 wells per square mm. The slide, containing approximately 1.6 million wells, is loaded with beads and mounted in a flow chamber designed to create a 300-μm high channel, above the well openings, through which the sequencing reagents flow (FIG. 29 d). The unetched base of the slide is in optical contact with a second fiber optic imaging bundle bonded to a charge-coupled device (CCD) sensor, allowing the capture of emitted photons from the bottom of each individual well (FIG. 29 d). A three-bead system has been developed and the components optimized to achieve high efficiency on solid support. The combination of picoliter-sized wells, enzyme loading uniformity allowed by the small beads and enhanced solid support chemistry enabled users to develop a method that extends the useful read length of sequencing-by-synthesis to 100 bases.
  • In the flow chamber cyclically delivered reagents flow perpendicularly to the wells. This configuration allows simultaneous extension reactions on template-carrying beads within the open wells and relies on convective and diffusive transport to control the addition or removal of reagents and by-products. The timescale for diffusion into and out of the wells is on the order of 10 s in the current configuration and is dependent on well depth and flow channel height. The timescales for the signal-generating enzymatic reactions are on the order of 0.02-1.5 s. The current reaction is dominated by mass transport effects, and improvements based on faster delivery of reagents are possible. Well depth was selected on the basis of a number of competing requirements: (1) wells need to be deep enough for the DNA-carrying beads to remain in the wells in the presence of convective transport past the wells; (2) they must be sufficiently deep to provide adequate isolation against diffusion of by-products from a well in which incorporation is taking place to a well where no incorporation is occurring; and (3) they must be shallow enough to allow rapid diffusion of nucleotides into the wells and rapid washing out of remaining nucleotides at the end of each flow cycle to enable high sequencing throughput and reduced reagent use. After the flow of each nucleotide, a wash containing a pyrase is used to ensure that nucleotides do not remain in any well before the next nucleotide being introduced.
  • Another example is base calling of individual reads. Nucleotide incorporation is detected by the associated release of inorganic pyrophosphate and the generation of photons. Wells containing template-carrying beads are identified by detecting a known four-nucleotide ‘key’ sequence at the beginning of the read. Raw signals are background-subtracted, normalized and corrected. The normalized signal intensity at each nucleotide flow, for a particular well, indicates the number of nucleotides, if any, that were incorporated. This linearity in signal is preserved to at least homopolymers of length eight. In sequencing by synthesis a very small number of templates on each bead lose synchronism (that is, either get ahead of, or fall behind, all other templates in sequence). The effect is primarily due to leftover nucleotides in a well (creating ‘carry forward’) or to incomplete extension. Typically, a carry forward rate of 1-2% and an incomplete extension rate of 0.1-0.3% is seen. Correction of these shifts is essential because the loss of synchronism is a cumulative effect that degrades the quality of sequencing at longer read lengths.
  • Methods have demonstrated the simultaneous acquisition of hundreds of thousands of sequence reads, 80-120 bases long, at 96% average accuracy in a single run of the instrument using a newly developed in vitro sample preparation methodology and sequencing technology. With Phred 20 as a cutoff, they are able to show that their instrument is able to produce over 47 million bases from test fragments and 25 million bases from genomic libraries. Recent work on the sequencing chemistry and algorithms that correct for crosstalk between wells suggests that the signal distributions will narrow, with an attendant reduction in errors and increase in read lengths. In preliminary experiments with genomic libraries that also include improvements in the emulsion protocol, one is able to achieve, using 84 cycles, read lengths of 200 bases with accuracies similar to those demonstrated here for 100 bases. On occasion, at 168 cycles, individual reads that are 100% accurate over greater then 400 bases have been generated.
  • Isothermal amplification reactions, as described above, have shown great promise generating high yields with great fidelity. However, an associated drawback arises from the tendency of the polymerase to generate spurious, non-template amplification products when reactions are conducted in the absence of template DNA. Additionally, our application utilizes high microfluidic throughput in conjunction with limiting DNA template dilutions to amplify single template molecules. As a result, the number of empty reaction droplets increases considerably, comprising 90% or more of the total droplet population following Poisson distributions. Non-template amplification (hereafter NTA) in even a small fraction of the total droplets can confound amplification detection strategies based on laser interrogation of intercalating dyes, thus this issue must be resolved. To address this problem in the art, the present invention provides the use of mixed modified and standard hexamer primers in microfluidic reactions to retard NTA while allowing template-based amplification to proceed.
  • Previous work has attempted to reduce NTA through incorporation of nitroindole bases (Loakes and Brown 1994; Loakes, Hill et al. 1997) in the random primers (Lage, Leamon et al. 2003) or reducing reaction volumes to 600 nL (Hutchison, Smith et al. 2005). Unfortunately, modified nitroindole primers have proven difficult to replicate, and often have the effect of significantly reducing the overall rate and yield of the amplifications in which they are incorporated. Low volume reactions conducted in multiplate wells have encountered difficulties stemming from dispensation of low volumes, and associated issues of sample evaporation, well to well contamination, etc.
  • The modified primers of the present invention containing nitroindoles and C3 non-replicable elements were studied in an effort to reduce NTA both in bulk and microfluidic reactions. Both nitroindoles and C3 non-replicable elements were found to be effective in reducing NTA, with primers containing two 5′ nitroindoles most effective in NTA suppression. However, increased NTA suppression was tightly linked with reduced yield in template amplification reactions. Amplifications using a ratio of nitroindole to random hexamer primers generated a range of both template and non-template amplification yields, with a 15:85 ratio of nitroindole to random hexamers generating template yields commensurate with random hexamers primers alone, but generating little if any spurious product in the absence of template.
  • Example 4
  • The PCR and isothermal amplifications described herein can be very useful in performing single nucleotide polymorphism analysis. A Single Nucleotide Polymorphism, or SNP, is a small genetic change, or variation, that can occur within a person's DNA sequence. The genetic code is specified by the four nucleotide “letters” A (adenine), C (cytosine), T (thymine), and G (guanine). SNP variation occurs when a single nucleotide, such as an A, replaces one of the other three nucleotide letters—C, G, or T.
  • An example of a SNP is the alteration of the DNA segment AAGGTTA to ATGGTTA, where the second “A” in the first snippet is replaced with a “T”. On average, SNPs occur in the human population more than 0.1 percent of the time. Because only about 3 to 5 percent of a person's DNA sequence codes for the production of proteins, most SNPs are found outside of “coding sequences”. SNPs found within a coding sequence are of particular interest to researchers because they are more likely to alter the biological function of a protein. Because of the recent advances in technology, coupled with the unique ability of these genetic variations to facilitate gene identification, there has been a recent flurry of SNP discovery and detection.
  • Although many SNPs do not produce physical changes in people, scientists believe that other SNPs may predispose people to disease and even influence their response to drug regimens. Currently, there is no simple way to determine how a patient will respond to a particular medication. A treatment proven effective in one patient may be ineffective in others. Worse yet, some patients may experience an adverse immunologic reaction to a particular drug. Today, pharmaceutical companies are limited to developing agents to which the “average” patient will respond. As a result, many drugs that might benefit a small number of patients never make it to market.
  • In the future, the most appropriate drug for an individual could be determined in advance of treatment by analyzing a patient's SNP profile. The ability to target a drug to those individuals most likely to benefit, referred to as “personalized medicine”, would allow pharmaceutical companies to bring many more drugs to market and allow doctors to prescribe individualized therapies specific to a patient's needs.
  • Finding single nucleotide changes in the human genome seems like a daunting prospect, but over the last 20 years, biomedical researchers have developed a number of techniques that make it possible to do just that. Each technique uses a different method to compare selected regions of a DNA sequence obtained from multiple individuals who share a common trait. In each test, the result shows a physical difference in the DNA samples only when a SNP is detected in one individual and not in the other. Currently, existing SNPs are most easily studied using microarrays. Microarrays allow the simultaneous testing of up to hundreds of thousands of separate SNPs and are quickly screened by computer.
  • The race among pharmaceutical companies today is to apply new system genomics approach to identify novel targets and validate these targets in the most efficient fashion. SNP research will provide fundamental understanding of many polygenic diseases, thus providing new therapeutic targets. As groups have performed genome-wide scans and other large studies that require the genotyping of thousands of SNPs and samples, a need for high-throughput SNP genotyping has become essential.
  • The microfluidic device of the present invention is capable of performing at least 10,000 SNP analysis per second, such that a full genome scan (i.e., 100K SNPs) with 10× overrepresentation can be performed in less than an hour. The speed and efficiency permitted using the devices and methods of the present invention will significantly lower the associated costs and reagent consumption for performing SNP analysis.
  • Example 5
  • The PCR and isothermal amplifications described herein can be very useful in providing necessary sample preparation processes for commercial available DNA sequencers, such as the Solexa's 1G sequencer, which relies upon immobilized DNA for a substrate. In one embodiment, single molecules can be amplified using PCR primers treated with moieties used for surface immobliziation, then flow the PCR positive droplets across the surface of the slide, forming a packed emulsion. The emulsion can then be broken, and the primers allowed to bind to the slide due the presence of the appropriate coating to bind the PCR products.
  • The Solexa 1G sequencer currently sequences amplified material that has been amplified in place, on primers bound to the slide, by bridge amplification. In this process, template DNA is flowed across the slide surface at very low concentrations, and adapters previously ligated to each template hybridize to complimentary primers attached to the slide in any one of eight lanes. Once hybridized, the primers are subjected to bridge amplification, a version of PCR amplification that utilizes immobilized primers—the product of the reaction is a patch of DNA immobilized to the slide. Several risks are encountered when amplifying DNA in this matter—the process is exquisitely sensitive to DNA concentration—if too much DNA is used, the DNA patches will be generated too closely, or even overlap, generating mixed signals during subsequent sequencing. If too little DNA is used, the DNA patches will be present at a very low density, and insufficient sequence may be generated during the run. As the sequencing reactions take 72 hours, and no titration runs are conducted to test the DNA concentrations prior to amplification, the potential loss of time and money is considerable. Additionally, neither single molecule amplification nor bridge amplification are very efficient, and bridge amplification has an upper and lower size limitation, generating products only within a particular length range.
  • The present invention provides methods to overcome this limitations. In one embodiment, a Solexa slide can be coated with any of the compounds commonly used to permit binding and immobilization (e.g., carboxy-esters, streptavidin, Igg, gold, etc). PCR reactions could be performed as described in the instant microfluidic device using primers modified with the appropriate binding moiety (5′ amines, 5′ biotins, 5′ DNPs or 5′ Thiols, respectively) to efficiently amplify PCR products in solution which could then be efficiently and easily bound to the Solexa slide for subsequent sequencing. The amplification is quite straight forward, conducted with a limiting dilution of template and a set of primer pairs compatible with the adapters ligated to the Solexa templates through their standard sample preparation process. One of the primer pairs would possess a 5′ binding moiety as described earlier, only one of the pair as this will permit removal of the opposing strand and the generation of single stranded immobilized templates on the slide. Once amplification has been conducted, and the positive droplets sorted, the droplets can be flowed onto each of the lanes of the Solexa slide. Proper spacing between the droplets can be obtained by mixing droplets containing only buffer or immiscible oil in with the PCR positive droplets at a ratio sufficient to ensure that the PCR positive droplets are rarely proximal to each other. Once each lane has been packed with droplets, the droplets can be broken through the application of electrical field and the PCR products allowed to bind to the slide in the same geographic area that the droplet had occupied. The immobilized templates can be rendered single stranded through the application of basic washes, temperature etc. This will permit the rapid amplification of PCR fragments and their subsequent density-controlled deposition onto the Solexa chip for sequencing.
  • Example 6
  • The present invention provides methods for detecting the presence and/or sequence of nucleic acids in low copy number in droplets on a microfluidic device. The detection of a specific nucleic acid sequence present in a sample by probing the sample with a complementary sequence of nucleic acids is a well known technique. Nucleic acids are highly specific in binding to complementary nucleic acids and are thus useful to determine whether a specific nucleic acid is present in a sample. One must know the sequence of the specific nucleic acid to be detected and then construct a probe having a complementary nucleic acid sequence to the specific nucleic acid sequence.
  • Since nucleic acid probes are highly specific, it is preferable in some situations to probe the nucleic acid sequence itself rather than the protein produced by the nucleic acid sequence. As a particular example, a diagnostic method based solely on protein detection would be unreliable for determining the presence of infectious particles of hepatitis B virus, due to the presence of significant levels of non-infectious antigen particles which lack the DNA genome. In another example, the various subtypes of human papilloma virus found in either pre-cancerous or benign cervical tumors can be distinguished only by the use of nucleic acid probe hybridization. Also, the specific genetic makeup of an AIDS virus makes it certain that an assay based on the presence of an AIDS virus specific nucleic acid sequence would be superior as a diagnostic.
  • The naturally-occurring high number of ribosomal RNA, up to 100,000 copies per cell, has been used by GenProbe to facilitate diagnosis of certain bacterial pathogens, such as Legionella and Mycoplasma, using nucleic acid probes. However, this strategy cannot be used with non-cellular pathogens, such as viruses, or with probed nucleic acid sequences with low copy numbers. Copy number is a particular problem with the development of a nucleic acid probe method for the detection of AIDS virus, where the integrated provirus may be present in less than one of ten thousand peripheral blood lymphocytes. Thus, if the particular nucleic acid sequence suspected to be present in a sample could be amplified, the copy number problem could be circumvented and probe assays could be more readily used.
  • In a normal biological sample, containing only a few cells, and consequently only a few copies of a particular gene, it is necessary to utilize an amplification process in order to overcome the copy number problem.
  • One method to amplify is to ‘grow out’ the sample, that is, to arrange conditions so that the living biological material present in the sample can replicate itself. Replication could increase the quantity of nucleic acid sequences to detectable levels. In the food industry, for example, in order to test processed food for the food-poisoning bacteria Salmonella, food samples must be incubated for a number of days to increase the quantity of nucleic acid copy numbers. In clinical samples, pathogens must also be allowed to increase their number by growing out over some considerable time.
  • Current methods utilize a process in which a sample suspected of containing a target DNA sequence is treated with oligonucleotide primers such that a primer extension product is synthesized which in turn serves as a template, resulting in amplification of the target a DNA sequence. The primer extension product is separated from the template using heat denaturation. Current methods also include a process for amplifying a target DNA sequence having two separate complementary strands. The process includes treating the strands with primers to synthesize extension products, separating the primer extension products from the templates, and in turn using the primer extension products as templates.
  • Both of the above methods require either manual or mechanical participation and multi-step operations by the user in the amplification process and are restricted to amplifying DNA only. The steps involved in these methods require the user to heat the sample, cool the sample, add appropriate enzymes and then repeat the steps. The temperature changes cause the enzymes to loose their activity. Hence, the user is required to repeatedly supplement the amplification mixture with aliquots of appropriate enzymes during the amplification process.
  • The present invention provides methods for detecting the presence and/or sequence of nucleic acids in low copy number in droplets on a microfluidic device. In one embodiment of the invention, a nucleotide/peptide nucleic acid (pna) probe (oligo probe) can be tethered such that when it binds to a template DNA molecule in low copy number located inside an aqueous emulsion (i.e., droplet) it turns on or activates an enzyme. By way of nonlimiting example, an alkaline phosphatase conjugate can be placed on one and of the low copy number nucleotide, and an inhibitor to alkaline phosphatase on the other end (like a molecular beacon). When the oligo probe binds to the low copy number template the inhibitor is removed from the enzyme and the enzyme turns over the substrate. The tethers can be a protein complementation assay wherein the binding of the oligo probe to the low copy number template causes the enzyme to be active.
  • Various other embodiments are described herein and should not be considered as limiting to the invention. Example 1: Taqman chews a beta-gal alpha protein attached at the 3′ end of an oligo probe thereby releasing free alpha subunit to bind to omega fragment in solution. Example 2: Two oligo probes sit down on the low copy number template adjacent to each other, thereby bringing two subunits of a protein complementing assay reagent together. Example 3: A Taqman-like enzyme that releases an active moiety can also be used. The active moiety can include, for example, an enzyme that becomes activated upon release from the oligo probe, or a fluorescent group that is quenched while tethered to the oligo probe. The use of a double-strand specific nuclease that will chew up the probe only when the probe is bound, thereby releasing active enzyme or fluorescent substrates. Example 4: The probe has a fluorescent group attached such that the detected hybridization causes the release of the fluorescent group a la Taqman. Example 5: The probe has an inactive enzyme attached such that the detected hybridization causes the release of the active enzyme by a Taqman-type release. Example 6: The probe has an inactive complementing enzyme attached such that the detected hybridization causes the release of the active moiety of the enzyme to be able to complement. Example 7: Two probes have inactive enzyme moieties attached such that the detected hybridization causes the complementation and activation of the enzyme. Example 8: Two probes come together and allow a Fluorescence Resonance Energy Transfer (FRET) reaction to occur. This would require a FRET-oligo library. Almost all SNP or transcriptional profiling method may be amenable to this concept.
  • Many assays are conducted on microfluidic devices are including, but not limited to, protein-protein, antibody-antigen, nucleic acid-protein, nucleic acid-nucleic acid, ligand-protein, ligand-nucleic acid, ligand-ligand, eukaryotic or prokaryotic cell surface moiety-second moiety, the measurement of two or more receptors on the surface of an eukaryotic or prokaryotic cell, the development of three-hybrid type systems using tandem fusions, interactor-cofactor, etc. Many other types of interactions are also known that can be adapted to the system. Assays incorporating complementation assays can be used in both proteomics and genomics.
  • Currently, these assay methods require methods require >10K fluorescein molecules to detect interaction/binding. In an embodiment, the present invention provides methods which reduce the number of interacting proteins by amplifying the signal of molecules that do interact. In preferred embodiments of some assays, stable emulsions may not be needed. An advantage is that we can eliminate surfactants and stabilizing additives which can affect protein activity within droplets.
  • Likewise, one could conceivably look for loss of complementation in any of the above assays. This loss or gain of complementation can be a function of physical (e.g., heat, light) or chemical additions to, or formulation of the droplets. These interactions include, but are not limited to, protein-protein, antibody-antigen, nucleic acid-protein, nucleic acid-nucleic acid, ligand-protein, ligand-nucleic acid, ligand-ligand, eukaryotic or prokaryotic cell surface moiety-second moiety, the measurement of two or more receptors on the surface of an eukaryotic or prokaryotic cell, the development of three-hybrid type systems using tandem fusions, interactor-cofactor, etc. Many other types of interactions are also known that can be adapted to the system. Assays incorporating complementation assays can be used in both proteomics and genomics.
  • The amount of each interacting partner may be with-in drop quantifiable by genetically or chemically coupling a reporter molecule (e.g., a dye or quantum dot, GFP protein) to one and or the other. Similarly, the complementation assay described for enzymatic amplification can use one of several different complementing proteins such that the concentration of each partner can be calculated within the droplet by using different enzyme substrates added to the droplets at the same or differing time. Timing of substrate addition is not critical and one of skill in the art would readily recognize that addition can be done at different times. ‘Killing’ by various denaturants, protease, etc is also within the purview of the skilled artisan.
  • In an embodiment, complementation assays can be used to add a specific addition to, for example, an IVT synthesized protein. As an example, the s-peptide of RNaseA and the S protein wherein the S peptide is genetically fused to the IVT-generated Ab and the S protein, upon binding to the S peptide activates the RNaseA activity and thereby stops further IVT synthesis. Similarly, any two complementing interactrs can be used to generate an activity.
  • By quantifying the amount of interactors in a droplet it may be possible to derive interaction kinetics (affinity and disassociation constants as examples). In an embodiment, the present invention provides methods which can allow the quantification of proteins below that which can be seen by fluorescent spectroscopy in the absence of amplification. Alternatively, amplification may also not be needed if one can genetically add a quantified number of reporters to the end of a molecule. For example, genetically fusing 10 GFP proteins onto the end of a protein would thereby increase the fluorescence intensity 10-fold. Similarly, a series of small complementing moieties can be fused onto the end of a protein and there obviate the need for long genetic fusions. As an example, a series of 10 s peptides spaced apart by a linker would be able to each ‘grab hold’ of an S protein to generate an increased signal over that which can be achieved by a single enzyme. Other examples include either biotin or biotin-binding protein mimetic and streptavidin or avidin, FLAG tags, poly histidines, complementing GFP, etc. Another example includes Qcoding the droplets.
  • FIGS. 30 and 31 show the current method for isolating antibodies on a microfluidic device. In the current method, DNA beads are made using bulk emulsion PCR; DNA-containing beads preferably isolated before loading onto a microfluidic device. The antigen can be <20 kD, (preferably <10 kD) and labeled with an appropriate dye, (preferably with several dye-molecules). It may even be preferable to put a poly-lysine tail onto the antigen to increase signal (but be concerned about quenching). The antigen is formulated into the bead-containing droplets at a concentration of 100 nm to allow proper sensitivity in the droplet (use of multiple/different dyes may allow this concentration to drop). In some situations it is possible to add antigen at the same time as the IVT solution.
  • FIG. 32 shows the method of the present invention for isolating antibodies on the microfluidic device. The right panel is a diagram of individual steps proposed to amplify signal of interacting antibody and antigen. The left panel is a schematic as would be designed for a chip to be used on microfluidic device.
  • FIG. 33 shows the genetic selection for full length antibody clones. A genetic selection can be used to enrich for full-length antibody clones by transforming E. coli and selecting for clones able to grow on medium in which a suitable sugar is the only carbon source.
  • Example 7
  • The present invention provides a microfluidic device topology and implementation that merges the functionality of a first microfluidic substrate with that of a second microfluidic substrate by using forced withdrawal and reinjection into the same fluidic port. In current multi-step assays, the following steps must be performed to complete a “two step” experiment: (1) Installation of the first microfluidic substrate; (2) Installation of all first stage reagents; (3) Priming of all fluid lines and stabilization of the device operation; (4) Passive connection of a storage container to the instrument after device has been stabilized; (5) Disconnection of the storage container from the first device when collection complete; (6) Incubation of the collected emulsion; (7) Removal of the first device from the instrument; (8) Cleaning of the fluid lines needed for the second step of the experiment; (9) Installation of a second device to the instrument; (10) Connection of all second stage reagents, including the emulsion collected during the first stage; (11) Priming of the fluidic connections to the second device prior to running the second half of the measurement; and (12) Collection/readout of the second stage.
  • The methods of the present invention replaces current methodology of multi-step assays with the following: (1) Installation of the first microfluidic substrate; (2) Installation of all first and second stage reagents, including the first stage storage container (if it is not already connected); (3) Priming and stabilization of the fluidic lines and device; (4) Controlled collection of the first stage combined droplets by actively withdrawing some fraction of the oil and all of the generated emulsion into the storage container; (5) Incubation of the collected emulsion; (6) Startup and reinjection of the collected droplets back into the second half of the device; and (8) Collection/readout of the second stage. FIG. 34 depicts a schematic representation of this device topology.
  • Elimination of the handling of the collected emulsion has significant benefits beyond simplifying the user interaction with the instrument. Contaminants have the potential to ruin the experiment, and any extra handling and connection/disconnection increase the probability that contaminants will be introduced into the instrument.
  • Example 8
  • Gene silencing through RNAi (RNA-interference) by use of short interfering RNA (siRNA) has emerged as a powerful tool for molecular biology and holds the potential to be used for therapeutic gene silencing. Short hairpin RNA (shRNA) transcribed from small DNA plasmids within the target cell has also been shown to mediate stable gene silencing and achieve gene knockdown at levels comparable to those obtained by transfection with chemically synthesized siRNA (T. R. Brummelkamp, R. Bemards, R. Agami, Science 296, 550 (2002), P. J. Paddison, A. A. Caudiy, G. J. Hannon, PNAS 99, 1443 (2002)). Possible applications of RNAi for therapeutic purposes are extensive and include silencing and knockdown of disease genes such as oncogenes or viral genes.
  • Many assays are conducted on microfluidic devices are including, but not limited to, protein-protein, antibody-antigen, nucleic acid-protein, nucleic acid-nucleic acid, ligand-protein, ligand-nucleic acid, ligand-ligand, eukaryotic or prokaryotic cell surface moiety-second moiety, the measurement of two or more receptors on the surface of an eukaryotic or prokaryotic cell, the development of three-hybrid type systems using tandem fusions, interactor-cofactor, etc. Many other types of interactions are also known that can be adapted to the system. However, there is a need in the art for improved methods of RNAi screening, quickly and accurately.
  • The present invention provides methods for the screening of lethal and synthetic lethal RNAi-induced phenotype on a microfluidic device. The present invention utilizes a lentiviral library of RNAi where each virus has a unique 60-nt identifying barcode bracketed on either side with nucleotide sequences common to all vectors.
  • The analysis of lethal and synthetic lethal RNAi-induced phenotypes occurs in two steps. In the first step, the viral library is combined in bulk and infected, also in bulk, into an appropriate host strain. The molar amount of each of the different lentivirus in the library is pre-determined by sequencing on, for example, an appropriate instrument or by gene expression analysis on a microfluidic device. Post infection, the treated cells are collected and the 60-nt barcode is amplified from chromosomal DNA using PCR primers based on the bracketing sequence. In the second step, the PCR amplification product is added to a microfluidic device and analyzed against a labeled droplet library wherein the labeled droplets contain lentiviral-barcode-quantification reagents (e.g., molecular beacons, Taqman probes, etc.) against each of said lentiviral barcodes. A gene-expression analysis-like analysis is performed to quantify the amount of each lentiviral barcode-type in the treated cells. An absence or significant decrease of any lentiviral barcode in the amplified product can be assumed to be due to the death of that barcode-containing lentivirus in the treated cells. In an embodiment, the products within the droplets can also be amplified.
  • GFP or additional transcription analysis can also occur in two steps. In step one, the viral library is combined in bulk and infected, also in bulk, into an appropriate host strain. The molar amount of each of the different lentivirus in the library is pre-determined by sequencing on, for example, an appropriate instrument or by gene expression analysis on a microfluidic device. Post infection, the treated cells are i) collected in bulk, ii) sorted using a phenotype able to be sorted in a microfluidic device (e.g, GFP expression, cell-surface marker, low-copy cell-surface marker, etc) and iii) the 60-nt barcode is amplified from chromosomal DNA using PCR primers based on the bracketing sequence. In the second step, the PCR amplification product is added to a microfludic device and analyzed against a labeled droplet library wherein the labeled droplets contain lentiviral-barcode-quantification reagents (e.g., molecular beacons, Taqman probes, etc.) against each of said lentiviral barcodes. A gene-expression analysis-like analysis is performed to quantify the amount of each lentiviral barcode-type in the treated cells. An absence or significant decrease of any lentiviral barcode in the amplified product can be assumed to be due to the death of that barcode-containing lentivirus in the treated cells.
  • Example 9
  • Many diseases are associated with particular chromosomal abnormalities. For example, chromosomes in cancerous cells frequently exhibit aberrations called translocations, where a piece of one chromosome breaks off and attaches to the end of another chromosome. Identifying such chromosome abnormalities and determining their role in disease is an important step in developing new methods for diagnosing many genetic disorders. Traditional karyotyping using Giemsa staining allows scientists to view the full set of human chromosomes in black and white, a technique that is useful for observing the number and size of the chromosomes. However, there is a need in the art for improved methods of karyotyping, quickly and accurately.
  • The present invention provides methods for karyotyping. Preferably, the karyotyping screens occur within droplets on a microfluidic device. Currently, scientists cannot accurately identify many translocations or other abnormalities using only a black and white karyotype. Spectral karyotyping (SKY) is a laboratory technique that allows scientists to visualize all 23 pairs of human chromosomes at one time, with each pair of chromosomes painted in a different fluorescent color. By using SKY, they can easily see instances where a chromosome, painted in one color, has a small piece of a different chromosome, painted in another color, attached to it.
  • Through the use of droplet-based methods, chromosomes can be captured within droplets without having to worry about shear-forces. The chromosomes can then be passed through a ‘neck-down’ to stretch them out. Labeling prior to loading, with either Giemsa stain or oligonucleotide probes, can be used to karyotype the DNA as it flows.
  • The present invention provides methods of using SKY probes to ‘paint’ individual chromosomes. Also provided is a method used by flow cytometrists for the preparation of chromosomes prior to flow analysis, including flow sorting. The present invention provides methods which allow the adaptation of these methods for use on a microfluidic device.
  • SKY involves the preparation of a large collection of short sequences of single-stranded DNA called probes. Each of the individual probes in this DNA library is complementary to a unique region of one chromosome; together, all of the probes make up a network of DNA that is complementary to all of the chromosomes within the human genome. Each probe is labeled with a fluorescent molecule that corresponds to the chromosome to which it is complementary. For example, probes that are complementary to chromosome 1 are labeled with yellow molecules, while those that are complementary to chromosome 2 are labeled with red molecules, and so on. When these probes are mixed with the chromosomes from a human cell, the probes hybridize, or bind, to the DNA in the chromosomes. As they hybridize, the fluorescent probes essentially paint the set of chromosomes in a rainbow of colors. Scientists can then use computers to analyze the painted chromosomes to determine whether any of them exhibit translocations or other structural abnormalities. See, FIG. 35.
  • Prior to analysis chromosomes can be prepared as described in Bee Ling Ng and Nigel P. Carter “Factors Affecting Flow Karyotype Resolution. Cytometry” Part A 69A: 1028-1036 (2006) as follows:
      • 1. Arrest cells at metaphase using 0.1 lg/ml demecolcine for optimal amount of time, dependent on the cell cycle time of the cell lines. (Approximately 5 h for suspension, 16 h for adherent cell lines and 4 h for LPS stimulated B lymphocyte culture).
      • 2. Harvest cells and centrifuge at 289 g for 5 min. Remove supernatant.
      • 3. Resuspend cell pellet in 5 ml of hypotonic solution (75 mM KCl, 10 mM MgSO4, 0.2 mM spermine, 0.5 mM spermidine, pH 8.0) and incubate at room temperature for 10 min.
      • 4. Centrifuge cell suspension at 289 g for 5 min. Remove supernatant.
      • 5. Resuspend cell pellet in 3 ml of ice cold polyamine isolation buffer (PAB, containing 15 mM Tris, 2 mM EDTA, 0.5 mM EGTA, 80 mM KCl 3 mM dithiothreitol, 0.25% Triton X-100, 0.2 mM spermine, 0.5 mM spermidine, pH 7.50) and vortex for 20 s.
      • 6. Briefly centrifuge chromosome suspensions at 201 g for 2 min. Filter supernatant through 20 lm mesh filter.
      • 7. Stain chromosomes overnight with 5 lg/ml of Hoechst, 40 lg/ml chromomycin A3 and 10 mM MgSO4.
      • 8. To the stained chromosome suspension, add 10 mM of sodium citrate and 25 mM of sodium sulphite 1 h before flow analysis.
  • The present invention also provides methods of sorting chromosomes for karyotyping wherein individual chromosomes are sorted. This sorting can be performed after chromosome-specific identification (such as hybridization of labeled probes) so as to enrich a population for one or more specific chromosomes. This enriched population can be used in DNA sequencing reactions.
  • The Giemsa-stained and/or labeled-probe-hybridized chromosomes can be sent through a constriction of a channel on a microfluidic device to detect the areas of stain and/or label as a genetic ‘bar-code’ to identify regions of translocation, etc. on individual chromosomes.
  • Identification of chromosomes and karyotyping can be used after enrichment of specific cell-types, for example i) fetal cells from maternal blood, or ii) cancer cells from human blood.
  • Example 10
  • The present invention provides microbial strains with improved biomass conversion and methods of preparing such strains. Biomass is organic matter such as plant matter, i.e., trees, grasses, agricultural crops, or other biological material such as animal material. It can be used as a solid fuel, or converted into liquid or gaseous forms, for the production of electric power, heat, chemicals, or fuels. Biomass can also be used in formulating other commercial products in other industrial sectors such as textiles, food supply, environmental, communication, housing, etc. For example, biofuel development seeks the development of new microbial strains with improved biomass conversion to ethanol.
  • Researchers have been applying sophisticated metabolic engineering techniques to develop microorganisms that can more effectively ferment the sugars in biomass. Lignocellulosic biomass contains five carbon sugars such as xylose (from the hemicellulose) as well as the more “common” six carbon sugars such as glucose found in grains. This makes fermentation and other bioprocessing far more challenging. While some biorefmery scenarios will take advantage of the different sugar streams to produce multiple products, others will be more cost effective if all the sugars can coferment in a single set of equipment. Accordingly, researchers are developing microorganisms that can coferment all the sugars in biomass in order to improve ethanol production economics. With industrial partners, researchers are working to develop designer strains of microorganisms for biomass conversion of specific feedstocks, feedstreams, and processes. Thus, there is a need for devices and methods for the rapid engineering of new microbial strains with improved biomass conversion.
  • The present invention provides a microfluidic device in which to formulate a mutant bacterial, yeast, or fungi strain which can be used for biomass energy conversion. The microorganism strain can be engineered, e.g., by recombinant methods, to include at least one nucleic acid sequence encoding one or more polypeptides of interest, wherein the mutant strain expresses the polypeptides of interest at a higher level than the corresponding non-mutant strain under the same conditions.
  • In one embodiment, the nucleic acid sequence can be operably linked to an expression-regulating region selected from the group consisting of a promoter sequence associated with cellulase expression, xylanase expression, or gpdA expression. In another embodiment, the nucleic acid sequence can further be optionally linked to a secretion signal sequence.
  • In one embodiment, the nucleic acid sequence can be a heterologous nucleic acid sequence selected from heterologous polypeptide-encoding nucleic acid sequences, heterologous signal sequences, or heterologous expression-regulating sequences, or combinations thereof. For instance, the nucleic acid sequence can be a heterologous signal sequence, e.g., a secretion signal sequence. Alternatively, the nucleic acid sequence can be a heterologous expression-regulating region, e.g. an inducible promoter or a high expression promoter.
  • The polypeptides of interest can be homologous peptides and are expressed in the mutant strain at a higher level than in the corresponding non-mutant strain under the same conditions. In one embodiment, the polypeptides of interest can be selected from one or more of carbohydrate-degrading enzymes, proteases, lipases, esterases, other hydrolases, oxidoreductases, and transferases. In yet another embodiment, the polypeptides of interest can be selected from one or more of fungal enzymes that allow production or overproduction of primary metabolites, organic acids, secondary metabolites, and antibiotics. These fungal sequences can include secretion signal sequences, for example, and can be selected from one or more of cellulase, β-galactosidase, xylanase, pectinase, esterase, protease, amylase, polygalacturonase or hydrophobin. Alternatively, the fungal sequences can include one or more fungal expression-regulating regions. Preferably, the polypeptides of interest exhibit optimal activity and/or stability at a pH above 6, and/or have more than 70% of its activity and/or stability at a pH above 6.
  • The mutant microorganism can further include a selectable marker. The selectable marker can confer resistance to a drug, for example, or relieve a nutritional defect.
  • In another embodiment, the microorganism can be mutated via mutagenesis. Mutagenesis can be achieved, for example, by one or both of UV irradiation or chemical mutagenesis. For example, in one embodiment mutagenesis can include exposing a microorganism to UV irraditation, exposing it to N-methyl-N′-nitro-N-nitrosoguanidine, and exposing it again to UV irradiation.
  • The present invention also provides methods for creating microbial strains with improved biomass conversion. In one embodiment, the method includes providing a microfluidic device made of a microfabricated substrate. The microfabricated substrate can have a plurality of electrically addressable, channel bearing modules integrally arranged so as to be in fluid communication with each other, thereby forming at least one main channel adapted to carry at least one continuous phase fluid. The method further includes flowing a buffer, a microbe library, and a media (either in separate solutions or all together in one solution) through a first inlet channel into the main channel of the microfabricated substrate such that one or more droplets is formed in the continuous phase fluid; flowing a substrate through a second inlet channel into the main channel of the microfabricated substrate such that one or more droplets is formed in said continuous phase fluid; coalescing the droplets containing the microbe library with the droplets containing the substrate as the droplets pass through a coalescence module, thereby producing a NanoRefinery; interrogating the NanoRefinery for a predetermined characteristic within a detection module on the microfabricated substrate; and collecting the NanoRefineries containing the microbes of interest in a collection module on the microfabricated substrate. An assay system, e.g., a means by which to determine whether a desired product has been produced, or a means by which to determine the absence of a starting substrate material, can also be incorporated into one of the droplets or the NanoRefinery. The assay system can be added either before, after, or simultaneously with the addition of the substrate.
  • In one embodiment, the assay system is a dye that can measure the amount of sugar in a solution. In another embodiment, the microbe library can include one or more of DNA, bacteria, yeast or fungi. The substrate can include biomass, which can include one or more of fermentation broth, cellulose or other polysaccharide, or plant lignan.
  • The present invention further provides methods for degrading or converting biomass into one or more products. In one embodiment, the method includes treating the biomass with an effective amount of a recombinant microorganism, wherein the recombinant microorganism expresses or overexpresses one or more heterologous sequences encoding enzymes that degrade or convert the biomass into one or more products. In one embodiment, the biomass can include plant cell wall polysaccharides. In another embodiment, the products can include one or more of the commodity chemicals or secondary commodity chemicals used to produce one or more of the Intermediates or finished products and consumer goods listed in FIGS. 36 and 37.
  • Example 11
  • The present invention provides methods of screening for enzymes with improved activity. As an example, at least one cell (prokaryotic or eukaryotic) is deposited into a droplet and the cells are allowed to secrete a substance for which a homogeneous assay is available. For a specific example, an individual Bacillus subtilis cell from a mutagen-treated culture is deposited within a 30 micron growth-medium containing droplet, the resulting droplets are collected and allowed to incubate overnight. The bacterium secretes a protease into the droplet. The droplets containing the Bacilli are then individually merged with an assay droplet containing a protease-cleavable dye-labeled peptide. The uncleaved peptide is colorless, while the cleaved peptide becomes red. The droplets are incubated on chip for a sufficient time such as to allow color formation. Droplets that are red are sorted. The collected sorted droplets are then plated onto solid growth medium and the resulting colonies, after overnight incubation, represent individual clonal isolates from the droplets.
  • In addition, the assay droplets can be sorted based upon a specific activity, for example enzymes that are more active, indicated by a more intense red droplet after a specific period of time. In addition, the conditions within the droplet can be changed during the merging of the two droplets to assay conditions which may itself not be permissive for the bacteria. For example the pH within the droplet is altered in order to find mutant enzymes that work better under either acidic or alkaline conditions. Or the droplets can be heated such that enzymes that are more heat resistant are identified.
  • The merging of the droplets can be right after the individual cell is placed into the droplet or after further incubation.
  • Another method of the instant invention is as described above, except that the cells are lysed before the assay step in order to release the contents of the cell into the droplet.
  • In other embodiments, the substrate can be added with- or simultaneously formulated from two or more separate reagent streams with the contained within a droplet. For a specific example, macrophage cells are washed in buffer and incubated with an enzyme-conjugated anti-cell-surface antibody. The cells are then individually loaded into droplets formulated at the same time, with the enzyme substrate. The amount of enzyme substrate turned over within the droplet will be proportional to the number of enzyme molecules within the droplet, which is proportional to the number of antibodies bound to the macrophage surface. By careful calibration it should be possible to estimate the number of cell-surface molecules attached to the cell surface.

Claims (199)

