US20070196820A1 - Devices and methods for enrichment and alteration of cells and other particles - Google Patents

Devices and methods for enrichment and alteration of cells and other particles Download PDF

Info

Publication number
US20070196820A1
US20070196820A1 US11/227,904 US22790405A US2007196820A1 US 20070196820 A1 US20070196820 A1 US 20070196820A1 US 22790405 A US22790405 A US 22790405A US 2007196820 A1 US2007196820 A1 US 2007196820A1
Authority
US
United States
Prior art keywords
analyte
sample
cells
magnetic
reagent
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US11/227,904
Inventor
Ravi Kapur
Mehmet Toner
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.)
GPB Scientific Inc
Original Assignee
Ravi Kapur
Mehmet Toner
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Ravi Kapur, Mehmet Toner filed Critical Ravi Kapur
Priority to US11/227,904 priority Critical patent/US20070196820A1/en
Priority to US11/323,971 priority patent/US20060223178A1/en
Priority to CA002622779A priority patent/CA2622779A1/en
Priority to EP06814753A priority patent/EP1934326A2/en
Priority to CNA2006800416665A priority patent/CN101305087A/en
Priority to AU2006292490A priority patent/AU2006292490A1/en
Priority to JP2008531362A priority patent/JP2009511001A/en
Priority to PCT/US2006/036061 priority patent/WO2007035498A2/en
Publication of US20070196820A1 publication Critical patent/US20070196820A1/en
Priority to IL190091A priority patent/IL190091A0/en
Assigned to GENERAL HOSPITAL CORPORATION, THE reassignment GENERAL HOSPITAL CORPORATION, THE ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: TONER, MEHMET
Assigned to GPB SCIENTIFIC, LLC, THE GENERAL HOSPITAL CORPORATION reassignment GPB SCIENTIFIC, LLC ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: TONER, MEHMET
Assigned to TONER, MEHMET reassignment TONER, MEHMET QUIT CLAIM ASSIGNMENT Assignors: THE GENERAL HOSPITAL CORPORATION
Abandoned legal-status Critical Current

Links

Images

Classifications

    • BPERFORMING OPERATIONS; TRANSPORTING
    • B03SEPARATION OF SOLID MATERIALS USING LIQUIDS OR USING PNEUMATIC TABLES OR JIGS; MAGNETIC OR ELECTROSTATIC SEPARATION OF SOLID MATERIALS FROM SOLID MATERIALS OR FLUIDS; SEPARATION BY HIGH-VOLTAGE ELECTRIC FIELDS
    • B03CMAGNETIC OR ELECTROSTATIC SEPARATION OF SOLID MATERIALS FROM SOLID MATERIALS OR FLUIDS; SEPARATION BY HIGH-VOLTAGE ELECTRIC FIELDS
    • B03C1/00Magnetic separation
    • B03C1/02Magnetic separation acting directly on the substance being separated
    • B03C1/30Combinations with other devices, not otherwise provided for
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01LCHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
    • B01L3/00Containers or dishes for laboratory use, e.g. laboratory glassware; Droppers
    • B01L3/50Containers for the purpose of retaining a material to be analysed, e.g. test tubes
    • B01L3/502Containers for the purpose of retaining a material to be analysed, e.g. test tubes with fluid transport, e.g. in multi-compartment structures
    • B01L3/5027Containers for the purpose of retaining a material to be analysed, e.g. test tubes with fluid transport, e.g. in multi-compartment structures by integrated microfluidic structures, i.e. dimensions of channels and chambers are such that surface tension forces are important, e.g. lab-on-a-chip
    • B01L3/502746Containers for the purpose of retaining a material to be analysed, e.g. test tubes with fluid transport, e.g. in multi-compartment structures by integrated microfluidic structures, i.e. dimensions of channels and chambers are such that surface tension forces are important, e.g. lab-on-a-chip characterised by the means for controlling flow resistance, e.g. flow controllers, baffles
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01LCHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
    • 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
    • B03SEPARATION OF SOLID MATERIALS USING LIQUIDS OR USING PNEUMATIC TABLES OR JIGS; MAGNETIC OR ELECTROSTATIC SEPARATION OF SOLID MATERIALS FROM SOLID MATERIALS OR FLUIDS; SEPARATION BY HIGH-VOLTAGE ELECTRIC FIELDS
    • B03CMAGNETIC OR ELECTROSTATIC SEPARATION OF SOLID MATERIALS FROM SOLID MATERIALS OR FLUIDS; SEPARATION BY HIGH-VOLTAGE ELECTRIC FIELDS
    • B03C1/00Magnetic separation
    • B03C1/32Magnetic separation acting on the medium containing the substance being separated, e.g. magneto-gravimetric-, magnetohydrostatic-, or magnetohydrodynamic separation
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12MAPPARATUS FOR ENZYMOLOGY OR MICROBIOLOGY; APPARATUS FOR CULTURING MICROORGANISMS FOR PRODUCING BIOMASS, FOR GROWING CELLS OR FOR OBTAINING FERMENTATION OR METABOLIC PRODUCTS, i.e. BIOREACTORS OR FERMENTERS
    • C12M47/00Means for after-treatment of the produced biomass or of the fermentation or metabolic products, e.g. storage of biomass
    • C12M47/04Cell isolation or sorting
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12MAPPARATUS FOR ENZYMOLOGY OR MICROBIOLOGY; APPARATUS FOR CULTURING MICROORGANISMS FOR PRODUCING BIOMASS, FOR GROWING CELLS OR FOR OBTAINING FERMENTATION OR METABOLIC PRODUCTS, i.e. BIOREACTORS OR FERMENTERS
    • C12M47/00Means for after-treatment of the produced biomass or of the fermentation or metabolic products, e.g. storage of biomass
    • C12M47/06Hydrolysis; Cell lysis; Extraction of intracellular or cell wall material
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6844Nucleic acid amplification reactions
    • C12Q1/686Polymerase chain reaction [PCR]
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N1/00Sampling; Preparing specimens for investigation
    • G01N1/28Preparing specimens for investigation including physical details of (bio-)chemical methods covered elsewhere, e.g. G01N33/50, C12Q
    • G01N1/40Concentrating samples
    • G01N1/4077Concentrating samples by other techniques involving separation of suspended solids
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N30/00Investigating or analysing materials by separation into components using adsorption, absorption or similar phenomena or using ion-exchange, e.g. chromatography or field flow fractionation
    • G01N30/0005Field flow fractionation
    • 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
    • G01N33/483Physical analysis of biological material
    • G01N33/487Physical analysis of biological material of liquid biological material
    • G01N33/49Blood
    • 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
    • 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
    • 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
    • B01L2300/00Additional constructional details
    • B01L2300/08Geometry, shape and general structure
    • B01L2300/0809Geometry, shape and general structure rectangular shaped
    • B01L2300/0816Cards, e.g. flat sample carriers usually with flow in two horizontal directions
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01LCHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
    • B01L2300/00Additional constructional details
    • B01L2300/08Geometry, shape and general structure
    • B01L2300/0861Configuration of multiple channels and/or chambers in a single devices
    • B01L2300/0864Configuration of multiple channels and/or chambers in a single devices comprising only one inlet and multiple receiving wells, e.g. for separation, splitting
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01LCHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
    • B01L2400/00Moving or stopping fluids
    • B01L2400/04Moving fluids with specific forces or mechanical means
    • B01L2400/0403Moving fluids with specific forces or mechanical means specific forces
    • B01L2400/0406Moving fluids with specific forces or mechanical means specific forces capillary forces
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01LCHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
    • B01L2400/00Moving or stopping fluids
    • B01L2400/04Moving fluids with specific forces or mechanical means
    • B01L2400/0403Moving fluids with specific forces or mechanical means specific forces
    • B01L2400/0409Moving fluids with specific forces or mechanical means specific forces centrifugal forces
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01LCHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
    • B01L2400/00Moving or stopping fluids
    • B01L2400/04Moving fluids with specific forces or mechanical means
    • B01L2400/0403Moving fluids with specific forces or mechanical means specific forces
    • B01L2400/0415Moving fluids with specific forces or mechanical means specific forces electrical forces, e.g. electrokinetic
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01LCHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
    • B01L2400/00Moving or stopping fluids
    • B01L2400/04Moving fluids with specific forces or mechanical means
    • B01L2400/0403Moving fluids with specific forces or mechanical means specific forces
    • B01L2400/043Moving fluids with specific forces or mechanical means specific forces magnetic forces
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01LCHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
    • B01L2400/00Moving or stopping fluids
    • B01L2400/04Moving fluids with specific forces or mechanical means
    • B01L2400/0403Moving fluids with specific forces or mechanical means specific forces
    • B01L2400/0472Diffusion
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01LCHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
    • B01L2400/00Moving or stopping fluids
    • B01L2400/04Moving fluids with specific forces or mechanical means
    • B01L2400/0475Moving fluids with specific forces or mechanical means specific mechanical means and fluid pressure
    • B01L2400/0487Moving fluids with specific forces or mechanical means specific mechanical means and fluid pressure fluid pressure, pneumatics
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01LCHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
    • B01L2400/00Moving or stopping fluids
    • B01L2400/08Regulating or influencing the flow resistance
    • B01L2400/084Passive control of flow resistance
    • B01L2400/086Passive control of flow resistance using baffles or other fixed flow obstructions
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B03SEPARATION OF SOLID MATERIALS USING LIQUIDS OR USING PNEUMATIC TABLES OR JIGS; MAGNETIC OR ELECTROSTATIC SEPARATION OF SOLID MATERIALS FROM SOLID MATERIALS OR FLUIDS; SEPARATION BY HIGH-VOLTAGE ELECTRIC FIELDS
    • B03CMAGNETIC OR ELECTROSTATIC SEPARATION OF SOLID MATERIALS FROM SOLID MATERIALS OR FLUIDS; SEPARATION BY HIGH-VOLTAGE ELECTRIC FIELDS
    • B03C2201/00Details of magnetic or electrostatic separation
    • B03C2201/18Magnetic separation whereby the particles are suspended in a liquid
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B33ADDITIVE MANUFACTURING TECHNOLOGY
    • B33YADDITIVE MANUFACTURING, i.e. MANUFACTURING OF THREE-DIMENSIONAL [3-D] OBJECTS BY ADDITIVE DEPOSITION, ADDITIVE AGGLOMERATION OR ADDITIVE LAYERING, e.g. BY 3-D PRINTING, STEREOLITHOGRAPHY OR SELECTIVE LASER SINTERING
    • B33Y80/00Products made by additive manufacturing
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N1/00Sampling; Preparing specimens for investigation
    • G01N1/28Preparing specimens for investigation including physical details of (bio-)chemical methods covered elsewhere, e.g. G01N33/50, C12Q
    • G01N1/40Concentrating samples
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N35/00Automatic analysis not limited to methods or materials provided for in any single one of groups G01N1/00 - G01N33/00; Handling materials therefor
    • G01N2035/00178Special arrangements of analysers
    • G01N2035/00237Handling microquantities of analyte, e.g. microvalves, capillary networks
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10TTECHNICAL SUBJECTS COVERED BY FORMER US CLASSIFICATION
    • Y10T137/00Fluid handling
    • Y10T137/0318Processes
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10TTECHNICAL SUBJECTS COVERED BY FORMER US CLASSIFICATION
    • Y10T137/00Fluid handling
    • Y10T137/8593Systems
    • 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
    • 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
    • Y10T436/25375Liberation or purification of sample or separation of material from a sample [e.g., filtering, centrifuging, etc.]

