US20040028567A1 - High throughput screening assay systems in microscale fluidic devices - Google Patents

High throughput screening assay systems in microscale fluidic devices Download PDF

Info

Publication number
US20040028567A1
US20040028567A1 US10/637,730 US63773003A US2004028567A1 US 20040028567 A1 US20040028567 A1 US 20040028567A1 US 63773003 A US63773003 A US 63773003A US 2004028567 A1 US2004028567 A1 US 2004028567A1
Authority
US
United States
Prior art keywords
channel
reservoir
fluid
channels
disposed
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US10/637,730
Inventor
J. Parce
Anne Kopf-Sill
Luc Bousse
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Caliper Life Sciences Inc
Original Assignee
Caliper Technologies Corp
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US08/671,987 external-priority patent/US5942443A/en
Application filed by Caliper Technologies Corp filed Critical Caliper Technologies Corp
Priority to US10/637,730 priority Critical patent/US20040028567A1/en
Assigned to CALIPER LIFE SCIENCES, INC. reassignment CALIPER LIFE SCIENCES, INC. CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: CALIPER TECHNOLOGIES CORP.
Publication of US20040028567A1 publication Critical patent/US20040028567A1/en
Priority to US11/050,900 priority patent/US20060000722A1/en
Priority to US11/176,805 priority patent/US20050241941A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/502Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing non-proliferative effects
    • 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
    • B01L3/50273Containers for the purpose of retaining a material to be analysed, e.g. test tubes with fluid transport, e.g. in multi-compartment structures by integrated microfluidic structures, i.e. dimensions of channels and chambers are such that surface tension forces are important, e.g. lab-on-a-chip characterised by the means or forces applied to move the fluids
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01LCHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
    • B01L3/00Containers or dishes for laboratory use, e.g. laboratory glassware; Droppers
    • B01L3/50Containers for the purpose of retaining a material to be analysed, e.g. test tubes
    • B01L3/502Containers for the purpose of retaining a material to be analysed, e.g. test tubes with fluid transport, e.g. in multi-compartment structures
    • B01L3/5027Containers for the purpose of retaining a material to be analysed, e.g. test tubes with fluid transport, e.g. in multi-compartment structures by integrated microfluidic structures, i.e. dimensions of channels and chambers are such that surface tension forces are important, e.g. lab-on-a-chip
    • B01L3/502753Containers 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 bulk separation arrangements on lab-on-a-chip devices, e.g. for filtration or centrifugation
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01LCHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
    • B01L3/00Containers or dishes for laboratory use, e.g. laboratory glassware; Droppers
    • B01L3/50Containers for the purpose of retaining a material to be analysed, e.g. test tubes
    • B01L3/502Containers for the purpose of retaining a material to be analysed, e.g. test tubes with fluid transport, e.g. in multi-compartment structures
    • B01L3/5027Containers for the purpose of retaining a material to be analysed, e.g. test tubes with fluid transport, e.g. in multi-compartment structures by integrated microfluidic structures, i.e. dimensions of channels and chambers are such that surface tension forces are important, e.g. lab-on-a-chip
    • B01L3/502761Containers for the purpose of retaining a material to be analysed, e.g. test tubes with fluid transport, e.g. in multi-compartment structures by integrated microfluidic structures, i.e. dimensions of channels and chambers are such that surface tension forces are important, e.g. lab-on-a-chip specially adapted for handling suspended solids or molecules independently from the bulk fluid flow, e.g. for trapping or sorting beads, for physically stretching molecules
    • 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
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N27/00Investigating or analysing materials by the use of electric, electrochemical, or magnetic means
    • G01N27/26Investigating or analysing materials by the use of electric, electrochemical, or magnetic means by investigating electrochemical variables; by using electrolysis or electrophoresis
    • G01N27/416Systems
    • G01N27/447Systems using electrophoresis
    • G01N27/44756Apparatus specially adapted therefor
    • G01N27/44791Microapparatus
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5011Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing antineoplastic activity
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5044Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics involving specific cell types
    • G01N33/5064Endothelial cells
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5091Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing the pathological state of an organism
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5097Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving plant cells
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/5302Apparatus specially adapted for immunological test procedures
    • G01N33/5304Reaction vessels, e.g. agglutination plates
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57407Specifically defined cancers
    • G01N33/57415Specifically defined cancers of breast
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00781Aspects relating to microreactors
    • B01J2219/00819Materials of construction
    • B01J2219/00824Ceramic
    • B01J2219/00828Silicon wafers or plates
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00781Aspects relating to microreactors
    • B01J2219/00819Materials of construction
    • B01J2219/00831Glass
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00781Aspects relating to microreactors
    • B01J2219/00819Materials of construction
    • B01J2219/00833Plastic
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00781Aspects relating to microreactors
    • B01J2219/00851Additional features
    • B01J2219/00853Employing electrode arrangements
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00781Aspects relating to microreactors
    • B01J2219/0095Control aspects
    • B01J2219/00952Sensing operations
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00781Aspects relating to microreactors
    • B01J2219/0095Control aspects
    • B01J2219/00952Sensing operations
    • B01J2219/00954Measured properties
    • B01J2219/00961Temperature
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00781Aspects relating to microreactors
    • B01J2219/0095Control aspects
    • B01J2219/00952Sensing operations
    • B01J2219/00968Type of sensors
    • B01J2219/0097Optical sensors
    • 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
    • B01L2200/0605Metering of fluids
    • 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
    • B01L2200/0647Handling flowable solids, e.g. microscopic beads, cells, particles
    • B01L2200/0668Trapping microscopic beads
    • 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
    • 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
    • B01L2300/08Geometry, shape and general structure
    • B01L2300/0861Configuration of multiple channels and/or chambers in a single devices
    • B01L2300/0877Flow chambers
    • 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
    • B01L2300/0887Laminated structure
    • 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
    • B01L2400/0418Moving fluids with specific forces or mechanical means specific forces electrical forces, e.g. electrokinetic electro-osmotic flow [EOF]
    • 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
    • B01L3/00Containers or dishes for laboratory use, e.g. laboratory glassware; Droppers
    • B01L3/50Containers for the purpose of retaining a material to be analysed, e.g. test tubes
    • B01L3/502Containers for the purpose of retaining a material to be analysed, e.g. test tubes with fluid transport, e.g. in multi-compartment structures
    • B01L3/5025Containers for the purpose of retaining a material to be analysed, e.g. test tubes with fluid transport, e.g. in multi-compartment structures for parallel transport of multiple samples
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01LCHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
    • B01L3/00Containers or dishes for laboratory use, e.g. laboratory glassware; Droppers
    • 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
    • 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
    • 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
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S366/00Agitating
    • Y10S366/02Micromixers: segmented laminar flow with boundary mixing orthogonal to the direction of fluid propagation with or without geometry influences from the pathway
    • 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
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S435/00Chemistry: molecular biology and microbiology
    • Y10S435/81Packaged device or kit
    • 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
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S435/00Chemistry: molecular biology and microbiology
    • Y10S435/817Enzyme or microbe electrode
    • 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
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S436/00Chemistry: analytical and immunological testing
    • Y10S436/804Radioisotope, e.g. radioimmunoassay
    • 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
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S436/00Chemistry: analytical and immunological testing
    • Y10S436/805Optical property
    • 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
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S436/00Chemistry: analytical and immunological testing
    • Y10S436/806Electrical property or magnetic property
    • 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
    • Y10T436/00Chemistry: analytical and immunological testing
    • Y10T436/11Automated chemical analysis
    • 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
    • Y10T436/00Chemistry: analytical and immunological testing
    • Y10T436/11Automated chemical analysis
    • Y10T436/110833Utilizing a moving indicator strip or tape
    • 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
    • Y10T436/00Chemistry: analytical and immunological testing
    • Y10T436/11Automated chemical analysis
    • Y10T436/113332Automated chemical analysis with conveyance of sample along a test line in a container or rack
    • 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
    • Y10T436/00Chemistry: analytical and immunological testing
    • Y10T436/11Automated chemical analysis
    • Y10T436/117497Automated chemical analysis with a continuously flowing sample or carrier stream
    • 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
    • Y10T436/00Chemistry: analytical and immunological testing
    • Y10T436/25Chemistry: analytical and immunological testing including sample preparation

