CA2389358C - Multiplexed molecular analysis apparatus and method - Google Patents

Multiplexed molecular analysis apparatus and method Download PDF

Info

Publication number
CA2389358C
CA2389358C CA002389358A CA2389358A CA2389358C CA 2389358 C CA2389358 C CA 2389358C CA 002389358 A CA002389358 A CA 002389358A CA 2389358 A CA2389358 A CA 2389358A CA 2389358 C CA2389358 C CA 2389358C
Authority
CA
Canada
Prior art keywords
capillary
array
reaction substrate
biosites
capillary tubes
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.)
Expired - Fee Related
Application number
CA002389358A
Other languages
French (fr)
Other versions
CA2389358A1 (en
Inventor
Mitchell D. Eggers
William J. Balch
Michael E. Hogan
Leopoldo G. Mendoza
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.)
HTG Molecular Diagnostics Inc
Original Assignee
Genometrix Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Genometrix Inc filed Critical Genometrix Inc
Publication of CA2389358A1 publication Critical patent/CA2389358A1/en
Application granted granted Critical
Publication of CA2389358C publication Critical patent/CA2389358C/en
Anticipated expiration legal-status Critical
Expired - Fee Related legal-status Critical Current

Links

Classifications

    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01LCHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
    • B01L3/00Containers or dishes for laboratory use, e.g. laboratory glassware; Droppers
    • B01L3/02Burettes; Pipettes
    • B01L3/0241Drop counters; Drop formers
    • B01L3/0268Drop counters; Drop formers using pulse dispensing or spraying, eg. inkjet type, piezo actuated ejection of droplets from capillaries
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J19/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J19/0046Sequential or parallel reactions, e.g. for the synthesis of polypeptides or polynucleotides; Apparatus and devices for combinatorial chemistry or for making molecular arrays
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01LCHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
    • B01L3/00Containers or dishes for laboratory use, e.g. laboratory glassware; Droppers
    • B01L3/02Burettes; Pipettes
    • B01L3/0241Drop counters; Drop formers
    • B01L3/0262Drop counters; Drop formers using touch-off at substrate or container
    • 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
    • B01L3/508Containers for the purpose of retaining a material to be analysed, e.g. test tubes rigid containers not provided for above
    • B01L3/5085Containers for the purpose of retaining a material to be analysed, e.g. test tubes rigid containers not provided for above for multiple samples, e.g. microtitration plates
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/543Immunoassay; Biospecific binding assay; Materials therefor with an insoluble carrier for immobilising immunochemicals
    • G01N33/54366Apparatus specially adapted for solid-phase testing
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N35/00Automatic analysis not limited to methods or materials provided for in any single one of groups G01N1/00 - G01N33/00; Handling materials therefor
    • G01N35/02Automatic analysis not limited to methods or materials provided for in any single one of groups G01N1/00 - G01N33/00; Handling materials therefor using a plurality of sample containers moved by a conveyor system past one or more treatment or analysis stations
    • G01N35/028Automatic analysis not limited to methods or materials provided for in any single one of groups G01N1/00 - G01N33/00; Handling materials therefor using a plurality of sample containers moved by a conveyor system past one or more treatment or analysis stations having reaction cells in the form of microtitration plates
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N35/00Automatic analysis not limited to methods or materials provided for in any single one of groups G01N1/00 - G01N33/00; Handling materials therefor
    • G01N35/10Devices for transferring samples or any liquids to, in, or from, the analysis apparatus, e.g. suction devices, injection devices
    • G01N35/1065Multiple transfer devices
    • G01N35/1074Multiple transfer devices arranged in a two-dimensional array
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00277Apparatus
    • B01J2219/00279Features relating to reactor vessels
    • B01J2219/00306Reactor vessels in a multiple arrangement
    • B01J2219/00313Reactor vessels in a multiple arrangement the reactor vessels being formed by arrays of wells in blocks
    • B01J2219/00315Microtiter plates
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00277Apparatus
    • B01J2219/00279Features relating to reactor vessels
    • B01J2219/00306Reactor vessels in a multiple arrangement
    • B01J2219/00313Reactor vessels in a multiple arrangement the reactor vessels being formed by arrays of wells in blocks
    • B01J2219/00315Microtiter plates
    • B01J2219/00317Microwell devices, i.e. having large numbers of wells
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00277Apparatus
    • B01J2219/00351Means for dispensing and evacuation of reagents
    • B01J2219/00364Pipettes
    • B01J2219/00367Pipettes capillary
    • B01J2219/00369Pipettes capillary in multiple or parallel arrangements
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00277Apparatus
    • B01J2219/00351Means for dispensing and evacuation of reagents
    • B01J2219/00378Piezo-electric or ink jet dispensers
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00277Apparatus
    • B01J2219/00497Features relating to the solid phase supports
    • B01J2219/00511Walls of reactor vessels
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00583Features relative to the processes being carried out
    • B01J2219/00585Parallel processes
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00583Features relative to the processes being carried out
    • B01J2219/00596Solid-phase processes
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00583Features relative to the processes being carried out
    • B01J2219/00603Making arrays on substantially continuous surfaces
    • B01J2219/00605Making arrays on substantially continuous surfaces the compounds being directly bound or immobilised to solid supports
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00583Features relative to the processes being carried out
    • B01J2219/00603Making arrays on substantially continuous surfaces
    • B01J2219/00605Making arrays on substantially continuous surfaces the compounds being directly bound or immobilised to solid supports
    • B01J2219/0061The surface being organic
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00583Features relative to the processes being carried out
    • B01J2219/00603Making arrays on substantially continuous surfaces
    • B01J2219/00605Making arrays on substantially continuous surfaces the compounds being directly bound or immobilised to solid supports
    • B01J2219/00612Making arrays on substantially continuous surfaces the compounds being directly bound or immobilised to solid supports the surface being inorganic
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00583Features relative to the processes being carried out
    • B01J2219/00603Making arrays on substantially continuous surfaces
    • B01J2219/00605Making arrays on substantially continuous surfaces the compounds being directly bound or immobilised to solid supports
    • B01J2219/00614Delimitation of the attachment areas
    • B01J2219/00617Delimitation of the attachment areas by chemical means
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00583Features relative to the processes being carried out
    • B01J2219/00603Making arrays on substantially continuous surfaces
    • B01J2219/00605Making arrays on substantially continuous surfaces the compounds being directly bound or immobilised to solid supports
    • B01J2219/00614Delimitation of the attachment areas
    • B01J2219/00621Delimitation of the attachment areas by physical means, e.g. trenches, raised areas
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00583Features relative to the processes being carried out
    • B01J2219/00603Making arrays on substantially continuous surfaces
    • B01J2219/00605Making arrays on substantially continuous surfaces the compounds being directly bound or immobilised to solid supports
    • B01J2219/00623Immobilisation or binding
    • B01J2219/00626Covalent
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00583Features relative to the processes being carried out
    • B01J2219/00603Making arrays on substantially continuous surfaces
    • B01J2219/00605Making arrays on substantially continuous surfaces the compounds being directly bound or immobilised to solid supports
    • B01J2219/00632Introduction of reactive groups to the surface
    • B01J2219/00637Introduction of reactive groups to the surface by coating it with another layer
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00583Features relative to the processes being carried out
    • B01J2219/00603Making arrays on substantially continuous surfaces
    • B01J2219/00659Two-dimensional arrays
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/0068Means for controlling the apparatus of the process
    • B01J2219/00702Processes involving means for analysing and characterising the products
    • B01J2219/00707Processes involving means for analysing and characterising the products separated from the reactor apparatus
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00718Type of compounds synthesised
    • B01J2219/0072Organic compounds
    • B01J2219/00722Nucleotides
    • 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
    • B01L2300/0893Geometry, shape and general structure having a very large number of wells, microfabricated wells
    • CCHEMISTRY; METALLURGY
    • C40COMBINATORIAL TECHNOLOGY
    • C40BCOMBINATORIAL CHEMISTRY; LIBRARIES, e.g. CHEMICAL LIBRARIES
    • C40B40/00Libraries per se, e.g. arrays, mixtures
    • C40B40/04Libraries containing only organic compounds
    • C40B40/06Libraries containing nucleotides or polynucleotides, or derivatives thereof
    • CCHEMISTRY; METALLURGY
    • C40COMBINATORIAL TECHNOLOGY
    • C40BCOMBINATORIAL CHEMISTRY; LIBRARIES, e.g. CHEMICAL LIBRARIES
    • C40B60/00Apparatus specially adapted for use in combinatorial chemistry or with libraries
    • C40B60/14Apparatus specially adapted for use in combinatorial chemistry or with libraries for creating libraries
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N35/00Automatic analysis not limited to methods or materials provided for in any single one of groups G01N1/00 - G01N33/00; Handling materials therefor
    • G01N2035/00178Special arrangements of analysers
    • G01N2035/00237Handling microquantities of analyte, e.g. microvalves, capillary networks
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N35/00Automatic analysis not limited to methods or materials provided for in any single one of groups G01N1/00 - G01N33/00; Handling materials therefor
    • G01N35/10Devices for transferring samples or any liquids to, in, or from, the analysis apparatus, e.g. suction devices, injection devices
    • G01N2035/1027General features of the devices
    • G01N2035/1034Transferring microquantities of liquid
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N35/00Automatic analysis not limited to methods or materials provided for in any single one of groups G01N1/00 - G01N33/00; Handling materials therefor
    • G01N35/10Devices for transferring samples or any liquids to, in, or from, the analysis apparatus, e.g. suction devices, injection devices
    • G01N2035/1027General features of the devices
    • G01N2035/1034Transferring microquantities of liquid
    • G01N2035/1039Micropipettes, e.g. microcapillary tubes
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N35/00Automatic analysis not limited to methods or materials provided for in any single one of groups G01N1/00 - G01N33/00; Handling materials therefor
    • G01N35/10Devices for transferring samples or any liquids to, in, or from, the analysis apparatus, e.g. suction devices, injection devices
    • G01N2035/1027General features of the devices
    • G01N2035/1034Transferring microquantities of liquid
    • G01N2035/1041Ink-jet like dispensers
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N35/00Automatic analysis not limited to methods or materials provided for in any single one of groups G01N1/00 - G01N33/00; Handling materials therefor
    • G01N35/0099Automatic analysis not limited to methods or materials provided for in any single one of groups G01N1/00 - G01N33/00; Handling materials therefor comprising robots or similar manipulators
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N35/00Automatic analysis not limited to methods or materials provided for in any single one of groups G01N1/00 - G01N33/00; Handling materials therefor
    • G01N35/10Devices for transferring samples or any liquids to, in, or from, the analysis apparatus, e.g. suction devices, injection devices
    • G01N35/1065Multiple transfer devices
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S435/00Chemistry: molecular biology and microbiology
    • Y10S435/808Optical sensing apparatus
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S436/00Chemistry: analytical and immunological testing
    • Y10S436/80Fluorescent dyes, e.g. rhodamine
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S436/00Chemistry: analytical and immunological testing
    • Y10S436/804Radioisotope, e.g. radioimmunoassay
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S436/00Chemistry: analytical and immunological testing
    • Y10S436/805Optical property
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S436/00Chemistry: analytical and immunological testing
    • Y10S436/807Apparatus included in process claim, e.g. physical support structures
    • Y10S436/809Multifield plates or multicontainer arrays
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10TTECHNICAL SUBJECTS COVERED BY FORMER US CLASSIFICATION
    • Y10T436/00Chemistry: analytical and immunological testing
    • Y10T436/11Automated chemical analysis
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10TTECHNICAL SUBJECTS COVERED BY FORMER US CLASSIFICATION
    • Y10T436/00Chemistry: analytical and immunological testing
    • Y10T436/11Automated chemical analysis
    • Y10T436/115831Condition or time responsive

Abstract

A method and device for preparing a reaction substrate for use as an assay device, comprising a step of parallel printing an array of biosites on said reaction substrate, wherein each biosite comprises a single rype of capture probe bound to said reaction substrate, and wherein the array of biosites is deposited on the reaction substrate by a capillary bundle printer device.

Description

WQ 98rs936 PC:,'LTS9'7: so98 MULTIPLEXED MOLECULAR ANALYSIS APPaRATUS AND i'YIETHOD
This is a divisional o.f Canadian patent application Serial No. 2,276,642 filed December 31, 1997.

ST:-kTE4iE'I'I' AS TO FEDERALLY SPONSORED RESE.ARCH
T'nis invention was made at least in part with funds from the National Ae:onautics and Space Adminisnation, Grant Number NAGw 4530.

TECHNICAZ. FIELD
.o I nis invention relates to a multipie:ced molecular anaiysis apparatus and method for the detection and quantincation of one or more molecular structures in a sample.

BACI{GROUND
It is very desirabie to rapidly detect and quantify one or more molecular Is structures in a sampie. i~xe molecular structures typically comprise Iigands, such as antibodies and anti-antibodies. Ligands aremolecuIes which are recognized by a particular receptor. L'.;ands may include, without limitation, agonists and antagonists for cell membrane receptors, toxins, venoms, oligosacchuides, proteins, bacteria and, monacional antibodies. For example, DNA or RNA sequence analysis is very useful in zo genetic and infectious disease diagnosis, toxicotogsr testing, genetic research, agriculture and pharmaceutical deveiopment Likewise, cell and antibody detection is important in numerous disease diagaostics.
In particular, nucleic acid-based anaiyses often require sequence identification andlor analysis such as in vitro diagnostic assays and methods development, high 25 throughput screening of natural products for biological activity, and rapid screening of ~ - ._.---_ w0 98/29736 PCT/US97/24898 perishabir items such as donated blood, tissues, or food products for a wide array of pathogens. In all of these cases there are fundamental constraints to the analysis. e.g., limited sample. time, or often both.
In these fields of use, a balance must be achieved between accuracy, speed, s and sensitivity in the context of the constraints mentioned earlier. i'vfost existing methodoloszies are generally not multiplexed. That is, optimization of analysis conditions and interpretation of results are performed in simplified single determination assays.
However. this can be problematic if a definitive diagnosis is required since nucleic acid hvbridization techniques requirr prior knowledQe of the pathogen to be screened. If symptoms are ambiguous, or indicative of any number of different disease organisms, an assay that would screen for numerous possible causative agents would be highly desirable. Moreover, if symptoms are complex, possibly caused by multiple pathogens, an assay that functioned as a "decision tree" which indicated with increasing specificity the orzanism invotved, would be of high diagnostic value.
Multiplexing requires additional controls to maintain accuracy. False positive or negative results due to contamination, degradation of sample, presence of inhibitors or cross reactants, and interfintra strand interactions should be considered when designing the analysis conditions.

Conventional Technologies and Limitations 2o Sanger Sequencing Of all the existing techniques, one of the most defuzitive is the traditional Sanger sequencing technique. This technique is invaluable for identifying previously unknown or unsuspected pathogens. It is also valuable in determining mutations that confer drug resistance to specific srtrains of disease organisms. These analyses are zs generally research oriented. The end result of this research, e.g., sequence determination of a specific pathogen, can be used to design probes for identification applications in a clinical setting.
~

However, there are constraints to employing this technique in a clinical lab.
The primarS constraints are cost and throughput due to the inherent labor intensive procedures. requiring multiple steps to be performed by skilled personnel. For example, typical analysis usually requires more than a day for completion. Of more concern is the s potential for ambiguity when multiple strains of a pathogen are present in one sample.
Virulence of the pathogen is often determined by the strain. An example is HPV, also known as human papilloma virus. Seventy strains of HPV are commonly known to exist.
Two strains, in particular, are strongly associated with an increased risk of cervical cancer, hence the aggressiveness of treatment or screening for tnalignancy is determined by the presence of an HPV strain. Multiple strains cause indeterminate results when using sequencing methodologies. The ideal assay would be multiplexed with the selectivity to identifv all strains involved.

Blotting Techniques Blotting techniques, such as those used in Southern and Northetn analyses, ts have been used extensively as the primary method of detection for clinically relevant nucleic acids. The samples are prepared quickly to protect them from endogenous nucleases and then subjected to a restriction enzyme digest or polymerase chain reaction (PCR) analysis to obtain nucleic acid fragments suitable for the assay.
Separation by size is carried out using gel electrophoresis. The denatured fragments are then made available for hybridization to labeled probes by blotting onto a membrane that binds the target nucleic acid. To identify multiple firagments, probes are applied sequentially with appropriate washing and hybridization steps. This can lead to a loss of signal and an increase in background due to non-specific binding. While blotting techniques are sensitive and inexpensive, they are labor intensive and dependent on the skill of the technician. They also do not allow for a high degree of multiplexing due to the problems associated with sequential applications of different probes.
~.e.~~,_n~.~.,~r..:~, .l-fir:roplate Assays Microplate assays have been developed to exploit binding assays, e.g., an ELISA assay. receptor binding and nucleic acid probe hybridization techniques.
Typically, with one microplate, e.g. micro-well titer plate, onlv one reading per well can s be taken. e.g.. by light emission analysis. These assays function in either one of two ways: (1) hybridization in solution; or (2) hybridization to a surface bound molecule. In the latter case, only a single element is immobilized per well. This, of course, limits the amount of information that can be detennined per unit of sample. Practical considerations, such as sample size, labor costs, and analysis time, place limits on the use to of microplates in multiplex analyses. With only a single analysis, reaction, or determination per well, a multiple pathogen screen with the appropr iate controls would consume a signifieant portion of a typical 96 well format microplate. In the case where strain detennination is to be made, multiple plates must be used. Distributing a patient sample over such a large number of wells becomes highly impractical due to limitations 15 on available sample material. Thus, available patient sample volumes inherently limit the analysis and dilution of the sample to increase volume seriously affects sensitivity.
Polymerase Chain Reaction Although, the polymerase chain reaction (PCR) can be used to amplify the target sequence and improve the sensitivity of the assay, there are practical limitations to 2o the number of sequences that can be amplified in a sample. For example, most multi-plexed PCR reactions for clinical use do not amplify more thart a few target sequences per reaction. The resulting amplicons must still be analyzed either by Sanger sequencing, gel electrophoresis, or hybridization techniques such as Southiern blotting or microplate assays. The sample components, by PCR's selective amplification, will be less likely to 25 have aberrant results due to cross reactants. This will not be totally eliminated and controls should be employed. In addition, PCR enhances the likelihood of false positive, results from contamination, thus requiring environmental controls. PCR
controls must x-~-.-.~.-_.~..,.

also include an amplification positive control to ensure against false negatives. Inhibitors to the PCR process such as hemoglobin are common in clinical samples. As a result, dze PCR process for multiplexed analysis is subject to most of the problems outlined previously. A high density of information needs to be acquired to ensure a correct s diagnoscic deternzination. Overall, PCR is not practical for quantitative assays, or for broad screenina of a large number of pathogens.

Probe-Based Hybridization Assays Recently, probe hybridization assays have been performed in array formats on solid surfaces, also called "chip formats." A large number of hybridization reactions using very small amounts of sample. can be conducted using these chip formats thereby facilitating information rich analyses utilizing reasonable sample volumes.
Various strategies have been implemented to enhance the accuracy of these probe-based hybridization assays. One strategy deals with the problems of maintaining selectivity with assays that have many nucleic acid probes with varying GC
content.
Stringency conditions used to eliminate single base mismatched cross reactants to GC
rich probes will strip AT rich probes of their perfect match. Strategies to combat this problem range from using electrical fields at individually addressable probe sites for stringency control to providing separate micro-volume reaction chambers so that separate wash conditions can be maintained. This latter example would be analogous to a zo miniaturized microplate. Other systems use enzymes as "proof readers" to allow for discrimination against mismatches while using less stringent conditions.
Although the above discussion addresses the problem of mismatches, nucleic acid hybridization is subject to other errors as well. False negatives pose a significant problem and are often caused by the following conditions:
1) Unavailability of the binding domain often caused by intra-strand folding in the target or probe molecule, protein binding, cross reactant DNA/RNA
competitive binding, or degradation of target molecule.
~

2) .\ iM-aniplifrcation qf target molecule due to the pcesence of small molecule inhibitors, degradation of sample, and/or high ionic strength.
Problems wich labeling systems are often problematic in sandwich assays.
Sandwich assays, consisting of labeled probes complementary to secondary sites on the bound target molecule, are commonly used in hybridization experiments. These sites are subject to the above mentioned binding domain problems. Enzymatic chemiluminescent systems are subject to inhibitors of the enzyme or substrate and endogenous peroxidases can cause false positives by oxidizing the chemiluminescent substrate.

SUib11-1ARY
An object of the present invention is to provide a multiplexed molecular analysis and method. In accordance with an aspect of the present invention, there is provided a device for preparing a reaction substrate having biosites for conducting multiplexed microassays to determine binding between a target analyte and a capture probe including:.
a plurality of capillary tubes, each tube having a proximal end and a distal end;
an attachment site for holding said capillary tubes at a point spaced from the distal ends of such capillary tubes;, an array template for slidably holding each capillary tube near its distal end, and for allowina the distal end of each capillarv tube to move with respect to the attachment site;
at least one manifold for positioning the proximal end of each capillary tube ithin a correspondine supply chamber, wherein each supply chamber is capable of supplvins a liquid reagent to at least one corresponding c3piilar,v rube; and a positioning device for precisely positioning the array template and said capillary tubes with respect to said reaction substrate and depositing liquid reagents from said capillary tubes onto said reaction substrate as biosites.

In accordance with another aspect of the invention, there is provided a reaction substrate for conducting multiplexed microassays to determine binding of a target molecule and a capture probe/target probe complex, said reaction substrate including an array of biosites, each biosite comprising a single type of capture probe bound to said substrate, each capture probe capable of binding to a corresponding target probe having a capture probe specific domain which specifically binds with a corresponding capture probe, and a target analyte specific domain which specifically binds with a target analyte.

In accordance with another aspect of the invention, there is provided a reaction substrate for use in conducting multiplexed microassays to determine binding between a target molecule and a capture probe, said reaction substrate having an array of parallel printed biosites, wherein each biosite comprises a single type of capture probe bound to said reaction substrate.

In accordance with another aspect of the invention, there is provided a method for preparing a reaction substrate for use as an assay device, comprising the step of parallel printing an array of biosites on said reaction substrate, wherein each biosite comprises a single type of capture probe bound to said reaction substrate.

In accordance with another aspect of the invention, there is provided a method for identifying target analytes in a plurality of separate samples, each target analyte capable of binding a corresponding capture probe, including the steps of:
parallel printing an array of biosites on a reaction substrate, each biosite comprising a single type of capture probe bound to said substrate;
contacting each biosite with a sample including at least one target analyte;
and determining the presence or absence of binding of a target analyte at each biosite in said array.

In accordance with another aspect of the invention, there is provided an apparatus for preparing a reaction substrate for use as an assay device, including:
an attachment site for holding a plurality of flexible capillary tubes at a point spaced from the distal ends of such capillary tubes;
a structure for positionin: the proximal end of each capillary tube within a correspondins supply chamber, each supply chamber capable of supplying a liquid rzagent to at least one corresponding capillary tube;
a print head for holding said plurality of capillary tubes near their distal ends and for precisely positioning the distal ends of said capillary tubes with respect to said reaction substrate to deposit liquid reagents onto said reaction substrate as biosites.

-6a-p~, In accordance with another aspect of the invention, there is provided a method for detecting labeled sample molecules, comprising the steps of:
providing a reaction substrate having a plurality of biosites, each biosite being attached to the reaction substrate, the reaction substrate having an embedded conductive material connected to an electrical source;
contacting each biosite with a sample of labeled naolecules;
initiating an electrochernical event within the labeled molecules which releases electromagnetic energy;
detecting the released electromagnetic energy.

In accordance with another aspect of the invention, there is provided a method for detecting particle emissions from labeled sample molecules, comprising the steps of:
providing a reaction substrate having a plurality cif biosites, each biosite comprising a single type of capture probe bound to said substrate;
providing a lensless array of particle detectors disposed in close proximity to said plurality of biosites;
contacting each biosite with a sample of labeled molecules;
converting, in said particle detectors, particle emissions emanating directly from said sample into corresponding electrical signals;
forming an image from said electrical signals representing the quantitative presence or absence of the labeled molecules on each biosite from said sample.

In accordance with another aspect of the invention, there is provided a set of nucleic acid capture probes, each configured to be bound to a reaction substrate, each capture probe having a length sufficient to provide dissimilarity among capture probes, each capture probe having a percentage base composition in the range of about 30-40%G, 30-40%C, 10-20%A, and 10-=20%T, wherein the set is selected such that the nucleic acid sequence of each capture probe in the set is substantially dissimilar from the nucleic acid sequence of all other capture probes in the set.

The instant invention provides for both a multiplexed environment to rapidly determine optimal assay parameters, as well as a fast, cost-effective, and accurate system for the quantitative analysis of target analytes, thereby circumventing the limitations of -6b-~ ~ ~

single determination assays. The optimization of a multiplexed assay can be carried out by experimental interrogation to determine the appropriate solution conditions for hybridization and stringency washes. The development c-f these optimal chemical environments will be highly dependent on the characteristics of the array of bound capture probe molecules, their complementary target molecules, and the nature of the sample matrix.
Multiplexed molecular analyses are often required to provide an answer for specific problems. For example, determining which infectious agent out of a panel of -possible organisms is causing a specific set of disease syznptoms requires many analyses.
Capture probe arrays offer the opportunity to provide these multivariate answers. How-ever, the use of single probe array platforms does not always provide enough information to solve these kinds of problems. Recent innovative adaptations of proximal charge-coupled device (CCD) tecluiology has made it feasible to quantitatively-detect and image molecular probe arrays incorporated into the bottom of microplate wells. This creates a -6c-~

WO 98l29736 - PCT/US97/24098 hi~,h throughput platform of exceptional utility, capabVe of addressina several applications witli very complex analysis parameters.

Uses The niultiplexed molecular analysis system of the instant invention is useful for s analyzing and quantifying several molecular targets within a sample substance using an array having a pluraiity of biosites upon which the sample substance is applied. For example. this invention can be used with microarrays in a microplate for multiplexed diagnostics. drug discovery and screening analysis, gene expression analysis, cell sorting, and microorganic monitoring (see examples below for each use).

Proximal CCD Imaging with lvlultiplexed Arrays One application of the microplate based arrays of this invention is in parallel processing of a large number of samples. Large clinical labs process thousands of samples a dav. A microplate configured with a four by fbur (4 x 4) matrix of biosites in each of the 96 wells would be able to perform a total of 1536 nearly simultaneous tests from 96 different patient samples utilizing the proximal CCD imager as illustrated in Fig.
1. Fig. I is a diagram showing a multiplexed molecular analysis detectionlimaging system. =Moreover, a microplate configured with 15 x 15 arrays of probe elements in each of 96 wells enables a total of 21,600 nearly simuitaneous hybridization analyses, which becomes significant for analyzing gene expression from specific cells.
Throughput is also important when screening natural products for biological activity. A matrix of biosites that model binding sites of interest may be placed in the bottom of each well and interrogated with an unknown product. Thousands of molecules may be screened per day against these biosite arrays.
,~~

Cr=ecrtrvrr vJ'Hierarchical Arrays Another use of the microplate based arrays is for the creation of hierarchical arrays for complex analyses. In this format. multiple arravs operate in parallel to provide an answer to a complex assay. The example of the diagnostic assay provided in the s Background section illustrates some of the parameters which should be considered in order to provide an accurate result. For any specific analysis, a set of probe elements must be chosen. The selected probe elements should be able to selectively associate with defined targets without significant cross association to other macromolecules expected from either the patient or other organisms commonly associated with a specific sample type. Controls must be designed to prevent false positive or negative results from the sources outlined in the Background section. Once this is done, a combinatorial process can be used to identify the optimal association and selectivity conditions for the defined analysis. For nucleic acid applications, these conditions are highly dependent on the capture probe length and composition, target base composition, and sample matrix. The number of arrays to be used depends on a number of different factors, e.g., the controls to be implemented and the differences in base composition of the capture probes.
Ultimately, a set of integrated chemical devices emerge that can rapidly, efficiently, and accurately provide an answer for the molecular analysis of interest.
Another use of the hierarchical arrays and the reaction vessel based arrays would 2o be for screening samples for a broad range of possible targets. In one case, a diagnostic test is performed to search for the cause of a defined set of symptoms. In most cases this narrows the range of possible organisms to a small number. Conversely, to screen donated blood or tissue for a broader range of disease organisms, a decision tree approach could be employed. Here an initial array or set of arrays could be chosen to screen for broader classes of pathogens using probes for highly conserved nucleic acid regions. Results from this would indicate which additional ariay sets within the microplate to sample next, moving to greater and greater specificity. If enough sample is available, as might be the case with donated blood or tissue, all of the decision tree .g_ WO 98/29736 PCT![7S97124098 elements could be interro,;,ated simultaneously. If sample quantity is limiting, the approach could be directed in a serial fashion.
Assay development for any multiplex analysis is time consuming. The microplate based arrays as described herein can be used to speed the process for capture probe/target binding or h~-bridization. A defined array can be deposited into each well of a microplate and then the association reactions are caried out using "gradients" of conditions that varY in nvo dimensions. For example, consider a 96 well microplate containing nucleic acids arranaed in 8 rows by 12 columns. In one step of the optimization, the effects of pH
on various substrate compositions might be examined to see how this affects la hybridization specificity. Twelve different pH's, one for each column, and 8 different surface chemistries, one for each row could be used under otherwise identical hybridization conditions to measure the effects on hybridization for each capture probe/target element in the array. This type of analysis will become essential as arrav technology becomes widely used and is amenable to any receptor/Iigand binding type experiment.
The hierarchical array format, consisting of defined sets of arrays with individually optimized chemical environments functioning in parallel to provide an answer to a complex analysis, can be implemented in other ways. Instead of a batch process, where a series of analysis sets are present in each microplate, a hierarchical array zo analysis set can be fashioned into a flow cell arrangement. This would be specific to a particular analysis and consist of the appropriate array sets and the necessary fluidics to take a single sample and deliver the appropriate aliquot to each array in the set. The fluidics will deliver the appropriate association and wash fluids to perform the reactions, as defined for each array in the set.
~~

Advantages The multiplexed molecular analysis system of the instant invention has many advantages over the conventional svstems. Some of these advantages are discussed below.

High TIvoughput Multiple DNAIRNA probe arrays can be fabricated in the bottom of 96 well microtiter plates which offer the potential of performing 1õ536 (96x 16) to 21,600 (96x225) hybridization tests per microtiter plate. Each well will contain a probe array of N elements dispensed onto plastic or glass and bonded to the microtiter plate.
Moreover, io by coupling the microtiter trays to a direct (lenstess) CCD
proximal/imager, all 1,536 to 21,600 hybridization tests can be quantitatively accessed w-ithin seconds at room temperature. Such proximal CCD detection approach enables unprecedented speed and resolution due to the inherently high collection efficiency and parallel imaging operation.
The upper limit to the hybridization tests per microtiter pl.ate exceeds 100,000 based on a 100 m center-to-center spacing of biosites.
Low Cost Since the capture probe volumes dispensed on the reaction substrate can be limited to about 50 picoliters (pL), only 150 nanoliters (nL) of capture probe reagent is required to produce over 1,500 distinct binding tests. The dispensing of the probe arrays zo on plastic rolls or on thin glass sheets can be efficiently performed in an assembly-line fashion with a modular ink-jet or capillary deposition systenn.

Airtomated Operation The multiplexed assay can be designed in a standard 96 well microtiter plate format for room temperature operation to accommodate conventional robotic systems utilized for sample delivery and preparation. Also, the proximal CCD-based imager with ._..,.,~.

WO 98129736 PCT/[TS97/24098 a~'raphicai user interface will enable the automation of the parallel acquisition of the numerous hybridization test results. The CCD imaging system software provides automated filterin~;~, thresholding, labeling. statistical analysis and quantitative graphical display of each probe/target binding area within seconds.

L'ersatilin.~
The proximal CCD detector/imager utilized in a particular embodiment of the multiplexed molecular analysis system accommodates numerous molecule labeling stratecies including fluorescence, chemiluminescence aiid radioisotopes.
Consequently, a single instrument can be employed for a variety of reporter groups used separately or tojether in a multiplexed manner for maximal information extraction.
High Resolution The accompanying proximal CCD detectorrmager offers high spatial and digital resolution. In the preferred embodiment, CCD pixel sizes of approximately 25 x2j m'-will support the imaging of hundreds to thousands of individual biosites on a reaction substrate. Together with 16 bit digital imaging, a highly iquantitative image of the high density of biosites is achieved.