Definitions

  • the invention relates to the fields of cell separation, medical diagnostics, and microfluidic devices.
  • Clinically or environmentally relevant information may often be present in a sample, but in quantities too low to detect. Thus, various enrichment or amplification methods are often employed in order to increase the detectability of such information.
  • the invention provides a device for producing a sample enriched in an analyte that includes a first channel (e.g., a microfluidic channel) including a structure that deterministically deflects particles having a hydrodynamic size above a critical size in a direction not parallel to the average direction of flow in the structure, wherein said particles are analyte particles or are a non-analyte component of the sample; and a reservoir fluidly coupled to an output of the first channel through which the analyte passes into the reservoir, the reservoir including a reagent that alters a magnetic property of the analyte.
  • a first channel e.g., a microfluidic channel
  • a structure that deterministically deflects particles having a hydrodynamic size above a critical size in a direction not parallel to the average direction of flow in the structure, wherein said particles are analyte particles or are a non-analyte component of the sample
  • a reservoir fluidly coupled to an output
  • the structure includes an array of obstacles that form a network of gaps, where a fluid passing through the gaps is divided unequally into a major flux and a minor flux so that the average direction of the major flux is not parallel to the average direction of fluidic flow in the channel.
  • the array of obstacles may include first and second rows, where the second row is displaced laterally relative to the first row so that fluid passing through a gap in the first row is divided unequally into two gaps in the second row.
  • the analyte may have a hydrodynamic size greater than or smaller than the critical size.
  • the device may include a magnetic force generator capable of generating a magnetic field, and may further include a region of magnetic obstacles (e.g., obstacles including a permanent magnet or obstacles including a non-permanent magnet) disposed in a second channel (e.g., a microfluidic channel).
  • the magnetic obstacles may be ordered in a two-dimensional array.
  • the reservoir of the device may further include a second channel including a magnet.
  • the reagent e.g., sodium nitrite
  • the reagent e.g., holo-transferrin or a magnetic particle, may bind to the one or more analytes.
  • a magnetic particle may further include an antibody (e.g., anti-CD71, anti-CD36, anti-CD45, anti-GPA, anti-antigen i, anti-CD34, anti-fetal hemoglobin, anti-EpCAM, anti-E-cadherin, or anti-Muc-1) or an antigen-binding fragment thereof.
  • an antibody e.g., anti-CD71, anti-CD36, anti-CD45, anti-GPA, anti-antigen i, anti-CD34, anti-fetal hemoglobin, anti-EpCAM, anti-E-cadherin, or anti-Muc-1
  • an antibody e.g., anti-CD71, anti-CD36, anti-CD45, anti-GPA, anti-antigen i, anti-CD34, anti-fetal hemoglobin, anti-EpCAM, anti-E-cadherin, or anti-Muc-1
  • an antigen-binding fragment thereof e.g., anti-CD71,
  • the invention provides a method for producing a sample enriched in a first analyte relative to a second analyte that includes applying at least a portion of the sample to a device including a structure that deterministically deflects particles having a hydrodynamic size above a critical size in a direction not parallel to the average direction of flow in the structure, thereby producing a second sample enriched in the first analyte and including the second analyte; combining the second sample with a reagent that alters a magnetic property of the first analyte to produce an altered first analyte; and applying a magnetic field to the second sample, where the magnetic field generates a differential force to physically separate the altered first analyte from the second analyte, thereby producing a sample enriched in the first analyte.
  • the reagent may bind to the first analyte.
  • the reagent e.g., sodium nitrite
  • the reagent may alter an intrinsic magnetic property of the first analyte.
  • the reagent may include a magnetic particle that binds to or is incorporated into the first analyte.
  • the magnetic particle may include an antibody (e.g., anti-CD71, anti-GPA, anti-antigen i, anti-CD45, anti-CD34, anti-fetal hemoglobin, anti-EpCAM, anti-E-cadherin, or anti-Muc-1) or an antigen-binding fragment thereof.
  • the analyte may have a hydrodynamic size greater than or less than said critical size.
  • the sample may be a maternal blood sample.
  • the first analyte may be a cell (e.g., bacterial cell, a fetal cell, or a blood cell such as a fetal red blood cell), an organelle (e.g., a nucleus), or a virus.
  • the invention provides a method of producing a sample enriched in red blood cells relative to a second blood component (e.g., maternal blood cells) that includes contacting the sample including red blood cells (e.g., fetal red blood cells) with a reagent that oxidizes iron to produce oxidized hemoglobin; and applying a magnetic field to the sample, where the red blood cells having oxidized hemoglobin are attracted to the magnetic field to a greater extent than the second blood component, thereby producing the sample enriched in the red blood cells.
  • a second blood component e.g., maternal blood cells
  • the method may further include performing prior to the contacting step, a step that enriches the sample with red blood cells (e.g., enriching fetal blood cells are enriched relative to maternal red blood cells), for example, by applying at least a portion of the sample to a device including a structure that deterministically deflects particles having a hydrodynamic size above a critical size in a direction not parallel to the average direction of flow in the structure.
  • a step that enriches the sample with red blood cells e.g., enriching fetal blood cells are enriched relative to maternal red blood cells
  • the invention provides a device for producing a sample enriched in red blood cells that includes an analytical device that enriches the red blood cells based on size, shape, deformability, or affinity; and a reservoir including a reagent that oxidizes iron, where the reagent (e.g., sodium nitrite) increases the magnetic responsiveness of the red blood cells.
  • the analytical device may include a first channel that includes a structure that deterministically deflects particles having a hydrodynamic size above a critical size in a direction not parallel to the average direction of flow in the structure.
  • the invention provides a reagent that includes a plurality of magnetic particles coupled to one or more binding moieties (e.g., an antibody such as a monoclonal antibody) that selectively binds GPA, fetal hemoglobin, CD34, CD45, CD71, EGFR, or EpCAm.
  • binding moieties e.g., an antibody such as a monoclonal antibody
  • the invention provides a method for separating one or more cells of interest from a mixture of cells that includes combining the mixture of cells with a reagent of the fifth aspect and incubating the mixture of cells and the reagent for a time sufficient to allow the binding moieties to selectively bind the one or more cells of interest in the mixture, and apply a magnetic field to the mixture thereby separating cells that bound the magnetic particles from cells that did not bind the magnetic particles.
  • the method may further include a step of enriching the mixture of cells for the one or more cells of interest.
  • the enriching step may include performing size-based separation with an array of obstacles or selectively lysing one or more cells that is not a cell of interest.
  • analyte is meant a molecule, other chemical species, e.g., an ion, or particle.
  • exemplary analytes include cells, viruses, nucleic acids, proteins, carbohydrates, and small organic molecules.
  • biological particle any species of biological origin that is insoluble in aqueous media on the time scale of sample acquisition, preparation, storage, and analysis. Examples include cells, particulate cell components, viruses, and complexes including proteins, lipids, nucleic acids, and carbohydrates.
  • biological sample is meant any sample of biological origin or containing, or potentially containing, biological particles.
  • Preferred biological samples are cellular samples.
  • blood component any component of whole blood, including host red blood cells, white blood cells, and platelets. Blood components also include the components of plasma, e.g., proteins, lipids, nucleic acids, and carbohydrates, and any other cells that may be present in blood, e.g., because of current or past pregnancy, organ transplant, or infection.
  • cellular sample is meant a sample containing cells or components thereof.
  • samples include naturally occurring fluids (e.g., blood, lymph, cerebrospinal fluid, urine, cervical lavage, and water samples), portions of such fluids, and fluids into which cells have been introduced (e.g., culture media, and liquefied tissue samples).
  • the term also includes a lysate.
  • capture moiety is meant a chemical species to which an analyte binds.
  • a capture moiety may be a compound coupled to a surface or the material making up the surface.
  • Exemplary capture moieties include antibodies, oligo- or polypeptides, nucleic acids, other proteins, synthetic polymers, and carbohydrates.
  • channel is meant a gap through which fluid may flow.
  • a channel may be a capillary, a conduit, or a strip of hydrophilic pattern on an otherwise hydrophobic surface wherein aqueous fluids are confined.
  • component of cell is meant any component of a cell that may be at least partially isolated upon lysis of the cell.
  • Cellular components may be organelles (e.g., nuclei, peri-nuclear compartments, nuclear membranes, mitochondria, chloroplasts, or cell membranes), polymers or molecular complexes (e.g., lipids, polysaccharides, proteins. (membrane, trans-membrane, or cytosolic), nucleic acids (native, therapeutic, or pathogenic), viral particles, or ribosomes), or other molecules (e.g., hormones, ions, cofactors, or drugs).
  • component of a cellular sample is meant a subset of cells contained within the sample.
  • enriched sample is meant a sample containing an analyte that has been processed to increase the relative amount of the analyte relative to other analytes typically present in a sample.
  • samples may be enriched by increasing the amount of the analyte of interest by at least 10%, 25%, 50%, 75%, 100% or by a factor of at least 1000, 10,000, 100,000, or 1,000,000.
  • depleted sample is meant a sample containing an analyte that has been processed to decrease the amount of the analyte relative to other analytes typically present in a sample.
  • samples may be depleted by decreasing the amount of the analyte of interest by at least 5%, 10%, 25%, 50%, 75%, 90%, 95%, 97%, 98%, 99%, or even 100%.
  • exchange buffer in the context of a sample (e.g., a cellular sample) is meant a medium distinct from the medium in which the sample is originally suspended, and into which one or more components of the sample are to be exchanged.
  • flow-extracting boundary is meant a boundary designed to remove fluid from an array.
  • flow-feeding boundary is meant a boundary designed to add fluid to an array.
  • gaps an opening through which fluids and/or particles may flow.
  • a gap may be a capillary, a space between two obstacles wherein fluids may flow, or a hydrophilic pattern on an otherwise hydrophobic surface wherein aqueous fluids are confined.
  • the network of gaps is defined by an array of obstacles.
  • the gaps are the spaces between adjacent obstacles.
  • the network of gaps is constructed with an array of obstacles on the surface of a substrate.
  • hydrodynamic size is meant the effective size of a particle when interacting with a flow, obstacles (e.g., posts), or other particles.
  • the obstacles or other particles may be in a microfluidic structure. It is used as a general term for particle volume, shape, and deformability in the flow.
  • Intracellular activation is meant activation of second messenger pathways, leading to transcription factor activation, or activation of kinases or other metabolic pathways. Intracellular activation through modulation of external cell membrane antigens can also lead to changes in receptor trafficking.
  • labeling reagent is meant a reagent that is capable of binding to an analyte, being internalized or otherwise absorbed, and being detected, e.g., through shape, morphology, color, fluorescence, luminescence, phosphorescence, absorbance, magnetic properties, or radioactive emission.
  • metabolome is meant the set of compounds within a cell, other than proteins and nucleic acids, that participate in metabolic reactions and that are required for the maintenance, growth or normal function of a cell.
  • microfluidic is meant having at least one dimension of less than 1 mm.
  • an obstacle is meant an impediment to flow in a channel, e.g., a protrusion from one surface.
  • an obstacle may refer to a post outstanding on a base substrate or a hydrophobic barrier for aqueous fluids.
  • the obstacle may be partially permeable.
  • an obstacle may be a post made of porous material, wherein the pores allow penetration of an aqueous component but are too small for the particles being separated to enter.
  • shrinking reagent is meant a reagent that decreases the hydrodynamic size of a particle.
  • Shrinking reagents may act by decreasing the volume, increasing the deformability, or changing the shape of a particle.
  • swelling reagent is meant a reagent that increases the hydrodynamic size of a particle. Swelling reagents may act by increasing the volume, reducing the deformability, or changing the shape of a particle.
  • substantially larger is meant at least 2-fold, 3-fold, 5-fold, 10-fold, 25-fold, 50-fold, or even 100-fold larger.
  • substantially smaller is meant at least 2-fold, 3-fold, 5-fold, 10-fold, 25-fold, 50-fold, or even 100-fold smaller.
  • FIGS. 1A-1E are schematic depictions of an array that separated cells based on deterministic lateral displacement: (A) illustrates the lateral displacement of subsequent rows; (B) illustrates how fluid flowing through a gap is divide unequally around obstacles in subsequent rows; (C) illustrates how an analyte with a hydrodynamic size above the critical size is displaced laterally in the device; (D) illustrates an array of cylindrical obstacles; and (E) illustrates an array of elliptical obstacles.
  • FIG. 2 is a schematic description illustrating the unequal division of the flux through a gap around obstacles in subsequent rows.
  • FIG. 3 is a schematic depiction of how the critical size depends on the flow profile, which is parabolic in this example.
  • FIG. 4 is an illustration of how shape affects the movement of analytes through a device.
  • FIG. 5 is an illustration of how deformability affects the movement of analytes through a device.
  • FIG. 6 is a schematic depiction of deterministic lateral displacement. Analytes having a hydrodynamic size above the critical size move to the edge of the array, while analytes having a hydrodynamic size below the critical size pass through the device without lateral displacement.
  • FIG. 7 is a schematic depiction of a three stage deterministic device.
  • FIG. 8 is a schematic depiction of the maximum size and cut-off size for the device of FIG. 7 .
  • FIG. 9 is a schematic depiction of a bypass channel.
  • FIG. 10 is a schematic depiction of a bypass channel.
  • FIG. 11 is a schematic depiction of a three stage deterministic device having a common bypass channel.
  • FIG. 12 is a schematic depiction of a three stage, duplex deterministic device having a common bypass channel.
  • FIG. 13 is a schematic depiction of a three stage deterministic device having a common bypass channel, where the flow through the device is substantially constant.
  • FIG. 14 is a schematic depiction of a three stage, duplex deterministic device having a common bypass channel, where the flow through the device is substantially constant.
  • FIG. 15 is a schematic depiction of a three stage deterministic device having a common bypass channel, where the fluidic resistance in the bypass channel and the adjacent stage are substantially constant.
  • FIG. 16 is a schematic depiction of a three stage, duplex deterministic device having a common bypass channel, where the fluidic resistance in the bypass channel and the adjacent stage are substantially constant.
  • FIG. 17 is a schematic depiction of a three stage deterministic device having two, separate bypass channels.
  • FIG. 18 is a schematic depiction of a three stage deterministic device having two, separate bypass channels, which are in arbitrary configuration.
  • FIG. 19 is a schematic depiction of a three stage, duplex deterministic device having three, separate bypass channels.
  • FIG. 20 is a schematic depiction of a three stage deterministic device having two, separate bypass channels, wherein the flow through each stage is substantially constant.
  • FIG. 21 is a schematic depiction of a three stage, duplex deterministic device having three, separate bypass channels, wherein the flow through each stage is substantially constant.
  • FIG. 22 is a schematic depiction of a flow-extracting boundary.
  • FIG. 23 is a schematic depiction of a flow-feeding boundary.
  • FIG. 24 is a schematic depiction of a flow-feeding boundary, including a bypass channel.
  • FIG. 25 is a schematic depiction of two flow-feeding boundaries flanking a central bypass channel.
  • FIG. 26 is a schematic depiction of a device having four channels that act as on-chip flow resistors.
  • FIGS. 27 and 28 are schematic depictions of the effect of on-chip resistors on the relative width of two fluids flowing in a device.
  • FIG. 29 is a schematic depiction of a duplex device having a common inlet for the two outer regions.
  • FIG. 30A is a schematic depiction of a multiple arrays on a device.
  • FIG. 30B is a schematic depiction of multiple arrays with common inlets and product outlets on a device.
  • FIG. 31 is a schematic depiction of a multi-stage device with a small footprint.
  • FIG. 32 is a schematic depiction of blood passing through a device.
  • FIG. 33 is a graph illustrating the hydrodynamic size distribution of blood cells.
  • FIGS. 34A-34D are schematic depictions of moving an analyte from a sample to a buffer in a single stage (A), three stage (B), duplex (C), or three stage duplex (D) deterministic device.
  • FIG. 35A is a schematic depiction of a two stage deterministic device employed to move a particle from blood to a buffer to produce three products.
  • FIG. 35B is a schematic graph of the maximum size and cut off size of the two stages.
  • FIG. 35C is a schematic graph of the composition of the three products.
  • FIG. 36 is a schematic depiction of a two stage deterministic device for alteration, where each stage has a bypass channel.
  • FIG. 37 is a schematic depiction of the use of fluidic channels to connect two stages in a device.
  • FIG. 38 is a schematic depiction of the use of fluidic channels to connect two stages in a device, wherein the two stages are configured as a small footprint array.
  • FIG. 39A is a schematic depiction of a two stage deterministic device having a bypass channel that accepts output from both stages.
  • FIG. 39B is a schematic graph of the size range of product achievable with this device.
  • FIG. 40 is a schematic depiction of a two stage deterministic device for alteration having bypass channels that flank each stage and empty into the same outlet.
  • FIG. 41 is a schematic depiction of a deterministic device for the sequential movement and alteration of particles.
  • FIG. 42A is a photograph of a deterministic device that may be incorporated into a device of the invention.
  • FIGS. 42B-42E are depictions the mask used to fabricate a device that may be incorporated into the invention.
  • FIG. 42F is a series of photographs of the device containing blood and buffer.
  • FIGS. 43A-43F are typical histograms generated by the hematology analyzer from a blood sample and the waste (buffer, plasma, red blood cells, and platelets) and product (buffer and nucleated cells) fractions generated by the device of FIG. 42 .
  • FIGS. 44A-44D are depictions the mask used to fabricate a deterministic device that may be incorporated into a device of the invention.
  • FIGS. 45A-45D are depictions the mask used to fabricate a deterministic device that may be incorporated a device of into the invention.
  • FIG. 46A is a micrograph of a sample enriched in fetal red blood cells.
  • FIG. 46B is a micrograph of maternal red blood cell waste.
  • FIG. 48 is a series of micrographs showing the positive identification of sex and trisomy 21.
  • FIGS. 49A-49D are depictions the mask used to fabricate a deterministic device that may be incorporated into a device of the invention.
  • FIGS. 50A-50G are electron micrographs of the device of FIG. 49 .
  • FIGS. 51A-51D are depictions the mask used to fabricate a deterministic device that may be incorporated into a device of the invention.
  • FIGS. 52A-52F are electron micrographs of the device of FIG. 51 .
  • FIGS. 53A-53F are electron micrographs of the device of FIG. 45 .
  • FIGS. 54A-54D are depictions the mask used to fabricate a deterministic device that may be incorporated a device of into the invention.
  • FIGS. 55A-55S are electron micrographs of the device of FIG. 54 .
  • FIGS. 56A-56C are electron micrographs of the device of FIG. 44 .
  • FIG. 57A is a schematic illustration of a deterministic device that may be incorporated into a device of the invention and its operation.
  • FIG. 57B is an illustration of the device of FIG. 57A and a further-schematized representation of this device.
  • FIGS. 58A and 58B are illustrations of two distinct configurations for joining two deterministic devices together.
  • a cascade configuration is shown, in which outlet 1 of one device is joined to a sample inlet of a second device.
  • FIG. 58B a bandpass configuration is shown, in which outlet 2 of one device is joined to a sample inlet of a second device.
  • FIG. 59 is an illustration of an enhanced method of size separation in which target cells are labeled with immunoaffinity beads.
  • FIG. 60 is an illustration of a method for performing size fractionation and for separating free labeling reagents, e.g., antibodies, from bound labeling reagents by using a device that may be incorporated into the invention.
  • FIG. 61 is an illustration of a method shown in FIG. 60 .
  • non-target cells may copurify with target cells, but these non-target cells do not interfere with quantification of target cells.
  • FIG. 62 is an illustration of a method for separating large cells from a mixture and producing a concentrated sample of these cells.
  • FIG. 63 is an illustration of a method for lysing cells inside a device of the invention and separating whole cells from organelles and other cellular components.
  • FIG. 64 is an illustration of two devices arrayed in a cascade configuration and used for performing size fractionation and for separating free labeling reagent from bound labeling reagents by using a device of the invention.
  • FIG. 65 is an illustration of two devices arrayed in a cascade configuration and used for performing size fractionation and for separating free labeling reagent from bound labeling reagents by using a device of the invention.
  • phage is utilized for binding and detection rather than antibodies.
  • FIG. 66 is an illustration of two devices arrayed in a bandpass configuration.
  • FIG. 67 is a graph of cell count versus hydrodynamic cell diameter for a microfluidic separation of normal whole blood.
  • FIG. 68 is a set of histograms from input, product, and waste samples generated with a Coulter “A C -T diff” clinical blood analyzer.
  • the x-axis depicts cell volume in femtomoles.
  • FIG. 69 is a pair of representative micrographs from product and waste streams of fetal blood processed with a cell enrichment module, showing clear separation of nucleated cells and red blood cells.
  • FIG. 70 is a pair of images showing cells fixed on a cell enrichment module with paraformaldehyde and observed by fluorescence microscopy. Target cells are bound to the obstacles and floor of the capture module.
  • FIG. 71A is a graph of cell count versus hydrodynamic cell diameter for a microfluidic separation of normal whole blood.
  • FIG. 71B is a graph of cell count versus hydrodynamic cell diameter for a microfluidic separation of whole blood including a population of circulating tumor cells.
  • FIG. 71C is the graph of FIG. 71B , additionally showing a size cutoff that excludes most native blood cells.
  • FIG. 71D is the graph of FIG. 71C , additionally showing that the population of cells larger than the size cutoff may include endothelial cell, endometrial cells, or trophoblasts indicative of a disease state.
  • FIG. 72 is a schematic illustration of a method that features isolating and counting large cells within a cellular sample, wherein the count is indicative of a patient's disease state, and subsequently further analyzing the large cell subpopulation.
  • FIG. 73A is a design for a preferred deterministic device that may be incorporated into the invention.
  • FIG. 73B is a table of design parameters corresponding to FIG. 73A .
  • FIG. 74 is a cross-sectional view of a magnetic separation device useful in a device of the invention and associated process flow for cell isolation followed by release for off-line analysis according to the present invention.
  • FIG. 75 is a schematic of the fabrication and functionalization of a magnetic separation device.
  • the magnetized posts enable post-packaging modification of the device.
  • FIG. 76 is a schematic of an application of a magnetic separation device to capture and release CD71+ cells from a complex mixture, such as blood, using monoclonal antibodies to the transferrin (CD71) receptor.
  • FIG. 77 is a schematic representation of an application of a magnetic separation device to capture and release CD71+ cells from a complex mixture, such as blood, using holotransferrin.
  • Holotransferrin is rich in iron content, commercially available, and has higher affinity constants and specificity of interaction with the CD71 receptor than its counterpart monoclonal antibody.
  • the invention provides analytical devices and methods useful for enriching analytes in a sample.
  • the invention provides a device that includes a channel that deterministically deflects particles based on hydrodynamic size and a reservoir containing a reagent capable of altering a magnetic property of the particle.
  • the invention also provides a method for producing a sample enriched in a first analyte relative to a second analyte by applying the sample to a device that includes a channel that deterministically deflects particles based on hydrodynamic size, thereby producing a second sample enriched in the first analyte, and combining the second sample with a reagent that alters a magnetic property of the first analyte, and applying a magnetic field thereby separating the first analyte from the second analyte.
  • the methods and devices of the present invention may be used to enrich samples for analytes such as red blood cells (e.g., fetal red blood cells).
  • fluid samples examples include biological fluid samples, such as, whole blood, sweat, tears, ear flow, sputum, lymph, bone marrow suspension, lymph, urine, saliva, semen, vaginal flow, cerebrospinal fluid, brain fluid, ascites, milk, secretions of the respiratory, intestinal and genitourinary tracts, and amniotic fluid.
  • biological fluid samples such as, whole blood, sweat, tears, ear flow, sputum, lymph, bone marrow suspension, lymph, urine, saliva, semen, vaginal flow, cerebrospinal fluid, brain fluid, ascites, milk, secretions of the respiratory, intestinal and genitourinary tracts, and amniotic fluid.
  • any other biological sample e.g., a biopsy sample
  • Analytes in biological fluid samples include, but are not limited to, foreign organisms such as bacteria, viruses, and protozoans.
  • the devices and methods of the invention may be employed in connection with any analytical device.
  • analytical device examples include affinity columns, cell counters, particle sorters, e.g., fluorescent activated cell sorters, capillary electrophoresis, microscopes, spectrophotometers, sample storage devices, and sample preparation devices.
  • particle sorters e.g., fluorescent activated cell sorters, capillary electrophoresis, microscopes, spectrophotometers, sample storage devices, and sample preparation devices.
  • Microfluidic devices are of particular interest in connection with the systems described herein.
  • Exemplary analytical devices include devices useful for size, shape, or deformability based separation of particles, including filters, sieves, and deterministic separation devices, e.g., those described in International Publication Nos. 2004/029221 and 2004/113877, Huang et al. Science 304, 987-990 (2004), U.S. Publication No. 2004/0144651, U.S. Pat. Nos. 5,837,115 and 6,692,952, and U.S. Application Nos. 60/703,833 and 60/704,067; devices useful for affinity capture, e.g., those described in International Publication No. 2004/029221 and U.S. application Ser. No.
  • 11/071,679 devices useful for preferential lysis of cells in a sample, e.g., those described in International Publication No. 2004/029221, U.S. Pat. No. 5,641,628, and U.S. Application No. 60/668,415; and devices useful for arraying cells, e.g., those described in International Publication No. 2004/029221, U.S. Pat. No. 6,692,952, and U.S. application Ser. Nos. 10/778,831 and 11/146,581.
  • Two or more devices, either the same or different devices, may be combined in series or integrated into a single device, e.g., as described in International Publication No. 2004/029221.
  • the analytical device may be used to enrich various analytes in a sample, e.g., for collection or further analysis.
  • Rare cells or components thereof can be retained in the device, or otherwise enriched, compared to other cells as described, e.g., in International Publication No. 2004/029221.
  • Exemplary rare cells include, depending on the sample, fetal cells (e.g., fetal red blood cells); stem cells (e.g., undifferentiated); cancer cells; immune system cells (host or graft); epithelial cells; connective tissue cells; bacteria; fungi; viruses; and pathogens (e.g., bacterial or protozoa).
  • Such rare cells may be isolated from samples including bodily fluids, e.g., blood, or environmental sources, e.g., water or air samples.
  • Fetal red blood cells may be enriched from maternal peripheral blood, e.g., for the purpose of determining sex and identifying aneuploidies or genetic characteristics, e.g., mutations, in the developing fetus.
  • Cancer cells may also be enriched from peripheral blood for the purpose of diagnosis and monitoring therapeutic progress.
  • Bodily fluids or environmental samples may also be screened for pathogens, e.g., for coliform bacteria, blood borne illnesses such as sepsis, or bacterial or viral meningitis.
  • Rare cells also include cells from one organism present in another organism, e.g., cells from a transplanted organ.
  • Analytes retained or otherwise enriched in the device may, for example, be labeled, e.g., with fluorescent or radioactive probes, subjected to chemical or genetic analysis (such as fluorescent in situ hybridization), if biological, cultured, or otherwise observed or probed.
  • Analytical devices may or may not include microfluidic channels, i.e., may or may not be microfluidic devices.
  • the dimensions of the channels of the device into which analytes are introduced may depend on the size or type of analytes employed.
  • a channel in an analytical device has at least one dimension (e.g., height, width, length, or radius) of no greater than 10, 9.5, 9, 8.5, 8, 7.5, 7, 6.5, 6, 5.5, 5, 4.5, 4, 3.5, 3, 2.5, 2, 1.5, or 1 mm.
  • Microfluidic devices employed in the systems and methods described herein preferably have at least one dimension of less than 1, 0.9, 0.8, 0.7, 0.6, 0.5, 0.4, 0.3, 0.2, 0.1, or even 0.05 mm.
  • the preferred dimensions of an analytical device can be determined by one skilled in the art based on the desired application.
  • devices of the invention may include additional elements, e.g., for isolating, collection, manipulation, or detection. Such elements are known in the art.
  • a device of the invention e.g., a device incorporating a deterministic device
  • Devices of the invention may also include a component for two-dimensional imaging of the output from the device, e.g., an array of wells or a planar surface.
  • devices described herein are employed in conjunction with an affinity enrichment.
  • Devices of the invention may also be employed in conjunction with other enrichment devices, either on the same device or in different devices.
  • Other enrichment techniques are described, e.g., in International Publication Nos. 2004/029221 and 2004/113877, U.S. Pat. No. 6,692,952, and U.S. application Ser. Nos. 11/071,270, 11/071,679, and 60/668,415, each of which is incorporated by reference.
  • the analytical device is a device that allows deterministic separation of an analyte based on the hydrodynamic size of the analyte.
  • Such devices will employ a channel, e.g., a microfluidic channel, containing a structure that enables deterministic separation.
  • the channel includes one or more arrays of obstacles that allow deterministic lateral displacement of components of fluids.
  • Such devices are described, e.g., in Huang et al. Science 304, 987-990 (2004) and U.S. Publication No. 20040144651, and U.S. Application No. 60/414,258.
  • These devices may further employ an array of a network of gaps, wherein a fluid passing through a gap is divided unequally into subsequent gaps.
  • fluid passing through a gap is divided unequally even though the gaps are identical in dimensions.
  • a flow carries particles to be separated through the array of gaps. The flow is aligned at a small angle (flow angle) with respect to a line-of-sight of the array. Particles having a hydrodynamic size larger than a critical size migrate along the line-of-sight in the array, whereas those having a hydrodynamic size smaller than the critical size follow the flow in a different direction. Flow in the device occurs under laminar flow conditions.
  • the critical size is a function of several design parameters.
  • each row of obstacles is shifted horizontally with respect to the previous row by ⁇ , where ⁇ is the center-to-center distance between the obstacles ( FIG. 1A ).
  • the parameter ⁇ / ⁇ (the “bifurcation ratio,” ⁇ ) determines the ratio of flow bifurcated to the left of the next obstacle.
  • is 1/3, for the convenience of illustration.
  • the flux through a gap between two obstacles is ⁇
  • the minor flux is ⁇
  • the major flux is (1- ⁇ ) ( FIG. 2 ).
  • the flux through a gap is divided essentially into thirds ( FIG. 1B ).
  • FIG. 1C illustrates the movement of an analyte sized above the critical size (e.g., 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 microns) through the array.
  • the critical size e.g. 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 microns
  • the critical size is approximately 2R critical , where R critical is the distance between the stagnant flow line and the obstacle. If the center of mass of a particle, e.g., a cell, falls within R critical , the particle would follow the major flux and move along the line-of-sight of the array.
  • R critical can be determined if the flow profile across the gap is known ( FIG. 3 ); it is the thickness of the layer of fluids that would make up the minor flux.
  • R critical can be tailored based on the bifurcation ratio, ⁇ . In general, the smaller ⁇ , the smaller R critical .
  • lymphocytes are spheres of ⁇ 5 ⁇ m diameter
  • erythrocytes are biconcave disks of ⁇ 7 ⁇ m diameter, and ⁇ 1.5 ⁇ m thick.
  • the long axis of erythrocytes (diameter) is larger than that of the lymphocytes, but the short axis (thickness) is smaller. If erythrocytes align their long axes to a flow when driven through an array of obstacles by the flow, their hydrodynamic size is effectively their thickness ( ⁇ 1.5 ⁇ m), which is smaller than lymphocytes.
  • an erythrocyte When an erythrocyte is driven through an array of obstacles by a hydrodynamic flow, it tends to align its long axis to the flow and behave like a ⁇ 1.5 ⁇ m-wide particle, which is effectively “smaller” than lymphocytes.
  • the method and device may therefore separate analytes according to their shapes, although the volumes of the analytes could be the same.
  • analytes having different deformabilities behave as if they have different sizes ( FIG. 5 ).
  • two analytes having the same undeformed shape may be separated by deterministic lateral displacement, as one analyte may deform more readily than the other analyte when it contacts an obstacle in the array and changes shape.
  • separation in the device may be achieved based on any parameter that affects hydrodynamic size including the physical dimensions, the shape, and the deformability of the analyte.
  • feeding a mixture of analytes, e.g., cells, of different hydrodynamic sizes from the top of the array and collecting the analytes at the bottom can produce two products, an output containing analytes larger than the critical size, 2R critical , and an output containing cells smaller than the critical size. Either output or both outputs may be collected, e.g., when fractionating a sample into two or more sub-samples. Analytes larger than the gap size will get trapped inside the array. Therefore, an array has a working size range. Cells have to be larger than a cut-off size (2R critical ) and smaller than a maximum pass-through size (array gap size) to be directed into the major flux.
  • the “size range” of an array is defined as the ratio of maximum pass-through size to cut-off size.
  • Deterministic devices may be employed in order to separate free, unreacted reagent from the altered analyte.
  • a labeling reagent such as an antibody may be pre-incubated with an analyte (e.g., a cellular sample) prior to introduction to or within the deterministic device.
  • the reagent specifically reacts with the analyte of interest, e.g., a cell population such as epithelial cells.
  • Exemplary labeling reagents include antibodies, quantum dots, phage, aptamers, fluorophore-containing molecules, enzymes capable of carrying out a detectable chemical reaction, sodium nitrite, or functionalized beads.
  • the reagent is smaller than the.
  • analyte e.g., a cell
  • analyte of interest e.g., a cell
  • an altered analyte e.g., an analyte bound to the reagent
  • this method achieves both size separation and separation of free, unreacted reagent from the analyte. Additionally, this method of separation facilitates downstream sample analysis, if desired, without the need for a release step or a potentially destructive method of analysis, as described below.
  • FIG. 61 shows a particular case in which the enriched, labeled sample contains a population of non-target cells that co-separate with the target cells due to similar size.
  • the non-target cells do not interfere with downstream sample analysis that relies on detection of the bound labeling reagent, because this reagent binds selectively to the cells of interest.
  • Deterministic separation may be achieved using an array of gaps and obstacles in a channel. Exemplary configurations of such arrays, bypass channels, and boundaries are described as follows.
  • a single stage contains an array of obstacles, e.g., cylindrical posts ( FIG. 1D ).
  • the array has a maximum pass-through size that is several times larger than the cut-off size, e.g., when separating white blood cells from red blood cells. This result may be achieved using a combination of a large gap size d and a small bifurcation ratio ⁇ .
  • the ⁇ is at most 1/2, e.g., at most 1/3, 1/10, 1/30, 1/100, 1/300, or 1/1000.
  • the obstacle shape may affect the flow profile in the gap; however, the obstacles can be compressed in the flow direction, in order to make the array short ( FIG. 1E ).
  • Single stage arrays may include bypass channels as described herein.
  • Multiple-stage arrays In another embodiment, multiple stages are employed to separate analytes over a wide size range.
  • An exemplary device is shown in FIG. 7 .
  • the device shown has three stages, but any number of stages may be employed, and an array can have as many stages as desired.
  • the cut-off size in the first stage is larger than the cut-off in the second stage, and the first stage cut-off size is smaller than the maximum pass-through size of the second stage ( FIG. 8 ).
  • the first stage will deflect (and remove) analytes, e.g., that would cause clogging in the second stage, before they reach the second stage.
  • the second stage will deflect (and remove) analytes that would cause clogging in the third stage, before they reach the third stage.
  • devices of the invention may include bypass channels that remove output from an array. Although described here in terms of removing analytes above the critical size, a bypass channel may also be employed to remove output from any portion of the array.
  • Single bypass channels In this design, all stages share one bypass channel, or there is only one stage.
  • the physical boundary of the bypass channel may be defined by the array boundary on one side and a sidewall on the other ( FIGS. 9-11 ).
  • Single bypass channels may also be employed with duplex arrays ( FIG. 12 ).
  • Single bypass channels may also be designed, in conjunction with an array, to maintain constant flux through a device ( FIG. 13 ). As shown, the bypass channel has varying width designed maintain constant flux through all the stages, so that the flow in the channel does not interfere with the flow in the arrays. Such a design may also be employed with an array duplex ( FIG. 14 ). Single bypass channels may also be designed in conjunction with the array in order to maintain substantially constant fluidic resistance through all stages ( FIG. 15 ). Such a design may also be employed with an array duplex ( FIG. 16 .)
  • each stage has its own bypass channel, and the channels are separated from each other by sidewalls. Large analytes, e.g., cells are deflected into the major flux to the lower right corner of the first stage and then into in the bypass channel (bypass channel 1 in FIG. 17 ). Smaller cells that would not cause clogging in the second stage proceed to the second stage, and cells above the critical size of the second stage are deflected to the lower right corner of the second stage and into in another bypass channel (bypass channel 2 in FIG. 17 ).
  • This design may be repeated for as many stages as desired.
  • the bypass channels are not fluidically connected, allowing for collection or other manipulation of multiple fractions.
  • the bypass channels do not need to be straight or be physically parallel to each other ( FIG. 18 ). Multiple bypass channels may also be employed with duplex arrays ( FIG. 19 ).
  • bypass channels may be designed, in conjunction with an array to maintain constant flux through a device ( FIG. 20 ).
  • bypass channels are designed to remove an amount of flow so the flow in the array is not perturbed, i.e., substantially constant.
  • Such a design may also be employed with an array duplex ( FIG. 21 ). In this design, the center bypass channel may be shared between the two arrays in the duplex.
  • the distance between the array and the sidewall gradually decreases to allow for the removal of ⁇ to each gap from the boundary.
  • the flow pattern inside this array is not affected by the bypass channel because of the boundary design.
  • a wide boundary may be desired if the boundary serves as a bypass channel, e.g., to allow for collection of analytes.
  • a boundary may be employed that uses part of its entire flow to feed the array and feeds ⁇ into each gap at the boundary (represented by arrows in FIG. 24 ).
  • the bypass channel includes two flow-feeding boundaries.
  • the flux across the dashed line 1 in the bypass channel is ⁇ bypass .
  • a flow ⁇ joins ⁇ bypass from a gap to the left of the dashed line.
  • the shapes of the obstacles at the boundaries are adjusted so that the flows going into the arrays are ⁇ at each gap at the boundaries.
  • the flux at dashed line 2 is again ⁇ bypass .
  • Deterministic separation may also employ fluidic resistors to define and stabilize flows within an array and to also define the flows collected from the array.
  • FIG. 26 shows a schematic of planar device; a sample, e.g., blood, inlet channel, a buffer inlet channel, a waste outlet channel, and a product outlet channel are each connected to an array. The inlets and outlets act as flow resistors. FIG. 26 also shows the corresponding fluidic resistances of these different device components.
  • FIGS. 27 and 28 show the currents and corresponding widths of the sample and buffer flows within the array when the device has a constant depth and is operated with a given pressure drop. The flow is determined by the pressure drop divided by the resistance. In this particular device, I blood and I buffer are equivalent, and this determines equivalent widths of the blood and buffer streams in the array.
  • Each of the inlet and outlet channels can be designed so that the pressure drops across the channels are appreciable to or greater than the fluctuations of the overall driving pressure. In typical cases, the inlet and outlet pressure drops are 0.001 to 0.99 times the driving pressure.
  • Deterministic separation may be achieved using multiplexed deterministic arrays. Putting multiple arrays on one device increases sample-processing throughput, and allows for parallel processing of multiple samples or portions of the sample for different fractions or manipulations. Multiplexing is further desirable for preparative applications.
  • the simplest multiplex device includes two devices attached in series, i.e., a cascade. For example, the output from the major flux of one device may be coupled to the input of a second device. Alternatively, the output from the minor flux of one device may be coupled to the input of the second device.
  • Two arrays can be disposed side-by-side, e.g., as mirror images ( FIG. 29 ).
  • the critical size of the two arrays may be the same or different.
  • the arrays may be arranged so that the major flux flows to the boundary of the two arrays, to the edge of each array, or a combination thereof.
  • Such a multiplexed array may also contain a central region disposed between the arrays, e.g., to collect analytes above the critical size or to alter the sample, e.g., through buffer exchange, reaction, or labeling.
  • two or more arrays that have separated inputs may be disposed on the same device ( FIG. 30A ). Such an arrangement could be employed for multiple samples, or the plurality of arrays may be connected to the same inlet for parallel processing of the same sample.
  • the outlets may or may not be fluidically connected. For example, when the plurality of arrays has the same critical size, the outlets may be connected for high throughput samples processing.
  • the arrays may not all have the same critical size or the analytes in the arrays may not all be treated in the same manner, and the outlets may not be fluidically connected.
  • Multiplexing may also be achieved by placing a plurality of duplex arrays on a single device ( FIG. 30B ).
  • a plurality of arrays, duplex or single, may be placed in any possible three-dimensional relationship to one another.
  • FIG. 58A shows the “cascade” configuration, in which outlet 1 of one device is joined to a sample inlet of a second device. This allows for an initial separation step using the first device so that the sample introduced to the second device is already enriched for cells of interest.
  • the two devices may have either identical or different critical sizes, depending on the intended application.
  • an unlabeled cellular sample is introduced to the first device in the cascade via a sample inlet, and a buffer containing labeling reagent is introduced to the first device via the fluid inlet.
  • Epithelial cells are deflected and emerge from the center outlet in the buffer containing labeling reagent.
  • This enriched labeled sample is then introduced to the second device in the cascade via a sample inlet, while buffer is added to the second device via the fluid inlet. Further enrichment of target cells and separation of free labeling reagent is achieved, and the enriched sample may be further analyzed.
  • labeling reagent may be added directly to the sample emerging from the center outlet of the first device before introduction to the second device.
  • the use of a cascade configuration may allow for the use of a smaller quantity or a higher concentration of labeling reagent at less expense than the single-device configuration of FIG. 60 ; in addition, any nonspecific binding that may occur is significantly reduced by the presence of an initial separation step using the first device.
  • FIG. 58B shows this configuration, in which outlet 2 of one device is joined to a sample inlet of a second device. This allows for an initial separation step using the first device so that the sample introduced to the second device contains cells that remained undeflected within the first device. This method may be useful when the cells of interest are not the largest cells in the sample; in this instance, the first stage may be used to reduce the number of large non-target cells by deflecting them to the center outlet.
  • the two devices may have either identical or different critical sizes, depending on the intended application. For example, different critical sizes are appropriate for an application requiring the separation of epithelial cells, in comparison with an application requiring the separation of smaller endothelial cells.
  • a cellular sample pre-incubated with labeling reagent is introduced to a sample inlet of the first device of the bandpass configuration, and a buffer is introduced to the first device via the fluid inlet.
  • the first device is disposed in such a manner that large, non-target cells are deflected and emerge from the center outlet, while a mixture of target cells, small non-target cells, and labeling reagent emerge from outlet 2 of the first device.
  • This mixture is then introduced to the second device via a sample inlet, while buffer is added to the second device via the fluid inlet. Enrichment of target cells and separation of free labeling reagent is achieved, and the enriched sample may be further analyzed.
  • Non-specific binding of labeling reagent to the deflected cells in the first stage is acceptable in this method, as the deflected cells and any bound labeling reagent are removed from the system.
  • the devices and the connections joining them may be integrated into a single device.
  • a single cascade device including two or more stages is possible, as is a single bandpass device including two or more stages.
  • the output of the multiple stages is then coupled to the input of the reservoir.
  • Small-footprint arrays Deterministic devices may also feature a small footprint. Reducing the footprint of an array can lower cost, and reduce the number of collisions with obstacles to eliminate any potential mechanical damage or other effects to analytes.
  • the length of a multiple stage array can be reduced if the boundaries between stages are not perpendicular to the direction of flow. The length reduction becomes significant as the number of stages increases.
  • FIG. 31 shows a small-footprint three-stage array.
  • An analytical device e.g., a deterministic device
  • a reservoir containing a reagent (e.g., magnetic particles having a binding moiety or sodium nitrite) capable of altering a magnetic property of an analyte (e.g., a cell such as a red blood cell).
  • the reservoir may include a channel, e.g., a microfluidic channel, a tube, or any other container capable of receiving the analyte and contacting it with the reagent.
  • the reservoir may be separable from the analytical device or may be integrated with it. Mixing of the reagent with the analyte may occur by any means including diffusion, mechanical mixing, or turbulent flow.
  • the reagent may be stored dry in the reservoir and liquefied upon introduction of a sample or stored in solution and mixed with the sample.
  • the reagent is added continuously or in a discrete bolus to the reservoir concomitant with the delivery of the sample.
  • the reservoir may also include structures that allow for the separation of the altered analyte from the unreacted reagent. For example, deterministic separation may be employed for this purpose as described herein. Alternatively, filters, rinses, or other means may be employed. Such a structure may or may not be included as part of the reservoir or analytical device.
  • the reservoir may also include an apparatus useful in enriching or depleting a sample in the magnetically altered analyte.
  • Such devices are described herein and include channels (e.g., microfluidic channels) which, in some embodiments include a magnetic field generator or a channel containing a magnet such as a MACS column (e.g., an MS or LD column from Miltenyi Biotec, Inc., Auburn, Calif.).
  • the reservoir includes a channel having a magnetic region to which a magnetic particle can magnetically attach, thereby creating a textured surface with which an analyte passing through the channel can come into contact.
  • a texture that enhances interactions between an analyte and the bound magnetic particles can be provided.
  • the magnetic particles may be coated with appropriate capture moieties such as antibodies (e.g., anti-CD71, anti-CD36, anti-CD45, anti-GPA, anti-antigen i, anti-CD34, anti-fetal hemoglobin, anti-EpCAM, anti-E-cadherin, or anti-Muc-1) that can bind to an analyte through affinity mechanisms.
  • the magnetic particles can be disposed uniformly throughout a device or in spatially resolved regions.
  • magnetic particles may be used to create structure within the device. For example, two magnetic regions on opposite sides of a channel can be used to attract magnetic particles to form a “bridge” linking the two regions.
  • the magnetic particles can be magnetically attached to hard magnetic regions of the channel or to soft magnetic regions that are actuated to produce a magnetic field.
  • the sample is treated with a reagent that includes magnetic particles prior to application of a magnetic field.
  • the magnetic particles may be coated with appropriate capture moieties such as antibodies to which an analyte can bind.
  • Application of a magnetic field to the treated sample will selectively bind an analyte bound to magnetic particles in the reservoir.
  • a sample is combined with a reagent that alters an intrinsic magnetic property of an analyte.
  • the altered analyte may be rendered more magnetically responsive, less magnetically responsive, or may be rendered magnetically unresponsive by the reagent as compared to the unaltered analyte.
  • a sample e.g., a maternal blood sample that has, for example, been depleted of maternal red blood cells
  • fRBCs fetal red blood cells
  • This oxidation alters the magnetic responsiveness of the fetal hemoglobin relative to other components of the sample, e.g., maternal white blood cells, thereby allowing separation of the fRBCs.
  • Any cell containing magnetically responsive components such as iron found in hemoglobin (e.g., adult or fetal), myoglobin, or cytochromes (e.g., cytochrome C) may be modified to alter intrinsic magnetic responsiveness of an analyte such as a cell, or a component thereof (e.g., an organelle).
  • any source of a magnetic field may be employed in the invention and may include hard magnets, soft magnets, or a combination thereof.
  • a spatially nonuniform permanent magnet or electromagnet may be used to create organized and in some cases periodic arrays of magnetic particles within an otherwise untextured microfluidic channel (Deng et al. Applied Physics Letters, 78, 1775 (2001)).
  • An electromagnet may be employed to create a non-uniform magnetic field in a device.
  • the non-uniform filed creates regions of higher and lower magnetic field strength, which, in turn, will attract magnetic particles in a periodic arrangement within the device.
  • Other external magnetic fields may be employed to create magnetic regions to which magnetic particles attach.
  • a hard magnetic material may also be used in the fabrication of the device, thereby obviating the need for electromagnets or external magnetic fields.
  • the device contains a plurality of channels having magnetic regions, e.g., to increase volumetric throughput. Further, these channels may be stacked vertically.
  • an analyte bound to a magnet can be released from defined locations within the channel, e.g., by increasing the overall flow rate of the fluid flowing through the device, decreasing the magnetic field, or through some combination of the two.
  • FIG. 74 An example of a reservoir is shown in FIG. 74 , which illustrates a reservoir geometry and functional process flow to isolate and then release target analytes, e.g., cells or molecules, from a complex mixture.
  • the reservoir contains obstacles that extend from one channel surface toward the opposing channel surface.
  • the obstacles may or may not extend the entire distance across the channel.
  • the obstacles are magnetic (e.g., contain hard or soft magnetic materials or are locations of high magnetic field in a non-uniform field) and attract and retain magnetic particles, which may be coated with capture moieties or may be cells attracted to a magnetic field.
  • the geometry of the reservoir, the distribution, shape, size of the obstacles and the flow parameters can be altered to optimize the efficiency of the enrichment of an analyte of interest, for example, by attracting an analyte bound to a magnetic particle with a capture moiety (e.g., as described in International Publication No. 2004/029221).
  • an anodic lidded silicon wafer with microtextured magnetic obstacles of varying shapes are arranged uniquely (spacing and density varied across equilateral triangular, diagonal, and random array distribution) to maximize the collision frequency of analytes, altered or not, with the obstacles within the confines of a continuous perfusion flow stream.
  • the exact geometry of the magnetic obstacles and the distribution of obstacles may depend on the type of analytes being isolated, enriched, or purified.
  • FIG. 75 illustrates an example of reservoir fabrication and functionalization.