1. A method of pairing sample fluids to form a droplet or nanoreactor comprising:
a) providing a microfluidic substrate comprising at least two inlet channels adapted to carry at least two dispersed phase sample fluids and at least one main channel adapted to carry at least one continuous phase fluid;
b) flowing a first sample fluid through a first inlet channel which is in fluid communication with said main channel at a junction, wherein said junction comprises a first fluidic nozzle designed for flow focusing such that said first sample fluid forms a plurality of highly uniform, monodisperse droplets of a first size in said continuous phase;
c) flowing a second sample fluid through a second inlet channel which is in fluid communication with said main channel at a junction, wherein said junction comprises a second fluidic nozzle designed for flow focusing such that said second sample fluid forms a plurality of highly uniform, monodisperse droplets of a second size in said continuous phase, wherein the size of the droplets of the second sample fluid are smaller than the size of the droplets of the first sample fluid;
d) providing a flow and droplet formation rate of the first and second sample fluids wherein the droplets are interdigitized such that a first sample fluid droplet is followed by and paired with a second sample fluid droplet;
e) providing channel dimensions such that the paired first sample fluid and the second sample fluid droplet are brought into proximity;
f) coalescing the paired first and second sample droplets as the paired droplets pass through an electric field, thereby producing a droplet or nanoreactor.
2. The method of claim 1, wherein said inlet and main channels are coated with an anti-wetting or blocking agent for the dispersed phase.
3. The method of claim 2, wherein said channels are coated with a silica primer layer followed by a perfluoroalkylalkylsilane compound, an amorphous soluble perfluoropolymer, BSA, PEG-silane or fluorosilane.
4. The method of claim 3, wherein the channels are coated with a silica primer layer followed by a perfluoroalkylalkylsilane compound.
5. The method of claim 1, wherein said plurality of droplets comprises a biological/chemical material.
6. The method of claim 5, wherein said biological/chemical material is selected from the group consisting of tissues, cells, particles, proteins, antibodies, amino acids, nucleotides, small molecules, and pharmaceuticals.
7. The method of claim 5, wherein said biological/chemical material comprises a label.
8. The method of claim 7, wherein each biological/chemical material is labeled with a unique label.
9. The method of claim 8, wherein said label is a protein, a DNA tag, a dye, a quantum dot or a radio frequency identification tag.
10. The method of claim 7, wherein said label can be detected by fluorescence polarization, fluorescence intensity, fluorescence lifetime, fluorescence energy transfer, pH, ionic content, temperature or combinations thereof.
11. The method of claim 1, wherein said continuous phase is a non-polar solvent.
12. The method of claim 1, wherein said continuous phase is a fluorocarbon oil.
13. The method of claim 1, wherein said continuous phase further comprises one or more additives.
14. The method of claim 13, wherein said additive is a fluorosurfactant.
15. The method of claim 14, wherein said fluorosurfactant is a perfluorinated polyether.
16. The method of claim 1, wherein the flow of said dispersed phase fluid and said continuous phase fluid is pressure driven.
17. The method of claim 1, wherein said coalescing step occurs within a coalescence module in fluid communication with the main channel on the microfluidic substrate.
18. The method of claim 17, wherein the coalescence module comprises one or more electrodes that generate an electric field.
19. The method of claim 1, wherein said channel dimensions comprise an expanded portion of the main channel between the electrodes to bring successive droplets into proximity.
20. The method of claim 1, wherein said channel dimensions comprise a narrowed portion of the main channel to center droplets within the main channel followed by a expanded portion of the main channel between the electrodes to bring successive droplets into proximity.
21. The method of claim 1, wherein the droplets have no charge.
22. The method of claim 1, further comprising interrogating the droplet or nanoreactor for at least one predetermined characteristic within a detection module in fluid communication with the main channel on the microfluidic substrate.
23. The method of claim 22, wherein said detection module comprising a detection apparatus for evaluating the contents or characteristics of the droplet or nanoreactor.
24. The method of claim 23, wherein said detection apparatus comprises an optical or electrical detector.
25. The method of claim 1, further comprising sorting the droplet or nanoreactor into or away from a collection module within a sorting module in fluid communication with the main channel on the microfluidic substrate in response to the contents or characterization of the droplet or nanoreactor evaluated in the detection module.
26. The method of claim 25, wherein said sorting module comprises a sorting apparatus adapted to direct the droplet or nanoreactor into or away from the collection module.
27. The method of claim 26, wherein the sorting apparatus comprises one or more electrodes that generate an electric field.
28. The method of claim 25, wherein the droplets or nanoreactors have no charge.
29. A method of forming a droplet emulsion library of a sample fluid comprising:
a) providing at least one first channel adapted to carry at least one dispersed phase sample fluid and at least one second channel adapted to carry at least one continuous phase fluid;
b) flowing said sample fluid through said first channel which is in fluid communication with said second channel at a junction, wherein said junction comprises a fluidic nozzle such that said sample fluid forms a plurality of highly uniform, monodisperse droplets of a predetermined size in said continuous phase, wherein the fluidic nozzle is isolated from the dispersed phase fluid, has a three dimensional design to permit flow focusing and eliminates surface wetting.
30. A method of forming a droplet emulsion library of a sample fluid comprising:
a) providing a microfluidic substrate comprising at least one first channel adapted to carry at least one dispersed phase sample fluid and at least one second channel adapted to carry at least one continuous phase fluid;
b) flowing said sample fluid through said first channel which is in fluid communication with said second channel at a junction, wherein said junction comprises a fluidic nozzle such that said sample fluid forms a plurality of highly uniform, monodisperse droplets of a predetermined size in said continuous phase.
31. A method of forming a droplet emulsion library of a sample fluid comprising:
a) providing a microfluidic substrate comprising at least one first channel adapted to carry at least one dispersed phase sample fluid and at least one channel adapted to carry at least one continuous phase fluid;
b) providing a means for storing said sample fluid wherein said storage means is in fluid communication with said first channel and provides means for introducing said sample fluid to said inlet channel, wherein the storage means contains an immiscible phase fluid with a density less than that of the sample fluid;
c) introducing the sample fluid into the storage means wherein said sample fluid flows through the less dense immiscible fluid such that the sample fluid settles at the bottom of the storage means and is subsequently introduced into the first channel;
d) flowing said sample fluid through said first channel which is in fluid communication with said second channel at a junction, wherein said junction comprises a fluidic nozzle such that said sample fluid forms a plurality of highly uniform, monodisperse droplets of a predetermined size in said continuous phase.
32. A method of forming a droplet emulsion library of a sample fluid comprising:
a) providing a microfluidic substrate comprising at least one first channel adapted to carry at least one dispersed phase sample fluid and at least one channel adapted to carry at least one continuous phase fluid;
b) providing a means for storing said sample fluid wherein said storage means is in fluid communication with said first channel and provides means for introducing said sample fluid to said inlet channel, wherein the storage means contains an immiscible phase fluid with a density greater than that of the sample fluid;
c) inserting a sample fluid introduction apparatus into the storage means wherein said sample fluid is forced through the more dense immiscible fluid by the introduction apparatus such that the sample fluid is subsequently introduced into the first channel;
d) flowing said sample fluid through said first channel which is in fluid communication with said second channel at a junction, wherein said junction comprises a fluidic nozzle such that said sample fluid forms a plurality of highly uniform, monodisperse droplets of a predetermined size in said continuous phase.
33. The method of claims 29-32, wherein said channels are coated with an anti-wetting or blocking agent for the dispersed phase.
34. The method of claim 33, wherein said channels are coated with a silica primer layer followed by a perfluoroalkylalkylsilane compound, an amorphous soluble perfluoropolymer, BSA, PEG-silane or fluorosilane.
35. The method of claim 34, wherein the channels are coated with a silica primer layer followed by a perfluoroalkylalkylsilane compound.
36. The method of claims 29-32, wherein said plurality of droplets comprises a biological/chemical material.
37. The method of claim 36, wherein said biological/chemical material is selected from the group consisting of tissues, cells, particles, proteins, antibodies, amino acids, nucleotides, small molecules, and pharmaceuticals.
38. The method of claim 36, wherein said biological/chemical material comprises a label.
39. The method of claim 38, wherein each biological/chemical material is labeled with a unique label.
40. The method of claim 39, wherein said label is a protein, a DNA tag, a dye, a quantum dot or a radio frequency identification tag.
41. The method of claim 38, wherein said label can be detected by fluorescence polarization, fluorescence intensity, fluorescence lifetime, fluorescence energy transfer, pH, ionic content, temperature or combinations thereof.
42. The method of claims 29-32, wherein said continuous phase is a non-polar solvent.
43. The method of claims 29-32, wherein said continuous phase is a fluorocarbon oil.
44. The method of claims 29-32, wherein said continuous phase further comprises one or more additives.
45. The method of claim 44, wherein said additive is a fluorosurfactant.
46. The method of claim 45, wherein said fluorosurfactant is a perfluorinated polyether.
47. The method of claims 29-32, wherein the flow of said dispersed phase fluid and said continuous phase fluid is pressure driven.
48. The method of claim 29, wherein the nozzle is formed from small bore tubing or from the tip of a molded ferrule.
49. The method of claim 31 and 32, wherein said storage means is a well or reservoir.
50. The method of claim 32, wherein the fluid introduction apparatus is a sample tip loading pump.
51. A method of forming a uniformed sized droplet emulsion library comprising:
a) providing a means for separating droplets of similar sizes, wherein said means comprises a periodic array of geometric parameters defining an obstacle matrix;
b) introducing at least one sample fluid containing various sized droplets to said separating means;
c) subjecting said sample fluid to laminar flow through the microscale obstacles within the separating means, wherein said sample fluids do not mix; and
d) separating and isolating uniformed sized droplets from within said sample fluid by deterministic lateral displacement.
52. The method of claim 51, wherein the means is selected from the group consisting of a microfluidic channel, microfluidic lateral diffusion device, tube, syringe, column and capillary.
53. A method for solidifying a droplet or nanoreactor comprising:
a) providing a microfluidic substrate comprising at least one inlet channel adapted to carry at least one dispersed phase sample fluid and at least one main channel adapted to carry at least one continuous phase fluid;
b) incorporating at least one solidifying agent within a sample fluid;
c) flowing the sample fluid through a first inlet channel which is in fluid communication with said main channel at a junction, such that said first sample fluid forms a plurality of highly uniform, monodisperse droplets in said continuous phase;
d) providing a means which activates the solidifying agent such that the droplet or nanoreactor forms a matrix.
54. A method for solidifying a droplet or nanoreactor comprising:
a) providing a microfluidic substrate comprising at least two inlet channels adapted to carry at least two dispersed phase sample fluids and at least one main channel adapted to carry at least one continuous phase fluid;
b) incorporating at least one solidifying agent within at least one sample fluid;
c) flowing a first sample fluid through a first inlet channel which is in fluid communication with said main channel at a junction, such that said first sample fluid forms a plurality of highly uniform, monodisperse droplets in said continuous phase;
d) flowing a second sample fluid through a second inlet channel which is in fluid communication with said main channel at a junction, such that said second sample fluid forms a plurality of highly uniform, monodisperse droplets in said continuous phase;
e) coalescing at least one droplet formed in step (c) with at least one droplet formed in step (d) as the droplets pass through an electric field, thereby producing a droplet or nanoreactor; and
f) providing a means which activates the solidifying agent such that the droplet or nanoreactor forms a matrix.
55. The method of claim 54, wherein said inlet and main channels are coated with an anti-wetting or blocking agent for the dispersed phase.
56. The method of claim 55, wherein said channels are coated with a silica primer layer followed by a perfluoroalkylalkylsilane compound, an amorphous soluble perfluoropolymer, BSA, PEG-silane or fluorosilane.
57. The method of claim 56, wherein the channels are coated with a silica primer layer followed by a perfluoroalkylalkylsilane compound.
58. The method of claim 54, wherein said plurality of droplets comprises a biological/chemical material.
59. The method of claim 58, wherein said biological/chemical material is selected from the group consisting of tissues, cells, particles, proteins, antibodies, amino acids, nucleotides, small molecules, and pharmaceuticals.
60. The method of claim 58, wherein said biological/chemical material comprises a label.
61. The method of claim 60, wherein each biological/chemical material is labeled with a unique label.
62. The method of claim 61, wherein said label is a protein, a DNA tag, a dye, a quantum dot or a radio frequency identification tag.
63. The method of claim 60, wherein said label can be detected by fluorescence polarization, fluorescence intensity, fluorescence lifetime, fluorescence energy transfer, pH, ionic content, temperature or combinations thereof.
64. The method of claim 54, wherein said continuous phase is a non-polar solvent.
65. The method of claim 54, wherein said continuous phase is a fluorocarbon oil.
66. The method of claim 54, wherein said continuous phase further comprises one or more additives.
67. The method of claim 66, wherein said additive is a fluorosurfactant.
68. The method of claim 67, wherein said fluorosurfactant is a perfluorinated polyether.
69. The method of claim 54, wherein the flow of said dispersed phase fluid and said continuous phase fluid is pressure driven.
70. The method of claim 54, wherein said coalescing step occurs within a coalescence module in fluid communication with the main channel on the microfluidic substrate.
71. The method of claim 70, wherein the coalescence module comprises one or more electrodes that generate an electric field.
72. The method of claim 70, wherein said coalescence module comprises an expanded portion of the main channel between the electrodes to bring successive droplets into proximity, whereby the paired droplets are coalesced within the electric field.
73. The method of claim 70, wherein said coalescence module comprises a narrowed portion of the main channel to center droplets within the main channel followed by a expanded portion of the main channel between the electrodes to bring successive droplets into proximity, whereby the paired droplets are coalesced within the electric field.
74. The method of claim 54, wherein the droplets have no charge.
75. The method of claim 54, further comprising interrogating the droplet or nanoreactor for at least one predetermined characteristic within a detection module in fluid communication with the main channel on the microfluidic substrate.
76. The method of claim 75, wherein said detection module comprising a detection apparatus for evaluating the contents or characteristics of the droplet or nanoreactor.
77. The method of claim 76, wherein said detection apparatus comprises an optical or electrical detector.
78. The method of claim 54, further comprising sorting the droplet or nanoreactor into or away from a collection module within a sorting module in fluid communication with the main channel on the microfluidic substrate in response to the contents or characterization of the droplet or nanoreactor evaluated in the detection module.
79. The method of claim 78, wherein said sorting module comprises a sorting apparatus adapted to direct the droplet or nanoreactor into or away from the collection module.
80. The method of claim 79, wherein the sorting apparatus comprises one or more electrodes that generate an electric field.
81. The method of claim 78, wherein the droplets or nanoreactors have no charge.
82. The method of claim 54, wherein the solidifying agent is a low temperature agarose or a polymerizing solution.
83. The method of claim 54, wherein the means for activating the solidifying agent is a physical or chemical means.
84. A method for introducing sample fluid to a microfluidic substrate comprising:
a) providing a microfluidic substrate comprising at least one inlet channel adapted to carry at least one dispersed phase sample fluid and at least one main channel adapted to carry at least one continuous phase fluid;
b) providing a means for storing said sample fluid, wherein said storage means is in fluid communication with said inlet channel and provides a means for introducing said sample fluid into said inlet channel;
c) combining the sample fluid with at least one immiscible phase fluid within the storage means, wherein the immiscible phase fluid has a density different from that of the sample fluid such that the fluids separate into distinct layers;
d) providing a force such that the immiscible phase fluid forces the sample fluid completely into the inlet channel of the microfluidic substrate.
85. A method for introducing sample fluid to a microfluidic substrate comprising:
a) providing a microfluidic substrate comprising at least one inlet channel adapted to carry at least one dispersed phase sample fluid and at least one main channel adapted to carry at least one continuous phase fluid;
b) providing a means for storing said sample fluid, wherein said storage means is in fluid communication with said inlet channel and provides a means for introducing said sample fluid into said inlet channel;
c) combining the sample fluid with at least two immiscible phase fluids within the storage means, wherein a first immiscible phase fluid has a density greater than that of the sample fluid and a second immiscible phase fluid has a density less than that of the sample fluid such that the fluids separate into distinct layers with the sample fluid layer residing between the two immiscible phase layers;
d) providing a force such that the more dense immiscible phase fluid forces the less dense immiscible phase fluid and the sample fluid completely into the inlet channel of the microfluidic substrate.
86. The method of claims 84 and 85, wherein said inlet and main channels are coated with an anti-wetting or blocking agent for the dispersed phase.
87. The method of claim 86, wherein said channels are coated with a silica primer layer followed by a perfluoroalkylalkylsilane compound, an amorphous soluble perfluoropolymer, BSA, PEG-silane or fluorosilane.
88. The method of claim 87, wherein the channels are coated with a silica primer layer followed by a perfluoroalkylalkylsilane compound.
89. The method of claims 84 and 85, wherein said plurality of sample fluid comprises a biological/chemical material.
90. The method of claim 89, wherein said biological/chemical material is selected from the group consisting of tissues, cells, particles, proteins, antibodies, amino acids, nucleotides, small molecules, and pharmaceuticals.
91. The method of claim 89, wherein said biological/chemical material comprises a label.
92. The method of claim 89, wherein each biological/chemical material is labeled with a unique label.
93. The method of claim 91, wherein said label is a protein, a DNA tag, a dye, a quantum dot or a radio frequency identification tag.
94. The method of claim 91, wherein said label can be detected by fluorescence polarization, fluorescence intensity, fluorescence lifetime, fluorescence energy transfer, pH, ionic content, temperature or combinations thereof.
95. The method of claims 84 and 85, wherein said continuous phase is a non-polar solvent.
96. The method of claims 84 and 85, wherein said continuous phase is a fluorocarbon oil.
97. The method of claims 84 and 85, wherein said continuous phase further comprises one or more additives.
98. The method of claim 97, wherein said additive is a fluorosurfactant.
99. The method of claim 98, wherein said fluorosurfactant is a perfluorinated polyether.
100. The method of claims 84 and 85, wherein the flow of said dispersed phase fluid and said continuous phase fluid is pressure driven.
101. The method of claims 84 and 85, wherein said storage means is a well or reservoir.
102. The method of claims 84 and 85, wherein said immiscible fluid is biologically and/or chemically inert.
103. A method of extracting biological or chemical material from within a droplet or nanoreactor comprising:
a) providing one or more droplets or nanoreactors formed within a microfluidic substrate;
b) combining the droplets or nanoreactors with a first immiscible fluid with a density greater than that of the droplets or nanoreactors such that said droplets or nanoreactors and immiscible fluid form separate layers;
c) removing the droplet or nanoreactor layer formed in step (b);
d) combining and mixing the droplet or nanoreactor layer with a second immiscible fluid comprising a destabilizing surfactant such that the droplets or nanoreactors and immiscible fluid form separate layers; and
e) removing the resulting aqueous layer formed step (d), thereby extracting the biological or chemical material from within the droplet or nanoreactor.
104. The method of claim 103, wherein said immiscible fluid is biologically and/or chemically inert.
105. The method of claim 103, wherein said destabilizing surfactant is a perfluorinated alcohol.
106. The method of claim 103, wherein the droplets or nanoreactors reside on top of the immiscible fluids within said separate layers.
107. The method of claim 103, wherein said biological/chemical material comprises a label.
108. The method of claim 107, wherein each biological/chemical material is labeled with a unique label.
109. The method of claim 107, wherein said label is a protein, a DNA tag, a dye, a quantum dot or a radio frequency identification tag.
110. The method of claim 107, wherein said label can be detected by fluorescence polarization, fluorescence intensity, fluorescence lifetime, fluorescence energy transfer, pH, ionic content, temperature or combinations thereof.
111. A method of amplifying DNA comprising:
a) providing a microfluidic substrate comprising at least one inlet channel adapted to carry at least one dispersed phase sample fluid and at least one main channel adapted to carry at least one continuous phase fluid, wherein the main channel comprises a serpentine line with heating and cooling regions;
b) flowing a sample fluid comprising one more target DNA molecules to be amplified, PCR primer pair sets, dNTPs, enzymes and buffer components effective to permit PCR amplification through an inlet channel which is in fluid communication with said main channel at a junction, such that said sample fluid forms a plurality of highly uniform, monodisperse droplets in said continuous phase, wherein said junction is heated to 95° C. to provide a hot start, wherein a portion of at least one of the PCR primer pair sets is attached to a semi-solid substrate;
c) reacting the contents of the droplets for at least twenty heating and cooling cycles to permit the PCR amplification of the target DNA molecules,
such that the amplified target DNA molecules are attached to a semi-solid substrate.
112. A method of amplifying DNA comprising:
a) providing a microfluidic substrate comprising at least two inlet channels adapted to carry at least two dispersed phase sample fluids and at least one main channel adapted to carry at least one continuous phase fluid, wherein the main channel comprises a serpentine line with heating and cooling regions;
b) flowing a first sample fluid comprising one more target DNA molecules to be amplified through a first inlet channel which is in fluid communication with said main channel at a junction, such that said first sample fluid forms a plurality of highly uniform, monodisperse droplets in said continuous phase;
c) flowing a second sample fluid comprising PCR primer pair sets, dNTPs, enzymes and buffer components effective to permit PCR amplification through a second inlet channel which is in fluid communication with said main channel at a junction, such that said second sample fluid forms a plurality of highly uniform, monodisperse droplets in said continuous phase, wherein said junction is heated to 95° C. to provide a hot start and wherein a portion of at least one of the PCR primer pair sets is attached to a semi-solid substrate;
d) coalescing the droplets comprising the DNA molecules from step (b) with the droplets comprising the PCR primer pair sets, dNTPs, enzymes and buffer components from step (c) within an electric field;
e) reacting the contents of the droplets for at least twenty heating and cooling cycles to permit the PCR amplification of the target DNA molecules,
such that the amplified target DNA molecules are attached to a semi-solid substrate.
113. A method of amplifying DNA comprising:
a) providing a microfluidic substrate comprising at least one inlet channel adapted to carry at least one dispersed phase sample fluid and at least one main channel adapted to carry at least one continuous phase fluid, wherein the main channel comprises a serpentine line with heating and cooling regions;
b) flowing a sample fluid comprising one more target DNA molecules to be sequenced, primer pair sets, dNTPs, enzymes and buffer components effective to permit isothermal amplification through an inlet channel which is in fluid communication with said main channel at a junction, such that said sample fluid forms a plurality of highly uniform, monodisperse droplets in said continuous phase, wherein a portion of at least one of the primer pair sets is attached to a semi-solid substrate;
c) reacting the contents of the droplets to permit the isothermal amplification of the target DNA molecules,
such that the amplified target DNA molecules are attached to a semi-solid substrate.
114. A method of amplifying DNA comprising:
a) providing a microfluidic substrate comprising at least two inlet channels adapted to carry at least two dispersed phase sample fluids and at least one main channel adapted to carry at least one continuous phase fluid, wherein the main channel comprises a serpentine line with heating and cooling regions;
b) flowing a first sample fluid comprising one more target DNA molecules to be amplified through a first inlet channel which is in fluid communication with said main channel at a junction, such that said first sample fluid forms a plurality of highly uniform, monodisperse droplets in said continuous phase;
c) flowing a second sample fluid comprising primer pair sets, dNTPs, enzymes and buffer components effective to permit isothermal amplification through a second inlet channel which is in fluid communication with said main channel at a junction, such that said second sample fluid forms a plurality of highly uniform, monodisperse droplets in said continuous phase, wherein a portion of at least one of the primer pair sets is attached to a semi-solid substrate;
d) coalescing the droplets comprising the DNA molecules from step (b) with the droplets comprising the primer pair sets, dNTPs, enzymes and buffer components from step (c) within an electric field;
e) reacting the contents of the droplets for at least twenty heating and cooling cycles to permit the isothermal amplification of the target DNA molecules,
such that the amplified target DNA molecules are attached to a semi-solid substrate.
115. A method of sequencing DNA comprising:
a) providing a microfluidic substrate comprising at least two inlet channels adapted to carry at least two dispersed phase sample fluids and at least one main channel adapted to carry at least one continuous phase fluid, wherein the main channel comprises a serpentine line with heating and cooling regions;
b) flowing a first sample fluid comprising one more target DNA molecules to be sequenced, PCR primer pair sets, dNTPs, enzymes and buffer components effective to permit PCR amplification through a first inlet channel which is in fluid communication with said main channel at a junction, such that said first sample fluid forms a plurality of highly uniform, monodisperse droplets in said continuous phase, wherein said junction is heated to 95° C. to provide a hot start;
c) reacting the contents of the droplets for at least twenty heating and cooling cycles to permit the PCR amplification of the target DNA molecules;
d) flowing a second sample fluid comprising shrimp alkaline phosphatase and exonuclease I through a second inlet channel which is in fluid communication with said main channel at a junction, such that said second sample fluid forms a plurality of highly uniform, monodisperse droplets in said continuous phase;
e) coalescing the droplets comprising the amplified DNA molecules from step (c) with the droplets comprising the shrimp alkaline phosphatase and exonuclease I from step (d) within an electric field and reacting the contents of the combined droplets at 37° C.;
f) inactivating the enzymes within the droplets to terminate the reaction from step (e) by heating the reacted droplets to 95° C.;
g) flowing a third sample fluid comprising universal sequencing primers, labeled ddNTPs and buffer effective to permit nucleotide sequencing through a third inlet channel which is in fluid communication with said main channel at a junction, such that said third sample fluid forms a plurality of highly uniform, monodisperse droplets in said continuous phase;
h) coalescing the droplets comprising the amplified DNA molecules from step (f) with the droplets comprising the universal sequencing primers, labeled ddNTPs and sequencing buffer from step (g) within an electric field;
i) reacting the contents of the coalesced droplets for at least twenty heating and cooling cycles to permit the sequencing reaction to proceed; and
j) analyzing the sequencing reaction to determine the nucleic acid sequence of the target DNA.
116. A method of sequencing DNA comprising:
a) providing a microfluidic substrate comprising at least two inlet channels adapted to carry at least two dispersed phase sample fluids and at least one main channel adapted to carry at least one continuous phase fluid, wherein the main channel comprises a serpentine line with heating and cooling regions;
b) flowing a first sample fluid comprising one more target DNA molecules to be sequenced, primer pair sets, dNTPs, enzymes and buffer components effective to permit isothermal amplification through a first inlet channel which is in fluid communication with said main channel at a junction, such that said first sample fluid forms a plurality of highly uniform, monodisperse droplets in said continuous phase;
c) reacting the contents of the droplets to permit the isothermal amplification of the target DNA molecules;
d) flowing a second sample fluid comprising shrimp alkaline phosphatase and exonuclease I through a second inlet channel which is in fluid communication with said main channel at a junction, such that said second sample fluid forms a plurality of highly uniform, monodisperse droplets in said continuous phase;
e) coalescing the droplets comprising the amplified DNA molecules from step (c) with the droplets comprising the shrimp alkaline phosphatase and exonuclease I from step (d) within an electric field and reacting the contents of the combined droplets at 37° C.;
f) inactivating the enzymes within the droplets to terminate the reaction from step (e) by heating the reacted droplets to 95° C.;
g) flowing a third sample fluid comprising universal sequencing primers, labeled ddNTPs and buffer effective to permit nucleotide sequencing through a third inlet channel which is in fluid communication with said main channel at a junction, such that said third sample fluid forms a plurality of highly uniform, monodisperse droplets in said continuous phase;
h) coalescing the droplets comprising the amplified DNA molecules from step (f) with the droplets comprising the universal sequencing primers, labeled ddNTPs and sequencing buffer from step (g) within an electric field;
i) reacting the contents of the coalesced droplets for at least twenty heating and cooling cycles to permit the sequencing reaction to proceed; and
j) analyzing the sequencing reaction to determine the nucleic acid sequence of the target DNA.
117. The method of claims 111-116, wherein said inlet and main channels are coated with an anti-wetting or blocking agent for the dispersed phase.
118. The method of claim 117, wherein said channels are coated with a silica primer layer followed by a perfluoroalkylalkylsilane compound, an amorphous soluble perfluoropolymer, BSA, PEG-silane or fluorosilane.
119. The method of claim 118, wherein the channels are coated with a silica primer layer followed by a perfluoroalkylalkylsilane compound.
120. The method of claims 111-116, wherein each ddNTP is labeled with a unique label.
121. The method of claims 111-116, wherein said label is a protein, a DNA tag, a dye, a quantum dot or a radio frequency identification tag.
122. The method of claim 121, wherein said label can be detected by fluorescence polarization, fluorescence intensity, fluorescence lifetime, fluorescence energy transfer, pH, ionic content, temperature or combinations thereof.
123. The method of claims 111-116, wherein said continuous phase is a non-polar solvent.
124. The method of claims 111-116, wherein said continuous phase is a fluorocarbon oil.
125. The method of claims 111-116, wherein said continuous phase further comprises one or more additives.
126. The method of claim 125, wherein said additive is a fluorosurfactant.
127. The method of claim 126, wherein said fluorosurfactant is a perfluorinated polyether.
128. The methods of claims 115 and 116, wherein a portion of at least one of the primer pair sets is attached to a semi-solid substrate.
129. The method of claims 111-116, wherein the flow of said dispersed phase fluid and said continuous phase fluid is pressure driven.
130. The method of claims 102 and 114-116, wherein at least one coalescing step occurs within a coalescence module in fluid communication with the main channel on the microfluidic substrate.
131. The method of claim 130, wherein the coalescence module comprises one or more electrodes that generate an electric field.
132. The method of claim 130 wherein said coalescence module comprises an expanded portion of the main channel between the electrodes to bring successive droplets into proximity.
133. The method of claim 130 wherein said coalescence module comprises a narrowed portion of the main channel to center droplets within the main channel followed by an expanded portion of the main channel between the electrodes to bring successive droplets into proximity.
134. The method of claims 112 and 114-116, wherein the droplets have no charge.
135. The method of claims 115 and 116, wherein analysis of the sequence reaction within the droplets occurs within a detection module in fluid communication with the main channel on the microfluidic substrate.
136. The method of claim 135, wherein said detection module comprising a detection apparatus for analyzing the sequence reactor within the droplets.
137. The method of claim 136, wherein said detection apparatus comprises an optical or electrical detector.
138. The method of claims 111-116, wherein the junction between the inlet channel and main channel comprises a fluidic nozzle for flow focusing.
139. A method of detecting a single nucleotide polymorphism (SNP) comprising:
a) providing a microfluidic substrate comprising at least one inlet channel adapted to carry at least one dispersed phase sample fluid and at least one main channel adapted to carry at least one continuous phase fluid, wherein the main channel comprises a serpentine line with heating and cooling regions;
b) flowing a sample fluid comprising one more target DNA molecules to be analyzed, labeled PCR primer pair sets, dNTPs, enzymes and buffer components effective to permit PCR amplification through an inlet channel which is in fluid communication with said main channel at a junction, such that said sample fluid forms a plurality of highly uniform, monodisperse droplets in said continuous phase, wherein said junction is heated to 95° C. to provide a hot start;
c) reacting the contents of the droplets for at least twenty heating and cooling cycles to permit the PCR amplification of the target DNA molecules; and
d) analyzing the reaction to determine the presence of absence of a SNP.
140. The method of claim 139, wherein said inlet and main channels are coated with an anti-wetting or blocking agent for the dispersed phase.
141. The method of claim 140, wherein said channels are coated with a silica primer layer followed by a perfluoroalkylalkylsilane compound, an amorphous soluble perfluoropolymer, BSA, PEG-silane or fluorosilane.
142. The method of claim 141, wherein the channels are coated with a silica primer layer followed by a perfluoroalkylalkylsilane compound.
143. The method of claim 139, wherein each primer is labeled with a unique label.
144. The method of claim 143, wherein said label is a protein, a DNA tag, a dye, a quantum dot or a radio frequency identification tag.
145. The method of claim 139, wherein said label can be detected by fluorescence polarization, fluorescence intensity, fluorescence lifetime, fluorescence energy transfer, pH, ionic content, temperature or combinations thereof.
146. The method of claim 139, wherein said continuous phase is a non-polar solvent.
147. The method of claim 139, wherein said continuous phase is a fluorocarbon oil.
148. The method of claim 139, wherein said continuous phase further comprises one or more additives.
149. The method of claim 148, wherein said additive is a fluorosurfactant.
150. The method of claim 149, wherein said fluorosurfactant is a perfluorinated polyether.
151. The methods of claim 139, wherein a portion of at least one of the primer pair sets is attached to a semi-solid substrate.
152. The method of claim 139, wherein the flow of said dispersed phase fluid and said continuous phase fluid is pressure driven.
153. The method of claim 139, wherein SNP analysis within the droplets occurs within a detection module in fluid communication with the main channel on the microfluidic substrate.
154. The method of claim 153, wherein said detection module comprising a detection apparatus for analyzing the sequence reactor within the droplets.
155. The method of claim 154, wherein said detection apparatus comprises an optical or electrical detector.
156. The method of claim 139, wherein the junction between the inlet channel and main channel comprises a fluidic nozzle for flow focusing.
157. A method of forming enzyme emulsions comprising:
a) providing a microfluidic substrate comprising at least one inlet channel adapted to carry at least one dispersed phase fluid and at least one main channel adapted to carry at least one continuous phase fluid;
b) flowing a sample fluid comprising at least one cell through an inlet channel which is in fluid communication with said main channel at a junction, such that said sample fluid forms a plurality of highly uniform, monodisperse droplets in said continuous phase, wherein said cells comprises at least one enzyme which can be secreted by said cells;
c) incubating the cells within the plurality of droplets such that the cells secrete at least one enzyme into the droplet,
wherein the cells within the droplets can be incubated within or outside of the microfluidic substrate.
158. A method of detecting enzyme activity comprising:
a) providing a microfluidic substrate comprising at least one inlet channel adapted to carry at least one dispersed phase sample fluid and at least one main channel adapted to carry at least one continuous phase fluid;
b) flowing a sample fluid comprising at least one cell and at least one labeled enzyme substrate through a inlet channel which is in fluid communication with said main channel at a junction, such that said sample fluid forms a plurality of highly uniform, monodisperse droplets in said continuous phase wherein said cells comprises at least one enzyme which can be secreted by said cells;
c) collecting the droplets and incubating them on the microfluidic substrate or off the microfluidic substrate at a temperature and duration appropriate to permit the enzyme/substrate reaction to occur;
d) reintroducing the droplets onto the microfluidic substrate if said droplets removed from the microfluidic substrate in step (c);
e) analyzing the contents of the coalesced droplets using the detection module to detect the presence or absence of an enzyme/substrate reaction; and,
f) selecting the droplets which contain the presence of an enzyme/substrate reaction.
159. A method of detecting enzyme activity comprising:
a) providing a microfluidic substrate comprising at least two inlet channels adapted to carry at least two dispersed phase sample fluids and at least one main channel adapted to carry at least one continuous phase fluid, wherein the main channel comprises a serpentine line with heating and cooling regions;
b) flowing a first sample fluid comprising at least one cell through a first inlet channel which is in fluid communication with said main channel at a junction, such that said first sample fluid forms a plurality of highly uniform, monodisperse droplets in said continuous phase, wherein said cells comprises at least one enzyme which can be secreted by said cells;
c) flowing a second sample fluid comprising at least one labeled enzyme substrate through a second inlet channel which is in fluid communication with said main channel at a junction, such that said second sample fluid forms a plurality of highly uniform, monodisperse droplets in said continuous phase;
d) coalescing the droplets comprising the secreted enzymes from step (b) with the droplets comprising the labeled substrate from step (c) within an electric field;
e) analyzing the contents of the coalesced droplets using a detection module to detect the presence or absence of an enzyme/substrate reaction; and,
f) selecting the coalesced droplets which contain the presence of an enzyme/substrate reaction.
160. The method of claim 159, further comprising incubating the cells within the plurality of droplets from step (b) such that the cells secrete at least one enzyme into the droplet, prior to flowing the second sample fluid in step (c).
161. The method of claim 160, wherein the cells are incubated within the plurality of droplets within the microfluidic substrate.
162. The method of claim 160, wherein the cells are incubated within the plurality of droplets outside the microfluidic substrate.
163. The method of claim 159, further comprising lysing the cells within the plurality of droplets from step (b) such that the cells release at least one enzyme into the plurality droplet, prior to flowing the second sample fluid in step (c).
164. The method of claim 159, further comprising selecting the coalesced droplets which contain the enzyme with the highest level of activity for the labeled substrate.
165. The method of claim 159, wherein said inlet and main channels are coated with an anti-wetting or blocking agent for the dispersed phase.
166. The method of claim 165, wherein said channels are coated with a silica primer layer followed by a perfluoroalkylalkylsilane compound, an amorphous soluble perfluoropolymer, BSA, PEG-silane or fluorosilane.
167. The method of claim 166, wherein the channels are coated with a silica primer layer followed by a perfluoroalkylalkylsilane compound.
168. The method of claim 159, wherein each enzyme substrate is labeled with a unique label.
169. The method of claim 159, wherein said label is a protein, a DNA tag, a dye, a quantum dot or a radio frequency identification tag.
170. The method of claim 159, wherein said label can be detected by fluorescence polarization, fluorescence intensity, fluorescence lifetime, fluorescence energy transfer, pH, ionic content, temperature or combinations thereof.
171. The method of claim 159, wherein said continuous phase is a non-polar solvent.
172. The method of claim 159, wherein said continuous phase is a fluorocarbon oil.
173. The method of claim 159, wherein said continuous phase further comprises one or more additives.
174. The method of claim 173, wherein said additive is a fluorosurfactant.
175. The method of claim 174, wherein said fluorosurfactant is a perfluorinated polyether.
176. The method of claim 159, wherein the flow of said dispersed phase fluid and said continuous phase fluid is pressure driven.
177. The method of claim 159, wherein said coalescing step occurs within a coalescence module in fluid communication with the main channel on the microfluidic substrate.
178. The method of claim 177, wherein the coalescence module comprises one or more electrodes that generate an electric field.
179. The method of claim 177 wherein said coalescence module comprises an expanded portion of the main channel between the electrodes to bring successive droplets into proximity.
180. The method of claim 177 wherein said coalescence module comprises a narrowed portion of the main channel to center droplets within the main channel followed by a expanded portion of the main channel between the electrodes to bring successive droplets into proximity.
181. The method of claim 159, wherein the droplets have no charge.
182. The method of claim 159, wherein analysis of the enzyme/substrate reaction within the droplets occurs within a detection module in fluid communication with the main channel on the microfluidic substrate.
183. The method of claim 182, wherein said detection module comprising a detection apparatus for analyzing the sequence reactor within the droplets.
184. The method of claim 183, wherein said detection apparatus comprises an optical or electrical detector.
185. The method of claim 159, wherein the junction between the inlet channel and main channel comprises a fluidic nozzle for flow focusing.
186. A method of detecting an aqueous solution comprising introducing a label into the aqueous solution such that said label alters a physical property of said aqueous solution thereby permitting the detection of aqueous solution comprising said label.
187. A method of tracking an aqueous solution comprising introducing a label into the aqueous solution such that said label alters a physical property of said aqueous solution thereby permitting the tracking of aqueous solution comprising said label.
188. The method of claims 186 and 187, wherein said label is a change in viscosity, a change in opacity, a change in volume, a change in density, a change in pH, a change in temperature, a change in dielectric constant, a change in conductivity, a change in the amount of beads present in the solution, a change in the amount of flocculent in the solution, a change in the amount of a selected solvent within the solution or the change in the amount of any measurable entity within the solution, or combinations thereof.
189. The method of claims 186 and 187, wherein said label is an inducible label.
190. The method of claim 189, wherein said inducible label is detectable when combined with at least one additional solution wherein said solution contains at least one entity to which can alter a property of the label such that the aqueous solution comprising said label is detectable.
191. The method of claims 186 and 187, wherein said labeled aqueous solution is a labeled aqueous droplet.
192. The method of claim 191, wherein said labeled aqueous droplet can be detected within a microfluidic substrate.
193. The method of claims 186 and 187, wherein said labels alter the light scattering properties of the aqueous solution.
194. The method of claims 186 and 187, wherein said aqueous solution further comprises a biological/chemical material.
195. The method of claim 194, wherein said biological/chemical material is selected from the group consisting of tissues, cells, particles, proteins, antibodies, amino acids, nucleotides, small molecules, and pharmaceuticals.
196. The method of claim 194, wherein said biological/chemical material comprises a label.
197. The method of claim 196, wherein each biological/chemical material is labeled with a unique label.
198. The method of claim 196, wherein said label is a protein, a DNA tag, a dye, a quantum dot or a radio frequency identification tag.
199. The method of claim 196, wherein said label can be detected by fluorescence polarization, fluorescence intensity, fluorescence lifetime, fluorescence energy transfer, pH, ionic content, temperature or combinations thereof.
US11/803,101 2006-05-11 2007-05-11 Microfluidic devices and methods of use thereof Abandoned US20080014589A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/803,101 US20080014589A1 (en) 2006-05-11 2007-05-11 Microfluidic devices and methods of use thereof

Applications Claiming Priority (27)

Application Number Priority Date Filing Date Title
US79983406P 2006-05-11 2006-05-11
US79983306P 2006-05-11 2006-05-11
US80861406P 2006-05-25 2006-05-25
US81509706P 2006-06-19 2006-06-19
US81973406P 2006-07-07 2006-07-07
US81973306P 2006-07-07 2006-07-07
US83315106P 2006-07-24 2006-07-24
US83498706P 2006-07-31 2006-07-31
US83787106P 2006-08-14 2006-08-14
US83769506P 2006-08-14 2006-08-14
US84171606P 2006-09-01 2006-09-01
US84332706P 2006-09-08 2006-09-08
US84337406P 2006-09-08 2006-09-08
US85644006P 2006-11-03 2006-11-03
US85654006P 2006-11-03 2006-11-03
US85827806P 2006-11-08 2006-11-08
US85827906P 2006-11-08 2006-11-08
US86066506P 2006-11-22 2006-11-22
US87376606P 2006-12-08 2006-12-08
US87464006P 2006-12-12 2006-12-12
US87456106P 2006-12-12 2006-12-12
US87620906P 2006-12-20 2006-12-20
US89925807P 2007-02-02 2007-02-02
US90315307P 2007-02-23 2007-02-23
US90429307P 2007-02-28 2007-02-28
US92033707P 2007-03-26 2007-03-26
US11/803,101 US20080014589A1 (en) 2006-05-11 2007-05-11 Microfluidic devices and methods of use thereof

Publications (1)

Publication Number Publication Date
US20080014589A1 true US20080014589A1 (en) 2008-01-17

Family

ID=38529633

Family Applications (11)

Application Number Title Priority Date Filing Date
US11/803,101 Abandoned US20080014589A1 (en) 2006-05-11 2007-05-11 Microfluidic devices and methods of use thereof
US11/803,104 Abandoned US20080003142A1 (en) 2006-05-11 2007-05-11 Microfluidic devices
US13/629,227 Active US9273308B2 (en) 2006-05-11 2012-09-27 Selection of compartmentalized screening method
US13/772,002 Abandoned US20130183659A1 (en) 2006-05-11 2013-02-20 Microfluidic devices
US13/779,943 Active US9981230B2 (en) 2006-05-11 2013-02-28 Microfluidic devices
US15/480,739 Pending US20170304785A1 (en) 2006-05-11 2017-04-06 Microfluidic devices
US15/886,212 Active US10639597B2 (en) 2006-05-11 2018-02-01 Microfluidic devices
US15/996,246 Active US10625220B2 (en) 2006-05-11 2018-06-01 Microfluidic devices
US15/996,253 Active US11351510B2 (en) 2006-05-11 2018-06-01 Microfluidic devices
US15/996,236 Pending US20180280897A1 (en) 2006-05-11 2018-06-01 Microfluidic devices
US16/985,603 Pending US20200360876A1 (en) 2006-05-11 2020-08-05 Microfluidic devices

Family Applications After (10)

Application Number Title Priority Date Filing Date
US11/803,104 Abandoned US20080003142A1 (en) 2006-05-11 2007-05-11 Microfluidic devices
US13/629,227 Active US9273308B2 (en) 2006-05-11 2012-09-27 Selection of compartmentalized screening method
US13/772,002 Abandoned US20130183659A1 (en) 2006-05-11 2013-02-20 Microfluidic devices
US13/779,943 Active US9981230B2 (en) 2006-05-11 2013-02-28 Microfluidic devices
US15/480,739 Pending US20170304785A1 (en) 2006-05-11 2017-04-06 Microfluidic devices
US15/886,212 Active US10639597B2 (en) 2006-05-11 2018-02-01 Microfluidic devices
US15/996,246 Active US10625220B2 (en) 2006-05-11 2018-06-01 Microfluidic devices
US15/996,253 Active US11351510B2 (en) 2006-05-11 2018-06-01 Microfluidic devices
US15/996,236 Pending US20180280897A1 (en) 2006-05-11 2018-06-01 Microfluidic devices
US16/985,603 Pending US20200360876A1 (en) 2006-05-11 2020-08-05 Microfluidic devices

Country Status (5)

Country Link
US (11) US20080014589A1 (en)
EP (9) EP4190448A3 (en)
JP (5) JP2010506136A (en)
AT (1) ATE540750T1 (en)
WO (2) WO2007133710A2 (en)

Cited By (347)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070044824A1 (en) * 2005-09-01 2007-03-01 Scott William Capeci Processing system and method of processing
US20070092914A1 (en) * 2004-03-31 2007-04-26 Medical Research Council, Harvard University Compartmentalised screening by microfluidic control
US20090026082A1 (en) * 2006-12-14 2009-01-29 Ion Torrent Systems Incorporated Methods and apparatus for measuring analytes using large scale FET arrays
US20090060797A1 (en) * 2002-12-30 2009-03-05 The Regents Of The University Of California Fluid control structures in microfluidic devices
US20090098555A1 (en) * 2007-09-26 2009-04-16 President And Fellows Of Harvard College Methods and applications for stitched dna barcodes
US20090111149A1 (en) * 2007-10-25 2009-04-30 Canon U.S. Life Sciences, Inc. Method of reducing cross-contamination in continuous amplification reactions in a channel
US20090131543A1 (en) * 2005-03-04 2009-05-21 Weitz David A Method and Apparatus for Forming Multiple Emulsions
US20090197248A1 (en) * 2004-10-08 2009-08-06 President And Fellows Of Harvard College Vitro evolution in microfluidic systems
WO2009100028A1 (en) * 2008-02-01 2009-08-13 The Regents Of The University Of California Microfluidic imaging cytometry
US20090217742A1 (en) * 2008-03-03 2009-09-03 University Of Washington Droplet compartmentalization for chemical separation and on-line sampling
US20090253181A1 (en) * 2008-01-22 2009-10-08 Microchip Biotechnologies, Inc. Universal sample preparation system and use in an integrated analysis system
US20090282901A1 (en) * 2008-05-17 2009-11-19 William Keith Leonard Method of high throughput viscometry
US20100022414A1 (en) * 2008-07-18 2010-01-28 Raindance Technologies, Inc. Droplet Libraries
US20100041046A1 (en) * 2008-08-15 2010-02-18 University Of Washington Method and apparatus for the discretization and manipulation of sample volumes
US20100068723A1 (en) * 2004-09-15 2010-03-18 Stevan Bogdan Jovanovich Microfluidic devices
WO2010036352A1 (en) * 2008-09-23 2010-04-01 Quantalife, Inc Droplet-based assay system
US7696002B1 (en) * 2007-01-25 2010-04-13 Pacesette, Inc. Electrolytic capacitor case design with insert molded elastomer seals
WO2010042744A1 (en) * 2008-10-08 2010-04-15 Universite De Strasbourg Microfluidic devices for reliable on-chip incubation of droplets in delay lines
US20100136544A1 (en) * 2007-03-07 2010-06-03 Jeremy Agresti Assays and other reactions involving droplets
US20100165784A1 (en) * 2008-12-31 2010-07-01 Microchip Biotechnologies, Inc., A California Corporation Instrument with microfluidic chip
US20100163109A1 (en) * 2007-02-06 2010-07-01 Brandeis University Manipulation of fluids and reactions in microfluidic systems
US20100173394A1 (en) * 2008-09-23 2010-07-08 Colston Jr Billy Wayne Droplet-based assay system
US20100184020A1 (en) * 2007-12-27 2010-07-22 Lawrence Livermore National Security, Llc. Chip-Based Sequencing Nucleic Acids
US20100188073A1 (en) * 2006-12-14 2010-07-29 Ion Torrent Systems Incorporated Methods and apparatus for measuring analytes using large scale fet arrays
US20100210479A1 (en) * 2003-03-31 2010-08-19 Medical Research Council Method of synthesis and testing of cominatorial libraries using microcapsules
US20100227767A1 (en) * 2007-07-26 2010-09-09 Boedicker James Q Stochastic confinement to detect, manipulate, and utilize molecules and organisms
WO2010111231A1 (en) 2009-03-23 2010-09-30 Raindance Technologies, Inc. Manipulation of microfluidic droplets
US20100252118A1 (en) * 2007-04-19 2010-10-07 Seth Fraden Manipulation of fluids, fluid components and reactions in microfluidic systems
US20100285975A1 (en) * 2007-07-24 2010-11-11 The Regents Of The University Of California Microfabricated droplet generator for single molecule/cell genetic analysis in engineered monodispersed emulsions
US20100303687A1 (en) * 2009-06-02 2010-12-02 Integenx Inc. Fluidic devices with diaphragm valves
US20100310299A1 (en) * 2005-08-01 2010-12-09 Silverbrook Research Pty Ltd Electronic image-sensing pen with force sensor and removeable ink cartridge
WO2010147942A1 (en) * 2009-06-16 2010-12-23 Massachusetts Institute Of Technology Multiphase non-linear electrokinetic devices
US20110005932A1 (en) * 2009-06-05 2011-01-13 Integenx Inc. Universal sample preparation system and use in an integrated analysis system
WO2011011172A1 (en) * 2009-07-21 2011-01-27 IntegenX, Inc. Microfluidic devices and uses thereof
US20110020920A1 (en) * 2004-06-01 2011-01-27 The Regents Of The University Of California Microfabricated integrated dna analysis system
US20110039303A1 (en) * 2007-02-05 2011-02-17 Stevan Bogdan Jovanovich Microfluidic and nanofluidic devices, systems, and applications
US20110036411A1 (en) * 2009-08-12 2011-02-17 Caliper Life Sciences, Inc. Pinching channels for fractionation of fragmented samples
WO2011028539A1 (en) * 2009-09-02 2011-03-10 Quantalife, Inc. System for mixing fluids by coalescence of multiple emulsions
US20110076735A1 (en) * 2004-09-15 2011-03-31 Jovanovich Stevan B Microfluidic Devices
US20110126911A1 (en) * 2009-12-01 2011-06-02 IntegenX Inc., a California Corporation Composite Plastic Articles
WO2011066476A1 (en) * 2009-11-25 2011-06-03 Quantalife, Inc. Methods and compositions for detecting genetic material
WO2011079176A2 (en) 2009-12-23 2011-06-30 Raindance Technologies, Inc. Microfluidic systems and methods for reducing the exchange of molecules between droplets
WO2011100604A2 (en) 2010-02-12 2011-08-18 Raindance Technologies, Inc. Digital analyte analysis
US20110217736A1 (en) * 2010-03-02 2011-09-08 Quantalife, Inc. System for hot-start amplification via a multiple emulsion
US20110218123A1 (en) * 2008-09-19 2011-09-08 President And Fellows Of Harvard College Creation of libraries of droplets and related species
US20110229545A1 (en) * 2010-03-17 2011-09-22 President And Fellows Of Harvard College Melt emulsification
US20110229897A1 (en) * 2009-06-04 2011-09-22 Lockheed Martin Corporation Optical approach for microfluidic DNA electrophoresis detection
US20110281737A1 (en) * 2008-10-22 2011-11-17 Life Technologies Corporation Method and Apparatus for Rapid Nucleic Acid Sequencing
US20110311978A1 (en) * 2008-09-23 2011-12-22 Quantalife, Inc. System for detection of spaced droplets
WO2011143075A3 (en) * 2010-05-08 2011-12-29 Veridex, Llc A simple and affordable method for immuophenotyping using a microfluidic chip sample preparation with image cytometry
USRE43122E1 (en) 1999-11-26 2012-01-24 The Governors Of The University Of Alberta Apparatus and method for trapping bead based reagents within microfluidic analysis systems
WO2012013316A1 (en) * 2010-07-26 2012-02-02 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. Method and device for passively separating and sorting drops, in particular in a microfluidic system, by using non-optical markers for reactions within the drops
WO2012021838A1 (en) * 2010-08-13 2012-02-16 University Of Connecticut Co-flow microfluidic device for polymersome formation
US20120052560A1 (en) * 2010-08-31 2012-03-01 Canon U.S. Life Sciences, Inc. System and method for serial processing of multiple nucleic acid assays
WO2012030878A1 (en) * 2010-08-31 2012-03-08 Canon U.S. Life Sciences, Inc. Composition and method for in-system priming microfluidic devices
WO2012045012A2 (en) 2010-09-30 2012-04-05 Raindance Technologies, Inc. Sandwich assays in droplets
WO2012078710A1 (en) * 2010-12-07 2012-06-14 Gnubio, Inc. Nucleic acid target detection using a detector, a probe and an inhibitor
US8217433B1 (en) 2010-06-30 2012-07-10 Life Technologies Corporation One-transistor pixel array
CN102612652A (en) * 2009-08-06 2012-07-25 康奈尔大学 Device and method for molecular analysis
US20120190040A1 (en) * 2009-08-02 2012-07-26 Qvella Corporation Cell concentration, capture and lysis devices and methods of use thereof
WO2012109604A1 (en) 2011-02-11 2012-08-16 Raindance Technologies, Inc. Thermocycling device for nucleic acid amplification and methods of use
WO2012109600A2 (en) 2011-02-11 2012-08-16 Raindance Technologies, Inc. Methods for forming mixed droplets
US8263336B2 (en) 2009-05-29 2012-09-11 Life Technologies Corporation Methods and apparatus for measuring analytes
WO2012139038A1 (en) * 2011-04-08 2012-10-11 Stokes Bio Limited Systems and methods for continuous flow pcr systems
US8286665B2 (en) 2006-03-22 2012-10-16 The Regents Of The University Of California Multiplexed latching valves for microfluidic devices and processors
US20120264646A1 (en) * 2008-07-18 2012-10-18 Raindance Technologies, Inc. Enzyme quantification
US8349167B2 (en) 2006-12-14 2013-01-08 Life Technologies Corporation Methods and apparatus for detecting molecular interactions using FET arrays
WO2013019491A1 (en) * 2011-08-01 2013-02-07 Denovo Sciences Cell capture system and method of use
US20130052648A1 (en) * 2010-03-12 2013-02-28 The General Hospital Corporation Amplifying rare cell surface markers
US20130059761A1 (en) * 2010-01-07 2013-03-07 Gen 9, Inc. Assembly of High Fidelity Polynucleotides
JP2013508156A (en) * 2009-10-27 2013-03-07 プレジデント アンド フェロウズ オブ ハーバード カレッジ Droplet generation technology
CN102985552A (en) * 2009-11-25 2013-03-20 伯乐生命医学产品有限公司 Methods and compositions for detecting genetic material
US8470164B2 (en) 2008-06-25 2013-06-25 Life Technologies Corporation Methods and apparatus for measuring analytes using large scale FET arrays
US8512538B2 (en) 2010-05-28 2013-08-20 Integenx Inc. Capillary electrophoresis device
WO2013126741A1 (en) 2012-02-24 2013-08-29 Raindance Technologies, Inc. Labeling and sample preparation for sequencing
WO2013143733A1 (en) * 2012-03-27 2013-10-03 Asml Netherlands B.V. Fuel system for lithographic apparatus, euv source,lithographic apparatus and fuel filtering method
US8552771B1 (en) 2012-05-29 2013-10-08 Life Technologies Corporation System for reducing noise in a chemical sensor array
WO2013159117A1 (en) 2012-04-20 2013-10-24 SlipChip, LLC Fluidic devices and systems for sample preparation or autonomous analysis
WO2013165748A1 (en) 2012-04-30 2013-11-07 Raindance Technologies, Inc Digital analyte analysis
US20130295568A1 (en) * 2010-02-12 2013-11-07 Darren Roy Link Digital analyte analysis
US20130344534A1 (en) * 2012-06-22 2013-12-26 Wistron Corporation Biological Particle Analyzer and Method of Analyzing Biological Particles
US8633015B2 (en) 2008-09-23 2014-01-21 Bio-Rad Laboratories, Inc. Flow-based thermocycling system with thermoelectric cooler
US8653567B2 (en) 2010-07-03 2014-02-18 Life Technologies Corporation Chemically sensitive sensor with lightly doped drains
US8658430B2 (en) 2011-07-20 2014-02-25 Raindance Technologies, Inc. Manipulating droplet size
US8663920B2 (en) 2011-07-29 2014-03-04 Bio-Rad Laboratories, Inc. Library characterization by digital assay
US8673627B2 (en) 2009-05-29 2014-03-18 Life Technologies Corporation Apparatus and methods for performing electrochemical reactions
WO2014043388A1 (en) 2012-09-12 2014-03-20 Gnubio, Inc. Integrated microfluidic system, method and kit for performing assays
WO2014043140A1 (en) 2012-09-12 2014-03-20 The Regents Of The University Of California Accurate genome sequencing of single cells by single-stranded amplification and sequencing
US8685324B2 (en) 2010-09-24 2014-04-01 Life Technologies Corporation Matched pair transistor circuits
US8730479B2 (en) 2010-03-25 2014-05-20 Bio-Rad Laboratories, Inc. Detection system for droplet-based assays
US8747748B2 (en) 2012-01-19 2014-06-10 Life Technologies Corporation Chemical sensor with conductive cup-shaped sensor surface
US8763642B2 (en) 2010-08-20 2014-07-01 Integenx Inc. Microfluidic devices with mechanically-sealed diaphragm valves
US8776573B2 (en) 2009-05-29 2014-07-15 Life Technologies Corporation Methods and apparatus for measuring analytes
US8778609B1 (en) 2013-03-14 2014-07-15 Good Start Genetics, Inc. Methods for analyzing nucleic acids
WO2014117088A1 (en) 2013-01-25 2014-07-31 Gnubio, Inc. System and method for performing droplet inflation
US8812422B2 (en) 2012-04-09 2014-08-19 Good Start Genetics, Inc. Variant database
US8821798B2 (en) 2012-01-19 2014-09-02 Life Technologies Corporation Titanium nitride as sensing layer for microwell structure
WO2014145555A1 (en) * 2013-03-15 2014-09-18 Lariat Biosciences, Inc. Microfluidic methods for manipulating dna
US8841116B2 (en) 2006-10-25 2014-09-23 The Regents Of The University Of California Inline-injection microdevice and microfabricated integrated DNA analysis system using same
US8841217B1 (en) 2013-03-13 2014-09-23 Life Technologies Corporation Chemical sensor with protruded sensor surface
US8841071B2 (en) 2011-06-02 2014-09-23 Raindance Technologies, Inc. Sample multiplexing
US8858782B2 (en) 2010-06-30 2014-10-14 Life Technologies Corporation Ion-sensing charge-accumulation circuits and methods
WO2014172288A2 (en) 2013-04-19 2014-10-23 Raindance Technologies, Inc. Digital analyte analysis
WO2014182835A1 (en) * 2013-05-09 2014-11-13 Bio-Rad Laboratories, Inc. Magnetic immuno digital pcr assay
US8951939B2 (en) 2011-07-12 2015-02-10 Bio-Rad Laboratories, Inc. Digital assays with multiplexed detection of two or more targets in the same optical channel
US8961764B2 (en) 2010-10-15 2015-02-24 Lockheed Martin Corporation Micro fluidic optic design
US8963216B2 (en) 2013-03-13 2015-02-24 Life Technologies Corporation Chemical sensor with sidewall spacer sensor surface
US8962366B2 (en) 2013-01-28 2015-02-24 Life Technologies Corporation Self-aligned well structures for low-noise chemical sensors
US8969257B1 (en) * 2007-12-28 2015-03-03 Intermolecular, Inc. Combinatorial flow system and method
WO2015031528A1 (en) 2013-08-27 2015-03-05 Gnubio, Inc. Microfluidic devices and methods of their use
DE102013217959A1 (en) * 2013-09-09 2015-03-12 Efficient Robotics Gmbh Microfluidic analysis device and manufacturing process
US8986628B2 (en) 2002-06-28 2015-03-24 President And Fellows Of Harvard College Method and apparatus for fluid dispersion
WO2015048798A1 (en) 2013-09-30 2015-04-02 Gnubio, Inc. Microfluidic cartridge device and methods of use and assembly
US9012390B2 (en) 2006-08-07 2015-04-21 Raindance Technologies, Inc. Fluorocarbon emulsion stabilizing surfactants
US9038919B2 (en) 2003-04-10 2015-05-26 President And Fellows Of Harvard College Formation and control of fluidic species
WO2015081102A1 (en) 2013-11-27 2015-06-04 Gnubio, Inc. Microfluidic droplet packing
US9056299B2 (en) 2009-03-13 2015-06-16 President And Fellows Of Harvard College Scale-up of flow-focusing microfluidic devices
WO2015103367A1 (en) 2013-12-31 2015-07-09 Raindance Technologies, Inc. System and method for detection of rna species
US9080968B2 (en) 2013-01-04 2015-07-14 Life Technologies Corporation Methods and systems for point of use removal of sacrificial material
US9089844B2 (en) 2010-11-01 2015-07-28 Bio-Rad Laboratories, Inc. System for forming emulsions
US9109251B2 (en) 2004-06-25 2015-08-18 University Of Hawaii Ultrasensitive biosensors
US9116117B2 (en) 2013-03-15 2015-08-25 Life Technologies Corporation Chemical sensor with sidewall sensor surface
US9121058B2 (en) 2010-08-20 2015-09-01 Integenx Inc. Linear valve arrays
US9128044B2 (en) 2013-03-15 2015-09-08 Life Technologies Corporation Chemical sensors with consistent sensor surface areas
US9127312B2 (en) 2011-02-09 2015-09-08 Bio-Rad Laboratories, Inc. Analysis of nucleic acids
US9150852B2 (en) 2011-02-18 2015-10-06 Raindance Technologies, Inc. Compositions and methods for molecular labeling
US9174216B2 (en) 2013-03-13 2015-11-03 DeNovo Science, Inc. System for capturing and analyzing cells
WO2015173658A2 (en) 2014-05-14 2015-11-19 Mark Davis Microfluidic devices that include channels that are slidable relative to each other and methods of use thereof
US9216414B2 (en) 2009-11-25 2015-12-22 Gen9, Inc. Microfluidic devices and methods for gene synthesis
WO2015195698A1 (en) 2014-06-16 2015-12-23 Gnubio, Inc. Size alternating injection into drops to facilitate sorting
US9222128B2 (en) 2011-03-18 2015-12-29 Bio-Rad Laboratories, Inc. Multiplexed digital assays with combinatorial use of signals
US9228898B2 (en) 2011-03-31 2016-01-05 Gnubio, Inc. Scalable spectroscopic detection and measurement
US9228233B2 (en) 2011-10-17 2016-01-05 Good Start Genetics, Inc. Analysis methods
US9238206B2 (en) 2011-05-23 2016-01-19 President And Fellows Of Harvard College Control of emulsions, including multiple emulsions
US9273308B2 (en) 2006-05-11 2016-03-01 Raindance Technologies, Inc. Selection of compartmentalized screening method
WO2016040476A1 (en) 2014-09-09 2016-03-17 The Broad Institute, Inc. A droplet-based method and apparatus for composite single-cell nucleic acid analysis
US9328344B2 (en) 2006-01-11 2016-05-03 Raindance Technologies, Inc. Microfluidic devices and methods of use in the formation and control of nanoreactors
US9347059B2 (en) 2011-04-25 2016-05-24 Bio-Rad Laboratories, Inc. Methods and compositions for nucleic acid analysis
US9366632B2 (en) 2010-02-12 2016-06-14 Raindance Technologies, Inc. Digital analyte analysis
WO2016100977A1 (en) 2014-12-19 2016-06-23 The Broad Institute Inc. Methods for profiling the t-cel- receptor repertoire
US20160177375A1 (en) * 2012-08-13 2016-06-23 The Regents Of The University Of California Methods and systems for detecting biological components
US9388465B2 (en) 2013-02-08 2016-07-12 10X Genomics, Inc. Polynucleotide barcode generation
US9393560B2 (en) 2010-03-25 2016-07-19 Bio-Rad Laboratories, Inc. Droplet transport system for detection
US9399797B2 (en) 2010-02-12 2016-07-26 Raindance Technologies, Inc. Digital analyte analysis
US9399215B2 (en) 2012-04-13 2016-07-26 Bio-Rad Laboratories, Inc. Sample holder with a well having a wicking promoter
US9404864B2 (en) 2013-03-13 2016-08-02 Denovo Sciences, Inc. System for imaging captured cells
US9410201B2 (en) 2012-12-14 2016-08-09 10X Genomics, Inc. Methods and systems for processing polynucleotides
US9415392B2 (en) 2009-03-24 2016-08-16 The University Of Chicago Slip chip device and methods
US9417190B2 (en) 2008-09-23 2016-08-16 Bio-Rad Laboratories, Inc. Calibrations and controls for droplet-based assays
WO2016138488A2 (en) 2015-02-26 2016-09-01 The Broad Institute Inc. T cell balance gene expression, compositions of matters and methods of use thereof
US9447461B2 (en) 2009-03-24 2016-09-20 California Institute Of Technology Analysis devices, kits, and related methods for digital quantification of nucleic acids and other analytes
US9448172B2 (en) 2003-03-31 2016-09-20 Medical Research Council Selection by compartmentalised screening
US9464319B2 (en) 2009-03-24 2016-10-11 California Institute Of Technology Multivolume devices, kits and related methods for quantification of nucleic acids and other analytes
US9492797B2 (en) 2008-09-23 2016-11-15 Bio-Rad Laboratories, Inc. System for detection of spaced droplets
US9500664B2 (en) 2010-03-25 2016-11-22 Bio-Rad Laboratories, Inc. Droplet generation for droplet-based assays
WO2016205728A1 (en) 2015-06-17 2016-12-22 Massachusetts Institute Of Technology Crispr mediated recording of cellular events
WO2016207721A1 (en) 2015-06-25 2016-12-29 University Of Limerick Mechanical device for generating combinatorial library
US9535920B2 (en) 2013-06-03 2017-01-03 Good Start Genetics, Inc. Methods and systems for storing sequence read data
US9562837B2 (en) 2006-05-11 2017-02-07 Raindance Technologies, Inc. Systems for handling microfludic droplets
US9574245B2 (en) 2011-07-25 2017-02-21 Qvella Corporation Methods and devices for electrical sample preparation
US9598725B2 (en) 2010-03-02 2017-03-21 Bio-Rad Laboratories, Inc. Emulsion chemistry for encapsulated droplets
US9606102B2 (en) 2013-01-26 2017-03-28 Denovo Sciences, Inc. System and method for capturing and analyzing cells
US9618475B2 (en) 2010-09-15 2017-04-11 Life Technologies Corporation Methods and apparatus for measuring analytes
WO2017075294A1 (en) 2015-10-28 2017-05-04 The Board Institute Inc. Assays for massively combinatorial perturbation profiling and cellular circuit reconstruction
WO2017075295A1 (en) 2015-10-27 2017-05-04 Berkeley Lights, Inc. Microfluidic electrowetting device apparatus having a covalently bound hydrophobic surface
WO2017075549A1 (en) 2015-10-28 2017-05-04 The Broad Institute, Inc. High-throughput dynamic reagent delivery system
WO2017079507A1 (en) * 2015-11-06 2017-05-11 Regents Of The University Of Michigan Droplet-based microfluidic rheometer system
US9671363B2 (en) 2013-03-15 2017-06-06 Life Technologies Corporation Chemical sensor with consistent sensor surface areas
US20170167981A1 (en) * 2015-12-11 2017-06-15 International Business Machines Corporation Smartphone compatible on-chip biodetection using integrated optical component and microfluidic channel with nanopillar array
US9689024B2 (en) 2012-08-14 2017-06-27 10X Genomics, Inc. Methods for droplet-based sample preparation
US9694361B2 (en) 2014-04-10 2017-07-04 10X Genomics, Inc. Fluidic devices, systems, and methods for encapsulating and partitioning reagents, and applications of same
US9701998B2 (en) 2012-12-14 2017-07-11 10X Genomics, Inc. Methods and systems for processing polynucleotides
US9707562B2 (en) 2013-03-13 2017-07-18 Denovo Sciences, Inc. System for capturing and analyzing cells
WO2017124101A2 (en) 2016-01-15 2017-07-20 The Broad Institute Inc. Semi-permeable arrays for analyzing biological systems and methods of using same
US9726653B2 (en) 2014-03-26 2017-08-08 Hamilton Sundstrand Corporation Chemical detector
WO2017136751A1 (en) 2016-02-05 2017-08-10 The Broad Institute Inc. Multi-stage, multiplexed target isolation and processing from heterogeneous populations
US20170227440A1 (en) * 2014-08-11 2017-08-10 Sphere Fluidics Limited Droplet sorting
US20170227479A1 (en) * 2014-08-21 2017-08-10 Schlumberger Technology Corporation Measurement of liquid parameters using a microfluidic device
WO2017147196A1 (en) 2016-02-22 2017-08-31 Massachusetts Institute Of Technology Methods for identifying and modulating immune phenotypes
US9764322B2 (en) 2008-09-23 2017-09-19 Bio-Rad Laboratories, Inc. System for generating droplets with pressure monitoring
WO2017161325A1 (en) 2016-03-17 2017-09-21 Massachusetts Institute Of Technology Methods for identifying and modulating co-occurant cellular phenotypes
WO2017168332A1 (en) 2016-03-28 2017-10-05 Boreal Genomics, Inc. Linked duplex target capture
US9789482B2 (en) 2003-08-27 2017-10-17 President And Fellows Of Harvard College Methods of introducing a fluid into droplets
US9797010B2 (en) 2007-12-21 2017-10-24 President And Fellows Of Harvard College Systems and methods for nucleic acid sequencing
US9803237B2 (en) 2012-04-24 2017-10-31 California Institute Of Technology Slip-induced compartmentalization
US9808798B2 (en) 2012-04-20 2017-11-07 California Institute Of Technology Fluidic devices for biospecimen preservation
US20170326549A1 (en) * 2014-11-26 2017-11-16 Ronald Jones Automated microscopic cell analysis
US9824068B2 (en) 2013-12-16 2017-11-21 10X Genomics, Inc. Methods and apparatus for sorting data
US9823217B2 (en) 2013-03-15 2017-11-21 Life Technologies Corporation Chemical device with thin conductive element
US9835585B2 (en) 2013-03-15 2017-12-05 Life Technologies Corporation Chemical sensor with protruded sensor surface
US9841398B2 (en) 2013-01-08 2017-12-12 Life Technologies Corporation Methods for manufacturing well structures for low-noise chemical sensors
US9856535B2 (en) 2013-05-31 2018-01-02 Denovo Sciences, Inc. System for isolating cells
WO2018009766A1 (en) * 2016-07-08 2018-01-11 President And Fellows Of Harvard College Formation of colloids or gels within droplets
WO2018013426A2 (en) 2016-07-08 2018-01-18 California Institute Of Technology Methods and devices for performing flow-through capture of low-concentration analytes
US9910020B1 (en) 2005-03-30 2018-03-06 Copilot Ventures Fund Iii Llc Methods and articles for identifying objects using encapsulated perfluorocarbon tracers
WO2018051242A1 (en) * 2016-09-14 2018-03-22 Ecole Polytechnique Federale De Lausanne (Epfl) Device for high throughput single-cell studies
US9925504B2 (en) 2004-03-31 2018-03-27 President And Fellows Of Harvard College Compartmentalised combinatorial chemistry by microfluidic control
US20180094255A1 (en) * 2015-03-20 2018-04-05 Novozymes A/S Droplet-Based Selection By Injection
US9944998B2 (en) 2013-07-25 2018-04-17 Bio-Rad Laboratories, Inc. Genetic assays
US9951386B2 (en) 2014-06-26 2018-04-24 10X Genomics, Inc. Methods and systems for processing polynucleotides
US9970984B2 (en) 2011-12-01 2018-05-15 Life Technologies Corporation Method and apparatus for identifying defects in a chemical sensor array
US9975122B2 (en) 2014-11-05 2018-05-22 10X Genomics, Inc. Instrument systems for integrated sample processing
US9981273B2 (en) 2012-09-28 2018-05-29 The Board Of Trustees Of The Leland Stanford Junior University Negative dielectrophoresis for selective elution of immuno-bound particles
US10000799B2 (en) 2014-11-04 2018-06-19 Boreal Genomics, Inc. Methods of sequencing with linked fragments
US10011872B1 (en) 2016-12-22 2018-07-03 10X Genomics, Inc. Methods and systems for processing polynucleotides
EP3351302A1 (en) 2017-01-18 2018-07-25 Biomillenia SAS Microfluidic system and method with tightly controlled incubation time and conditions
US10066259B2 (en) 2015-01-06 2018-09-04 Good Start Genetics, Inc. Screening for structural variants
US20180251771A1 (en) * 2011-02-16 2018-09-06 Glycosyn LLC Biosynthesis of human milk oligosaccharides in engineered bacteria
US10077472B2 (en) 2014-12-18 2018-09-18 Life Technologies Corporation High data rate integrated circuit with power management
WO2018170515A1 (en) 2017-03-17 2018-09-20 The Broad Institute, Inc. Methods for identifying and modulating co-occurant cellular phenotypes
US10100357B2 (en) 2013-05-09 2018-10-16 Life Technologies Corporation Windowed sequencing
WO2018195019A1 (en) 2017-04-18 2018-10-25 The Broad Institute Inc. Compositions for detecting secretion and methods of use
WO2018200872A1 (en) * 2017-04-26 2018-11-01 Berkeley Lights, Inc. Biological process systems and methods using microfluidic apparatus having an optimized electrowetting surface
US10191071B2 (en) 2013-11-18 2019-01-29 IntegenX, Inc. Cartridges and instruments for sample analysis
US10195571B2 (en) 2011-07-06 2019-02-05 President And Fellows Of Harvard College Multiple emulsions and techniques for the formation of multiple emulsions
US10196700B2 (en) 2009-03-24 2019-02-05 University Of Chicago Multivolume devices, kits and related methods for quantification and detection of nucleic acids and other analytes
US10207240B2 (en) 2009-11-03 2019-02-19 Gen9, Inc. Methods and microfluidic devices for the manipulation of droplets in high fidelity polynucleotide assembly
US10208332B2 (en) 2014-05-21 2019-02-19 Integenx Inc. Fluidic cartridge with valve mechanism
US10221436B2 (en) 2015-01-12 2019-03-05 10X Genomics, Inc. Processes and systems for preparation of nucleic acid sequencing libraries and libraries prepared using same
US10221442B2 (en) 2012-08-14 2019-03-05 10X Genomics, Inc. Compositions and methods for sample processing
US10227635B2 (en) 2012-04-16 2019-03-12 Molecular Loop Biosolutions, Llc Capture reactions
US10273541B2 (en) 2012-08-14 2019-04-30 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10287623B2 (en) 2014-10-29 2019-05-14 10X Genomics, Inc. Methods and compositions for targeted nucleic acid sequencing
EP3495817A1 (en) 2012-02-10 2019-06-12 Raindance Technologies, Inc. Molecular diagnostic screening assay
US10323279B2 (en) 2012-08-14 2019-06-18 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10351905B2 (en) 2010-02-12 2019-07-16 Bio-Rad Laboratories, Inc. Digital analyte analysis
EP3514246A1 (en) 2014-02-27 2019-07-24 The Broad Institute Inc. T cell balance gene expression and methods of use thereof
US10379079B2 (en) 2014-12-18 2019-08-13 Life Technologies Corporation Methods and apparatus for measuring analytes using large scale FET arrays
US10395758B2 (en) 2013-08-30 2019-08-27 10X Genomics, Inc. Sequencing methods
US10391490B2 (en) 2013-05-31 2019-08-27 Celsee Diagnostics, Inc. System and method for isolating and analyzing cells
US10400280B2 (en) 2012-08-14 2019-09-03 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10400235B2 (en) 2017-05-26 2019-09-03 10X Genomics, Inc. Single cell analysis of transposase accessible chromatin
US10429376B2 (en) * 2008-05-16 2019-10-01 Board Of Supervisors Of Louisiana State University And Agricultural And Mechanical College Microfluidic isolation of tumor cells or other rare cells from whole blood or other liquids
US10428326B2 (en) 2017-01-30 2019-10-01 10X Genomics, Inc. Methods and systems for droplet-based single cell barcoding
US10429399B2 (en) 2014-09-24 2019-10-01 Good Start Genetics, Inc. Process control for increased robustness of genetic assays
US10434507B2 (en) 2014-10-22 2019-10-08 The Regents Of The University Of California High definition microdroplet printer
US10451585B2 (en) 2009-05-29 2019-10-22 Life Technologies Corporation Methods and apparatus for measuring analytes
US10457977B2 (en) 2008-12-19 2019-10-29 President And Fellows Of Harvard College Particle-assisted nucleic acid sequencing
US10458942B2 (en) 2013-06-10 2019-10-29 Life Technologies Corporation Chemical sensor array having multiple sensors per well
US10466160B2 (en) 2011-08-01 2019-11-05 Celsee Diagnostics, Inc. System and method for retrieving and analyzing particles
US10471016B2 (en) 2013-11-08 2019-11-12 President And Fellows Of Harvard College Microparticles, methods for their preparation and use
GB201914537D0 (en) 2019-10-08 2019-11-20 Univ Southampton Transcript analysis
US10501739B2 (en) 2017-10-18 2019-12-10 Mission Bio, Inc. Method, systems and apparatus for single cell analysis
US10512910B2 (en) 2008-09-23 2019-12-24 Bio-Rad Laboratories, Inc. Droplet-based analysis method
US10520500B2 (en) 2009-10-09 2019-12-31 Abdeslam El Harrak Labelled silica-based nanomaterial with enhanced properties and uses thereof
US10525467B2 (en) 2011-10-21 2020-01-07 Integenx Inc. Sample preparation, processing and analysis systems
US10533221B2 (en) 2012-12-14 2020-01-14 10X Genomics, Inc. Methods and systems for processing polynucleotides
CN110740813A (en) * 2016-11-28 2020-01-31 亚利桑那州立大学董事会 Systems and methods involving continuous flow droplet reactions
US10550429B2 (en) 2016-12-22 2020-02-04 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10605767B2 (en) 2014-12-18 2020-03-31 Life Technologies Corporation High data rate integrated circuit with transmitter configuration
US10604799B2 (en) 2012-04-04 2020-03-31 Molecular Loop Biosolutions, Llc Sequence assembly
WO2020077236A1 (en) 2018-10-12 2020-04-16 The Broad Institute, Inc. Method for extracting nuclei or whole cells from formalin-fixed paraffin-embedded tissues
US10625259B1 (en) 2014-11-26 2020-04-21 Medica Corporation Automated microscopic cell analysis
US10632479B2 (en) 2015-05-22 2020-04-28 The Hong Kong University Of Science And Technology Droplet generator based on high aspect ratio induced droplet self-breakup
US10650912B2 (en) 2015-01-13 2020-05-12 10X Genomics, Inc. Systems and methods for visualizing structural variation and phasing information
US10647981B1 (en) 2015-09-08 2020-05-12 Bio-Rad Laboratories, Inc. Nucleic acid library generation methods and compositions
WO2020102608A2 (en) 2018-11-14 2020-05-22 President And Fellows Of Harvard College Multiplexing highly evolving viral variants with sherlock
WO2020102610A1 (en) 2018-11-14 2020-05-22 The Broad Institute, Inc. Crispr system based droplet diagnostic systems and methods
WO2020124050A1 (en) 2018-12-13 2020-06-18 The Broad Institute, Inc. Tiled assays using crispr-cas based detection
WO2020120442A2 (en) 2018-12-13 2020-06-18 Dna Script Direct oligonucleotide synthesis on cells and biomolecules
US10690627B2 (en) 2014-10-22 2020-06-23 IntegenX, Inc. Systems and methods for sample preparation, processing and analysis
WO2020131586A2 (en) 2018-12-17 2020-06-25 The Broad Institute, Inc. Methods for identifying neoantigens
US10697000B2 (en) 2015-02-24 2020-06-30 10X Genomics, Inc. Partition processing methods and systems
US10697007B2 (en) 2014-06-27 2020-06-30 The Regents Of The University Of California PCR-activated sorting (PAS)
US10704080B2 (en) 2014-06-11 2020-07-07 Samplix Aps Nucleotide sequence exclusion enrichment by droplet sorting (NEEDLS)
US10732649B2 (en) 2004-07-02 2020-08-04 The University Of Chicago Microfluidic system
US10745742B2 (en) 2017-11-15 2020-08-18 10X Genomics, Inc. Functionalized gel beads
US10752949B2 (en) 2012-08-14 2020-08-25 10X Genomics, Inc. Methods and systems for processing polynucleotides
EP3698871A1 (en) 2019-02-19 2020-08-26 Gottfried Wilhelm Leibniz Universität Hannover Laser based sorting of droplets in microfluidic streams
US10766033B2 (en) 2015-12-30 2020-09-08 Berkeley Lights, Inc. Droplet generation in a microfluidic device having an optoelectrowetting configuration
US10774370B2 (en) 2015-12-04 2020-09-15 10X Genomics, Inc. Methods and compositions for nucleic acid analysis
US10794925B2 (en) 2015-07-07 2020-10-06 University Of Washington Systems, methods, and devices for self-digitization of samples
US10801070B2 (en) 2013-11-25 2020-10-13 The Broad Institute, Inc. Compositions and methods for diagnosing, evaluating and treating cancer
US10815525B2 (en) 2016-12-22 2020-10-27 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10821440B2 (en) 2017-08-29 2020-11-03 Bio-Rad Laboratories, Inc. System and method for isolating and analyzing cells
WO2020223578A1 (en) * 2019-04-30 2020-11-05 Northeastern University Microfluidic chip for single cell pairing
US10829815B2 (en) 2017-11-17 2020-11-10 10X Genomics, Inc. Methods and systems for associating physical and genetic properties of biological particles
US10839939B2 (en) 2014-06-26 2020-11-17 10X Genomics, Inc. Processes and systems for nucleic acid sequence assembly
US10835585B2 (en) 2015-05-20 2020-11-17 The Broad Institute, Inc. Shared neoantigens
US10851414B2 (en) 2013-10-18 2020-12-01 Good Start Genetics, Inc. Methods for determining carrier status
US10854315B2 (en) 2015-02-09 2020-12-01 10X Genomics, Inc. Systems and methods for determining structural variation and phasing using variant call data
US10865440B2 (en) 2011-10-21 2020-12-15 IntegenX, Inc. Sample preparation, processing and analysis systems
US10874997B2 (en) 2009-09-02 2020-12-29 President And Fellows Of Harvard College Multiple emulsions created using jetting and other techniques
US10900032B2 (en) 2019-05-07 2021-01-26 Bio-Rad Laboratories, Inc. System and method for automated single cell processing
US10947581B2 (en) 2019-04-16 2021-03-16 Bio-Rad Laboratories, Inc. System and method for leakage control in a particle capture system
US10961573B2 (en) 2016-03-28 2021-03-30 Boreal Genomics, Inc. Linked duplex target capture
US10975442B2 (en) 2014-12-19 2021-04-13 Massachusetts Institute Of Technology Molecular biomarkers for cancer immunotherapy
US11041203B2 (en) 2013-10-18 2021-06-22 Molecular Loop Biosolutions, Inc. Methods for assessing a genomic region of a subject
US11041851B2 (en) 2010-12-23 2021-06-22 Molecular Loop Biosciences, Inc. Methods for maintaining the integrity and identification of a nucleic acid template in a multiplex sequencing reaction
US11047845B1 (en) 2017-11-15 2021-06-29 Medica Corporation Control material and methods for cell analyzers
US11045805B2 (en) 2017-11-01 2021-06-29 Bio-Rad Laboratories, Inc. Microfluidic system and method for arranging objects
US11053548B2 (en) 2014-05-12 2021-07-06 Good Start Genetics, Inc. Methods for detecting aneuploidy
US11072816B2 (en) 2017-05-03 2021-07-27 The Broad Institute, Inc. Single-cell proteomic assay using aptamers
US11081208B2 (en) 2016-02-11 2021-08-03 10X Genomics, Inc. Systems, methods, and media for de novo assembly of whole genome sequence data
US11084036B2 (en) 2016-05-13 2021-08-10 10X Genomics, Inc. Microfluidic systems and methods of use
US11092607B2 (en) 2015-10-28 2021-08-17 The Board Institute, Inc. Multiplex analysis of single cell constituents
US20210268497A1 (en) * 2018-07-29 2021-09-02 Koc Universitesi Microfluidic thromboelastometry instrument
US11110458B2 (en) 2013-02-01 2021-09-07 Bio-Rad Laboratories, Inc. System for detection of spaced droplets
US11111519B2 (en) 2015-02-04 2021-09-07 The Regents Of The University Of California Sequencing of nucleic acids via barcoding in discrete entities
US11124830B2 (en) 2016-12-21 2021-09-21 The Regents Of The University Of California Single cell genomic sequencing using hydrogel based droplets
US11123740B2 (en) 2015-06-29 2021-09-21 Arizona Board Of Regents On Behalf Of Arizona State University Systems and methods for continuous flow digital droplet polymerase chain reaction bioanalysis
US11123297B2 (en) 2015-10-13 2021-09-21 President And Fellows Of Harvard College Systems and methods for making and using gel microspheres
US11130128B2 (en) * 2008-09-23 2021-09-28 Bio-Rad Laboratories, Inc. Detection method for a target nucleic acid
US20210299619A1 (en) * 2009-11-12 2021-09-30 Stokes Bio Limited Methods of releasing and analyzing cellular components
US11142791B2 (en) 2016-08-10 2021-10-12 The Regents Of The University Of California Combined multiple-displacement amplification and PCR in an emulsion microdroplet
US11155881B2 (en) 2018-04-06 2021-10-26 10X Genomics, Inc. Systems and methods for quality control in single cell processing
US11174509B2 (en) 2013-12-12 2021-11-16 Bio-Rad Laboratories, Inc. Distinguishing rare variations in a nucleic acid sequence from a sample
US11179696B2 (en) 2012-11-30 2021-11-23 The Broad Institute Inc. High-throughput dynamic reagent delivery system
US11219896B2 (en) 2013-06-25 2022-01-11 University Of Washington Through Its Center For Commercialization Self-digitization of sample volumes
US11231451B2 (en) 2010-06-30 2022-01-25 Life Technologies Corporation Methods and apparatus for testing ISFET arrays
US11268137B2 (en) 2016-12-09 2022-03-08 Boreal Genomics, Inc. Linked ligation
US11274343B2 (en) 2015-02-24 2022-03-15 10X Genomics, Inc. Methods and compositions for targeted nucleic acid sequence coverage
US11273439B2 (en) 2019-05-07 2022-03-15 Bio-Rad Laboratories, Inc. System and method for target material retrieval from microwells
US11307166B2 (en) 2010-07-01 2022-04-19 Life Technologies Corporation Column ADC
US11332736B2 (en) 2017-12-07 2022-05-17 The Broad Institute, Inc. Methods and compositions for multiplexing single cell and single nuclei sequencing
US11339430B2 (en) 2007-07-10 2022-05-24 Life Technologies Corporation Methods and apparatus for measuring analytes using large scale FET arrays
US11365441B2 (en) 2019-05-22 2022-06-21 Mission Bio, Inc. Method and apparatus for simultaneous targeted sequencing of DNA, RNA and protein
US11389779B2 (en) 2007-12-05 2022-07-19 Complete Genomics, Inc. Methods of preparing a library of nucleic acid fragments tagged with oligonucleotide bar code sequences
US11389799B2 (en) 2019-01-17 2022-07-19 The Regents Of The University Of Michigan Microfluidic device for size and deformability measurements and applications thereof
US11389800B2 (en) 2011-09-28 2022-07-19 President And Fellows Of Harvard College Systems and methods for droplet production and/or fluidic manipulation
US11408024B2 (en) 2014-09-10 2022-08-09 Molecular Loop Biosciences, Inc. Methods for selectively suppressing non-target sequences
US11414702B2 (en) 2005-06-15 2022-08-16 Complete Genomics, Inc. Nucleic acid analysis by random mixtures of non-overlapping fragments
US11452768B2 (en) 2013-12-20 2022-09-27 The Broad Institute, Inc. Combination therapy with neoantigen vaccine
US11473136B2 (en) 2019-01-03 2022-10-18 Ncan Genomics, Inc. Linked target capture
WO2022219121A1 (en) * 2021-04-15 2022-10-20 Screensys Gmbh Method for developing and/or reprogramming plant cellular objects
US11482052B2 (en) 2014-10-22 2022-10-25 IntegenX, Inc. Systems and methods for biometric data collections
US11480778B2 (en) 2014-11-26 2022-10-25 Medica Corporation Automated microscopic cell analysis
WO2022232050A1 (en) 2021-04-26 2022-11-03 The Broad Institute, Inc. Compositions and methods for characterizing polynucleotide sequence alterations
US11492579B2 (en) * 2017-04-14 2022-11-08 Advanced Solutions Life Sciences, Llc Vascularized in vitro arrays of living cells
US11499183B2 (en) 2017-06-28 2022-11-15 Bio-Rad Laboratories, Inc. System and method for droplet detection
US11504719B2 (en) 2020-03-12 2022-11-22 Bio-Rad Laboratories, Inc. System and method for receiving and delivering a fluid for sample processing
US11530440B2 (en) 2013-03-14 2022-12-20 The Broad Institute, Inc. Methods for quantitating DNA using digital multiple displacement amplification
US11549149B2 (en) 2017-01-24 2023-01-10 The Broad Institute, Inc. Compositions and methods for detecting a mutant variant of a polynucleotide
US11591637B2 (en) 2012-08-14 2023-02-28 10X Genomics, Inc. Compositions and methods for sample processing
US11629344B2 (en) 2014-06-26 2023-04-18 10X Genomics, Inc. Methods and systems for processing polynucleotides
US11667954B2 (en) 2019-07-01 2023-06-06 Mission Bio, Inc. Method and apparatus to normalize quantitative readouts in single-cell experiments
US11725237B2 (en) 2013-12-05 2023-08-15 The Broad Institute Inc. Polymorphic gene typing and somatic change detection using sequencing data
US11724256B2 (en) 2019-06-14 2023-08-15 Bio-Rad Laboratories, Inc. System and method for automated single cell processing and analyses
US11732257B2 (en) 2017-10-23 2023-08-22 Massachusetts Institute Of Technology Single cell sequencing libraries of genomic transcript regions of interest in proximity to barcodes, and genotyping of said libraries
US11773389B2 (en) 2017-05-26 2023-10-03 10X Genomics, Inc. Single cell analysis of transposase accessible chromatin
US11841371B2 (en) 2018-03-13 2023-12-12 The Broad Institute, Inc. Proteomics and spatial patterning using antenna networks
US11840730B1 (en) 2009-04-30 2023-12-12 Molecular Loop Biosciences, Inc. Methods and compositions for evaluating genetic markers
US11859239B2 (en) 2018-03-19 2024-01-02 Ricoh Company, Ltd. Nucleic acid sample-contained container, method and apparatus for producing nucleic acid sample-contained container, non-transitory recording medium storing program for producing nucleic acid sample-contained container, and nucleic acid sample
US11873483B2 (en) 2015-03-11 2024-01-16 The Broad Institute, Inc. Proteomic analysis with nucleic acid identifiers
US11901041B2 (en) 2013-10-04 2024-02-13 Bio-Rad Laboratories, Inc. Digital analysis of nucleic acid modification
US11898206B2 (en) 2017-05-19 2024-02-13 10X Genomics, Inc. Systems and methods for clonotype screening
US11927519B2 (en) 2023-04-26 2024-03-12 Slingshot Biosciences, Inc. Synthetic human cell mimic hydrogel particle for cytometric or coulter device