Fast Time-to-Market Since the approach outlined is based on previously demonstrated proximal CCD
detection and imaging coupled with microarrays dispensed in conventional sized 2o microtiter plates, the overall molecular analysis system is expected to provide a fast time-to-market solution to complex multicomponent molecular-based analyses.

Overall, the invention disclosed provides a method and apparatus for both a multiplexed environment to rapidly determine the optimal assay parameters as well as a .,..

~
f3st, cost-effective, and accurate system for the quantitative analysis of molecules, thereby circumventing the limitations of single determination assays.

Other features and advantages of the invention will be apparent from the following detailed description, and from the claims.
-BRIEF DESCRIPTION OF THE DRAWINGS
Fig. I is a diagram showing a multiplexed molecular analysis detection/imaging system. Fig. I a is the reaction chamber circled as item I a in Figure 1.
Fig. 2 is a printed computer image obtained with the proximal CCD imager showing deposited DNA probe biosites with ink-jet printing.
Fig. 3 is a diagram showing the biosite deposition system using staggered ink-jet dispensing modules.
Fig. 4 is a diagram showing the biosite deposition system using multiple capillaries.
Fig. 4a is a diagram showing biosite deposition with array templates. Fig. 4b is a diagram showing biosite deposition into nanoliter wells.
Fig. 5a is a diagram illustrating the Universal Array concept.
Fig. 5b is a diagram showing direct binding for a target probe associated with the Universal Array.
Fig. 5c is a printed computer image showing a multi-microtiter well proximal CCD
image of a 4 x 4 Universal Array.
Fig. 5d is a printed computer image showing a single microtiter well proximal CCD
image of a 4 x 4 Universal Array.
Fig. 6 is a diagram illustrating an ECL implementation in a reaction vessel with proximal CCD imaging.
Fig. 7 is a diagram showing fabrication of ECL reaction vessels.
Fig. 8 is a chemical drawing showing lanthanide chelators.
Fig. 9 is a diagram showing the electronics schematic of a multiplexed molecular analysis system.
Fig. I Oa is a diagram showing a CCD sensor array for the proximal CCD imager.
Fig. I Ob is a diagram showing the tiling of CCD sensors to form a large format proximal CCD imager.
Fig. l Oc is a diagram showing an alternative tiling scheme of multiple CCD
sensors used to form a large format proximal CCD imager.
Fig. 11 is a printed computer image showing microarrays within a microplate reaction vessel. Fig. 11 a is one singe reaction chamber.
Fig. 12 is a diagram showing glass and polypropylene surface coupling chemistries.
Fig. 13 is a diagram showing genotyping by universal point mutation scanning.
Fig. 14 is a diagram showing microtiter-based throughput genotyping.
Fig. 15 is a diagram showing homogeneous in situ microarray detection of multiplexed PCR amplicons.
Fig. 16 is a diagram showing homogeneous in situ microarray detection of multiplexed gap-ligase chain reaction products.
Fig. 17 is a diagram showing small molecule universal array (drug screening/discovery).
Fig. 18 is a diagram illustrating spatial addresses of small molecules covalently immobilized on amino-derivitized thin-bottom, glass microtiter wells.
Fig. 19A is a printed computer image showing specific imaging of biotin-addressable biosites detected using streptavidin:HRP conjugate (4 x 4 single well microarray).
Fig. 19B is a printed computer image showing specific imaging of digoxigenin-addressable biosites detected using anti-digoxigenin:HRP conjugate (4 x 4 single well microarray).
Fig. 19C is a printed computer image showing specific imaging of fluorescein-addressable biosites detected using anti-fluorescein:HRP conjugate (4 x 4 single well microarray).
Fig. 19D is a printed computer image showing simultaneous imaging of fluorescein, biotin, and digoxigenin biosites detected using anti-fluorescein, anti-digoxigenin and streptavidin:HRP conjugates (4 x 4 single well microarray).
DETAILED DESCRIP'TION