  • the magnetized obstacles enable post-packaging modification of the reservoir. This is a very significant improvement over existing art.
  • the incompatibility of semiconductor processing parameters high heat, or solvent sealers to bond the lid
  • capture moieties sensitive to temperature and inorganic and organic solvents
  • the reservoir enables the end user to easily and rapidly charge the reservoir with a capture moiety, or mixture of capture moieties, of choice thereby increasing the versatility of use.
  • the capture moieties that can be loaded and retained on the obstacles include, but not limited to, all of the cluster of differentiation (CD) receptors on mammalian cells, synthetic and recombinant ligands for cell receptors, and any other organic, inorganic molecule, or compound of interest that can be attached to any magnetic particle.
  • CD cluster of differentiation
  • Such reagents include any reagent that is capable of altering a magnetic property of an analyte in a sample.
  • the exact nature of the reagent will depend on the nature of the analyte.
  • Exemplary reagents include agents that oxidize or reduce transition metals, magnetic beads capable of binding to an analyte, or reagents that are capable of chelating or otherwise binding iron, or other magnetic materials or particles.
  • Specific reagents include sodium nitrite.
  • the reagent may act to alter the magnetic properties of an analyte to enable or increase its attraction to a magnetic field, to enable or increase its repulsion to a magnetic field, or to eliminate a magnetic property such that the analyte is unaffected by a magnetic field.
  • any particle that responds to a magnetic field may be employed in the devices and methods of the invention.
  • Desirable particles are those that have surface chemistry that can be chemically or physically modified, e.g., by chemical reaction, physical adsorption, entanglement, or electrostatic interaction.
  • Magnetic particles of the present invention can come in any size and/or shape.
  • a magnetic particle has a diameter of less than 500 nm, 400 nm, 300 nm, 200 nm, 100 nm, 90 nm, 80 nm, 70 nm, 60 nm or 50 nm.
  • a magnetic particle has a diameter that is between 10-1000 nm, 20-800 nm, 30-600 nm, 40-400 nm, or 50-200 nm. In some embodiments, a magnetic particle has a diameter of more than 10 nm, 50 nm, 100 nm, 200 nm, 500 nm, 1000 nm, or 5000 nm.
  • the magnetic particles can be dry in liquid form. Mixing of a fluid sample with a second liquid medium containing magnetic particles can occur by any means known in the art.
  • Capture moieties can be bound to magnetic particles by any means known in the art. Examples include chemical reaction, physical adsorption, entanglement, or electrostatic interaction. The capture moiety bound to a magnetic particle will depend on the nature of the analyte targeted. Examples of capture moieties include, without limitation, proteins (such as antibodies, avidin, and cell-surface receptors), charged or uncharged polymers (such as polypeptides, nucleic acids, and synthetic polymers), hydrophobic or hydrophilic polymers, small molecules (such as biotin, receptor ligands, and chelating agents), and ions.
  • proteins such as antibodies, avidin, and cell-surface receptors
  • charged or uncharged polymers such as polypeptides, nucleic acids, and synthetic polymers
  • hydrophobic or hydrophilic polymers such as small molecules (such as biotin, receptor ligands, and chelating agents), and ions.
  • capture moieties can be used to specifically bind cells (e.g., bacterial, pathogenic, fetal cells, fetal blood cells, cancer cells, and blood cells), organelles (e.g., nuclei), viruses, peptides, protein, polymers, nucleic acids, supramolecular complexes, other biological molecules (e.g., organic or inorganic molecules), small molecules, ions, or combinations or fragments thereof.
  • cells e.g., bacterial, pathogenic, fetal cells, fetal blood cells, cancer cells, and blood cells
  • organelles e.g., nuclei
  • viruses e.g., bacterial, pathogenic, fetal cells, fetal blood cells, cancer cells, and blood cells
  • organelles e.g., nuclei
  • viruses e.g., bacterial, pathogenic, fetal cells, fetal blood cells, cancer cells, and blood cells
  • organelles e.g., nuclei
  • viruses e.g., bacterial
  • the isolation, enrichment, or depletion may include positive selection, i.e., a desired analyte is attracted to a magnetic field, or it may employ negative selection, i.e., a desired analyte is not attracted to the magnetic field. In either case, the population of analytes containing the desired analytes may be collected for analysis or further processing.
  • the device used to perform the magnetic separation may be any device that can produce a magnetic field.
  • a MACS column is used to effect separation of the magnetically altered analyte. If the analyte is rendered magnetically responsive by the reagent (e.g., using any reagent described herein), it may bind to the MACS column, thereby permitting enrichment of the desired analyte relative to other constituents of the sample.
  • separation may be achieved using a device, typically microfluidic, that contains a plurality of magnetic obstacles.
  • an analyte in the sample is modified to be magnetically responsive (e.g., through a reagent that enhances an intrinsic magnetic property of the analyte or by binding of a magnetically responsive particle to the analyte)
  • the analyte may bind to the obstacles, thereby permitting enrichment of the bound analyte.
  • negative selection may be employed.
  • the desired analyte may be rendered magnetically unresponsive, or an undesired analyte may be rendered magnetically responsive or bound to a magnetically responsive particle. In this case, an undesired analyte or analytes will be retained on the obstacles whereas the desire analyte will not, thus enriching the sample in the desired analyte.
  • Magnetic regions of the device can be fabricated with either hard or soft magnetic materials, such as, but not limited to, rare earth materials, neodymium-iron-boron, ferrous-chromium-cobalt, nickel-ferrous, cobalt-platinum, and strontium ferrite. Portions of the device may be fabricated directly out of magnetic materials, or the magnetic materials may be applied to another material.
  • hard magnetic materials can simplify the design of a device because they are capable of generating a magnetic field without other actuation.
  • Soft magnetic materials enable release and downstream processing of bound analytes simply by demagnetizing the material.
  • the application process can include cathodic sputtering, sintering, electrolytic deposition, or thin-film coating of composites of polymer binder-magnetic powder.
  • a preferred embodiment is a thin film coating of micromachined obstacles (e.g., silicon posts) by spin casting with a polymer composite, such as polyimide-strontium ferrite (the polyimide serves as the binder, and the strontium ferrite as the magnetic filler).
  • the polymer magnetic coating is cured to achieve stable mechanical properties.
  • the device is briefly exposed to an external induction field, which governs the preferred direction of permanent magnetism in the device.
  • the magnetic flux density and intrinsic coercivity of the magnetic fields from the obstacles can be controlled by the % volume of the magnetic filler.
  • an electrically conductive material is micropatterned on the outer surface of an enclosed microfluidic device.
  • the pattern may consist of a single, electrical circuit with a spatial periodicity of approximately 100 microns.
  • the magnetic field can be adjusted to influence supra and paramagnetic particles with magnetic mass susceptibility ranging from 0.1-200 ⁇ 10 ⁇ 6 m 3 /kg.
  • the paramagnetic particles of use can be classified based on size: particulates (1-5 ⁇ m in the size of a cell diameter); colloidal (on the order of 100 nm); and molecular (on the order of 2-10 nm).
  • F b is the magnetic force acting on the paramagnetic entity of volume V b
  • is the difference in magnetic susceptibility between the magnetic bead, ⁇ b, and the surrounding medium
  • ⁇ f is the magnetic permeability of free space
  • B is the external magnetic field
  • is the gradient operator.
  • the magnetic field can be controlled and regulated to enable attraction and retention of a wide spectrum of particulate, colloidal, and molecular paramagnetic entities typically coupled to capture moieties.
  • the invention features analytical devices for the enrichment of analytes such as particles, including bacteria, viruses, fungi, cells, cellular components, viruses, nucleic acids, proteins, and protein complexes.
  • analytes such as particles, including bacteria, viruses, fungi, cells, cellular components, viruses, nucleic acids, proteins, and protein complexes.
  • a device may also be used to effect various manipulations on analytes in a sample. Such manipulations include enrichment or concentration of an analyte, including size-based fractionization, or alteration of the analyte itself or the fluid carrying the analyte.
  • a device is employed to enrich rare analytes from a heterogeneous mixture or to alter a rare analyte, e.g., by exchanging the liquid in the sample or by contacting an analyte with a reagent.
  • Such devices allow for a high degree of enrichment with limited stress on a potentially fragile analyte such a cell, where devices of the invention provide reduced mechanical lysis or intracellular activation of cells.
  • the devices of the invention may be employed with any analyte whose size allows for separation in a device of the invention.
  • Deterministic devices may be employed in concentrated samples, e.g., where analytes are touching, hydrodynamically interacting with each other, or exerting an effect on the flow distribution around another analyte.
  • a deterministic device can separate white blood cells from red blood cells in whole blood from a human donor. Human blood typically contains ⁇ 45% of cells by volume. Cells are in physical contact and/or coupled to each other hydrodynamically when they flow through the array.
  • FIG. 32 shows schematically that cells densely packed inside an array can physically interact with each other.
  • the devices and methods of the invention may involve separating from a sample one or more analytes based on an intrinsic magnetic property of the one or more analytes.
  • the sample is treated with a reagent that alters a magnetic property of the analyte.
  • the alteration may be mediated by a magnetic particle or may be mediated by a reagent that alters an intrinsic magnetic property of the analyte.
  • a magnetically altered analyte may then bind to a surface of the device, and desired analytes (e.g., rare cells such as fetal cells, pathogenic cells, cancer cells, or bacterial cells) in a sample may be retained in the device.
  • the analyte of interest may then bind to the surfaces of the device.
  • desired analytes are retained in the device through size-, shape- or deformability-based mechanisms.
  • negative selection is employed, where a desired analyte is not bound to the device. Any of the embodiments may uses a MACS column for retention of an analyte (e.g., an analyte bound to a magnetic particle).
  • the analytes are retained in the device.
  • the surfaces of the device are desirably designed to minimize nonspecific binding of non-target analytes. For example, at least 99%, 98%, 95%, 90%, 80%, or 70% of non-target analytes are not retained in the device.
  • the selective retention in the device can result in the separation of a specific analyte population from a mixture, e.g., blood, sputum, urine, and soil, air, or water samples.
  • the selective retention of analytes may be obtained by introduction of magnetic particles (e.g., attached to obstacles present in the device or manipulated to create obstacles to increase surface area for an analyte to interact with to increase the likelihood of binding) into a device of the invention.
  • Capture moieties may be bound to the magnetic particles to effect specific binding of a target analyte.
  • the magnetic particles may be disposed such as to only allow analytes of a selected size, shape, or deformability to pass through the device. Combinations of these embodiments are also envisioned.
  • a device may be configured to retain certain analytes based on size and others based on binding.
  • a device may be designed to bind more than one analyte of interest, e.g., in a serial, parallel, or interspersed arrangement of regions within a device or where two or more capture moieties are disposed on the same magnetic particle or on adjacent particles, e.g., bound to the same obstacle or region.
  • multiple capture moieties that are specific for the same analyte e.g., anti-CD71 and anti-CD36
  • the flow conditions in the device are typically such that the analytes are very gently handled in the device to prevent damage.
  • Positive pressure or negative pressure pumping or flow from a column of fluid may be employed to transport analytes into and out of the microfluidic devices of the invention.
  • the device enables gentle processing, while maximizing the collision frequency of each analyte with one or more of the magnetic particles.
  • the target analytes interact with any capture moieties on collision with the magnetic particles.
  • the capture moieties can be co-localized with obstacles as a designed consequence of the magnetic field attraction in the device. This interaction leads to capture and retention of the target analytes in defined locations. Captured analyte can be released by demagnetizing the magnetic regions retaining the magnetic particles.
  • the demagnetization can be limited to selected obstacles or regions.
  • the magnetic field can be designed to be electromagnetic, enabling turn-on and turn-off off the magnetic fields for each individual region or obstacle at will.
  • the particles can be released by disrupting the bond between the analyte and the capture moiety, e.g., through chemical cleavage or interruption of a noncovalent interaction, or by decreasing the magnetic responsiveness of the bound analyte.
  • some ferrous particles are linked to monoclonal antibody via a DNA linker; the use of DNAse can cleave and release the analytes from the ferrous particle.
  • an antibody fragmenting protease e.g., papain
  • an antibody fragmenting protease can be used to engineer selective release. Increasing the sheer forces on the magnetic particles can also be used to release magnetic particles from magnetic regions, especially hard magnetic regions.
  • the captured analytes are not released and can be analyzed or further manipulated while retained.
  • FIG. 76 illustrates an example of a reservoir designed to capture and isolate cells expressing the transferrin receptor from a complex mixture.
  • Monoclonal antibodies to CD71 receptor are readily available off-the-shelf covalently coupled to magnetic materials, such as, but not limited to, ferrous doped polystyrene and ferroparticles or ferro-colloids (e.g., from Miltenyi and Dynal).
  • the mAB to CD71 bound to magnetic particles is flowed into the reservoir.
  • the antibody-coated particles are attracted to the obstacles (e.g., posts), floor, and walls and are retained by the strength of the magnetic field interaction between the particles and the magnetic field.
  • the particles between the obstacles and those loosely retained with the sphere of influence of the local magnetic fields away from the obstacles are removed by a rinse (the flow rate can be adjusted such that the hydrodynamic shear stress on the particles away from the obstacles is larger than the magnetic field strength).
  • FIG. 77 is a preferred embodiment for application of the reservoir to capture and release CD71+ cells from a complex mixture, e.g., blood, using holo-transferrin.
  • Holo-transferrin is rich in iron content, commercially available, and has higher affinity constants and specificity of interaction with the CD71 receptor than its counterpart monoclonal antibody.
  • the iron coupled to the transferrin ligand serves the dual purpose of retaining the conformation of the ligand for binding with the cell receptor, and as a molecular paramagnetic element for retaining the ligand on the obstacles.
  • devices of the invention can be used for isolation and detection of blood borne pathogens, bacterial and viral loads, airborne pathogens solubilized in aqueous medium, pathogen detection in food industry, and environmental sampling for chemical and biological hazards.
  • the magnetic particles can be co-localized with a capture moiety and a candidate drug compound. Capture of a cell of interest can further be analyzed for the interaction of the captured cell with the immobilized drug compound.
  • a device can thus be used to both isolate sub-populations of cells from a complex mixture and assay their reactivity with candidate drug compounds for use in the pharmaceutical drug discovery process for high throughput and secondary cell-based screening of candidate compounds.
  • receptor-ligand interaction studies for drug discovery can be accomplished in the device by localizing the capture moiety, i.e., the receptor, on a magnetic particle, and flowing in a complex mixture of candidate ligands (or agonists or antagonists).
  • the ligand of interest is captured, and the binding event can be detected, e.g., by secondary staining with a fluorescent probe.
  • This embodiment enables rapid identification of the absence or presence of known ligands from complex mixtures extracted from tissues or cell digests or identification of candidate drug compounds.
  • devices of the invention are employed to produce a sample enriched in a desired analyte, e.g., based at least in part on hydrodynamic size.
  • Applications of such enrichment include concentrating of an analyte such as particle including rare cells, and size fractionization, e.g., size filtering (selecting analytes in a particular size range).
  • Devices may also be used to enrich components of cells such as organelles (e.g., nuclei).
  • the devices and methods of the invention retain at least 1%, 10%, 30%, 50%, 75%, 80%, 90%, 95%, 98%, or 99% of the desired particles compared to the initial mixture, while potentially enriching the desired particles by a factor of at least 1, 10, 100, 1,000, 10,000, 100,000, or even 1,000,000 relative to one or more non-desired particles.
  • the enrichment may also result in a dilution of the enriched analytes compared to the original sample, although the concentration of the enriched analytes relative to other particles in the sample has increased.
  • the dilution is at most 90%, e.g., at most 75%, 50%, 33%, 25%, 1 0%, or 1%.
  • a device of the invention is used to produce a sample enriched in a rare analyte.
  • a rare analyte is an analyte that is present as less than 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, 0.1%, 0.01%, 0.001%, 0.0001%, 0.00001%, or 0.000001% of all analytes in a sample or whose mass is less than 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, 0.1%, 0.01%, 0.001%, 0.0001%, 0.00001%, or 0.000001% of total mass of a sample of a sample.
  • Exemplary rare analytes include, depending on the sample, fetal cells, stem cells (e.g., undifferentiated), cancer cells, immune system cells (host or graft), epithelial cells, connective tissue cells, bacteria, fungi, viruses, and pathogens (e.g., bacterial or protozoa).
  • Such rare analytes may be isolated from samples including bodily fluids, e.g., blood, or environmental sources, e.g., pathogens in water samples.
  • Fetal red blood cells may be enriched from maternal peripheral blood, e.g., for the purpose of determining sex and identifying aneuploidies or genetic characteristics, e.g., mutations, in the developing fetus.
  • Circulating tumor cells which are of epithelial cell type and origin, may also be enriched from peripheral blood for the purpose of diagnosis and monitoring therapeutic progress. Circulating endothelial cells may also be similarly enriched from peripheral blood.
  • Bodily fluids or environmental samples may also be screened for pathogens, e.g., for coliform bacteria, blood borne illnesses such as sepsis, or bacterial or viral meningitis.
  • pathogens e.g., for coliform bacteria, blood borne illnesses such as sepsis, or bacterial or viral meningitis.
  • Rare cells also include cells from one organism present in another organism, e.g., in cells from a transplanted organ.
  • a device may be employed for preparative applications.
  • An exemplary preparative application includes generation of cell packs from blood.
  • a device may be configured to produce fractions enriched in platelets, red blood cells, and white cells by deterministic deflection. By using multiplexed or multistage deterministic devices, all three cellular fractions may be produced in parallel or in series from the same sample.
  • the device may be employed to separate nucleated from non-nucleated cells, e.g., from cord blood sources.
  • deterministic devices may be designed to enrich analytes (e.g., a cell) with a minimum number of collisions between the analyte and obstacles. This minimization reduces mechanical damage to the analytes (e.g., a cell) and, in the case of cells, also prevents intracellular activation caused by the collisions.
  • Gentle handling preserves the limited number of rare analytes in a sample, in the case of cells, prevents rupture leading to contamination or degradation by intracellular components, and prevents maturation or activation of cells, e.g., stem cells or platelets.
  • the analyte is enriched such that fewer than 30%, 10%, 5%, 1%, 0.1%, or even 0.01% are damaged (e.g., activated or mechanically lysed).
  • FIG. 33 shows a typical size distribution of cells in human peripheral blood.
  • the white blood cells range from ⁇ 4 ⁇ m to ⁇ 18 ⁇ m, whereas the red blood cells are ⁇ 1.5 ⁇ m (short axis).
  • a deterministic device designed to separate white blood cells from red blood cells typically has a cut-off size of 2 to 4 ⁇ m and a maximum pass-through size of greater than 18 ⁇ m.
  • a deterministic device may function as a detector for abnormalities in red blood cells.
  • the deterministic principle of sorting enables a predictive outcome of, for example, % of enucleated cells deflected in the device.
  • % of enucleated cells deflected in the device In a disease state, such as malarial infection or sickle cell anemia, the distortion in shape and flexibility of the red cells would significantly change the % cells deflected. This change can be monitored as a first level sentry to alert to the potential of a diseased physiology to be followed by microscopy examination of shape and size of red cells to assign the disease.
  • the method is also generally applicable monitoring for any change in flexibility of particles in a sample.
  • a deterministic device may function as a detector for platelet aggregation.
  • the deterministic principle of sorting enables a predictive outcome of % free platelets deflected in the device. Activated platelets would form aggregates and the aggregates would be deflected. This change can be monitored as a first level sentry to alert the compromised efficacy of a platelet pack for reinfusion.
  • the method is also generally applicable monitoring for any change in size, e.g., through agglomeration, of particles in a sample.
  • FIG. 57A shows the operation of a deterministic device for purposes of enrichment.
  • a cellular sample is added through a sample inlet of the device, and buffer medium is added through the fluid inlet.
  • Cells below the critical size move through the device undeflected, emerging from the edge outlets in their original sample medium.
  • Cells above the critical size e.g., epithelial cells, are deflected and emerge from the center outlet contained in the buffer medium added through the fluid inlet. Operation of the device thus produces samples enriched in cells above and below the critical size.
  • the size and geometry of the gaps of the device may be chosen so as to direct virtually all other cell types to the edge outlets, while producing a sample from the center outlet that is substantially enriched in epithelial cells after a single pass through the device.
  • a deterministic device included in the invention need not be duplexed as shown in FIG. 57A in order to operate as described herein.
  • the schematized representation shown in FIG. 57B may represent either a duplexed device or a single array.
  • Enrichment may be enhanced in numerous ways.
  • target analytes e.g., cells
  • beads e.g., immunoaffinity beads
  • this may further increase their size, resulting in an even more efficient separation.
  • the size of smaller analytes e.g., cells
  • Beads may be made of polystyrene, magnetic material, or any other material that can be adhered to an analyte (e.g., cells). Desirably, such beads are neutrally buoyant so as not to disrupt the flow of labeled cells through a deterministic device.
  • an analyte of interest may be contacted with an altering reagent that may chemically or physically alter the analyte or the fluid in the sample.
  • altering reagent may chemically or physically alter the analyte or the fluid in the sample.
  • Such applications may include purification, buffer exchange, labeling (e.g., immunohistochemical, magnetic, and histochemical labeling, cell staining, and flow in-situ fluorescence hybridization (FISH)), cell fixation, cell stabilization, cell lysis, and cell activation.
  • labeling e.g., immunohistochemical, magnetic, and histochemical labeling, cell staining, and flow in-situ fluorescence hybridization (FISH)
  • FISH flow in-situ fluorescence hybridization
  • FIG. 34A shows this effect schematically for a single stage deterministic device
  • FIG. 34B shows this effect for a multistage deterministic device
  • FIG. 34C shows this effect for a duplex array of deterministic devices
  • FIG. 34D shows this effect for a multistage duplex array of deterministic devices.
  • analytes e.g., blood cells
  • Such transfers of an analyte from one liquid to another may be also employed to effect a series of alterations, e.g., Wright staining blood on-chip.
  • Such a series may include reacting an analyte with a first reagent and then transferring the particle to a wash buffer, and then another reagent.
  • FIGS. 35A-35C illustrates a further example of alteration in a two stage deterministic device having two bypass channels.
  • the larger analytes are moved from blood to buffer and collected in stage 1
  • intermediate sized analytes are moved from blood to buffer in stage 2
  • smaller analytes that are not moved from the blood in stage are collected also collected.
  • FIG. 35B illustrates the size cut-off of the two stages
  • FIG. 35C illustrates the size distribution of the three fractions collected.
  • FIG. 36 illustrates an example of alteration in a two stage deterministic device having bypass channels that are disposed between the lateral edge of the array and the channel wall.
  • FIG. 37 illustrates a deterministic device similar to that in FIG. 36 , except that the two stages are connected by fluidic channels.
  • FIG. 38 illustrates alteration in a deterministic device having two stages with a small footprint.
  • FIGS. 39A-39B illustrates alteration in a device in which the output from the first and second stages is captured in a single channel.
  • FIG. 40 illustrates another device for use in the methods of the invention.
  • FIG. 41 illustrates the use of a deterministic device to perform multiple, sequential alterations on an analyte.
  • an analyte is moved from the sample into a regent that reacts with the analyte, and the altered analyte is then moved into a buffer, thereby removing the unreacted reagent or reaction byproducts. Additional steps may be added (e.g., steps described herein).
  • Enrichment and alteration may also be combined.
  • desired cells may be contacted with a lysing reagent and cellular components, e.g., nuclei, are enriched based on size.
  • cellular components e.g., nuclei
  • analytes may be contacted with particulate labels, e.g., magnetic beads, which bind to the analytes. Unbound particulate labels may be removed based on size.
  • Deterministic devices may also be employed for purposes of buffer exchange. To achieve this result, a protocol similar to that used for enrichment is followed: a cellular sample is added through a sample inlet of a deterministic device, and the desired final buffer medium is added through a fluid inlet. As described above, cells above the critical size are deflected in the device and enter the buffer.
  • Devices of the invention may also be employed in order to concentrate a cellular sample of interest.
  • a cellular sample is introduced to the sample inlet of a deterministic device.
  • concentration of target cells in a smaller volume results. This concentration step may improve the results of any downstream analysis performed.
  • Devices of the invention may also be employed for purposes of cell lysis.
  • a protocol similar to that used for enrichment is followed: a cellular sample is added through a sample inlet of the device ( FIG. 63 ), and lysis buffer is added through the fluid inlet.
  • a cellular sample is added through a sample inlet of the device ( FIG. 63 )
  • lysis buffer is added through the fluid inlet.
  • cells above the critical size are deflected and enter the lysis buffer, leading to lysis of these cells.
  • the sample emerging from the center outlet includes lysed cell components including organelles, while undeflected whole cells emerge from the other outlet.
  • the device provides a method for selectively lysing target cells.
  • the reagent is a labeling reagent.
  • deterministic devices are able to separate free labeling reagent from labeling reagent bound to an analyte (e.g., a cell). It is then possible to perform a bulk measurement of the reacted sample without significant levels of background interference from free labeling reagent.
  • fluorescent antibodies selective for a particular epithelial cell marker such as EpCAM are used.
  • the fluorescent moiety may include Cy dyes, Alexa dyes, or other fluorophore-containing molecules.
  • the resulting labeled sample is then analyzed by measuring the fluorescence of the resulting sample of labeled enriched analytes such as cells using a fluorimeter.
  • a chromophore-containing label may be used in conjunction with a light spectrometer. The measurements obtained may be used to quantify the number of target analytes such as cells in a sample.
  • Labeling antibodies may possess covalently bound enzymes that cleave a substrate, altering its absorbance at a given wavelength; the extent of cleavage is then quantified with a spectrometer. Colorimetric or luminescent readouts are possible, depending on the substrate used.
  • an enzyme label allows for significant amplification of the measured signal, lowering the threshold of detectability.
  • Quantum dots e.g., Qdots® from QuantumDot Corp.
  • Qdots® may also be utilized as a labeling reagent that is covalently bound to a capture moiety such as an antibody.
  • Qdots are resistant to photobleaching and may be used in conjunction with two-photon excitation measurements.
  • Phage display is a technology in which binding peptides are displayed by engineered phage strains having strong binding affinities for a target, e.g., a protein found on the surface of cells of interest.
  • the peptide sequence corresponding to a given phage is encoded in that phage's DNA.
  • phage are useful labeling reagents in that they are small relative to an analyte such as a cell and thus may be easily separated, and they additionally carry DNA that may be analyzed and quantified using PCR or similar techniques, enabling a quantitative determination of the number of cells present in an enriched bound sample.
  • FIG. 65 depicts the use of phage as a labeling reagent in which two deterministic device stages are arrayed in a cascade configuration.
  • the method depicted in FIG. 65 fits the general description of FIG. 64 , with the exception of the labeling reagent employed.
  • Downstream analysis may include an accurate determination of the number of desired analytes (e.g., cells) in the sample being analyzed.
  • the amount of the target of a labeling reagent typically has to be known or predictable (e.g., based on expression levels in a cell), and the binding of the labeling reagent should also proceed in a predictable manner, free from interfering substances.
  • a device e.g., a deterministic devices
  • method that produces a highly enriched cellular samples prior to introduction of a labeling reagent are particularly useful.
  • labeling reagents that allow for amplification of the signal produced are preferred in the case of a rare desired analyte (e.g., epithelial cells in a blood sample).
  • Reagents that allow for signal amplification include enzymes and phage.
  • Other labeling reagents that do not allow for convenient amplification but nevertheless produce a strong signal, such as quantum dots, are also desirable.
  • the devices and methods of the invention are used to enrich cells contained in a sample, further quantification and molecular biology analysis may be performed on the same set of cells.
  • techniques that destroy the integrity of the cells may be performed subsequent to bulk measurement; such techniques include DNA or RNA analysis, proteome analysis, or metabolome analysis.
  • An example of such analysis is PCR, in which the cells are lysed and levels of particular DNA sequences are amplified. Such techniques are particularly useful when the number of target cells isolated is very low.
  • CTCs circulating tumor cells
  • CTCs which are of epithelial cell type and origin, have a short half-life of approximately one day, and their presence indicates a recent influx from a proliferating tumor (Patel et al., Ann Surg, 235:226-231, 2002). Therefore, CTCs can reflect the current clinical status of patient disease and therapeutic response. The enumeration and characterization of CTCs has potential value in assessing cancer prognosis and in monitoring therapeutic efficacy for early detection of treatment failure that can lead to disease relapse.
  • CTC analysis may detect early relapse in presymptomatic patients who have completed a course of therapy; at present, individuals without measurable disease are not eligible to participate in clinical trials of promising new treatments (Braun et al., N Engl J Med, 351:824-826, 2004).
  • the devices and methods of the invention may be used to evaluate cancer patients and those at risk for cancer.
  • a blood sample is drawn from the patient and introduced to a deterministic device of the invention with a critical size chosen appropriately to separate epithelial cells from other blood cells.
  • the number of epithelial cells in the blood sample is determined using a method described herein.
  • the cells may be labeled with an antibody that binds to EpCAM, and the antibody may have a covalently bound fluorescent label, or be bound to a magnetic particle.
  • a bulk measurement may then be made of the enriched sample produced by the device, and from this measurement, the number of epithelial cells present in the initial blood sample may be determined. Microscopic techniques may be used to visually quantify the cells in order to correlate the bulk measurement with the corresponding number of labeled cells in the blood sample.
  • this provides a useful indication of the progression of the disease and assists medical practitioners in making appropriate therapeutic choices based on the increase, decrease, or lack of change in circulating epithelial cells in the patient's bloodstream.
  • a sudden increase in the number of cells detected may provide an early warning that the patient has developed a tumor. This early diagnosis, coupled with subsequent therapeutic intervention, is likely to result in an improved patient outcome in comparison to an absence of diagnostic information.
  • Diagnostic methods include making bulk measurements of labeled epithelial cells isolated from blood, as well as techniques that destroy the integrity of the cells. For example, PCR may be performed on a sample in which the number of target cells isolated is very low; by using primers specific for particular cancer markers, information may be gained about the type of tumor from which the analyzed cells originated. Additionally, RNA analysis, proteome analysis, or metabolome analysis may be performed as a means of diagnosing the type or types of cancer present in the patient.
  • EGFR epidermal growth factor receptor
  • TK tyrosine kinase
  • EGFR inhibitors such as gefitinib (Iressa; AstraZeneca)
  • these cells may be analyzed by PCR to determine what mutations may be present in the EFGR gene expressed in the epithelial cells.
  • Certain mutations such as those clustered around the ATP-binding pocket of the EGFR TK domain, are known to make the cancer cells susceptible to gefitinib inhibition.
  • the presence of these mutations supports a diagnosis of cancer that is likely to respond to treatment using gefitinib.
  • many patients who respond to gefitinib eventually develop a second mutation, often a methionine-to-threonine substitution at position 790 in exon 20 of the TK domain, which renders them resistant to gefitinib.
  • one may test for this mutation as well, providing further diagnostic information about the course of the disease and the likelihood that it will respond to gefitinib or similar compounds.
  • Samples may be employed in the methods described herein with or without manipulation, e.g., stabilization and removal of certain components.
  • the sample is enriched in the cells of interest prior to introduction to a device of the invention.
  • Methods for enriching cell populations are described herein and known in the art, e.g., affinity mechanisms, agglutination, and size, shape, and deformability based enrichments.
  • Some samples may be diluted or concentrated prior to introduction into the device.
  • reagents are added to the sample, to selectively or nonselectively increase the hydrodynamic size of the particles within the sample.
  • This modified sample is, for example, then pumped through a deterministic device. Because the particles are swollen and have an increased hydrodynamic diameter, it will be possible to use deterministic devices with larger and more easily manufactured gap sizes.
  • the steps of swelling and size-based enrichment are performed in an integrated fashion on a deterministic device.
  • Suitable reagents include any hypotonic solution, e.g., deionized water, 2% sugar solution, or neat non-aqueous solvents.
  • Other reagents include beads, e.g., magnetic or polymer, that bind selectively (e.g., through antibodies or avidin-biotin) or non-selectively.
  • reagents are added to the sample to selectively or nonselectively decrease the hydrodynamic size of the particles within the sample.
  • Nonuniform decrease in particles in a sample will increase the difference in hydrodynamic size between particles.
  • nucleated cells are separated from enucleated cells by hypertonically shrinking the cells. The enucleated cells can shrink to a very small particle, while the nucleated cells cannot shrink below the size of the nucleus.
  • Exemplary shrinking reagents include hypertonic solutions.
  • affinity functionalized beads are used to increase the hydrodynamic size of an analyte of interest relative to other analytes present in a sample, thereby allowing for the operation of a deterministic device with a larger and more easily manufactured gap size.
  • Fluids may be driven through a device either actively or passively. Fluids may be pumped using electric field, a centrifugal field, pressure-driven fluid flow, an electro-osmotic flow, or capillary action. In preferred embodiments, the average direction of the field will be parallel to the walls of the channel that includes the deterministic device.
  • FIGS. 42A-42E show an exemplary device, characterized as follows.
  • the arrays and channels were fabricated in silicon using standard photolithography and deep silicon reactive etching techniques. The etch depth is 150 ⁇ m. Through holes for fluid access are made using KOH wet etching. The silicon substrate was sealed on the etched face to form enclosed fluidic channels using a blood compatible pressure sensitive adhesive (9795, 3M, St Paul, Minn.).
  • the device was mechanically mated to a plastic manifold with external fluidic reservoirs to deliver blood and buffer to the device and extract the generated fractions.
  • An external pressure source was used to apply a pressure of 2.4 PSI to the buffer and blood reservoirs to modulate fluidic delivery and extraction from the packaged device.
  • Measurement techniques Complete blood counts were determined using a Coulter impedance hematology analyzer (COULTER® Ac•T diffTM, Beckman Coulter, Fullerton, Calif.).
  • FIGS. 43A-43F shows typical histograms generated by the hematology analyzer from a blood sample and the waste (buffer, plasma, red blood cells, and platelets) and product (buffer and nucleated cells) fractions generated by the device.
  • Table 1 shows the performance over 5 different blood samples: TABLE 1 Performance Metrics Sample RBC Platelet WBC number Throughput removal removal loss 1 4 mL/hr 100% 99% ⁇ 1% 2 6 mL/hr 100% 99% ⁇ 1% 3 6 mL/hr 100% 99% ⁇ 1% 4 6 mL/hr 100% 97% ⁇ 1% 5 6 mL/hr 100% 98% ⁇ 1%
  • FIGS. 44A-44D show an exemplary device, characterized as follows.
  • the arrays and channels were fabricated in silicon using standard photolithography and deep silicon reactive etching techniques. The etch depth is 150 ⁇ m. Through holes for fluid access are made using KOH wet etching. The silicon substrate was sealed on the etched face to form enclosed fluidic channels using a blood compatible pressure sensitive adhesive (9795, 3M, St Paul, Minn.).
  • the device was mechanically mated to a plastic manifold with external fluidic reservoirs to deliver blood and buffer to the device and extract the generated fractions.
  • An external pressure source was used to apply a pressure of 2.4 PSI to the buffer and blood reservoirs to modulate fluidic delivery and extraction from the packaged device.
  • Measurement techniques Complete blood counts were determined using a Coulter impedance hematology analyzer (COULTER® Ac•T diffTM, Beckman Coulter, Fullerton, Calif.).
  • FIG. 45 shows a schematic of the device used to separate nucleated cells from fetal cord blood.
  • Bifurcation ratio 1/10.
  • Device design multiplexing 10 array duplexes; flow resistors for flow stability.
  • the arrays and channels were fabricated in silicon using standard photolithography and deep silicon reactive etching techniques. The etch depth is 140 ⁇ m. Through holes for fluid access are made using KOH wet etching. The silicon substrate was sealed on the etched face to form enclosed fluidic channels using a blood compatible pressure sensitive adhesive (9795, 3M, St Paul, Minn.).
  • the device was mechanically mated to a plastic manifold with external fluidic reservoirs to deliver blood and buffer to the device and extract the generated fractions.
  • An external pressure source was used to apply a pressure of 2.0 PSI to the buffer and blood reservoirs to modulate fluidic delivery and extraction from the packaged device.
  • Nucleated cells from the blood were separated from enucleated cells (red blood cells and platelets), and plasma delivered into a buffer stream of calcium and magnesium-free Dulbecco's Phosphate Buffered Saline (14190-144, Invitrogen, Carlsbad, Calif.) containing 1% Bovine Serum Albumin (BSA) (A8412-100ML, Sigma-Aldrich, St Louis, Mo.) and 2 mM EDTA (15575-020, Invitrogen, Carlsbad, Calif.).
  • BSA Bovine Serum Albumin
  • FIG. 46A-46B Cell smears of the product and waste fractions ( FIG. 46A-46B ) were prepared and stained with modified Wright-Giemsa (WG16, Sigma Aldrich, St. Louis, Mo.).
  • Example 1 The device and process described in detail in Example 1 were used in combination with immunomagnetic affinity enrichment techniques to demonstrate the feasibility of isolating fetal cells from maternal blood.
  • the nucleated cell fraction was labeled with anti-CD71 microbeads (130-046-201, Miltenyi Biotech Inc., Auburn, Calif.) and enriched using the MiniMACSTM MS column (130-042-201, Miltenyi Biotech Inc., Auburn, Calif.) according to the manufacturer's specifications. Finally, the CD71-positive fraction was spotted onto glass slides.
  • Measurement techniques Spotted slides were stained using fluorescence in situ hybridization (FISH) techniques according to the manufacturer's specifications using Vysis probes (Abbott Laboratories, Downer's Grove, Ill.). Samples were stained from the presence of X and Y chromosomes. In one case, a sample prepared from a known trisomy 21 pregnancy was also stained for chromosome 21.
  • FISH fluorescence in situ hybridization
  • each device provides the number of stages in series, the gap size for each stage, ⁇ (Flow Angle), and the number of channels per device (Arrays/Chip).
  • Flow Angle
  • Arrays/Chip the number of channels per device
  • This device includes five stages in a single array.
  • This device includes the stages, where each stage is a duplex having a bypass channel.
  • the height of the device was 125 ⁇ m.
  • FIG. 49A shows the mask employed to fabricate the device.
  • FIGS. B 1 B-B 1 D are enlargements of the portions of the mask that define the inlet, array, and outlet.
  • FIGS. 50A-50G show SEMs of the actual device.
  • This device includes the stages, where each stage is a duplex having a bypass channel. “Fins” were designed to flank the bypass channel to keep fluid from the bypass channel from re-entering the array.
  • the chip also included on-chip flow resistors, i.e., the inlets and outlets possessed greater fluidic resistance than the array. The height of the device was 117 ⁇ m.
  • FIG. 51 A shows the mask employed to fabricate the device.
  • FIGS. 51B-51D are enlargements of the portions of the mask that define the inlet, array, and outlet.
  • FIGS. 52A-52F show SEMs of the actual device.
  • This device includes the stages, where each stage is a duplex having a bypass channel. “Fins” were designed to flank the bypass channel to keep fluid from the bypass channel from re-entering the array. The edge of the fin closest to the array was designed to mimic the shape of the array.
  • the chip also included on-chip flow resistors, i.e., the inlets and outlets possessed greater fluidic resistance than the array. The height of the device was 138 ⁇ m.
  • Array Design 3 stage symmetric array Gap Sizes: Stage 1: 8 ⁇ m Stage 2: 12 ⁇ m Stage 3: 18 ⁇ m Stage 4: Stage 5: Flow Angle: 1/10 Arrays/Chip: 10 Other: alternate large fin central collection channel on-chip flow resistors
  • FIG. 45A shows the mask employed to fabricate the device.
  • FIGS. 45B-45D are enlargements of the portions of the mask that define the inlet, array, and outlet.
  • FIGS. 532A-532F show SEMs of the actual device.
  • This device includes the stages, where each stage is a duplex having a bypass channel. “Fins” were optimized using Femlab to flank the bypass channel to keep fluid from the bypass channel from re-entering the array. The edge of the fin closest to the array was designed to mimic the shape of the array.
  • the chip also included on-chip flow resistors, i.e., the inlets and outlets possessed greater fluidic resistance than the array. The height of the device was 139 or 142 ⁇ m.
  • FIG. 54A shows the mask employed to fabricate the device.
  • FIGS. 54 B-E 1 D are enlargements of the portions of the mask that define the inlet, array, and outlet.
  • FIGS. 55A-55S show SEMs of the actual device.
  • This device includes a single stage, duplex device having a bypass channel disposed to receive output from the ends of both arrays.
  • the obstacles in this device are elliptical.
  • the array boundary was modeled in Femlab to.
  • the chip also included on-chip flow resistors, i.e., the inlets and outlets possessed greater fluidic resistance than the array.
  • the height of the device was 152 ⁇ m.
  • FIG. 44A shows the mask employed to fabricate the device.
  • FIGS. 44B-44D are enlargements of the portions of the mask that define the inlet, array, and outlet.
  • FIGS. 56A-56C show SEMs of the actual device.
  • Deterministic devices incorporated into devices of the invention were designed by computer-aided design (CAD) and microfabricated by photolithography.
  • CAD computer-aided design
  • a two-step process was developed in which a blood sample is first debulked to remove the large population of small cells and then the rare target epithelial cells target cells are recovered by immunoaffinity capture.
  • the devices were defined by photolithography and etched into a silicon substrate based on a CAD-generated design.
  • the cell enrichment module which is approximately the size of a standard microscope slide, contains 14 parallel sample processing sections and associated sample handling channels that connect to common sample and buffer inlets and product and waste outlets. Each section contains an array of microfabricated obstacles that is optimized to separate the target cell type by size via displacement of the larger cells into the product stream.
  • the microchip was designed to separate red blood cells (RBCs) and platelets from the larger leukocytes and circulating tumor cells. Enriched populations of target cells were recovered from whole blood passed through the device. Performance of the cell enrichment microchip was evaluated by separating RBCs and platelets from white blood cells (WBCs) in normal whole blood ( FIG. 67 ). In cancer patients, circulating tumor cells are found in the larger WBC fraction. Blood was minimally diluted (30%), and a 6 ml sample was processed at a flow rate of up to 6 ml/hr. The product and waste stream were evaluated in a Coulter Model “A C -T diff” clinical blood analyzer, which automatically distinguishes, sizes and counts different blood cell populations.
  • RBCs red blood cells
  • WBCs white blood cells
  • the enrichment chip achieved separation of RBCs from WBCs, in which the WBC fraction had >99% retention of nucleated cells, >99% depletion of RBCs and >97% depletion of platelets. Representative histograms of these cell fractions are shown in FIG. 68 . Routine cytology confirmed the high degree of enrichment of the WBC RBC fractions ( FIG. 69 ).
  • epithelial cells were recovered by affinity capture in a microfluidic module that is functionalized with immobilized antibody.
  • a capture module with a single chamber containing a regular array of antibody-coated microfabricated obstacles was designed. These obstacles are disposed to maximize cell capture by increasing the capture area approximately four-fold, and by slowing the flow of cells under laminar flow adjacent to the obstacles to increase the contact time between the cells and the immobilized antibody.
  • the capture modules can be operated under conditions of relatively high flow rate but low shear to protect cells against damage.
  • the surface of the capture module was functionalized by sequential treatment with 10% silane, 0.5% gluteraldehyde and avidin, followed by biotinylated anti-EpCAM.
  • CMRA reagent Cell Tracker Orange
  • Molecular Probes Eugene, Oreg.
  • cell suspensions were processed directly in the capture modules without prior fractionation in the cell enrichment module to debulk the red blood cells; hence, the sample stream contained normal blood red cells and leukocytes as well as tumor cells.
  • the device was washed with buffer at a higher flow rate (3 ml/hr) to remove the nonspecifically bound cells.
  • NCI-H1650 cells that were spiked into whole blood and recovered by size fractionation and affinity capture as described above were successfully analyzed in situ.
  • a trial run to distinguish epithelial cells from leukocytes 0.5 ml of a stock solution of fluorescein-labeled CD45 pan-leukocyte monoclonal antibody was passed into the capture module and incubated at room temperature for 30 minutes. The module was washed with buffer to remove unbound antibody and the cells were fixed on the chip with 1% paraformaldehyde and observed by fluorescence microscopy. As shown in FIG. 70 , the epithelial cells were bound to the obstacles and floor of the capture module. Background staining of the flow passages with CD45 pan-leukocyte antibody is visible, as are several stained leukocytes, apparently due to a low level of non-specific capture.
  • FIG. 73A A design for preferred deterministic device is shown in FIG. 73A , and parameters corresponding to three preferred device embodiments associated with this design are shown in FIG. 73B . These embodiments are particularly useful for separating epithelial cells from blood.
  • a blood sample from a cancer patient is processed and analyzed using the devices and methods of Example 11, resulting in an enriched sample of epithelial cells containing CTCs. This sample is then analyzed to identify potential EGFR mutations.
  • genomic DNA is isolated from the target cells present in the enriched sample and amplified for use in allele-specific Real Time PCR assays. Since all EGFR mutations in NSC lung cancer reported to date that are known to confer sensitivity or resistance to gefitinib lie within the coding regions of exons 18 to 21, each of these four exons is PCR-amplified with a unique set of exon-specific primers. Next, multiplexed allele-specific quantitative PCR reactions are performed using the TaqMan 5′ nuclease assay PCR system (Applied Biosystems) and a model 7300 Applied Biosystems Real Time PCR machine. This allows the rapid identification of any of the known clinically relevant mutations.
  • a two-step PCR protocol is required for this method.
  • exons 18 through 21 are amplified in standard PCR reactions.
  • the resultant PCR products are split into separate aliquots for use in allele-specific multiplexed Real Time PCR assays.
  • the initial PCR reactions are stopped during the log phase in order to minimize possible loss of allele-specific information during amplification.
  • a second round of PCR amplifies subregions of the initial PCR product specific to each mutation of interest. Given the very high sensitivity of Real Time PCR, it is possible to obtain complete information on the mutation status of the EGFR gene even if as few as 10 CTCs are isolated.
  • Real Time PCR provides quantification of allelic sequences over 8 logs of input DNA concentrations; thus, even heterozygous mutations in impure populations are easily detected using this method.
  • Oligonucleotides are designed using the primer optimization software program Primer Express (Applied Biosystems), and hybridization conditions are optimized to distinguish the wild type EGFR DNA sequence from mutant alleles.
  • EGFR genomic DNA amplified from lung cancer cell lines that are known to carry EGFR mutations, such as H358 (wild type), H1650 (15-bp deletion, ⁇ 2235-2249), and H1975 (two point mutations, 2369 C ⁇ T, 2573 T ⁇ G), is used to optimize the allele-specific Real Time PCR reactions.
  • allele-specific labeled probes specific for wild type sequence or for known EGFR mutations are developed.
  • the oligonucleotides are designed to have melting temperatures that easily distinguish a match from a mismatch, and the Real Time PCR conditions are optimized to distinguish wild type and mutant alleles. All Real Time PCR reactions are carried out in triplicate.
  • labeled probes containing wild type sequence are multiplexed in the same reaction with a single mutant probe. Expressing the results as a ratio of one mutant allele sequence versus wild type sequence can identify samples containing or lacking a given mutation. After conditions are optimized for a given probe set, it is then possible to multiplex probes for all of the mutant alleles within a given exon within the same Real Time PCR assay, increasing the ease of use of this analytical tool in clinical settings.
  • the purity of the input sample of CTCs may vary, and the mutation status of the isolated CTCs may be heterogeneous. Nevertheless, the extremely high sensitivity of Real Time PCR enables the identification any and all mutant sequences present.
  • a diagnosis of the absence, presence, or progression of cancer may be based on the number of cells in a cellular sample that are larger than a particular cutoff size. For example, cells with a hydrodynamic cell diameter of 14 microns or larger may be selected. This cutoff size would eliminate most leukocytes. The nature of these cells may then be determined by downstream molecular or cytological analysis.
  • Cell types other than epithelial cells that would be useful to analyze include endothelial cells, endothelial progenitor cells, endometrial cells, or trophoblasts indicative of a disease state. Furthermore, determining separate counts for epithelial cells and other cell types, followed by a determination of the ratios between the number of epithelial cells and the number of other cell types, may provide useful diagnostic information.
  • a deterministic device may be configured to isolate targeted subpopulations of cells such as those described above, as shown in FIG. 71A -D.
  • a size cutoff may be selected such that most native blood cells, including red blood cells, white blood cells, and platelets, flow to waste, while non-native cells, which could include endothelial cells, endothelial progenitor cells, endometrial cells, or trophoblasts, are collected in an enriched sample. This enriched sample may be further analyzed.
  • a deterministic device may include counting means to determine the number of cells in the enriched sample, and further analysis of the cells in the enriched sample may provide additional information that is useful for diagnostic or other purposes.
  • the device includes a deterministic separation component, as described herein, capable of separated fetal nucleated red blood cells and maternal white blood cells from maternal enucleated red blood cells.
  • the deterministic component is connected to a reservoir containing sodium nitrite.
  • a maternal blood sample e.g., that has been diluted, is introduced into the device to produce a fraction enriched in fetal red blood cells and depleted of maternal red blood cells. This sample is directed into the reservoir where the sodium nitrite oxidizes the fetal heme iron, thereby increasing the magnetic responsiveness of the fetal red blood cells.
  • a magnetic field is then applied, e.g., via a MACS column, and the altered fetal red blood cells bind to the magnet, while maternal white blood cells are not bound by the magnet. Removing the white blood cells, e.g., by a rinse, and then eliminating the magnetic field allows recovery of the fetal red blood cells, e.g., for analysis, storage, or further manipulation.