Families Citing this family (299)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1438385A1 (en) * 2001-10-25 2004-07-21 Bar-Ilan University Interactive transparent individual cells biochip processor
US11243494B2 (en) 2002-07-31 2022-02-08 Abs Global, Inc. Multiple laminar flow-based particle and cellular separation with laser steering
US7329545B2 (en) 2002-09-24 2008-02-12 Duke University Methods for sampling a liquid flow
US6911132B2 (en) 2002-09-24 2005-06-28 Duke University Apparatus for manipulating droplets by electrowetting-based techniques
IL154677A0 (en) * 2003-02-27 2003-09-17 Univ Bar Ilan A method and apparatus for manipulating an individual cell
FR2856498B1 (en) * 2003-06-19 2005-09-30 Goulven Jean Alain Vernois DISTRIBUTION UNDER CONTROLLED ATMOSPHERE
US9200245B2 (en) 2003-06-26 2015-12-01 Seng Enterprises Ltd. Multiwell plate
US8597597B2 (en) * 2003-06-26 2013-12-03 Seng Enterprises Ltd. Picoliter well holding device and method of making the same
US7888110B2 (en) * 2003-06-26 2011-02-15 Seng Enterprises Ltd. Pico liter well holding device and method of making the same
WO2005023427A1 (en) 2003-09-05 2005-03-17 Stokes Bio Limited A microfluidic analysis system
GB0329220D0 (en) * 2003-12-17 2004-01-21 Inverness Medical Switzerland System
WO2006003664A1 (en) * 2004-07-07 2006-01-12 Seng Enterprises Ltd. Method and device for identifying an image of a well in an image of a well-bearing component
EP1781404A2 (en) * 2004-08-25 2007-05-09 Seng Enterprises Limited Method and device for isolating cells
US8038964B2 (en) * 2005-01-25 2011-10-18 Seng Enterprises Ltd. Device for studying individual cells
EP1859330B1 (en) 2005-01-28 2012-07-04 Duke University Apparatuses and methods for manipulating droplets on a printed circuit board
US20070054119A1 (en) * 2005-03-04 2007-03-08 Piotr Garstecki Systems and methods of forming particles
US20070141555A1 (en) * 2005-10-11 2007-06-21 Mordechai Deutsch Current damper for the study of cells
WO2007052245A1 (en) * 2005-11-03 2007-05-10 Seng Enterprises Ltd. Method and device for studying floating, living cells
CA2640024A1 (en) * 2006-01-27 2007-08-09 President And Fellows Of Harvard College Fluidic droplet coalescence
WO2007091228A1 (en) 2006-02-07 2007-08-16 Stokes Bio Limited A liquid bridge and system
US9476856B2 (en) 2006-04-13 2016-10-25 Advanced Liquid Logic, Inc. Droplet-based affinity assays
US20140193807A1 (en) 2006-04-18 2014-07-10 Advanced Liquid Logic, Inc. Bead manipulation techniques
US8492168B2 (en) 2006-04-18 2013-07-23 Advanced Liquid Logic Inc. Droplet-based affinity assays
US8613889B2 (en) 2006-04-13 2013-12-24 Advanced Liquid Logic, Inc. Droplet-based washing
US8637317B2 (en) 2006-04-18 2014-01-28 Advanced Liquid Logic, Inc. Method of washing beads
US7816121B2 (en) 2006-04-18 2010-10-19 Advanced Liquid Logic, Inc. Droplet actuation system and method
US7439014B2 (en) 2006-04-18 2008-10-21 Advanced Liquid Logic, Inc. Droplet-based surface modification and washing
WO2007123908A2 (en) * 2006-04-18 2007-11-01 Advanced Liquid Logic, Inc. Droplet-based multiwell operations
US8980198B2 (en) * 2006-04-18 2015-03-17 Advanced Liquid Logic, Inc. Filler fluids for droplet operations
US8716015B2 (en) 2006-04-18 2014-05-06 Advanced Liquid Logic, Inc. Manipulation of cells on a droplet actuator
US8470606B2 (en) 2006-04-18 2013-06-25 Duke University Manipulation of beads in droplets and methods for splitting droplets
US7901947B2 (en) 2006-04-18 2011-03-08 Advanced Liquid Logic, Inc. Droplet-based particle sorting
US8658111B2 (en) 2006-04-18 2014-02-25 Advanced Liquid Logic, Inc. Droplet actuators, modified fluids and methods
US7815871B2 (en) 2006-04-18 2010-10-19 Advanced Liquid Logic, Inc. Droplet microactuator system
US7763471B2 (en) 2006-04-18 2010-07-27 Advanced Liquid Logic, Inc. Method of electrowetting droplet operations for protein crystallization
US10078078B2 (en) 2006-04-18 2018-09-18 Advanced Liquid Logic, Inc. Bead incubation and washing on a droplet actuator
US7727723B2 (en) 2006-04-18 2010-06-01 Advanced Liquid Logic, Inc. Droplet-based pyrosequencing
US8637324B2 (en) 2006-04-18 2014-01-28 Advanced Liquid Logic, Inc. Bead incubation and washing on a droplet actuator
US8809068B2 (en) 2006-04-18 2014-08-19 Advanced Liquid Logic, Inc. Manipulation of beads in droplets and methods for manipulating droplets
US8041463B2 (en) 2006-05-09 2011-10-18 Advanced Liquid Logic, Inc. Modular droplet actuator drive
US7939021B2 (en) 2007-05-09 2011-05-10 Advanced Liquid Logic, Inc. Droplet actuator analyzer with cartridge
US9675972B2 (en) 2006-05-09 2017-06-13 Advanced Liquid Logic, Inc. Method of concentrating beads in a droplet
US7822510B2 (en) 2006-05-09 2010-10-26 Advanced Liquid Logic, Inc. Systems, methods, and products for graphically illustrating and controlling a droplet actuator
JP4968896B2 (en) * 2006-09-27 2012-07-04 富士フイルム株式会社 Dispersion manufacturing apparatus and dispersion manufacturing method
WO2008038258A1 (en) * 2006-09-28 2008-04-03 Stokes Bio Limited Microfluidic connector
US20100297748A1 (en) * 2006-09-28 2010-11-25 Stokes Bio Limited Integrated fluidic circuits
GB0701641D0 (en) * 2007-01-29 2007-03-07 Iti Scotland Ltd Analyte manipulation and detection
US8753894B2 (en) * 2007-02-01 2014-06-17 Diagnostic Biosensors, Llc Integrated membrane sensor
KR101431778B1 (en) 2007-02-09 2014-08-20 어드밴스드 리퀴드 로직, 아이엔씨. Droplet actuator devices and methods employing magnetic beads
WO2008121342A2 (en) * 2007-03-28 2008-10-09 President And Fellows Of Harvard College Emulsions and techniques for formation
JP2008245612A (en) * 2007-03-30 2008-10-16 Hitachi Ltd Method and device for preparing sample
US8691164B2 (en) * 2007-04-20 2014-04-08 Celula, Inc. Cell sorting system and methods
US20080268440A1 (en) * 2007-04-26 2008-10-30 Liu Timothy Z Biomolecule immobilization on surface via hydrophobic interactions
CN103418295B (en) 2007-06-21 2015-11-18 简.探针公司 For the instruments and methods of the content of hybrid detection chamber
US20090042737A1 (en) * 2007-08-09 2009-02-12 Katz Andrew S Methods and Devices for Correlated, Multi-Parameter Single Cell Measurements and Recovery of Remnant Biological Material
US8268246B2 (en) 2007-08-09 2012-09-18 Advanced Liquid Logic Inc PCB droplet actuator fabrication
WO2009032863A2 (en) 2007-09-04 2009-03-12 Advanced Liquid Logic, Inc. Droplet actuator with improved top substrate
US9744513B2 (en) 2007-09-20 2017-08-29 Jean-Louis Viovy Encapsulation microfluidic device
GB0720202D0 (en) 2007-10-16 2007-11-28 Cambridge Entpr Ltd Microfluidic systems
US9267918B2 (en) 2007-10-16 2016-02-23 Cambridge Enterprise Limited Microfluidic systems
US9145540B1 (en) 2007-11-15 2015-09-29 Seng Enterprises Ltd. Device for the study of living cells
WO2009081409A2 (en) * 2007-12-26 2009-07-02 Seng Enterprises Ltd. Device for the study of living cells
KR20100100974A (en) 2007-12-23 2010-09-15 어드밴스드 리퀴드 로직, 아이엔씨. Droplet actuator configurations and methods of conducting droplet operations
US9409177B2 (en) * 2008-03-21 2016-08-09 Lawrence Livermore National Security, Llc Chip-based device for parallel sorting, amplification, detection, and identification of nucleic acid subsequences
US9664619B2 (en) 2008-04-28 2017-05-30 President And Fellows Of Harvard College Microfluidic device for storage and well-defined arrangement of droplets
US8852952B2 (en) 2008-05-03 2014-10-07 Advanced Liquid Logic, Inc. Method of loading a droplet actuator
US20100075436A1 (en) * 2008-05-06 2010-03-25 Urdea Michael S Methods for use with nanoreactors
WO2009149257A1 (en) 2008-06-04 2009-12-10 The University Of Chicago The chemistrode: a plug-based microfluidic device and method for stimulation and sampling with high temporal, spatial, and chemical resolution
DK2315841T3 (en) 2008-08-21 2012-11-19 Novozymes As Method of screening in a microfluidic device
DE102008039117B3 (en) 2008-08-21 2010-05-20 Institut für Bioprozess- und Analysenmesstechnik e.V. Arrangement and method for generating, manipulating and analyzing compartments
US8179032B2 (en) * 2008-09-23 2012-05-15 The Board Of Trustees Of The University Of Illinois Ellipsoidal microcavity plasma devices and powder blasting formation
GB0818609D0 (en) 2008-10-10 2008-11-19 Univ Hull apparatus and method
WO2010053796A2 (en) * 2008-10-28 2010-05-14 The Regents Of The University Of Colorado, A Body Corporate Microfluidic cell sorter utilizing broadband coherent anti-stokes raman scattering
US9057568B2 (en) 2008-12-16 2015-06-16 California Institute Of Technology Temperature control devices and methods
WO2010085658A1 (en) * 2009-01-23 2010-07-29 Drexel University Apparatus and methods for detecting inflammation using quantum dots
CA2758382C (en) * 2009-04-13 2018-01-02 University Of Washington Ensemble-decision aliquot ranking
JP5881936B2 (en) * 2009-04-20 2016-03-09 ソニー株式会社 Sample solution introduction kit and sample solution injector
DE102009040151B4 (en) * 2009-05-26 2013-09-12 Analytik Jena Ag Arrangement for the detection of chemiluminescence on gases
US8574835B2 (en) 2009-05-29 2013-11-05 Life Technologies Corporation Scaffolded nucleic acid polymer particles and methods of making and using
WO2010138187A1 (en) * 2009-05-29 2010-12-02 Ion Torrent Systems Incorporated Scaffolded nucleic acid polymer particles and methods of making and using
US9524369B2 (en) 2009-06-15 2016-12-20 Complete Genomics, Inc. Processing and analysis of complex nucleic acid sequence data
CN102459592B (en) * 2009-06-15 2017-04-05 考利达基因组股份有限公司 For the method and composition of long fragment read sequencing
EP2446278B1 (en) * 2009-06-26 2021-11-17 President and Fellows of Harvard College Fluid injection
FR2948024B1 (en) * 2009-07-17 2020-01-10 Centre National De La Recherche Scientifique - Cnrs - ULTRASOUND ACTIVABLE EMULSION AND MANUFACTURING METHOD THEREOF.
WO2011011823A1 (en) * 2009-07-29 2011-02-03 Pyrobett Pte Ltd Method and apparatus for conducting an assay
US8926065B2 (en) 2009-08-14 2015-01-06 Advanced Liquid Logic, Inc. Droplet actuator devices and methods
US8188438B2 (en) * 2009-10-20 2012-05-29 Diagnostics Chips, LLC Electrokinetic microfluidic flow cytometer apparatuses with differential resistive particle counting and optical sorting
EP2494353B1 (en) 2009-10-30 2017-09-27 Illumina Inc. An apparatus comprising a plurality of encoded microvessels and a plurality of compartments and a method of reading a plurality of encoded micro vessels
US9091649B2 (en) 2009-11-06 2015-07-28 Advanced Liquid Logic, Inc. Integrated droplet actuator for gel; electrophoresis and molecular analysis
JP5873023B2 (en) 2009-12-01 2016-03-01 オックスフォード ナノポール テクノロジーズ リミテッド Biochemical analyzer
EP2516669B1 (en) 2009-12-21 2016-10-12 Advanced Liquid Logic, Inc. Enzyme assays on a droplet actuator
FR2958186A1 (en) 2010-03-30 2011-10-07 Ecole Polytech DEVICE FOR FORMING DROPS IN A MICROFLUID CIRCUIT.
ITTO20100068U1 (en) * 2010-04-20 2011-10-21 Eltek Spa MICROFLUID AND / OR EQUIPMENT DEVICES FOR MICROFLUID DEVICES
AU2011305445B2 (en) 2010-09-24 2017-03-16 The Board Of Trustees Of The Leland Stanford Junior University Direct capture, amplification and sequencing of target DNA using immobilized primers
US8486703B2 (en) 2010-09-30 2013-07-16 Ut-Battelle, Llc Surface sampling concentration and reaction probe
EP2436444A1 (en) * 2010-10-01 2012-04-04 Centre National de la Recherche Scientifique (C.N.R.S.) Microfluidic device for production and collection of droplets of a fluid
US10908066B2 (en) 2010-11-16 2021-02-02 1087 Systems, Inc. Use of vibrational spectroscopy for microfluidic liquid measurement
DE102010054879B4 (en) 2010-12-17 2013-07-18 Institut für Bioprozess- und Analysenmesstechnik e.V. Arrangement and method for conditioning fluid compartments
EP2654939A2 (en) * 2010-12-21 2013-10-30 President and Fellows of Harvard College Spray drying techniques
US8968585B2 (en) * 2010-12-23 2015-03-03 California Institute Of Technology Methods of fabrication of cartridges for biological analysis
US9233369B2 (en) 2010-12-23 2016-01-12 California Institute Of Technology Fluidic devices and fabrication methods for microfluidics
US8871500B2 (en) 2011-01-21 2014-10-28 Innovative Micro Technology MEMS particle sorting actuator and method of manufacturing
WO2012109138A1 (en) * 2011-02-07 2012-08-16 President And Fellows Of Harvard College Systems and methods for splitting droplets
EP3333269B1 (en) 2011-03-31 2021-05-05 Dana-Farber Cancer Institute, Inc. Methods to enable multiplex cold-pcr
KR101356279B1 (en) 2011-04-21 2014-01-28 충북대학교 산학협력단 System and method for detecting interaction between biomolecules
US9188615B2 (en) 2011-05-09 2015-11-17 Advanced Liquid Logic, Inc. Microfluidic feedback using impedance detection
WO2012174142A1 (en) * 2011-06-17 2012-12-20 Life Technologies Corporation Flat-field imaging system and methods of use
BR112014000257A2 (en) 2011-07-06 2017-03-01 Advanced Liquid Logic Inc reagent storage in a drop actuator
WO2013009927A2 (en) 2011-07-11 2013-01-17 Advanced Liquid Logic, Inc. Droplet actuators and techniques for droplet-based assays
WO2013016413A2 (en) 2011-07-25 2013-01-31 Advanced Liquid Logic Inc Droplet actuator apparatus and system
US20150205272A1 (en) * 2011-08-05 2015-07-23 Advanced Liquid Logic, Inc. Droplet actuator with improved waste disposal capability
GB201209924D0 (en) * 2012-06-05 2012-07-18 European Molecular Biology Lab Embl Microfluids for metagenomics
GB201115895D0 (en) 2011-09-14 2011-10-26 Embl Microfluidic device
US10130949B2 (en) * 2011-09-19 2018-11-20 Sorbonne Universite, Centre National De La Recherche Scientifique Microfluidic system
EP2761191A4 (en) * 2011-09-30 2015-08-26 Univ California Devices and methods for programming fluid flow using sequenced microstructures
US9896725B2 (en) * 2011-10-04 2018-02-20 The Regents Of The University Of California Corporation Real-time, label-free detection of macromolecules in droplets based on electrical measurements
US10731199B2 (en) 2011-11-21 2020-08-04 Advanced Liquid Logic, Inc. Glucose-6-phosphate dehydrogenase assays
US10222391B2 (en) 2011-12-07 2019-03-05 The Johns Hopkins University System and method for screening a library of samples
US9518291B2 (en) 2011-12-23 2016-12-13 California Institute Of Technology Devices and methods for biological sample-to-answer and analysis
US8883088B2 (en) 2011-12-23 2014-11-11 California Institute Of Technology Sample preparation devices and systems
CA2863121A1 (en) 2011-12-30 2013-07-04 Abbott Molecular Inc. Microorganism nucleic acid purification from host samples
BR112014019323A8 (en) * 2012-02-08 2017-07-11 Harvard College DROPLET FORMATION USING FLUID DECOMPOSITION
US9555382B2 (en) 2012-02-16 2017-01-31 National Research Council Of Canada Centrifugal microfluidic mixing apparatus with deflection element, and method of mixing
JP6168140B2 (en) * 2012-04-19 2017-07-26 和光純薬工業株式会社 Method for sampling reaction products in real time
US20130292247A1 (en) * 2012-05-01 2013-11-07 Commissariat A L'energie Atomique Et Aux Ene Alt Method and microsystem for the determination of clausius-mossotti factors for colloidal particles
JP2015516163A (en) * 2012-05-08 2015-06-11 セレクタ,インク Clone analysis of functional genomic analysis and composition for performing the clone analysis
WO2013192351A1 (en) 2012-06-20 2013-12-27 Bio-Rad Laboratories, Inc. Stabilized droplets for calibration and testing
CN104603595B (en) 2012-06-27 2017-08-08 先进流体逻辑公司 Technology and droplet actuator design for reducing bubble formation
US9790546B2 (en) 2012-08-31 2017-10-17 Roche Molecular Systems, Inc. Microfluidic chip, device and system for the generation of aqueous droplets in emulsion oil for nucleic acid amplification
EP2703497B1 (en) * 2012-08-31 2016-06-22 Roche Diagniostics GmbH Microfluidic chip, device and system for the generation of aqueous droplets in emulsion oil for nucleic acid amplification
JP5480455B1 (en) * 2012-09-06 2014-04-23 古河電気工業株式会社 Specimen identification sorting apparatus and specimen identification sorting method
US10000802B2 (en) 2012-10-04 2018-06-19 Base4 Innovation Ltd Sequencing method
GB201217772D0 (en) * 2012-10-04 2012-11-14 Base4 Innovation Ltd Sequencing method
WO2014071253A1 (en) 2012-11-05 2014-05-08 California Institute Of Technology Instruments for biological sample-to-answer devices
US8963095B2 (en) 2012-11-27 2015-02-24 Diagnostic Chips, LLC Electrokinetic microfluidic flow cytometer apparatuses with differential resistive particle counting and optical sorting
EP2925446A1 (en) 2012-11-29 2015-10-07 Koninklijke Philips N.V. Cartridge for uptake and processing of a sample
GB201303507D0 (en) * 2013-02-27 2013-04-10 Hollfelder Florian Assays
US10119134B2 (en) 2013-03-15 2018-11-06 Abvitro Llc Single cell bar-coding for antibody discovery
AU2014232882B2 (en) * 2013-03-15 2018-03-22 Hycor Biomedical, Inc. Automated immunoanalyzer system for performing diagnostic assays for allergies and autoimmune diseases
US20160038942A1 (en) * 2013-03-16 2016-02-11 Leslie Don Roberts Self-contained modular analytical cartridge and programmable reagent delivery system
EP2981349A4 (en) * 2013-04-02 2016-11-16 Raindance Technologies Inc Systems and methods for handling microfluidic droplets
GB201306444D0 (en) * 2013-04-09 2013-05-22 Base4 Innovation Ltd Single nucleotide detection method
CN105431553B (en) 2013-05-29 2020-02-07 生物辐射实验室股份有限公司 Systems and methods for sequencing in emulsion-based microfluidics
EP3004323B1 (en) * 2013-05-31 2019-04-17 3i Diagnostics, Inc. Rapid microbial detection
GB201311679D0 (en) 2013-06-28 2013-08-14 Ibm Microfluidic chip with dielectrophoretic electrodes extending in hydrophilic flow path
EP3017308A4 (en) 2013-07-05 2017-04-26 University Of Washington Through Its Center For Commercialization Methods, compositions and systems for microfluidic assays
US8961904B2 (en) 2013-07-16 2015-02-24 Premium Genetics (Uk) Ltd. Microfluidic chip
GB201313583D0 (en) 2013-07-30 2013-09-11 Sphere Fluidics Ltd Emulsion
GB2516684A (en) 2013-07-30 2015-02-04 Sphere Fluidics Ltd Microfluidic devices and systems
CA2959978A1 (en) * 2013-09-24 2015-04-02 The Regents Of The University Of California Encapsulated sensors and sensing systems for bioassays and diagnostics and methods for making and using them
FR3011932B1 (en) * 2013-10-10 2017-03-24 Univ Strasbourg TIME-RESOLVED FLUORESCENCE MEASUREMENT DEVICE AND METHOD FOR HIGH-SPEED SAMPLE SCREENING
US10845333B2 (en) * 2013-10-25 2020-11-24 The Foundation For The Promotion Of Industrial Science DNA detection method
US11796449B2 (en) 2013-10-30 2023-10-24 Abs Global, Inc. Microfluidic system and method with focused energy apparatus
WO2015073744A1 (en) * 2013-11-13 2015-05-21 Zhang Huidan Ultrasensitive detection and characterization of clustered kras mutations using peptide nucleic acid clamp pcr in drop-based microfluidics
US20150138337A1 (en) * 2013-11-15 2015-05-21 Schlumberger Technology Corporation Imaging-Based Measurement Device
WO2015074898A1 (en) * 2013-11-19 2015-05-28 Qiagen Gmbh Method for generating emulsions
TWI526392B (en) 2014-01-21 2016-03-21 國立清華大學 Method for forming microfluidic structure
WO2015112985A1 (en) * 2014-01-24 2015-07-30 The Johns Hopkins University System and device for high throughput generation of combinatorial droplets and methods of use
SG11201606193RA (en) 2014-01-31 2016-08-30 Toppan Printing Co Ltd Biomolecule analysis kit and biomolecule analysis method
CA2952013A1 (en) * 2014-06-10 2015-12-17 University Of Southern California Usc Discrete elements for 3d microfluidics
US11065348B2 (en) 2014-06-19 2021-07-20 The Trustees Of The University Of Pennsylvania Apparatus and methods for making recombinant protein-stabilized monodisperse microbubbles
JP6556220B2 (en) * 2014-08-04 2019-08-07 ジェンセル バイオシステムズ リミテッド Handling of three-phase fluid
US10590483B2 (en) 2014-09-15 2020-03-17 Abvitro Llc High-throughput nucleotide library sequencing
AU2015326473B2 (en) * 2014-09-29 2020-07-23 Chipcare Corporation A device for optical detection of cells comprising a cartridge and fluidic chip and methods thereof
JP6385450B2 (en) * 2014-09-30 2018-09-05 国立研究開発法人科学技術振興機構 Bubble ejection tip, local ablation device, local ablation method, injection device, and injection method
US11369962B2 (en) * 2014-10-24 2022-06-28 National Technology & Engineering Solutions Of Sandia, Llc Method and device for tracking and manipulation of droplets
US10040067B2 (en) * 2014-11-10 2018-08-07 Leibniz-Institut für Naturstoff-Forschung und Infektionsbiologie—Hans-Knöll-Institut Device and method for extracting individual picoliter droplets from microfluidic emulsions for further analysis and scale-up
JP6742311B2 (en) 2014-11-14 2020-08-19 アテナ ダイアグナスティクス,インコーポレイテッド Method for detecting silent carrier genotype
US9636675B2 (en) 2014-11-26 2017-05-02 International Business Machines Corporation Pillar array structure with uniform and high aspect ratio nanometer gaps
AU2015352119A1 (en) 2014-11-28 2017-07-13 Chipcare Corporation Multiplex bead array assay
CN107615038B (en) * 2015-03-06 2021-01-29 美铝澳大利亚有限公司 Online sampling device
EP3271713B1 (en) * 2015-03-18 2021-05-05 The Broad Institute, Inc. Massively parallel on-chip coalescence of microemulsions
US20180071981A1 (en) 2015-03-31 2018-03-15 The Regents Of The University Of California System and method for tunable patterning and assembly of particles via acoustophoresis
KR102384741B1 (en) 2015-04-22 2022-04-07 버클리 라잇츠, 인크. Microfluidic Cell Culture
JP6713730B2 (en) * 2015-05-20 2020-06-24 シスメックス株式会社 Cell detection device and cell detection method
GB201509640D0 (en) * 2015-06-03 2015-07-15 Sphere Fluidics Ltd Systems and methods
JP6527406B2 (en) * 2015-07-01 2019-06-05 本田技研工業株式会社 Analytical battery and method of manufacturing the same
GB201516447D0 (en) 2015-09-16 2015-10-28 Sphere Fluidics Ltd Microfluidic structure
BR112018005937A2 (en) 2015-09-24 2019-05-21 Abvitro Llc affinity oligonucleotide conjugates and uses of these
CA2999886A1 (en) 2015-09-24 2017-03-30 Abvitro Llc Single amplicon activated exclusion pcr
AU2016326734B2 (en) 2015-09-25 2022-07-07 Abvitro Llc High throughput process for T cell receptor target identification of natively-paired T cell receptor sequences
WO2017061619A1 (en) * 2015-10-09 2017-04-13 シスメックス株式会社 Specimen treatment chip, specimen treatment device, and specimen treatment method
EP3360953A4 (en) * 2015-10-09 2019-05-15 Sysmex Corporation Analyte treatment chip, analyte treatment device, and analyte treatment method
US10799865B2 (en) 2015-10-27 2020-10-13 Berkeley Lights, Inc. Microfluidic apparatus having an optimized electrowetting surface and related systems and methods
CN108603191A (en) * 2015-11-04 2018-09-28 塞勒密斯株式会社 Molecular cloning extracts and verification method
WO2017087505A1 (en) * 2015-11-17 2017-05-26 The Regents Of The University Of Michigan Macromolecular structures and uses thereof
US11371094B2 (en) 2015-11-19 2022-06-28 10X Genomics, Inc. Systems and methods for nucleic acid processing using degenerate nucleotides
CN108700601A (en) * 2015-11-25 2018-10-23 斯佩克特拉迪尼有限责任公司 System and device for microfluidic cartridge
JP6703824B2 (en) * 2015-11-30 2020-06-03 シスメックス株式会社 Cell selection method, cell detection method, cell selection device, and cell detection device
US20170157605A1 (en) * 2015-12-04 2017-06-08 Justin Lock Methods and compositions for low volume liquid handling
US20200330982A1 (en) * 2016-03-17 2020-10-22 Amin KAZEMZADEH Long-term storage and proportional dispensing device
GB201604876D0 (en) * 2016-03-22 2016-05-04 Uni I Tromsø Norges Arktiske Uni Marine DNA polymerases
ES2786974T3 (en) 2016-04-07 2020-10-14 Illumina Inc Methods and systems for the construction of standard nucleic acid libraries
FR3050212B1 (en) * 2016-04-15 2020-09-25 Chu Montpellier TUMOR CELL DETECTION AND / OR CHARACTERIZATION METHOD AND ASSOCIATED APPARATUS
FR3050269B1 (en) * 2016-04-15 2018-05-11 Ecole Superieure De Physique Et De Chimie Industrielles De La Ville De Paris (Espci) METHOD FOR SELECTING AND RECOVERING PRODUCTS AND ASSOCIATED SYSTEM
WO2017192286A1 (en) 2016-05-02 2017-11-09 Purdue Research Foundation Systems and methods for producing a chemical product
SG10202008265XA (en) 2016-05-26 2020-09-29 Berkeley Lights Inc Covalently modified surfaces, kits, and methods of preparation and use
US10124331B2 (en) * 2016-07-15 2018-11-13 Board Of Regents, The University Of Texas System Optofluidic lasers with surface gain and methods of making and using the same
US11339434B2 (en) * 2016-07-29 2022-05-24 The Regents Of The University Of California Methods for determining gene functions
WO2018031304A1 (en) * 2016-08-08 2018-02-15 Tru Tag Technologies, Inc. Identification of a tagged liquid
KR101816288B1 (en) * 2016-08-11 2018-01-09 한국기초과학지원연구원 Method of pre-treating sample with super absorbent polymer and device therefor
US10981163B2 (en) * 2016-09-12 2021-04-20 Abs Global, Inc. Method and system for hydrophobic coating of microfluidic chips
US20180073062A1 (en) * 2016-09-15 2018-03-15 The University Of Chicago Compositions and methods for identifying endogenous dna-dna interactions
WO2018054975A1 (en) * 2016-09-20 2018-03-29 The European Molecular Biology Laboratory Microfluidic sorting devices and methods
AU2017332495A1 (en) 2016-09-24 2019-04-11 Abvitro Llc Affinity-oligonucleotide conjugates and uses thereof
US11383235B2 (en) 2016-10-07 2022-07-12 The Regents Of The University Of California Device and method for microscale chemical reactions
US11857967B2 (en) 2016-10-07 2024-01-02 University Of Maryland, College Park Integration of porous monolithic structures within microfluidic systems
FR3057356B1 (en) * 2016-10-11 2021-12-17 Commissariat Energie Atomique METHOD AND SYSTEM FOR CHARACTERIZING A FLUID
EP3315963A1 (en) * 2016-10-26 2018-05-02 Fuchs Petrolub SE Sample receiving element, analyses set and method for analyzing a liquid, in particular a cooling lubricant emulsion
JP2020503846A (en) * 2016-11-08 2020-02-06 ナンナ セラピューティクス リミテッドNanna Therapeutics Limited Untagged coded chemical library
JP6884562B2 (en) * 2016-11-30 2021-06-09 シスメックス株式会社 Specimen processing method and sample processing equipment
JP7252899B2 (en) * 2016-12-01 2023-04-05 ノベル マイクロデバイシズ,インク. Automated point-of-care device and method of use for complex sample processing
CN110099968A (en) * 2016-12-28 2019-08-06 生物辐射实验室股份有限公司 The surface nature of microfluidic device is modified
US10995333B2 (en) 2017-02-06 2021-05-04 10X Genomics, Inc. Systems and methods for nucleic acid preparation
JP7071056B2 (en) * 2017-02-27 2022-05-18 シスメックス株式会社 Liquid delivery method and liquid delivery device
WO2018183744A1 (en) 2017-03-29 2018-10-04 The Research Foundation For The State University Of New York Microfluidic device and methods
US10508976B1 (en) 2017-03-31 2019-12-17 Advanced Micro Instruments, Inc. Gas sampling device and method
US20210060560A1 (en) * 2017-04-05 2021-03-04 Indian Institute Of Technology Madras (Itt Madras) Microfacs for detection and isolation of target cells
CN110621782B (en) * 2017-05-02 2023-10-24 国立大学法人东京大学 Method for monitoring dynamic changes in cells or substances derived therefrom and cell classification method using same
WO2018208447A1 (en) * 2017-05-11 2018-11-15 Emd Millipore Corporation Mechanical method of maintaining narrow residence time distributions in continuous flow systems
EP3621714A4 (en) 2017-05-11 2021-01-20 EMD Millipore Corporation Method of maintaining narrow residence time distributions in continuous flow systems
US10544413B2 (en) * 2017-05-18 2020-01-28 10X Genomics, Inc. Methods and systems for sorting droplets and beads
CN117143960A (en) * 2017-05-18 2023-12-01 10X基因组学有限公司 Method and system for sorting droplets and beads
CA3064205A1 (en) 2017-05-26 2018-11-29 Abvitro Llc High-throughput polynucleotide library sequencing and transcriptome analysis
US10639607B2 (en) * 2017-06-16 2020-05-05 Matralix Pte Ltd Systems and methods for preparing wax and lipid particles
GB2566002B (en) * 2017-06-22 2019-10-23 Sphere Fluidics Ltd Droplet dispensing systems
US10545516B2 (en) * 2017-08-02 2020-01-28 Schneider Electric Systems Usa, Inc. Industrial process control transmitter for determining solution concentration
SG11202000907UA (en) * 2017-08-22 2020-02-27 Elegen Corp Positional tracking and encoding in microfluidic devices
US10610865B2 (en) 2017-08-22 2020-04-07 10X Genomics, Inc. Droplet forming devices and system with differential surface properties
EP3679370A1 (en) 2017-09-07 2020-07-15 Juno Therapeutics, Inc. Methods of identifying cellular attributes related to outcomes associated with cell therapy
US10837047B2 (en) 2017-10-04 2020-11-17 10X Genomics, Inc. Compositions, methods, and systems for bead formation using improved polymers
GB201717103D0 (en) * 2017-10-18 2017-11-29 Tech Partnership Plc Fluid ejector system
WO2019084043A1 (en) 2017-10-26 2019-05-02 10X Genomics, Inc. Methods and systems for nuclecic acid preparation and chromatin analysis
WO2019083852A1 (en) 2017-10-26 2019-05-02 10X Genomics, Inc. Microfluidic channel networks for partitioning
EP3700672B1 (en) 2017-10-27 2022-12-28 10X Genomics, Inc. Methods for sample preparation and analysis
WO2019089810A1 (en) * 2017-11-01 2019-05-09 University Of Connecticut Two-dimensional sheet stabilized emulsion based inks
CN109746058A (en) * 2017-11-06 2019-05-14 北京新羿生物科技有限公司 Microlayer model detection chip
JP7030361B2 (en) * 2017-11-06 2022-03-07 北京新▲い▼生物科技有限公司 Microdroplet generator
WO2019108851A1 (en) 2017-11-30 2019-06-06 10X Genomics, Inc. Systems and methods for nucleic acid preparation and analysis
WO2019110591A1 (en) * 2017-12-06 2019-06-13 Samplix Aps A microfluidic device and a method for provision of emulsion droplets
WO2019157529A1 (en) 2018-02-12 2019-08-15 10X Genomics, Inc. Methods characterizing multiple analytes from individual cells or cell populations
US11639928B2 (en) 2018-02-22 2023-05-02 10X Genomics, Inc. Methods and systems for characterizing analytes from individual cells or cell populations
CN112236218B (en) 2018-04-02 2022-04-26 滴管公司 System and method for continuous flow emulsion processing
WO2019194799A1 (en) * 2018-04-04 2019-10-10 Hewlett-Packard Development Company, L.P. Microfluidic cellular membrane modification devices
EP3796998A1 (en) 2018-05-23 2021-03-31 ABS Global, Inc. Systems and methods for particle focusing in microchannels
US11703427B2 (en) 2018-06-25 2023-07-18 10X Genomics, Inc. Methods and systems for cell and bead processing
US20200032335A1 (en) 2018-07-27 2020-01-30 10X Genomics, Inc. Systems and methods for metabolome analysis
KR102083845B1 (en) * 2018-07-31 2020-03-03 광주과학기술원 Blood diagnostic element
WO2020031048A1 (en) 2018-08-08 2020-02-13 Inivata Ltd. Method of sequencing using variable replicate multiplex pcr
CN110819702B (en) * 2018-08-13 2023-03-21 上海新微技术研发中心有限公司 Digital PCR system and digital PCR liquid drop forming method
JP2020031606A (en) * 2018-08-31 2020-03-05 シスメックス株式会社 Nucleic acid detection device and nucleic acid detection method
CN110964628A (en) * 2018-09-29 2020-04-07 青岛华大智造普惠科技有限公司 Micro-fluidic chip system and preparation method of single-cell micro-droplets
US11130120B2 (en) * 2018-10-01 2021-09-28 Lifeng XIAO Micro-pipette tip for forming micro-droplets
GB201817321D0 (en) 2018-10-24 2018-12-05 Nanna Therapeutics Ltd Microbeads for tagless encoded chemical library screening
US11512356B2 (en) 2018-11-08 2022-11-29 Tokitae Llc Systems and methods for particle multiplexing in droplets
KR102089342B1 (en) * 2018-11-13 2020-04-20 (주)아프로텍 Precipitation Device having Dielectrophoresis Particle Separating Module
US11459607B1 (en) 2018-12-10 2022-10-04 10X Genomics, Inc. Systems and methods for processing-nucleic acid molecules from a single cell using sequential co-partitioning and composite barcodes
EP3899025A4 (en) * 2018-12-20 2023-02-22 Alveo Technologies Inc. Methods and compositions for detection of amplification products
US20220126298A1 (en) * 2019-01-07 2022-04-28 Elegen Corporation Methods of using microfluidic positional encoding devices
US11845983B1 (en) 2019-01-09 2023-12-19 10X Genomics, Inc. Methods and systems for multiplexing of droplet based assays
US20220072550A1 (en) * 2019-02-01 2022-03-10 Hewlett-Packard Development Company, L.P. Cell marking systems
US11851683B1 (en) 2019-02-12 2023-12-26 10X Genomics, Inc. Methods and systems for selective analysis of cellular samples
SG11202108788TA (en) 2019-02-12 2021-09-29 10X Genomics Inc Methods for processing nucleic acid molecules
US11467153B2 (en) 2019-02-12 2022-10-11 10X Genomics, Inc. Methods for processing nucleic acid molecules
US11655499B1 (en) 2019-02-25 2023-05-23 10X Genomics, Inc. Detection of sequence elements in nucleic acid molecules
WO2020175381A1 (en) * 2019-02-27 2020-09-03 京セラ株式会社 Particle separation and measurement device, and particle separation and measurement apparatus
EP4245140A3 (en) 2019-04-18 2024-01-17 ABS Global, Inc. System and process for continuous addition of cryoprotectant
EP3960842A4 (en) * 2019-04-26 2023-01-18 bitBiome, Inc. Selective detection, counting, and genomic analysis of living bacterium-derived nucleic acid on single-organism basis
US20220072549A1 (en) * 2019-04-30 2022-03-10 Hewlett-Packard Development Company, L.P. Microfluidic concentrating particlizers
WO2020223555A1 (en) 2019-04-30 2020-11-05 Berkeley Lights, Inc. Methods for encapsulating and assaying cells
WO2020223734A1 (en) * 2019-05-02 2020-11-05 The Board Of Trustees Of The University Of Illinois Atomic-to-nanoscale matter emission/flow regulation devices and methods
WO2021026176A1 (en) * 2019-08-05 2021-02-11 Academia Sinica Micro-reactor and method of use
AU2020361681A1 (en) 2019-10-10 2022-05-05 1859, Inc. Methods and systems for microfluidic screening
CN112710585B (en) * 2019-10-25 2023-02-07 南京工程学院 Evaluation method for dynamic stability of on-site mixed emulsion explosive
US11857981B2 (en) 2019-12-23 2024-01-02 10X Genomics, Inc. Magnetic separator for an automated single cell sequencing system
CN114761538A (en) * 2019-12-06 2022-07-15 伯乐实验室有限公司 Sample processing barcoded bead compositions, methods, preparations, and systems
WO2021127576A1 (en) * 2019-12-20 2021-06-24 Berkeley Lights, Inc. Methods of penning micro-objects using positive dielectrophoresis
WO2021142133A1 (en) * 2020-01-07 2021-07-15 Elegen Corporation Dna assembly in microfluidics device having integrated solid-phase columns
US11628439B2 (en) 2020-01-13 2023-04-18 Abs Global, Inc. Single-sheath microfluidic chip
US20230058381A1 (en) * 2020-01-29 2023-02-23 Massachusetts Institute Of Technology Systems and methods for the injection of viscous fluids
USD989342S1 (en) * 2020-02-04 2023-06-13 Ut-Battelle, Llc Microfluidic polymer chip interface bracket
USD993443S1 (en) * 2020-02-04 2023-07-25 Ut-Battelle, Llc Microfluidic glass chip interface bracket
KR102479966B1 (en) * 2020-02-20 2022-12-23 서울대학교산학협력단 Method for manufacturing organic solvent-free liposome using micro-fluidic chip
KR102408166B1 (en) * 2020-04-02 2022-06-10 아주대학교산학협력단 Biosensing chip to analyze cell-derived materials during cell culture, and method for quantitative analysis of cell activity using the same
WO2021222531A1 (en) * 2020-04-29 2021-11-04 Georgia Tech Research Corporation Systems and methods for specimen processing and storage
GB202006525D0 (en) 2020-05-04 2020-06-17 Sphere Fluidics Ltd Surfactant
US11851700B1 (en) 2020-05-13 2023-12-26 10X Genomics, Inc. Methods, kits, and compositions for processing extracellular molecules
CN111624180B (en) * 2020-05-22 2021-06-22 西安交通大学 Freezing fluorescence microscopic imaging system and method for capturing moving macromolecule form in microchannel
WO2022006239A1 (en) * 2020-06-30 2022-01-06 Plexium, Inc. Fluidic device and method
US11684920B2 (en) 2020-07-07 2023-06-27 International Business Machines Corporation Electrical tracking of a multiphase microfluidic flow
US20230303996A1 (en) * 2020-08-28 2023-09-28 Hewlett-Packard Development Company, L.P. Barriers in microfluidic channels
DE112021000024T5 (en) * 2020-09-29 2022-05-19 Jiangsu University PROCESS FOR MAKING A MICROSTRUCTURE ARRANGEMENT SURFACE WITH AN INCLINED SMOOTH FLOOR SURFACE BASED ON THE AIR FORMING METHOD
CN112578111A (en) * 2020-12-25 2021-03-30 扬州初心科技服务有限公司 Food antibiotic wriggling allies oneself with immunity detector more
WO2022231587A1 (en) * 2021-04-28 2022-11-03 Hewlett-Packard Development Company, L.P. Vibration of microfluidic device
GB202109969D0 (en) * 2021-07-09 2021-08-25 Lightcast Discovery Ltd Improvements in or relating to a method or an apparatus for detecting an interaction between a biological entity and a molecule
WO2023039389A1 (en) * 2021-09-07 2023-03-16 University Of Florida Research Foundation, Incorporated Efficient high-throughput electroporation for ev and exosome cargo loading
WO2023147334A1 (en) * 2022-01-25 2023-08-03 Berkeley Lights, Inc. Methods of assaying micro-objects in a microfluidic device
US20230241604A1 (en) * 2022-01-31 2023-08-03 Tdk Corporation Methods and Devices for Measuring Particle Properties
WO2024020671A1 (en) * 2022-07-25 2024-02-01 Interface Fluidics Ltd. Microfluidic devices, microfluidic systems, and methods for assessing thermophysical properties of a fluid