Definitions For the purpose of this invention, different words and phrases are defined as follows:
B~-"target molecules or target analyte" is meant the molecules of interest in a substance which are to be interrogated by binding to the capture probes immobilized in an arrav.
By "mRNA target molecule or mRNA target analyte"is meant a substance containing identical mIziNA components or a mixture of disparate mRNAs.
By "capture probe, probe molecules or probes" is meant the molecules which are deposited as biosites onto the reaction substrate for interrogating the target molecules.
Probes are meant to incliide nucleie acids, DNA, RNA, receptors, ligands, antibodies, anti-antibodies, antigens, proteins, and also small chem:ical compounds such as drugs, haptens, or peptides.
The term ' hapten binding polypeptide" includes intact antibody molecules as well as fragments thereof, such as Fab, F(ab'h, Fv, single chain antibody (SCA) , and single complementary-determining region (CDR). For purposes of the invention, "hapten" and "epitope" are considered interchangeable.
The term "array" refers to a two-dimensional spatial grouping or an arrangement.
By "hierarchical arr=y" is meant an array arranged in an hierarchy or arranged in a graded or ranked series. Examples of different "hierarchical arrays"
comprising the multiplexed assay of the invention.include, but are not limited to, an array of a 96 well microtiter plate, wherein there are N probe sites or biosiites per well, wherein there are 10" to 10'0 molecules for each probe site or biosite, wherein an array of M
depositors are as used to deposit probes in each probe site onto the film substrate that forms the bottom of the well in a 96 microtiter well reaction chamber. The depositors can deposit the probes via many different mechanisms, e.g., ink-jet deposition, capillary, and photolithography.

~~~~ . . ~ .~..._ The term "probe arrays" refers to the array of N different biosites deposited on a reaction substrate which serve to interrogate mixtures of taraet molecules or multiple sites on a single target molecule administered to the surface of the array.
The term "oligonucleotide probe arrays" refers to probe arrays wherein the probes are constructed of nucleic acids.
By "charae coupled device," also referred to as CCD, is meant a well-known electronic device which outputs an electrical signal proportional to the incident energy upon the CCD surface in a spatially addressable manner.
The term "CCD proximal detection" refers to the use of CCD technology for detection and imaging in which the CCD is proximal to the sample to be analyzed, thereby avoiding the need for conventional lenses.
By "ligands" is meant molecules which are recognized by a particular receptor.
"Ligands" may include, without limitation, agonists and antagonists for cell membrane receptors, toxins, venoms, oligosaccharides, proteins, bacteria and monoclonal is antibodies.
By "multiplexed" is meant many or a multiple number.
By "multiplexed diagnostic assay" is meant a method for performing in parallel a large set or number of diagnostic assays. Thus a set of parallel reactions can be handled with the same effort as a single sample in previously described methods.
Hence, a greater number of assays can be handled within a fixed period of time. The parallel set of reactions or multiplexed assay must be deciphered at the end of the process.
This is done by labeling or tagging the biosite, as defined herein.
The term "reaction vessel" refers to an array of reaction chambers as defined below. An example of a reaction vessel is a 96 well microtiter plate.
By "reaction chamber" is meant the environment in which the hybridization or other binding association takes place. Commercially available reaction vessels contain at least one reaction chamber, but can contain 8, 24, 96 or 384 reaction chambers. For this invention, "reaction chamber(s)," "well(s)," "reaction site(s)," "reaction substrate(s),"

~.T

WO 98/29736 rcT/irs97/24098 ==arrav hybridization site(s)," "hvbridization chamber(s)." and "hvbridization well(s)," are used intercliangeably. An example of a reaction chamber is one of the 96 microtiter wells in a 96 'ell microtiter plate.
By "biosite" is meant the biological molecules or capture probes that are deposited on the top surface of the reaction substrate, or base material.
Under appropriate conditions. an association or hvbridization can occur between the capture probe and a tarcet molecule. The component strands of the biological molecule form the biosite since there is the potential of a reaction occurring between each component strand of the biological molecule and the target molecule. For example, each reaction chamber can contain at least one biosite. The maximum number of biosites per reaction chamber will depend on the size of the reaction vessel and on the practical optical resolution of the accompanying detectorlimager. For example, an array of 16 (4 x 4 array) biosites may be deposited on the hybridization substrate or base material that eventually fosms the bottom of the entire reaction vessel. Each biosite comprises a circle of approximately 25-200 ts microns (}im) in diameter. Thus, for a 16 biosite array, each of the 16 x 200 m diameter area contains a uniform field of probes attached to the hybridization substrate (base material) in a concentration which is highly dependent on the probe size and the well size. Each 25-200 m diameter area can contain millions of probe molecules.
Also, each of the 16 different biosites (probe sites) can contain one type of probe.
Thus, 16 different probe types can be assayed in an array containing 16 biosites (4 x 4 array) per reaction chamber. As another example, four separate 10X 10 arrays (400 biosites) can be generated to fit into one well of a 96 well microtiter plate with sufficient spacing between each of the 400 biosites. For this I OX 10 format, 400 hybridization experiments are possible within a single reaction chamber corresponding to 38,400 (96 x 400) assays/hybridization that can be performed nearly simultaneously.
By "reaction substrate" is meant the substrate that the biosites or probe sites are deposited on by using the depositors. Examples of "reaction substrates"
include, without limitation, nylon membrane, polypropylene, polystyrene, vinyl, other plastics and glass.

:~ . ~ ~ ~

WO 98129736 PCTlQS9712409R
Bv "modular deposition array" is meant an array of depositors. The number of depositors depends primarily on the dimensions of the reaction substrate. For example, there can be four depositors fitted next to each other, stagQered regarding the front to back position of each depositor. Each depositor can be directly coupled to a housing reservoir. The housing reservoir holds a solution, e.g., a solution containing a desired probe at an appropriate concentration. The number of injection mechanisms again depends on the design of the depositor, e.g., ranging from one to several injection mechanisms per depositor.
By =-array formats on solid surfaces" is meant chip formats or microarrays.
By''throughput" is meant the number of analyses completed in a given unit of time.
By "decision tree approach" is meant a sequential routing approach in which at each step an assessmeiit is made which directs the subsequent step.
By "hybridization detection" is meant to include, without limitation, a means of 1s two or more components to interact through hybridization, an association, linking, coupling, chemical bonding, covalent association, lock and key association, and reaction.
For the purpose of this invention, these terms are used interchangeably.
By "methods of detecting (or detection) the association/hybridization" is meant to include, without limitation, fluorescent labeling, radioisotope labeling, chemilumines-cence labeling, bioluminescence labeling, colorimetric labeling. Labeling can be achieved by one of the many different methods known to those with skill in this art.
The term "luminescence" refers to, without liunitation, electrical (electro), chemical, fluorescence, phosphorescence, bioluminescence, and the like.
However, for this invention, electrochemiluminescence or electrical chemiluminescence (ECL) labeling is included as another method of detection which does not require a wash step to remove excess target molecules from the solution, and is highly sensitive. For the electrochemiluminescence or electrical chemiluminescence method of detection, once hybridization/association has occurred and a voltage has been applied, only the labeled ~

target molecules associated with the biosite will emit light and be detected.
The residual excess labrl in the solution not associated with the biosite will therefore not emit light.
Overview The multiplexed molecular analysis system of the invention can be divided into four aspects:
15 A. Preparing the sample for subsequent association to a probe array within the reaction chamber. This includes all front-end processes such as purification, isolation, denaturation and labeling required to e:ctract the target molecules from the sample.
B. Binding target molecules to the biosites within specialized reaction chambers in sufficient concentrations for association to occur. Following association, 20 non-specific binding of target molecules is often minimized by washing out the reaction chambers.
C. Detecting and/or imaging the association (hybridization) of the target molecules with the biosites within each reaction chamber by proximal detection/'inzaging.
D. Processing the images to determine information about the target molecules 25 such as the presence and amount of specific molecular constituents within a given sample that leads to the analysis output.

WO 98/29736 PCT/US97/2409y The advantage of the instant invention lies in the particular implementation of the above tour procedures/steps, in particular in the method and apparatus for:
STEP I. Biosite Deposition. Biosite deposition relates to constructing microarrays.
s STEP 2. Self Assembliny Arravs -- Universal Arrays. Creatiniz and constructing self assembling probe arrays or universal arrays enables on-line configuration of the biosites wherein an unvarying probe array (capture probes) is activated by binding to a cognate set of adapters (target probes) to yield a modified probe array which is specifically configured for analysis of a target or target mixture. For this invention, "cognate" is defined for nucleic acids as a sequence which is complementary by the means of Watson-Crick pairing to another sequence.
STEP 3. Molecular Labeling Strategies. Molecular labeling strategies relates to versatile labeling of the target molecules (fluorescence, chenuluminescence, etc.) consistent with proximal large area detection/ imaging.
STEP 4. Detection System. A detection system relates to parallel detection and/or imaging in the reaction vessel containing the reaction chambers using a proximal large area detector/imager.

STEP I -- BIOSITE DEPOSITION
Biosite deposition relates to constructing rnicroarrays. Tliere are many different methods that may be used for depositing biosites into/onto the reaction chamber. Three of these approaches are taught below.

l. Ink-Jet Deposition Ink-jet printing can be employed for printing the biological fluids to form the biosites. This approach provides very low droplet volumes (= 100 pL with 75 m diameter spot size) which minimizes reagents used and therefore cost.
Moreover, the printina process can be accelerated to thousands of droplets per second, thereby enabling io a high throughput production capability for the reaction vessels.
One method useful for this invention utilizes electromechanically driven ink-jets which produce a volumetric change in the fluid by appiving a voltage across a piezo-electric material (See Hansell, U.S. Patent No. 2,5I2,74:1, 1950). The volumetric changes causes a subsequent pressure transient in the fluid which can be directed to form a drop on demand (D. Bogg et al., IBMJour Res Develop (1984) 29:214-321.
Individual ink-jet devices can be integrated in a modular fashion to enable the =
printing of multiple fluids. For example, MicroFab Inc. has developed an ink-jet based DNA probe array printer constructed of eight modular dispensing units, each with an integral 500 mL reservoir independently addressed by respective drive and control electronics. ~
Fig. 2 depicts a printed computer image showing DNA probe biosites deposited with ink-jet printing. Fio. 2 illustrates actual biosite deposition whereby an array of 100 DNA probe biosites per 1 cm'- was ink-jet deposited onto a glass substrate.
The array consists of altemating columns of match and mismatch l2mer probes which were sub-sequently hybridized to a 12mer single-stranded DNA target. The mismatch columns correspond to an A-A mismatch in the probe/target complex. The actual image was captured within I second by the proximal CCD detector/imager described below.

L baiilcs of modular inl: jet devices containing M depositors per module can be assembled in a stagdered fashion to print LxM different biosites on the bottom surface of the reaction chambers as illustrated in Fig. 3. Fig. 3 is a diagram showing the biosite deposition system using staggered ink-jet dispensing modules. Here the reaction vessel is nioved by a precision motor-controlled stage underneath the ink-jet devices for rapid printinQ. Bv constructing additional banks of modular ink-jet devices and/or miniatur-izing the individual depositors. an arbitrarily large number of distinct biosites can be printed in the reaction vessels. Alternatively, the printing can be performed on thin substrates such as glass or plastics which are subsequently bonded to form the bottom of the reaction vessels.

2_ Capillary Deposition Another approach to biosite deposition involves the use of capillary tubing to dispense small amounts of the biosite solution onto the reaction substrate as illustrated in Fig. 4, 4a, and 4b.. Fig. 4 depicts a biosite deposition system using multiple capillaries.
Fig. 4a is a diagram showing biosite deposition with array templates. Fig. 4b is a diagram showing biosite deposition into nanoliter wells. As shown, a storage vessel which contains the appropriate solutions is pressurized momentarily to prime tubes held in appropriate position by a manifold to initiate the capillary dispensing action. With very small inner diameter capillaries (<50 m), continuous pressure may be applied.
Pressure pulses of varying duration can be utilized to deliver larger volumes of solution. Upon contact with the reaction substrate, the capillary tubes simultaneously deliver small volumes of the biosite solutions at precise locations controlled by spatial an-angement of the bundled capillaries.
In this invention, the storage vessel allows for sampling either from a standard format microtiter plate or a customized plate designed to hold small volumes of liquid, allowing the capillary to efficiently dispense picoliter volumes of liquid to many thou-sands of biosites with minimal loss to evaporation or possibility of cross contamination.

The lid of the storage vessel holder can be attached to a Z-axis motion control device to allow for automated chanaes of the biosite solutions contained in the micro-plates which may be delivered by a robotic arm. This is useful for printing sets of arrays containina larae numbers of solutions, such as small molecule libraries used in druo discovzrv.
Also in this invention, the capillarv tubing may be made of fused silica coated with an outer Iaver of polyimide. These tubes are available commercially in any length with ~-arious widths and internal diameters. The preferred dimensions are 80 to 500 m outer diameter (OD) and 10 to 200 m inner diameter (ID). The capillary bundles may be affixed to a robot arm and held in a precise pattern by threading the capillaries through array templates. An array template is a structure designed to maintain the capillaries in the desired configuration and spacing, and may consist of, without limitation, a metal grid or mesh, a rigidly-held fabric mesh, a bundle of "sleeve" tubes having an inner diameter sufficient to admit the fluid delivery capillaries, or a solid block having holes or is channels, e.o., a perforated aluminum block.
The embodiment depicted employs 190 m OD capillaries, which are threaded through an attachment site at the top of the printing fixture. The tubes extend down from the attachment site through an area that allows for the capillaries to flex during printing.
Below the flex region the capillaries are threaded through an array template or a set of fused silica sleeves held irt a grid pattem by the alumirium holder assemblies. The capillary sleeves/arrav template constitute an important innovation. The array templates/capillary sleeves also allow the capillary tubing to travel smoothly and independently with respect to each other in the verticall axis during printing.
The printing system can print high density probe arrays covering the bottom surface of microplate wells. To accomplish this, the printing system must be able to maintain a precise printing pattern and accommodate irregular surfaces. Rigid tubes could be used to maintain a precise pattern, however, they cannot readily accommodate irregular surfaces. Flexible tubes will print on uneven surfaces but will not maintain a - 23 ~

WO 98/29736 PCT1US97/2409' precise printing pattern_ The rigid sleeves, which extend below the aluminum liolder assembly approximately 2 cm, support the flexible 190 m OD fused silica capillary tubing and provide the structural rigidity necessary to maintain a precise grid pattern over this distance. The sleeves also allow the 190 m tubing to travel smoothly in the Z axis durin~; printing. This ability coupled with the flexibility of the small OD
capillarv tubint, allows for successful printing on surfaces that are not completely flat or absolutely perpendicular to the printing fixture. Since the robot arm extends 0.1 mm to 0.3 mm beyond the point where the capiIlary bundle contacts the surface, the capillaries flex in the deflection zone illustrated in Fig. 4 resulting in total surface contact among all capillaries in the bundle. When the printing fixture withdraws from the substrate, the capillaries straighten, returning to their original positions. The highly parallel nature of the capillary bundle printing technique allows for microarrays containing from two to over 10.000 chemically unique biosites to be created with a single "stamp."
The printer can print these arrays at a rate of approximately one per second. This represents a greater than 10-fold increase in speed over existing technologies such as photolithographic in situ synthesis or robotic deposition using conventional load and dispense technology.
In photolithographic microarray synthesis, a series of masks are sequentially applied to build the nucleic acid probes a base at a time. An array of oligonucleotide probes each 12 bases long would require 48 masks (12 nucleotide positions x 4 bases).
This process takes approximately 16 hours to complete a wafer containing 48 microarrays.
Current robotic microarray printing or gridding systems are universally based on various load and dispense techniques. These techniques can be split into two categories.
Active loading systems such as syringe needles or capillaries draw up enough solution to 2s dispense multiple biosites or array elements before retuming to reload or collect a new probe solution. Pin style printing or gridding systems can only print one biosite per pin at a time. The pins are dipped into the probe solutions momentarily and the amount of .

solution adhering to the pin is sufficient to print a single biosite. Both categories have limitations that are resolved by the capillary bundle printing system described herein.

Production capacity is a primary constraint in microarray manufacturing.
limiting the use of microarravs in high volume applications such as drug, discovery due to the cost and limited availability. For photolitho.graphic in situ synthesis. the constraint is the number of individual masks that must be applied to create an array of probes with the necessan' length to be effective. To increase capacity, the production systems must be duplicated. Current capacities for this approach (approximately 80,000 arrays for 1997) do not meet the needs for the drug discovery market, where a single company may screen over 100,000 samples per year.
Robotic printing systems currently manufacturE: microarrays in a largely serial fashion. The geometry of the fluid reservoir is often responsible for the limited degree of parallel biosite deposition. This can be explained by illustrating the process needed to produce a microarray. A"niicro" array has a small overall dimension, typically smaller Is than 2 cm by 2 cm. The actual size is determined by the number of array elements and their spacing, with an emphasis on reducing the overall size as much as possible to reduce reagent costs and sample requirements. If a parallel printing approach is implemented using multiple pins or depositiors, the geometries of these depositors must allow them to interface with the probe solution reservoirs and still be able to fit within the zo confines of the area to be occupied by the microarray. If a 1 cm3 100 element microarray (10 x 10) is to be constructed using a standard 384 well microplate with wells spaced 4.5 mm on center as the probe solution reservoir, only 4 depositors can be used to print simultaneously within the microarray. A total of 25 cycles of loading and printing would be required to complete the array. In comparison, this array would be manufactured with 25 a single print step for a capillary bundle printer with 100 capillaries.
This is 50 times faster than robotic depositors using a load and dispense technique. If the same array is condensed into a 0.5 cmz area, then only one depositor can be used, resuldng in a 200-fold differential in manufacturing time compared with the capillary bundle printer.

Wo 98l29736 PGT/US97/24098 ~, .
An important feature of the capillary bundle printer is the manner in which it interfac.s to the printing solution storage vessel. The capillat_v bundles have a printing (distal) end and a storage vessel end. The printing solutiori is held in a sealed container =
that positions every capillary in the printing bundle via a manifold so that each capillary dips into a specific well (supply chamber) of a microtiter plate, one capillary per well. Current multi-well microtiter plates are available with 96, 384, or 1536 we1ls, and can contain up to 96, 384, or 1536 individual probe solutions, respectively. For microarrays containing more probe elements, multiple printing solution reservoirs or storage vessels can be interfaced to a single print head, as illustrated in Fig. 4a. This design concept eliminates the geometry problems associated with load and dispense systems.
The flexible fused silica capillaries can be gathered together with the array templates or sleeves to create a print head with capillaries spaced as close as 200 m center to center.
The enclosed printing solution storage vessel is purged with an inert gas during the priming step of the printing process, which also serves to maintain an inert environment for the probe solutions. Contamination of the probe solutions is minimized because of the single direction of flow through the capillaries. The printing end does not dip into the reservoir after every print cycle as in the load and dispense techniques. This is important with contact printing where the depositors touch the surface of the chip or slide that will contain the microarray. These surfaces are chemically treated to interact or 2o bind to the probe solutions. Residual reactive chemicals, or even dust and dirt could be introduced into the probe solution supply chambers with load and dispense systems.
Often, the solution to be printed is available in limited quantity or is very expensive. This is often the case in pharmaceutical drug discover applications where small molecule libraries, containing hundreds of thousands of unique chemical structures that have been synthesized or collected and purified from natural sources, are used in high throughput screens of as many potential disease targets as possible. These libraries must be used as efficiently as possible. The amount of fluid that is required for each printing system varies depending on the design. Most require a minimum of 100 microliters ( L) and are ~

able to print less than 1.000 slides, with a significant amount of solution lost to washing between print cycles. The capillary array printer requires only 3 pL witll less than I L
tised for the initial priming. This volume of printing solution is sufficient to print between 20,000 to 30.000 microarrays vvith each capillary dispensinv 50 to 100 pL per array.
s Load and dispense systems deliver anywhere from 800 pL to several nL per array.
The highly paraliel approach allows probe solution deposition in a microarray _eometrv (less than 2 cm x 2 cm) independent of the geometry of the probe solution storaRe vessel. This permits production of an entire microarray containing from 2 to >
10,000 unique capture probes (biosites) in a single stamp of the print head.
The flexible fused silica tubing (or other suitable material such as glass.
Teflon or other relatively inert plastic or rubber, or thin, flexible cnetaI, such as stainless steel) oriainating at the printing storage vessel, pass through a series of arraying templates or sleeves that are held at specific locations in the print head. An attachment site holds the capillaries in a fixed position that does not generally allow horizontal or vertical movement. The capillaries extend down from this anchor point through an open area ("flexation zone") and into a set of array templates or sleeves. These lower array templates or sleeves serve to hold the printing capillaries in a geometry that matches the microarray to be printed. The array templates limit the lateral movement of the printing capillaries to preserve the correct printing pattern, while allowing unrestricted vertical movement of each printing capillary independently of each other. This feature allows the print head to print on slightly irregular or uneven surfaces. The print head moves downward to contact the substrate that is to receive the probe solutions, after the initial contact, the downward movement continues (the distance depends on the surface, from 100 m to a few mm) to ensure that all of the printing capillaries contact the surface. The flexation zone positioned between the attachment site (tlzat is holding the capillaries fixed) and the array templates or sleeves allows each capillary to bend so as to accommodate the "overdrive" of the print head. When the print head moves up away from the substrate, the printing capillaries straighten out again.

, , The capillary bundle ori;-inates in an enclosure containing discrete fluid supply chambe:s. such as the wells in a microtiter plate. Each capii4ary is inserted into a specific wztl. which usually contains a unique probe solution with respect to the other wells. The stora;;=z vessel can be momentarily pressurized to begin the fluid t1ow in all of the capillaries simultaneously to prime the printer. After primin', continuous flow of the probe solutions throueh the capillaries is thereafter facilitated bv adjusting the head height AH (the vertical distance from the upper fluid reservoir and the printing tips, as shown in Fig. 4), or by electro-osmotic or electrophoretic force (where the tubes, storage vessels. and reaction chambers are appropriately modified to maintain and modulate an electro-osmotic and/or electrophoretic potential). The chamber can maintain an inert environment by pressurizing the chamber with aninert aas, such as nitrogen or argon.
Fluid volumes deposited at each biosite can be modified by adjusting the head height. bv applying pressure to the printing solution storage vessel, by changing the length or inner dimension of the printing capillaries, or bv adjusting the surface tension of the probe solution or the substrate that is being printed.
The prime and continuous print with multiple capillaries prevents contamination of the probe solution that can occur with load and dispense systems, which must contact the surface and then return to the probe solution to draw more fluid. The continuous printing of the capillary bundle printer is extremely efficient and proves to be an enabling technique for applications that require the use of small volumes of probe solution. The small outer and inner diameters of the printing capillaries allow for printing as many as 10,000 spots per L from a total volume of less than 5 L.
In an alternative embodiment, the capillary tubes may be essentially rigid tubes (e.g., stainless steel) mounted in flexible or movable fashion at the attachment site, and 2s slidably held by an array template. In this embodiment, the plurality of capillary tubes can be pressed against a reaction substrate and "even up" at their distal ends by moving lengthwise through the array template, thus accommodating uneven deposition surfaces.

WO 98/29736 - PcT/US97n4098 3. Plruwlithr>>raph}vCapillmy Deposilion To increase the spatial resolution and precision of the capillary deposition approacil. a combined photolitho,--,raphic chemical masking and capillary approach is tauuht herein. The first photolithographic step selectively activates the precise biosite areas on the reaction substrate. Once selective activation has been achieved, the resulting capillary dzposition results in uniform biosite distribution.
N-lanv different substrates can be used for this invention. e.b , glass or plastic substrates. With glass substrates, the procedure begins by coating the surface with an aminosilane to aminate the surface. This amine is then reacted with a UV
sensitive protecting group, such as the succimidyl ester of a(=1,5-dimethoacy-2-nitrobenzyl) referred to as "caQed" succimidate. Discrete spots of free amine are revealed on the caged succimidate surface by local irradiation with a UV excitation source (LTV
laser or mercury arc). This reveals free acid groups which can then react with amine modified oligonucleotide probes. Such a process provides for local biosite modification, sur-ss rounded by substrate areas with a relatively high surface tension, unreacted sites.
W'hen using plastic substrates, the procedure begins by coating aminated plastic with an amine blocking group such as a trityl which is poorly water soluble and hence produces a coating with high surface tension. Next, an excitation source (eximer or IR
laser for example) is used to selectively remove trityl by light-induced heating. The biosite areas are then activated with bifunctional NHS ester or an equivalent.
The net result is similar for glass wherein the locally activated biosite areas will have low aqueous surface tension which are surrounded by relatively high surface tension, thereby constraining the capillary dispensing to the biosite area.

Creating and constructing self assembling probe arrays or universal arrays enables on-line configuration of the biosites wherein an unvarying probe array (capture probes) is activated by binding to a cognate set of adapters (target probes) to yield a moditied probe arrav which is specifically configured for analvsis of a target or target mixture.
The Universal Array format overcomes si';nificant obstacles that currently prevent probe array technolo~.Ty from being implemented in a commercially broad s mannzr. Fundamentally, probe arravs that allow for hiehly parallel analysis of bindins =
events require specialized equipment to manufacture and sophisticated instrumentation to interpret the binding patterns. Unfortunately, the current manufacturing processes for making biosite arrays, such as ink-jet, robotic deposition, or photolithographic in-situ svnthesis are relatively inflexible. These techniques are designed to make a large number of specific arrays to cover the cost of setup and operation. Hence, small volume custom arrays would be prohibitively expensive.
In contrast, the Universal Arrays system as taught herein solves this problem by taking advantage of efficient hiQh volume manufacturing techniques for the capture probe arrays only. In this fashion, each customer can use a pre-manufactured, high density is biosite capture array that is readily "tailored or customized" by the end-user for their specific target analyte screening. For this invention, "target analyte" is defined as the solution-state solute to be analyzed via binding to the probe array. In short, customization of the array can be perfon,ned in the customer's laboratory. The end-user synthesizes or produces bifunctional target probes containing two separate binding domains, one binding domain cognate to a specific member in the array (capture domain) and another binding domain specific for the target analytes of interest (target domain).
In an actual assay, end-users add their customized bifunctional probes to a solution phase mixture of analytes and incubate in a reaction chamber containing a pre-manufactured universal capture array. Altematively, the capture array is incubated first with the bifunctional probes followed by an addition of the analyte mixture (see Fig. 5). Fig. 5a is a diagram showing a Universal Array. The analytes self assemble onto the array in a sandwich-mode by selective binding of their bifunctional probes to both the complementary portions of the target and the capture array. The resulting addressable, self assembled WO 98129736 PC1'1[JS97/24098 atra}s is easi(}, analvzed with the complimentary proximal detector,timager.
Teachings for constructing the surface bound capture probes and target probes are outlined below.

1. Cerpttn=e Probes The surtace bound universal capture probes are: acranged in an array of biosites attached to a solid support. Each biosite consists of a niultitude of specific molecules distinct in function or composition from those found in every other biosite in the array.
These capture probes are designed to have a specific composition or sequence to provide rapid and efficient binding to the capture domain of the target probes. The specific composition is also chosen to minimize cross association betNveen capture probes and ia their specific target probes.
Specifically for a nucleic acid capture probe the surface bound capture array should be designed for optimum length, base composition, sequence, chetnistry, and dissimilarity between probes.
The length of the nucleic acid capture probe should be in the range of 2-30 bases and preferably in the range of 5-25 bases. More preferably, the length ranges from about 10-20 bases and most preferably is at or about 16 bases in length to allow for sufficient dissimilarity among capture probes. Length is also adjusted in this range to increase target probe binding affunity so that capture probe airays can be activated by addition of target probe mixtures as dilute as 10'9M. This allows target probes to be synthesized in Zo small scale and inexpensively. Also, length is adjusted to this range to reduce the rate of target probe dissociation from capture probe arrays. This allows the activated capture probe arrays to be washed thoroughly to remove unbound target probes, without dis-sociation of specifically bound target probes from the surface. With capture probes in such a size range, the complex formed by and between the target probe and capture probe interaction is stable throughout subsequent air drying, and can be stored indefinitely with refrigeration.

~

A preferred percentage base composition for capture probe array sets is in the rsn',e or at or around 30-40% G. 30-40% C, 10-20% A. I0-20 ,% T. RelativeIv G+C rich capture probes are desirable such that the thermodynamic stability of the 'resulting capture. ta r~-et probe pairin~~ will be high, thus allowing for surface activation at low s added target probe concentrations (e.g., in the ranve of 10'gNI). Nearest neighbor =
frequency in the capture probe set should minimize G-G or C-C nearest neighbors. This criterion minimizes the possibility of side reactions, mediated via G-quartet formation during capture probe attachment to the surface, or during the capture probe-target probe binding step.
For capture probe sets it is desirable to obtain a set structure such that each member of the capture probe set is maximally dissimilar from all others. To obtain such maximally dissimilar sets, the following algorithm can be employed.
I) The set size is defined. In a preferred embodiment, 16, 24, 36, 48, 49, 64, 81, 96 and 100 constitute useful sizes.
2) The overall sequence structure of the capture probe set is defined. The length and base composition as described above are used to define such parameters. In general, the number of G bases and C bases are held equal as are the number of A bases and T bases. This equality optimizes the configurational diversity of the final sets. Thus, such sets will be described by the equation G,,CõA.Tm.