Definitions

  • the apparatus comprise a substrate with one or more intersecting channels and an electroosmotic fluid movement component, or other component for moving fluid in the channels on the substrate.
  • the present invention meets these and a variety of other needs.
  • the present invention provides novel methods and apparatuses for performing screening assays which address and provide meaningful solutions to these problems.
  • the present invention provides methods of screening a plurality of test compounds for an effect on a biochemical system. These methods typically utilize microfabricated substrates which have at least a first surface, and at least two intersecting channels fabricated into that first surface. At least one of the intersecting channels will have at least one cross-sectional dimension in a range from 0.1 to 500 ⁇ m.
  • the methods involve flowing a first component of a biochemical system in a first of the at least two intersecting channels. At least a first test compound is flowed from a second channel into the first channel whereby the test compound contacts the first component of the biochemical system. An effect of the test compound on the biochemical system is then detected.
  • the method comprises continuously flowing the first component of a biochemical system in the first channel of the at least two intersecting channels. Different test compounds are periodically introduced into the first channel from a second channel. The effect, if any, of the test compound on the biochemical system is then detected.
  • the methods utilize a substrate having at least a first surface with a plurality of reaction channels fabricated into the first surface.
  • Each of the plurality of reaction channels is fluidly connected to at least two transverse channels also fabricated in the surface.
  • the at least first component of a biochemical system is introduced into the plurality of reaction channels, and a plurality of different test compounds is flowed through at least one of the at least two transverse channels. Further, each of the plurality of test compounds is introduced into the transverse channel in a discrete volume.
  • Each of the plurality of different test compounds is directed into a separate reaction channel and the effect of each of the test compounds on the biochemical system is then detected.
  • the present invention also provides apparatuses for practicing the above methods.
  • the present invention provides an apparatus for screening test compounds for an effect on a biochemical system.
  • the device comprises a substrate having at least one surface with at least two intersecting channels fabricated into the surface.
  • the at least two intersecting channels have at least one cross-sectional dimension in the range from about 0.1 to about 500 ⁇ m.
  • the device also comprises a source of different test compounds fluidly connected to a first of the at least two intersecting channels, and a source of at least one component of the biochemical system fluidly connected to a second of the at least two intersecting channels.
  • fluid direction systems for flowing the at least one component within the intersecting channels, and for introducing the different test compounds from the first to the second of the intersecting channels.
  • the apparatus also optionally comprises a detection zone in the second channel for detecting an effect of said test compound on said biochemical system.
  • the apparatus of the invention includes a fluid direction system which comprises at least three electrodes, each electrode being in electrical contact with the at least two intersecting channels on a different side of an intersection formed by the at least two intersecting channels.
  • the fluid direction system also includes a control system for concomitantly applying a variable voltage at each of the electrodes, whereby movement of the test compounds or the at least first component in the at least two intersecting channels are controlled.
  • the present invention provides an apparatus for detecting an effect of a test compound on a biochemical system, comprising a substrate having at least one surface with a plurality of reaction channels fabricated into the surface.
  • the apparatus also has at least two transverse channels fabricated into the surface, wherein each of the plurality of reaction channels is fluidly connected to a first of the at least two transverse channels at a first point in each of the reaction channels, and fluidly connected to a second transverse channel at a second point in each of the reaction channels.
  • the apparatus further includes a source of at least one component of the biochemical system fluidly connected to each of the reaction channels, a source of test compounds fluidly connected to the first of the transverse channels, and a fluid direction system for controlling movement of the test compound and the first component within the transverse channels and the plurality of reaction channels.
  • the apparatuses also optionally include a detection zone in the second transverse channel for detecting an effect of the test compound on the biochemical system.
  • FIG. 1 is a schematic illustration of one embodiment of a microlaboratory screening assay system of the present invention which can be used in running a continuous flow assay system.
  • FIGS. 2A and 2B show a schematic illustration of the apparatus shown in FIG. 1, operating in alternate assay systems.
  • FIG. 2A shows a system used for screening effectors of an enzyme-substrate interaction.
  • FIG. 2B illustrates the use of the apparatus in screening effectors of receptor-ligand interactions.
  • FIG. 3 is a schematic illustration of a “serial input parallel reaction” microlaboratory assay system in which compounds to be screened are serially introduced into the device but then screened in a parallel orientation within the device.
  • FIGS. 4 A- 4 F show a schematic illustration of the operation of the device shown in FIG. 3, in screening a plurality of bead based test compounds.
  • FIG. 5 shows a schematic illustration of a continuous flow assay device incorporating a sample shunt for performing prolonged incubation followed by a separation step.
  • FIG. 6A shows a schematic illustration of a serial input parallel reaction device for use with fluid based test compounds.
  • FIGS. 6B and 6C show a schematic illustration of fluid flow patterns within the device shown in FIG. 6A.
  • FIG. 7 shows a schematic illustration of one embodiment of an overall assay systems which employs multiple microlaboratory devices labeled as “LabChipsTM” for screening test compounds.
  • FIG. 8 is a schematic illustration of a chip layout used for a continuous-flow assay screening system.
  • FIGS. 9A and 9B shows fluorescence data from a continuous flow assay screen.
  • FIG. 9A shows fluorescence data from a test screen which periodically introduced a known inhibitor (IPTG) into a ⁇ -galactosidase assay system in a chip format.
  • FIG. 9B shows a superposition of two data segments from FIG. 9A, directly comparing the inhibitor data with control (buffer) data.
  • FIG. 10 illustrates the operating parameters of a fluid flow system on a small chip device for performing enzyme inhibitor screening.
  • FIG. 11 shows a schematic illustration of timing for sample/spacer loading in a microfluidic device channel.
  • FIG. 12 panels A-G schematically illustrate electrodes used in apparatuses of the invention.
  • the present invention provides novel microlaboratory systems and methods that are useful for performing high-throughput screening assays.
  • the present invention provides microfluidic devices and methods of using such devices in screening large numbers of different compounds for their effects on a variety of chemical, and preferably, biochemical systems.
  • biochemical system generally refers to a chemical interaction that involves molecules of the type generally found within living organisms. Such interactions include the full range of catabolic and anabolic reactions which occur in living systems including enzymatic, binding, signaling and other reactions. Further, biochemical systems, as defined herein, also include model systems which are mimetic of a particular biochemical interaction. Examples of biochemical systems of particular interest in practicing the present invention include, e.g., receptor-ligand interactions, enzyme-substrate interactions, cellular signaling pathways, transport reactions involving model barrier systems (e.g., cells or membrane fractions) for bioavailability screening, and a variety of other general systems.
  • model barrier systems e.g., cells or membrane fractions
  • Cellular or organismal viability or activity may also be screened using the methods and apparatuses of the present invention, e.g., in toxicology studies.
  • Biological materials which are assayed include, but are not limited to, cells, cellular fractions (membranes, cytosol preparations, etc.), agonists and antagonists of cell membrane receptors (e.g., cell receptor-ligand interactions such as e.g., transferring, c-kit, viral receptor ligands (e.g., CD4-HIV), cytokine receptors, chemokine receptors, interleukin receptors, immunoglobulin receptors and antibodies, the cadherein family, the integrin family, the selectin family, and the like; see, e.g., Pigott and Power (1993) The Adhesion Molecule FactsBook Academic Press New York and Hulme (ed) Receptor Ligand Interactions A Practical Approach Rickwood and Hames (series editors) IRL Press at Oxford Press NY
  • peptides retro-inverso peptides, polymers of ⁇ -, ⁇ -, or ⁇ -amino acids (D- or L-), enzymes, enzyme substrates, cofactors, drugs, lectins, sugars, nucleic acids (both linear and cyclic polymer configurations), oligosaccharides, proteins, phospholipids and antibodies.
  • Synthetic polymers such as heteropolymers in which a known drug is covalently bound to any of the above, such as polyurethanes, polyesters, polycarbonates, polyureas, polyamides, polyethyleneimines, polyarylene sulfides, polysiloxanes, polyimides, and polyacetates are also assayed. Other polymers are also assayed using the systems described herein, as would be apparent to one of skill upon review of this disclosure. One of skill will be generally familiar with the biological literature. For a general introduction to biological systems, see, Berger and Kimmel, Guide to Molecular Cloning Techniques, Methods in Enzymology volume 152 Academic Press, Inc., San Diego, Calif. (Berger); Sambrook et al.
  • the present invention generally incorporates model in vitro systems which mimic a given biochemical system in vivo for which effector compounds are desired.
  • the range of systems against which compounds can be screened and for which effector compounds are desired, is extensive.
  • compounds are optionally screened for effects in blocking, slowing or otherwise inhibiting key events associated with biochemical systems whose effect is undesirable.
  • test compounds are optionally screened for their ability to block systems that are responsible, at least in part, for the onset of disease or for the occurrence of particular symptoms of diseases, including, e.g., hereditary diseases, cancer, bacterial or viral infections and the like.
  • Compounds which show promising results in these screening assay methods can then be subjected to further testing to identify effective pharmacological agents for the treatment of disease or symptoms of a disease.
  • compounds can be screened for their ability to stimulate, enhance or otherwise induce biochemical systems whose function is believed to be desirable, e.g., to remedy existing deficiencies in a patient.
  • batteries of test compounds can then be applied against these model systems. By identifying those test compounds that have an effect on the particular biochemical system, in vitro, one can identify potential effectors of that system, in vivo.
  • the biochemical system models employed in the methods and apparatuses of the present invention will screen for an effect of a test compound on an interaction between two components of a biochemical system, e.g., receptor-ligand interaction, enzyme-substrate interaction, and the like.
  • the biochemical system model will typically include the two normally interacting components of the system for which an effector is sought, e.g., the receptor and its ligand or the enzyme and its substrate.
  • Determining whether a test compound has an effect on this interaction then involves contacting the system with the test compound and assaying for the functioning of the system, e.g., receptor-ligand binding or substrate turnover.
  • the assayed function is then compared to a control, e.g., the same reaction in the absence of the test compound or in the presence of a known effector.
  • a control e.g., the same reaction in the absence of the test compound or in the presence of a known effector.
  • such assays involve the measurement of a parameter of the biochemical system.
  • parameter of the biochemical system is meant some measurable evidence of the system's functioning, e.g., the presence or absence of a labeled group or a change in molecular weight (e.g., in binding reactions, transport screens), the presence or absence of a reaction product or substrate (in substrate turnover measurements), or an alteration in electrophoretic mobility (typically detected by a change in elution time of a labeled compound).
  • the methods and apparatuses may also be used to screen for effectors of much more complex systems, where the result or end product of the system is known and assayable at some level, e.g., enzymatic pathways, cell signaling pathways and the like.
  • the methods and apparatuses described herein are optionally used to screen for compounds that interact with a single component of a biochemical system, e.g., compounds that specifically bind to a particular biochemical compound, e.g., a receptor, ligand, enzyme, nucleic acid, structural macromolecule, etc.
  • Biochemical system models may also be embodied in whole cell systems. For example, where one is seeking to screen test compounds for an effect on a cellular response, whole cells are optionally utilized. Modified cell systems may also be employed in the screening systems encompassed herein.
  • chimeric reporter systems are optionally employed as indicators of an effect of a test compound on a particular biochemical system.
  • Chimeric reporter systems typically incorporate a heterogenous reporter system integrated into a signaling pathway which signals the binding of a receptor to its ligand.
  • a receptor is fused to a heterologous protein, e.g., an enzyme whose activity is readily assayable. Activation of the receptor by ligand binding then activates the heterologous protein which then allows for detection.
  • the surrogate reporter system produces an event or signal which is readily detectable, thereby providing an assay for receptor/ligand binding. Examples of such chimeric reporter systems have been previously described in the art.
  • biological barriers generally refers to cellular or membranous layers within biological systems, or synthetic models thereof. Examples of such biological barriers include the epithelial and endothelial layers, e.g. vascular endothelia and the like.
  • Biological responses are often triggered and/or controlled by the binding of a receptor to its ligand.
  • growth factors i.e., EGF epidermal growth actor, FGF fibroblast growth factor, PDGF platelet derived growth factor, etc.
  • their receptors stimulates a wide variety of biological responses including, e.g., cell proliferation and differentiation, activation of mediating enzymes, stimulation of messenger turnover, alterations in ion fluxes, activation of enzymes, changes in cell shape and the alteration in genetic expression levels.
  • control of the interaction of the receptor and its ligand may offer control of the biological responses caused by that interaction.
  • the present invention will be useful in screening for compounds that affect an interaction between a receptor molecule and its ligands.
  • the term “receptor” generally refers to one member of a pair of compounds which specifically recognize and bind to each other. The other member of the pair is termed a “ligand.”
  • a receptor/ligand pair may include a typical protein receptor, usually membrane associated, and its natural ligand, e.g., another protein or small molecule.
  • Receptor/ligand pairs may also include antibody/antigen binding pairs, complementary nucleic acids, nucleic acid associating proteins and their nucleic acid ligands.
  • a large number of specifically associating biochemical compounds are well known in the art and can be utilized in practicing the present invention.
  • screening assays have typically been set up in multiweli reaction plates, e.g., multi-well microplates, which allow for the simultaneous, parallel screening of large numbers of test compounds.
  • a similar, and perhaps overlapping, set of biochemical systems includes the interactions between enzymes and their substrates.
  • enzyme as used herein, generally refers to a protein which acts as a catalyst to induce a chemical change in other compounds or “substrates.”
  • effectors of an enzyme's activity toward its substrate are screened by contacting the enzyme with a substrate in the presence and absence of the compound to be screened and under conditions optimal for detecting changes in the enzyme's activity. After a set time for reaction, the mixture is assayed for the presence of reaction products or a decrease in the amount of substrate. The amount of substrate that has been catalyzed is them compared to a control, i.e., enzyme contacted with substrate in the absence of test compound or presence of a known effector.
  • a control i.e., enzyme contacted with substrate in the absence of test compound or presence of a known effector.
  • a compound that reduces the enzymes activity toward its substrate is termed an “inhibitor,” whereas a compound that accentuates that activity is termed an “inducer.”
  • the various screening methods encompassed by the present invention involve the serial introduction of a plurality of test compounds into a microfluidic device. Once injected into the device, the test compound is screened for effect on a biological system using a continuous serial or parallel assay orientation.
  • test compound refers to the collection of compounds that are to be screened for their ability to affect a particular biochemical system.
  • Test compounds may include a wide variety of different compounds, including chemical compounds, mixtures of chemical compounds, e.g., polysaccharides, small organic or inorganic molecules, biological macromolecules, e.g., peptides, proteins, nucleic acids, or an extract made from biological materials such as bacteria, plants, fungi, or animal cells or tissues, naturally occurring or synthetic compositions.
  • the test compounds are provided, e.g., injected, free in solution, or are optionally attached to a carrier, or a solid support, e.g., beads.
  • test compounds are screened individually, or in groups. Group screening is particularly useful where hit rates for effective test compounds are expected to be low such that one would not expect more than one positive result for a given group. Alternatively, such group screening is used where the effects of different test compounds are differentially detected in a single system, e.g., through electrophoretic separation of the effects, or differential labelling which enables separate detection.
  • Test compounds are commercially available, or derived from any of a variety of biological sources apparent to one of skill and as described, supra.
  • a tissue homogenate or blood sample from a patient is tested in the assay systems of the invention.
  • blood is tested for the presence or activity of a biologically relevant molecule.
  • the presence and activity level of an enzyme are detected by supplying and enzyme substrate to the biological sample and detecting the formation of a product using an assay systems of the invention.
  • infectious pathogens viruss, bacteria, fungi, or the like
  • cancerous tumors can be tested by monitoring binding of a labeled ligand to the pathogen or tumor cells, or a component of the pathogen or tumor such as a protein, cell membrane, cell extract or the like, or alternatively, by monitoring the presence of an antibody against the pathogen or tumor in the patient's blood.
  • a labeled ligand to the pathogen or tumor cells, or a component of the pathogen or tumor such as a protein, cell membrane, cell extract or the like
  • an antibody against the pathogen or tumor in the patient's blood for example, the binding of an antibody from a patient's blood to a viral protein such as an HIV protein is a common test for monitoring patient exposure to the virus.
  • a viral protein such as an HIV protein
  • Many assays for detecting pathogen infection are well known, and are adapted to the assay systems of the present invention.
  • Bio samples are derived from patients using well known techniques such as venipuncture or tissue biopsy. Where the biological material is derived from non-human animals, such as commercially relevant livestock, blood and tissue samples are conveniently obtained from livestock processing plants. Similarly, plant material used in the assays of the invention are conveniently derived from agricultural or horticultural sources. Alternatively, a biological sample can be from a cell or blood bank where tissue and/or blood are stored, or from an in vitro source such as a culture of cells. Techniques and methods for establishing a culture of cells for use as a source for biological materials are well known to those of skill in the art. Freshney Culture of Animal Cells, a Manual of Basic Technique, Third Edition Wiley-Liss, New York (1994) provides a general introduction to cell culture.
  • the screening methods of the present invention are generally carried out in microfluidic devices or “microlaboratory systems,” which allow for integration of the elements required for performing the assay, automation, and minimal environmental effects on the assay system, e.g., evaporation, contamination, human error, or the like.
  • a number of devices for carrying out the assay methods of the invention are described in substantial detail below. However, it will be recognized that the specific configuration of these devices will generally vary depending upon the type of assay and/or assay orientation desired.
  • the screening methods of the invention can be carried out using a microfluidic device having two intersecting channels. For more complex assays or assay orientations, multichannel/intersection devices are optionally employed. The small scale, integratability and self-contained nature of these devices allows for virtually any assay orientation to be realized within the context of the microlaboratory system.
  • the devices, methods and systems described herein employ electrokinetic material transport systems, and preferably, controlled electrokinetic material transport systems.
  • electrokinetic material transport systems include systems which transport and direct materials within an interconnected channel and/or chamber containing structure, through the application of electrical fields to the materials, thereby causing material movement through and among the channel and/or chambers, i.e., cations will move toward the negative electrode, while anions will move toward the positive electrode.
  • Such electrokinetic material transport and direction systems include those systems that rely upon the electrophoretic mobility of charged species within the electric field applied to the structure. Such systems are more particularly referred to as electrophoretic material transport systems.
  • Other electrolinetic material direction and transport systems rely upon the electroosmotic flow of fluid and material within a channel or chamber structure which results from the application of an electric field across such structures.
  • a fluid is placed into a channel which has a surface bearing charged functional groups, e.g., hydroxyl groups in etched glass channels or glass microcapillaries, those groups can ionize.
  • this ionization e.g., at neutral pH, results in the release of protons from the surface and into the fluid, creating a concentration of protons at near the fluid/surface interface, or a positively charged sheath surrounding the bulk fluid in the channel.
  • Application of a voltage gradient across the length of the channel will cause the proton sheath, as well as the fluid it surrounds, to move in the direction of the voltage drop, i.e., toward the negative electrode.
  • Controlled electrokinetic material transport and direction refers to electrokinetic systems as described above, which employ active control of the voltages applied at multiple, i.e., more than two, electrodes. Rephrased, such controlled electrokinetic systems concomitantly regulate voltage gradients applied across at least two intersecting channels. Controlled electrolinetic material transport is described in Published PCT Application No. WO 96/04547, to Ramsey, which is incorporated herein by reference in its entirety for all purposes.
  • the preferred microfluidic devices and systems described herein include a body structure which includes at least two intersecting channels or fluid conduits, e.g., interconnected, enclosed chambers, which channels include at least three unintersected termini.
  • intersection of two channels refers to a point at which two or more channels are in fluid communication with each other, and encompasses “T” intersections, cross intersections, “wagon wheel” intersections of multiple channels, or any other channel geometry where two or more channels are in such fluid communication.
  • An unintersected terminus of a channel is a point at which a channel terminates not as a result of that channel's intersection with another channel, e.g., a “T” intersection.
  • the devices will include at least three intersecting channels having at least four unintersected termini.
  • controlled electrokinetic material transport operates to controllably direct material flow through the intersection, by providing constraining flows from the other channels at the intersection. For example, assuming one was desirous of transporting a first material through the horizontal channel, e.g., from left to right, across the intersection with the vertical channel. Simple electrokinetic material flow of this material across the intersection could be accomplished by applying a voltage gradient across the length of the horizontal channel, i.e., applying a first voltage to the left terminus of this channel, and a second, lower voltage to the right terminus of this channel, or by allowing the right terminus to float (applying no voltage). However, this type of material flow through the intersection would result in a substantial amount of diffusion at the intersection, resulting from both the natural diffusive properties of the material being transported in the medium used, as well as convective effects at the intersection.
  • the material being transported across the intersection is constrained by low level flow from the side channels, e.g., the top and bottom channels. This is accomplished by applying a slight voltage gradient along the path of material flow, e.g., from the top or bottom termini of the vertical channel, toward the right terminus. The result is a “pinching” of the material flow at the intersection, which prevents the diffusion of the material into the vertical channel.
  • the pinched volume of material at the intersection may then be injected into the vertical channel by applying a voltage gradient across the length of the vertical channel, i.e., from the top terminus to the bottom terminus.
  • a low level of flow is directed back into the side channels, resulting in a “pull back” of the material from the intersection.
  • controlled electrokinetic material transport is readily utilized to create virtual valves which include no mechanical or moving parts.
  • flow of material from one channel segment to another e.g., the left arm to the right arm of the horizontal channel
  • a controlled flow from the vertical channel e.g., from the bottom arm to the top arm of the vertical channel.
  • the material is transported from the left arm, through the intersection and into the top arm by applying a voltage gradient across the left and top termini.
  • a constraining flow is directed from the bottom arm to the top arm by applying a similar voltage gradient along this path (from the bottom terminus to the top terminus).
  • Metered amounts of material are then dispensed from the left arm into the right arm of the horizontal channel by switching the applied voltage gradient from left to top, to left to right.
  • the amount of time and the voltage gradient applied dictates the amount of material that will be dispensed in this manner.
  • the methods and apparatuses of the invention are used in screening test compounds using a continuous flow assay system.
  • the continuous flow assay system can be readily used in screening for inhibitors or inducers of enzymatic activity, or for agonists or antagonists of receptor-ligand binding.
  • the continuous flow assay system involves the continuous flow of the particular biochemical system along a microfabricated channel.
  • continuous generally refers to an unbroken or contiguous stream of the particular composition that is being continuously flowed.
  • a continuous flow may include a constant fluid flow having a set velocity, or alternatively, a fluid flow which includes pauses in the flow rate of the overall system, such that the pause does not otherwise interrupt the flow stream.
  • the functioning of the system is indicated by the production of a detectable event or signal.
  • detectable signals include optically detectable chromophoric or fluorescent signals that are associated with the functioning of the particular model system used.
  • signals will generally be produced by products of the enzyme's catalytic action, e.g., on a chromogenic or fluorogenic substrate.
  • binding systems e.g., receptor ligand interactions, signals will typically involve the association of a labeled ligand with the receptor, or vice versa.
  • detectable signals and labels can also be used in the assays and apparatuses of the invention.
  • radioactive decay, electron density, changes in pH, solvent viscosity, temperature and salt concentration are also conveniently measured.
  • labels are commonly detectable by spectroscopic, photochemical, biochemical, immunochemical, or chemical means.
  • useful nucleic acid labels include 32P, 35S, fluorescent dyes, electron-dense reagents, enzymes (e.g., as commonly used in an ELISA), biotin, dioxigenin, or haptens and proteins for which antisera or monoclonal antibodies are available.
  • enzymes e.g., as commonly used in an ELISA
  • biotin e.g., as commonly used in an ELISA
  • haptens and proteins for which antisera or monoclonal antibodies are available.
  • Suitable labels include radionucleotides, enzymes, substrates, cofactors, inhibitors, fluorescent moieties, chemiluminiescent moieties, magnetic particles, and the like.
  • Labeling agents optionally include e.g., monoclonal antibodies, polyclonal antibodies, proteins, or other polymers such as affinity matrices, carbohydrates or lipids. Detection proceeds by any of a variety of known methods, including spectrophotometric or optical tracking of radioactive or fluorescent markers, or other methods which track a molecule based upon size, charge or affinity.
  • a detectable moiety can be of any material having a detectable physical or chemical property.
  • a label is any composition detectable by spectroscopic, photochemical, biochemical, immunochemical, electrical, optical thermal, or chemical means.
  • Useful labels in the present invention include fluorescent dyes (e.g., fluorescein isothiocyanate, Texas red, rhodamine, and the like), radiolabels (e.g., 3H, 125I, 35S, 14C, 32P or 33P), enzymes (e.g., LacZ, CAT, horse radish peroxidase, alkaline phosphatase and others, commonly used as detectable enzymes, either as marker products or as in an EUISA), nucleic acid intercalators (e.g., ethidium bromide) and colorimetric labels such as colloidal gold or colored glass or plastic (e.g. polystyrene, polypropylene, latex, etc.) beads.
  • fluorescent dyes e.g., fluorescein isothiocyanate, Texas red, rhodamine, and the like
  • radiolabels e.g., 3H, 125I, 35S, 14C, 32P or 33P
  • Fluorescent labels are particularly preferred labels.
  • Preferred labels are typically characterized by one or more of the following: high sensitivity, high stability, low background, low environmental sensitivity and high specificity in labeling.
  • Fluorescent moieties which are incorporated into the labels of the invention, are generally are known, including 1- and 2-amironaphthalene, p,p′-diaminostilbenes, pyrenes, quaternary phenanthridine salts, 9-aminoacridines, p,p′-diaminobenzophenone imines, anthracenes, oxacarbocyanine, merocyanine, 3-aminoequilenin, perylene, bis-benzoxazole, bis-p-oxazolyl benzene, 1,2-benzophenazin, retinol, bis-3-aminopyridiniuim salts, hellebrigenin, tetracycline, sterophenol, benzimidazolylphenylamine, 2-oxo-3-chromen, indole, xanthen, 7-hydroxycoumarin, phenoxazine, calicylate, stroph
  • Individual fluorescent compounds which have functionalities for linking to an element desirably detected in an apparatus or assay of the invention, or which can be modified to incorporate such functionalities include, e.g., dansyl chloride; fluoresceins such as 3,6-dihydroxy-9-phenylxanthhydrol; rhodamineisothiocyanate; N-phenyl 1-amino-8—sulfonatonaphthalene; N-phenyl 2-amino-6-sulfonatonaphthalene; 4-acetamido-4-isothiocyanato-stilbene-2,2′-disulfonic acid; pyrene-3-sulfonic acid; 2-toluidinonaphthalene6-sulfonate; N-phenyl-N-methyl-2-aminoaphthalene-6-sulfonate; ethidim bromide; stebrine; auromine-0,2-(9′-anthroyl)palmitate; dansy
  • fluorescent tags are commercially available from SIGMA chemical company (Saint Louis, Mo.), Molecular Probes, R&D systems (Minneapolis, Minn.), Pharmacia LKB Biotechnology (Piscataway, N.J.), CLONTECH Laboratories, Inc. (Palo Alto, Calif.), Chem Genes Corp., Aldrich Chemical Company (Milwaukee, Wis.), Glen Research, Inc., GIBCO BRL Life Technologies, Inc. (Gaithersberg, Md.), Fluka Chemica-Biochemika Analytika (Fluka Chemie AG, Buchs, Switzerland), and Applied Biosystems (Foster City, Calif.) as well as other commercial sources known to one of skill.
  • fluorescent labels absorb light above about 300 nm, preferably about 350 nm, and more preferably above about 400 nm, usually emitting at wavelengths greater than about 10 nm higher than the wavelength of the light absorbed. It should be noted that the absorption and emission characteristics of the bound label may differ from the unbound label. Therefore, when referring to the various wavelength ranges and characteristics of the labels, it is intended to indicate the labels as employed and not the label which is unconjugated and characterized in an arbitrary solvent.
  • Fluorescent labels are one preferred class of detectable labels, in part because by irradiating a fluorescent label with light, one can obtain a plurality of emissions. Thus, a single label can provide for a plurality of measurable events.
  • Detectable signal may also be provided by chemiluminescent and bioluminescent sources.
  • Chemiluminescent sources include a compound which becomes electronically excited by a chemical reaction and may then emit light which serves as the detectible signal or donates energy to a fluorescent acceptor.
  • a diverse number of families of compounds have been found to provide chemiluminescence under a variety or conditions.
  • One family of compounds is 2,3-dihydro-1,4—phthalazinedione.
  • the most popular compound is luminol, which is a 5-amino compound.
  • Other members of the family include the 5-amino-6,7,8-trimethoxy- and the dimethylamino[ca]benz analog. These compounds can be made to luminesce with alkaline hydrogen peroxide or calcium hypochlorite and base.
  • Another family of compounds is the 2,4,5-triphenylimidazoles, with lophine as the common name for the parent product.
  • Chemiluminescent analogs include para-dimethylamino and -methoxy substituents.
  • Chemiluminescence may also be obtained with oxalates, usually oxalyl active esters, e.g., p-nitrophenyl and a peroxide, e.g., hydrogen peroxide, under basic conditions.
  • oxalates usually oxalyl active esters, e.g., p-nitrophenyl and a peroxide, e.g., hydrogen peroxide, under basic conditions.
  • Other useful chemiluminescent compounds are also known and available, including —N-alkyl acridimim esters (basic H 2 O 2 ) and dioxetanes.
  • luciferins may be used in conjunction with luciferase or lucigenins to provide bioluminescence.
  • the label is coupled directly or indirectly to a molecule to be detected (a product, substrate, enzyme, or the like) according to methods well known in the art.
  • a molecule to be detected a product, substrate, enzyme, or the like
  • Non radioactive labels are often attached by indirect means.
  • a ligand molecule e.g., biotin
  • the ligand then binds to an anti-ligand (e.g., streptavidin) molecule which is either inherently detectable or covalently bound to a signal system, such as a detectable enzyme, a fluorescent compound, or a chemiluminescent compound.
  • a signal system such as a detectable enzyme, a fluorescent compound, or a chemiluminescent compound.
  • ligands and anti-ligands can be used. Where a ligand has a natural anti-ligand, for example, biotin, thyroxine, and cortisol, it can be used in conjunction with labeled, anti-ligands. Alternatively, any haptenic or antigenic compound can be used in combination with an antibody. Labels can also be conjugated directly to signal generating compounds, e.g., by conjugation with an enzyme or fluorophore.
  • Enzymes of interest as labels will primarily be hydrolases, particularly phosphatases, esterases and glycosidases, or oxidoreductases, particularly peroxidases.
  • Fluorescent compounds include fluorescein and its derivatives, rhodamine and its derivatives, dansyl, umbelliferone, etc.
  • Chemiluminescent compounds include luciferin, and 2,3-dihydrophthalazinediones, e.g., luminol.
  • Means of detecting labels are well known to those of skill in the art. Thus, for example, where the label is a radioactive label, means for detection include a scintillation counter or photographic film as in autoradiography.
  • the label is a fluorescent label
  • it may be detected by exciting the fluorochrome with the appropriate wavelength of light and detecting the resulting fluorescence, e.g., by microscopy, visual inspection, via photographic film, by the use of electronic detectors such as digital cameras, charge coupled devices (CCDs) or photomultipliers and phototubes, and the like. Fluorescent labels and detection techniques, particularly microscopy and spectroscopy are preferred. Similarly, enzymatic labels are detected by providing appropriate substrates for the enzyme and detecting the resulting reaction product. Finally, simple colorimetric labels are often detected simply by observing the color associated with the label. For example, conjugated gold often appears pink, while various conjugated beads appear the color of the bead.
  • the continuous system generates a constant signal which varies only when a test compound is introduced that affects the system. Specifically, as the system components flow along the channel, they will produce a relatively constant signal level at a detection zone or window of the channel. Test compounds are periodically introduced into the channel and mixed with the system components. Where those test compounds have an effect on the system, it will cause a deviation from the constant signal level at the detection window. This deviation may then be correlated to the particular test compound screened.
  • FIG. 1 One embodiment of a device for use in a serial or continuous assay geometry is shown in FIG. 1. As shown, the overall device 100 is fabricated in a planar substrate 102 . Suitable substrate materials are generally selected based upon their compatibility with the conditions present in the particular operation to be performed by the device. Such conditions can include extremes of pH, temperature, salt concentration, and application of electrical fields. Additionally, substrate materials are also selected for their inertness to critical components of an analysis or synthesis to be carried out by the device.
  • Examples of useful substrate materials include, e.g., glass, quartz and silicon as well as polymeric substrates, e.g. plastics.
  • conductive or semi-conductive substrates it will generally be desirable to include an insulating layer on the substrate. This is particularly important where the device incorporates electrical elements, e.g., electrical material and fluid direction systems, sensors and the like.
  • the substrate materials are optionally rigid, semi-rigid, or non-rigid, opaque, semi-opaque or transparent, depending upon the use for which they are intended.
  • devices which include an optical or visual detection element will generally be fabricated, at least in part, from transparent materials to allow, or at least, facilitate that detection.
  • the polymeric materials may have linear or branched backbones, and are optionally crosslinked or non-crosslinked.
  • particularly preferred polymeric materials include, e.g., polydimethylsiloxanes (PDMS), polyurethane, polyvinylchloride (PVC) polystyrene, polysulfone, polycarbonate and the like.
  • the device shown in FIG. 1 includes a series of channels 110 , 112 , and optional reagent channel 114 , fabricated into the surface of the substrate. At least one of these channels will typically have very small cross sectional dimensions, e.g., in the range of from about 0.1 ⁇ m to about 500 ⁇ m. Preferably the cross-sectional dimensions of the channels will be in the range of from about 0.1 to about 200 ⁇ m and more preferably in the range of from about 0.1 to about 100 ⁇ m. In particularly preferred aspects, each of the channels will have at least one cross-sectional dimension in the range of from about 0.1 ⁇ m to about 100 ⁇ m. Although generally shown as straight channels, it will be appreciated that in order to maximize the use of space on a substrate, serpentine, saw tooth or other channel geometries, to incorporate effectively longer channels in shorter distances.
  • Manufacturing of these microscale elements into the surface of the substrates may generally be carried out by any number of microfabrication techniques that are well known in the art.
  • lithographic techniques are optionally employed in fabricating, e.g., glass, quartz or silicon substrates, using methods well known in the semiconductor manufacturing industries such as photolithographic etching, plasma etching or wet chemical etching.
  • micromachining methods such as laser drilling, micromilling and the like are optionally employed.
  • polymeric substrates well known manufacturing techniques may also be used. These techniques include injection molding or stamp molding methods where large numbers of substrates are optionally produced using, e.g., rolling stamps to produce large sheets of microscale substrates or polymer microcasting techniques where the substrate is polymerized within a micromachined mold.
  • the devices will typically include an additional planar element which overlays the channeled substrate enclosing and fluidly sealing the various channels to form conduits.
  • Attaching the planar cover element is achieved by a variety of means, including, e.g., thermal bonding, adhesives or, in the case of certain substrates, e.g., glass, or semi-rigid and non-rigid polymeric substrates, a natural adhesion between the two components.
  • the planar cover element may additionally be provided with access ports and/or reservoirs for introducing the various fluid elements needed for a particular screen.
  • the device shown in FIG. 1 also includes reservoirs 104 , 106 and 108 , disposed and fluidly connected at the ends of the channels 110 and 114 .
  • sample channel 112 is used to introduce the plurality of different test compounds into the device. As such, this channel will generally be fluidly connected to a source of large numbers of separate test compounds that will be individually introduced into the sample channel 112 and subsequently into channel 110 .
  • micropipettors are optionally used to introduce the test compounds into the device.
  • an electropipettor is used which is fluidly connected to sample channel 112 .
  • An example of such an electropipettor is described in, e.g., U.S. patent application Ser. No. 08/671,986, filed Jun. 28, 1996, now U.S. Pat. No. 5,779,868, the disclosure of which is hereby incorporated herein by reference in its entirety for all purposes.
  • this electropipettor utilizes electroosmotic fluid direction as described herein, to alternately sample a number of test compounds, or “subject materials,” and spacer compounds.
  • the pipettor then delivers individual, physically isolated sample or test compound volumes in subject material regions, in series, into the sample channel for subsequent manipulation within the device.
  • Individual samples are typically separated by a spacer region of low ionic strength spacer fluid. These low ionic strength spacer regions have higher voltage drop over their length than do the higher ionic strength subject material or test compound regions, thereby driving the electrokinetic pumping.
  • first spacer regions On either side of the test compound or subject material region, which is typically in higher ionic strength solution, are fluid regions referred to as first spacer regions (also referred to as “guard bands”), that contact the interface of the subject material regions.
  • first spacer regions typically comprise a high ionic strength solution to prevent migration of the sample elements into the lower ionic strength fluid regions, or second spacer region, which would result in electrophoretic bias.
  • the use of such first and second spacer regions is described in greater detail in U.S. patent application Ser. No. 08/671,986, filed Jun. 28, 1996, now U.S. Pat. No. 5,779,868, which is incorporated herein by reference.
  • the sample channel 112 is optionally individually fluidly connected to a plurality of separate reservoirs via separate channels.
  • the separate reservoirs each contain a separate test compound with additional reservoirs being provided for appropriate spacer compounds.
  • the test compounds and/or spacer compounds are then transported from the various reservoirs into the sample channels using appropriate material direction schemes. In either case, it generally is desirable to separate the discrete sample volumes, or test compounds, with appropriate spacer regions.
  • the device also includes a detection window or zone 116 at which a signal from the biochemical system is optionally monitored.
  • This detection window typically will include a transparent cover allowing visual or optical observation and detection of the assay results, e.g., observation of a colorometric or fluorometric response.
  • monitoring of the signals at the detection window is achieved using an optical detection system.
  • fluorescence based signals are typically monitored using, e.g., laser activated fluorescence detection systems which employ a laser light source at an appropriate wavelength for activating the fluorescent indicator within the system. Fluorescence is then detected using an appropriate detector element, e.g., a photomultiplier tube (PMT).
  • PMT photomultiplier tube
  • spectrophotometric detection systems which direct a light source at the sample are optionally used, providing a measurement of absorbance or translnissivity of the sample.
  • the detection system may comprise non-optical detectors or sensors for detecting a particular characteristic of the system disposed within detection window 116 .
  • sensors may include temperature, conductivity, potentiometric (pH, ions), amperometric (for compounds that are oxidized or reduced, e.g., O 2 , H 2 O 2 , I 2 , oxidizable/reducible organic compounds, and the like).
  • a flowable first component of a biological system e.g., a fluid comprising a receptor or enzyme
  • This first component is flowed through main channel 110 , past the detection window, 116 , and toward waste reservoir 108 .
  • a second component of the biochemical system e.g., a ligand or substrate, is concurrently flowed into the main channel 110 from the side channel 114 , whereupon the first and second components mix and are able to interact.
  • Deposition of these elements within the device is carried out in a number of ways.
  • the enzyme and substrate, or receptor and ligand solutions can be introduced into the device through open or sealable access ports in the planar cover.
  • these components are optionally added to their respective reservoirs during manufacture of the device.
  • the enzyme/substrate or receptor/ligand components are optionally provided within the device in lyophilized form. Prior to use, these components are easily reconstituted by introducing a buffer solution into the reservoirs. Alternatively, the components are lyophilized with appropriate buffering salts, whereby simple water addition is all that is required for reconstitution.
  • the interaction of the first and second components is typically accompanied by a detectable signal.
  • the substrate is a chromogenic or fluorogenic substrate which produces an optically detectable signal when the enzyme acts upon the substrate.
  • the first component is a receptor and the second is a ligand
  • either the ligand or the receptor optionally includes a detectable signal.
  • the mixture and flow rate of compounds will typically remain constant such that the flow of the mixture of the first and second components past the detection window 116 will produce a steady-state signal.
  • steady state signal is generally meant a signal that has a regular, predictable signal intensity profile.
  • the steady-state signal may include signals having a constant signal intensity, or alternatively, a signal with a regular periodic intensity, against which variations in the normal signal profile is measured.
  • This latter signal is generated in cases where fluid flow is periodically interrupted for, e.g., loading additional test compounds, as described in the description of the continuous flow systems.
  • the signal produced in the above-described enzymatic system will vary along the length of the channel, i.e., increasing with time of exposure as the enzyme converts the fluorogenic substrate to the fluorescent product, the signal at any specific point along the channel will remain constant, given a constant flow rate.
  • test compounds is periodically or serially introduced into the main channel 110 and into the stream of first and second components as fluid regions containing the test compound, also referred to as the “subject material regions.” Where these test compounds have an effect on the interaction of the first and second elements, it will produce a deviation in the signal detected at the detection window corresponding to the subject material region.
  • the various different test compounds to be injected through channel 112 will be separated by a first and even second spacer fluid regions to allow differentiation of the effects, or lack of effects, from one test compound to another.
  • the spacer fluid regions may also function to reduce any electrophoretic bias that can occur within the test sample.
  • a steady, continuous flow of enzyme and fluorogenic substrate through main channel 110 will produce a constant fluorescent signal at the detection window 116 .
  • introduction of a test compound i.e., in a subject material region, will produce a momentary but detectable drop in the level of signal at the detection window corresponding with that subject material region.
  • the timing of the drop in signal can then be correlated with a particular test compound based upon a known injection to detection time-frame. Specifically, the time required for an injected compound to produce an observed effect can be readily determined using positive controls.
  • the receptor and its fluorescent ligand can be made to have different flow rates along the channel. This can be accomplished by incorporating size exclusion matrices within the channel, or, in the case of electroosmotic methods, altering the relative electrophoretic mobility of the two compounds so that the receptor flows more rapidly down the channel. Again, this is accomplished through the use of size exclusion matrices, or through the use of different surface charges in the channel which will result in differential flow rates of charge-varied compounds. Where a test compound binds to the receptor, it will result in a dark pulse in the fluorescent signal followed by a brighter pulse.
  • the steady state signal is a result of both free fluorescent ligand, and fluorescent ligand bound to the receptor.
  • the bound ligand is traveling at the same flow rate as the receptor while the unbound ligand is traveling more slowly.
  • the receptor will not ‘bring along’ the fluorescent ligand, thereby diluting the fluorescent ligand in the direction of flow, and leaving an excess of free fluorescent ligand behind. This results in a temporary reduction in the steady-state signal, followed by a temporary increase in fluorescence.
  • schemes similar to those employed for the enzymatic system is employed, where there is a signal that reflects the interaction of the receptor with its ligand.
  • pH indicators which indicate pH effects of receptor-ligand binding is incorporated into the device along with the biochemical system, i.e., in the form of encapsulated cells, whereby slight pH changes resulting from binding can be detected. See Weaver, et al., Bio/Technology (1988) 6:1084-1089. Additionally, one can monitor activation of enzymes resulting from receptor ligand binding, e.g., activation of kinases, or detect conformational changes in such enzymes upon activation, e.g., through incorporation of a fluorophore which is activated or quenched by the conformational change to the enzyme upon activation.
  • the devices may include integrated microfluidic structures, such as micropumps and microvalves, or external elements, e.g., pumps and switching valves, for the pumping and direction of the various fluids through the device.
  • integrated microfluidic structures such as micropumps and microvalves
  • external elements e.g., pumps and switching valves
  • microfluidic structures are described in, e.g., U.S. Pat. Nos. 5,271,724, 5,277,556, 5,171,132, and 5,375,979. See also, Published U.K. Patent Application No. 2 248 891 and Published European Patent Application No. 568 902.
  • the devices of the invention will typically include an electroosmotic fluid direction system.
  • electroosmotic fluid direction systems combine the elegance of a fluid direction system devoid of moving parts, with an ease of manufacturing, fluid control and disposability.
  • electroosmotic fluid direction systems include, e.g., those described in U.S. Pat. No. 5,858,195 to Ramsey et al., which is incorporated herein by reference in its entirety for all purposes.
  • these fluidic control systems typically include electrodes disposed within the reservoirs that are placed in fluid connection with the plurality of intersecting channels fabricated into the surface of the substrate.
  • the materials stored in the reservoirs are transported through the channel system delivering appropriate volumes of the various materials to one or more regions on the substrate in order to carry out a desired screening assay.
  • Fluid and materials transport and direction is accomplished through electroosmosis or electrokinesis.
  • an appropriate material typically comprising a fluid
  • those groups can ionize.
  • protons can leave the surface of the channel and enter the fluid.
  • the surface will possess a net negative charge
  • the fluid will possess an excess of protons or positive charge, particularly localized near the interface between the channel surface and the fluid.
  • cations will flow toward the negative electrode. Movement of the positively charged species in the fluid pulls the solvent with them.
  • v is the solvent velocity
  • is the dielectric constant of the fluid
  • is the zeta potential of the surface
  • E is the electric field strength
  • is the solvent viscosity
  • the system generally includes a voltage controller that is capable of applying selectable voltage levels, simultaneously, to each of the reservoirs, including ground.
  • a voltage controller can be implemented using multiple voltage dividers and multiple relays to obtain the selectable voltage levels. Alternatively, multiple, independent voltage sources are optionally used.
  • the voltage controller is electrically connected to each of the reservoirs via an electrode positioned or fabricated within each of the plurality of reservoirs.
  • Incorporating this electroosmotic fluid direction system into the device shown in FIG. 1 involves incorporation of an electrode within each of the reservoirs 104 , 106 and 108 , and at the terminus of sample channel 112 or at the terminus of any fluid channels connected thereto, whereby the electrode is in electrical contact with the fluid disposed in the respective reservoir or channel.
  • Substrate materials are also selected to produce channels having a desired surface charge. In the case of glass substrates, the etched channels will possess a net negative charge resulting from the ionized hydrolysis naturally present at the surface.
  • surface modifications are optionally employed to provide an appropriate surface charge, e.g., coatings, derivatization, e.g., silanation, or impregnation of the surface to provide appropriately charged groups on the surface. Examples of such treatments are described in, e.g., U.S. Pat. No. 5,885,470, which is hereby incorporated herein by reference in its entirety for all purposes.
  • suitable substrate materials are generally selected based upon their compatibility with the conditions present in the particular operation to be performed by the device. Such conditions can include extremes of pH, temperature and salt concentration. Additionally, substrate materials are also selected for their inertness to critical components of an analysis or synthesis to be carried out by the device.
  • Polymeric substrate materials may be rigid, semi-rigid, or non-rigid, opaque, semi-opaque or transparent, depending upon the use for which they are intended.
  • devices which include an optical or visual detection element will generally be fabricated, at least in part, from a transparent polymeric material to facilitate that detection. Alternatively, transparent windows of, e.g. glass or quartz, may be incorporated into the device for these detection elements.
  • polymeric materials may have linear or branched backbones, and may be crosslinked or non-crosslinked.
  • polymeric materials include, e.g., Acrylics, especially PMMAs (polymethylmethacrylates); exemplar acrylics include e.g., Acrylite M-30 or Acrylite L40 available from CYRO Industries, Rockaway, N.J., or PLEXIGLAS VS UVT available from Autohaas North America; polycarbonates (e.g., Makrolon CD-2005 available from The Plastics and Rubber division of Mobay Corporation (Pittsburg, Pa.) or Bayer Corporation, or LEXAN OQ 1020L or LEXAN OQ 1020, both available from GE Plastics) polydimethylsiloxanes (PDMS), polyurethane, polyvinylchloride (PVC) polystyrene, polysulfone, polycarbonate and the like.
  • PDMS polydimethylsiloxanes
  • PVC polyurethane
  • PVC polyvin
  • the electrokinetic fluid control systems employed in the devices of the present invention generally utilize a substrate having charged functional groups at its surface, such as the hydroxyl groups present on glass surfaces.
  • devices of the present invention can also employ plastic or other polymeric substrates. In general, these substrate materials have hydrophobic surfaces.
  • use of electrokinetic fluid control systems in devices utilizing polymeric substrates used in the present invention typically employs modification of the surfaces of the substrate that are in contact with fluids.
  • Surface modification of polymeric substrates may take on a variety of different forms.
  • surfaces may be coated with an appropriately charged material.
  • surfactants with charged groups and hydrophobic tails are desirable coating materials.
  • the hydrophobic tails will localize to the hydrophobic surface of the substrate, thereby presenting the charged head group at the fluid layer.
  • preparation of a charged surface on the substrate involves the exposure of the surface to be modified, e.g., the channels and/or reaction chambers, to an appropriate solvent which partially dissolves or softens the surface of the polymeric substrate.
  • a detergent is then contacted with the partially dissolved surface.
  • the hydrophobic portion of the detergent molecules will associate with the partially dissolve polymer.
  • the solvent is then washed from the surface, e.g., using water, whereupon the polymer surface hardens with the detergent embedded into the surface, presenting the charged head group to the fluid interface.
  • polymeric materials such as polydimethylsiloxane
  • plasma irradiation of PDMS oxidizes the methyl groups, liberating the carbons and leaving hydroxyl groups in their place, effectively creating a glass-like surface on the polymeric material, with its associated hydroxyl functional groups.
  • the polymeric substrate may be rigid, semi-rigid, nonrigid or a combination of rigid and nonrigid elements, depending upon the particular application for which the device is to be used.
  • a substrate is made up of at least one softer, flexible substrate element and at least one harder, more rigid substrate element, one of which includes the channels and chambers manufactured into its surface.
  • the inclusion of the soft element allows formation of an effective fluid seal for the channels and chambers, obviating the need and problems associated with gluing or melting more rigid plastic components together.
  • a number of additional elements are added to the polymeric substrate to provide for the electrokinetic fluid control systems. These elements may be added either during the substrate formation process, i.e., during the molding or stamping steps, or they may be added during a separate, subsequent step. These elements typically include electrodes for the application of voltages to the various fluid reservoirs, and in some embodiments, voltage sensors at the various channel intersections to monitor the voltage applied.
  • Electrodes may be incorporated as a portion of the molding process.
  • the electrodes may be patterned within the mold so that upon introduction of the polymeric material into the mold, the electrodes will be appropriately placed.
  • the electrodes and other elements may be added after the substrate is formed, using well known microfabrication methods, e.g., sputtering or controlled vapor deposition methods followed by chemical etching.
  • modulating voltages are concomitantly applied to the various reservoirs to affect a desired fluid flow characteristic, e.g., continuous flow of receptor/enzyme, ligand/substrate toward the waste reservoir with the periodic introduction of test compounds.
  • a desired fluid flow characteristic e.g., continuous flow of receptor/enzyme, ligand/substrate toward the waste reservoir with the periodic introduction of test compounds.
  • modulation of the voltages applied at the various reservoirs can move and direct fluid flow through the interconnected channel structure of the device in a controlled manner to effect the fluid flow for the desired screening assay and apparatus.
  • FIG. 2A shows a schematic illustration of fluid direction during a typical assay screen. Specifically, shown is the injection of a test compound (in a subject material region) into a continuous stream of an enzyme-fluorogenic substrate mixture. As shown in FIG. 2A, and with reference to FIG. 1, a continuous stream of enzyme is flowed from reservoir 104 , along main channel 110 . Test compounds 120 , separated by appropriate spacer regions 121 , e.g., low ionic strength spacer regions, are introduced from sample channel 112 into main channel 110 . Once introduced into the main channel, the test compounds will interact with the flowing enzyme stream. The mixed enzyme/test compound regions are then flowed along main channel 110 past the intersection with channel 114 .
  • spacer regions 121 e.g., low ionic strength spacer regions
  • a continuous stream of fluorogenic or chromogenic substrate which is contained in reservoir 106 is introduced into sample channel 110 , whereupon it contacts and mixes with the continuous stream of enzyme, including the subject material regions which include the test compounds 122 .
  • Action of the enzyme upon the substrate will produce an increasing level of the fluorescent or chromatic signal. This increasing signal is indicated by the increasing shading within the main channel as it approaches the detection window. This signal trend will also occur within those test compound or subject material regions which have no effect on the enzyme/substrate interaction, e.g., test compound 126 . Where a test compound does have an effect on the interaction of the enzyme and the substrate, a variation will appear in the signal produced.
  • a test compound which inhibits the interaction of the enzyme with its substrate will result in less fluorescent product being produced within that subject material region. This will result in a non-fluorescent, or detectably less fluorescent region within the flowing stream as it passes detection window 116 , which corresponds to the subject material region.
  • a subject material region including a test compound 128 which is a putative inhibitor of the enzyme-substrate interaction, shows detectably lower fluorescence than the surrounding stream. This is indicated by a lack of shading of subject material region 128 .
  • a detector adjacent to the detection window monitors the level of fluorescent signal being produced by the enzyme's activity on the fluorogenic or chromogenic substrate. This signal remains at a relatively constant level for those test compounds which have no effect on the enzyme-substrate interaction. When an inhibitory compound is screened, however, it will produce a momentary drop in the fluorescent signal representing the reduced or inhibited enzyme activity toward the substrate. Conversely, inducer compounds, upon screening, produce a momentary increase in the fluorescent signal, corresponding to the increased enzyme activity toward the substrate.
  • FIG. 2B provides a similar schematic illustration of a screen for effectors of a receptor-ligand interaction.
  • a continuous stream of receptor is flowed from reservoir 104 through main channel 110 .
  • Test compounds or subject material regions 150 separated by appropriate spacer fluid regions 121 are introduced into the main channel 110 from sample channel 112 , and a continuous stream of fluorescent ligand from reservoir 106 is introduced from side channel 114 . Fluorescence is indicated by shading within the channel.
  • the continuous stream of fluorescent ligand and receptor past the detection window 116 will provide a constant signal intensity.
  • the subject material regions in the stream containing the test compounds which have no effect on the receptor-ligand interaction, will provide the same or similar level of fluorescence as the rest of the surrounding stream, e.g., test compound or subject material region 152 .
  • the presence of test compounds which possess antagonistic or inhibitory activity toward the receptor-ligand interaction will result in lower levels of that interaction in those portions of the stream where those compounds are located, e.g., test compound or subject material region 154 .
  • differential flow rates for the receptor bound fluorescent ligand and free fluorescent ligand will result in a detectable drop in the level of fluorescence which corresponds to the dilution of the fluorescence resulting from unbound, faster moving receptor. The drop in fluorescence is then followed by an increase in fluorescence 156 which corresponds to an accumulation of the slower moving, unbound fluorescent ligand.
  • FIG. 5 A schematic illustration of one embodiment of a device 500 for performing this sample or subject material shunting or extraction is shown in FIG. 5.
  • the subject materials or test compounds 504 are introduced to the device or chip via the sample channel 512 . Again, these are typically introduced via an appropriate injection device 506 , e.g., a capillary pipettor.
  • the ionic strength and lengths of the first spacer regions 508 and second spacer regions 502 are selected such that those samples with the highest electrophoretic mobility will not migrate through the first spacer regions 508 into the second spacer regions 502 in the length of time that it takes the sample to travel down the reaction channel.
  • test compounds pass into the device 500 and into reaction channel 510 , where they are first combined with the receptor.
  • the test compound/receptor in the form of the subject material regions, are flowed along the reaction channel in the incubation zone 510 a .
  • the test compound/receptor mix is combined with a labelled ligand (e.g., fluorescent ligand) whereupon this mixture flows along the second incubation region 510 b of reaction channel 510 .
  • a labelled ligand e.g., fluorescent ligand
  • the lengths of the incubation regions and the flow rates of the system determine the time of incubation of the receptor with the fluorescent ligand and test compound.
  • the ionic strengths of the solutions containing the receptors and fluorescent ligands, as well as the flow rates of material from the reservoirs housing these elements into the sample channel are selected so as to not interfere with the first and second spacer regions.
  • the isolated subject material regions containing receptor, fluorescent ligand and test compound are flowed along the reaction channel 510 by the application of potentials at, e.g., reservoirs 514 , 516 , 518 and at the terminus of sample channel 512 .
  • Potentials are also applied at reservoirs 520 and 522 , at the opposite ends of separation channel 524 , to match the potentials at the two ends of the transfer channel, so that the net flow across the transfer channel is zero.
  • reaction channel 510 and transfer channel 526 the potentials are allowed to float at reservoirs 518 and 522 , whereupon the potentials applied at reservoirs 514 , 516 , 520 , and at the terminus of sample channel 512 , result in the subject material region being shunted through transfer channel 526 and into separation channel 524 .
  • the original potentials are reapplied to all of the reservoirs to stop the net fluid flow through transfer channel 526 .
  • the diversion of the subject material can then be repeated with each subsequent subject material region. Within the separation channel, the subject material region is exposed to different conditions than those of the reaction channel.
  • the subject material is shunted into the separation channel to place the subject material into a capillary filled with high ionic strength buffer, i.e., to remove the low ionic strength spacer regions, thereby allowing separation of the various sample components outside the confines of the original subject material region.
  • the receptor/ligand complex may have a different electrophoretic mobility from the ligand alone, in the transfer channel, thereby allowing more pronounced separation of the complex from the ligand, and its subsequent detection.
  • FIG. 3 a schematic illustration of one alternate embodiment employing a “serial input parallel reaction” geometry is shown in FIG. 3.
  • the device 300 again includes a planar substrate 302 as described previously. Fabricated into the surface of the substrate 302 are a series of parallel reaction channels 312 - 324 . Also shown are three transverse channels fluidly connected to each of these parallel reaction channels. The three transverse channels include a sample injection channel 304 , an optional seeding channel 306 and a collection channel 308 . Again, the substrate and channels are generally fabricated utilizing the materials and to the dimensions generally described above.
  • the reaction channels may also be fabricated in a variety of different orientations.
  • the channels are optionally fabricated connecting to and extending radially outward from a central reservoir, or are optionally arranged in some other non-parallel fashion.
  • the channels are optionally fabricated connecting to and extending radially outward from a central reservoir, or are optionally arranged in some other non-parallel fashion.
  • three transverse channels it will be recognized that fewer transverse channels are used where, e.g., the biochemical system components are predisposed within the device.
  • more transverse channels are optionally used to introduce further elements into a given assay screen.
  • the serial-in-parallel devices of the present invention will typically include at least two and preferably three, four, five or more transverse channels.
  • the microscale devices of the present invention will be capable of comprising more than 7 channels, depending upon the needs of the particular screen.
  • the devices will include from 10 to about 500 reaction channels, and more preferably, from 20 to about 200 reaction channels.
  • This device may be particularly useful for screening test compounds serially injected into the device, but employing a parallel assay geometry, once the samples are introduced into the device, to allow for increased throughput.
  • test compounds in discrete subject material regions are serially introduced into the device, separated as described above, and flowed along the transverse sample injection channel 304 until the separate subject material regions are adjacent the intersection of the sample channel 304 with the parallel reaction channels 310 - 324 .
  • the test compounds are optionally provided immobilized on individual beads.
  • the parallel channels are optionally fabricated to include bead resting wells 326 - 338 at the intersection of the reaction channels with the sample injection channel 304 .
  • Arrows 340 indicate the net fluid flow during this type of sample/bead injection. As individual beads settle into a resting well, fluid flow through that particular channel will be generally restricted. The next bead in the series following the unrestricted fluid flow, then flows to the next available resting well to settle in place.
  • the test compound is directed into its respective reaction channel by redirecting fluid flows down those channels.
  • the immobilization will typically be via a cleavable linker group, e.g., a photolabile, acid or base labile linker group.
  • the test compound will typically need to be released from the bead, e.g., by exposure to a releasing agent such as light, acid, base or the like prior to flowing the test compound down the reaction channel.
  • the test compound will be contacted with the biochemical system for which an effector compound is being sought.
  • the first component of the biochemical system is placed into the reaction channels using a similar technique to that described for the test compounds.
  • the biochemical system is typically introduced via one or more transverse seeding channels 306 .
  • Arrows 342 illustrate the direction of fluid flow within the seeding channel 306 .
  • the biochemical system are optionally solution based, e.g., a continuously flowing enzyme/substrate or receptor-ligand mixture, like that described above, or as shown in FIGS. 4 A- 4 F, may be a whole cell or bead based system, e.g., beads which have enzyme/substrate systems immobilized thereon.
  • the parallel channel may include a particle retention zone 344 .
  • retention zones will include a particle sieving or filtration matrix, e.g., a porous gel or microstructure which retains particulate material but allows the free flow of fluids.
  • microstructures for this filtration include, e.g., those described in U.S. Pat. No. 5,304,487, which is hereby incorporated by reference in its entirety for all purposes.
  • fluid direction within the more complex systems may be generally controlled using microfabricated fluid direction structures, e.g., pumps and valves.
  • electroosmotic systems are generally preferred for controlling fluid in these more complex systems.
  • such systems will incorporate electrodes within reservoirs disposed at the termini of the various transverse channels to control fluid flow thorough the device.
  • it may often be desirable in parallel systems e.g., where two fluids are desired to move at similar rates in parallel channels, to adjust the geometries of the various flow channels. In particular, as channel length increases, resistance along that channel will also increase.
  • flow lengths between electrodes should be designed to be substantially the same regardless of the parallel path chosen. This will generally prevent the generation of transverse electrical fields and thus promote equal flow in all parallel channels.
  • resistance of channels are optionally adjusted by varying the cross-sectional dimensions of the paths, thereby creating uniform resistance levels regardless of the path taken.
  • test compounds As the test compounds are drawn through their respective parallel reaction channels, they will contact the biochemical system in question.
  • the particular biochemical system will typically include a flowable indicator system which indicates the relative functioning of that system, e.g., a soluble indicator such as chromogenic or fluorogenic substrate, labelled ligand, or the like, or a particle based signal, such as a precipitate or bead bound signaling group.
  • the flowable indicator is then flowed through the respective parallel channel and into the collection channel 308 whereupon the signals from each of the parallel channels are flowed, in series, past the detection window, 116 .
  • FIGS. 4 A- 4 F show a schematic illustration of the progression of the injection of test compounds and biochemical system components into the “serial input parallel reaction” device, exposure of the system to the test compounds, and flowing of the resulting signal out of the parallel reaction channels and past the detection window.
  • FIG. 4A shows the introduction of test compounds immobilized on beads 346 through sample injection channel 304 .
  • the biochemical system components 348 are introduced into the reaction channels 312 - 324 through seeding channel 306 .
  • the components of the model system to be screened are optionally incorporated into the reaction channels during manufacture. Again, such components are optionally provided in liquid form or in lyophilized form for increased shelf life of the particular screening device.
  • the biochemical system components are embodied in a cellular or particle based system, however, fluid components may also be used as described herein. As the particulate components flow into the reaction channels, they are optionally retained upon an optional particle retaining matrix 344 , as described above.
  • FIG. 4B illustrates the release of test compounds from the beads 346 by exposing the beads to a releasing agent. As shown, the beads are exposed to light from an appropriate light source 352 , e.g., which is able to produce light in a wavelength sufficient to photolyze the linker group, thereby releasing compounds that are coupled to their respective beads via a photolabile linker group.
  • an appropriate light source 352 e.g., which is able to produce light in a wavelength sufficient to photolyze the linker group, thereby releasing compounds that are coupled to their respective beads via a photolabile linker group.
  • the released test compounds are flowed into and along the parallel reaction channels as shown by arrows 354 until they contact the biochemical system components.
  • the biochemical system components 348 are then allowed to perform their function, e.g., enzymatic reaction, receptor/ligand interaction, and the like, in the presence of the test compounds.
  • a soluble signal 356 which corresponds to the functioning of the biochemical system is then generated (FIG. 4D).
  • a variation in the level of signal produced is an indication that the particular test compound is an effector of the particular biochemical system. This is illustrated by the lighter shading of signal 358 .
  • the soluble signal is then flowed out of reactions channels 312 - 324 into the detection channel 308 , and along the detection channel past the detection window 116 .
  • the beads which bore the test compounds are optionally recovered to identify the test compounds which were present thereon. This is typically accomplished by incorporation of a tagging group during the synthesis of the test compound on the bead.
  • spent bead 360 i.e., from which a test compound has been released, is optionally transported out of the channel structure through port 362 for identification of the test compound that had been coupled to it.
  • identification are optionally accomplished outside of the device by directing the bead to a fraction collector, whereupon the test compounds present on the beads are optionally identified, either through identification of a tagging group, or through identification of residual compounds.
  • FIG. 6A shows an alternate embodiment of a “serial input parallel reaction” device which can be used for fluid based as opposed to bead based systems.
  • the device 600 generally incorporates at least two transverse channels as were shown in FIGS. 3 and 4, namely, sample injection channel 604 and detection channel 606 . These transverse channels are interconnected by the series of parallel channels 612 - 620 which connect sample channel 604 to detection channel 606 .
  • the device shown also includes an additional set of channels for directing the flow of fluid test compounds into the reaction channels.
  • an additional transverse pumping channel 634 is fluidly connected to sample channel 604 via a series of parallel pumping channels 636 - 646 .
  • the pumping channel includes reservoirs 650 and 652 at its termini.
  • the intersections of parallel channels 636 - 646 are staggered from the intersections of parallel channels 612 - 620 with sample channel 604 , e.g., half way between.
  • transverse pumping channel 608 is connected to detection channel 606 via parallel pumping channels 622 - 632 . Again, the intersections of parallel pumping channels 622 - 632 with detection channel 606 are staggered from the intersections of reaction channels 612 - 620 with the detection channel 606 .
  • FIGS. 6 B- 6 C A schematic illustration of the operation of this system is shown in FIGS. 6 B- 6 C.
  • a series of test compounds physically isolated from each other in separate subject material regions, are introduced into sample channel 604 using the methods described previously.
  • potentials are applied at the terminus of sample channel 604 , as well as reservoir 648 .
  • Potentials are also applied at reservoirs 650 : 652 , 654 : 656 , and 658 : 660 . This results in a fluid flow along the transverse channels 634 , 604 , 606 and 608 , as illustrated by the arrows, and a zero net flow through the parallel channel arrays interconnecting these transverse channels, as shown in FIG. 6B.
  • the geometry of the channels can be varied to maximize the use of space on the substrate. For example, where the sample channel is straight, the distance between reaction channels (and thus, the number of parallel reactions that can be carried out in a size limited substrate) is dictated by the distance between subject material regions. These restrictions, however, can be eliminated through the inclusion of altered channel geometries. For example, in some aspects, the length of a first and second spacer regions can be accommodated by a serpentine, square-wave, saw tooth or other reciprocating channel geometry. This allows packing a maximum number of reaction channels onto the limited area of the substrate surface.
  • the sample, or subject material is then moved into the parallel reaction channels 612 - 620 by applying a first potential to reservoirs 650 and 652 , while applying a second potential to reservoirs 658 and 660 , whereby fluid flow through parallel pumping channels 636 - 646 forces the subject material into parallel reaction channels 612 - 620 , as shown in FIG. 6C.
  • no potential is applied at reservoirs— 648 , 654 , 656 , or the terminus of sample channel 604 .
  • Parallel channels 636 - 646 and 622 - 632 are generally adjusted in length such that the total channel length, and thus the level of resistance, from reservoirs 650 and 652 to channel 604 and from reservoirs 658 and 660 to channel 606 , for any path taken will be the same.
  • Resistance can generally be adjusted by adjusting channel length or width.
  • channels can be lengthened by including folding or serpentine geometries.
  • channels 636 and 646 would be the longest and 640 and 642 the shortest, to create symmetric flow, thereby forcing the samples into the channels.
  • the resistance within these channels will be the same, as the individual channel length is the same.
  • the subject material region/signal element is moved into detection channel 606 by applying a potential from reservoirs 650 and 652 to reservoirs 658 and 660 , while the potentials at the remaining reservoirs are allowed to float.
  • the subject material regions/signal are then serially moved past the detection window/detector 662 by applying potentials to reservoirs 654 and 656 , while applying appropriate potentials at the termini of the other transverse channels to prevent any flow along the various parallel channels.
  • the current and thus the fluid flow, follows the path of least resistance, e.g., the shortest path.
  • this problem of transverse electrical fields is alleviated through the use of separate electrical systems, i.e., separate electrodes, at the termini of each and every parallel channel
  • production of devices incorporating all of these electrodes, and control systems for controlling the electrical potential applied at each of these electrodes can be complex, particularly where one is dealing with hundreds to thousands of parallel channels in a single small scale device, e.g., 1-2 cm 2 .
  • the present invention provides microfluidic devices for affecting serial to parallel conversion, by ensuring that current flow through each of a plurality of parallel channels is at an appropriate level to ensure a desired flow pattern through those channels or channel networks. A number of methods and substrate/channel designs for accomplishing these goals are appropriate.
  • the substrate includes a main channel.
  • a series of parallel channels terminate in a main channel.
  • the opposite termini of these parallel channels are connected to parabolic channels.
  • Electrodes are disposed at the termini of these parabolic channels.
  • the current flow in each of the parallel channels is maintained constant or equivalent, by adjusting the length of the parallel channels, resulting in a parabolic channel structure connecting each of the parallel channels to its respective electrodes.
  • the voltage drop within the parabolic channel between the parallel channels is maintained constant by adjusting the channel width to accommodate variations in the channel current resulting from the parallel current paths created by these parallel channels.
  • the parabolic design of the channels results in the resistance along all of the parallel channels being equal, resulting in an equal fluid flow, regardless of the path chosen.
  • determining the dimensions of channels to ensure that the resistances among the channels are controlled as desired may be carried out by well known methods, and generally depends upon factors such as the make-up of the fluids being moved through the substrates.
  • microlaboratory systems may also be used to screen for compounds which specifically interact with a component of a biochemical system without necessarily affecting an interaction between that component and another element of the biochemical system.
  • Such compounds typically include binding compounds which may generally be used in, e.g., diagnostic and therapeutic applications as targeting groups for therapeutics or marker groups, i.e. radionuclides, dyes and the like.
  • binding compounds may generally be used in, e.g., diagnostic and therapeutic applications as targeting groups for therapeutics or marker groups, i.e. radionuclides, dyes and the like.
  • these systems are optionally used to screen test compounds for the ability to bind to a given component of a biochemical system.
  • the system may include a test compound processing system 700 .
  • the system shown includes a platform 702 which can hold a number of separate assay chips or devices 704 .
  • each chip includes a number of discrete assay channels 706 , each having a separate interface 708 , e.g., pipettor, for introducing test compounds into the device.
  • interfaces are used to sip test compounds into the device, separated by sipping first and second spacer fluids, into the device.
  • the interfaces of the chip are inserted through an opening 710 in the bottom of the platform 702 , which is capable of being raised and lowered to place the interfaces in contact with test compounds or wash/first spacer fluids/second spacer fluids, which are contained in, e.g., multiwell micro plates 711 , positioned below the platform, e.g., on a conveyor system 712 .
  • multiwell plates containing large members of different test compounds are stacked 714 at one end of the conveyor system.
  • the plates are placed upon the conveyor separated by appropriate buffer reservoirs 716 and 718 , which may be filled by buffer system 720 .
  • the plates are stepped down the conveyor and the test compounds are sampled into the chips, interspersed by appropriate spacer fluid regions.
  • the overall control system includes a number of individual microlaboratory systems or devices, e.g., as shown in FIG. 7. Each device is connected to a computer system which is appropriately programmed to control fluid flow and direction within the various chips, and to monitor, record and analyze data resulting from the screening assays that are performed by the various devices.
  • the devices will typically be connected to the computer through an intermediate adapter module which provides an interface between the computer and the individual devices for implementing operational instructions from the computer to the devices, and for reporting data from the devices to the computer.
  • the adapter will generally include appropriate connections to corresponding elements on each device, e.g., electrical leads connected to the reservoir based electrodes that are used for electroosmotic fluid flow, power inputs and data outputs for detection systems, either electrical or fiberoptic, and data relays for other sensor elements incorporated into the devices.
  • the adapter device may also provide environmental control over the individual devices where such control is necessary, e.g., maintaining the individual devices at optimal temperatures for performing the particular screening assays.
  • each device is also equipped with appropriate fluid interfaces, e.g., micropipettors, for introducing test compounds into the individual devices.
  • the devices may readily be attached to robotic systems which allow test compounds to be sampled from a number of multiwell plates that are moved along a conveyor system. Intervening spacer fluid regions can also be introduced via a spacer solution reservoir.
  • FIG. 12A shows a typical electrode design, in which electrode 1211 is partially submerged in reservoir 1215 fluidly connected to fluid channel 1217 .
  • FIG. 12B utilizes a salt bridge between electrode with frit 1219 and fluid reservoir 1221 fluidly connected to fluid channel 1223 .
  • FIG. 12C reduces degradation of chemical species by providing electrode 1225 submersed in first fluid reservoir 1227 fluidly connected to second fluid reservoir 1229 by large channel 1231 which limits diffusion, but has a low electroosmotic flow.
  • FIG. 12D provides a similar two part reservoir, in which electrode 1235 is submersed in first fluid reservoir 1237 fluidly connected to second fluid reservoir 1241 by small channel 1243 which is treated to reduce or eliminate electroosmotic flow.
  • FIG. 12E provides another similar two part reservoir, in which electrode 1245 is submersed in first fluid reservoir 1247 fluidly connected to second fluid reservoir 1251 by channel 1253 .
  • Channel 1253 is filled with a material such as gel, Agar, glass beads or other matrix material for reducing electroosmotic flow.
  • FIG. 12F provides a variant two part reservoir system, in which electrode 1255 is submersed in first fluid reservoir 1257 fluidly connected to second fluid reservoir 1259 by channel 1261 .
  • the fluid level in second fluid reservoir 1259 is higher than the fluid level in first fluid reservoir 1257 , which forces fluid towards electrode 1255 .
  • FIG. 12G provides a second variant two part reservoir, in which electrode 1265 is submersed in first fluid reservoir 1267 fluidly connected to second fluid reservoir 1269 by channel 1271 .
  • the diameter on first fluid reservoir 1267 is small enough that capillary forces draw fluid into first fluid reservoir 1267 .
  • microfluidic system containing at least a first substrate having a first channel and a second channel intersecting said first channel, at least one of said channels having at least one cross-sectional dimension in a range from 0.1 to 500 ⁇ m, in order to test the effect of each of a plurality of test compounds on a biochemical system.
  • the invention provides, inter alia, an apparatus for detecting an effect of a test compound on a biochemical system, comprising a substrate having at least one surface with a plurality of reaction channels fabricated into the surface.
  • Apparatus as hereinbefore described having at least two transverse channels fabricated into the surface, wherein each of the plurality of reaction channels is fluidly connected to a first of the at least two transverse channels at a first point in each of the reaction channels, and fluidly connected to a second transverse channel at a second point in each of the reaction channels and an assay apparatus including an apparatus as hereinbefore described are also provided.
  • the efficacy of performing an enzyme inhibition assay screen was demonstrated in a planar chip format.
  • a 6-port planar chip was employed having the layout shown in FIG. 8. The numbers adjacent the channels represent the lengths of each channel in millimeters.
  • Two voltage states were applied to the ports of the chip.
  • the first state (State 1) resulted in flowing of enzyme with buffer from the top buffer well into the main channel.
  • the second voltage state resulted in the interruption of the flow of buffer from the top well, and the introduction of inhibitor from the inhibitor well, into the main channel along with the enzyme.
  • a control experiment was also run in which buffer was placed into the inhibitor well.
  • Applied voltages at each port for each of the two applied voltage states were as follows: State 1 State 2 Top Buffer Well (I) 1831 1498 Inhibitor Well(II) 1498 1900 Enzyme Well (III) 1891 1891 Substrate Well (IV) 1442 1442 Bottom Buffer Well (V) 1442 1442 Detect./Waste Well (VI) 0 0
  • Enzyme ⁇ -Galactosidase (180 U/ml in 50 mM Tris/300 ⁇ g/ml BSA
  • Buffer 20 mM Tris, pH 8.5
  • Enzyme and substrate were continually pumped through the main channel from their respective ports under both voltage states. Inhibitor or Buffer were delivered into the main channel alternately from their respective wells by alternating between voltage state 1 and voltage state 2. When no inhibitor was present at the detection end of the main channel, a base line level of fluorescent product was produced. Upon introduction of inhibitor, the fluorescent signal was greatly reduced, indicating inhibition of the enzyme/substrate interaction. Fluorescent data obtained from the alternating delivery of inhibitor and buffer into the main channel is shown in FIG. 9A. FIG. 9B a superposition of the two data segments from FIG. 9A, directly comparing the inhibitor data with control (buffer) data. The control shows only a minor fluctuation in the fluorescent signal that apparently resulted from a dilution of the enzyme substrate mixture, whereas the inhibitor screen shows a substantial reduction in the fluorescent signal, indicating clear inhibition.
  • FIG. 10 A schematic of the chip to be used is shown in FIG. 10.
  • the chip has a reaction channel 5 cm in length which includes a 1 cm incubation zone and a 4 cm reaction zone.
  • the reservoir at the beginning of the sample channel is filled with enzyme solution and the side reservoir is filled with the fluorogenic substrate.
  • Each of the enzyme and substrate are diluted to provide for a steady state signal in the linear signal range for the assay system, at the detector.
  • Potentials are applied at each of the reservoirs (sample source, enzyme, substrate and waste) to achieve an applied field of 200 V/cm. This applied field produces a flow rate of 2 mm/second.
  • Subject material regions containing test compounds in 150 mM NaCl are introduced into the sample channel separated by first spacer regions of 150 mM NaCl and second spacer regions of 5 mM borate buffer. Once introduced into the sample channel shown, the subject material region requires 12 seconds to travel the length of the sample channel and reach the incubation zone of the reaction channel. This is a result of the flow rate of 2 mm/sec, allowing for 1 second for moving the sample pipettor from the sample to the spacer compounds. Allowing for these interruptions, the net flow rate is 0.68 mm/sec. Another 12 seconds is required for the enzyme/test compound mixture to travel through the incubation zone to the intersection with the substrate channel where substrate is continuously flowing into the reaction zone of the reaction channel.
  • Each subject material region containing the test compounds then requires 48 seconds to travel the length of the reaction zone and past the fluorescence detector.
  • a schematic of timing for subject material region/spacer region loading is shown in FIG. 11.
  • the top panel shows the subject material/first spacer region/second spacer region distribution within a channel, whereas the lower panel shows the timing required for loading the channel.
  • the schematic includes the loading (sipping) of high salt (HS) first spacer fluid (“A”), moving the pipettor to the sample or subject material (“B”), sipping the sample or subject material (“C”), moving the pipettor to the high salt first spacer fluid (“D”) sipping the first spacer fluid (“E”), moving the pipettor to the low salt (LS) or second spacer fluid (“F”), sipping the second spacer fluid (“G”) and returning to the first spacer fluid (“H”).
  • the process is then repeated for each additional test compound.
  • a constant base fluorescent signal is established at the detector in the absence of test compounds.
  • a decrease in fluorescence is seen similar to that shown in FIGS. 9A and 9B, which, based upon time delays, corresponds to a specific individual test compound.
  • This test compound is tentatively identified as an inhibitor of the enzyme, and further testing is conducted to confirm this and quantitate the efficacy of this inhibitor.