Citations (83)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4279345A (en) * 1979-08-03 1981-07-21 Allred John C High speed particle sorter using a field emission electrode
US4508265A (en) * 1981-06-18 1985-04-02 Agency Of Industrial Science & Technology Method for spray combination of liquids and apparatus therefor
US4931225A (en) * 1987-12-30 1990-06-05 Union Carbide Industrial Gases Technology Corporation Method and apparatus for dispersing a gas into a liquid
US5204112A (en) * 1986-06-16 1993-04-20 The Liposome Company, Inc. Induction of asymmetry in vesicles
US5378957A (en) * 1989-11-17 1995-01-03 Charged Injection Corporation Methods and apparatus for dispersing a fluent material utilizing an electron beam
US5512131A (en) * 1993-10-04 1996-04-30 President And Fellows Of Harvard College Formation of microstamped patterns on surfaces and derivative articles
US5617997A (en) * 1994-06-13 1997-04-08 Praxair Technology, Inc. Narrow spray angle liquid fuel atomizers for combustion
US5733526A (en) * 1995-12-14 1998-03-31 Alliance Pharmaceutical Corp. Hydrocarbon oil/fluorochemical preparations and methods of use
US5762775A (en) * 1994-09-21 1998-06-09 Lockheed Martin Energy Systems, Inc. Method for electrically producing dispersions of a nonconductive fluid in a conductive medium
US5783431A (en) * 1996-04-24 1998-07-21 Chromaxome Corporation Methods for generating and screening novel metabolic pathways
US5858670A (en) * 1990-07-02 1999-01-12 The Arizona Board Of Regents Bio-oligomer libraries and a method of use thereof
US5904933A (en) * 1994-06-09 1999-05-18 Alliance Pharmaceutical Corp. Stable reverse and multiple fluorocarbon emulsions
US5935331A (en) * 1994-09-09 1999-08-10 Matsushita Electric Industrial Co., Ltd. Apparatus and method for forming films
US5942056A (en) * 1993-04-22 1999-08-24 Federalloy, Inc. Plumbing fixtures and fittings employing copper-bismuth casting alloys
US5942443A (en) * 1996-06-28 1999-08-24 Caliper Technologies Corporation High throughput screening assay systems in microscale fluidic devices
US6103537A (en) * 1997-10-02 2000-08-15 Aclara Biosciences, Inc. Capillary assays involving separation of free and bound species
US6174469B1 (en) * 1996-05-13 2001-01-16 Universidad De Sevilla Device and method for creating dry particles
US6187214B1 (en) * 1996-05-13 2001-02-13 Universidad De Seville Method and device for production of components for microfabrication
US6189803B1 (en) * 1996-05-13 2001-02-20 University Of Seville Fuel injection nozzle and method of use
US6197835B1 (en) * 1996-05-13 2001-03-06 Universidad De Sevilla Device and method for creating spherical particles of uniform size
US6196525B1 (en) * 1996-05-13 2001-03-06 Universidad De Sevilla Device and method for fluid aeration via gas forced through a liquid within an orifice of a pressure chamber
US6210896B1 (en) * 1998-08-13 2001-04-03 Us Genomics Molecular motors
US6221654B1 (en) * 1996-09-25 2001-04-24 California Institute Of Technology Method and apparatus for analysis and sorting of polynucleotides based on size
US6235383B1 (en) * 1997-01-24 2001-05-22 Samsung Corning Co., Ltd. Glass article having a durable water repellent surface
US6248378B1 (en) * 1998-12-16 2001-06-19 Universidad De Sevilla Enhanced food products
US6251661B1 (en) * 1997-05-14 2001-06-26 Morishita Jintan Co., Ltd. Seamless capsule for synthesizing biopolymer and method for producing the same
US6267858B1 (en) * 1996-06-28 2001-07-31 Caliper Technologies Corp. High throughput screening assay systems in microscale fluidic devices
US6268222B1 (en) * 1998-01-22 2001-07-31 Luminex Corporation Microparticles attached to nanoparticles labeled with flourescent dye
US20020004532A1 (en) * 2000-05-26 2002-01-10 Michelle Matathia Low emulsifier multiple emulsions
US20020005354A1 (en) * 1997-09-23 2002-01-17 California Institute Of Technology Microfabricated cell sorter
US20020008028A1 (en) * 2000-01-12 2002-01-24 Jacobson Stephen C. Microfluidic device and method for focusing, segmenting, and dispensing of a fluid stream
US20020022261A1 (en) * 1995-06-29 2002-02-21 Anderson Rolfe C. Miniaturized genetic analysis systems and methods
US6355198B1 (en) * 1996-03-15 2002-03-12 President And Fellows Of Harvard College Method of forming articles including waveguides via capillary micromolding and microtransfer molding
US20020036139A1 (en) * 1999-02-12 2002-03-28 Board Of Regents, The University Of Texas System Method and apparatus for programmable fluidic processing
US6386463B1 (en) * 1996-05-13 2002-05-14 Universidad De Sevilla Fuel injection nozzle and method of use
US20020058332A1 (en) * 2000-09-15 2002-05-16 California Institute Of Technology Microfabricated crossflow devices and methods
US6394429B2 (en) * 1996-05-13 2002-05-28 Universidad De Sevilla Device and method for fluid aeration via gas forced through a liquid within an orifice of a pressure chamber
US6405936B1 (en) * 1996-05-13 2002-06-18 Universidad De Sevilla Stabilized capillary microjet and devices and methods for producing same
US6408878B2 (en) * 1999-06-28 2002-06-25 California Institute Of Technology Microfabricated elastomeric valve and pump systems
US6432630B1 (en) * 1996-09-04 2002-08-13 Scandinanian Micro Biodevices A/S Micro-flow system for particle separation and analysis
US20020119459A1 (en) * 1999-01-07 2002-08-29 Andrew Griffiths Optical sorting method
US6506609B1 (en) * 1999-05-17 2003-01-14 Caliper Technologies Corp. Focusing of microparticles in microfluidic systems
US6508988B1 (en) * 2000-10-03 2003-01-21 California Institute Of Technology Combinatorial synthesis system
US20030015425A1 (en) * 2001-06-20 2003-01-23 Coventor Inc. Microfluidic system including a virtual wall fluid interface port for interfacing fluids with the microfluidic system
US20030029169A1 (en) * 2001-08-10 2003-02-13 Hanna William Thompson Integrated micro combined heat and power system
US6520425B1 (en) * 2001-08-21 2003-02-18 The University Of Akron Process and apparatus for the production of nanofibers
US6540895B1 (en) * 1997-09-23 2003-04-01 California Institute Of Technology Microfabricated cell sorter for chemical and biological materials
US6560030B2 (en) * 2000-08-16 2003-05-06 California Institute Of Technology Solid immersion lens structures and methods for producing solid immersion lens structures
US20030124586A1 (en) * 1997-07-07 2003-07-03 Andrew Griffiths In vitro sorting method
US6592821B1 (en) * 1999-05-17 2003-07-15 Caliper Technologies Corp. Focusing of microparticles in microfluidic systems
US20030148544A1 (en) * 2001-06-28 2003-08-07 Advanced Research And Technology Institute, Inc. Methods of preparing multicolor quantum dot tagged beads and conjugates thereof
US6610499B1 (en) * 2000-08-31 2003-08-26 The Regents Of The University Of California Capillary array and related methods
US6680178B2 (en) * 2000-06-02 2004-01-20 The Regents Of The University Of California Profiling of protease specificity using combinatorial fluorogenic substrate libraries
US20040068019A1 (en) * 2001-02-23 2004-04-08 Toshiro Higuchi Process for producing emulsion and microcapsules and apparatus therefor
US20040096515A1 (en) * 2001-12-07 2004-05-20 Bausch Andreas R. Methods and compositions for encapsulating active agents
US6752922B2 (en) * 2001-04-06 2004-06-22 Fluidigm Corporation Microfluidic chromatography
US6766817B2 (en) * 2001-07-25 2004-07-27 Tubarc Technologies, Llc Fluid conduction utilizing a reversible unsaturated siphon with tubarc porosity action
US20050032238A1 (en) * 2003-08-07 2005-02-10 Nanostream, Inc. Vented microfluidic separation devices and methods
US20050032240A1 (en) * 2003-02-11 2005-02-10 The Regents Of The University Of California Microfluidic devices for controlled viscous shearing and formation of amphiphilic vesicles
US20050084923A1 (en) * 2001-09-14 2005-04-21 Peter-Juergen Mueller Methods for cultivating and analyzing microbial individual cell cultures
US6890487B1 (en) * 1999-09-30 2005-05-10 Science & Technology Corporation ©UNM Flow cytometry for high throughput screening
US6926313B1 (en) * 2003-04-02 2005-08-09 Sandia National Laboratories High pressure capillary connector
US20050172476A1 (en) * 2002-06-28 2005-08-11 President And Fellows Of Havard College Method and apparatus for fluid dispersion
US20050183995A1 (en) * 2002-04-17 2005-08-25 Cytonome, Inc. Method and apparatus for sorting particles
US6935768B2 (en) * 2000-08-25 2005-08-30 Institut Fur Mikrotechnik Mainz Gmbh Method and statistical micromixer for mixing at least two liquids
US20060003442A1 (en) * 2004-06-30 2006-01-05 Gong Jian P Cell culture scaffold, methods of manufacturing the scaffold and cell culture methods
US20060051329A1 (en) * 2004-08-27 2006-03-09 The Regents Of The University Of California Microfluidic device for the encapsulation of cells with low and high cell densities
US20060078893A1 (en) * 2004-10-12 2006-04-13 Medical Research Council Compartmentalised combinatorial chemistry by microfluidic control
US20060078888A1 (en) * 2004-10-08 2006-04-13 Medical Research Council Harvard University In vitro evolution in microfluidic systems
US20060108012A1 (en) * 2002-11-14 2006-05-25 Barrow David A Microfluidic device and methods for construction and application
US7068874B2 (en) * 2000-11-28 2006-06-27 The Regents Of The University Of California Microfluidic sorting device
US20060147909A1 (en) * 2001-05-31 2006-07-06 Markus Rarbach Microstructures and use thereof for the directed evolution of biomolecules
US7081192B1 (en) * 2000-08-08 2006-07-25 Aviva Biosciences Corporation Methods for manipulating moieties in microfluidic systems
US20060163385A1 (en) * 2003-04-10 2006-07-27 Link Darren R Formation and control of fluidic species
US20070054119A1 (en) * 2005-03-04 2007-03-08 Piotr Garstecki Systems and methods of forming particles
US20070056853A1 (en) * 2005-09-15 2007-03-15 Lucnet Technologies Inc. Micro-chemical mixing
US7204431B2 (en) * 2003-10-31 2007-04-17 Agilent Technologies, Inc. Electrospray ion source for mass spectroscopy
US20070195127A1 (en) * 2006-01-27 2007-08-23 President And Fellows Of Harvard College Fluidic droplet coalescence
US20080003142A1 (en) * 2006-05-11 2008-01-03 Link Darren R Microfluidic devices
US20090012187A1 (en) * 2007-03-28 2009-01-08 President And Fellows Of Harvard College Emulsions and Techniques for Formation
US7514210B2 (en) * 2000-09-13 2009-04-07 Medical Research Council Compartmentalised self replication method for in vitro evolution of molecular libraries
US20090131543A1 (en) * 2005-03-04 2009-05-21 Weitz David A Method and Apparatus for Forming Multiple Emulsions
US7541383B2 (en) * 2002-12-20 2009-06-02 Amgen Inc. Asthma and allergic inflammation modulators