3) For a set structure defined by m and n, a random number generator is employed to produce a set of random sequence isomers.
4) One member of the random sequence set is selected to be used as element # 1 of the set.
5) The maximum similarity allowable among set members is defined.
Similarity is defined in terms of local pair-wise base comparison. For example, when two oligomer strands of identical length n are aligned such that 5' and 3' ends are in register, the lack of mismatches refers to the e,aur.-~m s:.,,_.,.r.. .,.azaz'w-'R~. .,~r.r m-~,,,v...M...
...~:_.,:xmw~::~,aa~a~saa~wr.c.aar~. . .. ,;. . .. ...c.~.v.,*-. n, . . .-: ..
. e:.~ ::..... .... .... . ... . ...~aamac. ..~=.. ,~,wn..~,-.,,,.,,....~....õ,.._--..~...-...__._ WO 98/29736 = PCT/US97/24098 situation where at all positions 1-n, bases in the two strands are identical.
Complete mismatching refers to the situation wherein, at all positions 1-n.
bases in the two strands are different. For example. a useful maximum similarity might be 10 or more mismatches within a set of 16, l 6mer capture probes.
6) A second member of the random sequence set is selected and its similarity to element # I is determined. If element #2 possesses less than the maximum allowable similarity to elennent # 1, it will be kept in the set. If element #2 possesses greater than the maximum allowable similarity, it is discarded and a new sequence is chosen for comparison. This process is repeated until a second element has been determined.
7) In a sequential manner, additional members of the random sequence set are chosen which satisfy the dissimilarity constraints with respect to all previously selected elements.
Standard deoxyribonucleic acid base homologues, or homologues with modified purine or pyrimine bases, or modified backbone chemistries such as phophoramidate, methyl phosphonate, or PNA may be employed in synthesis of capture probes.
The capture probe should be linked to a solid support. This can be done by coupling the probe by its 3' or 5' terminus. Attachment can be obtained via synthesis of the capture probe as a 3' or 5' biotinylated derivative, or as a 3'/5' ar.~+.ine modified derivative, a 3'/5' carboxylated derivative, a 3'/5' thiol derivative, or as a chemical equivalent. Such end-modified capture probes are chernically linked to an underlying microtiter substrate, via interaction with a streptaviden film (for biotin), coupling to surface carboxylic acids or epoxide groups or alkyl halides or isothiocyanates (for amines) to epoxides or alkyl halides (for thiols) or to surface amines (for carboxylic acids). Other attachment chemistries readily known to ithose skilled in the art can be substituted without altering general performance characteristics of the capture probe WO 98/29736 PCTlUS97/2409 arrays. Capture probe arravs can be fabricated by such chemistries using either robotic or nlicro ink jet technology.
In order to minimize cross hybridization during the target probe activation step.
capture probe sets are constructed such that every member of the capture probe set has a lzneth fiich is identical or differs by no more than I base from the average length of the set. and possesses an overall gross base composition which is identical or substantially similar to all other members of the set. These two criteria interact to allow the free energy of all target probe/capture probe pairings to be identicaI. The above described algorithm generates such sets of probes.
It is important that the sequence of each member of the capture probe set differ from every other member of the capture probe set by at least 20%, preferably 40%, more preferably 50% and most preferably 60%. This extent of sequence homology (less than 80% between any two members of the set) prohibits target probes from binding to members of the probe set other than that to which it has been designed.
1s There are numerous capture prabe sets that satisfy the general design criteria as outlined above. Presented below is a specific example of a 16 element capture probe set generated by the above described algorithm which adequately satisfies the above criteria.
For this example, capture probe length is held at 16 bases and base composition is fixed at G5C5T3A3 among all 16 members of the set. There are no more than 3 G-G or C-2o C pairings per capture probe element. This particular capture probe set is designed to be linked to microtiter support via an amine linkage at its 3' terminus. However, a 5' aniine linkage, or other chemistries could have been used as well.
The top-most array element (#1) has been chosen as a standard. Detailed inspection of this set shows that every member of the set differs from every other 25 member of the set by at least 10 base mismatches, thus satisfying the criterion of no more than 50% homology between capture probe set elements.

.F:,.

SEOLtEi~tCE 9 CAPTURE PROBES, 16 MERS
SEQ ID NO:1 5'-TGATTCAGACCGGCCG-3'a SEQ ID NO:2 5'-CCCGGGGCGTCTTAAC-3'a SEQ ID NO:3 5'-GGACGCCATATGCGCT-3'a SEQ ID NO:4 5'-TGAGGGCTCCGCCATA-3'a SEQ ID NO:5 5'-AACCCGTGACGTGTGC-3'a SEQ ID NO:6 5'-AGCATCGCCGGTCCTG-3'a SEQ ID NO:7 5'-CCTGCAAGGCTGACGT-3'a SEQ ID NO:3 5'-CAGTTGTCGACCCCGG-3'a SEQ ID NO:9 5'-C:GGCGCGTCCAATTCG-3'a SEQ ID NO:10 5-ATCGATCTGAGGGCCC-3'a SEQ ID NO:1 I 5'-GTACATGCGGCCTGCA-3'a SEQ ID NO:12 5'-TAGCCGCTCGCTAGAG-3'a SEQ ID NO:13 5'-CCTAGTGATGACCGGC-3'a SEQ ID NO:14 5'-GTCTGAGGGCAACCTC-3"a SEQ ID NO:15 5'-CTAGCTGGCTACGCAG-3''a SEQ ID NO:16 5'-GCCATCCGCTTGGAGC-3.a a = amine linkage to solid support, such as a 3' propanolamine, coupled to a carboxylate modified surface via amide linkage or epoxide modified surfaces.

, SEQUENCE # ELEMENTAL TARGET PROBES (coLynate to capture probes) SEQ ID NO: 17 3'-TTACTAAGTCTGGCCGGC-5' SEQ ID NO:18 3'-TTGGGCCCCGCAGAATTG-5' SEQ ID NO:19 3'-TTCCTGCGGTATACGCGA-5' SEQ ID NO:20 3'-TTACTCCCGAGGCGGTAT-5' SEQ ID NO:2 1 3'-1 I71 GGGCACTGCACACG-5' SEQ ID NO:22 3'-T'ITCGTAGCGGCCAGGAC-5' SEQ ID NO:23 3'-TTGGACGTTCCGACTGCA-5' SEQ ID NO:24 3'-TTGTCAACAGCTGGGGCC-5' SEQ ID NO:25 3'-TTGCCGCGCAGGTTAAGC=5' SEQ ID NO:26 3'-TI'TAGCTAGACTCCCGGG-5' SEQ ID NO:27 3'-TTCATGTACGCCGGACGT-5' SEQ ID NO:28 3'-TTATCGGCGAGCGATCTC-5' SEQ ID NO:29 3'-TTGGATCACTACTGGCCG-5' SEQ ID NO:30 3'-TTCAGACTCCCGTTGGAG-5' SEQ ID NO:31 3'-TTGATCGACCGATGCGTC-S' SEQ ID NO:32 3'-TTCGGTAGGCGAACCTCG-5' 2. Target Probes A target probe set is designed and constructed to bind to the capture probe set in a specific manner, i.e., each target probe element binds to only one element of the capture probe set. Thus, a mixture of target probes can be administered to a capture probe array formed on the bottom of a microtiter well, or equivalent surface. For the nucleic acid embodiment of the Universal Array, subsequent to binding, the target probe set will partition itself among capture probe set members via Watson-Crick base pairing, thereby delivering a unique binding domain (cognate to analyte) to each site in the probe array.
There are two general methods that can be employed by the end-user to synthesize customized nucleic acid-based bifunctional target probes. The simplest and _ niost direct method is to synthesize a sin3le oligonucleotide that contains the two dumains (capture and analvte) separated bv a(inker re,ion using a standard automated DNA synthesizer. As a class, the bifunctional target probes for a nucleic acid enibodiment possess a structural domain cognate to the capture probe which is the Watson-Crick complement to one element of the capture probe set. Its length and base sequence is thus defined by that of the capture probe via standard rules of antiparallel Watson-Crick duplex formation. In addition, the tar;et probe also contains one of the following structural domains:
a. Cognate to a Small Segment of a Solution State rVitcleic Acid Target Analyre This is the component of the target probe whiich is complementary via Watson-Crick pairing to the solution state target nucleic acid to be analyzed. In general, its sequence has no correlation to that of the domain which is cognate to the capture probe.
However, several general desip criteria should be met.
First, for ease of target probe synthesis, the ur.iique domain in the range of about 5-30 bases in length, and preferably in the range of about 10-25 bases in length. With shorter target probe domains, analyte binding affuiity is insufficient, and longer target probe domains present synthesis difficulties.
Second, when the unique sequence is equal in length or longer than the capture probe set, the unique elernent should possess a sequerice which is no more tLa.n 30%
homologous to the Watson-Crick complement of any capture probe element. This criterion eliminates inappropriate association of the unique target probe segment with members of the capture probe set.

r~~~~T~~ ~ ~.~ ~

WO 98/29736 pCT[US97/2409~
h. Cognate to a PriminJ Site (ised fnr Biochemical Amplification Such as I'CR and LCR
This domain essentially creates nucleic acid an--plification primers with taits complenientary to capture probe sites in a Universal Array. After amplification, the s resulting amplicon sets can be directly hybridized to the capture probe array and analyzed as described below.
c. Chemically tLfod fied f'vr Direct Linkage Another method of synthesizing bifunctional DNA target probes consists of individualiv and separately synthesizing analyte and capture sequence oligos that are chemically altered to incorporate a reactive funetionality which will allow subsequent chemical linlca.ae of the two domains into a single bifunctional molecule. In general, the 5' or 3' terminus of each oligo is chemically altered to facilitate condensation of the two sequences in a head to tail or tail to tail manner. A number of methods are known to those skilled in the art of nucleic acid synthesis that generate a variety of suitable func-ts tionalities for condensation of the two oligos. Preferred functionalities include carboxyl groups, phosphate groups, amino groups, thiol groups, and hydroxyl groups.
Further, chemical activation of these functionalities with homo- or heterobifunctional activating reagents allows for condensation of the activated oligo with the second functionalized oligo sequence. Some examples of the various functionalities and activating reagents that lead to condensation are listed below:

Terminal Functionali AC'TIN'ATING AGENT
(3' or 5') (Homo or Heterobifunctional) NHZ (amino) -NHS-NHS, NHS-maleimide, iodoacetic anhydride, EDC (carbo-diimide) SH, (thiol) maleimide-NHS
zs COOH (carboxyl) EDC (carbodiimide) OH (hydroxyl) carbodiimide (EDC) PO ( ho hate) N-meth liinidazole (EDC) POS al ha-thio hos hate maleimide-maleimide, mafeimide-NHS

~.~

A specilic example of the target probe domains that are cognate to the capture probe set of the Universal Array and can be moditied to allow for direct binding to a specitic3ll}- modified probe, nucleic acid or other molecule capable of selective binding to the analvte of interest is illustrated in Fisi. Sb. Fi'. 5b is a diagram showing direct binding for a tar'et probe. As shown in Fig. 5b, the target probe is constructed from two parts: the first is a presynthesized probe (TP1) complementary to a capture probe which has a Iinl:aize element for attaching the second target complex (TP2). Such embodiment yields a high degree of simplicity for the customer since the first tarQet component can be offered in a ready-to-use format.
to A sample protocol for the two piece approach is as follows:
I) Obtain TP I from conunercial source, e.g., Genometrix (synthesized as 3' amine, 5' thiol);
2) TP2 synthesized as an amine;
3) TP2 is mixed with iodoacetic acid anhydride in "Buffer A " to generate the iodoacetate derivative TP2*;
4) Ethanol ppt, run over G25 spin column and collect the excluded volume which contains TP2* only, with small inolecule reactants removed;
5) TPI + TP2* are mixed with "Buffer B";
6) Separate on G50 spin column.
For this invention, "Buffer A" consists of 10mIVI sodium citrate, pH 7.0, and "Buffer B" consists of l OmM sodium bicarbonate, pH 9Ø

3. Linker In some instances, a chemical linker may be needed to separate the two nucleic acid domains of the target probe, to minimize stearic interaction between the target probe and the solution state nucleic acid analyte. This linker may be constructed from nucleic acid building blocks. For example, the sequence T. (where n= 1-5) is preferred because stretches of T are readily synthesized and minimize the likelihood of sequence dependent interactions with capture probe. otlier target probe domains, or the solution phase nucleic acid analyte.
Hoti'ever, the linker is more preferably synthesized from an inert polymer, such as oli;,~o-zth-=lene (Ylvcolate linka-es (-O-CH?-CH?-O-), Linkkszes with n=3 are commer-s cially available as the phosphoramiodate for ready synthesis into oligonucleic acids via standard phosphodiester linkages. From one to five linkers can introduced as needed.
Detailed below is a specific example of the invention based upon the capture probe set described above. Here, the linker domain is listed as two repeats of a tri-ethvlene gl}-colate synthon, linked by a phosphodiester linkase into the target oligo-nucleotide backbone.

WO 98/29736 PCT/iJS97124098 NUCLEIC ACID ANALYTE (TP2) C'APTURE PROBE (TPI) 5'-----CCACACTGGAACTGAGA ------3' 5'-TGATTCAGACCGGCCG-3'a IIIIIIIIIIIIIIIII I:LIIIIIIIIIIIIII
3'-GGTGTGACCTTGACTCT-----(Tn)---ACTAAGTCTGGCCGGC-5' NUCLEIC ACID ANALYTE CAPTURE PROBE
5'----- CCACACTGGAAC.TGAGA------ 3' 5'-TGATTCAGACCGGCCG-31a IIIIIIIIIIITIIIII IIIIIIIIIIIIIIII
3'-GGTGTGACCTTGACTCT----- (X)----- ACTAAGTCTGGCCGGC-5' TARGET PROBE 2 X LINKER I'ARGET PROBE 1 X = -0?0--{O-CH;-CH,-O-OPO;-):-0 DIETHYLENE GLYCOLOATE LINKAGE

3 ' AMINE-----------5' -OP02-0-CH,-CHZ-CHZ=-CH,-CH2-CH,-SH
3' PROPYL AMINE TARGET PROBE 2 NH2-CH2-CH (OH) -CH2-OP02-12-0-3' -------------5' I-CH2-CO-NH-CHZ-CH(OH)-CH2-OP02-J2-O-3'------------- 5' COUPLED TP1 + TP2 PRODUCT

5' THIOL DERIVATIVE OF TP1 3' ALKYL HALIDE OF TP2 3'- [TP11 -OP02-0-CH2-CH2-CH2-CH2-CH2-CH2-S-CH2-CO-NH-CH2-CH (OH) -CH2-OPOZ-} Z-O- [TP2 ) -5' TP1 = target probe 1 TP2 = target probe 2 -4i-~~

WO 98/29736 PCTl1JS97t24098 A Universal Arrav having 16 capture probes within a single well of a 96 well microtiter pi.ite is shown in Fig. 5c. Fig. 5c is a printed computer image showing a multi-nticrotitzr -weil proximal CCD image of a=1 x-1 Universal Array. In Fig. 5c, tar'_et specific h}-bridization is observed in 15 out of the 16 oligo elements in the arrav. The s results of 15 target specific hybridizations conducted simultaneously in 3 separate reaction chambers in a multiwell reaction vessel are qtiantitativelv assessed from the digital image obtained from the proximal CCD imager. Hybrids are digoxigenin end-labeled olisonucieotide targets detected using anti-digoxigenin antibody-alkaline phosphatase conjugate and ELFTU fluorescence. In this assay (from Molecular Probes, Inc.) the antibodv binds to the digoxi~enin group, delivering alkaline phosphatase to the bound tarQet. The alkaline phosphatase converts the non-fluorescent ELF
precursor to a fluorescent product which can be detected by UV irradiation.
Fig. 5d is a printed computer ima-e showing a single microtiter well proximal CCD image of a 4 x 4 universal array. Fig. 5d shows the target specific hvbridization of 4 is of the 16 oligonucleotide elements in the array at positions A2, B2, C2, and D2. Note the desirable absence of significant cross hybridization, which has been specifically minimized by imposing the maximum dissimilarly design constraints. Hybrids are digoxigenin end-labeled oligonucleotide tarsets detected using anti-digoxigenin alkaline phosphatase conjugate and ELFTM fluorescence as described above.

4. Non-Nucleic Acid Embodiments Small molecule Universal Arrays can be employed for rapid, high throughput drug screening. In this format, surface bound capture probes consist of small haptens or molecules arranged in separated biosites attached to a solid support. Each biosite consists of specifically-addressable, covalently immobilized small molecules such as haptens, drugs and peptides. These organic capture molecules are designed to have a high affinity association with a bispecific ligand. These ligands contains both a domain cognate to the small immobilized organic molecule (capture probe) and cognate to the analyte of ,.~,,.~...-,.~s~,~ .,_ ...,.,-.

WO.98/29736 PCTIUS97/241198 interest. The domain cognate to the analyte can associate either directlv to this taraet or to a label on the analyte.
Specific examples of bispecific ligands include, without limitation, anti-body:antibody. antibody:receptor, antibody:Iectin, receptor:receptor.
bispecific anti-bodies. antibody:enzyme. antibody:streptavidin. and antibody:peptide conjugates.
Anah-tes can include, but are not limited to, dsDNA, ssDNA, total RNA, mRiNA, rRNA. peptides, antibodies, proteins. organic enzyme substrates, drugs, pesticides, insecticides and small organic molecules.
Conversely, the format for a small molecule Universal Array can be inverted so that the macromolecular ligand becomes the capture probe. Thus, a Universal Arrav (ivlacromolecular Universal Array) may contain large macromolecules such as, without limitation, antibodies, proteins, polysaccachrides, peptides, or receptors as the immobilized capture probe. In turn, unique small molecule tags having a specific, high affinity association for the macromolecular biosites are covalently attached to various 1s probes cognate to the analyte. These labeled probes novv represent the bispecific component cognate to both the capture macromolecule and the target analyte.
Some representative examples of small molecules (haptens or drugs) are listed in Table 1 below. This is only a partial list of commercially available antibodies to haptens, steroid hotmones and other small molecule drugs. Examples of'these bispecific, small molecule-labeled macromolecules include antibodies, receptors, peptides, oligonucleotides, dsDNA, ssDNA, RNA, polysaccharides, streptavidin, oi- lectins. A partial list of 48 representative compounds for which specific antibodies are available include:
fluorescein; dinitrophenol; amphetamine; barbiturate; acetaminophen;
acetohexamide;
desipramine; lidocaine; digitoxin; chloroquinine; quinine; ritalin;
phenobarbital;
phenytoin; fentanyl; phencyclidine; methamphetamine; metaniphrine; digoxin;
penicillin;
tetrahydrocannibinol; tobramycin; nitrazepam; morphine; Texas Red; TRITC;
primaquine; progesterone; bendazac; carbamazepine; estradiol; theophylline;
methadone;
methotrexate; aldosterone; norethisterone; salicylate; warfarin; cortisol;
testosterone;

WO 98/29736 ' PCT/17S97J2409F
nortrptyline: propanolol: estrone: androstenedione: dicoxi~;enin: biotin;
thyroxine; and triiodothyronine.
The general concept of'Universal Arrays, tivhether they be DNA-based, small molecule-based, or protein-based allows for great versatility and end-user friendliness.
s The various configurations described allow for highly parallel, simultaneous, multi-piexed. high throughput screening and analysis of a wide variety of analyte mixtures.
STEP 3-- iVIOLECULAR LABELING STRATEGIES
Molecular labeling strategies relate to versatile labeling of the target molecules (fluorescence, chemiluminescence, etc.) consistent with proximal large area detection/
imasinQ.

I. Introduction - Conventional Labeling Labeling can be achieved by one of the many different methods known to those skilled in the art. In general, labeling and detection of nucleic acid hybrids may be divided into two general types: direct and indirect. Direct methods employ either covalent attachment or direct enzymatic incorporation of the signal generating moiety (e.g., isotope, fluorophore, or enzyme) to the DiNA probe. Indirect labeling uses a hapten (e.g., biotin or digoxigenin) introduced into the nucleic acid probe (either chemically or enzyrnatically), followed by detection of the hapten with a secondary reagent such as streptavidin or antibody conjugated to a signal generating moiety (e.g., fluorophore or signal generating enzymes such as alkaline phosphatase or horseradish peroxidase).
For example, methods of detecting the association/hybridization include, without limitation, fluorescent labeling, radioisotope labeling, chemiluminescence labeling, bioluminescence labeling, colorimetric labeling and electrochemiluminescence labeling.
Many known labeling techniques require a wash step to remove excess target from the hybridization/association solution, e.g., fluorescent, radioisotope, chemiluminescence, bioluminescence and colorimetric labeling. Several of these will be described below.
_44-~~.:

WO 98/29736 ~ PCT/US97124098 2. Fluorescent Labeling Fluorescent labelin-,-, is suitable for this invention for several reasons.
First, potentially liazardous substances such as radioisotopes are avoided.
Furthermore, the fluorescent labeling procedures are simpler than chemiluminescent methods since the latter requims enzymatic reactions and detection in the solution state.
Finally, the fluor-escent labeling approach can be modified to achieve the highest sig-nal-to-nose ratio SNR
among the safest labeling techniques by utilizing secondary linl:er chemistries that enable the attachment of hundreds of fluorescent dye molecules per target molecule.
The particular fluorescent dyes to be considered include commercially available agents such as ethidium bromide, as well as the novel dyes proposed in the affiliated chemistrv component. These labeling agents have intense absorption bands in the near UV (300-3 -50 nm) range while their pririnciple emission band is in the visible (500-650 nm) ranee of the spectrum. Hence, these fluorescent IabeIs appear optimal for the proposed proximal CCD detection assay since the quantum efficiency of the device is several orders of magnitude lower at the excitation wavelength (337 nm) than at the fluorescent signal wavelength (545 nm). Therefore, from the perspective of detecting luminescence, the polysilicon CCD gates have the built-in capacity to filter away the contribution of incident light in the UV range, yet are very sensitive to the visible luminescence generated by the proposed fluorescent reporter groups. Such inherently large discrimination against LN excitation enables large SNRs (greater than 100) to be achieved by the CCDs.

;. Electrochemiluminescence Labeling Electrochemiluminescence or electrical chemihaminescence (ECL) labeling, e.g., ruthenium (Ru) does not require a wash step to remove excess target from the solution and is highly sensitive. Briefly, for electrochemiluminescence as a method of detection, the internal surface of the reaction chamber is coated with a conductive material, e.g., gold, and the biosite is attached to this conductive surface (See Fig. 6).
Fig. 6 is a WO 98/29736 PCTlUS97/24098 dia.'._*ram showing ECL implementation in reaction vessel with proximal CCD
imaging.
Usin~_ one niic:rotiter well (of a 96 microtiter well plate) as a reactioti chamber, the biosites are deposited onto the internal circumference of the microtiter well by one of several mzthocfs as described above (ink-jet, capiliary, or photolitho?raphy/capiltary).
s This conductive surface acts as a cathode (positive lead), and an anode (negative lead) is provided by inserting a metal cup with an electrode protruding through its center into the reaction chamber (microtiter well). The electrode is positioned such that it is inserted into the hybridization solution. The voltage applied to the anode induces an electrochemical event at the labeled molecule surface which releases energy in the form of photons (liQ.ht).
' The specific ECL label, e.g., Ru, is attached to the target molecule by the conventional means. The labeled target is added to the hybridization solution and once hybridization occurs between the Ru labeled target and biosite, e.g., after sufficient time has passed for hybridization to be completed, a voltage is applied and only Ru labeled target associated (hybridized) with the biosite will emit light and be detected. In order for the Ru labeled target to be detected, it must be in proximity to the cathode.
The residual excess Ru labeled target not associated with the biosite will therefore not emit light.
The ECL reaction vessel is diagramed in Fig. 7. In Fig. 7, the thin film substrate, e.g, plastic, glass, etc., is patterned with a conductive metal, e.g., gold, platinum, etc., to form electrodes within the reaction chambers. Next, the biosites are deposited with one of several methods described above (ink-jet, capillary, photolithographic/capillary) onto the patterned electrodes. Finally, the resulting thin film substrate is bonded onto the reaction vessel which serves as the bottom of the reaction chambers.

4. Lanthanide Chelate Labeling 2s As an altetnative to ethidium-based fluorescent reporter groups, which are known for their tendency to absorb nonspecifically to surfaces causing increased signal background, the use of aromatic lanthanide (Ln) chelators may be used in the instant invention. :\lthough the lanthanide ions (Th and Eu specifically) have luminescent yields near to one (1). and emission lifetimes year to 100 usec. they absorb light weaklv and are therefore poor luminescent dyes. However, when chelated by an appropriately chosen aromatic donor, energy transfer can occur resulting in hiah overall luminescent yields.
s DPA (dipiccolimic acid) is the prototype for such an aromatic Ln chelator, and has excellent pliotophysical properties. However, its absorbence maximum is near 260 nm, which overlaps the DNA absorption band and is therefore inappropriate for the proximal CCD approach. Thus, the synthesis of modified DPA derivatives with the correct absorption properties and which have the capacity to be linked directly or indirectly to the tamet molecules have been developed.
Since three DPA equivalents bind per Ln ion, the preferred approach is to link the modified DPA to a polymeric lattice, which provides for close spacing of chelators and can be designed to have usefui DNA or RNA binding properties. These results suggest that a fused bicyclic DPA derivative is the candidate of choice.
Fig. 8 is a chemical drawing showing lanthanide chelators. The two classes of polymeric latice as illustrated in Fig. 8 can be employed for attachment of DPA deriva-tives, both based upon the use of synthetic polypeptides in the 104 MW range.
Synthesis can be conducted as described for simple DPA-peptide conjugates. The first polymer is to be used for covalent attachment to RNA via the transamination reaction to cytosine. This peptide lattice can be simple poly-L-lys. The second approach involves the coupling of modified DPA to a DNA binding peptide, which can be used to deliver the Ln chelate to RNA by means of non-covalent nucleic acid binding. For example, peptides can be synthesized in solution as a Lys3Arg, random co-polymer (average mw t 04).
Subsequent to the conversion of Lys residues to the modified DPA conjugate, RNA binding can be 2s driven by association with multiple Arg equivalents, taking advantage of the known helix selectivity of polyarginine. As for ethidium bromide (EB), addition of the non-covalent chelator conjugate can be made after washing to retain hybridization stringency.

~.~

WO 98/29736 . PCTIUS97124098 A detection system relates to parallel detection and/or imaging in the reaction vessel rontaining the reaction chambers using a proximal large area detector/imager.
1. General Description 5 Following the hybridization process of the multiplexed molecular analysis system. the amount of hybridized target molecules bound to each biosite in the reaction chambers of the reaction vessel must be quantitatively determined. The preferred detection/imaging system for quantifying hybridization for the instant invention is proximal charge-coupled device (CCD) detection/imaging due to the inherent versatility (accommodates chemiluminescence, fluorescent and radioisotope target molecule reporter groups), high throughput, and high sensitivity as further detailed below.
The detection/imaging apparatus used for the multiplexed molecular analysis system is comprised of a lensless imaging array comprising a plurality of solid state imaging deviees, such as an array of CCDs, photoconductor-on-MOS arrays, is photoconductor-on-CMOS arrays, charge in,jection devices (CIDs), photoconductor on thin-film transistor arrays, amorphous silicon sensors, photodiode arrays, or the like. The array is disposed in pro:cimity to the sample (target molecules hybridized to the biosites) and is comparable in size to the reaction chambers. In this manner, a relatively large format digital image of the spatial distribution of the bound target molecules is produced without requiring the use of one or more lenses between the sample and the imaging array. This apparatus offers:
1) high sensitivity (subattomole DNA detection);
2) high throughput (seconds for complete image acquisition);
3) linear response over a wide dynamic range (3 to 5 orders of magnitude);
4) low noise;
5) high quantum efficiency; and 6) fast data acquisition.

,~.~ .

WO 98/29736 CA 02389358 2002-06-20 ~ ~ i i ~~~ ++~ +uya 'Moreover by placing tlie imaging array in proximity to the sample as illustrated in Fig. I. tliz collection efficiency is improved by a factor of at least ten (I00 over any lens-based tectuiiyue such as found in conventional CCD cameras). T11us. the sample (emitter or absorbzr) is in near contact with the detector (imaging, array), thereby eliminating s conventional imaging optics such as lenses and mirrors. This apparatus can be used for detectina and quantitatively imaging radioisotope, fluorescent, and chemiluminescent labeled molecules. since a Iensless CCD array apparatus is highly sensitive to both photons and x-ray particles. Hence a single ima;ing instrument can be used in conjunc-tion with numerous molecular labeling techniques, ranging from radioisotopes to fluorescent dyes.
The detection/imaging apparatus invention as tztu,ht herein can be divided into t-,vo subclasses. The first subclass entails a static platform, whereby a plurality of irnaoing devices are arranged in a relatively large format area comparable to the sample size.
~s The second subclass entails a dynamic platform that enables a smaller set of imaging devices to image a relatively large format sample by moving either the array of imaging devices or sample, relative to one another.
Thus, the dynamic embodiment of the detectionrlimager invention generally concerns a method and apparatus for ultrasensitive detection, high resolution quantitative digital imaging and spectroscopy of the spatial and/or temporal distribution of particle emissions or absorption from/by a sample (target molecules) in a relatively large format.
The apparatus of this invention includes:
a) a large area detector array for producing a relatively large image of detected particle distribution without the use of optical lenses;
b) a scanner for moving either the sensor an-ay or the sample in a manner for efficient imaging; and c) a source of energy for exciting the sample or providing absorption by the sample.

=49-w:~

UptimaIly, the ratio of detector array size to sarripie image is one (i) for a static format and less than one (1) for a dynamic format.
.an ztectronic schematic of the proximal detector/imager to be used with the multiplexed molecular analysis svstem is shown in Fig. 9. Fig. 9 is a diagram showina a multiplexed molecular analysis svstem electronics schematic. As illustrated in Fig. 9, the reaction vessel is placed directly on the fiber optic faceplate 22 which is bonded to the sensor array. The faceplate provides sensor isolation to accommodate routine cleaning, as well as atfording thermal isolation for ultrasensitive detection under cooled sensor operation. Also the optical faceplate can serve to filter excitation radiation by employing selective coatings. The sensor array is comprised of a plurality of smaller sensors such that the composite array approaches the surface area of the reaction vessel.
The excitation source serves to excite the fluorescent reporter groups attached to the target molecules.
Depending on the chosen reporter groups, the excitation source can be either a UV lamp, laser, or other commonly used light source used by those skilled in the art.
The sensor array driver circuitry includes clocking, biasing and gating the pixel electrodes within the sensors. The cooling circuitry controls the thermoelectric cooler beneath the sensor array to enable ultrasensitive detection by providing very low thermal noise.
Basically, the user selects the required temperature of operation and through feedback circuitry, the sensor array is held constant at such temperature. The image receive circuitry is responsible for obtaining the digital image from the sensor array and includes preamplification, amplification, analog to digital conversion, filtering, multiplexing, sampling and holding, and frame grabbing functions. Finally, the data processor processes the quantitative imaging data to provide the required parameters for the molecular analysis outcome.
Also, a computer display is included for displaying the digital image.

~:_ _. a ~:.

3. Sen.snr. trray Implementations A preferred embodiment of the detection/imaging sensor array of the invention consists of a plurality of CCD arrays CCD 1...CCDN assembled in a larae format module as illustrated in Figs. I QA-1 OC. Fig. I OA depicts a CCD array w-ith multiple pixels being exposed to a labeled bioloaical sample 32 which causes the collection of electrons 34 beneath the respective pixel gate 16. Individual CCD airrays are closely aligned and interconnected in particular geometries to form a relatively large (greater than 1cm'-) format imagin' sensors of the linear array type as shown in Fig. l OB or the two dimensional row and column type as shown in Fig. IOC..
Numerous CCD tiling strategies can be explored to determine the best tradeoff analysis between detection throughput and instrument cost. A large format tiled array with several wafer scale CCDs would provide simultaneous detection of all biosites within the reaction vessel within seconds. However the cost of the large (8.5 x 12.2 cm) CCD sensor array may be prohibitive. An engineering compromise is therefore preferred, is balancina the use of smaller devices to significantly reduce the cost of the tiled array, while also matching the throughput with the other processes in the overall multiplexed molecular analysis system.
As shown in Fig. 1OA, each CCD array CCD1...CCDN is formed, in the conventional manner, by groNving and patterning various oxide layers 14 on a Si wafer/
substrate 12. CCD gate electrodes 10 are then formed by deposition of polysilicon or other transparent gate material on the gate insulator or field oxide 14. A
dielectric or polymer layer 18, preferably of light transmissive material such as silicon nitride or glass, SiO, or polyamide is then formed over the electrodes 16.
Preferably, in a labeled molecule embodiment, a filter shown in dotted lines 17, which may be formed of an aluminum or tungsten metal grating, or dielectric multilayer interference filter, or absorption filter, is formed in the dielectric layer 18 between the surface and the metal electrode 16. The filter is adapted to block the excitation radiation . .. A. -,~ . -.~..~ . ,~..-.~-.. . . _ ~.-~ . _ . : ,~ .~,~...

WO 98/29736 ' PCT/US97/24098 and pass the secondary emission from the sample 20. In a static platform embodiment, the sensor nlodule remains fixed with respect to the sample. Hence to achieve the relativziv large imaging format, a plurality of imaeing devices CCD I...CCDN
should be arranRed in a module as illustrated in Figs. I OB and I OC. The module can be packaged for easv installation to facilitate multiple modules, each for specific applications. Various tiling strategies have been documented and can be employed to minimize the discontinuitv between devices, such as described in Burke, et al., "An Abuttable CCD
Imager for Visible and X-Ray Focal Plane Arrays," IEEE Trans On Electron Devices, 3~(5J:1069 (vlay, 1991).
As illustrated in Fig. 10A, a reaction vessel 20 is placed in proximity to the CCD
arrav sensor 10. The sample can be excited by an external energy source or can be internally labeled with radioisotopes emitting energetic particles or radiation, or photons may be emitted by the sample when labeled with fluorescent and chemiluminescent substances. Conversely, direct absorption may be used to determine their presence. In this case, the absence of illuminating radiation on the detector may constitute the presence of a particular molecule structure. Preferably the sample can be physically separated from the CCD detector by the faceplate which is trmsparent to the particle emission.
The CCD detection and imaging arrays CCD I... CCDN generate electron-hole pairs in the silicon 12 (see Fig. 1 A) when the charged particles or radiation of energy h v shown by the asterisk 32 arising from or transmitted by the sample are incident (arrows 30) on the CCD gates 16. Alternatively, the CCDs can be constructed in a back illumin-ation format whereby the charged particies are incident in the bulk silicon 12 for increased sensitivity. The liberated photoelectrons 34 are ther,L collected beneath adjacent CCD gates 16 and sequentially read out on a display conventionally.