Abstract

The present invention provides novel microfluidic devices and methods that are useful for performing high-throughput screening assays. In particular, the devices and methods of the invention are useful in screening large numbers of different compounds for their effects on a variety of chemical, and preferably, biochemical systems.

Description

    CROSS REFERENCE TO RELATED APPLICATIONS
  • This is a continuation of U.S. patent application Ser. No. 09/718,236 filed Nov. 21, 2000, which is a continuation of U.S. patent application Ser. No. 08/881,696 filed Jun. 24, 1997, now U.S. Pat. No. 6,267,858, which is a continuation-in-part of U.S. patent application Ser. No. 08/671,987 filed Jun. 28, 1996, now U.S. Pat. No. 5,942,443 and U.S. patent application Ser. No. 08/761,575 filed Dec. 6, 1996, now U.S. Pat. No. 6,046,056, all of which are hereby incorporated herein by reference in their entirety for all purposes. PCT Application No. PCT/US97/10894, which designates the United States of America and is substantially identical to the present application, was co-filed in the United States Receiving Office on Jun. 24, 1997. This application is also incorporated herein by reference.[0001]
  • FIELD OF THE INVENTION
  • This application relates to apparatus and assay systems for detecting molecular interactions. The apparatus comprise a substrate with one or more intersecting channels and an electroosmotic fluid movement component, or other component for moving fluid in the channels on the substrate. [0002]
  • BACKGROUND OF THE INVENTION
  • There has long been a need for the ability to rapidly assay compounds for their effects on various biological processes. For example, enzymologists have long sought better substrates, better inhibitors or better catalysts for enzymatic reactions. Similarly, in the pharmaceutical industries, attention has been focused on identifying compounds that may block, reduce, or even enhance the interactions between biological molecules. Specifically, in biological systems the interaction between a receptor and its ligand often may result, either directly or through some downstream event, in either a deleterious or beneficial effect on that system, and consequently, on a patient for whom treatment is sought. Accordingly, researchers have long sought after compounds or mixtures of compounds that can reduce, block or even enhance that interaction. Similarly, the ability to rapidly process samples for detection of biological molecules relevant to diagnostic or forensic analysis is of fundamental value for, e.g., diagnostic medicine, archaeology, anthropology, and modern criminal investigation. [0003]
  • Modern drug discovery is limited by the throughput of the assays that are used to screen compounds that possess these described effects. In particular, screening of a maximum number of different compounds necessitates reducing the time and labor requirements associated with each screen. [0004]
  • High throughput screening of collections of chemically synthesized molecules and of natural products (such as microbial fermentation broths) has thus played a central role in the search for lead compounds for the development of new pharmacological agents. The remarkable surge of interest in combinatorial chemistry and the associated technologies for generating and evaluating molecular diversity represent significant milestones in the evolution of this paradigm of drug discovery. See Pavia et al., 1993, Bioorg. Med. Chem. Lett. 3:387-396, incorporated herein by reference. To date, peptide chemistry has been the principle vehicle for exploring the utility of combinatorial methods in ligand identification. See Jung & Beck-Sickinger, 1992, Angew. Chem. Int. Ed. Eng. 31: 367-383, incorporated herein by reference. This may be ascribed to the availability of a large and structurally diverse range of amino acid monomers, a relatively generic, high-yielding solid phase coupling chemistry and the synergy with biological approaches for generating recombinant peptide libraries. Moreover, the potent and specific biological activities of many low molecular weight peptides make these molecules attractive starting points for therapeutic drug discovery. See Hirschmann, 1991, Angew. Chem. Int. Ed. Engl. 30: 1278-1301, and Wiley & Rich, 1993, Med. Res. Rev. 13: 327-384, each of which is incorporated herein by reference. Unfavorable pharmacodynamic properties such as poor oral bioavailability and rapid clearance in vivo have limited the more widespread development of peptidic compounds as drugs, however. This realization has recently inspired workers to extend the concepts of combinatorial organic synthesis beyond peptide chemistry to create libraries of known pharmacophores like benzodiazepines (see Bunin & Elhman, 1992, J. Amer. Chem. Soc. 114: 10997-10998, incorporated herein by reference) as well as polymeric molecules such as oligomeric N-substituted glycines (“peptoids”) and oligocarbamates. See Simon et al., 1992, Proc. Natl. Acad. Sci. USA 89: 9367-9371; Zuckermrann et al., 1992, J. Amer. Chem. Soc. 114: 10646-10647; and Cho et al., 1993, Science 261:1303-1305, each of which is incorporated herein by reference. [0005]
  • In similar developments, much as modern combinatorial chemistry has resulted in a dramatic increase in the number of test compounds that may be screened, human genome research has also uncovered large numbers of new target molecules (e.g., genes and gene products such as proteins and RNA) against which the efficacy of test compounds are screened. [0006]
  • Despite the improvements achieved using parallel screening methods and other technological advances, such as robotics and high throughput detection systems, current screening methods still have a number of associated problems. For example, screening large numbers of samples using existing parallel screening methods have high space requirements to accommodate the samples and equipment, e.g., robotics, etc., high costs associated with that equipment, and high reagent requirements necessary for performing the assays. Additionally, in many cases, reaction volumes must be very small to account for the small amounts of the test compounds that are available. Such small volumes compound errors associated with fluid handling and measurement, e.g., due to evaporation, small dispensing errors, or the like. Additionally, fluid-handling equipment and methods have typically been unable to handle these volume ranges with any acceptable level of accuracy due in part to surface tension effects in such small volumes. [0007]
  • The development of systems to address these problems must consider a variety of aspects of the assay process. Such aspects include target and compound sources, test compound and target handling, specific assay requirements, and data acquisition, reduction storage and analysis. In particular, there exists a need for high throughput screening methods and associated equipment and devices that are capable of performing repeated, accurate assay screens, and operating at very small volumes. [0008]
  • The present invention meets these and a variety of other needs. In particular, the present invention provides novel methods and apparatuses for performing screening assays which address and provide meaningful solutions to these problems. [0009]
  • BRIEF SUMMARY OF THE INVENTION
  • The present invention provides methods of screening a plurality of test compounds for an effect on a biochemical system. These methods typically utilize microfabricated substrates which have at least a first surface, and at least two intersecting channels fabricated into that first surface. At least one of the intersecting channels will have at least one cross-sectional dimension in a range from 0.1 to 500 μm. The methods involve flowing a first component of a biochemical system in a first of the at least two intersecting channels. At least a first test compound is flowed from a second channel into the first channel whereby the test compound contacts the first component of the biochemical system. An effect of the test compound on the biochemical system is then detected. [0010]
  • In a related aspect, the method comprises continuously flowing the first component of a biochemical system in the first channel of the at least two intersecting channels. Different test compounds are periodically introduced into the first channel from a second channel. The effect, if any, of the test compound on the biochemical system is then detected. [0011]
  • In an alternative aspect, the methods utilize a substrate having at least a first surface with a plurality of reaction channels fabricated into the first surface. Each of the plurality of reaction channels is fluidly connected to at least two transverse channels also fabricated in the surface. The at least first component of a biochemical system is introduced into the plurality of reaction channels, and a plurality of different test compounds is flowed through at least one of the at least two transverse channels. Further, each of the plurality of test compounds is introduced into the transverse channel in a discrete volume. Each of the plurality of different test compounds is directed into a separate reaction channel and the effect of each of the test compounds on the biochemical system is then detected. [0012]
  • The present invention also provides apparatuses for practicing the above methods. In one aspect, the present invention provides an apparatus for screening test compounds for an effect on a biochemical system. The device comprises a substrate having at least one surface with at least two intersecting channels fabricated into the surface. The at least two intersecting channels have at least one cross-sectional dimension in the range from about 0.1 to about 500 μm. The device also comprises a source of different test compounds fluidly connected to a first of the at least two intersecting channels, and a source of at least one component of the biochemical system fluidly connected to a second of the at least two intersecting channels. Also included are fluid direction systems for flowing the at least one component within the intersecting channels, and for introducing the different test compounds from the first to the second of the intersecting channels. The apparatus also optionally comprises a detection zone in the second channel for detecting an effect of said test compound on said biochemical system. [0013]
  • In preferred aspects, the apparatus of the invention includes a fluid direction system which comprises at least three electrodes, each electrode being in electrical contact with the at least two intersecting channels on a different side of an intersection formed by the at least two intersecting channels. The fluid direction system also includes a control system for concomitantly applying a variable voltage at each of the electrodes, whereby movement of the test compounds or the at least first component in the at least two intersecting channels are controlled. [0014]
  • In another aspect, the present invention provides an apparatus for detecting an effect of a test compound on a biochemical system, comprising a substrate having at least one surface with a plurality of reaction channels fabricated into the surface. The apparatus also has at least two transverse channels fabricated into the surface, wherein each of the plurality of reaction channels is fluidly connected to a first of the at least two transverse channels at a first point in each of the reaction channels, and fluidly connected to a second transverse channel at a second point in each of the reaction channels. The apparatus further includes a source of at least one component of the biochemical system fluidly connected to each of the reaction channels, a source of test compounds fluidly connected to the first of the transverse channels, and a fluid direction system for controlling movement of the test compound and the first component within the transverse channels and the plurality of reaction channels. As above, the apparatuses also optionally include a detection zone in the second transverse channel for detecting an effect of the test compound on the biochemical system.[0015]
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 is a schematic illustration of one embodiment of a microlaboratory screening assay system of the present invention which can be used in running a continuous flow assay system. [0016]
  • FIGS. 2A and 2B show a schematic illustration of the apparatus shown in FIG. 1, operating in alternate assay systems. FIG. 2A shows a system used for screening effectors of an enzyme-substrate interaction. FIG. 2B illustrates the use of the apparatus in screening effectors of receptor-ligand interactions. [0017]
  • FIG. 3 is a schematic illustration of a “serial input parallel reaction” microlaboratory assay system in which compounds to be screened are serially introduced into the device but then screened in a parallel orientation within the device. [0018]
  • FIGS. [0019] 4A-4F show a schematic illustration of the operation of the device shown in FIG. 3, in screening a plurality of bead based test compounds.
  • FIG. 5 shows a schematic illustration of a continuous flow assay device incorporating a sample shunt for performing prolonged incubation followed by a separation step. [0020]
  • FIG. 6A shows a schematic illustration of a serial input parallel reaction device for use with fluid based test compounds. [0021]
  • FIGS. 6B and 6C show a schematic illustration of fluid flow patterns within the device shown in FIG. 6A. [0022]
  • FIG. 7 shows a schematic illustration of one embodiment of an overall assay systems which employs multiple microlaboratory devices labeled as “LabChips™” for screening test compounds. [0023]
  • FIG. 8 is a schematic illustration of a chip layout used for a continuous-flow assay screening system. [0024]
  • FIGS. 9A and 9B shows fluorescence data from a continuous flow assay screen. FIG. 9A shows fluorescence data from a test screen which periodically introduced a known inhibitor (IPTG) into a β-galactosidase assay system in a chip format. FIG. 9B shows a superposition of two data segments from FIG. 9A, directly comparing the inhibitor data with control (buffer) data. [0025]
  • FIG. 10 illustrates the operating parameters of a fluid flow system on a small chip device for performing enzyme inhibitor screening. [0026]
  • FIG. 11 shows a schematic illustration of timing for sample/spacer loading in a microfluidic device channel. [0027]
  • FIG. 12, panels A-G schematically illustrate electrodes used in apparatuses of the invention. [0028]
  • DETAILED DESCRIPTION
  • I. Applications for the Invention [0029]
  • The present invention provides novel microlaboratory systems and methods that are useful for performing high-throughput screening assays. In particular, the present invention provides microfluidic devices and methods of using such devices in screening large numbers of different compounds for their effects on a variety of chemical, and preferably, biochemical systems. [0030]
  • As used herein, the phrase “biochemical system” generally refers to a chemical interaction that involves molecules of the type generally found within living organisms. Such interactions include the full range of catabolic and anabolic reactions which occur in living systems including enzymatic, binding, signaling and other reactions. Further, biochemical systems, as defined herein, also include model systems which are mimetic of a particular biochemical interaction. Examples of biochemical systems of particular interest in practicing the present invention include, e.g., receptor-ligand interactions, enzyme-substrate interactions, cellular signaling pathways, transport reactions involving model barrier systems (e.g., cells or membrane fractions) for bioavailability screening, and a variety of other general systems. Cellular or organismal viability or activity may also be screened using the methods and apparatuses of the present invention, e.g., in toxicology studies. Biological materials which are assayed include, but are not limited to, cells, cellular fractions (membranes, cytosol preparations, etc.), agonists and antagonists of cell membrane receptors (e.g., cell receptor-ligand interactions such as e.g., transferring, c-kit, viral receptor ligands (e.g., CD4-HIV), cytokine receptors, chemokine receptors, interleukin receptors, immunoglobulin receptors and antibodies, the cadherein family, the integrin family, the selectin family, and the like; see, e.g., Pigott and Power (1993) The Adhesion Molecule FactsBook Academic Press New York and Hulme (ed) Receptor Ligand Interactions A Practical Approach Rickwood and Hames (series editors) IRL Press at Oxford Press NY), toxins and venoms, viral epitopes, hormones (e.g., opiates, steroids, etc.), intracellular receptors (e.g. which mediate the effects of various small ligands, including steroids, thyroid hormone, retinoids and vitamin D; for reviews see, e.g., Evans (1988) Science, 240:889-895; Ham and Parker (1989) Curr. Opin. Cell Biol., 1:503-511; Burustein et al. (1989), Ann. Rev. Physiol., 51:683-699; Truss and Beato (1993) Endocr. Rev., 14:459-479), peptides, retro-inverso peptides, polymers of α-, β-, or ω-amino acids (D- or L-), enzymes, enzyme substrates, cofactors, drugs, lectins, sugars, nucleic acids (both linear and cyclic polymer configurations), oligosaccharides, proteins, phospholipids and antibodies. Synthetic polymers such as heteropolymers in which a known drug is covalently bound to any of the above, such as polyurethanes, polyesters, polycarbonates, polyureas, polyamides, polyethyleneimines, polyarylene sulfides, polysiloxanes, polyimides, and polyacetates are also assayed. Other polymers are also assayed using the systems described herein, as would be apparent to one of skill upon review of this disclosure. One of skill will be generally familiar with the biological literature. For a general introduction to biological systems, see, Berger and Kimmel, Guide to Molecular Cloning Techniques, Methods in [0031] Enzymology volume 152 Academic Press, Inc., San Diego, Calif. (Berger); Sambrook et al. (1989) Molecular Cloning—A Laboratory Manual (2nd ed.) Vol. 1-3, Cold Spring Harbor Laboratory, Cold Spring Harbor Press, NY, (Sambrook); Current Protocols in Molecular Biology, F. M. Ausubel et al., eds., Current Protocols, a joint venture between Greene Publishing Associates, Inc. and John Wiley & Sons, Inc., (through 1997 Supplement) (Ausubel); Watson et al. (1987) Molecular Biology of the Gene, Fourth Edition The Benjamin/Cummings Publishing Co., Menlo Park, Calif.; Watson et al. (1992) Recombinant DNA Second Edition Scientific American Books, NY; Alberts et al. (1989) Molecular Biology of the Cell Second Edition Garland Publishing, NY; Pattison (1994) Principles and Practice of Clinical Virology; Darnell et al., (1990) Molecular Cell Biology second edition, Scientific American Books, W. H. Freeman and Company; Berkow (ed.) The Merck Manual of Diagnosis and Therapy, Merck & Co., Rahway, N.J.; Harrison's Principles of Internal Medicine, Thirteenth Edition, Isselbacher et al. (eds). (1994) Lewin Genes, 5th Ed., Oxford University Press (1994); The “Practical Approach” Series of Books (Rickwood and Hames (series eds.) by IRL Press at Oxford University Press, NY; The “FactsBook Series” of books from Academic Press, NY,; Product information from manufacturers of biological reagents and experimental equipment also provide information useful in assaying biological systems. Such manufacturers include, e.g., the SIGMA chemical company (Saint Louis, Mo.), R&D systems (Minneapolis, Minn.), Pharmacia LKB Biotechnology (Piscataway, N.J.), CLONTECH Laboratories, Inc. (Palo Alto, Calif.), Chem Genes Corp., Aldrich Chemical Company (Milwaukee, Wis.), Glen Research, Inc., GIBCO BRL Life Technologies, Inc. (Gaithersberg, Md.), Fluka Chemica-Biochemika Analytika (Fluka Chemie AG, Buchs, Switzerland), Invitrogen, San Diego, Calif., and Applied Biosystems (Foster City, Calif.), as well as many other commercial sources known to one of skill.
  • In order to provide methods and devices for screening compounds for effects on biochemical systems, the present invention generally incorporates model in vitro systems which mimic a given biochemical system in vivo for which effector compounds are desired. The range of systems against which compounds can be screened and for which effector compounds are desired, is extensive. For example, compounds are optionally screened for effects in blocking, slowing or otherwise inhibiting key events associated with biochemical systems whose effect is undesirable. For example, test compounds are optionally screened for their ability to block systems that are responsible, at least in part, for the onset of disease or for the occurrence of particular symptoms of diseases, including, e.g., hereditary diseases, cancer, bacterial or viral infections and the like. Compounds which show promising results in these screening assay methods can then be subjected to further testing to identify effective pharmacological agents for the treatment of disease or symptoms of a disease. [0032]
  • Alternatively, compounds can be screened for their ability to stimulate, enhance or otherwise induce biochemical systems whose function is believed to be desirable, e.g., to remedy existing deficiencies in a patient. [0033]
  • Once a model system is selected, batteries of test compounds can then be applied against these model systems. By identifying those test compounds that have an effect on the particular biochemical system, in vitro, one can identify potential effectors of that system, in vivo. [0034]
  • In their simplest forms, the biochemical system models employed in the methods and apparatuses of the present invention will screen for an effect of a test compound on an interaction between two components of a biochemical system, e.g., receptor-ligand interaction, enzyme-substrate interaction, and the like. In this form, the biochemical system model will typically include the two normally interacting components of the system for which an effector is sought, e.g., the receptor and its ligand or the enzyme and its substrate. [0035]
  • Determining whether a test compound has an effect on this interaction then involves contacting the system with the test compound and assaying for the functioning of the system, e.g., receptor-ligand binding or substrate turnover. The assayed function is then compared to a control, e.g., the same reaction in the absence of the test compound or in the presence of a known effector. Typically, such assays involve the measurement of a parameter of the biochemical system. By “parameter of the biochemical system” is meant some measurable evidence of the system's functioning, e.g., the presence or absence of a labeled group or a change in molecular weight (e.g., in binding reactions, transport screens), the presence or absence of a reaction product or substrate (in substrate turnover measurements), or an alteration in electrophoretic mobility (typically detected by a change in elution time of a labeled compound). [0036]
  • Although described in terms of two-component biochemical systems, the methods and apparatuses may also be used to screen for effectors of much more complex systems, where the result or end product of the system is known and assayable at some level, e.g., enzymatic pathways, cell signaling pathways and the like. Alternatively, the methods and apparatuses described herein are optionally used to screen for compounds that interact with a single component of a biochemical system, e.g., compounds that specifically bind to a particular biochemical compound, e.g., a receptor, ligand, enzyme, nucleic acid, structural macromolecule, etc. [0037]
  • Biochemical system models may also be embodied in whole cell systems. For example, where one is seeking to screen test compounds for an effect on a cellular response, whole cells are optionally utilized. Modified cell systems may also be employed in the screening systems encompassed herein. For example, chimeric reporter systems are optionally employed as indicators of an effect of a test compound on a particular biochemical system. Chimeric reporter systems typically incorporate a heterogenous reporter system integrated into a signaling pathway which signals the binding of a receptor to its ligand. For example, a receptor is fused to a heterologous protein, e.g., an enzyme whose activity is readily assayable. Activation of the receptor by ligand binding then activates the heterologous protein which then allows for detection. Thus, the surrogate reporter system produces an event or signal which is readily detectable, thereby providing an assay for receptor/ligand binding. Examples of such chimeric reporter systems have been previously described in the art. [0038]
  • Additionally, where one is screening for bioavailability, e.g., transport, biological barriers are optionally included. The term “biological barriers” generally refers to cellular or membranous layers within biological systems, or synthetic models thereof. Examples of such biological barriers include the epithelial and endothelial layers, e.g. vascular endothelia and the like. [0039]
  • Biological responses are often triggered and/or controlled by the binding of a receptor to its ligand. For example, interaction of growth factors, i.e., EGF epidermal growth actor, FGF fibroblast growth factor, PDGF platelet derived growth factor, etc., with their receptors stimulates a wide variety of biological responses including, e.g., cell proliferation and differentiation, activation of mediating enzymes, stimulation of messenger turnover, alterations in ion fluxes, activation of enzymes, changes in cell shape and the alteration in genetic expression levels. Accordingly, control of the interaction of the receptor and its ligand may offer control of the biological responses caused by that interaction. [0040]
  • Accordingly, in one aspect, the present invention will be useful in screening for compounds that affect an interaction between a receptor molecule and its ligands. As used herein, the term “receptor” generally refers to one member of a pair of compounds which specifically recognize and bind to each other. The other member of the pair is termed a “ligand.” Thus, a receptor/ligand pair may include a typical protein receptor, usually membrane associated, and its natural ligand, e.g., another protein or small molecule. Receptor/ligand pairs may also include antibody/antigen binding pairs, complementary nucleic acids, nucleic acid associating proteins and their nucleic acid ligands. A large number of specifically associating biochemical compounds are well known in the art and can be utilized in practicing the present invention. [0041]
  • Traditionally, methods for screening for effectors of a receptor/ligand interaction have involved incubating a receptor/ligand binding pair in the presence of a test compound. The level of binding of the receptor/ligand pair is then compared to negative and/or positive controls. Where a decrease in normal binding is seen, the test compound is determined to be an inhibitor of the receptor/ligand binding. Where an increase in that binding is seen, the test compound is determined to be an enhancer or inducer of the interaction. [0042]
  • In the interest of efficiency, screening assays have typically been set up in multiweli reaction plates, e.g., multi-well microplates, which allow for the simultaneous, parallel screening of large numbers of test compounds. [0043]
  • A similar, and perhaps overlapping, set of biochemical systems includes the interactions between enzymes and their substrates. The term “enzyme” as used herein, generally refers to a protein which acts as a catalyst to induce a chemical change in other compounds or “substrates.”[0044]
  • Typically, effectors of an enzyme's activity toward its substrate are screened by contacting the enzyme with a substrate in the presence and absence of the compound to be screened and under conditions optimal for detecting changes in the enzyme's activity. After a set time for reaction, the mixture is assayed for the presence of reaction products or a decrease in the amount of substrate. The amount of substrate that has been catalyzed is them compared to a control, i.e., enzyme contacted with substrate in the absence of test compound or presence of a known effector. As above, a compound that reduces the enzymes activity toward its substrate is termed an “inhibitor,” whereas a compound that accentuates that activity is termed an “inducer.”[0045]
  • Generally, the various screening methods encompassed by the present invention involve the serial introduction of a plurality of test compounds into a microfluidic device. Once injected into the device, the test compound is screened for effect on a biological system using a continuous serial or parallel assay orientation. [0046]
  • As used herein, the term “test compound” refers to the collection of compounds that are to be screened for their ability to affect a particular biochemical system. Test compounds may include a wide variety of different compounds, including chemical compounds, mixtures of chemical compounds, e.g., polysaccharides, small organic or inorganic molecules, biological macromolecules, e.g., peptides, proteins, nucleic acids, or an extract made from biological materials such as bacteria, plants, fungi, or animal cells or tissues, naturally occurring or synthetic compositions. Depending upon the particular embodiment being practiced, the test compounds are provided, e.g., injected, free in solution, or are optionally attached to a carrier, or a solid support, e.g., beads. A number of suitable solid supports are employed for immobilization of the test compounds. Examples of suitable solid supports include agarose, cellulose, dextran (commercially available as, i.e., Sephadex, Sepharose) carboxymethyl cellulose, polystyrene, polyethylene glycol (PEG), filter paper, nitrocellulose, ion exchange resins, plastic films, glass beads, polyaminemethylvinylether maleic acid copolymer, amino acid copolymer, ethylene-maleic acid copolymer, nylon, silk, etc. Additionally, for the methods and apparatuses described herein, test compounds are screened individually, or in groups. Group screening is particularly useful where hit rates for effective test compounds are expected to be low such that one would not expect more than one positive result for a given group. Alternatively, such group screening is used where the effects of different test compounds are differentially detected in a single system, e.g., through electrophoretic separation of the effects, or differential labelling which enables separate detection. [0047]
  • Test compounds are commercially available, or derived from any of a variety of biological sources apparent to one of skill and as described, supra. In one aspect, a tissue homogenate or blood sample from a patient is tested in the assay systems of the invention. For example, in one aspect, blood is tested for the presence or activity of a biologically relevant molecule. For example, the presence and activity level of an enzyme are detected by supplying and enzyme substrate to the biological sample and detecting the formation of a product using an assay systems of the invention. Similarly, the presence of infectious pathogens (viruses, bacteria, fungi, or the like) or cancerous tumors can be tested by monitoring binding of a labeled ligand to the pathogen or tumor cells, or a component of the pathogen or tumor such as a protein, cell membrane, cell extract or the like, or alternatively, by monitoring the presence of an antibody against the pathogen or tumor in the patient's blood. For example, the binding of an antibody from a patient's blood to a viral protein such as an HIV protein is a common test for monitoring patient exposure to the virus. Many assays for detecting pathogen infection are well known, and are adapted to the assay systems of the present invention. [0048]
  • Biological samples are derived from patients using well known techniques such as venipuncture or tissue biopsy. Where the biological material is derived from non-human animals, such as commercially relevant livestock, blood and tissue samples are conveniently obtained from livestock processing plants. Similarly, plant material used in the assays of the invention are conveniently derived from agricultural or horticultural sources. Alternatively, a biological sample can be from a cell or blood bank where tissue and/or blood are stored, or from an in vitro source such as a culture of cells. Techniques and methods for establishing a culture of cells for use as a source for biological materials are well known to those of skill in the art. Freshney Culture of Animal Cells, a Manual of Basic Technique, Third Edition Wiley-Liss, New York (1994) provides a general introduction to cell culture. [0049]
  • II. Assay Systems [0050]
  • As described above, the screening methods of the present invention are generally carried out in microfluidic devices or “microlaboratory systems,” which allow for integration of the elements required for performing the assay, automation, and minimal environmental effects on the assay system, e.g., evaporation, contamination, human error, or the like. A number of devices for carrying out the assay methods of the invention are described in substantial detail below. However, it will be recognized that the specific configuration of these devices will generally vary depending upon the type of assay and/or assay orientation desired. For example, in some embodiments, the screening methods of the invention can be carried out using a microfluidic device having two intersecting channels. For more complex assays or assay orientations, multichannel/intersection devices are optionally employed. The small scale, integratability and self-contained nature of these devices allows for virtually any assay orientation to be realized within the context of the microlaboratory system. [0051]
  • A. Electrokinetic Material Transport [0052]
  • In preferred aspects, the devices, methods and systems described herein, employ electrokinetic material transport systems, and preferably, controlled electrokinetic material transport systems. As used herein, “electrokinetic material transport systems” include systems which transport and direct materials within an interconnected channel and/or chamber containing structure, through the application of electrical fields to the materials, thereby causing material movement through and among the channel and/or chambers, i.e., cations will move toward the negative electrode, while anions will move toward the positive electrode. [0053]
  • Such electrokinetic material transport and direction systems include those systems that rely upon the electrophoretic mobility of charged species within the electric field applied to the structure. Such systems are more particularly referred to as electrophoretic material transport systems. Other electrolinetic material direction and transport systems rely upon the electroosmotic flow of fluid and material within a channel or chamber structure which results from the application of an electric field across such structures. In brief, when a fluid is placed into a channel which has a surface bearing charged functional groups, e.g., hydroxyl groups in etched glass channels or glass microcapillaries, those groups can ionize. In the case of hydroxyl functional groups, this ionization, e.g., at neutral pH, results in the release of protons from the surface and into the fluid, creating a concentration of protons at near the fluid/surface interface, or a positively charged sheath surrounding the bulk fluid in the channel. Application of a voltage gradient across the length of the channel, will cause the proton sheath, as well as the fluid it surrounds, to move in the direction of the voltage drop, i.e., toward the negative electrode. [0054]
  • “Controlled electrokinetic material transport and direction,” as used herein, refers to electrokinetic systems as described above, which employ active control of the voltages applied at multiple, i.e., more than two, electrodes. Rephrased, such controlled electrokinetic systems concomitantly regulate voltage gradients applied across at least two intersecting channels. Controlled electrolinetic material transport is described in Published PCT Application No. WO 96/04547, to Ramsey, which is incorporated herein by reference in its entirety for all purposes. In particular, the preferred microfluidic devices and systems described herein, include a body structure which includes at least two intersecting channels or fluid conduits, e.g., interconnected, enclosed chambers, which channels include at least three unintersected termini. The intersection of two channels refers to a point at which two or more channels are in fluid communication with each other, and encompasses “T” intersections, cross intersections, “wagon wheel” intersections of multiple channels, or any other channel geometry where two or more channels are in such fluid communication. An unintersected terminus of a channel is a point at which a channel terminates not as a result of that channel's intersection with another channel, e.g., a “T” intersection. In preferred aspects, the devices will include at least three intersecting channels having at least four unintersected termini. In a basic cross channel structure, where a single horizontal channel is intersected and crossed by a single vertical channel, controlled electrokinetic material transport operates to controllably direct material flow through the intersection, by providing constraining flows from the other channels at the intersection. For example, assuming one was desirous of transporting a first material through the horizontal channel, e.g., from left to right, across the intersection with the vertical channel. Simple electrokinetic material flow of this material across the intersection could be accomplished by applying a voltage gradient across the length of the horizontal channel, i.e., applying a first voltage to the left terminus of this channel, and a second, lower voltage to the right terminus of this channel, or by allowing the right terminus to float (applying no voltage). However, this type of material flow through the intersection would result in a substantial amount of diffusion at the intersection, resulting from both the natural diffusive properties of the material being transported in the medium used, as well as convective effects at the intersection. [0055]
  • In controlled electrokinetic material trasort, the material being transported across the intersection is constrained by low level flow from the side channels, e.g., the top and bottom channels. This is accomplished by applying a slight voltage gradient along the path of material flow, e.g., from the top or bottom termini of the vertical channel, toward the right terminus. The result is a “pinching” of the material flow at the intersection, which prevents the diffusion of the material into the vertical channel. The pinched volume of material at the intersection may then be injected into the vertical channel by applying a voltage gradient across the length of the vertical channel, i.e., from the top terminus to the bottom terminus. In order to avoid any bleeding over of material from the horizontal channel during this injection, a low level of flow is directed back into the side channels, resulting in a “pull back” of the material from the intersection. [0056]
  • In addition to pinched injection schemes, controlled electrokinetic material transport is readily utilized to create virtual valves which include no mechanical or moving parts. Specifically, with reference to the cross intersection described above, flow of material from one channel segment to another, e.g., the left arm to the right arm of the horizontal channel, can be efficiently regulated, stopped and reinitiated, by a controlled flow from the vertical channel, e.g., from the bottom arm to the top arm of the vertical channel. Specifically, in the ‘off’ mode, the material is transported from the left arm, through the intersection and into the top arm by applying a voltage gradient across the left and top termini. A constraining flow is directed from the bottom arm to the top arm by applying a similar voltage gradient along this path (from the bottom terminus to the top terminus). Metered amounts of material are then dispensed from the left arm into the right arm of the horizontal channel by switching the applied voltage gradient from left to top, to left to right. The amount of time and the voltage gradient applied dictates the amount of material that will be dispensed in this manner. Although described for the purposes of illustration with respect to a four way, cross intersection, these controlled electrolinetic material transport systems can be readily adapted for more complex interconnected channel networks, e.g., arrays of interconnected parallel channels. [0057]
  • B. Continuous Flow Assay Systems [0058]
  • In one preferred aspect, the methods and apparatuses of the invention are used in screening test compounds using a continuous flow assay system. Generally, the continuous flow assay system can be readily used in screening for inhibitors or inducers of enzymatic activity, or for agonists or antagonists of receptor-ligand binding. In brief, the continuous flow assay system involves the continuous flow of the particular biochemical system along a microfabricated channel. As used herein, the term “continuous” generally refers to an unbroken or contiguous stream of the particular composition that is being continuously flowed. For example, a continuous flow may include a constant fluid flow having a set velocity, or alternatively, a fluid flow which includes pauses in the flow rate of the overall system, such that the pause does not otherwise interrupt the flow stream. The functioning of the system is indicated by the production of a detectable event or signal. In one preferred embodiment, such detectable signals include optically detectable chromophoric or fluorescent signals that are associated with the functioning of the particular model system used. For enzyme systems, such signals will generally be produced by products of the enzyme's catalytic action, e.g., on a chromogenic or fluorogenic substrate. For binding systems, e.g., receptor ligand interactions, signals will typically involve the association of a labeled ligand with the receptor, or vice versa. [0059]
  • A wide variety of other detectable signals and labels can also be used in the assays and apparatuses of the invention. In addition to the chromogenic and fluorogenic labels described above, radioactive decay, electron density, changes in pH, solvent viscosity, temperature and salt concentration are also conveniently measured. [0060]
  • More generally, labels are commonly detectable by spectroscopic, photochemical, biochemical, immunochemical, or chemical means. For example, useful nucleic acid labels include 32P, 35S, fluorescent dyes, electron-dense reagents, enzymes (e.g., as commonly used in an ELISA), biotin, dioxigenin, or haptens and proteins for which antisera or monoclonal antibodies are available. A wide variety of labels suitable for labeling biological components are known and are reported extensively in both the scientific and patent literature, and are generally applicable to the present invention for the labeling of biological components. Suitable labels include radionucleotides, enzymes, substrates, cofactors, inhibitors, fluorescent moieties, chemiluminiescent moieties, magnetic particles, and the like. Labeling agents optionally include e.g., monoclonal antibodies, polyclonal antibodies, proteins, or other polymers such as affinity matrices, carbohydrates or lipids. Detection proceeds by any of a variety of known methods, including spectrophotometric or optical tracking of radioactive or fluorescent markers, or other methods which track a molecule based upon size, charge or affinity. A detectable moiety can be of any material having a detectable physical or chemical property. Such detectable labels have been well-developed in the field of gel electrophoresis, column chromatograpy, solid substrates, spectroscopic techniques, and the like, and in general, labels useful in such methods can be applied to the present invention. Thus, a label is any composition detectable by spectroscopic, photochemical, biochemical, immunochemical, electrical, optical thermal, or chemical means. Useful labels in the present invention include fluorescent dyes (e.g., fluorescein isothiocyanate, Texas red, rhodamine, and the like), radiolabels (e.g., 3H, 125I, 35S, 14C, 32P or 33P), enzymes (e.g., LacZ, CAT, horse radish peroxidase, alkaline phosphatase and others, commonly used as detectable enzymes, either as marker products or as in an EUISA), nucleic acid intercalators (e.g., ethidium bromide) and colorimetric labels such as colloidal gold or colored glass or plastic (e.g. polystyrene, polypropylene, latex, etc.) beads. [0061]
  • Fluorescent labels are particularly preferred labels. Preferred labels are typically characterized by one or more of the following: high sensitivity, high stability, low background, low environmental sensitivity and high specificity in labeling. [0062]
  • Fluorescent moieties, which are incorporated into the labels of the invention, are generally are known, including 1- and 2-amironaphthalene, p,p′-diaminostilbenes, pyrenes, quaternary phenanthridine salts, 9-aminoacridines, p,p′-diaminobenzophenone imines, anthracenes, oxacarbocyanine, merocyanine, 3-aminoequilenin, perylene, bis-benzoxazole, bis-p-oxazolyl benzene, 1,2-benzophenazin, retinol, bis-3-aminopyridiniuim salts, hellebrigenin, tetracycline, sterophenol, benzimidazolylphenylamine, 2-oxo-3-chromen, indole, xanthen, 7-hydroxycoumarin, phenoxazine, calicylate, strophanthidin, porphyrins, triarylmethanes and flavin. Individual fluorescent compounds which have functionalities for linking to an element desirably detected in an apparatus or assay of the invention, or which can be modified to incorporate such functionalities include, e.g., dansyl chloride; fluoresceins such as 3,6-dihydroxy-9-phenylxanthhydrol; rhodamineisothiocyanate; N-phenyl 1-amino-8—sulfonatonaphthalene; N-phenyl 2-amino-6-sulfonatonaphthalene; 4-acetamido-4-isothiocyanato-stilbene-2,2′-disulfonic acid; pyrene-3-sulfonic acid; 2-toluidinonaphthalene6-sulfonate; N-phenyl-N-methyl-2-aminoaphthalene-6-sulfonate; ethidim bromide; stebrine; auromine-0,2-(9′-anthroyl)palmitate; dansyl phosphatidylethanolamine; N,N′-dioctadecyl oxacarbocyanine: N,N′-dihexyl oxacarbocyanine; merocyanine, 4-(3′pyrenyl)stearate; d-3-aminodesoxy-equilenin; 12-(9′-anthroyl)stearate; 2-methylanthracene; 9-vinylanracene; 2,2′(vinylene-p-phenylene)bisbenzoxazole; p-bis(2-(4-methyl-5-phenyl-oxazolyl))benzene; 6-dimethylamino-1,2-benzophenazin; retinol; bis(3′-aminopyridinium) 1,10-decandiyl diiodide; sulfonaphthylhydrazone of hellibrienin; chlorotetracycline; N-(7-dimethylamino4-methyl-2-oxo-3-chromenyl)maleinide; N-(p-(2-benzimidazolyl)-phenyl)maleimide; N-(4-fluoranthyl)maleimide; bis(homovanillic acid); resazarin; 4-chloro-7-nitro-2,1,3-benzooxadiazole; merocyanine 540; resorufm; rose bengal; and 2,4-diphenyl-3(2H)-furanone. Many fluorescent tags are commercially available from SIGMA chemical company (Saint Louis, Mo.), Molecular Probes, R&D systems (Minneapolis, Minn.), Pharmacia LKB Biotechnology (Piscataway, N.J.), CLONTECH Laboratories, Inc. (Palo Alto, Calif.), Chem Genes Corp., Aldrich Chemical Company (Milwaukee, Wis.), Glen Research, Inc., GIBCO BRL Life Technologies, Inc. (Gaithersberg, Md.), Fluka Chemica-Biochemika Analytika (Fluka Chemie AG, Buchs, Switzerland), and Applied Biosystems (Foster City, Calif.) as well as other commercial sources known to one of skill. [0063]
  • Desirably, fluorescent labels absorb light above about 300 nm, preferably about 350 nm, and more preferably above about 400 nm, usually emitting at wavelengths greater than about 10 nm higher than the wavelength of the light absorbed. It should be noted that the absorption and emission characteristics of the bound label may differ from the unbound label. Therefore, when referring to the various wavelength ranges and characteristics of the labels, it is intended to indicate the labels as employed and not the label which is unconjugated and characterized in an arbitrary solvent. [0064]
  • Fluorescent labels are one preferred class of detectable labels, in part because by irradiating a fluorescent label with light, one can obtain a plurality of emissions. Thus, a single label can provide for a plurality of measurable events. Detectable signal may also be provided by chemiluminescent and bioluminescent sources. Chemiluminescent sources include a compound which becomes electronically excited by a chemical reaction and may then emit light which serves as the detectible signal or donates energy to a fluorescent acceptor. A diverse number of families of compounds have been found to provide chemiluminescence under a variety or conditions. One family of compounds is 2,3-dihydro-1,4—phthalazinedione. The most popular compound is luminol, which is a 5-amino compound. Other members of the family include the 5-amino-6,7,8-trimethoxy- and the dimethylamino[ca]benz analog. These compounds can be made to luminesce with alkaline hydrogen peroxide or calcium hypochlorite and base. Another family of compounds is the 2,4,5-triphenylimidazoles, with lophine as the common name for the parent product. Chemiluminescent analogs include para-dimethylamino and -methoxy substituents. Chemiluminescence may also be obtained with oxalates, usually oxalyl active esters, e.g., p-nitrophenyl and a peroxide, e.g., hydrogen peroxide, under basic conditions. Other useful chemiluminescent compounds are also known and available, including —N-alkyl acridimim esters (basic H[0065] 2O2) and dioxetanes. Alternatively, luciferins may be used in conjunction with luciferase or lucigenins to provide bioluminescence.
  • The label is coupled directly or indirectly to a molecule to be detected (a product, substrate, enzyme, or the like) according to methods well known in the art. As indicated above, a wide variety of labels are used, with the choice of label depending on the sensitivity required, ease of conjugation of the compound, stability requirements, available instrumentation, and disposal provisions. Non radioactive labels are often attached by indirect means. Generally, a ligand molecule (e.g., biotin) is covalently bound to a polymer. The ligand then binds to an anti-ligand (e.g., streptavidin) molecule which is either inherently detectable or covalently bound to a signal system, such as a detectable enzyme, a fluorescent compound, or a chemiluminescent compound. A number of ligands and anti-ligands can be used. Where a ligand has a natural anti-ligand, for example, biotin, thyroxine, and cortisol, it can be used in conjunction with labeled, anti-ligands. Alternatively, any haptenic or antigenic compound can be used in combination with an antibody. Labels can also be conjugated directly to signal generating compounds, e.g., by conjugation with an enzyme or fluorophore. Enzymes of interest as labels will primarily be hydrolases, particularly phosphatases, esterases and glycosidases, or oxidoreductases, particularly peroxidases. Fluorescent compounds include fluorescein and its derivatives, rhodamine and its derivatives, dansyl, umbelliferone, etc. Chemiluminescent compounds include luciferin, and 2,3-dihydrophthalazinediones, e.g., luminol. Means of detecting labels are well known to those of skill in the art. Thus, for example, where the label is a radioactive label, means for detection include a scintillation counter or photographic film as in autoradiography. Where the label is a fluorescent label, it may be detected by exciting the fluorochrome with the appropriate wavelength of light and detecting the resulting fluorescence, e.g., by microscopy, visual inspection, via photographic film, by the use of electronic detectors such as digital cameras, charge coupled devices (CCDs) or photomultipliers and phototubes, and the like. Fluorescent labels and detection techniques, particularly microscopy and spectroscopy are preferred. Similarly, enzymatic labels are detected by providing appropriate substrates for the enzyme and detecting the resulting reaction product. Finally, simple colorimetric labels are often detected simply by observing the color associated with the label. For example, conjugated gold often appears pink, while various conjugated beads appear the color of the bead. [0066]
  • In preferred aspects, the continuous system generates a constant signal which varies only when a test compound is introduced that affects the system. Specifically, as the system components flow along the channel, they will produce a relatively constant signal level at a detection zone or window of the channel. Test compounds are periodically introduced into the channel and mixed with the system components. Where those test compounds have an effect on the system, it will cause a deviation from the constant signal level at the detection window. This deviation may then be correlated to the particular test compound screened. [0067]
  • One embodiment of a device for use in a serial or continuous assay geometry is shown in FIG. 1. As shown, the overall device [0068] 100 is fabricated in a planar substrate 102. Suitable substrate materials are generally selected based upon their compatibility with the conditions present in the particular operation to be performed by the device. Such conditions can include extremes of pH, temperature, salt concentration, and application of electrical fields. Additionally, substrate materials are also selected for their inertness to critical components of an analysis or synthesis to be carried out by the device.
  • Examples of useful substrate materials include, e.g., glass, quartz and silicon as well as polymeric substrates, e.g. plastics. In the case of conductive or semi-conductive substrates, it will generally be desirable to include an insulating layer on the substrate. This is particularly important where the device incorporates electrical elements, e.g., electrical material and fluid direction systems, sensors and the like. In the case of polymeric substrates, the substrate materials are optionally rigid, semi-rigid, or non-rigid, opaque, semi-opaque or transparent, depending upon the use for which they are intended. For example, devices which include an optical or visual detection element, will generally be fabricated, at least in part, from transparent materials to allow, or at least, facilitate that detection. Alternatively, transparent windows of, e.g., glass or quartz, are optionally incorporated into the device for these types detection elements. Additionally, the polymeric materials may have linear or branched backbones, and are optionally crosslinked or non-crosslinked. Examples of particularly preferred polymeric materials include, e.g., polydimethylsiloxanes (PDMS), polyurethane, polyvinylchloride (PVC) polystyrene, polysulfone, polycarbonate and the like. [0069]
  • The device shown in FIG. 1 includes a series of [0070] channels 110, 112, and optional reagent channel 114, fabricated into the surface of the substrate. At least one of these channels will typically have very small cross sectional dimensions, e.g., in the range of from about 0.1 μm to about 500 μm. Preferably the cross-sectional dimensions of the channels will be in the range of from about 0.1 to about 200 μm and more preferably in the range of from about 0.1 to about 100 μm. In particularly preferred aspects, each of the channels will have at least one cross-sectional dimension in the range of from about 0.1 μm to about 100 μm. Although generally shown as straight channels, it will be appreciated that in order to maximize the use of space on a substrate, serpentine, saw tooth or other channel geometries, to incorporate effectively longer channels in shorter distances.
  • Manufacturing of these microscale elements into the surface of the substrates may generally be carried out by any number of microfabrication techniques that are well known in the art. For example, lithographic techniques are optionally employed in fabricating, e.g., glass, quartz or silicon substrates, using methods well known in the semiconductor manufacturing industries such as photolithographic etching, plasma etching or wet chemical etching. Alternatively, micromachining methods such as laser drilling, micromilling and the like are optionally employed. Similarly, for polymeric substrates, well known manufacturing techniques may also be used. These techniques include injection molding or stamp molding methods where large numbers of substrates are optionally produced using, e.g., rolling stamps to produce large sheets of microscale substrates or polymer microcasting techniques where the substrate is polymerized within a micromachined mold. [0071]
  • The devices will typically include an additional planar element which overlays the channeled substrate enclosing and fluidly sealing the various channels to form conduits. Attaching the planar cover element is achieved by a variety of means, including, e.g., thermal bonding, adhesives or, in the case of certain substrates, e.g., glass, or semi-rigid and non-rigid polymeric substrates, a natural adhesion between the two components. The planar cover element may additionally be provided with access ports and/or reservoirs for introducing the various fluid elements needed for a particular screen. [0072]
  • The device shown in FIG. 1 also includes [0073] reservoirs 104, 106 and 108, disposed and fluidly connected at the ends of the channels 110 and 114. As shown, sample channel 112, is used to introduce the plurality of different test compounds into the device. As such, this channel will generally be fluidly connected to a source of large numbers of separate test compounds that will be individually introduced into the sample channel 112 and subsequently into channel 110.
  • The introduction of large numbers of individual, discrete volumes of test compounds into the sample is carried out by a number of methods. For example, micropipettors are optionally used to introduce the test compounds into the device. In preferred aspects, an electropipettor is used which is fluidly connected to sample [0074] channel 112. An example of such an electropipettor is described in, e.g., U.S. patent application Ser. No. 08/671,986, filed Jun. 28, 1996, now U.S. Pat. No. 5,779,868, the disclosure of which is hereby incorporated herein by reference in its entirety for all purposes. Generally, this electropipettor utilizes electroosmotic fluid direction as described herein, to alternately sample a number of test compounds, or “subject materials,” and spacer compounds. The pipettor then delivers individual, physically isolated sample or test compound volumes in subject material regions, in series, into the sample channel for subsequent manipulation within the device. Individual samples are typically separated by a spacer region of low ionic strength spacer fluid. These low ionic strength spacer regions have higher voltage drop over their length than do the higher ionic strength subject material or test compound regions, thereby driving the electrokinetic pumping. On either side of the test compound or subject material region, which is typically in higher ionic strength solution, are fluid regions referred to as first spacer regions (also referred to as “guard bands”), that contact the interface of the subject material regions. These first spacer regions typically comprise a high ionic strength solution to prevent migration of the sample elements into the lower ionic strength fluid regions, or second spacer region, which would result in electrophoretic bias. The use of such first and second spacer regions is described in greater detail in U.S. patent application Ser. No. 08/671,986, filed Jun. 28, 1996, now U.S. Pat. No. 5,779,868, which is incorporated herein by reference.
  • Alternatively, the [0075] sample channel 112 is optionally individually fluidly connected to a plurality of separate reservoirs via separate channels. The separate reservoirs each contain a separate test compound with additional reservoirs being provided for appropriate spacer compounds. The test compounds and/or spacer compounds are then transported from the various reservoirs into the sample channels using appropriate material direction schemes. In either case, it generally is desirable to separate the discrete sample volumes, or test compounds, with appropriate spacer regions.
  • As shown, the device also includes a detection window or [0076] zone 116 at which a signal from the biochemical system is optionally monitored. This detection window typically will include a transparent cover allowing visual or optical observation and detection of the assay results, e.g., observation of a colorometric or fluorometric response.
  • In particularly preferred aspects, monitoring of the signals at the detection window is achieved using an optical detection system. For example, fluorescence based signals are typically monitored using, e.g., laser activated fluorescence detection systems which employ a laser light source at an appropriate wavelength for activating the fluorescent indicator within the system. Fluorescence is then detected using an appropriate detector element, e.g., a photomultiplier tube (PMT). Similarly, for screens employing colorometric signals, spectrophotometric detection systems which direct a light source at the sample are optionally used, providing a measurement of absorbance or translnissivity of the sample. [0077]
  • In alternative aspects, the detection system may comprise non-optical detectors or sensors for detecting a particular characteristic of the system disposed within [0078] detection window 116. Such sensors may include temperature, conductivity, potentiometric (pH, ions), amperometric (for compounds that are oxidized or reduced, e.g., O2, H2O2, I2, oxidizable/reducible organic compounds, and the like).
  • In operation, a flowable first component of a biological system, e.g., a fluid comprising a receptor or enzyme, is placed in [0079] reservoir 104. This first component is flowed through main channel 110, past the detection window, 116, and toward waste reservoir 108. A second component of the biochemical system, e.g., a ligand or substrate, is concurrently flowed into the main channel 110 from the side channel 114, whereupon the first and second components mix and are able to interact. Deposition of these elements within the device is carried out in a number of ways. For example, the enzyme and substrate, or receptor and ligand solutions can be introduced into the device through open or sealable access ports in the planar cover. Alternatively, these components are optionally added to their respective reservoirs during manufacture of the device. In the case of such pre-added components, it is desirable to provide these components in a stabilized form to allow for prolonged shelf-life of the device. For example, the enzyme/substrate or receptor/ligand components are optionally provided within the device in lyophilized form. Prior to use, these components are easily reconstituted by introducing a buffer solution into the reservoirs. Alternatively, the components are lyophilized with appropriate buffering salts, whereby simple water addition is all that is required for reconstitution.