Family Cites Families (956)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2097692A (en) 1936-03-23 1937-11-02 Bohn Aluminium & Brass Corp Method and machine for forming bearing shells
US2164172A (en) 1938-04-30 1939-06-27 Gen Electric Liquid-dispensing apparatus
US2636855A (en) 1948-03-25 1953-04-28 Hilger & Watts Ltd Method of producing photoconductive coatings
US2656508A (en) 1949-08-27 1953-10-20 Wallace H Coulter Means for counting particles suspended in a fluid
US2692800A (en) 1951-10-08 1954-10-26 Gen Electric Nozzle flow control
US2797149A (en) 1953-01-08 1957-06-25 Technicon International Ltd Methods of and apparatus for analyzing liquids containing crystalloid and non-crystalloid constituents
US2879141A (en) 1955-11-16 1959-03-24 Technicon Instr Automatic analyzing apparatus
US2971700A (en) 1957-07-22 1961-02-14 Vilbiss Co Apparatus for coating articles with chemically reactive liquids
GB1143839A (en) 1965-10-15
CH455414A (en) 1966-01-10 1968-07-15 Bachofen Willy A Installation element for optical flow control on pipelines
US3479141A (en) 1967-05-17 1969-11-18 Technicon Corp Method and apparatus for analysis
US3980541A (en) 1967-06-05 1976-09-14 Aine Harry E Electrode structures for electric treatment of fluids and filters using same
US3621059A (en) 1969-07-30 1971-11-16 Du Pont Amides of hexafluoropropylene oxide polymer acids and polyalklene oxide
US3784471A (en) 1970-05-11 1974-01-08 Avco Corp Solid additives dispersed in perfluorinated liquids with perfluoroalkyl ether dispersants
US3828085A (en) 1970-07-09 1974-08-06 Allied Chem Novel amidoamine oxides
DE2100685C2 (en) 1971-01-08 1983-09-22 Basf Ag, 6700 Ludwigshafen Process for the preparation of pure 4-amino-5-halogen-pyridazonen- (6)
US3698635A (en) 1971-02-22 1972-10-17 Ransburg Electro Coating Corp Spray charging device
US3816331A (en) 1972-07-05 1974-06-11 Ncr Continuous encapsulation and device therefor
US3832646A (en) 1972-10-06 1974-08-27 Westinghouse Electric Corp Common mode noise suppressing circuit adjustment sequence
CH563807A5 (en) 1973-02-14 1975-07-15 Battelle Memorial Institute Fine granules and microcapsules mfrd. from liquid droplets - partic. of high viscosity requiring forced sepn. of droplets
CH564966A5 (en) 1974-02-25 1975-08-15 Sauter Fr Ag Fabrik Elektrisch
US3930061A (en) 1974-04-08 1975-12-30 Ransburg Corp Electrostatic method for forming structures and articles
US4059552A (en) 1974-06-21 1977-11-22 The Dow Chemical Company Cross-linked water-swellable polymer particles
US3960187A (en) 1974-07-23 1976-06-01 Usm Corporation Method and device for metering and dispersing fluid materials
US3982541A (en) 1974-07-29 1976-09-28 Esperance Jr Francis A L Eye surgical instrument
DK150802C (en) 1974-09-16 1988-02-01 Bifok Ab METHOD AND APPARATUS FOR CONTINUOUS HIGH-SPEED ANALYSIS OF A LIQUID TEST IN A BEARING FLOW
US4098897A (en) 1975-04-14 1978-07-04 Beecham Group Limited Anti bacterial agents
US4034966A (en) 1975-11-05 1977-07-12 Massachusetts Institute Of Technology Method and apparatus for mixing particles
US4014469A (en) 1975-11-17 1977-03-29 Kozo Sato Nozzle of gas cutting torch
JPS5372016A (en) 1976-12-08 1978-06-27 Toyo Tire & Rubber Co Ltd Apparatus for preparation and supply of heavy oil w/o emulsion fuel
US4117550A (en) 1977-02-14 1978-09-26 Folland Enertec Ltd. Emulsifying system
US4091042A (en) 1977-08-19 1978-05-23 American Cyanamid Company Continuous adiabatic process for the mononitration of benzene
US4130394A (en) 1977-10-03 1978-12-19 Technicon Instruments Corporation Short sample detection
AU531759B2 (en) 1978-04-17 1983-09-08 Ici Ltd. Electrostatic spraying
SU1226392A1 (en) 1978-08-11 1986-04-23 Научно-исследовательский институт часовой промышленности Reduction gear box for electronic-mechanical clock with step motor
US4210809A (en) 1979-03-16 1980-07-01 Technicon Instruments Corporation Method and apparatus for the non-invasive determination of the characteristics of a segmented fluid stream
US4315754A (en) 1979-08-28 1982-02-16 Bifok Ab Flow injection analysis with intermittent flow
US4266721A (en) 1979-09-17 1981-05-12 Ppg Industries, Inc. Spray application of coating compositions utilizing induction and corona charging means
JPS5665627A (en) 1979-11-05 1981-06-03 Agency Of Ind Science & Technol Method of combining particles of liquid, etc.
US4253846A (en) 1979-11-21 1981-03-03 Technicon Instruments Corporation Method and apparatus for automated analysis of fluid samples
DE3168903D1 (en) 1980-08-28 1985-03-28 Du Pont Flow analysis
GB2097692B (en) 1981-01-10 1985-05-22 Shaw Stewart P D Combining chemical reagents
GB2092497B (en) 1981-02-06 1985-01-16 Honda Motor Co Ltd Welding torch assembly
US4439980A (en) 1981-11-16 1984-04-03 The United States Of America As Represented By The Secretary Of The Navy Electrohydrodynamic (EHD) control of fuel injection in gas turbines
DE3230289A1 (en) 1982-08-14 1984-02-16 Bayer Ag, 5090 Leverkusen PRODUCTION OF PHARMACEUTICAL OR COSMETIC DISPERSIONS
DE3379448D1 (en) 1982-10-13 1989-04-27 Ici Plc Electrostatic sprayhead assembly
US4853336A (en) 1982-11-15 1989-08-01 Technicon Instruments Corporation Single channel continuous flow system
CA1238900A (en) 1982-11-15 1988-07-05 Stephen Saros Single channel continuous slug flow mixing of discrete fluid components
US4533634A (en) 1983-01-26 1985-08-06 Amf Inc. Tissue culture medium
US4585209A (en) 1983-10-27 1986-04-29 Harry E. Aine Miniature valve and method of making same
US4618476A (en) 1984-02-10 1986-10-21 Eastman Kodak Company Capillary transport device having speed and meniscus control means
US4566908A (en) 1984-02-24 1986-01-28 Mita Industrial Company, Limited Azoic pigments having a silica core
US4865444A (en) 1984-04-05 1989-09-12 Mobil Oil Corporation Apparatus and method for determining luminosity of hydrocarbon fuels
US4675285A (en) 1984-09-19 1987-06-23 Genetics Institute, Inc. Method for identification and isolation of DNA encoding a desired protein
US4883750A (en) 1984-12-13 1989-11-28 Applied Biosystems, Inc. Detection of specific sequences in nucleic acids
GB8504254D0 (en) 1985-02-19 1985-03-20 Ici Plc Spraying apparatus
GB8504916D0 (en) 1985-02-26 1985-03-27 Isc Chemicals Ltd Emulsions of perfluorocarbons in aqueous media
US4676274A (en) 1985-02-28 1987-06-30 Brown James F Capillary flow control
US4683195A (en) 1986-01-30 1987-07-28 Cetus Corporation Process for amplifying, detecting, and/or-cloning nucleic acid sequences
US4683202A (en) 1985-03-28 1987-07-28 Cetus Corporation Process for amplifying nucleic acid sequences
US5333675C1 (en) 1986-02-25 2001-05-01 Perkin Elmer Corp Apparatus and method for performing automated amplification of nucleic acid sequences and assays using heating and cooling steps
US5656493A (en) 1985-03-28 1997-08-12 The Perkin-Elmer Corporation System for automated performance of the polymerase chain reaction
US4739044A (en) 1985-06-13 1988-04-19 Amgen Method for derivitization of polynucleotides
US4801529A (en) 1985-06-18 1989-01-31 Brandeis University Methods for isolating mutant microoganisms using microcapsules coated with indicator material
US4963498A (en) 1985-08-05 1990-10-16 Biotrack Capillary flow device
US4757141A (en) 1985-08-26 1988-07-12 Applied Biosystems, Incorporated Amino-derivatized phosphite and phosphate linking agents, phosphoramidite precursors, and useful conjugates thereof
GB8604328D0 (en) 1986-02-21 1986-03-26 Ici Plc Producing spray of droplets of liquid
CA1284931C (en) 1986-03-13 1991-06-18 Henry A. Erlich Process for detecting specific nucleotide variations and genetic polymorphisms present in nucleic acids
US4916070A (en) 1986-04-14 1990-04-10 The General Hospital Corporation Fibrin-specific antibodies and method of screening for the antibodies
US4767929A (en) 1986-10-06 1988-08-30 The United States Of America As Represented By The United State Department Of Energy Extended range radiation dose-rate monitor
US4767515A (en) 1987-07-30 1988-08-30 The United States Of America As Represented By The United States Department Of Energy Surface area generation and droplet size control in solvent extraction systems utilizing high intensity electric fields
US5149625A (en) 1987-08-11 1992-09-22 President And Fellows Of Harvard College Multiplex analysis of DNA
EP0304312B1 (en) 1987-08-21 1992-10-21 Sharp Kabushiki Kaisha An optical disk for use in optical memory devices
JPS6489884A (en) 1987-09-30 1989-04-05 Sony Corp White balance correction circuit
US5180662A (en) 1988-01-05 1993-01-19 The United States Of America As Represented By The Department Of Health And Human Services Cytotoxic T lymphocyte activation assay
US4856363A (en) 1988-02-10 1989-08-15 Wickes Manufacturing Company Parking brake assembly
US5185099A (en) 1988-04-20 1993-02-09 Institut National De Recherche Chimique Appliquee Visco-elastic, isotropic materials based on water, fluorinate sufactants and fluorinated oils, process for their preparation, and their use in various fields, such as optics, pharmacology and electrodynamics
US5055390A (en) 1988-04-22 1991-10-08 Massachusetts Institute Of Technology Process for chemical manipulation of non-aqueous surrounded microdroplets
US4908112A (en) 1988-06-16 1990-03-13 E. I. Du Pont De Nemours & Co. Silicon semiconductor wafer for analyzing micronic biological samples
US5498523A (en) 1988-07-12 1996-03-12 President And Fellows Of Harvard College DNA sequencing with pyrophosphatase
US5096615A (en) 1988-07-19 1992-03-17 The United States Of America As Represented By The United States Department Of Energy Solid aerosol generator
US5104813A (en) 1989-04-13 1992-04-14 Biotrack, Inc. Dilution and mixing cartridge
US4981580A (en) 1989-05-01 1991-01-01 Coulter Corporation Coincidence arbitration in a flow cytomery sorting system
NZ229355A (en) 1989-05-31 1991-12-23 Nz Ministry Forestry Spray nozzle assembly; flexible fluid outlet within nozzle to atomise fluid
US5143854A (en) 1989-06-07 1992-09-01 Affymax Technologies N.V. Large scale photolithographic solid phase synthesis of polypeptides and receptor binding screening thereof
CA2016981C (en) 1989-06-12 1994-09-27 Mark Joseph Devaney, Jr. Temperature control device and reaction vessel
EP0478686B1 (en) 1989-06-22 1993-08-11 Applications Et Transferts De Technologies Avancees Atta Fluorine and phosphorous-containing amphiphilic molecules with surfactant properties
CA2020958C (en) 1989-07-11 2005-01-11 Daniel L. Kacian Nucleic acid sequence amplification methods
GB8917963D0 (en) 1989-08-05 1989-09-20 Scras Apparatus for repeated automatic execution of a thermal cycle for treatment of biological samples
US5192659A (en) 1989-08-25 1993-03-09 Genetype Ag Intron sequence analysis method for detection of adjacent and remote locus alleles as haplotypes
DE3930376A1 (en) 1989-09-12 1991-03-21 Biotest Ag ENZYME IMMUNOMETRIC DETERMINATION METHOD USING PEROXIDASE AS A MARKING ENZYME
ATE168416T1 (en) 1989-10-05 1998-08-15 Optein Inc CELL-FREE SYNTHESIS AND ISOLATION OF GENES AND POLYPEPTIDES
US5310653A (en) 1989-10-24 1994-05-10 Board Of Regents, The University Of Texas System Tumor marker protein and antibodies thereto for cancer risk assessment or diagnosis
US5207973A (en) 1989-11-27 1993-05-04 Martin Marietta Energy Systems, Inc. Method and apparatus for the production of metal oxide powder
US5122360A (en) 1989-11-27 1992-06-16 Martin Marietta Energy Systems, Inc. Method and apparatus for the production of metal oxide powder
US4941959A (en) 1989-11-27 1990-07-17 Martin Marietta Energy Systems, Inc. Electric field-driven, magnetically-stabilized ferro-emulsion phase contactor
US5313009A (en) 1990-01-04 1994-05-17 Nrm International Technologies C.V. Nitration process
US5091652A (en) 1990-01-12 1992-02-25 The Regents Of The University Of California Laser excited confocal microscope fluorescence scanner and method
JP3176607B2 (en) 1990-02-07 2001-06-18 群馬大学長 Method for forming uniform droplets
EP0442019B1 (en) 1990-02-16 1994-02-09 J. Wagner Gmbh Method of operating an electrostatic and pneumatic paint spray gun
US5523162A (en) 1990-04-03 1996-06-04 Ppg Industries, Inc. Water repellent surface treatment for plastic and coated plastic substrates
SE470347B (en) 1990-05-10 1994-01-31 Pharmacia Lkb Biotech Microstructure for fluid flow systems and process for manufacturing such a system
US5270163A (en) 1990-06-11 1993-12-14 University Research Corporation Methods for identifying nucleic acid ligands
EP0786469B1 (en) 1990-06-11 2006-03-01 Gilead Sciences, Inc. Methods of use of nucleic acid ligands
WO1992003734A1 (en) 1990-08-20 1992-03-05 Alain De Weck A method for measuring t-lymphocyte responses by chemiluminescent assays
DE476178T1 (en) 1990-09-21 1992-07-23 Bioplex Medical B.V., Vaals, Nl DEVICE FOR THE APPLICATION OF ANTI-BLOODING FABRIC ON PERFORATED BLOOD VESSELS.
US6149789A (en) 1990-10-31 2000-11-21 Fraunhofer Gesellschaft Zur Forderung Der Angewandten Forschung E.V. Process for manipulating microscopic, dielectric particles and a device therefor
FR2669028B1 (en) 1990-11-13 1992-12-31 Rhone Poulenc Chimie PROCESS FOR THE MANUFACTURE OF DOUBLE RARE EARTH AND AMMONIUM OXALATES AND THEIR USES FOR THE MANUFACTURE OF RARE EARTH OXIDES.
KR100236506B1 (en) 1990-11-29 2000-01-15 퍼킨-엘머시터스인스트루먼츠 Apparatus for polymerase chain reaction
US5490505A (en) 1991-03-07 1996-02-13 Masimo Corporation Signal processing apparatus
US6110700A (en) 1991-03-11 2000-08-29 The General Hospital Corporation PRAD1 cyclin and its cDNA
US5262027A (en) 1991-03-22 1993-11-16 Martin Marietta Energy Systems, Inc. Method of using an electric field controlled emulsion phase contactor
GB9107628D0 (en) 1991-04-10 1991-05-29 Moonbrook Limited Preparation of diagnostic agents
NZ242896A (en) 1991-05-30 1996-05-28 Blood Res Center Apparatus and methods for analysing blood components especially leukocyte content
US5460945A (en) 1991-05-30 1995-10-24 Center For Blood Research, Inc. Device and method for analysis of blood components and identifying inhibitors and promoters of the inflammatory response
NZ264353A (en) 1991-05-30 1996-05-28 For Blood Research Inc Centre Method of collecting or purifying leukocytes from a fluid sample, apparatus, immune response inhibitor test
DE4119955C2 (en) 1991-06-18 2000-05-31 Danfoss As Miniature actuator
EP0546174B1 (en) 1991-06-29 1997-10-29 Miyazaki-Ken Monodisperse single and double emulsions and production thereof
GB9117191D0 (en) 1991-08-08 1991-09-25 Tioxide Chemicals Limited Preparation of titanium derivatives
CA2114950A1 (en) 1991-08-10 1993-02-11 Michael J. Embleton Treatment of cell populations
DE4127405C2 (en) 1991-08-19 1996-02-29 Fraunhofer Ges Forschung Process for the separation of mixtures of microscopic dielectric particles suspended in a liquid or a gel and device for carrying out the process
DE69223980T2 (en) 1991-10-15 1998-05-28 Multilyte Ltd BINDING TEST USING A MARKED REAGENT
US5270170A (en) 1991-10-16 1993-12-14 Affymax Technologies N.V. Peptide library and screening method
JP3164919B2 (en) 1991-10-29 2001-05-14 ゼロックス コーポレーション Method of forming dichroic balls
US6048690A (en) 1991-11-07 2000-04-11 Nanogen, Inc. Methods for electronic fluorescent perturbation for analysis and electronic perturbation catalysis for synthesis
US5344489A (en) 1991-11-15 1994-09-06 Manfred R. Kuehnle Synthetic, monodispersed color pigments for the coloration of media such as printing inks, and method and apparatus for making same
US5612188A (en) 1991-11-25 1997-03-18 Cornell Research Foundation, Inc. Automated, multicompartmental cell culture system
WO1993013216A1 (en) 1991-12-24 1993-07-08 The President And Fellows Of Harvard College Site-directed mutagenesis of dna
US5413924A (en) 1992-02-13 1995-05-09 Kosak; Kenneth M. Preparation of wax beads containing a reagent for release by heating
US5241159A (en) 1992-03-11 1993-08-31 Eastman Kodak Company Multi-zone heating for a fuser roller
JPH05265447A (en) 1992-03-18 1993-10-15 Kawai Musical Instr Mfg Co Ltd Electronic musical instrument capable of deciding finger of hand operating keyboard
US6107059A (en) 1992-04-29 2000-08-22 Affymax Technologies N.V. Peptide library and screening method
US5304487A (en) 1992-05-01 1994-04-19 Trustees Of The University Of Pennsylvania Fluid handling in mesoscale analytical devices
US5726026A (en) 1992-05-01 1998-03-10 Trustees Of The University Of Pennsylvania Mesoscale sample preparation device and systems for determination and processing of analytes
AU680195B2 (en) 1992-05-01 1997-07-24 Trustees Of The University Of Pennsylvania, The Analysis based on flow restriction
US5587128A (en) 1992-05-01 1996-12-24 The Trustees Of The University Of Pennsylvania Mesoscale polynucleotide amplification devices
US5498392A (en) 1992-05-01 1996-03-12 Trustees Of The University Of Pennsylvania Mesoscale polynucleotide amplification device and method
US5744366A (en) 1992-05-01 1998-04-28 Trustees Of The University Of Pennsylvania Mesoscale devices and methods for analysis of motile cells
US5296375A (en) 1992-05-01 1994-03-22 Trustees Of The University Of Pennsylvania Mesoscale sperm handling devices
US5486335A (en) 1992-05-01 1996-01-23 Trustees Of The University Of Pennsylvania Analysis based on flow restriction
US5397605A (en) 1992-05-29 1995-03-14 Barbieri; Girolamo Method and apparatus for electrostatically coating a workpiece with paint
SE500071C2 (en) 1992-06-25 1994-04-11 Vattenfall Utveckling Ab Device for mixing two fluids, in particular liquids of different temperature
DE4223169C1 (en) 1992-07-10 1993-11-25 Ferring Arzneimittel Gmbh Process for the microencapsulation of water-soluble active substances
JPH0665609A (en) 1992-08-25 1994-03-08 Mitsubishi Materials Corp Production of ferrous sintered and forged parts
RU2048522C1 (en) 1992-10-14 1995-11-20 Институт белка РАН Method of nucleic acid copying, method of their expression and a medium for their realization
GB9225098D0 (en) 1992-12-01 1993-01-20 Coffee Ronald A Charged droplet spray mixer
US6105571A (en) 1992-12-22 2000-08-22 Electrosols, Ltd. Dispensing device
IL104384A (en) 1993-01-13 1996-11-14 Yeda Res & Dev Method for screening catalytic non-enzyme polypeptides and proteins
US5436149A (en) 1993-02-19 1995-07-25 Barnes; Wayne M. Thermostable DNA polymerase with enhanced thermostability and enhanced length and efficiency of primer extension
JPH06265447A (en) 1993-03-16 1994-09-22 Hitachi Ltd Trace quantity reactor and trace element measuring instrument therewith
DE4308839C2 (en) 1993-03-19 1997-04-30 Jordanow & Co Gmbh Device for mixing flow media
FR2703263B1 (en) 1993-03-31 1995-05-19 Rhone Poulenc Nutrition Animal Process for the preparation of spherules of active principles.
EP1296134B1 (en) 1993-04-15 2013-05-29 Bayer Intellectual Property GmbH Sampling device and its use for controlling sample introduction in microcolumn separation techniques
EP0696200A4 (en) 1993-04-19 1998-04-15 Medisorb Technologies Internat Encapsulation of nucleic acids with conjugates that facilitate and target cellular uptake and gene expression
CA2160457A1 (en) 1993-04-19 1994-10-27 Stuart A. Kauffman Random chemistry for the generation of new compounds
DE69431719T2 (en) 1993-06-25 2003-09-18 Affymetrix Inc N D Ges D Staat HYBRIDIZATION AND SEQUENCING OF NUCLEIC ACIDS
US7229770B1 (en) 1998-10-01 2007-06-12 The Regents Of The University Of California YKL-40 as a marker and prognostic indicator for cancers
US20040091923A1 (en) 1993-07-23 2004-05-13 Bio-Rad Laboratories, Inc. Linked linear amplification of nucleic acids
US5417235A (en) 1993-07-28 1995-05-23 Regents Of The University Of Michigan Integrated microvalve structures with monolithic microflow controller
US5403617A (en) 1993-09-15 1995-04-04 Mobium Enterprises Corporation Hybrid pulsed valve for thin film coating and method
US6776094B1 (en) 1993-10-04 2004-08-17 President & Fellows Of Harvard College Kit For Microcontact Printing
WO1995011922A1 (en) 1993-10-29 1995-05-04 Affymax Technologies N.V. In vitro peptide and antibody display libraries
US6165778A (en) 1993-11-02 2000-12-26 Affymax Technologies N.V. Reaction vessel agitation apparatus
US6316208B1 (en) 1994-01-07 2001-11-13 Memorial Sloan-Kettering Cancer Center Methods for determining isolated p27 protein levels and uses thereof
DE4402038A1 (en) 1994-01-25 1995-07-27 Borries Horst Von Blister pack
PH31414A (en) 1994-02-24 1998-10-29 Boehringer Ingelheim Int Method of diagnosing cancer precancerous state, orsusceptibility to other forms of diseases by anal ysis of irf-1 specific rna in biopsy samples.
WO1995024929A2 (en) 1994-03-15 1995-09-21 Brown University Research Foundation Polymeric gene delivery system
US5989815A (en) 1994-03-18 1999-11-23 University Of Utah Research Foundation Methods for detecting predisposition to cancer at the MTS gene
GB9406171D0 (en) 1994-03-29 1994-05-18 Electrosols Ltd Dispensing device
JPH07270319A (en) 1994-03-30 1995-10-20 Mochida Pharmaceut Co Ltd Method for measuring substance containing adenyl group using heteropoly acid
US5587081A (en) 1994-04-26 1996-12-24 Jet-Tech, Inc. Thermophilic aerobic waste treatment process
GB9411671D0 (en) 1994-06-10 1994-08-03 Univ Singapore Tumor diagnosis and prognosis
US5750988A (en) 1994-07-11 1998-05-12 Hewlett-Packard Company Orthogonal ion sampling for APCI mass spectrometry
US6653626B2 (en) 1994-07-11 2003-11-25 Agilent Technologies, Inc. Ion sampling for APPI mass spectrometry
US5641658A (en) 1994-08-03 1997-06-24 Mosaic Technologies, Inc. Method for performing amplification of nucleic acid with two primers bound to a single solid support
US6124439A (en) 1994-08-17 2000-09-26 The Rockefeller University OB polypeptide antibodies and method of making
US5680283A (en) 1994-09-30 1997-10-21 Kabushiki Kaisha Toshiba Magnetic head and magnetic disk drive
US5846719A (en) 1994-10-13 1998-12-08 Lynx Therapeutics, Inc. Oligonucleotide tags for sorting and identification
US5604097A (en) 1994-10-13 1997-02-18 Spectragen, Inc. Methods for sorting polynucleotides using oligonucleotide tags
US5695934A (en) 1994-10-13 1997-12-09 Lynx Therapeutics, Inc. Massively parallel sequencing of sorted polynucleotides
JPH08153669A (en) 1994-11-30 1996-06-11 Hitachi Ltd Thin film forming method and formation device
US5813988A (en) 1995-02-03 1998-09-29 Research Foundation Time-resolved diffusion tomographic imaging in highly scattering turbid media
US5661222A (en) 1995-04-13 1997-08-26 Dentsply Research & Development Corp. Polyvinylsiloxane impression material
WO1996034112A1 (en) 1995-04-24 1996-10-31 Chromaxome Corp. Methods for generating and screening novel metabolic pathways
US5840254A (en) 1995-06-02 1998-11-24 Cdc Technologies, Inc. Apparatus for mixing fluids for analysis
EP0957772A2 (en) 1995-06-06 1999-11-24 Quantic Biomedical Partners Wound sealant preparation and application device and method
US5756122A (en) 1995-06-07 1998-05-26 Georgetown University Liposomally encapsulated nucleic acids having high entrapment efficiencies, method of manufacturer and use thereof for transfection of targeted cells
KR19990022354A (en) 1995-06-07 1999-03-25 펠루라, 티모시, 제이 Reverse fluorocarbon emulsion composition for drug delivery
US5882856A (en) 1995-06-07 1999-03-16 Genzyme Corporation Universal primer sequence for multiplex DNA amplification
US5910408A (en) 1995-06-07 1999-06-08 The General Hospital Corporation Catalytic DNA having ligase activity
EP0748860B1 (en) 1995-06-14 2001-08-29 Tonen Corporation Demulsification by microorganisms
DE69628016T2 (en) 1995-06-16 2004-04-01 University Of Washington, Seattle MINIATURIZED DIFFERENTIAL EXTRACTION DEVICE AND METHOD
TW293783B (en) 1995-06-16 1996-12-21 Ciba Geigy Ag
US5589136A (en) 1995-06-20 1996-12-31 Regents Of The University Of California Silicon-based sleeve devices for chemical reactions
US5789206A (en) 1995-07-07 1998-08-04 Myriad Genetics, Inc. Method for ligating adaptors to nucleic acids which methods are useful for obtaining the ends of genes
DE69626579T2 (en) 1995-07-19 2003-11-20 Nippon Telegraph & Telephone Water repellent composition, fluorocarbon polymer coating composition and coating film
US5872010A (en) 1995-07-21 1999-02-16 Northeastern University Microscale fluid handling system
WO1997004748A2 (en) 1995-08-01 1997-02-13 Advanced Therapies, Inc. Enhanced artificial viral envelopes for cellular delivery of therapeutic substances
US5636400A (en) 1995-08-07 1997-06-10 Young; Keenan L. Automatic infant bottle cleaner
JP3232525B2 (en) 1995-08-22 2001-11-26 信越化学工業株式会社 Water repellent agent
US6130098A (en) 1995-09-15 2000-10-10 The Regents Of The University Of Michigan Moving microdroplets
WO1997012991A1 (en) 1995-09-22 1997-04-10 Terragen Diversity Inc. Method for isolating xylanase gene sequences from soil dna, compositions useful in such method and compositions obtained thereby
US5851769A (en) 1995-09-27 1998-12-22 The Regents Of The University Of California Quantitative DNA fiber mapping
US6243373B1 (en) 1995-11-01 2001-06-05 Telecom Internet Ltd. Method and apparatus for implementing a computer network/internet telephone system
US6562605B1 (en) 1995-11-13 2003-05-13 Genencor International, Inc. Extraction of water soluble biomaterials from fluids using a carbon dioxide/surfactant mixture
US20030215798A1 (en) 1997-06-16 2003-11-20 Diversa Corporation High throughput fluorescence-based screening for novel enzymes
JP3759986B2 (en) 1995-12-07 2006-03-29 フロイント産業株式会社 Seamless capsule and manufacturing method thereof
US5808691A (en) 1995-12-12 1998-09-15 Cirrus Logic, Inc. Digital carrier synthesis synchronized to a reference signal that is asynchronous with respect to a digital sampling clock
US5681600A (en) 1995-12-18 1997-10-28 Abbott Laboratories Stabilization of liquid nutritional products and method of making
US5670325A (en) 1996-08-14 1997-09-23 Exact Laboratories, Inc. Method for the detection of clonal populations of transformed cells in a genomically heterogeneous cellular sample
US6261797B1 (en) 1996-01-29 2001-07-17 Stratagene Primer-mediated polynucleotide synthesis and manipulation techniques
US5868322A (en) 1996-01-31 1999-02-09 Hewlett-Packard Company Apparatus for forming liquid droplets having a mechanically fixed inner microtube
JP2975943B2 (en) 1996-02-20 1999-11-10 農林水産省食品総合研究所長 Emulsion manufacturing method and emulsion manufacturing apparatus
AU2290897A (en) 1996-04-04 1997-10-29 Novartis Ag Device for counting small particles and a sorting apparatus comprising such a device
WO1997039359A1 (en) 1996-04-15 1997-10-23 Dade International Inc. Apparatus and method for analysis
US6207397B1 (en) 1996-04-18 2001-03-27 Ariad Pharmaceuticals, Inc. In vitro fluorescence polarization assay
GB9608129D0 (en) 1996-04-19 1996-06-26 Central Research Lab Ltd Method and apparatus for diffusive transfer between immiscible fluids
GB9608540D0 (en) 1996-04-25 1996-07-03 Medical Res Council Isolation of enzymes
EP2369007B1 (en) 1996-05-29 2015-07-29 Cornell Research Foundation, Inc. Detection of nucleic acid sequence differences using coupled ligase detection and polymerase chain reactions
US5726404A (en) 1996-05-31 1998-03-10 University Of Washington Valveless liquid microswitch
US5840506A (en) 1996-06-05 1998-11-24 Thomas Jefferson University Methods for the diagnosis and prognosis of cancer
US6083693A (en) 1996-06-14 2000-07-04 Curagen Corporation Identification and comparison of protein-protein interactions that occur in populations
US5876771A (en) 1996-06-20 1999-03-02 Tetra Laval Holdings & Finance, Sa Process and article for determining the residence time of a food particle
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
AU729537B2 (en) 1996-06-28 2001-02-01 Caliper Technologies Corporation High throughput screening assay systems in microscale fluidic devices
US5779868A (en) 1996-06-28 1998-07-14 Caliper Technologies Corporation Electropipettor and compensation means for electrophoretic bias
CA2258481C (en) 1996-06-28 2006-05-23 Caliper Technologies Corporation Electropipettor and compensation means for electrophoretic bias
EP0912238B1 (en) 1996-07-15 2001-10-10 CalCiTech Ltd. Production of powders
US6252129B1 (en) 1996-07-23 2001-06-26 Electrosols, Ltd. Dispensing device and method for forming material
US6100029A (en) 1996-08-14 2000-08-08 Exact Laboratories, Inc. Methods for the detection of chromosomal aberrations
US6203993B1 (en) 1996-08-14 2001-03-20 Exact Science Corp. Methods for the detection of nucleic acids
US6146828A (en) 1996-08-14 2000-11-14 Exact Laboratories, Inc. Methods for detecting differences in RNA expression levels and uses therefor
US5928870A (en) 1997-06-16 1999-07-27 Exact Laboratories, Inc. Methods for the detection of loss of heterozygosity
US5884846A (en) 1996-09-19 1999-03-23 Tan; Hsiaoming Sherman Pneumatic concentric nebulizer with adjustable and capillaries
US6120666A (en) 1996-09-26 2000-09-19 Ut-Battelle, Llc Microfabricated device and method for multiplexed electrokinetic focusing of fluid streams and a transport cytometry method using same
US5858187A (en) 1996-09-26 1999-01-12 Lockheed Martin Energy Systems, Inc. Apparatus and method for performing electrodynamic focusing on a microchip
GB9620209D0 (en) 1996-09-27 1996-11-13 Cemu Bioteknik Ab Method of sequencing DNA
CA2236867A1 (en) 1996-09-27 1998-04-02 Icos Corporation Method to identify compounds for disrupting protein/protein interactions
US6140053A (en) 1996-11-06 2000-10-31 Sequenom, Inc. DNA sequencing by mass spectrometry via exonuclease degradation
US7054674B2 (en) 1996-11-19 2006-05-30 Astron Clinica Limited Method of and apparatus for investigating tissue histology
GB9624003D0 (en) 1996-11-19 1997-01-08 Univ Birmingham Method and apparatus for measurement of skin histology
US6379929B1 (en) 1996-11-20 2002-04-30 The Regents Of The University Of Michigan Chip-based isothermal amplification devices and methods
WO1998023733A2 (en) 1996-11-27 1998-06-04 University Of Washington Thermostable polymerases having altered fidelity
US6310354B1 (en) 1996-12-03 2001-10-30 Erkki Soini Method and a device for monitoring nucleic acid amplification reactions
US5958703A (en) 1996-12-03 1999-09-28 Glaxo Group Limited Use of modified tethers in screening compound libraries
US20030104372A1 (en) * 1996-12-23 2003-06-05 Pyrosequencing Ab. Allele specific primer extension
GB9626815D0 (en) 1996-12-23 1997-02-12 Cemu Bioteknik Ab Method of sequencing DNA
US20020034737A1 (en) 1997-03-04 2002-03-21 Hyseq, Inc. Methods and compositions for detection or quantification of nucleic acid species
KR100566859B1 (en) 1997-01-21 2006-04-03 제너럴 하스피톨 코포레이션 Selection of proteins using rna-protein fusions
US5890745A (en) * 1997-01-29 1999-04-06 The Board Of Trustees Of The Leland Stanford Junior University Micromachined fluidic coupler
CA2196496A1 (en) 1997-01-31 1998-07-31 Stephen William Watson Michnick Protein fragment complementation assay for the detection of protein-protein interactions
AU6152498A (en) 1997-02-05 1998-08-25 California Institute Of Technology Microfluidic sub-millisecond mixers
JPH10217477A (en) 1997-02-07 1998-08-18 Fuji Xerox Co Ltd Ink jet recording device
JP3935509B2 (en) 1997-02-12 2007-06-27 ワイ. チャン,ユージーン Methods and products for polymer analysis
GB9703369D0 (en) 1997-02-18 1997-04-09 Lindqvist Bjorn H Process
US6208749B1 (en) 1997-02-28 2001-03-27 Electro-Optical Sciences, Inc. Systems and methods for the multispectral imaging and characterization of skin tissue
US6081612A (en) 1997-02-28 2000-06-27 Electro Optical Sciences Inc. Systems and methods for the multispectral imaging and characterization of skin tissue
US6307957B1 (en) 1997-02-28 2001-10-23 Electro-Optical Sciences Inc Multispectral imaging and characterization of biological tissue
US6045755A (en) 1997-03-10 2000-04-04 Trega Biosciences,, Inc. Apparatus and method for combinatorial chemistry synthesis
US5994068A (en) 1997-03-11 1999-11-30 Wisconsin Alumni Research Foundation Nucleic acid indexing
US6023540A (en) 1997-03-14 2000-02-08 Trustees Of Tufts College Fiber optic sensor with encoded microspheres
EP0975969A4 (en) 1997-03-18 2002-07-31 Chromaxome Corp Methods for screening compounds using encapsulated cells
US6294344B1 (en) 1997-03-19 2001-09-25 The Board Of Trustees Of The University Of Arkansas Methods for the early diagnosis of ovarian cancer
US6316213B1 (en) 1997-03-19 2001-11-13 The Board Of Trustees Of The University Of Arkansas Methods for the early diagnosis of ovarian, breast and lung cancer
US6268165B1 (en) 1997-03-19 2001-07-31 The Board Of Trustees Of The University Of Arkansas Methods for the early diagnosis of ovarian cancer
US6090800A (en) 1997-05-06 2000-07-18 Imarx Pharmaceutical Corp. Lipid soluble steroid prodrugs
US6048551A (en) 1997-03-27 2000-04-11 Hilfinger; John M. Microsphere encapsulation of gene transfer vectors
JPH10288131A (en) 1997-04-11 1998-10-27 Yanmar Diesel Engine Co Ltd Injection nozzle of diesel engine
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
DE19717085C2 (en) 1997-04-23 1999-06-17 Bruker Daltonik Gmbh Processes and devices for extremely fast DNA multiplication using polymerase chain reactions (PCR)
US5879892A (en) 1997-04-25 1999-03-09 Ludwig Institute For Cancer Research Leukemia associated genes
US6632619B1 (en) 1997-05-16 2003-10-14 The Governors Of The University Of Alberta Microfluidic system and methods of use
US6004025A (en) 1997-05-16 1999-12-21 Life Technologies, Inc. Automated liquid manufacturing system
WO1998052691A1 (en) 1997-05-16 1998-11-26 Alberta Research Council Microfluidic system and methods of use
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
US20020015997A1 (en) 1997-06-16 2002-02-07 Lafferty William Michael Capillary array-based sample screening
US5888778A (en) 1997-06-16 1999-03-30 Exact Laboratories, Inc. High-throughput screening method for identification of genetic mutations or disease-causing microorganisms using segmented primers
US6074879A (en) 1997-06-23 2000-06-13 Bayer Corporation Synthetic polymer particles for use as standards and calibrators in flow cytometry
JP2843319B1 (en) 1997-06-27 1999-01-06 科学技術振興事業団 Microstrip gas chamber high-speed data acquisition system and sample measurement method using the same
JP3557859B2 (en) 1997-07-15 2004-08-25 コニカミノルタホールディングス株式会社 Silver halide photographic emulsion, production method thereof and silver halide photographic light-sensitive material
US6403373B1 (en) 1997-10-10 2002-06-11 Ludwig Institute For Cancer Research Isolated nucleic acid molecules associated with colon, renal, and stomach cancer and methods of using these
US6974669B2 (en) 2000-03-28 2005-12-13 Nanosphere, Inc. Bio-barcodes based on oligonucleotide-modified nanoparticles
US20050037397A1 (en) 2001-03-28 2005-02-17 Nanosphere, Inc. Bio-barcode based detection of target analytes
US6165578A (en) 1997-07-23 2000-12-26 Matsushita Electric Industrial Co., Ltd. Optical information recording medium and method for producing the same
FR2767064B1 (en) 1997-08-07 1999-11-12 Centre Nat Rech Scient METHOD FOR RELEASING AN ACTIVE INGREDIENT CONTAINED IN A MULTIPLE EMULSION
US5980936A (en) 1997-08-07 1999-11-09 Alliance Pharmaceutical Corp. Multiple emulsions comprising a hydrophobic continuous phase
NZ328751A (en) 1997-09-16 1999-01-28 Bernard Charles Sherman Solid medicament containing an anionic surfactant and cyclosporin
US6833242B2 (en) 1997-09-23 2004-12-21 California Institute Of Technology Methods for detecting and sorting polynucleotides based on size
US6511803B1 (en) 1997-10-10 2003-01-28 President And Fellows Of Harvard College Replica amplification of nucleic acid arrays
US6008003A (en) 1997-10-28 1999-12-28 Promega Corporation Non-invasive diagnostic method for interstitial cystitis and bladder cancer
GB9723262D0 (en) 1997-11-05 1998-01-07 British Nuclear Fuels Plc Reactions of aromatic compounds
US6162421A (en) 1997-11-17 2000-12-19 Revlon Consumer Products Corporation Pigmented water-in-oil emulsion cosmetic sticks
US5927852A (en) 1997-12-01 1999-07-27 Minnesota Mining And Manfacturing Company Process for production of heat sensitive dispersions or emulsions
US6972170B1 (en) 1997-12-01 2005-12-06 Sloan-Kettering Institute For Cancer Research Markers for prostate cancer
JP2002508250A (en) 1997-12-17 2002-03-19 ユニバーシィダッド デ セビリヤ Device and method for producing spherical particles of uniform size
US5972615A (en) 1998-01-21 1999-10-26 Urocor, Inc. Biomarkers and targets for diagnosis, prognosis and management of prostate disease
GB2334271B (en) 1998-02-17 2000-09-20 Sofitech Nv Water based drilling fluid with shale swelling inhibiting agent and phosphonate
TW575562B (en) 1998-02-19 2004-02-11 Agrevo Uk Ltd Fungicides
US7022821B1 (en) 1998-02-20 2006-04-04 O'brien Timothy J Antibody kit for the detection of TADG-15 protein
US6064149A (en) 1998-02-23 2000-05-16 Micron Technology Inc. Field emission device with silicon-containing adhesion layer
US6897018B1 (en) 1998-02-25 2005-05-24 The United States Of America As Represented By The Department Of Health And Human Services DLC-1 gene deleted in cancers
US6292756B1 (en) 1998-02-26 2001-09-18 Premier Instruments, Inc. Narrow band infrared water fraction apparatus for gas well and liquid hydrocarbon flow stream use
FR2776538B1 (en) 1998-03-27 2000-07-21 Centre Nat Rech Scient ELECTROHYDRODYNAMIC SPRAYING MEANS
JP3081880B2 (en) 1998-03-30 2000-08-28 農林水産省食品総合研究所長 Microsphere continuous manufacturing equipment
JP3109471B2 (en) 1998-03-31 2000-11-13 日本電気株式会社 Cleaning / drying equipment and semiconductor device manufacturing line
FI980874A (en) * 1998-04-20 1999-10-21 Wallac Oy Method and apparatus for conducting chemical analysis on small amounts of liquid
US6395253B2 (en) 1998-04-23 2002-05-28 The Regents Of The University Of Michigan Microspheres containing condensed polyanionic bioactive agents and methods for their production
US20060269558A1 (en) 1998-04-27 2006-11-30 Murphy Gerald P Nr-CAM gene, nucleic acids and nucleic acid products for therapeutic and diagnostic uses for tumors
US5997636A (en) 1998-05-01 1999-12-07 Instrumentation Technology Associates, Inc. Method and apparatus for growing crystals
DE19822674A1 (en) 1998-05-20 1999-12-09 Gsf Forschungszentrum Umwelt Gas inlet for an ion source
ATE530891T1 (en) 1998-05-22 2011-11-15 California Inst Of Techn MINIATURIZED CELL SORTER
WO1999061163A1 (en) 1998-05-25 1999-12-02 Fuji Koeki Co., Ltd. Liquid spray device and cutting method
US6209928B1 (en) * 1998-06-04 2001-04-03 The Regents Of The University Of California Microfluidic interconnects
CA2332919A1 (en) 1998-06-08 1999-12-16 Caliper Technologies Corporation Microfluidic devices, systems and methods for performing integrated reactions and separations
GB9812768D0 (en) 1998-06-13 1998-08-12 Zeneca Ltd Methods
US6576420B1 (en) 1998-06-23 2003-06-10 Regents Of The University Of California Method for early diagnosis of, and determination of prognosis in, cancer
US7700568B2 (en) 1998-06-30 2010-04-20 Sloan-Kettering Institute For Cancer Research Uses of DNA-PK
JP2981547B1 (en) 1998-07-02 1999-11-22 農林水産省食品総合研究所長 Cross-flow type microchannel device and method for producing or separating emulsion using the device
EP1100889A4 (en) 1998-07-17 2002-03-06 Mirus Corp Micellar systems
US6003794A (en) 1998-08-04 1999-12-21 Progressive Grower Technologies, Inc. Electrostatic spray module
AU764633B2 (en) 1998-08-07 2003-08-28 Cellay, Llc Gel microdrops in genetic analysis
EP1876442A3 (en) 1998-09-17 2008-03-05 Advion BioSciences, Inc. Integrated monolithic microfabricated liquid chromatography system and method
EP2306195A3 (en) 1998-09-18 2012-04-25 Massachusetts Institute of Technology Biological applications of semiconductor nanocrystals
DE19845078A1 (en) 1998-09-30 2000-04-06 Basf Ag Polymer particles containing dye
US6637463B1 (en) 1998-10-13 2003-10-28 Biomicro Systems, Inc. Multi-channel microfluidic system design with balanced fluid flow distribution
US6591852B1 (en) 1998-10-13 2003-07-15 Biomicro Systems, Inc. Fluid circuit components based upon passive fluid dynamics
US6601613B2 (en) 1998-10-13 2003-08-05 Biomicro Systems, Inc. Fluid circuit components based upon passive fluid dynamics
WO2000022436A1 (en) 1998-10-13 2000-04-20 Biomicro Systems, Inc. Fluid circuit components based upon passive fluid dynamics
US6902892B1 (en) 1998-10-19 2005-06-07 Diadexus, Inc. Method of diagnosing, monitoring, staging, imaging and treating prostate cancer
US6960433B1 (en) 1998-10-19 2005-11-01 Diadexus, Inc. Method of diagnosing, monitoring, staging, imaging and treating prostate cancer
US7022472B2 (en) 1998-10-22 2006-04-04 Diadexus, Inc. Mutations in human MLH1 and human MSH2 genes useful in diagnosing colorectal cancer
US6086740A (en) 1998-10-29 2000-07-11 Caliper Technologies Corp. Multiplexed microfluidic devices and systems
US20030045491A1 (en) 2001-02-23 2003-03-06 Christoph Reinhard TTK in diagnosis and as a therapeutic target in cancer
US6614598B1 (en) 1998-11-12 2003-09-02 Institute Of Technology, California Microlensing particles and applications
US6569631B1 (en) 1998-11-12 2003-05-27 3-Dimensional Pharmaceuticals, Inc. Microplate thermal shift assay for ligand development using 5-(4″dimethylaminophenyl)-2-(4′-phenyl)oxazole derivative fluorescent dyes
US6450189B1 (en) 1998-11-13 2002-09-17 Universidad De Sevilla Method and device for production of components for microfabrication
US6139303A (en) 1998-11-20 2000-10-31 United Technologies Corporation Fixture for disposing a laser blocking material in an airfoil
US6353226B1 (en) 1998-11-23 2002-03-05 Abbott Laboratories Non-invasive sensor capable of determining optical parameters in a sample having multiple layers
US6465193B2 (en) 1998-12-11 2002-10-15 The Regents Of The University Of California Targeted molecular bar codes and methods for using the same
DE19857302C2 (en) 1998-12-14 2000-10-26 Forschungszentrum Juelich Gmbh Process for the enantioselective reduction of 3,5-dioxocarboxylic acids, their salts and esters
US20030069601A1 (en) 1998-12-15 2003-04-10 Closys Corporation Clotting cascade initiating apparatus and methods of use
US6205353B1 (en) 1998-12-22 2001-03-20 Research Foundation Of Cuny Time-resolved optical backscattering tomographic image reconstruction in scattering turbid media
AU2849800A (en) 1999-01-15 2000-08-01 Ljl Biosystems, Inc. Methods and apparatus for detecting polynucleotide hybridization
US6565727B1 (en) 1999-01-25 2003-05-20 Nanolytics, Inc. Actuators for microfluidics without moving parts
US6600077B1 (en) 1999-01-29 2003-07-29 Board Of Trustees Operating Michigan State University Biocatalytic synthesis of quinic acid and conversion to hydroquinone
GB9903841D0 (en) 1999-02-20 1999-04-14 Imp College Innovations Ltd Diagnosis and treatment of cancer
WO2000052204A2 (en) 1999-02-22 2000-09-08 Orntoft Torben F Gene expression in bladder tumors
US7615373B2 (en) 1999-02-25 2009-11-10 Virginia Commonwealth University Intellectual Property Foundation Electroprocessed collagen and tissue engineering
US6633031B1 (en) 1999-03-02 2003-10-14 Advion Biosciences, Inc. Integrated monolithic microfabricated dispensing nozzle and liquid chromatography-electrospray system and method
US6942978B1 (en) 1999-03-03 2005-09-13 The Board Of Trustees Of The University Of Arkansas Transmembrane serine protease overexpressed in ovarian carcinoma and uses thereof
US6171850B1 (en) 1999-03-08 2001-01-09 Caliper Technologies Corp. Integrated devices and systems for performing temperature controlled reactions and analyses
US6428053B1 (en) * 1999-03-12 2002-08-06 California Institute Of Technology Micromachined fluidic coupler and method of making the same
DE19911777A1 (en) 1999-03-17 2000-09-21 Merck Patent Gmbh Process for the preparation of cosmetic formulations
JP2000271475A (en) 1999-03-23 2000-10-03 Shinji Katsura Finely controlling method of chemical reaction by fine operation of water-in-oil emulsion
US6174160B1 (en) 1999-03-25 2001-01-16 University Of Washington Staged prevaporizer-premixer
US7153700B1 (en) 1999-03-26 2006-12-26 Dana-Farber Cancer Institute, Inc. Methods and compositions for diagnosing and predicting the behavior of cancer
WO2000061275A2 (en) 1999-04-08 2000-10-19 Bernd Penth Method and device for carrying out chemical and physical processes
US6267353B1 (en) 1999-04-19 2001-07-31 Pbm, Inc. Self draining valve
US20030207295A1 (en) 1999-04-20 2003-11-06 Kevin Gunderson Detection of nucleic acid reactions on bead arrays
IT1309792B1 (en) * 1999-04-22 2002-01-30 Eltek Spa HOUSEHOLD APPLIANCES USING WATER, IN PARTICULAR A WASHING MACHINE, WITH PERFECTED DEVICE FOR BLAST CHILLING
IL146034A0 (en) 1999-04-23 2002-07-25 Battelle Memorial Institute High mass transfer electrosprayer
US6682940B2 (en) 1999-05-04 2004-01-27 Dan A. Pankowsky Products and methods for single parameter and multiparameter phenotyping of cells
JP3815969B2 (en) 1999-05-12 2006-08-30 アクララ バイオサイエンシーズ, インコーポレイテッド Multiplex fluorescence detection in microfluidic devices
US6738502B1 (en) 1999-06-04 2004-05-18 Kairos Scientific, Inc. Multispectral taxonomic identification
ES2424713T4 (en) 1999-06-11 2014-01-23 Aradigm Corporation Method of producing a spray
US20060169800A1 (en) 1999-06-11 2006-08-03 Aradigm Corporation Aerosol created by directed flow of fluids and devices and methods for producing same
US6296673B1 (en) 1999-06-18 2001-10-02 The Regents Of The University Of California Methods and apparatus for performing array microcrystallizations
US6630006B2 (en) 1999-06-18 2003-10-07 The Regents Of The University Of California Method for screening microcrystallizations for crystal formation
JP3623479B2 (en) 1999-06-22 2005-02-23 テカン トレーディング アーゲー Apparatus and method for performing miniaturized in vitro amplification assays
US6210396B1 (en) 1999-06-24 2001-04-03 Medtronic, Inc. Guiding catheter with tungsten loaded band
US7195670B2 (en) 2000-06-27 2007-03-27 California Institute Of Technology High throughput screening of crystallization of materials
US6818395B1 (en) 1999-06-28 2004-11-16 California Institute Of Technology Methods and apparatus for analyzing polynucleotide sequences
US6964847B1 (en) 1999-07-14 2005-11-15 Packard Biosciences Company Derivative nucleic acids and uses thereof
EP1242813A4 (en) * 1999-07-28 2002-10-30 Univ Washington Fluidic interconnect, interconnect manifold and microfluidic devices for internal delivery of gases and application of vacuum
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
US6524456B1 (en) 1999-08-12 2003-02-25 Ut-Battelle, Llc Microfluidic devices for the controlled manipulation of small volumes
AU6788100A (en) 1999-08-20 2001-03-19 Luminex Corporation Liquid array technology
US7163801B2 (en) 1999-09-01 2007-01-16 The Burnham Institute Methods for determining the prognosis for cancer patients using tucan
US6439103B1 (en) 1999-09-07 2002-08-27 Vector Engineering Co. Hydraulic and pneumatic cylinder construction
GB9921155D0 (en) 1999-09-08 1999-11-10 Medical Res Council Selection system
US7090983B1 (en) 1999-09-10 2006-08-15 Takashi Muramatsu Methods for detecting early cancer
US6274320B1 (en) 1999-09-16 2001-08-14 Curagen Corporation Method of sequencing a nucleic acid
TW507305B (en) 1999-09-18 2002-10-21 Samsung Electronics Co Ltd Method of measuring etched state of semiconductor wafer
US20010050881A1 (en) 1999-09-20 2001-12-13 Depaoli David W. Continuous flow, electrohydrodynamic micromixing apparatus and methods
US6998232B1 (en) 1999-09-27 2006-02-14 Quark Biotech, Inc. Methods of diagnosing bladder cancer
DE19947496C2 (en) 1999-10-01 2003-05-22 Agilent Technologies Inc Microfluidic microchip
US6506551B1 (en) 1999-10-08 2003-01-14 North Shore - Long Island Jewish Research Institute CD38 as a prognostic indicator in B cell chronic lymphocytic leukemia
US7393634B1 (en) 1999-10-12 2008-07-01 United States Of America As Represented By The Secretary Of The Air Force Screening for disease susceptibility by genotyping the CCR5 and CCR2 genes
CA2386858C (en) 1999-10-28 2011-12-20 Agensys, Inc. 36p6d5: secreted tumor antigen
US20020048777A1 (en) 1999-12-06 2002-04-25 Shujath Ali Method of diagnosing monitoring, staging, imaging and treating prostate cancer
DE19961257C2 (en) * 1999-12-18 2002-12-19 Inst Mikrotechnik Mainz Gmbh micromixer
US7510707B2 (en) 1999-12-20 2009-03-31 New York University Mt. Sinai School Of Medicine PAR, a novel marker gene for breast and prostate cancers
DE59904983D1 (en) 1999-12-23 2003-05-15 Muehlbauer Ernst Gmbh & Co Kg Dynamic mixer for dental impression materials
AU2610101A (en) 2000-01-06 2001-07-16 Caliper Technologies Corporation Methods and systems for monitoring intracellular binding reactions
EP1259545A2 (en) 2000-01-21 2002-11-27 Ludwig Institute For Cancer Research Small cell lung cancer associated antigens and uses therefor
US20010032053A1 (en) 2000-01-24 2001-10-18 Hielscher Andreas H. Imaging of a scattering medium using the equation of radiative transfer
EP1254367A4 (en) 2000-02-03 2006-07-05 Nanoscale Combinatorial Synthe Structure identification methods using mass measurements
US7582420B2 (en) 2001-07-12 2009-09-01 Illumina, Inc. Multiplex nucleic acid reactions
US6530944B2 (en) 2000-02-08 2003-03-11 Rice University Optically-active nanoparticles for use in therapeutic and diagnostic methods
US6355193B1 (en) 2000-03-01 2002-03-12 Gale Stott Method for making a faux stone concrete panel
GB2359765B (en) 2000-03-02 2003-03-05 Univ Newcastle Capillary reactor distribution device and method
US7485454B1 (en) 2000-03-10 2009-02-03 Bioprocessors Corp. Microreactor
JP2003527299A (en) 2000-03-10 2003-09-16 フロー・フォーカシング・インコーポレーテッド A method for fabricating optical fibers by converging highly viscous liquids
ITPR20000017A1 (en) 2000-03-15 2001-09-15 Lino Lanfranchi APPARATUS FOR THE CONTROL OF CONTAINERS, IN PARTICULAR PREFORMS
JP3442338B2 (en) 2000-03-17 2003-09-02 株式会社日立製作所 DNA analyzer, DNA base sequencer, DNA base sequence determination method, and reaction module
US20020012971A1 (en) 2000-03-20 2002-01-31 Mehta Tammy Burd PCR compatible nucleic acid sieving medium
US6565010B2 (en) 2000-03-24 2003-05-20 Praxair Technology, Inc. Hot gas atomization
WO2001071027A2 (en) 2000-03-24 2001-09-27 Micromet Ag mRNA AMPLIFICATION
AU2001249503A1 (en) 2000-03-27 2001-10-08 Thomas Jefferson University High specificity marker detection
DE10015109A1 (en) 2000-03-28 2001-10-04 Peter Walzel Processes and devices for producing drops of equal size
JP4927287B2 (en) 2000-03-31 2012-05-09 マイクロニックス、インコーポレーテッド Microfluidic device for protein crystallization
US7867763B2 (en) 2004-01-25 2011-01-11 Fluidigm Corporation Integrated chip carriers with thermocycler interfaces and methods of using the same
DK2206791T3 (en) 2000-04-10 2016-10-24 Taxon Biosciences Inc Methods of study and genetic analysis of populations
US6481453B1 (en) 2000-04-14 2002-11-19 Nanostream, Inc. Microfluidic branch metering systems and methods
AU2001255458A1 (en) 2000-04-18 2001-10-30 Waters Investments Limited Improved electrospray and other lc/ms interfaces
JP2001301154A (en) 2000-04-20 2001-10-30 Dainippon Printing Co Ltd Field jet sticking method of liquid having surface tension lowering upon application of voltage
US6469094B1 (en) 2000-04-28 2002-10-22 Xerox Corporation Polymerization processes
CN1189159C (en) 2000-05-05 2005-02-16 欧莱雅 Micro-capsule contg. water soluble beauty-care activity component water nuclear, and composition contg. same
WO2001090415A2 (en) 2000-05-20 2001-11-29 The Regents Of The University Of Michigan Method of producing a dna library using positional amplification
DE10025290B4 (en) 2000-05-22 2005-03-24 Fico I.T.M. S.A. Sun visor outer surfaces
JP2004502926A (en) 2000-05-24 2004-01-29 マイクロニックス、インコーポレーテッド Microfluidic device that produces a concentration gradient
US6686184B1 (en) 2000-05-25 2004-02-03 President And Fellows Of Harvard College Patterning of surfaces utilizing microfluidic stamps including three-dimensionally arrayed channel networks
US6645432B1 (en) 2000-05-25 2003-11-11 President & Fellows Of Harvard College Microfluidic systems including three-dimensionally arrayed channel networks
JP3939077B2 (en) 2000-05-30 2007-06-27 大日本スクリーン製造株式会社 Substrate cleaning device
US20060263888A1 (en) 2000-06-02 2006-11-23 Honeywell International Inc. Differential white blood count on a disposable card
US7049072B2 (en) 2000-06-05 2006-05-23 University Of South Florida Gene expression analysis of pluri-differentiated mesenchymal progenitor cells and methods for diagnosing a leukemic disease state
US7351376B1 (en) 2000-06-05 2008-04-01 California Institute Of Technology Integrated active flux microfluidic devices and methods
US6974667B2 (en) 2000-06-14 2005-12-13 Gene Logic, Inc. Gene expression profiles in liver cancer
US6592321B2 (en) 2000-08-03 2003-07-15 Demag Cranes & Components Gmbh Control and guiding device for manually operating a handling unit, and modular construction kit for making such devices of different configuration
FR2812942B1 (en) 2000-08-08 2002-10-31 Commissariat Energie Atomique POLARIZED LIGHT FLUORESCENCE IMAGING DEVICE
US20040005582A1 (en) 2000-08-10 2004-01-08 Nanobiodynamics, Incorporated Biospecific desorption microflow systems and methods for studying biospecific interactions and their modulators
US6682890B2 (en) 2000-08-17 2004-01-27 Protein Design Labs, Inc. Methods of diagnosing and determining prognosis of colorectal cancer
US20030148273A1 (en) 2000-08-26 2003-08-07 Shoulian Dong Target enrichment and amplification
JP2002071687A (en) 2000-08-31 2002-03-12 Canon Inc Screening method for variant gene
JP3993372B2 (en) 2000-09-13 2007-10-17 独立行政法人理化学研究所 Reactor manufacturing method
US6739036B2 (en) 2000-09-13 2004-05-25 Fuji Machine Mfg., Co., Ltd. Electric-component mounting system
DE10045586C2 (en) 2000-09-15 2002-07-18 Alstom Power Boiler Gmbh Process and device for cleaning smoke gases containing sulfur dioxide
US7829276B2 (en) 2000-09-18 2010-11-09 Thomas Jefferson University Methods of using CRCA-1 as a stomach and esophageal cancer marker
US6775405B1 (en) 2000-09-29 2004-08-10 Koninklijke Philips Electronics, N.V. Image registration system and method using cross-entropy optimization
EP1364052A2 (en) 2000-10-10 2003-11-26 Diversa Corporation High throughput or capillary-based screening for a bioactivity or biomolecule
JP2004537712A (en) 2000-10-18 2004-12-16 バーチャル・アレイズ・インコーポレーテッド Multiple cell analysis system
JP2004526949A (en) 2000-10-19 2004-09-02 ストラクチュラル ジェノミックス,インコーポレーテッド Apparatus and method for crystal identification by in situ X-ray diffraction
JP3946430B2 (en) 2000-10-20 2007-07-18 株式会社日立製作所 Valve timing control device for internal combustion engine
GB0026424D0 (en) 2000-10-28 2000-12-13 Ncimb Ltd Genetic analysis of microorganisms
EP1343973B2 (en) * 2000-11-16 2020-09-16 California Institute Of Technology Apparatus and methods for conducting assays and high throughput screening
KR100426453B1 (en) 2000-11-28 2004-04-13 김진우 Human cervical cancer 2 protooncogene and protein encoded by same, expression vector containing same, and cell transformed by said vector
AU2002236507A1 (en) 2000-11-29 2002-06-11 Cangen International Dap-kinase and hoxa9, two human genes associated with genesis, progression, and aggressiveness of non-small cell lung cancer
US6849423B2 (en) 2000-11-29 2005-02-01 Picoliter Inc Focused acoustics for detection and sorting of fluid volumes
AU2002243277A1 (en) 2000-12-07 2002-06-24 President And Fellows Of Harvard College Methods and compositions for encapsulating active agents
EP1350029B1 (en) 2001-01-08 2014-09-10 President and Fellows of Harvard College Valves and pumps for microfluidic systems and method for making microfluidic systems
KR100475649B1 (en) 2001-01-29 2005-03-10 배석철 RUNX3 gene showing anti-tumor activity and use thereof
ES2180405B1 (en) 2001-01-31 2004-01-16 Univ Sevilla DEVICE AND PROCEDURE FOR PRODUCING MULTICOMPONENT COMPOSITE LIQUID JEANS AND MULTICOMPONENT AND / OR MULTI-PAPER MICRO AND NANOMETRIC SIZE CAPSULES.
EP1355537A4 (en) 2001-01-31 2010-04-07 Kraft Foods Global Brands Llc Production of capsules and particles for improvement of food products
EP1371989A4 (en) 2001-02-23 2006-10-25 Japan Science & Tech Agency Small liquid particle handling method, and device therefor
EP1741482B1 (en) 2001-02-23 2008-10-15 Japan Science and Technology Agency Process and apparatus for producing microcapsules
US6936264B2 (en) 2001-03-05 2005-08-30 The Procter & Gamble Company Delivery of reactive agents via multiple emulsions for use in shelf stable products
NO325061B1 (en) 2001-03-06 2008-01-28 Photosense As Method and arrangement for determining the optical property of a multilayer tissue
EP1372848A4 (en) 2001-03-09 2006-08-09 Biomicro Systems Inc Method and system for microfluidic interfacing to arrays
US7297518B2 (en) 2001-03-12 2007-11-20 California Institute Of Technology Methods and apparatus for analyzing polynucleotide sequences by asynchronous base extension
US6717136B2 (en) 2001-03-19 2004-04-06 Gyros Ab Microfludic system (EDI)
US7010391B2 (en) 2001-03-28 2006-03-07 Handylab, Inc. Methods and systems for control of microfluidic devices
US20030064414A1 (en) 2001-03-30 2003-04-03 Benecky Michael J. Rapid assessment of coagulation activity in whole blood
AU2002307152A1 (en) 2001-04-06 2002-10-21 California Institute Of Technology Nucleic acid amplification utilizing microfluidic devices
US7318642B2 (en) 2001-04-10 2008-01-15 Essilor International (Compagnie Générale d'Optique) Progressive addition lenses with reduced unwanted astigmatism
US7756558B2 (en) 2004-05-24 2010-07-13 Trutouch Technologies, Inc. Apparatus and methods for mitigating the effects of foreign interferents on analyte measurements in spectroscopy
ES2287351T3 (en) 2001-04-25 2007-12-16 Cornell Research Foundation, Inc. DEVICES AND METHODS FOR CELL SYSTEMS BASED ON PHARMACOCINETICS.
US20020164271A1 (en) 2001-05-02 2002-11-07 Ho Winston Z. Wavelength-coded bead for bioassay and signature recogniton
KR100917731B1 (en) 2001-05-11 2009-09-15 파나소닉 주식회사 Biomolecular substrate and method and apparatus for examination and diagnosis using the same
US7320027B1 (en) 2001-05-14 2008-01-15 At&T Corp. System having generalized client-server computing
AU2002339871A1 (en) 2001-05-24 2002-12-03 New Objective, Inc. Method and apparatus for feedback controlled electrospray
JP3570714B2 (en) 2001-05-24 2004-09-29 株式会社リコー Developer container and image forming apparatus
US6806058B2 (en) 2001-05-26 2004-10-19 One Cell Systems, Inc. Secretions of proteins by encapsulated cells
US6719840B2 (en) 2001-06-08 2004-04-13 Syrrx, Inc. In situ crystal growth and crystallization
US6797056B2 (en) 2001-06-08 2004-09-28 Syrrx, Inc. Microfluidic method employing delivery of plural different fluids to same lumen
JP2002372016A (en) 2001-06-18 2002-12-26 Mitsubishi Heavy Ind Ltd Bolt connection structure
US7514209B2 (en) 2001-06-18 2009-04-07 Rosetta Inpharmatics Llc Diagnosis and prognosis of breast cancer patients
GB0114856D0 (en) 2001-06-18 2001-08-08 Medical Res Council Selection by avidity capture
GB0114854D0 (en) 2001-06-18 2001-08-08 Medical Res Council Selective gene amplification
US7171311B2 (en) 2001-06-18 2007-01-30 Rosetta Inpharmatics Llc Methods of assigning treatment to breast cancer patients
US6553944B1 (en) 2001-07-03 2003-04-29 Virginia A. Allen Wrist worn leash retaining device
US6656267B2 (en) 2001-07-10 2003-12-02 Structural Genomix, Inc. Tray for macromolecule crystallization and method of using the same
AU2002327220A1 (en) 2001-07-10 2003-01-29 Wisconsin Alumni Research Foundation Surface plasmon resonance imaging of micro-arrays
CA2353030A1 (en) 2001-07-13 2003-01-13 Willem Jager Caster mounted reel mower
US7314599B2 (en) 2001-07-17 2008-01-01 Agilent Technologies, Inc. Paek embossing and adhesion for microfluidic devices
JP2004535816A (en) 2001-07-20 2004-12-02 ボード オブ リージェンツ,ザ ユニバーシティ オブ テキサス システム Methods and compositions for HPV-related precancerous and cancerous growth comprising CIN
EP1412065A2 (en) 2001-07-27 2004-04-28 President And Fellows Of Harvard College Laminar mixing apparatus and methods
US7700293B2 (en) 2001-08-02 2010-04-20 The Regents Of The University Of Michigan Expression profile of prostate cancer
DE60234467D1 (en) 2001-08-16 2009-12-31 Us Health MOLECULAR PROPERTIES OF NON-SMALL CELL LUNG CANCER
WO2003015890A1 (en) 2001-08-20 2003-02-27 President And Fellows Of Harvard College Fluidic arrays and method of using
JP2003053996A (en) * 2001-08-22 2003-02-26 Canon Inc Ink supply mechanism and ink jet recorder comprising it
AU2002339865A1 (en) 2001-09-05 2003-03-18 The Children's Hospital Of Philadelphia Methods and compositions useful for diagnosis, staging, and treatment of cancers and tumors
US7390463B2 (en) 2001-09-07 2008-06-24 Corning Incorporated Microcolumn-based, high-throughput microfluidic device
FR2829948B1 (en) 2001-09-21 2004-07-09 Commissariat Energie Atomique METHOD FOR MOVING A FLUID OF INTEREST INTO A CAPILLARY AND FLUIDIC MICROSYSTEM
US6429148B1 (en) 2001-10-09 2002-08-06 Promos Technologies, Inc. Anisotropic formation process of oxide layers for vertical transistors
US6670142B2 (en) 2001-10-26 2003-12-30 The Regents Of The University Of California Method for screening combinatorial bead library, capturing cells from body fluids, and ligands for cancer cells
US20040076966A1 (en) 2001-10-30 2004-04-22 J. Brian Windsor Method and system for the co-isolation of cognate DNA, RNA and protein sequences and method for screening co-isolates for defined activities
US6464336B1 (en) 2001-10-31 2002-10-15 Eastman Kodak Company Ink jet printing with color-balanced ink drops mixed using bleached ink
US7371736B2 (en) 2001-11-07 2008-05-13 The Board Of Trustees Of The University Of Arkansas Gene expression profiling based identification of DKK1 as a potential therapeutic targets for controlling bone loss
US7308364B2 (en) 2001-11-07 2007-12-11 The University Of Arkansas For Medical Sciences Diagnosis of multiple myeloma on gene expression profiling
EP1451365A4 (en) 2001-11-13 2006-09-13 Rubicon Genomics Inc Dna amplification and sequencing using dna molecules generated by random fragmentation
JP2003149136A (en) 2001-11-13 2003-05-21 Shimadzu Corp Optical image measuring method
GB0127564D0 (en) 2001-11-16 2002-01-09 Medical Res Council Emulsion compositions
JP4381142B2 (en) 2001-11-16 2009-12-09 ザ ジョーンズ ホプキンス ユニバーシティー スクール オブ メディシン Prostate cancer detection
WO2003046208A2 (en) 2001-11-28 2003-06-05 Mj Bioworks Incorporated Parallel polymorphism scoring by amplification and error correction
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
US7057026B2 (en) 2001-12-04 2006-06-06 Solexa Limited Labelled nucleotides
GB0129374D0 (en) 2001-12-07 2002-01-30 Univ Brunel Test apparatus
US6800849B2 (en) 2001-12-19 2004-10-05 Sau Lan Tang Staats Microfluidic array devices and methods of manufacture and uses thereof
US20030198972A1 (en) 2001-12-21 2003-10-23 Erlander Mark G. Grading of breast cancer
US6949342B2 (en) 2001-12-21 2005-09-27 Whitehead Institute For Biomedical Research Prostate cancer diagnosis and outcome prediction by expression analysis
US20030144260A1 (en) 2002-01-03 2003-07-31 Yissum Research Development Company Of The Hebrew University Of Jerusalem Heterocyclic compounds, method of developing new drug leads and combinatorial libraries used in such method
CA2472649A1 (en) * 2002-01-08 2003-07-17 Japan Science And Technology Agency Pcr and hybridization methods utilizing electrostatic transportation and devices therefor
JPWO2003062418A1 (en) 2002-01-25 2005-05-19 オリンパス株式会社 Method and apparatus for detecting nucleic acid information
JP2003222633A (en) 2002-01-30 2003-08-08 Nippon Sheet Glass Co Ltd Microchip
ATE392262T1 (en) 2002-02-04 2008-05-15 Univ Sevilla DEVICE FOR GENERATING CAPILLARY BEAMS AND MICRO AND NANOMETER PARTICLES
US20030232356A1 (en) 2002-02-08 2003-12-18 Dooley Thomas P. Skin cell biomarkers and methods for identifying biomarkers using nucleic acid microarrays
AU2003211688A1 (en) 2002-02-11 2003-09-04 Rhodia Chimie Method for controlling the stability of emulsions and stabilized emulsions
US7101467B2 (en) 2002-03-05 2006-09-05 Caliper Life Sciences, Inc. Mixed mode microfluidic systems
EP2581739B1 (en) 2002-03-05 2015-11-04 Caliper Life Sciences, Inc. Microfluidic separation method with combined pressure and voltage control
EP1488007A4 (en) 2002-03-13 2006-05-03 Genomic Health Inc Gene expression profiling in biopsied tumor tissues
AU2003226679A1 (en) 2002-03-20 2003-09-29 Innovativebio.Biz Microcapsules with controlable permeability encapsulating a nucleic acid amplification reaction mixture and their use as reaction compartments for parallels reactions
US7348142B2 (en) 2002-03-29 2008-03-25 Veridex, Lcc Cancer diagnostic panel
US7147763B2 (en) 2002-04-01 2006-12-12 Palo Alto Research Center Incorporated Apparatus and method for using electrostatic force to cause fluid movement
WO2003085379A2 (en) 2002-04-01 2003-10-16 Fluidigm Corporation Microfluidic particle-analysis systems
GB0207533D0 (en) 2002-04-02 2002-05-08 Oxford Glycosciences Uk Ltd Protein
US7479371B2 (en) 2002-04-09 2009-01-20 Tokai University Method of judging leukemia, pre-leukemia or aleukemic malignant blood disease and diagnostic therefor
EP2031395A1 (en) 2002-05-08 2009-03-04 Panasonic Corporation Biomolecular substrate and test device
US7901939B2 (en) 2002-05-09 2011-03-08 University Of Chicago Method for performing crystallization and reactions in pressure-driven fluid plugs
EP2302389B1 (en) * 2002-05-09 2018-01-24 The University of Chicago Device and method for pressure-driven plug transport and reaction
CA2485169C (en) 2002-05-20 2013-03-26 Joseph C. Mcauliffe Peptide derivatives, and their use for the synthesis of silicon-based composite materials
US20040018525A1 (en) 2002-05-21 2004-01-29 Bayer Aktiengesellschaft Methods and compositions for the prediction, diagnosis, prognosis, prevention and treatment of malignant neoplasma
US20030219754A1 (en) 2002-05-23 2003-11-27 Oleksy Jerome E. Fluorescence polarization detection of nucleic acids
WO2003099407A2 (en) 2002-05-24 2003-12-04 Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung e.V. Method for transferring heterogeneous liquids in microchannels without the occurrence of mixing
US7198899B2 (en) 2002-06-03 2007-04-03 Chiron Corporation Use of NRG4, or inhibitors thereof, in the treatment of colon and pancreatic cancers
US7218959B2 (en) 2002-06-05 2007-05-15 Research Foundation Of City University Hybrid-dual-fourier tomographic algorithm for a fast three-dimensionial optical image reconstruction in turbid media
JP3883060B2 (en) 2002-06-17 2007-02-21 株式会社リガク Crystal evaluation equipment
US7776348B2 (en) 2002-06-26 2010-08-17 L'oreal S.A. Water-in-oil emulsion foundation
US20050019776A1 (en) 2002-06-28 2005-01-27 Callow Matthew James Universal selective genome amplification and universal genotyping system
US7244961B2 (en) 2002-08-02 2007-07-17 Silicon Valley Scientific Integrated system with modular microfluidic components
US7150412B2 (en) 2002-08-06 2006-12-19 Clean Earth Technologies Llc Method and apparatus for electrostatic spray
DK3587433T3 (en) 2002-08-23 2020-05-18 Illumina Cambridge Ltd MODIFIED NUCLEOTIDES
GB0220063D0 (en) 2002-08-29 2002-10-09 Isis Innovation Magnetic particle and process for preparation
FR2844039B1 (en) 2002-09-04 2005-04-29 Air Liquide PROCESS AND PLANT FOR PRODUCING OXYGEN AND RARE GASES BY CRYOGENIC AIR DISTILLATION
AU2003288902A1 (en) 2002-09-06 2004-04-08 Genteric, Inc. Microcapsules and methods of use
GB0221053D0 (en) 2002-09-11 2002-10-23 Medical Res Council Single-molecule in vitro evolution
US20050208495A1 (en) 2002-09-17 2005-09-22 Joseph Richard A Real-time detection of nucleic acid reactions
US7078681B2 (en) 2002-09-18 2006-07-18 Agilent Technologies, Inc. Multimode ionization source
US7357937B2 (en) 2002-09-24 2008-04-15 Therox, Inc. Perfluorocarbon emulsions with non-fluorinated surfactants
US7329545B2 (en) 2002-09-24 2008-02-12 Duke University Methods for sampling a liquid flow
WO2004031408A1 (en) 2002-09-30 2004-04-15 F.Hoffmann-La Roche Ag Oligonucleotides for genotyping thymidylate synthase gene
US6966990B2 (en) 2002-10-11 2005-11-22 Ferro Corporation Composite particles and method for preparing
ES2341103T3 (en) * 2002-10-23 2010-06-15 The Trustees Of Princeton University CONTINUOUS SEPARATION PROCEDURE OF PARTICLES USING RETICULATES OF ASYMETRICALLY ALIGNED OBSTACLES REGARDING FIELDS.
US20040136497A1 (en) 2002-10-30 2004-07-15 Meldrum Deirdre R Preparation of samples and sample evaluation
US20050048669A1 (en) * 2003-08-26 2005-03-03 Nanostream, Inc. Gasketless microfluidic device interface
US20040181343A1 (en) 2002-11-01 2004-09-16 Cellectricon Ab Computer program products and systems for rapidly changing the solution environment around sensors
US20040086892A1 (en) 2002-11-06 2004-05-06 Crothers Donald M. Universal tag assay
EP1563097A4 (en) * 2002-11-21 2006-03-15 Primera Biosystems Sampling method and apparatus for amplification reaction analysis
DE10254601A1 (en) 2002-11-22 2004-06-03 Ganymed Pharmaceuticals Ag Gene products differentially expressed in tumors and their use
US20040101822A1 (en) 2002-11-26 2004-05-27 Ulrich Wiesner Fluorescent silica-based nanoparticles
JP2004354364A (en) * 2002-12-02 2004-12-16 Nec Corp Fine particle manipulating unit, chip mounted with the same and detector, and method for separating, capturing and detecting protein
US7166575B2 (en) 2002-12-17 2007-01-23 Nastech Pharmaceutical Company Inc. Compositions and methods for enhanced mucosal delivery of peptide YY and methods for treating and preventing obesity
WO2004061410A2 (en) 2002-12-18 2004-07-22 Ciphergen Biosystems, Inc. Serum biomarkers in lung cancer
CA2510166A1 (en) 2002-12-20 2004-09-30 Caliper Life Sciences, Inc. Single molecule amplification and detection of dna
US20050042639A1 (en) 2002-12-20 2005-02-24 Caliper Life Sciences, Inc. Single molecule amplification and detection of DNA length
US8275554B2 (en) 2002-12-20 2012-09-25 Caliper Life Sciences, Inc. System for differentiating the lengths of nucleic acids of interest in a sample
EP1594694A4 (en) 2002-12-30 2010-01-20 Univ California Methods and apparatus for pathogen detection and analysis
CA2513308A1 (en) 2003-01-17 2004-08-05 Josef Michl Pancreatic cancer associated antigen, antibody thereto, and diagnostic and treatment methods
US20040142329A1 (en) 2003-01-17 2004-07-22 Ingeneus Corporation Probe conjugation to increase multiplex binding motif preference
WO2004065628A1 (en) 2003-01-21 2004-08-05 Guoliang Fu Quantitative multiplex detection of nucleic acids
US6832787B1 (en) * 2003-01-24 2004-12-21 Sandia National Laboratories Edge compression manifold apparatus
CA2514187A1 (en) 2003-01-24 2004-08-12 Bayer Pharmaceuticals Corporation Expression profiles for colon cancer and methods of use
US7323305B2 (en) * 2003-01-29 2008-01-29 454 Life Sciences Corporation Methods of amplifying and sequencing nucleic acids
US7575865B2 (en) * 2003-01-29 2009-08-18 454 Life Sciences Corporation Methods of amplifying and sequencing nucleic acids
US7361474B2 (en) 2003-02-24 2008-04-22 United States Of America As Represented By The Department Of Veterans Affairs Serum macrophage migration inhibitory factor (MIF) as marker for prostate cancer
WO2004075734A2 (en) 2003-02-25 2004-09-10 Inlight Solutions, Inc. DETERMINATION OF pH INCLUDING HEMOGLOBIN CORRECTION
EP1627045A2 (en) 2003-02-28 2006-02-22 Plexxikon, Inc. Pyk2 crystal structure and uses
US20040209299A1 (en) 2003-03-07 2004-10-21 Rubicon Genomics, Inc. In vitro DNA immortalization and whole genome amplification using libraries generated from randomly fragmented DNA
US20050123937A1 (en) 2003-03-07 2005-06-09 Thorp H. H. Methods for the electrochemical detection of target compounds
US7041481B2 (en) 2003-03-14 2006-05-09 The Regents Of The University Of California Chemical amplification based on fluid partitioning
US7045040B2 (en) 2003-03-20 2006-05-16 Asm Nutool, Inc. Process and system for eliminating gas bubbles during electrochemical processing
KR100620303B1 (en) 2003-03-25 2006-09-13 도요다 지도샤 가부시끼가이샤 Gas storage tank and its manufacturing method
GB0307428D0 (en) 2003-03-31 2003-05-07 Medical Res Council Compartmentalised combinatorial chemistry
GB0307403D0 (en) 2003-03-31 2003-05-07 Medical Res Council Selection by compartmentalised screening
US7378233B2 (en) 2003-04-12 2008-05-27 The Johns Hopkins University BRAF mutation T1796A in thyroid cancers
WO2004099379A2 (en) 2003-05-02 2004-11-18 Health Research, Inc. Use of jag2 expression in diagnosis of plasma cell disorders
WO2004099432A2 (en) 2003-05-02 2004-11-18 The Johns Hopkins University Identification of biomarkers for detecting pancreatic cancer
US7262059B2 (en) 2003-05-06 2007-08-28 Thrombodyne, Inc. Systems and methods for measuring fluid properties
US20060275915A1 (en) 2003-05-16 2006-12-07 Global Technologies (Nz) Ltd. Method and apparatus for mixing sample and reagent in a suspension fluid
DE10322893A1 (en) 2003-05-19 2004-12-16 Hans-Knöll-Institut für Naturstoff-Forschung e.V. Equipment for microtechnological structuring of fluids used in analytical or combinatorial biology or chemistry, has dosing, splitting and fusion devices in fluid pathway
WO2004103565A2 (en) 2003-05-19 2004-12-02 Hans-Knöll-Institut für Naturstoff-Forschung e.V. Device and method for structuring liquids and for dosing reaction liquids into liquid compartments immersed in a separation medium
JP4466991B2 (en) 2003-05-22 2010-05-26 英明 森山 Crystal growth apparatus and method
RU2005141456A (en) 2003-06-06 2006-06-10 Майкроникс, Инк. (Us) SYSTEM AND METHOD OF HEATING, COOLING AND THERMAL CYCLING IN A MICRO-LIQUID DEVICE
CA2528253A1 (en) 2003-06-12 2004-12-23 Spencer B. Gibson Methods for detecting cancer and monitoring cancer progression
PL3470535T3 (en) 2003-06-24 2020-08-24 Genomic Health, Inc. Prediction of likelihood of cancer recurrence
JP2005037346A (en) 2003-06-25 2005-02-10 Aisin Seiki Co Ltd Micro fluid control system
US7115230B2 (en) 2003-06-26 2006-10-03 Intel Corporation Hydrodynamic focusing devices
WO2004113571A2 (en) 2003-06-26 2004-12-29 Exonhit Therapeutics Sa Prostate specific genes and the use thereof as targets for prostate cancer therapy and diagnosis
AU2003903296A0 (en) 2003-06-30 2003-07-10 Raustech Pty Ltd Chemical compositions of matter
GB0315438D0 (en) * 2003-07-02 2003-08-06 Univ Manchester Analysis of mixed cell populations
US8048627B2 (en) 2003-07-05 2011-11-01 The Johns Hopkins University Method and compositions for detection and enumeration of genetic variations
CN102899398B (en) 2003-07-17 2015-09-30 环太平洋生物技术有限公司 For the marker that cancer of the stomach detects
WO2005008248A2 (en) 2003-07-18 2005-01-27 Georgetown University Diagnosis and treatment of cervical cancer
US20050014165A1 (en) 2003-07-18 2005-01-20 California Pacific Medical Center Biomarker panel for colorectal cancer
EP1654066B1 (en) 2003-07-31 2014-11-12 Handylab, Inc. Processing particle-containing samples
US7473531B1 (en) 2003-08-08 2009-01-06 Colora Corporation Pancreatic cancer targets and uses thereof
EP2662136A3 (en) * 2003-08-27 2013-12-25 President and Fellows of Harvard College Method for handling and mixing droplets
CN1860363B (en) 2003-08-28 2011-12-28 赛路拉公司 Methods and apparatus for sorting cells using an optical switch in a microfluidic channel network
WO2005023427A1 (en) 2003-09-05 2005-03-17 Stokes Bio Limited A microfluidic analysis system
US20070053896A1 (en) 2003-09-05 2007-03-08 Royal Women's Hospital Diagnostic marker for ovarian cancer
WO2005026691A2 (en) 2003-09-08 2005-03-24 Health Research, Inc. Detection of 13q14 chromosomal alterations
US7354706B2 (en) 2003-09-09 2008-04-08 The Regents Of The University Of Colorado, A Body Corporate Use of photopolymerization for amplification and detection of a molecular recognition event
EP1660674B1 (en) 2003-09-10 2010-03-17 Althea Technologies, Inc. Expression profiling using microarrays
US7504214B2 (en) 2003-09-19 2009-03-17 Biotheranostics, Inc. Predicting outcome with tamoxifen in breast cancer
WO2005028629A2 (en) 2003-09-19 2005-03-31 Applera Corporation Whole genome expression analysis system
CA2540141C (en) 2003-09-22 2012-09-04 Trisogen Biotechnology Limited Partnership Methods and kits useful for detecting an alteration in a locus copy number
US20060269971A1 (en) 2003-09-26 2006-11-30 Mount Sinai Hospital Methods for detecting prostate cancer
US7332280B2 (en) 2003-10-14 2008-02-19 Ronald Levy Classification of patients having diffuse large B-cell lymphoma based upon gene expression
WO2005039389A2 (en) 2003-10-22 2005-05-06 454 Corporation Sequence-based karyotyping
WO2005041884A2 (en) 2003-10-31 2005-05-12 Engineered Release Systems, Inc Polymer-based microstructures
GB0325653D0 (en) 2003-11-03 2003-12-10 Medical Res Council CST emulsions
CN101039951A (en) 2003-11-03 2007-09-19 基因信息公司 Liver cancer biomarkers
JP2007512811A (en) 2003-11-10 2007-05-24 インベスチゲン, インコーポレイテッド Methods for preparing nucleic acids for detection
US7169560B2 (en) 2003-11-12 2007-01-30 Helicos Biosciences Corporation Short cycle methods for sequencing polynucleotides
EP1533605A3 (en) 2003-11-19 2006-05-31 Aisin Seiki Kabushiki Kaisha Micro control system for transfer of liquids
WO2005049787A2 (en) 2003-11-24 2005-06-02 Yeda Research And Development Co.Ltd. Compositions and methods for in vitro sorting of molecular and cellular libraries
JP4949038B2 (en) 2003-12-01 2012-06-06 ダコ デンマーク アクティーゼルスカブ Methods and compositions for immunohistochemical detection
WO2005066613A1 (en) 2003-12-31 2005-07-21 President And Fellows Of Harvard College Assay device and method
JP4437202B2 (en) 2004-01-09 2010-03-24 学校法人慶應義塾 Telemedicine system for pigmentation site
US7569662B2 (en) 2004-01-27 2009-08-04 Compugen Ltd Nucleotide and amino acid sequences, and assays and methods of use thereof for diagnosis of lung cancer
US7368548B2 (en) 2004-01-27 2008-05-06 Compugen Ltd. Nucleotide and amino acid sequences, and assays and methods of use thereof for diagnosis of prostate cancer
CA2553833C (en) 2004-01-28 2012-10-02 454 Corporation Nucleic acid amplification with continuous flow emulsion
US20050186215A1 (en) 2004-02-04 2005-08-25 Kwok Tim T. CUDR as biomarker for cancer progression and therapeutic response
US20060195266A1 (en) 2005-02-25 2006-08-31 Yeatman Timothy J Methods for predicting cancer outcome and gene signatures for use therein
US7507532B2 (en) 2004-03-08 2009-03-24 Medigen Biotechnology Corporation Cancer specific gene MH15
KR100552706B1 (en) * 2004-03-12 2006-02-20 삼성전자주식회사 Method and apparatus for nucleic acid amplification
JP4938451B2 (en) 2004-03-23 2012-05-23 オンコセラピー・サイエンス株式会社 Methods for diagnosis of non-small cell lung cancer
BRPI0509193B8 (en) 2004-03-24 2021-07-27 Tripath Imaging Inc method for diagnosing high-grade cervical disease in a patient, and kit
US20050221339A1 (en) 2004-03-31 2005-10-06 Medical Research Council Harvard University Compartmentalised screening by microfluidic control
KR100885177B1 (en) 2004-04-12 2009-02-23 학교법인 포항공과대학교 Oligonucleotide for detecting target dna or rna
JP2007533798A (en) 2004-04-23 2007-11-22 クマチェヴァ、ユージニア Method for producing polymer particles having specific particle size, shape, form and composition
AU2005241093B2 (en) 2004-05-04 2008-06-05 Institute Of Virology Of Slovak Academy Of Sciences MN/CA IX/ CA9 and renal cancer prognosis
US7622281B2 (en) * 2004-05-20 2009-11-24 The Board Of Trustees Of The Leland Stanford Junior University Methods and compositions for clonal amplification of nucleic acid
US7828175B2 (en) 2004-05-21 2010-11-09 Pepsico, Inc. Beverage dispensing system with a head capable of dispensing plural different beverages
WO2006007207A2 (en) 2004-05-25 2006-01-19 Helicos Biosciences Corporation Methods and devices for nucleic acid sequence determination
US7799553B2 (en) 2004-06-01 2010-09-21 The Regents Of The University Of California Microfabricated integrated DNA analysis system
WO2005118138A1 (en) 2004-06-04 2005-12-15 Crystal Vision Microsystems Llc Device and process for continuous on-chip flow injection analysis
WO2006009910A2 (en) 2004-06-17 2006-01-26 The Regents Of The University Of California Time-resolved optometric fluorescence detection for skin diagnostics
US7991557B2 (en) 2004-06-19 2011-08-02 Genenews Corporation Computer system and methods for constructing biological classifiers and uses thereof
US20070154889A1 (en) 2004-06-25 2007-07-05 Veridex, Llc Methods and reagents for the detection of melanoma
US7079415B2 (en) 2004-06-30 2006-07-18 Stmicroelectronics, Inc. Magnetic random access memory element
US9477233B2 (en) 2004-07-02 2016-10-25 The University Of Chicago Microfluidic system with a plurality of sequential T-junctions for performing reactions in microdroplets
US7655470B2 (en) 2004-10-29 2010-02-02 University Of Chicago Method for manipulating a plurality of plugs and performing reactions therein in microfluidic systems
WO2006002641A1 (en) 2004-07-02 2006-01-12 Versamatrix A/S Spherical radiofrequency-encoded beads
JP2008506123A (en) 2004-07-09 2008-02-28 トリパス イメージング, インコーポレイテッド Methods and compositions for detection of ovarian cancer
US20060100788A1 (en) 2004-07-14 2006-05-11 Invitrogen Corporation Collections of matched biological reagents and methods for identifying matched reagents
US7670792B2 (en) 2004-07-14 2010-03-02 The Regents Of The University Of California Biomarkers for early detection of ovarian cancer
EP1985715A3 (en) 2004-07-16 2008-12-31 Oncomethylome Sciences, S.A. ESR1 and cervical cancer
US20060078475A1 (en) 2004-07-29 2006-04-13 Yu-Chong Tai Modular microfluidic packaging system
US20060021280A1 (en) 2004-07-30 2006-02-02 Hamilton Daniel B Reformer, and methods of making and using the same
US7405002B2 (en) 2004-08-04 2008-07-29 Agency For Science, Technology And Research Coated water-soluble nanoparticles comprising semiconductor core and silica coating
JP2006058652A (en) 2004-08-20 2006-03-02 Toshiba Corp Toner
US9566558B2 (en) 2004-09-09 2017-02-14 Institut Curie Device for manipulation of packets in micro-containers, in particular in microchannels
US20060068398A1 (en) 2004-09-24 2006-03-30 Cepheid Universal and target specific reagent beads for nucleic acid amplification
US7698287B2 (en) 2004-09-30 2010-04-13 Microsoft Corporation Design of spreadsheet functions for working with tables of data
WO2006035773A1 (en) 2004-09-30 2006-04-06 Ngk Insulators, Ltd. Liquid drop discharge piezoelectric device
US20060078894A1 (en) 2004-10-12 2006-04-13 Winkler Matthew M Methods and compositions for analyzing nucleic acids
US7482123B2 (en) 2004-11-05 2009-01-27 The Regents Of The University Of California Biomarkers for prostate cancer metastasis
US20130071836A9 (en) 2004-11-08 2013-03-21 Sungwhan An Colon cancer biomarker discovery
US7416851B2 (en) 2004-11-08 2008-08-26 Institut Pasteur Method of diagnosis/prognosis of human chronic lymphocytic leukemia comprising the profiling of LPL/ADAM genes
US20080004436A1 (en) 2004-11-15 2008-01-03 Yeda Research And Development Co. Ltd. At The Weizmann Institute Of Science Directed Evolution and Selection Using in Vitro Compartmentalization
JP2008521412A (en) 2004-11-30 2008-06-26 ベリデックス・エルエルシー Lung cancer prognosis judging means
US8103331B2 (en) 2004-12-06 2012-01-24 Cambridge Research & Instrumentation, Inc. Systems and methods for in-vivo optical imaging and measurement
US20060160762A1 (en) 2004-12-13 2006-07-20 Children's Medical Center Corporation Methods for the treatment, diagnosis, and prognosis of cancer
WO2006074430A2 (en) 2005-01-07 2006-07-13 The Johins Hopkins University Biomarkers for melanoma
US20090270702A1 (en) 2005-01-21 2009-10-29 Haishan Zeng Method and apparatus for measuring cancerous changes from reflectance spectral measurements obtained during endoscopic imaging
WO2006078841A1 (en) 2005-01-21 2006-07-27 President And Fellows Of Harvard College Systems and methods for forming fluidic droplets encapsulated in particles such as colloidal particles
US7442507B2 (en) 2005-01-24 2008-10-28 New York University School Of Medicine Methods for detecting circulating mutant BRAF DNA
WO2006081473A2 (en) 2005-01-28 2006-08-03 Children's Medical Center Corporation Methods for diagnosis and prognosis of epithelial cancers
EP2239342A3 (en) * 2005-02-01 2010-11-03 AB Advanced Genetic Analysis Corporation Reagents, methods and libraries for bead-based sequencing
US7407757B2 (en) 2005-02-10 2008-08-05 Population Genetics Technologies Genetic analysis by sequence-specific sorting
US7393665B2 (en) 2005-02-10 2008-07-01 Population Genetics Technologies Ltd Methods and compositions for tagging and identifying polynucleotides
US8486648B2 (en) 2005-02-16 2013-07-16 Dana-Farber Cancer Institute, Inc. Methods of detecting ovarian cancer
CN101120016A (en) 2005-02-17 2008-02-06 儿童医疗中心有限公司 Adamts-7 as a biomarker for cancers of epithelial origin
JP4982387B2 (en) 2005-02-18 2012-07-25 キヤノン ユー.エス. ライフ サイエンシズ, インコーポレイテッド Device and method for identifying genomic DNA of microorganisms
AU2006214105A1 (en) 2005-02-18 2006-08-24 Children's Medical Center Corporation Cyr61 as a biomarker for diagnosis and prognosis of cancers of epithelial origin
WO2006091776A2 (en) 2005-02-25 2006-08-31 The Brigham And Women's Hospital, Inc. Biomarkers for predicting prostate cancer progression
CN101133529A (en) 2005-03-04 2008-02-27 独立行政法人科学技术振兴机构 Wide-band optical amplifier
US7510842B2 (en) 2005-03-11 2009-03-31 Vermilllion, Inc. Biomarker for ovarian and endometrial cancer: hepcidin
FR2882939B1 (en) 2005-03-11 2007-06-08 Centre Nat Rech Scient FLUIDIC SEPARATION DEVICE
US20060234264A1 (en) 2005-03-14 2006-10-19 Affymetrix, Inc. Multiplex polynucleotide synthesis
US7604940B1 (en) * 2005-03-16 2009-10-20 Applied Biosystems, Llc Compositions and methods for analyzing isolated polynucleotides
US20060269934A1 (en) * 2005-03-16 2006-11-30 Applera Corporation Compositions and methods for clonal amplification and analysis of polynucleotides
WO2007084163A2 (en) 2005-04-06 2007-07-26 President And Fellows Of Harvard College Molecular characterization with carbon nanotube control
US7918244B2 (en) 2005-05-02 2011-04-05 Massachusetts Institute Of Technology Microfluidic bubble logic devices
US7570988B2 (en) 2005-05-02 2009-08-04 Wisconsin Alumni Research Foundation Method for extraction of optical properties from diffuse reflectance spectra
US8084001B2 (en) 2005-05-02 2011-12-27 Cornell Research Foundation, Inc. Photoluminescent silica-based sensors and methods of use
US7473530B2 (en) 2005-05-04 2009-01-06 Wayne State University Method to detect lung cancer
WO2006124458A2 (en) 2005-05-11 2006-11-23 Nanolytics, Inc. Method and device for conducting biochemical or chemical reactions at multiple temperatures
WO2006122311A2 (en) 2005-05-11 2006-11-16 The Trustees Of The University Of Pennsylvania Microfluidic chip
CA2607965A1 (en) 2005-05-18 2007-02-22 Cornell Research Foundation, Inc. Pharmacokinetic-based culture system with biological barriers
WO2007044091A2 (en) 2005-06-02 2007-04-19 Fluidigm Corporation Analysis using microfluidic partitioning devices
US8407013B2 (en) 2005-06-07 2013-03-26 Peter K. Rogan AB initio generation of single copy genomic probes
US7368242B2 (en) 2005-06-14 2008-05-06 Affymetrix, Inc. Method and kits for multiplex hybridization assays
US7494776B2 (en) 2005-07-07 2009-02-24 Beckman Coulter, Inc. Labeled complementary oligonucleotides to detect oligonucleotide-linked ligands
WO2007011867A2 (en) 2005-07-15 2007-01-25 Applera Corporation Fluid processing device and method
GB0514936D0 (en) 2005-07-20 2005-08-24 Solexa Ltd Preparation of templates for nucleic acid sequencing
FR2888912B1 (en) 2005-07-25 2007-08-24 Commissariat Energie Atomique METHOD FOR CONTROLLING COMMUNICATION BETWEEN TWO ZONES BY ELECTROWRINKING, DEVICE COMPRISING ISOLABLE ZONES AND OTHERS AND METHOD FOR PRODUCING SUCH DEVICE
US7632562B2 (en) 2005-08-04 2009-12-15 Eastman Kodak Company Universal print media
JP4756948B2 (en) 2005-08-08 2011-08-24 ベイバイオサイエンス株式会社 Flow cytometer and flow cytometry method
FR2893626B1 (en) 2005-11-18 2008-01-04 Inst Francais Du Petrole WELL FLUID COMPRISING A FLUORINATED LIQUID PHASE
EP1928571B1 (en) * 2005-08-22 2013-03-13 Life Technologies Corporation Device and method for microfluidic control of a first fluid in contact with a second fluid, wherein the first and second fluids are immiscible
US7666593B2 (en) 2005-08-26 2010-02-23 Helicos Biosciences Corporation Single molecule sequencing of captured nucleic acids
US7915030B2 (en) 2005-09-01 2011-03-29 Canon U.S. Life Sciences, Inc. Method and molecular diagnostic device for detection, analysis and identification of genomic DNA
KR20080050404A (en) 2005-09-02 2008-06-05 포라 가세이 고교 가부시키가이샤 Method of evaluating skin conditions adn method of estimating skin thickness
US20090098057A1 (en) 2007-10-16 2009-04-16 Shiying Zheng Silica-cored carrier particle
US7556776B2 (en) 2005-09-08 2009-07-07 President And Fellows Of Harvard College Microfluidic manipulation of fluids and reactions
GB2429385C (en) 2005-09-23 2008-04-24 Astron Clinica Ltd Image processing method and apparatus.
US9360526B2 (en) 2005-10-24 2016-06-07 The Johns Hopkins University Methods for beaming
JP2009515167A (en) 2005-11-02 2009-04-09 バイエル ヘルスケア エルエルシー Cancer prediction and prognosis testing methods, and cancer treatment monitoring
US20120021930A1 (en) 2005-11-22 2012-01-26 Stichting Dienst Landbouwkundig Onderzoek Multiplex Nucleic Acid Detection
US7358231B1 (en) 2005-12-01 2008-04-15 Applera Corporation Pancreatic cancer secreted targets and uses thereof
US7846664B2 (en) 2005-12-07 2010-12-07 The Regents Of The University Of California Diagnosis and treatment of chronic lymphocytic leukemia (CLL)
WO2007070542A2 (en) 2005-12-12 2007-06-21 The Government Of The United States Of America As Represented By The Secretary Of The Deptartment Ofhealth And Human Services Nanoprobes for detection or modification of molecules
ES2277785B1 (en) 2005-12-21 2008-06-16 Oryzon Genomics, S.A. METHOD OF DIFFERENTIAL EXPRESSION ANALYSIS IN COLORECTAL CANCER.
CA2629071A1 (en) 2005-12-21 2007-06-28 F. Hoffmann-La Roche Ag Method of assessing colorectal cancer by measuring hemoglobin and m2-pk in a stool sample
US20100137163A1 (en) 2006-01-11 2010-06-03 Link Darren R Microfluidic Devices and Methods of Use in The Formation and Control of Nanoreactors
EP1987162A4 (en) 2006-01-23 2009-11-25 Population Genetics Technologi Nucleic acid analysis using sequence tokens
US7537897B2 (en) 2006-01-23 2009-05-26 Population Genetics Technologies, Ltd. Molecular counting
JP2009524438A (en) 2006-01-27 2009-07-02 トライパス イメージング インコーポレイテッド Methods and compositions for identifying patients likely to have ovarian cancer
HUE045358T2 (en) 2006-02-02 2019-12-30 Univ Leland Stanford Junior Non-invasive fetal genetic screening by digital analysis
WO2007091228A1 (en) 2006-02-07 2007-08-16 Stokes Bio Limited A liquid bridge and system
US8735169B2 (en) 2006-02-07 2014-05-27 Stokes Bio Limited Methods for analyzing agricultural and environmental samples
WO2007092627A2 (en) 2006-02-09 2007-08-16 University Of South Florida Detection of cancer by elevated levels of bcl-2
JP5237126B2 (en) 2006-03-01 2013-07-17 キージーン ナムローゼ フェンノートシャップ Methods for detecting gene-related sequences based on high-throughput sequences using ligation assays
US20070292869A1 (en) 2006-03-02 2007-12-20 Ppd Biomarker Discovery Sciences, Llc Compositions and Methods for Analyzing Renal Cancer
MX2008011356A (en) 2006-03-03 2008-09-15 Veridex Llc Molecular assay to predict recurrence of dukes' b colon cancer.
WO2007107358A1 (en) 2006-03-21 2007-09-27 Dsm Ip Assets B.V. Microparticles comprising a crosslinked polymer
SG170061A1 (en) 2006-03-24 2011-04-29 Phenomenome Discoveries Inc Biomarkers useful for diagnosing prostate cancer, and methods thereof
US20090181864A1 (en) 2006-03-31 2009-07-16 Nam Trung Nguyen Active control for droplet-based microfluidics
JP5122555B2 (en) 2006-03-31 2013-01-16 ソレクサ・インコーポレイテッド Synthetic sequencing system and apparatus
US8492168B2 (en) 2006-04-18 2013-07-23 Advanced Liquid Logic Inc. Droplet-based affinity assays
US8613889B2 (en) 2006-04-13 2013-12-24 Advanced Liquid Logic, Inc. Droplet-based washing
US7282337B1 (en) 2006-04-14 2007-10-16 Helicos Biosciences Corporation Methods for increasing accuracy of nucleic acid sequencing
US8980198B2 (en) 2006-04-18 2015-03-17 Advanced Liquid Logic, Inc. Filler fluids for droplet operations
US7901947B2 (en) 2006-04-18 2011-03-08 Advanced Liquid Logic, Inc. Droplet-based particle sorting
US7439014B2 (en) * 2006-04-18 2008-10-21 Advanced Liquid Logic, Inc. Droplet-based surface modification and washing
US20070259368A1 (en) 2006-05-03 2007-11-08 Genomictree, Inc. Gastric cancer biomarker discovery
US7702468B2 (en) 2006-05-03 2010-04-20 Population Diagnostics, Inc. Evaluating genetic disorders
US9562837B2 (en) 2006-05-11 2017-02-07 Raindance Technologies, Inc. Systems for handling microfludic droplets
USRE49362E1 (en) 2006-05-18 2023-01-10 Illumina Cambridge Limited Dye compounds and the use of their labelled conjugates
EP2021515A4 (en) 2006-05-26 2010-06-02 Meltzer Stephen J Methylated promoters as biomarkers of colon cancer
EP2636755A1 (en) 2006-05-26 2013-09-11 AltheaDx Incorporated Biochemical analysis of partitioned cells
FR2901717A1 (en) 2006-05-30 2007-12-07 Centre Nat Rech Scient METHOD FOR TREATING DROPS IN A MICROFLUIDIC CIRCUIT
EP2589668A1 (en) 2006-06-14 2013-05-08 Verinata Health, Inc Rare cell analysis using sample splitting and DNA tags
CA2656022C (en) 2006-06-19 2017-10-17 The Johns Hopkins University Single-molecule pcr on microparticles in water-in-oil emulsions
US7629124B2 (en) * 2006-06-30 2009-12-08 Canon U.S. Life Sciences, Inc. Real-time PCR in micro-channels
KR100813169B1 (en) 2006-07-21 2008-03-17 삼성전자주식회사 Optical sensor module having tilt and body fat measurement appratus of having the optical sensor module
WO2008011709A1 (en) 2006-07-24 2008-01-31 Miraculins Inc. Biomarkers for use in the diagnosis and treatment of colorectal cancer
WO2008021123A1 (en) 2006-08-07 2008-02-21 President And Fellows Of Harvard College Fluorocarbon emulsion stabilizing surfactants
US20080050723A1 (en) 2006-08-23 2008-02-28 Nabil Belacel Molecular method for diagnosis of colon cancer
US8932994B2 (en) 2006-08-24 2015-01-13 Illumina, Inc. Method for retaining even coverage of short insert libraries
EP2058396A4 (en) 2006-08-31 2010-06-30 Toyo Seikan Kaisha Ltd Nucleic acid amplification method
US7811778B2 (en) 2006-09-06 2010-10-12 Vanderbilt University Methods of screening for gastrointestinal cancer
DE102006042040B4 (en) 2006-09-07 2013-04-18 Siemens Audiologische Technik Gmbh A method of adapting a hearing aid using a genetic feature and arrangement for performing the method
US20080081330A1 (en) 2006-09-28 2008-04-03 Helicos Biosciences Corporation Method and devices for analyzing small RNA molecules
CA2665053C (en) 2006-09-29 2016-08-23 The Administrators Of The Tulane Educational Fund Methods and devices for simultaneously monitoring the characteristics of microscopic particles in suspension and the characteristics of soluble components during reactions
AU2007324128B2 (en) 2006-10-10 2013-10-10 The Henry M. Jackson Foundation For The Advancement Of Military Medicine, Inc. Prostate cancer-specific alterations in ERG gene expression and detection and treatment methods based on those alterations
TWM319361U (en) 2006-10-20 2007-09-21 Tai Sol Electronics Co Ltd Flexible heat pipe
US8841116B2 (en) 2006-10-25 2014-09-23 The Regents Of The University Of California Inline-injection microdevice and microfabricated integrated DNA analysis system using same
WO2008069906A2 (en) 2006-11-14 2008-06-12 The Regents Of The University Of California Digital expression of gene analysis
WO2008058384A1 (en) 2006-11-15 2008-05-22 University Health Network Materials and methods for prognosing lung cancer survival
US20090093551A1 (en) 2006-12-08 2009-04-09 Bhatia Sangeeta N Remotely triggered release from heatable surfaces
WO2008073290A1 (en) 2006-12-08 2008-06-19 The Board Of Trustees Of The University Of Arkansas Tp53 gene expression and uses thereof
TW200825414A (en) 2006-12-08 2008-06-16 Univ Nat Taiwan Biomarker molecule of gastrointestinal disease and measurement method thereof
US8349167B2 (en) 2006-12-14 2013-01-08 Life Technologies Corporation Methods and apparatus for detecting molecular interactions using FET arrays
US8262900B2 (en) 2006-12-14 2012-09-11 Life Technologies Corporation Methods and apparatus for measuring analytes using large scale FET arrays
EP2639579B1 (en) 2006-12-14 2016-11-16 Life Technologies Corporation Apparatus for measuring analytes using large scale FET arrays
US8198027B2 (en) 2006-12-21 2012-06-12 Gen-Probe Incorporated Methods and compositions for nucleic acid amplification
US8338166B2 (en) 2007-01-04 2012-12-25 Lawrence Livermore National Security, Llc Sorting, amplification, detection, and identification of nucleic acid subsequences in a complex mixture
US20080171078A1 (en) 2007-01-12 2008-07-17 Mark Gray Uniformly sized liposomes
US7807393B2 (en) 2007-01-29 2010-10-05 Northwestern University Biomarkers for prostate cancer
EP2121983A2 (en) 2007-02-02 2009-11-25 Illumina Cambridge Limited Methods for indexing samples and sequencing multiple nucleotide templates
US20090075264A1 (en) 2007-02-02 2009-03-19 Orion Genomics Llc Gene methylation in liver cancer diagnosis
WO2008115626A2 (en) 2007-02-05 2008-09-25 Microchip Biotechnologies, Inc. Microfluidic and nanofluidic devices, systems, and applications
WO2008097559A2 (en) 2007-02-06 2008-08-14 Brandeis University Manipulation of fluids and reactions in microfluidic systems
US9029085B2 (en) 2007-03-07 2015-05-12 President And Fellows Of Harvard College Assays and other reactions involving droplets
JP2010521670A (en) 2007-03-12 2010-06-24 ダナ ファーバー キャンサー インスティテュート,インコーポレイテッド Use of FANCI and drugs that modulate FANCI in the prognosis, in diagnosis, and in the treatment of cancer
EP2664916B1 (en) 2007-04-02 2017-02-08 Acoustic Cytometry Systems, Inc. Method for manipulating a fluid medium within a flow cell using acoustic focusing
US20090062144A1 (en) 2007-04-03 2009-03-05 Nancy Lan Guo Gene signature for prognosis and diagnosis of lung cancer
US8592221B2 (en) 2007-04-19 2013-11-26 Brandeis University Manipulation of fluids, fluid components and reactions in microfluidic systems
US8691164B2 (en) 2007-04-20 2014-04-08 Celula, Inc. Cell sorting system and methods
US20100130369A1 (en) 2007-04-23 2010-05-27 Advanced Liquid Logic, Inc. Bead-Based Multiplexed Analytical Methods and Instrumentation
CA2686212A1 (en) 2007-05-04 2008-11-13 Dermtech International Diagnosis of melanoma by nucleic acid analysis
US7901888B2 (en) 2007-05-09 2011-03-08 The Regents Of The University Of California Multigene diagnostic assay for malignant thyroid neoplasm
US20090029372A1 (en) 2007-05-14 2009-01-29 Kobenhavns Universitet Adam12 as a biomarker for bladder cancer
CN101720359A (en) 2007-06-01 2010-06-02 454生命科学公司 System and meth0d for identification of individual samples from a multiplex mixture
WO2009045579A2 (en) 2007-06-14 2009-04-09 The Regents Of The University Of California Multimodal imaging probes for in vivo targeted and non-targeted imaging and therapeutics
US7820386B2 (en) 2007-06-15 2010-10-26 National Defense Medical Center Cancer screening method
CN103418295B (en) 2007-06-21 2015-11-18 简.探针公司 For the instruments and methods of the content of hybrid detection chamber
CN101855553B (en) 2007-06-29 2014-06-11 韩国安国药品株式会社 Predictive markers for ovarian cancer
US20090017463A1 (en) 2007-07-10 2009-01-15 Vanderbilt University Methods for predicting prostate cancer recurrence
US20090068170A1 (en) 2007-07-13 2009-03-12 President And Fellows Of Harvard College Droplet-based selection
EP2167634A4 (en) 2007-07-16 2013-11-06 California Inst Of Techn Arrays, substrates, devices, methods and systems for detecting target molecules
DE102007034020A1 (en) 2007-07-20 2009-01-22 Biotronik Crm Patent Ag Active element and battery and method of making same
US8454906B2 (en) 2007-07-24 2013-06-04 The Regents Of The University Of California Microfabricated droplet generator for single molecule/cell genetic analysis in engineered monodispersed emulsions
US20090042737A1 (en) 2007-08-09 2009-02-12 Katz Andrew S Methods and Devices for Correlated, Multi-Parameter Single Cell Measurements and Recovery of Remnant Biological Material
WO2009029229A2 (en) 2007-08-24 2009-03-05 President And Fellows Of Harvard College Ferrofluid emulsions, particles, and systems and methods for making and using the same
US20090087849A1 (en) 2007-09-06 2009-04-02 Tripath Imaging, Inc. Nucleic acid-based methods and compositions for the detection of ovarian cancer
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
WO2009037266A2 (en) 2007-09-17 2009-03-26 Universite Louis Pasteur Method for detecting or quantifying a truncating mutation
CA2697640C (en) 2007-09-21 2016-06-21 Katholieke Universiteit Leuven Tools and methods for genetic tests using next generation sequencing
US8268564B2 (en) 2007-09-26 2012-09-18 President And Fellows Of Harvard College Methods and applications for stitched DNA barcodes
WO2009049214A2 (en) 2007-10-12 2009-04-16 Northwestern University Inhibition and treatment of prostate cancer metastasis
JP2011500041A (en) 2007-10-16 2011-01-06 エフ.ホフマン−ラ ロシュ アーゲー High resolution and high efficiency HLA genotyping by clonal sequencing
US20100086914A1 (en) 2008-10-03 2010-04-08 Roche Molecular Systems, Inc. High resolution, high throughput hla genotyping by clonal sequencing
CN101910415B (en) 2007-11-07 2015-02-25 不列颠哥伦比亚大学 Microfluidic device and method of using same
US7923517B2 (en) 2007-11-09 2011-04-12 Ricoh Company, Ltd. Polymer microparticles and production method for the same
US8462269B2 (en) 2007-11-16 2013-06-11 Mediatek Inc. Devices and methods for extracting a synchronization signal from a video signal
US8592150B2 (en) 2007-12-05 2013-11-26 Complete Genomics, Inc. Methods and compositions for long fragment read sequencing
EP2247638A1 (en) 2007-12-20 2010-11-10 DSM IP Assets B.V. Hybrid polyurethane block copolymers with thermoplastic processability and thermoset properties
WO2009085215A1 (en) 2007-12-21 2009-07-09 President And Fellows Of Harvard College Systems and methods for nucleic acid sequencing
CN101646786A (en) 2007-12-26 2010-02-10 爱科来株式会社 Method for amplifying target nucleic acid sequence and probe used for the same
US20090226971A1 (en) 2008-01-22 2009-09-10 Neil Reginald Beer Portable Rapid Microfluidic Thermal Cycler for Extremely Fast Nucleic Acid Amplification
US9170060B2 (en) 2008-01-22 2015-10-27 Lawrence Livermore National Security, Llc Rapid microfluidic thermal cycler for nucleic acid amplification
JP5255072B2 (en) 2008-01-24 2013-08-07 ボールター インク Method for identifying malignant and benign tissue lesions
NZ588545A (en) 2008-03-18 2013-05-31 Balter Inc Optical method for determining morphological parameters and physiological properties of a lesion
US20090246788A1 (en) 2008-04-01 2009-10-01 Roche Nimblegen, Inc. Methods and Assays for Capture of Nucleic Acids
JP2009265751A (en) 2008-04-22 2009-11-12 Oki Electric Ind Co Ltd Character recognition device, optical character recognition system and character recognition program
US9664619B2 (en) 2008-04-28 2017-05-30 President And Fellows Of Harvard College Microfluidic device for storage and well-defined arrangement of droplets
US20110104725A1 (en) 2008-05-02 2011-05-05 Advanced Liquid Logic, Inc. Method of Effecting Coagulation in a Droplet
US8852952B2 (en) 2008-05-03 2014-10-07 Advanced Liquid Logic, Inc. Method of loading a droplet actuator
US20100075436A1 (en) 2008-05-06 2010-03-25 Urdea Michael S Methods for use with nanoreactors
US9068181B2 (en) 2008-05-23 2015-06-30 The General Hospital Corporation Microfluidic droplet encapsulation
AU2009262959A1 (en) 2008-06-27 2009-12-30 Massachusetts Institute Of Technology Microfluidic droplets for metabolic engineering and other applications
US8004661B2 (en) 2008-06-30 2011-08-23 Microbix Biosystems Inc. Method and apparatus for sorting cells
US7888034B2 (en) 2008-07-01 2011-02-15 454 Life Sciences Corporation System and method for detection of HIV tropism variants
US20110274706A1 (en) 2010-05-04 2011-11-10 General Electric Company Nucleic acid delivery vehicle and uses thereof
US20110275063A1 (en) 2008-07-11 2011-11-10 President And Fellows Of Harvard College Systems and methods of droplet-based selection
FR2934050B1 (en) 2008-07-15 2016-01-29 Univ Paris Curie METHOD AND DEVICE FOR READING EMULSION
EP4047367A1 (en) 2008-07-18 2022-08-24 Bio-Rad Laboratories, Inc. Method for detecting target analytes with droplet libraries
US20100035252A1 (en) 2008-08-08 2010-02-11 Ion Torrent Systems Incorporated Methods for sequencing individual nucleic acids under tension
DK3663411T3 (en) 2008-08-12 2022-01-10 Stokes Bio Ltd PROCEDURES FOR DIGITAL PCR
WO2010033200A2 (en) 2008-09-19 2010-03-25 President And Fellows Of Harvard College Creation of libraries of droplets and related species
WO2011120024A1 (en) 2010-03-25 2011-09-29 Quantalife, Inc. Droplet generation for droplet-based assays
US9156010B2 (en) 2008-09-23 2015-10-13 Bio-Rad Laboratories, Inc. Droplet-based assay system
US9132394B2 (en) 2008-09-23 2015-09-15 Bio-Rad Laboratories, Inc. System for detection of spaced droplets
CA2914882C (en) 2008-10-02 2017-07-25 T&M Inventions, Llc Support for roof penetrating structures
US20100137143A1 (en) 2008-10-22 2010-06-03 Ion Torrent Systems Incorporated Methods and apparatus for measuring analytes
US20100301398A1 (en) 2009-05-29 2010-12-02 Ion Torrent Systems Incorporated Methods and apparatus for measuring analytes
US8546128B2 (en) 2008-10-22 2013-10-01 Life Technologies Corporation Fluidics system for sequential delivery of reagents
US9080211B2 (en) 2008-10-24 2015-07-14 Epicentre Technologies Corporation Transposon end compositions and methods for modifying nucleic acids
US20110301042A1 (en) 2008-11-11 2011-12-08 Helicos Biosciences Corporation Methods of sample encoding for multiplex analysis of samples by single molecule sequencing
EP2365997B1 (en) 2008-11-26 2019-03-20 University College Cork - National University of Ireland, Cork A process for preparing silica microparticles
US9404924B2 (en) 2008-12-04 2016-08-02 Massachusetts Institute Of Technology Method of performing one-step, single cell RT-PCR
EP3150724A1 (en) 2008-12-19 2017-04-05 President and Fellows of Harvard College Particle-assisted nucleic acid sequencing
US11634747B2 (en) 2009-01-21 2023-04-25 Streck Llc Preservation of fetal nucleic acids in maternal plasma
JP5457222B2 (en) 2009-02-25 2014-04-02 エフ.ホフマン−ラ ロシュ アーゲー Miniaturized high-throughput nucleic acid analysis
JP2010198393A (en) 2009-02-26 2010-09-09 Alpine Electronics Inc Map display device
EP2230312A1 (en) 2009-03-19 2010-09-22 Helmholtz-Zentrum für Infektionsforschung GmbH Probe compound for detecting and isolating enzymes and means and methods using the same
US8481698B2 (en) 2009-03-19 2013-07-09 The President And Fellows Of Harvard College Parallel proximity ligation event analysis
EP3415235A1 (en) 2009-03-23 2018-12-19 Raindance Technologies Inc. Manipulation of microfluidic droplets
SG10201402770YA (en) 2009-04-02 2014-08-28 Fluidigm Corp Multi-primer amplification method for barcoding of target nucleic acids
FR2945545B1 (en) 2009-05-14 2011-08-05 Univ Aix Marseille Ii METHOD FOR DETECTION OF PROCARYOTE DNA EXTRACTED FROM A SAMPLE SAMPLE
US8574835B2 (en) 2009-05-29 2013-11-05 Life Technologies Corporation Scaffolded nucleic acid polymer particles and methods of making and using
US8673627B2 (en) 2009-05-29 2014-03-18 Life Technologies Corporation Apparatus and methods for performing electrochemical reactions
EP2446278B1 (en) 2009-06-26 2021-11-17 President and Fellows of Harvard College Fluid injection
KR101378214B1 (en) 2009-06-29 2014-03-27 가부시끼가이샤 도시바 Sample analysis method and assay kit for use in the method
US20130288254A1 (en) 2009-08-13 2013-10-31 Advanced Liquid Logic, Inc. Droplet Actuator and Droplet-Based Techniques
EP2473625B1 (en) 2009-09-01 2018-03-07 Koninklijke Philips N.V. Devices and methods for microarray selection
EP2473618B1 (en) 2009-09-02 2015-03-04 Bio-Rad Laboratories, Inc. System for mixing fluids by coalescence of multiple emulsions
US9625454B2 (en) 2009-09-04 2017-04-18 The Research Foundation For The State University Of New York Rapid and continuous analyte processing in droplet microfluidic devices
US10520500B2 (en) 2009-10-09 2019-12-31 Abdeslam El Harrak Labelled silica-based nanomaterial with enhanced properties and uses thereof
JP2013511991A (en) 2009-11-25 2013-04-11 クアンタライフ, インコーポレイテッド Methods and compositions for detecting genetic material
EP2336354A1 (en) 2009-12-18 2011-06-22 Roche Diagnostics GmbH A method for the detection of a RNA molecule, a kit and a use related thereof
WO2011079176A2 (en) 2009-12-23 2011-06-30 Raindance Technologies, Inc. Microfluidic systems and methods for reducing the exchange of molecules between droplets
WO2011090556A1 (en) 2010-01-19 2011-07-28 Verinata Health, Inc. Methods for determining fraction of fetal nucleic acid in maternal samples
US20110257031A1 (en) 2010-02-12 2011-10-20 Life Technologies Corporation Nucleic acid, biomolecule and polymer identifier codes
US9494520B2 (en) 2010-02-12 2016-11-15 Raindance Technologies, Inc. Digital analyte analysis
US10351905B2 (en) 2010-02-12 2019-07-16 Bio-Rad Laboratories, Inc. Digital analyte analysis
US8535889B2 (en) 2010-02-12 2013-09-17 Raindance Technologies, Inc. Digital analyte analysis
US9366632B2 (en) 2010-02-12 2016-06-14 Raindance Technologies, Inc. Digital analyte analysis
US9399797B2 (en) 2010-02-12 2016-07-26 Raindance Technologies, Inc. Digital analyte analysis
JP5901046B2 (en) 2010-02-19 2016-04-06 国立大学法人 千葉大学 Novel alternative splicing variant of OATP1B3 mRNA
HUE027972T2 (en) 2010-02-25 2016-11-28 Advanced Liquid Logic Inc Method of making nucleic acid libraries
US20110223314A1 (en) 2010-03-10 2011-09-15 Xiaoxiao Zhang Efficient microencapsulation
US8865675B2 (en) 2010-05-12 2014-10-21 Protiva Biotherapeutics, Inc. Compositions and methods for silencing apolipoprotein B
EP2596127A2 (en) 2010-07-23 2013-05-29 Esoterix Genetic Laboratories, LLC Identification of differentially represented fetal or maternal genomic regions and uses thereof
GB2482911A (en) 2010-08-20 2012-02-22 Sphere Fluidics Ltd Microdroplet emulsion system
EP2617061B1 (en) 2010-09-15 2021-06-30 Life Technologies Corporation Methods and apparatus for measuring analytes
US9562897B2 (en) 2010-09-30 2017-02-07 Raindance Technologies, Inc. Sandwich assays in droplets
US20120088691A1 (en) 2010-10-08 2012-04-12 Gao Chen Highly multiplexed real-time pcr using encoded microbeads
GB2512213B (en) 2010-10-08 2015-02-11 Harvard College High-throughput single cell barcoding
GB2498163B (en) 2010-10-08 2015-07-01 Harvard College High-throughput immune sequencing
US20130225623A1 (en) 2010-10-27 2013-08-29 Mount Sinai School Of Medicine Methods of Treating Psychiatric or Neurological Disorders with MGLUR Antagonists
US8829171B2 (en) 2011-02-10 2014-09-09 Illumina, Inc. Linking sequence reads using paired code tags
US8278711B2 (en) 2010-11-23 2012-10-02 General Electric Company Semiconductor device and method of making the same
WO2012083225A2 (en) 2010-12-16 2012-06-21 Gigagen, Inc. System and methods for massively parallel analysis of nycleic acids in single cells
US20120167142A1 (en) 2010-12-23 2012-06-28 Eldon Technology Limited Methods and apparatuses to facilitate preselection of programming preferences
CA2965074A1 (en) 2010-12-27 2012-07-05 Abbott Molecular Inc. Quantitating high titer samples by digital pcr
US9156326B2 (en) 2011-01-26 2015-10-13 Firestone Industrial Products Company, Llc Gas spring piston as well as gas spring assembly, suspension system, kit and method including same
US20120244043A1 (en) 2011-01-28 2012-09-27 Sean Leblanc Elastomeric gasket for fluid interface to a microfluidic chip
JP6069224B2 (en) 2011-01-31 2017-02-01 アプライズ バイオ, インコーポレイテッド Methods for identifying multiple epitopes in a cell
WO2012106668A2 (en) 2011-02-03 2012-08-09 Fluidigm Corporation Multifunctional probe-primers
CA2826748C (en) 2011-02-09 2020-08-04 Bio-Rad Laboratories, Inc. Method of detecting variations in copy number of a target nucleic acid
WO2012109604A1 (en) 2011-02-11 2012-08-16 Raindance Technologies, Inc. Thermocycling device for nucleic acid amplification and methods of use
EP3412778A1 (en) 2011-02-11 2018-12-12 Raindance Technologies, Inc. Methods for forming mixed droplets
EP3736281A1 (en) 2011-02-18 2020-11-11 Bio-Rad Laboratories, Inc. Compositions and methods for molecular labeling
WO2012129363A2 (en) 2011-03-24 2012-09-27 President And Fellows Of Harvard College Single cell nucleic acid detection and analysis
JP5711583B2 (en) 2011-03-28 2015-05-07 株式会社タムラ製作所 Reflow device
US9017993B2 (en) 2011-04-07 2015-04-28 Life Technologies Corporation System and methods for making and processing emulsions
EP3246416A1 (en) 2011-04-15 2017-11-22 The Johns Hopkins University Safe sequencing system
US9110026B2 (en) 2011-05-05 2015-08-18 Biopico Systems Inc Microfluidic devices and methods based on massively parallel picoreactors for cell and molecular diagnostics
WO2012167142A2 (en) 2011-06-02 2012-12-06 Raindance Technolgies, Inc. Enzyme quantification
US8841071B2 (en) * 2011-06-02 2014-09-23 Raindance Technologies, Inc. Sample multiplexing
US20130178378A1 (en) 2011-06-09 2013-07-11 Andrew C. Hatch Multiplex digital pcr
US9150916B2 (en) 2011-06-24 2015-10-06 Beat Christen Compositions and methods for identifying the essential genome of an organism
US8658430B2 (en) 2011-07-20 2014-02-25 Raindance Technologies, Inc. Manipulating droplet size
FR2978498B1 (en) 2011-07-28 2018-03-02 Valeo Equipements Electriques Moteur MOTOR VEHICLE STARTER CIRCUIT COMPRISING A VOLTAGE-INCREASING DEVICE AND EQUIPPED STARTER
GB2496016B (en) 2011-09-09 2016-03-16 Univ Leland Stanford Junior Methods for obtaining a sequence
WO2013056241A2 (en) 2011-10-14 2013-04-18 Pacific Biosciences Of California, Inc. Real-time redox sequencing
WO2013101783A2 (en) 2011-12-30 2013-07-04 Bio-Rad Laboratories, Inc. Methods and compositions for performing nucleic acid amplification reactions
WO2013116698A2 (en) 2012-02-02 2013-08-08 Invenra, Inc. High throughput screen for biologically active polypeptides
EP3495817A1 (en) 2012-02-10 2019-06-12 Raindance Technologies, Inc. Molecular diagnostic screening assay
US20130210638A1 (en) 2012-02-10 2013-08-15 Jeffrey Charles Olson Methods for sequencing nucleic acid
US9176031B2 (en) 2012-02-24 2015-11-03 Raindance Technologies, Inc. Labeling and sample preparation for sequencing
EP3524693A1 (en) 2012-04-30 2019-08-14 Raindance Technologies, Inc. Digital analyte analysis
US20160115532A1 (en) 2012-08-10 2016-04-28 Sequenta, Inc. High sensitivity mutation detection using sequence tags
US20150376609A1 (en) 2014-06-26 2015-12-31 10X Genomics, Inc. Methods of Analyzing Nucleic Acids from Individual Cells or Cell Populations
US9790546B2 (en) 2012-08-31 2017-10-17 Roche Molecular Systems, Inc. Microfluidic chip, device and system for the generation of aqueous droplets in emulsion oil for nucleic acid amplification
CN104797718B (en) 2012-09-12 2020-05-08 加利福尼亚大学董事会 Accurate genome sequencing of single cells by single-stranded amplification and sequencing
US9840734B2 (en) 2012-10-22 2017-12-12 Raindance Technologies, Inc. Methods for analyzing DNA
GB201218909D0 (en) 2012-10-22 2012-12-05 Univ Singapore Assay for the parallel detection of biological material based on PCR
WO2014085802A1 (en) 2012-11-30 2014-06-05 The Broad Institute, Inc. High-throughput dynamic reagent delivery system
WO2014138688A1 (en) 2013-03-07 2014-09-12 Bio-Rad Laboratories, Inc. Repetitive reverse transcription partition assay
US9273349B2 (en) 2013-03-14 2016-03-01 Affymetrix, Inc. Detection of nucleic acids
EP2971138B1 (en) 2013-03-15 2020-05-13 Bio-rad Laboratories, Inc. Digital assays with associated targets
EP2981349A4 (en) 2013-04-02 2016-11-16 Raindance Technologies Inc Systems and methods for handling microfluidic droplets
CN105431553B (en) 2013-05-29 2020-02-07 生物辐射实验室股份有限公司 Systems and methods for sequencing in emulsion-based microfluidics
US20150011397A1 (en) 2013-06-17 2015-01-08 Kim Lewis Methods for quantitative determination of multiple proteins in complex mixtures
CN105555972B (en) 2013-07-25 2020-07-31 伯乐生命医学产品有限公司 Genetic assay
US11901041B2 (en) 2013-10-04 2024-02-13 Bio-Rad Laboratories, Inc. Digital analysis of nucleic acid modification
CN104946737B (en) 2013-12-11 2019-02-22 安可济控股有限公司 For detecting the composition and method of rare sequence variants
US9944977B2 (en) 2013-12-12 2018-04-17 Raindance Technologies, Inc. Distinguishing rare variations in a nucleic acid sequence from a sample
US11193176B2 (en) 2013-12-31 2021-12-07 Bio-Rad Laboratories, Inc. Method for detecting and quantifying latent retroviral RNA species
US20150197790A1 (en) 2014-01-10 2015-07-16 Bio-Rad Laboratories, Inc. Intercalating dyes for differential detection
US20150298091A1 (en) 2014-04-21 2015-10-22 President And Fellows Of Harvard College Systems and methods for barcoding nucleic acids
CN107075543B (en) 2014-04-21 2021-11-16 哈佛学院院长及董事 Systems and methods for barcoding nucleic acids
US11155809B2 (en) 2014-06-24 2021-10-26 Bio-Rad Laboratories, Inc. Digital PCR barcoding
WO2016092372A1 (en) 2014-12-12 2016-06-16 Marcella Chiari New clickable polymers and gels for microarray and other applications
JP6518515B2 (en) 2015-05-28 2019-05-22 山洋電気株式会社 Motor sensor
US10647981B1 (en) 2015-09-08 2020-05-12 Bio-Rad Laboratories, Inc. Nucleic acid library generation methods and compositions
WO2017100350A1 (en) 2015-12-07 2017-06-15 Raindance Technologies, Inc. Multiplexing in partitions using microparticles
WO2017117358A1 (en) 2015-12-30 2017-07-06 Bio-Rad Laboratories, Inc. Digital protein quantification
US10036024B2 (en) 2016-06-03 2018-07-31 Purdue Research Foundation siRNA compositions that specifically downregulate expression of a variant of the PNPLA3 gene and methods of use thereof for treating a chronic liver disease or alcoholic liver disease (ALD)
US10858699B2 (en) 2016-08-30 2020-12-08 Integrated Dna Technologies, Inc. Cleavable hairpin primers