Silicon based CCDs are preferred as the solid state detection and imaging sensor primarily due to the high sensitivity of the devices over a wide wavelength range of from I to 10,000 A wavelengths. That is, silicon is very responsive to electromagnetic ~,~xr,~~:,~, radiation from the visible spectrum to soft x-rays. Specifically for silicon, only 1.1 eV of energy is required to generate an electron-hole pair in the 3.000 to 11.000 A
wavelength ran'e. Tlius for visible tight, a single photon incident on the CCD gate 16 will result in a sinLfle electron charge packet beneath the gate, whereas for soft x-rays, a single beta particle (typically KeV to MeV range) will generate thousands to tens of thousands of electrons. Hence the silicon CCD device provides ultrisensitive detection and imaging for low energy alpha or beta emitting isotopes (3H, "C, 35S) as well as high energy alpha or beta emitting isotopes (=''-P, 'I). Consequently, the CCD is both a visible imager (applicable to fluorescent and chemiluminescent labeled molecular samples) and a particle spectrometer (applicable to radioisotope labeled samples as well as external x-ray radiated samples). Thus, the CCD can provide simuitarieous imaging and spectroscopy in the same image.
In addition to the high sensitivity, the CCDs offer a wide dynamic range (up to 5 orders of magnitude) since the charge packet collected 'beneath each pixel or gate 16 can 1s range from a few to a million electrons. Furthermore, the detection response is linear over the wide dynamic range which facilitates the spectroscopy function, since the amount of charge collected is directly proportional to the incident photon energy.
Hence, no reciprocity breakdown occurs in CCDs, a well-known limitation in photographic film.

3. Scanning Mechanics To image the reaction vessels with a smaller sized and less expensive sensor array, the reaction vessel can be imaged in a column-by--column manner as it is moved across the sensor array with a scanning mechanism. A plurality of imaging devices can be arranged in a module of columns to minimize discontinuity. Also, the scanning can be accomplished with intentional overlapping to provide continuous high resolution imaging across the entire large format sample area.

WO 98/29736 PCT/US97/24tMP
EXAMPLE I
DIFFERENTIAL DETECTION OF THREE NHS-IMNIOBILIZED
HAPTENS USING UNIVERSAL ARRAYS
This example demonstrates reduction to practice of small molecule universal s arrays as iitustrated in Figs. 18 and 19. Fig. 18 is a graphical schematic layout of a microarray that will be printed on glass slides using the Hamilton 2200 Microlab robot.
This schematic layout illustrates the relative spatial location/addresses of three separate covalently immobilized haptens on to the glass substrate (e.g., digoxigenin, fluorescein, and biotin). The robot will print the array by depositing 10 nL volumes of each activated hapten (N-hydroxysuccinimide activated) on to an amino-silanized glass surface thus cerating a 4 x 4 matrix microarray. Each hapten will be deposited by the robot 4 times as illustrated by the schematic. For example, digoxigenin will be deposited at array addresses indicated by address locations A1, B2, C3, and D4. Similarly, fluorescin can be found at address locations A4, B3, C2, and D I and biotin at B2, B3, B4, and C3 as illustrated in the schematic. A buffer blank (control) will be deposited at locations A2, A3, D2, and D3. These buffer blanks should not generate a signal on the CCD
proximal detector in the presence of hapten detecting conjugates.
Incubation of these covalently immobilized hapten microarrays with an appropriate bispecific molecule (e.g., hapten recognition site and enzyme reporter site) such as an antibody/enzyme conjugate and subsequent detection of the appropriate chemiluminescent substrate should generate an image "pattern" on the CCD
detector as predicted by the schematic addresses shown in Fig. 1$. In this example, specific light generating substrate molecules are localized at predictable/addressable biosites in the array either individually or in a multiplexed fashion.
zs Briefly, in order to covalently immobilize the above described hapten microarray the following protocol was developed. First, several 22 x 22 mm square glass microscope cover slides (150 m thick) were washed in a container containing ALCONOX
detergent solution, and subsequently transferred to a clean container containing wann tap water to ~~

w0 ygMm PGT/US97/24098 rinse otf the detcrbent. This rinse step was followed bv two separate brief rinses, first in a container containino 100% acetone, then the slides were transferred to a rinse in a solution of I00'% methanol. The slides were rinsed one final time in deionized H,0 to remove traces of organic solvent. The clean glass slides were then oven dried at 37 C.
After dning. the clean slides were then surface derivitized bv vacuum deposition of a solution of 3-aminopropyltrimethoxysilane in a vacuum oven. The slides were laid do~vn in a metal tray on clean lint free paper towels: A I:3 solution of 3-aminopropyltri-methoxti=sliane and xylene was freshly prepared by mixing I ml of 3-aminopropyltri-methox}=silane (Aldrich) with 3 ml of dry p-xylene solvent in a small glass petri dish. The dish was covered with aluminum foil and a small needle puncture was made in the foil.
This solution was placed in the tray with the glass slides. The trav was subsequently covered with aluminum foil and placed in a NAPCO vacuum oven at 75 C under 25" of I-ia vacuum overnight. The next day the amino-silanized glass slides were removed from the vacuum oven and stored in a dry place until used.
i5 In order to robotically dispense and print hapten microarrays, four separate activated hapten solutions were made as follows. First, approximately 1 mg of the following compounds were weighed out into separate weigh boats: fluorescein-5-(and-6)-carbixamido)hexanoic acid, succinimidyl ester, followeci by 1 mg of sulfosuccinimidyl 6-(biotinamido) hexanoate and then I mg of digoxigenin-3-O-methylcarbonyl-y-amino-caproic acid-N-hydroxysuccinimide ester. Each activated hapten was dissolved in 100 L
DMSO. Subsequently 50 ul of each hapten was mixed into separate tubes containing 950 ul of 0.1 M Na.HCO3/NaCO3 buffer at pH 8.05. A blank solution containing 50 L
of DMSO into this buffer was also, made as a control for dispensing on to the array as described above. Each of the four solutions (100 L) was placed into 16 wells of a microtiter plate. The microtiter plate was then placed on the Hamilton 2200 Microlab robot and 10 nL aliquots were collected and dispensed by the robotic dispensing needle onto the amino-silanized glass cover slides at known address locations illustrated by the schematic layout in Fig. 18.

r~ ,.:~ .~. T ._.

WO 98/29736 PCTl1JS97124098 FoIto =ina microfluid dispensing of four separate (identical) glass cover slides by thz con:puter controlled robot needle the arravs were air dried for 15 minutes. To detect the imr; obilized haptens the glass slides were rinsed for 10 minutes in 10 ml of I x TBS +
0.1 %o Twezn-9 20 (Tris-Buffered Saline, 100 mlVl Tris-HCI, 150 mVi NaCi, pH
7.5).
Individual slides were then incubated with appropriate conjugate dilutions.
Image A was aeneraze3 by incubating one of the slides in 10 ml of a 1:5000 dilution of streptavidin:
horseradish peroxidase conjugate in 1 x TBS + 0.1 % TweenC 20. Image B was generated by incubating one of the slides in a 1:5000 dilution of anti-digoxigenin:
horseradish peroxidase conjugate. Image C was generated by incubating in a 1:1000 dilution of antifluorescin:horseradish peroxidase conjuaate. Finally, Image D was generated by incubating a fourth slide simultaneously with all three horseradish peroxidase conjugates at the above dilutions. Following conjugate incubation all slides were washed by a 10 minute rinse on a platform shaker in 10 ml I x TBS + 0.1% Tween 20 to remove excess conjugates. The slides were then imaged by adding 200 L of freshly made chemiluminescent substrate (SuperSignalT1' Substrate from Pierce Chemical) as reconunended by the manufacturer. The slides containing substrate were imaged by a 10 second integration time at room temperature on the proximal CCD detector described above.
Fig. 19A is a printed computer image showing specific imaging of biotin-2a addressable biosites detected using streptavidin:HRP conjugate (4 x 4 single well nzicro-array). In Fig. 19A, Image A was generated by incubating the small molecule 4 x 4 universal array with a streptavidin:HRP conjugate specific for biotin. As seen in this image, only biosites with addresses B1, C1, B4, and C4 known to contain biotin (refer to Fig. 18) are detected using proximal CCD imaging of chemiluminescent signals.
Specific addressing of these biosites generates a "box" image pattern.
As shown in Figs 19B and 19C, Image B and Image C are two additional 4 x 4 microarrays incubated with the indicated antibody conjugate. Fig. 19B is a printed computer image showing specific imaging of digoxigenin-addressable biosites detected ~~~~

usina anti-di'.:oxibenin:HRP conjugate (4 x 4 single well nlicroarra}=). As seen in 19B, onI}= biosites with addresses Al. B2, C3, and D4 known to contain diaoxigenin (refer to 18) are detected using proximal CCD imaginD of cherriiluminescent signals.
Fig. 19C
is a printed computer ima'e showing specific imaging of fluorescein-addressable biosites s detected usin< anti-fluorescein:HRP conju'ate (4 x 4 single welI
microarray). As seen in I 9C, only biosites with addresses A4, B3, C2, and D 1 k:nown to contain fluorescein (refer to Fig. 18) are detected usin; proximal CCD imaging of chemiluminescent signals.
Thus, the signals from these two small molecules aenerate the predicted "diagonals" as iilustrated in Fig. 18.
Additionally, in Fig. 19D, Image D illustrates simultaneous detection of all three haptens in a single well by simultaneously incubating a single 4 x 4 array with all three conju!zates. Fig. 19D is a printed computer image showin~ simultaneous imaginas of fluorescein, biotin, and di4oxigenin biosites detected using anti-fluorescein, anti-di'o:ci?enin and streptavidin:HRP conjugates (4 x 4 single well microarray).
This image generates the predicted "H" pattern as expected because wells A2, A3, D2 and D3 were blank (see Fig. 18).

EXAMPLE II
USE - MICROARRAYS IN A MICROPLATE
Several applications of the multiplexed molecular analysis system are detailed below which can be accommodated with a multiple well microplate serving as the particular reaction vessel. The novelty, however, is the plurality of biosites within each well. That is, each well in the multiple well microtiter plate contains N
biosites where N
ranges from 2 through 1,000. The upper bound is based on the resolution limitations posed by the bottom substrate of the microtiter plate used in conjunction with the proximal CCD detector/imager.
For example, each well or reaction chamber can contain 96 biosites as shown in Fig. 11. Fig. 1 I is a printed computer image showing microarrays within a microplate .:

reaction vessel. One single reaction chamber is shown as Figure 11 a. Thus, the reaction vessel essentially consists of microarrays within a microplate which cumulatively affords 9.216 (96 x 96) hybridization experiments per microtiter plate --- a 100 to 1 multiplexing capacity.
The specific multiplexed microtiter plate reaction vessels to be used with proximal imaging are constructed by bonding thin films (typically glass or plastics) to conventional bottomless microtiter plates. All commercially available microtiter plates tested to date are incompatible to proximal imaging due to the thickness and composition of the bottom substrates.
The biosites are deposited by one of the several methods disclosed, either before or after the bottoms are bonded to the plate. In both situations, the probe molecules comprising the individual biosites must be attached to the glass or plastic surfaces.
In a preferred embodiment, thin (50-300 m) vinyl substrates are amino or epoxy functionalized with silanes similar to glass substrates. Thin vinyl substrates are immersed in a 1-2% aqueous solution of polyvinyl alcohol at 65 C. The adsorbed polyvinyl alcohol is then reacted with either epoxy silane or amino silane, thus functionalizing the polymeric hydroxyl groups. Such optically clear vinyl substrates have the distinct advantage of blocking a large amount of the UV excitation source incident on the proximal CCD detector, but allowing the longer wavelengths (e.g. 500-650 nm) to pass through efficiently. This allows for greater sensitivity of labelled detector molecules that emit in such wavelength region.

Nucleic acid probe attachment to glass employs well-known epoxy silane methods (see Fig. 12) described by Southern and others (U. Maskos et al., Nucleic Acids Res (1992) 20:1679-84; S.C. Case-Green et al., Nucleic Acids Res (1994) 22:131-36; and Z.
Guo et al., Nucleic Acids Res (1994) 22:5456-65). Fig. 12 is a diagram showing glass and polypropylene surface coupling chemistries. With 3' amine-modified probes, covalent surface densities can be obtained having 1011 molecules/mm' which is near the theoretical packing density limit. Amino-modified polypropylene is a convenient altLtnatix-e tt.) a glass substrate since it is inexpensive and opticallv clear above 300 nm.
.Amine-moditic d polypropylene can be converted to a c:arboxvlic acid-modified surface bv treaunznt with concentrated succinic anhvdride in acetonitrile. Amine-modified probe is then coupled to this surface by standard carbodiimide chemistry in H,O to yield probes = 5 at densities near I 09/mm= (see Fig. 12).

EXAMPLE III
USE - MULTIPLEXED DIAGNOSTICS
The multiplexed molecular analysis system can be employed for immunoassay and probe-based diagnostics. For immunoassavs, the throughput of conventional ELISA
assays can be increased with the multiplexed microplate format wherein a patient sample can be simultaneously interrogated by numerous antigens/antibodies within a single reaction chamber (well).
Similariy for probe-based diagnostics, target molecules derived from a patient sample can be dispensed into a single well containing numerous biosites for diagnosing genetic or infectious diseases. For example, single-stranded nucleic acid probes which are complementary to 96 known mutations of cystic fibirosis are arranged within a single well in a microplate. Upon hybridization with the patient's DNA sample, the resulting binding pattern obtained from the proximal CCD detector/imager indicates the presence of such known mutations.
The system can also be employed for high throughput, probe-based infectious disease diagnostics. Here the array of biosites within a s.ingle well in the microtiter plate can comprise DNA probes complementary to known viral strains. For example, a panel of probes (biosites) is arranged to diagnose a number of sexually transmittable diseases within a single well (reaction chamber). Consequently for a single microtiter plate, numerous patient samples can be simultaneously interrogated each against a panel of numerous probes to provide a very rapid, cost effective diagnostic testing platform.

,.,.,,.
~ ,,~;<< . ~~=~ ... __,., Universal Arrays are perfectly suited for analysis and detection of multiple point mutations witliin a single PCR template. Often technical constraints are encountered when attempting to analvze multiple point mutations from a single PCR amplicon reaction. iViost point mutation analysis techniques such as iibonuclease protection assay.
s SSCP_ or CLEAVASET r' are well suited for detectinp a single point mutation per amplicon or DNA template and require lengthy del-based separation techniques.
The simultaneous. rapid detection of numerous point mutations within a single PCR
amplicon without an expensive, lengthy gel separation step is well beyond the capability of these technoloeies. Other newer, non-gel based technologies such as TAQMANTM are also poorly suited for multiplexed analysis within a single reaction vessel. Fig.
13 illustrates the concept of using Universal Arrays for point mutation analysis (genotyping) at a single loci. Fig. 13 is a dia4Qram showing genotyping by universal point mutation scanning. For example purposes only, Fig. 13 uses a single point mutation biosite to illustrate this type of analysis but could just as easily be simultaneously carried out on 25 different loci on a single PCR template as illustrated in Fig. 14.
Briefly, as shown in Fig. 13, the PCR template is aliquoted into 4 separate tubes (one for each dNTP) containing a standard sequencing mix, with the exception that dideoxynucleotides are not included. Instead, a single alpha-thio dNTP is substituted in each of the four separate mixes. Each mix also contains a single labeled primer with a universal sequence or "handle" at the 5' end which anneals just one nucleotide away from the mutation site on the PCR template (note: multiple primers each with unique universal sequences and complementary to different loci on the template is readily accomplished).
After standard thermal cycle extension reactions are complete each tube is briefly incubated with snake venom phosphdiesterase. Only primers and templates that were not extended during the sequencing reaction are vulnerable to digestion by this 3'-specific exonuclease. Mutation primers containing a 3' thiophosphate ester linkage are highly resistant to digestion.

'~rv.~ia....TG. ...., '.e?M: .-tcS4 ,:.':t...r:9+T..#S' :P.;==:~....F.n.x=-..e...--....._..ea, m. . -. :-. p.:.... iS t91>.3xR+~a'.. A, .;.,n. .K .. . .
;. .. . .As;tt~'._SlRR:. VS'Ãt%alRttinAi..:S:..~

In tlZis specific example. only the A reaction extended since a T was the next complementary base on the PCR template. Each digested, sequencing reaction mix in tuni is then hybridized to four niicrotiter wells each containin;,, identical immobilized microarra}=s complementary to the universal primer sequences. In this case, only the s microtiter -ell hybridized to the A reaction mix nives a positive siznal at a biosite loci complementary to the universal handle. In this fashion,, up to 96 loci could be probed for point mutatiotis on a single PCR template. Both strands in the PCR amplicon could be ''scanned" in this manner simultaneously to allow more room for many primers to anneal without competition for the same hybridization loci on the template. In Fig.
13, "5-DIG"
means 5' digoxigenin labeled.
For probe based diagnostics where both multiplexing within a single target molecule and low target concentrations are a problem, amplification with either PCR or LCR usina the microtiter plate in a microtiter well concept conjoined to the Universal Array has distinct advantages. In a preferred embodiment, universal "handles"
can be synthesized directly on the 5' end of Polymerase Chain Reaction or Ligase Chain Reaction primers and following in situ thermal cycling the amplified products can be simultaneously hvbridized to 96 separate biosites. This format has other diagnostic advantages such as homogeneous detection of amplified products without having to open or expose the sample well to the ambient environment.
Fig. 14 is a diagram showing microtiter-based tFaroughput genotyping. Briefly, Fig. 14 illustrates the concept of high throughput genotyping using microarrays. In practice, 96 separate PCR amplification reactions woulti be carried out using genomic DNA templates isolated from 96 different patient samples. The figure illustrates the concept of genotyping starting with 96 previously robotically purified PCR
templates from these reactions. Each purified PCR product from each of the 96 wells is split into 4 separate aliquots/wells of a 384 well plate. Each well in this new plate would contain a pre-made sequencing buffer mixture, 25 individual primers, a thermostable DNA
polymerase, and only one of the four ccthio-dNTP's. The primers would anneal in a WO 98/29736 -. PCT/US97/24098 juxtaposed fashion to the PCR template just one nucleoticle away from the nucleotide locus bzin__ genotyped. In each ot'the four wells, those primers juxtaposed next to the inctude-u nucleotide in the sequencing mix would be extended. Following the simul-taneous extension of 384 reactions, each of the 334 wells is in situ digested with snalce ; venom phosphodiesterase. Only primers in each reaction that had been extended by a single base are protected from digestion. All other DNA is degraded to mononucleotides.
Following a brief thermal denaturation of the exonuclease, the contents of all the wells is roboticallr transferred to a new 384-well microtiter plate containing sequence comple-ments microarrayed in a 5 x 5 microarray attached to the bottom of each well.
Each of the 25 primers that had not been digested would hybridize to its corresponding comple-ment in the airay and imaged on the CCD detector to define the genotype at each loci.
Fiz. 15 illustrates this homogenous multiplexed approach for the Polymerase Chain Reaction (PCR) simultaneously at 3 different loci. Fig. 15 is a diagram showing homogeneous in situ microarrav detection of multiplexed PCR amplicons. Ficy.

illustrates specific multiplex hybridization detection of PCR products using microtiter-based microarrays. Briefly, in this figure three separate amplification loci are being detected simultaneously. Each locus (e.g., PCR LOCUS 1) is defined by two specially modified amplification primers that define the ends of the amplified PCR
product. One primer in the pair, contains a fluorescently detectable label such as fluorescein. The other primer in the pair contains two domains, one is a unique universal sequence comple-mentary to a capture probe arrayed at the bottom of a single imicrotiter well and the other domain specific for template amplification. The universal sequence is attached to the amplification primer in a 5' to 5' linkage so that when the polymerase is amplifying the region of interest it does notjump over this specialized juncture, leaving the universal sequence as a single stranded motif. If a particular template in a sample well being amplified contains both primer loci (i.e., detection and capture sites), then a PCR product will be generated that can simultaneously hybridize and be detected to a complementary member of a universal capture array by the CCD proximal detector. Since only PCR

anlplll:oils hybridized to ii2eniblrs of the uiliversal arraV at the bottoill of eacll -,t'ZI' are proximal to thc detector, tlle assay requires no special separation step to detect hybridized ampticons and thus becomes homoQenous in nature.
Similarlv. Fig. 16 illustrates this multiplexed concept with Gap-Ligase Chain Reaction (G-LCR). Fig. 16 is a diagram showing homogeneous in situ mieroarrav detection of multiplexed gap-ligase chain reaction products. The ability to detect hybrid-ization events homo;,~eneously is provided by the fact that onlv molecules proximally associated with specific biosites can be ima-ed by the detector. Fia. 16 illustrates specific multiplex hybridization detection of Gap-Ligase Chain Reaction products using microtiter-based microassavs. Similarly, as described previously for PCR
products (see Fig. 15). this figure illustrates the assav at three separate l'tgation-dependent amplification loci simultaneously. Each locus (e.g., LOCUS 1) is defined by two specially modified primers that define the ends of the gap ligase chain reaction product. One primer in the pair, contains a fluorescently detectable label such as fluorescein. T'ne other primer in the pair contains two domains, one is a unique universal sequence complementary to a capture probe arrayed at the bottom of a sin=le microtiter well and the other domain is specific for a region on the template being detected. The universal sequence attached to this primer serves as a sequence specific single stranded handle. When the template is present in the sample then sequence directed ligation will join both the label and the universal handle into a single product. After many cycles this amplified ligated product can be simultaneously hybridized and detected to its complementary member on a universal capture array immobilized to the bottom of a microtiter well and imaged by the CCD proximal detector. Since only ligated products hybridized to members of the universal array at the bottom of each well are proximal to the detector, the assay requires no special separation step to detect hybridized amplicons and thus becomes homogenous in nature.

EXAMPLE IV
DRUG DISCOVERY/SCREENING ANALYSIS
In tflis example. a small molecule Universal Array could use high affnity, commercially available antibodies to numerous haptens. steroids, or small molecule druRs. A partial list of 48 representative compounds are enumerated in Table 1 for which specific antibodies are available. This table is only a partial list of commercially available antibodies to haptens, steroid hormones and other small molecule drugs.
TABLE I
tluorescein dinitrophenol amphetamine barbiturate acetaminophen acetoheYamide desi ramine lidocaine disitoxin chloro uinine quinine ritalin phenobaribital phenytoin fentanyl phencyclidine methamphetamine metaniphrine diLroxin penicillin tetrahvdrocannibinol tobramvcin nitraze atn morphine Texas Red TRITC primaguine progesterone bendazac carbamazepine estradiol theophylline methadone methotrexate aldosterone norethisterone salicvlate warfarin cortisol testosterone nortrptyline propanolol estrone androstenedione digoxigenin biotin thyroxine triiodoth onine Small molecule Universal Arrays are made by covalent attachment of small molecules such as those found in Table 1 to substrate surfaces. Immobilization of haptens, steroids, or drugs is accomplished by introducing a functionalized moiety at one end of the small molecule. These moieties are well known to those skilled in the art (e.g.
N-hydroxv-succinimide, maleimide, isothiocyanate, iodoacetamide or other amine or sulfur reactive moieties). Small function-alized molecules or drugs can then be reacted with NH2 or SH, derivitized plastic or glass substrates. Some specific examples of such commercially available activated haptens include NHS-fluorescein, NHS-biotin, NHS-digoxigenin, maleimide-biotin, and maleimide-tetramethylrhodamine.
Following deposition of the individual small molecule biosites, a bispecific ligand can be used to spatially localize specific binding events to given biosites.
The bispecific ~.

li~and c:an cUniprise, but is not limited to, antibodv-antibody conju~~ates, antibody-receptor. antibodv-streptavidin, antibody-peptide. antibodv-small molecule conjugates or bispecitic antibodies.
The bispecific liQand is specific to both the immobilized hapten or drug on the = s substrate surface (biosite) and the analyte being screened. Examples of Universal Array screenina are dia-uramed in Fin. 17. Fi-. 17 is a diagram showine small molecule universal arrav (drug screening/discovery). Fig. 17 illustrates the basic small molecule Universal Array concept using four different immobilized haptens in a single well.
Various bispecific molecules are diagramed for illustration purposes. Fig. 17 iIlustrates te four separate and distinct haptens immobilized at the botzom of each of 96 wells of a microtiter plate. Each locus or biosite in the array is defuied by four unique immobilized haptens illustrated in this example by fluorescein, digoxigenin, 2.4 dinitrophenol, and TRITC. Bispecific molecules uniquely specific for both the immobilized hapten and another labeled analyte in the sample are added to each well. In this fashion, different is multiple analytes can be simultaneously detected and their presence indicated by signals at specific hapten biosites. In this example, 96 individual samples can be assayed for four different analytes simultaneously. As shown, the fluorescein biosite detects a labeled receptor (protein) analyte, both the 2,4 dinitrophenol and digoxigenin haptens allow for the simultaneous detection or presence of two additional types of protein receptors in the 20 sample. Finally, the TRITC hapten allows for detection and presence of a specific enzyme substrate via an intervening enzyme conjugate. Once again, the proximal mode of detection allows for homogenous imaging of only those binding events at the surface of the array. The advantages of sucha multiplexed immunological approach is the exquisite specificity and variety of small molecules that comprise such a Universal Array 25 using non-DNA based recognition of biosites.
Actual reduction to practice of small molecule Universal Arrays is illustrated in Figs. 18 and 19 and described in Example I above.

EXAMPLE V
USE - GENE EXPRESSION ANALYSIS
The multiplexed molecular analvsis svstem is also useful for analvzing, the expression of hundreds of different mRNA species in a single tissue sample within a sin~~le -etl of a microtiter plate. Here synthetic nucleic acids form the distinct biosites which constitute numerous hiahly sensitive and selective hybridization analyses per samplz, employing only 50 L of sample extract. Such massive hybridization analyses enables the discovery and employment of numerous biomar.kers for specific diseases such as canc-Ir. Essentially, the search for biomarkers of early phase lung cancer becomes an iterative, combinatorial process. For lung cancer and other epithelial disease, several hundred mR_NAs are analyzed for their value as biomarkers at relatively low cost. In such an iterative process, the biostatistician becomes the end-user of the technology and a central component in the development of the final set of mRNA biornarkers.
Once an mRi~IA biomarker set is discovered by this iterative approach, the technology is naturally 1s suited for low cost, high throughput screening of large patient populations with the mRNA biomarker set of choice.

EXA1tiIPLE VI
USE - CELL SORTING
Conversely, intact cells are analyzed utilizing the multiplexed format of this invention. Specifically, most "cell enrichment" protocols involve either double label flow cytometry, or physical separation of cells via affinity chromatography of some kind. Both require access to an antibody which is specific to the cell type of interest.
In a multiplexed microplate format, the cell-specific antibodies are arranged in a matrix fashion within the reaction chamber (single well in the 96 well microplate). The key to making the cellular analysis work is creating a situation wherein such antibody arrays retain the capacity for high affinity and high selectivity binding of intact cells.

... .~.~9,,T~,re*rR!..:,.>?:'R, The procedure is to add a complex cellular mixture, e.,-, a biological saniple (for exainple. blood or sputum), to such an antibodv matrix, then with some local mixing, = allov.-in,, the cells to bind to the surface. If cells bind to such a matrix witli -ood affinitv and selzctiviny. they are then fixed to the matrix, permeabilized, and subjected to labeled = s probe hybridization (or PCR) in a fashion which is nearly identical to the methods which are currentlv used to anal-vze DNA or RNA in cells for microscopy or flow cytometry.
The principle benefit of the multiplexed format is that many different cell types are separated in a single well of a microtiter plate.

EXAMPLE VII
USE - MiCROORGANIC MONITORING
Microorganism monitoring applications can also be addressed by the multiplexed molecular analysis system. In particular for monitoring air, water, and food for micro-orQanisms. the system can rapidly and cost effectively provide detection and quantifica-tion of complex biological mixtures. An example would be a ribosomal RNA probe-based assay in Nvhich nucleic acid probes serving as the biosites are chosen to selectively capture RNA of characteristic microorganisms.
Basically, the procedure is initiated by preparing the microbial rR1VA sample for hybridization to the biosite array within the reaction chanlber. Following specific binding of the fluorescently labeled microbial RNA to the probe array, a two dimensional image results that uniquely characterizes the sample. The analyzer output is the microbial spectrum, consisting of the amount and type of microorganisms present in the sample.
The rationale for the proposed approach to simultaneous monitoring of microorganisms includes:
1) Fast microbial analysis can be achieved due to the avoidance of standard cell cultivation procedures which require days to perform. Moreover, the proposed highly sensitive proximal CCD detection procedure, combined with the inherent amplification ,dPt. _.R'...-:::y.x. s1iN~..,:..,'u9~l,..z.,.v~t..Y~rrra.n xi'a33LQ':.F.;riY.'Ah.'+.:3 -{'..tl5y{'Pf';:Ye..:.FM~'v.=tr'+'I~91vT.:CYe~,.~~+er.,Km.Fx,m=~-~rw..,Wa,~~-~hay.. ."K'RFTt'z~.ttMNp~:.o-gk2?t'.iMMf-B"~:.. .. .M'F~ t'1FW+*':o..k-BtNRFe:w.m~ 'FN'>c. ...

property of rILNf1,, reduces the combined sample preparation, assay, and detection time froni davs to hours.
Siniultaneous microbial monitoring can be achieved due to the high density arrays that support hundreds of immobilized probes per cm= to facilitate multiple microorg:uzism detection and identification in a high throughput manner.
Minimal labor ancl training is required since no cell culturing or gel-based sequencing is required. Instead, an operator merely subjects the prepared sample to auto-mated hybridization, washing, and drying processes to obtain the microbial spectrum.
4) tLlinimal equipment is necessary since the probe-based assav is integrated with the proximal CCD detection device, therebv alleviatinig traditional macro-detection techniques such as epifluorescent and confocal microscopy.

Tne following references may facilitate the understa.nding or practice of the certain aspects and/or embodiments of this invention. Inclusion of a reference in this list is not intended to and does not constitute an admission that the reference represents prior art with respect to the present invention.
Hansell, US 2,512,743 D. Bogg, F. Talke, IBiL1Jour. Res. Develop. (1984) 29:214-321 Burke, et al., "An Abuttable CCD Imager for Visible and X-Ray Focal Plane An:ays," IEEE Trans. On Electron Devices, 38(5):1069 (May, 1991).
Maskos, U., et al., Nucleic Acids Res. 20:1679-1684 (1992).
Stephen C. Case-Green, et al., Nucleic Acids Res. 22:131-136 (1994).
Guo, Z., et al., Nucleic Acids Res. 22:5456-5465 (1994).

A number of embodiments of the present invention have been described.
Nevertheless, it will be understood that various modifications may be made without departing from the spirit and scope of the invention. Accordingly, other embodiments are within the scope of the following claims.

~

Claims (62)

What is claimed is:
1. A method for preparing a reaction substrate for use as an assay device, comprising a step of parallel printing an array of biosites on said reaction substrate, wherein each biosite comprises a single type of capture probe bound to said reaction substrate, and wherein the array of biosites is deposited on the reaction substrate by a capillary bundle printer device, wherein the capillary bundle device comprises:

a plurality of bundled capillary tubes, each tube having a proximal end and a distal end;

an attachment site for holding the bundled capillary tubes at a point spaced from the distal ends of the capillary tubes;

an array template for slidably holding each capillary tube near its distal end, and for allowing the distal end of each capillary tube to move with respect to the attachment site;

at least one manifold for positioning the proximal end of each capillary tube within a corresponding supply chamber, wherein each supply chamber is capable of supplying a liquid reagent to at least one corresponding capillary tube; and a positioning device for precisely positioning the array template and the bundled capillary tubes with respect to the reaction substrate and depositing liquid reagents from the distal end of the capillary tubes onto the reaction substrates as biosites.
2. The method of claim 1, wherein the plurality of bundled capillary tubes comprises about 2 to about 10,000 tubes, having a center-to center spacing of about 80 µm to about 5 mm.
3. The method of claim 1, wherein each supply chamber supplies only one capillary tube.
4. The method of claim 1, wherein one supply chamber supplies more than one capillary tube of the plurality of bundled capillary tubes.
5. The method of claim 1, wherein the bundled capillary tubes comprise stainless steel, plastic, rubber, glass TEFLON.TM., flexible metal, or fused silica coated with polyimide.
6. The method of claim 1, wherein the device includes a plurality of array templates.
7. The method of claim 1, further comprising a positioning device for precisely positioning the reaction substrate.
8. The method of claim 1, wherein the capillaries have an inside diameter of about 10 µm to about 200 µm and an outside diameter of about 80 µm to about 500 µm.