Abstract

The invention features a device for the deterministic separation of analytes coupled to a reservoir containing a reagent that alters a magnetic propert of the analyte. Exemplary methods include the enrichment of a sample in a desired analyte (e.g., using deterministic separation) or the alteration of a desired analyte in the device. The devices and methods may be advantageously employed to enrich for rare cells, e.g., fetal cells or epithelial cells, present in a sample, e.g., maternal blood.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • The present application claims the benefit of U.S. Provisional Application Ser. Nos. 60/668,415, filed Apr. 5, 2005 and 60/704,067, filed Jul. 29, 2005, each of which is hereby incorporated by reference in its entirety.
  • BACKGROUND OF THE INVENTION
  • The invention relates to the fields of cell separation, medical diagnostics, and microfluidic devices.
  • Clinically or environmentally relevant information may often be present in a sample, but in quantities too low to detect. Thus, various enrichment or amplification methods are often employed in order to increase the detectability of such information.
  • For cells, different flow cytometry and cell sorting methods are available, but these techniques typically employ large and expensive pieces of equipment, which require large volumes of sample and skilled operators. These cytometers and sorters use methods like electrostatic deflection, centrifugation, fluorescence activated cell sorting (FACS), and magnetic activated cell sorting (MACS) to achieve cell separation. These methods often suffer from the inability to enrich a sample sufficiently to allow analysis of rare components of the sample. Furthermore, such techniques may result in unacceptable losses of such rare components, e.g., through inefficient separation or degradation of the components.
  • Thus, there is a need for new devices and methods for enriching samples.
  • SUMMARY OF THE INVENTION
  • In a first aspect, the invention provides a device for producing a sample enriched in an analyte that includes a first channel (e.g., a microfluidic channel) including a structure that deterministically deflects particles having a hydrodynamic size above a critical size in a direction not parallel to the average direction of flow in the structure, wherein said particles are analyte particles or are a non-analyte component of the sample; and a reservoir fluidly coupled to an output of the first channel through which the analyte passes into the reservoir, the reservoir including a reagent that alters a magnetic property of the analyte. In a first embodiment, the structure includes an array of obstacles that form a network of gaps, where a fluid passing through the gaps is divided unequally into a major flux and a minor flux so that the average direction of the major flux is not parallel to the average direction of fluidic flow in the channel. In the first embodiment, the array of obstacles may include first and second rows, where the second row is displaced laterally relative to the first row so that fluid passing through a gap in the first row is divided unequally into two gaps in the second row. The analyte may have a hydrodynamic size greater than or smaller than the critical size. The device may include a magnetic force generator capable of generating a magnetic field, and may further include a region of magnetic obstacles (e.g., obstacles including a permanent magnet or obstacles including a non-permanent magnet) disposed in a second channel (e.g., a microfluidic channel). The magnetic obstacles may be ordered in a two-dimensional array. The reservoir of the device may further include a second channel including a magnet. The reagent (e.g., sodium nitrite) may alter an intrinsic magnetic property of one or more analytes. In one embodiment, the reagent, e.g., holo-transferrin or a magnetic particle, may bind to the one or more analytes. A magnetic particle may further include an antibody (e.g., anti-CD71, anti-CD36, anti-CD45, anti-GPA, anti-antigen i, anti-CD34, anti-fetal hemoglobin, anti-EpCAM, anti-E-cadherin, or anti-Muc-1) or an antigen-binding fragment thereof.
  • In a second aspect, the invention provides a method for producing a sample enriched in a first analyte relative to a second analyte that includes applying at least a portion of the sample to a device including a structure that deterministically deflects particles having a hydrodynamic size above a critical size in a direction not parallel to the average direction of flow in the structure, thereby producing a second sample enriched in the first analyte and including the second analyte; combining the second sample with a reagent that alters a magnetic property of the first analyte to produce an altered first analyte; and applying a magnetic field to the second sample, where the magnetic field generates a differential force to physically separate the altered first analyte from the second analyte, thereby producing a sample enriched in the first analyte. The reagent may bind to the first analyte. In another embodiment, the reagent (e.g., sodium nitrite) may alter an intrinsic magnetic property of the first analyte. In yet another embodiment, the reagent may include a magnetic particle that binds to or is incorporated into the first analyte. The magnetic particle may include an antibody (e.g., anti-CD71, anti-GPA, anti-antigen i, anti-CD45, anti-CD34, anti-fetal hemoglobin, anti-EpCAM, anti-E-cadherin, or anti-Muc-1) or an antigen-binding fragment thereof. The analyte may have a hydrodynamic size greater than or less than said critical size. The sample may be a maternal blood sample. The first analyte may be a cell (e.g., bacterial cell, a fetal cell, or a blood cell such as a fetal red blood cell), an organelle (e.g., a nucleus), or a virus.
  • In a third aspect, the invention provides a method of producing a sample enriched in red blood cells relative to a second blood component (e.g., maternal blood cells) that includes contacting the sample including red blood cells (e.g., fetal red blood cells) with a reagent that oxidizes iron to produce oxidized hemoglobin; and applying a magnetic field to the sample, where the red blood cells having oxidized hemoglobin are attracted to the magnetic field to a greater extent than the second blood component, thereby producing the sample enriched in the red blood cells. The method may further include performing prior to the contacting step, a step that enriches the sample with red blood cells (e.g., enriching fetal blood cells are enriched relative to maternal red blood cells), for example, by applying at least a portion of the sample to a device including a structure that deterministically deflects particles having a hydrodynamic size above a critical size in a direction not parallel to the average direction of flow in the structure.
  • In a fourth aspect, the invention provides a device for producing a sample enriched in red blood cells that includes an analytical device that enriches the red blood cells based on size, shape, deformability, or affinity; and a reservoir including a reagent that oxidizes iron, where the reagent (e.g., sodium nitrite) increases the magnetic responsiveness of the red blood cells. The analytical device may include a first channel that includes a structure that deterministically deflects particles having a hydrodynamic size above a critical size in a direction not parallel to the average direction of flow in the structure.
  • In a fifth aspect, the invention provides a reagent that includes a plurality of magnetic particles coupled to one or more binding moieties (e.g., an antibody such as a monoclonal antibody) that selectively binds GPA, fetal hemoglobin, CD34, CD45, CD71, EGFR, or EpCAm.
  • In a sixth aspect, the invention provides a method for separating one or more cells of interest from a mixture of cells that includes combining the mixture of cells with a reagent of the fifth aspect and incubating the mixture of cells and the reagent for a time sufficient to allow the binding moieties to selectively bind the one or more cells of interest in the mixture, and apply a magnetic field to the mixture thereby separating cells that bound the magnetic particles from cells that did not bind the magnetic particles. The method may further include a step of enriching the mixture of cells for the one or more cells of interest. The enriching step may include performing size-based separation with an array of obstacles or selectively lysing one or more cells that is not a cell of interest.
  • By “analyte” is meant a molecule, other chemical species, e.g., an ion, or particle. Exemplary analytes include cells, viruses, nucleic acids, proteins, carbohydrates, and small organic molecules.
  • By “biological particle” is meant any species of biological origin that is insoluble in aqueous media on the time scale of sample acquisition, preparation, storage, and analysis. Examples include cells, particulate cell components, viruses, and complexes including proteins, lipids, nucleic acids, and carbohydrates.
  • By “biological sample” is meant any sample of biological origin or containing, or potentially containing, biological particles. Preferred biological samples are cellular samples.
  • By “blood component” is meant any component of whole blood, including host red blood cells, white blood cells, and platelets. Blood components also include the components of plasma, e.g., proteins, lipids, nucleic acids, and carbohydrates, and any other cells that may be present in blood, e.g., because of current or past pregnancy, organ transplant, or infection.
  • By “cellular sample” is meant a sample containing cells or components thereof. Such samples include naturally occurring fluids (e.g., blood, lymph, cerebrospinal fluid, urine, cervical lavage, and water samples), portions of such fluids, and fluids into which cells have been introduced (e.g., culture media, and liquefied tissue samples). The term also includes a lysate.
  • By “capture moiety” is meant a chemical species to which an analyte binds. A capture moiety may be a compound coupled to a surface or the material making up the surface. Exemplary capture moieties include antibodies, oligo- or polypeptides, nucleic acids, other proteins, synthetic polymers, and carbohydrates.
  • By “channel” is meant a gap through which fluid may flow. A channel may be a capillary, a conduit, or a strip of hydrophilic pattern on an otherwise hydrophobic surface wherein aqueous fluids are confined.
  • By “component” of cell is meant any component of a cell that may be at least partially isolated upon lysis of the cell. Cellular components may be organelles (e.g., nuclei, peri-nuclear compartments, nuclear membranes, mitochondria, chloroplasts, or cell membranes), polymers or molecular complexes (e.g., lipids, polysaccharides, proteins. (membrane, trans-membrane, or cytosolic), nucleic acids (native, therapeutic, or pathogenic), viral particles, or ribosomes), or other molecules (e.g., hormones, ions, cofactors, or drugs). By “component” of a cellular sample is meant a subset of cells contained within the sample.
  • By “enriched sample” is meant a sample containing an analyte that has been processed to increase the relative amount of the analyte relative to other analytes typically present in a sample. For example, samples may be enriched by increasing the amount of the analyte of interest by at least 10%, 25%, 50%, 75%, 100% or by a factor of at least 1000, 10,000, 100,000, or 1,000,000.
  • By “depleted sample” is meant a sample containing an analyte that has been processed to decrease the amount of the analyte relative to other analytes typically present in a sample. For example, samples may be depleted by decreasing the amount of the analyte of interest by at least 5%, 10%, 25%, 50%, 75%, 90%, 95%, 97%, 98%, 99%, or even 100%.
  • By “exchange buffer” in the context of a sample (e.g., a cellular sample) is meant a medium distinct from the medium in which the sample is originally suspended, and into which one or more components of the sample are to be exchanged.
  • By “flow-extracting boundary” is meant a boundary designed to remove fluid from an array.
  • By “flow-feeding boundary” is meant a boundary designed to add fluid to an array.
  • By “gap” is meant an opening through which fluids and/or particles may flow. For example, a gap may be a capillary, a space between two obstacles wherein fluids may flow, or a hydrophilic pattern on an otherwise hydrophobic surface wherein aqueous fluids are confined. In a preferred embodiment of the invention, the network of gaps is defined by an array of obstacles. In this embodiment, the gaps are the spaces between adjacent obstacles. In a preferred embodiment, the network of gaps is constructed with an array of obstacles on the surface of a substrate.
  • By “hydrodynamic size” is meant the effective size of a particle when interacting with a flow, obstacles (e.g., posts), or other particles. The obstacles or other particles may be in a microfluidic structure. It is used as a general term for particle volume, shape, and deformability in the flow.
  • By “intracellular activation” is meant activation of second messenger pathways, leading to transcription factor activation, or activation of kinases or other metabolic pathways. Intracellular activation through modulation of external cell membrane antigens can also lead to changes in receptor trafficking.
  • By “labeling reagent” is meant a reagent that is capable of binding to an analyte, being internalized or otherwise absorbed, and being detected, e.g., through shape, morphology, color, fluorescence, luminescence, phosphorescence, absorbance, magnetic properties, or radioactive emission.
  • By “metabolome” is meant the set of compounds within a cell, other than proteins and nucleic acids, that participate in metabolic reactions and that are required for the maintenance, growth or normal function of a cell.
  • By “microfluidic” is meant having at least one dimension of less than 1 mm.
  • By “obstacle” is meant an impediment to flow in a channel, e.g., a protrusion from one surface. For example, an obstacle may refer to a post outstanding on a base substrate or a hydrophobic barrier for aqueous fluids. In some embodiments, the obstacle may be partially permeable. For example, an obstacle may be a post made of porous material, wherein the pores allow penetration of an aqueous component but are too small for the particles being separated to enter.
  • By “shrinking reagent” is meant a reagent that decreases the hydrodynamic size of a particle. Shrinking reagents may act by decreasing the volume, increasing the deformability, or changing the shape of a particle.
  • By “swelling reagent” is meant a reagent that increases the hydrodynamic size of a particle. Swelling reagents may act by increasing the volume, reducing the deformability, or changing the shape of a particle.
  • By “substantially larger” is meant at least 2-fold, 3-fold, 5-fold, 10-fold, 25-fold, 50-fold, or even 100-fold larger.
  • By “substantially smaller” is meant at least 2-fold, 3-fold, 5-fold, 10-fold, 25-fold, 50-fold, or even 100-fold smaller.
  • Other features and advantages will be apparent from the following description and the claims.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIGS. 1A-1E are schematic depictions of an array that separated cells based on deterministic lateral displacement: (A) illustrates the lateral displacement of subsequent rows; (B) illustrates how fluid flowing through a gap is divide unequally around obstacles in subsequent rows; (C) illustrates how an analyte with a hydrodynamic size above the critical size is displaced laterally in the device; (D) illustrates an array of cylindrical obstacles; and (E) illustrates an array of elliptical obstacles.
  • FIG. 2 is a schematic description illustrating the unequal division of the flux through a gap around obstacles in subsequent rows.
  • FIG. 3 is a schematic depiction of how the critical size depends on the flow profile, which is parabolic in this example.
  • FIG. 4 is an illustration of how shape affects the movement of analytes through a device.
  • FIG. 5 is an illustration of how deformability affects the movement of analytes through a device.
  • FIG. 6 is a schematic depiction of deterministic lateral displacement. Analytes having a hydrodynamic size above the critical size move to the edge of the array, while analytes having a hydrodynamic size below the critical size pass through the device without lateral displacement.
  • FIG. 7 is a schematic depiction of a three stage deterministic device.
  • FIG. 8 is a schematic depiction of the maximum size and cut-off size for the device of FIG. 7.
  • FIG. 9 is a schematic depiction of a bypass channel.
  • FIG. 10 is a schematic depiction of a bypass channel.
  • FIG. 11 is a schematic depiction of a three stage deterministic device having a common bypass channel.
  • FIG. 12 is a schematic depiction of a three stage, duplex deterministic device having a common bypass channel.
  • FIG. 13 is a schematic depiction of a three stage deterministic device having a common bypass channel, where the flow through the device is substantially constant.
  • FIG. 14 is a schematic depiction of a three stage, duplex deterministic device having a common bypass channel, where the flow through the device is substantially constant.
  • FIG. 15 is a schematic depiction of a three stage deterministic device having a common bypass channel, where the fluidic resistance in the bypass channel and the adjacent stage are substantially constant.
  • FIG. 16 is a schematic depiction of a three stage, duplex deterministic device having a common bypass channel, where the fluidic resistance in the bypass channel and the adjacent stage are substantially constant.
  • FIG. 17 is a schematic depiction of a three stage deterministic device having two, separate bypass channels.
  • FIG. 18 is a schematic depiction of a three stage deterministic device having two, separate bypass channels, which are in arbitrary configuration.
  • FIG. 19 is a schematic depiction of a three stage, duplex deterministic device having three, separate bypass channels.
  • FIG. 20 is a schematic depiction of a three stage deterministic device having two, separate bypass channels, wherein the flow through each stage is substantially constant.
  • FIG. 21 is a schematic depiction of a three stage, duplex deterministic device having three, separate bypass channels, wherein the flow through each stage is substantially constant.
  • FIG. 22 is a schematic depiction of a flow-extracting boundary.
  • FIG. 23 is a schematic depiction of a flow-feeding boundary.
  • FIG. 24 is a schematic depiction of a flow-feeding boundary, including a bypass channel.
  • FIG. 25 is a schematic depiction of two flow-feeding boundaries flanking a central bypass channel.
  • FIG. 26 is a schematic depiction of a device having four channels that act as on-chip flow resistors.
  • FIGS. 27 and 28 are schematic depictions of the effect of on-chip resistors on the relative width of two fluids flowing in a device.
  • FIG. 29 is a schematic depiction of a duplex device having a common inlet for the two outer regions.
  • FIG. 30A is a schematic depiction of a multiple arrays on a device. FIG. 30B is a schematic depiction of multiple arrays with common inlets and product outlets on a device.
  • FIG. 31 is a schematic depiction of a multi-stage device with a small footprint.
  • FIG. 32 is a schematic depiction of blood passing through a device.
  • FIG. 33 is a graph illustrating the hydrodynamic size distribution of blood cells.
  • FIGS. 34A-34D are schematic depictions of moving an analyte from a sample to a buffer in a single stage (A), three stage (B), duplex (C), or three stage duplex (D) deterministic device.
  • FIG. 35A is a schematic depiction of a two stage deterministic device employed to move a particle from blood to a buffer to produce three products. FIG. 35B is a schematic graph of the maximum size and cut off size of the two stages. FIG. 35C is a schematic graph of the composition of the three products.
  • FIG. 36 is a schematic depiction of a two stage deterministic device for alteration, where each stage has a bypass channel.
  • FIG. 37 is a schematic depiction of the use of fluidic channels to connect two stages in a device.
  • FIG. 38 is a schematic depiction of the use of fluidic channels to connect two stages in a device, wherein the two stages are configured as a small footprint array.
  • FIG. 39A is a schematic depiction of a two stage deterministic device having a bypass channel that accepts output from both stages. FIG. 39B is a schematic graph of the size range of product achievable with this device.
  • FIG. 40 is a schematic depiction of a two stage deterministic device for alteration having bypass channels that flank each stage and empty into the same outlet.
  • FIG. 41 is a schematic depiction of a deterministic device for the sequential movement and alteration of particles.
  • FIG. 42A is a photograph of a deterministic device that may be incorporated into a device of the invention. FIGS. 42B-42E are depictions the mask used to fabricate a device that may be incorporated into the invention. FIG. 42F is a series of photographs of the device containing blood and buffer.
  • FIGS. 43A-43F are typical histograms generated by the hematology analyzer from a blood sample and the waste (buffer, plasma, red blood cells, and platelets) and product (buffer and nucleated cells) fractions generated by the device of FIG. 42.
  • FIGS. 44A-44D are depictions the mask used to fabricate a deterministic device that may be incorporated into a device of the invention.
  • FIGS. 45A-45D are depictions the mask used to fabricate a deterministic device that may be incorporated a device of into the invention.
  • FIG. 46A is a micrograph of a sample enriched in fetal red blood cells. FIG. 46B is a micrograph of maternal red blood cell waste.
  • FIG. 47 is a series of micrographs showing the positive identification of male fetal cells (Blue=nucleus, Red=X chromosome, Green=Y chromosome).
  • FIG. 48 is a series of micrographs showing the positive identification of sex and trisomy 21.
  • FIGS. 49A-49D are depictions the mask used to fabricate a deterministic device that may be incorporated into a device of the invention.
  • FIGS. 50A-50G are electron micrographs of the device of FIG. 49.
  • FIGS. 51A-51D are depictions the mask used to fabricate a deterministic device that may be incorporated into a device of the invention.
  • FIGS. 52A-52F are electron micrographs of the device of FIG. 51.
  • FIGS. 53A-53F are electron micrographs of the device of FIG. 45.
  • FIGS. 54A-54D are depictions the mask used to fabricate a deterministic device that may be incorporated a device of into the invention.
  • FIGS. 55A-55S are electron micrographs of the device of FIG. 54.
  • FIGS. 56A-56C are electron micrographs of the device of FIG. 44.
  • FIG. 57A is a schematic illustration of a deterministic device that may be incorporated into a device of the invention and its operation. FIG. 57B is an illustration of the device of FIG. 57A and a further-schematized representation of this device.
  • FIGS. 58A and 58B are illustrations of two distinct configurations for joining two deterministic devices together. In FIG. 58A, a cascade configuration is shown, in which outlet 1 of one device is joined to a sample inlet of a second device. In FIG. 58B, a bandpass configuration is shown, in which outlet 2 of one device is joined to a sample inlet of a second device.
  • FIG. 59 is an illustration of an enhanced method of size separation in which target cells are labeled with immunoaffinity beads.
  • FIG. 60 is an illustration of a method for performing size fractionation and for separating free labeling reagents, e.g., antibodies, from bound labeling reagents by using a device that may be incorporated into the invention.
  • FIG. 61 is an illustration of a method shown in FIG. 60. In this case, non-target cells may copurify with target cells, but these non-target cells do not interfere with quantification of target cells.
  • FIG. 62 is an illustration of a method for separating large cells from a mixture and producing a concentrated sample of these cells.
  • FIG. 63 is an illustration of a method for lysing cells inside a device of the invention and separating whole cells from organelles and other cellular components.
  • FIG. 64 is an illustration of two devices arrayed in a cascade configuration and used for performing size fractionation and for separating free labeling reagent from bound labeling reagents by using a device of the invention.
  • FIG. 65 is an illustration of two devices arrayed in a cascade configuration and used for performing size fractionation and for separating free labeling reagent from bound labeling reagents by using a device of the invention. In this figure, phage is utilized for binding and detection rather than antibodies.
  • FIG. 66 is an illustration of two devices arrayed in a bandpass configuration.
  • FIG. 67 is a graph of cell count versus hydrodynamic cell diameter for a microfluidic separation of normal whole blood.
  • FIG. 68 is a set of histograms from input, product, and waste samples generated with a Coulter “AC-T diff” clinical blood analyzer. The x-axis depicts cell volume in femtomoles.
  • FIG. 69 is a pair of representative micrographs from product and waste streams of fetal blood processed with a cell enrichment module, showing clear separation of nucleated cells and red blood cells.
  • FIG. 70 is a pair of images showing cells fixed on a cell enrichment module with paraformaldehyde and observed by fluorescence microscopy. Target cells are bound to the obstacles and floor of the capture module.
  • FIG. 71A is a graph of cell count versus hydrodynamic cell diameter for a microfluidic separation of normal whole blood. FIG. 71B is a graph of cell count versus hydrodynamic cell diameter for a microfluidic separation of whole blood including a population of circulating tumor cells. FIG. 71C is the graph of FIG. 71B, additionally showing a size cutoff that excludes most native blood cells. FIG. 71D is the graph of FIG. 71C, additionally showing that the population of cells larger than the size cutoff may include endothelial cell, endometrial cells, or trophoblasts indicative of a disease state.
  • FIG. 72 is a schematic illustration of a method that features isolating and counting large cells within a cellular sample, wherein the count is indicative of a patient's disease state, and subsequently further analyzing the large cell subpopulation.
  • FIG. 73A is a design for a preferred deterministic device that may be incorporated into the invention. FIG. 73B is a table of design parameters corresponding to FIG. 73A.
  • FIG. 74 is a cross-sectional view of a magnetic separation device useful in a device of the invention and associated process flow for cell isolation followed by release for off-line analysis according to the present invention.
  • FIG. 75 is a schematic of the fabrication and functionalization of a magnetic separation device. The magnetized posts enable post-packaging modification of the device.
  • FIG. 76 is a schematic of an application of a magnetic separation device to capture and release CD71+ cells from a complex mixture, such as blood, using monoclonal antibodies to the transferrin (CD71) receptor.
  • FIG. 77 is a schematic representation of an application of a magnetic separation device to capture and release CD71+ cells from a complex mixture, such as blood, using holotransferrin. Holotransferrin is rich in iron content, commercially available, and has higher affinity constants and specificity of interaction with the CD71 receptor than its counterpart monoclonal antibody.
  • Figures are not necessarily to scale.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The invention provides analytical devices and methods useful for enriching analytes in a sample. In one embodiment, the invention provides a device that includes a channel that deterministically deflects particles based on hydrodynamic size and a reservoir containing a reagent capable of altering a magnetic property of the particle. The invention also provides a method for producing a sample enriched in a first analyte relative to a second analyte by applying the sample to a device that includes a channel that deterministically deflects particles based on hydrodynamic size, thereby producing a second sample enriched in the first analyte, and combining the second sample with a reagent that alters a magnetic property of the first analyte, and applying a magnetic field thereby separating the first analyte from the second analyte. The methods and devices of the present invention may be used to enrich samples for analytes such as red blood cells (e.g., fetal red blood cells). Examples of fluid samples that are contemplated by the present invention include biological fluid samples, such as, whole blood, sweat, tears, ear flow, sputum, lymph, bone marrow suspension, lymph, urine, saliva, semen, vaginal flow, cerebrospinal fluid, brain fluid, ascites, milk, secretions of the respiratory, intestinal and genitourinary tracts, and amniotic fluid. Moreover, any other biological sample (e.g., a biopsy sample) which may be solubilized is also contemplated by the systems and methods herein. Analytes in biological fluid samples include, but are not limited to, foreign organisms such as bacteria, viruses, and protozoans.
  • Analytical Devices
  • The devices and methods of the invention may be employed in connection with any analytical device. Examples include affinity columns, cell counters, particle sorters, e.g., fluorescent activated cell sorters, capillary electrophoresis, microscopes, spectrophotometers, sample storage devices, and sample preparation devices. Microfluidic devices are of particular interest in connection with the systems described herein.
  • Exemplary analytical devices include devices useful for size, shape, or deformability based separation of particles, including filters, sieves, and deterministic separation devices, e.g., those described in International Publication Nos. 2004/029221 and 2004/113877, Huang et al. Science 304, 987-990 (2004), U.S. Publication No. 2004/0144651, U.S. Pat. Nos. 5,837,115 and 6,692,952, and U.S. Application Nos. 60/703,833 and 60/704,067; devices useful for affinity capture, e.g., those described in International Publication No. 2004/029221 and U.S. application Ser. No. 11/071,679; devices useful for preferential lysis of cells in a sample, e.g., those described in International Publication No. 2004/029221, U.S. Pat. No. 5,641,628, and U.S. Application No. 60/668,415; and devices useful for arraying cells, e.g., those described in International Publication No. 2004/029221, U.S. Pat. No. 6,692,952, and U.S. application Ser. Nos. 10/778,831 and 11/146,581. Two or more devices, either the same or different devices, may be combined in series or integrated into a single device, e.g., as described in International Publication No. 2004/029221.
  • In particular embodiments, the analytical device may be used to enrich various analytes in a sample, e.g., for collection or further analysis. Rare cells or components thereof can be retained in the device, or otherwise enriched, compared to other cells as described, e.g., in International Publication No. 2004/029221. Exemplary rare cells include, depending on the sample, fetal cells (e.g., fetal red blood cells); stem cells (e.g., undifferentiated); cancer cells; immune system cells (host or graft); epithelial cells; connective tissue cells; bacteria; fungi; viruses; and pathogens (e.g., bacterial or protozoa). Such rare cells may be isolated from samples including bodily fluids, e.g., blood, or environmental sources, e.g., water or air samples. Fetal red blood cells may be enriched from maternal peripheral blood, e.g., for the purpose of determining sex and identifying aneuploidies or genetic characteristics, e.g., mutations, in the developing fetus. Cancer cells may also be enriched from peripheral blood for the purpose of diagnosis and monitoring therapeutic progress. Bodily fluids or environmental samples may also be screened for pathogens, e.g., for coliform bacteria, blood borne illnesses such as sepsis, or bacterial or viral meningitis. Rare cells also include cells from one organism present in another organism, e.g., cells from a transplanted organ. Analytes retained or otherwise enriched in the device may, for example, be labeled, e.g., with fluorescent or radioactive probes, subjected to chemical or genetic analysis (such as fluorescent in situ hybridization), if biological, cultured, or otherwise observed or probed.
  • Analytical devices may or may not include microfluidic channels, i.e., may or may not be microfluidic devices. The dimensions of the channels of the device into which analytes are introduced may depend on the size or type of analytes employed. Preferably, a channel in an analytical device has at least one dimension (e.g., height, width, length, or radius) of no greater than 10, 9.5, 9, 8.5, 8, 7.5, 7, 6.5, 6, 5.5, 5, 4.5, 4, 3.5, 3, 2.5, 2, 1.5, or 1 mm. Microfluidic devices employed in the systems and methods described herein preferably have at least one dimension of less than 1, 0.9, 0.8, 0.7, 0.6, 0.5, 0.4, 0.3, 0.2, 0.1, or even 0.05 mm. The preferred dimensions of an analytical device can be determined by one skilled in the art based on the desired application.
  • Additional Components
  • In addition to an analytical device and a reservoir containing a reagent capable of altering a magnetic property of an analyte, devices of the invention may include additional elements, e.g., for isolating, collection, manipulation, or detection. Such elements are known in the art. For example, a device of the invention (e.g., a device incorporating a deterministic device) may also include components for other types of separation, including affinity, magnetic, electrophoretic, centrifugal, and dielectrophoretic separation. Devices of the invention may also include a component for two-dimensional imaging of the output from the device, e.g., an array of wells or a planar surface. Preferably, devices described herein are employed in conjunction with an affinity enrichment.
  • Devices of the invention may also be employed in conjunction with other enrichment devices, either on the same device or in different devices. Other enrichment techniques are described, e.g., in International Publication Nos. 2004/029221 and 2004/113877, U.S. Pat. No. 6,692,952, and U.S. application Ser. Nos. 11/071,270, 11/071,679, and 60/668,415, each of which is incorporated by reference.
  • Deterministic Separation
  • In certain embodiments, the analytical device is a device that allows deterministic separation of an analyte based on the hydrodynamic size of the analyte. Such devices will employ a channel, e.g., a microfluidic channel, containing a structure that enables deterministic separation. In one example, the channel includes one or more arrays of obstacles that allow deterministic lateral displacement of components of fluids. Such devices are described, e.g., in Huang et al. Science 304, 987-990 (2004) and U.S. Publication No. 20040144651, and U.S. Application No. 60/414,258. These devices may further employ an array of a network of gaps, wherein a fluid passing through a gap is divided unequally into subsequent gaps. In one embodiment, fluid passing through a gap is divided unequally even though the gaps are identical in dimensions. A flow carries particles to be separated through the array of gaps. The flow is aligned at a small angle (flow angle) with respect to a line-of-sight of the array. Particles having a hydrodynamic size larger than a critical size migrate along the line-of-sight in the array, whereas those having a hydrodynamic size smaller than the critical size follow the flow in a different direction. Flow in the device occurs under laminar flow conditions.
  • The critical size is a function of several design parameters. With reference to the obstacle array in FIG. 1, each row of obstacles is shifted horizontally with respect to the previous row by Δλ, where λ is the center-to-center distance between the obstacles (FIG. 1A). The parameter Δλ/λ (the “bifurcation ratio,”ε) determines the ratio of flow bifurcated to the left of the next obstacle. In FIG. 1, ε is 1/3, for the convenience of illustration. In general, if the flux through a gap between two obstacles is φ, the minor flux is εφ, and the major flux is (1-εφ) (FIG. 2). In this example, the flux through a gap is divided essentially into thirds (FIG. 1B). While each of the three fluxes through a gap weaves around the array of obstacles, the average direction of each flux is in the overall direction of flow. FIG. 1C illustrates the movement of an analyte sized above the critical size (e.g., 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 microns) through the array. Such analytes move with the major flux, being transferred sequentially to the major flux passing through each gap.
  • Referring to FIG. 2, the critical size is approximately 2Rcritical, where Rcritical is the distance between the stagnant flow line and the obstacle. If the center of mass of a particle, e.g., a cell, falls within Rcritical, the particle would follow the major flux and move along the line-of-sight of the array. Rcritical can be determined if the flow profile across the gap is known (FIG. 3); it is the thickness of the layer of fluids that would make up the minor flux. For a given gap size, d, Rcritical can be tailored based on the bifurcation ratio, ε. In general, the smaller ε, the smaller Rcritical.
  • In an array for deterministic lateral displacement, particles of different shapes behave as if they have different sizes (FIG. 4). For example, lymphocytes are spheres of ˜5 μm diameter, and erythrocytes are biconcave disks of ˜7 μm diameter, and ˜1.5 μm thick. The long axis of erythrocytes (diameter) is larger than that of the lymphocytes, but the short axis (thickness) is smaller. If erythrocytes align their long axes to a flow when driven through an array of obstacles by the flow, their hydrodynamic size is effectively their thickness (˜1.5 μm), which is smaller than lymphocytes. When an erythrocyte is driven through an array of obstacles by a hydrodynamic flow, it tends to align its long axis to the flow and behave like a ˜1.5 μm-wide particle, which is effectively “smaller” than lymphocytes. The method and device may therefore separate analytes according to their shapes, although the volumes of the analytes could be the same. In addition, analytes having different deformabilities behave as if they have different sizes (FIG. 5). For example, two analytes having the same undeformed shape may be separated by deterministic lateral displacement, as one analyte may deform more readily than the other analyte when it contacts an obstacle in the array and changes shape. Thus, separation in the device may be achieved based on any parameter that affects hydrodynamic size including the physical dimensions, the shape, and the deformability of the analyte.
  • Referring to FIG. 6, feeding a mixture of analytes, e.g., cells, of different hydrodynamic sizes from the top of the array and collecting the analytes at the bottom, as shown schematically, can produce two products, an output containing analytes larger than the critical size, 2Rcritical, and an output containing cells smaller than the critical size. Either output or both outputs may be collected, e.g., when fractionating a sample into two or more sub-samples. Analytes larger than the gap size will get trapped inside the array. Therefore, an array has a working size range. Cells have to be larger than a cut-off size (2Rcritical) and smaller than a maximum pass-through size (array gap size) to be directed into the major flux. The “size range” of an array is defined as the ratio of maximum pass-through size to cut-off size.
  • Separation of Free, Unreacted Reagent from Altered Analyte
  • Deterministic devices may be employed in order to separate free, unreacted reagent from the altered analyte. As shown in FIG. 60, a labeling reagent such as an antibody may be pre-incubated with an analyte (e.g., a cellular sample) prior to introduction to or within the deterministic device. Desirably, the reagent specifically reacts with the analyte of interest, e.g., a cell population such as epithelial cells. Exemplary labeling reagents include antibodies, quantum dots, phage, aptamers, fluorophore-containing molecules, enzymes capable of carrying out a detectable chemical reaction, sodium nitrite, or functionalized beads. Generally, the reagent is smaller than the. analyte (e.g., a cell) of interest, or the analyte of interest bound to a bead; thus, when the sample combined with the reagent is introduced to the device, unreacted reagent moves through the device undeflected, while an altered analyte (e.g., an analyte bound to the reagent) is deflected, thereby separating the unreacted reagent from the altered analyte. Advantageously, this method achieves both size separation and separation of free, unreacted reagent from the analyte. Additionally, this method of separation facilitates downstream sample analysis, if desired, without the need for a release step or a potentially destructive method of analysis, as described below.
  • FIG. 61 shows a particular case in which the enriched, labeled sample contains a population of non-target cells that co-separate with the target cells due to similar size. The non-target cells do not interfere with downstream sample analysis that relies on detection of the bound labeling reagent, because this reagent binds selectively to the cells of interest.
  • Array Design
  • Deterministic separation may be achieved using an array of gaps and obstacles in a channel. Exemplary configurations of such arrays, bypass channels, and boundaries are described as follows.
  • Single-stage array. In one embodiment, a single stage contains an array of obstacles, e.g., cylindrical posts (FIG. 1D). In certain embodiments, the array has a maximum pass-through size that is several times larger than the cut-off size, e.g., when separating white blood cells from red blood cells. This result may be achieved using a combination of a large gap size d and a small bifurcation ratio ε. In preferred embodiments, the ε is at most 1/2, e.g., at most 1/3, 1/10, 1/30, 1/100, 1/300, or 1/1000. In such embodiments, the obstacle shape may affect the flow profile in the gap; however, the obstacles can be compressed in the flow direction, in order to make the array short (FIG. 1E). Single stage arrays may include bypass channels as described herein.
  • Multiple-stage arrays. In another embodiment, multiple stages are employed to separate analytes over a wide size range. An exemplary device is shown in FIG. 7. The device shown has three stages, but any number of stages may be employed, and an array can have as many stages as desired. Typically, the cut-off size in the first stage is larger than the cut-off in the second stage, and the first stage cut-off size is smaller than the maximum pass-through size of the second stage (FIG. 8). The same is true for the following stages. The first stage will deflect (and remove) analytes, e.g., that would cause clogging in the second stage, before they reach the second stage. Similarly, the second stage will deflect (and remove) analytes that would cause clogging in the third stage, before they reach the third stage.
  • As described, in a multiple-stage array, large analytes, e.g., cells, that could cause clogging downstream are deflected first, and these deflected analytes need to bypass the downstream stages to avoid clogging. Thus, devices of the invention may include bypass channels that remove output from an array. Although described here in terms of removing analytes above the critical size, a bypass channel may also be employed to remove output from any portion of the array.
  • Different designs for bypass channels are as follows.
  • Single bypass channels. In this design, all stages share one bypass channel, or there is only one stage. The physical boundary of the bypass channel may be defined by the array boundary on one side and a sidewall on the other (FIGS. 9-11). Single bypass channels may also be employed with duplex arrays (FIG. 12).
  • Single bypass channels may also be designed, in conjunction with an array, to maintain constant flux through a device (FIG. 13). As shown, the bypass channel has varying width designed maintain constant flux through all the stages, so that the flow in the channel does not interfere with the flow in the arrays. Such a design may also be employed with an array duplex (FIG. 14). Single bypass channels may also be designed in conjunction with the array in order to maintain substantially constant fluidic resistance through all stages (FIG. 15). Such a design may also be employed with an array duplex (FIG. 16.)
  • Multiple bypass channels. In this design (FIG. 17), each stage has its own bypass channel, and the channels are separated from each other by sidewalls. Large analytes, e.g., cells are deflected into the major flux to the lower right corner of the first stage and then into in the bypass channel (bypass channel 1 in FIG. 17). Smaller cells that would not cause clogging in the second stage proceed to the second stage, and cells above the critical size of the second stage are deflected to the lower right corner of the second stage and into in another bypass channel (bypass channel 2 in FIG. 17). This design may be repeated for as many stages as desired. In this embodiment, the bypass channels are not fluidically connected, allowing for collection or other manipulation of multiple fractions. The bypass channels do not need to be straight or be physically parallel to each other (FIG. 18). Multiple bypass channels may also be employed with duplex arrays (FIG. 19).
  • Multiple bypass channels may be designed, in conjunction with an array to maintain constant flux through a device (FIG. 20). In this example, bypass channels are designed to remove an amount of flow so the flow in the array is not perturbed, i.e., substantially constant. Such a design may also be employed with an array duplex (FIG. 21). In this design, the center bypass channel may be shared between the two arrays in the duplex.
  • Optimal boundary design. If the array were infinitely large, the flow distribution would be the same at every gap. The flux φ going through a gap would be the same, and the minor flux would be εφ for every gap. In practice, the boundaries of the array perturb this infinite flow pattern. Portions of the boundaries of arrays may be designed to generate the flow pattern of an infinite array. Boundaries may be flow-feeding, i.e., the boundary injects fluid into the array or flow-extracting, i.e., the boundary extracts fluid from the array.
  • A preferred flow-extracting boundary widens gradually to extract εφ (represented by arrows in FIG. 22) from each gap at the boundary (d=24 μm, ε=1/60). For example, the distance between the array and the sidewall gradually increases to allow for the addition of εφ from each gap to the boundary. The flow pattern inside this array is not affected by the bypass channel because of the boundary design.
  • A preferred flow-feeding boundary narrows gradually to feed exactly εφ (represented by arrows in FIG. 23) into each gap at the boundary (d=24 μm, ε=1/60). For example, the distance between the array and the sidewall gradually decreases to allow for the removal of εφ to each gap from the boundary. Again, the flow pattern inside this array is not affected by the bypass channel because of the boundary design.
  • A flow-feeding boundary may also be as wide as or wider than the gaps of an array (FIG. 24) (d=24 μm, ε=1/60). A wide boundary may be desired if the boundary serves as a bypass channel, e.g., to allow for collection of analytes. A boundary may be employed that uses part of its entire flow to feed the array and feeds εφ into each gap at the boundary (represented by arrows in FIG. 24).
  • FIG. 25 shows a single bypass channel in a duplex array (ε=1/10, d=8 μm). The bypass channel includes two flow-feeding boundaries. The flux across the dashed line 1 in the bypass channel is Φbypass. A flow φ joins Φbypass from a gap to the left of the dashed line. The shapes of the obstacles at the boundaries are adjusted so that the flows going into the arrays are εφ at each gap at the boundaries. The flux at dashed line 2 is again Φbypass.
  • On-chip Flow Resistor for Defining and Stabilizing Flow
  • Deterministic separation may also employ fluidic resistors to define and stabilize flows within an array and to also define the flows collected from the array. FIG. 26 shows a schematic of planar device; a sample, e.g., blood, inlet channel, a buffer inlet channel, a waste outlet channel, and a product outlet channel are each connected to an array. The inlets and outlets act as flow resistors. FIG. 26 also shows the corresponding fluidic resistances of these different device components.
  • Flow Definition within the Array
  • FIGS. 27 and 28 show the currents and corresponding widths of the sample and buffer flows within the array when the device has a constant depth and is operated with a given pressure drop. The flow is determined by the pressure drop divided by the resistance. In this particular device, Iblood and Ibuffer are equivalent, and this determines equivalent widths of the blood and buffer streams in the array.
  • Definition of Collection Fraction
  • By controlling the relative resistance of the product and waste outlet channels, one can modulate the collection tolerance for each fraction. For example, in this particular set of schematics, when Rproduct is greater than Rwaste, a more concentrated product fraction will result at the expense of a potentially increased loss to and dilution of waste fraction. Conversely, when Rproduct is less than Rwaste, a more dilute and higher yield product fraction will be collected at the expense of potential contamination from the waste stream.
  • Flow Stabilization
  • Each of the inlet and outlet channels can be designed so that the pressure drops across the channels are appreciable to or greater than the fluctuations of the overall driving pressure. In typical cases, the inlet and outlet pressure drops are 0.001 to 0.99 times the driving pressure.
  • Multiplexed Deterministic Arrays
  • Deterministic separation may be achieved using multiplexed deterministic arrays. Putting multiple arrays on one device increases sample-processing throughput, and allows for parallel processing of multiple samples or portions of the sample for different fractions or manipulations. Multiplexing is further desirable for preparative applications. The simplest multiplex device includes two devices attached in series, i.e., a cascade. For example, the output from the major flux of one device may be coupled to the input of a second device. Alternatively, the output from the minor flux of one device may be coupled to the input of the second device.
  • Duplexing. Two arrays can be disposed side-by-side, e.g., as mirror images (FIG. 29). In such an arrangement, the critical size of the two arrays may be the same or different. Moreover, the arrays may be arranged so that the major flux flows to the boundary of the two arrays, to the edge of each array, or a combination thereof. Such a multiplexed array may also contain a central region disposed between the arrays, e.g., to collect analytes above the critical size or to alter the sample, e.g., through buffer exchange, reaction, or labeling.
  • Multiplexing on a device. In addition to forming a duplex, two or more arrays that have separated inputs may be disposed on the same device (FIG. 30A). Such an arrangement could be employed for multiple samples, or the plurality of arrays may be connected to the same inlet for parallel processing of the same sample. In parallel processing of the same sample, the outlets may or may not be fluidically connected. For example, when the plurality of arrays has the same critical size, the outlets may be connected for high throughput samples processing. In another example, the arrays may not all have the same critical size or the analytes in the arrays may not all be treated in the same manner, and the outlets may not be fluidically connected.
  • Multiplexing may also be achieved by placing a plurality of duplex arrays on a single device (FIG. 30B). A plurality of arrays, duplex or single, may be placed in any possible three-dimensional relationship to one another.
  • Exemplary multiple stage devices. In addition to those described above, the following exemplary multiple stage deterministic devices may also be included in devices of the invention. For example, FIG. 58A shows the “cascade” configuration, in which outlet 1 of one device is joined to a sample inlet of a second device. This allows for an initial separation step using the first device so that the sample introduced to the second device is already enriched for cells of interest. The two devices may have either identical or different critical sizes, depending on the intended application.
  • In FIG. 60, an unlabeled cellular sample is introduced to the first device in the cascade via a sample inlet, and a buffer containing labeling reagent is introduced to the first device via the fluid inlet. Epithelial cells are deflected and emerge from the center outlet in the buffer containing labeling reagent. This enriched labeled sample is then introduced to the second device in the cascade via a sample inlet, while buffer is added to the second device via the fluid inlet. Further enrichment of target cells and separation of free labeling reagent is achieved, and the enriched sample may be further analyzed. Alternatively, labeling reagent may be added directly to the sample emerging from the center outlet of the first device before introduction to the second device. The use of a cascade configuration may allow for the use of a smaller quantity or a higher concentration of labeling reagent at less expense than the single-device configuration of FIG. 60; in addition, any nonspecific binding that may occur is significantly reduced by the presence of an initial separation step using the first device.
  • An alternative configuration of two or more device stages is the “bandpass” configuration. FIG. 58B shows this configuration, in which outlet 2 of one device is joined to a sample inlet of a second device. This allows for an initial separation step using the first device so that the sample introduced to the second device contains cells that remained undeflected within the first device. This method may be useful when the cells of interest are not the largest cells in the sample; in this instance, the first stage may be used to reduce the number of large non-target cells by deflecting them to the center outlet. As in the cascade configuration, the two devices may have either identical or different critical sizes, depending on the intended application. For example, different critical sizes are appropriate for an application requiring the separation of epithelial cells, in comparison with an application requiring the separation of smaller endothelial cells.
  • In FIG. 66, a cellular sample pre-incubated with labeling reagent is introduced to a sample inlet of the first device of the bandpass configuration, and a buffer is introduced to the first device via the fluid inlet. The first device is disposed in such a manner that large, non-target cells are deflected and emerge from the center outlet, while a mixture of target cells, small non-target cells, and labeling reagent emerge from outlet 2 of the first device. This mixture is then introduced to the second device via a sample inlet, while buffer is added to the second device via the fluid inlet. Enrichment of target cells and separation of free labeling reagent is achieved, and the enriched sample may be further analyzed. Non-specific binding of labeling reagent to the deflected cells in the first stage is acceptable in this method, as the deflected cells and any bound labeling reagent are removed from the system.
  • In any of the multiple deterministic device configurations described above, the devices and the connections joining them may be integrated into a single device. For example, a single cascade device including two or more stages is possible, as is a single bandpass device including two or more stages. The output of the multiple stages is then coupled to the input of the reservoir.
  • Small-footprint arrays. Deterministic devices may also feature a small footprint. Reducing the footprint of an array can lower cost, and reduce the number of collisions with obstacles to eliminate any potential mechanical damage or other effects to analytes. The length of a multiple stage array can be reduced if the boundaries between stages are not perpendicular to the direction of flow. The length reduction becomes significant as the number of stages increases. FIG. 31 shows a small-footprint three-stage array.
  • Reservoir Containing a Reagent That Alters a Magnetic Property
  • An analytical device (e.g., a deterministic device) is coupled to, or otherwise includes, a reservoir containing a reagent (e.g., magnetic particles having a binding moiety or sodium nitrite) capable of altering a magnetic property of an analyte (e.g., a cell such as a red blood cell). The reservoir may include a channel, e.g., a microfluidic channel, a tube, or any other container capable of receiving the analyte and contacting it with the reagent. The reservoir may be separable from the analytical device or may be integrated with it. Mixing of the reagent with the analyte may occur by any means including diffusion, mechanical mixing, or turbulent flow. The reagent may be stored dry in the reservoir and liquefied upon introduction of a sample or stored in solution and mixed with the sample. In another embodiment, the reagent is added continuously or in a discrete bolus to the reservoir concomitant with the delivery of the sample.
  • The reservoir may also include structures that allow for the separation of the altered analyte from the unreacted reagent. For example, deterministic separation may be employed for this purpose as described herein. Alternatively, filters, rinses, or other means may be employed. Such a structure may or may not be included as part of the reservoir or analytical device.
  • The reservoir may also include an apparatus useful in enriching or depleting a sample in the magnetically altered analyte. Such devices are described herein and include channels (e.g., microfluidic channels) which, in some embodiments include a magnetic field generator or a channel containing a magnet such as a MACS column (e.g., an MS or LD column from Miltenyi Biotec, Inc., Auburn, Calif.).
  • In one embodiment, the reservoir includes a channel having a magnetic region to which a magnetic particle can magnetically attach, thereby creating a textured surface with which an analyte passing through the channel can come into contact. Through the appropriate choice of magnetic particle size and shape relative to the dimensions of the channel, a texture that enhances interactions between an analyte and the bound magnetic particles can be provided. The magnetic particles may be coated with appropriate capture moieties such as antibodies (e.g., anti-CD71, anti-CD36, anti-CD45, anti-GPA, anti-antigen i, anti-CD34, anti-fetal hemoglobin, anti-EpCAM, anti-E-cadherin, or anti-Muc-1) that can bind to an analyte through affinity mechanisms. The magnetic particles can be disposed uniformly throughout a device or in spatially resolved regions. In addition, magnetic particles may be used to create structure within the device. For example, two magnetic regions on opposite sides of a channel can be used to attract magnetic particles to form a “bridge” linking the two regions. The magnetic particles can be magnetically attached to hard magnetic regions of the channel or to soft magnetic regions that are actuated to produce a magnetic field.
  • In another embodiment, the sample is treated with a reagent that includes magnetic particles prior to application of a magnetic field. As described above, the magnetic particles may be coated with appropriate capture moieties such as antibodies to which an analyte can bind. Application of a magnetic field to the treated sample will selectively bind an analyte bound to magnetic particles in the reservoir.
  • In yet another embodiment, a sample is combined with a reagent that alters an intrinsic magnetic property of an analyte. The altered analyte may be rendered more magnetically responsive, less magnetically responsive, or may be rendered magnetically unresponsive by the reagent as compared to the unaltered analyte. In one example, a sample (e.g., a maternal blood sample that has, for example, been depleted of maternal red blood cells) containing fetal red blood cells (fRBCs) is treated with sodium nitrite, thereby causing oxidation of fetal hemoglobin contained within the fRBCs. This oxidation alters the magnetic responsiveness of the fetal hemoglobin relative to other components of the sample, e.g., maternal white blood cells, thereby allowing separation of the fRBCs. Any cell containing magnetically responsive components such as iron found in hemoglobin (e.g., adult or fetal), myoglobin, or cytochromes (e.g., cytochrome C) may be modified to alter intrinsic magnetic responsiveness of an analyte such as a cell, or a component thereof (e.g., an organelle).
  • For any of the above embodiments, any source of a magnetic field may be employed in the invention and may include hard magnets, soft magnets, or a combination thereof. In one embodiment, a spatially nonuniform permanent magnet or electromagnet may be used to create organized and in some cases periodic arrays of magnetic particles within an otherwise untextured microfluidic channel (Deng et al. Applied Physics Letters, 78, 1775 (2001)). An electromagnet may be employed to create a non-uniform magnetic field in a device. The non-uniform filed creates regions of higher and lower magnetic field strength, which, in turn, will attract magnetic particles in a periodic arrangement within the device. Other external magnetic fields may be employed to create magnetic regions to which magnetic particles attach. A hard magnetic material may also be used in the fabrication of the device, thereby obviating the need for electromagnets or external magnetic fields. In one embodiment, the device contains a plurality of channels having magnetic regions, e.g., to increase volumetric throughput. Further, these channels may be stacked vertically.
  • In the above embodiments, an analyte bound to a magnet can be released from defined locations within the channel, e.g., by increasing the overall flow rate of the fluid flowing through the device, decreasing the magnetic field, or through some combination of the two.
  • An example of a reservoir is shown in FIG. 74, which illustrates a reservoir geometry and functional process flow to isolate and then release target analytes, e.g., cells or molecules, from a complex mixture. As shown, the reservoir contains obstacles that extend from one channel surface toward the opposing channel surface. The obstacles may or may not extend the entire distance across the channel. In the present example, the obstacles are magnetic (e.g., contain hard or soft magnetic materials or are locations of high magnetic field in a non-uniform field) and attract and retain magnetic particles, which may be coated with capture moieties or may be cells attracted to a magnetic field. The geometry of the reservoir, the distribution, shape, size of the obstacles and the flow parameters can be altered to optimize the efficiency of the enrichment of an analyte of interest, for example, by attracting an analyte bound to a magnetic particle with a capture moiety (e.g., as described in International Publication No. 2004/029221). In one specific example, an anodic lidded silicon wafer with microtextured magnetic obstacles of varying shapes (cylindrical, rectangular, trapezoidal, or pleomorphic) and size (10-999 microns) are arranged uniquely (spacing and density varied across equilateral triangular, diagonal, and random array distribution) to maximize the collision frequency of analytes, altered or not, with the obstacles within the confines of a continuous perfusion flow stream. The exact geometry of the magnetic obstacles and the distribution of obstacles may depend on the type of analytes being isolated, enriched, or purified.
  • FIG. 75 illustrates an example of reservoir fabrication and functionalization. The magnetized obstacles enable post-packaging modification of the reservoir. This is a very significant improvement over existing art. The incompatibility of semiconductor processing parameters (high heat, or solvent sealers to bond the lid) with capture moieties (sensitive to temperature and inorganic and organic solvents) makes this device universal and compatible for functionalization with all capture moieties. Retention of the capture moieties on the obstacles (e.g., posts) by use of magnetic fields, is an added advantage over prior art that uses complex surface chemistry for immobilization. The reservoir enables the end user to easily and rapidly charge the reservoir with a capture moiety, or mixture of capture moieties, of choice thereby increasing the versatility of use. On-demand and ‘just-in-time’ one step functionalization is enabled by this reservoir, thereby circumventing issues of on-the-shelf stability of the capture moieties if they were chemically cross-linked at production. The capture moieties that can be loaded and retained on the obstacles include, but not limited to, all of the cluster of differentiation (CD) receptors on mammalian cells, synthetic and recombinant ligands for cell receptors, and any other organic, inorganic molecule, or compound of interest that can be attached to any magnetic particle.
  • Reagents Capable of Altering a Magnetic Property
  • Such reagents include any reagent that is capable of altering a magnetic property of an analyte in a sample. The exact nature of the reagent will depend on the nature of the analyte. Exemplary reagents include agents that oxidize or reduce transition metals, magnetic beads capable of binding to an analyte, or reagents that are capable of chelating or otherwise binding iron, or other magnetic materials or particles. Specific reagents include sodium nitrite. The reagent may act to alter the magnetic properties of an analyte to enable or increase its attraction to a magnetic field, to enable or increase its repulsion to a magnetic field, or to eliminate a magnetic property such that the analyte is unaffected by a magnetic field.
  • Any particle that responds to a magnetic field may be employed in the devices and methods of the invention. Desirable particles are those that have surface chemistry that can be chemically or physically modified, e.g., by chemical reaction, physical adsorption, entanglement, or electrostatic interaction. Magnetic particles of the present invention can come in any size and/or shape. In some embodiments, a magnetic particle has a diameter of less than 500 nm, 400 nm, 300 nm, 200 nm, 100 nm, 90 nm, 80 nm, 70 nm, 60 nm or 50 nm. In some embodiments, a magnetic particle has a diameter that is between 10-1000 nm, 20-800 nm, 30-600 nm, 40-400 nm, or 50-200 nm. In some embodiments, a magnetic particle has a diameter of more than 10 nm, 50 nm, 100 nm, 200 nm, 500 nm, 1000 nm, or 5000 nm. The magnetic particles can be dry in liquid form. Mixing of a fluid sample with a second liquid medium containing magnetic particles can occur by any means known in the art.