  • As noted above, the interaction of the first and second components is typically accompanied by a detectable signal. For example, in those embodiments where the first component is an enzyme and the second a substrate, the substrate is a chromogenic or fluorogenic substrate which produces an optically detectable signal when the enzyme acts upon the substrate. In the case where the first component is a receptor and the second is a ligand, either the ligand or the receptor optionally includes a detectable signal. In either event, the mixture and flow rate of compounds will typically remain constant such that the flow of the mixture of the first and second components past the [0080] detection window 116 will produce a steady-state signal. By “steady state signal” is generally meant a signal that has a regular, predictable signal intensity profile. As such, the steady-state signal may include signals having a constant signal intensity, or alternatively, a signal with a regular periodic intensity, against which variations in the normal signal profile is measured. This latter signal is generated in cases where fluid flow is periodically interrupted for, e.g., loading additional test compounds, as described in the description of the continuous flow systems. Although the signal produced in the above-described enzymatic system will vary along the length of the channel, i.e., increasing with time of exposure as the enzyme converts the fluorogenic substrate to the fluorescent product, the signal at any specific point along the channel will remain constant, given a constant flow rate.
  • From [0081] sample channel 112, test compounds is periodically or serially introduced into the main channel 110 and into the stream of first and second components as fluid regions containing the test compound, also referred to as the “subject material regions.” Where these test compounds have an effect on the interaction of the first and second elements, it will produce a deviation in the signal detected at the detection window corresponding to the subject material region. As noted above, typically, the various different test compounds to be injected through channel 112 will be separated by a first and even second spacer fluid regions to allow differentiation of the effects, or lack of effects, from one test compound to another. In those embodiments where electroosmotic fluid direction systems are employed, the spacer fluid regions may also function to reduce any electrophoretic bias that can occur within the test sample. The use of these spacer regions to drive the electroosmotic flow of fluids, as well as in the general elimination of electrophoretic bias within the sample or test compound or subject material regions is substantially described in U.S. patent application Ser. No. 08/671,986, filed Jun. 28, 1996, now U.S. Pat. No. 5,779,868, previously incorporated herein by reference.
  • By way of example, a steady, continuous flow of enzyme and fluorogenic substrate through [0082] main channel 110 will produce a constant fluorescent signal at the detection window 116. Where a test compound inhibits the enzyme, introduction of a test compound, i.e., in a subject material region, will produce a momentary but detectable drop in the level of signal at the detection window corresponding with that subject material region. The timing of the drop in signal can then be correlated with a particular test compound based upon a known injection to detection time-frame. Specifically, the time required for an injected compound to produce an observed effect can be readily determined using positive controls.
  • For receptor/ligand systems, a similar variation in the steady state signal may also be observed. Specifically, the receptor and its fluorescent ligand can be made to have different flow rates along the channel. This can be accomplished by incorporating size exclusion matrices within the channel, or, in the case of electroosmotic methods, altering the relative electrophoretic mobility of the two compounds so that the receptor flows more rapidly down the channel. Again, this is accomplished through the use of size exclusion matrices, or through the use of different surface charges in the channel which will result in differential flow rates of charge-varied compounds. Where a test compound binds to the receptor, it will result in a dark pulse in the fluorescent signal followed by a brighter pulse. Without being bound to a particular theory of operation, it is believed that the steady state signal is a result of both free fluorescent ligand, and fluorescent ligand bound to the receptor. The bound ligand is traveling at the same flow rate as the receptor while the unbound ligand is traveling more slowly. Where the test compound inhibits the receptor-ligand interaction, the receptor will not ‘bring along’ the fluorescent ligand, thereby diluting the fluorescent ligand in the direction of flow, and leaving an excess of free fluorescent ligand behind. This results in a temporary reduction in the steady-state signal, followed by a temporary increase in fluorescence. Alternatively, schemes similar to those employed for the enzymatic system is employed, where there is a signal that reflects the interaction of the receptor with its ligand. For example, pH indicators which indicate pH effects of receptor-ligand binding is incorporated into the device along with the biochemical system, i.e., in the form of encapsulated cells, whereby slight pH changes resulting from binding can be detected. See Weaver, et al., Bio/Technology (1988) 6:1084-1089. Additionally, one can monitor activation of enzymes resulting from receptor ligand binding, e.g., activation of kinases, or detect conformational changes in such enzymes upon activation, e.g., through incorporation of a fluorophore which is activated or quenched by the conformational change to the enzyme upon activation. [0083]
  • Flowing and direction of fluids within the microscale fluidic devices is carried out by a variety of methods. For example, the devices may include integrated microfluidic structures, such as micropumps and microvalves, or external elements, e.g., pumps and switching valves, for the pumping and direction of the various fluids through the device. Examples of microfluidic structures are described in, e.g., U.S. Pat. Nos. 5,271,724, 5,277,556, 5,171,132, and 5,375,979. See also, Published U.K. Patent Application No. 2 248 891 and Published European Patent Application No. 568 902. [0084]
  • Although microfabricated fluid pumping and valving systems are readily employed in the devices of the invention, the cost and complexity associated with their manufacture and operation can generally prohibit their use in mass-produced disposable devices as are envisioned by the present invention. For that reason, in particularly preferred aspects, the devices of the invention will typically include an electroosmotic fluid direction system. Such fluid direction systems combine the elegance of a fluid direction system devoid of moving parts, with an ease of manufacturing, fluid control and disposability. Examples of particularly preferred electroosmotic fluid direction systems include, e.g., those described in U.S. Pat. No. 5,858,195 to Ramsey et al., which is incorporated herein by reference in its entirety for all purposes. [0085]
  • In brief, these fluidic control systems typically include electrodes disposed within the reservoirs that are placed in fluid connection with the plurality of intersecting channels fabricated into the surface of the substrate. The materials stored in the reservoirs are transported through the channel system delivering appropriate volumes of the various materials to one or more regions on the substrate in order to carry out a desired screening assay. [0086]
  • Fluid and materials transport and direction is accomplished through electroosmosis or electrokinesis. In brief, when an appropriate material, typically comprising a fluid, is placed in a channel or other fluid conduit having functional groups present at the surface, those groups can ionize. For example, where the surface of the channel includes hydroxyl functional groups at the surface, protons can leave the surface of the channel and enter the fluid. Under such conditions, the surface will possess a net negative charge, whereas the fluid will possess an excess of protons or positive charge, particularly localized near the interface between the channel surface and the fluid. By applying an electric field along the length of the channel, cations will flow toward the negative electrode. Movement of the positively charged species in the fluid pulls the solvent with them. The steady state velocity of this fluid movement is generally given by the equation. [0087] = ɛξ E 4 πη
    Figure US20040028567A1-20040212-M00001
  • where v is the solvent velocity, ε is the dielectric constant of the fluid, ξ is the zeta potential of the surface, E is the electric field strength, and η is the solvent viscosity. Thus, as can be easily seen from this equation, the solvent velocity is directly proportional to the surface potential. [0088]
  • To provide appropriate electric fields, the system generally includes a voltage controller that is capable of applying selectable voltage levels, simultaneously, to each of the reservoirs, including ground. Such a voltage controller can be implemented using multiple voltage dividers and multiple relays to obtain the selectable voltage levels. Alternatively, multiple, independent voltage sources are optionally used. The voltage controller is electrically connected to each of the reservoirs via an electrode positioned or fabricated within each of the plurality of reservoirs. [0089]
  • Incorporating this electroosmotic fluid direction system into the device shown in FIG. 1 involves incorporation of an electrode within each of the [0090] reservoirs 104, 106 and 108, and at the terminus of sample channel 112 or at the terminus of any fluid channels connected thereto, whereby the electrode is in electrical contact with the fluid disposed in the respective reservoir or channel. Substrate materials are also selected to produce channels having a desired surface charge. In the case of glass substrates, the etched channels will possess a net negative charge resulting from the ionized hydrolysis naturally present at the surface. Alternatively, surface modifications are optionally employed to provide an appropriate surface charge, e.g., coatings, derivatization, e.g., silanation, or impregnation of the surface to provide appropriately charged groups on the surface. Examples of such treatments are described in, e.g., U.S. Pat. No. 5,885,470, which is hereby incorporated herein by reference in its entirety for all purposes.
  • In brief, suitable substrate materials are generally selected based upon their compatibility with the conditions present in the particular operation to be performed by the device. Such conditions can include extremes of pH, temperature and salt concentration. Additionally, substrate materials are also selected for their inertness to critical components of an analysis or synthesis to be carried out by the device. Polymeric substrate materials may be rigid, semi-rigid, or non-rigid, opaque, semi-opaque or transparent, depending upon the use for which they are intended. For example, devices which include an optical or visual detection element, will generally be fabricated, at least in part, from a transparent polymeric material to facilitate that detection. Alternatively, transparent windows of, e.g. glass or quartz, may be incorporated into the device for these detection elements. Additionally, the polymeric materials may have linear or branched backbones, and may be crosslinked or non-crosslinked. Examples of polymeric materials include, e.g., Acrylics, especially PMMAs (polymethylmethacrylates); exemplar acrylics include e.g., Acrylite M-30 or Acrylite L40 available from CYRO Industries, Rockaway, N.J., or PLEXIGLAS VS UVT available from Autohaas North America; polycarbonates (e.g., Makrolon CD-2005 available from The Plastics and Rubber division of Mobay Corporation (Pittsburg, Pa.) or Bayer Corporation, or LEXAN OQ 1020L or LEXAN OQ 1020, both available from GE Plastics) polydimethylsiloxanes (PDMS), polyurethane, polyvinylchloride (PVC) polystyrene, polysulfone, polycarbonate and the like. Optical, mechanical, thermal, electrical, and chemical resistance properties for many plastics are well known (and are generally available from the manufacturer), or can easily be determined by standard assays. [0091]
  • As described herein, the electrokinetic fluid control systems employed in the devices of the present invention generally utilize a substrate having charged functional groups at its surface, such as the hydroxyl groups present on glass surfaces. As described, devices of the present invention can also employ plastic or other polymeric substrates. In general, these substrate materials have hydrophobic surfaces. As a result, use of electrokinetic fluid control systems in devices utilizing polymeric substrates used in the present invention typically employs modification of the surfaces of the substrate that are in contact with fluids. [0092]
  • Surface modification of polymeric substrates may take on a variety of different forms. For example, surfaces may be coated with an appropriately charged material. For example, surfactants with charged groups and hydrophobic tails are desirable coating materials. In short, the hydrophobic tails will localize to the hydrophobic surface of the substrate, thereby presenting the charged head group at the fluid layer. [0093]
  • In one embodiment, preparation of a charged surface on the substrate involves the exposure of the surface to be modified, e.g., the channels and/or reaction chambers, to an appropriate solvent which partially dissolves or softens the surface of the polymeric substrate. A detergent is then contacted with the partially dissolved surface. The hydrophobic portion of the detergent molecules will associate with the partially dissolve polymer. The solvent is then washed from the surface, e.g., using water, whereupon the polymer surface hardens with the detergent embedded into the surface, presenting the charged head group to the fluid interface. [0094]
  • In alternative aspects, polymeric materials, such as polydimethylsiloxane, may be modified by plasma irradiation. In particular, plasma irradiation of PDMS oxidizes the methyl groups, liberating the carbons and leaving hydroxyl groups in their place, effectively creating a glass-like surface on the polymeric material, with its associated hydroxyl functional groups. [0095]
  • The polymeric substrate may be rigid, semi-rigid, nonrigid or a combination of rigid and nonrigid elements, depending upon the particular application for which the device is to be used. In one embodiment, a substrate is made up of at least one softer, flexible substrate element and at least one harder, more rigid substrate element, one of which includes the channels and chambers manufactured into its surface. Upon mating the two substrates, the inclusion of the soft element allows formation of an effective fluid seal for the channels and chambers, obviating the need and problems associated with gluing or melting more rigid plastic components together. [0096]
  • A number of additional elements are added to the polymeric substrate to provide for the electrokinetic fluid control systems. These elements may be added either during the substrate formation process, i.e., during the molding or stamping steps, or they may be added during a separate, subsequent step. These elements typically include electrodes for the application of voltages to the various fluid reservoirs, and in some embodiments, voltage sensors at the various channel intersections to monitor the voltage applied. [0097]
  • Electrodes may be incorporated as a portion of the molding process. In particular, the electrodes may be patterned within the mold so that upon introduction of the polymeric material into the mold, the electrodes will be appropriately placed. Alternatively, the electrodes and other elements may be added after the substrate is formed, using well known microfabrication methods, e.g., sputtering or controlled vapor deposition methods followed by chemical etching. [0098]
  • Whether polymeric or other substrates are used, modulating voltages are concomitantly applied to the various reservoirs to affect a desired fluid flow characteristic, e.g., continuous flow of receptor/enzyme, ligand/substrate toward the waste reservoir with the periodic introduction of test compounds. Particularly, modulation of the voltages applied at the various reservoirs can move and direct fluid flow through the interconnected channel structure of the device in a controlled manner to effect the fluid flow for the desired screening assay and apparatus. [0099]
  • FIG. 2A shows a schematic illustration of fluid direction during a typical assay screen. Specifically, shown is the injection of a test compound (in a subject material region) into a continuous stream of an enzyme-fluorogenic substrate mixture. As shown in FIG. 2A, and with reference to FIG. 1, a continuous stream of enzyme is flowed from [0100] reservoir 104, along main channel 110. Test compounds 120, separated by appropriate spacer regions 121, e.g., low ionic strength spacer regions, are introduced from sample channel 112 into main channel 110. Once introduced into the main channel, the test compounds will interact with the flowing enzyme stream. The mixed enzyme/test compound regions are then flowed along main channel 110 past the intersection with channel 114. A continuous stream of fluorogenic or chromogenic substrate which is contained in reservoir 106, is introduced into sample channel 110, whereupon it contacts and mixes with the continuous stream of enzyme, including the subject material regions which include the test compounds 122. Action of the enzyme upon the substrate will produce an increasing level of the fluorescent or chromatic signal. This increasing signal is indicated by the increasing shading within the main channel as it approaches the detection window. This signal trend will also occur within those test compound or subject material regions which have no effect on the enzyme/substrate interaction, e.g., test compound 126. Where a test compound does have an effect on the interaction of the enzyme and the substrate, a variation will appear in the signal produced. For example, assuming a fluorogenic substrate, a test compound which inhibits the interaction of the enzyme with its substrate will result in less fluorescent product being produced within that subject material region. This will result in a non-fluorescent, or detectably less fluorescent region within the flowing stream as it passes detection window 116, which corresponds to the subject material region. For example, as shown, a subject material region including a test compound 128, which is a putative inhibitor of the enzyme-substrate interaction, shows detectably lower fluorescence than the surrounding stream. This is indicated by a lack of shading of subject material region 128.
  • A detector adjacent to the detection window monitors the level of fluorescent signal being produced by the enzyme's activity on the fluorogenic or chromogenic substrate. This signal remains at a relatively constant level for those test compounds which have no effect on the enzyme-substrate interaction. When an inhibitory compound is screened, however, it will produce a momentary drop in the fluorescent signal representing the reduced or inhibited enzyme activity toward the substrate. Conversely, inducer compounds, upon screening, produce a momentary increase in the fluorescent signal, corresponding to the increased enzyme activity toward the substrate. [0101]
  • FIG. 2B provides a similar schematic illustration of a screen for effectors of a receptor-ligand interaction. As in FIG. 2A, a continuous stream of receptor is flowed from [0102] reservoir 104 through main channel 110. Test compounds or subject material regions 150 separated by appropriate spacer fluid regions 121 are introduced into the main channel 110 from sample channel 112, and a continuous stream of fluorescent ligand from reservoir 106 is introduced from side channel 114. Fluorescence is indicated by shading within the channel. As in FIG. 2A, the continuous stream of fluorescent ligand and receptor past the detection window 116 will provide a constant signal intensity. The subject material regions in the stream, containing the test compounds which have no effect on the receptor-ligand interaction, will provide the same or similar level of fluorescence as the rest of the surrounding stream, e.g., test compound or subject material region 152. However, the presence of test compounds which possess antagonistic or inhibitory activity toward the receptor-ligand interaction will result in lower levels of that interaction in those portions of the stream where those compounds are located, e.g., test compound or subject material region 154. Further, differential flow rates for the receptor bound fluorescent ligand and free fluorescent ligand will result in a detectable drop in the level of fluorescence which corresponds to the dilution of the fluorescence resulting from unbound, faster moving receptor. The drop in fluorescence is then followed by an increase in fluorescence 156 which corresponds to an accumulation of the slower moving, unbound fluorescent ligand.
  • In some embodiments, it is desirable to provide an additional channel for shunting off or extracting the subject material region reaction mixture from the running buffer and/or spacer regions. This may be the case where one wishes to keep the reaction elements contained within the a discrete fluid region during the reaction, while allowing these elements to be separated during a data acquisition stage. As described previously, one can keep the various elements of the reaction together in the subject material region that is moving through the reaction channel by incorporating appropriate spacer fluid regions between samples. Such spacer fluid regions are generally selected to retain the samples within their original subject material regions, i.e., not allowing smearing of the sample into the spacer regions, even during prolonged reaction periods. However, this goal can be at odds with those assays which are based upon the separation of elements of the assay, e.g., ligand-receptor assays described above, or where a reaction product must be separated in a capillary. Thus, it may be desirable to remove those elements which prevented such separation during the initial portions of the fluid direction. [0103]
  • A schematic illustration of one embodiment of a [0104] device 500 for performing this sample or subject material shunting or extraction is shown in FIG. 5. As shown, the subject materials or test compounds 504 are introduced to the device or chip via the sample channel 512. Again, these are typically introduced via an appropriate injection device 506, e.g., a capillary pipettor. The ionic strength and lengths of the first spacer regions 508 and second spacer regions 502 are selected such that those samples with the highest electrophoretic mobility will not migrate through the first spacer regions 508 into the second spacer regions 502 in the length of time that it takes the sample to travel down the reaction channel.
  • Assuming a receptor ligand assay system, test compounds pass into the [0105] device 500 and into reaction channel 510, where they are first combined with the receptor. The test compound/receptor, in the form of the subject material regions, are flowed along the reaction channel in the incubation zone 510 a. Following this initial incubation, the test compound/receptor mix is combined with a labelled ligand (e.g., fluorescent ligand) whereupon this mixture flows along the second incubation region 510 b of reaction channel 510. The lengths of the incubation regions and the flow rates of the system (determined by the potentials applied at each of the reservoirs 514, 516, 518, 520, 522, and at the terminus of sample channel 512) determine the time of incubation of the receptor with the fluorescent ligand and test compound. The ionic strengths of the solutions containing the receptors and fluorescent ligands, as well as the flow rates of material from the reservoirs housing these elements into the sample channel are selected so as to not interfere with the first and second spacer regions.
  • The isolated subject material regions containing receptor, fluorescent ligand and test compound are flowed along the [0106] reaction channel 510 by the application of potentials at, e.g., reservoirs 514, 516, 518 and at the terminus of sample channel 512. Potentials are also applied at reservoirs 520 and 522, at the opposite ends of separation channel 524, to match the potentials at the two ends of the transfer channel, so that the net flow across the transfer channel is zero. As the subject material region passes the intersection of reaction channel 510 and transfer channel 526, the potentials are allowed to float at reservoirs 518 and 522, whereupon the potentials applied at reservoirs 514, 516, 520, and at the terminus of sample channel 512, result in the subject material region being shunted through transfer channel 526 and into separation channel 524. Once in the separation channel, the original potentials are reapplied to all of the reservoirs to stop the net fluid flow through transfer channel 526. The diversion of the subject material can then be repeated with each subsequent subject material region. Within the separation channel, the subject material region is exposed to different conditions than those of the reaction channel. For example, a different flow rate may be used, capillary treatments may allow for separation of differentially charged or different sized species, and the like. In a preferred aspect, the subject material is shunted into the separation channel to place the subject material into a capillary filled with high ionic strength buffer, i.e., to remove the low ionic strength spacer regions, thereby allowing separation of the various sample components outside the confines of the original subject material region. For example, in the case of the above-described receptor/ligand screen, the receptor/ligand complex may have a different electrophoretic mobility from the ligand alone, in the transfer channel, thereby allowing more pronounced separation of the complex from the ligand, and its subsequent detection.
  • Such modifications have a wide variety of uses, particularly where it is desirable to separate reaction products following reaction, e.g., in cleavage reactions, fragmentation reactions, PCR reactions, and the like. [0107]
  • C. Serial in Parallel Assay Systems [0108]
  • More complex systems can also be produced within the scope of the present invention. For example, a schematic illustration of one alternate embodiment employing a “serial input parallel reaction” geometry is shown in FIG. 3. As shown, the [0109] device 300 again includes a planar substrate 302 as described previously. Fabricated into the surface of the substrate 302 are a series of parallel reaction channels 312-324. Also shown are three transverse channels fluidly connected to each of these parallel reaction channels. The three transverse channels include a sample injection channel 304, an optional seeding channel 306 and a collection channel 308. Again, the substrate and channels are generally fabricated utilizing the materials and to the dimensions generally described above. Although shown and described in terms of a series of parallel channels, the reaction channels may also be fabricated in a variety of different orientations. For example, rather than providing a series of parallel channels fluidly connected to a single transverse channel, the channels are optionally fabricated connecting to and extending radially outward from a central reservoir, or are optionally arranged in some other non-parallel fashion. Additionally, although shown with three transverse channels, it will be recognized that fewer transverse channels are used where, e.g., the biochemical system components are predisposed within the device. Similarly, where desired, more transverse channels are optionally used to introduce further elements into a given assay screen. Accordingly, the serial-in-parallel devices of the present invention will typically include at least two and preferably three, four, five or more transverse channels. Similarly, although shown with 7 reaction channels, it will be readily appreciated that the microscale devices of the present invention will be capable of comprising more than 7 channels, depending upon the needs of the particular screen. In preferred aspects, the devices will include from 10 to about 500 reaction channels, and more preferably, from 20 to about 200 reaction channels.
  • This device may be particularly useful for screening test compounds serially injected into the device, but employing a parallel assay geometry, once the samples are introduced into the device, to allow for increased throughput. [0110]
  • In operation, test compounds in discrete subject material regions, are serially introduced into the device, separated as described above, and flowed along the transverse [0111] sample injection channel 304 until the separate subject material regions are adjacent the intersection of the sample channel 304 with the parallel reaction channels 310-324. As shown in FIGS. 4A-4F, the test compounds are optionally provided immobilized on individual beads. In those cases where the test compounds are immobilized on beads, the parallel channels are optionally fabricated to include bead resting wells 326-338 at the intersection of the reaction channels with the sample injection channel 304. Arrows 340 indicate the net fluid flow during this type of sample/bead injection. As individual beads settle into a resting well, fluid flow through that particular channel will be generally restricted. The next bead in the series following the unrestricted fluid flow, then flows to the next available resting well to settle in place.
  • Once in position adjacent to the intersection of the parallel reaction channel and the sample injection channel, the test compound is directed into its respective reaction channel by redirecting fluid flows down those channels. Again, in those instances where the test compound is immobilized on a bead, the immobilization will typically be via a cleavable linker group, e.g., a photolabile, acid or base labile linker group. Accordingly, the test compound will typically need to be released from the bead, e.g., by exposure to a releasing agent such as light, acid, base or the like prior to flowing the test compound down the reaction channel. [0112]
  • Within the parallel channel, the test compound will be contacted with the biochemical system for which an effector compound is being sought. As shown, the first component of the biochemical system is placed into the reaction channels using a similar technique to that described for the test compounds. In particular, the biochemical system is typically introduced via one or more [0113] transverse seeding channels 306. Arrows 342 illustrate the direction of fluid flow within the seeding channel 306. The biochemical system are optionally solution based, e.g., a continuously flowing enzyme/substrate or receptor-ligand mixture, like that described above, or as shown in FIGS. 4A-4F, may be a whole cell or bead based system, e.g., beads which have enzyme/substrate systems immobilized thereon.
  • In those instances where the biochemical system is incorporated in a particle, e.g., a cell or bead, the parallel channel may include a [0114] particle retention zone 344. Typically, such retention zones will include a particle sieving or filtration matrix, e.g., a porous gel or microstructure which retains particulate material but allows the free flow of fluids. Examples of microstructures for this filtration include, e.g., those described in U.S. Pat. No. 5,304,487, which is hereby incorporated by reference in its entirety for all purposes. As with the continuous system, fluid direction within the more complex systems may be generally controlled using microfabricated fluid direction structures, e.g., pumps and valves. However, as the systems grow more complex, such systems become largely unmanageable. Accordingly, electroosmotic systems, as described above, are generally preferred for controlling fluid in these more complex systems. Typically, such systems will incorporate electrodes within reservoirs disposed at the termini of the various transverse channels to control fluid flow thorough the device. In some aspects, it is desirable to include electrodes at the termini of all the various channels. This generally provides for more direct control, but also grows less manageable as systems grow more complex. In order to utilize fewer electrodes and thus reduce the potential complexity, it may often be desirable in parallel systems, e.g., where two fluids are desired to move at similar rates in parallel channels, to adjust the geometries of the various flow channels. In particular, as channel length increases, resistance along that channel will also increase. As such, flow lengths between electrodes should be designed to be substantially the same regardless of the parallel path chosen. This will generally prevent the generation of transverse electrical fields and thus promote equal flow in all parallel channels. To accomplish substantially the same resistance between the electrodes, one can alter the geometry of the channel structure to provide for the same channel length, and thus, the channel resistance, regardless of the path travelled. Alternatively, resistance of channels are optionally adjusted by varying the cross-sectional dimensions of the paths, thereby creating uniform resistance levels regardless of the path taken.
  • As the test compounds are drawn through their respective parallel reaction channels, they will contact the biochemical system in question. As described above, the particular biochemical system will typically include a flowable indicator system which indicates the relative functioning of that system, e.g., a soluble indicator such as chromogenic or fluorogenic substrate, labelled ligand, or the like, or a particle based signal, such as a precipitate or bead bound signaling group. The flowable indicator is then flowed through the respective parallel channel and into the [0115] collection channel 308 whereupon the signals from each of the parallel channels are flowed, in series, past the detection window, 116.
  • FIGS. [0116] 4A-4F, with reference to FIG. 3, show a schematic illustration of the progression of the injection of test compounds and biochemical system components into the “serial input parallel reaction” device, exposure of the system to the test compounds, and flowing of the resulting signal out of the parallel reaction channels and past the detection window. In particular, FIG. 4A shows the introduction of test compounds immobilized on beads 346 through sample injection channel 304. Similarly, the biochemical system components 348 are introduced into the reaction channels 312-324 through seeding channel 306. Although shown as being introduced into the device along with the test compounds, as described above, the components of the model system to be screened are optionally incorporated into the reaction channels during manufacture. Again, such components are optionally provided in liquid form or in lyophilized form for increased shelf life of the particular screening device.
  • As shown, the biochemical system components are embodied in a cellular or particle based system, however, fluid components may also be used as described herein. As the particulate components flow into the reaction channels, they are optionally retained upon an optional [0117] particle retaining matrix 344, as described above.
  • FIG. 4B illustrates the release of test compounds from the [0118] beads 346 by exposing the beads to a releasing agent. As shown, the beads are exposed to light from an appropriate light source 352, e.g., which is able to produce light in a wavelength sufficient to photolyze the linker group, thereby releasing compounds that are coupled to their respective beads via a photolabile linker group.
  • In FIG. 4C, the released test compounds are flowed into and along the parallel reaction channels as shown by [0119] arrows 354 until they contact the biochemical system components. The biochemical system components 348 are then allowed to perform their function, e.g., enzymatic reaction, receptor/ligand interaction, and the like, in the presence of the test compounds. Where the various components of the biochemical system are immobilized on a solid support, release of the components from their supports can provide the initiating event for the system. A soluble signal 356 which corresponds to the functioning of the biochemical system is then generated (FIG. 4D). As described previously, a variation in the level of signal produced is an indication that the particular test compound is an effector of the particular biochemical system. This is illustrated by the lighter shading of signal 358.
  • In FIGS. 4E and 4F, the soluble signal is then flowed out of reactions channels [0120] 312-324 into the detection channel 308, and along the detection channel past the detection window 116.
  • Again, a detection system as described above, located adjacent the detection window will monitor the signal levels. In some embodiments, the beads which bore the test compounds are optionally recovered to identify the test compounds which were present thereon. This is typically accomplished by incorporation of a tagging group during the synthesis of the test compound on the bead. As shown, spent [0121] bead 360, i.e., from which a test compound has been released, is optionally transported out of the channel structure through port 362 for identification of the test compound that had been coupled to it. Such identification are optionally accomplished outside of the device by directing the bead to a fraction collector, whereupon the test compounds present on the beads are optionally identified, either through identification of a tagging group, or through identification of residual compounds. Incorporation of tagging groups in combinatorial chemistry methods has been previously described using encrypted nucleotide sequences or chlorinated/fluorinated aromatic compounds as tagging groups. See, e.g., Published PCT Application No. WO 95/12608. Alternatively, the beads are optionally transported to a separate assay system within the device itself whereupon the identification is carried out.
  • FIG. 6A shows an alternate embodiment of a “serial input parallel reaction” device which can be used for fluid based as opposed to bead based systems. As shown the [0122] device 600 generally incorporates at least two transverse channels as were shown in FIGS. 3 and 4, namely, sample injection channel 604 and detection channel 606. These transverse channels are interconnected by the series of parallel channels 612-620 which connect sample channel 604 to detection channel 606.
  • The device shown also includes an additional set of channels for directing the flow of fluid test compounds into the reaction channels. In particular, an additional [0123] transverse pumping channel 634 is fluidly connected to sample channel 604 via a series of parallel pumping channels 636-646. The pumping channel includes reservoirs 650 and 652 at its termini. The intersections of parallel channels 636-646 are staggered from the intersections of parallel channels 612-620 with sample channel 604, e.g., half way between. Similarly, transverse pumping channel 608 is connected to detection channel 606 via parallel pumping channels 622-632. Again, the intersections of parallel pumping channels 622-632 with detection channel 606 are staggered from the intersections of reaction channels 612-620 with the detection channel 606.
  • A schematic illustration of the operation of this system is shown in FIGS. [0124] 6B-6C. As shown, a series of test compounds, physically isolated from each other in separate subject material regions, are introduced into sample channel 604 using the methods described previously. For electroosmotic systems, potentials are applied at the terminus of sample channel 604, as well as reservoir 648. Potentials are also applied at reservoirs 650:652, 654:656, and 658:660. This results in a fluid flow along the transverse channels 634, 604, 606 and 608, as illustrated by the arrows, and a zero net flow through the parallel channel arrays interconnecting these transverse channels, as shown in FIG. 6B. Once the subject material regions containing the test compounds are aligned with parallel reaction channels 612-620, connecting sample channel 604 to detection channel 606, as shown by the shaded areas in FIG. 6B, flow is stopped in all transverse directions by removing the potentials applied to the reservoirs at the ends of these channels. As described above, the geometry of the channels can be varied to maximize the use of space on the substrate. For example, where the sample channel is straight, the distance between reaction channels (and thus, the number of parallel reactions that can be carried out in a size limited substrate) is dictated by the distance between subject material regions. These restrictions, however, can be eliminated through the inclusion of altered channel geometries. For example, in some aspects, the length of a first and second spacer regions can be accommodated by a serpentine, square-wave, saw tooth or other reciprocating channel geometry. This allows packing a maximum number of reaction channels onto the limited area of the substrate surface.
  • Once aligned with the parallel reaction channels, the sample, or subject material, is then moved into the parallel reaction channels [0125] 612-620 by applying a first potential to reservoirs 650 and 652, while applying a second potential to reservoirs 658 and 660, whereby fluid flow through parallel pumping channels 636-646 forces the subject material into parallel reaction channels 612-620, as shown in FIG. 6C. During this process, no potential is applied at reservoirs—648, 654, 656, or the terminus of sample channel 604. Parallel channels 636-646 and 622-632 are generally adjusted in length such that the total channel length, and thus the level of resistance, from reservoirs 650 and 652 to channel 604 and from reservoirs 658 and 660 to channel 606, for any path taken will be the same. Resistance can generally be adjusted by adjusting channel length or width. For example, channels can be lengthened by including folding or serpentine geometries. Although not shown as such, to accomplish this same channel length, channels 636 and 646 would be the longest and 640 and 642 the shortest, to create symmetric flow, thereby forcing the samples into the channels. As can be seen, during flowing of the samples through channels 612-620, the resistance within these channels will be the same, as the individual channel length is the same.
  • Following the reaction to be screened, the subject material region/signal element is moved into [0126] detection channel 606 by applying a potential from reservoirs 650 and 652 to reservoirs 658 and 660, while the potentials at the remaining reservoirs are allowed to float. The subject material regions/signal are then serially moved past the detection window/detector 662 by applying potentials to reservoirs 654 and 656, while applying appropriate potentials at the termini of the other transverse channels to prevent any flow along the various parallel channels.
  • Although shown with channels which intersect at right angles, it will be appreciated that other geometries are also appropriate for serial input parallel reactions. For example, U.S. Ser. No. 08,835,101, filed Apr. 4, 1997, describes advantages to parabolic geometries and channels which vary in width for control of fluid flow. In brief, fluid flow in electroosmotic systems is controlled by and therefore related to current flow between electrodes. Resistance in the fluid channels varies as a function of path length and width, and thus, different length channels have different resistances. If this differential in resistance is not corrected for, it results in the creation of transverse electrical fields which can inhibit the ability of the devices to direct fluid flow to particular regions. The current, and thus the fluid flow, follows the path of least resistance, e.g., the shortest path. While this problem of transverse electrical fields is alleviated through the use of separate electrical systems, i.e., separate electrodes, at the termini of each and every parallel channel, production of devices incorporating all of these electrodes, and control systems for controlling the electrical potential applied at each of these electrodes can be complex, particularly where one is dealing with hundreds to thousands of parallel channels in a single small scale device, e.g., 1-2 cm[0127] 2. Accordingly, the present invention provides microfluidic devices for affecting serial to parallel conversion, by ensuring that current flow through each of a plurality of parallel channels is at an appropriate level to ensure a desired flow pattern through those channels or channel networks. A number of methods and substrate/channel designs for accomplishing these goals are appropriate.
  • In one example of parabolic geometry for the channels in an apparatus of the invention, the substrate includes a main channel. A series of parallel channels terminate in a main channel. The opposite termini of these parallel channels are connected to parabolic channels. Electrodes are disposed at the termini of these parabolic channels. The current flow in each of the parallel channels is maintained constant or equivalent, by adjusting the length of the parallel channels, resulting in a parabolic channel structure connecting each of the parallel channels to its respective electrodes. The voltage drop within the parabolic channel between the parallel channels is maintained constant by adjusting the channel width to accommodate variations in the channel current resulting from the parallel current paths created by these parallel channels. The parabolic design of the channels, in combination with their tapering structures, results in the resistance along all of the parallel channels being equal, resulting in an equal fluid flow, regardless of the path chosen. Generally, determining the dimensions of channels to ensure that the resistances among the channels are controlled as desired, may be carried out by well known methods, and generally depends upon factors such as the make-up of the fluids being moved through the substrates. [0128]
  • Although generally described in terms of screening assays for identification of compounds which affect a particular interaction, based upon the present disclosure, it will be readily appreciated that the above described microlaboratory systems may also be used to screen for compounds which specifically interact with a component of a biochemical system without necessarily affecting an interaction between that component and another element of the biochemical system. Such compounds typically include binding compounds which may generally be used in, e.g., diagnostic and therapeutic applications as targeting groups for therapeutics or marker groups, i.e. radionuclides, dyes and the like. For example, these systems are optionally used to screen test compounds for the ability to bind to a given component of a biochemical system. [0129]
  • II. Microlaboratory System [0130]
  • Although generally described in terms of individual discrete devices, for ease of operation, the systems described will typically be a part of a larger system which can monitor and control the functioning of the devices, either on an individual basis, or in parallel, multi-device screens. An example of such a system is shown in FIG. 7. [0131]
  • As shown in FIG. 7, the system may include a test [0132] compound processing system 700. The system shown includes a platform 702 which can hold a number of separate assay chips or devices 704. As shown, each chip includes a number of discrete assay channels 706, each having a separate interface 708, e.g., pipettor, for introducing test compounds into the device. These interfaces are used to sip test compounds into the device, separated by sipping first and second spacer fluids, into the device. In the system shown, the interfaces of the chip are inserted through an opening 710 in the bottom of the platform 702, which is capable of being raised and lowered to place the interfaces in contact with test compounds or wash/first spacer fluids/second spacer fluids, which are contained in, e.g., multiwell micro plates 711, positioned below the platform, e.g., on a conveyor system 712. In operation, multiwell plates containing large members of different test compounds are stacked 714 at one end of the conveyor system. The plates are placed upon the conveyor separated by appropriate buffer reservoirs 716 and 718, which may be filled by buffer system 720. The plates are stepped down the conveyor and the test compounds are sampled into the chips, interspersed by appropriate spacer fluid regions. After loading the test compounds into the chips, the multiwell plates are then collected or stacked 722 at the opposite end of the system. The overall control system includes a number of individual microlaboratory systems or devices, e.g., as shown in FIG. 7. Each device is connected to a computer system which is appropriately programmed to control fluid flow and direction within the various chips, and to monitor, record and analyze data resulting from the screening assays that are performed by the various devices. The devices will typically be connected to the computer through an intermediate adapter module which provides an interface between the computer and the individual devices for implementing operational instructions from the computer to the devices, and for reporting data from the devices to the computer. For example, the adapter will generally include appropriate connections to corresponding elements on each device, e.g., electrical leads connected to the reservoir based electrodes that are used for electroosmotic fluid flow, power inputs and data outputs for detection systems, either electrical or fiberoptic, and data relays for other sensor elements incorporated into the devices. The adapter device may also provide environmental control over the individual devices where such control is necessary, e.g., maintaining the individual devices at optimal temperatures for performing the particular screening assays.
  • As shown, each device is also equipped with appropriate fluid interfaces, e.g., micropipettors, for introducing test compounds into the individual devices. The devices may readily be attached to robotic systems which allow test compounds to be sampled from a number of multiwell plates that are moved along a conveyor system. Intervening spacer fluid regions can also be introduced via a spacer solution reservoir. [0133]
  • III. Fluid Electrode Interface to Prevent Degdation of Chemical Species in a Microchip [0134]
  • When pumping fluids or other materials electroosmotically or electrophoretically through an apparatus of the invention, chemical species in the fluid can be degraded if high voltages or currents are applied, or if voltages are applied for a long period of time. Designs which retard movement of chemical species from the electrode to a channel entrance or retard the movement of chemical species to the electrode improve performance of chemical assays by reducing unwanted degradation of chemical species within the sample. These designs are particularly preferred in assay systems where voltages are applied for long periods, e.g., several hours to several days. [0135]
  • Electrode designs which reduce degradation of chemical species in the assays of the invention are illustrated by consideration of FIG. 12, panels A-G. The designs retard the moving of chemical species from the electrode to the channel entrance or retard the movement of chemical species to the electrode improve performance of chemical assays. FIG. 12A shows a typical electrode design, in which electrode [0136] 1211 is partially submerged in reservoir 1215 fluidly connected to fluid channel 1217.
  • In comparison, FIG. 12B utilizes a salt bridge between electrode with frit [0137] 1219 and fluid reservoir 1221 fluidly connected to fluid channel 1223.
  • FIG. 12C reduces degradation of chemical species by providing [0138] electrode 1225 submersed in first fluid reservoir 1227 fluidly connected to second fluid reservoir 1229 by large channel 1231 which limits diffusion, but has a low electroosmotic flow.
  • FIG. 12D provides a similar two part reservoir, in which electrode [0139] 1235 is submersed in first fluid reservoir 1237 fluidly connected to second fluid reservoir 1241 by small channel 1243 which is treated to reduce or eliminate electroosmotic flow.
  • FIG. 12E provides another similar two part reservoir, in which electrode [0140] 1245 is submersed in first fluid reservoir 1247 fluidly connected to second fluid reservoir 1251 by channel 1253. Channel 1253 is filled with a material such as gel, Agar, glass beads or other matrix material for reducing electroosmotic flow.
  • FIG. 12F provides a variant two part reservoir system, in which electrode [0141] 1255 is submersed in first fluid reservoir 1257 fluidly connected to second fluid reservoir 1259 by channel 1261. The fluid level in second fluid reservoir 1259 is higher than the fluid level in first fluid reservoir 1257, which forces fluid towards electrode 1255.
  • FIG. 12G provides a second variant two part reservoir, in which electrode [0142] 1265 is submersed in first fluid reservoir 1267 fluidly connected to second fluid reservoir 1269 by channel 1271. The diameter on first fluid reservoir 1267 is small enough that capillary forces draw fluid into first fluid reservoir 1267.
  • Modifications can be made to the method and apparatus as hereinbefore described without departing from the spirit or scope of the invention as claimed, and the invention can be put to a number of different uses, including: [0143]
  • The use of a microfluidic system containing at least a first substrate having a first channel and a second channel intersecting said first channel, at least one of said channels having at least one cross-sectional dimension in a range from 0.1 to 500 μm, in order to test the effect of each of a plurality of test compounds on a biochemical system. [0144]
  • The use of a microfluidic system as hereinbefore described, wherein said biochemical system flows through one of said channels substantially continuously, enabling sequential testing of said plurality of test compounds. [0145]
  • The use of a microfluidic system as hereinbefore described, wherein the provision of a plurality of reaction channels in said first substrate enables parallel exposure of a plurality of test compounds to at least one biochemical system. [0146]
  • The use of a microfluidic system as hereinbefore described, wherein each test compound is physically isolated from adjacent test compounds. [0147]
  • The use of a substrate carrying intersecting channels in screening test materials for effect on a biochemical system by flowing said test materials and biochemical system together using said channels. [0148]
  • The use of a substrate as hereinbefore described, wherein at least one of said channels has at least one cross-sectional dimension of range 0.1 to 500 μm. [0149]
  • An assay utilizing a use of any one of the microfluidic systems or substrates hereinbefore described. [0150]
  • The invention provides, inter alia, an apparatus for detecting an effect of a test compound on a biochemical system, comprising a substrate having at least one surface with a plurality of reaction channels fabricated into the surface. Apparatus as hereinbefore described, having at least two transverse channels fabricated into the surface, wherein each of the plurality of reaction channels is fluidly connected to a first of the at least two transverse channels at a first point in each of the reaction channels, and fluidly connected to a second transverse channel at a second point in each of the reaction channels and an assay apparatus including an apparatus as hereinbefore described are also provided. [0151]
  • EXAMPLES
  • The following examples are provided by way of illustration only and not by way of limitation. Those of skill will readily recognize a variety of noncritical parameters which can be changed or modified to yield essentially similar results. [0152]
  • Example 1
  • Enzyme Inhibitor Screen [0153]
  • The efficacy of performing an enzyme inhibition assay screen was demonstrated in a planar chip format. A 6-port planar chip was employed having the layout shown in FIG. 8. The numbers adjacent the channels represent the lengths of each channel in millimeters. Two voltage states were applied to the ports of the chip. The first state (State 1) resulted in flowing of enzyme with buffer from the top buffer well into the main channel. The second voltage state (State 2) resulted in the interruption of the flow of buffer from the top well, and the introduction of inhibitor from the inhibitor well, into the main channel along with the enzyme. A control experiment was also run in which buffer was placed into the inhibitor well. [0154]
  • Applied voltages at each port for each of the two applied voltage states were as follows: [0155]
    State 1 State 2
    Top Buffer Well (I) 1831 1498
    Inhibitor Well(II) 1498 1900
    Enzyme Well (III) 1891 1891
    Substrate Well (IV) 1442 1442
    Bottom Buffer Well (V) 1442 1442
    Detect./Waste Well (VI) 0 0
  • To demonstrate the efficacy of the system, an assay was designed to screen inhibitors of β-galactosidase using the following enzyme/substrate/inhibitor reagents: [0156]
  • Enzyme: β-Galactosidase (180 U/ml in 50 mM Tris/300 μg/ml BSA [0157]
  • Substrate: Fluorescein-digalactoside (FDG) 400 μM [0158]
  • Inhibitor: IPTG, 200 mM [0159]
  • Buffer: 20 mM Tris, pH 8.5 [0160]
  • Enzyme and substrate were continually pumped through the main channel from their respective ports under both voltage states. Inhibitor or Buffer were delivered into the main channel alternately from their respective wells by alternating between [0161] voltage state 1 and voltage state 2. When no inhibitor was present at the detection end of the main channel, a base line level of fluorescent product was produced. Upon introduction of inhibitor, the fluorescent signal was greatly reduced, indicating inhibition of the enzyme/substrate interaction. Fluorescent data obtained from the alternating delivery of inhibitor and buffer into the main channel is shown in FIG. 9A. FIG. 9B a superposition of the two data segments from FIG. 9A, directly comparing the inhibitor data with control (buffer) data. The control shows only a minor fluctuation in the fluorescent signal that apparently resulted from a dilution of the enzyme substrate mixture, whereas the inhibitor screen shows a substantial reduction in the fluorescent signal, indicating clear inhibition.
  • Example 2
  • Screening of Multiple Test Compounds [0162]
  • An assay screen is performed to identify inhibitors of an enzymatic reaction. A schematic of the chip to be used is shown in FIG. 10. The chip has a [0163] reaction channel 5 cm in length which includes a 1 cm incubation zone and a 4 cm reaction zone. The reservoir at the beginning of the sample channel is filled with enzyme solution and the side reservoir is filled with the fluorogenic substrate. Each of the enzyme and substrate are diluted to provide for a steady state signal in the linear signal range for the assay system, at the detector. Potentials are applied at each of the reservoirs (sample source, enzyme, substrate and waste) to achieve an applied field of 200 V/cm. This applied field produces a flow rate of 2 mm/second. During passage of a given sample through the chip, there will generally be a diffusive broadening of the sample. For example, in the case of a small molecule sample, e.g., 1 mM benzoic acid diffusive broadening of approximately 0.38 mm and an electrophoretic shift of 0.4 mm is seen.
  • Subject material regions containing test compounds in 150 mM NaCl are introduced into the sample channel separated by first spacer regions of 150 mM NaCl and second spacer regions of 5 mM borate buffer. Once introduced into the sample channel shown, the subject material region requires 12 seconds to travel the length of the sample channel and reach the incubation zone of the reaction channel. This is a result of the flow rate of 2 mm/sec, allowing for 1 second for moving the sample pipettor from the sample to the spacer compounds. Allowing for these interruptions, the net flow rate is 0.68 mm/sec. Another 12 seconds is required for the enzyme/test compound mixture to travel through the incubation zone to the intersection with the substrate channel where substrate is continuously flowing into the reaction zone of the reaction channel. Each subject material region containing the test compounds then requires 48 seconds to travel the length of the reaction zone and past the fluorescence detector. A schematic of timing for subject material region/spacer region loading is shown in FIG. 11. The top panel shows the subject material/first spacer region/second spacer region distribution within a channel, whereas the lower panel shows the timing required for loading the channel. As shown, the schematic includes the loading (sipping) of high salt (HS) first spacer fluid (“A”), moving the pipettor to the sample or subject material (“B”), sipping the sample or subject material (“C”), moving the pipettor to the high salt first spacer fluid (“D”) sipping the first spacer fluid (“E”), moving the pipettor to the low salt (LS) or second spacer fluid (“F”), sipping the second spacer fluid (“G”) and returning to the first spacer fluid (“H”). The process is then repeated for each additional test compound. [0164]
  • A constant base fluorescent signal is established at the detector in the absence of test compounds. Upon introduction of the test compounds, a decrease in fluorescence is seen similar to that shown in FIGS. 9A and 9B, which, based upon time delays, corresponds to a specific individual test compound. This test compound is tentatively identified as an inhibitor of the enzyme, and further testing is conducted to confirm this and quantitate the efficacy of this inhibitor. [0165]
  • While tie foregoing invention has been described in some detail for purposes of clarity and understanding, it will be clear to one skilled in the art from a reading of this disclosure that various changes in form and detail can be made without departing from the true scope of the invention. All publications and patent documents cited in this application are incorporated by reference in their entirety for all purposes to the same extent as if each individual publication or patent document were so individually denoted. [0166]