9. The method of claim 1, wherein the array template comprises an array of sleeves, each sleeve having an inside diameter sufficient to permit a capillary to slide through, and a length sufficient to allow a precise pattern to be maintained while depositing fluids onto the reaction substrate.
10. The method of claim 1, wherein the array template comprises a plurality of holes formed in a rigid material.
11. The method of claim 1, wherein the array template comprises a rigidly formed or held mesh.
12. The method of claim 1, further comprising a housing for containing the supply chambers.
13. The method of claim 12, wherein the housing is capable of maintaining an inert atmosphere.
14. The method of claim 12, wherein the housing is capable of maintaining an elevated or reduced temperature.
15. The method of claim 12, wherein the housing may be pressurized to a predetermined pressure.
16. The method of claim 15, wherein the pressure is modulated to control the flow of liquid reagents through the capillary tubes.
17. The method of claim 1, wherein the supply chambers are positioned higher than the distal end of the capillary tubes to provide a pressure head.
18. The method of claim 1, wherein deposit of liquid reagent is controlled by a means for electrophoresis.
19. The method of claim 1, wherein supply of liquid reagent is controlled by a means for electro-osmosis.
20. The method of claim 1, wherein the device includes multiple sets of supply chambers, each for supplying liquid reagent to a subset of the bundled capillary tubes.
21. The method of claim 1, wherein the bundled capillary tubes are free to flex between the attachment site and the array template.
22. The method of claim 1, wherein the reaction substrate is pre-etched to define a pattern of reactive areas matching the geometry of the deposited biosites.
23. The method of claim 1, wherein the biosites are deposited simultaneously.
24. The method of claim 1, wherein each biosite is fluidically isolated from each other biosite.
25. The method of claim 1, wherein the array of biosites is deposited on the reaction substrate in a single print step by the capillary bundle printer device.
26. The method of claim 1, wherein the positioning device further comprises a Z-axis motion control.
27. The method of claim 1, wherein the reaction substrate is delivered to the capillary bundle by a robotic arm.
28. The method of claim 1, wherein the capillary bundles are affixed to a robotic arm.
29. The method of claim 1, wherein the reaction substrate comprises a nylon membrane, polypropylene, polystyrene, vinyl, plastic or glass.
30. The method of claim 1, wherein the array template comprises a metal grid, a mesh, a rigidly-held fabric mesh, a bundle of sleeve tubes having an inner diameter sufficient to admit the fluid delivery capillaries, or a solid block having holes or channels.
31. The method of claim 1, wherein each biosite comprises a spot of about 25 to about 200 µm in diameter.
32. The method of claim 1, wherein said reaction substrate is optically clear and has a thickness of about 50 to about 300 µm.
33. A method for preparing a reaction substrate for use as an assay device, comprising a step of parallel printing an array of biosites on said reaction substrate, wherein each biosite comprises a single type of capture probe bound to said reaction substrate, and wherein the array of biosites is deposited on the reaction substrate by a capillary bundle printer device, wherein the capillary bundle device comprises:

a plurality of bundled capillary tubes, each tube having a proximal end and a distal end;

an attachment site for holding the bundled capillary tubes at a point spaced from the distal ends of the capillary tubes;

a structure for positioning the proximal end of each capillary tube within a corresponding supply chamber, each supply chamber capable of supplying a liquid reagent to at least one corresponding capillary tube;

a print head for holding the plurality of bundled capillary tubes near their distal ends and for precisely positioning the distal ends of the capillary tubes with respect to the reaction substrate to deposit liquid reagents onto the reaction substrate as biosites.
34. The method of claim 33, where the print head includes an ink-jet deposition device.
35. The method of claim 33, wherein the liquid reagent comprises at lease one capture probe.
36. A device for preparing a reaction substrate having biosites for conducting multiplexed microassays to determine binding between a target analyte and a capture probe, including:

a plurality of capillary tubes, each tube having a proximal end and a distal end;

an attachment site for holding said capillary tubes at a point spaced from the distal ends of such capillary tubes;

an array template for slidably holding each capillary tube near its distal end, and for allowing the distal end of each capillary tube to move with respect to the attachment site;

at least one manifold for positioning the proximal end of each capillary tube within a corresponding supply chamber, wherein each supply chamber is capable of supplying a liquid reagent to at least one corresponding capillary tube; and a positioning device for precisely positioning the array template and said capillary tubes with respect to said reaction substrate and depositing liquid reagents from said distal end of said capillary tubes onto said reaction substrate as biosites.
37. The device of claim 36, wherein said plurality of capillary tubes comprises about 2 to about 10,000 tubes, having a center to center spacing of about 80 m to about 5 mm.
38. The device of claim 36, wherein each supply'chamber supplies only one capillary tube.,
39. The device of claim 36, wherein one supply chamber supplies more than one capillary tube of a plurality of capillary tubes.
40. The device of claim 36, wherein said capillary tubes comprise stainless steel, plastic, rubber, glass, or fused silica coated with polyimide.
41. The device of claim 36, wherein said device includes a plurality of array templates.
42. The device of claim 36, further comprising a positioning device for precisely positioning said reaction substrate.
43. The device of claim 36, wherein said capillaries have an inside diameter of about 10 to about 200 µm, and an outside diameter of about 80 to about 500 µm.
44. The device of claim 36, wherein said array template comprises an array of sleeves, each sleeve having an inside diameter sufficient to permit a capillary to slide through, and a length sufficient to allow a precise pattern to be maintained while depositing fluids onto said reaction substrate.
45. The device of claim 36, wherein said array template comprises a plurality of holes formed in a rigid material.
46. The device of claim 36, wherein said array template comprises a rigidly formed or held mesh.
47. The device of claim 36, further comprising a housing for containing said supply chambers.
48. The device of claim 47, wherein said housing is capable of maintaining an inert atmosphere.
49. The device of claim 47, wherein said housing is capable of maintaining an elevated or reduced temperature.
50. The device of claim 47, wherein said housing may be pressurized to a predetermined pressure.
51. The device of claim 50, wherein said pressure is modulated to control the flow of liquid reagents through said capillary tubes.
52. The device of claim 36, wherein said supply chambers are positioned higher than the distal end of said capillary tubes to provide a pressure head.
53. The device of claim 36, wherein deposit of liquid reagent is controlled by a means for electrophoresis.
54. The device of claim 36, wherein deposit of liquid reagent is controlled by a means for electro-osmosis.
55. The device of claim 36, wherein said device includes multiple sets of supply chambers, each for supplying liquid reagent to a subset of said capillary tubes.
56. The device of claim 36, wherein said capillary tubes are free to flex between said attachment site and said array template.
57. The device of claim 36, wherein the reaction substrate is pre-etched to define a pattern of reactive areas matching the geometry of the deposited biosites.
58. The device of claim 26, wherein said biosites are deposited simultaneously.
59. The device of claim 36, wherein each biosite is fluidically isolated from each other biosite.
60. The device of claim 36, wherein the liquid reagent comprises at least one capture probe.
61. An apparatus for preparing a reaction substrate for use as an assay device, including:
an attachment site for holding a plurality of flexible capillary tubes each flexible capillary tube having a distal end and a proximal end at a point spaced from the distal end of each said capillary tube;
a structure for positioning the proximal end of each said capillary tube within a corresponding supply chamber, each supply chamber capable of supplying a liquid reagent to at least one corresponding capillary tube;
a print head for holding said plurality of capillary tubes near their distal ends and for precisely positioning the distal ends of said capillary tubes with respect to said reaction substrate to deposit liquid reagents onto said reaction substrate as biosites.
62. The apparatus of claim 61, where the print head includes an ink-jet deposition device.
CA002389358A 1996-12-31 1997-12-31 Multiplexed molecular analysis apparatus and method Expired - Fee Related CA2389358C (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US3462796P 1996-12-31 1996-12-31
US60/034,627 1996-12-31
CA002276462A CA2276462C (en) 1996-12-31 1997-12-31 Multiplexed molecular analysis system apparatus and method

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
CA002276462A Division CA2276462C (en) 1996-12-31 1997-12-31 Multiplexed molecular analysis system apparatus and method

Publications (2)

Publication Number Publication Date
CA2389358A1 CA2389358A1 (en) 1998-07-09
CA2389358C true CA2389358C (en) 2008-07-15

Family

ID=21877589

Family Applications (2)

Application Number Title Priority Date Filing Date
CA002389358A Expired - Fee Related CA2389358C (en) 1996-12-31 1997-12-31 Multiplexed molecular analysis apparatus and method
CA002276462A Expired - Lifetime CA2276462C (en) 1996-12-31 1997-12-31 Multiplexed molecular analysis system apparatus and method

Family Applications After (1)

Application Number Title Priority Date Filing Date
CA002276462A Expired - Lifetime CA2276462C (en) 1996-12-31 1997-12-31 Multiplexed molecular analysis system apparatus and method

Country Status (6)

Country Link
US (7) US6083763A (en)
EP (3) EP2295988A2 (en)
JP (5) JP4663824B2 (en)
AU (1) AU6646398A (en)
CA (2) CA2389358C (en)
WO (1) WO1998029736A1 (en)

Families Citing this family (668)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6090555A (en) * 1997-12-11 2000-07-18 Affymetrix, Inc. Scanned image alignment systems and methods
US5631734A (en) 1994-02-10 1997-05-20 Affymetrix, Inc. Method and apparatus for detection of fluorescently labeled materials
US6741344B1 (en) * 1994-02-10 2004-05-25 Affymetrix, Inc. Method and apparatus for detection of fluorescently labeled materials
US6207369B1 (en) * 1995-03-10 2001-03-27 Meso Scale Technologies, Llc Multi-array, multi-specific electrochemiluminescence testing
US20020048809A1 (en) * 1997-06-16 2002-04-25 Lafferty William Micharl Capillary array-based sample screening
US6972183B1 (en) 1997-06-16 2005-12-06 Diversa Corporation Capillary array-based enzyme screening
US6794127B1 (en) 1997-06-16 2004-09-21 Diversa Corporation Capillary array-based sample screening
US20010041333A1 (en) * 1997-06-16 2001-11-15 Short Jay M. High throughput screening for a bioactivity or biomolecule
US6958245B2 (en) 1996-04-25 2005-10-25 Bioarray Solutions Ltd. Array cytometry
US7041510B2 (en) 1996-04-25 2006-05-09 Bioarray Solutions Ltd. System and method for programmable illumination pattern generation
US7144119B2 (en) * 1996-04-25 2006-12-05 Bioarray Solutions Ltd. System and method for programmable illumination pattern generation
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
US6136311A (en) 1996-05-06 2000-10-24 Cornell Research Foundation, Inc. Treatment and diagnosis of cancer
AU6646398A (en) * 1996-12-31 1998-07-31 Genometrix Incorporated Multiplexed molecular analysis apparatus and method
US7622294B2 (en) * 1997-03-14 2009-11-24 Trustees Of Tufts College Methods for detecting target analytes and enzymatic reactions
US20030027126A1 (en) 1997-03-14 2003-02-06 Walt David R. Methods for detecting target analytes and enzymatic reactions
US6327410B1 (en) 1997-03-14 2001-12-04 The Trustees Of Tufts College Target analyte sensors utilizing Microspheres
AU751956B2 (en) * 1997-03-20 2002-09-05 University Of Washington Solvent for biopolymer synthesis, solvent microdroplets and methods of use
US6391622B1 (en) * 1997-04-04 2002-05-21 Caliper Technologies Corp. Closed-loop biochemical analyzers
AUPO625497A0 (en) * 1997-04-16 1997-05-15 Macquarie Research Limited Analysis of molecules
DE69838067T2 (en) * 1997-05-23 2008-03-13 Bioarray Solutions Ltd. COLOR CODING AND SITU INQUIRY OF MATRIX-COUPLED CHEMICAL COMPOUNDS
US5876946A (en) * 1997-06-03 1999-03-02 Pharmacopeia, Inc. High-throughput assay
US20020015997A1 (en) * 1997-06-16 2002-02-07 Lafferty William Michael Capillary array-based sample screening
US20050070005A1 (en) * 1997-06-16 2005-03-31 Martin Keller High throughput or capillary-based screening for a bioactivity or biomolecule
US20030013115A1 (en) * 1997-06-16 2003-01-16 Diversa Corporation, A Delaware Corporation Capillary array-based sample screening
US20050118707A1 (en) * 1997-06-16 2005-06-02 Diversa Corp Positioning system for moving a selected station of a holding plate to a predetermined location for interaction with a probe
US20020102598A1 (en) * 1997-06-16 2002-08-01 Lafferty William Michael Positioning system for moving a selected station of a holding plate to a predetermined location for interaction with a probe
US20030092022A1 (en) * 1997-06-16 2003-05-15 Diversa Corporation, A Delaware Corporation High throughput screening for sequences of interest
US20040241759A1 (en) * 1997-06-16 2004-12-02 Eileen Tozer High throughput screening of libraries
JP4313861B2 (en) * 1997-08-01 2009-08-12 キヤノン株式会社 Manufacturing method of probe array
US6004925A (en) * 1997-09-29 1999-12-21 J. L. Dasseux Apolipoprotein A-I agonists and their use to treat dyslipidemic disorders
US7115884B1 (en) 1997-10-06 2006-10-03 Trustees Of Tufts College Self-encoding fiber optic sensor
US7348181B2 (en) * 1997-10-06 2008-03-25 Trustees Of Tufts College Self-encoding sensor with microspheres
US6485944B1 (en) 1997-10-10 2002-11-26 President And Fellows Of Harvard College Replica amplification of nucleic acid arrays
US6511803B1 (en) 1997-10-10 2003-01-28 President And Fellows Of Harvard College Replica amplification of nucleic acid arrays
US6232066B1 (en) 1997-12-19 2001-05-15 Neogen, Inc. High throughput assay system
US6238869B1 (en) 1997-12-19 2001-05-29 High Throughput Genomics, Inc. High throughput assay system
US20100105572A1 (en) * 1997-12-19 2010-04-29 Kris Richard M High throughput assay system
US20030039967A1 (en) * 1997-12-19 2003-02-27 Kris Richard M. High throughput assay system using mass spectrometry
US20030096232A1 (en) 1997-12-19 2003-05-22 Kris Richard M. High throughput assay system
US6458533B1 (en) 1997-12-19 2002-10-01 High Throughput Genomics, Inc. High throughput assay system for monitoring ESTs
US6893877B2 (en) 1998-01-12 2005-05-17 Massachusetts Institute Of Technology Methods for screening substances in a microwell array
US7794946B1 (en) 1998-02-04 2010-09-14 Life Technologies Corporation Microarray and uses therefor
CA2322202C (en) * 1998-03-10 2010-11-30 Large Scale Proteomics Corporation Detection and characterization of microorganisms
FR2776389B1 (en) * 1998-03-20 2000-06-16 Fondation Jean Dausset Ceph AUTOMATIC DEVICE FOR PRODUCING SAMPLES FOR THE IMPLEMENTATION OF CHEMICAL OR BIOLOGICAL REACTIONS IN A LIQUID MEDIUM
US6872537B1 (en) * 1998-04-14 2005-03-29 Regents Of The University Of California Assays for the detection of microtubule depolymerization inhibitors
US6699969B1 (en) * 1998-04-14 2004-03-02 The Regents Of The University Of California Assays for the detection of microtubule depolymerization inhibitors
DE69835342T2 (en) * 1998-04-27 2007-08-23 Corning Inc. Method for storing biological samples with the aid of a redrawn capillary store
US6350618B1 (en) 1998-04-27 2002-02-26 Corning Incorporated Redrawn capillary imaging reservoir
US6884626B1 (en) 1998-04-27 2005-04-26 Corning Incorporated Redrawn capillary imaging reservoir
EP1078245B1 (en) * 1998-05-16 2008-08-06 Applera Corporation Instrument for monitoring polymerase chain reaction of dna
US7498164B2 (en) 1998-05-16 2009-03-03 Applied Biosystems, Llc Instrument for monitoring nucleic acid sequence amplification reaction
US6818437B1 (en) * 1998-05-16 2004-11-16 Applera Corporation Instrument for monitoring polymerase chain reaction of DNA
DE19823660C1 (en) * 1998-05-20 1999-10-07 Nanomont Ges Fuer Nanotechnolo Process and assembly to apply and fix biochemically-active micro- and nano- micro-spheres on a substrate, especially useful in biochemical analysis such as DNA, viral and genetic testing
EP1090293B2 (en) 1998-06-24 2019-01-23 Illumina, Inc. Decoding of array sensors with microspheres
US6235473B1 (en) * 1998-07-02 2001-05-22 Orchid Biosciences, Inc. Gene pen devices for array printing
US6762061B1 (en) 1998-07-03 2004-07-13 Corning Incorporated Redrawn capillary imaging reservoir
US6576478B1 (en) * 1998-07-14 2003-06-10 Zyomyx, Inc. Microdevices for high-throughput screening of biomolecules
US6794197B1 (en) * 1998-07-14 2004-09-21 Zyomyx, Inc. Microdevice and method for detecting a characteristic of a fluid
EP1105529B2 (en) * 1998-07-30 2013-05-29 Illumina Cambridge Limited Arrayed biomolecules and their use in sequencing
US6107038A (en) * 1998-08-14 2000-08-22 Agilent Technologies Inc. Method of binding a plurality of chemicals on a substrate by electrophoretic self-assembly
EP1250955A1 (en) * 1998-09-08 2002-10-23 Tibotec N.V. Method for the rapid screening of analytes
US6461812B2 (en) * 1998-09-09 2002-10-08 Agilent Technologies, Inc. Method and multiple reservoir apparatus for fabrication of biomolecular arrays
US6309891B1 (en) 1998-09-09 2001-10-30 Incyte Genomics, Inc. Capillary printing systems
US6703228B1 (en) 1998-09-25 2004-03-09 Massachusetts Institute Of Technology Methods and products related to genotyping and DNA analysis
EP1001037A3 (en) * 1998-09-28 2003-10-01 Whitehead Institute For Biomedical Research Pre-selection and isolation of single nucleotide polymorphisms
EP1124939B1 (en) 1998-10-08 2003-04-23 AstraZeneca AB Microfabricated cell injector
US6591852B1 (en) 1998-10-13 2003-07-15 Biomicro Systems, Inc. Fluid circuit components based upon passive fluid dynamics
BR9914554A (en) 1998-10-13 2001-06-26 Biomicro Systems Inc Fluid circuit components based on passive fluid dynamics
US6637463B1 (en) 1998-10-13 2003-10-28 Biomicro Systems, Inc. Multi-channel microfluidic system design with balanced fluid flow distribution
US6601613B2 (en) 1998-10-13 2003-08-05 Biomicro Systems, Inc. Fluid circuit components based upon passive fluid dynamics
US7199809B1 (en) * 1998-10-19 2007-04-03 Symyx Technologies, Inc. Graphic design of combinatorial material libraries
US6426231B1 (en) * 1998-11-18 2002-07-30 The Texas A&M University System Analyte sensing mediated by adapter/carrier molecules
US6309828B1 (en) * 1998-11-18 2001-10-30 Agilent Technologies, Inc. Method and apparatus for fabricating replicate arrays of nucleic acid molecules
EP1002570A1 (en) * 1998-11-20 2000-05-24 Corning Incorporated Capillary transfer device for high density arrays
US20020006359A1 (en) * 1998-11-25 2002-01-17 Affymetrix, Inc. Microplate sample and reagent loading system
FR2786787B1 (en) * 1998-12-08 2002-04-19 Proteus METHOD OF IN VITRO ANALYSIS OF A KNOWN PHENOTYPE FROM A SAMPLE OF NUCLEIC ACIDS
US6333499B1 (en) * 1998-12-15 2001-12-25 Fuji Photo Film Co., Ltd. Method of detecting a scanning start point, scanner, method of reading out image information, and image information reader
US7510841B2 (en) 1998-12-28 2009-03-31 Illumina, Inc. Methods of making and using composite arrays for the detection of a plurality of target analytes
US6429027B1 (en) 1998-12-28 2002-08-06 Illumina, Inc. Composite arrays utilizing microspheres
FR2789401B1 (en) * 1999-02-08 2003-04-04 Cis Bio Int METHOD FOR MANUFACTURING LIGAND DIES ADDRESSED ON A MEDIUM
US6627157B1 (en) * 1999-03-04 2003-09-30 Ut-Battelle, Llc Dual manifold system and method for fluid transfer
GB9906477D0 (en) * 1999-03-19 1999-05-12 Pyrosequencing Ab Liquid dispensing apparatus
US6306578B1 (en) 1999-03-19 2001-10-23 Genencor International, Inc. Multi-through hole testing plate for high throughput screening
EP1230383A2 (en) * 1999-04-08 2002-08-14 Sir Mortimer B. Davis Jewish General Hospital Quantitative assay for expression of genes in microarray
US8114850B2 (en) 1999-04-08 2012-02-14 Advanced Cancer Therapeutics, Llc Antiproliferative activity of G-rich oligonucleotides and method of using same to bind to nucleolin
US7960540B2 (en) 1999-04-08 2011-06-14 Advanced Cancer Therapeutics, Llc Antiproliferative activity of G-rich oligonucleotides and method of using same to bind to nucleolin
EP1181304B1 (en) 1999-04-08 2007-10-10 Antisoma Research Limited Antiproliferative activity of g-righ oligonucleotides and method of using same to bind to nucleolin
US20030129654A1 (en) * 1999-04-15 2003-07-10 Ilya Ravkin Coded particles for multiplexed analysis of biological samples
US20030134330A1 (en) * 1999-04-15 2003-07-17 Ilya Ravkin Chemical-library composition and method
US20030166015A1 (en) * 1999-04-15 2003-09-04 Zarowitz Michael A. Multiplexed analysis of cell-substrate interactions
US20030207249A1 (en) * 1999-04-15 2003-11-06 Beske Oren E. Connection of cells to substrates using association pairs
US6908737B2 (en) * 1999-04-15 2005-06-21 Vitra Bioscience, Inc. Systems and methods of conducting multiplexed experiments
JP2002542463A (en) * 1999-04-15 2002-12-10 バーチャル・アレイズ・インコーポレーテッド Combinatorial chemical library with indices at code locations
US7253435B2 (en) * 1999-04-15 2007-08-07 Millipore Corporation Particles with light-polarizing codes
US6355431B1 (en) 1999-04-20 2002-03-12 Illumina, Inc. Detection of nucleic acid amplification reactions using bead arrays
US20050181440A1 (en) * 1999-04-20 2005-08-18 Illumina, Inc. Nucleic acid sequencing using microsphere arrays
US20060275782A1 (en) 1999-04-20 2006-12-07 Illumina, Inc. Detection of nucleic acid reactions on bead arrays
US7655443B1 (en) * 1999-05-07 2010-02-02 Siemens Healthcare Diagnostics, Inc. Nucleic acid sequencing with simultaneous quantitation
US6690399B1 (en) * 1999-05-07 2004-02-10 Tropix, Inc. Data display software for displaying assay results
WO2000068670A1 (en) * 1999-05-07 2000-11-16 Anslyn Eric V Method and system for remotely collecting and evaluating chemical/biochemical information
US20020182749A1 (en) * 1999-05-11 2002-12-05 Aclara Biosciences, Inc. Sample evaporative control
US7932213B2 (en) * 1999-05-11 2011-04-26 President And Fellows Of Harvard College Small molecule printing
US6824987B1 (en) * 1999-05-11 2004-11-30 President And Fellows Of Harvard College Small molecule printing
US6651008B1 (en) 1999-05-14 2003-11-18 Cytokinetics, Inc. Database system including computer code for predictive cellular bioinformatics
US6876760B1 (en) 2000-12-04 2005-04-05 Cytokinetics, Inc. Classifying cells based on information contained in cell images
US6743576B1 (en) * 1999-05-14 2004-06-01 Cytokinetics, Inc. Database system for predictive cellular bioinformatics
US7151847B2 (en) * 2001-02-20 2006-12-19 Cytokinetics, Inc. Image analysis of the golgi complex
US7410793B2 (en) 1999-05-17 2008-08-12 Applera Corporation Optical instrument including excitation source
US20050279949A1 (en) * 1999-05-17 2005-12-22 Applera Corporation Temperature control for light-emitting diode stabilization
US7423750B2 (en) * 2001-11-29 2008-09-09 Applera Corporation Configurations, systems, and methods for optical scanning with at least one first relative angular motion and at least one second angular motion or at least one linear motion
US6620584B1 (en) 1999-05-20 2003-09-16 Illumina Combinatorial decoding of random nucleic acid arrays
US6544732B1 (en) 1999-05-20 2003-04-08 Illumina, Inc. Encoding and decoding of array sensors utilizing nanocrystals
US6573369B2 (en) * 1999-05-21 2003-06-03 Bioforce Nanosciences, Inc. Method and apparatus for solid state molecular analysis
US20030186311A1 (en) * 1999-05-21 2003-10-02 Bioforce Nanosciences, Inc. Parallel analysis of molecular interactions
US8080380B2 (en) * 1999-05-21 2011-12-20 Illumina, Inc. Use of microfluidic systems in the detection of target analytes using microsphere arrays
US20030073250A1 (en) * 1999-05-21 2003-04-17 Eric Henderson Method and apparatus for solid state molecular analysis
US8481268B2 (en) 1999-05-21 2013-07-09 Illumina, Inc. Use of microfluidic systems in the detection of target analytes using microsphere arrays
US20020042081A1 (en) * 2000-10-10 2002-04-11 Eric Henderson Evaluating binding affinities by force stratification and force panning
JP3911909B2 (en) 1999-06-09 2007-05-09 株式会社日立製作所 DNA sample preparation method and DNA sample preparation apparatus
WO2001006253A2 (en) 1999-07-16 2001-01-25 Board Of Regents, The University Of Texas System Detection system based on an analyte reactive particle
EP1218545B1 (en) 1999-08-18 2012-01-25 Illumina, Inc. Methods for preparing oligonucleotide solutions
US6492118B1 (en) 1999-08-27 2002-12-10 Matrix Technologies Corporation Methods of immobilizing ligands on solid supports
JP3865107B2 (en) * 2000-05-26 2007-01-10 横河電機株式会社 Biochip manufacturing method and biochip manufacturing apparatus using the same
WO2001019678A1 (en) * 1999-09-14 2001-03-22 Pharmacopeia, Inc. Article comprising a multi-channel dispensing head
WO2001027327A2 (en) 1999-10-08 2001-04-19 Protogene Laboratories, Inc. Method and apparatus for performing large numbers of reactions using array assembly
WO2001030498A1 (en) * 1999-10-26 2001-05-03 Genometrix Genomics Incorporated A storage card for hosting a biological specimen
US6428957B1 (en) 1999-11-08 2002-08-06 Agilent Technologies, Inc. Systems tools and methods of assaying biological materials using spatially-addressable arrays
US6892141B1 (en) * 2000-03-17 2005-05-10 Hitachi, Ltd. Primer design system
US6878517B1 (en) * 1999-12-15 2005-04-12 Congra Grocery Products Company Multispecies food testing and characterization organoleptic properties
US7429466B2 (en) * 2000-01-24 2008-09-30 Hypromatrix, Inc Methods and arrays for detecting biological molecules
US8143195B2 (en) * 2000-01-24 2012-03-27 Yingjian Wang Arrays for bringing two or more reagents in contact with one or more biological targets and methods for making and using the arrays
US6399396B1 (en) * 2000-01-28 2002-06-04 Agilent Technologies, Inc. Compressed loading apparatus and method for liquid transfer
AU2001234709A1 (en) * 2000-01-31 2001-08-07 Board Of Regents, The University Of Texas System Method of preparing a sensor array
US6913884B2 (en) * 2001-08-16 2005-07-05 Illumina, Inc. Compositions and methods for repetitive use of genomic DNA
US7582420B2 (en) 2001-07-12 2009-09-01 Illumina, Inc. Multiplex nucleic acid reactions
CA2399733C (en) 2000-02-07 2011-09-20 Illumina, Inc. Nucleic acid detection methods using universal priming
US8076063B2 (en) 2000-02-07 2011-12-13 Illumina, Inc. Multiplexed methylation detection methods
US20050214825A1 (en) * 2000-02-07 2005-09-29 John Stuelpnagel Multiplex sample analysis on universal arrays
WO2001057268A2 (en) 2000-02-07 2001-08-09 Illumina, Inc. Nucleic acid detection methods using universal priming
US7955794B2 (en) 2000-09-21 2011-06-07 Illumina, Inc. Multiplex nucleic acid reactions
WO2001059151A2 (en) * 2000-02-10 2001-08-16 Tm Bioscience Corporation Method of designing and selecting polynucleotide sequences
US20020039728A1 (en) * 2000-02-10 2002-04-04 Robert Kain Alternative substrates and formats for bead-based array of arrays
US6770441B2 (en) * 2000-02-10 2004-08-03 Illumina, Inc. Array compositions and methods of making same
ATE412774T1 (en) 2000-02-16 2008-11-15 Illumina Inc PARALLEL GENOTYPING OF MULTIPLE PATIENT SAMPLES
US20020151040A1 (en) 2000-02-18 2002-10-17 Matthew O' Keefe Apparatus and methods for parallel processing of microvolume liquid reactions
CA2400644C (en) * 2000-02-18 2009-07-14 Board Of Trustees Of The Leland Stanford Junior University Apparatus and methods for parallel processing of micro-volume liquid reactions
US20040014102A1 (en) * 2000-02-22 2004-01-22 Shiping Chen High density parallel printing of microarrays
CA2399189A1 (en) * 2000-02-22 2001-08-30 Genospectra, Inc. Microarray fabrication techniques and apparatus
JP2003529056A (en) * 2000-02-22 2003-09-30 ジェノスペクトラ,インコーポレイティド Microarray fabrication technology and equipment
US20040009514A1 (en) * 2000-02-28 2004-01-15 Frutos Anthony G. Assembly for label-free detection of hybridized nucleic targets
WO2001064831A1 (en) * 2000-02-29 2001-09-07 The Board Of Trustees Of The Leland Stanford Junior University Microarray substrate with integrated photodetector and methods of use thereof
JP3960802B2 (en) * 2000-03-02 2007-08-15 マイクロチップス・インコーポレーテッド Microfabricated devices for storing and selectively exposing chemicals and devices
US6897015B2 (en) * 2000-03-07 2005-05-24 Bioforce Nanosciences, Inc. Device and method of use for detection and characterization of pathogens and biological materials
US6447723B1 (en) 2000-03-13 2002-09-10 Packard Instrument Company, Inc. Microarray spotting instruments incorporating sensors and methods of using sensors for improving performance of microarray spotting instruments
US6878554B1 (en) 2000-03-20 2005-04-12 Perkinelmer Las, Inc. Method and apparatus for automatic pin detection in microarray spotting instruments
US6756232B1 (en) * 2000-03-20 2004-06-29 Perkinelmer Las, Inc. Method and apparatus for producing compact microarrays
US7829313B2 (en) * 2000-03-24 2010-11-09 Eppendorf Array Technologies Identification and quantification of a plurality of biological (micro)organisms or their components
US7202026B2 (en) * 2000-03-24 2007-04-10 Eppendorf Array Technologies Sa (Eat) Identification of a large number of biological (micro)organisms groups at different levels by their detection on a same array
US7205104B2 (en) * 2000-03-24 2007-04-17 Eppendorf Array Technologies Sa (Eat) Identification of biological (micro) organisms by detection of their homologous nucleotide sequences on arrays
US20080085515A1 (en) * 2000-03-24 2008-04-10 Eppendorf Array Technologies Sa (Eat) Identification of multiple biological (micro) organisms by detection of their nucleotide sequences on arrays
US7875442B2 (en) * 2000-03-24 2011-01-25 Eppendorf Array Technologies Identification and quantification of a plurality of biological (micro)organisms or their components
DE10015448A1 (en) * 2000-03-29 2001-10-11 November Ag Molekulare Medizin Detection and quantification of binding between affinity molecules useful in screening for drugs, does not require removal of reaction solution prior to measurement
DE60131735T2 (en) * 2000-04-03 2008-11-06 Parabol Technologies S.A. DEVICE FOR DISTRIBUTING EXACTLY CONTROLLED SMALL FLUID QUANTITIES
DE10017791A1 (en) * 2000-04-10 2001-10-11 Basf Ag Method and device for microdosing small amounts of liquid for biopolymer arrays
US6432365B1 (en) * 2000-04-14 2002-08-13 Discovery Partners International, Inc. System and method for dispensing solution to a multi-well container
JP2001304816A (en) * 2000-04-26 2001-10-31 Kenichiro Kobayashi Travel measuring system and apparatus using granular dot pattern by laser reflected light
DE10020704B4 (en) * 2000-04-27 2006-09-28 Bioref Gmbh Biochip for archiving and laboratory medical analysis of biological sample material, process for its production and its use in diagnostic procedures
AU2001257454A1 (en) * 2000-04-28 2001-11-12 Quantum Dot Corporation Methods and compositions for polynucleotide analysis using generic capture sequences
AU2001259512B2 (en) * 2000-05-04 2007-03-01 Yale University High density protein arrays for screening of protein activity
CA2412302A1 (en) * 2000-05-11 2002-10-30 Matsushita Seiko Co., Ltd. Chemical sensor device
JP3532829B2 (en) * 2000-05-17 2004-05-31 日立ソフトウエアエンジニアリング株式会社 Biochip preparation solution and biochip preparation method
US6547943B1 (en) * 2000-05-25 2003-04-15 Spectrumedix Llc Capillary system providing multiple analysis of sample from same body of liquid
WO2001091902A2 (en) 2000-05-30 2001-12-06 Massachusetts Institute Of Technology Methods and devices for sealing microchip reservoir devices
US7396675B2 (en) 2000-06-02 2008-07-08 Bayer Technology Services Gmbh Kit and method for determining a plurality of analytes
US6994827B2 (en) * 2000-06-03 2006-02-07 Symyx Technologies, Inc. Parallel semicontinuous or continuous reactors
US7521245B1 (en) 2000-06-05 2009-04-21 Perkinelmer Las, Inc. Method for washing and drying pins in microarray spotting instruments
US6591196B1 (en) * 2000-06-06 2003-07-08 Agilent Technologies Inc. Method and system for extracting data from surface array deposited features
EP1164201A1 (en) * 2000-06-14 2001-12-19 Facultés Universitaires Notre-Dame de la Paix Reverse detection for identification and/or quantification of nucleotide target sequences on biochips
ATE319087T1 (en) * 2000-06-21 2006-03-15 Bioarray Solutions Ltd MULTI-ANALYTICAL MOLECULAR ANALYSIS BY USING APPLICATION-SPECIFIC RANDOM PARTICLE ARRAYS
US9709559B2 (en) 2000-06-21 2017-07-18 Bioarray Solutions, Ltd. Multianalyte molecular analysis using application-specific random particle arrays
US20020049544A1 (en) * 2000-06-23 2002-04-25 Cytokinetics, Inc. Image analysis for phenotyping sets of mutant cells
DE10036457A1 (en) * 2000-07-26 2002-02-14 Giesing Michael Use of an imaging photoelectric area sensor for evaluating biochips and imaging processes therefor
JP4833495B2 (en) * 2000-08-03 2011-12-07 アレイジェット リミテッド Highly parallel structure of microarray by inkjet printhead
GB0018963D0 (en) * 2000-08-03 2000-09-20 Manning Howard J Highly parallel fabrication of microarrays by ink jet printheads
DE10040857A1 (en) * 2000-08-11 2002-02-28 Jens P Fuerste Nucleic acid library or protein or peptide library
US7008769B2 (en) * 2000-08-15 2006-03-07 Bioforce Nanosciences, Inc. Nanoscale molecular arrayer
US6902702B1 (en) 2000-08-16 2005-06-07 University Health Network Devices and methods for producing microarrays of biological samples
US6789040B2 (en) 2000-08-22 2004-09-07 Affymetrix, Inc. System, method, and computer software product for specifying a scanning area of a substrate
US6640891B1 (en) * 2000-09-05 2003-11-04 Kevin R. Oldenburg Rapid thermal cycling device
US20020100582A1 (en) * 2000-09-05 2002-08-01 Oldenburg Kevin R. Rapid thermal cycling device
AU2002223529B2 (en) * 2000-09-07 2006-08-24 Tibotec Bvba Automated system for filling capillaries
GB0022069D0 (en) * 2000-09-08 2000-10-25 Pyrosequencing Ab Method
US20040185464A1 (en) * 2000-09-15 2004-09-23 Kris Richard M. High throughput assay system
US7057704B2 (en) * 2000-09-17 2006-06-06 Bioarray Solutions Ltd. System and method for programmable illumination pattern generation
US7731904B2 (en) * 2000-09-19 2010-06-08 Canon Kabushiki Kaisha Method for making probe support and apparatus used for the method
US7070740B1 (en) 2000-09-28 2006-07-04 Beckman Coulter, Inc. Method and apparatus for processing biomolecule arrays
US6911181B1 (en) * 2000-10-03 2005-06-28 Isis Pharmaceuticals, Inc. Self-dispensing storage device
WO2002030561A2 (en) * 2000-10-10 2002-04-18 Biotrove, Inc. Apparatus for assay, synthesis and storage, and methods of manufacture, use, and manipulation thereof
WO2002030264A2 (en) * 2000-10-10 2002-04-18 Microchips, Inc. Microchip reservoir devices using wireless transmission of power and data
US20100261159A1 (en) 2000-10-10 2010-10-14 Robert Hess Apparatus for assay, synthesis and storage, and methods of manufacture, use, and manipulation thereof
US6852291B1 (en) * 2000-10-11 2005-02-08 Innovadyne Technologies, Inc. Hybrid valve apparatus and method for fluid handling
US7135146B2 (en) * 2000-10-11 2006-11-14 Innovadyne Technologies, Inc. Universal non-contact dispense peripheral apparatus and method for a primary liquid handling device
US7456028B2 (en) * 2000-10-16 2008-11-25 Board Of Trustees Of The University Of Arkansas, N.A. Electrochemical method for detecting water born pathogens
US20030045005A1 (en) * 2000-10-17 2003-03-06 Michael Seul Light-controlled electrokinetic assembly of particles near surfaces
AU2002236691A1 (en) * 2000-10-18 2002-05-21 Virtual Arrays, Inc. Multiplexed cell analysis system
JP2004537712A (en) * 2000-10-18 2004-12-16 バーチャル・アレイズ・インコーポレーテッド Multiple cell analysis system
US6829376B2 (en) * 2000-10-24 2004-12-07 Affymetrix, Inc. Computer software system, method, and product for scanned image alignment
US20040018491A1 (en) * 2000-10-26 2004-01-29 Kevin Gunderson Detection of nucleic acid reactions on bead arrays
US6656740B1 (en) * 2000-10-31 2003-12-02 Agilent Technologies, Inc. Pressure variation in array fabrication
US7371563B2 (en) * 2000-11-08 2008-05-13 Surface Logix, Inc. Peelable and resealable devices for biochemical assays