  • Capture moieties can be bound to magnetic particles by any means known in the art. Examples include chemical reaction, physical adsorption, entanglement, or electrostatic interaction. The capture moiety bound to a magnetic particle will depend on the nature of the analyte targeted. Examples of capture moieties include, without limitation, proteins (such as antibodies, avidin, and cell-surface receptors), charged or uncharged polymers (such as polypeptides, nucleic acids, and synthetic polymers), hydrophobic or hydrophilic polymers, small molecules (such as biotin, receptor ligands, and chelating agents), and ions. Such capture moieties can be used to specifically bind cells (e.g., bacterial, pathogenic, fetal cells, fetal blood cells, cancer cells, and blood cells), organelles (e.g., nuclei), viruses, peptides, protein, polymers, nucleic acids, supramolecular complexes, other biological molecules (e.g., organic or inorganic molecules), small molecules, ions, or combinations or fragments thereof. Specific examples of capture moieties include anti-CD71, anti-CD36, anti-GPA, anti-EpCAM, anti-E-cadherin, anti-Muc-1, and holo-transferrin. In another embodiment, the capture moiety is fetal cell specific.
  • Magnetic Separation
  • Once a magnetic property of an analyte has been altered, that alteration may be used to effect an isolation, enrichment, or depletion of the analyte relative to other constituents of a sample. The isolation, enrichment, or depletion may include positive selection, i.e., a desired analyte is attracted to a magnetic field, or it may employ negative selection, i.e., a desired analyte is not attracted to the magnetic field. In either case, the population of analytes containing the desired analytes may be collected for analysis or further processing.
  • The device used to perform the magnetic separation may be any device that can produce a magnetic field. In one embodiment, a MACS column is used to effect separation of the magnetically altered analyte. If the analyte is rendered magnetically responsive by the reagent (e.g., using any reagent described herein), it may bind to the MACS column, thereby permitting enrichment of the desired analyte relative to other constituents of the sample.
  • In another embodiment, separation may be achieved using a device, typically microfluidic, that contains a plurality of magnetic obstacles. If an analyte in the sample is modified to be magnetically responsive (e.g., through a reagent that enhances an intrinsic magnetic property of the analyte or by binding of a magnetically responsive particle to the analyte), the analyte may bind to the obstacles, thereby permitting enrichment of the bound analyte. Alternatively, negative selection may be employed. In this example, the desired analyte may be rendered magnetically unresponsive, or an undesired analyte may be rendered magnetically responsive or bound to a magnetically responsive particle. In this case, an undesired analyte or analytes will be retained on the obstacles whereas the desire analyte will not, thus enriching the sample in the desired analyte.
  • Magnetic regions of the device can be fabricated with either hard or soft magnetic materials, such as, but not limited to, rare earth materials, neodymium-iron-boron, ferrous-chromium-cobalt, nickel-ferrous, cobalt-platinum, and strontium ferrite. Portions of the device may be fabricated directly out of magnetic materials, or the magnetic materials may be applied to another material. The use of hard magnetic materials can simplify the design of a device because they are capable of generating a magnetic field without other actuation. Soft magnetic materials, however, enable release and downstream processing of bound analytes simply by demagnetizing the material. Depending on the magnetic material, the application process can include cathodic sputtering, sintering, electrolytic deposition, or thin-film coating of composites of polymer binder-magnetic powder. A preferred embodiment is a thin film coating of micromachined obstacles (e.g., silicon posts) by spin casting with a polymer composite, such as polyimide-strontium ferrite (the polyimide serves as the binder, and the strontium ferrite as the magnetic filler). After coating, the polymer magnetic coating is cured to achieve stable mechanical properties. After curing, the device is briefly exposed to an external induction field, which governs the preferred direction of permanent magnetism in the device. The magnetic flux density and intrinsic coercivity of the magnetic fields from the obstacles can be controlled by the % volume of the magnetic filler.
  • In another embodiment, an electrically conductive material is micropatterned on the outer surface of an enclosed microfluidic device. The pattern may consist of a single, electrical circuit with a spatial periodicity of approximately 100 microns. By controlling the layout of this electrical circuit and the magnitude of the electrical current that passes through the circuit, one can develop periodic regions of higher and lower magnetic strength within the enclosed microfluidic device.
  • The magnetic field can be adjusted to influence supra and paramagnetic particles with magnetic mass susceptibility ranging from 0.1-200×10−6 m3/kg. The paramagnetic particles of use can be classified based on size: particulates (1-5 μm in the size of a cell diameter); colloidal (on the order of 100 nm); and molecular (on the order of 2-10 nm). The fundamental force acting on a paramagnetic entity is: F b = 1 2 μ o Δχ V G B 2
    where Fb is the magnetic force acting on the paramagnetic entity of volume Vb, Δχ is the difference in magnetic susceptibility between the magnetic bead, χb, and the surrounding medium, χf, μo is the magnetic permeability of free space, B is the external magnetic field, and ∇ is the gradient operator. The magnetic field can be controlled and regulated to enable attraction and retention of a wide spectrum of particulate, colloidal, and molecular paramagnetic entities typically coupled to capture moieties.
    Uses of Devices of the Invention
  • As described, the invention features analytical devices for the enrichment of analytes such as particles, including bacteria, viruses, fungi, cells, cellular components, viruses, nucleic acids, proteins, and protein complexes. In addition to altering a magnetic property, a device may also be used to effect various manipulations on analytes in a sample. Such manipulations include enrichment or concentration of an analyte, including size-based fractionization, or alteration of the analyte itself or the fluid carrying the analyte. Preferably, a device is employed to enrich rare analytes from a heterogeneous mixture or to alter a rare analyte, e.g., by exchanging the liquid in the sample or by contacting an analyte with a reagent. Such devices allow for a high degree of enrichment with limited stress on a potentially fragile analyte such a cell, where devices of the invention provide reduced mechanical lysis or intracellular activation of cells.
  • Although primarily described in terms of cells, the devices of the invention may be employed with any analyte whose size allows for separation in a device of the invention.
  • Deterministic devices, and other analytical devices, may be employed in concentrated samples, e.g., where analytes are touching, hydrodynamically interacting with each other, or exerting an effect on the flow distribution around another analyte. For example, a deterministic device can separate white blood cells from red blood cells in whole blood from a human donor. Human blood typically contains ˜45% of cells by volume. Cells are in physical contact and/or coupled to each other hydrodynamically when they flow through the array. FIG. 32 shows schematically that cells densely packed inside an array can physically interact with each other.
  • As described, the devices and methods of the invention may involve separating from a sample one or more analytes based on an intrinsic magnetic property of the one or more analytes. In one embodiment, the sample is treated with a reagent that alters a magnetic property of the analyte. The alteration may be mediated by a magnetic particle or may be mediated by a reagent that alters an intrinsic magnetic property of the analyte. A magnetically altered analyte may then bind to a surface of the device, and desired analytes (e.g., rare cells such as fetal cells, pathogenic cells, cancer cells, or bacterial cells) in a sample may be retained in the device. Thus, the analyte of interest may then bind to the surfaces of the device. In another embodiment, desired analytes are retained in the device through size-, shape- or deformability-based mechanisms. In another embodiment, negative selection is employed, where a desired analyte is not bound to the device. Any of the embodiments may uses a MACS column for retention of an analyte (e.g., an analyte bound to a magnetic particle).
  • In embodiments of the invention using positive selection, it is desirable that at least 60%, 70%, 80%, 90%, 95%, 98%, or 99% of the analytes are retained in the device. The surfaces of the device are desirably designed to minimize nonspecific binding of non-target analytes. For example, at least 99%, 98%, 95%, 90%, 80%, or 70% of non-target analytes are not retained in the device. The selective retention in the device can result in the separation of a specific analyte population from a mixture, e.g., blood, sputum, urine, and soil, air, or water samples.
  • The selective retention of analytes may be obtained by introduction of magnetic particles (e.g., attached to obstacles present in the device or manipulated to create obstacles to increase surface area for an analyte to interact with to increase the likelihood of binding) into a device of the invention. Capture moieties may be bound to the magnetic particles to effect specific binding of a target analyte. In another embodiment, the magnetic particles may be disposed such as to only allow analytes of a selected size, shape, or deformability to pass through the device. Combinations of these embodiments are also envisioned. For example, a device may be configured to retain certain analytes based on size and others based on binding. In addition, a device may be designed to bind more than one analyte of interest, e.g., in a serial, parallel, or interspersed arrangement of regions within a device or where two or more capture moieties are disposed on the same magnetic particle or on adjacent particles, e.g., bound to the same obstacle or region. Further, multiple capture moieties that are specific for the same analyte (e.g., anti-CD71 and anti-CD36) may be employed in the device, either on the same or different magnetic particles, e.g., disposed on the same or different obstacle or region.
  • The flow conditions in the device are typically such that the analytes are very gently handled in the device to prevent damage. Positive pressure or negative pressure pumping or flow from a column of fluid may be employed to transport analytes into and out of the microfluidic devices of the invention. The device enables gentle processing, while maximizing the collision frequency of each analyte with one or more of the magnetic particles. The target analytes interact with any capture moieties on collision with the magnetic particles. The capture moieties can be co-localized with obstacles as a designed consequence of the magnetic field attraction in the device. This interaction leads to capture and retention of the target analytes in defined locations. Captured analyte can be released by demagnetizing the magnetic regions retaining the magnetic particles. For selective release of analytes from regions, the demagnetization can be limited to selected obstacles or regions. For example, the magnetic field can be designed to be electromagnetic, enabling turn-on and turn-off off the magnetic fields for each individual region or obstacle at will. In other embodiments, the particles can be released by disrupting the bond between the analyte and the capture moiety, e.g., through chemical cleavage or interruption of a noncovalent interaction, or by decreasing the magnetic responsiveness of the bound analyte. For example, some ferrous particles are linked to monoclonal antibody via a DNA linker; the use of DNAse can cleave and release the analytes from the ferrous particle. Alternatively, an antibody fragmenting protease (e.g., papain) can be used to engineer selective release. Increasing the sheer forces on the magnetic particles can also be used to release magnetic particles from magnetic regions, especially hard magnetic regions. In other embodiments, the captured analytes are not released and can be analyzed or further manipulated while retained.
  • FIG. 76 illustrates an example of a reservoir designed to capture and isolate cells expressing the transferrin receptor from a complex mixture. Monoclonal antibodies to CD71 receptor are readily available off-the-shelf covalently coupled to magnetic materials, such as, but not limited to, ferrous doped polystyrene and ferroparticles or ferro-colloids (e.g., from Miltenyi and Dynal). The mAB to CD71 bound to magnetic particles is flowed into the reservoir. The antibody-coated particles are attracted to the obstacles (e.g., posts), floor, and walls and are retained by the strength of the magnetic field interaction between the particles and the magnetic field. The particles between the obstacles and those loosely retained with the sphere of influence of the local magnetic fields away from the obstacles, are removed by a rinse (the flow rate can be adjusted such that the hydrodynamic shear stress on the particles away from the obstacles is larger than the magnetic field strength).
  • FIG. 77 is a preferred embodiment for application of the reservoir to capture and release CD71+ cells from a complex mixture, e.g., blood, using holo-transferrin. Holo-transferrin is rich in iron content, commercially available, and has higher affinity constants and specificity of interaction with the CD71 receptor than its counterpart monoclonal antibody. The iron coupled to the transferrin ligand serves the dual purpose of retaining the conformation of the ligand for binding with the cell receptor, and as a molecular paramagnetic element for retaining the ligand on the obstacles.
  • In addition to the above embodiments, devices of the invention can be used for isolation and detection of blood borne pathogens, bacterial and viral loads, airborne pathogens solubilized in aqueous medium, pathogen detection in food industry, and environmental sampling for chemical and biological hazards. Additionally, the magnetic particles can be co-localized with a capture moiety and a candidate drug compound. Capture of a cell of interest can further be analyzed for the interaction of the captured cell with the immobilized drug compound. A device can thus be used to both isolate sub-populations of cells from a complex mixture and assay their reactivity with candidate drug compounds for use in the pharmaceutical drug discovery process for high throughput and secondary cell-based screening of candidate compounds. In other embodiments, receptor-ligand interaction studies for drug discovery can be accomplished in the device by localizing the capture moiety, i.e., the receptor, on a magnetic particle, and flowing in a complex mixture of candidate ligands (or agonists or antagonists). The ligand of interest is captured, and the binding event can be detected, e.g., by secondary staining with a fluorescent probe. This embodiment enables rapid identification of the absence or presence of known ligands from complex mixtures extracted from tissues or cell digests or identification of candidate drug compounds.
  • Enrichment
  • In one embodiment, devices of the invention are employed to produce a sample enriched in a desired analyte, e.g., based at least in part on hydrodynamic size. Applications of such enrichment include concentrating of an analyte such as particle including rare cells, and size fractionization, e.g., size filtering (selecting analytes in a particular size range). Devices may also be used to enrich components of cells such as organelles (e.g., nuclei). Desirably, the devices and methods of the invention retain at least 1%, 10%, 30%, 50%, 75%, 80%, 90%, 95%, 98%, or 99% of the desired particles compared to the initial mixture, while potentially enriching the desired particles by a factor of at least 1, 10, 100, 1,000, 10,000, 100,000, or even 1,000,000 relative to one or more non-desired particles. The enrichment may also result in a dilution of the enriched analytes compared to the original sample, although the concentration of the enriched analytes relative to other particles in the sample has increased. Preferably, the dilution is at most 90%, e.g., at most 75%, 50%, 33%, 25%, 1 0%, or 1%.
  • In another embodiment, a device of the invention is used to produce a sample enriched in a rare analyte. In general, a rare analyte is an analyte that is present as less than 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, 0.1%, 0.01%, 0.001%, 0.0001%, 0.00001%, or 0.000001% of all analytes in a sample or whose mass is less than 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, 0.1%, 0.01%, 0.001%, 0.0001%, 0.00001%, or 0.000001% of total mass of a sample of a sample. Exemplary rare analytes include, depending on the sample, fetal cells, stem cells (e.g., undifferentiated), cancer cells, immune system cells (host or graft), epithelial cells, connective tissue cells, bacteria, fungi, viruses, and pathogens (e.g., bacterial or protozoa). Such rare analytes may be isolated from samples including bodily fluids, e.g., blood, or environmental sources, e.g., pathogens in water samples. Fetal red blood cells may be enriched from maternal peripheral blood, e.g., for the purpose of determining sex and identifying aneuploidies or genetic characteristics, e.g., mutations, in the developing fetus. Circulating tumor cells, which are of epithelial cell type and origin, may also be enriched from peripheral blood for the purpose of diagnosis and monitoring therapeutic progress. Circulating endothelial cells may also be similarly enriched from peripheral blood.
  • Bodily fluids or environmental samples may also be screened for pathogens, e.g., for coliform bacteria, blood borne illnesses such as sepsis, or bacterial or viral meningitis. Rare cells also include cells from one organism present in another organism, e.g., in cells from a transplanted organ.
  • In addition to enrichment of a rare analyte, a device may be employed for preparative applications. An exemplary preparative application includes generation of cell packs from blood. In one example, a device may be configured to produce fractions enriched in platelets, red blood cells, and white cells by deterministic deflection. By using multiplexed or multistage deterministic devices, all three cellular fractions may be produced in parallel or in series from the same sample. In other embodiments, the device may be employed to separate nucleated from non-nucleated cells, e.g., from cord blood sources.
  • Using devices which incorporate deterministic deflection is advantageous in situations where the particles being enriched are subject to damage or other degradation. As described herein, deterministic devices may be designed to enrich analytes (e.g., a cell) with a minimum number of collisions between the analyte and obstacles. This minimization reduces mechanical damage to the analytes (e.g., a cell) and, in the case of cells, also prevents intracellular activation caused by the collisions. Gentle handling preserves the limited number of rare analytes in a sample, in the case of cells, prevents rupture leading to contamination or degradation by intracellular components, and prevents maturation or activation of cells, e.g., stem cells or platelets. In preferred embodiments, the analyte is enriched such that fewer than 30%, 10%, 5%, 1%, 0.1%, or even 0.01% are damaged (e.g., activated or mechanically lysed).
  • FIG. 33 shows a typical size distribution of cells in human peripheral blood. The white blood cells range from ˜4 μm to ˜18 μm, whereas the red blood cells are ˜1.5 μm (short axis). A deterministic device designed to separate white blood cells from red blood cells typically has a cut-off size of 2 to 4 μm and a maximum pass-through size of greater than 18 μm.
  • In an alternative embodiment, a deterministic device may function as a detector for abnormalities in red blood cells. The deterministic principle of sorting enables a predictive outcome of, for example, % of enucleated cells deflected in the device. In a disease state, such as malarial infection or sickle cell anemia, the distortion in shape and flexibility of the red cells would significantly change the % cells deflected. This change can be monitored as a first level sentry to alert to the potential of a diseased physiology to be followed by microscopy examination of shape and size of red cells to assign the disease. The method is also generally applicable monitoring for any change in flexibility of particles in a sample.
  • In an alternative embodiment, a deterministic device may function as a detector for platelet aggregation. The deterministic principle of sorting enables a predictive outcome of % free platelets deflected in the device. Activated platelets would form aggregates and the aggregates would be deflected. This change can be monitored as a first level sentry to alert the compromised efficacy of a platelet pack for reinfusion. The method is also generally applicable monitoring for any change in size, e.g., through agglomeration, of particles in a sample.
  • FIG. 57A shows the operation of a deterministic device for purposes of enrichment. A cellular sample is added through a sample inlet of the device, and buffer medium is added through the fluid inlet. Cells below the critical size move through the device undeflected, emerging from the edge outlets in their original sample medium. Cells above the critical size, e.g., epithelial cells, are deflected and emerge from the center outlet contained in the buffer medium added through the fluid inlet. Operation of the device thus produces samples enriched in cells above and below the critical size. Because epithelial cells are among the largest cells in the bloodstream, the size and geometry of the gaps of the device may be chosen so as to direct virtually all other cell types to the edge outlets, while producing a sample from the center outlet that is substantially enriched in epithelial cells after a single pass through the device.
  • A deterministic device included in the invention need not be duplexed as shown in FIG. 57A in order to operate as described herein. The schematized representation shown in FIG. 57B may represent either a duplexed device or a single array.
  • Enrichment may be enhanced in numerous ways. For example, target analytes (e.g., cells) may be labeled with beads (e.g., immunoaffinity beads), thereby increasing their size (as depicted in FIG. 59). In the case of epithelial cells, this may further increase their size, resulting in an even more efficient separation. Alternatively, the size of smaller analytes (e.g., cells) may be increased to the extent that they become the largest objects in the sample or occupy a unique size range in comparison to the other components of the sample, or so that they copurify with other analytes. Beads may be made of polystyrene, magnetic material, or any other material that can be adhered to an analyte (e.g., cells). Desirably, such beads are neutrally buoyant so as not to disrupt the flow of labeled cells through a deterministic device.
  • Alteration
  • In other embodiments, in addition to enrichment, an analyte of interest may be contacted with an altering reagent that may chemically or physically alter the analyte or the fluid in the sample. Such applications may include purification, buffer exchange, labeling (e.g., immunohistochemical, magnetic, and histochemical labeling, cell staining, and flow in-situ fluorescence hybridization (FISH)), cell fixation, cell stabilization, cell lysis, and cell activation.
  • Such methods may allow for the transfer of analytes from a sample into a different liquid (e.g., buffer exchange). FIG. 34A shows this effect schematically for a single stage deterministic device, FIG. 34B shows this effect for a multistage deterministic device, FIG. 34C shows this effect for a duplex array of deterministic devices, and FIG. 34D shows this effect for a multistage duplex array of deterministic devices. By using such methods, analytes (e.g., blood cells) may be enriched in the sample. Such transfers of an analyte from one liquid to another may be also employed to effect a series of alterations, e.g., Wright staining blood on-chip. Such a series may include reacting an analyte with a first reagent and then transferring the particle to a wash buffer, and then another reagent.
  • FIGS. 35A-35C illustrates a further example of alteration in a two stage deterministic device having two bypass channels. In this example, the larger analytes are moved from blood to buffer and collected in stage 1, intermediate sized analytes are moved from blood to buffer in stage 2, and smaller analytes that are not moved from the blood in stage are collected also collected. FIG. 35B illustrates the size cut-off of the two stages, and FIG. 35C illustrates the size distribution of the three fractions collected.
  • FIG. 36 illustrates an example of alteration in a two stage deterministic device having bypass channels that are disposed between the lateral edge of the array and the channel wall. FIG. 37 illustrates a deterministic device similar to that in FIG. 36, except that the two stages are connected by fluidic channels. FIG. 38 illustrates alteration in a deterministic device having two stages with a small footprint. FIGS. 39A-39B illustrates alteration in a device in which the output from the first and second stages is captured in a single channel. FIG. 40 illustrates another device for use in the methods of the invention.
  • FIG. 41 illustrates the use of a deterministic device to perform multiple, sequential alterations on an analyte. In this device an analyte is moved from the sample into a regent that reacts with the analyte, and the altered analyte is then moved into a buffer, thereby removing the unreacted reagent or reaction byproducts. Additional steps may be added (e.g., steps described herein).
  • Enrichment and alteration may also be combined. For example, desired cells may be contacted with a lysing reagent and cellular components, e.g., nuclei, are enriched based on size. In another example, analytes may be contacted with particulate labels, e.g., magnetic beads, which bind to the analytes. Unbound particulate labels may be removed based on size.
  • Buffer Exchange
  • Deterministic devices may also be employed for purposes of buffer exchange. To achieve this result, a protocol similar to that used for enrichment is followed: a cellular sample is added through a sample inlet of a deterministic device, and the desired final buffer medium is added through a fluid inlet. As described above, cells above the critical size are deflected in the device and enter the buffer.
  • Concentration
  • Devices of the invention may also be employed in order to concentrate a cellular sample of interest. In one example shown in FIG. 62, a cellular sample is introduced to the sample inlet of a deterministic device. By reducing the volume of buffer introduced into the fluid inlet so that this volume is significantly smaller than the volume of the cellular sample, concentration of target cells in a smaller volume results. This concentration step may improve the results of any downstream analysis performed.
  • Cell Lysis
  • Devices of the invention may also be employed for purposes of cell lysis. To achieve this in a deterministic device, a protocol similar to that used for enrichment is followed: a cellular sample is added through a sample inlet of the device (FIG. 63), and lysis buffer is added through the fluid inlet. As described above, cells above the critical size are deflected and enter the lysis buffer, leading to lysis of these cells. As a result, the sample emerging from the center outlet includes lysed cell components including organelles, while undeflected whole cells emerge from the other outlet. Thus, the device provides a method for selectively lysing target cells.
  • Downstream Analysis
  • A key prerequisite for many diagnostic assays is the removal of a free or unreacted altering reagent from the sample to be analyzed. In one embodiment, the reagent is a labeling reagent. As described above, deterministic devices are able to separate free labeling reagent from labeling reagent bound to an analyte (e.g., a cell). It is then possible to perform a bulk measurement of the reacted sample without significant levels of background interference from free labeling reagent. In one example, fluorescent antibodies selective for a particular epithelial cell marker such as EpCAM are used. The fluorescent moiety may include Cy dyes, Alexa dyes, or other fluorophore-containing molecules. The resulting labeled sample is then analyzed by measuring the fluorescence of the resulting sample of labeled enriched analytes such as cells using a fluorimeter. Alternatively, a chromophore-containing label may be used in conjunction with a light spectrometer. The measurements obtained may be used to quantify the number of target analytes such as cells in a sample.
  • Many other methods of measurement and labeling reagents are useful in the methods and devices of the invention. Labeling antibodies may possess covalently bound enzymes that cleave a substrate, altering its absorbance at a given wavelength; the extent of cleavage is then quantified with a spectrometer. Colorimetric or luminescent readouts are possible, depending on the substrate used. Advantageously, the use of an enzyme label allows for significant amplification of the measured signal, lowering the threshold of detectability.
  • Quantum dots, e.g., Qdots® from QuantumDot Corp., may also be utilized as a labeling reagent that is covalently bound to a capture moiety such as an antibody. Qdots are resistant to photobleaching and may be used in conjunction with two-photon excitation measurements.
  • Another possible labeling reagent useful in the methods of the invention is phage. Phage display is a technology in which binding peptides are displayed by engineered phage strains having strong binding affinities for a target, e.g., a protein found on the surface of cells of interest. The peptide sequence corresponding to a given phage is encoded in that phage's DNA. Thus, phage are useful labeling reagents in that they are small relative to an analyte such as a cell and thus may be easily separated, and they additionally carry DNA that may be analyzed and quantified using PCR or similar techniques, enabling a quantitative determination of the number of cells present in an enriched bound sample.
  • FIG. 65 depicts the use of phage as a labeling reagent in which two deterministic device stages are arrayed in a cascade configuration. The method depicted in FIG. 65 fits the general description of FIG. 64, with the exception of the labeling reagent employed.
  • Downstream analysis may include an accurate determination of the number of desired analytes (e.g., cells) in the sample being analyzed. In order to produce accurate quantitative results, the amount of the target of a labeling reagent (e.g., a surface antigen on a cell of interest) typically has to be known or predictable (e.g., based on expression levels in a cell), and the binding of the labeling reagent should also proceed in a predictable manner, free from interfering substances. Thus, a device (e.g., a deterministic devices) or method that produces a highly enriched cellular samples prior to introduction of a labeling reagent are particularly useful. In addition, labeling reagents that allow for amplification of the signal produced are preferred in the case of a rare desired analyte (e.g., epithelial cells in a blood sample). Reagents that allow for signal amplification include enzymes and phage. Other labeling reagents that do not allow for convenient amplification but nevertheless produce a strong signal, such as quantum dots, are also desirable.
  • When the devices and methods of the invention are used to enrich cells contained in a sample, further quantification and molecular biology analysis may be performed on the same set of cells. The gentle treatment of the cells in the devices of the invention, coupled with the described methods of bulk measurement, maintain the integrity of the cells so that further analysis may be performed if desired. For example, techniques that destroy the integrity of the cells may be performed subsequent to bulk measurement; such techniques include DNA or RNA analysis, proteome analysis, or metabolome analysis. An example of such analysis is PCR, in which the cells are lysed and levels of particular DNA sequences are amplified. Such techniques are particularly useful when the number of target cells isolated is very low.
  • Cancer Diagnosis
  • Epithelial cells exfoliated from solid tumors have been found in the circulation of patients with cancers of the breast, colon, liver, ovary, prostate, and lung. In general, the presence of circulating tumor cells (CTCs) after therapy has been associated with tumor progression and spread, poor response to therapy, relapse of disease, and/or decreased survival. Therefore, enumeration of CTCs offers a means to stratify patients for baseline characteristics that predict initial risk and subsequent risk based upon response to therapy.
  • Unlike tumor-derived cells in bone marrow, which can be dormant and long-lived, CTCs, which are of epithelial cell type and origin, have a short half-life of approximately one day, and their presence indicates a recent influx from a proliferating tumor (Patel et al., Ann Surg, 235:226-231, 2002). Therefore, CTCs can reflect the current clinical status of patient disease and therapeutic response. The enumeration and characterization of CTCs has potential value in assessing cancer prognosis and in monitoring therapeutic efficacy for early detection of treatment failure that can lead to disease relapse. In addition, CTC analysis may detect early relapse in presymptomatic patients who have completed a course of therapy; at present, individuals without measurable disease are not eligible to participate in clinical trials of promising new treatments (Braun et al., N Engl J Med, 351:824-826, 2004).
  • The devices and methods of the invention may be used to evaluate cancer patients and those at risk for cancer. For example, a blood sample is drawn from the patient and introduced to a deterministic device of the invention with a critical size chosen appropriately to separate epithelial cells from other blood cells. The number of epithelial cells in the blood sample is determined using a method described herein. For example, the cells may be labeled with an antibody that binds to EpCAM, and the antibody may have a covalently bound fluorescent label, or be bound to a magnetic particle. A bulk measurement may then be made of the enriched sample produced by the device, and from this measurement, the number of epithelial cells present in the initial blood sample may be determined. Microscopic techniques may be used to visually quantify the cells in order to correlate the bulk measurement with the corresponding number of labeled cells in the blood sample.
  • By making a series of measurements over days, weeks, months, or years, one may track the level of epithelial cells present in a patient's bloodstream as a function of time. In the case of existing cancer patients, this provides a useful indication of the progression of the disease and assists medical practitioners in making appropriate therapeutic choices based on the increase, decrease, or lack of change in circulating epithelial cells in the patient's bloodstream. For those at risk of cancer, a sudden increase in the number of cells detected may provide an early warning that the patient has developed a tumor. This early diagnosis, coupled with subsequent therapeutic intervention, is likely to result in an improved patient outcome in comparison to an absence of diagnostic information.
  • Diagnostic methods include making bulk measurements of labeled epithelial cells isolated from blood, as well as techniques that destroy the integrity of the cells. For example, PCR may be performed on a sample in which the number of target cells isolated is very low; by using primers specific for particular cancer markers, information may be gained about the type of tumor from which the analyzed cells originated. Additionally, RNA analysis, proteome analysis, or metabolome analysis may be performed as a means of diagnosing the type or types of cancer present in the patient.
  • One important diagnostic indicator for lung cancer and other cancers is the presence or absence of certain mutations in epidermal growth factor receptor (EGFR). EGFR consists of an extracellular ligand-binding domain, a transmembrane portion, and an intracellular tyrosine kinase (TK) domain. The normal physiologic role of EGFR is to bind ErbB ligands, including epidermal growth factor (EGF), at the extracellular binding site to trigger a cascade of downstream intracellular signals leading to cell proliferation, survival, motility and other related activities. Many non-small cell lung tumors with EGFR mutations respond to small molecule EGFR inhibitors, such as gefitinib (Iressa; AstraZeneca), but often eventually acquire secondary mutations that make them drug resistant. Using the devices and methods of the invention, one may monitor patients taking such drugs by taking frequent samples of blood and determining the number of epithelial cells in each sample as a function of time. This provides information as to the course of the disease. For example, a decreasing number of circulating epithelial cells over time suggests a decrease in the severity of the disease and the size of the tumor or tumors. Immediately following quantification of epithelial cells, these cells may be analyzed by PCR to determine what mutations may be present in the EFGR gene expressed in the epithelial cells. Certain mutations, such as those clustered around the ATP-binding pocket of the EGFR TK domain, are known to make the cancer cells susceptible to gefitinib inhibition. Thus, the presence of these mutations supports a diagnosis of cancer that is likely to respond to treatment using gefitinib. However, many patients who respond to gefitinib eventually develop a second mutation, often a methionine-to-threonine substitution at position 790 in exon 20 of the TK domain, which renders them resistant to gefitinib. By using the devices and method of the invention, one may test for this mutation as well, providing further diagnostic information about the course of the disease and the likelihood that it will respond to gefitinib or similar compounds.
  • Sample Preparation
  • Samples may be employed in the methods described herein with or without manipulation, e.g., stabilization and removal of certain components. In one embodiment, the sample is enriched in the cells of interest prior to introduction to a device of the invention. Methods for enriching cell populations are described herein and known in the art, e.g., affinity mechanisms, agglutination, and size, shape, and deformability based enrichments. Some samples may be diluted or concentrated prior to introduction into the device.
  • In one embodiment, reagents are added to the sample, to selectively or nonselectively increase the hydrodynamic size of the particles within the sample. This modified sample is, for example, then pumped through a deterministic device. Because the particles are swollen and have an increased hydrodynamic diameter, it will be possible to use deterministic devices with larger and more easily manufactured gap sizes. In a preferred embodiment, the steps of swelling and size-based enrichment are performed in an integrated fashion on a deterministic device. Suitable reagents include any hypotonic solution, e.g., deionized water, 2% sugar solution, or neat non-aqueous solvents. Other reagents include beads, e.g., magnetic or polymer, that bind selectively (e.g., through antibodies or avidin-biotin) or non-selectively.
  • In another embodiment, reagents are added to the sample to selectively or nonselectively decrease the hydrodynamic size of the particles within the sample. Nonuniform decrease in particles in a sample will increase the difference in hydrodynamic size between particles. For example, nucleated cells are separated from enucleated cells by hypertonically shrinking the cells. The enucleated cells can shrink to a very small particle, while the nucleated cells cannot shrink below the size of the nucleus. Exemplary shrinking reagents include hypertonic solutions.
  • In an alternative embodiment, affinity functionalized beads are used to increase the hydrodynamic size of an analyte of interest relative to other analytes present in a sample, thereby allowing for the operation of a deterministic device with a larger and more easily manufactured gap size.
  • Fluids may be driven through a device either actively or passively. Fluids may be pumped using electric field, a centrifugal field, pressure-driven fluid flow, an electro-osmotic flow, or capillary action. In preferred embodiments, the average direction of the field will be parallel to the walls of the channel that includes the deterministic device.
  • Any of the following exemplary deterministic devices and methods may be incorporated into devices of the invention.
  • EXAMPLES Example 1 A Silicon Device Multiplexing 14 3-stage Array Duplexes
  • FIGS. 42A-42E show an exemplary device, characterized as follows.
  • Dimension: 90 mm×34 mm×1 mm
  • Array design: 3 stages, gap size=18, 12 and 8 μm for the first, second and third stage, respectively. Bifurcation ratio=1/10. Duplex; single bypass channel
  • Device design: multiplexing 14 array duplexes; flow resistors for flow stability
  • Device fabrication: The arrays and channels were fabricated in silicon using standard photolithography and deep silicon reactive etching techniques. The etch depth is 150 μm. Through holes for fluid access are made using KOH wet etching. The silicon substrate was sealed on the etched face to form enclosed fluidic channels using a blood compatible pressure sensitive adhesive (9795, 3M, St Paul, Minn.).
  • Device Packaging: The device was mechanically mated to a plastic manifold with external fluidic reservoirs to deliver blood and buffer to the device and extract the generated fractions.
  • Device Operation: An external pressure source was used to apply a pressure of 2.4 PSI to the buffer and blood reservoirs to modulate fluidic delivery and extraction from the packaged device.
  • Experimental conditions: human blood from consenting adult donors was collected into K2EDTA vacutainers (366643, Becton Dickinson, Franklin Lakes, N.J.). The undiluted blood was processed using the exemplary device described above (FIG. 42F) at room temperature and within 9 hrs of draw. Nucleated cells from the blood were separated from enucleated cells (red blood cells and platelets), and plasma delivered into a buffer stream of calcium and magnesium-free Dulbecco's Phosphate Buffered Saline (14190-144, Invitrogen, Carlsbad, Calif.) containing 1% Bovine Serum Albumin (BSA) (A8412-100ML, Sigma-Aldrich, St Louis, Mo.).
  • Measurement techniques: Complete blood counts were determined using a Coulter impedance hematology analyzer (COULTER® Ac•T diff™, Beckman Coulter, Fullerton, Calif.).
  • Performance: FIGS. 43A-43F shows typical histograms generated by the hematology analyzer from a blood sample and the waste (buffer, plasma, red blood cells, and platelets) and product (buffer and nucleated cells) fractions generated by the device. Table 1 shows the performance over 5 different blood samples:
    TABLE 1
    Performance
    Metrics
    Sample RBC Platelet WBC
    number Throughput removal removal loss
    1 4 mL/hr 100% 99% <1%
    2 6 mL/hr 100% 99% <1%
    3 6 mL/hr 100% 99% <1%
    4 6 mL/hr 100% 97% <1%
    5 6 mL/hr 100% 98% <1%
  • Example 2 A Silicon Device Multiplexing 14 Single-Stage Array Duplexes
  • FIGS. 44A-44D show an exemplary device, characterized as follows.
  • Dimension: 90 mm×34 mm×1 mm
  • Array design: 1 stage, gap size=24 μm. Bifurcation ratio=1/60. Duplex; double bypass channel
  • Device design: multiplexing 14 array duplexes; flow resistors for flow stability
  • Device fabrication: The arrays and channels were fabricated in silicon using standard photolithography and deep silicon reactive etching techniques. The etch depth is 150 μm. Through holes for fluid access are made using KOH wet etching. The silicon substrate was sealed on the etched face to form enclosed fluidic channels using a blood compatible pressure sensitive adhesive (9795, 3M, St Paul, Minn.).
  • Device Packaging: The device was mechanically mated to a plastic manifold with external fluidic reservoirs to deliver blood and buffer to the device and extract the generated fractions.
  • Device Operation: An external pressure source was used to apply a pressure of 2.4 PSI to the buffer and blood reservoirs to modulate fluidic delivery and extraction from the packaged device.
  • Experimental conditions: human blood from consenting adult donors was collected into K2EDTA vacutainers (366643, Becton Dickinson, Franklin Lakes, N.J.). The undiluted blood was processed using the exemplary device described above at room temperature and within 9 hrs of draw. Nucleated cells from the blood were separated from enucleated cells (red blood cells and platelets), and plasma delivered into a buffer stream of calcium and magnesium-free Dulbecco's Phosphate Buffered Saline (14190-144, Invitrogen, Carlsbad, Calif.) containing 1% Bovine Serum Albumin (BSA) (A8412-100ML, Sigma-Aldrich, St Louis, Mo.).
  • Measurement techniques: Complete blood counts were determined using a Coulter impedance hematology analyzer (COULTER® Ac•T diff™, Beckman Coulter, Fullerton, Calif.).
  • Performance: The device operated at 17 mL/hr and achieved >99% red blood cell removal, >95% nucleated cell retention, and >98% platelet removal.
  • Example 3 Separation of Fetal Cord Blood
  • FIG. 45 shows a schematic of the device used to separate nucleated cells from fetal cord blood.
  • Dimension: 100 mm×28 mm×1 mm
  • Array design: 3 stages, gap size=18, 12 and 8 μm for the first, second and third stage, respectively. Bifurcation ratio 1/10. Duplex; single bypass channel.
  • Device design: multiplexing 10 array duplexes; flow resistors for flow stability.
  • Device fabrication: The arrays and channels were fabricated in silicon using standard photolithography and deep silicon reactive etching techniques. The etch depth is 140 μm. Through holes for fluid access are made using KOH wet etching. The silicon substrate was sealed on the etched face to form enclosed fluidic channels using a blood compatible pressure sensitive adhesive (9795, 3M, St Paul, Minn.).
  • Device Packaging: The device was mechanically mated to a plastic manifold with external fluidic reservoirs to deliver blood and buffer to the device and extract the generated fractions.
  • Device Operation: An external pressure source was used to apply a pressure of 2.0 PSI to the buffer and blood reservoirs to modulate fluidic delivery and extraction from the packaged device.
  • Experimental conditions: Human fetal cord blood was drawn into phosphate buffered saline containing Acid Citrate Dextrose anticoagulants. 1 mL of blood was processed at 3 mL/hr using the device described above at room temperature and within 48 hrs of draw. Nucleated cells from the blood were separated from enucleated cells (red blood cells and platelets), and plasma delivered into a buffer stream of calcium and magnesium-free Dulbecco's Phosphate Buffered Saline (14190-144, Invitrogen, Carlsbad, Calif.) containing 1% Bovine Serum Albumin (BSA) (A8412-100ML, Sigma-Aldrich, St Louis, Mo.) and 2 mM EDTA (15575-020, Invitrogen, Carlsbad, Calif.).
  • Measurement techniques: Cell smears of the product and waste fractions (FIG. 46A-46B) were prepared and stained with modified Wright-Giemsa (WG16, Sigma Aldrich, St. Louis, Mo.).
  • Performance: Fetal nucleated red blood cells were observed in the product fraction (FIG. 46A) and absent from the waste fraction (FIG. 46B).
  • Example 4 Isolation of Fetal Cells from Maternal Blood
  • The device and process described in detail in Example 1 were used in combination with immunomagnetic affinity enrichment techniques to demonstrate the feasibility of isolating fetal cells from maternal blood.
  • Experimental conditions: blood from consenting maternal donors carrying male fetuses was collected into K2EDTA vacutainers (366643, Becton Dickinson, Franklin Lakes, N.J.) immediately following elective termination of pregnancy. The undiluted blood was processed using the device described in Example 1 at room temperature and within 9 hrs of draw. Nucleated cells from the blood were separated from enucleated cells (red blood cells and platelets), and plasma delivered into a buffer stream of calcium and magnesium-free Dulbecco's Phosphate Buffered Saline (14190-144, Invitrogen, Carlsbad, Calif.) containing 1% Bovine Serum Albumin (BSA) (A8412-100ML, Sigma-Aldrich, St Louis, Mo.). Subsequently, the nucleated cell fraction was labeled with anti-CD71 microbeads (130-046-201, Miltenyi Biotech Inc., Auburn, Calif.) and enriched using the MiniMACS™ MS column (130-042-201, Miltenyi Biotech Inc., Auburn, Calif.) according to the manufacturer's specifications. Finally, the CD71-positive fraction was spotted onto glass slides.
  • Measurement techniques: Spotted slides were stained using fluorescence in situ hybridization (FISH) techniques according to the manufacturer's specifications using Vysis probes (Abbott Laboratories, Downer's Grove, Ill.). Samples were stained from the presence of X and Y chromosomes. In one case, a sample prepared from a known trisomy 21 pregnancy was also stained for chromosome 21.
  • Performance: Isolation of fetal cells was confirmed by the reliable presence of male cells in the CD71-positive population prepared from the nucleated cell fractions (FIG. 47). In the single abnormal case tested, the trisomy 21 pathology was also identified (FIG. 48).
  • The following examples show specific embodiments of devices. The description for each device provides the number of stages in series, the gap size for each stage, ε (Flow Angle), and the number of channels per device (Arrays/Chip). Each device was fabricated out of silicon using DRIE, and each device had a thermal oxide layer.
  • Example 5
  • This device includes five stages in a single array.
    • Array Design: 5 stage, asymmetric array
    • Gap Sizes: Stage 1: 8 μm
    • Stage 2: 10 μm
      • Stage 3: 12 μm
      • Stage 4: 14 μm
      • Stage 5: 16 μm
    • FlowAngle: 1/10
    • Arrays/Chip: 1
    Example 6
  • This device includes the stages, where each stage is a duplex having a bypass channel. The height of the device was 125 μm.
    • Array Design: symmetric 3 stage array with central collection channel
    • Gap Sizes: Stage 1: 8 μm
    • Stage 2: 12 μm
    • Stage 3: 18 μm
    • Stage 4:
    • Stage 5:
    • Flow Angle: 1/10
    • Arrays/Chip: 1
    • Other: central collection channel
  • FIG. 49A shows the mask employed to fabricate the device. FIGS. B1B-B1D are enlargements of the portions of the mask that define the inlet, array, and outlet. FIGS. 50A-50G show SEMs of the actual device.
  • Example 7
  • This device includes the stages, where each stage is a duplex having a bypass channel. “Fins” were designed to flank the bypass channel to keep fluid from the bypass channel from re-entering the array. The chip also included on-chip flow resistors, i.e., the inlets and outlets possessed greater fluidic resistance than the array. The height of the device was 117 μm.
    • Array Design: 3 stage symmetric array
    • Gap Sizes: Stage 1: 8 μm
      • Stage 2: 12 μm
      • Stage 3: 18 μm
      • Stage 4:
      • Stage 5:
    • Flow Angle: 1/10
    • Arrays/Chip: 10
      Other: large fin central collection channel on-chip flow resistors
  • FIG. 51 A shows the mask employed to fabricate the device. FIGS. 51B-51D are enlargements of the portions of the mask that define the inlet, array, and outlet. FIGS. 52A-52F show SEMs of the actual device.
  • Example 8
  • This device includes the stages, where each stage is a duplex having a bypass channel. “Fins” were designed to flank the bypass channel to keep fluid from the bypass channel from re-entering the array. The edge of the fin closest to the array was designed to mimic the shape of the array. The chip also included on-chip flow resistors, i.e., the inlets and outlets possessed greater fluidic resistance than the array. The height of the device was 138 μm.
    Array Design: 3 stage symmetric array
    Gap Sizes: Stage 1: 8 μm
    Stage 2: 12 μm
    Stage 3: 18 μm
    Stage 4:
    Stage 5:
    Flow Angle: 1/10
    Arrays/Chip: 10
    Other: alternate large fin central collection channel
    on-chip flow resistors
  • FIG. 45A shows the mask employed to fabricate the device. FIGS. 45B-45D are enlargements of the portions of the mask that define the inlet, array, and outlet. FIGS. 532A-532F show SEMs of the actual device.
  • Example 9
  • This device includes the stages, where each stage is a duplex having a bypass channel. “Fins” were optimized using Femlab to flank the bypass channel to keep fluid from the bypass channel from re-entering the array. The edge of the fin closest to the array was designed to mimic the shape of the array. The chip also included on-chip flow resistors, i.e., the inlets and outlets possessed greater fluidic resistance than the array. The height of the device was 139 or 142 μm.
    • Array Design: 3 stage symmetric array
    • Gap Sizes: Stage 1: 8 μm
      • Stage 2: 12 μm
      • Stage 3: 18 μm
      • Stage 4:
      • Stage 5:
    • Flow Angle: 1/10
    • Arrays/Chip: 10
    • Other: Femlab optimized central collection channel (Femiab I) on-chip flow resistors
  • FIG. 54A shows the mask employed to fabricate the device. FIGS. 54B-E1D are enlargements of the portions of the mask that define the inlet, array, and outlet. FIGS. 55A-55S show SEMs of the actual device.
  • Example 10
  • This device includes a single stage, duplex device having a bypass channel disposed to receive output from the ends of both arrays. The obstacles in this device are elliptical. The array boundary was modeled in Femlab to. The chip also included on-chip flow resistors, i.e., the inlets and outlets possessed greater fluidic resistance than the array. The height of the device was 152 μm.
    • Array Design: single stage symmetric array
    • Gap Sizes: Stage 1: 24 μm
    • Stage 2:
      • Stage 3:
      • Stage 4:
      • Stage 5:
    • Flow Angle: 1/60
    • Arrays/Chip: 14
    • Other: central barrier
      • ellipsoid posts
      • on-chip resistors
      • Femlab modeled array boundary
  • FIG. 44A shows the mask employed to fabricate the device. FIGS. 44B-44D are enlargements of the portions of the mask that define the inlet, array, and outlet. FIGS. 56A-56C show SEMs of the actual device.
  • Example 11
  • Deterministic devices incorporated into devices of the invention were designed by computer-aided design (CAD) and microfabricated by photolithography. A two-step process was developed in which a blood sample is first debulked to remove the large population of small cells and then the rare target epithelial cells target cells are recovered by immunoaffinity capture. The devices were defined by photolithography and etched into a silicon substrate based on a CAD-generated design. The cell enrichment module, which is approximately the size of a standard microscope slide, contains 14 parallel sample processing sections and associated sample handling channels that connect to common sample and buffer inlets and product and waste outlets. Each section contains an array of microfabricated obstacles that is optimized to separate the target cell type by size via displacement of the larger cells into the product stream. In this example, the microchip was designed to separate red blood cells (RBCs) and platelets from the larger leukocytes and circulating tumor cells. Enriched populations of target cells were recovered from whole blood passed through the device. Performance of the cell enrichment microchip was evaluated by separating RBCs and platelets from white blood cells (WBCs) in normal whole blood (FIG. 67). In cancer patients, circulating tumor cells are found in the larger WBC fraction. Blood was minimally diluted (30%), and a 6 ml sample was processed at a flow rate of up to 6 ml/hr. The product and waste stream were evaluated in a Coulter Model “AC-T diff” clinical blood analyzer, which automatically distinguishes, sizes and counts different blood cell populations. The enrichment chip achieved separation of RBCs from WBCs, in which the WBC fraction had >99% retention of nucleated cells, >99% depletion of RBCs and >97% depletion of platelets. Representative histograms of these cell fractions are shown in FIG. 68. Routine cytology confirmed the high degree of enrichment of the WBC RBC fractions (FIG. 69).
  • Next, epithelial cells were recovered by affinity capture in a microfluidic module that is functionalized with immobilized antibody. A capture module with a single chamber containing a regular array of antibody-coated microfabricated obstacles was designed. These obstacles are disposed to maximize cell capture by increasing the capture area approximately four-fold, and by slowing the flow of cells under laminar flow adjacent to the obstacles to increase the contact time between the cells and the immobilized antibody. The capture modules can be operated under conditions of relatively high flow rate but low shear to protect cells against damage. The surface of the capture module was functionalized by sequential treatment with 10% silane, 0.5% gluteraldehyde and avidin, followed by biotinylated anti-EpCAM. Active sites were blocked with 3% bovine serum albumin in PBS, quenched with dilute Tris HCl and stabilized with dilute L-histidine. Modules were washed in PBS after each stage and finally dried and stored at room temperature. Capture performance was measured with the human advanced lung cancer cell line NCI-H1650 (ATCC Number CRL-5883). This cell line has a heterozygous 15 bp in-frame deletion in exon 19 of EGFR that renders it susceptible to gefitinib. Cells from confluent cultures were harvested with trypsin, stained with the vital dye Cell Tracker Orange (CMRA reagent, Molecular Probes, Eugene, Oreg.), resuspended in fresh whole blood and fractionated in the microfluidic chip at various flow rates. In these initial feasibility experiments, cell suspensions were processed directly in the capture modules without prior fractionation in the cell enrichment module to debulk the red blood cells; hence, the sample stream contained normal blood red cells and leukocytes as well as tumor cells. After the cells were processed in the capture module, the device was washed with buffer at a higher flow rate (3 ml/hr) to remove the nonspecifically bound cells. The adhesive top was removed and the adherent cells were fixed on the chip with paraformaldehyde and observed by fluorescence microscopy. Cell recovery was calculated from hemacytometer counts; representative capture results are shown in Table 2. Initial yields in reconstitution studies with unfractionated blood were greater than 60% with less than 5% of non-specific binding.
    TABLE 2
    Run Avg. flow Length of No. cells No. cells
    number rate run processed captured Yield
    1 3.0 1 hr 150,000 38,012 25%
    2 1.5 2 hr 150,000 30,000/ml 60%
    3 1.08 2 hr 108,000 68,661 64%
    4 1.21 2 hr 121,000 75,491 62%
  • Next, NCI-H1650 cells that were spiked into whole blood and recovered by size fractionation and affinity capture as described above were successfully analyzed in situ. In a trial run to distinguish epithelial cells from leukocytes, 0.5 ml of a stock solution of fluorescein-labeled CD45 pan-leukocyte monoclonal antibody was passed into the capture module and incubated at room temperature for 30 minutes. The module was washed with buffer to remove unbound antibody and the cells were fixed on the chip with 1% paraformaldehyde and observed by fluorescence microscopy. As shown in FIG. 70, the epithelial cells were bound to the obstacles and floor of the capture module. Background staining of the flow passages with CD45 pan-leukocyte antibody is visible, as are several stained leukocytes, apparently due to a low level of non-specific capture.
  • Example 12 Device Embodiments
  • A design for preferred deterministic device is shown in FIG. 73A, and parameters corresponding to three preferred device embodiments associated with this design are shown in FIG. 73B. These embodiments are particularly useful for separating epithelial cells from blood.
  • Example 13 PCR Assay for EGFR Mutations
  • A blood sample from a cancer patient is processed and analyzed using the devices and methods of Example 11, resulting in an enriched sample of epithelial cells containing CTCs. This sample is then analyzed to identify potential EGFR mutations.
  • To perform this analysis, genomic DNA is isolated from the target cells present in the enriched sample and amplified for use in allele-specific Real Time PCR assays. Since all EGFR mutations in NSC lung cancer reported to date that are known to confer sensitivity or resistance to gefitinib lie within the coding regions of exons 18 to 21, each of these four exons is PCR-amplified with a unique set of exon-specific primers. Next, multiplexed allele-specific quantitative PCR reactions are performed using the TaqMan 5′ nuclease assay PCR system (Applied Biosystems) and a model 7300 Applied Biosystems Real Time PCR machine. This allows the rapid identification of any of the known clinically relevant mutations.
  • A two-step PCR protocol is required for this method. First, exons 18 through 21 are amplified in standard PCR reactions. The resultant PCR products are split into separate aliquots for use in allele-specific multiplexed Real Time PCR assays. The initial PCR reactions are stopped during the log phase in order to minimize possible loss of allele-specific information during amplification. Next, a second round of PCR amplifies subregions of the initial PCR product specific to each mutation of interest. Given the very high sensitivity of Real Time PCR, it is possible to obtain complete information on the mutation status of the EGFR gene even if as few as 10 CTCs are isolated. Real Time PCR provides quantification of allelic sequences over 8 logs of input DNA concentrations; thus, even heterozygous mutations in impure populations are easily detected using this method.
  • Oligonucleotides are designed using the primer optimization software program Primer Express (Applied Biosystems), and hybridization conditions are optimized to distinguish the wild type EGFR DNA sequence from mutant alleles. EGFR genomic DNA amplified from lung cancer cell lines that are known to carry EGFR mutations, such as H358 (wild type), H1650 (15-bp deletion, Δ2235-2249), and H1975 (two point mutations, 2369 C→T, 2573 T→G), is used to optimize the allele-specific Real Time PCR reactions. Using the TaqMan 5′ nuclease assay, allele-specific labeled probes specific for wild type sequence or for known EGFR mutations are developed. The oligonucleotides are designed to have melting temperatures that easily distinguish a match from a mismatch, and the Real Time PCR conditions are optimized to distinguish wild type and mutant alleles. All Real Time PCR reactions are carried out in triplicate.
  • Initially, labeled probes containing wild type sequence are multiplexed in the same reaction with a single mutant probe. Expressing the results as a ratio of one mutant allele sequence versus wild type sequence can identify samples containing or lacking a given mutation. After conditions are optimized for a given probe set, it is then possible to multiplex probes for all of the mutant alleles within a given exon within the same Real Time PCR assay, increasing the ease of use of this analytical tool in clinical settings.
  • The purity of the input sample of CTCs may vary, and the mutation status of the isolated CTCs may be heterogeneous. Nevertheless, the extremely high sensitivity of Real Time PCR enables the identification any and all mutant sequences present.
  • Example 14 Determining Counts for Non-Epithelial Cell Types
  • Using the methods of the invention, one may make a diagnosis based on counting cell types other than epithelial cells. A diagnosis of the absence, presence, or progression of cancer may be based on the number of cells in a cellular sample that are larger than a particular cutoff size. For example, cells with a hydrodynamic cell diameter of 14 microns or larger may be selected. This cutoff size would eliminate most leukocytes. The nature of these cells may then be determined by downstream molecular or cytological analysis.
  • Cell types other than epithelial cells that would be useful to analyze include endothelial cells, endothelial progenitor cells, endometrial cells, or trophoblasts indicative of a disease state. Furthermore, determining separate counts for epithelial cells and other cell types, followed by a determination of the ratios between the number of epithelial cells and the number of other cell types, may provide useful diagnostic information.
  • A deterministic device may be configured to isolate targeted subpopulations of cells such as those described above, as shown in FIG. 71A-D. A size cutoff may be selected such that most native blood cells, including red blood cells, white blood cells, and platelets, flow to waste, while non-native cells, which could include endothelial cells, endothelial progenitor cells, endometrial cells, or trophoblasts, are collected in an enriched sample. This enriched sample may be further analyzed.
  • Using a deterministic device, therefore, it is possible to isolate a subpopulation of cells from blood or other bodily fluids based on size, which conveniently allows for the elimination of a large proportion of native blood cells when large cell types are targeted. As shown schematically in FIG. 72, a deterministic device may include counting means to determine the number of cells in the enriched sample, and further analysis of the cells in the enriched sample may provide additional information that is useful for diagnostic or other purposes.
  • Example 15 Enrichment of Fetal Nucleated Red Blood Cells from Maternal Blood
  • For this example, the device includes a deterministic separation component, as described herein, capable of separated fetal nucleated red blood cells and maternal white blood cells from maternal enucleated red blood cells. The deterministic component is connected to a reservoir containing sodium nitrite. A maternal blood sample, e.g., that has been diluted, is introduced into the device to produce a fraction enriched in fetal red blood cells and depleted of maternal red blood cells. This sample is directed into the reservoir where the sodium nitrite oxidizes the fetal heme iron, thereby increasing the magnetic responsiveness of the fetal red blood cells. A magnetic field is then applied, e.g., via a MACS column, and the altered fetal red blood cells bind to the magnet, while maternal white blood cells are not bound by the magnet. Removing the white blood cells, e.g., by a rinse, and then eliminating the magnetic field allows recovery of the fetal red blood cells, e.g., for analysis, storage, or further manipulation.
  • Other Embodiments
  • All publications, patents, and patent applications mentioned in the above specification are hereby incorporated by reference. Various modifications and variations of the described method and system of the invention will be apparent to those skilled in the art without departing from the scope and spirit of the invention. Although the invention has been described in connection with specific embodiments, it should be understood that the invention as claimed should not be unduly limited to such specific embodiments. Indeed, various modifications of the described modes for carrying out the invention that are obvious to those skilled in the art are intended to be within the scope of the invention.
  • Other embodiments are in the claims.