Claims (14)

What is claimed is:
1. A microfluidic device, comprising:
a body having at least a first microscale channel disposed therein;
at least a first reservoir in fluid communication with the first microscale channel, the first reservoir having no electrodes disposed therein;
at least a second reservoir connected to the first reservoir by a second channel; and
at least a first electrode place into contact with a fluid disposed in the second reservoir:
wherein the second channel is configured to retard the movement of one or more degradable components between the electrode and the first microscale channel, the second channel comprising one or more of:
a cross-sectional dimension that is smaller than a cross-sectional dimension of the first channel; and
a cross-sectional dimension that is smaller than a cross-sectional dimension of the first channel.
2. A microfluidic device, comprising:
a body having at least a first microscale channel disposed therein;
at least a first reservoir in fluid communication with the first microscale channel, the first reservoir having no electrodes disposed therein;
at least a second reservoir connected to the first reservoir by a second channel, wherein the second channel comprises a frit disposed therein; and
at least a first electrode place into contact with a fluid disposed in the second reservoir.
3. A microfluidic device, comprising:
a body having at least a first microscale channel disposed therein;
at least a first reservoir in fluid communication with the first microscale channel, the first reservoir having no electrodes disposed therein;
at least a second reservoir connected to the first reservoir by a second channel; and
at least a first electrode place into contact with a fluid disrosed in the second reservoir;
wherein a fluid level in the first reservoir is higher than a fluid level in the second reservoir.
4. A microfluidic device, comprising:
a body having at least a first channel disposed therein;
first and second reservoirs in fluid communication with first and second ends of the first channel, respectively;
first and second electrodes electrically connected to a fluid in the first and second reservoirs, respectively, to provide a voltage gradient through the first channel between the first and second reservoirs; and
wherein the first electrode is separated from the first end of the first channel by a first structure which prevents migration of an electrically degraded chemical species from a surface of the first electrode to the first end of the first channel.
5. The microfluidic device of claim 4, wherein the first structure comprises a third reservoir disposed in the body, and a second channel fluidly connecting the third reservoir to the first reservoir, the first electrode being disposed in contact with a fluid in the third reservoir, but not disposed in the first reservoir.
6. The microfluidic device of claim 5, wherein the third reservoir comprises a level of fluid that is lower than a level of fluid in the first reservoir.
7. The microfluidic device of claim 5, wherein the second channel comprises a cross section that is smaller than a cross section of the first channel.
8. The microfluidic device of claim 5, wherein the second channel comprises a cross section that is larger than a cross section of the first channel.
9. The microfluidic device of claim 4, wherein the first structure comprises a frit disposed between the first electrode and the first end of the first channel.
10. The microfluidic device of claim 9, wherein the frit surrounds the first electrode.
11. The microfluidic device of claim 9, further comprising a third reservoir disposed in the body, and a second channel fluidly connecting the third reservoir to the first reservoir, the first electrode being disposed in contact with a fluid in the third reservoir, and the frit being disposed within the second channel.
12. The microfluidic device of claim 4, wherein the first structure comprises a salt bridge disposed between the first electrode and the first end of the first channel.
13. The microfluidic device of claim 12, wherein the salt bridge comprises a gel disposed within the second channel.
14. The microfluidic device of claim 13, wherein the salt bridge comprises beads disposed within the second channel.
US10/637,730 1996-06-28 2003-08-07 High throughput screening assay systems in microscale fluidic devices Abandoned US20040028567A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US10/637,730 US20040028567A1 (en) 1996-06-28 2003-08-07 High throughput screening assay systems in microscale fluidic devices
US11/050,900 US20060000722A1 (en) 1996-06-28 2005-02-03 High throughput screening assay systems in microscale fluidic devices
US11/176,805 US20050241941A1 (en) 1996-06-28 2005-07-06 High throughput screening assay systems in microscale fluidic devices

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US08/671,987 US5942443A (en) 1996-06-28 1996-06-28 High throughput screening assay systems in microscale fluidic devices
US08/761,575 US6046056A (en) 1996-06-28 1996-12-06 High throughput screening assay systems in microscale fluidic devices
US08/881,696 US6267858B1 (en) 1996-06-28 1997-06-24 High throughput screening assay systems in microscale fluidic devices
US09/718,236 US6630353B1 (en) 1996-06-28 2000-11-21 High throughput screening assay systems in microscale fluidic devices
US10/637,730 US20040028567A1 (en) 1996-06-28 2003-08-07 High throughput screening assay systems in microscale fluidic devices

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/718,236 Continuation US6630353B1 (en) 1996-06-28 2000-11-21 High throughput screening assay systems in microscale fluidic devices

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US11/050,900 Continuation-In-Part US20060000722A1 (en) 1996-06-28 2005-02-03 High throughput screening assay systems in microscale fluidic devices
US11/176,805 Continuation US20050241941A1 (en) 1996-06-28 2005-07-06 High throughput screening assay systems in microscale fluidic devices

Publications (1)

Publication Number Publication Date
US20040028567A1 true US20040028567A1 (en) 2004-02-12

Family

ID=27100659

Family Applications (13)

Application Number Title Priority Date Filing Date
US09/214,035 Expired - Lifetime US6429025B1 (en) 1996-06-28 1997-06-24 High-throughput screening assay systems in microscale fluidic devices
US08/881,696 Expired - Lifetime US6267858B1 (en) 1996-06-28 1997-06-24 High throughput screening assay systems in microscale fluidic devices
US09/044,587 Expired - Lifetime US6413782B1 (en) 1996-06-28 1998-03-19 Methods of manufacturing high-throughput screening systems
US09/044,557 Expired - Lifetime US6274337B1 (en) 1996-06-28 1998-03-19 High throughput screening assay systems in microscale fluidic devices
US09/132,963 Expired - Lifetime US6479299B1 (en) 1996-06-28 1998-08-12 Pre-disposed assay components in microfluidic devices and methods
US09/196,535 Expired - Lifetime US6306659B1 (en) 1996-06-28 1998-11-20 High throughput screening assay systems in microscale fluidic devices
US09/612,587 Expired - Lifetime US6399389B1 (en) 1996-06-28 2000-07-07 High throughput screening assay systems in microscale fluidic devices
US09/718,235 Expired - Lifetime US6558960B1 (en) 1996-06-28 2000-11-21 High throughput screening assay systems in microscale fluidic devices
US09/718,236 Expired - Lifetime US6630353B1 (en) 1996-06-28 2000-11-21 High throughput screening assay systems in microscale fluidic devices
US10/279,496 Expired - Lifetime US7091048B2 (en) 1996-06-28 2002-10-24 High throughput screening assay systems in microscale fluidic devices
US10/637,730 Abandoned US20040028567A1 (en) 1996-06-28 2003-08-07 High throughput screening assay systems in microscale fluidic devices
US10/821,503 Expired - Fee Related US7067263B2 (en) 1996-06-28 2004-04-08 High throughput screening assay systems in microscale fluidic devices
US11/176,805 Abandoned US20050241941A1 (en) 1996-06-28 2005-07-06 High throughput screening assay systems in microscale fluidic devices

Family Applications Before (10)

Application Number Title Priority Date Filing Date
US09/214,035 Expired - Lifetime US6429025B1 (en) 1996-06-28 1997-06-24 High-throughput screening assay systems in microscale fluidic devices
US08/881,696 Expired - Lifetime US6267858B1 (en) 1996-06-28 1997-06-24 High throughput screening assay systems in microscale fluidic devices
US09/044,587 Expired - Lifetime US6413782B1 (en) 1996-06-28 1998-03-19 Methods of manufacturing high-throughput screening systems
US09/044,557 Expired - Lifetime US6274337B1 (en) 1996-06-28 1998-03-19 High throughput screening assay systems in microscale fluidic devices
US09/132,963 Expired - Lifetime US6479299B1 (en) 1996-06-28 1998-08-12 Pre-disposed assay components in microfluidic devices and methods
US09/196,535 Expired - Lifetime US6306659B1 (en) 1996-06-28 1998-11-20 High throughput screening assay systems in microscale fluidic devices
US09/612,587 Expired - Lifetime US6399389B1 (en) 1996-06-28 2000-07-07 High throughput screening assay systems in microscale fluidic devices
US09/718,235 Expired - Lifetime US6558960B1 (en) 1996-06-28 2000-11-21 High throughput screening assay systems in microscale fluidic devices
US09/718,236 Expired - Lifetime US6630353B1 (en) 1996-06-28 2000-11-21 High throughput screening assay systems in microscale fluidic devices
US10/279,496 Expired - Lifetime US7091048B2 (en) 1996-06-28 2002-10-24 High throughput screening assay systems in microscale fluidic devices

Family Applications After (2)

Application Number Title Priority Date Filing Date
US10/821,503 Expired - Fee Related US7067263B2 (en) 1996-06-28 2004-04-08 High throughput screening assay systems in microscale fluidic devices
US11/176,805 Abandoned US20050241941A1 (en) 1996-06-28 2005-07-06 High throughput screening assay systems in microscale fluidic devices

Country Status (8)

Country Link
US (13) US6429025B1 (en)
EP (1) EP0907412B1 (en)
JP (1) JP3788519B2 (en)
CN (1) CN1173776C (en)
BR (1) BR9710054A (en)
CA (1) CA2258489C (en)
NZ (1) NZ333346A (en)
WO (1) WO1998000231A1 (en)

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020108860A1 (en) * 2001-01-15 2002-08-15 Staats Sau Lan Tang Fabrication of polymeric microfluidic devices
US20030124736A1 (en) * 1998-08-05 2003-07-03 Caliper Technologies Corp. Open-field serial to parallel converter
US20040053422A1 (en) * 2002-09-17 2004-03-18 Selena Chan Microfluidic devices with porous membranes for molecular sieving, metering, and separations
WO2005114223A3 (en) * 2004-05-21 2006-05-04 Caliper Life Sciences Inc Automat system for handling microfluidic devices
US20070018116A1 (en) * 2005-07-21 2007-01-25 Felix Lustenberger Apparatus and method for all-solid-state fluorescence lifetime imaging
US20070243523A1 (en) * 2006-03-31 2007-10-18 Fluxion Biosciences Inc. Methods and Apparatus for the Manipulation of Particle Suspensions and Testing Thereof
US20090101559A1 (en) * 2005-01-21 2009-04-23 Anand Bala Subramaniam Microconcentrator/Microfilter
US20110101241A1 (en) * 2004-07-26 2011-05-05 Mesa Imaging Ag Solid-State Photodetector Pixel and Photodetecting Method
US10167502B2 (en) 2015-04-03 2019-01-01 Fluxion Biosciences, Inc. Molecular characterization of single cells and cell populations for non-invasive diagnostics