Patent Citations (99)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4279345A (en) * 1979-08-03 1981-07-21 Allred John C High speed particle sorter using a field emission electrode
US4508265A (en) * 1981-06-18 1985-04-02 Agency Of Industrial Science & Technology Method for spray combination of liquids and apparatus therefor
US5204112A (en) * 1986-06-16 1993-04-20 The Liposome Company, Inc. Induction of asymmetry in vesicles
US4931225A (en) * 1987-12-30 1990-06-05 Union Carbide Industrial Gases Technology Corporation Method and apparatus for dispersing a gas into a liquid
US5378957A (en) * 1989-11-17 1995-01-03 Charged Injection Corporation Methods and apparatus for dispersing a fluent material utilizing an electron beam
US5858670A (en) * 1990-07-02 1999-01-12 The Arizona Board Of Regents Bio-oligomer libraries and a method of use thereof
US5942056A (en) * 1993-04-22 1999-08-24 Federalloy, Inc. Plumbing fixtures and fittings employing copper-bismuth casting alloys
US5512131A (en) * 1993-10-04 1996-04-30 President And Fellows Of Harvard College Formation of microstamped patterns on surfaces and derivative articles
US5904933A (en) * 1994-06-09 1999-05-18 Alliance Pharmaceutical Corp. Stable reverse and multiple fluorocarbon emulsions
US5617997A (en) * 1994-06-13 1997-04-08 Praxair Technology, Inc. Narrow spray angle liquid fuel atomizers for combustion
US5935331A (en) * 1994-09-09 1999-08-10 Matsushita Electric Industrial Co., Ltd. Apparatus and method for forming films
US5762775A (en) * 1994-09-21 1998-06-09 Lockheed Martin Energy Systems, Inc. Method for electrically producing dispersions of a nonconductive fluid in a conductive medium
US20020022261A1 (en) * 1995-06-29 2002-02-21 Anderson Rolfe C. Miniaturized genetic analysis systems and methods
US5733526A (en) * 1995-12-14 1998-03-31 Alliance Pharmaceutical Corp. Hydrocarbon oil/fluorochemical preparations and methods of use
US6355198B1 (en) * 1996-03-15 2002-03-12 President And Fellows Of Harvard College Method of forming articles including waveguides via capillary micromolding and microtransfer molding
US5783431A (en) * 1996-04-24 1998-07-21 Chromaxome Corporation Methods for generating and screening novel metabolic pathways
US6187214B1 (en) * 1996-05-13 2001-02-13 Universidad De Seville Method and device for production of components for microfabrication
US6557834B2 (en) * 1996-05-13 2003-05-06 Universidad De Seville Device and method for fluid aeration via gas forced through a liquid within an orifice of a pressure chamber
US6386463B1 (en) * 1996-05-13 2002-05-14 Universidad De Sevilla Fuel injection nozzle and method of use
US6189803B1 (en) * 1996-05-13 2001-02-20 University Of Seville Fuel injection nozzle and method of use
US6197835B1 (en) * 1996-05-13 2001-03-06 Universidad De Sevilla Device and method for creating spherical particles of uniform size
US6196525B1 (en) * 1996-05-13 2001-03-06 Universidad De Sevilla Device and method for fluid aeration via gas forced through a liquid within an orifice of a pressure chamber
US6174469B1 (en) * 1996-05-13 2001-01-16 Universidad De Sevilla Device and method for creating dry particles
US6432148B1 (en) * 1996-05-13 2002-08-13 Universidad De Sevilla Fuel injection nozzle and method of use
US6357670B2 (en) * 1996-05-13 2002-03-19 Universidad De Sevilla Stabilized capillary microjet and devices and methods for producing same
US6234402B1 (en) * 1996-05-13 2001-05-22 Universidad De Sevilla Stabilized capillary microjet and devices and methods for producing same
US6241159B1 (en) * 1996-05-13 2001-06-05 Universidad De Sevilla Liquid atomization procedure
US6554202B2 (en) * 1996-05-13 2003-04-29 Universidad De Sevilla Fuel injection nozzle and method of use
US6394429B2 (en) * 1996-05-13 2002-05-28 Universidad De Sevilla Device and method for fluid aeration via gas forced through a liquid within an orifice of a pressure chamber
US6405936B1 (en) * 1996-05-13 2002-06-18 Universidad De Sevilla Stabilized capillary microjet and devices and methods for producing same
US6558960B1 (en) * 1996-06-28 2003-05-06 Caliper Technologies Corp. High throughput screening assay systems in microscale fluidic devices
US6274337B1 (en) * 1996-06-28 2001-08-14 Caliper Technologies Corp. High throughput screening assay systems in microscale fluidic devices
US6429025B1 (en) * 1996-06-28 2002-08-06 Caliper Technologies Corp. High-throughput screening assay systems in microscale fluidic devices
US6267858B1 (en) * 1996-06-28 2001-07-31 Caliper Technologies Corp. High throughput screening assay systems in microscale fluidic devices
US6399389B1 (en) * 1996-06-28 2002-06-04 Caliper Technologies Corp. High throughput screening assay systems in microscale fluidic devices
US6046056A (en) * 1996-06-28 2000-04-04 Caliper Technologies Corporation High throughput screening assay systems in microscale fluidic devices
US5942443A (en) * 1996-06-28 1999-08-24 Caliper Technologies Corporation High throughput screening assay systems in microscale fluidic devices
US6558944B1 (en) * 1996-06-28 2003-05-06 Caliper Technologies Corp. High throughput screening assay systems in microscale fluidic devices
US6432630B1 (en) * 1996-09-04 2002-08-13 Scandinanian Micro Biodevices A/S Micro-flow system for particle separation and analysis
US6221654B1 (en) * 1996-09-25 2001-04-24 California Institute Of Technology Method and apparatus for analysis and sorting of polynucleotides based on size
US6344325B1 (en) * 1996-09-25 2002-02-05 California Institute Of Technology Methods for analysis and sorting of polynucleotides
US6235383B1 (en) * 1997-01-24 2001-05-22 Samsung Corning Co., Ltd. Glass article having a durable water repellent surface
US6251661B1 (en) * 1997-05-14 2001-06-26 Morishita Jintan Co., Ltd. Seamless capsule for synthesizing biopolymer and method for producing the same
US20030124586A1 (en) * 1997-07-07 2003-07-03 Andrew Griffiths In vitro sorting method
US20020005354A1 (en) * 1997-09-23 2002-01-17 California Institute Of Technology Microfabricated cell sorter
US6540895B1 (en) * 1997-09-23 2003-04-01 California Institute Of Technology Microfabricated cell sorter for chemical and biological materials
US6103537A (en) * 1997-10-02 2000-08-15 Aclara Biosciences, Inc. Capillary assays involving separation of free and bound species
US6268222B1 (en) * 1998-01-22 2001-07-31 Luminex Corporation Microparticles attached to nanoparticles labeled with flourescent dye
US6210896B1 (en) * 1998-08-13 2001-04-03 Us Genomics Molecular motors
US6248378B1 (en) * 1998-12-16 2001-06-19 Universidad De Sevilla Enhanced food products
US20020119459A1 (en) * 1999-01-07 2002-08-29 Andrew Griffiths Optical sorting method
US20020036139A1 (en) * 1999-02-12 2002-03-28 Board Of Regents, The University Of Texas System Method and apparatus for programmable fluidic processing
US6506609B1 (en) * 1999-05-17 2003-01-14 Caliper Technologies Corp. Focusing of microparticles in microfluidic systems
US6592821B1 (en) * 1999-05-17 2003-07-15 Caliper Technologies Corp. Focusing of microparticles in microfluidic systems
US6408878B2 (en) * 1999-06-28 2002-06-25 California Institute Of Technology Microfabricated elastomeric valve and pump systems
US6890487B1 (en) * 1999-09-30 2005-05-10 Science & Technology Corporation ©UNM Flow cytometry for high throughput screening
US20020008028A1 (en) * 2000-01-12 2002-01-24 Jacobson Stephen C. Microfluidic device and method for focusing, segmenting, and dispensing of a fluid stream
US20020004532A1 (en) * 2000-05-26 2002-01-10 Michelle Matathia Low emulsifier multiple emulsions
US6680178B2 (en) * 2000-06-02 2004-01-20 The Regents Of The University Of California Profiling of protease specificity using combinatorial fluorogenic substrate libraries
US7081192B1 (en) * 2000-08-08 2006-07-25 Aviva Biosciences Corporation Methods for manipulating moieties in microfluidic systems
US6560030B2 (en) * 2000-08-16 2003-05-06 California Institute Of Technology Solid immersion lens structures and methods for producing solid immersion lens structures
US6608726B2 (en) * 2000-08-16 2003-08-19 California Institute Of Technology Solid immersion lens structures and methods for producing solid immersion lens structures
US6935768B2 (en) * 2000-08-25 2005-08-30 Institut Fur Mikrotechnik Mainz Gmbh Method and statistical micromixer for mixing at least two liquids
US6610499B1 (en) * 2000-08-31 2003-08-26 The Regents Of The University Of California Capillary array and related methods
US7514210B2 (en) * 2000-09-13 2009-04-07 Medical Research Council Compartmentalised self replication method for in vitro evolution of molecular libraries
US20020058332A1 (en) * 2000-09-15 2002-05-16 California Institute Of Technology Microfabricated crossflow devices and methods
US6508988B1 (en) * 2000-10-03 2003-01-21 California Institute Of Technology Combinatorial synthesis system
US7068874B2 (en) * 2000-11-28 2006-06-27 The Regents Of The University Of California Microfluidic sorting device
US20040068019A1 (en) * 2001-02-23 2004-04-08 Toshiro Higuchi Process for producing emulsion and microcapsules and apparatus therefor
US6752922B2 (en) * 2001-04-06 2004-06-22 Fluidigm Corporation Microfluidic chromatography
US20060147909A1 (en) * 2001-05-31 2006-07-06 Markus Rarbach Microstructures and use thereof for the directed evolution of biomolecules
US20030015425A1 (en) * 2001-06-20 2003-01-23 Coventor Inc. Microfluidic system including a virtual wall fluid interface port for interfacing fluids with the microfluidic system
US20030148544A1 (en) * 2001-06-28 2003-08-07 Advanced Research And Technology Institute, Inc. Methods of preparing multicolor quantum dot tagged beads and conjugates thereof
US6766817B2 (en) * 2001-07-25 2004-07-27 Tubarc Technologies, Llc Fluid conduction utilizing a reversible unsaturated siphon with tubarc porosity action
US7066586B2 (en) * 2001-07-25 2006-06-27 Tubarc Technologies, Llc Ink refill and recharging system
US6918404B2 (en) * 2001-07-25 2005-07-19 Tubarc Technologies, Llc Irrigation and drainage based on hydrodynamic unsaturated fluid flow
US20030029169A1 (en) * 2001-08-10 2003-02-13 Hanna William Thompson Integrated micro combined heat and power system
US6520425B1 (en) * 2001-08-21 2003-02-18 The University Of Akron Process and apparatus for the production of nanofibers
US20050084923A1 (en) * 2001-09-14 2005-04-21 Peter-Juergen Mueller Methods for cultivating and analyzing microbial individual cell cultures
US20040096515A1 (en) * 2001-12-07 2004-05-20 Bausch Andreas R. Methods and compositions for encapsulating active agents
US20050183995A1 (en) * 2002-04-17 2005-08-25 Cytonome, Inc. Method and apparatus for sorting particles
US20050172476A1 (en) * 2002-06-28 2005-08-11 President And Fellows Of Havard College Method and apparatus for fluid dispersion
US20060108012A1 (en) * 2002-11-14 2006-05-25 Barrow David A Microfluidic device and methods for construction and application
US7541383B2 (en) * 2002-12-20 2009-06-02 Amgen Inc. Asthma and allergic inflammation modulators
US20050032240A1 (en) * 2003-02-11 2005-02-10 The Regents Of The University Of California Microfluidic devices for controlled viscous shearing and formation of amphiphilic vesicles
US6926313B1 (en) * 2003-04-02 2005-08-09 Sandia National Laboratories High pressure capillary connector
US20060163385A1 (en) * 2003-04-10 2006-07-27 Link Darren R Formation and control of fluidic species
US20050032238A1 (en) * 2003-08-07 2005-02-10 Nanostream, Inc. Vented microfluidic separation devices and methods
US7204431B2 (en) * 2003-10-31 2007-04-17 Agilent Technologies, Inc. Electrospray ion source for mass spectroscopy
US20060003442A1 (en) * 2004-06-30 2006-01-05 Gong Jian P Cell culture scaffold, methods of manufacturing the scaffold and cell culture methods
US20060051329A1 (en) * 2004-08-27 2006-03-09 The Regents Of The University Of California Microfluidic device for the encapsulation of cells with low and high cell densities
US20060078888A1 (en) * 2004-10-08 2006-04-13 Medical Research Council Harvard University In vitro evolution in microfluidic systems
US20060078893A1 (en) * 2004-10-12 2006-04-13 Medical Research Council Compartmentalised combinatorial chemistry by microfluidic control
US20070054119A1 (en) * 2005-03-04 2007-03-08 Piotr Garstecki Systems and methods of forming particles
US20090131543A1 (en) * 2005-03-04 2009-05-21 Weitz David A Method and Apparatus for Forming Multiple Emulsions
US20070056853A1 (en) * 2005-09-15 2007-03-15 Lucnet Technologies Inc. Micro-chemical mixing
US20070195127A1 (en) * 2006-01-27 2007-08-23 President And Fellows Of Harvard College Fluidic droplet coalescence
US20080003142A1 (en) * 2006-05-11 2008-01-03 Link Darren R Microfluidic devices
US20090012187A1 (en) * 2007-03-28 2009-01-08 President And Fellows Of Harvard College Emulsions and Techniques for Formation