Claims (43)

1. A device for producing a sample enriched in an analyte, said device comprising:
(a) a first channel comprising a structure that deterministically deflects particles having a hydrodynamic size above a critical size in a direction not parallel to the average direction of flow in said structure, wherein said particles are analyte particles or are a non-analyte component of said sample; and
(b) a reservoir fluidly coupled to an output of said first channel through which said analyte passes into said reservoir, wherein said reservoir comprises a reagent that alters a magnetic property of said analyte.
2. The device of claim 1, wherein said first channel is a microfluidic channel.
3. The device of claim 1, wherein said structure comprises an array of obstacles that form a network of gaps, wherein a fluid passing through said gaps is divided unequally into a major flux and a minor flux so that the average direction of the major flux is not parallel to the average direction of fluidic flow in said channel.
4. The device of claim 3, wherein said array of obstacles comprises first and second rows, wherein the second row is displaced laterally relative to the first row so that fluid passing through a gap in the first row is divided unequally into two gaps in the second row.
5. The device of claim 1, wherein said analyte has a hydrodynamic size greater than said critical size.
6. The device of claim 1, wherein said analyte has a hydrodynamic size smaller than said critical size.
7. The device of claim 1, further comprising a magnetic force generator capable of generating a magnetic field.
8. The device of claim 7, wherein said magnetic force generator comprises a region of magnetic obstacles disposed in a second channel.
9. The device of claim 8, wherein at least a portion of said magnetic obstacles comprise a permanent magnet.
10. The device of claim 8, wherein at least a portion of said magnetic obstacles comprise a non-permanent magnet.
11. The device of claim 8, wherein said obstacles are ordered in a two-dimensional array.
12. The device of claim 8, wherein said second channel is a microfluidic channel.
13. The device of claim 1, wherein said reservoir further comprises a second channel comprising a magnet.
14. The device of claim 1, wherein said reagent alters an intrinsic magnetic property of said one or more analytes.
15. The device of claim 14, wherein said reagent comprises sodium nitrite.
16. The device of claim 1, wherein said reagent binds to said one or more analytes.
17. The device of claim 16, wherein said reagent comprises a magnetic particle.
18. The device of claim 17, wherein said magnetic particle comprises an antibody or an antigen-binding fragment thereof.
19. The device of claim 18, wherein said antibody is anti-CD71, anti-CD36, anti-CD45, anti-GPA, anti-antigen i, anti-CD34, or anti-fetal hemoglobin.
20. The device of claim 16, wherein said reagent comprises holo-transferrin.
21. A method for producing a sample enriched in a first analyte relative to a second analyte, said method comprising:
(a) applying at least a portion of said sample to a device comprising a structure that deterministically deflects particles having a hydrodynamic size above a critical size in a direction not parallel to the average direction of flow in said structure, thereby producing a second sample enriched in said first analyte and comprising said second analyte;
(b) combining said second sample with a reagent that alters a magnetic property of said first analyte to produce an altered first analyte; and
(c) applying a magnetic field to said second sample, wherein said magnetic field generates a differential force to physically separate said altered first analyte from said second analyte, thereby producing a sample enriched in said first analyte.
22. The method of claim 21, wherein said reagent binds to said first analyte.
23. The method of claim 21, wherein said reagent alters an intrinsic magnetic property of said first analyte.
24. The method of claim 23, wherein said reagent comprises sodium nitrite.
25. The method of claim 21, wherein said reagent comprises a magnetic particle that binds to or is incorporated into said first analyte.
26. The method of claim 25, wherein said magnetic particle comprises an antibody or an antigen-binding fragment thereof.
27. The method of claim 26, wherein said antibody is anti-CD71, anti-GPA, anti-antigen i, anti-CD45, anti-CD34, or anti-fetal hemoglobin.
28. The method of claim 21, wherein said analyte has a hydrodynamic size greater than said critical size.
29. The method of claim 21, wherein said analyte has a hydrodynamic size smaller than said critical size.
30. The method of claim 21, wherein said sample comprises a maternal blood sample.
31. The method of claim 21, wherein said first analyte is a cell, an organelle, or a virus.
32. The method of claim 31, wherein said cell is a bacterial cell, a fetal cell, or a blood cell.
33. The method of claim 32, wherein said blood cell is a fetal red blood cell.
34. The method of claim 31, wherein said organelle is a nucleus.
35. A method of producing a sample enriched in red blood cells relative to a second blood component, said method comprising:
(a) contacting a sample comprising red blood cells with a reagent that oxidizes iron to produce oxidized hemoglobin; and
(b) applying a magnetic field to said sample, wherein said red blood cells having oxidized hemoglobin are attracted to said magnetic field to a greater extent than said second blood component, thereby producing said sample enriched in said red blood cells.
36. The method of claim 35, wherein said red blood cells are fetal red blood cells.
37. The method of claim 36, wherein said second blood component is a maternal blood cell.
38. The method of claim 35, wherein prior to said step (a), said sample is enriched for said red blood cells.
39. The method of claim 38, wherein said enriching is performed by applying at least a portion of said sample to a device comprising a structure that deterministically deflects particles having a hydrodynamic size above a critical size in a direction not parallel to the average direction of flow in said structure.
40. The method of claim 39, wherein fetal red blood cells are enriched relative to maternal red blood cells.
41. A device for producing a sample enriched in red blood cells, said device comprising:
(a) an analytical device that enriches said red blood cells based on size, shape, deformability, or affinity; and
(b) a reservoir comprising a reagent that oxidizes iron, wherein said reagent increases the magnetic responsiveness of said red blood cells.
42. The device of claim 41, wherein said analytical device comprises a first channel comprising a structure that deterministically deflects particles having a hydrodynamic size above a critical size in a direction not parallel to the average direction of flow in said structure.
43. The device of claim 41, wherein said reagent is sodium nitrite.
US11/227,904 2005-04-05 2005-09-15 Devices and methods for enrichment and alteration of cells and other particles Abandoned US20070196820A1 (en)

Priority Applications (9)

Application Number Priority Date Filing Date Title
US11/227,904 US20070196820A1 (en) 2005-04-05 2005-09-15 Devices and methods for enrichment and alteration of cells and other particles
US11/323,971 US20060223178A1 (en) 2005-04-05 2005-12-29 Devices and methods for magnetic enrichment of cells and other particles
AU2006292490A AU2006292490A1 (en) 2005-09-15 2006-09-15 Devices and methods for magnetic enrichment of cells and other particles
EP06814753A EP1934326A2 (en) 2005-09-15 2006-09-15 Devices and methods for magnetic enrichment of cells and other particles
CNA2006800416665A CN101305087A (en) 2005-09-15 2006-09-15 Devices and methods for magnetic enrichment of cells and other particles
CA002622779A CA2622779A1 (en) 2005-09-15 2006-09-15 Devices and methods for magnetic enrichment of cells and other particles
JP2008531362A JP2009511001A (en) 2005-09-15 2006-09-15 Device and method for magnetic concentration of cells and other particles
PCT/US2006/036061 WO2007035498A2 (en) 2005-09-15 2006-09-15 Devices and methods for magnetic enrichment of cells and other particles
IL190091A IL190091A0 (en) 2005-09-15 2008-03-11 Devices and methods for magnetic enrichment of cells and other particles

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US66841505P 2005-04-05 2005-04-05
US70406705P 2005-07-29 2005-07-29
US11/227,904 US20070196820A1 (en) 2005-04-05 2005-09-15 Devices and methods for enrichment and alteration of cells and other particles

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US11/323,971 Continuation-In-Part US20060223178A1 (en) 2005-04-05 2005-12-29 Devices and methods for magnetic enrichment of cells and other particles

Publications (1)

Publication Number Publication Date
US20070196820A1 true US20070196820A1 (en) 2007-08-23

Family

ID=37071048

Family Applications (7)

Application Number Title Priority Date Filing Date
US11/227,904 Abandoned US20070196820A1 (en) 2005-04-05 2005-09-15 Devices and methods for enrichment and alteration of cells and other particles
US11/449,149 Active 2027-12-28 US8021614B2 (en) 2005-04-05 2006-06-08 Devices and methods for enrichment and alteration of cells and other particles
US13/232,781 Active 2028-01-30 US9174222B2 (en) 2005-04-05 2011-09-14 Devices and method for enrichment and alteration of cells and other particles
US13/452,054 Active US8585971B2 (en) 2005-04-05 2012-04-20 Devices and method for enrichment and alteration of cells and other particles
US14/930,313 Active 2026-12-01 US9956562B2 (en) 2005-04-05 2015-11-02 Devices and method for enrichment and alteration of cells and other particles
US15/965,128 Active US10786817B2 (en) 2005-04-05 2018-04-27 Devices and method for enrichment and alteration of cells and other particles
US17/021,100 Pending US20210178403A1 (en) 2005-04-05 2020-09-15 Devices And Method For Enrichment And Alteration Of Cells And Other Particles

Family Applications After (6)

Application Number Title Priority Date Filing Date
US11/449,149 Active 2027-12-28 US8021614B2 (en) 2005-04-05 2006-06-08 Devices and methods for enrichment and alteration of cells and other particles
US13/232,781 Active 2028-01-30 US9174222B2 (en) 2005-04-05 2011-09-14 Devices and method for enrichment and alteration of cells and other particles
US13/452,054 Active US8585971B2 (en) 2005-04-05 2012-04-20 Devices and method for enrichment and alteration of cells and other particles
US14/930,313 Active 2026-12-01 US9956562B2 (en) 2005-04-05 2015-11-02 Devices and method for enrichment and alteration of cells and other particles
US15/965,128 Active US10786817B2 (en) 2005-04-05 2018-04-27 Devices and method for enrichment and alteration of cells and other particles
US17/021,100 Pending US20210178403A1 (en) 2005-04-05 2020-09-15 Devices And Method For Enrichment And Alteration Of Cells And Other Particles

Country Status (9)

Country Link
US (7) US20070196820A1 (en)
EP (3) EP2492339B1 (en)
JP (1) JP2008538283A (en)
CN (1) CN101918527B (en)
AU (1) AU2006232103A1 (en)
CA (1) CA2601480A1 (en)
GB (1) GB2429774A (en)
HK (1) HK1175494A1 (en)
WO (1) WO2006108101A2 (en)

Cited By (46)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060160243A1 (en) 2005-01-18 2006-07-20 Biocept, Inc. Recovery of rare cells using a microchannel apparatus with patterned posts
US20060223178A1 (en) * 2005-04-05 2006-10-05 Tom Barber Devices and methods for magnetic enrichment of cells and other particles
US20060252087A1 (en) * 2005-01-18 2006-11-09 Biocept, Inc. Recovery of rare cells using a microchannel apparatus with patterned posts
US20070161051A1 (en) * 2006-01-12 2007-07-12 Biocept, Inc. Device for cell separation and analysis and method of using
US20070172903A1 (en) * 2002-09-27 2007-07-26 The General Hospital Corporation Microfluidic device for cell separation and uses thereof
WO2008130655A1 (en) * 2007-04-20 2008-10-30 The General Hospital Corporation Methods for counting cells
US20090014360A1 (en) * 2007-04-16 2009-01-15 The General Hospital Corporation D/B/A Massachusetts General Hospital Systems and methods for particle focusing in microchannels
US20090136982A1 (en) * 2005-01-18 2009-05-28 Biocept, Inc. Cell separation using microchannel having patterned posts
WO2010041231A2 (en) 2008-10-10 2010-04-15 Cnrs-Dae Cell sorting device
US20100304978A1 (en) * 2009-01-26 2010-12-02 David Xingfei Deng Methods and compositions for identifying a fetal cell
WO2011071912A1 (en) * 2009-12-07 2011-06-16 Yale University Label-free cellular manipulation and sorting via biocompatible ferrofluids
US8021614B2 (en) 2005-04-05 2011-09-20 The General Hospital Corporation Devices and methods for enrichment and alteration of cells and other particles
US8137912B2 (en) 2006-06-14 2012-03-20 The General Hospital Corporation Methods for the diagnosis of fetal abnormalities
WO2012048113A2 (en) 2010-10-07 2012-04-12 The General Hospital Corporation Biomarkers of cancer
US8162149B1 (en) 2009-01-21 2012-04-24 Sandia Corporation Particle sorter comprising a fluid displacer in a closed-loop fluid circuit
US8168389B2 (en) 2006-06-14 2012-05-01 The General Hospital Corporation Fetal cell analysis using sample splitting
US8195415B2 (en) 2008-09-20 2012-06-05 The Board Of Trustees Of The Leland Stanford Junior University Noninvasive diagnosis of fetal aneuploidy by sequencing
WO2013063035A1 (en) 2011-10-24 2013-05-02 The General Hospital Corporation Biomarkers of cancer
US20130302797A1 (en) * 2007-04-16 2013-11-14 Anne R. Kopf-Sill Methods for diagnosing, prognosing, or theranosing a condition using rare cells
US9217131B2 (en) 2010-01-21 2015-12-22 Biocep Ltd. Magnetic separation of rare cells
WO2016073448A1 (en) 2014-11-03 2016-05-12 The General Hospital Corporation Combined sorting and concentrating particles in a microfluidic device
CN106053456A (en) * 2016-06-24 2016-10-26 许毅 Urine concentrating device
US20160320389A1 (en) * 2015-04-30 2016-11-03 International Business Machines Corporation Immunoassay for Detection of Virus-Antibody Nanocomplexes in Solution by Chip-Based Pillar Array
AU2015268583B2 (en) * 2010-10-28 2017-06-15 Yale University Microfluidic Processing Of Target Species In Ferrofluids
US20180154319A1 (en) * 2016-12-01 2018-06-07 Imagine Tf, Llc Microstructure flow mixing devices
US9999855B2 (en) 2010-10-28 2018-06-19 Yale University Microfluidic processing of target species in ferrofluids
US10203332B2 (en) 2014-08-22 2019-02-12 Samsung Electronics Co., Ltd. Apparatus for and method of separating target matter
WO2019046052A1 (en) 2017-09-01 2019-03-07 Gpb Scientific, Llc Methods for preparing therapeutically active cells using microfluidics
US10324011B2 (en) 2013-03-15 2019-06-18 The Trustees Of Princeton University Methods and devices for high throughput purification
US10359429B2 (en) 2008-02-25 2019-07-23 Gpb Scientific, Llc Tagged ligands for enrichment of rare analytes from a mixed sample
US10369568B2 (en) 2005-01-18 2019-08-06 Biocept, Inc. Cell separation using microchannel having patterned posts
US10386276B2 (en) * 2016-09-20 2019-08-20 International Business Machines Corporation Phosphoprotein detection using a chip-based pillar array
US10591391B2 (en) 2006-06-14 2020-03-17 Verinata Health, Inc. Diagnosis of fetal abnormalities using polymorphisms including short tandem repeats
WO2020097048A1 (en) * 2018-11-05 2020-05-14 Micromedicine, Inc. Systems and methods for sorting particles using hydrodynamic sizing
US10677779B2 (en) * 2015-02-24 2020-06-09 Korea University Research And Business Foundation Platelet test chip
US10704090B2 (en) 2006-06-14 2020-07-07 Verinata Health, Inc. Fetal aneuploidy detection by sequencing
WO2021011907A1 (en) 2019-07-18 2021-01-21 Gpb Scientific, Inc. Ordered processing of blood products to produce therapeutically active cells
US10976232B2 (en) 2015-08-24 2021-04-13 Gpb Scientific, Inc. Methods and devices for multi-step cell purification and concentration
WO2021133897A1 (en) 2019-12-28 2021-07-01 Gpb Scientific, Inc. Microfluidic cartridges for processing particles and cells
US11142746B2 (en) 2013-03-15 2021-10-12 University Of Maryland, Baltimore High efficiency microfluidic purification of stem cells to improve transplants
US11162065B2 (en) 2015-06-05 2021-11-02 Novartis Ag Flow-through paramagnetic particle-based cell separation and paramagnetic particle removal
US11204350B2 (en) 2013-03-15 2021-12-21 Ancera, Llc Systems and methods for bead-based assays in ferrofluids
US11285490B2 (en) 2015-06-26 2022-03-29 Ancera, Llc Background defocusing and clearing in ferrofluid-based capture assays
US11383247B2 (en) 2013-03-15 2022-07-12 Ancera, Llc Systems and methods for active particle separation
US11493428B2 (en) 2013-03-15 2022-11-08 Gpb Scientific, Inc. On-chip microfluidic processing of particles
US11768137B2 (en) 2017-04-14 2023-09-26 Ventana Medical Systems, Inc. Size-based separation of dissociated fixed tissues

Families Citing this family (164)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6692952B1 (en) * 1999-11-10 2004-02-17 Massachusetts Institute Of Technology Cell analysis and sorting apparatus for manipulation of cells
US6913697B2 (en) 2001-02-14 2005-07-05 Science & Technology Corporation @ Unm Nanostructured separation and analysis devices for biological membranes
EP1604184A4 (en) * 2003-02-27 2010-10-27 Stephen A Lesko Standardized evaluation of therapeutic efficacy based on cellular biomarkers
EP1636564A1 (en) * 2003-06-13 2006-03-22 The General Hospital Corporation Microfluidic systems for size based removal of red blood cells and platelets from blood
US20070026417A1 (en) * 2005-07-29 2007-02-01 Martin Fuchs Devices and methods for enrichment and alteration of circulating tumor cells and other particles
US20070026414A1 (en) * 2005-07-29 2007-02-01 Martin Fuchs Devices and methods for enrichment and alteration of circulating tumor cells and other particles
EP1874920A4 (en) * 2005-04-05 2009-11-04 Cellpoint Diagnostics Devices and methods for enrichment and alteration of circulating tumor cells and other particles
US20070026415A1 (en) * 2005-07-29 2007-02-01 Martin Fuchs Devices and methods for enrichment and alteration of circulating tumor cells and other particles
US20070026413A1 (en) * 2005-07-29 2007-02-01 Mehmet Toner Devices and methods for enrichment and alteration of circulating tumor cells and other particles
US20070059680A1 (en) * 2005-09-15 2007-03-15 Ravi Kapur System for cell enrichment
US20090181421A1 (en) * 2005-07-29 2009-07-16 Ravi Kapur Diagnosis of fetal abnormalities using nucleated red blood cells
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
US20070026416A1 (en) * 2005-07-29 2007-02-01 Martin Fuchs Devices and methods for enrichment and alteration of circulating tumor cells and other particles
US20070059719A1 (en) * 2005-09-15 2007-03-15 Michael Grisham Business methods for prenatal Diagnosis
US20070059716A1 (en) * 2005-09-15 2007-03-15 Ulysses Balis Methods for detecting fetal abnormality
US20070059683A1 (en) * 2005-09-15 2007-03-15 Tom Barber Veterinary diagnostic system
US20070059718A1 (en) * 2005-09-15 2007-03-15 Mehmet Toner Systems and methods for enrichment of analytes
US20070059774A1 (en) * 2005-09-15 2007-03-15 Michael Grisham Kits for Prenatal Testing
US20070059781A1 (en) * 2005-09-15 2007-03-15 Ravi Kapur System for size based separation and analysis
EP2385143B1 (en) 2006-02-02 2016-07-13 The Board of Trustees of the Leland Stanford Junior University Non-invasive fetal genetic screening by digital analysis
BRPI0711896A2 (en) 2006-05-22 2012-07-17 Univ Columbia microfluid membrane-free exchange systems and methods using filtration of fluid extraction stream extraction.
US20080124721A1 (en) * 2006-06-14 2008-05-29 Martin Fuchs Analysis of rare cell-enriched samples
US20080007838A1 (en) * 2006-07-07 2008-01-10 Omnitech Partners, Inc. Field-of-view indicator, and optical system and associated method employing the same
US20100288689A1 (en) * 2006-08-22 2010-11-18 Agency For Science, Technology And Research Microfluidic filtration unit, device and methods thereof
US20100112590A1 (en) 2007-07-23 2010-05-06 The Chinese University Of Hong Kong Diagnosing Fetal Chromosomal Aneuploidy Using Genomic Sequencing With Enrichment
HUE054639T2 (en) 2007-07-23 2021-09-28 Univ Hong Kong Chinese Determining a nucleic acid sequence imbalance
US20090053799A1 (en) * 2007-08-23 2009-02-26 Cynvenio Biosystems, Llc Trapping magnetic sorting system for target species
US20110300608A1 (en) * 2007-09-21 2011-12-08 Streck, Inc. Nucleic acid isolation in preserved whole blood
US20110143340A1 (en) * 2007-11-01 2011-06-16 Biocept, Inc. Non-invasive isolation of fetal nucleic acid
US8496606B2 (en) * 2008-02-04 2013-07-30 The Trustees Of Columbia University In The City Of New York Fluid separation devices, systems and methods
WO2009117611A2 (en) * 2008-03-19 2009-09-24 Cynvenio Biosystems, Llc Trapping magnetic cell sorting system
US20110137018A1 (en) * 2008-04-16 2011-06-09 Cynvenio Biosystems, Inc. Magnetic separation system with pre and post processing modules
FR2931141B1 (en) 2008-05-13 2011-07-01 Commissariat Energie Atomique MICROFLUIDIC SYSTEM AND METHOD FOR THE SORTING OF AMAS FROM CELLS AND PREFERENCE FOR CONTINUOUS ENCAPSULATION THROUGH THEIR SORTING
FR2931085B1 (en) 2008-05-13 2011-05-27 Commissariat Energie Atomique METHOD OF SORTING PARTICLES OR AMAS FROM PARTICLES IN A FLUID CIRCULATING IN A CHANNEL
WO2010006174A2 (en) 2008-07-10 2010-01-14 Reichenbach Steven H Method and apparatus for sorting particles using asymmetrical particle shifting
EP2324063A4 (en) * 2008-07-18 2012-01-18 Novartis Ag Non-invasive fetal rhd genotyping from maternal whole blood
JP5795255B2 (en) 2008-07-18 2015-10-14 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Canon U.S. Life Sciences, Inc. Methods and systems for microfluidic DNA sample preparation
IT1391408B1 (en) * 2008-10-02 2011-12-23 Silicon Biosystems Spa CHAMBER OF SEPARATION
JP2010099052A (en) * 2008-10-27 2010-05-06 Olympus Corp Method for isolating cell
JP5750661B2 (en) * 2009-01-23 2015-07-22 学校法人 芝浦工業大学 3D dielectrophoresis device
EP2421955A4 (en) * 2009-04-21 2012-10-10 Genetic Technologies Ltd Methods for obtaining fetal genetic material
GB2483402B (en) 2009-06-04 2014-04-09 Lockheed Corp Multiple-sample microfluidic chip for DNA analysis
PL2440941T3 (en) * 2009-06-10 2017-10-31 Cynvenio Biosystems Inc Sheath flow devices and methods
FR2946895A1 (en) 2009-06-19 2010-12-24 Commissariat Energie Atomique MICROFLUIDIC SYSTEM AND CORRESPONDING METHOD FOR THE TRANSFER OF ELEMENTS BETWEEN PHASES LIQUID AND USE OF SAID SYSTEM FOR EXTRACTING THESE ELEMENTS
WO2011025986A1 (en) * 2009-08-28 2011-03-03 The Trustees Of Columbia University In The City Of New York Multi-layer blood component exchange devices, systems, and methods
US8584535B2 (en) * 2009-09-17 2013-11-19 Innova Prep LLC Liquid to liquid biological particle concentrator with disposable fluid path
CN102713919B (en) * 2009-11-13 2016-07-06 贝克曼考尔特公司 For using the system and method clustering the existence detecting biological condition
US9034658B2 (en) 2009-11-23 2015-05-19 The General Hospital Corporation Microfluidic devices for the capture of biological sample components
CA2785390C (en) 2009-12-23 2016-02-09 Cytovera, Inc. A system and method for particle filtration
US8187979B2 (en) * 2009-12-23 2012-05-29 Varian Semiconductor Equipment Associates, Inc. Workpiece patterning with plasma sheath modulation
WO2011109762A1 (en) * 2010-03-04 2011-09-09 National University Of Singapore Microfluidics sorter for cell detection and isolation
US8774488B2 (en) 2010-03-11 2014-07-08 Cellscape Corporation Method and device for identification of nucleated red blood cells from a maternal blood sample
US20110293558A1 (en) * 2010-03-22 2011-12-01 Massachusetts Institute Of Technology Material properties of t cells and related methods and compositions
JP5728778B2 (en) * 2010-06-01 2015-06-03 国立大学法人九州工業大学 Analysis device and method of manufacturing analysis device
US8590710B2 (en) * 2010-06-10 2013-11-26 Samsung Electronics Co., Ltd. Target particles-separating device and method using multi-orifice flow fractionation channel
MX2013004184A (en) 2010-10-15 2013-07-29 Lockheed Corp Micro fluidic optic design.
KR101768123B1 (en) * 2010-12-03 2017-08-16 삼성전자주식회사 Hydrodynamic filter, filtering apparatus including the same and filtering method by the same
AU2012211957B2 (en) * 2011-02-04 2017-02-23 The Trustees Of The University Of Pennsylvania A method for detecting chromosome structure and gene expression simultaneously in single cells
EP2490005A1 (en) * 2011-02-18 2012-08-22 Koninklijke Philips Electronics N.V. Microfluidic resistance network and microfluidic device
EP2490020A1 (en) * 2011-02-18 2012-08-22 Koninklijke Philips Electronics N.V. Measurement chip, microfluidic device and method of measurement chip manufacture
KR101882864B1 (en) 2011-06-24 2018-08-27 삼성전자주식회사 Hydrodynamic filter unit, hydrodynamic filter including the same and method of filtering target materials by using them
CN103998932B (en) 2011-06-29 2017-06-06 中央研究院 Capture, purifying and release using face coat to biological substance
US10466160B2 (en) 2011-08-01 2019-11-05 Celsee Diagnostics, Inc. System and method for retrieving and analyzing particles
US9174216B2 (en) 2013-03-13 2015-11-03 DeNovo Science, Inc. System for capturing and analyzing cells
ES2797448T3 (en) 2011-08-01 2020-12-02 Bio Rad Laboratories Cell capture system
US9404864B2 (en) 2013-03-13 2016-08-02 Denovo Sciences, Inc. System for imaging captured cells
WO2013020089A2 (en) * 2011-08-04 2013-02-07 Sage Science, Inc. Systems and methods for processing fluids
KR101933618B1 (en) 2011-11-29 2018-12-31 삼성전자주식회사 Device for detecting and separating target biomolecules and method for detecting and separating target biomolecules using the same
BR112014013756A2 (en) 2011-12-07 2017-06-13 Cytovera Inc Method and device for sample processing
US9322054B2 (en) 2012-02-22 2016-04-26 Lockheed Martin Corporation Microfluidic cartridge
FR2987282B1 (en) * 2012-02-24 2017-12-29 Fonds De L'espci Georges Charpak MICROCANAL WITH OPENING AND / OR CLOSING AND / OR PUMPING DEVICE
AU2013318647B2 (en) 2012-09-21 2017-10-26 Massachusetts Institute Of Technology Micro-fluidic device and uses thereof
AU2013329110B2 (en) 2012-10-12 2017-05-04 Sage Science, Inc. Side-eluting molecular fractionator
WO2014065861A1 (en) 2012-10-26 2014-05-01 The Trustees Of The University Of Pennsylvania Compositions, methods and microfluidics device for telomerase based in vitro diagnostic assays for detecting circulating tumor cells (ctc)
US9494500B2 (en) 2012-10-29 2016-11-15 Academia Sinica Collection and concentration system for biologic substance of interest and use thereof
US9606102B2 (en) 2013-01-26 2017-03-28 Denovo Sciences, Inc. System and method for capturing and analyzing cells
US8934700B2 (en) * 2013-03-04 2015-01-13 Caliper Life Sciences, Inc. High-throughput single-cell imaging, sorting, and isolation
JP2016514047A (en) * 2013-03-06 2016-05-19 プレジデント アンド フェローズ オブ ハーバード カレッジ Device and method for forming relatively monodisperse droplets
US9707562B2 (en) 2013-03-13 2017-07-18 Denovo Sciences, Inc. System for capturing and analyzing cells
US9888283B2 (en) 2013-03-13 2018-02-06 Nagrastar Llc Systems and methods for performing transport I/O
USD758372S1 (en) * 2013-03-13 2016-06-07 Nagrastar Llc Smart card interface
WO2014145237A1 (en) * 2013-03-15 2014-09-18 Dialyflux, Llc Surfaces for manipulating particle flow
WO2016019393A1 (en) 2014-08-01 2016-02-04 Gpb Scientific, Llc Methods and systems for processing particles
US10391490B2 (en) 2013-05-31 2019-08-27 Celsee Diagnostics, Inc. System and method for isolating and analyzing cells
US9856535B2 (en) 2013-05-31 2018-01-02 Denovo Sciences, Inc. System for isolating cells
US9656262B2 (en) * 2013-06-11 2017-05-23 Euveda Biosciences, Inc. Microfluidic grid-based design for high throughput assays
DE102013215570A1 (en) 2013-08-07 2015-02-12 Robert Bosch Gmbh A method and apparatus for processing a sample of biological material containing target cells and companion cells for extracting nucleic acids of the target cells
DE102013215575A1 (en) * 2013-08-07 2015-02-12 Robert Bosch Gmbh A method and apparatus for processing a sample of biological material containing target cells and companion cells for extracting nucleic acids of the target cells
US10202577B2 (en) 2013-10-18 2019-02-12 The General Hospital Corporation Microfluidic sorting using high gradient magnetic fields
EP3071327A1 (en) * 2013-11-19 2016-09-28 Platod Fluidic device for producing platelets
CN109289945A (en) 2013-11-22 2019-02-01 通用医疗公司 For separating the microfluidic methods and system of cluster of particle
US10047344B2 (en) 2014-02-18 2018-08-14 National University Of Singapore Biophysically sorted osteoprogenitors from culture expanded bone marrow derived mesenchymal stromal cells (MSCs)
JP2015166707A (en) * 2014-03-04 2015-09-24 キヤノン株式会社 microchannel device
EP3126814B1 (en) 2014-04-01 2019-06-12 Academia Sinica Methods and systems for cancer diagnosis and prognosis
JP2017515472A (en) 2014-05-01 2017-06-15 キング アブドラ ユニバーシティ オブ サイエンス アンド テクノロジー Microfluidic device for separating cells
US10391491B2 (en) 2014-08-07 2019-08-27 The General Hospital Corporation Platelet-targeted microfluidic isolation of cells
CN105381824B (en) 2014-08-26 2019-04-23 中央研究院 Collector framework layout designs
WO2016044555A1 (en) 2014-09-17 2016-03-24 Massachusetts Institute Of Technology System and method for inertial focusing microfiltration for intra-operative blood salvage autotransfusion
CN104212708B (en) * 2014-09-19 2015-12-02 青岛市中心医院 A kind of intestinal contents target component extraction element
CA2963293A1 (en) 2014-10-06 2016-04-14 The Trustees Of The University Of Pennsylvania Compositions and methods for isolation of circulating tumor cells (ctc)
CA2964487A1 (en) 2014-10-15 2016-04-21 Sage Science, Inc. Apparatuses, methods and systems for automated processing of nucleic acids and electrophoretic sample preparation
US9835538B2 (en) * 2014-11-26 2017-12-05 International Business Machines Corporation Biopolymer separation using nanostructured arrays
US9636675B2 (en) * 2014-11-26 2017-05-02 International Business Machines Corporation Pillar array structure with uniform and high aspect ratio nanometer gaps
KR102360072B1 (en) * 2014-12-08 2022-02-08 삼성전자주식회사 Apparatus for classifying micro-particles
EP3242946A4 (en) * 2015-01-07 2018-09-26 Indee. Inc. A method for mechanical and hydrodynamic microfluidic transfection and apparatus therefor
KR101583633B1 (en) * 2015-01-12 2016-01-08 한국항공대학교산학협력단 Negative dielectrophoresis force(n-dep) based cell sorting platform and cell sorting method using the same
US10364467B2 (en) 2015-01-13 2019-07-30 The Chinese University Of Hong Kong Using size and number aberrations in plasma DNA for detecting cancer
US20180015470A1 (en) * 2015-01-23 2018-01-18 Unimed Biotech (Shanghai) Co., Ltd. Microfluidics based fetal cell detection and isolation for non-invasive prenatal testing
SG11201706826VA (en) * 2015-02-27 2017-09-28 Toppan Printing Co Ltd Method for separating cells, and device therefor
USD864968S1 (en) 2015-04-30 2019-10-29 Echostar Technologies L.L.C. Smart card interface
CN108025304B (en) * 2015-07-22 2022-01-21 北卡罗来纳-查佩尔山大学 Fluidic devices containing bead well geometries with spatially separated bead retention and signal detection segments and related methods
JP7054678B2 (en) 2015-11-20 2022-04-14 ワシントン・ユニバーシティ Preparation Electrophoresis Method for Targeted Purification of Genomic DNA Fragments
WO2017127775A1 (en) * 2016-01-22 2017-07-27 The Board Of Trustees Of The Leland Stanford Junior University A micro-fluidic device for selective sorting of highly motile and morphologically normal sperm from unprocessed semen
US10107726B2 (en) 2016-03-16 2018-10-23 Cellmax, Ltd. Collection of suspended cells using a transferable membrane
CA3021722A1 (en) * 2016-04-22 2017-10-26 Cagri Abdullah Savran High-throughput particle capture and analysis
JP6933212B2 (en) * 2016-06-20 2021-09-08 凸版印刷株式会社 Liquid medium replacement method and flow path device for the method
CN106119189B (en) * 2016-07-11 2020-04-10 山东亚大药业有限公司 Separation and purification method and kit for fetal nucleated red blood cells
KR102456488B1 (en) 2016-07-21 2022-10-19 버클리 라잇츠, 인크. Classification of T lymphocytes in microfluidic devices
CA3032340A1 (en) * 2016-08-02 2018-02-08 Imec Vzw Method and arrangement for focusing objects in a flow
JP6843420B2 (en) * 2016-08-22 2021-03-17 国立大学法人東京工業大学 Fine particle separation device and fine particle separation method
EP3957997A1 (en) 2016-08-26 2022-02-23 Juno Therapeutics, Inc. Methods of enumerating particles present in a cell composition
US20190225930A1 (en) * 2016-09-13 2019-07-25 HysOcean, Inc. Microfluidic filter devices and methods
US10253350B2 (en) * 2016-09-20 2019-04-09 International Business Machines Corporation Separation of molecules using nanopillar arrays
US10010883B2 (en) * 2016-09-20 2018-07-03 International Business Machines Corporation Deterministic lateral displacement arrays
CN114755412A (en) * 2016-10-23 2022-07-15 伯克利之光生命科技公司 Method for screening B cell lymphocytes
US11384327B2 (en) 2016-11-01 2022-07-12 California Institute Of Technology Microfluidic devices and methods for purifying rare antigen-specific T cell populations
US10471425B2 (en) 2017-02-16 2019-11-12 International Business Machines Corporation Automated machine for sorting of biological fluids
CA3057681A1 (en) 2017-03-28 2018-10-04 Chromatan Inc. Continuous countercurrent spiral chromatography
EP3607308A1 (en) 2017-04-07 2020-02-12 Sage Science, Inc. Systems and methods for detection of genetic structural variation using integrated electrophoretic dna purification
DE102017003455B4 (en) * 2017-04-10 2020-12-10 Forschungszentrum Jülich GmbH Device and method for converting heat, chemical energy or electrical energy into kinetic energy and use of the device
US11648559B2 (en) * 2017-08-04 2023-05-16 University Of Georgia Research Foundation, Inc. Devices and methods for separating circulating tumor cells from biological samples
WO2019046307A1 (en) 2017-08-29 2019-03-07 Celsee Diagnostics, Inc. System and method for isolating and analyzing cells
JP6620269B2 (en) * 2017-10-03 2019-12-11 Nok株式会社 Cell capture device
CN107723207B (en) * 2017-11-01 2019-01-01 深圳市瑞格生物科技有限公司 A kind of chip of separating trap cell and its application in tumour cell sorting
US11458474B2 (en) 2018-01-19 2022-10-04 International Business Machines Corporation Microfluidic chips with one or more vias
US10946380B2 (en) * 2018-01-19 2021-03-16 International Business Machines Corporation Microfluidic chips for particle purification and fractionation
US20190226953A1 (en) 2018-01-19 2019-07-25 International Business Machines Corporation Microscale and mesoscale condenser devices
WO2019153032A1 (en) * 2018-02-07 2019-08-15 University Of South Australia Pre-natal cell isolation
WO2019190488A1 (en) * 2018-03-27 2019-10-03 Hewlett-Packard Development Company, L.P. Particle separation
WO2019190490A1 (en) * 2018-03-27 2019-10-03 Hewlett-Packard Development Company, L.P. Nucleic acid separation
JP7048940B2 (en) * 2018-03-30 2022-04-06 株式会社日立製作所 Cell manufacturing equipment
CN109874316B (en) * 2018-05-25 2022-10-11 江苏汇先医药技术有限公司 Device for enriching a screening target, such as cells, bacteria or biomolecules, from a sample
US11185861B2 (en) 2018-06-13 2021-11-30 International Business Machines Corporation Multistage deterministic lateral displacement device for particle separation
DE102018210665A1 (en) * 2018-06-29 2020-01-02 Robert Bosch Gmbh Microfluidic flow cell and method for separating cells
US10697719B2 (en) * 2018-08-09 2020-06-30 International Business Machines Corporation Monitoring a recirculating cooling system for bacterial growth
US10898895B2 (en) * 2018-09-13 2021-01-26 Talis Biomedical Corporation Vented converging capillary biological sample port and reservoir
AU2019351130A1 (en) 2018-09-27 2021-04-08 Grail, Llc Methylation markers and targeted methylation probe panel
US11440002B2 (en) * 2018-10-23 2022-09-13 International Business Machines Corporation Microfluidic chips with one or more vias filled with sacrificial plugs
US10967375B2 (en) * 2018-10-23 2021-04-06 International Business Machines Corporation Microfluidic devices with multiple inlets and outlets
EP3872488A4 (en) * 2018-10-25 2022-11-09 TL Genomics Inc. Blood pretreatment for microfluidic blood cell sorting
US10633693B1 (en) 2019-04-16 2020-04-28 Celsee Diagnostics, Inc. System and method for leakage control in a particle capture system
US11273439B2 (en) 2019-05-07 2022-03-15 Bio-Rad Laboratories, Inc. System and method for target material retrieval from microwells
CA3138881A1 (en) 2019-05-07 2020-11-12 Bio-Rad Laboratories, Inc. System and method for automated single cell processing
EP3982716A4 (en) 2019-06-14 2023-09-06 Bio-Rad Laboratories, Inc. System and method for automated single cell processing and analyses
CN110862905B (en) * 2020-01-09 2023-03-31 北京航空航天大学合肥创新研究院 Chip device for cell migration experiment, preparation method and experiment method
US11211144B2 (en) 2020-02-18 2021-12-28 Tempus Labs, Inc. Methods and systems for refining copy number variation in a liquid biopsy assay
US11475981B2 (en) 2020-02-18 2022-10-18 Tempus Labs, Inc. Methods and systems for dynamic variant thresholding in a liquid biopsy assay
US11211147B2 (en) 2020-02-18 2021-12-28 Tempus Labs, Inc. Estimation of circulating tumor fraction using off-target reads of targeted-panel sequencing
US11504719B2 (en) 2020-03-12 2022-11-22 Bio-Rad Laboratories, Inc. System and method for receiving and delivering a fluid for sample processing
US11738288B2 (en) 2020-06-29 2023-08-29 Jacques Chammas Automated system and method to isolate specific cells from blood or bone marrow
CN111733138B (en) * 2020-07-30 2021-03-30 首都医科大学附属北京友谊医院 High-flux magnetic sorting method for circulating tumor cells
FR3117884B1 (en) 2020-12-21 2024-02-16 Commissariat Energie Atomique System for sorting by size of particles expelled by centrifugation and method of configuring such a system
CN113214959B (en) * 2021-04-06 2022-08-26 深圳市儿童医院 Chip for separating and capturing Ewing sarcoma circulating tumor cells
CN114196521A (en) * 2021-12-30 2022-03-18 中国科学院上海微系统与信息技术研究所 Fluorescence in situ hybridization chip and fluorescence in situ hybridization method