Families Citing this family (663)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5770029A (en) * 1996-07-30 1998-06-23 Soane Biosciences Integrated electrophoretic microdevices
US6048734A (en) 1995-09-15 2000-04-11 The Regents Of The University Of Michigan Thermal microvalves in a fluid flow method
US5885470A (en) * 1997-04-14 1999-03-23 Caliper Technologies Corporation Controlled fluid transport in microfabricated polymeric substrates
US5942443A (en) 1996-06-28 1999-08-24 Caliper Technologies Corporation High throughput screening assay systems in microscale fluidic devices
DE69737883T2 (en) 1996-04-25 2008-03-06 Bioarray Solutions Ltd. LIGHT-REGULATED, ELECTROKINETIC COMPOSITION OF PARTICLES TO SURFACES
BR9710054A (en) 1996-06-28 2000-01-11 Caliper Techn Corp Apparatus for separating test compounds for an effect on a biochemical system and for detecting a effect of a test compound on a biochemical system, procedures for determining whether a sample contains a compound capable of affecting a biochemical system, for separating a plurality of test compounds for an effect on a biochemical system and uses of a microfluidic system and a test substrate.
CN1329729C (en) * 1996-06-28 2007-08-01 卡钳生命科学股份有限公司 Electropipettor and compensation means for electrophoretic bias
US6235471B1 (en) 1997-04-04 2001-05-22 Caliper Technologies Corp. Closed-loop biochemical analyzers
US6391622B1 (en) 1997-04-04 2002-05-21 Caliper Technologies Corp. Closed-loop biochemical analyzers
AU760310B2 (en) * 1997-04-14 2003-05-15 Caliper Life Sciences, Inc. Controlled fluid transport in microfabricated polymeric substrates
AU7121198A (en) * 1997-04-18 1998-11-13 California Institute Of Technology Multifunctional polymeric tissue coatings
US7033474B1 (en) 1997-04-25 2006-04-25 Caliper Life Sciences, Inc. Microfluidic devices incorporating improved channel geometries
EP0988529B1 (en) * 1997-04-25 2013-06-12 Caliper Life Sciences, Inc. Microfluidic devices incorporating improved channel geometries
US5976336A (en) 1997-04-25 1999-11-02 Caliper Technologies Corp. Microfluidic devices incorporating improved channel geometries
EP1021566A4 (en) 1997-06-09 2002-06-19 Caliper Techn Corp Apparatus and methods for correcting for variable velocity in microfluidic systems
US6001231A (en) 1997-07-15 1999-12-14 Caliper Technologies Corp. Methods and systems for monitoring and controlling fluid flow rates in microfluidic systems
US6907921B2 (en) 1998-06-18 2005-06-21 3M Innovative Properties Company Microchanneled active fluid heat exchanger
US6514412B1 (en) 1998-06-18 2003-02-04 3M Innovative Properties Company Microstructured separation device
US6375871B1 (en) * 1998-06-18 2002-04-23 3M Innovative Properties Company Methods of manufacturing microfluidic articles
US6431695B1 (en) 1998-06-18 2002-08-13 3M Innovative Properties Company Microstructure liquid dispenser
US5989402A (en) 1997-08-29 1999-11-23 Caliper Technologies Corp. Controller/detector interfaces for microfluidic systems
US6540895B1 (en) 1997-09-23 2003-04-01 California Institute Of Technology Microfabricated cell sorter for chemical and biological materials
US6893877B2 (en) 1998-01-12 2005-05-17 Massachusetts Institute Of Technology Methods for screening substances in a microwell array
US6756019B1 (en) 1998-02-24 2004-06-29 Caliper Technologies Corp. Microfluidic devices and systems incorporating cover layers
US6251343B1 (en) 1998-02-24 2001-06-26 Caliper Technologies Corp. Microfluidic devices and systems incorporating cover layers
US7497994B2 (en) 1998-02-24 2009-03-03 Khushroo Gandhi Microfluidic devices and systems incorporating cover layers
DE19815882A1 (en) * 1998-04-08 1999-10-14 Fuhr Guenther Method and device for manipulating microparticles in fluid flows
US6123798A (en) 1998-05-06 2000-09-26 Caliper Technologies Corp. Methods of fabricating polymeric structures incorporating microscale fluidic elements
ATE530891T1 (en) * 1998-05-22 2011-11-15 California Inst Of Techn MINIATURIZED CELL SORTER
US6274089B1 (en) 1998-06-08 2001-08-14 Caliper Technologies Corp. Microfluidic devices, systems and methods for performing integrated reactions and separations
CA2332919A1 (en) * 1998-06-08 1999-12-16 Caliper Technologies Corporation Microfluidic devices, systems and methods for performing integrated reactions and separations
US6306590B1 (en) * 1998-06-08 2001-10-23 Caliper Technologies Corp. Microfluidic matrix localization apparatus and methods
US6149882A (en) 1998-06-09 2000-11-21 Symyx Technologies, Inc. Parallel fixed bed reactor and fluid contacting apparatus
WO1999067639A1 (en) * 1998-06-25 1999-12-29 Caliper Technologies Corporation High throughput methods, systems and apparatus for performing cell based screening assays
US7155344B1 (en) 1998-07-27 2006-12-26 Caliper Life Sciences, Inc. Distributed database for analytical instruments
US6132685A (en) * 1998-08-10 2000-10-17 Caliper Technologies Corporation High throughput microfluidic systems and methods
US6716394B2 (en) 1998-08-11 2004-04-06 Caliper Technologies Corp. DNA sequencing using multiple fluorescent labels being distinguishable by their decay times
US6447724B1 (en) 1998-08-11 2002-09-10 Caliper Technologies Corp. DNA sequencing using multiple fluorescent labels being distinguishable by their decay times
US6647358B2 (en) 1998-09-14 2003-11-11 Lion Bioscience Ag Pharmacokinetic-based drug design tool and method
AU6216199A (en) 1998-10-08 2000-04-26 Astrazeneca Ab Microfabricated cell injector
WO2000022426A1 (en) * 1998-10-09 2000-04-20 Hitachi, Ltd. Capillary electrophoretic system, sample analyzer and liquid sample cassette for electrophoretic separation
GB9822185D0 (en) * 1998-10-13 1998-12-02 Zeneca Ltd Device
US6149787A (en) 1998-10-14 2000-11-21 Caliper Technologies Corp. External material accession systems and methods
AU2003204460B2 (en) * 1998-10-14 2005-11-10 Caliper Life Sciences, Inc. Microfluidic controller and detector system with self-calibration
US6498497B1 (en) * 1998-10-14 2002-12-24 Caliper Technologies Corp. Microfluidic controller and detector system with self-calibration
NZ511560A (en) * 1998-11-05 2002-11-26 Chemometec As A method for the assessment of particles and a system and a device for use in the method
US6150119A (en) * 1999-01-19 2000-11-21 Caliper Technologies Corp. Optimized high-throughput analytical system
US6416642B1 (en) 1999-01-21 2002-07-09 Caliper Technologies Corp. Method and apparatus for continuous liquid flow in microscale channels using pressure injection, wicking, and electrokinetic injection
JP3441993B2 (en) * 1999-01-27 2003-09-02 松下電器産業株式会社 Cholesterol sensor
US20020019059A1 (en) 1999-01-28 2002-02-14 Calvin Y.H. Chow Devices, systems and methods for time domain multiplexing of reagents
WO2000050642A1 (en) 1999-02-23 2000-08-31 Caliper Technologies Corp. Sequencing by incorporation
US7150994B2 (en) 1999-03-03 2006-12-19 Symyx Technologies, Inc. Parallel flow process optimization reactor
DE19910392B4 (en) * 1999-03-05 2005-03-17 Clondiag Chip Technologies Gmbh Micro column reactor
US6326083B1 (en) 1999-03-08 2001-12-04 Calipher Technologies Corp. Surface coating for microfluidic devices that incorporate a biopolymer resistant moiety
US6303343B1 (en) 1999-04-06 2001-10-16 Caliper Technologies Corp. Inefficient fast PCR
AU4128500A (en) * 1999-04-09 2000-11-14 University Of Manchester Institute Of Science & Technology, The Electrophoresis method and apparatus with time- or space-modulated sample injection
US6322683B1 (en) * 1999-04-14 2001-11-27 Caliper Technologies Corp. Alignment of multicomponent microfabricated structures
AU769571B2 (en) * 1999-04-28 2004-01-29 Universitat Zurich Polyionic coatings in analytic and sensor devices
US6458259B1 (en) 1999-05-11 2002-10-01 Caliper Technologies Corp. Prevention of surface adsorption in microchannels by application of electric current during pressure-induced flow
WO2000070353A2 (en) * 1999-05-13 2000-11-23 Caliper Technologies Corp. High throughput assay systems and methods
WO2000070080A1 (en) 1999-05-17 2000-11-23 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
US6472141B2 (en) 1999-05-21 2002-10-29 Caliper Technologies Corp. Kinase assays using polycations
US6287774B1 (en) 1999-05-21 2001-09-11 Caliper Technologies Corp. Assay methods and system
WO2000073799A1 (en) * 1999-06-01 2000-12-07 Caliper Technologies Corp. Microscale assays and microfluidic devices for transporter, gradient induced, and binding activities
DE19927534B4 (en) * 1999-06-16 2008-01-31 Merck Patent Gmbh Apparatus for sample application
JP2001004628A (en) * 1999-06-18 2001-01-12 Kanagawa Acad Of Sci & Technol Immunoassay and its method
GB2351245B (en) * 1999-06-21 2003-07-16 Univ Hull Method of controlling liquid movement in a chemical device
FR2795476B1 (en) * 1999-06-22 2001-07-27 Biomerieux Sa VALVE FOR DIRECTING A FLUID IN AN ANALYSIS CARD
DE19928412C2 (en) * 1999-06-22 2002-03-21 Agilent Technologies Inc Supply element for a laboratory microchip
US8709153B2 (en) 1999-06-28 2014-04-29 California Institute Of Technology Microfludic protein crystallography techniques
US7195670B2 (en) 2000-06-27 2007-03-27 California Institute Of Technology High throughput screening of crystallization of materials
US7052545B2 (en) * 2001-04-06 2006-05-30 California Institute Of Technology High throughput screening of crystallization of materials
DE19935433A1 (en) 1999-08-01 2001-03-01 Febit Ferrarius Biotech Gmbh Microfluidic reaction carrier
US7138254B2 (en) * 1999-08-02 2006-11-21 Ge Healthcare (Sv) Corp. Methods and apparatus for performing submicroliter reactions with nucleic acids or proteins
US6524456B1 (en) * 1999-08-12 2003-02-25 Ut-Battelle, Llc Microfluidic devices for the controlled manipulation of small volumes
DE60013848T2 (en) * 1999-08-12 2005-09-22 UT-Battelle, LLC., Oak Ridge MICROFLUID DEVICES FOR THE CONTROLLED HANDLING OF SMALL VOLUMES
US6858185B1 (en) 1999-08-25 2005-02-22 Caliper Life Sciences, Inc. Dilutions in high throughput systems with a single vacuum source
US6613581B1 (en) 1999-08-26 2003-09-02 Caliper Technologies Corp. Microfluidic analytic detection assays, devices, and integrated systems
US6613211B1 (en) 1999-08-27 2003-09-02 Aclara Biosciences, Inc. Capillary electrokinesis based cellular assays
US6468763B1 (en) 1999-09-29 2002-10-22 Caliper Technologies Corp. Optical detection of transmembrane potential changes
WO2001027253A1 (en) 1999-10-08 2001-04-19 Caliper Technologies Corp. Use of nernstein voltage sensitive dyes in measuring transmembrane voltage
DE19949551C2 (en) * 1999-10-14 2001-12-13 Agilent Technologies Inc Microfluidic microchip, energy supply device and method for operating a microfluidic microchip
US6908594B1 (en) * 1999-10-22 2005-06-21 Aclara Biosciences, Inc. Efficient microfluidic sealing
US6432290B1 (en) 1999-11-26 2002-08-13 The Governors Of The University Of Alberta Apparatus and method for trapping bead based reagents within microfluidic analysis systems
CA2290731A1 (en) 1999-11-26 2001-05-26 D. Jed Harrison Apparatus and method for trapping bead based reagents within microfluidic analysis system
US6271038B1 (en) 1999-12-15 2001-08-07 Glaxo Wellcome Inc. Methods for high throughout determination and ranking of formulations and solubility
ATE340025T1 (en) * 2000-01-06 2006-10-15 Caliper Life Sciences Inc APPARATUS AND METHODS FOR HIGH-THROUGHPUT SAMPLING AND ANALYSIS
AU2610101A (en) 2000-01-06 2001-07-16 Caliper Technologies Corporation Methods and systems for monitoring intracellular binding reactions
US6468761B2 (en) 2000-01-07 2002-10-22 Caliper Technologies, Corp. Microfluidic in-line labeling method for continuous-flow protease inhibition analysis
US7037416B2 (en) 2000-01-14 2006-05-02 Caliper Life Sciences, Inc. Method for monitoring flow rate using fluorescent markers
US6556923B2 (en) * 2000-01-26 2003-04-29 Caliper Technologies Corp. Software for high throughput microfluidic systems
US6589729B2 (en) 2000-02-04 2003-07-08 Caliper Technologies Corp. Methods, devices, and systems for monitoring time dependent reactions
DE60032113T2 (en) * 2000-02-11 2007-06-28 Stmicroelectronics S.R.L., Agrate Brianza Integrated device for microfluidic temperature control and its manufacturing method
AU3499701A (en) * 2000-02-11 2001-08-20 Aclara Biosciences Inc Microfluidic device with sample injector and method
US7332271B2 (en) * 2000-02-18 2008-02-19 Board Of Trustees Of The Leland Stanford Junior University Apparatus and methods for parallel processing of micro-volume liquid reactions
US20020151040A1 (en) * 2000-02-18 2002-10-17 Matthew O' Keefe Apparatus and methods for parallel processing of microvolume liquid reactions
US6681616B2 (en) 2000-02-23 2004-01-27 Caliper Technologies Corp. Microfluidic viscometer
US7040144B2 (en) * 2000-02-23 2006-05-09 Caliper Life Sciences, Inc. Microfluidic viscometer
AU4907101A (en) 2000-02-23 2001-09-03 Caliper Technologies Inc Multi-reservoir pressure control system
US7230315B2 (en) * 2000-02-29 2007-06-12 Stmicroelectronics S.R.L. Integrated chemical microreactor with large area channels and manufacturing process thereof
US7732192B2 (en) * 2000-02-29 2010-06-08 Stmicroelectronics S.R.L. Integrated chemical microreactor with large area channels and manufacturing process thereof
US7452713B2 (en) * 2000-02-29 2008-11-18 Stmicroelectronics S.R.L. Process for manufacturing a microfluidic device with buried channels
DE60108482T2 (en) 2000-03-07 2006-02-16 Symyx Technologies, Inc., Santa Clara PROCESS OPTIMIZING REACTOR WITH PARALLEL FLOW
US7485454B1 (en) 2000-03-10 2009-02-03 Bioprocessors Corp. Microreactor
JP4648513B2 (en) * 2000-03-15 2011-03-09 関東化学株式会社 Concentration detection method, concentration detection device, and drug dilution blending device
US20020012971A1 (en) 2000-03-20 2002-01-31 Mehta Tammy Burd PCR compatible nucleic acid sieving medium
JP2001340753A (en) * 2000-03-29 2001-12-11 Sumitomo Chem Co Ltd Reaction method and reactor
US7867763B2 (en) 2004-01-25 2011-01-11 Fluidigm Corporation Integrated chip carriers with thermocycler interfaces and methods of using the same
US6436722B1 (en) * 2000-04-18 2002-08-20 Idexx Laboratories, Inc. Device and method for integrated diagnostics with multiple independent flow paths
US6733645B1 (en) 2000-04-18 2004-05-11 Caliper Technologies Corp. Total analyte quantitation
AU5935501A (en) 2000-05-03 2001-11-12 Caliper Techn Corp Multi depth substrate fabrication processes
EP1281002A4 (en) 2000-05-11 2006-08-09 Caliper Life Sciences Inc Microfluidic devices and methods to regulate hydrodynamic and electrical resistance utilizing bulk viscosity enhancers
CA2407141A1 (en) 2000-05-12 2001-11-22 Caliper Technologies Corporation Detection of nucleic acid hybridization by fluorescence polarization
US6635487B1 (en) 2000-05-17 2003-10-21 Caliper Technologies Corp. Fluorescence standard for use in microfluidic instruments
AU2001275138A1 (en) * 2000-06-02 2001-12-17 The University Of Utah Research Foundation Active needle devices with integrated functionality
DE60023464T2 (en) * 2000-06-05 2006-07-20 Stmicroelectronics S.R.L., Agrate Brianza Process for the production of integrated chemical microreactors made of semiconductor material and integrated microreactor
WO2001098785A2 (en) * 2000-06-19 2001-12-27 Arizona Board Of Regents Rapid flow-based immunoassay microchip
US9709559B2 (en) 2000-06-21 2017-07-18 Bioarray Solutions, Ltd. Multianalyte molecular analysis using application-specific random particle arrays
EP1311839B1 (en) 2000-06-21 2006-03-01 Bioarray Solutions Ltd Multianalyte molecular analysis using application-specific random particle arrays
US6885982B2 (en) 2000-06-27 2005-04-26 Fluidigm Corporation Object oriented microfluidic design method and system
AU2001273057A1 (en) * 2000-06-27 2002-01-08 Fluidigm Corporation A microfluidic design automation method and system
US6627159B1 (en) * 2000-06-28 2003-09-30 3M Innovative Properties Company Centrifugal filling of sample processing devices
JP2002022752A (en) * 2000-07-13 2002-01-23 Suzuki Motor Corp Specimen testing device
AU2001284700B2 (en) 2000-08-03 2005-12-08 Caliper Life Sciences, Inc. Methods and devices for high throughput fluid delivery
US6670153B2 (en) * 2000-09-14 2003-12-30 Caliper Technologies Corp. Microfluidic devices and methods for performing temperature mediated reactions
EP1193214B1 (en) * 2000-09-27 2007-01-03 STMicroelectronics S.r.l. Integrated chemical microreactor, thermally insulated from detection electrodes, and manufacturing method therefor
US20100261159A1 (en) 2000-10-10 2010-10-14 Robert Hess Apparatus for assay, synthesis and storage, and methods of manufacture, use, and manipulation thereof
US6716629B2 (en) * 2000-10-10 2004-04-06 Biotrove, Inc. Apparatus for assay, synthesis and storage, and methods of manufacture, use, and manipulation thereof
US20090118139A1 (en) 2000-11-07 2009-05-07 Caliper Life Sciences, Inc. Microfluidic method and system for enzyme inhibition activity screening
US20050221385A1 (en) * 2000-11-07 2005-10-06 Caliper Life Sciences, Inc. Pressure based mobility shift assays
US7105304B1 (en) 2000-11-07 2006-09-12 Caliper Life Sciences, Inc. Pressure-based mobility shift assays
US8097471B2 (en) * 2000-11-10 2012-01-17 3M Innovative Properties Company Sample processing devices
EP1343973B2 (en) 2000-11-16 2020-09-16 California Institute Of Technology Apparatus and methods for conducting assays and high throughput screening
CA2364132C (en) * 2000-12-12 2010-06-01 Bayer Corporation Method of making a capillary channel
US7070682B2 (en) 2001-01-16 2006-07-04 Cheng Lee Microfluidic apparatus for performing gel protein extractions and methods for using the apparatus
US6875403B2 (en) * 2001-02-09 2005-04-05 Microchem Solutions Method and apparatus for reproducible sample injection on microfabricated devices
AU2002256998A1 (en) * 2001-02-09 2002-09-19 Microchem Solutions Apparatus and method for small-volume fluid manipulation and transportation
US6692700B2 (en) 2001-02-14 2004-02-17 Handylab, Inc. Heat-reduction methods and systems related to microfluidic devices
US6913697B2 (en) * 2001-02-14 2005-07-05 Science & Technology Corporation @ Unm Nanostructured separation and analysis devices for biological membranes
JP2004530894A (en) * 2001-02-15 2004-10-07 カリパー・ライフ・サイエンシズ・インコーポレーテッド Microfluidic system with enhanced detection system
US7016560B2 (en) * 2001-02-28 2006-03-21 Lightwave Microsystems Corporation Microfluidic control for waveguide optical switches, variable attenuators, and other optical devices
WO2002068821A2 (en) 2001-02-28 2002-09-06 Lightwave Microsystems Corporation Microfluidic control using dieletric pumping
US6949377B2 (en) * 2001-03-05 2005-09-27 Ho Winston Z Chemiluminescence-based microfluidic biochip
US20020127740A1 (en) * 2001-03-06 2002-09-12 Ho Winston Z. Quantitative microfluidic biochip and method of use
US7118917B2 (en) 2001-03-07 2006-10-10 Symyx Technologies, Inc. Parallel flow reactor having improved thermal control
EP1372848A4 (en) * 2001-03-09 2006-08-09 Biomicro Systems Inc Method and system for microfluidic interfacing to arrays
US7429354B2 (en) 2001-03-19 2008-09-30 Gyros Patent Ab Structural units that define fluidic functions
US7759067B2 (en) 2001-03-19 2010-07-20 Gyros Patent Ab Method for determining the amount of an analyte with a disc-shaped microfluidic device
US6852287B2 (en) 2001-09-12 2005-02-08 Handylab, Inc. Microfluidic devices having a reduced number of input and output connections
US7829025B2 (en) 2001-03-28 2010-11-09 Venture Lending & Leasing Iv, Inc. Systems and methods for thermal actuation of microfluidic devices
US7010391B2 (en) 2001-03-28 2006-03-07 Handylab, Inc. Methods and systems for control of microfluidic devices
US7323140B2 (en) 2001-03-28 2008-01-29 Handylab, Inc. Moving microdroplets in a microfluidic device
US8895311B1 (en) 2001-03-28 2014-11-25 Handylab, Inc. Methods and systems for control of general purpose microfluidic devices
US7314718B1 (en) 2001-04-03 2008-01-01 Bioarray Solutions Ltd. Method and apparatus for maintaining multiple planar fluid flows
AU2002307152A1 (en) 2001-04-06 2002-10-21 California Institute Of Technology Nucleic acid amplification utilizing microfluidic devices
JP2004527247A (en) * 2001-04-10 2004-09-09 バイオプロセッサーズ コーポレイション Microfermentor devices and cell-based screening methods
US7476533B2 (en) * 2002-04-19 2009-01-13 Adhesives Research, Inc. Diagnostic devices for use in the assaying of biological fluids
EP2214015B2 (en) * 2001-04-19 2023-12-27 Adhesives Research, Inc. Hydrophilic diagnostic devices for use in the assaying of biological fluids
US6727479B2 (en) * 2001-04-23 2004-04-27 Stmicroelectronics S.R.L. Integrated device based upon semiconductor technology, in particular chemical microreactor
US7641780B2 (en) 2001-05-01 2010-01-05 Calibrant Biosystems, Inc. Two-dimensional microfluidics for protein separations and gene analysis
US6974526B2 (en) * 2001-05-01 2005-12-13 Calibrant Biosystems, Inc. Plastic microfluidics enabling two-dimensional protein separations in proteome analysis
US6929730B2 (en) * 2001-05-01 2005-08-16 Cheng Sheng Lee Two dimensional microfluidic gene scanner
JP4566456B2 (en) * 2001-05-31 2010-10-20 独立行政法人理化学研究所 Trace liquid control mechanism and trace liquid control method
US7723123B1 (en) 2001-06-05 2010-05-25 Caliper Life Sciences, Inc. Western blot by incorporating an affinity purification zone
US20030077570A1 (en) * 2001-09-20 2003-04-24 Coventor, Inc. Small molecule substrate based enzyme activity assays
US7262063B2 (en) 2001-06-21 2007-08-28 Bio Array Solutions, Ltd. Directed assembly of functional heterostructures
US20050149304A1 (en) * 2001-06-27 2005-07-07 Fluidigm Corporation Object oriented microfluidic design method and system
CA2652991A1 (en) * 2001-07-16 2003-11-13 Caprotec Bioanalytics Gmbh Capture compounds, collections thereof and methods for analyzing the proteome and complex compositions
US20060062696A1 (en) 2001-07-27 2006-03-23 Caliper Life Sciences, Inc. Optimized high throughput analytical systems
US7019831B2 (en) * 2001-08-24 2006-03-28 Applera Corporation Separation device substrate including non-fluorescent quencher dye
US7666661B2 (en) 2001-08-27 2010-02-23 Platypus Technologies, Llc Substrates, devices, and methods for quantitative liquid crystal assays
WO2003025115A1 (en) * 2001-09-20 2003-03-27 Cytonome, Inc. Small molecule substrate based enzyme activity assays
ES2661167T3 (en) 2001-10-15 2018-03-27 Bioarray Solutions Ltd. Multiplexed analysis of polymorphic loci by simultaneous consultation and enzyme-mediated detection
DE60200822T2 (en) * 2001-10-18 2005-09-15 Aida Engineering, Ltd., Sagamihara Microdosing and sampling device and microchip with this device
US20030082632A1 (en) * 2001-10-25 2003-05-01 Cytoprint, Inc. Assay method and apparatus
US8440093B1 (en) 2001-10-26 2013-05-14 Fuidigm Corporation Methods and devices for electronic and magnetic sensing of the contents of microfluidic flow channels
US6882500B2 (en) * 2001-10-26 2005-04-19 Seagate Technology Llc Airflow channel within a disc drive housing
US20030085952A1 (en) * 2001-11-05 2003-05-08 Williams Roger O Apparatus and method for controlling the free surface of liquid in a well plate
SE526185C2 (en) * 2001-11-07 2005-07-19 Prolight Diagnostics Ab Method and apparatus for immunoassay
WO2003042697A1 (en) * 2001-11-14 2003-05-22 Genospectra, Inc. Biochemical analysis system with combinatorial chemistry applications
US20030098661A1 (en) * 2001-11-29 2003-05-29 Ken Stewart-Smith Control system for vehicle seats
JP4355210B2 (en) 2001-11-30 2009-10-28 フルイディグム コーポレイション Microfluidic device and method of using microfluidic device
US7691333B2 (en) 2001-11-30 2010-04-06 Fluidigm Corporation Microfluidic device and methods of using same
US6889468B2 (en) 2001-12-28 2005-05-10 3M Innovative Properties Company Modular systems and methods for using sample processing devices
US7470518B2 (en) * 2002-02-12 2008-12-30 Cellectricon Ab Systems and method for rapidly changing the solution environment around sensors
EP2347824A3 (en) * 2002-02-12 2012-03-07 Cellectricon Ab Systems and methods for rapidly changing the solution environment around sensors
US6814859B2 (en) * 2002-02-13 2004-11-09 Nanostream, Inc. Frit material and bonding method for microfluidic separation devices
JP3933058B2 (en) 2002-02-25 2007-06-20 日立化成工業株式会社 Support unit for microfluidic system and method for manufacturing the same
US7419821B2 (en) 2002-03-05 2008-09-02 I-Stat Corporation Apparatus and methods for analyte measurement and immunoassay
EP2581739B1 (en) 2002-03-05 2015-11-04 Caliper Life Sciences, Inc. Microfluidic separation method with combined pressure and voltage control
ATE421088T1 (en) * 2002-03-11 2009-01-15 Caprotec Bioanalytics Gmbh COMPOUNDS AND METHODS FOR ANALYZING THE PROTEOME
US20030215941A1 (en) * 2002-03-12 2003-11-20 Stewart Campbell Assay device that analyzes the absorption, metabolism, permeability and/or toxicity of a candidate compound
US7252928B1 (en) 2002-03-12 2007-08-07 Caliper Life Sciences, Inc. Methods for prevention of surface adsorption of biological materials to capillary walls in microchannels
US7524462B2 (en) * 2002-03-29 2009-04-28 Agilent Technologies, Inc. Capillary flow for a heterogenous assay in a micro-channel environment
WO2003085379A2 (en) * 2002-04-01 2003-10-16 Fluidigm Corporation Microfluidic particle-analysis systems
US9943847B2 (en) 2002-04-17 2018-04-17 Cytonome/St, Llc Microfluidic system including a bubble valve for regulating fluid flow through a microchannel
AU2003225281A1 (en) 2002-04-30 2003-11-17 University Of South Florida Materials and methods for prevention and treatment of rna viral diseases
EP2302389B1 (en) * 2002-05-09 2018-01-24 The University of Chicago Device and method for pressure-driven plug transport and reaction
US20030208936A1 (en) * 2002-05-09 2003-11-13 Lee Charles Hee Method for manufacturing embroidery decorated cards and envelopes
CA2486812A1 (en) * 2002-05-22 2004-05-21 Platypus Technologies, Llc Substrates, devices, and methods for cellular assays
US20030228566A1 (en) * 2002-06-11 2003-12-11 Biotechplex Corporation Method of and apparatus for screening for drug candidates
JP2006507921A (en) 2002-06-28 2006-03-09 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 Method and apparatus for fluid dispersion
US7517440B2 (en) 2002-07-17 2009-04-14 Eksigent Technologies Llc Electrokinetic delivery systems, devices and methods
US7235164B2 (en) * 2002-10-18 2007-06-26 Eksigent Technologies, Llc Electrokinetic pump having capacitive electrodes
US7364647B2 (en) * 2002-07-17 2008-04-29 Eksigent Technologies Llc Laminated flow device
US7214348B2 (en) 2002-07-26 2007-05-08 Applera Corporation Microfluidic size-exclusion devices, systems, and methods
EP1535061A2 (en) 2002-08-21 2005-06-01 Shell Internationale Researchmaatschappij B.V. Method for measuring fluid chemistry in drilling and production operations
US8277753B2 (en) 2002-08-23 2012-10-02 Life Technologies Corporation Microfluidic transfer pin
US7534601B2 (en) * 2002-08-27 2009-05-19 Vanderbilt University Capillary perfused bioreactors with multiple chambers
WO2004020342A2 (en) * 2002-08-27 2004-03-11 Vanderbilt University Capillary perfused bioreactors with multiple chambers
US7790443B2 (en) * 2002-08-27 2010-09-07 Vanderbilt University Bioreactors with substance injection capacity
US7595303B1 (en) 2002-09-05 2009-09-29 University Of South Florida Genetic adjuvants for immunotherapy
US7094345B2 (en) * 2002-09-09 2006-08-22 Cytonome, Inc. Implementation of microfluidic components, including molecular fractionation devices, in a microfluidic system
ITTO20020808A1 (en) * 2002-09-17 2004-03-18 St Microelectronics Srl INTEGRATED DNA ANALYSIS DEVICE.
ITTO20020809A1 (en) * 2002-09-17 2004-03-18 St Microelectronics Srl MICROPUMP, IN PARTICULAR FOR AN INTEGRATED DNA ANALYSIS DEVICE.
US7960184B2 (en) * 2002-09-20 2011-06-14 George Mason Intellectual Properties, Inc. Methods and devices for active bioassay
US20040115830A1 (en) * 2002-09-25 2004-06-17 Igor Touzov Components for nano-scale Reactor
EP2213615A3 (en) 2002-09-25 2012-02-29 California Institute of Technology Microfluidic Large Scale Integration
ES2375724T3 (en) 2002-09-27 2012-03-05 The General Hospital Corporation MICROFLUDE DEVICE FOR SEPERATION OF CELLS AND ITS USES.
JP5695287B2 (en) 2002-10-02 2015-04-01 カリフォルニア インスティテュート オブ テクノロジー Nucleic acid analysis of microfluids
US8232074B2 (en) 2002-10-16 2012-07-31 Cellectricon Ab Nanoelectrodes and nanotips for recording transmembrane currents in a plurality of cells
US20060183106A1 (en) * 2002-10-18 2006-08-17 Adam Siddiqui-Jain Processes for identifying quadruplex-targeted antiviral molecules
US7179357B2 (en) * 2002-10-25 2007-02-20 Beckman Coulter, Inc. Serial sample injection in capillary electrophoresis
GB2395006A (en) * 2002-10-29 2004-05-12 Micro Chemical Systems Ltd Apparatus and method for performing an assay
JP4346893B2 (en) * 2002-11-01 2009-10-21 株式会社日立製作所 Chemical reactor
ATE364840T1 (en) * 2002-11-04 2007-07-15 Transform Pharmaceuticals Inc ANALYSIS OF PHARMACEUTICAL SOLUBILITY AND STABILITY
US7108775B2 (en) * 2002-11-08 2006-09-19 Applera Corporation Apparatus and method for confining eluted samples in electrophoresis systems
KR20020097093A (en) * 2002-11-09 2002-12-31 신세현 Natural Convection Microfluidic Mixer
AU2003298655A1 (en) 2002-11-15 2004-06-15 Bioarray Solutions, Ltd. Analysis, secure access to, and transmission of array images
US6759713B2 (en) * 2002-11-22 2004-07-06 Xerox Corporation Electronic device structures including interconnecting microfluidic channels
AU2003293399A1 (en) * 2002-12-04 2004-06-23 Spinx, Inc. Devices and methods for programmable microscale manipulation of fluids
WO2004052540A2 (en) * 2002-12-05 2004-06-24 Protasis Corporation Configurable microfluidic substrate assembly
US7507376B2 (en) * 2002-12-19 2009-03-24 3M Innovative Properties Company Integrated sample processing devices
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
US20060094108A1 (en) * 2002-12-20 2006-05-04 Karl Yoder Thermal cycler for microfluidic array assays
US20050042639A1 (en) 2002-12-20 2005-02-24 Caliper Life Sciences, Inc. Single molecule amplification and detection of DNA length
CA2510166A1 (en) 2002-12-20 2004-09-30 Caliper Life Sciences, Inc. Single molecule amplification and detection of dna
US7682565B2 (en) 2002-12-20 2010-03-23 Biotrove, Inc. Assay apparatus and method using microfluidic arrays
DE50310049D1 (en) * 2002-12-23 2008-08-07 Febit Biotech Gmbh INSTALLATION OF HAPTEN GROUPS IN THE MANUFACTURE OF SUPPORTS FOR ANALYTICAL ASSESSMENT
EP1594694A4 (en) * 2002-12-30 2010-01-20 Univ California Methods and apparatus for pathogen detection and analysis
US20050118704A1 (en) * 2002-12-31 2005-06-02 Brana Malobabic System and method for real-time detection and remote monitoring of pathogens
US20040129676A1 (en) * 2003-01-07 2004-07-08 Tan Roy H. Apparatus for transfer of an array of liquids and methods for manufacturing same
US7122153B2 (en) * 2003-01-08 2006-10-17 Ho Winston Z Self-contained microfluidic biochip and apparatus
US20050221281A1 (en) * 2003-01-08 2005-10-06 Ho Winston Z Self-contained microfluidic biochip and apparatus
US7332129B2 (en) * 2003-01-09 2008-02-19 3M Innovative Properties Company Sample processing device having process chambers with bypass slots
WO2004064972A2 (en) * 2003-01-16 2004-08-05 Hk Pharmaceuticals, Inc. Capture compounds, collections thereof and methods for analyzing the proteome and complex compositions
TW571101B (en) 2003-01-21 2004-01-11 Ind Tech Res Inst Fluid analysis apparatus
US20040150217A1 (en) * 2003-01-23 2004-08-05 Heffelfinger David M. Identifying indicia and focusing target
US7041481B2 (en) 2003-03-14 2006-05-09 The Regents Of The University Of California Chemical amplification based on fluid partitioning
US7329538B2 (en) * 2003-03-17 2008-02-12 Charles River Laboratories, Inc. Methods and compositions for the detection of microbial contaminants
SE0300823D0 (en) 2003-03-23 2003-03-23 Gyros Ab Preloaded Microscale Devices
SE0300822D0 (en) 2003-03-23 2003-03-23 Gyros Ab A collection of Micro Scale Devices
US20060078893A1 (en) 2004-10-12 2006-04-13 Medical Research Council Compartmentalised combinatorial chemistry by microfluidic control
EP1473084B1 (en) * 2003-03-31 2015-07-29 Canon Kabushiki Kaisha Biochemical reaction cartridge
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
US20050145496A1 (en) 2003-04-03 2005-07-07 Federico Goodsaid Thermal reaction device and method for using the same
US7476363B2 (en) 2003-04-03 2009-01-13 Fluidigm Corporation Microfluidic devices and methods of using same
JP5419248B2 (en) 2003-04-03 2014-02-19 フルイディグム コーポレイション Microfluidic device and method of use thereof
US7604965B2 (en) 2003-04-03 2009-10-20 Fluidigm Corporation Thermal reaction device and method for using the same
US8828663B2 (en) 2005-03-18 2014-09-09 Fluidigm Corporation Thermal reaction device and method for using the same
EP3023140B1 (en) * 2003-04-10 2019-10-09 President and Fellows of Harvard College Formation and control of fluidic species
US7422725B2 (en) * 2003-05-01 2008-09-09 Enplas Corporation Sample handling unit applicable to microchip, and microfluidic device having microchips
US7217396B2 (en) * 2003-05-05 2007-05-15 The Board Of Trustees Of The University Of Illinois Microfabricated micro fluid channels
DE10320869A1 (en) * 2003-05-09 2004-12-16 Evotec Technologies Gmbh Methods and devices for liquid treatment of suspended particles
US7038472B1 (en) 2003-05-12 2006-05-02 Caliper Life Sciences, Inc. Methods and systems for measuring internal dimensions of microscale structures
US20040228962A1 (en) * 2003-05-16 2004-11-18 Chang Liu Scanning probe microscopy probe and method for scanning probe contact printing
US7578916B2 (en) * 2004-05-25 2009-08-25 Applied Biosystems, Llc Emulsions of ionic liquids
US7572409B2 (en) * 2003-05-23 2009-08-11 Applied Biosystems, Llc Ionic liquid apparatus and method for biological samples
EP1629285A2 (en) * 2003-06-05 2006-03-01 Bioprocessors Corporation System and method for process automation
US20040248306A1 (en) * 2003-06-09 2004-12-09 Hernandez Juan J. Microfluidic water analytical device
US7736889B2 (en) * 2003-06-10 2010-06-15 The United States Of America As Represented By The Secretary Of The Navy Fluidic force discrimination
US7672791B2 (en) * 2003-06-13 2010-03-02 International Business Machines Corporation Method of performing three-dimensional molecular superposition and similarity searches in databases of flexible molecules
JP2005007529A (en) * 2003-06-19 2005-01-13 Dainippon Screen Mfg Co Ltd Micro fluid device and manufacturing method of micro fluid device
WO2004113491A1 (en) * 2003-06-20 2004-12-29 Nitto Denko Corporation Cell microchip
EP1654390B1 (en) 2003-07-25 2009-11-11 Platypus Technologies, LLC Liquid crystal based analyte detection
US7727752B2 (en) 2003-07-29 2010-06-01 Life Technologies Corporation Kinase and phosphatase assays
US20050103713A1 (en) * 2003-07-30 2005-05-19 Ramsey J. M. Devices with small-scale channels and the fabrication thereof by etching
EP1654066B1 (en) 2003-07-31 2014-11-12 Handylab, Inc. Processing particle-containing samples
US7413712B2 (en) 2003-08-11 2008-08-19 California Institute Of Technology Microfluidic rotary flow reactor matrix
US20050037508A1 (en) * 2003-08-12 2005-02-17 Juan Hernandez Microfluidic titration apparatus
KR101166180B1 (en) * 2003-08-13 2012-07-18 루미넥스 코포레이션 Methods for controlling one or more parameters of a flow cytometer type measurement system
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
WO2005074417A2 (en) * 2003-09-03 2005-08-18 Salk Institute For Biological Studies Multiple antigen detection assays and reagents
WO2005029705A2 (en) 2003-09-18 2005-03-31 Bioarray Solutions, Ltd. Number coding for identification of subtypes of coded types of solid phase carriers
US7695688B2 (en) * 2003-09-19 2010-04-13 Applied Biosystems, Llc High density plate filler
US20060233673A1 (en) * 2003-09-19 2006-10-19 Beard Nigel P High density plate filler
US7998435B2 (en) * 2003-09-19 2011-08-16 Life Technologies Corporation High density plate filler
US20060272738A1 (en) * 2003-09-19 2006-12-07 Gary Lim High density plate filler
US20050220675A1 (en) * 2003-09-19 2005-10-06 Reed Mark T High density plate filler
US9492820B2 (en) 2003-09-19 2016-11-15 Applied Biosystems, Llc High density plate filler
US8277760B2 (en) * 2003-09-19 2012-10-02 Applied Biosystems, Llc High density plate filler
US20050226782A1 (en) * 2003-09-19 2005-10-13 Reed Mark T High density plate filler
US7407630B2 (en) * 2003-09-19 2008-08-05 Applera Corporation High density plate filler
US20060233671A1 (en) * 2003-09-19 2006-10-19 Beard Nigel P High density plate filler
US20050237528A1 (en) * 2003-09-19 2005-10-27 Oldham Mark F Transparent heater for thermocycling
WO2005031305A2 (en) 2003-09-22 2005-04-07 Bioarray Solutions, Ltd. Surface immobilized polyelectrolyte with multiple functional groups capable of covalently bonding to biomolecules
US8268446B2 (en) 2003-09-23 2012-09-18 The University Of North Carolina At Chapel Hill Photocurable perfluoropolyethers for use as novel materials in microfluidic devices
US7166212B2 (en) * 2003-09-30 2007-01-23 Chromba, Inc. Multicapillary column for chromatography and sample preparation
US20050069949A1 (en) * 2003-09-30 2005-03-31 International Business Machines Corporation Microfabricated Fluidic Structures
US20050069462A1 (en) * 2003-09-30 2005-03-31 International Business Machines Corporation Microfluidics Packaging
US20070017870A1 (en) 2003-09-30 2007-01-25 Belov Yuri P Multicapillary device for sample preparation
US7718133B2 (en) * 2003-10-09 2010-05-18 3M Innovative Properties Company Multilayer processing devices and methods
EP1692298A4 (en) 2003-10-28 2008-08-13 Bioarray Solutions Ltd Optimization of gene expression analysis using immobilized capture probes
CN1882703B (en) 2003-10-29 2011-07-06 佰尔瑞溶液有限公司 Multiplexed nucleic acid analysis by fragmentation of double-stranded DNA
EP1687396A2 (en) 2003-11-10 2006-08-09 Platypus Technologies, LLC Substrates, devices, and methods for cellular assays
EP1535665A1 (en) * 2003-11-28 2005-06-01 STMicroelectronics S.r.l. Integrated chemical microreactor with separated channels for confining liquids inside the channels and manufacturing process thereof
EP1541991A1 (en) * 2003-12-12 2005-06-15 STMicroelectronics S.r.l. Integrated semiconductor chemical microreactor for real-time monitoring of biological reactions
KR100519672B1 (en) * 2003-12-22 2005-10-11 주식회사 디지탈바이오테크놀러지 Channel Apparatus for Focusing a Fluid Flow
US20050161327A1 (en) * 2003-12-23 2005-07-28 Michele Palmieri Microfluidic device and method for transporting electrically charged substances through a microchannel of a microfluidic device
EP1547688A1 (en) * 2003-12-23 2005-06-29 STMicroelectronics S.r.l. Microfluidic device and method of locally concentrating electrically charged substances in a microfluidic device
WO2005066613A1 (en) * 2003-12-31 2005-07-21 President And Fellows Of Harvard College Assay device and method
JP3991034B2 (en) * 2004-01-23 2007-10-17 キヤノン株式会社 Detection method
CA2554240A1 (en) * 2004-01-25 2005-08-11 Fluidigm Corporation Crystal forming devices and systems and methods for making and using the same
US8030057B2 (en) * 2004-01-26 2011-10-04 President And Fellows Of Harvard College Fluid delivery system and method
PT1776181E (en) * 2004-01-26 2014-01-02 Harvard College Fluid delivery system and method
SE0400181D0 (en) * 2004-01-29 2004-01-29 Gyros Ab Segmented porous and preloaded microscale devices
US8158728B2 (en) 2004-02-13 2012-04-17 The University Of North Carolina At Chapel Hill Methods and materials for fabricating microfluidic devices
US8058056B2 (en) * 2004-03-12 2011-11-15 The Regents Of The University Of California Method and apparatus for integrated cell handling and measurements
US8105554B2 (en) 2004-03-12 2012-01-31 Life Technologies Corporation Nanoliter array loading
US7432106B2 (en) * 2004-03-24 2008-10-07 Applied Biosystems Inc. Liquid processing device including gas trap, and system and method
US20050214737A1 (en) * 2004-03-26 2005-09-29 Dejneka Matthew J Transparent filtered capillaries
US20050221339A1 (en) 2004-03-31 2005-10-06 Medical Research Council Harvard University Compartmentalised screening by microfluidic control
KR100597870B1 (en) * 2004-04-01 2006-07-06 한국과학기술원 Microfluidic chip for high-throughput screening and high-throughput assay
US7559356B2 (en) * 2004-04-19 2009-07-14 Eksident Technologies, Inc. Electrokinetic pump driven heat transfer system
WO2005105136A1 (en) * 2004-04-27 2005-11-10 University Of South Florida Nanogene therapy for cell proliferation disorders
US8470586B2 (en) 2004-05-03 2013-06-25 Handylab, Inc. Processing polynucleotide-containing samples
US8852862B2 (en) 2004-05-03 2014-10-07 Handylab, Inc. Method for processing polynucleotide-containing samples
JP4683538B2 (en) 2004-05-06 2011-05-18 セイコーインスツル株式会社 Analysis system and analysis method including microchip for analysis
WO2005108571A1 (en) * 2004-05-07 2005-11-17 Konica Minolta Medical & Graphic, Inc. Micro-reactor for testing, genetic testing apparatus, and genetic testing method
US7799553B2 (en) 2004-06-01 2010-09-21 The Regents Of The University Of California Microfabricated integrated DNA analysis system
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
EP1779111B1 (en) * 2004-07-16 2010-07-07 Gyros Patent Ab Grading of immune responses
EP1618955B1 (en) * 2004-07-19 2010-12-22 STMicroelectronics Srl Biological molecules detection device having increased detection rate, and method for quick detection of biological molecules
JP4990771B2 (en) 2004-07-28 2012-08-01 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 Method for monitoring microbial genomic DNA
US7848889B2 (en) 2004-08-02 2010-12-07 Bioarray Solutions, Ltd. Automated analysis of multiplexed probe-target interaction patterns: pattern matching and allele identification
US20060105453A1 (en) 2004-09-09 2006-05-18 Brenan Colin J Coating process for microfluidic sample arrays
US7932090B2 (en) * 2004-08-05 2011-04-26 3M Innovative Properties Company Sample processing device positioning apparatus and methods
US7618524B1 (en) 2004-08-10 2009-11-17 Sandia Corporation Polymeric salt bridges for conducting electric current in microfluidic devices
US20060070880A1 (en) * 2004-08-31 2006-04-06 Applera Corporation Methods and apparatus for manipulating separation media
CN102759466A (en) 2004-09-15 2012-10-31 英特基因有限公司 Microfluidic devices
WO2006039293A2 (en) * 2004-09-29 2006-04-13 University Of Virginia Patent Foundation Localized control of thermal properties on microdevices and applications thereof
US7968287B2 (en) 2004-10-08 2011-06-28 Medical Research Council Harvard University In vitro evolution in microfluidic systems
US7674545B2 (en) * 2004-10-19 2010-03-09 Korea Institute Of Science & Technology Electrokinetic micro power cell using microfluidic-chip with multi-channel type
WO2006059649A1 (en) 2004-11-30 2006-06-08 Hitachi Chemical Co., Ltd. Component for preanalytical treatment
WO2006060739A2 (en) 2004-12-01 2006-06-08 Proteologics, Inc. Ubiquitin ligase assays and related reagents
WO2007078268A2 (en) 2004-12-02 2007-07-12 Charles River Laboratories, Inc. Methods and compositions for the detection and/or quantification of gram positive bacterial contaminants
US9260693B2 (en) 2004-12-03 2016-02-16 Cytonome/St, Llc Actuation of parallel microfluidic arrays
WO2006076617A2 (en) 2005-01-13 2006-07-20 Charles River Laboratories, Inc. Method for classifying a microorganism in a biological sample
EP1846567A4 (en) * 2005-01-20 2009-12-09 Univ California Cellular microarrays for screening differentiation factors
KR100738071B1 (en) * 2005-01-21 2007-07-12 삼성전자주식회사 A dielectrophoresis apparatus disposed of means for concentration gradient generation, method for separating a material and method for screening a suitable conditions for separating a material
CA2595840C (en) * 2005-01-25 2013-12-10 Massachusetts Institute Of Technology Electrokinetic concentration device and methods of use thereof
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
US20070054119A1 (en) * 2005-03-04 2007-03-08 Piotr Garstecki Systems and methods of forming particles
AU2006220816A1 (en) 2005-03-04 2006-09-14 President And Fellows Of Harvard College Method and apparatus for forming multiple emulsions
JP2008538077A (en) * 2005-03-16 2008-10-09 ユニバーシティ オブ シカゴ Micro fluidic system
JP4548174B2 (en) * 2005-03-24 2010-09-22 コニカミノルタエムジー株式会社 Microchip for inspection and inspection apparatus using the same
US20070196820A1 (en) 2005-04-05 2007-08-23 Ravi Kapur Devices and methods for enrichment and alteration of cells and other particles
US8501416B2 (en) 2005-04-19 2013-08-06 President And Fellows Of Harvard College Fluidic structures including meandering and wide channels
EP1896610A2 (en) * 2005-05-03 2008-03-12 Handylab, Inc. Lyophilized pellets
EP1888790A2 (en) * 2005-05-06 2008-02-20 The Regents of the University of California Microfluidic system for identifying or sizing individual particles passing through a channel
US20060264783A1 (en) * 2005-05-09 2006-11-23 Holmes Elizabeth A Systems and methods for monitoring pharmacological parameters
US8097222B2 (en) * 2005-05-12 2012-01-17 Stmicroelectronics, S.R.L. Microfluidic device with integrated micropump, in particular biochemical microreactor, and manufacturing method thereof
US8486629B2 (en) 2005-06-01 2013-07-16 Bioarray Solutions, Ltd. Creation of functionalized microparticle libraries by oligonucleotide ligation or elongation
US7263874B2 (en) * 2005-06-08 2007-09-04 Bioscale, Inc. Methods and apparatus for determining properties of a fluid
FR2890578B3 (en) * 2005-09-09 2007-11-30 Rhodia Chimie Sa MICROFLUIDIC FLOW DEVICE FOR DETERMINING PARAMETERS OF PHYSICAL AND / OR CHEMICAL TRANSFORMATION AND USE THEREOF
JP4892219B2 (en) * 2005-06-17 2012-03-07 凸版印刷株式会社 Reaction chip, reaction apparatus, and reaction chip manufacturing method
US7323660B2 (en) * 2005-07-05 2008-01-29 3M Innovative Properties Company Modular sample processing apparatus kits and modules
US7754474B2 (en) 2005-07-05 2010-07-13 3M Innovative Properties Company Sample processing device compression systems and methods
US7763210B2 (en) 2005-07-05 2010-07-27 3M Innovative Properties Company Compliant microfluidic sample processing disks
US9354156B2 (en) 2007-02-08 2016-05-31 Emd Millipore Corporation Microfluidic particle analysis method, device and system
US9637715B2 (en) 2005-07-07 2017-05-02 Emd Millipore Corporation Cell culture and invasion assay method and system
WO2007008609A2 (en) 2005-07-07 2007-01-18 The Regents Of The University Of California Methods and apparatus for cell culture array
US8257964B2 (en) 2006-01-04 2012-09-04 Cell ASIC Microwell cell-culture device and fabrication method
US9388374B2 (en) 2005-07-07 2016-07-12 Emd Millipore Corporation Microfluidic cell culture systems
US8921102B2 (en) 2005-07-29 2014-12-30 Gpb Scientific, Llc Devices and methods for enrichment and alteration of circulating tumor cells and other particles
AU2006279763A1 (en) * 2005-08-11 2007-02-22 Biotrove, Inc. Apparatus for assay, synthesis and storage, and methods of manufacture, use, and manipulation thereof
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
WO2007025129A2 (en) 2005-08-25 2007-03-01 Platypus Technologies, Llc. Compositions and liquid crystals
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
US7281419B2 (en) * 2005-09-21 2007-10-16 The Board Of Trustees Of The University Of Illinois Multifunctional probe array system
US8822206B2 (en) * 2005-10-11 2014-09-02 The Johns Hopkins University Device for high-throughput stimulation, immunostaining, and visualization of single cells
WO2007044888A2 (en) * 2005-10-11 2007-04-19 The Johns Hopkins Univerisity Microfluidic device and method for high-throughput cellular gradient and dose response studies
WO2007047336A2 (en) * 2005-10-12 2007-04-26 University Of Virginia Patent Foundation Integrated microfluidic analysis systems
US20090155894A1 (en) * 2005-10-17 2009-06-18 Soper Steven A Electrokinetic Thermal Cycler and Reactor
WO2007056113A2 (en) * 2005-11-02 2007-05-18 Cylene Pharmaceuticals, Inc. Methods for targeting quadruplex sequences
EP1957794B1 (en) 2005-11-23 2014-07-02 Eksigent Technologies, LLC Electrokinetic pump designs and drug delivery systems
US20100137163A1 (en) 2006-01-11 2010-06-03 Link Darren R Microfluidic Devices and Methods of Use in The Formation and Control of Nanoreactors
CA2640024A1 (en) * 2006-01-27 2007-08-09 President And Fellows Of Harvard College Fluidic droplet coalescence
WO2008030631A2 (en) 2006-02-03 2008-03-13 Microchip Biotechnologies, Inc. Microfluidic devices
WO2007098065A2 (en) * 2006-02-17 2007-08-30 The United States Of America As Represented By The Department Of Veterans Affairs Human sodium channel isoforms
US7815868B1 (en) 2006-02-28 2010-10-19 Fluidigm Corporation Microfluidic reaction apparatus for high throughput screening
EP1830186A1 (en) * 2006-03-01 2007-09-05 ETH Zürich High-throughput cell-based screening system
US7766033B2 (en) 2006-03-22 2010-08-03 The Regents Of The University Of California Multiplexed latching valves for microfluidic devices and processors
US7998708B2 (en) 2006-03-24 2011-08-16 Handylab, Inc. Microfluidic system for amplifying and detecting polynucleotides in parallel
US8088616B2 (en) 2006-03-24 2012-01-03 Handylab, Inc. Heater unit for microfluidic diagnostic system
US11806718B2 (en) 2006-03-24 2023-11-07 Handylab, Inc. Fluorescence detector for microfluidic diagnostic system
US11287421B2 (en) 2006-03-24 2022-03-29 Labrador Diagnostics Llc Systems and methods of sample processing and fluid control in a fluidic system
US10900066B2 (en) 2006-03-24 2021-01-26 Handylab, Inc. Microfluidic system for amplifying and detecting polynucleotides in parallel
EP1997888A4 (en) * 2006-03-24 2010-07-28 Toshiba Kk Nucleic acid detection cassette and nucleic acid detection apparatus
US8741230B2 (en) 2006-03-24 2014-06-03 Theranos, Inc. Systems and methods of sample processing and fluid control in a fluidic system
DK2001990T3 (en) 2006-03-24 2016-10-03 Handylab Inc Integrated microfluidic sample processing system and method for its use
GB2436616A (en) * 2006-03-29 2007-10-03 Inverness Medical Switzerland Assay device and method
US8007999B2 (en) 2006-05-10 2011-08-30 Theranos, Inc. Real-time detection of influenza virus
WO2007133710A2 (en) * 2006-05-11 2007-11-22 Raindance Technologies, Inc. Microfluidic devices and methods of use thereof
US9562837B2 (en) 2006-05-11 2017-02-07 Raindance Technologies, Inc. Systems for handling microfludic droplets
CN101074963B (en) * 2006-05-17 2011-07-06 聿新生物科技股份有限公司 Electrode testing strip for inspecting cholesterine by electrochemical method and its production
US7674924B2 (en) 2006-05-22 2010-03-09 Third Wave Technologies, Inc. Compositions, probes, and conjugates and uses thereof
EP2636755A1 (en) 2006-05-26 2013-09-11 AltheaDx Incorporated Biochemical analysis of partitioned cells
EP2589668A1 (en) 2006-06-14 2013-05-08 Verinata Health, Inc Rare cell analysis using sample splitting and DNA tags
EP2029779A4 (en) 2006-06-14 2010-01-20 Living Microsystems Inc Use of highly parallel snp genotyping for fetal diagnosis
US20080050739A1 (en) 2006-06-14 2008-02-28 Roland Stoughton Diagnosis of fetal abnormalities using polymorphisms including short tandem repeats
US8137912B2 (en) 2006-06-14 2012-03-20 The General Hospital Corporation Methods for the diagnosis of fetal abnormalities
JP4292279B2 (en) * 2006-07-18 2009-07-08 ファルマ・アクセス株式会社 Crystal production tool
WO2008021123A1 (en) 2006-08-07 2008-02-21 President And Fellows Of Harvard College Fluorocarbon emulsion stabilizing surfactants
US7842499B2 (en) * 2006-08-07 2010-11-30 Platypus Technologies, Llc Substrates, devices, and methods for cellular assays
US8656949B2 (en) * 2006-08-15 2014-02-25 University Of Maryland College Park Microfluidic devices and methods of fabrication
US20100178204A1 (en) * 2006-08-21 2010-07-15 Anp Technologies Automated Self-Contained Liquid Handling and Detection System Device
US7674616B2 (en) * 2006-09-14 2010-03-09 Hemosense, Inc. Device and method for measuring properties of a sample
CA2665490C (en) * 2006-10-03 2014-06-17 The Trustees Of The University Of Pennsylvania Method for treatment of macular degeneration
US8012744B2 (en) 2006-10-13 2011-09-06 Theranos, Inc. Reducing optical interference in a fluidic device
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
US8592220B2 (en) 2006-10-26 2013-11-26 Intermolecular, Inc. High pressure parallel fixed bed reactor and method
EP2091647A2 (en) 2006-11-14 2009-08-26 Handylab, Inc. Microfluidic system for amplifying and detecting polynucleotides in parallel
US20080113391A1 (en) 2006-11-14 2008-05-15 Ian Gibbons Detection and quantification of analytes in bodily fluids
WO2008060604A2 (en) 2006-11-14 2008-05-22 Handylab, Inc. Microfluidic system for amplifying and detecting polynucleotides in parallel
US8202491B2 (en) * 2006-11-21 2012-06-19 Bioscale, Inc. Apparatus for analyte processing
US20080118402A1 (en) * 2006-11-21 2008-05-22 David Brancazio Method and apparatus for analyte processing
US20080153152A1 (en) * 2006-11-22 2008-06-26 Akira Wakabayashi Microfluidic chip
CN1996009B (en) * 2007-01-10 2010-05-19 博奥生物有限公司 Microfluid device for multi-sample analysis and application method therefor
US7867592B2 (en) 2007-01-30 2011-01-11 Eksigent Technologies, Inc. Methods, compositions and devices, including electroosmotic pumps, comprising coated porous surfaces
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
WO2008121342A2 (en) * 2007-03-28 2008-10-09 President And Fellows Of Harvard College Emulsions and techniques for formation
ES2797951T3 (en) * 2007-04-04 2020-12-04 Ande Corp Integrated nucleic acid analysis
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
WO2008137008A2 (en) 2007-05-04 2008-11-13 Claros Diagnostics, Inc. Fluidic connectors and microfluidic systems
US20080297169A1 (en) * 2007-05-31 2008-12-04 Greenquist Alfred C Particle Fraction Determination of A Sample
US8097422B2 (en) 2007-06-20 2012-01-17 Salk Institute For Biological Studies Kir channel modulators
EP2014761B1 (en) * 2007-06-22 2016-09-28 Sony Deutschland GmbH A device for processing an analyte and a method of processing and/or detecting an analyte using said device
US8921121B2 (en) * 2007-06-29 2014-12-30 The Trustees Of Columbia University In The City Of New York Methods, devices, and systems for chemiluminescence-based microfluidic cell counting
US8287820B2 (en) 2007-07-13 2012-10-16 Handylab, Inc. Automated pipetting apparatus having a combined liquid pump and pipette head system
US8133671B2 (en) 2007-07-13 2012-03-13 Handylab, Inc. Integrated apparatus for performing nucleic acid extraction and diagnostic testing on multiple biological samples
US8182763B2 (en) 2007-07-13 2012-05-22 Handylab, Inc. Rack for sample tubes and reagent holders
USD621060S1 (en) 2008-07-14 2010-08-03 Handylab, Inc. Microfluidic cartridge
US20090136385A1 (en) 2007-07-13 2009-05-28 Handylab, Inc. Reagent Tube
US9618139B2 (en) 2007-07-13 2017-04-11 Handylab, Inc. Integrated heater and magnetic separator
JP5651011B2 (en) 2007-07-13 2015-01-07 ハンディーラブ インコーポレイテッド Polynucleotide capture material and method of use thereof
US8105783B2 (en) 2007-07-13 2012-01-31 Handylab, Inc. Microfluidic cartridge
US9186677B2 (en) 2007-07-13 2015-11-17 Handylab, Inc. Integrated apparatus for performing nucleic acid extraction and diagnostic testing on multiple biological samples
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
US8158430B1 (en) 2007-08-06 2012-04-17 Theranos, Inc. Systems and methods of fluidic sample processing
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
WO2009026359A2 (en) * 2007-08-20 2009-02-26 Platypus Technologies, Llc Improved devices for cell assays
US20090120796A1 (en) * 2007-09-26 2009-05-14 Jongyoon Han Electrokinetic concentration device and methods of use thereof
CN101874205B (en) 2007-10-02 2014-10-01 赛拉诺斯股份有限公司 Modular point-of-care devices and uses thereof
EP2217721A4 (en) * 2007-10-29 2013-01-09 Univ California Osteoarthritis gene therapy
CN101910415B (en) 2007-11-07 2015-02-25 不列颠哥伦比亚大学 Microfluidic device and method of using same
WO2009076134A1 (en) * 2007-12-11 2009-06-18 Eksigent Technologies, Llc Electrokinetic pump with fixed stroke volume
ES2882661T3 (en) * 2008-01-03 2021-12-02 Emd Millipore Corp Cell culture matrix system for automated assays and methods of operation and manufacturing thereof
KR20110030415A (en) 2008-01-22 2011-03-23 인터젠엑스 인크. Universal sample preparation system and use in an integrated analysis system
JP4556194B2 (en) * 2008-02-01 2010-10-06 セイコーエプソン株式会社 Biological sample reaction method
US8397762B2 (en) * 2008-02-04 2013-03-19 Bioscale, Inc. Fluidic system with improved flow characteristics
US20110092686A1 (en) * 2008-03-28 2011-04-21 Pelican Group Holdings, Inc. Multicapillary sample preparation devices and methods for processing analytes
US20090286692A1 (en) * 2008-04-15 2009-11-19 Wainwright Norman R Cartridge and Method for Sample Analysis
KR20110042152A (en) 2008-04-25 2011-04-25 노쓰웨스턴유니버시티 Nanostructures suitable for sequestering cholesterol
WO2009131677A1 (en) * 2008-04-25 2009-10-29 Claros Diagnostics, Inc. Flow control in microfluidic systems
US9664619B2 (en) 2008-04-28 2017-05-30 President And Fellows Of Harvard College Microfluidic device for storage and well-defined arrangement of droplets
US8472683B2 (en) * 2008-05-09 2013-06-25 General Electric Company Motion correction in tomographic images
US9879360B2 (en) * 2008-06-20 2018-01-30 International Business Machines Corporation Microfluidic selection of library elements
US9393566B2 (en) 2008-06-23 2016-07-19 Canon U.S. Life Sciences, Inc. System and method for temperature referencing for melt curve data collection
USD618820S1 (en) 2008-07-11 2010-06-29 Handylab, Inc. Reagent holder
USD787087S1 (en) 2008-07-14 2017-05-16 Handylab, Inc. Housing
EP4047367A1 (en) 2008-07-18 2022-08-24 Bio-Rad Laboratories, Inc. Method for detecting target analytes with droplet libraries
US20110151500A1 (en) * 2008-08-11 2011-06-23 Kazuya Hosokawa Blood-Platelet Test Method and Blood-Platelet Test Device
US8178355B2 (en) * 2008-09-15 2012-05-15 Platypus Technologies, Llc. Detection of vapor phase compounds by changes in physical properties of a liquid crystal
EP3378951B1 (en) 2008-09-20 2020-05-13 The Board of Trustees of the Leland Stanford Junior University Noninvasive diagnosis of aneuploidy by sequencing
US9132394B2 (en) 2008-09-23 2015-09-15 Bio-Rad Laboratories, Inc. System for detection of spaced droplets
US8709762B2 (en) 2010-03-02 2014-04-29 Bio-Rad Laboratories, Inc. System for hot-start amplification via a multiple emulsion
US9492797B2 (en) 2008-09-23 2016-11-15 Bio-Rad Laboratories, Inc. System for detection of spaced droplets
WO2011120006A1 (en) 2010-03-25 2011-09-29 Auantalife, Inc. A Delaware Corporation Detection system for droplet-based assays
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
US9156010B2 (en) 2008-09-23 2015-10-13 Bio-Rad Laboratories, Inc. Droplet-based assay system
US11130128B2 (en) 2008-09-23 2021-09-28 Bio-Rad Laboratories, Inc. Detection method for a target nucleic acid
US9417190B2 (en) 2008-09-23 2016-08-16 Bio-Rad Laboratories, Inc. Calibrations and controls for droplet-based assays
US8633015B2 (en) 2008-09-23 2014-01-21 Bio-Rad Laboratories, Inc. Flow-based thermocycling system with thermoelectric cooler
US9764322B2 (en) 2008-09-23 2017-09-19 Bio-Rad Laboratories, Inc. System for generating droplets with pressure monitoring
WO2011120024A1 (en) 2010-03-25 2011-09-29 Quantalife, Inc. Droplet generation for droplet-based assays
US10512910B2 (en) 2008-09-23 2019-12-24 Bio-Rad Laboratories, Inc. Droplet-based analysis method
US8591829B2 (en) 2008-12-18 2013-11-26 Opko Diagnostics, Llc Reagent storage in microfluidic systems and related articles and methods
US8448499B2 (en) 2008-12-23 2013-05-28 C A Casyso Ag Cartridge device for a measuring system for measuring viscoelastic characteristics of a sample liquid, a corresponding measuring system, and a corresponding method
US8672532B2 (en) 2008-12-31 2014-03-18 Integenx Inc. Microfluidic methods
EP2391714B2 (en) 2009-01-30 2019-07-24 Whitehead Institute for Biomedical Research Methods for ligation and uses thereof
JP5586631B2 (en) * 2009-01-30 2014-09-10 ジェン−プローブ・インコーポレーテッド System and method for detecting a signal and applying thermal energy to a signal transmission element
DE202010018623U1 (en) 2009-02-02 2018-12-07 Opko Diagnostics, Llc Structures for controlling the light interaction with microfluidic devices
EP2394175B1 (en) * 2009-02-09 2016-02-03 caprotec bioanalytics GmbH Devices, systems and methods for separating magnetic particles
US9068921B2 (en) * 2009-03-07 2015-06-30 Hewlett-Packard Development Company, L.P. Analyzer and method for sensing using the same
EP2409138B1 (en) * 2009-03-16 2017-08-02 Clondiag GmbH Fluorescence-based assays on microfluidic devices
EP3415235A1 (en) 2009-03-23 2018-12-19 Raindance Technologies Inc. Manipulation of microfluidic droplets
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
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
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
KR101796906B1 (en) 2009-03-24 2017-11-10 유니버시티 오브 시카고 Method for carrying out a reaction
WO2010118427A1 (en) 2009-04-10 2010-10-14 Canon U.S. Life Sciences, Inc. Fluid interface cartridge for a microfluidic chip
US8790916B2 (en) 2009-05-14 2014-07-29 Genestream, Inc. Microfluidic method and system for isolating particles from biological fluid
DE102009040151B4 (en) * 2009-05-26 2013-09-12 Analytik Jena Ag Arrangement for the detection of chemiluminescence on gases
US8673627B2 (en) * 2009-05-29 2014-03-18 Life Technologies Corporation Apparatus and methods for performing electrochemical reactions
KR20120030130A (en) 2009-06-02 2012-03-27 인터젠엑스 인크. Fluidic devices with diaphragm valves
CA2764678C (en) 2009-06-04 2017-12-12 Lockheed Martin Corporation Multiple-sample microfluidic chip for dna analysis
SG176669A1 (en) 2009-06-05 2012-01-30 Integenx Inc Universal sample preparation system and use in an integrated analysis system
US9550985B2 (en) 2009-06-15 2017-01-24 Netbio, Inc. Methods for forensic DNA quantitation
AU2010276403A1 (en) 2009-07-20 2012-03-08 Siloam Biosciences, Inc. Microfluidic assay platforms
WO2011028764A2 (en) 2009-09-02 2011-03-10 President And Fellows Of Harvard College Multiple emulsions created using jetting and other techniques
EP2473618B1 (en) 2009-09-02 2015-03-04 Bio-Rad Laboratories, 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
CN105825049A (en) 2009-10-19 2016-08-03 提拉诺斯公司 Integrated health data capture and analysis system
AU2010315867A1 (en) 2009-11-03 2012-06-21 University Of Virginia Patent Foundation Versatile, visible method for detecting polymeric analytes
JP5823405B2 (en) 2009-11-04 2015-11-25 ザ ユニバーシティ オブ ブリティッシュ コロンビア Nucleic acid-containing lipid particles and related methods
US20110143378A1 (en) * 2009-11-12 2011-06-16 CyVek LLC. Microfluidic method and apparatus for high performance biological assays
USD638550S1 (en) 2009-11-13 2011-05-24 3M Innovative Properties Company Sample processing disk cover
US8834792B2 (en) 2009-11-13 2014-09-16 3M Innovative Properties Company Systems for processing sample processing devices
USD667561S1 (en) 2009-11-13 2012-09-18 3M Innovative Properties Company Sample processing disk cover
USD638951S1 (en) 2009-11-13 2011-05-31 3M Innovative Properties Company Sample processing disk cover
US9700889B2 (en) 2009-11-23 2017-07-11 Cyvek, Inc. Methods and systems for manufacture of microarray assay systems, conducting microfluidic assays, and monitoring and scanning to obtain microfluidic assay results
US9759718B2 (en) 2009-11-23 2017-09-12 Cyvek, Inc. PDMS membrane-confined nucleic acid and antibody/antigen-functionalized microlength tube capture elements, and systems employing them, and methods of their use
US9651568B2 (en) 2009-11-23 2017-05-16 Cyvek, Inc. Methods and systems for epi-fluorescent monitoring and scanning for microfluidic assays
US10065403B2 (en) 2009-11-23 2018-09-04 Cyvek, Inc. Microfluidic assay assemblies and methods of manufacture
US9500645B2 (en) 2009-11-23 2016-11-22 Cyvek, Inc. Micro-tube particles for microfluidic assays and methods of manufacture
US9855735B2 (en) 2009-11-23 2018-01-02 Cyvek, Inc. Portable microfluidic assay devices and methods of manufacture and use
WO2011063408A1 (en) 2009-11-23 2011-05-26 Cyvek, Inc. Method and apparatus for performing assays
US9216412B2 (en) 2009-11-23 2015-12-22 Cyvek, Inc. Microfluidic devices and methods of manufacture and use
WO2013134745A1 (en) 2012-03-08 2013-09-12 Cyvek, Inc Portable microfluidic assay devices and methods of manufacture and use
US8567425B2 (en) 2009-11-24 2013-10-29 Opko Diagnostics, Llc Fluid mixing and delivery in microfluidic systems
US8584703B2 (en) * 2009-12-01 2013-11-19 Integenx Inc. Device with diaphragm valve
WO2011079176A2 (en) 2009-12-23 2011-06-30 Raindance Technologies, Inc. Microfluidic systems and methods for reducing the exchange of molecules between droplets
JP5863670B2 (en) 2010-01-19 2016-02-17 ノースウェスタン ユニバーシティ Synthetic nanostructures containing nucleic acids and / or other components
US9353342B2 (en) 2010-01-21 2016-05-31 Emd Millipore Corporation Cell culture and gradient migration assay methods and devices
EP2535721B1 (en) 2010-02-10 2017-04-19 Fujimori Kogyo Co., Ltd. Microchip for platelet examination and platelet examination device using same
US10351905B2 (en) 2010-02-12 2019-07-16 Bio-Rad Laboratories, Inc. Digital analyte analysis
US9366632B2 (en) 2010-02-12 2016-06-14 Raindance Technologies, Inc. Digital analyte analysis
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
US8399198B2 (en) 2010-03-02 2013-03-19 Bio-Rad Laboratories, Inc. Assays with droplets transformed into capsules
AR080405A1 (en) * 2010-03-17 2012-04-04 Basf Se EMULSIFICATION TO FOUND
CN101817495B (en) * 2010-03-25 2012-03-14 湖南大学 Micro fluid control chip and preparation method and application thereof
WO2011119920A2 (en) 2010-03-25 2011-09-29 Oregon Health & Science University Cmv glycoproteins and recombinant vectors
CA2767114A1 (en) 2010-03-25 2011-09-29 Bio-Rad Laboratories, Inc. Droplet transport system for detection
JP2011220947A (en) * 2010-04-14 2011-11-04 Hitachi Engineering & Services Co Ltd Microbiological testing apparatus and microbiological testing chip
CN105381825A (en) 2010-04-16 2016-03-09 欧普科诊断有限责任公司 Feedback control in microfluidic systems
USD645971S1 (en) 2010-05-11 2011-09-27 Claros Diagnostics, Inc. Sample cassette
US20130203045A1 (en) 2010-05-26 2013-08-08 University Of Virginia Patent Foundation Method for detecting nucleic acids based on aggregate formation
US8512538B2 (en) 2010-05-28 2013-08-20 Integenx Inc. Capillary electrophoresis device
EP2596347B1 (en) 2010-07-22 2017-09-06 Hach Company Alkalinity analysis using a lab-on-a-chip
WO2012024658A2 (en) 2010-08-20 2012-02-23 IntegenX, Inc. Integrated analysis system
US8763642B2 (en) 2010-08-20 2014-07-01 Integenx Inc. Microfluidic devices with mechanically-sealed diaphragm valves
US9562897B2 (en) 2010-09-30 2017-02-07 Raindance Technologies, Inc. Sandwich assays in droplets
US10114020B2 (en) 2010-10-11 2018-10-30 Mbio Diagnostics, Inc. System and device for analyzing a fluidic sample
CA2814720C (en) 2010-10-15 2016-12-13 Lockheed Martin Corporation Micro fluidic optic design
DE202011110979U1 (en) 2010-11-01 2017-12-04 Bio-Rad Laboratories, Inc. System for forming emulsions
CA2825196C (en) 2011-01-21 2021-01-05 Theranos, Inc. Systems and methods for sample use maximization
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
EP2686449B1 (en) 2011-03-18 2020-11-18 Bio-Rad Laboratories, Inc. Multiplexed digital assays with combinatorial use of signals
DE112012001473B4 (en) * 2011-03-28 2016-08-25 Kake Educational Institution A method for measuring a sample solution concentration and apparatus for measuring a sample solution concentration
US10526572B2 (en) 2011-04-01 2020-01-07 EMD Millipore Corporaticn Cell culture and invasion assay method and system
RU2690374C2 (en) 2011-04-15 2019-06-03 Бектон, Дикинсон Энд Компани Scanning in real time microfluid thermal cycler and methods of synchronized thermal cycling and scanning optical detection
EP3395957B1 (en) 2011-04-25 2020-08-12 Bio-Rad Laboratories, Inc. Methods and compositions for nucleic acid analysis
WO2012149285A1 (en) 2011-04-28 2012-11-01 Claire Mitchell Method for treatment of macular degeneration by modulating p2y12 or p2x7 receptors
WO2012151289A2 (en) 2011-05-02 2012-11-08 University Of Virginia Patent Foundation Method and system to detect aggregate formation on a substrate
WO2012151268A1 (en) 2011-05-02 2012-11-08 University Of Virginia Patent Foundation Method and system for high throughput optical and label free detection of analytes
EP2704759A4 (en) 2011-05-05 2015-06-03 Eksigent Technologies Llc Gel coupling for electrokinetic delivery systems
JP2014515926A (en) 2011-05-12 2014-07-07 ネットバイオ・インコーポレーテッド Methods and compositions for rapid multiplex amplification of STR loci
US8931331B2 (en) 2011-05-18 2015-01-13 3M Innovative Properties Company Systems and methods for volumetric metering on a sample processing device
EP2709762B1 (en) 2011-05-18 2021-03-31 DiaSorin S.p.A. Systems and methods for detecting the presence of a selected volume of material in a sample processing device
KR101963721B1 (en) 2011-05-18 2019-03-29 디아소린 에스.피.에이. Systems and methods for valving on a sample processing device
US9238206B2 (en) 2011-05-23 2016-01-19 President And Fellows Of Harvard College Control of emulsions, including multiple emulsions
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
PT2691530T (en) 2011-06-10 2018-05-10 Univ Oregon Health & Science Cmv glycoproteins and recombinant vectors
WO2013066418A1 (en) * 2011-06-14 2013-05-10 Corning Incorporated Hybrid microfluidic assemblies
CN106268389A (en) 2011-07-06 2017-01-04 哈佛学院院长等 Multiple Emulsion and for preparing the technology of multiple Emulsion
US8658430B2 (en) 2011-07-20 2014-02-25 Raindance Technologies, Inc. Manipulating droplet size
EP2737089B1 (en) 2011-07-29 2017-09-06 Bio-rad Laboratories, Inc. Library characterization by digital assay
US20130189754A1 (en) 2011-09-12 2013-07-25 International Aids Vaccine Initiative Immunoselection of recombinant vesicular stomatitis virus expressing hiv-1 proteins by broadly neutralizing antibodies
CA2849917C (en) 2011-09-30 2020-03-31 Becton, Dickinson And Company Unitized reagent strip
USD692162S1 (en) 2011-09-30 2013-10-22 Becton, Dickinson And Company Single piece reagent holder
US20150136604A1 (en) 2011-10-21 2015-05-21 Integenx Inc. Sample preparation, processing and analysis systems
US10865440B2 (en) 2011-10-21 2020-12-15 IntegenX, Inc. Sample preparation, processing and analysis systems
US20140328759A1 (en) 2011-10-25 2014-11-06 The University Of British Columbia Limit size lipid nanoparticles and related methods
US9402894B2 (en) 2011-10-27 2016-08-02 International Aids Vaccine Initiative Viral particles derived from an enveloped virus
EP2773892B1 (en) 2011-11-04 2020-10-07 Handylab, Inc. Polynucleotide sample preparation device
EP3441142A1 (en) 2011-11-16 2019-02-13 Becton, Dickinson and Company Methods and systems for detecting an analyte in a sample
ES2870857T3 (en) 2011-12-03 2021-10-27 Emd Millipore Corp Microfluidic system for cell culture
JP6108657B2 (en) * 2011-12-28 2017-04-05 日本バイリーン株式会社 Structure and manufacturing method thereof
BR112014018995B1 (en) 2012-02-03 2021-01-19 Becton, Dickson And Company systems to perform automated testing
US9322054B2 (en) 2012-02-22 2016-04-26 Lockheed Martin Corporation Microfluidic cartridge
US9304065B2 (en) 2012-02-29 2016-04-05 Fluidigm Corporation Methods, systems and devices for multiple single-cell capturing and processing using microfluidics
MY171654A (en) 2012-03-05 2019-10-22 Oy Arctic Partners Ab Methods and apparatuses for predicting risk of prostate cancer and prostate gland volume
WO2013155531A2 (en) 2012-04-13 2013-10-17 Bio-Rad Laboratories, Inc. Sample holder with a well having a wicking promoter
CN104379261A (en) * 2012-04-16 2015-02-25 昆南诺股份有限公司 A nanocapillary device for biomolecule detection, a fluidic network structure and a method of manufacturing thereof
US9180449B2 (en) 2012-06-12 2015-11-10 Hach Company Mobile water analysis
EP2679596B1 (en) 2012-06-27 2017-04-12 International Aids Vaccine Initiative HIV-1 env glycoprotein variant
DE102012109026A1 (en) * 2012-09-25 2014-03-27 Eads Deutschland Gmbh Detection device and detection method for the automatic determination of biomass
EP2906928A4 (en) 2012-10-15 2016-11-09 Nanocellect Biomedical Inc Systems, apparatus, and methods for sorting particles
WO2014070235A1 (en) 2012-10-29 2014-05-08 Mbio Diagnostics, Inc. Biological particle identification system, cartridge and associated methods
USD768872S1 (en) 2012-12-12 2016-10-11 Hach Company Cuvette for a water analysis instrument
BR112015010695B1 (en) 2013-01-11 2023-03-07 Becton, Dickinson And Company MICROFLUID DEVICE AND METHOD FOR PERFORMING A LIQUID SAMPLE TEST, METHOD FOR FORMING A MICROFLUID DEVICE, SYSTEM AND KIT
US9995412B2 (en) 2013-03-01 2018-06-12 Wave 80 Biosciences, Inc. Long-throw microfluidic actuator
CA2903382A1 (en) * 2013-03-01 2014-09-12 Wave 80 Biosciences, Inc. Methods and systems for enhanced microfluidic processing
ES2741001T3 (en) 2013-03-13 2020-02-07 Opko Diagnostics Llc Mixing fluids in fluid systems
CN105143456A (en) 2013-03-15 2015-12-09 不列颠哥伦比亚大学 Lipid nanoparticles for transfection and related methods
WO2014144789A2 (en) 2013-03-15 2014-09-18 Fluidigm Corporation Methods and devices for analysis of defined multicellular combinations
JP2014240065A (en) * 2013-05-15 2014-12-25 公立大学法人大阪府立大学 Flow channel structure and production method of flow channel structure
EP2848937A1 (en) 2013-09-05 2015-03-18 International Aids Vaccine Initiative Methods of identifying novel HIV-1 immunogens
US11901041B2 (en) 2013-10-04 2024-02-13 Bio-Rad Laboratories, Inc. Digital analysis of nucleic acid modification
US10058604B2 (en) 2013-10-07 2018-08-28 International Aids Vaccine Initiative Soluble HIV-1 envelope glycoprotein trimers
US9815059B2 (en) * 2013-10-07 2017-11-14 Devrim PESEN OKVUR Microfluidic device for investigation of distance dependent interactions in cell biology
EP3066190B1 (en) 2013-11-06 2020-12-30 Becton, Dickinson and Company Microfluidic devices, and methods of using the same
CN105899936B (en) 2013-11-13 2019-12-24 贝克顿·迪金森公司 Optical imaging system and method of using the same
EP3071333A4 (en) 2013-11-18 2017-11-15 IntegenX Inc. Cartridges and instruments for sample analysis
US9944977B2 (en) 2013-12-12 2018-04-17 Raindance Technologies, Inc. Distinguishing rare variations in a nucleic acid sequence from a sample
JP6509230B2 (en) 2013-12-23 2019-05-08 ビーエーエスエフ ソシエタス・ヨーロピアBasf Se Methods for determining modulators of insect transient receptor potential V (TRPV) channels
US11193176B2 (en) 2013-12-31 2021-12-07 Bio-Rad Laboratories, Inc. Method for detecting and quantifying latent retroviral RNA species
GB2544198B (en) 2014-05-21 2021-01-13 Integenx Inc Fluidic cartridge with valve mechanism
US11305236B2 (en) 2014-06-13 2022-04-19 Gattaco Inc. Surface tension driven filtration
CN106573201B (en) * 2014-06-13 2020-05-22 迈克尔·瑞安·麦克尼利 Capillary pressure resetting mechanism and application
EP3201596A4 (en) * 2014-10-01 2018-07-11 University of Tasmania Extraction and concentration device
CA3044748C (en) 2014-10-14 2021-10-26 Becton, Dickinson And Company Blood sample management using open cell foam
US11298061B2 (en) 2014-10-14 2022-04-12 Becton, Dickinson And Company Blood sample management using open cell foam
US10210410B2 (en) 2014-10-22 2019-02-19 Integenx Inc. Systems and methods for biometric data collections
CN107106983B (en) 2014-10-22 2021-04-16 尹特根埃克斯有限公司 Systems and methods for sample preparation, processing, and analysis
AU2015360401B2 (en) 2014-12-12 2021-05-06 Opko Diagnostics, Llc Fluidic systems comprising an incubation channel, including fluidic systems formed by molding
US10605767B2 (en) 2014-12-18 2020-03-31 Life Technologies Corporation High data rate integrated circuit with transmitter configuration
WO2016145057A1 (en) 2015-03-10 2016-09-15 Becton, Dickinson And Company Biological fluid micro-sample management device
EP3069730A3 (en) 2015-03-20 2017-03-15 International Aids Vaccine Initiative Soluble hiv-1 envelope glycoprotein trimers
US9931394B2 (en) 2015-03-23 2018-04-03 International Aids Vaccine Initiative Soluble HIV-1 envelope glycoprotein trimers
JP6466775B2 (en) * 2015-04-30 2019-02-06 シスメックス株式会社 Sample analysis method using sample analysis cartridge, sample analysis cartridge, and sample analyzer
CN106248929A (en) * 2015-06-11 2016-12-21 宁波大学 A kind of AIDS diagnosis multi-channel chip device making substrate with PDMS
NL2015130B1 (en) * 2015-07-09 2017-02-01 Mimetas B V Barrier function measurements.
USD804682S1 (en) 2015-08-10 2017-12-05 Opko Diagnostics, Llc Multi-layered sample cassette
CA3109854C (en) 2015-09-01 2023-07-25 Becton, Dickinson And Company Depth filtration device for separating specimen phases
US10227556B2 (en) 2015-09-04 2019-03-12 Wayne State University Cell culture devices for biomimetic and pathomimetic cell cultures
US10647981B1 (en) 2015-09-08 2020-05-12 Bio-Rad Laboratories, Inc. Nucleic acid library generation methods and compositions
JP6998862B2 (en) 2015-09-25 2022-02-04 エフ.ホフマン-ラ ロシュ アーゲー Soluble sortase A
WO2017050874A1 (en) 2015-09-25 2017-03-30 F. Hoffmann-La Roche Ag Process for producing thioesters employing a sortase a
CN108026560A (en) 2015-09-25 2018-05-11 豪夫迈·罗氏有限公司 Reacted in eutectic solvent using the acid amides that turns of sorting enzyme
US10228367B2 (en) 2015-12-01 2019-03-12 ProteinSimple Segmented multi-use automated assay cartridge
CA3005084A1 (en) 2015-12-11 2017-06-15 Opko Diagnostics, Llc Fluidic systems involving incubation of samples and/or reagents
EP3436578B1 (en) 2016-03-30 2022-01-19 F. Hoffmann-La Roche AG Improved sortase
US11426580B2 (en) 2016-08-12 2022-08-30 Health Discovery Labs Llc Systems and methods for low intensity high efficiency electrical stimulation
CN106362811B (en) * 2016-08-29 2018-07-13 北京工业大学 A kind of micro-fluidic chip measuring pressure change in the deformable channel of wall surface
EP3519578B1 (en) 2016-10-03 2021-12-22 Precision Nanosystems Inc Compositions for transfecting resistant cell types
CN113607719A (en) * 2016-11-29 2021-11-05 普诺森公司 Method and apparatus for simultaneously detecting a wide range of protein concentrations
US11525152B2 (en) 2017-04-07 2022-12-13 Acenxion Biosystems, Inc. System and method for rapid detection of viable microorganisms in liquid media
JP7227568B2 (en) 2017-07-27 2023-02-22 ユニバーシティー オブ サウザン カリフォルニア Otopetrin proton channel as a target for drug development
CN108181458B (en) * 2018-02-26 2019-05-14 北京华科泰生物技术股份有限公司 A kind of micro-fluidic chip and its preparation method and application based on fluorescence immunoassay joint-detection
EP3566772A1 (en) * 2018-05-11 2019-11-13 Agilent Technologies, Inc. (A Delaware Corporation) Immobilizing fluidic sample for inhibiting spatial broadening
GB201820870D0 (en) * 2018-12-20 2019-02-06 Cambridge Entpr Ltd Detection of components
CN109647557B (en) * 2019-02-27 2021-04-02 哈尔滨工业大学 Direct particle separation chip based on induced charge electroosmosis micro vortex and application and separation method thereof
US20230405590A1 (en) * 2020-11-22 2023-12-21 Gattaco Inc. Sample transfer devices, and components and methods thereof