US6803205B2 (en) * 2000-11-08 2004-10-12 Surface Logix, Inc. Methods of measuring enzyme activity using peelable and resealable devices
US6967074B2 (en) 2000-11-08 2005-11-22 Surface Logix, Inc. Methods of detecting immobilized biomolecules
US7351575B2 (en) * 2000-11-08 2008-04-01 Surface Logix, Inc. Methods for processing biological materials using peelable and resealable devices
US7439056B2 (en) 2000-11-08 2008-10-21 Surface Logix Inc. Peelable and resealable devices for arraying materials
US7001740B2 (en) * 2000-11-08 2006-02-21 Surface Logix, Inc. Methods of arraying biological materials using peelable and resealable devices
AU2002235128A1 (en) 2000-11-14 2002-05-27 Genetag Technology, Inc. Expression miniarrays and uses thereof
FR2816711B1 (en) * 2000-11-15 2003-03-07 Biomerieux Sa METHOD FOR DEPOSITING AN ANALYTE SOLUTION
GB2369086B (en) 2000-11-20 2005-06-08 Herfurth Laser Technology Ltd Reaction plate
JP3596868B2 (en) * 2000-11-24 2004-12-02 キヤノン株式会社 Method for evaluating the amount of probe in end-labeled probe array, and method for evaluating amount of target substance using labeled probe array
DE60141171D1 (en) 2000-11-27 2010-03-11 Intelligent Medical Devices Ll CLINICAL INTELLIGENT DIAGNOSTIC DEVICES AND METHOD
US6905816B2 (en) 2000-11-27 2005-06-14 Intelligent Medical Devices, Inc. Clinically intelligent diagnostic devices and methods
US7218764B2 (en) * 2000-12-04 2007-05-15 Cytokinetics, Inc. Ploidy classification method
US7776571B2 (en) * 2000-12-12 2010-08-17 Autogenomics, Inc. Multi-substrate biochip unit
US6599694B2 (en) * 2000-12-18 2003-07-29 Cytokinetics, Inc. Method of characterizing potential therapeutics by determining cell-cell interactions
EP1362126A4 (en) * 2001-01-18 2004-07-21 Kemmons A Tubbs An integrated high throughput system for the analysis of biomolecules
CA2437558A1 (en) * 2001-01-31 2002-08-08 The Board Of Regents Of The University Of Texas System Method and apparatus for the confinement of materials in a micromachined chemical sensor array
US7202392B2 (en) * 2001-01-31 2007-04-10 The General Hospital Corporation In vivo screening array
US7112305B2 (en) * 2001-01-31 2006-09-26 Agilent Technologies, Inc. Automation-optimized microarray package
US6635441B2 (en) * 2001-02-08 2003-10-21 Irm, Llc Multi-sample fermentor and method of using same
AU2002255581A1 (en) * 2001-02-16 2002-10-15 Genospectra, Inc. Bundled capillaries apparatus for high throughput screening
US6956961B2 (en) * 2001-02-20 2005-10-18 Cytokinetics, Inc. Extracting shape information contained in cell images
US7016787B2 (en) * 2001-02-20 2006-03-21 Cytokinetics, Inc. Characterizing biological stimuli by response curves
US20030190671A1 (en) * 2001-02-28 2003-10-09 Mcgill University Use of metabolic phenotyping in individualized treatment with amonafide
WO2002073180A1 (en) * 2001-03-08 2002-09-19 Ruo-Pan Huang An antibody-based protein array system
WO2002072867A2 (en) * 2001-03-08 2002-09-19 Chromavision Medical Systems, Inc. Apparatus and method for labeling rows and columns in an irregular array
WO2002072264A1 (en) * 2001-03-09 2002-09-19 Biomicro Systems, Inc. Method and system for microfluidic interfacing to arrays
AUPR378001A0 (en) * 2001-03-16 2001-04-12 Proteome Systems Ltd Protein chip
AU2002223010B2 (en) * 2001-03-16 2006-07-27 Shimadzu Corporation Array-based biomolecule analysis
US20050019942A1 (en) * 2001-03-16 2005-01-27 Proteome Systems Ltd. Array-based biomolecule analysis
US20020146815A1 (en) * 2001-03-28 2002-10-10 Hidenori Watanabe Manufacturing method and apparatus for probe carriers
EP1248108B1 (en) * 2001-03-29 2011-08-31 Canon Kabushiki Kaisha method of cellular screening and substrates suitable for it
AU2002307090A1 (en) * 2001-04-03 2002-10-21 Surromed, Inc. Methods and reagents for multiplexed analyte capture, surface array self-assembly, and analysis of complex biological samples
NL1017989C2 (en) * 2001-05-03 2002-11-05 Univ Delft Tech Method for performing an assay, device for that, as well as a method for manufacturing a device.
AU2002340641A1 (en) * 2001-05-03 2002-11-18 Sigma-Genosys, Ltd. Methods for assembling protein microarrays
US7338805B2 (en) 2001-05-04 2008-03-04 Bio Merieux Labeling reagents, methods for synthesizing such reagents and methods for detecting biological molecules
US20050009204A1 (en) * 2003-07-11 2005-01-13 Ye Fang Multiplexed binding assays for receptor arrays
DE10126630A1 (en) * 2001-05-31 2003-01-09 Peter Und Traudl Engelhorn Sti Cell sorting method
US6570158B2 (en) * 2001-06-02 2003-05-27 Hya Feygin Method and apparatus for infrared-spectrum imaging
US20020187478A1 (en) * 2001-06-07 2002-12-12 Childers Winthrop D. Rapid pharmaceutical component screening devices and methods
US20030124659A1 (en) * 2001-06-07 2003-07-03 Graves David James Multiplexing method
US6824974B2 (en) * 2001-06-11 2004-11-30 Genorx, Inc. Electronic detection of biological molecules using thin layers
US20040048241A1 (en) * 2001-06-11 2004-03-11 Freeman Beverly Annette Methods for attaching molecules
US20030077570A1 (en) * 2001-09-20 2003-04-24 Coventor, Inc. Small molecule substrate based enzyme activity assays
US20020197729A1 (en) * 2001-06-21 2002-12-26 Fuji Photo Film Co., Ltd. Biochemical analysis unit and method for manufacturing the same
US7981844B2 (en) * 2001-06-21 2011-07-19 Verenium Corporation Methods for the manufacture of pure single enantiomer compounds and for selecting enantioselective enzymes
US7262063B2 (en) 2001-06-21 2007-08-28 Bio Array Solutions, Ltd. Directed assembly of functional heterostructures
US6855538B2 (en) * 2001-06-27 2005-02-15 The Regents Of The University Of California High-efficiency microarray printing device
US7402286B2 (en) * 2001-06-27 2008-07-22 The Regents Of The University Of California Capillary pins for high-efficiency microarray printing device
US6993172B2 (en) * 2001-06-29 2006-01-31 Agilent Technologies, Inc. Method and system for automated outlying feature and outlying feature background detection during processing of data scanned from a molecular array
CN100465619C (en) 2001-06-29 2009-03-04 梅索磅秤技术有限公司 Assay plates, reader systems and methods for luminescence test measurements
EP1273346A1 (en) * 2001-07-05 2003-01-08 Seyonic SA Multi-channel fluid dispensing apparatus
FR2827199B1 (en) * 2001-07-10 2004-07-09 Centre Nat Rech Scient PROCESS AND MACHINE FOR THE EX SITU MANUFACTURE OF LOW AND MEDIUM INTEGRATION BIOPE NETWORKS
US20030134273A1 (en) * 2001-07-17 2003-07-17 Eric Henderson Combined molecular binding detection through force microscopy and mass spectrometry
DE60236732D1 (en) * 2001-07-30 2010-07-29 Meso Scale Technologies Llc ASSAY ELECTRODES WITH REAL-ESTABLISHED LIPID / PROTEIN LAYERS AND PROCESS FOR THEIR PREPARATION AND USE
US7008788B2 (en) * 2001-07-30 2006-03-07 Agilent Technologies, Inc. Containers for supports comprising biopolymers
US20030032198A1 (en) * 2001-08-13 2003-02-13 Symyx Technologies, Inc. High throughput dispensing of fluids
DE10141650C1 (en) * 2001-08-24 2002-11-28 Lohmann Therapie Syst Lts Safe transdermal therapeutic system for administration of fentanyl or analogous analgesics, having matrix layer of carboxy group-free polyacrylate adhesive providing high permeation rate
US6682702B2 (en) * 2001-08-24 2004-01-27 Agilent Technologies, Inc. Apparatus and method for simultaneously conducting multiple chemical reactions
US20030087309A1 (en) * 2001-08-27 2003-05-08 Shiping Chen Desktop drug screening system
US20030045002A1 (en) * 2001-08-28 2003-03-06 Fuji Photo Film Co., Ltd. Cartridge for biochemical analysis unit and method for recording biochemical analysis data in biochemical analysis unit
US20030044320A1 (en) * 2001-08-31 2003-03-06 Shun Luo High throughput screening micro array platform
US6929944B2 (en) * 2001-08-31 2005-08-16 Beckman Coulter, Inc. Analysis using a distributed sample
US20040071328A1 (en) * 2001-09-07 2004-04-15 Vaisberg Eugeni A. Classifying cells based on information contained in cell images
EP1425090A2 (en) * 2001-09-07 2004-06-09 Corning Incorporated MICROCOLUMN&minus;PLATFORM BASED ARRAY FOR HIGH&minus;THROUGHPUT ANALYSIS
WO2003023410A1 (en) * 2001-09-07 2003-03-20 Innovadyne Technologies, Inc. Secondary liquid dispensing module for liquid handling system
US6902894B2 (en) * 2001-09-07 2005-06-07 Genetel Pharmaceuticals Ltd. Mutation detection on RNA polmerase beta subunit gene having rifampin resistance
EP1438582A1 (en) * 2001-09-16 2004-07-21 ChemoMetec A/S Method and a system for detecting and optionally isolating a rare event particle
US7042488B2 (en) 2001-09-27 2006-05-09 Fujinon Corporation Electronic endoscope for highlighting blood vessel
AU2002334887A1 (en) * 2001-10-05 2003-04-22 Vanderbilt University Capillary tube printing tips for microarray printing
WO2003034029A2 (en) 2001-10-15 2003-04-24 Bioarray Solutions, Ltd. Multiplexed analysis of polymorphic loci by concurrent interrogation and enzyme-mediated detection
US20030219800A1 (en) * 2001-10-18 2003-11-27 Beske Oren E. Multiplexed cell transfection using coded carriers
US6765490B2 (en) 2001-10-23 2004-07-20 Steven W. Lopez System and methods for detecting harmful agents within contents of mail
US6867005B2 (en) * 2001-10-24 2005-03-15 Beckman Coulter, Inc. Method and apparatus for increasing the dynamic range and accuracy of binding assays
US7584240B2 (en) 2001-11-07 2009-09-01 Genvault Corporation Automated biological sample archive for storage, retrieval and analysis of large numbers of samples for remote clients
US6764651B2 (en) * 2001-11-07 2004-07-20 Varian, Inc. Fiber-optic dissolution systems, devices, and methods
US20030087425A1 (en) * 2001-11-07 2003-05-08 Eggers Mitchell D Sample carrier
AU2002360361A1 (en) * 2001-11-09 2003-06-10 Biomicroarrays, Inc. High surface area substrates for microarrays and methods to make same
US20050003360A1 (en) * 2001-11-13 2005-01-06 Ruo-Pang Huang Array systems and methods
WO2003042697A1 (en) * 2001-11-14 2003-05-22 Genospectra, Inc. Biochemical analysis system with combinatorial chemistry applications
US6815178B1 (en) * 2001-11-19 2004-11-09 Antony R. Shoaf Endospore detection method
US6797393B2 (en) 2001-11-30 2004-09-28 Eastman Kodak Company Method for making biochip substrate
FR2832941B1 (en) * 2001-11-30 2004-09-24 Gemplus Card Int CLEANING MATERIAL JET HEADS
US20050221283A1 (en) * 2001-12-11 2005-10-06 Mahant Vijay K Biochip
EP1770429A3 (en) * 2001-12-11 2007-04-11 Autogenomics, Inc. Multi-substrate biochip unit
US7223538B2 (en) 2001-12-14 2007-05-29 Ge Healthcare Bio-Sciences Ab Post-synthesis labeling of nucleic acids, assays using nucleic acids that are labeled post-synthetically, single nucleotide polymorphism detection, and associated compounds and microarrays
US6756223B2 (en) * 2001-12-18 2004-06-29 Motorola, Inc. Electro-chemical analysis device with integrated thermal sensor and method for monitoring a sample using the device
CN1281324C (en) * 2001-12-19 2006-10-25 阿菲梅特里克斯公司 Manufacturing process for array plate assembly
WO2003060070A2 (en) * 2001-12-21 2003-07-24 Applied Research Systems Ars Holding N.V. Method of identifying novel proteins
US7282183B2 (en) * 2001-12-24 2007-10-16 Agilent Technologies, Inc. Atmospheric control in reaction chambers
US6846454B2 (en) * 2001-12-24 2005-01-25 Agilent Technologies, Inc. Fluid exit in reaction chambers
JP2003189869A (en) * 2001-12-27 2003-07-08 Nisshinbo Ind Inc Method for judging biological species contained in specimen and kit to be used for the method
US7335153B2 (en) * 2001-12-28 2008-02-26 Bio Array Solutions Ltd. Arrays of microparticles and methods of preparation thereof
EP1323474A1 (en) * 2001-12-31 2003-07-02 Corning Incorporated Device and process for simultaneous transfer of liquids
US7373968B2 (en) * 2002-01-08 2008-05-20 Kevin R. Oldenburg Method and apparatus for manipulating an organic liquid sample
US7614444B2 (en) 2002-01-08 2009-11-10 Oldenburg Kevin R Rapid thermal cycling device
DE10201463B4 (en) * 2002-01-16 2005-07-21 Clondiag Chip Technologies Gmbh Reaction vessel for performing array method
ATE377763T1 (en) * 2002-01-25 2007-11-15 Innovadyne Technologies Inc CONTACTLESS METHOD FOR DISTRIBUTING SMALL LIQUID VOLUME
JP3721334B2 (en) * 2002-02-01 2005-11-30 富士写真フイルム株式会社 Receptor-ligand association reaction method and reactor used therefor
JP3836379B2 (en) * 2002-02-01 2006-10-25 富士写真フイルム株式会社 Receptor-ligand association reaction method
JP3818926B2 (en) * 2002-02-04 2006-09-06 富士写真フイルム株式会社 Receptor-ligand association reaction method
KR100718397B1 (en) * 2002-02-13 2007-05-14 피피지 인더스트리즈 오하이오 인코포레이티드 Coating line and process for forming a multilayer composite coating on a substrate
WO2003069333A1 (en) 2002-02-14 2003-08-21 Illumina, Inc. Automated information processing in randomly ordered arrays
US7504364B2 (en) * 2002-03-01 2009-03-17 Receptors Llc Methods of making arrays and artificial receptors
US20030165994A1 (en) * 2002-03-04 2003-09-04 Kozmin Sergey A. Device and method for multiparallel synthesis and screening
US6815078B2 (en) 2002-03-06 2004-11-09 Eastman Kodak Company Substrate for protein microarray containing functionalized polymer
AU2003218727A1 (en) * 2002-03-11 2003-09-22 Axaron Bioscience Ag Method for applying a substance to a substrate
DE10211818B4 (en) * 2002-03-16 2006-07-06 peS Gesellschaft für medizinische Diagnose-Systeme mbH Method for the quantitative determination of several analytes
US7179654B2 (en) * 2002-03-18 2007-02-20 Agilent Technologies, Inc. Biochemical assay with programmable array detection
CA2480990A1 (en) * 2002-04-11 2003-10-23 Sequenom, Inc. Methods and devices for performing chemical reactions on a solid support
US20060013736A1 (en) * 2002-04-19 2006-01-19 Blok Herman J System, substrate plate and incubation device for conducting bioassays
US20030198967A1 (en) * 2002-04-23 2003-10-23 Matson Robert S. Multi-functional microarrays and methods
AU2003228711C1 (en) 2002-04-26 2010-01-07 Board Of Regents, The University Of Texas System Method and system for the detection of cardiac risk factors
AU2003225281A1 (en) 2002-04-30 2003-11-17 University Of South Florida Materials and methods for prevention and treatment of rna viral diseases
US20030207099A1 (en) * 2002-05-01 2003-11-06 Gillmor Susan D. Low-contact-angle polymer membranes and method for fabricating micro-bioarrays
EP1506403A2 (en) * 2002-05-13 2005-02-16 Zeptosens AG Kit zur assay-entwicklung und für serien-analysen
US6841379B2 (en) * 2002-05-15 2005-01-11 Beckman Coulter, Inc. Conductive microplate
DE10221564A1 (en) * 2002-05-15 2003-11-27 Evotec Ag Photoluminescence analyzer for chemical and biological sample screening uses a high intensity homogeneous line image as illumination source, and has multiple sensor scanning of the image
JP2005526253A (en) * 2002-05-17 2005-09-02 アプレラ コーポレイション Apparatus and method for differentiating multiple fluorescent signals by excitation wavelength
WO2003100382A1 (en) * 2002-05-20 2003-12-04 Northrop Grumman Corporation Automatic point source biological agent detection system
US20040126773A1 (en) * 2002-05-23 2004-07-01 Beske Oren E. Assays with coded sensor particles to sense assay conditions
US20030219755A1 (en) * 2002-05-24 2003-11-27 Nanibhushan Dattagupta Compositions and methods for performing hybridization assays using target enhanced signal amplification (TESA)
WO2003100012A2 (en) * 2002-05-24 2003-12-04 Nimblegen Systems, Inc. Microarrays and method for running hybridization reaction for multiple samples on a single microarray
JP4409426B2 (en) * 2002-05-28 2010-02-03 オウトジエノミクス・インコーポレーテツド Microarray detector and method
EP1508029A4 (en) * 2002-05-28 2010-04-14 Autogenomics Inc Multi-reagent pack
US20050239193A1 (en) * 2002-05-30 2005-10-27 Bioforce Nanosciences, Inc. Device and method of use for detection and characterization of microorganisms and microparticles
US6789965B2 (en) * 2002-05-31 2004-09-14 Agilent Technologies, Inc. Dot printer with off-axis loading
AU2003249681A1 (en) * 2002-05-31 2003-12-19 Diversa Corporation Multiplexed systems for nucleic acid sequencing
US20030232384A1 (en) * 2002-06-13 2003-12-18 Eastman Kodak Company Microarray system utilizing microtiter plates
EP2107120A1 (en) * 2002-07-24 2009-10-07 Board of Regents, The University of Texas System Capture and detection of microbes by membrane methods
US7452712B2 (en) 2002-07-30 2008-11-18 Applied Biosystems Inc. Sample block apparatus and method of maintaining a microcard on a sample block
US8277753B2 (en) 2002-08-23 2012-10-02 Life Technologies Corporation Microfluidic transfer pin
JP3883934B2 (en) * 2002-08-29 2007-02-21 富士フイルムホールディングス株式会社 Chemiluminescence method using biochemical analysis unit
AU2003258683A1 (en) * 2002-09-03 2004-03-29 Zeptosens Ag Analytical platform and detection method with analytes which are to be detected in a sample in the form of immobilized specific binding partners
US7595303B1 (en) 2002-09-05 2009-09-29 University Of South Florida Genetic adjuvants for immunotherapy
US20050037428A1 (en) * 2002-09-16 2005-02-17 Receptors Llc Artificial receptors including reversibly immobilized building blocks, the building blocks, and methods
US20060051802A1 (en) * 2002-09-16 2006-03-09 Receptors Llc Artificial receptors, building blocks, and methods
US20060057625A1 (en) * 2002-09-16 2006-03-16 Carlson Robert E Scaffold-based artificial receptors and methods
US20050170385A1 (en) * 2002-09-16 2005-08-04 Receptors Llc Artificial receptors including gradients
US20040137481A1 (en) * 2002-09-16 2004-07-15 Receptors Llc Artificial receptor building blocks, components, and kits
US7595883B1 (en) 2002-09-16 2009-09-29 The Board Of Trustees Of The Leland Stanford Junior University Biological analysis arrangement and approach therefor
US7469076B2 (en) * 2003-09-03 2008-12-23 Receptors Llc Sensors employing combinatorial artificial receptors
US20050037381A1 (en) * 2002-09-16 2005-02-17 Receptors Llc Artificial receptors, building blocks, and methods
US20050136483A1 (en) * 2003-09-03 2005-06-23 Receptors Llc Nanodevices employing combinatorial artificial receptors
US20050037429A1 (en) * 2003-03-28 2005-02-17 Receptors Llc Artificial receptors including reversibly immobilized building blocks and methods
US7534241B2 (en) * 2002-09-23 2009-05-19 Microchips, Inc. Micro-reservoir osmotic release systems and microtube array device
US7482170B2 (en) * 2002-09-30 2009-01-27 Roche Nimblegen, Inc. Parallel loading of arrays
US9701754B1 (en) 2002-10-23 2017-07-11 City Of Hope Covalent disulfide-linked diabodies and uses thereof
US20040081969A1 (en) * 2002-10-29 2004-04-29 Ilsley Diane D. Devices and methods for evaulating the quality of a sample for use in an array assay
DE10251668A1 (en) * 2002-11-06 2004-05-27 Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung e.V. Sample holder unit, for cryogenic storage under very low temperatures, comprises a data store to be scanned for identification of the samples held in the suspended sample chambers
US20030153013A1 (en) * 2002-11-07 2003-08-14 Ruo-Pan Huang Antibody-based protein array system
US20040095615A1 (en) * 2002-11-14 2004-05-20 Chung-Hua Tsai Biological scanner with two dimensional scanning operation
US7526114B2 (en) 2002-11-15 2009-04-28 Bioarray Solutions Ltd. Analysis, secure access to, and transmission of array images
US7718442B2 (en) 2002-11-22 2010-05-18 Genvault Corporation Sealed sample storage element system and method
IL159113A (en) * 2002-11-27 2008-11-26 Given Imaging Ltd Method and device of imaging with an imager having a fiber plate cover
US20080045788A1 (en) * 2002-11-27 2008-02-21 Zvika Gilad Method and device of imaging with an in vivo imager
US20060094108A1 (en) * 2002-12-20 2006-05-04 Karl Yoder Thermal cycler for microfluidic array assays
JP4395133B2 (en) * 2002-12-20 2010-01-06 カリパー・ライフ・サイエンシズ・インク. Single molecule amplification and detection of DNA
WO2004074818A2 (en) 2002-12-20 2004-09-02 Biotrove, Inc. Assay apparatus and method using microfluidic arrays
AU2003290429A1 (en) * 2002-12-25 2004-07-22 Casio Computer Co., Ltd. Optical dna sensor, dna reading apparatus, identification method of dna and manufacturing method of optical dna sensor
EP1583845A4 (en) * 2003-01-02 2006-06-28 Bioforce Nanosciences Inc Method and apparatus for molecular analysis in small sample volumes
KR100537504B1 (en) * 2003-01-20 2005-12-19 삼성전자주식회사 An automated fluidic system for protein detection in biological samples
CA2513985C (en) 2003-01-21 2012-05-29 Illumina Inc. Chemical reaction monitor
US6966336B1 (en) 2003-01-24 2005-11-22 Sandia National Laboratories Fluid injection microvalve
US6832787B1 (en) 2003-01-24 2004-12-21 Sandia National Laboratories Edge compression manifold apparatus
US7311882B1 (en) 2003-01-24 2007-12-25 Sandia National Laboratories Capillary interconnect device
US6918573B1 (en) 2003-01-27 2005-07-19 Sandia National Laboratories Microvalve
JP2004290171A (en) * 2003-02-07 2004-10-21 Akira Hiraishi Molecular biological identification technique of microorganism
US6970240B2 (en) 2003-03-10 2005-11-29 Applera Corporation Combination reader
EP1613737A4 (en) * 2003-03-28 2008-12-03 Receptors Llc Artificial receptors including reversibly immobilized building blocks and methods
US6926313B1 (en) 2003-04-02 2005-08-09 Sandia National Laboratories High pressure capillary connector
US7553455B1 (en) 2003-04-02 2009-06-30 Sandia Corporation Micromanifold assembly
US7161334B1 (en) 2003-04-16 2007-01-09 Sandia National Laboratories Modular high voltage power supply for chemical analysis
EP1477814A1 (en) * 2003-05-13 2004-11-17 The Automation Partnership (Cambridge) Limited Fluid dispenser
CA2524194C (en) * 2003-05-13 2015-07-21 Gen-Probe Incorporated Method and kit for identifying antibiotic-resistant microorganisms
US9317922B2 (en) 2003-05-16 2016-04-19 Board Of Regents The University Of Texas System Image and part recognition technology
US7651850B2 (en) 2003-05-16 2010-01-26 Board Of Regents, The University Of Texas System Image and part recognition technology
CN1280428C (en) * 2003-05-19 2006-10-18 清华大学 Biochip system based on minute particle and its application
US20060147962A1 (en) * 2003-06-16 2006-07-06 Mars, Inc. Genotype test
US8409528B2 (en) * 2003-06-19 2013-04-02 Abbott Laboratories Apparatus and method for handling fluids for analysis
US7452678B2 (en) * 2003-06-24 2008-11-18 Bristol-Myers Squibb Company Identification of biomarkers for liver toxicity
US20050014217A1 (en) * 2003-07-18 2005-01-20 Cytokinetics, Inc. Predicting hepatotoxicity using cell based assays
US7235353B2 (en) * 2003-07-18 2007-06-26 Cytokinetics, Inc. Predicting hepatotoxicity using cell based assays
GB2423988A (en) * 2003-07-18 2006-09-13 Cytokinetics Inc Characterizing biological stimuli by response curves
US20050055014A1 (en) * 2003-08-04 2005-03-10 Coppeta Jonathan R. Methods for accelerated release of material from a reservoir device
US20120122103A1 (en) 2003-09-11 2012-05-17 Rangarajan Sampath Compositions for use in identification of bacteria
US7235163B2 (en) * 2003-09-12 2007-06-26 Applera Corporation Loading features for channel array
US7488451B2 (en) * 2003-09-15 2009-02-10 Millipore Corporation Systems for particle manipulation
US20050084914A1 (en) * 2003-09-15 2005-04-21 Foulkes J. G. Assays with primary cells
US7927796B2 (en) 2003-09-18 2011-04-19 Bioarray Solutions, Ltd. Number coding for identification of subtypes of coded types of solid phase carriers
US20050221358A1 (en) * 2003-09-19 2005-10-06 Carrillo Albert L Pressure chamber clamp mechanism
US20070015289A1 (en) * 2003-09-19 2007-01-18 Kao H P Dispenser array spotting
US20050226779A1 (en) * 2003-09-19 2005-10-13 Oldham Mark F Vacuum assist for a microplate
US20050226771A1 (en) * 2003-09-19 2005-10-13 Lehto Dennis A High speed microplate transfer
WO2005028629A2 (en) * 2003-09-19 2005-03-31 Applera Corporation Whole genome expression analysis system
US20050233472A1 (en) * 2003-09-19 2005-10-20 Kao H P Spotting high density plate using a banded format
TW200521436A (en) 2003-09-22 2005-07-01 Bioarray Solutions Ltd Surface immobilized polyelectrolyte with multiple functional groups capable of covalently bonding to biomolecules
US20060008815A1 (en) * 2003-10-24 2006-01-12 Metamorphix, Inc. Compositions, methods, and systems for inferring canine breeds for genetic traits and verifying parentage of canine animals
WO2005042763A2 (en) 2003-10-28 2005-05-12 Bioarray Solutions Ltd. Optimization of gene expression analysis using immobilized capture probes
JP2007509629A (en) 2003-10-29 2007-04-19 バイオアレイ ソリューションズ リミテッド Complex nucleic acid analysis by cleavage of double-stranded DNA
US7981362B2 (en) 2003-11-04 2011-07-19 Meso Scale Technologies, Llc Modular assay plates, reader systems and methods for test measurements
US8080097B2 (en) 2003-11-06 2011-12-20 Hewlett-Packard Development Company, L.P. System and a method for the creation of edible, optically invisible images
EP1687406B1 (en) * 2003-11-10 2010-01-27 Geneohm Sciences, Inc. Nucleic acid detection method having increased sensitivity
EP1695082A2 (en) * 2003-12-11 2006-08-30 Board of Regents, The University of Texas System Method and system for the analysis of saliva using a sensor array
US7341834B2 (en) * 2003-12-15 2008-03-11 Geneohn Sciences, Inc. Multiplexed electrochemical detection system and method
US20050136538A1 (en) * 2003-12-17 2005-06-23 Srikant Pathak Lithographic method for attaching biological cells to a solid substrate using a small molecule linker
US7729863B2 (en) * 2003-12-17 2010-06-01 Fred Hutchinson Cancer Research Center Methods and materials for canine breed identification
ATE520983T1 (en) 2003-12-24 2011-09-15 Shinichiro Isobe METHOD FOR DETECTING A BIOMOLECULE, MARKING DYE AND MARKING KIT USED THEREFOR
US7781226B2 (en) * 2004-02-27 2010-08-24 The Board Of Regents Of The University Of Texas System Particle on membrane assay system
US20060257854A1 (en) * 2004-02-27 2006-11-16 Mcdevitt John T Membrane assay system including preloaded particles
US20060257991A1 (en) * 2004-02-27 2006-11-16 Mcdevitt John T Integration of fluids and reagents into self-contained cartridges containing particle-based sensor elements and membrane-based sensor elements
US8105849B2 (en) * 2004-02-27 2012-01-31 Board Of Regents, The University Of Texas System Integration of fluids and reagents into self-contained cartridges containing sensor elements
US20060257941A1 (en) * 2004-02-27 2006-11-16 Mcdevitt John T Integration of fluids and reagents into self-contained cartridges containing particle and membrane sensor elements
US8101431B2 (en) 2004-02-27 2012-01-24 Board Of Regents, The University Of Texas System Integration of fluids and reagents into self-contained cartridges containing sensor elements and reagent delivery systems
AU2005222618A1 (en) 2004-03-12 2005-09-29 Biotrove, Inc. Nanoliter array loading
US20050214737A1 (en) * 2004-03-26 2005-09-29 Dejneka Matthew J Transparent filtered capillaries
FR2868071B1 (en) * 2004-03-26 2006-06-09 Biomerieux Sa MARKING REAGENTS, METHODS FOR SYNTHESIZING SUCH REAGENTS AND METHODS FOR DETECTING BIOLOGICAL MOLECULES
KR101147147B1 (en) 2004-04-01 2012-05-25 머크 샤프 앤드 돔 코포레이션 Modified polynucleotides for reducing off-target effects in rna interference
US20050273271A1 (en) * 2004-04-05 2005-12-08 Aibing Rao Method of characterizing cell shape
JP4170947B2 (en) * 2004-04-09 2008-10-22 株式会社日立ハイテクノロジーズ Biological sample component detection method and apparatus
US20050238542A1 (en) * 2004-04-22 2005-10-27 Applera Corporation Pins for spotting nucleic acids
US20050266093A1 (en) * 2004-04-27 2005-12-01 Mohapatra Shyam S Nanogene therapy for cell proliferation disorders
WO2005115630A2 (en) * 2004-04-30 2005-12-08 Bioforce Nanosciences, Inc. Method and apparatus for depositing material onto a surface
US7364898B2 (en) * 2004-05-04 2008-04-29 Eppendorf Ag Customized micro-array construction and its use for target molecule detection
GB2414298B (en) * 2004-05-18 2008-09-10 Michael Strachan Walker Diagnostic method and apparatus
ATE441669T1 (en) 2004-05-24 2009-09-15 Genvault Corp STABLE STORAGE OF PROTEIN AND STABLE STORAGE OF NUCLEIC ACID IN RECOVERABLE FORM
JP2008501037A (en) * 2004-06-01 2008-01-17 マイクロチップス・インコーポレーテッド Devices and methods for measuring and enhancing transport of drugs or analytes to / from medical implants
US7338763B2 (en) * 2004-06-02 2008-03-04 Eppendorf Array Technologies S.A. Method and kit for the detection and/or quantification of homologous nucleotide sequences on arrays
US7323318B2 (en) * 2004-07-15 2008-01-29 Cytokinetics, Inc. Assay for distinguishing live and dead cells
US20070031818A1 (en) * 2004-07-15 2007-02-08 Cytokinetics, Inc., A Delaware Corporation Assay for distinguishing live and dead cells
US7537590B2 (en) * 2004-07-30 2009-05-26 Microchips, Inc. Multi-reservoir device for transdermal drug delivery and sensing
US7848889B2 (en) 2004-08-02 2010-12-07 Bioarray Solutions, Ltd. Automated analysis of multiplexed probe-target interaction patterns: pattern matching and allele identification
US20060024204A1 (en) * 2004-08-02 2006-02-02 Oldenburg Kevin R Well plate sealing apparatus and method
US20060105453A1 (en) 2004-09-09 2006-05-18 Brenan Colin J Coating process for microfluidic sample arrays
EP1623996A1 (en) * 2004-08-06 2006-02-08 Deutsches Krebsforschungszentrum Stiftung des öffentlichen Rechts Improved method of selecting a desired protein from a library
ATE424928T1 (en) * 2004-09-01 2009-03-15 Microchips Inc CONTAINER DEVICES WITH MULTIPLE CLOSURES FOR CONTROLLED DISPENSING OR EXPOSURE OF CONTAINERS CONTENTS
JP4576557B2 (en) * 2004-09-01 2010-11-10 カシオ計算機株式会社 Biopolymer analysis chip and analysis support device
WO2006028930A2 (en) * 2004-09-03 2006-03-16 Receptors Llc Combinatorial artificial receptors including tether building blocks on scaffolds
EP1789792A2 (en) 2004-09-11 2007-05-30 Receptors LLC Combinatorial artificial receptors including peptide building blocks
US7825972B2 (en) * 2004-10-08 2010-11-02 Cooper Allan J Processing method device and system to produce a focused image signal from an unfocused image
TWI353341B (en) * 2004-10-14 2011-12-01 Ibm Programmable molecular manipulating processes
US7208730B2 (en) * 2004-10-14 2007-04-24 International Business Machines Corporation Programmable molecular manipulating devices
US7733298B2 (en) * 2004-10-19 2010-06-08 Hewlett-Packard Development Company, L.P. Display device
FI118192B (en) * 2004-11-03 2007-08-15 Medicel Oy reactor device
ATE390562T1 (en) * 2004-11-19 2008-04-15 Ebm Papst St Georgen Gmbh & Co ARRANGEMENT WITH A FAN AND A PUMP
US7935811B2 (en) 2004-11-22 2011-05-03 Dharmacon, Inc. Apparatus and system having dry gene silencing compositions
US7923207B2 (en) * 2004-11-22 2011-04-12 Dharmacon, Inc. Apparatus and system having dry gene silencing pools
US20060166234A1 (en) 2004-11-22 2006-07-27 Barbara Robertson Apparatus and system having dry control gene silencing compositions
US20090042280A1 (en) * 2004-12-13 2009-02-12 Geneohm Sciences, Inc., Fluidic cartridges for electrochemical detection of dna
JP2006177837A (en) * 2004-12-24 2006-07-06 Hitachi Ltd Luminescence detection apparatus
WO2006081279A2 (en) * 2005-01-25 2006-08-03 Microchips, Inc. Control of drug release by transient modification of local microenvironments
US7298009B2 (en) * 2005-02-01 2007-11-20 Infineon Technologies Ag Semiconductor method and device with mixed orientation substrate
JP4214124B2 (en) * 2005-03-14 2009-01-28 株式会社バイオエコーネット Ear thermometer
US7622079B2 (en) * 2005-03-22 2009-11-24 Applied Biosystems, Llc Dual nest microplate spotter
US20070020650A1 (en) * 2005-04-01 2007-01-25 Avak Kahvejian Methods for detecting proteins
US20060246576A1 (en) 2005-04-06 2006-11-02 Affymetrix, Inc. Fluidic system and method for processing biological microarrays in personal instrumentation
EP1888780A4 (en) 2005-05-06 2009-11-11 Applera Corp Multiple capillary device and method for synthesis and dispensing
US7738086B2 (en) * 2005-05-09 2010-06-15 The Trustees Of Columbia University In The City Of New York Active CMOS biosensor chip for fluorescent-based detection
US20070097364A1 (en) * 2005-05-09 2007-05-03 The Trustees Of Columbia University In The City Of New York Active CMOS biosensor chip for fluorescent-based detection
AU2006309284B2 (en) 2005-05-31 2012-08-02 Board Of Regents, The University Of Texas System Methods and compositions related to determination and use of white blood cell counts
US8486629B2 (en) 2005-06-01 2013-07-16 Bioarray Solutions, Ltd. Creation of functionalized microparticle libraries by oligonucleotide ligation or elongation
CA2627360C (en) 2005-10-29 2014-06-10 Bayer Technology Services Gmbh Process for determining one or more analytes in samples of biological origin having complex composition, and use thereof
GB2432217A (en) * 2005-11-09 2007-05-16 Seiko Epson Corp Application of biosensor chips
US20080309701A1 (en) * 2005-11-28 2008-12-18 Koninklijke Philips Electronics, N.V. Ink Jet Device for Releasing Controllably a Plurality of Substances Onto a Substrate, Method of Discrimination Between a Plurality of Substances and Use of an Ink Jet Device
KR101157176B1 (en) * 2005-12-20 2012-06-20 삼성전자주식회사 Microfluidic device and method for concentration or purification of cells or viruses
US7537934B2 (en) * 2005-12-30 2009-05-26 Intel Corporation Chemiluminescence sensor array
SG176453A1 (en) * 2006-01-03 2011-12-29 Harvard College Small molecule printing
US20090176208A1 (en) * 2006-01-04 2009-07-09 Simon Brodie Methods for detecting, identifying and reporting the presence of animal pathological agents
US20090305238A1 (en) * 2006-01-23 2009-12-10 Applera Corporation Microarray Microcard
US9445025B2 (en) 2006-01-27 2016-09-13 Affymetrix, Inc. System, method, and product for imaging probe arrays with small feature sizes
US8055098B2 (en) 2006-01-27 2011-11-08 Affymetrix, Inc. System, method, and product for imaging probe arrays with small feature sizes
US8261598B2 (en) * 2006-03-09 2012-09-11 Agency For Science, Technology And Research Apparatus for performing a reaction in a droplet and method of using the same
EP1996716B1 (en) 2006-03-20 2011-05-11 The Regents of the University of California Engineered anti-prostate stem cell antigen (psca) antibodies for cancer targeting
WO2008054480A2 (en) * 2006-03-22 2008-05-08 Northrop Grumman Corporation Enhanced biohazard detection system
US20080076189A1 (en) * 2006-03-30 2008-03-27 Visigen Biotechnologies, Inc. Modified surfaces for the detection of biomolecules at the single molecule level
US11001881B2 (en) 2006-08-24 2021-05-11 California Institute Of Technology Methods for detecting analytes
US11098345B2 (en) * 2006-06-05 2021-08-24 California Institute Of Technology Methods for detecting target analytes
WO2007143669A2 (en) 2006-06-05 2007-12-13 California Institute Of Technology Real time micro arrays
US8637436B2 (en) 2006-08-24 2014-01-28 California Institute Of Technology Integrated semiconductor bioarray
EP2032246A1 (en) * 2006-06-13 2009-03-11 Koninklijke Philips Electronics N.V. Ink jet device for producing a biological assay substrate by releasing a plurality of substances onto the substrate, and method for monitoring the ink jet device
JP2008020412A (en) * 2006-07-14 2008-01-31 Sony Corp Bioassay-use substrate and bioassay method
US8048626B2 (en) 2006-07-28 2011-11-01 California Institute Of Technology Multiplex Q-PCR arrays
US11525156B2 (en) * 2006-07-28 2022-12-13 California Institute Of Technology Multiplex Q-PCR arrays
TWI316873B (en) * 2006-08-08 2009-11-11 Ind Tech Res Inst Apparatus of manufacturing biochip and manufacture method of biochip tip array
US11560588B2 (en) * 2006-08-24 2023-01-24 California Institute Of Technology Multiplex Q-PCR arrays
CA2663601C (en) 2006-09-22 2014-11-25 Dharmacon, Inc. Duplex oligonucleotide complexes and methods for gene silencing by rna interference
EP2084517A1 (en) 2006-11-23 2009-08-05 Nanoident Technologies AG Device for optoelectronically characterizing samples
US9874501B2 (en) 2006-11-24 2018-01-23 Curiox Biosystems Pte Ltd. Use of chemically patterned substrate for liquid handling, chemical and biological reactions
WO2008063135A1 (en) 2006-11-24 2008-05-29 Agency For Science, Technology And Research Apparatus for processing a sample in a liquid droplet and method of using the same
JP2008241447A (en) * 2007-03-27 2008-10-09 Fujifilm Corp Apparatus and method for reading image
EP2140275B1 (en) * 2007-05-02 2017-12-20 Siemens Healthcare Diagnostics Inc. Piezo dispensing of a diagnostic liquid into microfluidic devices
WO2008137213A1 (en) * 2007-05-02 2008-11-13 Siemens Healthcare Diagnostics Inc. Piezo dispensing of a diagnostic liquid onto a reagent surface
EP2157907B1 (en) * 2007-06-07 2013-03-27 MicroCHIPS, Inc. Electrochemical biosensors and arrays
FR2917090B1 (en) * 2007-06-11 2012-06-15 Biomerieux Sa MARKING REAGENTS HAVING DIAZO AND NITRO FUNCTIONS, METHODS FOR SYNTHESIZING SUCH REAGENTS AND METHODS FOR DETECTING BIOLOGICAL MOLECULES
ES2567094T3 (en) 2007-07-12 2016-04-19 The Brigham And Women's Hospital, Inc. Compositions and methods to diagnose and evaluate inflammatory myopathies
JP6126773B2 (en) 2007-09-04 2017-05-10 ザ リージェンツ オブ ザ ユニバーシティ オブ カリフォルニア High affinity anti-prostatic stem cell antigen (PSCA) antibody for cancer targeting and detection
US20090121476A1 (en) * 2007-11-08 2009-05-14 The Government Of The Us, As Represented By The Secretary Of The Navy Microfluidic Bus for Interconnecting Multiple Fluid Conduits