Cited By (876)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
USRE43122E1 (en) 1999-11-26 2012-01-24 The Governors Of The University Of Alberta Apparatus and method for trapping bead based reagents within microfluidic analysis systems
US8986628B2 (en) 2002-06-28 2015-03-24 President And Fellows Of Harvard College Method and apparatus for fluid dispersion
US9651039B2 (en) 2002-12-30 2017-05-16 The Regents Of The University Of California Fluid control structures in microfluidic devices
US20100224255A1 (en) * 2002-12-30 2010-09-09 The Regents Of The University Of California Fluid control structures in microfluidic devices
US20090060797A1 (en) * 2002-12-30 2009-03-05 The Regents Of The University Of California Fluid control structures in microfluidic devices
US9644623B2 (en) 2002-12-30 2017-05-09 The Regents Of The University Of California Fluid control structures in microfluidic devices
US11187702B2 (en) 2003-03-14 2021-11-30 Bio-Rad Laboratories, Inc. Enzyme quantification
US9857303B2 (en) 2003-03-31 2018-01-02 Medical Research Council Selection by compartmentalised screening
US9448172B2 (en) 2003-03-31 2016-09-20 Medical Research Council Selection by compartmentalised screening
US20100210479A1 (en) * 2003-03-31 2010-08-19 Medical Research Council Method of synthesis and testing of cominatorial libraries using microcapsules
US10052605B2 (en) 2003-03-31 2018-08-21 Medical Research Council Method of synthesis and testing of combinatorial libraries using microcapsules
US20150283546A1 (en) 2003-04-10 2015-10-08 President And Fellows Of Harvard College Formation and control of fluidic species
US11141731B2 (en) 2003-04-10 2021-10-12 President And Fellows Of Harvard College Formation and control of fluidic species
US10293341B2 (en) 2003-04-10 2019-05-21 President And Fellows Of Harvard College Formation and control of fluidic species
US9038919B2 (en) 2003-04-10 2015-05-26 President And Fellows Of Harvard College Formation and control of fluidic species
US10625256B2 (en) 2003-08-27 2020-04-21 President And Fellows Of Harvard College Electronic control of fluidic species
US9878325B2 (en) 2003-08-27 2018-01-30 President And Fellows Of Harvard College Electronic control of fluidic species
US9789482B2 (en) 2003-08-27 2017-10-17 President And Fellows Of Harvard College Methods of introducing a fluid into droplets
US11383234B2 (en) 2003-08-27 2022-07-12 President And Fellows Of Harvard College Electronic control of fluidic species
US11821109B2 (en) 2004-03-31 2023-11-21 President And Fellows Of Harvard College Compartmentalised combinatorial chemistry by microfluidic control
US9925504B2 (en) 2004-03-31 2018-03-27 President And Fellows Of Harvard College Compartmentalised combinatorial chemistry by microfluidic control
US20070092914A1 (en) * 2004-03-31 2007-04-26 Medical Research Council, Harvard University Compartmentalised screening by microfluidic control
US8420318B2 (en) 2004-06-01 2013-04-16 The Regents Of The University Of California Microfabricated integrated DNA analysis system
US20110020920A1 (en) * 2004-06-01 2011-01-27 The Regents Of The University Of California Microfabricated integrated dna analysis system
US9109251B2 (en) 2004-06-25 2015-08-18 University Of Hawaii Ultrasensitive biosensors
US10563252B2 (en) 2004-06-25 2020-02-18 University Of Hawaii Ultrasensitive biosensors
US10732649B2 (en) 2004-07-02 2020-08-04 The University Of Chicago Microfluidic system
US8431390B2 (en) 2004-09-15 2013-04-30 Integenx Inc. Systems of sample processing having a macro-micro interface
US8431340B2 (en) 2004-09-15 2013-04-30 Integenx Inc. Methods for processing and analyzing nucleic acid samples
US8476063B2 (en) 2004-09-15 2013-07-02 Integenx Inc. Microfluidic devices
US20100068723A1 (en) * 2004-09-15 2010-03-18 Stevan Bogdan Jovanovich Microfluidic devices
US20110076735A1 (en) * 2004-09-15 2011-03-31 Jovanovich Stevan B Microfluidic Devices
US8551714B2 (en) 2004-09-15 2013-10-08 Integenx Inc. Microfluidic devices
US9752185B2 (en) 2004-09-15 2017-09-05 Integenx Inc. Microfluidic devices
US20090197248A1 (en) * 2004-10-08 2009-08-06 President And Fellows Of Harvard College Vitro evolution in microfluidic systems
US9029083B2 (en) 2004-10-08 2015-05-12 Medical Research Council Vitro evolution in microfluidic systems
US9186643B2 (en) 2004-10-08 2015-11-17 Medical Research Council In vitro evolution in microfluidic systems
US8871444B2 (en) 2004-10-08 2014-10-28 Medical Research Council In vitro evolution in microfluidic systems
US11786872B2 (en) 2004-10-08 2023-10-17 United Kingdom Research And Innovation Vitro evolution in microfluidic systems
US9498759B2 (en) 2004-10-12 2016-11-22 President And Fellows Of Harvard College Compartmentalized screening by microfluidic control
US20090005254A1 (en) * 2004-10-12 2009-01-01 Andrew Griffiths Compartmentalized Screening by Microfluidic Control
US10316873B2 (en) 2005-03-04 2019-06-11 President And Fellows Of Harvard College Method and apparatus for forming multiple emulsions
US20090131543A1 (en) * 2005-03-04 2009-05-21 Weitz David A Method and Apparatus for Forming Multiple Emulsions
US9039273B2 (en) 2005-03-04 2015-05-26 President And Fellows Of Harvard College Method and apparatus for forming multiple emulsions
US9910020B1 (en) 2005-03-30 2018-03-06 Copilot Ventures Fund Iii Llc Methods and articles for identifying objects using encapsulated perfluorocarbon tracers
US11414702B2 (en) 2005-06-15 2022-08-16 Complete Genomics, Inc. Nucleic acid analysis by random mixtures of non-overlapping fragments
US20100310299A1 (en) * 2005-08-01 2010-12-09 Silverbrook Research Pty Ltd Electronic image-sensing pen with force sensor and removeable ink cartridge
US20070044824A1 (en) * 2005-09-01 2007-03-01 Scott William Capeci Processing system and method of processing
US9410151B2 (en) 2006-01-11 2016-08-09 Raindance Technologies, Inc. Microfluidic devices and methods of use in the formation and control of nanoreactors
US9328344B2 (en) 2006-01-11 2016-05-03 Raindance Technologies, Inc. Microfluidic devices and methods of use in the formation and control of nanoreactors
US9534216B2 (en) 2006-01-11 2017-01-03 Raindance Technologies, Inc. Microfluidic devices and methods of use in the formation and control of nanoreactors
US8286665B2 (en) 2006-03-22 2012-10-16 The Regents Of The University Of California Multiplexed latching valves for microfluidic devices and processors
US9562837B2 (en) 2006-05-11 2017-02-07 Raindance Technologies, Inc. Systems for handling microfludic droplets
US9273308B2 (en) 2006-05-11 2016-03-01 Raindance Technologies, Inc. Selection of compartmentalized screening method
US11351510B2 (en) 2006-05-11 2022-06-07 Bio-Rad Laboratories, Inc. Microfluidic devices
US9012390B2 (en) 2006-08-07 2015-04-21 Raindance Technologies, Inc. Fluorocarbon emulsion stabilizing surfactants
US9498761B2 (en) 2006-08-07 2016-11-22 Raindance Technologies, Inc. Fluorocarbon emulsion stabilizing surfactants
US8841116B2 (en) 2006-10-25 2014-09-23 The Regents Of The University Of California Inline-injection microdevice and microfabricated integrated DNA analysis system using same
US8540867B2 (en) 2006-12-14 2013-09-24 Life Technologies Corporation Methods and apparatus for detecting molecular interactions using FET arrays
US8262900B2 (en) 2006-12-14 2012-09-11 Life Technologies Corporation Methods and apparatus for measuring analytes using large scale FET arrays
US9989489B2 (en) 2006-12-14 2018-06-05 Life Technnologies Corporation Methods for calibrating an array of chemically-sensitive sensors
US8658017B2 (en) 2006-12-14 2014-02-25 Life Technologies Corporation Methods for operating an array of chemically-sensitive sensors
US9269708B2 (en) 2006-12-14 2016-02-23 Life Technologies Corporation Methods and apparatus for measuring analytes using large scale FET arrays
US8685230B2 (en) 2006-12-14 2014-04-01 Life Technologies Corporation Methods and apparatus for high-speed operation of a chemically-sensitive sensor array
US20090026082A1 (en) * 2006-12-14 2009-01-29 Ion Torrent Systems Incorporated Methods and apparatus for measuring analytes using large scale FET arrays
US8692298B2 (en) 2006-12-14 2014-04-08 Life Technologies Corporation Chemical sensor array having multiple sensors per well
US20110230375A1 (en) * 2006-12-14 2011-09-22 Life Technologies Corporation Methods and apparatus for measuring analytes using large scale fet arrays
US8575664B2 (en) 2006-12-14 2013-11-05 Life Technologies Corporation Chemically-sensitive sensor array calibration circuitry
US8426899B2 (en) 2006-12-14 2013-04-23 Life Technologies Corporation Methods and apparatus for measuring analytes using large scale FET arrays
US10415079B2 (en) 2006-12-14 2019-09-17 Life Technologies Corporation Methods and apparatus for detecting molecular interactions using FET arrays
US8558288B2 (en) 2006-12-14 2013-10-15 Life Technologies Corporation Methods and apparatus for measuring analytes using large scale FET arrays
US10203300B2 (en) 2006-12-14 2019-02-12 Life Technologies Corporation Methods and apparatus for measuring analytes using large scale FET arrays
US10633699B2 (en) 2006-12-14 2020-04-28 Life Technologies Corporation Methods and apparatus for measuring analytes using large scale FET arrays
US8540868B2 (en) 2006-12-14 2013-09-24 Life Technologies Corporation Methods and apparatus for detecting molecular interactions using FET arrays
US9134269B2 (en) 2006-12-14 2015-09-15 Life Technologies Corporation Methods and apparatus for measuring analytes using large scale FET arrays
US10816506B2 (en) 2006-12-14 2020-10-27 Life Technologies Corporation Method for measuring analytes using large scale chemfet arrays
US8540865B2 (en) 2006-12-14 2013-09-24 Life Technologies Corporation Methods and apparatus for detecting molecular interactions using FET arrays
US8540866B2 (en) 2006-12-14 2013-09-24 Life Technologies Corporation Methods and apparatus for detecting molecular interactions using FET arrays
US8535513B2 (en) 2006-12-14 2013-09-17 Life Technologies Corporation Methods and apparatus for measuring analytes using large scale FET arrays
US9404920B2 (en) 2006-12-14 2016-08-02 Life Technologies Corporation Methods and apparatus for detecting molecular interactions using FET arrays
US8530941B2 (en) 2006-12-14 2013-09-10 Life Technologies Corporation Methods and apparatus for measuring analytes using large scale FET arrays
US8742472B2 (en) 2006-12-14 2014-06-03 Life Technologies Corporation Chemically sensitive sensors with sample and hold capacitors
US8766328B2 (en) 2006-12-14 2014-07-01 Life Technologies Corporation Chemically-sensitive sample and hold sensors
US8764969B2 (en) 2006-12-14 2014-07-01 Life Technologies Corporation Methods for operating chemically sensitive sensors with sample and hold capacitors
US8519448B2 (en) 2006-12-14 2013-08-27 Life Technologies Corporation Chemically-sensitive array with active and reference sensors
US20100188073A1 (en) * 2006-12-14 2010-07-29 Ion Torrent Systems Incorporated Methods and apparatus for measuring analytes using large scale fet arrays
US8264014B2 (en) 2006-12-14 2012-09-11 Life Technologies Corporation Methods and apparatus for measuring analytes using large scale FET arrays
US8502278B2 (en) 2006-12-14 2013-08-06 Life Technologies Corporation Chemically-sensitive sample and hold sensors
US8269261B2 (en) 2006-12-14 2012-09-18 Life Technologies Corporation Methods and apparatus for measuring analytes using large scale FET arrays
US8496802B2 (en) 2006-12-14 2013-07-30 Life Technologies Corporation Methods for operating chemically-sensitive sample and hold sensors
US9039888B2 (en) 2006-12-14 2015-05-26 Life Technologies Corporation Methods and apparatus for detecting molecular interactions using FET arrays
US8890216B2 (en) 2006-12-14 2014-11-18 Life Technologies Corporation Methods and apparatus for measuring analytes using large scale FET arrays
US8492799B2 (en) 2006-12-14 2013-07-23 Life Technologies Corporation Methods and apparatus for detecting molecular interactions using FET arrays
US9023189B2 (en) 2006-12-14 2015-05-05 Life Technologies Corporation High density sensor array without wells
US8293082B2 (en) 2006-12-14 2012-10-23 Life Technologies Corporation Methods and apparatus for measuring analytes using large scale FET arrays
US8306757B2 (en) 2006-12-14 2012-11-06 Life Technologies Corporation Methods and apparatus for measuring analytes using large scale FET arrays
US8313625B2 (en) 2006-12-14 2012-11-20 Life Technologies Corporation Methods and apparatus for measuring analytes using large scale FET arrays
US8313639B2 (en) 2006-12-14 2012-11-20 Life Technologies Corporation Methods and apparatus for measuring analytes using large scale FET arrays
US8317999B2 (en) 2006-12-14 2012-11-27 Life Technologies Corporation Methods and apparatus for measuring analytes using large scale FET arrays
US8492800B2 (en) 2006-12-14 2013-07-23 Life Technologies Corporation Chemically sensitive sensors with sample and hold capacitors
US8349167B2 (en) 2006-12-14 2013-01-08 Life Technologies Corporation Methods and apparatus for detecting molecular interactions using FET arrays
US10502708B2 (en) 2006-12-14 2019-12-10 Life Technologies Corporation Chemically-sensitive sensor array calibration circuitry
US8450781B2 (en) 2006-12-14 2013-05-28 Life Technologies Corporation Methods and apparatus for measuring analytes using large scale FET arrays
US8445945B2 (en) 2006-12-14 2013-05-21 Life Technologies Corporation Low noise chemically-sensitive field effect transistors
US8441044B2 (en) 2006-12-14 2013-05-14 Life Technologies Corporation Methods for manufacturing low noise chemically-sensitive field effect transistors
US8435395B2 (en) 2006-12-14 2013-05-07 Life Technologies Corporation Methods and apparatus for measuring analytes using large scale FET arrays
US11435314B2 (en) 2006-12-14 2022-09-06 Life Technologies Corporation Chemically-sensitive sensor array device
US9951382B2 (en) 2006-12-14 2018-04-24 Life Technologies Corporation Methods and apparatus for measuring analytes using large scale FET arrays
US20100197507A1 (en) * 2006-12-14 2010-08-05 Ion Torrent Systems Incorporated Methods and apparatus for measuring analytes using large scale fet arrays
US8426898B2 (en) 2006-12-14 2013-04-23 Life Technologies Corporation Methods and apparatus for measuring analytes using large scale FET arrays
US8415716B2 (en) 2006-12-14 2013-04-09 Life Technologies Corporation Chemically sensitive sensors with feedback circuits
US7696002B1 (en) * 2007-01-25 2010-04-13 Pacesette, Inc. Electrolytic capacitor case design with insert molded elastomer seals
US8557518B2 (en) 2007-02-05 2013-10-15 Integenx Inc. Microfluidic and nanofluidic devices, systems, and applications
US20110039303A1 (en) * 2007-02-05 2011-02-17 Stevan Bogdan Jovanovich Microfluidic and nanofluidic devices, systems, and applications
US9440232B2 (en) 2007-02-06 2016-09-13 Raindance Technologies, Inc. Manipulation of fluids and reactions in microfluidic systems
US9017623B2 (en) 2007-02-06 2015-04-28 Raindance Technologies, Inc. Manipulation of fluids and reactions in microfluidic systems
US8772046B2 (en) 2007-02-06 2014-07-08 Brandeis University Manipulation of fluids and reactions in microfluidic systems
US10603662B2 (en) 2007-02-06 2020-03-31 Brandeis University Manipulation of fluids and reactions in microfluidic systems
US20100163109A1 (en) * 2007-02-06 2010-07-01 Brandeis University Manipulation of fluids and reactions in microfluidic systems
US11819849B2 (en) 2007-02-06 2023-11-21 Brandeis University Manipulation of fluids and reactions in microfluidic systems
US9816121B2 (en) 2007-03-07 2017-11-14 President And Fellows Of Harvard College Assays and other reactions involving droplets
US10738337B2 (en) 2007-03-07 2020-08-11 President And Fellows Of Harvard College Assays and other reactions involving droplets
US10221437B2 (en) 2007-03-07 2019-03-05 President And Fellows Of Harvard College Assays and other reactions involving droplets
US10941430B2 (en) 2007-03-07 2021-03-09 President And Fellows Of Harvard College Assays and other reactions involving droplets
US9068210B2 (en) 2007-03-07 2015-06-30 President And Fellows Of Harvard College Assay and other reactions involving droplets
US20100136544A1 (en) * 2007-03-07 2010-06-03 Jeremy Agresti Assays and other reactions involving droplets
US9850526B2 (en) 2007-03-07 2017-12-26 President And Fellows Of Harvard College Assays and other reactions involving droplets
US9017948B2 (en) 2007-03-07 2015-04-28 President And Fellows Of Harvard College Assays and other reactions involving droplets
US9029085B2 (en) 2007-03-07 2015-05-12 President And Fellows Of Harvard College Assays and other reactions involving droplets
US10683524B2 (en) 2007-03-07 2020-06-16 President And Fellows Of Harvard College Assays and other reactions involving droplets
US10508294B2 (en) 2007-03-07 2019-12-17 President And Fellows Of Harvard College Assays and other reactions involving droplets
US8592221B2 (en) 2007-04-19 2013-11-26 Brandeis University Manipulation of fluids, fluid components and reactions in microfluidic systems
US10357772B2 (en) 2007-04-19 2019-07-23 President And Fellows Of Harvard College Manipulation of fluids, fluid components and reactions in microfluidic systems
US9068699B2 (en) 2007-04-19 2015-06-30 Brandeis University Manipulation of fluids, fluid components and reactions in microfluidic systems
US20100252118A1 (en) * 2007-04-19 2010-10-07 Seth Fraden Manipulation of fluids, fluid components and reactions in microfluidic systems
US10960397B2 (en) 2007-04-19 2021-03-30 President And Fellows Of Harvard College Manipulation of fluids, fluid components and reactions in microfluidic systems
US11618024B2 (en) 2007-04-19 2023-04-04 President And Fellows Of Harvard College Manipulation of fluids, fluid components and reactions in microfluidic systems
US10675626B2 (en) 2007-04-19 2020-06-09 President And Fellows Of Harvard College Manipulation of fluids, fluid components and reactions in microfluidic systems
US11224876B2 (en) 2007-04-19 2022-01-18 Brandeis University Manipulation of fluids, fluid components and reactions in microfluidic systems
US11339430B2 (en) 2007-07-10 2022-05-24 Life Technologies Corporation Methods and apparatus for measuring analytes using large scale FET arrays
US8454906B2 (en) 2007-07-24 2013-06-04 The Regents Of The University Of California Microfabricated droplet generator for single molecule/cell genetic analysis in engineered monodispersed emulsions
US20100285975A1 (en) * 2007-07-24 2010-11-11 The Regents Of The University Of California Microfabricated droplet generator for single molecule/cell genetic analysis in engineered monodispersed emulsions
US9090885B2 (en) 2007-07-26 2015-07-28 The University Of Chicago Co-incubating confined microbial communities
US20100227767A1 (en) * 2007-07-26 2010-09-09 Boedicker James Q Stochastic confinement to detect, manipulate, and utilize molecules and organisms
US20100317085A1 (en) * 2007-07-26 2010-12-16 Boedicker James Q Co-incubating confined microbial communities
US8268564B2 (en) * 2007-09-26 2012-09-18 President And Fellows Of Harvard College Methods and applications for stitched DNA barcodes
US20090098555A1 (en) * 2007-09-26 2009-04-16 President And Fellows Of Harvard College Methods and applications for stitched dna barcodes
US9114397B2 (en) 2007-10-25 2015-08-25 Canon U.S. Life Sciences, Inc. Method of reducing cross-contamination in continuous amplification reactions in a channel
US20090111149A1 (en) * 2007-10-25 2009-04-30 Canon U.S. Life Sciences, Inc. Method of reducing cross-contamination in continuous amplification reactions in a channel
US11389779B2 (en) 2007-12-05 2022-07-19 Complete Genomics, Inc. Methods of preparing a library of nucleic acid fragments tagged with oligonucleotide bar code sequences
US9797010B2 (en) 2007-12-21 2017-10-24 President And Fellows Of Harvard College Systems and methods for nucleic acid sequencing
US10633701B2 (en) 2007-12-21 2020-04-28 President And Fellows Of Harvard College Systems and methods for nucleic acid sequencing
US8815576B2 (en) * 2007-12-27 2014-08-26 Lawrence Livermore National Security, Llc. Chip-based sequencing nucleic acids
US20100184020A1 (en) * 2007-12-27 2010-07-22 Lawrence Livermore National Security, Llc. Chip-Based Sequencing Nucleic Acids
US8969257B1 (en) * 2007-12-28 2015-03-03 Intermolecular, Inc. Combinatorial flow system and method
US20090253181A1 (en) * 2008-01-22 2009-10-08 Microchip Biotechnologies, Inc. Universal sample preparation system and use in an integrated analysis system
US8748165B2 (en) 2008-01-22 2014-06-10 Integenx Inc. Methods for generating short tandem repeat (STR) profiles
WO2009100028A1 (en) * 2008-02-01 2009-08-13 The Regents Of The University Of California Microfluidic imaging cytometry
US20090217742A1 (en) * 2008-03-03 2009-09-03 University Of Washington Droplet compartmentalization for chemical separation and on-line sampling
US8062903B2 (en) 2008-03-03 2011-11-22 University Of Washington Droplet compartmentalization for chemical separation and on-line sampling
US10429376B2 (en) * 2008-05-16 2019-10-01 Board Of Supervisors Of Louisiana State University And Agricultural And Mechanical College Microfluidic isolation of tumor cells or other rare cells from whole blood or other liquids
US7900503B2 (en) * 2008-05-17 2011-03-08 William Keith Leonard Method of high throughput viscometry
US20090282901A1 (en) * 2008-05-17 2009-11-19 William Keith Leonard Method of high throughput viscometry
US8470164B2 (en) 2008-06-25 2013-06-25 Life Technologies Corporation Methods and apparatus for measuring analytes using large scale FET arrays
US8524057B2 (en) 2008-06-25 2013-09-03 Life Technologies Corporation Methods and apparatus for measuring analytes using large scale FET arrays
US9194000B2 (en) 2008-06-25 2015-11-24 Life Technologies Corporation Methods and apparatus for measuring analytes using large scale FET arrays
US20100022414A1 (en) * 2008-07-18 2010-01-28 Raindance Technologies, Inc. Droplet Libraries
US11511242B2 (en) 2008-07-18 2022-11-29 Bio-Rad Laboratories, Inc. Droplet libraries
US11534727B2 (en) 2008-07-18 2022-12-27 Bio-Rad Laboratories, Inc. Droplet libraries
US11596908B2 (en) 2008-07-18 2023-03-07 Bio-Rad Laboratories, Inc. Droplet libraries
US10533998B2 (en) * 2008-07-18 2020-01-14 Bio-Rad Laboratories, Inc. Enzyme quantification
US20120264646A1 (en) * 2008-07-18 2012-10-18 Raindance Technologies, Inc. Enzyme quantification
US20100041046A1 (en) * 2008-08-15 2010-02-18 University Of Washington Method and apparatus for the discretization and manipulation of sample volumes
US9180453B2 (en) * 2008-08-15 2015-11-10 University Of Washington Method and apparatus for the discretization and manipulation of sample volumes
US10421070B2 (en) 2008-08-15 2019-09-24 University Of Washington Method and apparatus for the discretization and manipulation of sample volumes
CN102187216A (en) * 2008-08-15 2011-09-14 华盛顿大学 Method and apparatus for the discretization and manipulation of sample volumes
US20110218123A1 (en) * 2008-09-19 2011-09-08 President And Fellows Of Harvard College Creation of libraries of droplets and related species
US11401550B2 (en) 2008-09-19 2022-08-02 President And Fellows Of Harvard College Creation of libraries of droplets and related species
US20110092373A1 (en) * 2008-09-23 2011-04-21 Quantalife, Inc. System for transporting emulsions from an array to a detector
US9623384B2 (en) 2008-09-23 2017-04-18 Bio-Rad Laboratories, Inc. System for transporting emulsions from an array to a detector
US11633739B2 (en) 2008-09-23 2023-04-25 Bio-Rad Laboratories, Inc. Droplet-based assay system
US20100173394A1 (en) * 2008-09-23 2010-07-08 Colston Jr Billy Wayne Droplet-based assay system
US11130134B2 (en) 2008-09-23 2021-09-28 Bio-Rad Laboratories, Inc. Method of performing droplet-based assays
US9156010B2 (en) 2008-09-23 2015-10-13 Bio-Rad Laboratories, Inc. Droplet-based assay system
AU2009297108B2 (en) * 2008-09-23 2015-02-12 Bio-Rad Laboratories, Inc. Droplet-based assay system
US9248417B2 (en) * 2008-09-23 2016-02-02 Bio-Rad Laboratories, Inc. System for droplet-based assays using an array of emulsions
US9132394B2 (en) * 2008-09-23 2015-09-15 Bio-Rad Laboratories, Inc. System for detection of spaced droplets
GB2502409B (en) * 2008-09-23 2014-03-12 Bio Rad Laboratories Droplet-Based assay system
US9243288B2 (en) 2008-09-23 2016-01-26 Bio-Rad Laboratories, Inc. Cartridge with lysis chamber and droplet generator
US9126160B2 (en) 2008-09-23 2015-09-08 Bio-Rad Laboratories, Inc. System for forming an array of emulsions
US9492797B2 (en) 2008-09-23 2016-11-15 Bio-Rad Laboratories, Inc. System for detection of spaced droplets
US10258989B2 (en) 2008-09-23 2019-04-16 Bio-Rad Laboratories, Inc. Method of making a device for generating droplets
US10512910B2 (en) 2008-09-23 2019-12-24 Bio-Rad Laboratories, Inc. Droplet-based analysis method
GB2502409A (en) * 2008-09-23 2013-11-27 Bio Rad Laboratories Droplet based assay system
US11612892B2 (en) 2008-09-23 2023-03-28 Bio-Rad Laboratories, Inc. Method of performing droplet-based assays
GB2504241B (en) * 2008-09-23 2014-03-12 Bio Rad Laboratories Droplet-based assay system
US11130128B2 (en) * 2008-09-23 2021-09-28 Bio-Rad Laboratories, Inc. Detection method for a target nucleic acid
US9649635B2 (en) 2008-09-23 2017-05-16 Bio-Rad Laboratories, Inc. System for generating droplets with push-back to remove oil
GB2477053B (en) * 2008-09-23 2013-11-13 Quantalife Inc Droplet-based assay system
US9417190B2 (en) 2008-09-23 2016-08-16 Bio-Rad Laboratories, Inc. Calibrations and controls for droplet-based assays
US10258988B2 (en) 2008-09-23 2019-04-16 Bio-Rad Laboratories, Inc. Device for generating droplets
US9764322B2 (en) 2008-09-23 2017-09-19 Bio-Rad Laboratories, Inc. System for generating droplets with pressure monitoring
US9216392B2 (en) 2008-09-23 2015-12-22 Bio-Rad Laboratories, Inc. System for forming an array of emulsions
US20110092392A1 (en) * 2008-09-23 2011-04-21 Quantalife, Inc. System for forming an array of emulsions
US20110092376A1 (en) * 2008-09-23 2011-04-21 Quantalife, Inc. System for droplet-based assays using an array of emulsions
US20110311978A1 (en) * 2008-09-23 2011-12-22 Quantalife, Inc. System for detection of spaced droplets
US10279350B2 (en) 2008-09-23 2019-05-07 Bio-Rad Laboratories, Inc. Method of generating droplets
GB2477053A (en) * 2008-09-23 2011-07-20 Quantalife Inc Droplet-based System
GB2504241A (en) * 2008-09-23 2014-01-22 Bio Rad Laboratories Droplet based assay system
US8633015B2 (en) 2008-09-23 2014-01-21 Bio-Rad Laboratories, Inc. Flow-based thermocycling system with thermoelectric cooler
US9636682B2 (en) 2008-09-23 2017-05-02 Bio-Rad Laboratories, Inc. System for generating droplets—instruments and cassette
WO2010036352A1 (en) * 2008-09-23 2010-04-01 Quantalife, Inc Droplet-based assay system
US20120121480A1 (en) * 2008-10-08 2012-05-17 Universite De Strasbourg Microfluidic devices for reliable on-chip incubation of droplets in delay lines
WO2010042744A1 (en) * 2008-10-08 2010-04-15 Universite De Strasbourg Microfluidic devices for reliable on-chip incubation of droplets in delay lines
US11137369B2 (en) 2008-10-22 2021-10-05 Life Technologies Corporation Integrated sensor arrays for biological and chemical analysis
US9964515B2 (en) 2008-10-22 2018-05-08 Life Technologies Corporation Integrated sensor arrays for biological and chemical analysis
US11448613B2 (en) 2008-10-22 2022-09-20 Life Technologies Corporation ChemFET sensor array including overlying array of wells
US8936763B2 (en) 2008-10-22 2015-01-20 Life Technologies Corporation Integrated sensor arrays for biological and chemical analysis
US20110281737A1 (en) * 2008-10-22 2011-11-17 Life Technologies Corporation Method and Apparatus for Rapid Nucleic Acid Sequencing
US11874250B2 (en) 2008-10-22 2024-01-16 Life Technologies Corporation Integrated sensor arrays for biological and chemical analysis
US9944981B2 (en) 2008-10-22 2018-04-17 Life Technologies Corporation Methods and apparatus for measuring analytes
US10457977B2 (en) 2008-12-19 2019-10-29 President And Fellows Of Harvard College Particle-assisted nucleic acid sequencing
US20100165784A1 (en) * 2008-12-31 2010-07-01 Microchip Biotechnologies, Inc., A California Corporation Instrument with microfluidic chip
US8672532B2 (en) 2008-12-31 2014-03-18 Integenx Inc. Microfluidic methods
US9056299B2 (en) 2009-03-13 2015-06-16 President And Fellows Of Harvard College Scale-up of flow-focusing microfluidic devices
US9486757B2 (en) 2009-03-13 2016-11-08 President And Fellows Of Harvard College Scale-up of microfluidic devices
US10518230B2 (en) 2009-03-13 2019-12-31 President And Fellows Of Harvard College Scale-up of microfluidic devices
US11517864B2 (en) 2009-03-13 2022-12-06 President And Fellows Of Harvard College Scale-up of microfluidic devices
US8528589B2 (en) 2009-03-23 2013-09-10 Raindance Technologies, Inc. Manipulation of microfluidic droplets
US11268887B2 (en) 2009-03-23 2022-03-08 Bio-Rad Laboratories, Inc. Manipulation of microfluidic droplets
EP3415235A1 (en) 2009-03-23 2018-12-19 Raindance Technologies Inc. Manipulation of microfluidic droplets
WO2010111231A1 (en) 2009-03-23 2010-09-30 Raindance Technologies, Inc. Manipulation of microfluidic droplets
US9493826B2 (en) 2009-03-24 2016-11-15 California Institute Of Technology Multivolume devices, kits and related methods for quantification and detection of nucleic acids and other analytes
US10370705B2 (en) 2009-03-24 2019-08-06 University Of Chicago Analysis devices, kits, and related methods for digital quantification of nucleic acids and other analytes
US10543485B2 (en) 2009-03-24 2020-01-28 University Of Chicago Slip chip device and methods
US10196700B2 (en) 2009-03-24 2019-02-05 University Of Chicago Multivolume devices, kits and related methods for quantification and detection of nucleic acids and other analytes
US9464319B2 (en) 2009-03-24 2016-10-11 California Institute Of Technology Multivolume devices, kits and related methods for quantification of nucleic acids and other analytes
US9415392B2 (en) 2009-03-24 2016-08-16 The University Of Chicago Slip chip device and methods
US9447461B2 (en) 2009-03-24 2016-09-20 California Institute Of Technology Analysis devices, kits, and related methods for digital quantification of nucleic acids and other analytes
US11840730B1 (en) 2009-04-30 2023-12-12 Molecular Loop Biosciences, Inc. Methods and compositions for evaluating genetic markers
US8766327B2 (en) 2009-05-29 2014-07-01 Life Technologies Corporation Active chemically-sensitive sensors with in-sensor current sources
US10718733B2 (en) 2009-05-29 2020-07-21 Life Technologies Corporation Methods and apparatus for measuring analytes
US8592154B2 (en) 2009-05-29 2013-11-26 Life Technologies Corporation Methods and apparatus for high speed operation of a chemically-sensitive sensor array
US8592153B1 (en) 2009-05-29 2013-11-26 Life Technologies Corporation Methods for manufacturing high capacitance microwell structures of chemically-sensitive sensors
US8742469B2 (en) 2009-05-29 2014-06-03 Life Technologies Corporation Active chemically-sensitive sensors with correlated double sampling
US8698212B2 (en) 2009-05-29 2014-04-15 Life Technologies Corporation Active chemically-sensitive sensors
US10451585B2 (en) 2009-05-29 2019-10-22 Life Technologies Corporation Methods and apparatus for measuring analytes
US8748947B2 (en) 2009-05-29 2014-06-10 Life Technologies Corporation Active chemically-sensitive sensors with reset switch
US8822205B2 (en) 2009-05-29 2014-09-02 Life Technologies Corporation Active chemically-sensitive sensors with source follower amplifier
US11768171B2 (en) 2009-05-29 2023-09-26 Life Technologies Corporation Methods and apparatus for measuring analytes
US8776573B2 (en) 2009-05-29 2014-07-15 Life Technologies Corporation Methods and apparatus for measuring analytes
US9927393B2 (en) 2009-05-29 2018-03-27 Life Technologies Corporation Methods and apparatus for measuring analytes
US8912580B2 (en) 2009-05-29 2014-12-16 Life Technologies Corporation Active chemically-sensitive sensors with in-sensor current sources
US10809226B2 (en) 2009-05-29 2020-10-20 Life Technologies Corporation Methods and apparatus for measuring analytes
US8994076B2 (en) 2009-05-29 2015-03-31 Life Technologies Corporation Chemically-sensitive field effect transistor based pixel array with protection diodes
US8263336B2 (en) 2009-05-29 2012-09-11 Life Technologies Corporation Methods and apparatus for measuring analytes
US11692964B2 (en) 2009-05-29 2023-07-04 Life Technologies Corporation Methods and apparatus for measuring analytes
US8673627B2 (en) 2009-05-29 2014-03-18 Life Technologies Corporation Apparatus and methods for performing electrochemical reactions
US20100303687A1 (en) * 2009-06-02 2010-12-02 Integenx Inc. Fluidic devices with diaphragm valves
US8388908B2 (en) 2009-06-02 2013-03-05 Integenx Inc. Fluidic devices with diaphragm valves
US9067207B2 (en) 2009-06-04 2015-06-30 University Of Virginia Patent Foundation Optical approach for microfluidic DNA electrophoresis detection
US20110229897A1 (en) * 2009-06-04 2011-09-22 Lockheed Martin Corporation Optical approach for microfluidic DNA electrophoresis detection
US20110005932A1 (en) * 2009-06-05 2011-01-13 Integenx Inc. Universal sample preparation system and use in an integrated analysis system
US8394642B2 (en) 2009-06-05 2013-03-12 Integenx Inc. Universal sample preparation system and use in an integrated analysis system
US8562918B2 (en) 2009-06-05 2013-10-22 Integenx Inc. Universal sample preparation system and use in an integrated analysis system
US9012236B2 (en) 2009-06-05 2015-04-21 Integenx Inc. Universal sample preparation system and use in an integrated analysis system
WO2010147942A1 (en) * 2009-06-16 2010-12-23 Massachusetts Institute Of Technology Multiphase non-linear electrokinetic devices
WO2011011172A1 (en) * 2009-07-21 2011-01-27 IntegenX, Inc. Microfluidic devices and uses thereof
US9063136B2 (en) * 2009-08-02 2015-06-23 Qvella Corporation Cell concentration, capture and lysis devices and methods of use thereof
US20120190040A1 (en) * 2009-08-02 2012-07-26 Qvella Corporation Cell concentration, capture and lysis devices and methods of use thereof
US11371988B2 (en) 2009-08-02 2022-06-28 Qvella Corporation Cell concentration, capture and lysis devices and methods of use thereof
US9988667B2 (en) 2009-08-06 2018-06-05 Cornell University Device and methods for epigenetic analysis
US8735065B2 (en) 2009-08-06 2014-05-27 Cornell University Device and methods for epigenetic analysis
US20120245047A1 (en) * 2009-08-06 2012-09-27 Craighead Harold G Device and methods for molecular analysis
CN102612652A (en) * 2009-08-06 2012-07-25 康奈尔大学 Device and method for molecular analysis
US9447451B2 (en) 2009-08-06 2016-09-20 Cornell University Device and methods for epigenetic analysis
US9605298B2 (en) * 2009-08-06 2017-03-28 Cornell University Device and methods for molecular analysis
US20120222959A1 (en) * 2009-08-12 2012-09-06 Caliper Life Sciences, Inc. Pinching channels for fractionation of fragmented samples
US20110036411A1 (en) * 2009-08-12 2011-02-17 Caliper Life Sciences, Inc. Pinching channels for fractionation of fragmented samples
US8202486B2 (en) * 2009-08-12 2012-06-19 Caliper Life Sciences, Inc. Pinching channels for fractionation of fragmented samples
US8277744B2 (en) * 2009-08-12 2012-10-02 Caliper Life Sciences, Inc. Pinching channels for fractionation of fragmented samples
EP2940153B1 (en) * 2009-09-02 2020-05-13 Bio-Rad Laboratories, Inc. System for mixing fluids by coalescence of multiple emulsions
JP2013503630A (en) * 2009-09-02 2013-02-04 クァンタライフ・インコーポレーテッド System for mixing fluids by combining multiple emulsions
US10166522B2 (en) 2009-09-02 2019-01-01 Bio-Rad Laboratories, Inc. System for mixing fluids by coalescence of multiple emulsions
EP2473618A1 (en) * 2009-09-02 2012-07-11 Quantalife, Inc System for mixing fluids by coalescence of multiple emulsions
US9194861B2 (en) 2009-09-02 2015-11-24 Bio-Rad Laboratories, Inc. Method of mixing fluids by coalescence of multiple emulsions
US10677693B2 (en) 2009-09-02 2020-06-09 Bio-Rad Laboratories, Inc. System for mixing fluids by coalescence of multiple emulsions
WO2011028539A1 (en) * 2009-09-02 2011-03-10 Quantalife, Inc. System for mixing fluids by coalescence of multiple emulsions
US10874997B2 (en) 2009-09-02 2020-12-29 President And Fellows Of Harvard College Multiple emulsions created using jetting and other techniques
EP2473618A4 (en) * 2009-09-02 2013-01-23 Quantalife Inc System for mixing fluids by coalescence of multiple emulsions
US10520500B2 (en) 2009-10-09 2019-12-31 Abdeslam El Harrak Labelled silica-based nanomaterial with enhanced properties and uses thereof
JP2013508156A (en) * 2009-10-27 2013-03-07 プレジデント アンド フェロウズ オブ ハーバード カレッジ Droplet generation technology
US11000849B2 (en) 2009-10-27 2021-05-11 President And Fellows Of Harvard College Droplet creation techniques
US9839911B2 (en) 2009-10-27 2017-12-12 President And Fellows Of Harvard College Droplet creation techniques
US10207240B2 (en) 2009-11-03 2019-02-19 Gen9, Inc. Methods and microfluidic devices for the manipulation of droplets in high fidelity polynucleotide assembly
US20210299619A1 (en) * 2009-11-12 2021-09-30 Stokes Bio Limited Methods of releasing and analyzing cellular components
CN102985552A (en) * 2009-11-25 2013-03-20 伯乐生命医学产品有限公司 Methods and compositions for detecting genetic material
US9968902B2 (en) 2009-11-25 2018-05-15 Gen9, Inc. Microfluidic devices and methods for gene synthesis
US20110159499A1 (en) * 2009-11-25 2011-06-30 Quantalife, Inc. Methods and compositions for detecting genetic material
US9216414B2 (en) 2009-11-25 2015-12-22 Gen9, Inc. Microfluidic devices and methods for gene synthesis
WO2011066476A1 (en) * 2009-11-25 2011-06-03 Quantalife, Inc. Methods and compositions for detecting genetic material
US8584703B2 (en) 2009-12-01 2013-11-19 Integenx Inc. Device with diaphragm valve
US20110126911A1 (en) * 2009-12-01 2011-06-02 IntegenX Inc., a California Corporation Composite Plastic Articles
WO2011079176A2 (en) 2009-12-23 2011-06-30 Raindance Technologies, Inc. Microfluidic systems and methods for reducing the exchange of molecules between droplets
US10837883B2 (en) 2009-12-23 2020-11-17 Bio-Rad Laboratories, Inc. Microfluidic systems and methods for reducing the exchange of molecules between droplets
WO2011079176A3 (en) * 2009-12-23 2011-11-10 Raindance Technologies, Inc. Microfluidic systems and methods for reducing the exchange of molecules between droplets
US9217144B2 (en) * 2010-01-07 2015-12-22 Gen9, Inc. Assembly of high fidelity polynucleotides
US20130059761A1 (en) * 2010-01-07 2013-03-07 Gen 9, Inc. Assembly of High Fidelity Polynucleotides
US20160144333A1 (en) * 2010-01-07 2016-05-26 Gen9, Inc. Assembly of High Fidelity Polynucleotides
US9925510B2 (en) * 2010-01-07 2018-03-27 Gen9, Inc. Assembly of high fidelity polynucleotides
US11071963B2 (en) 2010-01-07 2021-07-27 Gen9, Inc. Assembly of high fidelity polynucleotides
US20130295568A1 (en) * 2010-02-12 2013-11-07 Darren Roy Link Digital analyte analysis
US9228229B2 (en) 2010-02-12 2016-01-05 Raindance Technologies, Inc. Digital analyte analysis
US10351905B2 (en) 2010-02-12 2019-07-16 Bio-Rad Laboratories, Inc. Digital analyte analysis
US9494520B2 (en) * 2010-02-12 2016-11-15 Raindance Technologies, Inc. Digital analyte analysis
US8535889B2 (en) * 2010-02-12 2013-09-17 Raindance Technologies, Inc. Digital analyte analysis
US9399797B2 (en) 2010-02-12 2016-07-26 Raindance Technologies, Inc. Digital analyte analysis
US20110244455A1 (en) * 2010-02-12 2011-10-06 Raindance Technologies, Inc. Digital analyte analysis
US9074242B2 (en) * 2010-02-12 2015-07-07 Raindance Technologies, Inc. Digital analyte analysis
US20110250597A1 (en) * 2010-02-12 2011-10-13 Raindance Technologies, Inc. Digital analyte analysis
US10808279B2 (en) 2010-02-12 2020-10-20 Bio-Rad Laboratories, Inc. Digital analyte analysis
EP3392349A1 (en) 2010-02-12 2018-10-24 Raindance Technologies, Inc. Digital analyte analysis
US11254968B2 (en) 2010-02-12 2022-02-22 Bio-Rad Laboratories, Inc. Digital analyte analysis
WO2011100604A2 (en) 2010-02-12 2011-08-18 Raindance Technologies, Inc. Digital analyte analysis
US9366632B2 (en) 2010-02-12 2016-06-14 Raindance Technologies, Inc. Digital analyte analysis
US11390917B2 (en) 2010-02-12 2022-07-19 Bio-Rad Laboratories, Inc. Digital analyte analysis
US11866771B2 (en) 2010-03-02 2024-01-09 Bio-Rad Laboratories, Inc. Emulsion chemistry for encapsulated droplets
US10378048B2 (en) 2010-03-02 2019-08-13 Bio-Rad Laboratories, Inc. Emulsion chemistry for encapsulated droplets
US11060136B2 (en) 2010-03-02 2021-07-13 Bio-Rad Laboratories, Inc. Emulsion chemistry for encapsulated droplets
US20110217736A1 (en) * 2010-03-02 2011-09-08 Quantalife, Inc. System for hot-start amplification via a multiple emulsion
US8709762B2 (en) * 2010-03-02 2014-04-29 Bio-Rad Laboratories, Inc. System for hot-start amplification via a multiple emulsion
US9598725B2 (en) 2010-03-02 2017-03-21 Bio-Rad Laboratories, Inc. Emulsion chemistry for encapsulated droplets
US20130052648A1 (en) * 2010-03-12 2013-02-28 The General Hospital Corporation Amplifying rare cell surface markers
US20190056405A1 (en) * 2010-03-12 2019-02-21 Massachusetts Gen Hospital Amplifying rare cell surface markers
US10054591B2 (en) 2010-03-12 2018-08-21 The General Hospital Corporation Amplifying rare cell surface markers
US20110229545A1 (en) * 2010-03-17 2011-09-22 President And Fellows Of Harvard College Melt emulsification
US9500664B2 (en) 2010-03-25 2016-11-22 Bio-Rad Laboratories, Inc. Droplet generation for droplet-based assays
US10099219B2 (en) 2010-03-25 2018-10-16 Bio-Rad Laboratories, Inc. Device for generating droplets
US9393560B2 (en) 2010-03-25 2016-07-19 Bio-Rad Laboratories, Inc. Droplet transport system for detection
US10744506B2 (en) 2010-03-25 2020-08-18 Bio-Rad Laboratories, Inc. Device for generating droplets
US10272432B2 (en) 2010-03-25 2019-04-30 Bio-Rad Laboratories, Inc. Device for generating droplets
US8730479B2 (en) 2010-03-25 2014-05-20 Bio-Rad Laboratories, Inc. Detection system for droplet-based assays
US9442106B2 (en) * 2010-05-08 2016-09-13 Universiteit Twente Simple and affordable method for immunophenotyping using a microfluidic chip sample preparation with image cytometry
EP2585578A2 (en) * 2010-05-08 2013-05-01 Veridex, LLC A simple and affordable method for immuophenotyping using a microfluidic chip sample preparation with image cytometry
WO2011143075A3 (en) * 2010-05-08 2011-12-29 Veridex, Llc A simple and affordable method for immuophenotyping using a microfluidic chip sample preparation with image cytometry
EP2585578A4 (en) * 2010-05-08 2014-01-08 Univ Twente A simple and affordable method for immuophenotyping using a microfluidic chip sample preparation with image cytometry
US20140038230A1 (en) * 2010-05-08 2014-02-06 Veridex, Llc A simple and affordable method for immunophenotyping using a microfluidic chip sample preparation with image cytometry
US8512538B2 (en) 2010-05-28 2013-08-20 Integenx Inc. Capillary electrophoresis device
US10641729B2 (en) 2010-06-30 2020-05-05 Life Technologies Corporation Column ADC
US8772698B2 (en) 2010-06-30 2014-07-08 Life Technologies Corporation CCD-based multi-transistor active pixel sensor array
US8731847B2 (en) 2010-06-30 2014-05-20 Life Technologies Corporation Array configuration and readout scheme
US9239313B2 (en) 2010-06-30 2016-01-19 Life Technologies Corporation Ion-sensing charge-accumulation circuits and methods
US8415177B2 (en) 2010-06-30 2013-04-09 Life Technologies Corporation Two-transistor pixel array
US8983783B2 (en) 2010-06-30 2015-03-17 Life Technologies Corporation Chemical detection device having multiple flow channels
US8823380B2 (en) 2010-06-30 2014-09-02 Life Technologies Corporation Capacitive charge pump
US9164070B2 (en) 2010-06-30 2015-10-20 Life Technologies Corporation Column adc
US8415176B2 (en) 2010-06-30 2013-04-09 Life Technologies Corporation One-transistor pixel array
US8217433B1 (en) 2010-06-30 2012-07-10 Life Technologies Corporation One-transistor pixel array
US8421437B2 (en) 2010-06-30 2013-04-16 Life Technologies Corporation Array column integrator
US8247849B2 (en) 2010-06-30 2012-08-21 Life Technologies Corporation Two-transistor pixel array
US8741680B2 (en) 2010-06-30 2014-06-03 Life Technologies Corporation Two-transistor pixel array
US8742471B2 (en) 2010-06-30 2014-06-03 Life Technologies Corporation Chemical sensor array with leakage compensation circuit
US8432149B2 (en) 2010-06-30 2013-04-30 Life Technologies Corporation Array column integrator
US11231451B2 (en) 2010-06-30 2022-01-25 Life Technologies Corporation Methods and apparatus for testing ISFET arrays
US8487790B2 (en) 2010-06-30 2013-07-16 Life Technologies Corporation Chemical detection circuit including a serializer circuit
US8432150B2 (en) 2010-06-30 2013-04-30 Life Technologies Corporation Methods for operating an array column integrator
US8858782B2 (en) 2010-06-30 2014-10-14 Life Technologies Corporation Ion-sensing charge-accumulation circuits and methods
US10481123B2 (en) 2010-06-30 2019-11-19 Life Technologies Corporation Ion-sensing charge-accumulation circuits and methods
US8455927B2 (en) 2010-06-30 2013-06-04 Life Technologies Corporation One-transistor pixel array with cascoded column circuit
US8524487B2 (en) 2010-06-30 2013-09-03 Life Technologies Corporation One-transistor pixel array with cascoded column circuit
US11307166B2 (en) 2010-07-01 2022-04-19 Life Technologies Corporation Column ADC
US8653567B2 (en) 2010-07-03 2014-02-18 Life Technologies Corporation Chemically sensitive sensor with lightly doped drains
US9960253B2 (en) 2010-07-03 2018-05-01 Life Technologies Corporation Chemically sensitive sensor with lightly doped drains
WO2012013316A1 (en) * 2010-07-26 2012-02-02 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. Method and device for passively separating and sorting drops, in particular in a microfluidic system, by using non-optical markers for reactions within the drops
US8968873B2 (en) * 2010-08-13 2015-03-03 University Of Connecticut Co-flow microfluidic device for polymersome formation
WO2012021838A1 (en) * 2010-08-13 2012-02-16 University Of Connecticut Co-flow microfluidic device for polymersome formation
US20120141796A1 (en) * 2010-08-13 2012-06-07 University Of Connecticut Co-Flow Microfluidic Device for Polymersome Formation
US8763642B2 (en) 2010-08-20 2014-07-01 Integenx Inc. Microfluidic devices with mechanically-sealed diaphragm valves
US9731266B2 (en) 2010-08-20 2017-08-15 Integenx Inc. Linear valve arrays
US9121058B2 (en) 2010-08-20 2015-09-01 Integenx Inc. Linear valve arrays
US9701997B2 (en) 2010-08-31 2017-07-11 Canon U.S. Life Sciences, Inc. Composition and method for in-system priming microfluidic devices
US20120052560A1 (en) * 2010-08-31 2012-03-01 Canon U.S. Life Sciences, Inc. System and method for serial processing of multiple nucleic acid assays
US9114399B2 (en) * 2010-08-31 2015-08-25 Canon U.S. Life Sciences, Inc. System and method for serial processing of multiple nucleic acid assays
US10363558B2 (en) 2010-08-31 2019-07-30 Canon U.S. Life Sciences, Inc. System and method for serial processing of multiple nucleic acid assays
WO2012030878A1 (en) * 2010-08-31 2012-03-08 Canon U.S. Life Sciences, Inc. Composition and method for in-system priming microfluidic devices
US9618475B2 (en) 2010-09-15 2017-04-11 Life Technologies Corporation Methods and apparatus for measuring analytes
US9958414B2 (en) 2010-09-15 2018-05-01 Life Technologies Corporation Apparatus for measuring analytes including chemical sensor array
US9958415B2 (en) 2010-09-15 2018-05-01 Life Technologies Corporation ChemFET sensor including floating gate
US8912005B1 (en) 2010-09-24 2014-12-16 Life Technologies Corporation Method and system for delta double sampling
US9110015B2 (en) 2010-09-24 2015-08-18 Life Technologies Corporation Method and system for delta double sampling
US8796036B2 (en) 2010-09-24 2014-08-05 Life Technologies Corporation Method and system for delta double sampling
US8685324B2 (en) 2010-09-24 2014-04-01 Life Technologies Corporation Matched pair transistor circuits
EP3447155A1 (en) 2010-09-30 2019-02-27 Raindance Technologies, Inc. Sandwich assays in droplets
US9562897B2 (en) 2010-09-30 2017-02-07 Raindance Technologies, Inc. Sandwich assays in droplets
WO2012045012A2 (en) 2010-09-30 2012-04-05 Raindance Technologies, Inc. Sandwich assays in droplets
US11635427B2 (en) 2010-09-30 2023-04-25 Bio-Rad Laboratories, Inc. Sandwich assays in droplets
US8961764B2 (en) 2010-10-15 2015-02-24 Lockheed Martin Corporation Micro fluidic optic design
US9089844B2 (en) 2010-11-01 2015-07-28 Bio-Rad Laboratories, Inc. System for forming emulsions
WO2012078710A1 (en) * 2010-12-07 2012-06-14 Gnubio, Inc. Nucleic acid target detection using a detector, a probe and an inhibitor
US9581549B2 (en) 2010-12-07 2017-02-28 Gnubio, Inc. Nucleic acid target detection using a detector, a probe and an inhibitor
US10533212B2 (en) 2010-12-07 2020-01-14 Bio-Rad Laboratories, Inc. Nucleic acid target detection using a detector, a probe and an inhibitor
US11041851B2 (en) 2010-12-23 2021-06-22 Molecular Loop Biosciences, Inc. Methods for maintaining the integrity and identification of a nucleic acid template in a multiplex sequencing reaction
US11041852B2 (en) 2010-12-23 2021-06-22 Molecular Loop Biosciences, Inc. Methods for maintaining the integrity and identification of a nucleic acid template in a multiplex sequencing reaction
US11768200B2 (en) 2010-12-23 2023-09-26 Molecular Loop Biosciences, Inc. Methods for maintaining the integrity and identification of a nucleic acid template in a multiplex sequencing reaction
US11499181B2 (en) 2011-02-09 2022-11-15 Bio-Rad Laboratories, Inc. Analysis of nucleic acids
US9127312B2 (en) 2011-02-09 2015-09-08 Bio-Rad Laboratories, Inc. Analysis of nucleic acids
US10167509B2 (en) 2011-02-09 2019-01-01 Bio-Rad Laboratories, Inc. Analysis of nucleic acids
US11077415B2 (en) 2011-02-11 2021-08-03 Bio-Rad Laboratories, Inc. Methods for forming mixed droplets
EP3412778A1 (en) 2011-02-11 2018-12-12 Raindance Technologies, Inc. Methods for forming mixed droplets
WO2012109600A3 (en) * 2011-02-11 2013-11-28 Raindance Technologies, Inc. Methods for forming mixed droplets
WO2012109600A2 (en) 2011-02-11 2012-08-16 Raindance Technologies, Inc. Methods for forming mixed droplets
EP3859011A1 (en) 2011-02-11 2021-08-04 Bio-Rad Laboratories, Inc. Methods for forming mixed droplets
US9364803B2 (en) 2011-02-11 2016-06-14 Raindance Technologies, Inc. Methods for forming mixed droplets
WO2012109604A1 (en) 2011-02-11 2012-08-16 Raindance Technologies, Inc. Thermocycling device for nucleic acid amplification and methods of use
US20180251771A1 (en) * 2011-02-16 2018-09-06 Glycosyn LLC Biosynthesis of human milk oligosaccharides in engineered bacteria
EP3736281A1 (en) 2011-02-18 2020-11-11 Bio-Rad Laboratories, Inc. Compositions and methods for molecular labeling
US9150852B2 (en) 2011-02-18 2015-10-06 Raindance Technologies, Inc. Compositions and methods for molecular labeling
US11747327B2 (en) 2011-02-18 2023-09-05 Bio-Rad Laboratories, Inc. Compositions and methods for molecular labeling
US11768198B2 (en) 2011-02-18 2023-09-26 Bio-Rad Laboratories, Inc. Compositions and methods for molecular labeling
US11168353B2 (en) 2011-02-18 2021-11-09 Bio-Rad Laboratories, Inc. Compositions and methods for molecular labeling
US9222128B2 (en) 2011-03-18 2015-12-29 Bio-Rad Laboratories, Inc. Multiplexed digital assays with combinatorial use of signals
US9228898B2 (en) 2011-03-31 2016-01-05 Gnubio, Inc. Scalable spectroscopic detection and measurement
US20140193800A1 (en) * 2011-04-08 2014-07-10 Stokes Bio Limited Systems and Methods for Continuous Flow PCR Systems
WO2012139038A1 (en) * 2011-04-08 2012-10-11 Stokes Bio Limited Systems and methods for continuous flow pcr systems
US10190115B2 (en) 2011-04-25 2019-01-29 Bio-Rad Laboratories, Inc. Methods and compositions for nucleic acid analysis
US9347059B2 (en) 2011-04-25 2016-05-24 Bio-Rad Laboratories, Inc. Methods and compositions for nucleic acid analysis
US9885034B2 (en) 2011-04-25 2018-02-06 Bio-Rad Laboratories, Inc. Methods and compositions for nucleic acid analysis
US10760073B2 (en) 2011-04-25 2020-09-01 Bio-Rad Laboratories, Inc. Methods and compositions for nucleic acid analysis
US9573099B2 (en) 2011-05-23 2017-02-21 President And Fellows Of Harvard College Control of emulsions, including multiple emulsions
US9238206B2 (en) 2011-05-23 2016-01-19 President And Fellows Of Harvard College Control of emulsions, including multiple emulsions
US20140295421A1 (en) * 2011-06-02 2014-10-02 Raindance Technologies, Inc. Enzyme quantification
EP3709018A1 (en) 2011-06-02 2020-09-16 Bio-Rad Laboratories, Inc. Microfluidic apparatus for identifying components of a chemical reaction
EP2714970A4 (en) * 2011-06-02 2015-10-14 Raindance Technologies Inc Enzyme quantification
WO2012167142A3 (en) * 2011-06-02 2014-05-08 Raindance Technolgies, Inc. Enzyme quantification
WO2012167142A2 (en) 2011-06-02 2012-12-06 Raindance Technolgies, Inc. Enzyme quantification
US8841071B2 (en) 2011-06-02 2014-09-23 Raindance Technologies, Inc. Sample multiplexing
US9556470B2 (en) * 2011-06-02 2017-01-31 Raindance Technologies, Inc. Enzyme quantification
EP3216872A1 (en) 2011-06-02 2017-09-13 Raindance Technologies, Inc. Enzyme quantification
DE202012013668U1 (en) 2011-06-02 2019-04-18 Raindance Technologies, Inc. enzyme quantification
US11754499B2 (en) 2011-06-02 2023-09-12 Bio-Rad Laboratories, Inc. Enzyme quantification
US10195571B2 (en) 2011-07-06 2019-02-05 President And Fellows Of Harvard College Multiple emulsions and techniques for the formation of multiple emulsions
US8951939B2 (en) 2011-07-12 2015-02-10 Bio-Rad Laboratories, Inc. Digital assays with multiplexed detection of two or more targets in the same optical channel
US11898193B2 (en) 2011-07-20 2024-02-13 Bio-Rad Laboratories, Inc. Manipulating droplet size
US8658430B2 (en) 2011-07-20 2014-02-25 Raindance Technologies, Inc. Manipulating droplet size
US9574245B2 (en) 2011-07-25 2017-02-21 Qvella Corporation Methods and devices for electrical sample preparation
US8663920B2 (en) 2011-07-29 2014-03-04 Bio-Rad Laboratories, Inc. Library characterization by digital assay
US10408737B1 (en) 2011-08-01 2019-09-10 Celsee Diagnostics, Inc. Cell capture system and method of use
US9103754B2 (en) 2011-08-01 2015-08-11 Denovo Sciences, Inc. Cell capture system and method of use
US10746648B2 (en) 2011-08-01 2020-08-18 Bio-Rad Laboratories, Inc. Cell capture and method of use
US9513195B2 (en) 2011-08-01 2016-12-06 Denovo Sciences, Inc. Cell capture system and method of use
WO2013019491A1 (en) * 2011-08-01 2013-02-07 Denovo Sciences Cell capture system and method of use
US10466160B2 (en) 2011-08-01 2019-11-05 Celsee Diagnostics, Inc. System and method for retrieving and analyzing particles
US10921237B2 (en) 2011-08-01 2021-02-16 Bio-Rad Laboratories, Inc. Cell capture system and method of use
US11635365B2 (en) 2011-08-01 2023-04-25 Bio-Rad Laboratories, Inc. Cell capture system and method of use
US10914672B2 (en) 2011-08-01 2021-02-09 Bio-Rad Laboratories, Inc. System and method for retrieving and analyzing particles
US10401277B2 (en) 2011-08-01 2019-09-03 Celsee Diagnostics, Inc. Cell capture system and method of use
US10345219B2 (en) 2011-08-01 2019-07-09 Celsee Diagnostics, Inc. Cell capture system and method of use
US10408736B1 (en) 2011-08-01 2019-09-10 Celsee Diagnostics, Inc. Cell capture system and method of use
US10533936B1 (en) 2011-08-01 2020-01-14 Celsee Diagnostics, Inc. Cell capture system and method of use
US10416070B1 (en) 2011-08-01 2019-09-17 Celsee Diagnostics, Inc. Cell capture system and method of use
US10641700B2 (en) 2011-08-01 2020-05-05 Celsee Diagnostics, Inc. Cell capture system and method of use
US11073468B2 (en) 2011-08-01 2021-07-27 Bio-Rad Laboratories, Inc. Cell capture system and method of use
US10782226B1 (en) 2011-08-01 2020-09-22 Bio-Rad Laboratories, Inc. Cell capture system and method of use
US9746413B2 (en) 2011-08-01 2017-08-29 Denovo Sciences, Inc. Cell capture system and method of use
US10564090B2 (en) 2011-08-01 2020-02-18 Celsee Diagnostics, Inc. System and method for retrieving and analyzing particles
US10794817B1 (en) 2011-08-01 2020-10-06 Bio-Rad Laboratories, Inc. Cell capture system and method of use
US10190965B2 (en) 2011-08-01 2019-01-29 Celsee Diagnostics, Inc. Cell capture system and method of use
US10591404B1 (en) 2011-08-01 2020-03-17 Celsee Diagnostics, Inc. Cell capture system and method of use
US11300496B2 (en) 2011-08-01 2022-04-12 Bio-Rad Laboratories, Inc. Cell capture system and method of use
US10481077B1 (en) 2011-08-01 2019-11-19 Celsee Diagnostics, Inc. Cell capture system and method of use
US11231355B2 (en) 2011-08-01 2022-01-25 Bio-Rad Laboratories, Inc. Cell capture system and method of use
US11275015B2 (en) 2011-08-01 2022-03-15 Bio-Rad Laboratories, Inc. System and method for retrieving and analyzing particles
US11237096B2 (en) 2011-08-01 2022-02-01 Bio-Rad Laboratories, Inc. Cell capture system and method of use
US10436700B1 (en) 2011-08-01 2019-10-08 Celsee Diagnostics, Inc. Cell capture system and method of use
US11389800B2 (en) 2011-09-28 2022-07-19 President And Fellows Of Harvard College Systems and methods for droplet production and/or fluidic manipulation
US9228233B2 (en) 2011-10-17 2016-01-05 Good Start Genetics, Inc. Analysis methods
US9822409B2 (en) 2011-10-17 2017-11-21 Good Start Genetics, Inc. Analysis methods
US10370710B2 (en) 2011-10-17 2019-08-06 Good Start Genetics, Inc. Analysis methods
US10865440B2 (en) 2011-10-21 2020-12-15 IntegenX, Inc. Sample preparation, processing and analysis systems
US11684918B2 (en) 2011-10-21 2023-06-27 IntegenX, Inc. Sample preparation, processing and analysis systems
US10525467B2 (en) 2011-10-21 2020-01-07 Integenx Inc. Sample preparation, processing and analysis systems
US10598723B2 (en) 2011-12-01 2020-03-24 Life Technologies Corporation Method and apparatus for identifying defects in a chemical sensor array
US9970984B2 (en) 2011-12-01 2018-05-15 Life Technologies Corporation Method and apparatus for identifying defects in a chemical sensor array
US10365321B2 (en) 2011-12-01 2019-07-30 Life Technologies Corporation Method and apparatus for identifying defects in a chemical sensor array
US8821798B2 (en) 2012-01-19 2014-09-02 Life Technologies Corporation Titanium nitride as sensing layer for microwell structure
US8747748B2 (en) 2012-01-19 2014-06-10 Life Technologies Corporation Chemical sensor with conductive cup-shaped sensor surface
EP3495817A1 (en) 2012-02-10 2019-06-12 Raindance Technologies, Inc. Molecular diagnostic screening assay
EP3309262A1 (en) 2012-02-24 2018-04-18 Raindance Technologies, Inc. Labeling and sample preparation for sequencing
WO2013126741A1 (en) 2012-02-24 2013-08-29 Raindance Technologies, Inc. Labeling and sample preparation for sequencing
US9648714B2 (en) 2012-03-27 2017-05-09 Asml Netherlands B.V. Fuel system for lithographic apparatus, EUV source, lithographic apparatus and fuel filtering method
WO2013143733A1 (en) * 2012-03-27 2013-10-03 Asml Netherlands B.V. Fuel system for lithographic apparatus, euv source,lithographic apparatus and fuel filtering method
US11149308B2 (en) 2012-04-04 2021-10-19 Invitae Corporation Sequence assembly
US10604799B2 (en) 2012-04-04 2020-03-31 Molecular Loop Biosolutions, Llc Sequence assembly
US11155863B2 (en) 2012-04-04 2021-10-26 Invitae Corporation Sequence assembly
US11667965B2 (en) 2012-04-04 2023-06-06 Invitae Corporation Sequence assembly
US8812422B2 (en) 2012-04-09 2014-08-19 Good Start Genetics, Inc. Variant database
US9298804B2 (en) 2012-04-09 2016-03-29 Good Start Genetics, Inc. Variant database
US9399215B2 (en) 2012-04-13 2016-07-26 Bio-Rad Laboratories, Inc. Sample holder with a well having a wicking promoter
US10683533B2 (en) 2012-04-16 2020-06-16 Molecular Loop Biosolutions, Llc Capture reactions
US10227635B2 (en) 2012-04-16 2019-03-12 Molecular Loop Biosolutions, Llc Capture reactions
WO2013159117A1 (en) 2012-04-20 2013-10-24 SlipChip, LLC Fluidic devices and systems for sample preparation or autonomous analysis
US9808798B2 (en) 2012-04-20 2017-11-07 California Institute Of Technology Fluidic devices for biospecimen preservation
US9822356B2 (en) 2012-04-20 2017-11-21 California Institute Of Technology Fluidic devices and systems for sample preparation or autonomous analysis
US9803237B2 (en) 2012-04-24 2017-10-31 California Institute Of Technology Slip-induced compartmentalization
WO2013165748A1 (en) 2012-04-30 2013-11-07 Raindance Technologies, Inc Digital analyte analysis
EP3524693A1 (en) 2012-04-30 2019-08-14 Raindance Technologies, Inc. Digital analyte analysis
US9270264B2 (en) 2012-05-29 2016-02-23 Life Technologies Corporation System for reducing noise in a chemical sensor array
US8786331B2 (en) 2012-05-29 2014-07-22 Life Technologies Corporation System for reducing noise in a chemical sensor array
US10404249B2 (en) 2012-05-29 2019-09-03 Life Technologies Corporation System for reducing noise in a chemical sensor array
US9985624B2 (en) 2012-05-29 2018-05-29 Life Technologies Corporation System for reducing noise in a chemical sensor array
US8552771B1 (en) 2012-05-29 2013-10-08 Life Technologies Corporation System for reducing noise in a chemical sensor array
CN103512841B (en) * 2012-06-22 2015-12-02 纬创资通股份有限公司 Biological sample pick-up unit and biological sample detection method
US9410178B2 (en) * 2012-06-22 2016-08-09 Wistron Corporation Biological particle analyzer and method of analyzing biological particles
US20130344534A1 (en) * 2012-06-22 2013-12-26 Wistron Corporation Biological Particle Analyzer and Method of Analyzing Biological Particles
CN103512841A (en) * 2012-06-22 2014-01-15 纬创资通股份有限公司 Biological particle analyzer and method of analyzing biological particles
US10745762B2 (en) 2012-08-13 2020-08-18 The Regents Of The University Of California Method and system for synthesizing a target polynucleotide within a droplet
US20160177375A1 (en) * 2012-08-13 2016-06-23 The Regents Of The University Of California Methods and systems for detecting biological components
US11203787B2 (en) 2012-08-13 2021-12-21 The Regents Of The University Of California Methods and systems for detecting biological components
US11891666B2 (en) 2012-08-13 2024-02-06 The Regents Of The University Of California Methods and systems for detecting biological components
US10161007B2 (en) 2012-08-13 2018-12-25 The Regents Of The University Of California Methods and systems for detecting biological components
US11001896B2 (en) 2012-08-13 2021-05-11 The Regents Of The University Of California System and method to synthesize a target molecule within a droplet
US9695468B2 (en) 2012-08-14 2017-07-04 10X Genomics, Inc. Methods for droplet-based sample preparation
US10597718B2 (en) 2012-08-14 2020-03-24 10X Genomics, Inc. Methods and systems for sample processing polynucleotides
US10053723B2 (en) 2012-08-14 2018-08-21 10X Genomics, Inc. Capsule array devices and methods of use
US11359239B2 (en) 2012-08-14 2022-06-14 10X Genomics, Inc. Methods and systems for processing polynucleotides
US11021749B2 (en) 2012-08-14 2021-06-01 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10669583B2 (en) 2012-08-14 2020-06-02 10X Genomics, Inc. Method and systems for processing polynucleotides
US10752950B2 (en) 2012-08-14 2020-08-25 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10400280B2 (en) 2012-08-14 2019-09-03 10X Genomics, Inc. Methods and systems for processing polynucleotides
US11591637B2 (en) 2012-08-14 2023-02-28 10X Genomics, Inc. Compositions and methods for sample processing
US10752949B2 (en) 2012-08-14 2020-08-25 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10584381B2 (en) 2012-08-14 2020-03-10 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10221442B2 (en) 2012-08-14 2019-03-05 10X Genomics, Inc. Compositions and methods for sample processing
US11078522B2 (en) 2012-08-14 2021-08-03 10X Genomics, Inc. Capsule array devices and methods of use
US11035002B2 (en) 2012-08-14 2021-06-15 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10273541B2 (en) 2012-08-14 2019-04-30 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10323279B2 (en) 2012-08-14 2019-06-18 10X Genomics, Inc. Methods and systems for processing polynucleotides
US11441179B2 (en) 2012-08-14 2022-09-13 10X Genomics, Inc. Methods and systems for processing polynucleotides
US9689024B2 (en) 2012-08-14 2017-06-27 10X Genomics, Inc. Methods for droplet-based sample preparation
US10626458B2 (en) 2012-08-14 2020-04-21 10X Genomics, Inc. Methods and systems for processing polynucleotides
US20170321252A1 (en) * 2012-08-14 2017-11-09 10X Genomics, Inc. Capsule array devices and methods of use
US10450607B2 (en) 2012-08-14 2019-10-22 10X Genomics, Inc. Methods and systems for processing polynucleotides
CN104797718A (en) * 2012-09-12 2015-07-22 加利福尼亚大学董事会 Accurate genome sequencing of single cells by single-stranded amplification and sequencing
WO2014043140A1 (en) 2012-09-12 2014-03-20 The Regents Of The University Of California Accurate genome sequencing of single cells by single-stranded amplification and sequencing
EP2895626B1 (en) * 2012-09-12 2019-12-18 The Regents of The University of California Accurate genome sequencing of single cells by single-stranded amplification and sequencing
WO2014043388A1 (en) 2012-09-12 2014-03-20 Gnubio, Inc. Integrated microfluidic system, method and kit for performing assays
US10752945B2 (en) 2012-09-12 2020-08-25 The Regents Of The University Of California Accurate genome sequencing of single cells by single-stranded amplification and sequencing
EP2895591A4 (en) * 2012-09-12 2016-10-12 Gnubio Inc Integrated microfluidic system, method and kit for performing assays
US10343167B2 (en) 2012-09-12 2019-07-09 Bio-Rad Laboratories, Inc. Integrated microfluidic system, method and kit for performing assays
US9821312B2 (en) 2012-09-12 2017-11-21 Bio-Rad Laboratories, Inc. Integrated microfluidic system, method and kit for performing assays
US9981273B2 (en) 2012-09-28 2018-05-29 The Board Of Trustees Of The Leland Stanford Junior University Negative dielectrophoresis for selective elution of immuno-bound particles
US11179696B2 (en) 2012-11-30 2021-11-23 The Broad Institute Inc. High-throughput dynamic reagent delivery system
US9567631B2 (en) 2012-12-14 2017-02-14 10X Genomics, Inc. Methods and systems for processing polynucleotides
US9701998B2 (en) 2012-12-14 2017-07-11 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10612090B2 (en) 2012-12-14 2020-04-07 10X Genomics, Inc. Methods and systems for processing polynucleotides
US11421274B2 (en) 2012-12-14 2022-08-23 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10676789B2 (en) 2012-12-14 2020-06-09 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10227648B2 (en) 2012-12-14 2019-03-12 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10253364B2 (en) 2012-12-14 2019-04-09 10X Genomics, Inc. Method and systems for processing polynucleotides
US10533221B2 (en) 2012-12-14 2020-01-14 10X Genomics, Inc. Methods and systems for processing polynucleotides
US11473138B2 (en) 2012-12-14 2022-10-18 10X Genomics, Inc. Methods and systems for processing polynucleotides
US9410201B2 (en) 2012-12-14 2016-08-09 10X Genomics, Inc. Methods and systems for processing polynucleotides
US9856530B2 (en) 2012-12-14 2018-01-02 10X Genomics, Inc. Methods and systems for processing polynucleotides
US9080968B2 (en) 2013-01-04 2015-07-14 Life Technologies Corporation Methods and systems for point of use removal of sacrificial material
US9852919B2 (en) 2013-01-04 2017-12-26 Life Technologies Corporation Methods and systems for point of use removal of sacrificial material
US9841398B2 (en) 2013-01-08 2017-12-12 Life Technologies Corporation Methods for manufacturing well structures for low-noise chemical sensors
US10436742B2 (en) 2013-01-08 2019-10-08 Life Technologies Corporation Methods for manufacturing well structures for low-noise chemical sensors
WO2014117088A1 (en) 2013-01-25 2014-07-31 Gnubio, Inc. System and method for performing droplet inflation
EP3473905A1 (en) 2013-01-25 2019-04-24 Bio-rad Laboratories, Inc. System and method for performing droplet inflation
US11345951B2 (en) 2013-01-26 2022-05-31 Bio-Rad Laboratories, Inc. System and method for capturing and analyzing cells
US10975422B2 (en) 2013-01-26 2021-04-13 Bio-Rad Laboratories, Inc. System and method for capturing and analyzing cells
US9752181B2 (en) 2013-01-26 2017-09-05 Denovo Sciences, Inc. System and method for capturing and analyzing cells
US9606102B2 (en) 2013-01-26 2017-03-28 Denovo Sciences, Inc. System and method for capturing and analyzing cells
US10718007B2 (en) 2013-01-26 2020-07-21 Bio-Rad Laboratories, Inc. System and method for capturing and analyzing cells
US8962366B2 (en) 2013-01-28 2015-02-24 Life Technologies Corporation Self-aligned well structures for low-noise chemical sensors
US11110458B2 (en) 2013-02-01 2021-09-07 Bio-Rad Laboratories, Inc. System for detection of spaced droplets
US11878302B2 (en) 2013-02-01 2024-01-23 Bio-Rad Laboratories, Inc. System for detection of spaced droplets
US10150963B2 (en) 2013-02-08 2018-12-11 10X Genomics, Inc. Partitioning and processing of analytes and other species
US11193121B2 (en) 2013-02-08 2021-12-07 10X Genomics, Inc. Partitioning and processing of analytes and other species
US10150964B2 (en) 2013-02-08 2018-12-11 10X Genomics, Inc. Partitioning and processing of analytes and other species
US9644204B2 (en) 2013-02-08 2017-05-09 10X Genomics, Inc. Partitioning and processing of analytes and other species
US9388465B2 (en) 2013-02-08 2016-07-12 10X Genomics, Inc. Polynucleotide barcode generation
US9802193B2 (en) 2013-03-13 2017-10-31 Denovo Sciences, Inc. System and method for capturing and analyzing cells
US9995708B2 (en) 2013-03-13 2018-06-12 Life Technologies Corporation Chemical sensor with sidewall spacer sensor surface
US9610581B2 (en) 2013-03-13 2017-04-04 Denovo Sciences, Inc. System and method for capturing and analyzing cells
US10509022B2 (en) 2013-03-13 2019-12-17 Celsee Diagnostics, Inc. System for imaging captured cells
US9404864B2 (en) 2013-03-13 2016-08-02 Denovo Sciences, Inc. System for imaging captured cells
US8963216B2 (en) 2013-03-13 2015-02-24 Life Technologies Corporation Chemical sensor with sidewall spacer sensor surface
US9612199B2 (en) 2013-03-13 2017-04-04 Denovo Sciences, Inc. System for imaging captured cells
US10350601B2 (en) 2013-03-13 2019-07-16 Celsee Diagnostics, Inc. System and method for capturing and analyzing cells
US9925538B2 (en) 2013-03-13 2018-03-27 DeNovo Sciecnes, Inc. System and method for capturing and analyzing cells
US9821311B2 (en) 2013-03-13 2017-11-21 Denovo Sciences, Inc. System for capturing and analyzing cells
US9707562B2 (en) 2013-03-13 2017-07-18 Denovo Sciences, Inc. System for capturing and analyzing cells
US11199532B2 (en) 2013-03-13 2021-12-14 Bio-Rad Laboratories, Inc. System for imaging captured cells
US9174216B2 (en) 2013-03-13 2015-11-03 DeNovo Science, Inc. System for capturing and analyzing cells
US10690650B2 (en) 2013-03-13 2020-06-23 Bio-Rad Laboratories, Inc. System for imaging captured cells
US8841217B1 (en) 2013-03-13 2014-09-23 Life Technologies Corporation Chemical sensor with protruded sensor surface
US11530440B2 (en) 2013-03-14 2022-12-20 The Broad Institute, Inc. Methods for quantitating DNA using digital multiple displacement amplification
US9677124B2 (en) 2013-03-14 2017-06-13 Good Start Genetics, Inc. Methods for analyzing nucleic acids
US9115387B2 (en) 2013-03-14 2015-08-25 Good Start Genetics, Inc. Methods for analyzing nucleic acids
US10202637B2 (en) 2013-03-14 2019-02-12 Molecular Loop Biosolutions, Llc Methods for analyzing nucleic acid
US8778609B1 (en) 2013-03-14 2014-07-15 Good Start Genetics, Inc. Methods for analyzing nucleic acids
US9823217B2 (en) 2013-03-15 2017-11-21 Life Technologies Corporation Chemical device with thin conductive element
US10105702B2 (en) 2013-03-15 2018-10-23 Lariat Biosciences, Inc. Microfluidic methods for manipulating DNA
US9116117B2 (en) 2013-03-15 2015-08-25 Life Technologies Corporation Chemical sensor with sidewall sensor surface
US9671363B2 (en) 2013-03-15 2017-06-06 Life Technologies Corporation Chemical sensor with consistent sensor surface areas
US9128044B2 (en) 2013-03-15 2015-09-08 Life Technologies Corporation Chemical sensors with consistent sensor surface areas
US10481124B2 (en) 2013-03-15 2019-11-19 Life Technologies Corporation Chemical device with thin conductive element
WO2014145555A1 (en) * 2013-03-15 2014-09-18 Lariat Biosciences, Inc. Microfluidic methods for manipulating dna
US9835585B2 (en) 2013-03-15 2017-12-05 Life Technologies Corporation Chemical sensor with protruded sensor surface
US10422767B2 (en) 2013-03-15 2019-09-24 Life Technologies Corporation Chemical sensor with consistent sensor surface areas
WO2014172288A2 (en) 2013-04-19 2014-10-23 Raindance Technologies, Inc. Digital analyte analysis
US10655175B2 (en) 2013-05-09 2020-05-19 Life Technologies Corporation Windowed sequencing
US11028438B2 (en) 2013-05-09 2021-06-08 Life Technologies Corporation Windowed sequencing
WO2014182835A1 (en) * 2013-05-09 2014-11-13 Bio-Rad Laboratories, Inc. Magnetic immuno digital pcr assay
US9896717B2 (en) 2013-05-09 2018-02-20 Bio-Rad Laboratories, Inc. Magnetic immuno digital PCR assay
US10100357B2 (en) 2013-05-09 2018-10-16 Life Technologies Corporation Windowed sequencing
CN105431575A (en) * 2013-05-09 2016-03-23 生物辐射实验室股份有限公司 Magnetic immuno digital PCR assay
US10851426B2 (en) 2013-05-31 2020-12-01 Bio-Rad Laboratories, Inc. System and method for isolating and analyzing cells
US11052396B2 (en) 2013-05-31 2021-07-06 Bio-Rad Laboratories, Inc. System and method for isolating and analyzing cells
US10449543B2 (en) 2013-05-31 2019-10-22 Celsee Diagnostics, Inc. System and method for isolating and analyzing cells
US10391490B2 (en) 2013-05-31 2019-08-27 Celsee Diagnostics, Inc. System and method for isolating and analyzing cells
US11358147B2 (en) 2013-05-31 2022-06-14 Bio-Rad Laboratories, Inc. System and method for isolating and analyzing cells
US10512914B2 (en) 2013-05-31 2019-12-24 Celsee Diagnostics, Inc. System for isolating and analyzing cells in a single-cell format
US9856535B2 (en) 2013-05-31 2018-01-02 Denovo Sciences, Inc. System for isolating cells
US10706017B2 (en) 2013-06-03 2020-07-07 Good Start Genetics, Inc. Methods and systems for storing sequence read data
US9535920B2 (en) 2013-06-03 2017-01-03 Good Start Genetics, Inc. Methods and systems for storing sequence read data
US10458942B2 (en) 2013-06-10 2019-10-29 Life Technologies Corporation Chemical sensor array having multiple sensors per well
US10816504B2 (en) 2013-06-10 2020-10-27 Life Technologies Corporation Chemical sensor array having multiple sensors per well
US11499938B2 (en) 2013-06-10 2022-11-15 Life Technologies Corporation Chemical sensor array having multiple sensors per well
US11774401B2 (en) 2013-06-10 2023-10-03 Life Technologies Corporation Chemical sensor array having multiple sensors per well
US11219896B2 (en) 2013-06-25 2022-01-11 University Of Washington Through Its Center For Commercialization Self-digitization of sample volumes
US9944998B2 (en) 2013-07-25 2018-04-17 Bio-Rad Laboratories, Inc. Genetic assays
US10022721B2 (en) * 2013-08-27 2018-07-17 Bio-Rad Laboratories, Inc. Microfluidic devices and methods of their use
EP3039119A4 (en) * 2013-08-27 2017-04-05 GnuBIO, Inc. Microfluidic devices and methods of their use
EP3039119A1 (en) * 2013-08-27 2016-07-06 GnuBIO, Inc. Microfluidic devices and methods of their use
WO2015031528A1 (en) 2013-08-27 2015-03-05 Gnubio, Inc. Microfluidic devices and methods of their use
US10589274B2 (en) 2013-08-27 2020-03-17 Bio-Rad Laboratories, Inc. Microfluidic devices and methods of their use
US10395758B2 (en) 2013-08-30 2019-08-27 10X Genomics, Inc. Sequencing methods
DE102013217959A1 (en) * 2013-09-09 2015-03-12 Efficient Robotics Gmbh Microfluidic analysis device and manufacturing process
US10427157B2 (en) 2013-09-09 2019-10-01 Efficient Robotics Gmbh Microfluidic analysis component and production method
US11268632B2 (en) 2013-09-09 2022-03-08 Efficient Robotics Gmbh Microfluidic analysis component and production method
WO2015048798A1 (en) 2013-09-30 2015-04-02 Gnubio, Inc. Microfluidic cartridge device and methods of use and assembly
US9555411B2 (en) 2013-09-30 2017-01-31 Gnubio, Inc. Microfluidic cartridge devices and methods of use and assembly
US9776183B2 (en) 2013-09-30 2017-10-03 Bio-Rad Laboratories, Inc. Microfluidic cartridge devices and methods of use and assembly
US11901041B2 (en) 2013-10-04 2024-02-13 Bio-Rad Laboratories, Inc. Digital analysis of nucleic acid modification
US11041203B2 (en) 2013-10-18 2021-06-22 Molecular Loop Biosolutions, Inc. Methods for assessing a genomic region of a subject
US10851414B2 (en) 2013-10-18 2020-12-01 Good Start Genetics, Inc. Methods for determining carrier status
US10471016B2 (en) 2013-11-08 2019-11-12 President And Fellows Of Harvard College Microparticles, methods for their preparation and use
US10989723B2 (en) 2013-11-18 2021-04-27 IntegenX, Inc. Cartridges and instruments for sample analysis
US10191071B2 (en) 2013-11-18 2019-01-29 IntegenX, Inc. Cartridges and instruments for sample analysis
US10801070B2 (en) 2013-11-25 2020-10-13 The Broad Institute, Inc. Compositions and methods for diagnosing, evaluating and treating cancer
US11834718B2 (en) 2013-11-25 2023-12-05 The Broad Institute, Inc. Compositions and methods for diagnosing, evaluating and treating cancer by means of the DNA methylation status
WO2015081102A1 (en) 2013-11-27 2015-06-04 Gnubio, Inc. Microfluidic droplet packing
US11725237B2 (en) 2013-12-05 2023-08-15 The Broad Institute Inc. Polymorphic gene typing and somatic change detection using sequencing data
US11174509B2 (en) 2013-12-12 2021-11-16 Bio-Rad Laboratories, Inc. Distinguishing rare variations in a nucleic acid sequence from a sample
US9824068B2 (en) 2013-12-16 2017-11-21 10X Genomics, Inc. Methods and apparatus for sorting data
US11452768B2 (en) 2013-12-20 2022-09-27 The Broad Institute, Inc. Combination therapy with neoantigen vaccine
US11193176B2 (en) 2013-12-31 2021-12-07 Bio-Rad Laboratories, Inc. Method for detecting and quantifying latent retroviral RNA species
WO2015103367A1 (en) 2013-12-31 2015-07-09 Raindance Technologies, Inc. System and method for detection of rna species
US11209440B2 (en) 2014-02-27 2021-12-28 The Broad Institute, Inc. T cell balance gene expression, compositions of matters and methods of use thereof
EP3514246A1 (en) 2014-02-27 2019-07-24 The Broad Institute Inc. T cell balance gene expression and methods of use thereof
US9726653B2 (en) 2014-03-26 2017-08-08 Hamilton Sundstrand Corporation Chemical detector
US10071377B2 (en) 2014-04-10 2018-09-11 10X Genomics, Inc. Fluidic devices, systems, and methods for encapsulating and partitioning reagents, and applications of same
US9694361B2 (en) 2014-04-10 2017-07-04 10X Genomics, Inc. Fluidic devices, systems, and methods for encapsulating and partitioning reagents, and applications of same
US10137449B2 (en) 2014-04-10 2018-11-27 10X Genomics, Inc. Fluidic devices, systems, and methods for encapsulating and partitioning reagents, and applications of same
US10150117B2 (en) 2014-04-10 2018-12-11 10X Genomics, Inc. Fluidic devices, systems, and methods for encapsulating and partitioning reagents, and applications of same
US10343166B2 (en) 2014-04-10 2019-07-09 10X Genomics, Inc. Fluidic devices, systems, and methods for encapsulating and partitioning reagents, and applications of same
US11053548B2 (en) 2014-05-12 2021-07-06 Good Start Genetics, Inc. Methods for detecting aneuploidy
WO2015173651A1 (en) 2014-05-14 2015-11-19 Mark Davies Microfluidic device with channel plates
WO2015173658A2 (en) 2014-05-14 2015-11-19 Mark Davis Microfluidic devices that include channels that are slidable relative to each other and methods of use thereof
US10208332B2 (en) 2014-05-21 2019-02-19 Integenx Inc. Fluidic cartridge with valve mechanism
US11891650B2 (en) 2014-05-21 2024-02-06 IntegenX, Inc. Fluid cartridge with valve mechanism
US10961561B2 (en) 2014-05-21 2021-03-30 IntegenX, Inc. Fluidic cartridge with valve mechanism
US11390906B2 (en) 2014-06-11 2022-07-19 Samplix Aps Nucleotide sequence exclusion enrichment by droplet sorting (NEEDLS)
US10704080B2 (en) 2014-06-11 2020-07-07 Samplix Aps Nucleotide sequence exclusion enrichment by droplet sorting (NEEDLS)
WO2015195698A1 (en) 2014-06-16 2015-12-23 Gnubio, Inc. Size alternating injection into drops to facilitate sorting
US10760124B2 (en) 2014-06-26 2020-09-01 10X Genomics, Inc. Methods and systems for processing polynucleotides
US11629344B2 (en) 2014-06-26 2023-04-18 10X Genomics, Inc. Methods and systems for processing polynucleotides
US9951386B2 (en) 2014-06-26 2018-04-24 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10344329B2 (en) 2014-06-26 2019-07-09 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10030267B2 (en) 2014-06-26 2018-07-24 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10839939B2 (en) 2014-06-26 2020-11-17 10X Genomics, Inc. Processes and systems for nucleic acid sequence assembly
US10208343B2 (en) 2014-06-26 2019-02-19 10X Genomics, Inc. Methods and systems for processing polynucleotides
US11713457B2 (en) 2014-06-26 2023-08-01 10X Genomics, Inc. Methods and systems for processing polynucleotides
US11133084B2 (en) 2014-06-26 2021-09-28 10X Genomics, Inc. Systems and methods for nucleic acid sequence assembly
US10480028B2 (en) 2014-06-26 2019-11-19 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10457986B2 (en) 2014-06-26 2019-10-29 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10041116B2 (en) 2014-06-26 2018-08-07 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10337061B2 (en) 2014-06-26 2019-07-02 10X Genomics, Inc. Methods and systems for processing polynucleotides
US11312990B2 (en) 2014-06-27 2022-04-26 The Regents Of The University Of California PCR-activated sorting (PAS)
US10697007B2 (en) 2014-06-27 2020-06-30 The Regents Of The University Of California PCR-activated sorting (PAS)
US20170227440A1 (en) * 2014-08-11 2017-08-10 Sphere Fluidics Limited Droplet sorting
US11656167B2 (en) 2014-08-11 2023-05-23 Sphere Fluidics Limited Droplet sorting based on ionization mass spectrometry
US10545083B2 (en) * 2014-08-11 2020-01-28 Sphere Fluidics Limited Droplet sorting based on ionization mass spectrometry
US10895544B2 (en) * 2014-08-21 2021-01-19 Schlumberger Technology Corporation Measurement of liquid parameters using a microfluidic device
US20170227479A1 (en) * 2014-08-21 2017-08-10 Schlumberger Technology Corporation Measurement of liquid parameters using a microfluidic device
EP4105337A1 (en) 2014-09-09 2022-12-21 The Broad Institute, Inc. A droplet-based method and apparatus for composite single-cell nucleic acid analysis
WO2016040476A1 (en) 2014-09-09 2016-03-17 The Broad Institute, Inc. A droplet-based method and apparatus for composite single-cell nucleic acid analysis
US11597964B2 (en) 2014-09-09 2023-03-07 The Broad Institute, Inc. Droplet-based method and apparatus for composite single-cell nucleic acid analysis
US11566279B2 (en) 2014-09-09 2023-01-31 The Broad Institute, Inc. Droplet-based method and apparatus for composite single-cell nucleic acid analysis
US11408024B2 (en) 2014-09-10 2022-08-09 Molecular Loop Biosciences, Inc. Methods for selectively suppressing non-target sequences
US10429399B2 (en) 2014-09-24 2019-10-01 Good Start Genetics, Inc. Process control for increased robustness of genetic assays
US11482052B2 (en) 2014-10-22 2022-10-25 IntegenX, Inc. Systems and methods for biometric data collections
US10434507B2 (en) 2014-10-22 2019-10-08 The Regents Of The University Of California High definition microdroplet printer
US10690627B2 (en) 2014-10-22 2020-06-23 IntegenX, Inc. Systems and methods for sample preparation, processing and analysis
US11020736B2 (en) 2014-10-22 2021-06-01 The Regents Of The University Of California High definition microdroplet printer
US11739368B2 (en) 2014-10-29 2023-08-29 10X Genomics, Inc. Methods and compositions for targeted nucleic acid sequencing
US10287623B2 (en) 2014-10-29 2019-05-14 10X Genomics, Inc. Methods and compositions for targeted nucleic acid sequencing
US10000799B2 (en) 2014-11-04 2018-06-19 Boreal Genomics, Inc. Methods of sequencing with linked fragments
US11827930B2 (en) 2014-11-04 2023-11-28 Ncan Genomics, Inc. Methods of sequencing with linked fragments
US10829813B2 (en) 2014-11-04 2020-11-10 Boreal Genomics, Inc. Methods of sequencing with linked fragments
US10245587B2 (en) 2014-11-05 2019-04-02 10X Genomics, Inc. Instrument systems for integrated sample processing
US11135584B2 (en) 2014-11-05 2021-10-05 10X Genomics, Inc. Instrument systems for integrated sample processing
US9975122B2 (en) 2014-11-05 2018-05-22 10X Genomics, Inc. Instrument systems for integrated sample processing
US20170326549A1 (en) * 2014-11-26 2017-11-16 Ronald Jones Automated microscopic cell analysis
US11590496B2 (en) 2014-11-26 2023-02-28 Medica Corporation Automated microscopic cell analysis
US10625259B1 (en) 2014-11-26 2020-04-21 Medica Corporation Automated microscopic cell analysis
US11478789B2 (en) * 2014-11-26 2022-10-25 Medica Corporation Automated microscopic cell analysis
US11480778B2 (en) 2014-11-26 2022-10-25 Medica Corporation Automated microscopic cell analysis
US11536688B2 (en) 2014-12-18 2022-12-27 Life Technologies Corporation High data rate integrated circuit with transmitter configuration
US10379079B2 (en) 2014-12-18 2019-08-13 Life Technologies Corporation Methods and apparatus for measuring analytes using large scale FET arrays
US10605767B2 (en) 2014-12-18 2020-03-31 Life Technologies Corporation High data rate integrated circuit with transmitter configuration
US10767224B2 (en) 2014-12-18 2020-09-08 Life Technologies Corporation High data rate integrated circuit with power management
US10077472B2 (en) 2014-12-18 2018-09-18 Life Technologies Corporation High data rate integrated circuit with power management
EP3757211A1 (en) 2014-12-19 2020-12-30 The Broad Institute, Inc. Methods for profiling the t-cell-receptor repertoire
US10993997B2 (en) 2014-12-19 2021-05-04 The Broad Institute, Inc. Methods for profiling the t cell repertoire
WO2016100977A1 (en) 2014-12-19 2016-06-23 The Broad Institute Inc. Methods for profiling the t-cel- receptor repertoire
US10975442B2 (en) 2014-12-19 2021-04-13 Massachusetts Institute Of Technology Molecular biomarkers for cancer immunotherapy
US11680284B2 (en) 2015-01-06 2023-06-20 Moledular Loop Biosciences, Inc. Screening for structural variants
US10066259B2 (en) 2015-01-06 2018-09-04 Good Start Genetics, Inc. Screening for structural variants
US11414688B2 (en) 2015-01-12 2022-08-16 10X Genomics, Inc. Processes and systems for preparation of nucleic acid sequencing libraries and libraries prepared using same
US10221436B2 (en) 2015-01-12 2019-03-05 10X Genomics, Inc. Processes and systems for preparation of nucleic acid sequencing libraries and libraries prepared using same
US10557158B2 (en) 2015-01-12 2020-02-11 10X Genomics, Inc. Processes and systems for preparation of nucleic acid sequencing libraries and libraries prepared using same
US10650912B2 (en) 2015-01-13 2020-05-12 10X Genomics, Inc. Systems and methods for visualizing structural variation and phasing information
US11111519B2 (en) 2015-02-04 2021-09-07 The Regents Of The University Of California Sequencing of nucleic acids via barcoding in discrete entities
US11732287B2 (en) 2015-02-04 2023-08-22 The Regents Of The University Of California Sequencing of nucleic acids via barcoding in discrete entities
US10854315B2 (en) 2015-02-09 2020-12-01 10X Genomics, Inc. Systems and methods for determining structural variation and phasing using variant call data
US10697000B2 (en) 2015-02-24 2020-06-30 10X Genomics, Inc. Partition processing methods and systems
US11603554B2 (en) 2015-02-24 2023-03-14 10X Genomics, Inc. Partition processing methods and systems
US11274343B2 (en) 2015-02-24 2022-03-15 10X Genomics, Inc. Methods and compositions for targeted nucleic acid sequence coverage
WO2016138488A2 (en) 2015-02-26 2016-09-01 The Broad Institute Inc. T cell balance gene expression, compositions of matters and methods of use thereof
US11427869B2 (en) 2015-02-26 2022-08-30 The Broad Institute, Inc. T cell balance gene expression, compositions of matters and methods of use thereof
US11873483B2 (en) 2015-03-11 2024-01-16 The Broad Institute, Inc. Proteomic analysis with nucleic acid identifiers
US20180094255A1 (en) * 2015-03-20 2018-04-05 Novozymes A/S Droplet-Based Selection By Injection
US10883102B2 (en) * 2015-03-20 2021-01-05 Novozymes A/S Droplet-based selection by injection
US10835585B2 (en) 2015-05-20 2020-11-17 The Broad Institute, Inc. Shared neoantigens
US10632479B2 (en) 2015-05-22 2020-04-28 The Hong Kong University Of Science And Technology Droplet generator based on high aspect ratio induced droplet self-breakup
US11643669B2 (en) 2015-06-17 2023-05-09 Massachusetts Institute Of Technology CRISPR mediated recording of cellular events
WO2016205728A1 (en) 2015-06-17 2016-12-22 Massachusetts Institute Of Technology Crispr mediated recording of cellular events
EP3587629A1 (en) 2015-06-25 2020-01-01 Hooke Bio Limited Method for generating combinatiorial library
WO2016207721A1 (en) 2015-06-25 2016-12-29 University Of Limerick Mechanical device for generating combinatorial library
US11618030B2 (en) 2015-06-29 2023-04-04 Arizona Board Of Regents On Behalf Of Arizona State University Systems and methods for continuous flow digital droplet polymerase chain reaction bioanalysis
US11123740B2 (en) 2015-06-29 2021-09-21 Arizona Board Of Regents On Behalf Of Arizona State University Systems and methods for continuous flow digital droplet polymerase chain reaction bioanalysis
US10794925B2 (en) 2015-07-07 2020-10-06 University Of Washington Systems, methods, and devices for self-digitization of samples
US11408903B2 (en) 2015-07-07 2022-08-09 University Of Washington Systems, methods, and devices for self-digitization of samples
US10647981B1 (en) 2015-09-08 2020-05-12 Bio-Rad Laboratories, Inc. Nucleic acid library generation methods and compositions
US11123297B2 (en) 2015-10-13 2021-09-21 President And Fellows Of Harvard College Systems and methods for making and using gel microspheres
WO2017075295A1 (en) 2015-10-27 2017-05-04 Berkeley Lights, Inc. Microfluidic electrowetting device apparatus having a covalently bound hydrophobic surface
EP3862088A1 (en) 2015-10-27 2021-08-11 Berkeley Lights, Inc. Method of manufcturing microfluidic electrowetting device having a covalently bound hydrophobic surface
WO2017075294A1 (en) 2015-10-28 2017-05-04 The Board Institute Inc. Assays for massively combinatorial perturbation profiling and cellular circuit reconstruction
WO2017075549A1 (en) 2015-10-28 2017-05-04 The Broad Institute, Inc. High-throughput dynamic reagent delivery system
US11904310B2 (en) 2015-10-28 2024-02-20 The Broad Institute, Inc. High-throughput dynamic reagent delivery system
US11092607B2 (en) 2015-10-28 2021-08-17 The Board Institute, Inc. Multiplex analysis of single cell constituents
US11214797B2 (en) 2015-10-28 2022-01-04 The Broad Institute, Inc. Assays for massively combinatorial perturbation profiling and cellular circuit reconstruction
US20180321123A1 (en) * 2015-11-06 2018-11-08 Regents Of The University Of Michigan Droplet-based microfluidic rheometer system
CN108463708A (en) * 2015-11-06 2018-08-28 密歇根大学董事会 Microfluid rheometer system based on droplet
US11879820B2 (en) 2015-11-06 2024-01-23 Regents Of The University Of Michigan Droplet-based microfluidic rheometer system
US10845284B2 (en) 2015-11-06 2020-11-24 Regents Of The University Of Michigan Droplet-based microfluidic rheometer system
WO2017079507A1 (en) * 2015-11-06 2017-05-11 Regents Of The University Of Michigan Droplet-based microfluidic rheometer system
US10774370B2 (en) 2015-12-04 2020-09-15 10X Genomics, Inc. Methods and compositions for nucleic acid analysis
US11873528B2 (en) 2015-12-04 2024-01-16 10X Genomics, Inc. Methods and compositions for nucleic acid analysis
US11624085B2 (en) 2015-12-04 2023-04-11 10X Genomics, Inc. Methods and compositions for nucleic acid analysis
US11473125B2 (en) 2015-12-04 2022-10-18 10X Genomics, Inc. Methods and compositions for nucleic acid analysis
US20170167981A1 (en) * 2015-12-11 2017-06-15 International Business Machines Corporation Smartphone compatible on-chip biodetection using integrated optical component and microfluidic channel with nanopillar array
US11009464B2 (en) * 2015-12-11 2021-05-18 International Business Machines Corporation Smartphone compatible on-chip biodetection using integrated optical component and microfluidic channel with nanopillar array
US10766033B2 (en) 2015-12-30 2020-09-08 Berkeley Lights, Inc. Droplet generation in a microfluidic device having an optoelectrowetting configuration
WO2017124101A2 (en) 2016-01-15 2017-07-20 The Broad Institute Inc. Semi-permeable arrays for analyzing biological systems and methods of using same
WO2017136751A1 (en) 2016-02-05 2017-08-10 The Broad Institute Inc. Multi-stage, multiplexed target isolation and processing from heterogeneous populations
US11081208B2 (en) 2016-02-11 2021-08-03 10X Genomics, Inc. Systems, methods, and media for de novo assembly of whole genome sequence data
WO2017147196A1 (en) 2016-02-22 2017-08-31 Massachusetts Institute Of Technology Methods for identifying and modulating immune phenotypes
US11427861B2 (en) 2016-03-17 2022-08-30 Massachusetts Institute Of Technology Methods for identifying and modulating co-occurant cellular phenotypes
WO2017161325A1 (en) 2016-03-17 2017-09-21 Massachusetts Institute Of Technology Methods for identifying and modulating co-occurant cellular phenotypes
WO2017168332A1 (en) 2016-03-28 2017-10-05 Boreal Genomics, Inc. Linked duplex target capture
US10961573B2 (en) 2016-03-28 2021-03-30 Boreal Genomics, Inc. Linked duplex target capture
US10801059B2 (en) 2016-03-28 2020-10-13 Boreal Genomics, Inc. Droplet-based linked-fragment sequencing
EP4282974A2 (en) 2016-03-28 2023-11-29 Ncan Genomics, Inc. Linked duplex target capture
US11905556B2 (en) 2016-03-28 2024-02-20 Ncan Genomics, Inc. Linked target capture
US11021742B2 (en) 2016-03-28 2021-06-01 Boreal Genomics, Inc. Linked-fragment sequencing
US10961568B2 (en) 2016-03-28 2021-03-30 Boreal Genomics, Inc. Linked target capture
US11084036B2 (en) 2016-05-13 2021-08-10 10X Genomics, Inc. Microfluidic systems and methods of use
US11607658B2 (en) 2016-07-08 2023-03-21 President And Fellows Of Harvard College Formation of colloids or gels within droplets
WO2018013426A2 (en) 2016-07-08 2018-01-18 California Institute Of Technology Methods and devices for performing flow-through capture of low-concentration analytes
WO2018009766A1 (en) * 2016-07-08 2018-01-11 President And Fellows Of Harvard College Formation of colloids or gels within droplets
US11142791B2 (en) 2016-08-10 2021-10-12 The Regents Of The University Of California Combined multiple-displacement amplification and PCR in an emulsion microdroplet
WO2018051242A1 (en) * 2016-09-14 2018-03-22 Ecole Polytechnique Federale De Lausanne (Epfl) Device for high throughput single-cell studies
US11872559B2 (en) 2016-09-14 2024-01-16 Ecole Polytechnique Federale De Lausanne (Epfl) Device for high throughput single-cell studies
CN110740813A (en) * 2016-11-28 2020-01-31 亚利桑那州立大学董事会 Systems and methods involving continuous flow droplet reactions
US11413616B2 (en) * 2016-11-28 2022-08-16 Arizona Board Of Regents On Behalf Of Arizona State University Systems and methods related to continuous flow droplet reaction
US11607689B2 (en) 2016-11-28 2023-03-21 Arizona Board Of Regents On Behalf Of Arizona State University Systems and methods related to continuous flow droplet reaction
CN110740813B (en) * 2016-11-28 2022-06-03 亚利桑那州立大学董事会 Systems and methods involving continuous flow droplet reactions
US11268137B2 (en) 2016-12-09 2022-03-08 Boreal Genomics, Inc. Linked ligation
US11879151B2 (en) 2016-12-09 2024-01-23 Ncan Genomics, Inc. Linked ligation
US11124830B2 (en) 2016-12-21 2021-09-21 The Regents Of The University Of California Single cell genomic sequencing using hydrogel based droplets
US11180805B2 (en) 2016-12-22 2021-11-23 10X Genomics, Inc Methods and systems for processing polynucleotides
US10815525B2 (en) 2016-12-22 2020-10-27 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10793905B2 (en) 2016-12-22 2020-10-06 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10323278B2 (en) 2016-12-22 2019-06-18 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10858702B2 (en) 2016-12-22 2020-12-08 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10011872B1 (en) 2016-12-22 2018-07-03 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10480029B2 (en) 2016-12-22 2019-11-19 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10550429B2 (en) 2016-12-22 2020-02-04 10X Genomics, Inc. Methods and systems for processing polynucleotides
EP3351302A1 (en) 2017-01-18 2018-07-25 Biomillenia SAS Microfluidic system and method with tightly controlled incubation time and conditions
WO2018134311A1 (en) 2017-01-18 2018-07-26 Biomillenia Sas Microfluidic system and method with tightly controlled incubation time and conditions
US11549149B2 (en) 2017-01-24 2023-01-10 The Broad Institute, Inc. Compositions and methods for detecting a mutant variant of a polynucleotide
US10428326B2 (en) 2017-01-30 2019-10-01 10X Genomics, Inc. Methods and systems for droplet-based single cell barcoding
US11193122B2 (en) 2017-01-30 2021-12-07 10X Genomics, Inc. Methods and systems for droplet-based single cell barcoding
WO2018170515A1 (en) 2017-03-17 2018-09-20 The Broad Institute, Inc. Methods for identifying and modulating co-occurant cellular phenotypes
US11492579B2 (en) * 2017-04-14 2022-11-08 Advanced Solutions Life Sciences, Llc Vascularized in vitro arrays of living cells
WO2018195019A1 (en) 2017-04-18 2018-10-25 The Broad Institute Inc. Compositions for detecting secretion and methods of use
WO2018200872A1 (en) * 2017-04-26 2018-11-01 Berkeley Lights, Inc. Biological process systems and methods using microfluidic apparatus having an optimized electrowetting surface
US11072816B2 (en) 2017-05-03 2021-07-27 The Broad Institute, Inc. Single-cell proteomic assay using aptamers
US11898206B2 (en) 2017-05-19 2024-02-13 10X Genomics, Inc. Systems and methods for clonotype screening
US10400235B2 (en) 2017-05-26 2019-09-03 10X Genomics, Inc. Single cell analysis of transposase accessible chromatin
US11198866B2 (en) 2017-05-26 2021-12-14 10X Genomics, Inc. Single cell analysis of transposase accessible chromatin
US11773389B2 (en) 2017-05-26 2023-10-03 10X Genomics, Inc. Single cell analysis of transposase accessible chromatin
US10927370B2 (en) 2017-05-26 2021-02-23 10X Genomics, Inc. Single cell analysis of transposase accessible chromatin
US11155810B2 (en) 2017-05-26 2021-10-26 10X Genomics, Inc. Single cell analysis of transposase accessible chromatin
US10844372B2 (en) 2017-05-26 2020-11-24 10X Genomics, Inc. Single cell analysis of transposase accessible chromatin
US11499183B2 (en) 2017-06-28 2022-11-15 Bio-Rad Laboratories, Inc. System and method for droplet detection
US10821440B2 (en) 2017-08-29 2020-11-03 Bio-Rad Laboratories, Inc. System and method for isolating and analyzing cells
US11865542B2 (en) 2017-08-29 2024-01-09 Bio-Rad Laboratories, Inc. System and method for isolating and analyzing cells
US11504714B2 (en) 2017-08-29 2022-11-22 Bio-Rad Laboratories, Inc. System and method for isolating and analyzing cells
US11358146B2 (en) 2017-08-29 2022-06-14 Bio-Rad Laboratories, Inc. System and method for isolating and analyzing cells
US11781129B2 (en) 2017-10-18 2023-10-10 Mission Bio, Inc. Method, systems and apparatus for single cell analysis
US10501739B2 (en) 2017-10-18 2019-12-10 Mission Bio, Inc. Method, systems and apparatus for single cell analysis
US11732257B2 (en) 2017-10-23 2023-08-22 Massachusetts Institute Of Technology Single cell sequencing libraries of genomic transcript regions of interest in proximity to barcodes, and genotyping of said libraries
US11045805B2 (en) 2017-11-01 2021-06-29 Bio-Rad Laboratories, Inc. Microfluidic system and method for arranging objects
US11857968B2 (en) 2017-11-01 2024-01-02 Bio-Rad Laboratories, Inc. Microfluidic system and method for arranging objects
US10745742B2 (en) 2017-11-15 2020-08-18 10X Genomics, Inc. Functionalized gel beads
US10876147B2 (en) 2017-11-15 2020-12-29 10X Genomics, Inc. Functionalized gel beads
US11047845B1 (en) 2017-11-15 2021-06-29 Medica Corporation Control material and methods for cell analyzers
US11884962B2 (en) 2017-11-15 2024-01-30 10X Genomics, Inc. Functionalized gel beads
US10829815B2 (en) 2017-11-17 2020-11-10 10X Genomics, Inc. Methods and systems for associating physical and genetic properties of biological particles
US11332736B2 (en) 2017-12-07 2022-05-17 The Broad Institute, Inc. Methods and compositions for multiplexing single cell and single nuclei sequencing
US11841371B2 (en) 2018-03-13 2023-12-12 The Broad Institute, Inc. Proteomics and spatial patterning using antenna networks
US11859239B2 (en) 2018-03-19 2024-01-02 Ricoh Company, Ltd. Nucleic acid sample-contained container, method and apparatus for producing nucleic acid sample-contained container, non-transitory recording medium storing program for producing nucleic acid sample-contained container, and nucleic acid sample
US11155881B2 (en) 2018-04-06 2021-10-26 10X Genomics, Inc. Systems and methods for quality control in single cell processing
US20210268497A1 (en) * 2018-07-29 2021-09-02 Koc Universitesi Microfluidic thromboelastometry instrument
WO2020077236A1 (en) 2018-10-12 2020-04-16 The Broad Institute, Inc. Method for extracting nuclei or whole cells from formalin-fixed paraffin-embedded tissues
WO2020102610A1 (en) 2018-11-14 2020-05-22 The Broad Institute, Inc. Crispr system based droplet diagnostic systems and methods
WO2020102608A2 (en) 2018-11-14 2020-05-22 President And Fellows Of Harvard College Multiplexing highly evolving viral variants with sherlock
WO2020124050A1 (en) 2018-12-13 2020-06-18 The Broad Institute, Inc. Tiled assays using crispr-cas based detection
WO2020120442A2 (en) 2018-12-13 2020-06-18 Dna Script Direct oligonucleotide synthesis on cells and biomolecules
US11268091B2 (en) 2018-12-13 2022-03-08 Dna Script Sas Direct oligonucleotide synthesis on cells and biomolecules
WO2020131586A2 (en) 2018-12-17 2020-06-25 The Broad Institute, Inc. Methods for identifying neoantigens
US11473136B2 (en) 2019-01-03 2022-10-18 Ncan Genomics, Inc. Linked target capture
US11896974B2 (en) 2019-01-17 2024-02-13 Regents Of The University Of Michigan Microfluidic device for size and deformability measurements and applications thereof
US11389799B2 (en) 2019-01-17 2022-07-19 The Regents Of The University Of Michigan Microfluidic device for size and deformability measurements and applications thereof
EP3698871A1 (en) 2019-02-19 2020-08-26 Gottfried Wilhelm Leibniz Universität Hannover Laser based sorting of droplets in microfluidic streams
US11814671B2 (en) 2019-04-16 2023-11-14 Bio-Rad Laboratories, Inc. System and method for leakage control in a particle capture system
US10947581B2 (en) 2019-04-16 2021-03-16 Bio-Rad Laboratories, Inc. System and method for leakage control in a particle capture system
US11866766B2 (en) 2019-04-16 2024-01-09 Bio-Rad Laboratories, Inc. System and method for leakage control in a particle capture system
WO2020223578A1 (en) * 2019-04-30 2020-11-05 Northeastern University Microfluidic chip for single cell pairing
US11273439B2 (en) 2019-05-07 2022-03-15 Bio-Rad Laboratories, Inc. System and method for target material retrieval from microwells
US11833507B2 (en) 2019-05-07 2023-12-05 Bio-Rad Laboratories, Inc. System and method for target material retrieval from microwells
US10900032B2 (en) 2019-05-07 2021-01-26 Bio-Rad Laboratories, Inc. System and method for automated single cell processing
US11578322B2 (en) 2019-05-07 2023-02-14 Bio-Rad Laboratories, Inc. System and method for automated single cell processing
US11365441B2 (en) 2019-05-22 2022-06-21 Mission Bio, Inc. Method and apparatus for simultaneous targeted sequencing of DNA, RNA and protein
US11724256B2 (en) 2019-06-14 2023-08-15 Bio-Rad Laboratories, Inc. System and method for automated single cell processing and analyses
US11667954B2 (en) 2019-07-01 2023-06-06 Mission Bio, Inc. Method and apparatus to normalize quantitative readouts in single-cell experiments
GB201914537D0 (en) 2019-10-08 2019-11-20 Univ Southampton Transcript analysis
US11504719B2 (en) 2020-03-12 2022-11-22 Bio-Rad Laboratories, Inc. System and method for receiving and delivering a fluid for sample processing
WO2022219121A1 (en) * 2021-04-15 2022-10-20 Screensys Gmbh Method for developing and/or reprogramming plant cellular objects
WO2022232050A1 (en) 2021-04-26 2022-11-03 The Broad Institute, Inc. Compositions and methods for characterizing polynucleotide sequence alterations
US11927519B2 (en) 2023-04-26 2024-03-12 Slingshot Biosciences, Inc. Synthetic human cell mimic hydrogel particle for cytometric or coulter device