Citations (41)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3014848A (en) * 1958-03-07 1961-12-26 Technicon Instr Method of performing biological assays
US3018224A (en) * 1957-08-23 1962-01-23 Technicon Instr Analysis and production-monitoring methods and apparatus
US3065148A (en) * 1959-07-14 1962-11-20 Technicon Instr Method and apparatus for use in conducting studies on cells
US3615241A (en) * 1970-02-27 1971-10-26 Nasa Firefly pump-metering system
US3699004A (en) * 1970-08-31 1972-10-17 Technicon Instr Method and apparatus for sample analysis on a continuous flow basis
US3701716A (en) * 1969-04-15 1972-10-31 Beckman Instruments Inc Liquid analysis apparatus
US4618586A (en) * 1983-04-08 1986-10-21 Endotronics, Inc. Apparatus for administering a controlled dosage of a chemical substance having an improved culture chamber
US4629686A (en) * 1982-02-19 1986-12-16 Endotronics, Inc. Apparatus for delivering a controlled dosage of a chemical substance
US4675300A (en) * 1985-09-18 1987-06-23 The Board Of Trustees Of The Leland Stanford Junior University Laser-excitation fluorescence detection electrokinetic separation
US4737464A (en) * 1985-09-26 1988-04-12 Molecular Devices Corporation Solid-state optical assay imaging apparatus
US4783413A (en) * 1986-07-30 1988-11-08 Contraves Ag Apparatus for supplying a medium to a reaction chamber
US4908112A (en) * 1988-06-16 1990-03-13 E. I. Du Pont De Nemours & Co. Silicon semiconductor wafer for analyzing micronic biological samples
US4963498A (en) * 1985-08-05 1990-10-16 Biotrack Capillary flow device
US4978503A (en) * 1984-06-13 1990-12-18 Ares-Serono Research & Development Limited Partnership Devices for use in chemical test procedures
US5032381A (en) * 1988-12-20 1991-07-16 Tropix, Inc. Chemiluminescence-based static and flow cytometry
US5096807A (en) * 1985-03-06 1992-03-17 Murex Corporation Imaging immunoassay detection system with background compensation and its use
US5104804A (en) * 1990-06-04 1992-04-14 Molecular Devices Corporation Cell assay device used in a microphysiometer
US5126022A (en) * 1990-02-28 1992-06-30 Soane Tecnologies, Inc. Method and device for moving molecules by the application of a plurality of electrical fields
US5140161A (en) * 1985-08-05 1992-08-18 Biotrack Capillary flow device
US5144139A (en) * 1985-08-05 1992-09-01 Biotrack, Inc. Capillary flow device
US5164598A (en) * 1985-08-05 1992-11-17 Biotrack Capillary flow device
US5171534A (en) * 1984-01-16 1992-12-15 California Institute Of Technology Automated DNA sequencing technique
US5171132A (en) * 1989-12-27 1992-12-15 Seiko Epson Corporation Two-valve thin plate micropump
US5188963A (en) * 1989-11-17 1993-02-23 Gene Tec Corporation Device for processing biological specimens for analysis of nucleic acids
US5192405A (en) * 1991-01-11 1993-03-09 Millipore Corporation Process for effecting high efficiency separations by capillary electrophoresis
US5271724A (en) * 1990-08-31 1993-12-21 Westonbridge International Limited Valve equipped with a position detector and a micropump incorporating said valve
US5278048A (en) * 1988-10-21 1994-01-11 Molecular Devices Corporation Methods for detecting the effect of cell affecting agents on living cells
US5277556A (en) * 1990-07-10 1994-01-11 Westonbridge International Limited Valve and micropump incorporating said valve
US5296114A (en) * 1991-12-06 1994-03-22 Ciba-Geigy Corporation Electrophoretic separating device and electrophoretic separating method
US5296375A (en) * 1992-05-01 1994-03-22 Trustees Of The University Of Pennsylvania Mesoscale sperm handling devices
US5304487A (en) * 1992-05-01 1994-04-19 Trustees Of The University Of Pennsylvania Fluid handling in mesoscale analytical devices
US5312731A (en) * 1989-10-05 1994-05-17 Engstroem Gunnar Method and apparatus for studying a reaction pattern of a cell or cell aggregates during perfusion with different media
US5324591A (en) * 1987-03-06 1994-06-28 Geo-Centers, Inc. Deep ultraviolet photolithographically defined ultra-thin films for selective cell adhesion and outgrowth and method of manufacturing the same and devices containing the same
US5375979A (en) * 1992-06-19 1994-12-27 Robert Bosch Gmbh Thermal micropump with values formed from silicon plates
US5384261A (en) * 1991-11-22 1995-01-24 Affymax Technologies N.V. Very large scale immobilized polymer synthesis using mechanically directed flow paths
US5389524A (en) * 1989-07-28 1995-02-14 Kemisk Vaerk Koge A/S Method and a system for quantitatively monitoring a chemical component dissolved in a liquid medium
US5395503A (en) * 1986-06-20 1995-03-07 Molecular Devices Corporation Zero volume electrochemical cell
US5607565A (en) * 1995-03-27 1997-03-04 Coulter Corporation Apparatus for measuring analytes in a fluid sample
US5637458A (en) * 1994-07-20 1997-06-10 Sios, Inc. Apparatus and method for the detection and assay of organic molecules
US5637469A (en) * 1992-05-01 1997-06-10 Trustees Of The University Of Pennsylvania Methods and apparatus for the detection of an analyte utilizing mesoscale flow systems
US5645702A (en) * 1995-06-07 1997-07-08 Hewlett-Packard Company Low voltage miniaturized column analytical apparatus and method