US10725020B2 (en) 2007-11-14 2020-07-28 Curiox Biosystems Pte Ltd. High throughput miniaturized assay system and methods
WO2013114217A1 (en) 2012-02-05 2013-08-08 Curiox Biosystems Pte Ltd. Array plates and methods for making and using same
US8811763B2 (en) * 2007-12-06 2014-08-19 The United States Of America As Represented By The Secretary Of The Army Method and system for producing image frames using quantum properties
US8707897B1 (en) * 2007-12-28 2014-04-29 Intermolecular, Inc. Combinatorial processing chamber having electrical contacts
US8808625B2 (en) * 2008-01-11 2014-08-19 National Institute Of Advanced Industrial Science And Technology Dispensing apparatus and a dispensing method
US8188060B2 (en) 2008-02-11 2012-05-29 Dharmacon, Inc. Duplex oligonucleotides with enhanced functionality in gene regulation
FR2932885B1 (en) 2008-06-24 2010-08-27 Genewave METHOD AND DEVICE FOR DETECTING THE FLUORESCENCE OF A BIOPUCE
FR2934595B1 (en) * 2008-07-29 2013-04-05 Biomerieux Sa MARKING REAGENTS HAVING A PYRIDINE CORE HAVING DIAZOMETHYL FUNCTION, METHODS FOR SYNTHESIZING SUCH REAGENTS AND METHODS FOR DETECTING BIOLOGICAL MOLECULES
US20100069616A1 (en) * 2008-08-06 2010-03-18 The Regents Of The University Of California Engineered antibody-nanoparticle conjugates
JP2012501681A (en) 2008-09-12 2012-01-26 ジェンボールト コーポレイション Matrix and media for storage and stabilization of biomolecules
WO2010080822A1 (en) * 2009-01-06 2010-07-15 1366 Technologies Inc. Dispensing liquid containing material to patterned surfaces using a dispensing tube
ES2712732T3 (en) 2009-02-17 2019-05-14 Cornell Res Foundation Inc Methods and kits for the diagnosis of cancer and the prediction of therapeutic value
US20100216225A1 (en) * 2009-02-25 2010-08-26 Ag-Defense Systems, Inc. Portable microorganism detection unit
JP2010237074A (en) * 2009-03-31 2010-10-21 Toyama Univ Contact dispensing method of low surface tension trace solution and device for use in dispensing
US8784752B2 (en) 2009-04-17 2014-07-22 Curiox Biosystems Pte Ltd Use of chemically patterned substrate for liquid handling, chemical and biological reactions
CN101892291A (en) * 2009-05-08 2010-11-24 中国科学院苏州纳米技术与纳米仿生研究所 Universal tag, probe and detection method for biomolecule multi-target detection
US20100313995A1 (en) * 2009-06-15 2010-12-16 Cory Gerdts Robot for orchestrating microfluidics experiments
US9594239B1 (en) * 2009-06-16 2017-03-14 Lester F. Ludwig Optical tomography for microscopy, cell cytometry, microplate array instrumentation, crystallography, and other applications
WO2010148365A2 (en) * 2009-06-19 2010-12-23 The Arizona Board Of Regents, A Body Corporate Of The State Of Arizona For And On Behalf Of Arizona State University Compound arrays for sample profiling
GB0913258D0 (en) 2009-07-29 2009-09-02 Dynex Technologies Inc Reagent dispenser
US9523701B2 (en) 2009-07-29 2016-12-20 Dynex Technologies, Inc. Sample plate systems and methods
FR2951542B1 (en) * 2009-10-16 2011-12-02 Commissariat Energie Atomique METHOD FOR OPTICALLY DETECTING MICROMETRIC OBJECTS IN SOLUTION
EP2494400B1 (en) 2009-10-28 2021-12-08 Alentic Microscience Inc. Microscopy imaging
US9075225B2 (en) 2009-10-28 2015-07-07 Alentic Microscience Inc. Microscopy imaging
EP2496929B1 (en) * 2009-11-05 2013-07-03 Waterford Institute Of Technology A nanohole array biosensor
US9700889B2 (en) 2009-11-23 2017-07-11 Cyvek, Inc. Methods and systems for manufacture of microarray assay systems, conducting microfluidic assays, and monitoring and scanning to obtain microfluidic assay results
WO2011063408A1 (en) 2009-11-23 2011-05-26 Cyvek, Inc. Method and apparatus for performing assays
US9855735B2 (en) 2009-11-23 2018-01-02 Cyvek, Inc. Portable microfluidic assay devices and methods of manufacture and use
WO2013134741A2 (en) 2012-03-08 2013-09-12 Cyvek, Inc. Methods and systems for manufacture of microarray assay systems, conducting microfluidic assays, and monitoring and scanning to obtain microfluidic assay results
US9500645B2 (en) 2009-11-23 2016-11-22 Cyvek, Inc. Micro-tube particles for microfluidic assays and methods of manufacture
US9759718B2 (en) 2009-11-23 2017-09-12 Cyvek, Inc. PDMS membrane-confined nucleic acid and antibody/antigen-functionalized microlength tube capture elements, and systems employing them, and methods of their use
US9651568B2 (en) 2009-11-23 2017-05-16 Cyvek, Inc. Methods and systems for epi-fluorescent monitoring and scanning for microfluidic assays
US10065403B2 (en) 2009-11-23 2018-09-04 Cyvek, Inc. Microfluidic assay assemblies and methods of manufacture
CN102753194B (en) 2009-12-02 2015-07-08 伊麦吉纳博公司 J591 minibodies and cys-diabodies for targeting human prostate specific membrane antigen (psma)
US8501122B2 (en) 2009-12-08 2013-08-06 Affymetrix, Inc. Manufacturing and processing polymer arrays
JP5498772B2 (en) * 2009-12-24 2014-05-21 花王株式会社 Liquid dripping device and liquid dripping prevention method in liquid dripping device
EP2933629B1 (en) 2010-02-19 2019-04-10 Pacific Biosciences Of California, Inc. System for measuring analytical reactions comprising a socket for an optode array chip
WO2011106315A1 (en) * 2010-02-23 2011-09-01 Rheonix, Inc. Self-contained biological assay apparatus, methods, and applications
US9102979B2 (en) * 2010-02-23 2015-08-11 Rheonix, Inc. Self-contained biological assay apparatus, methods, and applications
WO2011158759A1 (en) * 2010-06-16 2011-12-22 矢部川電気工業株式会社 Production device, production method, antibody chip, programme, and recording medium
DE102010024964B4 (en) * 2010-06-24 2012-01-26 Siemens Aktiengesellschaft Cell monitoring by means of scattered light measurement
WO2012011877A2 (en) 2010-07-23 2012-01-26 Curiox Biosystems Pte Ltd Apparatus and method for multiple reactions in small volumes
CN103459611B (en) 2010-09-17 2016-11-02 哈佛大学校长及研究员协会 The functional genomics research that effectiveness and the safety of pluripotent stem cell are characterized
US8651138B2 (en) * 2010-12-02 2014-02-18 The United States Of America, As Represented By The Secretary Of The Navy Tubular array for fluidic focusing with integrated optical access region
JP6087293B2 (en) 2011-01-06 2017-03-01 メソ スケール テクノロジーズ エルエルシー Assay cartridge and method of using the same
CN103649759B (en) 2011-03-22 2016-08-31 西维克公司 Microfluidic device and manufacture method and purposes
US20130025690A1 (en) * 2011-07-29 2013-01-31 Intermolecular, Inc. No-Contact Wet Processing Tool with Liquid Barrier
US9425027B2 (en) * 2011-05-15 2016-08-23 Varian Semiconductor Equipment Associates, Inc. Methods of affecting material properties and applications therefor
EP2737082A1 (en) * 2011-07-27 2014-06-04 Scienion AG Labeled device
WO2013049231A1 (en) 2011-09-28 2013-04-04 Htg Molecular Diagnostics, Inc. Methods of co-detecting mrna and small non-coding rna
WO2013060482A1 (en) * 2011-10-28 2013-05-02 Torsten Matthias Device and method for detecting substances present in biological or chemical samples
US20140155297A1 (en) * 2012-02-24 2014-06-05 Cambrian Genomics, Inc. Method and apparatus for light based recovery of sequence verified dna
US11497681B2 (en) 2012-02-28 2022-11-15 Corning Incorporated Glass articles with low-friction coatings
US10737973B2 (en) 2012-02-28 2020-08-11 Corning Incorporated Pharmaceutical glass coating for achieving particle reduction
MX2014010334A (en) 2012-02-28 2014-09-22 Corning Inc Glass articles with low-friction coatings.
US8906320B1 (en) 2012-04-16 2014-12-09 Illumina, Inc. Biosensors for biological or chemical analysis and systems and methods for same
CA2870716C (en) * 2012-04-18 2021-01-19 David E. Jones Microspotting device
US9217179B2 (en) 2012-05-24 2015-12-22 The Governing Council Of The University Of Toronto Systems and methods for multiplexed electrochemical detection
US9095833B2 (en) * 2012-05-31 2015-08-04 Biolytic Lab Performance, Inc. System for performing automated solid phase extractions
US10273048B2 (en) 2012-06-07 2019-04-30 Corning Incorporated Delamination resistant glass containers with heat-tolerant coatings
US9034442B2 (en) 2012-11-30 2015-05-19 Corning Incorporated Strengthened borosilicate glass containers with improved damage tolerance
FR2993276B1 (en) * 2012-07-12 2015-07-24 Commissariat Energie Atomique COMPOSITION FOR OPTICALLY DETECTING PARTICLES, COMPRISING WATER, PARTICLES, SURFACTANT AND HYDROPHILIC POLYMER, SYSTEM AND METHOD FOR OPTICAL DETECTION THEREOF
SG10201804101XA (en) * 2012-09-10 2018-07-30 Biofire Diagnostics Llc Multiple amplification cycle detection
US20150338401A1 (en) * 2012-11-16 2015-11-26 Ayal Ram Multiplexed bioassay techniques
US10117806B2 (en) 2012-11-30 2018-11-06 Corning Incorporated Strengthened glass containers resistant to delamination and damage
CN103869075A (en) * 2012-12-14 2014-06-18 仓怀兴 Preparation device of protein chip by real-time quantitative detection and capillary electrophoresis
DE102013000682B3 (en) * 2013-01-08 2013-10-31 Rainer Hintsche Detecting molecules, involves immobilizing mixtures of two or more different biochemical capture molecules on two or more analytical array positions, and binding two or more different analytes on each array position
EP2759602A1 (en) 2013-01-25 2014-07-30 Charité - Universitätsmedizin Berlin Non-invasive prenatal genetic diagnostic methods
US9624532B2 (en) 2013-02-05 2017-04-18 Neil Gordon Ultra-sensitive detection of extremely low level biological analytes using electrochemical signal amplification and biosensor
US10502666B2 (en) 2013-02-06 2019-12-10 Alentic Microscience Inc. Sample processing improvements for quantitative microscopy
US9518920B2 (en) 2013-06-26 2016-12-13 Alentic Microscience Inc. Sample processing improvements for microscopy
WO2014138028A1 (en) * 2013-03-04 2014-09-12 The Scripps Research Institute Liquid array platform for multiplexed analysis of molecule-protein interactions
US10436778B2 (en) * 2013-03-15 2019-10-08 Plexbio Co., Ltd. Methods and systems for multiplex assays
WO2014200905A2 (en) 2013-06-10 2014-12-18 President And Fellows Of Harvard College Early developmental genomic assay for characterizing pluripotent stem cell utility and safety
JP2015014507A (en) * 2013-07-04 2015-01-22 株式会社ニコン Dispenser, biochip screening device, dispensing method, and biochip screening method
US9557318B2 (en) 2013-07-09 2017-01-31 Curiox Biosystems Pte Ltd. Array plates for washing samples
US9153024B2 (en) * 2013-08-02 2015-10-06 CRIXlabs, Inc. Method and system for predicting spatial and temporal distributions of therapeutic substance carriers
TWI646230B (en) 2013-08-05 2019-01-01 扭轉生物科技有限公司 Re-synthesized gene bank
US9594019B1 (en) * 2013-08-09 2017-03-14 Lester F. Ludwig Optical tomography for microscopy, cell cytometry, microplate array instrumentation, crystallography, and other applications
EP4220137A1 (en) 2013-12-10 2023-08-02 Illumina, Inc. Biosensors for biological or chemical analysis and methods of manufacturing the same
CA2970734C (en) 2013-12-17 2019-09-24 Alentic Microscience Inc. Dosimeters including lensless imaging systems
US20150173664A1 (en) * 2013-12-20 2015-06-25 Richard C. Fuisz Method, device and kit to improve blood samples size from lancet or needle
US20150174547A1 (en) * 2013-12-20 2015-06-25 Neil Emans Transfection systems, methods and media
US10260111B1 (en) 2014-01-20 2019-04-16 Brett Eric Etchebarne Method of detecting sepsis-related microorganisms and detecting antibiotic-resistant sepsis-related microorganisms in a fluid sample
KR102535489B1 (en) 2014-06-13 2023-05-22 큐-리네아 에이비 Method for detecting and characterising a microorganism
EP3189017B1 (en) 2014-09-05 2022-11-16 Corning Incorporated Glass articles and methods for improving the reliability of glass articles
JP6757316B2 (en) * 2014-10-24 2020-09-16 クアンタポール, インコーポレイテッド Efficient optical analysis of polymers using nanostructured arrays
JP6784671B2 (en) 2014-11-26 2020-11-11 コーニング インコーポレイテッド How to make reinforced and durable glass containers
CN107209120B (en) 2015-02-02 2020-05-15 株式会社日立高新技术 Multicolor fluorescence analysis device
CA2975855A1 (en) 2015-02-04 2016-08-11 Twist Bioscience Corporation Compositions and methods for synthetic gene assembly
US10669304B2 (en) 2015-02-04 2020-06-02 Twist Bioscience Corporation Methods and devices for de novo oligonucleic acid assembly
US9708647B2 (en) 2015-03-23 2017-07-18 Insilixa, Inc. Multiplexed analysis of nucleic acid hybridization thermodynamics using integrated arrays
CN107430144B (en) 2015-04-09 2021-11-09 简·探针公司 Sample testing system and method with automatic cleaning
US9981239B2 (en) 2015-04-21 2018-05-29 Twist Bioscience Corporation Devices and methods for oligonucleic acid library synthesis
GB201507026D0 (en) 2015-04-24 2015-06-10 Linea Ab Q Medical sample transportation container
WO2016181466A1 (en) * 2015-05-11 2016-11-17 株式会社安川電機 Dispensing system, controller, and control method
US10837977B2 (en) 2015-05-11 2020-11-17 Kabushiki Kaisha Yaskawa Denki Rack for dispensing and dispensing system
US10545139B2 (en) 2015-06-16 2020-01-28 Curiox Biosystems Pte Ltd. Methods and devices for performing biological assays using magnetic components
CN104897657A (en) * 2015-06-26 2015-09-09 朱金国 Method for detecting electrochemiluminescence by aid of suspending electrodes and microporous plates
EP3325943B1 (en) * 2015-07-17 2021-05-12 Bio-Rad Laboratories, Inc. Contact imaging devices for fluorescence applications
US9952122B2 (en) * 2015-08-03 2018-04-24 Palo Alto Research Center Incorporated Polysensing bioelectronic test plate
MX2018001566A (en) 2015-08-07 2019-04-25 Imaginab Inc Antigen binding constructs to target molecules.
US9499861B1 (en) 2015-09-10 2016-11-22 Insilixa, Inc. Methods and systems for multiplex quantitative nucleic acid amplification
CN108351349B (en) 2015-09-10 2020-09-29 因斯利克萨公司 Methods and systems for multiplexed quantitative nucleic acid amplification
US10758886B2 (en) 2015-09-14 2020-09-01 Arizona Board Of Regents On Behalf Of Arizona State University Conditioned surfaces for in situ molecular array synthesis
CA2998169A1 (en) 2015-09-18 2017-03-23 Twist Bioscience Corporation Oligonucleic acid variant libraries and synthesis thereof
CN108698012A (en) 2015-09-22 2018-10-23 特韦斯特生物科学公司 Flexible substrates for nucleic acid synthesis
EP3150564B1 (en) 2015-09-30 2018-12-05 Corning Incorporated Halogenated polyimide siloxane chemical compositions and glass articles with halogenated polylmide siloxane low-friction coatings
WO2017075435A1 (en) 2015-10-30 2017-05-04 Corning Incorporated Glass articles with mixed polymer and metal oxide coatings
US10228367B2 (en) 2015-12-01 2019-03-12 ProteinSimple Segmented multi-use automated assay cartridge
US9895673B2 (en) 2015-12-01 2018-02-20 Twist Bioscience Corporation Functionalized surfaces and preparation thereof
US10513745B2 (en) * 2015-12-28 2019-12-24 PathogenDX Inc Microarray based multiplex pathogen analysis and uses thereof
US10501814B2 (en) * 2015-12-28 2019-12-10 PathogenDX Inc Microarray based multiplex pathogen analysis and uses thereof
US10612075B2 (en) * 2015-12-28 2020-04-07 PathogenDX Inc Microarray based multiplex pathogen analysis and uses thereof
WO2017155858A1 (en) 2016-03-07 2017-09-14 Insilixa, Inc. Nucleic acid sequence identification using solid-phase cyclic single base extension
US10883926B2 (en) 2016-04-08 2021-01-05 Universal Bio Research Co., Ltd. General-purpose optical measuring device and method of same
GB2554767A (en) 2016-04-21 2018-04-11 Q Linea Ab Detecting and characterising a microorganism
KR20190019190A (en) 2016-06-20 2019-02-26 헬스텔 인크. Diagnosis and treatment of autoimmune diseases
CN109791151A (en) 2016-06-20 2019-05-21 健康之语公司 The method of antidiastole for autoimmune disease
AU2017315294B2 (en) 2016-08-22 2023-12-21 Twist Bioscience Corporation De novo synthesized nucleic acid libraries
JP6871364B2 (en) 2016-09-21 2021-05-12 ツイスト バイオサイエンス コーポレーション Nucleic acid-based data storage
EP3516370A4 (en) * 2016-09-26 2020-06-03 Azure Biosystems, Inc. Methods and devices for photometric analysis
WO2018089858A1 (en) 2016-11-11 2018-05-17 Healthtell Inc. Methods for identifying candidate biomarkers
GB2573069A (en) 2016-12-16 2019-10-23 Twist Bioscience Corp Variant libraries of the immunological synapse and synthesis thereof
WO2018147960A1 (en) 2017-02-08 2018-08-16 Imaginab, Inc. Extension sequences for diabodies
KR20190119107A (en) 2017-02-22 2019-10-21 트위스트 바이오사이언스 코포레이션 Nucleic Acid Based Data Storage
CA3056388A1 (en) 2017-03-15 2018-09-20 Twist Bioscience Corporation Variant libraries of the immunological synapse and synthesis thereof
EP3607580B1 (en) 2017-04-05 2023-05-31 Curiox Biosystems Pte Ltd. Methods, devices, and apparatus for washing samples on array plates
WO2018231864A1 (en) 2017-06-12 2018-12-20 Twist Bioscience Corporation Methods for seamless nucleic acid assembly
CN111566209A (en) 2017-06-12 2020-08-21 特韦斯特生物科学公司 Seamless nucleic acid assembly method
JP7125986B2 (en) * 2017-08-14 2022-08-25 シグマ-アルドリッチ・カンパニー・リミテッド・ライアビリティ・カンパニー Multi-pin solid-phase microextraction device
CN107764884B (en) * 2017-08-30 2021-06-15 成都安普利菲能源技术有限公司 High-throughput electrochemical detection device and high-throughput electrochemical detection method
JP2020536504A (en) 2017-09-11 2020-12-17 ツイスト バイオサイエンス コーポレーション GPCR-coupled protein and its synthesis
WO2019069372A1 (en) * 2017-10-03 2019-04-11 株式会社ニコン Detection target measurement method, capture probe-anchoring carrier, detection kit, and fluid device
GB2583590A (en) 2017-10-20 2020-11-04 Twist Bioscience Corp Heated nanowells for polynucleotide synthesis
TWI646508B (en) * 2017-11-07 2019-01-01 思創影像科技股份有限公司 Equipment for detecting dna amplifying reaction
EP3724664B1 (en) * 2017-12-15 2023-08-23 Bio-Rad Laboratories, Inc. Combination imaging of assays
US10861829B2 (en) 2017-12-26 2020-12-08 Illumina, Inc. Sensor system
EP3735459A4 (en) 2018-01-04 2021-10-06 Twist Bioscience Corporation Dna-based digital information storage
KR102066119B1 (en) * 2018-02-09 2020-02-17 제노플랜코리아 주식회사 Dispenser mapping apparatus for multi-analysis and the operation method thereof
US10153317B1 (en) 2018-04-26 2018-12-11 Alentic Microscience Inc. Image sensors comprising a chamber to confine a sample at a sensor surface of successive light sensitive subareas and non-light sensitive areas
GB2590196A (en) 2018-05-18 2021-06-23 Twist Bioscience Corp Polynucleotides, reagents, and methods for nucleic acid hybridization
JP2021528068A (en) 2018-06-21 2021-10-21 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft Hybridization of all LNA oligonucleotides
US11543356B2 (en) * 2018-10-29 2023-01-03 Hewlett-Packard Development Company, L.P. Rotation and flat-form imaging for microscopic objects
JP7307793B2 (en) 2018-11-16 2023-07-12 エフ. ホフマン-ラ ロシュ アーゲー Streptavidin-coated solid phase with binding pair members
CN109680270B (en) * 2018-12-28 2020-11-24 河海大学常州校区 Preparation device and preparation method of array electrode made of IPMC material
TW202043486A (en) 2019-01-29 2020-12-01 美商伊路米納有限公司 Sequencing kits
TW202045709A (en) * 2019-01-29 2020-12-16 美商伊路米納有限公司 Flow cells
WO2020176680A1 (en) 2019-02-26 2020-09-03 Twist Bioscience Corporation Variant nucleic acid libraries for antibody optimization
EP3930753A4 (en) 2019-02-26 2023-03-29 Twist Bioscience Corporation Variant nucleic acid libraries for glp1 receptor
JP2022525322A (en) 2019-03-14 2022-05-12 インシリクサ, インコーポレイテッド Methods and systems for time-gate fluorescence-based detection
JP2022535580A (en) 2019-06-07 2022-08-09 エフ.ホフマン-ラ ロシュ アーゲー Hybridization of all LNA oligonucleotides
US11332738B2 (en) 2019-06-21 2022-05-17 Twist Bioscience Corporation Barcode-based nucleic acid sequence assembly
DE102019134846B4 (en) * 2019-12-18 2023-08-17 Hamilton Bonaduz Ag laboratory system
DE102020108432A1 (en) 2020-03-25 2021-09-30 Jenoptik Optical Systems Gmbh Device and method for luminescence analysis of several samples
CN111337468B (en) * 2020-04-24 2022-12-09 新乡医学院 Multi-analysis snake venom mixture analysis fluorescent sensor
GB2599162A (en) * 2020-09-29 2022-03-30 Ffei Ltd Cell deposition and imaging apparatus
WO2022256244A1 (en) * 2021-06-04 2022-12-08 Nutcracker Therapeutics, Inc. Systems and methods to communicate fluids
NL2028535B1 (en) * 2021-06-04 2022-12-15 Nutcracker Therapeutics Inc Systems and methods to communicate fluids
GB2607603A (en) * 2021-06-08 2022-12-14 Spectrum Logic Ltd Imaging apparatus

Family Cites Families (122)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2512743A (en) 1946-04-01 1950-06-27 Rca Corp Jet sprayer actuated by supersonic waves
LU77642A1 (en) * 1976-07-09 1977-10-03
US4228237A (en) 1978-09-21 1980-10-14 Calbiochem-Behring Corp. Methods for the detection and determination of ligands
US4280815A (en) * 1979-06-18 1981-07-28 Technicon Instruments Corporation Electrochemiluminescent immunoassay and apparatus therefor
DE3269567D1 (en) 1981-04-29 1986-04-10 Ciba Geigy Ag New devices and kits for immunological analysis
FI63596C (en) 1981-10-16 1983-07-11 Orion Yhtymae Oy MICROBIA DIAGNOSIS FOERFARANDE SOM GRUNDAR SIG PAO SKIKTSHYBRIDISERING AV NUCLEINSYROR OCH VID FOERFARANDET ANVAENDA KOMBINATIONER AV REAGENSER
US5241060A (en) 1982-06-23 1993-08-31 Enzo Diagnostics, Inc. Base moiety-labeled detectable nucleatide
US4994373A (en) 1983-01-27 1991-02-19 Enzo Biochem, Inc. Method and structures employing chemically-labelled polynucleotide probes
GB8405437D0 (en) 1984-03-01 1984-04-04 Amersham Int Plc Detecting polynucleotide sequences
DE3410508C2 (en) * 1984-03-22 1986-06-26 Kernforschungsanlage Jülich GmbH, 5170 Jülich Serial filling device for filling the cups of a micro cup plate
FI68420C (en) 1984-03-29 1985-09-10 Rainer Jalo Kalevi Kouvunen SPAENNBRAEDES IN SKYDDSPAOS
US5288609A (en) 1984-04-27 1994-02-22 Enzo Diagnostics, Inc. Capture sandwich hybridization method and composition
EP0190205B1 (en) * 1984-07-24 1992-10-21 Coselco Mimotopes Pty. Ltd. Method for determining mimotopes
US5221605A (en) * 1984-10-31 1993-06-22 Igen, Inc. Luminescent metal chelate labels and means for detection
US5310687A (en) * 1984-10-31 1994-05-10 Igen, Inc. Luminescent metal chelate labels and means for detection
US4751177A (en) 1985-06-13 1988-06-14 Amgen Methods and kits for performing nucleic acid hybridization assays
US4868105A (en) 1985-12-11 1989-09-19 Chiron Corporation Solution phase nucleic acid sandwich assay
US4925785A (en) 1986-03-07 1990-05-15 Biotechnica Diagnostics, Inc. Nucleic acid hybridization assays
US4851331A (en) * 1986-05-16 1989-07-25 Allied Corporation Method and kit for polynucleotide assay including primer-dependant DNA polymerase
US5175270A (en) 1986-09-10 1992-12-29 Polyprobe, Inc. Reagents for detecting and assaying nucleic acid sequences
US4844298A (en) * 1987-03-11 1989-07-04 Sumitomo Electric Industries, Ltd. Apparatus for automatically dispensing accurately a predetermined quantity of liquids required to be stored at constant temperatures
US5935779A (en) 1988-11-03 1999-08-10 Igen International Inc. Methods for improved particle electrochemiluminescence assay
WO1989000725A1 (en) * 1987-07-16 1989-01-26 Cavro Scientific Instruments, Inc. Xyz positioner
US5093268A (en) 1988-04-28 1992-03-03 Igen, Inc. Apparatus for conducting a plurality of simultaneous measurements of electrochemiluminescent phenomena
US5700637A (en) 1988-05-03 1997-12-23 Isis Innovation Limited Apparatus and method for analyzing polynucleotide sequences and method of generating oligonucleotide arrays
US6054270A (en) 1988-05-03 2000-04-25 Oxford Gene Technology Limited Analying polynucleotide sequences
US5374524A (en) 1988-05-10 1994-12-20 E. I. Du Pont De Nemours And Company Solution sandwich hybridization, capture and detection of amplified nucleic acids
JP2897959B2 (en) 1988-05-20 1999-05-31 エフ.ホフマン―ラ ロシュ アクチェンゲゼルシャフト Immobilized sequence-specific probe
US4883760A (en) * 1988-06-20 1989-11-28 Adi Diagnostics Inc. Device for performing enzyme immunoassays
US5188963A (en) 1989-11-17 1993-02-23 Gene Tec Corporation Device for processing biological specimens for analysis of nucleic acids
US5068088A (en) 1988-11-03 1991-11-26 Igen, Inc. Method and apparatus for conducting electrochemiluminescent measurements
US5061445A (en) 1988-11-03 1991-10-29 Igen, Inc. Apparatus for conducting measurements of electrochemiluminescent phenomena
US5063081A (en) 1988-11-14 1991-11-05 I-Stat Corporation Method of manufacturing a plurality of uniform microfabricated sensing devices having an immobilized ligand receptor
US5547839A (en) 1989-06-07 1996-08-20 Affymax Technologies N.V. Sequencing of surface immobilized polymers utilizing microflourescence detection
US5527681A (en) * 1989-06-07 1996-06-18 Affymax Technologies N.V. Immobilized molecular synthesis of systematically substituted compounds
US6040138A (en) 1995-09-15 2000-03-21 Affymetrix, Inc. Expression monitoring by hybridization to high density oligonucleotide arrays
US5143854A (en) * 1989-06-07 1992-09-01 Affymax Technologies N.V. Large scale photolithographic solid phase synthesis of polypeptides and receptor binding screening thereof
US5744101A (en) 1989-06-07 1998-04-28 Affymax Technologies N.V. Photolabile nucleoside protecting groups
US5262128A (en) * 1989-10-23 1993-11-16 The United States Of America As Represented By The Department Of Health And Human Services Array-type multiple cell injector
US5252743A (en) * 1989-11-13 1993-10-12 Affymax Technologies N.V. Spatially-addressable immobilization of anti-ligands on surfaces
ES2103795T3 (en) 1989-12-04 1997-10-01 Microprobe Corp IMPROVEMENT OF THE CAPTURE OF A NUCLEIC ACID DIANA THROUGH THE USE OF UNITED OLIGONUCLEOTIDES COVALENTLY TO POLYMERS.
US5212295A (en) 1990-01-11 1993-05-18 Isis Pharmaceuticals Monomers for preparation of oligonucleotides having chiral phosphorus linkages
AU7762091A (en) 1990-03-30 1991-10-30 City Of Hope Detection of minimal residual disease in lymphoid malignancies
DE4024544A1 (en) * 1990-08-02 1992-02-06 Boehringer Mannheim Gmbh ANALYZING ELEMENT AND METHOD FOR THE PRODUCTION THEREOF
DK0558671T3 (en) * 1990-11-21 1999-09-13 Iterex Pharma Lp Synthesis of equimolar multiple oligomer mixtures, especially of oligopeptide mixtures
US5525302A (en) * 1991-02-01 1996-06-11 Astle; Thomas W. Method and device for simultaneously transferring plural samples
IT1252715B (en) * 1991-06-13 1995-06-26 Nevio Recinelli MEANS FOR DETERMINING THE PRESENCE OF TOTAL OR DIRECT IMMUNOGLOBULINS AGAINST ALLERGENS OR OTHER MOLECULES (ANTIBODIES OR ALLERGENS) IN AQUEOUS SAMPLES IN A COMPLETELY AUTOMATIC WAY
US5422241A (en) 1991-07-03 1995-06-06 Ambion, Inc. Methods for the recovery of nucleic acids from reaction mixtures
US5226462A (en) * 1991-07-26 1993-07-13 Carl Richard A Introducing measured amounts of liquid into receptacles
US5556748A (en) 1991-07-30 1996-09-17 Xenopore Corporation Methods of sandwich hybridization for the quantitative analysis of oligonucleotides
US5449754A (en) 1991-08-07 1995-09-12 H & N Instruments, Inc. Generation of combinatorial libraries
US5474796A (en) 1991-09-04 1995-12-12 Protogene Laboratories, Inc. Method and apparatus for conducting an array of chemical reactions on a support surface
EP0534640B1 (en) 1991-09-23 1996-10-02 Pfizer Inc. Process for detecting specific mRNA and DNA in cells
WO1993009245A1 (en) 1991-10-31 1993-05-13 University Of Pittsburgh Reverse dot blot hybridization using tandem head-to-tail monomers containing probes synthesized by staggered complementary primers
US5605662A (en) * 1993-11-01 1997-02-25 Nanogen, Inc. Active programmable electronic devices for molecular biological analysis and diagnostics
US6051380A (en) * 1993-11-01 2000-04-18 Nanogen, Inc. Methods and procedures for molecular biological analysis and diagnostics
JP3509859B2 (en) 1991-11-07 2004-03-22 ナノトロニクス,インコーポレイテッド Hybridization of chromophore and fluorophore conjugated polynucleotides to create donor-donor energy transfer systems
IL103674A0 (en) * 1991-11-19 1993-04-04 Houston Advanced Res Center Method and apparatus for molecule detection
US5846708A (en) 1991-11-19 1998-12-08 Massachusetts Institiute Of Technology Optical and electrical methods and apparatus for molecule detection
US5324633A (en) 1991-11-22 1994-06-28 Affymax Technologies N.V. Method and apparatus for measuring binding affinity
WO1993009668A1 (en) 1991-11-22 1993-05-27 Affymax Technology N.V. Combinatorial strategies for polymer synthesis
US5310469A (en) 1991-12-31 1994-05-10 Abbott Laboratories Biosensor with a membrane containing biologically active material
US5376313A (en) 1992-03-27 1994-12-27 Abbott Laboratories Injection molding a plastic assay cuvette having low birefringence
US5470705A (en) 1992-04-03 1995-11-28 Applied Biosystems, Inc. Probe composition containing a binding domain and polymer chain and methods of use
CA2115342C (en) 1992-06-17 2003-08-26 Robert B. Wallace A method of detecting and discriminating between nucleic acid sequences
US5795714A (en) * 1992-11-06 1998-08-18 Trustees Of Boston University Method for replicating an array of nucleic acid probes
US5556749A (en) * 1992-11-12 1996-09-17 Hitachi Chemical Research Center, Inc. Oligoprobe designstation: a computerized method for designing optimal DNA probes
EP1197568B1 (en) 1992-11-27 2011-04-27 Innogenetics N.V. Detection and typing of HCV using 5'UTR and NS5 nucleic acid sequences
US5605798A (en) 1993-01-07 1997-02-25 Sequenom, Inc. DNA diagnostic based on mass spectrometry
JP3197277B2 (en) 1993-05-10 2001-08-13 日水製薬株式会社 Method for analyzing one or more immunological ligands, analytical reagents and kits
US5834256A (en) 1993-06-11 1998-11-10 Cell Genesys, Inc. Method for production of high titer virus and high efficiency retroviral mediated transduction of mammalian cells
US5650274A (en) * 1993-06-25 1997-07-22 Hitachi, Ltd. DNA analyzing method
GB9321650D0 (en) * 1993-10-20 1993-12-08 Genome Research Limited Improvements in or relating to electrophoresis
US5472672A (en) 1993-10-22 1995-12-05 The Board Of Trustees Of The Leland Stanford Junior University Apparatus and method for polymer synthesis using arrays
US6156501A (en) 1993-10-26 2000-12-05 Affymetrix, Inc. Arrays of modified nucleic acid probes and methods of use
ATE424927T1 (en) * 1993-10-28 2009-03-15 Houston Advanced Res Ct MICROFABRICATED POROUS FLOW DEVICE FOR DISCRETE DETERMINATION OF BONDING REACTIONS
DK145493D0 (en) * 1993-12-23 1993-12-23 Dako As ANTIBODY
US5561043A (en) 1994-01-31 1996-10-01 Trustees Of Boston University Self-assembling multimeric nucleic acid constructs
DE19518217A1 (en) 1994-05-11 1995-11-30 Imtec Immundiagnostika Gmbh Determn of DNA after capture by probe immobilised on solid phase
EP0695941B1 (en) 1994-06-08 2002-07-31 Affymetrix, Inc. Method and apparatus for packaging a chip
US5807522A (en) * 1994-06-17 1998-09-15 The Board Of Trustees Of The Leland Stanford Junior University Methods for fabricating microarrays of biological samples
JP2771900B2 (en) * 1994-07-25 1998-07-02 ベーリンガー マンハイム ゲーエムベーハー Metal complexes containing charged linkers
US6060288A (en) * 1994-08-03 2000-05-09 Mosaic Technologies Method for performing amplification of nucleic acid on supports
US6121048A (en) 1994-10-18 2000-09-19 Zaffaroni; Alejandro C. Method of conducting a plurality of reactions
WO1997027317A1 (en) 1996-01-23 1997-07-31 Affymetrix, Inc. Nucleic acid analysis techniques
US6974666B1 (en) 1994-10-21 2005-12-13 Appymetric, Inc. Methods of enzymatic discrimination enhancement and surface-bound double-stranded DNA
US5556752A (en) 1994-10-24 1996-09-17 Affymetrix, Inc. Surface-bound, unimolecular, double-stranded DNA
JP3481705B2 (en) * 1994-12-12 2003-12-22 株式会社モリテックス Automatic solid-phase extraction device
JPH08233710A (en) * 1995-02-24 1996-09-13 Hitachi Ltd Sample preparation apparatus
US5599695A (en) 1995-02-27 1997-02-04 Affymetrix, Inc. Printing molecular library arrays using deprotection agents solely in the vapor phase
BR9607193B1 (en) * 1995-03-10 2009-01-13 multispecific multispecific electrochemical test.
DE19516196A1 (en) 1995-05-08 1996-11-14 Boehringer Mannheim Gmbh Method for the quantitative detection of nucleic acids
US5753439A (en) * 1995-05-19 1998-05-19 Trustees Of Boston University Nucleic acid detection methods
US5545531A (en) * 1995-06-07 1996-08-13 Affymax Technologies N.V. Methods for making a device for concurrently processing multiple biological chip assays
US5676472A (en) 1995-07-10 1997-10-14 Smart Machines Rotary labyrinth seal
FR2737502B1 (en) 1995-07-31 1997-10-24 Genset Sa NUCLEIC ACID DETECTION METHOD USING NUCLEOTIDE PROBES ALLOWING BOTH SPECIFIC CAPTURE AND DETECTION
WO1997007245A1 (en) 1995-08-14 1997-02-27 Ely Michael Rabani Methods and devices for parallel multiplex polynucleotide sequencing
US5843655A (en) 1995-09-18 1998-12-01 Affymetrix, Inc. Methods for testing oligonucleotide arrays
US5661028A (en) 1995-09-29 1997-08-26 Lockheed Martin Energy Systems, Inc. Large scale DNA microsequencing device
FR2739935B1 (en) * 1995-10-12 1997-12-19 Genomic Sa DEVICE FOR TRANSFERRING MICRO QUANTITY SAMPLES OF LIQUIDS
US5763263A (en) * 1995-11-27 1998-06-09 Dehlinger; Peter J. Method and apparatus for producing position addressable combinatorial libraries
US6051377A (en) * 1995-11-30 2000-04-18 Pharmaseq, Inc. Multiplex assay for nucleic acids employing transponders
EP2332958B1 (en) 1996-02-09 2016-04-20 Cornell Research Foundation, Inc. Detection of nucleic and sequence differences using the ligase detection reaction with addressable arrays
US5849598A (en) * 1996-03-15 1998-12-15 Washington University Method for transferring micro quantities of liquid samples to discrete locations
EP0912761A4 (en) 1996-05-29 2004-06-09 Cornell Res Foundation Inc Detection of nucleic acid sequence differences using coupled ligase detection and polymerase chain reactions
ATE408824T1 (en) * 1996-05-30 2008-10-15 Cellomics Inc MINIATURIZED CELL ARRANGEMENT AND METHOD AND DEVICE FOR SCREENING USING CELLS
AU3568897A (en) * 1996-06-07 1998-01-05 Eos Biotechnology, Inc. Immobilised linear oligonucleotide arrays
US5770370A (en) 1996-06-14 1998-06-23 David Sarnoff Research Center, Inc. Nuclease protection assays
US5804384A (en) 1996-12-06 1998-09-08 Vysis, Inc. Devices and methods for detecting multiple analytes in samples
US5942397A (en) 1996-12-11 1999-08-24 Tarlov; Michael J. Surface immobilization of biopolymers
US5877580A (en) * 1996-12-23 1999-03-02 Regents Of The University Of California Micromachined chemical jet dispenser
AU6646398A (en) * 1996-12-31 1998-07-31 Genometrix Incorporated Multiplexed molecular analysis apparatus and method
GB2324370B (en) 1997-04-14 1999-03-03 Stuart Harbron Detection of hybrid double-stranded DNA with antibody after enzyme degradation of excess single-standed DNA
WO1999028494A1 (en) 1997-12-04 1999-06-10 Packard Bioscience Company Methods of using probes for analyzing polynucleotide sequence
US6238869B1 (en) 1997-12-19 2001-05-29 High Throughput Genomics, Inc. High throughput assay system
US20030096232A1 (en) 1997-12-19 2003-05-22 Kris Richard M. High throughput assay system
US6458533B1 (en) 1997-12-19 2002-10-01 High Throughput Genomics, Inc. High throughput assay system for monitoring ESTs
US6232066B1 (en) 1997-12-19 2001-05-15 Neogen, Inc. High throughput assay system
US6033911A (en) * 1998-02-27 2000-03-07 Hamilton Company Automated assaying device
TW382789B (en) * 1998-04-22 2000-02-21 United Microelectronics Corp Method for manufacturing CMOS
AU2375600A (en) 1998-12-22 2000-07-12 Stephen Felder High throughput assay system using mass spectrometry
US6200781B1 (en) 1999-06-25 2001-03-13 Integrated Genetic Devices, Ltd. Apparatus, system and method for automated execution and analysis of biological and chemical reactions

Also Published As

Publication number Publication date
EP0990142A4 (en) 2000-09-27
JP2011050390A (en) 2011-03-17
AU6646398A (en) 1998-07-31
EP1249705A2 (en) 2002-10-16
EP0990142A1 (en) 2000-04-05
JP4663824B2 (en) 2011-04-06
US6479301B1 (en) 2002-11-12
US6312960B1 (en) 2001-11-06
US6803238B1 (en) 2004-10-12
CA2389358A1 (en) 1998-07-09
EP2295988A2 (en) 2011-03-16
EP1249705A3 (en) 2003-11-05
WO1998029736A1 (en) 1998-07-09
US6083763A (en) 2000-07-04
US7413852B2 (en) 2008-08-19
US6331441B1 (en) 2001-12-18
US20090239759A1 (en) 2009-09-24
JP4334823B2 (en) 2009-09-30
JP2001510339A (en) 2001-07-31
JP2009002956A (en) 2009-01-08
CA2276462A1 (en) 1998-07-09
US20040023249A1 (en) 2004-02-05
JP2003107097A (en) 2003-04-09
CA2276462C (en) 2007-06-12
JP2010078605A (en) 2010-04-08

Similar Documents

Publication Publication Date Title
CA2389358C (en) Multiplexed molecular analysis apparatus and method
US5874219A (en) Methods for concurrently processing multiple biological chip assays
US8273304B2 (en) Analytical biochemistry system with robotically carried bioarray
US6720149B1 (en) Methods for concurrently processing multiple biological chip assays
US6620625B2 (en) Ultra high throughput sampling and analysis systems and methods
US6929944B2 (en) Analysis using a distributed sample
US20050287590A1 (en) Multi-functional microarrays and methods
EP1073903B1 (en) Detection of very low quantities of analyte bound to a solid phase
EP1718411A1 (en) A device for analysing an interaction between target and probe molecules
US20070141576A1 (en) Biological chip and use thereof
JPWO2005056145A1 (en) Biological chips and uses thereof

Legal Events

Date Code Title Description
EEER Examination request
MKLA Lapsed

Effective date: 20180102