Also Published As

Publication number Publication date
US20180280897A1 (en) 2018-10-04
US9981230B2 (en) 2018-05-29
US20200360876A1 (en) 2020-11-19
EP3335782A1 (en) 2018-06-20
JP6023252B2 (en) 2016-11-09
EP2530167A1 (en) 2012-12-05
EP2047910A3 (en) 2009-11-11
JP2013015536A (en) 2013-01-24
EP4190448A3 (en) 2023-09-20
ATE540750T1 (en) 2012-01-15
EP2530168B1 (en) 2015-09-16
US20130210639A1 (en) 2013-08-15
EP2481815A1 (en) 2012-08-01
US20130090248A1 (en) 2013-04-11
US20180178174A1 (en) 2018-06-28
JP2015142586A (en) 2015-08-06
EP2530168A1 (en) 2012-12-05
JP2010506136A (en) 2010-02-25
US20080003142A1 (en) 2008-01-03
US20180272295A1 (en) 2018-09-27
US9273308B2 (en) 2016-03-01
WO2008063227A3 (en) 2008-11-06
US11351510B2 (en) 2022-06-07
EP2021113A2 (en) 2009-02-11
EP2481815B1 (en) 2016-01-27
US10639597B2 (en) 2020-05-05
EP3031918A1 (en) 2016-06-15
JP6316369B2 (en) 2018-04-25
WO2008063227A2 (en) 2008-05-29
US20180272296A1 (en) 2018-09-27
WO2007133710A2 (en) 2007-11-22
EP3782722A1 (en) 2021-02-24
EP4190448A2 (en) 2023-06-07
JP2017006145A (en) 2017-01-12
EP3782722B1 (en) 2022-07-06
US20130183659A1 (en) 2013-07-18
EP2047910A2 (en) 2009-04-15
EP2047910B1 (en) 2012-01-11
EP3335782B1 (en) 2020-09-09
US20170304785A1 (en) 2017-10-26
WO2007133710A3 (en) 2008-02-21
US10625220B2 (en) 2020-04-21
EP3031918B1 (en) 2018-03-14
JP2018113981A (en) 2018-07-26

Similar Documents

Publication Publication Date Title
US20200360876A1 (en) Microfluidic devices
US20210262020A1 (en) Systems and methods for handling microfluidic droplets
WO2014165559A2 (en) Systems and methods for handling microfluidic droplets

Legal Events

Date Code Title Description
AS Assignment

Owner name: RAINDANCE TECHNOLOGIES, INC., CONNECTICUT

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:LINK, DARREN R.;WEINER, MICHAEL;MARRAN, DAVID;AND OTHERS;REEL/FRAME:019623/0935;SIGNING DATES FROM 20070706 TO 20070716

AS Assignment

Owner name: CAPITAL ROYALTY PARTNERS II - PARALLEL FUND "A" L.P., TEXAS

Free format text: SHORT-FORM PATENT SECURITY AGREEMENT;ASSIGNOR:RAINDANCE TECHNOLOGIES, INC.;REEL/FRAME:031267/0376

Effective date: 20130917

Owner name: PARALLEL INVESTMENT OPPORTUNITIES PARTNERS II L.P., TEXAS

Free format text: SHORT-FORM PATENT SECURITY AGREEMENT;ASSIGNOR:RAINDANCE TECHNOLOGIES, INC.;REEL/FRAME:031267/0376

Effective date: 20130917

Owner name: PARALLEL INVESTMENT OPPORTUNITIES PARTNERS II L.P.

Free format text: SHORT-FORM PATENT SECURITY AGREEMENT;ASSIGNOR:RAINDANCE TECHNOLOGIES, INC.;REEL/FRAME:031267/0376

Effective date: 20130917

Owner name: CAPITAL ROYALTY PARTNERS II - PARALLEL FUND "A" L.

Free format text: SHORT-FORM PATENT SECURITY AGREEMENT;ASSIGNOR:RAINDANCE TECHNOLOGIES, INC.;REEL/FRAME:031267/0376

Effective date: 20130917

Owner name: CAPITAL ROYALTY PARTNERS II L.P., TEXAS

Free format text: SHORT-FORM PATENT SECURITY AGREEMENT;ASSIGNOR:RAINDANCE TECHNOLOGIES, INC.;REEL/FRAME:031267/0376

Effective date: 20130917

STCB Information on status: application discontinuation

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

AS Assignment

Owner name: RAINDANCE TECHNOLOGIES, INC., MASSACHUSETTS

Free format text: RELEASE BY SECURED PARTY;ASSIGNORS:CAPITAL ROYALTY PARTNERS II L.P.;CAPITAL ROYALTY PARTNERS II (CAYMAN) L.P.;PARALLEL INVESTMENT OPPORTUNITIES PARTNERS II L.P.;AND OTHERS;REEL/FRAME:041290/0585

Effective date: 20170216

AS Assignment

Owner name: BIO-RAD LABORATORIES, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:RAINDANCE TECHNOLOGIES, INC.;REEL/FRAME:049109/0498

Effective date: 20190503