Family Cites Families (92)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3918908A (en) 1974-05-17 1975-11-11 Geomet Method for prothrombin testing
US4443408A (en) 1981-07-09 1984-04-17 International Technidyne, Inc. Apparatus for analyzing the influence of additive reagents upon the coagulation of blood
US4390403A (en) 1981-07-24 1983-06-28 Batchelder J Samuel Method and apparatus for dielectrophoretic manipulation of chemical species
US4554839A (en) 1983-10-14 1985-11-26 Cetus Corporation Multiple trough vessel for automated liquid handling apparatus
US4952518A (en) 1984-10-01 1990-08-28 Cetus Corporation Automated assay machine and assay tray
US4756884A (en) * 1985-08-05 1988-07-12 Biotrack, Inc. Capillary flow device
US5108703A (en) 1986-03-26 1992-04-28 Beckman Instruments, Inc. Automated multi-purpose analytical chemistry processing center and laboratory work station
CA1296622C (en) * 1986-08-12 1992-03-03 Jeffrey E. Anderson Method and apparatus for automated assessment of the immunoregulatory status of the mononuclear leukocyte immune system
US4740708A (en) 1987-01-06 1988-04-26 International Business Machines Corporation Semiconductor wafer surface inspection apparatus and method
US4849340A (en) * 1987-04-03 1989-07-18 Cardiovascular Diagnostics, Inc. Reaction system element and method for performing prothrombin time assay
WO1990004645A1 (en) * 1988-10-21 1990-05-03 Molecular Devices Corporation Methods and apparatus for detecting the effect of cell affecting agents on living cells
IE903930A1 (en) 1989-11-06 1991-05-08 Akzo Nv Immunoassays for and monoclonal antibodies to prothrombin¹activation peptides and their degradation products
US5279936A (en) * 1989-12-22 1994-01-18 Syntex (U.S.A.) Inc. Method of separation employing magnetic particles and second medium
US5858188A (en) * 1990-02-28 1999-01-12 Aclara Biosciences, Inc. Acrylic microchannels and their use in electrophoretic applications
US5770029A (en) 1996-07-30 1998-06-23 Soane Biosciences Integrated electrophoretic microdevices
US5750015A (en) 1990-02-28 1998-05-12 Soane Biosciences Method and device for moving molecules by the application of a plurality of electrical fields
US5147606A (en) 1990-08-06 1992-09-15 Miles Inc. Self-metering fluid analysis device
GB2248891A (en) 1990-10-18 1992-04-22 Westonbridge Int Ltd Membrane micropump
US5488469A (en) 1991-08-30 1996-01-30 Omron Corporation Cell analyzing apparatus
GB9119382D0 (en) 1991-09-11 1991-10-23 Knight Scient Ltd Apparatus for monitoring liquids
US5605662A (en) 1993-11-01 1997-02-25 Nanogen, Inc. Active programmable electronic devices for molecular biological analysis and diagnostics
US5296144A (en) * 1992-01-02 1994-03-22 World Trade Corporation Composite membrane of a hydrophilic asymmetric membrane coated with an organosiloxane block copolymer
US5451525A (en) 1992-02-14 1995-09-19 Coulter Corporation Method and materials for determining particle count in a flow cytometer
SE500702C2 (en) 1992-04-07 1994-08-15 Staffan Birnbaum Methods and apparatus for optical analysis of samples separated into thin capillaries
US5223219A (en) 1992-04-10 1993-06-29 Biotrack, Inc. Analytical cartridge and system for detecting analytes in liquid samples
US5486335A (en) * 1992-05-01 1996-01-23 Trustees Of The University Of Pennsylvania Analysis based on flow restriction
US5744366A (en) * 1992-05-01 1998-04-28 Trustees Of The University Of Pennsylvania Mesoscale devices and methods for analysis of motile cells
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
US5498392A (en) * 1992-05-01 1996-03-12 Trustees Of The University Of Pennsylvania Mesoscale polynucleotide amplification device and method
US5587128A (en) 1992-05-01 1996-12-24 The Trustees Of The University Of Pennsylvania Mesoscale polynucleotide amplification devices
AU680195B2 (en) 1992-05-01 1997-07-24 Trustees Of The University Of Pennsylvania, The Analysis based on flow restriction
GB2266751A (en) 1992-05-02 1993-11-10 Westonbridge Int Ltd Piezoelectric micropump excitation voltage control.
US5598052A (en) 1992-07-28 1997-01-28 Philips Electronics North America Vacuum microelectronic device and methodology for fabricating same
US5639423A (en) 1992-08-31 1997-06-17 The Regents Of The University Of Calfornia Microfabricated reactor
US5547849A (en) 1993-02-17 1996-08-20 Biometric Imaging, Inc. Apparatus and method for volumetric capillary cytometry
US5441894A (en) 1993-04-30 1995-08-15 Abbott Laboratories Device containing a light absorbing element for automated chemiluminescent immunoassays
US5460709A (en) 1993-06-21 1995-10-24 Helena Laboratories Corporation Automatic electrophoresis method and apparatus
US5395587A (en) * 1993-07-06 1995-03-07 Smithkline Beecham Corporation Surface plasmon resonance detector having collector for eluted ligate
US5429734A (en) * 1993-10-12 1995-07-04 Massachusetts Institute Of Technology Monolithic capillary electrophoretic device
EP0726906A4 (en) 1993-11-02 1998-10-07 Affymax Tech Nv Synthesizing and screening molecular diversity
DE4411268C2 (en) 1994-03-31 2001-02-01 Danfoss As Analysis method and device
DE4411266C2 (en) * 1994-03-31 2001-05-17 Danfoss As Analysis method and device
US5540889A (en) 1994-05-11 1996-07-30 Whitehead Institute For Biomedical Research Apparatus and method for a highly parallel pipetter
DE69527585T2 (en) 1994-06-08 2003-04-03 Affymetrix Inc Method and device for packaging chips
US6001229A (en) 1994-08-01 1999-12-14 Lockheed Martin Energy Systems, Inc. Apparatus and method for performing microfluidic manipulations for chemical analysis
US5571410A (en) * 1994-10-19 1996-11-05 Hewlett Packard Company Fully integrated miniaturized planar liquid sample handling and analysis device
US5632876A (en) 1995-06-06 1997-05-27 David Sarnoff Research Center, Inc. Apparatus and methods for controlling fluid flow in microchannels
US5744367A (en) 1994-11-10 1998-04-28 Igen International, Inc. Magnetic particle based electrochemiluminescent detection apparatus and method
US5585069A (en) 1994-11-10 1996-12-17 David Sarnoff Research Center, Inc. Partitioned microelectronic and fluidic device array for clinical diagnostics and chemical synthesis
US5603351A (en) 1995-06-07 1997-02-18 David Sarnoff Research Center, Inc. Method and system for inhibiting cross-contamination in fluids of combinatorial chemistry device
JP3398749B2 (en) * 1994-11-10 2003-04-21 オーキッド バイオ サイエンシズ, インコーポレイテッド Liquid distribution system
DE69533159T2 (en) 1994-11-14 2005-07-21 Trustees Of The University Of Pennsylvania MINIATURIZED SAMPLE PREPARATION DEVICES AND SYSTEMS FOR DETERMINING AND TREATING ANALYTES
JP3909859B2 (en) 1994-11-14 2007-04-25 トラステイーズ・オブ・ザ・ユニバーシテイ・オブ・ペンシルベニア Medium-scale polynucleotide amplification equipment
US5731212A (en) 1994-12-20 1998-03-24 International Technidyne Corporation Test apparatus and method for testing cuvette accommodated samples
US5560811A (en) 1995-03-21 1996-10-01 Seurat Analytical Systems Incorporated Capillary electrophoresis apparatus and method
US5650075A (en) * 1995-05-30 1997-07-22 Motorola, Inc. Method for etching photolithographically produced quartz crystal blanks for singulation
US5605262A (en) 1995-06-01 1997-02-25 Bond; James R. Adjustable wrist band
US5856174A (en) 1995-06-29 1999-01-05 Affymetrix, Inc. Integrated nucleic acid diagnostic device
JPH11510389A (en) 1995-07-31 1999-09-14 アメリカ合衆国 Extending protein-protein interaction surfaces to inactivate cellular protein functions
US5716825A (en) 1995-11-01 1998-02-10 Hewlett Packard Company Integrated nucleic acid analysis system for MALDI-TOF MS
WO1997017426A1 (en) 1995-11-08 1997-05-15 Trustees Of Boston University Cellular physiology workstations for automated data acquisition and perfusion control
US5783397A (en) 1995-12-11 1998-07-21 Northeastern University Screening natural samples for new therapeutic compounds using capillary electrophoresis
US5942443A (en) * 1996-06-28 1999-08-24 Caliper Technologies Corporation High throughput screening assay systems in microscale fluidic devices
US6399023B1 (en) 1996-04-16 2002-06-04 Caliper Technologies Corp. Analytical system and method
US5885470A (en) 1997-04-14 1999-03-23 Caliper Technologies Corporation Controlled fluid transport in microfabricated polymeric substrates
US5824204A (en) * 1996-06-27 1998-10-20 Ic Sensors, Inc. Micromachined capillary electrophoresis device
US5779868A (en) * 1996-06-28 1998-07-14 Caliper Technologies Corporation Electropipettor and compensation means for electrophoretic bias
BR9710054A (en) * 1996-06-28 2000-01-11 Caliper Techn Corp Apparatus for separating test compounds for an effect on a biochemical system and for detecting a effect of a test compound on a biochemical system, procedures for determining whether a sample contains a compound capable of affecting a biochemical system, for separating a plurality of test compounds for an effect on a biochemical system and uses of a microfluidic system and a test substrate.
CN1329729C (en) 1996-06-28 2007-08-01 卡钳生命科学股份有限公司 Electropipettor and compensation means for electrophoretic bias
US5800690A (en) 1996-07-03 1998-09-01 Caliper Technologies Corporation Variable control of electroosmotic and/or electrophoretic forces within a fluid-containing structure via electrical forces
US5699157A (en) 1996-07-16 1997-12-16 Caliper Technologies Corp. Fourier detection of species migrating in a microchannel
US5804436A (en) 1996-08-02 1998-09-08 Axiom Biotechnologies, Inc. Apparatus and method for real-time measurement of cellular response
US5858187A (en) 1996-09-26 1999-01-12 Lockheed Martin Energy Systems, Inc. Apparatus and method for performing electrodynamic focusing on a microchip
US6447727B1 (en) 1996-11-19 2002-09-10 Caliper Technologies Corp. Microfluidic systems
US5964995A (en) 1997-04-04 1999-10-12 Caliper Technologies Corp. Methods and systems for enhanced fluid transport
JP2001521622A (en) 1997-04-04 2001-11-06 カリパー テクノロジーズ コーポレイション Closed-loop biochemical analyzer
EP0988529B1 (en) 1997-04-25 2013-06-12 Caliper Life Sciences, Inc. Microfluidic devices incorporating improved channel geometries
US6090251A (en) 1997-06-06 2000-07-18 Caliper Technologies, Inc. Microfabricated structures for facilitating fluid introduction into microfluidic devices
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
EP1021566A4 (en) 1997-06-09 2002-06-19 Caliper Techn Corp Apparatus and methods for correcting for variable velocity in microfluidic systems
US5882465A (en) 1997-06-18 1999-03-16 Caliper Technologies Corp. Method of manufacturing microfluidic devices
US5959291A (en) 1997-06-27 1999-09-28 Caliper Technologies Corporation Method and apparatus for measuring low power signals
US5876675A (en) 1997-08-05 1999-03-02 Caliper Technologies Corp. Microfluidic devices and systems
US5989402A (en) 1997-08-29 1999-11-23 Caliper Technologies Corp. Controller/detector interfaces for microfluidic systems
JP2001515204A (en) 1997-09-02 2001-09-18 カリパー テクノロジーズ コーポレイション Microfluidic system with electrofluid control and electrothermal control
US6012902A (en) 1997-09-25 2000-01-11 Caliper Technologies Corp. Micropump
US5842787A (en) 1997-10-09 1998-12-01 Caliper Technologies Corporation Microfluidic systems incorporating varied channel dimensions
US5958694A (en) 1997-10-16 1999-09-28 Caliper Technologies Corp. Apparatus and methods for sequencing nucleic acids in microfluidic systems
US6022733A (en) 1997-12-02 2000-02-08 Tam; Yun K. Simulated biological dissolution and absorption system
US6074725A (en) 1997-12-10 2000-06-13 Caliper Technologies Corp. Fabrication of microfluidic circuits by printing techniques
US5948227A (en) 1997-12-17 1999-09-07 Caliper Technologies Corp. Methods and systems for performing electrophoretic molecular separations
US6132685A (en) 1998-08-10 2000-10-17 Caliper Technologies Corporation High throughput microfluidic systems and methods

Patent Citations (43)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3018224A (en) * 1957-08-23 1962-01-23 Technicon Instr Analysis and production-monitoring methods and apparatus
US3014848A (en) * 1958-03-07 1961-12-26 Technicon Instr Method of performing biological assays
US3065148A (en) * 1959-07-14 1962-11-20 Technicon Instr Method and apparatus for use in conducting studies on cells
US3701716A (en) * 1969-04-15 1972-10-31 Beckman Instruments Inc Liquid analysis apparatus
US3615241A (en) * 1970-02-27 1971-10-26 Nasa Firefly pump-metering system
US3699004A (en) * 1970-08-31 1972-10-17 Technicon Instr Method and apparatus for sample analysis on a continuous flow basis
US3700562A (en) * 1970-08-31 1972-10-24 Technicon Instr Method and apparatus for sample analysis on a continuous flow basis
US4629686A (en) * 1982-02-19 1986-12-16 Endotronics, Inc. Apparatus for delivering a controlled dosage of a chemical substance
US4618586A (en) * 1983-04-08 1986-10-21 Endotronics, Inc. Apparatus for administering a controlled dosage of a chemical substance having an improved culture chamber
US5171534A (en) * 1984-01-16 1992-12-15 California Institute Of Technology Automated DNA sequencing technique
US4978503A (en) * 1984-06-13 1990-12-18 Ares-Serono Research & Development Limited Partnership Devices for use in chemical test procedures
US5096807A (en) * 1985-03-06 1992-03-17 Murex Corporation Imaging immunoassay detection system with background compensation and its use
US5140161A (en) * 1985-08-05 1992-08-18 Biotrack Capillary flow device
US5164598A (en) * 1985-08-05 1992-11-17 Biotrack Capillary flow device
US4963498A (en) * 1985-08-05 1990-10-16 Biotrack Capillary flow device
US5144139A (en) * 1985-08-05 1992-09-01 Biotrack, Inc. Capillary flow device
US4675300A (en) * 1985-09-18 1987-06-23 The Board Of Trustees Of The Leland Stanford Junior University Laser-excitation fluorescence detection electrokinetic separation
US4737464A (en) * 1985-09-26 1988-04-12 Molecular Devices Corporation Solid-state optical assay imaging apparatus
US5395503A (en) * 1986-06-20 1995-03-07 Molecular Devices Corporation Zero volume electrochemical cell
US4783413A (en) * 1986-07-30 1988-11-08 Contraves Ag Apparatus for supplying a medium to a reaction chamber
US5324591A (en) * 1987-03-06 1994-06-28 Geo-Centers, Inc. Deep ultraviolet photolithographically defined ultra-thin films for selective cell adhesion and outgrowth and method of manufacturing the same and devices containing the same
US4908112A (en) * 1988-06-16 1990-03-13 E. I. Du Pont De Nemours & Co. Silicon semiconductor wafer for analyzing micronic biological samples
US5278048A (en) * 1988-10-21 1994-01-11 Molecular Devices Corporation Methods for detecting the effect of cell affecting agents on living cells
US5032381A (en) * 1988-12-20 1991-07-16 Tropix, Inc. Chemiluminescence-based static and flow cytometry
US5389524A (en) * 1989-07-28 1995-02-14 Kemisk Vaerk Koge A/S Method and a system for quantitatively monitoring a chemical component dissolved in a liquid medium
US5312731A (en) * 1989-10-05 1994-05-17 Engstroem Gunnar Method and apparatus for studying a reaction pattern of a cell or cell aggregates during perfusion with different media
US5188963A (en) * 1989-11-17 1993-02-23 Gene Tec Corporation Device for processing biological specimens for analysis of nucleic acids
US5171132A (en) * 1989-12-27 1992-12-15 Seiko Epson Corporation Two-valve thin plate micropump
US5126022A (en) * 1990-02-28 1992-06-30 Soane Tecnologies, Inc. Method and device for moving molecules by the application of a plurality of electrical fields
US5104804A (en) * 1990-06-04 1992-04-14 Molecular Devices Corporation Cell assay device used in a microphysiometer
US5277556A (en) * 1990-07-10 1994-01-11 Westonbridge International Limited Valve and micropump incorporating said valve
US5271724A (en) * 1990-08-31 1993-12-21 Westonbridge International Limited Valve equipped with a position detector and a micropump incorporating said valve
US5192405A (en) * 1991-01-11 1993-03-09 Millipore Corporation Process for effecting high efficiency separations by capillary electrophoresis
US5384261A (en) * 1991-11-22 1995-01-24 Affymax Technologies N.V. Very large scale immobilized polymer synthesis using mechanically directed flow paths
US5296114A (en) * 1991-12-06 1994-03-22 Ciba-Geigy Corporation Electrophoretic separating device and electrophoretic separating method
US5304487A (en) * 1992-05-01 1994-04-19 Trustees Of The University Of Pennsylvania Fluid handling in mesoscale analytical devices
US5296375A (en) * 1992-05-01 1994-03-22 Trustees Of The University Of Pennsylvania Mesoscale sperm handling devices
US5427946A (en) * 1992-05-01 1995-06-27 Trustees Of The University Of Pennsylvania Mesoscale sperm handling devices
US5637469A (en) * 1992-05-01 1997-06-10 Trustees Of The University Of Pennsylvania Methods and apparatus for the detection of an analyte utilizing mesoscale flow systems
US5375979A (en) * 1992-06-19 1994-12-27 Robert Bosch Gmbh Thermal micropump with values formed from silicon plates
US5637458A (en) * 1994-07-20 1997-06-10 Sios, Inc. Apparatus and method for the detection and assay of organic molecules
US5607565A (en) * 1995-03-27 1997-03-04 Coulter Corporation Apparatus for measuring analytes in a fluid sample
US5645702A (en) * 1995-06-07 1997-07-08 Hewlett-Packard Company Low voltage miniaturized column analytical apparatus and method

Cited By (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030124736A1 (en) * 1998-08-05 2003-07-03 Caliper Technologies Corp. Open-field serial to parallel converter
US7208320B2 (en) * 1998-08-05 2007-04-24 Caliper Life Sciences, Inc. Open-field serial to parallel converter
US20020108860A1 (en) * 2001-01-15 2002-08-15 Staats Sau Lan Tang Fabrication of polymeric microfluidic devices
US20040053422A1 (en) * 2002-09-17 2004-03-18 Selena Chan Microfluidic devices with porous membranes for molecular sieving, metering, and separations
US7279134B2 (en) * 2002-09-17 2007-10-09 Intel Corporation Microfluidic devices with porous membranes for molecular sieving, metering, and separations
EP2093571A3 (en) * 2004-05-21 2009-09-02 Caliper Life Sciences, Inc. Automated system for handling microfluidic devices
WO2005114223A3 (en) * 2004-05-21 2006-05-04 Caliper Life Sciences Inc Automat system for handling microfluidic devices
US20060165559A1 (en) * 2004-05-21 2006-07-27 Caliper Life Sciences, Inc. Automated system for handling microfluidic devices
US9207249B2 (en) 2004-05-21 2015-12-08 Caliper Life Sciences, Inc. Automated system for handling microfluidic devices
US8496875B2 (en) 2004-05-21 2013-07-30 Caliper Life Sciences, Inc. Automated system for handling microfluidic devices
US20110101241A1 (en) * 2004-07-26 2011-05-05 Mesa Imaging Ag Solid-State Photodetector Pixel and Photodetecting Method
US9209327B2 (en) 2004-07-26 2015-12-08 Heptagon Micro Optics Pte. Ltd. Solid-state photodetector pixel and photodetecting method
US20090101559A1 (en) * 2005-01-21 2009-04-23 Anand Bala Subramaniam Microconcentrator/Microfilter
US7508505B2 (en) * 2005-07-21 2009-03-24 Mesa Imaging Ag Apparatus and method for all-solid-state fluorescence lifetime imaging
US20070018116A1 (en) * 2005-07-21 2007-01-25 Felix Lustenberger Apparatus and method for all-solid-state fluorescence lifetime imaging
US8293524B2 (en) * 2006-03-31 2012-10-23 Fluxion Biosciences Inc. Methods and apparatus for the manipulation of particle suspensions and testing thereof
US20070243523A1 (en) * 2006-03-31 2007-10-18 Fluxion Biosciences Inc. Methods and Apparatus for the Manipulation of Particle Suspensions and Testing Thereof
US10167502B2 (en) 2015-04-03 2019-01-01 Fluxion Biosciences, Inc. Molecular characterization of single cells and cell populations for non-invasive diagnostics
US11098352B2 (en) 2015-04-03 2021-08-24 Fluxion Biosciences, Inc. Molecular characterization of single cells and cell populations for non-invasive diagnostics

Also Published As

Publication number Publication date
US7091048B2 (en) 2006-08-15
US6399389B1 (en) 2002-06-04
US6630353B1 (en) 2003-10-07
JP2001502790A (en) 2001-02-27
US20030134431A1 (en) 2003-07-17
US6413782B1 (en) 2002-07-02
EP0907412A1 (en) 1999-04-14
US6267858B1 (en) 2001-07-31
US20050241941A1 (en) 2005-11-03
US6479299B1 (en) 2002-11-12
BR9710054A (en) 2000-01-11
US20040241733A1 (en) 2004-12-02
US6306659B1 (en) 2001-10-23
CN1173776C (en) 2004-11-03
EP0907412B1 (en) 2008-08-27
NZ333346A (en) 2000-03-27
US7067263B2 (en) 2006-06-27
US6558960B1 (en) 2003-05-06
JP3788519B2 (en) 2006-06-21
CA2258489A1 (en) 1998-01-08
CA2258489C (en) 2004-01-27
CN1262629A (en) 2000-08-09
WO1998000231A1 (en) 1998-01-08
US6274337B1 (en) 2001-08-14
US6429025B1 (en) 2002-08-06

Similar Documents

Publication Publication Date Title
US6267858B1 (en) High throughput screening assay systems in microscale fluidic devices
EP1271148B1 (en) High-throughput screening assay systems in microscale fluidic devices
WO1998000231A9 (en) High-throughput screening assay systems in microscale fluidic devices
AU729537B2 (en) High throughput screening assay systems in microscale fluidic devices
US20060000722A1 (en) High throughput screening assay systems in microscale fluidic devices
KR100453100B1 (en) High throughput screening assay system in microfluidic devices
AU759505B2 (en) High-throughput screening assay systems in microscale fluidic devices
AU2003213516B2 (en) High-throughput Screening Assay Systems in Microscale Fluidic Devices
AU2005234635B2 (en) High-throughput screening assay systems in microscale fluidic devices

Legal Events

Date Code Title Description
AS Assignment

Owner name: CALIPER LIFE SCIENCES, INC., CALIFORNIA

Free format text: CHANGE OF NAME;ASSIGNOR:CALIPER TECHNOLOGIES CORP.;REEL/FRAME:014326/0407

Effective date: 20040123

Owner name: CALIPER LIFE SCIENCES, INC.,CALIFORNIA

Free format text: CHANGE OF NAME;ASSIGNOR:CALIPER TECHNOLOGIES CORP.;REEL/FRAME:014326/0407

Effective date: 20040123

STCB Information on status: application discontinuation

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