Citations (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5300779A (en) * 1985-08-05 1994-04-05 Biotrack, Inc. Capillary flow device
US5837200A (en) * 1995-06-02 1998-11-17 Bayer Aktiengesellschaft Sorting device for biological cells or viruses
US5840502A (en) * 1994-08-31 1998-11-24 Activated Cell Therapy, Inc. Methods for enriching specific cell-types by density gradient centrifugation
US6027623A (en) * 1998-04-22 2000-02-22 Toyo Technologies, Inc. Device and method for electrophoretic fraction
US6030581A (en) * 1997-02-28 2000-02-29 Burstein Laboratories Laboratory in a disk
US6368871B1 (en) * 1997-08-13 2002-04-09 Cepheid Non-planar microstructures for manipulation of fluid samples
US20020119482A1 (en) * 1996-07-30 2002-08-29 Aclara Biosciences, Inc. Microfluidic method for nucleic acid purification and processing
US6511967B1 (en) * 1999-04-23 2003-01-28 The General Hospital Corporation Use of an internalizing transferrin receptor to image transgene expression
US20030040119A1 (en) * 2001-04-11 2003-02-27 The Regents Of The University Of Michigan Separation devices and methods for separating particles
US20030091476A1 (en) * 2000-07-03 2003-05-15 Xiaochuan Zhou Fluidic methods and devices for parallel chemical reactions
US20030134416A1 (en) * 2001-10-11 2003-07-17 Douglas Yamanishi Methods, compositions, and automated systems for separating rare cells from fluid samples
US6632655B1 (en) * 1999-02-23 2003-10-14 Caliper Technologies Corp. Manipulation of microparticles in microfluidic systems
US6636498B1 (en) * 1999-01-08 2003-10-21 Cisco Technology, Inc. Mobile IP mobile router
US20040005582A1 (en) * 2000-08-10 2004-01-08 Nanobiodynamics, Incorporated Biospecific desorption microflow systems and methods for studying biospecific interactions and their modulators
US20040019300A1 (en) * 2002-07-26 2004-01-29 Leonard Leslie Anne Microfluidic blood sample separations
US20040048360A1 (en) * 1999-08-26 2004-03-11 Caliper Technologies Corp. Microfluidic analytic detection assays, devices, and integrated systems
US20040053352A1 (en) * 1998-09-28 2004-03-18 Tianmei Ouyang Diagnostics based on tetrazolium compounds
US20040142463A1 (en) * 2001-10-11 2004-07-22 George Walker Methods, compositions, and automated systems for separating rare cells from fluid samples
US20060000772A1 (en) * 2002-11-29 2006-01-05 Toru Sano Separation apparatus and separation method
US20070202525A1 (en) * 2006-02-02 2007-08-30 The Board Of Trustees Of The Leland Stanford Junior University Non-invasive fetal genetic screening by digital analysis
US7277876B2 (en) * 2004-01-23 2007-10-02 Solomon Research Llc Dynamic adaptive distributed computer system
US20080023399A1 (en) * 2006-06-01 2008-01-31 Inglis David W Apparatus and method for continuous particle separation
US20080038733A1 (en) * 2006-03-28 2008-02-14 Baylor College Of Medicine Screening for down syndrome

Family Cites Families (441)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3924947A (en) 1973-10-19 1975-12-09 Coulter Electronics Apparatus for preservation and identification of particles analyzed by flow-through apparatus
US4009435A (en) 1973-10-19 1977-02-22 Coulter Electronics, Inc. Apparatus for preservation and identification of particles analyzed by flow-through apparatus
US3906929A (en) 1973-11-23 1975-09-23 Lynn Lawrence Augspurger Processes for reproduction of cellular bodies
DE2502621C3 (en) 1975-01-23 1978-09-14 Kernforschungsanlage Juelich Gmbh, 5170 Juelich Measurement of elastic and dielectric properties of the membrane of living cells
US4115534A (en) 1976-08-19 1978-09-19 Minnesota Mining And Manufacturing Company In vitro diagnostic test
US4190535A (en) 1978-02-27 1980-02-26 Corning Glass Works Means for separating lymphocytes and monocytes from anticoagulated blood
EP0057907B1 (en) * 1981-02-05 1986-12-30 Asahi Kasei Kogyo Kabushiki Kaisha Apparatus for separating blood components
US4415405A (en) 1981-08-19 1983-11-15 Yale University Method for engraving a grid pattern on microscope slides and slips
US4584268A (en) 1981-10-13 1986-04-22 Ceriani Roberto Luis Method and compositions for carcinoma diagnosis
US4434156A (en) 1981-10-26 1984-02-28 The Salk Institute For Biological Studies Monoclonal antibodies specific for the human transferrin receptor glycoprotein
IL68507A (en) 1982-05-10 1986-01-31 Univ Bar Ilan System and methods for cell selection
US5310674A (en) 1982-05-10 1994-05-10 Bar-Ilan University Apertured cell carrier
US4508625A (en) 1982-10-18 1985-04-02 Graham Marshall D Magnetic separation using chelated magnetic ions
AU594641B2 (en) 1983-11-08 1990-03-15 Bar-Ilan University Apparatus & methods for cell selection
US4675286A (en) 1985-01-28 1987-06-23 Aspen Diagnostics, Inc. Fetal cell separation and testing
WO1986006170A1 (en) 1985-04-10 1986-10-23 Immunicon Corporation Direct homogeneous assay
US4990326A (en) * 1985-05-31 1991-02-05 Summa Medical Corporation Method for detecting blood platelets
US4963498A (en) 1985-08-05 1990-10-16 Biotrack Capillary flow device
US4664796A (en) 1985-09-16 1987-05-12 Coulter Electronics, Inc. Flux diverting flow chamber for high gradient magnetic separation of particles from a liquid medium
US4790640A (en) 1985-10-11 1988-12-13 Nason Frederic L Laboratory slide
US4999283A (en) 1986-01-10 1991-03-12 University Of Kentucky Research Foundation Method for x and y spermatozoa separation
US5447841A (en) 1986-01-16 1995-09-05 The Regents Of The Univ. Of California Methods for chromosome-specific staining
US4800159A (en) 1986-02-07 1989-01-24 Cetus Corporation Process for amplifying, detecting, and/or cloning nucleic acid sequences
JPS62217973A (en) 1986-03-20 1987-09-25 東レ株式会社 Apparatus for fractionating liquid
US4906439A (en) 1986-03-25 1990-03-06 Pb Diagnostic Systems, Inc. Biological diagnostic device and method of use
US4789628A (en) 1986-06-16 1988-12-06 Vxr, Inc. Devices for carrying out ligand/anti-ligand assays, methods of using such devices and diagnostic reagents and kits incorporating such devices
US4814098A (en) 1986-09-06 1989-03-21 Bellex Corporation Magnetic material-physiologically active substance conjugate
US4925788A (en) 1986-10-24 1990-05-15 Immunicon Corporation Immunoassay system and procedure based on precipitin-like interaction between immune complex and Clq or other non-immunospecific factor
JP2662215B2 (en) 1986-11-19 1997-10-08 株式会社日立製作所 Cell holding device
US4886761A (en) 1987-03-26 1989-12-12 Yellowstone Diagnostics Corporation Polysilicon binding assay support and methods
US4971904A (en) 1987-06-26 1990-11-20 E. I. Du Pont De Nemours And Company Heterogeneous immunoassay
JP2559760B2 (en) 1987-08-31 1996-12-04 株式会社日立製作所 Cell delivery method
US4977078A (en) 1987-12-22 1990-12-11 Olympus Optical Co., Ltd. Plate substrate immunoassay device and method for performing a multi-test immunoassay on a specimen
US5039426A (en) 1988-05-17 1991-08-13 University Of Utah Process for continuous particle and polymer separation in split-flow thin cells using flow-dependent lift forces
US5215926A (en) 1988-06-03 1993-06-01 Cellpro, Inc. Procedure for designing efficient affinity cell separation processes
DE68928082T2 (en) 1988-08-31 1998-01-15 Aprogenex Inc MANUAL IN SITU HYBRIDIZATION PROCESS
US5183744A (en) 1988-10-26 1993-02-02 Hitachi, Ltd. Cell handling method for cell fusion processor
US5101825A (en) 1988-10-28 1992-04-07 Blackbox, Inc. Method for noninvasive intermittent and/or continuous hemoglobin, arterial oxygen content, and hematocrit determination
DE68927059T3 (en) 1988-11-15 2001-05-10 Univ Yale IN SITU SUPPRESSION HYBRIDIZATION AND USES
US4984574A (en) 1988-11-23 1991-01-15 Seth Goldberg Noninvasive fetal oxygen monitor using NMR
DE68928535T2 (en) 1988-12-06 1998-04-16 Flinders Technologies Pty Ltd ISOLATION OF FETAL CELLS FROM MATERNAL BLOOD FOR CARRYING OUT PRENATAL DIAGNOSTICS
CA1340565C (en) 1989-06-29 1999-05-25 Thomas B. Okarma Device and process for cell capture and recovery
US5698271A (en) 1989-08-22 1997-12-16 Immunivest Corporation Methods for the manufacture of magnetically responsive particles
AU658132B2 (en) 1989-11-13 1995-04-06 Children's Medical Center Corporation Non-invasive method for isolation and detection of fetal DNA
US5641628A (en) 1989-11-13 1997-06-24 Children's Medical Center Corporation Non-invasive method for isolation and detection of fetal DNA
WO1991007661A1 (en) 1989-11-20 1991-05-30 Hill Vincent E A method of detecting drugs in living and post-mortem skin and a kit therefor
EP0502037A1 (en) 1989-11-24 1992-09-09 Isis Innovation Limited Prenatal genetic determination
GB8926781D0 (en) 1989-11-27 1990-01-17 Nat Res Dev Identification of micro-organisms
AU647741B2 (en) 1989-12-01 1994-03-31 Regents Of The University Of California, The Methods and compositions for chromosome-specific staining
GB8929057D0 (en) 1989-12-22 1990-02-28 Gen Electric Co Plc Sensor
FR2657543B1 (en) 1990-01-26 1992-12-18 Biocom Sa MODULAR DEVICE FOR COLLECTING, INCUBATING, FILTERING MULTIPLE SAMPLES.
US5327777A (en) 1990-02-24 1994-07-12 University Of Hertfordshire Biorheological measurement
JPH03247276A (en) 1990-02-27 1991-11-05 Hitachi Ltd Arrangement of cell and apparatus therefor
US6054034A (en) 1990-02-28 2000-04-25 Aclara Biosciences, Inc. Acrylic microchannels and their use in electrophoretic applications
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
US5858188A (en) 1990-02-28 1999-01-12 Aclara Biosciences, Inc. Acrylic microchannels and their use in electrophoretic applications
US6176962B1 (en) 1990-02-28 2001-01-23 Aclara Biosciences, Inc. Methods for fabricating enclosed microchannel structures
US5770029A (en) 1996-07-30 1998-06-23 Soane Biosciences Integrated electrophoretic microdevices
FR2659347B1 (en) 1990-03-12 1994-09-02 Agronomique Inst Nat Rech DEVICE FOR CULTURING CELLS PROVIDING THEIR IMMOBILIZATION.
US5153117A (en) 1990-03-27 1992-10-06 Genetype A.G. Fetal cell recovery method
CA2081203A1 (en) 1990-04-23 1991-10-24 Ronald J. Berenson Method for enriching fetal cells from maternal blood
US5147606A (en) 1990-08-06 1992-09-15 Miles Inc. Self-metering fluid analysis device
CA2091764C (en) 1990-09-20 2003-04-08 Michael Louis Bittner Probe compositions for chromosome identification and methods
US6277569B1 (en) 1990-09-20 2001-08-21 Vysis, Inc. Methods for multiple direct label probe detection of multiple chromosomes or regions thereof by in situ hybridization
US5622831A (en) 1990-09-26 1997-04-22 Immunivest Corporation Methods and devices for manipulation of magnetically collected material
US5541072A (en) 1994-04-18 1996-07-30 Immunivest Corporation Method for magnetic separation featuring magnetic particles in a multi-phase system
US5135627A (en) 1990-10-15 1992-08-04 Soane Technologies, Inc. Mosaic microcolumns, slabs, and separation media for electrophoresis and chromatography
US5217627A (en) 1990-11-06 1993-06-08 Pall Corporation System and method for processing biological fluid
US5186827A (en) 1991-03-25 1993-02-16 Immunicon Corporation Apparatus for magnetic separation featuring external magnetic means
US5646001A (en) 1991-03-25 1997-07-08 Immunivest Corporation Affinity-binding separation and release of one or more selected subset of biological entities from a mixed population thereof
US5466574A (en) 1991-03-25 1995-11-14 Immunivest Corporation Apparatus and methods for magnetic separation featuring external magnetic means
US5173158A (en) 1991-07-22 1992-12-22 Schmukler Robert E Apparatus and methods for electroporation and electrofusion
US5240856A (en) 1991-10-23 1993-08-31 Cellpro Incorporated Apparatus for cell separation
US5672481A (en) 1991-10-23 1997-09-30 Cellpro, Incorporated Apparatus and method for particle separation in a closed field
US5846708A (en) 1991-11-19 1998-12-08 Massachusetts Institiute Of Technology Optical and electrical methods and apparatus for molecule detection
AU4292893A (en) 1992-04-21 1993-11-18 Regents Of The University Of California, The Multicolor in situ hybridization methods for genetic testing
US5296375A (en) 1992-05-01 1994-03-22 Trustees Of The University Of Pennsylvania Mesoscale sperm handling devices
US5498392A (en) 1992-05-01 1996-03-12 Trustees Of The University Of Pennsylvania Mesoscale polynucleotide amplification device and method
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
US5304487A (en) 1992-05-01 1994-04-19 Trustees Of The University Of Pennsylvania Fluid handling in mesoscale analytical 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
AU674685B2 (en) 1992-05-01 1997-01-09 Trustees Of The University Of Pennsylvania, The Fluid handling in microfabricated analytical devices
US5486335A (en) 1992-05-01 1996-01-23 Trustees Of The University Of Pennsylvania Analysis based on flow restriction
US6143576A (en) 1992-05-21 2000-11-07 Biosite Diagnostics, Inc. Non-porous diagnostic devices for the controlled movement of reagents
US6156270A (en) 1992-05-21 2000-12-05 Biosite Diagnostics, Inc. Diagnostic devices and apparatus for the controlled movement of reagents without membranes
US5629147A (en) 1992-07-17 1997-05-13 Aprogenex, Inc. Enriching and identifying fetal cells in maternal blood for in situ hybridization
AU2593192A (en) 1992-09-14 1994-04-12 Oystein Fodstad Detection of specific target cells in specialized or mixed cell population and solutions containing mixed cell populations
US5275933A (en) 1992-09-25 1994-01-04 The Board Of Trustees Of The Leland Stanford Junior University Triple gradient process for recovering nucleated fetal cells from maternal blood
US5489506A (en) 1992-10-26 1996-02-06 Biolife Systems, Inc. Dielectrophoretic cell stream sorter
US6953668B1 (en) 1992-11-05 2005-10-11 Sloan-Kettering Institute For Cancer Research Prostate-specific membrane antigen
US5457024A (en) 1993-01-22 1995-10-10 Aprogenex, Inc. Isolation of fetal erythrocytes
US5427663A (en) 1993-06-08 1995-06-27 British Technology Group Usa Inc. Microlithographic array for macromolecule and cell fractionation
US5714325A (en) 1993-09-24 1998-02-03 New England Medical Center Hospitals Prenatal diagnosis by isolation of fetal granulocytes from maternal blood
US5472842A (en) 1993-10-06 1995-12-05 The Regents Of The University Of California Detection of amplified or deleted chromosomal regions
ATE424927T1 (en) 1993-10-28 2009-03-15 Houston Advanced Res Ct MICROFABRICATED POROUS FLOW DEVICE FOR DISCRETE DETERMINATION OF BONDING REACTIONS
US6315953B1 (en) 1993-11-01 2001-11-13 Nanogen, Inc. Devices for molecular biological analysis and diagnostics including waveguides
US6068818A (en) 1993-11-01 2000-05-30 Nanogen, Inc. Multicomponent devices for molecular biological analysis and diagnostics
US6331274B1 (en) 1993-11-01 2001-12-18 Nanogen, Inc. Advanced active circuits and devices for molecular biological analysis and diagnostics
NL9401260A (en) 1993-11-12 1995-06-01 Cornelis Johannes Maria Van Ri Membrane for microfiltration, ultrafiltration, gas separation and catalysis, method for manufacturing such a membrane, mold for manufacturing such a membrane, as well as various separation systems comprising such a membrane.
US5432054A (en) 1994-01-31 1995-07-11 Applied Imaging Method for separating rare cells from a population of cells
NO180658C (en) 1994-03-10 1997-05-21 Oeystein Fodstad Method and Device for Detecting Specific Target Cells in Specialized or Mixed Cell Populations and Solutions Containing Mixed Cell Populations
US5563067A (en) 1994-06-13 1996-10-08 Matsushita Electric Industrial Co., Ltd. Cell potential measurement apparatus having a plurality of microelectrodes
US6071394A (en) 1996-09-06 2000-06-06 Nanogen, Inc. Channel-less separation of bioparticles on a bioelectronic chip by dielectrophoresis
US5637458A (en) 1994-07-20 1997-06-10 Sios, Inc. Apparatus and method for the detection and assay of organic molecules
US5648222A (en) 1994-07-27 1997-07-15 The Trustees Of Columbia University In The City Of New York Method for preserving cells, and uses of said method
US6001229A (en) 1994-08-01 1999-12-14 Lockheed Martin Energy Systems, Inc. Apparatus and method for performing microfluidic manipulations for chemical analysis
US5707799A (en) 1994-09-30 1998-01-13 Abbott Laboratories Devices and methods utilizing arrays of structures for analyte capture
US5662813A (en) 1994-10-21 1997-09-02 Bioseparations, Inc. Method for separation of nucleated fetal erythrocytes from maternal blood samples
CA2181190C (en) 1994-11-14 2001-02-06 Peter Wilding Mesoscale sample preparation device and systems for determination and processing of analytes
US5750339A (en) 1994-11-30 1998-05-12 Thomas Jefferson University Methods for identifying fetal cells
US5648220A (en) 1995-02-14 1997-07-15 New England Medical Center Hospitals, Inc. Methods for labeling intracytoplasmic molecules
US6207369B1 (en) 1995-03-10 2001-03-27 Meso Scale Technologies, Llc Multi-array, multi-specific electrochemiluminescence testing
FR2733055B1 (en) 1995-04-12 1997-12-19 Chemodyne Sa NEW DEVICE FOR STUDYING ORGANOTYPICAL CULTURES AND ITS APPLICATIONS IN ELECTROPHYSIOLOGY
US5709943A (en) 1995-05-04 1998-01-20 Minnesota Mining And Manufacturing Company Biological adsorption supports
US5715946A (en) * 1995-06-07 1998-02-10 Reichenbach; Steven H. Method and apparatus for sorting particles suspended in a fluid
US5639669A (en) 1995-06-07 1997-06-17 Ledley; Robert Separation of fetal cells from maternal blood
DE69619400T2 (en) 1995-06-16 2002-09-26 Univ Washington Seattle FLAT MICROPRODUCED CROSS-FLOW FILTER FOR LIQUIDS
US6454945B1 (en) 1995-06-16 2002-09-24 University Of Washington Microfabricated devices and methods
US5856174A (en) 1995-06-29 1999-01-05 Affymetrix, Inc. Integrated nucleic acid diagnostic device
US6130098A (en) 1995-09-15 2000-10-10 The Regents Of The University Of Michigan Moving microdroplets
CA2237589A1 (en) 1995-11-16 1997-05-22 Michael W. Dahm Method of quantifying tumour cells in a body fluid and a suitable test kit
US5863502A (en) 1996-01-24 1999-01-26 Sarnoff Corporation Parallel reaction cassette and associated devices
US5830679A (en) 1996-03-01 1998-11-03 New England Medical Center Hospitals, Inc. Diagnostic blood test to identify infants at risk for sepsis
US5972721A (en) 1996-03-14 1999-10-26 The United States Of America As Represented By The Secretary Of The Air Force Immunomagnetic assay system for clinical diagnosis and other purposes
US5891651A (en) 1996-03-29 1999-04-06 Mayo Foundation For Medical Education And Research Methods of recovering colorectal epithelial cells or fragments thereof from stool
EP0891550A1 (en) 1996-04-05 1999-01-20 The Johns Hopkins University School Of Medicine A method of enriching rare cells
US6399023B1 (en) 1996-04-16 2002-06-04 Caliper Technologies Corp. Analytical system and method
US6958245B2 (en) 1996-04-25 2005-10-25 Bioarray Solutions Ltd. Array cytometry
US6387707B1 (en) 1996-04-25 2002-05-14 Bioarray Solutions Array Cytometry
US6251691B1 (en) 1996-04-25 2001-06-26 Bioarray Solutions, Llc Light-controlled electrokinetic assembly of particles near surfaces
US5989835A (en) 1997-02-27 1999-11-23 Cellomics, Inc. System for cell-based screening
US6890426B2 (en) 1996-06-07 2005-05-10 Immunivest Corporation Magnetic separation apparatus and methods
EP0920627B1 (en) 1996-06-07 2004-05-12 Immunivest Corporation Magnetic separation employing external and internal gradients
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.
IT1294964B1 (en) 1996-07-12 1999-04-23 Domenico Valerio INSULATION AND CULTURE OF FETAL CELLS FROM THE MATERNAL PERIPHERAL BLOOD
US6280967B1 (en) 1996-08-02 2001-08-28 Axiom Biotechnologies, Inc. Cell flow apparatus and method for real-time of cellular responses
US6100029A (en) 1996-08-14 2000-08-08 Exact Laboratories, Inc. Methods for the detection of chromosomal aberrations
WO1998008931A1 (en) 1996-08-26 1998-03-05 Princeton University Reversibly sealable microstructure sorting devices
EP0925494B1 (en) 1996-09-04 2001-12-19 Scandinavian Micro Biodevices A/S A micro flow system for particle separation and analysis
US6120666A (en) 1996-09-26 2000-09-19 Ut-Battelle, Llc Microfabricated device and method for multiplexed electrokinetic focusing of fluid streams and a transport cytometry method using same
US5858187A (en) 1996-09-26 1999-01-12 Lockheed Martin Energy Systems, Inc. Apparatus and method for performing electrodynamic focusing on a microchip
US6110343A (en) 1996-10-04 2000-08-29 Lockheed Martin Energy Research Corporation Material transport method and apparatus
US5731156A (en) 1996-10-21 1998-03-24 Applied Imaging, Inc. Use of anti-embryonic hemoglobin antibodies to identify fetal cells
US6008010A (en) 1996-11-01 1999-12-28 University Of Pittsburgh Method and apparatus for holding cells
EP0938674B1 (en) 1996-11-16 2005-06-01 NMI Naturwissenschaftliches und Medizinisches Institut an der Universität Tübingen in Reutlingen Stiftung Bürgerlichen Rechts Array of microelements, method of contacting cells in a liquid environment and method for the production of an array of microelements
DE19712309A1 (en) 1996-11-16 1998-05-20 Nmi Univ Tuebingen Microelement arrangement, method for contacting cells in a liquid environment and method for producing a microelement arrangement
US6083761A (en) 1996-12-02 2000-07-04 Glaxo Wellcome Inc. Method and apparatus for transferring and combining reagents
JP2001506005A (en) * 1996-12-11 2001-05-08 ナイコメッド・アマーシャム・ピーエルシー Selective cell lysis
WO1998028623A1 (en) 1996-12-20 1998-07-02 Gamera Bioscience Corporation An affinity binding-based system for detecting particulates in a fluid
US6235474B1 (en) 1996-12-30 2001-05-22 The Johns Hopkins University Methods and kits for diagnosing and determination of the predisposition for diseases
US6087134A (en) 1997-01-14 2000-07-11 Applied Imaging Corporation Method for analyzing DNA from a rare cell in a cell population
US5879624A (en) 1997-01-15 1999-03-09 Boehringer Laboratories, Inc. Method and apparatus for collecting and processing blood
US6306584B1 (en) 1997-01-21 2001-10-23 President And Fellows Of Harvard College Electronic-property probing of biological molecules at surfaces
US6008007A (en) 1997-01-31 1999-12-28 Oncotech, Inc. Radiation resistance assay for predicting treatment response and clinical outcome
US6056859A (en) 1997-02-12 2000-05-02 Lockheed Martin Energy Research Corporation Method and apparatus for staining immobilized nucleic acids
GB9704444D0 (en) 1997-03-04 1997-04-23 Isis Innovation Non-invasive prenatal diagnosis
CN1260046A (en) 1997-03-08 2000-07-12 邓迪大学 Prenatal diagnostic methods
GB9704876D0 (en) 1997-03-08 1997-04-23 Univ Dundee Diagnostic methods
US6623983B1 (en) 1997-03-25 2003-09-23 Immunivest Corporation Apparatus and methods for capture and analysis of particulate entities
US6391622B1 (en) 1997-04-04 2002-05-21 Caliper Technologies Corp. Closed-loop biochemical analyzers
US6066449A (en) 1997-04-15 2000-05-23 The Trustees Of Columbia University In The City Of New York Method of detecting metastatic thyroid cancer
KR100351531B1 (en) 1997-04-25 2002-09-11 캘리퍼 테크놀로지스 코포레이션 Microfludic devices incorporating improved channel geometries
WO1998049344A1 (en) 1997-04-28 1998-11-05 Lockheed Martin Energy Research Corporation Method and apparatus for analyzing nucleic acids
US6169816B1 (en) 1997-05-14 2001-01-02 Applied Imaging, Inc. Identification of objects of interest using multiple illumination schemes and finding overlap of features in corresponding multiple images
US6632619B1 (en) 1997-05-16 2003-10-14 The Governors Of The University Of Alberta Microfluidic system and methods of use
US6156273A (en) 1997-05-27 2000-12-05 Purdue Research Corporation Separation columns and methods for manufacturing the improved separation columns
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
JP4124830B2 (en) 1997-06-12 2008-07-23 クリニカル・マイクロ・センサーズ・インコーポレイテッド Electrical method for analyte detection
US5882465A (en) 1997-06-18 1999-03-16 Caliper Technologies Corp. Method of manufacturing microfluidic devices
SG81234A1 (en) 1997-07-04 2001-06-19 Toyko Electron Ltd Process solution supplying apparatus
US5876675A (en) 1997-08-05 1999-03-02 Caliper Technologies Corp. Microfluidic devices and systems
CA2301309A1 (en) 1997-08-13 1999-02-25 Cepheid Microstructures for the manipulation of fluid samples
US6540895B1 (en) 1997-09-23 2003-04-01 California Institute Of Technology Microfabricated cell sorter for chemical and biological materials
US7214298B2 (en) 1997-09-23 2007-05-08 California Institute Of Technology Microfabricated cell sorter
US5842787A (en) 1997-10-09 1998-12-01 Caliper Technologies Corporation Microfluidic systems incorporating varied channel dimensions
US6241894B1 (en) 1997-10-10 2001-06-05 Systemix High gradient magnetic device and method for cell separation or purification
US5962234A (en) 1997-10-20 1999-10-05 Applied Imaging Corporation Use of anti-embryonic epsilon hemoglobin antibodies to identify fetal cells
US5962250A (en) 1997-10-28 1999-10-05 Glaxo Group Limited Split multi-well plate and methods
AU9233698A (en) 1997-11-22 1999-06-17 Robert A. Levine Method for the detection, identification, enumeration and confirmation of circulating cancer cells and/or hematologic progenitor cells in whole blood
US6197523B1 (en) 1997-11-24 2001-03-06 Robert A. Levine Method for the detection, identification, enumeration and confirmation of circulating cancer and/or hematologic progenitor cells in whole blood
AU746580B2 (en) 1997-12-17 2002-05-02 Ecole Polytechnique Federale De Lausanne Positioning and electrophysiological characterization of individual cells and reconstituted membrane systems on microstructured carriers
US6210889B1 (en) 1998-01-28 2001-04-03 The Universite Laval Method for enrichment of fetal cells from maternal blood and use of same in determination of fetal sex and detection of chromosomal abnormalities
US20010018192A1 (en) 1998-02-12 2001-08-30 Terstappen Leon W.M.M. Labeled cells for use as an internal functional control in rare cell detection assays
US20020172987A1 (en) 1998-02-12 2002-11-21 Terstappen Leon W.M.M. Methods and reagents for the rapid and efficient isolation of circulating cancer cells
DE69941689D1 (en) 1998-02-12 2010-01-07 Univ Texas METHOD AND REAGENTS FOR RAPID AND EFFICIENT INSULATION OF CIRCULATING CANCER CELLS
US6537505B1 (en) 1998-02-20 2003-03-25 Bio Dot, Inc. Reagent dispensing valve
US6036857A (en) 1998-02-20 2000-03-14 Florida State University Research Foundation, Inc. Apparatus for continuous magnetic separation of components from a mixture
US6251343B1 (en) 1998-02-24 2001-06-26 Caliper Technologies Corp. Microfluidic devices and systems incorporating cover layers
CA2322282A1 (en) 1998-02-27 1999-09-02 Cli Oncology, Inc. Method and compositions for differential detection of primary tumor cells and metastatic cells
US6210910B1 (en) 1998-03-02 2001-04-03 Trustees Of Tufts College Optical fiber biosensor array comprising cell populations confined to microcavities
US6100033A (en) 1998-04-30 2000-08-08 The Regents Of The University Of California Diagnostic test for prenatal identification of Down's syndrome and mental retardation and gene therapy therefor
US6200765B1 (en) 1998-05-04 2001-03-13 Pacific Northwest Cancer Foundation Non-invasive methods to detect prostate cancer
JP2002528699A (en) 1998-05-22 2002-09-03 カリフォルニア インスティチュート オブ テクノロジー Microfabricated cell sorter
US6306589B1 (en) 1998-05-27 2001-10-23 Vysis, Inc. Biological assays for analyte detection
US6296752B1 (en) 1998-06-05 2001-10-02 Sarnoff Corporation Apparatus for separating molecules
US6529835B1 (en) 1998-06-25 2003-03-04 Caliper Technologies Corp. High throughput methods, systems and apparatus for performing cell based screening assays
EP1092144A1 (en) 1998-06-29 2001-04-18 Evotec BioSystems AG Method and device for manipulating particles in microsystems
US6045990A (en) 1998-07-09 2000-04-04 Baust; John M. Inclusion of apoptotic regulators in solutions for cell storage at low temperature
US6897073B2 (en) 1998-07-14 2005-05-24 Zyomyx, Inc. Non-specific binding resistant protein arrays and methods for making the same
US6576478B1 (en) 1998-07-14 2003-06-10 Zyomyx, Inc. Microdevices for high-throughput screening of biomolecules
US6274339B1 (en) 1999-02-05 2001-08-14 Millennium Pharmaceuticals, Inc. Methods and compositions for the diagnosis and treatment of body weight disorders, including obesity
FR2782730B1 (en) 1998-08-25 2002-05-17 Biocom Sa CELL SEPARATION PROCESS FOR THE ISOLATION OF PATHOGENIC CELLS, PARTICULARLY RARE CANCERES, EQUIPMENT AND REAGENT FOR IMPLEMENTING THE PROCESS AND APPLICATION OF THE PROCESS
US20030166132A1 (en) 1998-08-26 2003-09-04 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
US6245227B1 (en) 1998-09-17 2001-06-12 Kionix, Inc. Integrated monolithic microfabricated electrospray and liquid chromatography system and method
US6673541B1 (en) 1998-09-18 2004-01-06 Micromet Ag DNA amplification of a single cell
US6637463B1 (en) 1998-10-13 2003-10-28 Biomicro Systems, Inc. Multi-channel microfluidic system design with balanced fluid flow distribution
US6086740A (en) 1998-10-29 2000-07-11 Caliper Technologies Corp. Multiplexed microfluidic devices and systems
US6277489B1 (en) 1998-12-04 2001-08-21 The Regents Of The University Of California Support for high performance affinity chromatography and other uses
US6062261A (en) 1998-12-16 2000-05-16 Lockheed Martin Energy Research Corporation MicrofluIdic circuit designs for performing electrokinetic manipulations that reduce the number of voltage sources and fluid reservoirs
WO2000037163A1 (en) 1998-12-23 2000-06-29 Nanogen, Inc. Integrated portable biological detection system
US6150119A (en) 1999-01-19 2000-11-21 Caliper Technologies Corp. Optimized high-throughput analytical system
WO2000047998A1 (en) 1999-02-10 2000-08-17 Cell Works Inc. Class characterization of circulating cancer cells isolated from body fluids and methods of use
AU3609900A (en) 1999-03-02 2000-09-21 Qualigen, Inc. Methods and apparatus for separation of biological fluids
US6942978B1 (en) 1999-03-03 2005-09-13 The Board Of Trustees Of The University Of Arkansas Transmembrane serine protease overexpressed in ovarian carcinoma and uses thereof
TW496775B (en) 1999-03-15 2002-08-01 Aviva Bioscience Corp Individually addressable micro-electromagnetic unit array chips
CN1185492C (en) 1999-03-15 2005-01-19 清华大学 Single-point strobed micro electromagnetic units array chip or electromagnetic biologic chip and application thereof
US6858439B1 (en) 1999-03-15 2005-02-22 Aviva Biosciences Compositions and methods for separation of moieties on chips
US6306578B1 (en) 1999-03-19 2001-10-23 Genencor International, Inc. Multi-through hole testing plate for high throughput screening
US6368562B1 (en) 1999-04-16 2002-04-09 Orchid Biosciences, Inc. Liquid transportation system for microfluidic device
US6942771B1 (en) 1999-04-21 2005-09-13 Clinical Micro Sensors, Inc. Microfluidic systems in the electrochemical detection of target analytes
US6174683B1 (en) 1999-04-26 2001-01-16 Biocept, Inc. Method of making biochips and the biochips resulting therefrom
US6589791B1 (en) 1999-05-20 2003-07-08 Cartesian Technologies, Inc. State-variable control system
JP2003500098A (en) 1999-05-21 2003-01-07 ザ ボード オブ トラスティーズ オブ レランド スタンフォード ジュニア ユニバーシティ Microfluidic device and method of generating pulsed micro liquid jet in liquid environment
US6635163B1 (en) 1999-06-01 2003-10-21 Cornell Research Foundation, Inc. Entropic trapping and sieving of molecules
US6664104B2 (en) 1999-06-25 2003-12-16 Cepheid Device incorporating a microfluidic chip for separating analyte from a sample
US6818395B1 (en) 1999-06-28 2004-11-16 California Institute Of Technology Methods and apparatus for analyzing polynucleotide sequences
US6395232B1 (en) 1999-07-09 2002-05-28 Orchid Biosciences, Inc. Fluid delivery system for a microfluidic device using a pressure pulse
US6524456B1 (en) 1999-08-12 2003-02-25 Ut-Battelle, Llc Microfluidic devices for the controlled manipulation of small volumes
US6762059B2 (en) 1999-08-13 2004-07-13 U.S. Genomics, Inc. Methods and apparatuses for characterization of single polymers
US6623945B1 (en) 1999-09-16 2003-09-23 Motorola, Inc. System and method for microwave cell lysing of small samples
FR2798673B1 (en) 1999-09-16 2004-05-28 Exonhit Therapeutics Sa METHODS AND COMPOSITIONS FOR DETECTION OF PATHOLOGICAL EVENTS
US20030113528A1 (en) 1999-09-17 2003-06-19 Wilson Moya Patterned porous structures
EP1218547A4 (en) 1999-10-15 2005-04-20 Ventana Med Syst Inc Method of detecting single gene copies in-situ
AU4504001A (en) 1999-11-04 2001-06-04 Princeton University Electrodeless dielectrophoresis for polarizable particles
AU1592501A (en) 1999-11-10 2001-06-06 Massachusetts Institute Of Technology Cell analysis and sorting apparatus for manipulation of cells
US20060128006A1 (en) 1999-11-10 2006-06-15 Gerhardt Antimony L Hydrodynamic capture and release mechanisms for particle manipulation
US6692952B1 (en) 1999-11-10 2004-02-17 Massachusetts Institute Of Technology Cell analysis and sorting apparatus for manipulation of cells
US6361958B1 (en) 1999-11-12 2002-03-26 Motorola, Inc. Biochannel assay for hybridization with biomaterial
US6875619B2 (en) 1999-11-12 2005-04-05 Motorola, Inc. Microfluidic devices comprising biochannels
WO2001040786A1 (en) 1999-12-01 2001-06-07 The Regents Of The University Of California Electric-field-assisted fluidic assembly of inorganic and organic materials, molecules and like small things including living cells
US6309889B1 (en) 1999-12-23 2001-10-30 Glaxo Wellcome Inc. Nano-grid micro reactor and methods
WO2001049874A1 (en) 2000-01-06 2001-07-12 Caliper Technologies Corp. Methods and systems for monitoring intracellular binding reactions
WO2001051668A1 (en) 2000-01-13 2001-07-19 Immunivest Corporation Ferrofluid based arrays
WO2001071026A2 (en) 2000-03-20 2001-09-27 Adnagen Ag Kit, method and microarray for determining the sex of a human foetus
CA2404431C (en) 2000-03-27 2011-06-07 Thomas Jefferson University Guanylyl cyclase c in the detection of stomach and esophageal cancers
EP1268554A2 (en) 2000-03-31 2003-01-02 IPF Pharmaceuticals GmbH Diagnostic and medicament for analysing the cell surface proteome of tumour and inflammatory cells and for treating tumorous and inflammatory diseases, preferably using a specific chemokine receptor analysis and the chemokine receptor-ligand interaction
US20030170631A1 (en) 2000-04-03 2003-09-11 Corixa Corporation Methods, compositions and kits for the detection and monitoring of breast cancer
AU2001253079A1 (en) 2000-04-03 2001-10-15 Corixa Corporation Methods, compositions and kits for the detection and monitoring of breast cancer
US6716620B2 (en) 2000-04-17 2004-04-06 Purdue Research Foundation Biosensor and related method
GB0009784D0 (en) 2000-04-20 2000-06-07 Simeg Limited Methods for clinical diagnosis
WO2001081621A2 (en) 2000-04-20 2001-11-01 Adnagen Ag Method, diagnostic kit and microarray for determining the rhesus factor
DE10019877A1 (en) 2000-04-20 2001-10-25 Clariant Gmbh Detergents and cleaning agents containing bleach-active dendrimer ligands and their metal complexes
US7641856B2 (en) 2004-05-14 2010-01-05 Honeywell International Inc. Portable sample analyzer with removable cartridge
JP2004532968A (en) 2000-06-14 2004-10-28 ボード・オブ・リージェンツ,ザ・ユニヴァーシティ・オヴ・テキサス・システム Systems and methods for cell subpopulation analysis
ATE319087T1 (en) 2000-06-21 2006-03-15 Bioarray Solutions Ltd MULTI-ANALYTICAL MOLECULAR ANALYSIS BY USING APPLICATION-SPECIFIC RANDOM PARTICLE ARRAYS
KR100845638B1 (en) 2000-07-17 2008-07-10 엡슨 토요콤 가부시키가이샤 Piezoelectric oscillator
DE10035433C2 (en) 2000-07-20 2002-07-18 Tuma Wolfgang Gentle high enrichment of fetal cells from peripheral blood and use of the same
EP1309863A1 (en) * 2000-08-08 2003-05-14 Aviva Biosciences Corporation Methods for manipulating moieties in microfluidic systems
US6610499B1 (en) 2000-08-31 2003-08-26 The Regents Of The University Of California Capillary array and related methods
US6818424B2 (en) 2000-09-01 2004-11-16 E. I. Du Pont De Nemours And Company Production of cyclic terpenoids
JP5433840B2 (en) 2000-09-09 2014-03-05 ザ・リサーチ・ファウンデーション・フォー・ザ・ステイト・ユニヴァーシティ・オブ・ニューヨーク Methods and compositions for isolating metastatic cancer cells and their use in measuring the metastatic potential of cancer
US20020164825A1 (en) 2000-09-09 2002-11-07 Wen-Tien Chen Cell separation matrix
EP1334347A1 (en) 2000-09-15 2003-08-13 California Institute Of Technology Microfabricated crossflow devices and methods
CA2421828A1 (en) 2000-09-30 2002-04-11 Xiao-Bo Wang Apparatuses containing multiple force generating elements and uses thereof
US6689615B1 (en) 2000-10-04 2004-02-10 James Murto Methods and devices for processing blood samples
CA2424996A1 (en) 2000-10-09 2002-04-18 Aviva Biosciences Corporation Compositions and methods for separation of moieties on chips
CA2424941A1 (en) 2000-10-10 2002-04-18 Aviva Biosciences Corporation An integrated biochip system for sample preparation and analysis
US20050100951A1 (en) 2000-10-26 2005-05-12 Biocept, Inc. 3D format biochips and method of use
US20020115163A1 (en) 2000-11-13 2002-08-22 Genoptix Methods for sorting particles by size and elasticity
US20020123112A1 (en) 2000-11-13 2002-09-05 Genoptix Methods for increasing detection sensitivity in optical dielectric sorting systems
US6744038B2 (en) 2000-11-13 2004-06-01 Genoptix, Inc. Methods of separating particles using an optical gradient
US6784420B2 (en) 2000-11-13 2004-08-31 Genoptix, Inc. Method of separating particles using an optical gradient
US20030007894A1 (en) 2001-04-27 2003-01-09 Genoptix Methods and apparatus for use of optical forces for identification, characterization and/or sorting of particles
US20020108859A1 (en) 2000-11-13 2002-08-15 Genoptix Methods for modifying interaction between dielectric particles and surfaces
US6833542B2 (en) 2000-11-13 2004-12-21 Genoptix, Inc. Method for sorting particles
AU2002243263A1 (en) 2000-11-15 2002-07-24 Roche Diagnostics Corporation Methods and reagents for identifying rare fetal cells in the material circulation
US6521188B1 (en) 2000-11-22 2003-02-18 Industrial Technology Research Institute Microfluidic actuator
WO2002044689A2 (en) 2000-11-28 2002-06-06 The Regents Of The University Of California Storing microparticles in optical switch which is transported by micro-fluidic device
US6495340B2 (en) 2000-11-28 2002-12-17 Medis El Ltd. Cell carrier grids
US6778724B2 (en) 2000-11-28 2004-08-17 The Regents Of The University Of California Optical switching and sorting of biological samples and microparticles transported in a micro-fluidic device, including integrated bio-chip devices
US6849423B2 (en) 2000-11-29 2005-02-01 Picoliter Inc Focused acoustics for detection and sorting of fluid volumes
US6893836B2 (en) 2000-11-29 2005-05-17 Picoliter Inc. Spatially directed ejection of cells from a carrier fluid
US20020064808A1 (en) 2000-11-29 2002-05-30 Mutz Mitchell W. Focused acoustic energy for ejecting cells from a fluid
US20020076354A1 (en) * 2000-12-01 2002-06-20 Cohen David Samuel Apparatus and methods for separating components of particulate suspension
FR2817967B1 (en) 2000-12-08 2003-02-28 Diagast PROCESS FOR MAGNETIZING CHEMICAL OR BIOLOGICAL MARKERS
US6770434B2 (en) 2000-12-29 2004-08-03 The Provost, Fellows And Scholars Of The College Of The Holy & Undivided Trinity Of Queen Elizabeth Near Dublin Biological assay method
US7205157B2 (en) 2001-01-08 2007-04-17 Becton, Dickinson And Company Method of separating cells from a sample
US6453928B1 (en) 2001-01-08 2002-09-24 Nanolab Ltd. Apparatus, and method for propelling fluids
US20020160363A1 (en) 2001-01-31 2002-10-31 Mcdevitt John T. Magnetic-based placement and retention of sensor elements in a sensor array
US20020106715A1 (en) 2001-02-02 2002-08-08 Medisel Ltd System and method for collecting data from individual cells
US20020110835A1 (en) 2001-02-13 2002-08-15 Rajan Kumar Microfluidic devices and methods
WO2002065515A2 (en) 2001-02-14 2002-08-22 Science & Technology Corporation @ Unm Nanostructured devices for separation and analysis
US6913697B2 (en) 2001-02-14 2005-07-05 Science & Technology Corporation @ Unm Nanostructured separation and analysis devices for biological membranes
US20030190602A1 (en) 2001-03-12 2003-10-09 Monogen, Inc. Cell-based detection and differentiation of disease states
CA2440753A1 (en) 2001-03-12 2002-09-19 Monogen, Inc Cell-based detection and differentiation of disease states
US7323140B2 (en) 2001-03-28 2008-01-29 Handylab, Inc. Moving microdroplets in a microfluidic device
EP1377811B1 (en) 2001-04-03 2008-07-16 Micronics, Inc. Split focusing cytometer
US20020173043A1 (en) 2001-04-04 2002-11-21 Eddine Merabet Cyanide-free reagent, and method for detecting hemoglobin
US20030036100A1 (en) 2001-04-10 2003-02-20 Imperial College Innovations Ltd. Simultaneous determination of phenotype and genotype
US7713705B2 (en) 2002-12-24 2010-05-11 Biosite, Inc. Markers for differential diagnosis and methods of use thereof
FR2824144B1 (en) 2001-04-30 2004-09-17 Metagenex S A R L METHOD OF PRENATAL DIAGNOSIS ON FETAL CELLS ISOLATED FROM MATERNAL BLOOD
US6805841B2 (en) 2001-05-09 2004-10-19 The Provost Fellows And Scholars Of The College Of The Holy And Undivided Trinity Of Queen Elizabeth Near Dublin Liquid pumping system
WO2002090505A2 (en) 2001-05-09 2002-11-14 Virginia Commonwealth University Multiple sequencible and ligatible structures for genomic analysis
US20020166760A1 (en) * 2001-05-11 2002-11-14 Prentiss Mara G. Micromagentic systems and methods for microfluidics
US6743636B2 (en) 2001-05-24 2004-06-01 Industrial Technology Research Institute Microfluid driving device
DE10127079A1 (en) 2001-06-02 2002-12-12 Ulrich Pachmann Method for the quantitative detection of vital epithelial tumor cells in a body fluid
AU2002310501A1 (en) 2001-06-20 2003-01-08 Cytonome, Inc. Microfluidic system including a virtual wall fluid interface port for interfacing fluids with the microfluidic system
US7419574B2 (en) 2001-06-20 2008-09-02 Cummings Eric B Dielectrophoresis device and method having non-uniform arrays for manipulating particles
US20060019235A1 (en) 2001-07-02 2006-01-26 The Board Of Trustees Of The Leland Stanford Junior University Molecular and functional profiling using a cellular microarray
US20040108208A1 (en) 2001-08-03 2004-06-10 Kazuhiro Iida Separation apparatus and process for fabricating separation apparatus
CA2396408C (en) 2001-08-03 2006-03-28 Nec Corporation Fractionating apparatus having colonies of pillars arranged in migration passage at interval and process for fabricating pillars
DE60227678D1 (en) 2001-08-23 2008-08-28 Immunivest Corp ANALYSIS OF CIRCULATING TUMOR CELLS, FRAGMENTS AND TRUMS
US7863012B2 (en) 2004-02-17 2011-01-04 Veridex, Llc Analysis of circulating tumor cells, fragments, and debris
EP1483052B1 (en) 2001-08-28 2010-12-22 Gyros Patent Ab Retaining microfluidic microcavity and other microfluidic structures
ES2284911T3 (en) 2001-09-04 2007-11-16 Iq Corporation B.V. DETERMINATION AND QUANTIFICATION OF RED BALLOON POPULATIONS IN SAMPLES.
WO2003023057A2 (en) 2001-09-06 2003-03-20 Adnagen Ag Method and diagnosis kit for selecting and or qualitative and/or quantitative detection of cells
DE10143776A1 (en) 2001-09-06 2003-04-03 Adnagen Ag Selection and determination of specific cells, useful particularly for diagnosis and monitoring of tumors, by antibody-mediated selection then detecting specific mRNA
US7202045B2 (en) 2001-09-19 2007-04-10 Regents Of The University Of Michigan Detection and treatment of cancers of the lung
US8980568B2 (en) 2001-10-11 2015-03-17 Aviva Biosciences Corporation Methods and compositions for detecting non-hematopoietic cells from a blood sample
US20030072682A1 (en) 2001-10-11 2003-04-17 Dan Kikinis Method and apparatus for performing biochemical testing in a microenvironment
US6783647B2 (en) 2001-10-19 2004-08-31 Ut-Battelle, Llc Microfluidic systems and methods of transport and lysis of cells and analysis of cell lysate
AU2002360305A1 (en) 2001-10-26 2003-05-06 Immunivest Corporation Multiparameter analysis of comprehensive nucleic acids and morphological features on the same sample
AU2002359340A1 (en) 2001-10-31 2003-05-12 Ventrigraft Inc Methods and device compositions for the recruitment of cells to blood contacting surfaces in vivo
EP1527193A2 (en) 2001-11-07 2005-05-04 Origene Technologies Inc. Prostate cancer genes
US20030232350A1 (en) 2001-11-13 2003-12-18 Eos Biotechnology, Inc. Methods of diagnosis of cancer, compositions and methods of screening for modulators of cancer
US20050244843A1 (en) 2001-11-16 2005-11-03 Wen-Tien Chen Blood test prototypes and methods for the detection of circulating tumor and endothelial cells
WO2003044519A1 (en) * 2001-11-20 2003-05-30 Nec Corporation Separation apparatus, method of separation, and process for producing separation apparatus
US20050118591A1 (en) 2001-11-22 2005-06-02 Adnagen Ag Diagnosis kit, dna chip, and methods for diagnosing or supervising the treatment of testicular cancer
AU2002357588B2 (en) 2001-12-11 2008-11-20 Netech Inc. Blood cell separation system
US20040241653A1 (en) 2001-12-31 2004-12-02 Elena Feinstein Methods for identifying marker genes for cancer
EP1466162A1 (en) 2002-01-10 2004-10-13 Board Of Regents The University Of Texas System Flow sorting system and methods regarding same
US7383134B2 (en) 2002-01-15 2008-06-03 Piper James R Method and/or system for analyzing biological samples using a computer system
AU2003216175A1 (en) 2002-02-04 2003-09-02 Colorado School Of Mines Laminar flow-based separations of colloidal and cellular particles
EP1474772A4 (en) 2002-02-14 2005-11-09 Immunivest Corp Methods and algorithms for cell enumeration in a low-cost cytometer
US7053967B2 (en) 2002-05-23 2006-05-30 Planar Systems, Inc. Light sensitive display
FR2836072B1 (en) 2002-02-21 2004-11-12 Commissariat Energie Atomique COMPONENT USING COMPOSITE MATERIAL FOR MICROSYSTEM FOR BIOLOGICAL OR BIOCHEMICAL ANALYSIS
FR2836071B1 (en) 2002-02-21 2005-02-04 Commissariat Energie Atomique COMPONENT FOR MICROSYSTEM FOR BIOLOGICAL OR BIOCHEMICAL ANALYSIS
US6958119B2 (en) 2002-02-26 2005-10-25 Agilent Technologies, Inc. Mobile phase gradient generation microfluidic device
US7223371B2 (en) 2002-03-14 2007-05-29 Micronics, Inc. Microfluidic channel network device
SE0200860D0 (en) 2002-03-20 2002-03-20 Monica Almqvist Microfluidic cell and method for sample handling
WO2004023105A2 (en) 2002-03-20 2004-03-18 Advanced Sensor Technologies, Inc. Personal monitor to detect exposure to toxic agents
EP2666849A3 (en) 2002-04-01 2014-05-28 Fluidigm Corporation Microfluidic particle-analysis systems
US7312085B2 (en) 2002-04-01 2007-12-25 Fluidigm Corporation Microfluidic particle-analysis systems
US20040241707A1 (en) 2002-04-01 2004-12-02 Gao Chun L. Enhanced diagnostic potential of prostate-specific antigen expressing cells
US7141369B2 (en) 2002-04-25 2006-11-28 Semibio Technology, Inc. Measuring cellular metabolism of immobilized cells
AU2003234459A1 (en) 2002-05-03 2003-11-17 Immunivest Corporation Device and method for analytical cell imaging
WO2003099312A1 (en) 2002-05-27 2003-12-04 Östergötlands Läns Landsting Method for determining immune system affecting compounds
SE0201738D0 (en) * 2002-06-07 2002-06-07 Aamic Ab Micro-fluid structures
US20040005247A1 (en) 2002-07-03 2004-01-08 Nanostream, Inc. Microfluidic closed-end metering systems and methods
US20040101444A1 (en) 2002-07-15 2004-05-27 Xeotron Corporation Apparatus and method for fluid delivery to a hybridization station
US20040018611A1 (en) 2002-07-23 2004-01-29 Ward Michael Dennis Microfluidic devices for high gradient magnetic separation
US7214348B2 (en) 2002-07-26 2007-05-08 Applera Corporation Microfluidic size-exclusion devices, systems, and methods
US7118676B2 (en) * 2003-09-04 2006-10-10 Arryx, Inc. Multiple laminar flow-based particle and cellular separation with laser steering
US9435799B2 (en) 2002-07-31 2016-09-06 Janssen Diagnostics, Inc. Methods and reagents for improved selection of biological materials
WO2004015411A1 (en) 2002-08-08 2004-02-19 Nanostream, Inc. Systems and methods for high-throughput microfluidic sample analysis
US20060008807A1 (en) 2002-08-23 2006-01-12 O'hara Shawn M Multiparameter analysis of comprehensive nucleic acids and morphological features on the same sample
US20040043506A1 (en) 2002-08-30 2004-03-04 Horst Haussecker Cascaded hydrodynamic focusing in microfluidic channels
US6878271B2 (en) 2002-09-09 2005-04-12 Cytonome, Inc. Implementation of microfluidic components in a microfluidic system
US7455770B2 (en) 2002-09-09 2008-11-25 Cytonome, Inc. Implementation of microfluidic components in a microfluidic system
US7094345B2 (en) 2002-09-09 2006-08-22 Cytonome, Inc. Implementation of microfluidic components, including molecular fractionation devices, in a microfluidic system
US6806543B2 (en) 2002-09-12 2004-10-19 Intel Corporation Microfluidic apparatus with integrated porous-substrate/sensor for real-time (bio)chemical molecule detection
AU2003277153A1 (en) 2002-09-27 2004-04-19 The General Hospital Corporation Microfluidic device for cell separation and uses thereof
JP2006500074A (en) 2002-09-27 2006-01-05 オリディス・ビオメド・フォルシュングス−ウント・エントヴィックルングス・ゲーエムベーハー Polypeptides and nucleic acids encoding them and their use for prevention, diagnosis or treatment of liver damage and epithelial cancer
DE60332046D1 (en) 2002-10-23 2010-05-20 Univ Princeton CONTINUOUS PARTICLE SEPARATION METHOD USING FIELD ASYMMETRICALLY LOCATED OBSTACLE ARRAYS
US6811385B2 (en) 2002-10-31 2004-11-02 Hewlett-Packard Development Company, L.P. Acoustic micro-pump
US7122384B2 (en) 2002-11-06 2006-10-17 E. I. Du Pont De Nemours And Company Resonant light scattering microparticle methods
AU2002952696A0 (en) 2002-11-14 2002-11-28 Genomics Research Partners Pty Ltd Status determination
JP2004354364A (en) * 2002-12-02 2004-12-16 Nec Corp Fine particle manipulating unit, chip mounted with the same and detector, and method for separating, capturing and detecting protein
DE10259703A1 (en) 2002-12-19 2004-07-08 Ivonex Gmbh separation process
US6746503B1 (en) 2003-01-30 2004-06-08 The Regents Of The University Of California Precision gap particle separator
JP4593557B2 (en) 2003-02-27 2010-12-08 ベリデックス・リミテッド・ライアビリティ・カンパニー Circulating tumor cells (CTC): early assessment of time to progression, survival and response to therapy in patients with metastatic cancer
DE10313201A1 (en) 2003-03-21 2004-10-07 Steag Microparts Gmbh Microstructured separator and microfluidic process for separating liquid components from a liquid containing particles
US20040197832A1 (en) 2003-04-03 2004-10-07 Mor Research Applications Ltd. Non-invasive prenatal genetic diagnosis using transcervical cells
US7319010B2 (en) 2003-05-12 2008-01-15 The Regents Of The University Of Michigan Detection and treatment of cancers of the colon
US6962658B2 (en) 2003-05-20 2005-11-08 Eksigent Technologies, Llc Variable flow rate injector
RU2005141456A (en) 2003-06-06 2006-06-10 Майкроникс, Инк. (Us) SYSTEM AND METHOD OF HEATING, COOLING AND THERMAL CYCLING IN A MICRO-LIQUID DEVICE
EP1636564A1 (en) 2003-06-13 2006-03-22 The General Hospital Corporation Microfluidic systems for size based removal of red blood cells and platelets from blood
JP2005037346A (en) * 2003-06-25 2005-02-10 Aisin Seiki Co Ltd Micro fluid control system
US7745221B2 (en) 2003-08-28 2010-06-29 Celula, Inc. Methods and apparatus for sorting cells using an optical switch in a microfluidic channel network
WO2005042759A2 (en) 2003-09-10 2005-05-12 Althea Technologies, Inc. Expression profiling using microarrays
WO2005028663A2 (en) 2003-09-18 2005-03-31 Immunivest Corporation Operator independent programmable sample preparation and analysis system
US8062849B2 (en) 2003-10-28 2011-11-22 The Johns Hopkins University Quantitative multiplex methylation-specific PCR
BRPI0415886A (en) 2003-10-29 2007-01-09 Mec Dynamics Corp disposable strips and method of determining hemoglobin percentage of hbalc
WO2005043121A2 (en) 2003-10-31 2005-05-12 Vitatex, Inc. Blood test prototypes and methods for the detection of circulating tumor and endothelial cells
WO2005049168A2 (en) 2003-11-17 2005-06-02 Immunivest Corporation Method and apparatus for pre-enrichment and recovery of cells from densified whole blood
US7329391B2 (en) 2003-12-08 2008-02-12 Applera Corporation Microfluidic device and material manipulating method using same
EP1697399B1 (en) 2003-12-12 2016-11-23 GOVERNMENT OF THE UNITED STATES OF AMERICA, as repr. by THE SECR. OF THE DEPT. OF HEALTH AND HUMAN SERVICES AND HIS SUCCESSORS A human cytotoxic t-lymphocyte epitope and its agonist epitope from the non-variable number of tandem repeat sequence of muc-1
US7939249B2 (en) 2003-12-24 2011-05-10 3M Innovative Properties Company Methods for nucleic acid isolation and kits using a microfluidic device and concentration step
US20050147977A1 (en) 2003-12-29 2005-07-07 Tae-Woong Koo Methods and compositions for nucleic acid detection and sequence analysis
ES2543833T3 (en) 2004-01-07 2015-08-24 Novartis Vaccines And Diagnostics, Inc. M-CSF-specific monoclonal antibody and uses thereof
EP1561507A1 (en) 2004-01-27 2005-08-10 Future Diagnostics B.V. System for characterising a fluid, microfluidic device for characterising or analysing concentration components, a method of characterising or analysing such concentrations and a measurement device
WO2005072399A2 (en) 2004-01-29 2005-08-11 Massachusetts Institute Of Technology Microscale sorting cytometer
US20050181353A1 (en) 2004-02-17 2005-08-18 Rao Galla C. Stabilization of cells and biological specimens for analysis
US20050191636A1 (en) 2004-03-01 2005-09-01 Biocept, Inc. Detection of STRP, such as fragile X syndrome
CA2557819A1 (en) 2004-03-03 2005-09-15 The General Hospital Corporation Magnetic device for isolation of cells and biomolecules in a microfluidic environment
US20060121624A1 (en) 2004-03-03 2006-06-08 Huang Lotien R Methods and systems for fluid delivery
WO2005085861A2 (en) 2004-03-03 2005-09-15 Oridis Biomed Forschungs- Und Entwicklungs Gmbh Nucleic acids and encoded polypeptides for use in liver disorders and epithelial cancer
EP1765503A2 (en) 2004-03-03 2007-03-28 The General Hospital Corporation System for delivering a diluted solution
JP2007529203A (en) 2004-03-12 2007-10-25 ザ・レジェンツ・オブ・ザ・ユニバーシティ・オブ・カリフォルニア Integrated cell manipulation and measurement method and apparatus
SE0400662D0 (en) * 2004-03-24 2004-03-24 Aamic Ab Assay device and method
US7390388B2 (en) 2004-03-25 2008-06-24 Hewlett-Packard Development Company, L.P. Method of sorting cells on a biodevice
WO2005098046A2 (en) 2004-04-01 2005-10-20 Immunivest Corporation Methods for the determination of cell specific biomarkers
WO2005108621A1 (en) 2004-04-30 2005-11-17 Yale University Methods and compositions for cancer diagnosis
JP2008500611A (en) 2004-05-03 2008-01-10 サイジーン ラボラトリーズ インク. Method and system for providing anonymous testing and reporting based on a comprehensive knowledge database and selective access to testing results and reports
US7468249B2 (en) 2004-05-05 2008-12-23 Biocept, Inc. Detection of chromosomal disorders
WO2005108963A1 (en) 2004-05-06 2005-11-17 Nanyang Technological University Microfluidic cell sorter system
US7858040B2 (en) 2004-05-07 2010-12-28 Saryna Biotechnologies Llc Direct mixing and injection for high throughput fluidic systems
US20050252840A1 (en) 2004-05-13 2005-11-17 Eksigent Technologies, Llc Micromixer
US7622281B2 (en) 2004-05-20 2009-11-24 The Board Of Trustees Of The Leland Stanford Junior University Methods and compositions for clonal amplification of nucleic acid
WO2005116264A2 (en) 2004-05-24 2005-12-08 Immunivest Corporation A blood test to monitor the genetic changes of progressive cancer using immunomagnetic enrichment and fluorescence in situ hybridization (fish)
EP1607485A1 (en) 2004-06-14 2005-12-21 Institut National De La Sante Et De La Recherche Medicale (Inserm) Method for quantifying VEGF121 isoform in a biological sample
DE102004036669A1 (en) 2004-07-28 2006-03-23 Otto Bock Healthcare Gmbh Pump with a closed with at least one flexible wall fluid volume
US20060051265A1 (en) 2004-09-08 2006-03-09 Health Research, Inc. Apparatus and method for sorting microstructures in a fluid medium
WO2006035846A1 (en) 2004-09-30 2006-04-06 Dainippon Ink And Chemicals, Inc. Process for producing porous sintered metal
DE102004047953A1 (en) 2004-10-01 2006-04-20 Rudolf Rigler Selection of particle possessing predetermined property from population encompassing multiplicity of different particles, comprises providing population of different particles, and labeling particles which possess predetermined property
WO2006076567A2 (en) 2005-01-13 2006-07-20 Micronics, Inc. Microfluidic rare cell detection device
JP2008526251A (en) 2005-01-18 2008-07-24 バイオセプト インコーポレイティッド Cell separation using microchannels with patterned posts
US20060252087A1 (en) 2005-01-18 2006-11-09 Biocept, Inc. Recovery of rare cells using a microchannel apparatus with patterned posts
US8158410B2 (en) * 2005-01-18 2012-04-17 Biocept, Inc. Recovery of rare cells using a microchannel apparatus with patterned posts
US7981696B2 (en) 2005-02-18 2011-07-19 The United States of America, as represented by the Secretary of Commerce, The National Institute of Standards and Technology Microfluidic platform of arrayed switchable spin-valve elements for high-throughput sorting and manipulation of magnetic particles and biomolecules
EP1874920A4 (en) 2005-04-05 2009-11-04 Cellpoint Diagnostics Devices and methods for enrichment and alteration of circulating tumor cells and other particles
US20070026414A1 (en) 2005-07-29 2007-02-01 Martin Fuchs Devices and methods for enrichment and alteration of circulating tumor cells and other particles
US20070026413A1 (en) 2005-07-29 2007-02-01 Mehmet Toner Devices and methods for enrichment and alteration of circulating tumor cells and other particles
US20070026418A1 (en) 2005-07-29 2007-02-01 Martin Fuchs Devices and methods for enrichment and alteration of circulating tumor cells and other particles
US20070196820A1 (en) 2005-04-05 2007-08-23 Ravi Kapur Devices and methods for enrichment and alteration of cells and other particles
US20070026417A1 (en) 2005-07-29 2007-02-01 Martin Fuchs Devices and methods for enrichment and alteration of circulating tumor cells and other particles
US20070026415A1 (en) 2005-07-29 2007-02-01 Martin Fuchs Devices and methods for enrichment and alteration of circulating tumor cells and other particles
US20060223178A1 (en) 2005-04-05 2006-10-05 Tom Barber Devices and methods for magnetic enrichment of cells and other particles
WO2006119569A1 (en) * 2005-05-11 2006-11-16 Genetic Technologies Limtied Methods of enriching fetal cells
US20070026419A1 (en) 2005-07-29 2007-02-01 Martin Fuchs Devices and methods for enrichment and alteration of circulating tumor cells and other particles
US20070059680A1 (en) 2005-09-15 2007-03-15 Ravi Kapur System for cell enrichment
US20070026416A1 (en) 2005-07-29 2007-02-01 Martin Fuchs Devices and methods for enrichment and alteration of circulating tumor cells and other particles
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
US20070059716A1 (en) 2005-09-15 2007-03-15 Ulysses Balis Methods for detecting fetal abnormality
CA2622745A1 (en) 2005-09-15 2007-03-29 Artemis Health, Inc. Systems and methods for enrichment of analytes
US20070059683A1 (en) 2005-09-15 2007-03-15 Tom Barber Veterinary diagnostic system
US20070059781A1 (en) 2005-09-15 2007-03-15 Ravi Kapur System for size based separation and analysis
US20070059718A1 (en) 2005-09-15 2007-03-15 Mehmet Toner Systems and methods for enrichment of analytes
US20070059774A1 (en) 2005-09-15 2007-03-15 Michael Grisham Kits for Prenatal Testing
US20070059719A1 (en) 2005-09-15 2007-03-15 Michael Grisham Business methods for prenatal Diagnosis
EP1943354A2 (en) * 2005-10-21 2008-07-16 Monaliza Medical Ltd. Methods and kits for analyzing genetic material of a fetus

Patent Citations (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5300779A (en) * 1985-08-05 1994-04-05 Biotrack, Inc. Capillary flow device
US5840502A (en) * 1994-08-31 1998-11-24 Activated Cell Therapy, Inc. Methods for enriching specific cell-types by density gradient centrifugation
US5837200A (en) * 1995-06-02 1998-11-17 Bayer Aktiengesellschaft Sorting device for biological cells or viruses
US20020119482A1 (en) * 1996-07-30 2002-08-29 Aclara Biosciences, Inc. Microfluidic method for nucleic acid purification and processing
US6030581A (en) * 1997-02-28 2000-02-29 Burstein Laboratories Laboratory in a disk
US6368871B1 (en) * 1997-08-13 2002-04-09 Cepheid Non-planar microstructures for manipulation of fluid samples
US6027623A (en) * 1998-04-22 2000-02-22 Toyo Technologies, Inc. Device and method for electrophoretic fraction
US20040053352A1 (en) * 1998-09-28 2004-03-18 Tianmei Ouyang Diagnostics based on tetrazolium compounds
US6636498B1 (en) * 1999-01-08 2003-10-21 Cisco Technology, Inc. Mobile IP mobile router
US6632655B1 (en) * 1999-02-23 2003-10-14 Caliper Technologies Corp. Manipulation of microparticles in microfluidic systems
US6511967B1 (en) * 1999-04-23 2003-01-28 The General Hospital Corporation Use of an internalizing transferrin receptor to image transgene expression
US20040048360A1 (en) * 1999-08-26 2004-03-11 Caliper Technologies Corp. Microfluidic analytic detection assays, devices, and integrated systems
US20030091476A1 (en) * 2000-07-03 2003-05-15 Xiaochuan Zhou Fluidic methods and devices for parallel chemical reactions
US20040005582A1 (en) * 2000-08-10 2004-01-08 Nanobiodynamics, Incorporated Biospecific desorption microflow systems and methods for studying biospecific interactions and their modulators
US20030040119A1 (en) * 2001-04-11 2003-02-27 The Regents Of The University Of Michigan Separation devices and methods for separating particles
US20030134416A1 (en) * 2001-10-11 2003-07-17 Douglas Yamanishi Methods, compositions, and automated systems for separating rare cells from fluid samples
US20040142463A1 (en) * 2001-10-11 2004-07-22 George Walker Methods, compositions, and automated systems for separating rare cells from fluid samples
US20040019300A1 (en) * 2002-07-26 2004-01-29 Leonard Leslie Anne Microfluidic blood sample separations
US20060000772A1 (en) * 2002-11-29 2006-01-05 Toru Sano Separation apparatus and separation method
US7277876B2 (en) * 2004-01-23 2007-10-02 Solomon Research Llc Dynamic adaptive distributed computer system
US20070202525A1 (en) * 2006-02-02 2007-08-30 The Board Of Trustees Of The Leland Stanford Junior University Non-invasive fetal genetic screening by digital analysis
US20080038733A1 (en) * 2006-03-28 2008-02-14 Baylor College Of Medicine Screening for down syndrome
US20080023399A1 (en) * 2006-06-01 2008-01-31 Inglis David W Apparatus and method for continuous particle separation

Cited By (110)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8304230B2 (en) 2002-09-27 2012-11-06 The General Hospital Corporation Microfluidic device for cell separation and uses thereof
US8895298B2 (en) 2002-09-27 2014-11-25 The General Hospital Corporation Microfluidic device for cell separation and uses thereof
US11052392B2 (en) 2002-09-27 2021-07-06 The General Hospital Corporation Microfluidic device for cell separation and uses thereof
US8986966B2 (en) 2002-09-27 2015-03-24 The General Hospital Corporation Microfluidic device for cell separation and uses thereof
US20070172903A1 (en) * 2002-09-27 2007-07-26 The General Hospital Corporation Microfluidic device for cell separation and uses thereof
US20070231851A1 (en) * 2002-09-27 2007-10-04 The General Hospital Corporation Microfluidic device for cell separation and uses thereof
US10081014B2 (en) 2002-09-27 2018-09-25 The General Hospital Corporation Microfluidic device for cell separation and uses thereof
US10369568B2 (en) 2005-01-18 2019-08-06 Biocept, Inc. Cell separation using microchannel having patterned posts
US20060252087A1 (en) * 2005-01-18 2006-11-09 Biocept, Inc. Recovery of rare cells using a microchannel apparatus with patterned posts
US20060160243A1 (en) 2005-01-18 2006-07-20 Biocept, Inc. Recovery of rare cells using a microchannel apparatus with patterned posts
US20090136982A1 (en) * 2005-01-18 2009-05-28 Biocept, Inc. Cell separation using microchannel having patterned posts
US9212977B2 (en) 2005-01-18 2015-12-15 Biocept, Inc. Cell separation using microchannel having patterned posts
US8158410B2 (en) 2005-01-18 2012-04-17 Biocept, Inc. Recovery of rare cells using a microchannel apparatus with patterned posts
US8585971B2 (en) 2005-04-05 2013-11-19 The General Hospital Corporation Devices and method for enrichment and alteration of cells and other particles
US10786817B2 (en) 2005-04-05 2020-09-29 The General Hospital Corporation Devices and method for enrichment and alteration of cells and other particles
US9956562B2 (en) 2005-04-05 2018-05-01 The General Hospital Corporation Devices and method for enrichment and alteration of cells and other particles
US8021614B2 (en) 2005-04-05 2011-09-20 The General Hospital Corporation Devices and methods for enrichment and alteration of cells and other particles
US9174222B2 (en) 2005-04-05 2015-11-03 The General Hospital Corporation Devices and method for enrichment and alteration of cells and other particles
US20060223178A1 (en) * 2005-04-05 2006-10-05 Tom Barber Devices and methods for magnetic enrichment of cells and other particles
US20100167337A1 (en) * 2006-01-12 2010-07-01 Biocept Inc. Device for cell separation and analysis and method of using
US20070161051A1 (en) * 2006-01-12 2007-07-12 Biocept, Inc. Device for cell separation and analysis and method of using
US7695956B2 (en) 2006-01-12 2010-04-13 Biocept, Inc. Device for cell separation and analysis and method of using
US11781187B2 (en) 2006-06-14 2023-10-10 The General Hospital Corporation Rare cell analysis using sample splitting and DNA tags
US8168389B2 (en) 2006-06-14 2012-05-01 The General Hospital Corporation Fetal cell analysis using sample splitting
US10155984B2 (en) 2006-06-14 2018-12-18 The General Hospital Corporation Rare cell analysis using sample splitting and DNA tags
US10591391B2 (en) 2006-06-14 2020-03-17 Verinata Health, Inc. Diagnosis of fetal abnormalities using polymorphisms including short tandem repeats
US8372584B2 (en) 2006-06-14 2013-02-12 The General Hospital Corporation Rare cell analysis using sample splitting and DNA tags
US9347100B2 (en) 2006-06-14 2016-05-24 Gpb Scientific, Llc Rare cell analysis using sample splitting and DNA tags
US10704090B2 (en) 2006-06-14 2020-07-07 Verinata Health, Inc. Fetal aneuploidy detection by sequencing
US9273355B2 (en) 2006-06-14 2016-03-01 The General Hospital Corporation Rare cell analysis using sample splitting and DNA tags
US8137912B2 (en) 2006-06-14 2012-03-20 The General Hospital Corporation Methods for the diagnosis of fetal abnormalities
US9017942B2 (en) 2006-06-14 2015-04-28 The General Hospital Corporation Rare cell analysis using sample splitting and DNA tags
US11674176B2 (en) 2006-06-14 2023-06-13 Verinata Health, Inc Fetal aneuploidy detection by sequencing
US8784012B2 (en) 2007-04-16 2014-07-22 The General Hospital Corporation Systems and methods for particle focusing in microchannels
US9347595B2 (en) 2007-04-16 2016-05-24 The General Hospital Corporation Systems and methods for particle focusing in microchannels
US9808803B2 (en) 2007-04-16 2017-11-07 The General Hospital Corporation Systems and methods for particle focusing in microchannels
US20090014360A1 (en) * 2007-04-16 2009-01-15 The General Hospital Corporation D/B/A Massachusetts General Hospital Systems and methods for particle focusing in microchannels
US20170101680A1 (en) * 2007-04-16 2017-04-13 Gpb Scientific, Llc Methods for diagnosing, prognosing, or theranosing a condition using rare cells
EP2482055A2 (en) 2007-04-16 2012-08-01 The General Hospital Corporation d/b/a Massachusetts General Hospital Systems and methods for particle focusing in microchannels
US10549278B2 (en) 2007-04-16 2020-02-04 The General Hospital Corporation Systems and methods for particle focusing in microchannels
US8186913B2 (en) * 2007-04-16 2012-05-29 The General Hospital Corporation Systems and methods for particle focusing in microchannels
US8807879B2 (en) 2007-04-16 2014-08-19 The General Hospital Corporation Systems and methods for particle focusing in microchannels
EP2562531A2 (en) 2007-04-16 2013-02-27 The General Hospital Corporation d/b/a Massachusetts General Hospital Systems and methods for particle focusing in microchannels
US20130302797A1 (en) * 2007-04-16 2013-11-14 Anne R. Kopf-Sill Methods for diagnosing, prognosing, or theranosing a condition using rare cells
US11498071B2 (en) 2007-04-16 2022-11-15 The General Hospital Corporation Systems and methods for particle focusing in microchannels
WO2008130655A1 (en) * 2007-04-20 2008-10-30 The General Hospital Corporation Methods for counting cells
US8852875B2 (en) 2007-04-20 2014-10-07 The General Hospital Corporation Methods for counting cells
AP2937A (en) * 2007-04-20 2014-07-31 Gen Hospital Corp Method for counting cells
US10359429B2 (en) 2008-02-25 2019-07-23 Gpb Scientific, Llc Tagged ligands for enrichment of rare analytes from a mixed sample
US9404157B2 (en) 2008-09-20 2016-08-02 The Board Of Trustees Of The Leland Stanford Junior University Noninvasive diagnosis of fetal aneuploidy by sequencing
US8195415B2 (en) 2008-09-20 2012-06-05 The Board Of Trustees Of The Leland Stanford Junior University Noninvasive diagnosis of fetal aneuploidy by sequencing
US8682594B2 (en) 2008-09-20 2014-03-25 The Board Of Trustees Of The Leland Stanford Junior University Noninvasive diagnosis of fetal aneuploidy by sequencing
US10669585B2 (en) 2008-09-20 2020-06-02 The Board Of Trustees Of The Leland Stanford Junior University Noninvasive diagnosis of fetal aneuploidy by sequencing
US9353414B2 (en) 2008-09-20 2016-05-31 The Board Of Trustees Of The Leland Stanford Junior University Noninvasive diagnosis of fetal aneuploidy by sequencing
US8296076B2 (en) 2008-09-20 2012-10-23 The Board Of Trustees Of The Leland Stanford Junior University Noninvasive diagnosis of fetal aneuoploidy by sequencing
WO2010041231A2 (en) 2008-10-10 2010-04-15 Cnrs-Dae Cell sorting device
US20110212440A1 (en) * 2008-10-10 2011-09-01 Cnrs-Dae Cell sorting device
US8162149B1 (en) 2009-01-21 2012-04-24 Sandia Corporation Particle sorter comprising a fluid displacer in a closed-loop fluid circuit
US20100304978A1 (en) * 2009-01-26 2010-12-02 David Xingfei Deng Methods and compositions for identifying a fetal cell
US9726592B2 (en) 2009-12-07 2017-08-08 Yale University Label-free cellular manipulation and sorting via biocompatible ferrofluids
US10782223B2 (en) 2009-12-07 2020-09-22 Yale University Label-free cellular manipulation and sorting via biocompatible ferrofluids
WO2011071912A1 (en) * 2009-12-07 2011-06-16 Yale University Label-free cellular manipulation and sorting via biocompatible ferrofluids
US8961878B2 (en) 2009-12-07 2015-02-24 Yale University Label-free cellular manipulation and sorting via biocompatible ferrofluids
US9217131B2 (en) 2010-01-21 2015-12-22 Biocep Ltd. Magnetic separation of rare cells
US9546392B2 (en) 2010-01-21 2017-01-17 Biocep Ltd. Magnetic separation of rare cells
WO2012048113A2 (en) 2010-10-07 2012-04-12 The General Hospital Corporation Biomarkers of cancer
AU2015268583B2 (en) * 2010-10-28 2017-06-15 Yale University Microfluidic Processing Of Target Species In Ferrofluids
US9999855B2 (en) 2010-10-28 2018-06-19 Yale University Microfluidic processing of target species in ferrofluids
WO2013063035A1 (en) 2011-10-24 2013-05-02 The General Hospital Corporation Biomarkers of cancer
US10324011B2 (en) 2013-03-15 2019-06-18 The Trustees Of Princeton University Methods and devices for high throughput purification
US11486802B2 (en) 2013-03-15 2022-11-01 University Of Maryland, Baltimore Methods and devices for high throughput purification
US11383247B2 (en) 2013-03-15 2022-07-12 Ancera, Llc Systems and methods for active particle separation
US11204350B2 (en) 2013-03-15 2021-12-21 Ancera, Llc Systems and methods for bead-based assays in ferrofluids
US11142746B2 (en) 2013-03-15 2021-10-12 University Of Maryland, Baltimore High efficiency microfluidic purification of stem cells to improve transplants
US11493428B2 (en) 2013-03-15 2022-11-08 Gpb Scientific, Inc. On-chip microfluidic processing of particles
US10852220B2 (en) 2013-03-15 2020-12-01 The Trustees Of Princeton University Methods and devices for high throughput purification
US10203332B2 (en) 2014-08-22 2019-02-12 Samsung Electronics Co., Ltd. Apparatus for and method of separating target matter
US10875021B2 (en) 2014-11-03 2020-12-29 The General Hospital Corporation Sorting particles in a microfluidic device
US10583438B2 (en) 2014-11-03 2020-03-10 The General Hospital Corporation Combined sorting and concentrating particles in a microfluidic device
WO2016073448A1 (en) 2014-11-03 2016-05-12 The General Hospital Corporation Combined sorting and concentrating particles in a microfluidic device
US10478819B2 (en) 2014-11-03 2019-11-19 The General Hospital Corporation Sorting particles in a microfluidic device
US11052393B2 (en) 2014-11-03 2021-07-06 The General Hospital Corporation Concentrating particles in a microfluidic device
US10150116B2 (en) 2014-11-03 2018-12-11 The General Hospital Corporation Concentrating particles in a microfluidic device
US9895694B2 (en) 2014-11-03 2018-02-20 The General Hospital Corporation Sorting particles in a microfluidic device
US9610582B2 (en) 2014-11-03 2017-04-04 The General Hospital Corporation Combined sorting and concentrating particles in a microfluidic device
US11027280B2 (en) 2014-11-03 2021-06-08 The General Hospital Corporation Sorting particles in a microfluidic device
US11446664B2 (en) 2014-11-03 2022-09-20 The General Hospital Corporation Combined sorting and concentrating particles in a microfluidic device
US10677779B2 (en) * 2015-02-24 2020-06-09 Korea University Research And Business Foundation Platelet test chip
US10156568B2 (en) * 2015-04-30 2018-12-18 International Business Machines Corporation Immunoassay for detection of virus-antibody nanocomplexes in solution by chip-based pillar array
US20160320389A1 (en) * 2015-04-30 2016-11-03 International Business Machines Corporation Immunoassay for Detection of Virus-Antibody Nanocomplexes in Solution by Chip-Based Pillar Array
US11162948B2 (en) 2015-04-30 2021-11-02 International Business Machines Corporation Immunoassay for detection of virus-antibody nanocomplexes in solution by chip-based pillar array
US11912978B2 (en) 2015-06-05 2024-02-27 Novartis Ag Flow-through paramagnetic particle-based cell separation and paramagnetic particle removal
US11162065B2 (en) 2015-06-05 2021-11-02 Novartis Ag Flow-through paramagnetic particle-based cell separation and paramagnetic particle removal
US11833526B2 (en) 2015-06-26 2023-12-05 Ancera Inc. Background defocusing and clearing in ferrofluid-based capture assays
US11285490B2 (en) 2015-06-26 2022-03-29 Ancera, Llc Background defocusing and clearing in ferrofluid-based capture assays
US10976232B2 (en) 2015-08-24 2021-04-13 Gpb Scientific, Inc. Methods and devices for multi-step cell purification and concentration
CN106053456A (en) * 2016-06-24 2016-10-26 许毅 Urine concentrating device
US11161112B2 (en) 2016-09-20 2021-11-02 International Business Machines Corporation Phosphoprotein detection using a chip-based pillar array
US10386276B2 (en) * 2016-09-20 2019-08-20 International Business Machines Corporation Phosphoprotein detection using a chip-based pillar array
US20180154319A1 (en) * 2016-12-01 2018-06-07 Imagine Tf, Llc Microstructure flow mixing devices
US10603647B2 (en) * 2016-12-01 2020-03-31 Imagine Tf, Llc Microstructure flow mixing devices
US11768137B2 (en) 2017-04-14 2023-09-26 Ventana Medical Systems, Inc. Size-based separation of dissociated fixed tissues
WO2019046052A1 (en) 2017-09-01 2019-03-07 Gpb Scientific, Llc Methods for preparing therapeutically active cells using microfluidics
US10988734B2 (en) 2017-09-01 2021-04-27 Gpb Scientific, Inc. Methods for preparing therapeutically active cells using microfluidics
US10844353B2 (en) 2017-09-01 2020-11-24 Gpb Scientific, Inc. Methods for preparing therapeutically active cells using microfluidics
US11306288B2 (en) 2017-09-01 2022-04-19 Gpb Scientific, Inc. Methods for preparing therapeutically active cells using microfluidics
US11149251B2 (en) 2017-09-01 2021-10-19 Gpb Scientific, Inc. Methods for preparing therapeutically active cells using microfluidics
WO2020097048A1 (en) * 2018-11-05 2020-05-14 Micromedicine, Inc. Systems and methods for sorting particles using hydrodynamic sizing
WO2021011907A1 (en) 2019-07-18 2021-01-21 Gpb Scientific, Inc. Ordered processing of blood products to produce therapeutically active cells
WO2021133897A1 (en) 2019-12-28 2021-07-01 Gpb Scientific, Inc. Microfluidic cartridges for processing particles and cells

Also Published As

Publication number Publication date
US20210178403A1 (en) 2021-06-17
EP1866410A2 (en) 2007-12-19
CN101918527A (en) 2010-12-15
US20070026381A1 (en) 2007-02-01
HK1175494A1 (en) 2013-07-05
JP2008538283A (en) 2008-10-23
US20190001344A1 (en) 2019-01-03
CA2601480A1 (en) 2006-10-12
WO2006108101A3 (en) 2009-04-16
WO2006108101A2 (en) 2006-10-12
US10786817B2 (en) 2020-09-29
US20160144378A1 (en) 2016-05-26
US9174222B2 (en) 2015-11-03
US8585971B2 (en) 2013-11-19
EP2664666B1 (en) 2017-06-07
US20120225473A1 (en) 2012-09-06
CN101918527B (en) 2012-06-27
EP2492339B1 (en) 2014-03-19
US8021614B2 (en) 2011-09-20
EP2492339A1 (en) 2012-08-29
EP2664666A3 (en) 2014-03-12
AU2006232103A1 (en) 2006-10-12
EP2664666A2 (en) 2013-11-20
EP1866410B1 (en) 2012-08-08
EP1866410A4 (en) 2009-11-04
US9956562B2 (en) 2018-05-01
GB0612648D0 (en) 2006-08-16
US20120196273A1 (en) 2012-08-02
GB2429774A (en) 2007-03-07

Similar Documents

Publication Publication Date Title
US20070196820A1 (en) Devices and methods for enrichment and alteration of cells and other particles
US20060223178A1 (en) Devices and methods for magnetic enrichment of cells and other particles
US11725180B2 (en) Microfluidic sorting using high gradient magnetic fields
US20070059716A1 (en) Methods for detecting fetal abnormality
US20070059683A1 (en) Veterinary diagnostic system
US20070059718A1 (en) Systems and methods for enrichment of analytes
US20070059774A1 (en) Kits for Prenatal Testing
US20070059781A1 (en) System for size based separation and analysis
US20070059680A1 (en) System for cell enrichment
JP2009511001A (en) Device and method for magnetic concentration of cells and other particles
US20070059719A1 (en) Business methods for prenatal Diagnosis
CA2622745A1 (en) Systems and methods for enrichment of analytes
EP2838581A1 (en) Apparatus and method for separating a biological entity from a sample volume
Liu Isolation and Characterization of Circulating Tumor Cells and Tumor-derived Exosomes Using Ferrohydrodynamic Techniques in Microfluidic Systems

Legal Events

Date Code Title Description
AS Assignment

Owner name: GENERAL HOSPITAL CORPORATION, THE,MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:TONER, MEHMET;REEL/FRAME:024047/0827

Effective date: 20100210

AS Assignment

Owner name: GPB SCIENTIFIC, LLC, VIRGINIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:TONER, MEHMET;REEL/FRAME:027044/0055

Effective date: 20110920

Owner name: TONER, MEHMET, MASSACHUSETTS

Free format text: QUIT CLAIM ASSIGNMENT;ASSIGNOR:THE GENERAL HOSPITAL CORPORATION;REEL/FRAME:027046/0173

Effective date: 20110928

Owner name: THE GENERAL HOSPITAL CORPORATION, MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:TONER, MEHMET;REEL/FRAME:027044/0055

Effective date: 20110920

STCB Information on status: application discontinuation

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